Uploaded by Jorge Enrique Lara Camargo

CHRONIC KIDNEY DISEASE AND PROGRESSION

advertisement
Volume 14 • Number 4 • October 2015
NephSAP
®
Nephrology Self-Assessment Program
Chronic Kidney Disease
and Progression
Co-Editors:
Michael J. Choi, MD
Linda F. Fried, MD
EDUCATION DIRECTOR,
NephSAP
Gerald A. Hladik, MD
University of North Carolina at Chapel Hill
Chapel Hill, NC
DEPUTY EDUCATION DIRECTOR,
NephSAP
Jerry Yee, MD, FASN
Henry Ford Hospital
Detroit, MI
MANAGING EDITOR
Gisela Deuter, BSN, MSA
Washington, DC
ASSOCIATE EDITORS
Michael J. Choi, MD
Johns Hopkins University School of Medicine
Baltimore, MD
Debbie L. Cohen, MD
University of Pennsylvania School of Medicine
Philadelphia, PA
Linda F. Fried, MD, MPH
University of Pittsburgh
Pittsburgh, PA
Richard J. Glassock, MD
Professor Emeritus, The David Geffen School of
Medicine at the University of California
Los Angeles, CA
Stanley Goldfarb, MD
University of Pennsylvania School of Medicine
Philadelphia, PA
Ruediger W. Lehrich, MD
Duke University
Durham, NC
Kevin J. Martin, MBBCh
St. Louis University School of Medicine
St. Louis, MO
John P. Middleton, MD
Duke University
Durham, NC
Patrick T. Murray, MD
University College Dublin
Dublin, Ireland
Aldo J. Peixoto, MD
Yale University
West Haven, CT
Preface
NephSAPÒ is one of the premiere educational activities of the American Society of
Nephrology (ASN). Its primary goals are self-assessment, education, and the provision of
Continuing Medical Education (CME) credits and Maintenance of Certification (MOC) points
for individuals certified by the American Board of Internal Medicine. Members of the ASN
receive NephSAP electronically through the ASN website by clicking on the NephSAP link
under “Education and Meetings” tab.
EDUCATION: Medical and nephrologic information continually accrues at a rapid pace.
Bombarded from all sides with demands on their time, busy practitioners, academicians, and
trainees at all levels are increasingly challenged to review and understand new and evolving
evidence. Each bimonthly issue of NephSAP is dedicated to a specific theme, i.e., to a specific
area of clinical nephrology, hypertension, dialysis, and transplantation, and consists of an
editorial, a syllabus, and self-assessment questions, to serve as a self-study device. Over the
course of 24 months, all clinically relevant and key elements of nephrology will be reviewed
and updated. The authors of each issue digest, assimilate, and interpret key studies published
since the release of the previous issues and integrate this new material with the body of
existing information. Occasionally a special edition is produced to cover an area not ordinarily
addressed by core issues of NephSAP.
SELF-ASSESSMENT: Thirty, single-best-answer questions will follow the 80 to 100 pages of
syllabus text. The examination is available online with immediate feedback. Those answering
75% correctly will receive MOC and CME credit, and receive the answers to all the questions
along with brief discussions and an updated bibliography. Members will find a new area
reviewed every 2 months, and they will be able to test their understanding with our quiz. This
format will help readers stay up to date in developing areas of clinical nephrology,
hypertension, dialysis, and transplantation, and the review and update will support those
taking certification and recertification examinations.
CONTINUING MEDICAL EDUCATION: Most state and local medical agencies as well as
hospitals are demanding documentation of requisite CME credits for licensure and for staff
appointments. A maximum of 48 credits annually can be obtained by successfully completing
the NephSAP examinations. In addition, individuals enrolled in Maintenance of Certification
(MOC) through the American Board of Internal Medicine may obtain points toward MOC by
successfully completing the self-assessment examination of NephSAP.
N This paper meets the requirements of ANSI/NISO Z39.48-1921 (Permanence of Paper),
effective with July 2002, Vol. 1, No. 1.
Asghar Rastegar, MD
Yale University
New Haven, CT
Brad H. Rovin, MD
Ohio State University Medical Center
Columbus, OH
Manoocher Soleimani, MD
University of Cincinnati
Cincinnati, OH
Charuhas V. Thakar, MD
University of Cincinnati
Cincinnati, OH
John P. Vella, MD
Maine Medical Center
Portland, ME
Alexander C. Wiseman, MD
University of Colorado at Denver
Denver, CO
FOUNDING EDITORS
Richard J. Glassock, MD
Editor-in-Chief Emeritus
Robert G. Narins, MD
CONTRIBUTING AUTHOR FOR THIS ISSUE
Sumeska Thavarajah, MD
Johns Hopkins University School of Medicine
Baltimore, MD
NephSAPÒ
Ó2015 by The American Society of Nephrology
Volume 14, Number 4, October 2015
283
Editorial
317
Volume Overload
Obesity and Kidney Disease
317
Vascular Disease
Holly Kramer, Karen Griffin
318
Pulsatility and Pulse Pressure
318
Retinopathy
318
Preexisting CVD
318
Hypoxemia
319
Hyperuricemia/Allopurinol
319
Pentoxifylline in Diabetes
320
Microbiome
320
Prediction Models
321
Conclusions
Syllabus
294
NephSAP, Volume 14, Number 4, October 2015—
CKD and Progression
Michael Choi, Linda Fried, Sumeska Thavarajah
294
Learning Objectives
294
CKD Surveillance and Geographic Variability
294
CKD Prevalence and Lifetime Risk in the United
States
295
CKD Screening and Monitoring in the United
States
324
Diabetic Kidney Disease and Kidney Biopsy
298
Worldwide Surveillance
325
Renin-Angiotensin-Aldosterone System Blockers
298
Mesoamerican Nephropathy
329
Cardiovascular Disease
300
301
Issues in GFR and Albuminuria Estimation
Specific Populations
301
Patients with Diabetes and High Normal GFR
302
Asian Populations
303
Pediatrics
303
Severe Obesity
304
Advanced CKD
304
Elderly
304
305
GFR and Albumin Excretion Variability
Risk Factors for Progression of Kidney Disease
329
Coronary Plaque Characteristics in CKD
330
Association of Kidney Disease with CVD in Type 2
Diabetes
330
Interaction of Age and CKD on Mortality and
Coronary Event
331
Lipid Guidelines
333
Cardiac Biomarkers
334
Inflammatory Markers and the Microbiome
335
Urine Biomarkers
337
Echocardiographic Changes in CKD
337
Percutaneous Cardiovascular Intervention and
Drug-Eluting Stents
305
Biomarkers
306
Genetics
338
Atrial Fibrillation
306
Obesity
339
Cardioverter Defibrillator
310
Diet
341
Exercise in CKD
310
Fluid
343
Depression in CKD
310
Dietary Patterns
344
CKD and Safety
310
Protein Intake
348
Systems of Care and Effect on CKD
311
Sodium
348
Multidisciplinary Clinics
313
Bicarbonate
349
Economics
315
Rate of Decline of GFR and GFR Variability
349
Electronic Health Records
Volume 14, Number 4, October 2015
350
351
CKD Education
Disparities
351
Race and Sex
352
Access to Care
Upcoming Issues
Transplantation
John P. Vella, MD and Alexander C. Wiseman, MD
November 2015
Hypertension
353
Geriatric Nephrology Issues
353
Physical Function and Frailty
Aldo J. Peixoto, MD and Debbie L. Cohen, MD
March 2016
354
Cognitive Impairment
Pregnancy and the Kidney
355
Palliative Care
CME Self-Assessment Questions
358
NephSAP, Volume 14, Number 4, October 2015—
Chronic Kidney Disease and Progression
Belinda Jim, MD, Kate Bramham, MBBS, Sharon E. Maynard, MD,
and Michelle A. Hladunewich, MD
May 2016
Glomerular, Vascular, and Tubulointerstitial Diseases
Richard J. Glassock, MD and Brad H. Rovin, MD
July 2016
Volume 14, Number 4, October 2015
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
Alok Agrawal, MD, FASN
Wright State University
Dayton, OH
Mustafa Ahmad, MD, FASN
King Fahad Medical City
Riyadh, Saudi Arabia
Kamal E. Ahmed, MD, FASN
Yuma Nephrology
Yuma, AZ
Sadiq Ahmed, MD
University of Kentucky
Lexington, KY
Nasimul Ahsan, MD, FASN
North Florida/South Georgia
VA Health System
Gainesville, FL
Jafar Al-Said, MD, FASN
Bahrain Specialist Hospital
Manama, Bahrain
Christopher A. Dyer, MD
University of Texas Health Science
Center at San Antonio
San Antonio, TX
Mahmoud El-Khatib, MD
University of Cincinnati
Cincinnati, OH
Lynda A. Frassetto, MD, FASN
University of California at
San Francisco
San Francisco, CA
Nitin V. Kolhe, MD, FASN
Royal Derby Hospital
Derby, Derbyshire, UK
Claude Mabry Galphin, MD
Nephrology Associates
Chattanooga, TN
Nicolae Leca, MD
University of Washington
Seattle, WA
Mohammad Reza Ganji, MD
Tehran University
Tehran, Iran
Paolo Lentini, MD, PhD
San Bassiano Hospital
Bassano del Grappa, Italy
Naheed Ansari, MD, FASN
Jacobi Medical Center/Albert Einstein
College of Medicine
Bronx, NY
Duvuru Geetha, MD, FASN
Johns Hopkins University
Baltimore, MD
Carl S. Goldstein, MD, FASN
Robert Wood Johnson
Medical School
New Brunswick, NJ
Gopal Basu, MD
Christian Medical College
Vellore, Tamil Nadu, India
Steven M. Gorbatkin, MD, PhD
Emory University,
Atlanta, GA
Mona B. Brake, MD, FASN
Robert J. Dole VA Medical Center
Wichita, KS
Ashik Hayat, MD, FASN
Taranaki Base Hospital
Newplymouth, Taranaki, NZ
Ruth C. Campbell, MD
Medical University of South Carolina
Charleston, SC
Ekambaram Ilamathi, MD, FASN
State University of New York
Stony Brook, NY
Chokchai Chareandee, MD, FASN
University of Minnesota
Minneapolis, MN
Dalila B. Corry, MD, FASN
UCLA School of Medicine
Northridge, CA
Bulent Cuhaci, MD, FASN
American Hastanesi
Istanbul, Turkey
Kevin A. Curran, MD
Fresenius Medical Care & US Renal
Care Dialysis Facilities
Canton, TX
Rajiv Dhamija, MD
Rancho Los Amigos National
Rehabilitation Center
Downey, CA
Talha Hassan Imam, MD
Kaiser Permanente
Fontana, CA
Pradeep V. Kadambi, MD
University of Arizona
Tucson, AZ
Sharon L. Karp, MD
Indiana University
Indianapolis, IN
Amir Kazory, MD, FASN
University of Florida
Gainesville, FL
Rahul Koushik, MD
University of Texas
Health Science Center
San Antonio, TX
Lalathaksha Murthy Kumbar, MBBS
Henry Ford Hospital
Detroit, MI
Edgar V. Lerma, MD, FASN
University of Illinois at Chicago
College of Medicine
Chicago, IL
Orfeas Liangos, MD, FASN
Klinikum Coburg
Coburg, Bayern, Germany
Meyer Lifschitz, MD
Shaare Zedek Medical Center
Jerusalem, Israel
Jolanta Malyszko, MD, PhD
Medical University
Bialystok, Poland
Christopher Mariat, MD, PhD
University Jean Monnet
Saint-Etienne, France
Naveed Masani, MD
Winthrop University Hospital
Mineola, NY
Hanna W. Mawad, MD, FASN
University of Kentucky
Lexington, KY
Kevin McConnell, MD
Jefferson Nephrology, Ltd
Charlottesville, VA
Apurv Khanna, MD
SUNY Upstate Medical University
Syracuse, NY
Pascal Meier, MD, FASN
Centre Hospitalier du Valais Romand
Sion, Switzerland
Istvan Kiss, MD, PhD
Semmelweis University
Budapest, Hungary
Ashraf Mikhail, MBBCh
Morriston Hospital
Swansea, Wales, UK
Tanuja Mishra, MD
Kaiser Permanente
Mid-Atlantic Region
Ellicott City, MD
Wajeh Y. Qunibi, MD
University of Texas
Health Science Center
San Antonio, TX
Lawrence S. Moffatt, Jr., MD
Carolinas Medical Center
Charlotte, NC
Pawan K. Rao, MD, FASN
St. Joseph Hospital
Syracuse, NY
Sumit Mohan, MD
Columbia University College of
Physicians and Surgeons
New York, NY
Bharathi V. Reddy, MD
University of Chicago
Medical Center
Chicago, IL
Shahriar Moossavi, MD, PhD, FASN
Wake Forest School of Medicine
Winston-Salem, NC
Joel C. Reynolds, MD, FASN
Internal Medicine Clinic
Meridian, MS
Koosha Mortazavi, MD
Vista Del Mar Medical Group
Oxnard, CA
Brian S. Rifkin, MD
Hattiesburg Clinic
Hattiesburg, MS
Tariq Mubin, MD
Kern Nephrology Medical Group
Bakersfield, CA
Helbert Rondon-Berrios, MD, FASN
University of Pittsburgh
School of Medicine
Pittsburgh, PA
Narayana S. Murali, MD
Marshfield Clinic
Marshfield, WI
Thangamani Muthukumar, MD
Cornell University
New York, NY
Mohanram Narayanan, MD, FASN
Scott and White Healthcare
Temple, TX
Macaulay A. Onuigbo, MD, FASN
Mayo Clinic
Rochester, MN
Kevin P. O'Reilly, MD
Ohio State University
Columbus, OH
Carlos E. Palant, MD
Washington DC VA Medical Center
Washington, DC
Malvinder Parmar, MB, MS, FASN
Northern Ontario School of Medicine
Timmins, ON, Canada
Bijan Roshan, MD, FASN
Kidney Associates of Colorado
Denver, CO
Mario F. Rubin, MD, FASN
University of Arizona
Tucson, AZ
Ehab R. Saad, MD, FASN
Medical College of Wisconsin
Milwaukee, WI
Bharat Sachdeva, MBBS
Louisiana State University
Health Sciences Center
Shreveport, LA
Mark C. Saddler, MBChB
Durango Nephrology Associates
Durango, CO
Mohammad G. Saklayen, MBBS
Wright State University
Boonshoft School of Medicine
Dayton, OH
Pairach Pintavorn, MD, FASN
East Georgia Kidney and Hypertension
Augusta, GA
Muwaffaq Salameh, MBBS
St. Martha Regional Hospital
Antigonish, NS, Canada
James M. Pritsiolas, MD, FASN
CarePoint Health Medical Group
Bayonne and Chatham, NJ
Mohammad N. Saqib, MD
Lehigh Valley Hospital
Orefield, PA
Paul H. Pronovost, MD, FASN
Yale University School of Medicine
Waterbury, CT
Henry L. Schairer, Jr., MD, FASN
Lehigh Valley Health Network
Allentown, PA
Mohammad A. Quasem, MD
Universal Health Services Hospitals
Binghamton, NY
Gaurang M. Shah, MD, FASN
Long Beach VA Healthcare System
Long Beach, CA
Nita K. Shah, MD
St. Barnabas Health Center
Livingston, NJ
Arif Showkat, MD, FASN
University of Tennessee
Memphis, TN
Sandeep S. Soman, MD
Henry Ford Hospital
Detroit, MI
Manish M. Sood, MD, FASN
University of Manitoba
Winnipeg, MB, Canada
Susan P. Steigerwalt, MD
St. John Providence Hospital
Detroit, MI
Ignatius Yun-Sang Tang, MD
University of Illinois Hospital and
Health Sciences System
Chicago, IL
Ahmad R. Tarakji, MD, FASN
College of Medicine, King Saud University
Riyadh, Saudi Arabia
Hung-Bin Tsai, MD
National Taiwan University
Hospital
Taipei, Taiwan
Anthony M. Valeri, MD
Columbia University
New York, NY
Allen W. Vander, MD, FASN
Kidney Center of South
Louisiana
Thibodaux, LA
Juan Carlos Q. Velez, MD
Medical University of South
Carolina
Charleston, SC
Anitha Vijayan, MD, FASN
Washington University
in St. Louis
St. Louis, MO
Shefali Vyas, MD
St. Barnabas Health Center
Livingston, NJ
Nand K. Wadhwa, MD
Stony Brook University
Stony Brook, NY
Sameer Yaseen, MD
Nephrology PC
Des Moines, IA
Mario Javier Zarama, MD
Kidney Specialists of
Minnesota, PA
Saint Paul, MN
Volume 14, Number 4, October 2015
Program Mission and Objectives
The Nephrology Self-Assessment Program (NephSAP) provides a learning vehicle for clinical nephrologists to renew and
refresh their clinical knowledge, diagnostic, and therapeutic skills. This enduring material provides nephrologists challenging,
clinically oriented questions based on case vignettes, a detailed syllabus that reviews recent publications, and an editorial on an
important and evolving topic. This combination of materials enables clinicians to rigorously assess their strengths and
weaknesses in the broad domain of nephrology.
Accreditation Statement
The American Society of Nephrology (ASN) is accredited by the Accreditation Council for Continuing Medical Education to
provide continuing medical education for physicians.
AMA Credit Designation Statement
The ASN designates this enduring material for a maximum of 8.0 AMA PRA Category 1 Credits™. Physicians should claim
only the credit commensurate with the extent of their participation in the activity.
Original Release Date
October 2015
CME Credit Termination Date
September 30, 2017
Examination Available Online
On or before Thursday, October 15, 2015
Estimated Time for Completion
8 hours
Answers with Explanations
Provided with a passing score after the first and/or after the second attempt
October 2017: posted on the ASN website when the issue is archived.
•
•
Target Audience
Nephrology certification and recertification candidates
Practicing nephrologists
Internists
•
•
•
Method of Participation
Read the syllabus that is supplemented by original articles in the reference lists.
Complete the online self-assessment examination.
Each participant is allowed two attempts to pass the examination (.75% correct) for CME credit.
Upon completion, review your score and incorrect answers and print your certificate.
Answers and explanations are provided with a passing score or after the second attempt.
•
•
•
•
•
Volume 14, Number 4, October 2015
Activity Evaluation and CME Credit Instructions
Go to www.asn-online.org/cme, and enter your ASN login on the right.
Click the ASN CME Center.
Locate the activity name and click the corresponding ENTER ACTIVITY button.
Read all front matter information.
On the left-hand side, click and complete the Demographics & General Evaluations.
Complete and pass the examination for CME credit.
Upon completion, click Claim Your Credits, check the Attestation Statement box, and enter the number of CME
credits commensurate with the extent of your participation in the activity.
If you need a certificate, Print Your Certificate on the left.
•
•
•
•
•
•
•
•
For your complete ASN transcript, click the ASN CME Center banner, and click View/Print Transcript on the left.
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 enrolled in the ABIM–MOC program
Enroll for MOC via the ABIM website (www.abim.org).
Enter your (ABIM) Candidate Number and Date of Birth prior to completing the examination.
Take the self-assessment examination within the timeframe specified in this issue of NephSAP.
Below your score select “Click here to post to ABIM.”
•
•
•
•
•
MOC points will be applied to only those ABIM candidates who have enrolled in the MOC program. It is your responsibility to
complete the ABIM MOC enrollment process.
System Requirements
Compatible Browser and Software
The ASN website (asn-online.org) has been formatted for cross-browser functionality, and should display correctly in all
modern web browsers. To view the interactive version of NephSAP, your browser must have Adobe Flash Player installed or
have HTML5 capabilities. NephSAP is also available in Portable Document Format (PDF), which requires Adobe Reader or
comparable PDF viewing software.
Monitor Settings
The ASN website was designed to be viewed in a 1024 · 768 or higher resolution.
Medium or Combination of Media Used
The media used include an electronic syllabus and online evaluation and examination.
Technical Support
If you have difficulty viewing any of the pages, please refer to the ASN technical support page for possible solutions. If you
continue having problems, contact ASN at email@asn-online.org.
Volume 14, Number 4, October 2015
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 content, and are independent of the
control from commercial interests and free of 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), individuals who are
in a position to control the content 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 relationships of a spouse
or partner. Peer reviewers are asked to abstain from reviewing topics if they have a conflict of interest. Disclosure information is made available to learners
prior to the start of any ASN educational activity.
EDITORIAL BOARD:
Michael J. Choi, MD—Current Employer: Johns Hopkins University School of Medicine; Consultancy: GlaxoSmithKline-Data monitoring safety
board, Relypsa-Advisory board; Scientific Advisor/Membership: Editorial Board: Clinical Journal of the American Society of Nephrology,
Clinical Nephrology, National Kidney Foundation Board of Directors
Linda F. Fried, MD, MPH, FASN—Current Employer: VA Pittsburgh Healthcare System; Research Funding: Site investigator in study: Abbvie,
NephroGenex, Merck (study drug donation); Ownership Interest: Pfizer expert witness
Gerald A. Hladik, MD—Current Employer: University of North Carolina at Chapel Hill; Scientific Advisor/Membership: Education Director for
NephSAP, American Society of Nephrology
John P. Middleton, MD—Current Employer: Duke University; Consultancy: Relypsa, AstraZeneca; Research Funding: Keryx, Bristol-Myers
Squibb, Janssen, NephroGenex; Scientific Advisor/Membership: Editorial Board: Journal of Human Hypertension, Advances in CKD; Steering
committee NIDDK CKD Consortium
Asghar Rastegar, MD—Current Employer: Yale University School of Medicine
Jerry Yee, MD, FASN—Current Employer: Henry Ford Hospital; Consultancy: Amgen, Vasc-Alert, Alexion, ZS Pharma; Ownership Interest:
Merck, Gilead; Honoraria: Amgen, Alexion, Gerson, Drexel University, University of California at San Diego, ZS Pharma; Patents/Inventions: VascAlert; Scientific Advisor/Membership: NKF: Editor-In-Chief of Advances in CKD (journal); Editorial Board: CJASN, American Journal of Nephrology,
Deputy Editor, NephSAP; Other Interests/Relationships: Chief Medical Director, Greenfield Health Systems
CONTRIBUTING AUTHOR:
Sumeska Thavarajah, MD—Current Employer: Johns Hopkins Bayview Medical Center; Scientific Advisor/Membership: Medical Advisory BoardNKF of Maryland, Current Chair
EDITORIAL AUTHORS:
Karen Griffin, MD, FASN—Current Employer: Loyola University Medical Center, Hines VA Hospital; Research Funding: Complexa; Scientific
Advisor/Membership: Member, Editorial Board of Hypertension; Chair, Professional & Public Education Committee of the American Heart
Association Hypertension Council Member, NIH PBKD Study Section; Other Interests/Relationships: Member of the Scientific Advisory Board of the
National Kidney Foundation of Illinois
Holly J. Kramer, MD—Current Employer: Loyola University Medical Center; Scientific Advisor/Membership: Editorial Board: American Journal of
Kidney Disease, Advances in Chronic Kidney Disease, Clinical Journal of the American Society of Nephrology
ASN STAFF:
Gisela A. Deuter, BSN, MSA—Nothing to disclose
Commercial Support
There is no commercial support for this issue.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Editorial
Obesity and Kidney Disease
Holly Kramer, MD, MPH,*†‡ and Karen Griffin, MD†‡
*Departments of Public Health Sciences and †Medicine, Division of Nephrology and
Hypertension, Loyola University Chicago, Maywood, Illinois; and Section of Nephrology, ‡Hines
Veterans Affairs Medical Center, Hines, Illinois
“No disease that can be treated by diet should be
treated with any other means,” Mosheh ben Maimon
(Maimonides, circa 1200 AD).
activity” to manage a patient with moderate CKD
and obesity (5). Similar to caloric restriction, specific
aspects of nutritional management, which may deter
the development and progression of CKD in the
setting of obesity, are not widely used. These nutritional aspects include salt and protein restriction and
increasing the fiber content of the diet. Behavioral
change, however, is extremely difficult to initiate and
sustain.
The industrialization of food, gradual increases in
portion sizes, and concomitant changes in the cost
structure of foods over the past three decades have
largely mediated the obesity epidemic and impeded
public health efforts. Since the 1980s, healthier foods
have become relatively more expensive compared with
processed foods, and portion sizes for processed foods
have increased disproportionately to those of healthy
foods (6). Despite the ubiquity and quantity of calorically dense and palatable processed foods in the 21st
century, marked interindividual differences in obesity
risk remain. These interindividual differences in obesity risk have been attributed to genetic factors, with
numerous variants across the genome, each contributing very small effects (7). However, such genetic effects
cannot be operative if caloric intake does not exceed
caloric expenditure. The answer to why some individuals consume food in great excess of caloric needs
may be found in the vast array of neuroimaging studies
that have been reported in obese and nonobese
individuals.
Overall, comparisons of brain imaging studies
by obesity status have shown increased activity in
the areas of the brain that modulate the neural
circuits for somatosensory processes, memory, and
mesolimbic pathways (8). A meta-analysis of neuroimaging studies in obese and nonobese individuals
reported that obese individuals in general have increased
Obesity
Within the United States adult population, approximately one in three is overweight (body mass index
[BMI] ¼25–29.9 kg/m2), one in three is obese (BMI$30
kg/m2), and approximately 1 in 20 is morbidly obese
(BMI$40 kg/m2) (1,2). Thus, approximately only one in
three United States adults has an ideal BMI (18.5–24.9
kg/m2). Estimates of the costs of obesity in the US health
care system are staggering, with up to $150 billion spent
on obesity-related comorbidities annually, including
CKD (3). Currently, it is estimated that up to 24% of
CKD can be attributed to obesity among industrialized
countries (4). However, the majority of diabetes and
hypertension incidence remains a function of excessive
caloric intake combined with low physical activity.
Thus, it stands to reason that the United States obesity
epidemic contributed to the concurrent and rapid rise
in ESRD over the past 25 years and that most ESRD
in the United States originates directly or indirectly
from obesity.
Public health efforts to address the obesity epidemic have been extremely slow to respond. Even today,
the overwhelming majority of physicians are not
addressing obesity with their patients and/or providing
nutrition and weight management counseling for management of chronic diseases, including CKD. Within
the nephrology community, the role of obesity in CKD
management remains contentious. Among 399 nephrologists (57% from Europe and 12% from Central and
South America), only 65% stated that obesity per se is
a risk factor for CKD, and only 32% said that they
would “prescribe a calorically restricted diet and try to
motivate the patient to increase his/her degree of physical
283
284
activation of certain prefrontal cortical regions after
viewing images of food (9). The increased activation
of these prefrontal cortical regions when viewing
food images compared with that in nonobese individuals suggests that obese individuals are more
likely to focus on the hedonic or reward aspects of
the food (8,9). In other words, the amount of ingested
food required to make an individual feel rewarded
differs by obesity status. While viewing food images,
obese individuals also have higher and more consistent activation of prefrontal cortical regions that
modulate neural circuits involved with habit formation (mesolimbic pathways) but reduced activation of
cortical areas that regulate feelings of satiety and
cognitive control compared with nonobese individuals
(8,9). However, it remains unclear whether differences in mesolimbic pathway activation between
individuals are a function of genetic variants, diet,
or gene-environment interactions. Consumption of
energy-dense and highly palatable foods elicits neurologic responses very similar to neurologic responses
elicited by illicit drug use. Repeated consumption of
energy-dense foods and fast foods may alter mesolimbic pathways and lead to addictive behaviors. It
has been hypothesized that consumption of salty
processed foods activates areas in the brain that elicit
a hedonic reward followed by cravings for more
salty, processed foods, similar to responses of opiate
use (10). The scientific community has not reached
consensus that certain foods, such as fast food, are
addictive (11); however, the cortical activation patterns after viewing food among obese individuals
are similar to patterns seen in drug-addicted individuals (8).
Is Obesity an Independent Risk Factor for CKD?
Obesity, especially morbid obesity, seems to be
a very strong risk factor for future risk of ESRD when
obesity is present during young adulthood. Between
1964 and 1985, 320,252 adults ages 18–34 years old
volunteered for screening health evaluations in a large
health care system. When participants had been followed
for 15–34 years and were 33–68 years old, morbid
obesity (BMI$40 kg/m2) was associated with a 6-fold
higher risk of ESRD compared with that in individuals
with an ideal BMI (18.5–24.9 kg/m2). Among the young
adults with baseline CKD, morbid obesity was associated with a 3-fold higher risk of ESRD compared with
that in individuals with an ideal BMI (18.5–24.9 kg/m2)
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
and baseline CKD (12). Similar associations were noted
between overweight and obesity during adolescence and
risk for developing ESRD during midlife (13). Studies
that used older cohorts with more recent follow-up time
periods have found much weaker associations between
obesity and ESRD risk (14–16) and suggested that the
association between obesity and ESRD risk is limited to
individuals with metabolic syndrome and/or elevated BP
(14,16). Among 21,840 participants in the Reasons for
Geographic and Racial Differences in Stroke Study,
a cohort with a mean baseline age of approximately
65 years old followed for 6.361.3 years, 247 developed ESRD. Participants who were obese defined by
a BMI$30 kg/m2 had an approximately 2-fold higher risk
for ESRD compared with participants with an ideal
BMI after adjustment for covariates, but this association was limited to individuals with metabolic syndrome (14).
Measures of abdominal obesity, such as increased
waist circumference or waist to hip ratio, are more
consistently associated with ESRD risk compared with
BMI (17–19). BMI reflects muscle mass and visceral
and peripheral adipose tissue, whereas measures of
abdominal adiposity reflect visceral fat, a strong risk
factor for development of insulin resistance (20). However, regardless of the measurement methods, the
presence of obesity among older adults (e.g., 60 years
old and older) only modestly increases the risk of
ESRD, and associations are largely caused by the
confounding effects of hypertension and diabetes
(14,15). In contrast, obesity, especially morbid obesity,
during adolescence or young adulthood extrapolates
to a prolonged period of exposure to obesity-related
comorbidities that increase CKD risk (e.g., metabolic
syndrome, diabetes, and hypertension). This prolonged
effect of obesity takes place along with the potentially
independent effects of obesity on kidney functional
parameters as discussed below (Figure 1). Thus, public
health efforts to curb the epidemic of CKD should
include obesity prevention and treatment programs that
target adolescents and young adults. Such programs that
target young adults will not only help retard CKD
incidence (21) but also, will reduce cardiovascular
disease frequency and lengthen the life expectancy of
young adults (22–24). Weight loss for older obese adults
(age .60 years old) may also provide health benefits,
but the overall public health benefits of obesity prevention are much larger for young individuals who
have several decades of life ahead of them.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 1. Hypothesized model for time of obesity onset and
future risk for ESRD. Obesity during young adulthood versus
older age translates to a longer cumulative exposure to
obesity-related factors. On the basis of data from refs. 12–15.
Obesity and Hyperfiltration
The kidneys are remarkable organs that adapt
readily to changes in caloric intake and metabolic rate
(25,26). With weight gain, metabolic rate, glomerular
diameter, GFR, and renal plasma flow (RPF) all increase
(27) along with regional blood flow, arterial pressure,
and cardiac output (28,29). The greater GFR frequently
noted in obese individuals has been called hyperfiltration, a term previously described in association with
diabetes and pregnancy, where GFR increases to a level
higher than normal, generally two SDs above the normal
for healthy individuals (30). Indexing GFR for body
surface area usually eliminates the higher GFR noted in
the obese state; therefore, the higher GFR noted with
obesity is not a pathophysiologic state per se. However,
nephron number is set at birth, and therefore the increase
in total GFR with weight gain occurs at the expense of
increasing single-nephron GFR (SNGFR).
It is often suggested that obesity-related hyperfiltration is analogous to the 5/6th remnant kidney
model, in which hyperfiltration is thought to be
mediated by increases in both glomerular capillary
RPF rate and mean glomerular capillary hydraulic
pressure caused by adaptive reductions in preglomerular and postglomerular arteriolar resistances. The
increase in glomerular capillary pressure (PGC) has
been ascribed to a greater dilation of the afferent than
the efferent arteriole that results in a relative efferent
vasoconstriction, giving rise to glomerular hypertension (31–34). However, hyperfiltration can also be
achieved through coordinated increases in glomerular
filtration surface area (hypertrophy) and increases in
single-nephron RPF through proportionate afferent and
efferent vasodilation without significant glomerular
285
hypertension. Indeed, recent data suggest that glomerular hypertrophy along with increases in RPF are the
dominant mechanisms by which hyperfiltration is
achieved in normotensive states. In fact, glomerular
hypertension may not even be effective in increasing
SNGFR because of the inverse relationship between
PGC and the ultrafiltration coefficient (Kf), the product
of hydraulic conductivity and glomerular capillary
surface area (35).
The link between glomerular hypertrophy and
hyperfiltration was recently addressed by Lenihan et al.
(36). Using kidney donation as a model, the increased
GFR in the remaining kidney after kidney donation was
shown to be mainly a function of increasing Kf as
determined by renal cortical volume measured pre- and
postkidney donation by magnetic resonance imaging
and an estimation of nephron number from a core biopsy
specimen obtained from the donated kidney. Hence, the
expected increase in Kf when accompanied by increased
RPF facilitates the requisite increase in GFR without
a pathologic increase in PGC. In other words, it is like
growing glomeruli in situ. However, it should be noted
that, in the obese state, increases in RPF tend to lag
behind the increase in GFR.
Kwakernaak et al. (37) previously summarized
the numerous studies that examined GFR, effective
renal plasma flow (ERPF), and the GFR to RPF ratio
or filtration fraction (FF) by obesity status and found
fairly consistent associations between obesity status
and greater FF across studies. Increased FF reflects
a disproportionate increase in GFR relative to RPF and
has been considered a proxy for glomerular hypertension. The theory that increased FF equates with glomerular hypertension is on the basis of micropuncture data
in certain colonies of Munich Wistar rats. In these
animals, ultrafiltration concludes before the end of the
glomerular capillaries when filtration pressure equilibrium has been achieved and net ultrafiltration pressure
equals zero. In other words, these rats do not completely
use the available glomerular capillary surface area for
ultrafiltration. In contrast, humans, large animals
(dogs), and other rat strains seem to use the entire
glomerular capillary surface area for filtration, a state
of filtration pressure disequilibrium. In filtration
pressure disequilibrium states, any decrease in RPF,
even if caused by proportional increases in afferent
and efferent resistance, may increase FF without an
increase in PGC. Additionally, assuming that greater
FF in humans reflects glomerular hypertension is
286
problematic, because PGC correlates poorly with SNGFR
because of the aforementioned inverse relationship with
Kf (35).
Susceptibility to Obesity-Mediated CKD
The increase in SNGFR as a result of unilateral
nephrectomy for kidney donation or during pregnancy
exceeds the more modest increases in SNGFR as a result
of the obese state. This modest increase in SNGFR in
the obese state may explain the overall low risk of
kidney disease among obese individuals given the
absence of diabetes or hypertension. Similarly, PGC
unlikely increases in most patients after kidney donation,
and this may explain why the absolute risk of ESRD
among kidney donors is low, albeit higher than in
nonkidney donors, despite the presence of glomerular
hyperfiltration in the nondonated kidney (36,38,39).
Hence, obesity-associated hyperfiltration itself is likely
not sufficient for development of CKD, and additional
factors are required to induce susceptibility and injury,
such as reduced nephron mass. Total GFR is the product
of SNGFR, with total nephron number having a Gaussian distribution with a mean centered around 1,000,000
(40). After kidney donation, the remaining kidney hypertrophies and renocortical volumes increase, reflecting
an increase in glomerular capillary surface area to augment
filtration surface area and GFR. Similarly, as an individual
gains weight, GFR must increase further to match metabolic demands. Consequentially, individuals at the low end
of the spectrum of nephron number may be at particular
risk for obesity-related nephropathy.
Augmentation of SNGFR with weight gain in the
setting of reduced nephron number may require a degree
of vasodilation of the afferent arteriole that compromises
the normal vasoconstrictive response to elevations in
systemic BP. This impairment in renal autoregulatory
capacity/efficiency results in enhanced transmission of
systemic pressure to the glomerular capillaries, and
glomerular hypertension results in obesity–related glomerular sclerosis (32,41). Similarly, the greater increases
in glomerular hypertrophy require that terminally differentiated podocytes envelop a larger glomerular
capillary surface area. It has been postulated that podocytes provide mechanical support against PGC. Accordingly, a reduction in podocyte density may compromise
the ability to withstand mechanical stress, particularly
during PGC elevations (42,43). In moderate glomerular
hypertrophy, the increase in glomerular capillary surface
area is usually accomplished by increases in glomerular
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
capillary length without an increase in diameter. However, with more extreme glomerular enlargement, increases in both glomerular capillary length and diameter
occur. Thus, on the basis of the Law of LaPlace, in which
wall tension increases as a function of glomerular diameter at constant PGC, there is increased susceptibility
for the development of glomerulosclerosis (44). In
addition, when the glomerular radius can increase no
further in the face of increasing PGC, wall tension must
increase with equivalent pathobiology.
Management of Obese Patients with CKD
Because obesity has been proposed as a high-risk
state for glomerular hypertension, blockade of the
renin-angiotensin-aldosterone system has been suggested as the primary treatment for obesity-associated
hypertension. The relatively lower ERPF observed in
the obese state may be more dependent on angiotensin, because changes in ERPF observed with use of
angiotensin–converting enzyme inhibitors (ACEIs) are
significantly higher among obese individuals compared
with lean or overweight individuals (BMI,30 kg/m2) (45).
Medications that block the renin-angiotensin-aldosterone
system show renal protective effects for obese and
nonobese adults with established proteinuria. However,
superior benefits from ACEIs or angiotensin receptor
blockers (ARBs) versus diuretics or calcium channel
blockers have not been shown in large–scale clinical
trials in populations not enriched with adults with CKD.
Focusing solely on use of ACEI/ARB medications for BP
management in obese adults has limitations. Obesity is
a volume-expanded state, and obese individuals frequently have a high salt intake, which will attenuate
the BP-lowering effects of drugs that block the reninangiotensin system, such as ACEIs or ARBs (46).
The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) included
over 33,000 individuals 55 years of age and older with
mean BMI ¼29.7 kg/m2 (47). ALLHAT showed no
difference in the primary outcome of fatal coronary heart
disease or nonfatal myocardial infarction by allocation
status to chlorthalidone, lisinopril, or amlodipine, regardless of baseline GFR. Risks of ESRD and mortality
also did not differ by allocation status to chlorthalidone,
lisinopril, or amlodipine (48). However, the lisinopril
arm had a 1.15-fold higher relative risk for stroke (95%
confidence interval [95% CI], 1.05 to 1.16) and a 1.19fold higher relative risk for heart failure (95% CI, 1.07 to
1.31) compared with chlorthalidone (47). During all of
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
the years of the trial, mean systolic BP was lowest in the
chlorthalidone arm. Study outcomes for the amlodipine
arm were similar to the chlorthalidone arm, except for an
increased risk of heart failure with amlodipine compared
with chlorthalidone (relative risk, 1.38; 95% CI, 1.25 to
1.52). Because of impairment of autoregulation, use of
calcium channel blockers can accelerate kidney damage
in obese adults with hypertension by transmission of
systemic pressures to the glomerular capillary, especially
if the hypertension is inconsistently controlled (49,50).
Salt Restriction
The obesity epidemic has been fueled by the
industrialization of our diet, and processed foods are not
only calorically dense but also, high in salt. Obese
individuals will, in general, have a higher salt intake
compared with lean individuals (51,52). High salt intake
will heighten BP and lead to volume expansion, making
BP management more difficult. High salt intake may also
accelerate kidney injury by amplifying glomerular hypertrophy with weight gain. Although no study has
examined the effect of salt intake on glomerular volume
in humans, animal models of renal mass reduction show
that salt restriction reduces glomerular hypertrophy and
susceptibility to hypertensive injury (53,54). Additionally, research suggests that high salt intake impairs renal
autoregulatory responses (55–57), but this remains
controversial (58,59). High salt intake may also affect
kidney disease through nonhemodynamic pathways.
High salt intake magnifies oxidative stress and contributes to renal injury through stimulation of renal cortical
NADH– and NADPH–induced superoxide generation
(60,61). High salt intake has also been shown to induce
fibrogenic pathways through upregulation of TGF-b
(62,63). The effect of a high-salt diet on GFR, RPF,
and FF may be most operative in overweight or obese
adults (44,64). Krikken et al. (64) showed that, when
healthy young men transition from a low-sodium
(50 mmol/d) to a high-sodium diet (200 mmol/d), GFR,
ERPF, and FF increase only among men with BMI .25
kg/m2. It should be noted that only two men in this study
had a BMI .30 kg/m2. Thus, dietary recommendations
for both overweight and obese individuals with CKD
should include salt restriction.
Protein Restriction
The typical United States diet contains about two
times the protein intake recommended by the United
States dietary guidelines (65). For patients with CKD,
287
managing proper protein intake remains an important
modifiable factor for slowing the rate of CKD progression, but the role of dietary protein in CKD progression
may be most operative in adults with obesity and CKD.
The current recommended daily allowance for protein
intake is 0.8 g/kg healthy body wt (not total body weight)
(66). Among obese adults, mean protein intake exceeds
100 g protein per day. Assuming a healthy body weight of
70 kg, 100 g of protein intake per day equates to an
obese individual consuming at least 1.43 g protein per
1 kg healthy body wt or approximately 80% more protein
intake than the recommended daily allowance (67). The
effect of animal protein intake on glomerular hemodynamics was shown several decades ago (68). Both ERPF
and GFR increase by at least 30% when healthy persons
transition from a low–animal protein diet to a high–
animal protein diet (68). These hemodynamic effects are
limited to animal protein, because increasing vegetable
protein intake does not increase RPF (69). Friedman
et al. (70) elegantly showed that hyperfiltration in obese
patients is not mediated by protein intake. However,
high–animal protein intake may heighten PGC through
afferent dilation as a function of amino acids triggering
multiple humoral and local mediators (69,71–73). Afferent vasodilation will enhance transmission of systemic
pressures to the glomerular capillary in obese individuals
who are frequently hypertensive. Among adults with
established CKD, moderate protein restriction (approximately 0.7 g/kg per day) compared with a standard protein intake (approximately 1.0 g/kg per day) is associated
with a 0.53-ml/min per year (95% CI, 0.08 to 0.98) lower
GFR decline compared with a standard protein intake,
but stronger effects are noted in adults with diabetes,
a group with a high prevalence of obesity (74).
High intake of animal protein combined with low
intake of fruits and vegetables is the most common
dietary pattern among United States adults (75), and it
translates to high net endogenous acid production (76).
Higher net endogenous acid production increases reninangiotensin system activity and the amount of hydrogen
ions that individual nephrons must excrete to maintain
acid-base balance (77). Sodium bicarbonate supplementation will help counteract higher net endogenous acid
production. However, increasing fruit and vegetable
intake will not only reduce net endogenous acid production but also increase the fiber content of the diet.
Dietary fibers are generally the plant cell wall polysaccharides that remain undigested by gastrointestinal
tract enzymes (78). High–fiber diet content reduces total
288
caloric intake without major disruptions in satiety, thereby
facilitating weight loss and/or weight maintenance (79).
High-fiber foods, such as whole grains, legumes, fruits,
and vegetables, have the added benefit of reducing
systemic inflammation (80,81), which may potentially
affect CKD incidence and progression (82–85).
It has been postulated that diets high in fiber
reduce systemic inflammation by facilitating the growth
of endosymbiotic bacteria colonizers of the gut, such as
Bifidobacterium, which inhibit intestinal colonization by
gram-negative pathogens and reduce the formation of
endotoxins and end products of urease-producing bacteria (79,86,87). Reduction of bacterial end products
leads to more junctional proteins at intestinal epithelial
zonula occludens, thereby decreasing paracellular gut
wall permeability and the likelihood of translocation of
bacteria and endotoxins from intestinal lumen to the
systemic circulation (88). Although no specific recommendations for dietary fiber intake exist for adults with
CKD, the American Dietetic Association recommends
14 g dietary fiber per 1000 kcal per day (78). These
reference intakes may confer benefits for patients with
CKD, but serum potassium and phosphate levels should
be monitored.
Caloric Restriction
Multiple studies have consistently shown a link
between abdominal obesity and increased urine protein
(mainly albumin) excretion across a wide range of
patient populations (89–92). Higher urine protein excretion generally correlates with a faster rate of GFR
decline for the majority of kidney diseases (93–95).
Although intensive BP lowering reduces urine protein
excretion (96,97), caloric restriction and weight loss are
also effective interventions that lower urine protein
excretion among adult patients, regardless of BP
(98,99). For example, caloric intake during the first
6 months after bariatric surgery is markedly limited.
Notably, although this weight loss does not reverse
morbid obesity by 6 months, GFR, ERPF, and urine
protein excretion decline significantly compared with
prebariatric surgery levels (98,100).
One pathway that mechanistically reconciles how
caloric restriction reduces protein excretion is the Sirt1Foxo1 pathway (101). Sirt1 is an NAD1-dependent
deacteylase (102) that is abundantly expressed in the
renal inner medulla and activates the cell cycle protein
Foxo1 (103). Activated Foxo1 forms a transcriptional
complex with CCAAT/enhancer binding protein a
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
(C/EBP-a) the adiponectin gene promoter site, thereby
upregulating gene expression of adiponectin (104).
Adiponectin receptors reside on podocytes and likely
play a role in podocyte morphology, function, and response to injury (105–107). In a study of 30 overweight
and obese adults with proteinuria and proteinuric
kidney diseases (both with and without diabetes),
eliminating 500 kcal/d for 5 months decreased urine
protein excretion by 30%, despite achieved weight loss
of only 4% of baseline weight (108). Moreover, the decline in urine protein excretion occurred as early as
1 month after initiation of caloric restriction (108,109).
Consistent caloric restriction combined with an
increased or stable physical activity level will lead to
weight loss. Drastic weight loss is not required to
positively affect CKD, and in some patients, the
detrimental effects of obesity, especially with morbid
obesity, may be reversible. A systematic review that
included 522 adults from clinical trials and cohort
studies of dietary interventions and bariatric surgery
disclosed that every 1 kg of intentional weight loss,
regardless of intervention, led to a decrease in urine
protein excretion of approximately 110 mg (95% CI, 60
to 160 mg) and a decrease in urine albumin excretion of
1.1 mg (95% CI, 0.5 to 2.4 mg) (110). Clinically
meaningful changes in GFR measures are limited to
individuals with baseline morbid obesity, hyperfiltration,
and substantial weight loss with bariatric surgery (110).
Interventions for Obese Patients with CKD
It remains controversial whether persons with
established CKD, especially advanced CKD and dialysisdependent CKD, will benefit from caloric restriction.
The effect of obesity on CKD and mortality differs by
age, presence of obesity-related comorbidities, such as
hypertension and diabetes, and expected years of
survival. Other factors, including patient preference
and nutritional status, must also be considered when
determining the risks and benefits of weight loss for an
individual patient. In an older patient with limited
years of remaining life, the benefits of weight loss will
unlikely outweigh the risks. Several review articles have
provided outlines for weight management strategies in
obese patients with CKD and may help guide clinicians
to initiate discussions with their patients (111–113).
Certainly, weight management interventions should include approaches that increase physical activity levels.
However, given the limited time for a clinic visit and the
multiple aspects of CKD care that must be discussed,
289
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
most clinicians will struggle to adequately discuss nutritional interventions with their obese patients with CKD.
Fortunately, the Affordable Care Act expanded
preventive services, including medical nutritional therapies, for people receiving Medicare. These preventive
services are now provided with no out-of-pocket
expenses. Medicare will pay for three visits per year
for medical nutritional counseling services with a registered dietician or nutritionist for Medicare Part B
enrollees with diabetes, with CKD, or in persons who
received a kidney transplant within 3 years of the clinic
visit (114). During the first year, patients are eligible for
3 hours of either face-to-face counseling or counseling
through an interactive telecommunications system. During subsequent years, patients are eligible for 2 hours of
annual counseling. Unfortunately, younger patients with
nondialysis CKD are not covered by Medicare. These
individuals may benefit greatly from nutritional counseling
services, but cost frequently precludes the acquisition of
these services. Peer-to-peer support facilitates weight loss,
and a multitude of online support systems exist and are
relatively inexpensive. Weight Watchers and other weight
loss programs that include peer support systems can be
effective but may not be affordable for some patients.
Weight Loss and Bariatric Surgery
For morbidly obese individuals, dietary interventions fail and fail consistently. For morbidly obese
patients with CKD, many clinicians are hesitant to
pursue bariatric surgery for obesity management given
the lack of large–scale clinical trials showing safety in
this population. During years 2002–2011, in total, 1114
patients with dialysis-dependent CKD and 395 recipients of kidney transplants underwent bariatric surgery
for obesity management in the United States. Most
surgeries were done by open or laparoscopic bypass
procedures. The mean BMI of these patients before
surgery was 42.1 kg/m2, and the maximum weight loss
was achieved at 18 months, with mean BMI plateauing
around 35 kg/m2 (115). The 30-day postoperative
mortality rate was 1.3%, similar to mortality rates
among patients without CKD. After 3 years, survival
rates were 92% among the recipients of transplants and
79% among patients receiving dialysis (115). Without
clinical trial data, evaluating the long-term benefits
versus risks of bariatric surgery for obesity management in adults with CKD remains extremely difficult.
AKI may occur in up to 8% of patients who have
undergone bariatric surgery, and hospital stay, post-
operative complications, and kidney stones may be
higher in patients with CKD than patients without
CKD (116–118). However, bariatric surgery does lead
to sustained weight loss (over 3 years) and facilitates
kidney transplantation in obese patients otherwise excluded from transplantation because of body size (115).
In a decision analysis of weight loss strategies comparing
Roux-en-Y gastric bypass (RYGB) to diet and exercise
in patients precluded from transplantation because of
obesity, Choudhury et al. (119) determined that RYGB
led to shorter transplantation wait times and longer
survival compared with diet and exercise. Patients with
RYGB and BMI$40 kg/m2 gained 5.4 years of life
compared with only 2.8 years of life gained through
sustained diet and exercise of at least 2 years. Data from
the US Renal Data System showed that .70% of
patients with ESRD who underwent bariatric surgery
while on a transplant waiting list received a kidney
transplant (120). Thus, bariatric surgery alone may
facilitate kidney transplantation in a timely manner and
improve survival for patients precluded from kidney
transplantation because of obesity.
Conclusions
Obesity remains one of the most important
modifiable risk factors for CKD, which is attributable
to its tremendous effect on diabetes, hypertension, and
multiple aspects of glomerular hemodynamics as well
as podocyte function and the response to injury of
these cells. At a minimum, patients with CKD should
receive medical nutritional counseling with an individualized plan to optimize their diet and reduce
intake of processed foods and salt. Diet and exercise
frequently fail to enhance weight loss in morbidly
obese patients, and bariatric surgery should be considered, especially if morbid obesity precludes kidney
transplantation. Public policy efforts to address the
obesity epidemic must include the nephrology community, because obesity effects CKD risks and outcomes, kidney transplantation rates, and incidence and
prevalence of ESRD.
Acknowledgments
The authors thank Tom Mattix for his graphical expertise and
contribution of a figure to this manuscript.
Disclosures
None.
290
References
1. Flegal KM, Carroll MD, Ogden CL, Curtin LR: Prevalence and trends
in obesity among US adults, 1999-2008. JAMA 303: 235–241, 2010
PubMed
2. Sturm R: Increases in clinically severe obesity in the United States,
1986-2000. Arch Intern Med 163: 2146–2148, 2003 PubMed
3. Finkelstein EA, Trogdon JG, Cohen JW, Dietz W: Annual medical
spending attributable to obesity: Payer-and service-specific estimates.
Health Aff (Millwood) 28: w822–w831, 2009 PubMed
4. Wang Y, Chen X, Song Y, Caballero B, Cheskin LJ: Association
between obesity and kidney disease: A systematic review and metaanalysis. Kidney Int 73: 19–33, 2008 PubMed
5. Stenvinkel P, Ikizler TA, Mallamaci F, Zoccali C: Obesity and
nephrology: Results of a knowledge and practice pattern survey.
Nephrol Dial Transplant 28[Suppl 4]: iv99–iv104, 2013 PubMed
6. Finkelstein EA, Zuckerman L: The Fattening of America, Hoboken,
NJ, John Wiley and Sons, Inc., 2008
7. Mägi R, Manning S, Yousseif A, Pucci A, Santini F, Karra E, Querci
G, Pelosini C, McCarthy MI, Lindgren CM, Batterham RL: Contribution of 32 GWAS-identified common variants to severe obesity in
European adults referred for bariatric surgery. PLoS One 8: e70735,
2013 PubMed
8. Burger KS, Stice E: Variability in reward responsivity and obesity:
Evidence from brain imaging studies. Curr Drug Abuse Rev 4: 182–
189, 2011 PubMed
9. Brooks SJ, Cedernaes J, Schiöth HB: Increased prefrontal and parahippocampal activation with reduced dorsolateral prefrontal and
insular cortex activation to food images in obesity: A meta-analysis
of fMRI studies. PLoS One 8: e60393, 2013 PubMed
10. Cocores JA, Gold MS: The Salted Food Addiction Hypothesis may
explain overeating and the obesity epidemic. Med Hypotheses 73:
892–899, 2009 PubMed
11. Garber AK, Lustig RH: Is fast food addictive? Curr Drug Abuse Rev 4:
146–162, 2011 PubMed
12. Hsu CY, McCulloch CE, Iribarren C, Darbinian J, Go AS: Body mass
index and risk for end-stage renal disease. Ann Intern Med 144: 21–28,
2006 PubMed
13. Vivante A, Golan E, Tzur D, Leiba A, Tirosh A, Skorecki K, CalderonMargalit R: Body mass index in 1.2 million adolescents and risk for endstage renal disease. Arch Intern Med 172: 1644–1650, 2012 PubMed
14. Panwar B, Hanks LJ, Tanner RM, Muntner P, Kramer H, McClellan
WM, Warnock DG, Judd SE, Gutiérrez OM: Obesity, metabolic
health, and the risk of end-stage renal disease. Kidney Int 87: 1216–
1222, 2015 PubMed
15. Franceschini N, Gouskova NA, Reiner AP, Bostom A, Howard BV,
Pettinger M, Umans JG, Brookhart MA, Winkelmayer WC, Eaton CB,
Heiss G, Fine JP: Adiposity patterns and the risk for ESRD in
postmenopausal women. Clin J Am Soc Nephrol 10: 241–250, 2015
PubMed
16. Munkhaugen J, Lydersen S, Widerøe TE, Hallan S: Prehypertension,
obesity, and risk of kidney disease: 20-year follow-up of the HUNT I
study in Norway. Am J Kidney Dis 54: 638–646, 2009 PubMed
17. Kramer H, Shoham D, McClure LA, Durazo-Arvizu R, Howard G,
Judd S, Muntner P, Safford M, Warnock DG, McClellan W:
Association of waist circumference and body mass index with allcause mortality in CKD: The REGARDS (Reasons for Geographic and
Racial Differences in Stroke) Study. Am J Kidney Dis 58: 177–185,
2011 PubMed
18. Elsayed EF, Sarnak MJ, Tighiouart H, Griffith JL, Kurth T, Salem DN,
Levey AS, Weiner DE: Waist-to-hip ratio, body mass index, and
subsequent kidney disease and death. Am J Kidney Dis 52: 29–38,
2008 PubMed
19. Kovesdy CP, Czira ME, Rudas A, Ujszaszi A, Rosivall L, Novak M,
Kalantar-Zadeh K, Molnar MZ, Mucsi I: Body mass index, waist
circumference and mortality in kidney transplant recipients. Am J
Transplant 10: 2644–2651, 2010 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
20. Guilherme A, Virbasius JV, Puri V, Czech MP: Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev
Mol Cell Biol 9: 367–377, 2008 PubMed
21. Flegal KM, Graubard BI, Williamson DF, Gail MH: Cause-specific
excess deaths associated with underweight, overweight, and obesity.
JAMA 298: 2028–2037, 2007 PubMed
22. Grabowski DC, Ellis JE: High body mass index does not predict
mortality in older people: Analysis of the Longitudinal Study of
Aging. J Am Geriatr Soc 49: 968–979, 2001 PubMed
23. Peeters A, Barendregt JJ, Willekens F, Mackenbach JP, Al Mamun A,
Bonneux L; NEDCOM, the Netherlands Epidemiology and
Demography Compression of Morbidity Research Group: Obesity
in adulthood and its consequences for life expectancy: A life-table
analysis. Ann Intern Med 138: 24–32, 2003 PubMed
24. Schmidt M, Johannesdottir SA, Lemeshow S, Lash TL, Ulrichsen SP,
Botker HE, Sorensen HT: Obesity in young men, and individual and
combined risks of type 2 diabetes, cardiovascular morbidity and death
before 55 years of age: A Danish 33-year follow-up study. BMJ Open
3: e002698, 2013
25. Owen OE, Felig P, Morgan AP, Wahren J, Cahill GF Jr.: Liver and
kidney metabolism during prolonged starvation. J Clin Invest 48: 574–
583, 1969 PubMed
26. Klahr S, Alleyne GA: Effects of chronic protein-calorie malnutrition
on the kidney. Kidney Int 3: 129–141, 1973 PubMed
27. Levey AS, Kramer H: Obesity, glomerular hyperfiltration, and the
surface area correction. Am J Kidney Dis 56: 255–258, 2010 PubMed
28. Hall JE, Jones DW, Kuo JJ, da Silva A, Tallam LS, Liu J: Impact of the
obesity epidemic on hypertension and renal disease. Curr Hypertens
Rep 5: 386–392, 2003 PubMed
29. Hall JE, Kuo JJ, da Silva AA, de Paula RB, Liu J, Tallam L: Obesityassociated hypertension and kidney disease. Curr Opin Nephrol
Hypertens 12: 195–200, 2003 PubMed
30. Helal I, Fick-Brosnahan GM, Reed-Gitomer B, Schrier RW: Glomerular hyperfiltration: Definitions, mechanisms and clinical implications.
Nat Rev Nephrol 8: 293–300, 2012 PubMed
31. Hostetter TH, Rennke HG, Brenner BM: Compensatory renal hemodynamic injury: A final common pathway of residual nephron destruction. Am J Kidney Dis 1: 310–314, 1982 PubMed
32. Hostetter TH, Rennke HG, Brenner BM: The case for intrarenal
hypertension in the initiation and progression of diabetic and other
glomerulopathies. Am J Med 72: 375–380, 1982 PubMed
33. Brenner BM, Hostetter TH, Olson JL, Rennke HG, Venkatachalam
MA: The role of glomerular hyperfiltration in the initiation and
progression of diabetic nephropathy. Acta Endocrinol Suppl (Copenh)
242: 7–10, 1981 PubMed
34. Hostetter TH, Olson JL, Rennke HG, Venkatachalam MA, Brenner
BM: Hyperfiltration in remnant nephrons: A potentially adverse
response to renal ablation. Am J Physiol 241: F85–F93, 1981 PubMed
35. Blantz RC, Gabbai FB: Glomerular hemodynamics in pathophysiologic conditions. Am J Hypertens 2: 208S–212S, 1989 PubMed
36. Lenihan CR, Busque S, Derby G, Blouch K, Myers BD, Tan JC:
Longitudinal study of living kidney donor glomerular dynamics after
nephrectomy. J Clin Invest 125: 1311–1318, 2015 PubMed
37. Kwakernaak AJ, Toering TJ, Navis G: Body mass index and body fat
distribution as renal risk factors: A focus on the role of renal haemodynamics. Nephrol Dial Transplant 28[Suppl 4]: iv42–iv49, 2013 PubMed
38. Ibrahim HN, Foley R, Tan L, Rogers T, Bailey RF, Guo H, Gross CR,
Matas AJ: Long-term consequences of kidney donation. N Engl J Med
360: 459–469, 2009 PubMed
39. Muzaale AD, Massie AB, Wang MC, Montgomery RA, McBride MA,
Wainright JL, Segev DL: Risk of end-stage renal disease following
live kidney donation. JAMA 311: 579–586, 2014 PubMed
40. Hoy WE, Douglas-Denton RN, Hughson MD, Cass A, Johnson K,
Bertram JF: A stereological study of glomerular number and volume:
Preliminary findings in a multiracial study of kidneys at autopsy.
Kidney Int Suppl 83: S31–S37, 2003 PubMed
291
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
41. Kasiske BL, Napier J: Glomerular sclerosis in patients with massive
obesity. Am J Nephrol 5: 45–50, 1985 PubMed
42. Rennke HG, Klein PS: Pathogenesis and significance of nonprimary
focal and segmental glomerulosclerosis. Am J Kidney Dis 13: 443–
456, 1989 PubMed
43. Wiggins JE, Goyal M, Sanden SK, Wharram BL, Shedden KA, Misek
DE, Kuick RD, Wiggins RC: Podocyte hypertrophy, “adaptation,” and
“decompensation” associated with glomerular enlargement and glomerulosclerosis in the aging rat: Prevention by calorie restriction. J Am
Soc Nephrol 16: 2953–2966, 2005 PubMed
44. Griffin KA, Kramer H, Bidani AK: Adverse renal consequences of
obesity. Am J Physiol Renal Physiol 294: F685–F696, 2008 PubMed
45. Ahmed SB, Fisher ND, Stevanovic R, Hollenberg NK: Body mass
index and angiotensin-dependent control of the renal circulation in
healthy humans. Hypertension 46: 1316–1320, 2005 PubMed
46. Krikken JA, Laverman GD, Navis G: Benefits of dietary sodium
restriction in the management of chronic kidney disease. Curr Opin
Nephrol Hypertens 18: 531–538, 2009 PubMed
47. ALLHAT Officers and Coordinators for the ALLHAT Collaborative
Research Group. The Antihypertensive and Lipid-Lowering Treatment
to Prevent Heart Attack Trial: Major outcomes in moderately hypercholesterolemic, hypertensive patients randomized to pravastatin vs
usual care: The Antihypertensive and Lipid-Lowering Treatment to
Prevent Heart Attack Trial (ALLHAT-LLT). JAMA 288: 2998–3007,
2002 PubMed
48. Rahman M, Ford CE, Cutler JA, Davis BR, Piller LB, Whelton PK,
Wright JT Jr., Barzilay JI, Brown CD, Colon PJ Sr., Fine LJ, Grimm
RH Jr., Gupta AK, Baimbridge C, Haywood LJ, Henriquez MA,
Ilamaythi E, Oparil S, Preston R; ALLHAT Collaborative Research
Group: Long-term renal and cardiovascular outcomes in Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial
(ALLHAT) participants by baseline estimated GFR. Clin J Am Soc
Nephrol 7: 989–1002, 2012 PubMed
49. Griffin KA, Bidani AK: Potential risks of calcium channel blockers in
chronic kidney disease. Curr Cardiol Rep 10: 448–455, 2008 PubMed
50. Toto RD: Lessons from the African-American Study of Kidney
Disease and Hypertension: An update. Curr Hypertens Rep 8: 409–
412, 2006 PubMed
51. Tayo BO, Luke A, McKenzie CA, Kramer H, Cao G, Durazo-Arvizu
R, Forrester T, Adeyemo AA, Cooper RS: Patterns of sodium and
potassium excretion and blood pressure in the African Diaspora.
J Hum Hypertens 26: 315–324, 2012
52. Song HJ, Cho YG, Lee HJ: Dietary sodium intake and prevalence of
overweight in adults. Metabolism 62: 703–708, 2013 PubMed
53. Lax DS, Benstein JA, Tolbert E, Dworkin LD: Effects of salt
restriction on renal growth and glomerular injury in rats with remnant
kidneys. Kidney Int 41: 1527–1534, 1992 PubMed
54. Dworkin LD, Benstein JA, Tolbert E, Feiner HD: Salt restriction
inhibits renal growth and stabilizes injury in rats with established renal
disease. J Am Soc Nephrol 7: 437–442, 1996 PubMed
55. Saeed A, Dibona GF, Marcussen N, Guron G: High-NaCl intake
impairs dynamic autoregulation of renal blood flow in ANG II-infused
rats. Am J Physiol Regul Integr Comp Physiol 299: R1142–R1149,
2010 PubMed
56. Saeed A, DiBona GF, Grimberg E, Nguy L, Mikkelsen ML,
Marcussen N, Guron G: High-NaCl diet impairs dynamic renal blood
flow autoregulation in rats with adenine-induced chronic renal failure.
Am J Physiol Regul Integr Comp Physiol 306: R411–R419, 2014
PubMed
57. Fellner RC, Cook AK, O’Connor PM, Zhang S, Pollock DM, Inscho
EW: High-salt diet blunts renal autoregulation by a reactive oxygen
species-dependent mechanism. Am J Physiol Renal Physiol 307: F33–
F40, 2014 PubMed
58. Abu-Amarah I, Bidani AK, Hacioglu R, Williamson GA, Griffin KA:
Differential effects of salt on renal hemodynamics and potential pressure transmission in stroke-prone and stroke-resistant spontaneously
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
hypertensive rats. Am J Physiol Renal Physiol 289: F305–F313, 2005
PubMed
Aviv A, Hollenberg NK, Weder AB: Sodium glomerulopathy:
Tubuloglomerular feedback and renal injury in African Americans.
Kidney Int 65: 361–368, 2004 PubMed
Kitiyakara C, Chabrashvili T, Chen Y, Blau J, Karber A, Aslam S,
Welch WJ, Wilcox CS: Salt intake, oxidative stress, and renal
expression of NADPH oxidase and superoxide dismutase. J Am Soc
Nephrol 14: 2775–2782, 2003 PubMed
Weir MR, Fink JC: Salt intake and progression of chronic kidney
disease: An overlooked modifiable exposure? A commentary. Am J
Kidney Dis 45: 176–188, 2005 PubMed
Ying WZ, Sanders PW: Dietary salt modulates renal production of
transforming growth factor-beta in rats. Am J Physiol 274: F635–F641,
1998 PubMed
Ying WZ, Sanders PW: Dietary salt increases endothelial nitric oxide
synthase and TGF-beta1 in rat aortic endothelium. Am J Physiol 277:
H1293–H1298, 1999 PubMed
Krikken JA, Lely AT, Bakker SJ, Navis G: The effect of a shift in
sodium intake on renal hemodynamics is determined by body mass
index in healthy young men. Kidney Int 71: 260–265, 2007 PubMed
Moshfegh A, Goldman J, Cleveland L: What We Eat in America,
NHANES 2001–2002. Usual Nutrient Intake from Foods as Compared
to Dietary Reference Intakes. US Department of Agriculture, Agricultural Research Service, 2005. Available at: http://www.Ars.Usda.gov/
SP2UserFiles/Place/12355000/pdf/usualintaketables2001-02. Accessed
April 30, 2015
Institute of Medicine of the National Academies: Dietary Reference
Intakes for Energy, Carbohydrate, Fat, Fatty Acids, Cholesterol,
Protein, and Amino Acids. Panel on Macronutrients, Panel on the
Definition of Dietary Fiber, Subcommittee on Upper Reference Levels
of Nutrients, Subcommitte on Interpretation and Uses of Dietary
Reference Intakes, and the Standing Committee on the Scientific
Evaluation of Dietary Reference Intakes, Food and Nutrition Board,
Washington, DC, The National Academies Press, 2005
Austin GL, Ogden LG, Hill JO: Trends in carbohydrate, fat, and
protein intakes and association with energy intake in normal-weight,
overweight, and obese individuals: 1971-2006. Am J Clin Nutr 93:
836–843, 2011 PubMed
Ando A, Kawata T, Hara Y, Yaegashi M, Arai J, Sugino N: Effects of
dietary protein intake on renal function in humans. Kidney Int Suppl
27: S64–S67, 1989 PubMed
Landau D, Rabkin R: Effect of nutritional status and changes in protein
intake on renal function. In: Nutritional Management of Renal
Disease, 3rd Ed., edited by Kopple JD, Massry SG, Kalantar-Zadeh
K, Amsterdam, Elsevier, 2013, pp 197–207
Friedman AN, Yu Z, Juliar BE, Nguyen JT, Strother M, Quinney SK,
Li L, Inman M, Gomez G, Shihabi Z, Moe S: Independent influence of
dietary protein on markers of kidney function and disease in obesity.
Kidney Int 78: 693–697, 2010 PubMed
Don BR, Blake S, Hutchison FN, Kaysen GA, Schambelan M: Dietary
protein intake modulates glomerular eicosanoid production in the rat.
Am J Physiol 256: F711–F718, 1989 PubMed
King AJ, Troy JL, Anderson S, Neuringer JR, Gunning M, Brenner BM:
Nitric oxide: A potential mediator of amino acid-induced renal hyperemia
and hyperfiltration. J Am Soc Nephrol 1: 1271–1277, 1991 PubMed
Murakami M, Suzuki H, Ichihara A, Naitoh M, Nakamoto H, Saruta T:
Effects of L-arginine on systemic and renal haemodynamics in
conscious dogs. Clin Sci (Lond) 81: 727–732, 1991 PubMed
Kasiske BL, Lakatua JD, Ma JZ, Louis TA: A meta-analysis of the
effects of dietary protein restriction on the rate of decline in renal
function. Am J Kidney Dis 31: 954–961, 1998 PubMed
Centers for Disease Control and Prevention (CDC): State-specific
trends in fruit and vegetable consumption among adults — United
States, 2000-2009. MMWR Morb Mortal Wkly Rep 59: 1125–1130,
2010 PubMed
292
76. Scialla JJ, Anderson CA: Dietary acid load: A novel nutritional target
in chronic kidney disease? Adv Chronic Kidney Dis 20: 141–149, 2013
PubMed
77. Wesson DE, Jo CH, Simoni J: Angiotensin II receptors mediate
increased distal nephron acidification caused by acid retention. Kidney
Int 82: 1184–1194, 2012 PubMed
78. Slavin JL: Position of the American Dietetic Association: Health
implications of dietary fiber. J Am Diet Assoc 108: 1716–1731, 2008
PubMed
79. Kuo SM: The interplay between fiber and the intestinal microbiome in
the inflammatory response. Adv Nutr 4: 16–28, 2013 PubMed
80. Ajani UA, Ford ES, Mokdad AH: Dietary fiber and C-reactive protein:
Findings from national health and nutrition examination survey data.
J Nutr 134: 1181–1185, 2004 PubMed
81. Ma Y, Hébert JR, Li W, Bertone-Johnson ER, Olendzki B, Pagoto SL,
Tinker L, Rosal MC, Ockene IS, Ockene JK, Griffith JA, Liu S:
Association between dietary fiber and markers of systemic inflammation in the Women’s Health Initiative Observational Study. Nutrition
24: 941–949, 2008 PubMed
82. Lin J, Hu FB, Mantzoros C, Curhan GC: Lipid and inflammatory
biomarkers and kidney function decline in type 2 diabetes. Diabetologia 53: 263–267, 2010 PubMed
83. Shankar A, Sun L, Klein BE, Lee KE, Muntner P, Nieto FJ, Tsai MY,
Cruickshanks KJ, Schubert CR, Brazy PC, Coresh J, Klein R: Markers
of inflammation predict the long-term risk of developing chronic
kidney disease: A population-based cohort study. Kidney Int 80: 1231–
1238, 2011 PubMed
84. Bash LD, Erlinger TP, Coresh J, Marsh-Manzi J, Folsom AR, Astor
BC: Inflammation, hemostasis, and the risk of kidney function decline
in the Atherosclerosis Risk in Communities (ARIC) Study. Am J
Kidney Dis 53: 596–605, 2009 PubMed
85. Hiramoto JS, Katz R, Peralta CA, Ix JH, Fried L, Cushman M, Siscovick
D, Palmas W, Sarnak M, Shlipak MG: Inflammation and coagulation
markers and kidney function decline: The Multi-Ethnic Study of
Atherosclerosis (MESA). Am J Kidney Dis 60: 225–232, 2012 PubMed
86. Liévin V, Peiffer I, Hudault S, Rochat F, Brassart D, Neeser JR, Servin
AL: Bifidobacterium strains from resident infant human gastrointestinal
microflora exert antimicrobial activity. Gut 47: 646–652, 2000 PubMed
87. Langlands SJ, Hopkins MJ, Coleman N, Cummings JH: Prebiotic
carbohydrates modify the mucosa associated microflora of the human
large bowel. Gut 53: 1610–1616, 2004 PubMed
88. Sabatino A, Regolisti G, Brusasco I, Cabassi A, Morabito S,
Fiaccadori E: Alterations of intestinal barrier and microbiota in chronic
kidney disease. Nephrol Dial Transplant 30: 924–933, 2015
89. Sibley SD, de Boer IH, Steffes MW, Brunzell JD; Diabetes Control
and Complications Trial/Epidemiology of Diabetes Interventions and
Complications (DCCT/EDIC) Research Study Group: Intra-abdominal
fat and elevated urine albumin excretion in men with type 1 diabetes.
Diabetes Care 30: 1898–1900, 2007 PubMed
90. Kramer H, Reboussin D, Bertoni AG, Marcovina S, Lipkin E,
Greenway FL 3rd, Brancati FL; Look Ahead Research Group: Obesity
and albuminuria among adults with type 2 diabetes: The Look
AHEAD (Action for Health in Diabetes) Study. Diabetes Care 32:
851–853, 2009 PubMed
91. Foster MC, Hwang SJ, Massaro JM, Hoffmann U, DeBoer IH, Robins
SJ, Vasan RS, Fox CS: Association of subcutaneous and visceral
adiposity with albuminuria: The Framingham Heart Study. Obesity
(Silver Spring) 19: 1284–1289, 2011 PubMed
92. Cirillo M, Senigalliesi L, Laurenzi M, Alfieri R, Stamler J, Stamler R,
Panarelli W, De Santo NG: Microalbuminuria in nondiabetic adults:
Relation of blood pressure, body mass index, plasma cholesterol
levels, and smoking: The Gubbio Population Study. Arch Intern Med
158: 1933–1939, 1998 PubMed
93. Rossing P, Hommel E, Smidt UM, Parving HH: Impact of arterial
blood pressure and albuminuria on the progression of diabetic
nephropathy in IDDM patients. Diabetes 42: 715–719, 1993 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
94. Ruggenenti P, Perna A, Mosconi L, Pisoni R, Remuzzi G: Urinary
protein excretion rate is the best independent predictor of ESRF in nondiabetic proteinuric chronic nephropathies. “Gruppo Italiano di Studi
Epidemiologici in Nefrologia” (GISEN). Kidney Int 53: 1209–1216,
1998 PubMed
95. Hoy WE, Wang Z, VanBuynder P, Baker PR, Mathews JD: The
natural history of renal disease in Australian Aborigines. Part 1.
Changes in albuminuria and glomerular filtration rate over time.
Kidney Int 60: 243–248, 2001 PubMed
96. Klahr S, Levey AS, Beck GJ, Caggiula AW, Hunsicker L, Kusek JW,
Striker G; Modification of Diet in Renal Disease Study Group: The
effects of dietary protein restriction and blood-pressure control on the
progression of chronic renal disease. N Engl J Med 330: 877–884,
1994 PubMed
97. Cushman WC, Evans GW, Byington RP, Goff DC Jr., Grimm RH Jr.,
Cutler JA, Simons-Morton DG, Basile JN, Corson MA, Probstfield JL,
Katz L, Peterson KA, Friedewald WT, Buse JB, Bigger JT, Gerstein
HC, Ismail-Beigi F; ACCORD Study Group: Effects of intensive
blood-pressure control in type 2 diabetes mellitus. N Engl J Med 362:
1575–1585, 2010 PubMed
98. Afshinnia F, Wilt TJ, Duval S, Esmaeili A, Ibrahim HN: Weight loss
and proteinuria: Systematic review of clinical trials and comparative
cohorts. Nephrol Dial Transplant 25: 1173–1183, 2010 PubMed
99. Shen WW, Chen HM, Chen H, Xu F, Li LS, Liu ZH: Obesity-related
glomerulopathy: Body mass index and proteinuria. Clin J Am Soc
Nephrol 5: 1401–1409, 2010 PubMed
100. Amor A, Jiménez A, Moizé V, Ibarzabal A, Flores L, Lacy AM, Vidal
J: Weight loss independently predicts urinary albumin excretion
normalization in morbidly obese type 2 diabetic patients undergoing
bariatric surgery. Surg Endosc 27: 2046–2051, 2013 PubMed
101. Chagnac A, Weinstein T, Herman M, Hirsh J, Gafter U, Ori Y: The
effects of weight loss on renal function in patients with severe obesity.
J Am Soc Nephrol 14: 1480–1486, 2003 PubMed
102. Blum CA, Ellis JL, Loh C, Ng PY, Perni RB, Stein RL: SIRT1
modulation as a novel approach to the treatment of diseases of aging.
J Med Chem 54: 417–432, 2011 PubMed
103. He W, Wang Y, Zhang MZ, You L, Davis LS, Fan H, Yang HC, Fogo
AB, Zent R, Harris RC, Breyer MD, Hao CM: Sirt1 activation protects
the mouse renal medulla from oxidative injury. J Clin Invest 120:
1056–1068, 2010 PubMed
104. Qiao L, Shao J: SIRT1 regulates adiponectin gene expression through
Foxo1-C/enhancer-binding protein alpha transcriptional complex.
J Biol Chem 281: 39915–39924, 2006 PubMed
105. Rutkowski JM, Wang ZV, Park AS, Zhang J, Zhang D, Hu MC, Moe
OW, Susztak K, Scherer PE: Adiponectin promotes functional recovery after podocyte ablation. J Am Soc Nephrol 24: 268–282, 2013
PubMed
106. Sharma K: The link between obesity and albuminuria: Adiponectin
and podocyte dysfunction. Kidney Int 76: 145–148, 2009 PubMed
107. Sharma K, Ramachandrarao S, Qiu G, Usui HK, Zhu Y, Dunn SR,
Ouedraogo R, Hough K, McCue P, Chan L, Falkner B, Goldstein BJ:
Adiponectin regulates albuminuria and podocyte function in mice.
J Clin Invest 118: 1645–1656, 2008 PubMed
108. Morales E, Valero MA, León M, Hernández E, Praga M: Beneficial
effects of weight loss in overweight patients with chronic proteinuric
nephropathies. Am J Kidney Dis 41: 319–327, 2003 PubMed
109. Praga M, Morales E: Obesity-related renal damage: Changing diet to
avoid progression. Kidney Int 78: 633–635, 2010 PubMed
110. Bolignano D, Zoccali C: Effects of weight loss on renal function in
obese CKD patients: A systematic review. Nephrol Dial Transplant 28
[Suppl 4]: iv82–iv98, 2013 PubMed
111. Stenvinkel P, Zoccali C, Ikizler TA: Obesity in CKD–what should
nephrologists know? J Am Soc Nephrol 24: 1727–1736, 2013 PubMed
112. Kramer H, Tuttle KR, Leehey D, Luke A, Durazo-Arvizu R, Shoham
D, Cooper R, Beddhu S: Obesity management in adults with CKD. Am
J Kidney Dis 53: 151–165, 2009 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
113. Tuttle KR, Sunwold D, Kramer H: Can comprehensive lifestyle
change alter the course of chronic kidney disease? Semin Nephrol
29: 512–523, 2009 PubMed
114. Centers for Medicare & Medicaid Services: Available at: http://www.
Medicare.gov/coverage/nutrition-Therapy-Services. Accessed August
4, 2015
115. US Renal Data System: USRDS 2013 Annual Data Report: Atlas of
Chronic Kidney Disease and End-Stage Renal Disease in the United
States, Bethesda, MD, National Institutes of Health, National Institute
of Diabetes and Digestive and Kidney Diseases, 2010
116. Matlaga BR, Shore AD, Magnuson T, Clark JM, Johns R, Makary
MA: Effect of gastric bypass surgery on kidney stone disease. J Urol
181: 2573–2577, 2009 PubMed
117. Turgeon NA, Perez S, Mondestin M, Davis SS, Lin E, Tata S, Kirk
AD, Larsen CP, Pearson TC, Sweeney JF: The impact of renal function
293
on outcomes of bariatric surgery. J Am Soc Nephrol 23: 885–894, 2012
PubMed
118. Thakar CV, Kharat V, Blanck S, Leonard AC: Acute kidney injury
after gastric bypass surgery. Clin J Am Soc Nephrol 2: 426–430, 2007
PubMed
119. Choudhury RA, Murayama KM, Abt PL, Glick HA, Naji A, Williams
NN, Dumon KR: Roux-en-Y gastric bypass compared with aggressive
diet and exercise therapy for morbidly obese patients awaiting renal
transplant: A decision analysis. Surg Obes Relat Dis 10: 79–87, 2014
PubMed
120. Modanlou KA, Muthyala U, Xiao H, Schnitzler MA, Salvalaggio PR,
Brennan DC, Abbott KC, Graff RJ, Lentine KL: Bariatric surgery
among kidney transplant candidates and recipients: Analysis of the
United States renal data system and literature review. Transplantation
87: 1167–1173, 2009 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Syllabus
NephSAP, Volume 14, Number 4, October 2015—CKD and Progression
Michael Choi, MD,* Linda Fried, MD, MPH, FASN,†‡ and Sumeska Thavarajah, MD*
*Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Marland;
†
Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania; and
‡
University of Pittsburgh Graduate School of Public Health, University of Pittsburgh School of
Medicine, Pittsburgh, Pennsylvania
and Nutrition Examination Survey (NHANES III) in
1988–1994 and the NHANES in 1999–2002. By the
CKD-EPI creatinine equation, the prevalence of reduced eGFR increased from 4.7% to 6.5% between
these two periods. By CKD-EPI creatinine, the prevalence increased from 5.5% to 8.7%, and using
CKD-EPI creatinine-cystatin C formula, prevalence
increased from 4.4% to 7.1%. After adjustments for
changes in the United States population, prevalence
ratios for reduced eGFR in the latter versus former
surveys were 1.24 (95% confidence interval [95% CI],
1.09 to 1.45), 1.34 (95% CI, 1.15 to 1.67), and 1.33
(95% CI, 1.17 to 1.65) by creatinine, cystatin C, and
creatinine-cystatin C, respectively. The prevalence
changes of CKD did not include changes in increased
urine albumin excretion between time periods. Also, in
an accompanying editorial, Hsu and Hsu (2) point out
that, more recently, the prevalence of stages 3 and 4
CKD in the NHANES was stable (8.1% in 2001–2002
and 7.8% in 2009–2010).
Grams et al. (3) examined the lifetime risks for
CKD and ESRD using Markov Monte Carlo models to
simulate kidney disease development. Mortality rates
were obtained from the National Vitals Statistics Report,
mortality risk was obtained from the 2 million person
meta-analyses, CKD prevalence was obtained from the
NHANES, and ESRD incidence, prevalence, and mortality were obtained from the US Renal Data System.
Overall lifetime risks for stages 3a, 3b, 4, and ESRD were
59.1%, 33.6%, 11.5%, and 3.6%, respectively. Women
had greater CKD risk but lower ESRD risk. One half
of the patients with stage 3a CKD developed it over
age 70. Lifetime risks of stages 4 and 5 CKD and
ESRD were higher in blacks and developed 10–15
years earlier than in whites. ESRD lifetime risks for
white women, white men, black women, and black
men were 2.2%, 3.3%, 7.8%, and 8.5%, respectively
Learning Objectives
1. To review new information on risk factors for
development and progression of CKD
2. To examine new information on screening for
CKD and approaches to care for patients with
CKD
3. To discuss new research on clinical risk
factors for and treatment of cardiovascular
disease in CKD
4. To review and describe the new Kidney
Disease: Improving Global Outcomes guidelines for lipid management in nondialysis
CKD
This issue of NephSAP presents an overview of the
new information in nondialysis CKD from January of
2013 to October of 2014. The topic of CKD is broad, and
one issue cannot cover all new research in CKD. This
issue focuses on important areas of clinical relevance to
nephrologists. Not covered in this issue of NephSAP are
glomerular and tubulointerstitial diseases, transplantation,
and divalent ion metabolism, because they have been
recently reviewed in other issues of NephSAP.
CKD Surveillance and Geographic Variability
CKD Prevalence and Lifetime Risk in the United
States
It is unclear if the prevalence of reduced eGFR,
defined as eGFR,60 ml/min per 1.73 m2, in the United
State is increasing. Grams et al. (1) examined trends of
prevalence of CKD by eGFR calculated using Chronic
Kidney Disease Epidemiology Collaboration (CKD-EPI)
creatinine, CKD-EPI cystatin C, and CKD-EPI creatininecystatin C equations. CKD prevalence in adult participants was compared between the Third National Health
294
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
295
Figure 1. Lifetime risks for CKD from stage 3a to end- stage renal disease from Markov Monte Carlo models. Cumulative
incidence from birth by race and sex of CKD stages (A) 3a1, (B) 3b1, and (C) 3b1 and (D) ESRD. Reprinted with permission
from Grams ME, Chow EK, Segev DL, Coresh J: Lifetime incidence of CKD stages 3-5 in the United States. Am J Kidney Dis
62: 245–252, 2013.
(Figure 1). Accompanying editorials to that study ask
readers to consider CKD screening for blacks (4) and
the elderly (5). For those .80 years old, an analysis of
the NHANES data between 1988–1994 and 2005–2010
showed that prevalence of eGFR by CKD-EPI creatinine ,45 ml/min per 1.73 m2 increased from 14.3% to
21.7% (6).
CKD Screening and Monitoring in the United
States
CKD is usually asymptomatic, and awareness of
CKD is low among providers and patients with CKD
(7). Komenda et al. (8) performed a systematic review of
the cost-effectiveness of CKD screening in the general
population as well as in hypertensive and diabetic
populations. There were eight studies that evaluated
the cost-effectiveness of proteinuria and two studies that
evaluated screening with eGFR. In general, the threshold
for an intervention considered as cost-effective is
,$50,000 per quality–adjusted life year (QALY)
gained. Incremental cost–effectiveness ratios for proteinuria screening had ranges from $14,063 to $160,018
per QALY in the general population, from $5,298 to
$54,943 per QALY in the diabetic population, and from
$23,680 to $73,939 per QALY in the hypertensive
population. In patients with diabetes, one study for
eGFR screening reported a cost of $23,680 per QALY
versus $100,253–$109,912 per QALY in two studies of
the general population. The cost-effectiveness of screening various populations was determined by estimates of
the incidence of CKD in the population studied, the
estimated rate of progression in that population, and the
effectiveness of drug therapy for slowing progression.
Komenda et al. (8) concluded that CKD screening is
cost-effective in patients with diabetes and hypertension.
The US Preventive Task Force’s systematic review concluded that there was insufficient evidence to
screen asymptomatic adults (9). The American College
296
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 1. American College of Physicians clinical practice guidelines for CKD
Reprinted with permission from Qaseem A, Hopkins RH Jr., Sweet DE, Starkey M, Shekelle P; Clinical Guidelines Committee of the American College of Physicians: Screening,
monitoring, and treatment of stage 1 to 3 chronic kidney disease: A clinical practice guideline from the American College of Physicians. Ann Intern Med 159: 835–847, 2013.
of Physicians (ACP) published clinical practice guidelines for screening, monitoring, and treatment of adults
with stages 1–3 CKD on the basis of a systematic
evidence review (Table 1) (10). The first guideline did
not recommend screening for CKD in asymptomatic
adults without risk factors for CKD. The major risk
factors for CKD listed included diabetes, hypertension,
and cardiovascular disease. Other risk factors listed were
older age, obesity, family history, and black, NativeAmerican, or Hispanic ethnicity. The ACP guideline
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
statement was on the basis of the lack of randomized,
controlled trials (RCTs) that compared the effect of
systematic CKD screening versus no CKD screening
on clinical outcomes, sources of variability for measuring single urinary albumin excretion, and lack of effectiveness of treatment on clinical outcomes of CKD
identified through screening. There were no RCTs evaluating harms of systematic screening, but expert opinion–
based harms included misclassification with false-positive
tests, adverse effects of unnecessary testing, psychologic
effects of being labeled with a diagnosis of CKD, potential
adverse effects of pharmacologic treatment changes after
diagnosis with CKD, and possible financial ramifications
of CKD diagnosis. The second guideline opposed the
recommendation of testing for proteinuria in adults
with or without diabetes currently taking an angiotensin–
converting enzyme inhibitor (ACEI) or an angiotensin II
receptor blocker (ARB). As above, the guidelines found
no trials that evaluated clinical outcomes for patients with
stages 1 and 2 CKD who were systematically monitored
for worsening kidney function versus no CKD monitoring, usual care, or an alternative CKD monitoring
regimen. There were no RCTs that compared harm for
those monitored versus no monitoring, usual care, or an
alternative CKD monitoring regimen. Potential harms
included incorrect reclassification with possibly increased financial costs attributable to a diagnosis of more
advanced CKD, adverse effects of unnecessary testing,
psychologic effects of being labeled as having CKD, and
adverse events associated with medication changes.
Other published guidelines agree on targeted
screening for those at risk rather than general population
screening. A letter to the editor of the ACP guidelines
asked why the language for screening was written as
a negative (who not to screen rather than a recommendation as a declarative statement as to who should be
screened) (11). Perhaps this was because the ACP
guideline statement found insufficient evidence to
directly recommend screening for CKD in asymptomatic adults, even those with CKD risk factors, which
Qassem et al. (12) reiterated in a separate article on CKD
screening. However, the ACP guideline statement did
not go so far as to suggest limiting screening to only
symptomatic patients with CKD risk factors. The
United States Preventive Services Task Force systematic review of the ACP guideline did acknowledge
that most persons with CKD stages 1 to 3, even those
with diabetes and hypertension, are not clinically
recognized and do not have CKD testing in usual care.
297
Hellemons et al. (13) found low adherence by
primary care providers to nationwide screening guidelines
for annual albuminuria testing and treatment of albuminuria with renin-angiotensin-aldosterone system (RAAS)
blockers. This issue was studied in a cohort of 14,210
patients with type 2 diabetes from The Netherlands. The
urine albumin to creatinine ratio was measured in 56.8% of
patients in 2009; ,25% had annual screening, and .20%
were never screened. RAAS treatment was prescribed in
78% with prevalent microalbuminuria/macroalbuminuria,
66.5% with incident microalbuminuria/macroalbuminuria,
59.3% with normoalbuminuria, and 52.1% of those
without albumin to creatinine ratio measurements.
The second ACP guideline to forego monitoring
proteinuria for those on RAAS blockade with or without
diabetes contrasts with recommendations from other
societies. The American Diabetes Association recommends annual screening for albuminuria in patients with
diabetes and continued monitoring for albuminuria with
treatment (14). The international Kidney Disease: Improving Global Outcomes (KDIGO) clinical practice
guideline for CKD also advocates for at least yearly
monitoring for CKD progression (15). The KDIGO
CKD guideline did not specifically address screening,
but the national Kidney Disease Outcomes Quality
Initiative (KDOQI) commentary on the KDIGO CKD
clinical practice guideline specifically disagreed with the
ACP recommendation to cease monitoring proteinuria
after RAAS blockers had been initiated. The KDOQI
group felt that monitoring proteinuria for prognosis and
not just treatment was important (16). Editorials pointed
out that monitoring proteinuria would allow for possibly
increasing RAAS blockade if significant proteinuria
persisted after the initial dose (17,18). Although hyperkalemia and AKI were acknowledged as potential harms
of RAAS blockade, there was no formal ACP guideline
statement about rechecking potassium and eGFR after
patients had begun RAAS blockade. The prudence of
rechecking these values in patients taking a combination
of RAAS blockers, diuretics, and nonsteroidal anti–
inflammatory drugs, the combination of which can increase the incidence of AKI, was also advocated in
response to the ACP guideline (17,19). Qaseem et al.
(12) did acknowledge that SCr levels should checked for
adverse effects after RAAS blockade in a separate
screening article. In addition, the ACP guideline recommends the use of ACEIs or ARBs in patients with
hypertension and stages 1–3 CKD, regardless of the level
of albumin excretion. The KDIGO guidelines differ in
298
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
recommending ACEIs or ARBs for adults with diabetes
with urine albumin .30 mg/24 h or adults without
diabetes with urine albumin .300 mg/24 h (15). The
ACP guideline recommended initiating a statin if elevated LDL values are above goal. This contrasts with the
KDIGO Work Group recommendations on lipid management of patients of CKD and will be reviewed in the
cardiovascular section of this NephSAP (20).
The ACP guidelines correctly emphasize the indirect costs of screening in those who have false-positive
tests. However, the reliance on RCTs for judging the
efficacy of CKD screening by changes in mortality and
ESRD may be problematic in a population screened with
early to moderate CKD, because CKD is generally
a slowly progressive disease, in which rapid progression
is often defined as loss of eGFR.3–5 ml/min per 1.73 m2
per year. Interventions in well established CKD require
many years of follow-up to show changes in outcome.
Recommending screening to primary care providers for
asymptomatic patients at risk for CKD seems prudent as
does the continued monitoring of proteinuria with or
without RAAS blockade.
countries. In Taiwan in 2003, with a budget of $15
million/yr, a campaign was initiated to ban herbs with
aristolochic acid, increase public awareness, educate
patients, and set up integrated care teams for patients with
CKD. Annual incidence for ESRD decreased from 432 per
million population in 2005 to 361 per million population
in 2010 (25). The program resulted in a savings of $36
million annually with reduced dialysis costs and improved
quality of life. In Uruguay and Chile, the initiation of
prevention programs has resulted in dramatic decreases in
the incidence and prevalence of ESRD. The details of the
surveillance programs in developed countries, such as
the United States, Europe, Australia, England, and Japan,
are discussed in a review by Radhakrishnan et al. (24). An
increase in patients referred for planned initiation of
dialysis was observed after implementation of a CKD
screening and intervention program in England in 2006.
Other countries have demonstrated lower rates of ESRD
after implementation of similar screening and intervention programs. Hopefully, these programs will reduce
complications, deaths, disabilities, and economic burdens associated with CKD throughout the world.
Worldwide Surveillance
Mesoamerican Nephropathy
The number of worldwide deaths between 1990 and
2010 from CKD has increased by 82%, the third largest
increase behind HIV and diabetes according to the Global
Burden of Disease study (21). This figure may be even
higher, because Rao et al. (22) concluded that mortality
from diabetic kidney disease was underestimated by 4- to
9-fold. Jha et al. (23) reviewed the global dimension of
CKD. Prevalence, which is highest in developed countries, is estimated to be 8%–16% worldwide. CKD risk
factors, such as hypertension, diabetes, and obesity,
continue to grow in developing countries. It is projected
that the number of patients with ESRD will increase
disproportionally in the developing countries of India and
China, where the number of elderly is increasing. Worldwide, diabetes is still the leading cause of CKD in
developed and many developing countries. However, in
many parts of the world, CKD affects younger people,
40% of who do not have diabetes or hypertension (24).
GN and other causes, such as herbal and environmental
toxins, are important causes of CKD in developing
countries. The poorest populations are at the highest risk
for CKD. Unfortunately, many of those who develop
ESRD in these countries cannot afford RRT. Screening
and intervention strategies have been implemented,
resulting in reduction of incidence of ESRD in developing
Unexplained kidney disease was first noted in the
early 1990s in Costa Rican sugarcane workers. This
epidemic of CKD has been named Mesoamerican
Nephropathy, and findings of the First International
Research Workshop on Mesoamerican Nephropathy
were reported (26). In their extensive review of the
literature, Correa-Rotter et al. (27) describe this disease,
which affects young men who are agricultural workers,
especially those working in sugarcane fields, and reside
in communities along the Pacific coast of Central
America. Working in extreme heat is a common condition, and the patients with this progressive form of CKD
are often asymptomatic, with normal to slightly elevated
BP and low-grade or non-nephrotic proteinuria. These
patients often manifest hyperuricemia and/or surprisingly,
hypokalemia. A series of eight patients with eGFR
creatinine of 27–79 ml/min per 1.73 m2 underwent renal
biopsy (28). There was extensive glomerulosclerosis
(29%–78%) with signs of chronic glomerular ischemia,
tubular atrophy, and interstitial fibrosis with vascular
lesions. Podocytes showed glomerular injury, and six
of eight specimens revealed the variable presence of
podocyte vacuolization. These features are similar to
a CKD epidemic in Sri Lanka (29). Also, Mesoamerican
Nephropathy has more extensive glomerular involvement
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
compared with Balkan and Chinese Herb Nephropathy.
Correa-Rotter et al. (27) concluded that the histologic
patterns were consistent with glomerular ischemia. Potential causes for the epidemic include heat stress with
volume depletion. In an animal model of CKD generated
by repeated episodes of dehydration, increased serum
osmolality–stimulated aldose reductase activity converted glucose to sorbitol and fructose for additional
metabolism by fructokinase to oxidant mediators (30).
Notably, sugar cane workers often take high fructose–
containing beverages as well as nonsteroidal anti–
inflammatory drugs. Arsenic in drinking water, pesticides,
Leptopirosis, and genetic susceptibility were also possible
mediators of the disease.
References
1. Grams ME, Juraschek SP, Selvin E, Foster MC, Inker LA, Eckfeldt JH,
Levey AS, Coresh J: Trends in the prevalence of reduced GFR in the
United States: A comparison of creatinine- and cystatin C-based
estimates. Am J Kidney Dis 62: 253–260, 2013 PubMed
2. Hsu RK, Hsu CY: Temporal trends in prevalence of CKD: The glass is half
full and not half empty. Am J Kidney Dis 62: 214–216, 2013 PubMed
3. Grams ME, Chow EK, Segev DL, Coresh J: Lifetime incidence of CKD
stages 3-5 in the United States. Am J Kidney Dis 62: 245–252, 2013 PubMed
4. Kiberd BA: Death, taxes, and now CKD. Am J Kidney Dis 62: 220–222,
2013 PubMed
5. Vassalotti JA, Piraino B: Loss of kidney function with aging is a patient
safety hazard. Am J Kidney Dis 62: 217–219, 2013 PubMed
6. Bowling CB, Sharma P, Fox CS, O’Hare AM, Muntner P: Prevalence of
reduced estimated glomerular filtration rate among the oldest old from
1988-1994 through 2005-2010. JAMA 310: 1284–1286, 2013 PubMed
7. Tuot DSPL, Plantinga LC, Hsu CY, Powe NR: Is awareness of chronic
kidney disease associated with evidence-based guideline-concordant
outcomes? Am J Nephrol 35: 191–197, 2012 PubMed
8. Komenda P, Ferguson TW, Macdonald K, Rigatto C, Koolage C, Sood
MM, Tangri N: Cost-effectiveness of primary screening for CKD: A
systematic review. Am J Kidney Dis 63: 789–797, 2014 PubMed
9. Fink HAIA, Ishani A, Taylor BC, Greer NL, MacDonald R, Rossini D,
Sadiq S, Lankireddy S, Kane RL, Wilt TJ: Screening for, monitoring,
and treatment of chronic kidney disease stages 1 to 3: A systematic
review for the U.S. Preventive Services Task Force and for an American
College of Physicians Clinical Practice Guideline. Ann Intern Med 156:
570–581, 2012 PubMed
10. Qaseem A, Wilt T, Denberg TD: Screening, monitoring, and treatment
of stage 1 to 3 chronic kidney disease. Ann Intern Med 161: 83–84,
2014 PubMed
11. Stefan N, Artunc F, Heyne N, Machann J, Schleicher ED, Häring HU:
Obesity and renal disease: Not all fat is created equal and not all obesity
is harmful to the kidneys [published online ahead of print April 20,
2014]. Nephrol Dial Transplant PubMed
12. Qaseem A, Wilt TJ, Cooke M, Denberg TD: The paucity of evidence
supporting screening for stages 1-3 CKD in asymptomatic patients with or
without risk factors. Clin J Am Soc Nephrol 9: 1993–1995, 2014 PubMed
13. Hellemons ME, Denig P, de Zeeuw D, Voorham J, Lambers Heerspink
HJ: Is albuminuria screening and treatment optimal in patients with type
2 diabetes in primary care? Observational data of the GIANTT cohort.
Nephrol Dial Transplant 28: 706–715, 2013 PubMed
14. American Diabetes Association: Executive Summary: Standards of
Medical Care in Diabetes –2014 Diabetes Care 37: Supplement 1 S5–
S13; 2014 PubMed
299
15. KDIGO CKD Work Group: KDIGO 2012 Clinical Practice Guideline
for the Evaluation and Management of Chronic Kidney Disease. Kidney
Int Suppl 3: 1–150, 2013
16. Inker LAAB, Astor BC, Fox CH, Isakova T, Lash JP, Peralta CA,
Kurella Tamura M, Feldman HI: KDOQI US commentary on the 2012
KDIGO clinical practice guideline for the evaluation and management
of CKD. Am J Kidney Dis 63: 713–735, 2014 PubMed
17. Abdel-Kader K: Screening, monitoring, and treatment of stage 1 to 3
chronic kidney disease. Ann Intern Med 161: 83, 2014 PubMed
18. Lambers Heerspink HJ, Gaillard CJ, Gansevoort RT: Screening,
monitoring, and treatment of stage 1 to 3 chronic kidney disease. Ann
Intern Med 161: 82–83, 2014 PubMed
19. Lapi F, Azoulay L, Yin H, Nessim SJ, Suissa S: Concurrent use of
diuretics, angiotensin converting enzyme inhibitors, and angiotensin
receptor blockers with non-steroidal anti-inflammatory drugs and risk
of acute kidney injury: Nested case-control study. BMJ 346: e8525,
2013 PubMed
20. KDIGO Lipid Work Group: KDIGO Clinical Practice Guideline for
Lipid Management in Chronic Kidney Disease. Kidney Int Suppl 3:
259–305, 2013
21. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V,
Abraham J, Adair T, Aggarwal R, Ahn SY, Alvarado M, Anderson HR,
Anderson LM, Andrews KG, Atkinson C, Baddour LM, Barker-Collo
S, Bartels DH, Bell ML, Benjamin EJ, Bennett D, Bhalla K, Bikbov B,
Bin Abdulhak A, Birbeck G, Blyth F, Bolliger I, Boufous S, Bucello C,
Burch M, Burney P, Carapetis J, Chen H, Chou D, Chugh SS, Coffeng
LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD,
Cooper LT, Corriere M, Cortinovis M, de Vaccaro KC, Couser W,
Cowie BC, Criqui MH, Cross M, Dabhadkar KC, Dahodwala N, De
Leo D, Degenhardt L, Delossantos A, Denenberg J, Des Jarlais DC,
Dharmaratne SD, Dorsey ER, Driscoll T, Duber H, Ebel B, Erwin PJ,
Espindola P, Ezzati M, Feigin V, Flaxman AD, Forouzanfar MH,
Fowkes FG, Franklin R, Fransen M, Freeman MK, Gabriel SE, Gakidou
E, Gaspari F, Gillum RF, Gonzalez-Medina D, Halasa YA, Haring D,
Harrison JE, Havmoeller R, Hay RJ, Hoen B, Hotez PJ, Hoy D,
Jacobsen KH, James SL, Jasrasaria R, Jayaraman S, Johns N,
Karthikeyan G, Kassebaum N, Keren A, Khoo JP, Knowlton LM,
Kobusingye O, Koranteng A, Krishnamurthi R, Lipnick M, Lipshultz SE,
Ohno SL, Mabweijano J, MacIntyre MF, Mallinger L, March L, Marks
GB, Marks R, Matsumori A, Matzopoulos R, Mayosi BM, McAnulty
JH, McDermott MM, McGrath J, Mensah GA, Merriman TR, Michaud
C, Miller M, Miller TR, Mock C, Mocumbi AO, Mokdad AA, Moran A,
Mulholland K, Nair MN, Naldi L, Narayan KM, Nasseri K, Norman P,
O’Donnell M, Omer SB, Ortblad K, Osborne R, Ozgediz D, Pahari B,
Pandian JD, Rivero AP, Padilla RP, Perez-Ruiz F, Perico N, Phillips D,
Pierce K, Pope CA 3rd, Porrini E, Pourmalek F, Raju M, Ranganathan
D, Rehm JT, Rein DB, Remuzzi G, Rivara FP, Roberts T, De León FR,
Rosenfeld LC, Rushton L, Sacco RL, Salomon JA, Sampson U, Sanman
E, Schwebel DC, Segui-Gomez M, Shepard DS, Singh D, Singleton J,
Sliwa K, Smith E, Steer A, Taylor JA, Thomas B, Tleyjeh IM, Towbin
JA, Truelsen T, Undurraga EA, Venketasubramanian N, Vijayakumar
L, Vos T, Wagner GR, Wang M, Wang W, Watt K, Weinstock MA,
Weintraub R, Wilkinson JD, Woolf AD, Wulf S, Yeh PH, Yip P,
Zabetian A, Zheng ZJ, Lopez AD, Murray CJ, AlMazroa MA, Memish
ZA: Global and regional mortality from 235 causes of death for 20 age
groups in 1990 and 2010: A systematic analysis for the Global Burden
of Disease Study 2010. Lancet 380: 2095–2128, 2012 PubMed
22. Rao C, Adair T, Bain C, Doi SA: Mortality from diabetic renal disease:
A hidden epidemic. Eur J Public Health 22: 280–284, 2012 PubMed
23. Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, Saran R,
Wang AY, Yang CW: Chronic kidney disease: Global dimension and
perspectives. Lancet 382: 260–272, 2013 PubMed
24. Radhakrishnan J, Remuzzi G, Saran R, Williams DE, Rios-Burrows N,
Powe N, Brück K, Wanner C, Stel VS, Venuthurupalli SK, Hoy WE,
Healy HG, Salisbury A, Fassett RG, O’Donoghue D, Roderick P,
Matsuo S, Hishida A, Imai E, Iimuro S; CDC-CKD Surveillance Team;
300
25.
26.
27.
28.
29.
30.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
European CKD Burden Consortium; CKD.QLD group: Taming the
chronic kidney disease epidemic: A global view of surveillance efforts.
Kidney Int 86: 246–250, 2014 PubMed
Wei SYCY, Chang YY, Mau LW, Lin MY, Chiu HC, Tsai JC, Huang
CJ, Chen HC, Hwang SJ: Chronic kidney disease care program
improves quality of pre-end-stage renal disease care and reduces
medical costs. Nephrology (Carlton) 15: 108–115, 2010 PubMed
Wesseling C, Crowe J, Hogstedt C, Jakobsson K, Lucas R, Wegman
DH; First International Research Workshop on the Mesoamerican
Nephropathy: Resolving the enigma of the mesoamerican nephropathy: A research workshop summary. Am J Kidney Dis 63: 396–404,
2014 PubMed
Correa-Rotter R, Wesseling C, Johnson RJ: CKD of unknown origin in
Central America: The case for a Mesoamerican nephropathy. Am J
Kidney Dis 63: 506–520, 2014 PubMed
Wijkström J, Leiva R, Elinder CG, Leiva S, Trujillo Z, Trujillo L,
Söderberg M, Hultenby K, Wernerson A: Clinical and pathological
characterization of Mesoamerican nephropathy: A new kidney disease
in Central America. Am J Kidney Dis 62: 908–918, 2013 PubMed
Athuraliya NTAT, Abeysekera TD, Amerasinghe PH, Kumarasiri R,
Bandara P, Karunaratne U, Milton AH, Jones AL: Uncertain etiologies
of proteinuric-chronic kidney disease in rural Sri Lanka. Kidney Int 80:
1212–1221, 2011 PubMed
Roncal Jimenez CAIT, Ishimoto T, Lanaspa MA, Rivard CJ, Nakagawa
T, Ejaz AA, Cicerchi C, Inaba S, Le M, Miyazaki M, Glaser J, CorreaRotter R, González MA, Aragón A, Wesseling C, Sánchez-Lozada LG,
Johnson RJ: Fructokinase activity mediates dehydration-induced renal
injury. Kidney Int 86: 294–302, 2014 PubMed
Issues in GFR and Albuminuria Estimation
The exact role for cystatin C in GFR estimation is
debated (1,2). As reviewed in the last NephSAP CKD
issue, the Kidney Disease: Improving Global Outcomes
(KDIGO) 2012 Clinical Practice Guidelines for the
Evaluation and Management of Chronic Kidney Disease recommended using the Chronic Kidney Disease
Epidemiology Collaboration (CKD-EPI) equation instead of the four–variable Modification of Diet in Renal
Disease (MDRD) equation. KDIGO also recommended
the use of cystatin C alone or combined with creatinine
in the 2012 CKD-EPI equations to avoid overdiagnosis
of CKD in individuals with stage 3a CKD without other
markers of kidney damage (3). An important goal for an
eGFR equation using an endogenous filtration marker,
such as creatinine or cystatin C, is how well an equation
predicts clinical outcomes. An equally important but not
necessarily parallel criterion is the formula’s accuracy
compared with measured GFR with an exogenous marker.
Risk prediction by eGFR with cystatin C seems
superior to that by eGFR with creatinine. The Chronic
Kidney Disease Prognosis Consortium included 90,750
participants from the general population and 2960
participants with CKD. Shlipak et al. (2) compared
the association of eGFR using creatinine, cystatin C, or
both with the outcomes of death, cardiovascular death,
and ESRD as well as reclassification. Relative accuracy
between these eGFR formulas could not be assessed,
because measured GFR was not available. Cystatin
C assay was calibrated to the international reference
standard. Reclassification of eGFR to an improved
GFR category by cystatin C compared with creatinine
was associated with reduced risk in all outcomes, and
similarly, reclassification to a worse eGFR category by
cystatin C was associated with increased risk. The net
reclassification improvement for combining all cohorts
for cystatin C versus creatinine was impressively 21%
(95% confidence interval [95% CI], 17% to 26%) for
death but curiously, not significant for ESRD at 3%
(95% CI, 23% to 8%, P¼0.46) and barely significant for
predicting ESRD, thereby limiting analysis to general
population cohorts at 10% (0%–21%, P¼0.05). Interestingly, the combined cystatin C and creatinine equation
did not predict risk of death as well as the cystatin C alone
formula, but the combined equation more accurately
predicted ESRD. Cystatin C seems to be a better biomarker for mortality than ESRD (Figure 2).
eGFR formulas may be superior for predicting
adverse clinical outcomes in patients with CKD compared with measured GFR values. Factors other than
GFR that alter SCr and cystatin C levels include
generation, metabolism, and nonglomerular excretion.
There are also surrogates for these determinants, which
may track with outcomes better than measured GFR. For
example, age is a variable in all eGFR formulas, with the
CKD-EPI cystatin C equation having the largest contribution from age. Non-GFR determinants of cystatin C
may be altered by factors such as inflammation, obesity,
and atherosclerosis. Rule and Glassock (4) evaluated the
degree to which risk factors interacted with different
eGFR formulas relative to their interaction with measured GFR. There were 1150 participants with iothalamate clearance who had CKD-EPI eGFR calculated by
creatinine, cystatin C, or both. One explanation for an
improved risk prediction with certain eGFR formulas
over measured GFR is that there is variability between
measured GFR values compared with the relative
stability of eGFR values. However, in the analysis by
Rule and Glassock (4), the mean variation for measured
GFR in 40 participants who had repeat measurements
was similar to the variation in all of the eGFR formulas
(8.2%, 6.4%, 8.2%, and 10.7% for measured GFR and
CKD-EPI creatinine, creatinine-cystatin C, and cystatin
C alone, respectively). Measured GFR was most accurately estimated by the CKD-EPI creatinine-cystatin C
301
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 3. The association of chronic kidney disease risk factors
with percentage decrease in glomerular filtration rate (Y GFR)
by each method (measured GFR (mGFR), eGFRCr, eGFRCr-CysC,
and eGFRCysC. The * identifies P,0.05 for eGFR compared with
mGFR using model 1 (generalized estimating equations). Risk
factor associations with mGFR were more similar to those with
eGFRCr than with eGFRCr-CysC or eGFRCysC. The one exception
was 24-h urine creatinine, for which eGFRCysC was more similar
to the same association with mGFR. Alb., albumin; BMI, body
mass index; Cr, creatinine; CRP, C-reactive protein; CysC,
cystatin C; eGFR, estimated GFR. Reprinted with permission
from Rule AD, Bailey KR, Lieske JC, Peyser PA, Turner ST:
Estimating the glomerular filtration rate from serum creatinine is
better than from cystatin C for evaluating risk factors associated
with chronic kidney disease. Kidney Int 83: 1173, 2013.
Figure 2. Risk prediction of death, cardiovascular death and endstage renal disease by eGFR from serum creatinine, cystatin C or
combination. Adjusted hazard ratios for (A) all-cause mortality,
(B) cardiovascular death, and (C) ESRD in the general population
cohort studies. Reprinted with permission from Shlipak MG,
Coresh J, Gansevoort RT: Cystatin C versus creatinine for kidney
function-based risk. N Engl J Med 369: 2459, 2013.
formula. Body mass index (BMI), albuminuria .30
mg/g, hypertension, diabetes, and C-reactive protein
tracked with decreased CKD-EPI by cystatin C and
creatinine-cystatin C to a significantly greater degree
than their influence on decreased measured GFR. The
association of risk factors to lower measured GFR was
best approximated by the CKD-EPI creatinine formula
(Figure 3). Rule and Glassock (4) maintain that an eGFR
equation should be judged on how well its associations
with CKD risk factors and outcomes approximate those
measured GFR.
Cystatin C–based eGFR formulas seem better at
predicting CKD complications, perhaps because of nonGFR determinants of serum cystatin C levels. However,
unless combined with SCr, eGFR formulas with cystatin
C are not more accurate in estimating GFR.
Specific Populations
Patients with Diabetes and High Normal GFR.
As discussed in the last NephSAP CKD issue, the
creatinine–based CKD-EPI formula is less likely to misclassify an individual with CKD than the MDRD formula
when measured GFR is close to 60 ml/min per 1.73 m2.
However, it seems to overestimate GFR in older individuals. There are questions regarding this formula’s
302
accuracy in patients with diabetes on the basis of a small
cross–sectional study (5,6). One study compared the
performance of creatinine-based formulas with longitudinal iohexol clearance measurements in 600 Italian patients
with type 2 diabetes over a median of 4 years (6). The
participants followed in the Bergamo Nephrologic Diabetes Complications Trial B Study and the Delapril and
Manidpine for Nephroprotection in Diabetic Nephropathy
Study were .40 years old and had normo- or microalbuminuria. They underwent serial iohexol measurements every 6 months. Overall, there was weak
agreement between measured GFR and eGFR by all of
the creatinine-based formulas. The mean biases between
eGFR and measured eGFR for the MDRD and CKD-EPI
formulas were 216 and 212.5 ml/min per 1.73 m2,
respectively. Detection of participants who were hyperfiltering (.120 ml/min per 1.73 m2) at baseline and
change in short-term GFR was poor. At baseline, 90 of
600 participants had hyperfiltration by measured GFR,
but only 9 were detected by MDRD and 0 were detected
by CKD-EPI formulas. The underestimation by eGFR
formulas at baseline was greatest with the highest
measured GFR values. In addition, eGFR formulas could
not predict short- or long-term declines in measured GFR
over time. In general, the eGFR formulas underestimated
the magnitude of declines in measured GFRs.
In 1441 young participants with type 1 diabetes
from the Diabetes Control and Complications Trial/
Epidemiology of Diabetes Interventions and Complications Study, de Boer et al. (7) compared the accuracy
of longitudinal changes between iothalamate–based
measured GFRs and eGFRs by CKD-EPI creatinine,
CKD-EPI cystatin C, and creatinine-cystatin C combined formulas. Mean baseline iothalamate GFR was
122.7621 ml/min per 1.73 m2. In cross-sectional
analyses, CKD-EPI creatinine-cystatin C had the highest
correlation, precision, and accuracy with measured
GFR. In longitudinal analyses, the change in CKD-EPI
creatinine-cystatin C correlated best with the change in
measured GFR, but the differences between estimating
formulas were small. Over a median of 32 years of
follow-up, mean rates of change between CKD-EPI
creatinine, cystatin, and creatinine-cystatin were 21.37,
21.11, and 21.29 ml/min per 1.73 m2 per year,
respectively. de Boer et al. (7) concluded that adding
cystatin C to creatinine had a modestly greater precision
and accuracy versus eGFR on the basis of creatinine
alone, but this difference was subtle and may not be
meaningful in improving GFR tracking.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
The ability to predict progression to ESRD using
1/SCr, MDRD creatinine, CKD-EPI creatinine, 1/serum
cystatin C, CKD-EPI cystatin C, CKD-EPI creatininecystatin, and measured GFR was assessed in 234 Pima
Indians with type 2 diabetes. Mean baseline measured GFR
was 156 ml/min, with hemoglobin A1C of 9.7, albumin to
creatinine ratio (ACR) .30 mg/g, and age of 43 years old
(8). During median follow-up of 10.7 years, 29% developed
ESRD. After adjustment for age, sex, diabetes duration,
weight, hemoglobin A1C, and ACR, the 1/serum cystatin
C had the best ability to predict ESRD (receiver operating
characteristic- area under the curve¼0.84510.026), whereas
measured GFR and 1/SCr had significantly lower predictive
abilities (0.81460.28 and 0.81560.28, respectively). After
adjustment, CKD-EPI creatinine-cystatin C and cystatin
alone formulas were also significantly better predictors than
measured GFR. Given the risk with hyperfiltration, it is not
surprising that measured GFR would show a J-shaped curve
for association with ESRD. Pavkov et al. (8) concluded that
increased risk prediction using cystatin C was related to
non-GFR determinants, such as inflammation and vascular
remodeling.
In a study of 778 participants with diabetes in the
National Health and Nutrition Examination Survey
(NHANES), which compared CKD-EPI cystatin C
with the CKD-EPI creatinine formula, the cystatin C
equation classified more participants as having reduced
kidney function (22.0% versus 16.5%) (9). For those
with eGFR values of 15 to 30 ml/min per 1.73 m2 by the
cystatin C equation, there was a significantly increased
risk of cardiovascular mortality that was not detected by
the creatinine equation for those in the same category.
In patients with diabetes, the ability of cystatinbased formulas to predict CKD complications seems to
be superior to those of creatinine–based eGFR formulas
and measured GFR. As above, non-GFR determinants of
cystatin C track with risk factors for progression. The
CKD-EPI creatinine-cystatin C formula may best approximate measured GFR, but no formula seems to be ideal for
detecting hyperfiltration and short-term changes.
Asian Populations.
The MDRD and CKD-EPI
creatinine–based equations may not be as accurate in
different populations because of differences in muscle
mass from the cohorts used to develop the equations.
There are modifications of the MDRD equation for
Chinese, Japanese, Thai, and Korean populations (10–
13). The CKD-EPI four–variable ethnicity equation
(black, Asian, Native American and Hispanic, white,
and other) seemed to show increased accuracy in
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Asian populations, especially in those with eGFR
values .60 ml/min per 1.73 m2 (14). In 977 Chinese
participants, which included those with CKD as well as
healthy volunteers, the 2009 CKD-EPI creatinine equation
and modified MDRD creatinine equation for the Chinese
population (10) showed less bias, more precision, and
greater accuracy than the CKD-EPI four–variable or
MDRD creatinine equation compared with measured
99mTc-diethylenetriamine pentaacetic acid clearance
(15). It was unclear why the standard CKD-EPI equation outperformed the four–variable MDRD equation,
although Kong et al. (15) point out that there were only
100 Asians in the four–level development dataset.
There is also a Chinese modification of the CKDEPI creatinine formula (16) that was tested for risk
prediction compared with the modified MDRD creatinine equation in Chinese patients who had a stroke. In
a large cohort of 15,791 individuals with short follow-up,
there was improved risk prediction for all-cause mortality,
stroke recurrence, death, or stroke recurrence and disability with the CKD-EPI modified formula (17).
There have been attempts to modify cystatin C
eGFR equations in the Japanese population. The standard
CKD-EPI cystatin C and CKD-EPI creatinine-cystatin C
formulas as well as modifications for cystatin C–based
formulas were compared with measured inulin clearance.
Interestingly, in 763 Japanese individuals with a mean
measured GFR of 58 ml/min per 1.73 m2, the standard
CKD-EPI cystatin C formula without ethnic modification
seemed to perform as well or better than the other
formulas (18).
In studies reviewed for this issue, population–
modified, creatinine–based formulas seem to be better
for accuracy of GFR and risk prediction. A modified
cystatin C–based formula was not more accurate for GFR
estimation compared with the standard CKD-EPI cystatin C formula.
Pediatrics.
The updated Schwartz equation developed
in the Chronic Kidney Disease in Children (CKiD) Cohort of 349 children ages 1–16 years old with a median
iohexol measured GFR of 41.3 ml/min per 1.73 m2 has
been used to estimate GFR in children with a practical
bedside formula of eGFR¼0.413·(height[cm]/SCr) (19).
Hoste et al. (20) developed an eGFR formula that could
be used in adolescents and young adults with a wider
range of GFR values. The formula was validated in
a European cohort of 750 individuals ages 10–25 years
old with a mean inulin measured GFR of 95 ml/min per
1.73 m2. The formula was eGFR¼107.3/(Scr/(3.94·ht[m]1
303
17.6·ht[m]229.84·ht[m]312.04·ht[m]4 for boys and
girls. The new eGFR equation outperformed other
creatinine–based formulas, including the Schwartz
formula, overall for the spectrum of measured GFR
values from ,60 to .90 ml/min per 1.73 m2; however,
the updated Schwartz equation performed better in the
,60 ml/min per 1.73 m2 range.
Schwartz et al. (21) developed a different combined creatinine and cystatin C equation using the CKiD
Cohort, and this new equation was touted to be more
accurate than the modified Schwartz creatinine formula.
This new formula performs well for GFRs from 15 to
75 ml/min per 1.73 m2 (21), with limitation at higher
GFR levels. The combined Schwartz formula eGFR¼
39.8·(ht[m]/Scr)0.456·(1.8/cystatin C)0.418·(30/BUN)0.0791/
76male·(ht[m]/1.4)0.179 was compared with a combined
serum creatinine-cystatin C formula in 238 children
with a median GFR of 86 ml/min per 1.73 m2 measured by sinistrin clearance (22). The equations are
0.42 · (ht[m]/Scr)20.04 · (ht[m]/Scr) 2 214.5 ·
CysC10.69 · age118.25 for girls and 0.42 · (ht[m]/
Scr)20.04 · (ht[m]/Scr)2214.5 · CysC10.69 · age1
21.88 for boys. The new quadratic combined formula was
felt to outperform the combined Schwartz formula, which
had poor accuracy for sinistrin-based GFRs .91 ml/min
per 1.73 m2.
Severe Obesity.
Although the KDIGO Clinical Practice Guidelines for the Evaluation and Management of
Chronic Kidney Disease advocate the use of the CKDEPI creatinine formula over the MDRD formula, it is
unclear which formula is superior in obese patients with
BMI levels predominantly in the 30- to 35-kg/m2 range
(23,24). These two formulas were compared in 366
obese individuals from Europe (50 of African descent)
with mean BMIs in the severely obese range of 36 kg/m2
and mean measured chromium-51 ethylenediamine tetraacetic acid GFR of 56 ml/min per 1.73 m2 (2,25). In the
overall group, MDRD performed better with less mean
bias (11.9614.3 versus 14.6614.7 ml/min per 1.73 m2
for the MDRD and CKD-EPI equations, respectively)
and greater accuracy (within 30% of the measured value;
80% for MDRD versus 76% with CKD-EPI). In participants with measured GFR .60 ml/min per 1.73 m2,
MDRD again was closer to measured GFR (bias of
14.6618.4 versus 9.3617.2 ml/min per 1.73 m2 and
accuracy within 30% [81% versus 79% for MDRD and
CKD-EPI, respectively]). This study was performed in
a predominantly European cohort with more severe obesity
than prior studies that had compared these two formulas.
304
Advanced CKD.
The MDRD and CKD-EPI equations were derived from cohorts with mean GFRs of 40
and 69 ml/min per 1.73 m2, respectively. Using the
Swedish Renal Registry, Evans et al. (26) examined the
performance of various creatinine–based formulas, among
them the Cockcroft–Gault, MDRD, and CKD-EPI equations, with measured GFRs in 2098 individuals with
stages 4 and 5 CKD. There were 398 participants with
measured GFRs ,10 ml/min per 1.73 m2 and 1974
participants with measured GFRs of 11–20 ml/min per
1.73 m2. For the entire cohort, median bias was 1.2, 1.6,
and 4.6 ml/min per 1.73 m2 for the CKD-EPI, MDRD,
and Cockcroft–Gault formulas, respectively. There was
poor accuracy within 30% of measured GFR for all
formulas (67%, 65%, and 54%, respectively).
Elderly.
The accuracy of estimating equations for the
elderly is unclear, because cohorts used to develop and
validate the MDRD and CKD-EPI formulas had few
individuals .65 years old. In previous studies of the
elderly, the CKD-EPI creatinine formula seemed to
underestimate GFR, resulting in overdiagnosis of CKD.
The accuracy of the MDRD, CKD-EPI creatinine, CKDEPI cystatin C, and CKD-EPI creatinine-cystatin C
formulas was compared with measured GFR by iohexol
clearance in 395 participants from Kent, England who
were .74 years old (27). Mean measured GFR was 53.4
ml/min per 1.73 m2. All of the CKD-EPI formulas
performed slightly better than the MDRD; however, there
was not a tremendous difference. Accuracy within 30%
was not optimal for any formula. Bias was slightly less
with formulas that included cystatin (3.5 [1.9–4.8], 1.7
[0.3–3.2], 21.2 [22.2 to 0], and 0.8 [20.4 to 1.9] ml/min
per 1.73 m2 for the MDRD, CKD-EPI creatinine, CKDEPI cystatin C, and CKD-EPI creatinine-cystatin C
formulas, respectively, with corresponding accuracies
within 30% of measured values of 81%, 83%, 86%, and
86%, respectively). All formulas overestimated GFR for
measured values .60 ml/min per 1.73 m2 to a similar
degree (3.4–5.5 ml/min per 1.73 m2). The differences in
bias for those with measured GFRs ,60 ml/min per
1.73 m2 were smaller: 2 (95% CI, 0.8 to 3.9), 0.6
(95% CI, 20.7 to 2.3), 22.9 (95% CI, 23.7 to 1.9),
and 21.6 (95% CI, 22.8 to 0.2) ml/min per 1.73 m2
for MDRD, CKD-EPI creatinine, CKD-EPI cystatin
C, and CKD-EPI creatinine-cystatin C, respectively.
Kilbride et al. (27) concluded that both creatininebased formulas worked fairly well. This was a
European cohort, which limits application of these
findings to other populations. Improved risk prediction
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
of clinical outcomes for different formulas was not
ascertained.
GFR and Albumin Excretion Variability
The extent of day to day variability of markers for
glomerular filtration and albumin excretion is not well
described. Selvin et al. (28) examined the within-person
variation of serum markers creatinine, cystatin C, b-trace
protein, and b2-microglobulin and urine markers albumin,
creatinine, and ACR in participants of the NHANES III.
These values were separated about 18 days apart. Serum
creatinine (SCr) and cystatin C had the lowest withinperson variability (7.6% and 6.8%, respectively), and although urine albumin alone had a variability of .30%,
the ACR was only 11.3%. If repeat values of ,60 ml/min
per 1.73 m2 were required to define CKD, CKD prevalence would decrease by nearly 20% (17.6% by
CKD-EPI creatinine and 21.1% by CKD-EPI creatininecystatin C formulas). For increased albumin excretion .30 mg/g, requiring two tests to be above this
threshold would decrease the prevalence for albuminuria by about 33% from having only one test above the
threshold.
The requirement for repeat eGFR values ,60
ml/min per 1.73 m2 .3 months apart to define CKD
could also be applied to urine ACR values to define
albuminuria.
References
1. Rule AD, Glassock RJ: GFR estimating equations: Getting closer to the
truth? Clin J Am Soc Nephrol 8: 1414–1420, 2013 PubMed
2. Shlipak MG, Coresh J, Gansevoort RT: Cystatin C versus creatinine for
kidney function-based risk. N Engl J Med 369: 2459, 2013 PubMed
3. KDIGO CKD Work Group: KDIGO 2012 Clinical Practice Guideline
for the Evaluation and Management of Chronic Kidney Disease. Kidney
Int Suppl 3: 1–150, 2013
4. Rule AD, Bailey KR, Lieske JC, Peyser PA, Turner ST: Estimating the
glomerular filtration rate from serum creatinine is better than from
cystatin C for evaluating risk factors associated with chronic kidney
disease. Kidney Int 83: 1169–1176, 2013 PubMed
5. Silveiro SP, Araújo GN, Ferreira MN, Souza FD, Yamaguchi HM,
Camargo EG: Chronic Kidney Disease Epidemiology Collaboration
(CKD-EPI) equation pronouncedly underestimates glomerular filtration
rate in type 2 diabetes. Diabetes Care 34: 2353–2355, 2011 PubMed
6. Gaspari F, Ruggenenti P, Porrini E, Motterlini N, Cannata A, Carrara F,
Jiménez Sosa A, Cella C, Ferrari S, Stucchi N, Parvanova A, Iliev I,
Trevisan R, Bossi A, Zaletel J, Remuzzi G; GFR Study Investigators:
The GFR and GFR decline cannot be accurately estimated in type 2
diabetics. Kidney Int 84: 164–173, 2013 PubMed
7. de Boer IH, Sun W, Cleary PA, Lachin JM, Molitch ME, Zinman B,
Steffes MW; Diabetes Control and Complications Trial/Epidemiology
of Diabetes Interventions and Complications Study Research Group:
Longitudinal changes in estimated and measured GFR in type 1
diabetes. J Am Soc Nephrol 25: 810–818, 2014 PubMed
8. Pavkov ME, Knowler WC, Hanson RL, Williams DE, Lemley KV,
Myers BD, Nelson RG: Comparison of serum cystatin C, serum
305
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
creatinine, measured GFR, and estimated GFR to assess the risk of
kidney failure in American Indians with diabetic nephropathy. Am J
Kidney Dis 62: 33–41, 2013 PubMed
Tsai CW, Grams ME, Inker LA, Coresh J, Selvin E: Cystatin C- and
creatinine-based estimated glomerular filtration rate, vascular disease,
and mortality in persons with diabetes in the U.S. Diabetes Care 37:
1002–1008, 2014 PubMed
Ma YC, Zuo L, Chen JH, Luo Q, Yu XQ, Li Y, Xu JS, Huang SM,
Wang LN, Huang W, Wang M, Xu GB, Wang HY: Modified
glomerular filtration rate estimating equation for Chinese patients
with chronic kidney disease. J Am Soc Nephrol 17: 2937–2944,
2006 PubMed
Imai E, Horio M, Nitta K, Yamagata K, Iseki K, Hara S, Ura N,
Kiyohara Y, Hirakata H, Watanabe T, Moriyama T, Ando Y, Inaguma
D, Narita I, Iso H, Wakai K, Yasuda Y, Tsukamoto Y, Ito S, Makino H,
Hishida A, Matsuo S: Estimation of glomerular filtration rate by the
MDRD study equation modified for Japanese patients with chronic
kidney disease. Clin Exp Nephrol 11: 41–50, 2007 PubMed
Praditpornsilpa K, Townamchai N, Chaiwatanarat T, Tiranathanagul K,
Katawatin P, Susantitaphong P, Trakarnvanich T, Kanjanabuch T,
Avihingsanon Y, Tungsanga K, Eiam-Ong S: The need for robust
validation for MDRD-based glomerular filtration rate estimation in
various CKD populations. Nephrol Dial Transplant 26: 2780–2785,
2011 PubMed
Lee CS, Cha RH, Lim YH, Kim H, Song KH, Gu N, Yu KS, Lim CS,
Han JS, Kim S, Kim YS: Ethnic coefficients for glomerular filtration
rate estimation by the Modification of Diet in Renal Disease study
equations in the Korean population. J Korean Med Sci 25: 1616–1625,
2010 PubMed
Stevens LA, Claybon MA, Schmid CH, Chen J, Horio M, Imai E,
Nelson RG, Van Deventer M, Wang HY, Zuo L, Zhang YL, Levey AS:
Evaluation of the Chronic Kidney Disease Epidemiology Collaboration
equation for estimating the glomerular filtration rate in multiple
ethnicities. Kidney Int 79: 555–562, 2011 PubMed
Kong X, Ma Y, Chen J, Luo Q, Yu X, Li Y, Xu J, Huang S, Wang L,
Huang W, Wang M, Xu G, Zhang L, Zuo L, Wang H; Chinese eGFR
Investigation Collaboration: Evaluation of the Chronic Kidney Disease
Epidemiology Collaboration equation for estimating glomerular filtration rate in the Chinese population. Nephrol Dial Transplant 28: 641–
651, 2013 PubMed
Teo BW, Xu H, Wang D, Li J, Sinha AK, Shuter B, Sethi S, Lee EJ:
GFR estimating equations in a multiethnic Asian population. Am J
Kidney Dis 58: 56–63, 2011 PubMed
Wang X, Luo Y, Wang Y, Wang C, Zhao X, Wang D, Liu L, Liu G,
Wang Y; China National Stroke Registry Investigators: Comparison of
associations of outcomes after stroke with estimated GFR using Chinese
modifications of the MDRD study and CKD-EPI creatinine equations:
Results from the China National Stroke Registry. Am J Kidney Dis 63:
59–67, 2014 PubMed
Horio M, Imai E, Yasuda Y, Watanabe T, Matsuo S; Collaborators
Developing the Japanese Equation for Estimated GFR: GFR estimation
using standardized serum cystatin C in Japan. Am J Kidney Dis 61: 197–
203, 2013 PubMed
Schwartz GJ, Muñoz A, Schneider MF, Mak RH, Kaskel F, Warady
BA, Furth SL: New equations to estimate GFR in children with CKD.
J Am Soc Nephrol 20: 629–637, 2009 PubMed
Hoste L, Dubourg L, Selistre L, De Souza VC, Ranchin B, Hadj-Aïssa
A, Cochat P, Martens F, Pottel H: A new equation to estimate the
glomerular filtration rate in children, adolescents and young adults.
Nephrol Dial Transplant 29: 1082–1091, 2014 PubMed
Schwartz GJ, Schneider MF, Maier PS, Moxey-Mims M, Dharnidharka
VR, Warady BA, Furth SL, Muñoz A: Improved equations estimating
GFR in children with chronic kidney disease using an immunonephelometric determination of cystatin C. Kidney Int 82: 445–453, 2012 PubMed
Chehade H, Cachat F, Jannot AS, Meyrat BJ, Mosig D, Bardy D, Parvex
P, Girardin E: New combined serum creatinine and cystatin C quadratic
23.
24.
25.
26.
27.
28.
formula for GFR assessment in children. Clin J Am Soc Nephrol 9: 54–
63, 2014 PubMed
Stevens LA, Schmid CH, Greene T, Zhang YL, Beck GJ, Froissart M,
Hamm LL, Lewis JB, Mauer M, Navis GJ, Steffes MW, Eggers PW,
Coresh J, Levey AS: Comparative performance of the CKD Epidemiology Collaboration (CKD-EPI) and the Modification of Diet
in Renal Disease (MDRD) Study equations for estimating GFR
levels above 60 mL/min/1.73 m2. Am J Kidney Dis 56: 486–495,
2010 PubMed
Nyman U, Grubb A, Sterner G, Björk J: The CKD-EPI and MDRD
equations to estimate GFR. Validation in the Swedish Lund-Malmö
Study cohort. Scand J Clin Lab Invest 71: 129–138, 2011 PubMed
Bouquegneau A, Vidal-Petiot E, Vrtovsnik F, Cavalier E, Rorive M,
Krzesinski JM, Delanaye P, Flamant M: Modification of Diet in Renal
Disease versus Chronic Kidney Disease Epidemiology Collaboration
equation to estimate glomerular filtration rate in obese patients. Nephrol
Dial Transplant 28[Suppl 4]: iv122–iv130, 2013 PubMed
Evans M, van Stralen KJ, Schön S, Prütz K-G, Stendahl M, Rippe B,
Jager KJ; ERA-EDTA Registry; Swedish Renal Registry Collaboration:
Glomerular filtration rate-estimating equations for patients with advanced chronic kidney disease. Nephrol Dial Transplant 28: 2518–
2526, 2013 PubMed
Kilbride HS, Stevens PE, Eaglestone G, Knight S, Carter JL, Delaney
MP, Farmer CK, Irving J, O’Riordan SE, Dalton RN, Lamb EJ:
Accuracy of the MDRD (Modification of Diet in Renal Disease)
study and CKD-EPI (CKD Epidemiology Collaboration) equations
for estimation of GFR in the elderly. Am J Kidney Dis 61: 57–66,
2013 PubMed
Selvin E, Juraschek SP, Eckfeldt J, Levey AS, Inker LA, Coresh J:
Within-person variability in kidney measures. Am J Kidney Dis 61:
716–722, 2013 PubMed
Risk Factors for Progression of Kidney Disease
This NephSAP section reviews risk factors for
CKD progression. The role of AKI and important
studies in pediatrics were reviewed in prior NephSAP
editions.
Biomarkers
Promising biomarkers that accurately predict
CKD progression are not ready for clinical use at the
time of this edition of NephSAP. An albumin to creatinine
ratio (ACR) is routinely used to predict the onset of
diabetic nephropathy. However, patients with type 1
diabetes can have severe glomerular lesions, and patients
with type 2 diabetes can develop CKD with normal
albumin excretion (1,2).
Haptoglobin binds to hemoglobin, and its increased
appearance in the urine may be caused by increased
glomerular permeability, tubular damage, or a response
to oxidative injury. Bhensdadia et al. (3) assessed if the
baseline urine haptoglobin to creatinine ratio (HCR)
would predict early renal functional decline in patients
with type 2 diabetes more accurately than ACR. HCR
was tested in 204 patients with type 2 diabetes enrolled in
the Veterans Affairs Diabetes Trial with an average
306
follow-up of 3.7 years. Early renal functional decline was
defined as $3.3% annual loss of GFR. Urine proteomic
analysis showed that haptoglobin levels were markedly
increased in preliminary screening of patients with type 2
diabetes with increasing SCr versus those who had a stable
SCr. Comparing highest with lowest tertiles of HCR,
there was an adjusted 2.7-fold increased risk (95%
confidence interval [95% CI], 1.15 to 6.32) for predicting
early functional decline versus a 2.5-fold risk (95% CI,
1.14 to 5.48) using ACR. The ability to predict ESRD
was fair. Using both markers did not significantly
improve performance. (receiver operating characteristic
[ROC] area under the curves were 0.613, 0.621, and
0.664 for HCR, ACR, and both, respectively). HCR was
equal to ACR in predicting other renal outcomes, such as
a .50% increase in SCr. The HCR shows some
promise as an additional marker to ACR in type 2
diabetes in predicting rapid progression, especially in
those without increased albumin excretion, but it
requires confirmation in future studies. It is unclear if
this marker will predict less rapid disease progression
in patients with diabetes compared with ACR. In
addition, the threshold level for HCR to identify
progression is not established.
Genetics
The increased risk of nondiabetic CKD and
FSGS in blacks has been partially attributed to variants
of the apolipoprotein L1 (APOL1) gene. The G1 and
G2 variants appear with increased frequency in
individuals of West African ancestry by virtue of their
protective carrier state against Trypanosoma brucei
rhodesiense. Parsa et al. (4) investigated the effects of
having two copies of APOL1 gene variants (high risk)
versus zero or one copy (low risk) on progression of
CKD in two cohorts: the black Study of Kidney
Disease and Hypertension (AASK) Trial and the Chronic
Renal Insufficiency Cohort (CRIC) Study. AASK participants in this study included 693 blacks genotyped for
APOL1 who had CKD attributed to hypertension without
diabetes. The AASK end points were the composite of
ESRD or doubling of SCr. In the CRIC Study, outcomes
were slope in eGFR and the composite of ESRD or a 50%
reduction of measured GFR from baseline. Of 2955 White
and black individuals in the CRIC Study, 46% had
diabetes.
In the AASK, those with two gene variants had
an adjusted 88% increased risk for ESRD or doubling
of SCr (hazard ratio [HR], 1.88; 95% CI, 1.46 to 2.41;
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
P,0.001) (Figure 4). CRIC participants with two risk
variants had a more rapid decline in eGFR and higher risk
of the composite renal outcome versus White patients
with or without diabetes. After adjustment, blacks with
diabetes and high–risk APOL1 status had faster progression of CKD compared with Whites with diabetes
and blacks with diabetes and low–risk APOL1 status.
Patients with diabetes and high–risk APOL1 variants had
a difference in eGFR slope of 21.32 ml/min per 1.73 m2
per year compared with Whites with diabetes and
21.07 ml/min per 1.73 m2 per year compared with
blacks with diabetes and low–risk APOL1 status. In
patients without diabetes, blacks with high–risk
APOL1 status displayed faster loss by 21.05 and
21.21 ml/min per 1.73 m2 per year compared with
Whites and blacks with low risk, respectively. After
4.4 years of follow-up, there was a 46% increased
chance of reaching the composite end point in blacks
with diabetes and high–risk APOL1 status versus
blacks with diabetes and low–risk APOL1 status. In
blacks without diabetes and with high–risk APOL1
status, there was a 61% increased risk of reaching the
composite end point versus blacks without diabetes
and with low–risk APOL1 status (Figure 4).
The mechanism for progressive CKD in individuals with APOL1 high–risk variants is unclear. APOL1
variants may reduce podocyte autophagy, a process that
eliminates toxic protein aggregates and damaged organelles from cells. In addition, only a minority of
patients with two APOL1 risk alleles develop CKD.
Other genes, inflammation, viral infection, or other
environmental factors are hypothesized to act as
a second hit to cause progressive CKD.
Blacks both with and without diabetes
with two APOL1 risk alleles developed
more rapid CKD progression and increased
chance of ESRD or .50% decrease in
measured GFR. Other genes, inflammation, viral infection, or other environmental
factors most likely act as a second hit to
cause progressive CKD.
Obesity
The connection between obesity and CKD is
complex and comprehensively reviewed by Stenvinkel
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
307
Figure 4. High risk APOL1 status increases risk for doubling of serum creatinine or ESRD independent of proteinuria, blood
pressure goal, or ACE inhbitor use in the black Study of Kidney Disease and Hypertension. (A) Entire Cohort. (B) Level of
proteinuria. Proteinuria defined as urine protein to creatinine ratio equal to or . 220 mg/g. (C) Blood pressure goal, standard
defined as ,102-107 mmHg and intensive defined as , or equal to 92 mmHg. (D) Use of initial anti hypertensive medication.
ACE inhibitor or other (metoprolol or amlodipine). Proportion of patients free from progression of CKD in the black Study of
Kidney Disease and Hypertension. ACE, angiotensin-converting enzyme; APOL1, apolipoprotein L1. Reprinted with
permission from Parsa A, Kao WH, Xie D, Astor BC, Li M, Hsu CY, Feldman HI, Parekh RS, Kusek JW, Greene TH, Fink
JC, Anderson AH, Choi MJ, Wright JT Jr., Lash JP, Freedman BI, Ojo A, Winkler CA, Raj DS, Kopp JB, He J, Jensvold NG,
Tao K, Lipkowitz MS, Appel LJ; AASK Study Investigators; CRIC Study Investigators: APOL1 risk variants, race, and
progression of chronic kidney disease. N Engl J Med 369: 2183–2196, 2013.
308
et al. (5). Independent of hypertension and diabetes, the
postulated mechanisms for how obesity and lipid accumulation in the kidney could directly cause CKD
progression include hyperfiltration, cell maladaptation,
albuminuria, inflammation, and tubulointerstitial injury
and fibrosis. These mechanisms have been recently
reviewed (6,7). In addition, the risk for CKD is less in
the 10%–30% of patients who are obese but metabolically healthy (8).
Increased body mass index (BMI), the metric used
by the World Health Organization to define obesity, was
reported to be linked to new-onset CKD in a large
European cohort (9). In the Coronary Artery Risk
Development in Young Adults (CARDIA) Longitudinal
Cohort Study of 2839 participants with cystatin C eGFR
values .90 ml/min per 1.73 m2 at baseline, Grubbs et al.
(10) studied the association of BMI with the trajectory of
kidney function decline, rapid decline (.3% loss of eGFR
per year), and incident CKD (estimated glomerular filtration rate from serum cystatin C [eGFRcys],60 ml/min
per 1.73 m2). For those .30 years old, higher BMI
category was associated with increased odds of rapid
decline. For BMI categories of 25.0–29.9, 30–39.9, and
$40 kg/m2, adjusted odds ratios (ORs) were 1.50 (95%
CI, 1.21 to 1.87), 2.01 (95% CI, 1.57 to 2.87), and 2.57
(95% CI, 1.67 to 3.94), respectively, compared with the
reference BMI of 18.5–24.9 kg/m2. However, increasing
BMI was not related to incident CKD. In participants of
the CARDIA Study without albuminuria or decreased
eGFR at baseline, obesity ($30 kg/m2) was found to
be associated with incident albuminuria by ACR after
adjustment (OR, 1.9; 95% CI, 1.1 to 3.3) (11).
Although traditionally, BMI has been used to
define obesity (.30 kg/m2), this may not be the
optimal estimate of fat mass in patients with CKD
(12). Central or visceral body fat distribution has been
linked to CKD. Central deposition of body fat can be
measured by waist-to-hip ratios (WHRs). Disease risk
increases with high waist circumference and WHR
(.102 cm and .0.9 in men and .88 cm and .0.8
in women, respectively) (5). Kwakernaak et al. (13)
examined if WHR was related to kidney outcomes.
This study had 315 participants with a baseline mean
BMI of 24.9 kg/m2 and 125I-iothalamate GFR of 109
ml/min per 1.73 m2. After adjustment (including BMI),
increasing WHR was associated with lower baseline
measured GFR, lower effective renal plasma flow
measured by 131I-hippurate, and higher filtration fraction (GFR/effective renal plasma flow), a marker for
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
glomerular hyperfiltration in both crude and BSAnormalized measurements. This cross-sectional study
showed these associations with increasing WHR, independent of BMI. Of note, there was no association
between increased WHR and albumin excretion.
Accumulation of lipid in nonadipose tissue, such as
the liver, could play an important role in the pathogenesis
of obesity–related kidney disease. Nonalcoholic fatty
liver disease (NAFLD) has been shown experimentally
to release inflammatory, coagulant, oxidant, and fibrogenic meditators (14). Targher et al. (14) studied the
relationship of NAFLD diagnosed by liver ultrasound and
incident CKD in 261 patients with type 1 diabetes. These
participants had a mean eGFR of 92 ml/min per 1.73 m2
without macroalbuminuria (10.3% had microalbuminuria). For those individuals with NAFLD, there was
a doubling of risk for incident CKD (Modification of
Diet in Renal Disease [MDRD] eGFR ,60 ml/min per
1.73 m2) or the occurrence of macroalbuminuria (adjusted
HR, 2.03; 95% CI, 1.10 to 3.77). Liver ultrasound may be
insensitive in determining NAFLD, although one-half of
the cohort had NAFLD identified by ultrasound criteria.
Self-reporting was used to determine the nonalcoholic
state.
The relationship between increasing BMI and
kidney outcomes in those with prevalent rather than
incident CKD is not straightforward. Lu et al. (15)
examined renal outcomes in 453,946 veterans with
eGFRs,60 ml/min per 1.73 m2 stratified by BMI
(,20, 20–24.9, 25–29.9, 30–34.9, 35–39.9, 40–44.9,
45–49.9, and .50 kg/m2). Mean age was 74 years old,
87% were white, and 94% were men in this observational
study. Obese patients were younger, had higher BP, had
greater medication use, had greater prevalence of heart
failure (HF) and diabetes, and had lower prevalence of
malignancies and lung, liver, and rheumatologic diseases. After adjustment, BMI showed a U-shaped association with clinical outcomes for all-cause mortality and
various measures of CKD progression, such as incidence
of ESRD, doubling of SCr, and slope of eGFR. Kidney
outcomes were worst for those with BMIs,25 kg/m2 and
best for those in the 25–29.9- and 30–34.9-kg/m2 categories. Those with BMIs.35 kg/m2 had worse outcomes for progression of CKD if they had a baseline
eGFR.30 ml/min per 1.73 m2, but worse outcomes
were attenuated if baseline eGFR was ,30 ml/min
per 1.73 m2 (Figure 5). This observational study of
older men with prevalent CKD neither shows causality nor
describes a relationship with incident CKD. Lu et al. (15)
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
309
Figure 5. All-cause mortality and CKD progression in veterans with baseline eGFR,60 ml/min per 1.73 m2 by BMI. Body
mass index (BMI) categories showing a U-shaped association with progression of CKD in 453,946 United States veterans with
eGFR,60 ml/min per 1.73 m2. Association of BMI categories with four different outcomes representing progression of CKD:
(A) steeper slopes of eGFR versus time (defined as slopes ,24 ml/min per 1.73 m2 per year), (B) doubling of serum creatinine
or ESRD, (C) projected eGFR ,10 ml/min per 1.73 m2 or ESRD, and (D) ESRD. Estimates were calculated from a logistic
regression model (for steeper slopes) and Cox models (for all other outcomes) adjusted for age, race, comorbidities, medications,
and baseline eGFR. Reprinted with permission from Lu JL, Kalantar-Zadeh K, Ma JZ, Quarles LD, Kovesdy CP: Association of
body mass index with outcomes in patients with CKD. J Am Soc Nephrol 25: 2088–2096, 2014
provided explanations for the relative slowing of CKD
progression in individuals with moderate to advanced
CKD with BMIs in the 25- to 35-kg/m2 range. Those
with greater BMIs may have increased nutritional reserves,
which would be advantageous with advanced CKD, where
there may be malnutrition. A higher BMI may be caused
by increased muscle or bone mass rather than more
adiposity. In addition, a normal BMI may not represent
the metabolic effects of obesity caused by increased
visceral adiposity as described above.
Abou-Mrad et al. (16) reviewed weight reduction
regimens and bariatric surgery in the obese with CKD. An
earlier study by Navaneethan and Yehnert (17) reviewed
here described the effects of both restrictive and
310
malabsorptive bariatric surgery in 25 severely obese
participants (mean BMI ¼49.8 kg/m2) with stage 3
CKD. There was an improved eGFR from a mean of
47.9 ml/min per 1.73 m2 presurgery to 61.6 ml/min per
1.73 m2 2 years after surgery. However, Navaneethan
and Yehnert (17) excluded those who developed AKI
postbariatric surgery
Diet
Fluid.
A high fluid intake has been postulated to
lower CKD incidence by increasing solute clearance
and preventing kidney stones, whereas a lower fluid
intake may increase metabolic demand in the kidney
from urinary concentration. Clark et al. (18) had previously shown that drinking larger volumes of fluid
(24-hour urine volumes .3 L/d) was associated with
a reduced incidence of CKD. Palmer et al. (19) also
examined the relationship between fluid intake (evaluated by a food frequency questionnaire) and all-cause
and cardiovascular mortality and eGFR. This longitudinal, elderly, Australian cohort had 3858 participants
with a mean age of 70 years old. There were 1479
participants who had measurements of SCr at 10 years
of follow-up. No relationship was found between
higher fluid intake and mortality or changes in eGFR
after adjustment.
Dietary Patterns.
Dietary patterns may be more
important than specific macronutrients, such as protein,
or micronutrients, such as sodium, in modifying CKD
progression. In 3972 participants from the Reasons for
Geographic and Racial Difference in Stroke (REGARDS)
Study with eGFR,60 ml/min per 1.73 m2 or ACR$30
mg/g, Gutiérrez et al. (20) examined the effect of dietary
patterns on incident ESRD over 6 years of follow-up. A
participant’s dietary pattern was characterized by food
type predominance on a food frequency questionnaire.
Classifications were convenience (Chinese and Mexican
foods, pizza, and frozen or take-out dishes), plant based
(fruits, vegetables, and fish), sweets/fats (sugary foods
and carbohydrate heavy), Southern (fried foods, organ
meats, greens typical of Southern cuisines, and sweetened
beverages), and alcohol/salads (alcohol, green leafy
vegetables, and salad dressing). There was no association
of any dietary pattern with incident ESRD in adjusted
models. The Southern diet was associated with higher
mortality, whereas the plant-based diet was associated with improved mortality. The lack of influence of
various dietary patterns on incident ESRD may be
because there were nearly six times as many deaths as
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
ESRD events, and the study may have been underpowered to detect CKD progression on the basis of the
predominance of ESRD events. The accuracy of the
food reporting questionnaires employed in this study is
not clear.
Tirosh et al. (21) examined the effects of three diets
on eGFR and albumin excretion. The diets consisted of
low fat, Mediterranean, or low carbohydrate/increased
protein. This 2-year randomized, controlled trial (21)
included 318 participants with or without type 2 diabetes
with BMI.27 kg/m2 (mean 31 kg/m2) and eGFR.30
ml/min per 1.73 m2 (mean eGFR 70.5 ml/min per
1.73 m2). All diets resulted in significant improvements in
2-year eGFR. The low–carbohydrate/increased protein
diet raised eGFR by 15.4% (95% CI, 2.1 to 8.5), the
Mediterranean raised eGFR by 15.2% (range 3.0–7.4),
and the low-fat diet raised eGFR by 14% (range 0.9–
7.1). The eGFR increased by the same magnitude for
those with or without diabetes and for those with eGFR
values above or below 60 ml/min per 1.73 m2. Urine
albumin excretion among those with microalbuminuria
did not decrease significantly in any diet group. The
participants with eGFRs in the 30–60 ml/min per 1.73 m2
range on the low–carbohydrate/higher protein diet had
a 10% increase in eGFR over 2 years. Interestingly, after
2 years, there was no significant change in weight, and
2-year weight loss was not associated with increased eGFR.
Tirosh et al. (21) concluded that the low–carbohydrate/
increased protein diet was as safe as the other diets in
moderately obese patients with or without diabetes
with a SCr of ,2 mg/dl. The differences in protein
intake between diets were not large (22% of energy
from protein with the low-carbohydrate diet verse
19% in the other two diets). However, this was only
a 2- year study, in which participants had relatively
preserved GFRs.
Protein Intake.
Higher protein intake increases
glomerular hyperfiltration and may eventually lead to
incident CKD. In an ancillary study of the Optimal
Macro-Nutrient Intake Trial, a randomized, cross-over
trial of three separate diets emphasized carbohydrates,
unsaturated fats, or protein during 6-week periods. The
protein content varied from 15% in the carbohydrateand unsaturated fat–emphasized diets to 25% in the
protein-emphasized diet. In 164 participants, mean
cystatin C eGFR was 92.0 ml/min per 1.73 m2. This
increased by 3.81 ml/min per 1.73 m2 in the higher
protein diet independent of BP, with no change in the
other diets in the short term (22).
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
A longer post hoc analysis over 5.5 years by
Dunkler et al. (23) examined the association of the
healthiness of a diet as well as protein content on incident
CKD and progression. The latter was defined as newonset micro- or macroalbuminuria or eGFR decline of
.5% per year (23). This cohort of 6213 patients with type
2 diabetes without macroalbuminuria ages 55 years old
and older was enrolled in the Ongoing Telmisartan Alone
and in Combination with Ramipril Global Endpoint Trial
(ONTARGET). Mean eGFR was about 72 ml/min per
1.73 m2. Diet was recorded from a food questionnaire at
baseline, and the healthiness of diets was assessed using
a standardized index. The healthiest tertile had a 27%
lower adjusted risk of CKD (adjusted OR, 0.73; 95% CI,
0.64 to 0.84). Participants in the lowest tertile of total and
animal protein intake surprisingly had a slightly increased
risk of CKD compared with those in the highest tertile
(OR, 1.167; 95% CI, 1.05 to 1.30); however, median total
protein intake was only 0.58 g/kg per day (interquartile
range 0.42–0.82). The majority of participants in this
cohort had only mild CKD at baseline (Chronic Kidney
Disease Epidemiology Collaboration [CKD-EPI] eGFR
of approximately 72 ml/min per 1.73 m2), and 21.1%
manifested microalbuminuria.
In those with more severe CKD, Piccoli et al. (24)
reported the feasibility of using a low-protein diet supplemented with a-keto analogues of essential amino acids
(LPD-KA) to slow CKD progression. This diet consisted
of 0.6 g/kg per day supplemented with Ketsteril (1 pill per
10 kg), one to three free choice meals per week, and a list
of allowed and forbidden foods. These keto analogues of
amino acids do not have the nitrogen load of the essential
amino acids. This was a vegan diet, except for the free
choice meals. Patients had either stage 4 or nondialysis
stage 5 CKD (n¼129), rapidly progressive stage 3 CKD
(n¼29), and/or refractory proteinuria (n¼10) (24). This
older cohort had severe CKD (mean age 70 years old;
median GFR 17 ml/min per 1.73 m2 with median proteinuria of 1.4 g/d) and prevalent comorbidities comparable with patients on dialysis. Of those who started the
LPD-KA, there was a low mortality rate (4.4% per year).
The diet was discontinued for a variety of reasons: 40
participants started dialysis, 8 participants died, 7 participants discontinued the diet because of clinical reasons, 14
participants discontinued the diet because of personal
preference (monotony of diet), and 8 participants discontinued the diet because of improvement in eGFR. Although this study was not sufficiently powered to show an
effect on CKD progression rate, eGFR loss before this diet
311
was 28 ml/min per year, which improved to no reduction
in eGFR after 6 months of the diet in 95 participants. This
was not a uniform study population because of the
inclusion of 10 subjects with severe proteinuria and more
normal eGFR.
In pregnant women with CKD, higher protein
intake could exacerbate glomerular hypertension, but
there also may be a requirement for higher protein
intake for the fetus during pregnancy. Piccoli et al. (25)
compared the LPD-KA diet (0.6–0.8 g/kg per day with
keto supplementation and one to three protein–unrestricted
meals per week) in 21 pregnant women with stages 3–5
CKD or nephrotic-range proteinuria with 16 pregnant
women with CKD not on this diet. There was significantly less likelihood of small for gestational age
babies (3 of 21) compared with that in the control diet
group (7 of 16), but there were no other significant
differences as far as maternal kidney function or proteinuria. Piccoli et al. (25) concluded that the diet was
safe during pregnancy and reduced the likelihood of
small for gestational age children, although it did not
seem to affect long-term growth.
Sodium.
The effect of dietary sodium restriction on
CKD progression is controversial, and studies reported
here do not clarify our goal sodium intake for preventing
CKD progression. Excess sodium intake could cause
kidney vascular damage through volume retention with
increased shear stress and nonhemodynamic oxidative stress and inflammation (26). However, there are
concerns that a low-sodium diet could result in increased activity of the renin-angiotensin-aldosterone
system (RAAS), increased activity of the sympathetic
nervous system, or insulin resistance, which potentially
would accelerate CKD progression. In patients with
CKD, excess sodium intake has been associated with
CKD progression that is not independent of proteinuria.
McQuarrie et al. (27) studied the association
between urinary sodium excretion and urine sodium
to creatinine ratio, time to death, or requirement for
RRT in 423 participants with mean GFR of 48 ml/min
per 1.73 m2. There were 102 deaths and 99 patients
who required RRT during follow-up. The overall
cohort’s mean age was 51 years old, mean eGFR was
48625 ml/min per 1.73 m2, mean ACR was 97.3 mg/g,
and mean sodium intake was 155 mmol/d (3.56 g/d).
After adjustment, a high versus low dietary sodium
intake measured by either 24-hour urine sodium or
24-hour urine sodium to creatinine ratio did not independently predict mortality or ESRD. However, the mean
312
24-hour urine sodium was 160 mmol/d (3.68 g/d) in
the high–sodium diet group compared with 127
mmol/d (2.92 g/d) in the low-sodium group. This lack
of effect between dietary sodium and CKD progression was supported by the findings by Dunkler
et al. (23) in the previously reported study of patients
with type 2 diabetes. Mean urinary sodium excretion
was higher in this study at 4.89 g/d (212 mmol/d),
with a wide range of dietary sodium intake that had
little association with CKD progression (23). The
mean eGFR was about 72 ml/min per 1.73 m2. Those
with a urine sodium excretion ,3 g/d had slightly
higher odds of CKD progression with increased odds
of death.
A complex relationship between sodium intake,
proteinuria, and kidney failure was described by Fan
et al. (28). The 840 participants in the MDRD Study
had a mean measured GFR of 32.5 ml/min per 1.73 m2
with median urine protein excretion of 320 mg/d
(0.07–1.510). This was a post hoc analysis of the
association of unrestricted sodium intake with kidney
failure, defined as onset of dialysis or transplant, which
developed in 617 participants, and the composite outcome of kidney failure or death achieved in 723
participants over a median follow-up of 6 years. Mean
baseline 24-hour urine sodium excretion was 3.46 g/d.
After adjustment, there was no relation with either
kidney failure (HR, 0.99; 95% CI, 0.91 to 1.08) or the
composite of kidney failure and death (OR, 1.01; 95%
CI, 0.93 to 1.09) for every 1-g/d greater urinary sodium
excretion. On additional analysis, when urine sodium
was .3 g/d, there was no association with kidney
failure. In individuals with ,1 g/d proteinuria with
urine sodium excretion ,3 g/d, dietary sodium restriction was related to protection against kidney failure
(adjusted HR, 0.61; 95% CI, 0.42 to 0.89). However,
there seemed to be a J-shaped relationship; if urine
sodium was ,3 g/d in participants with .1 g/d proteinuria, the risk for kidney failure increased (adjusted
HR, 1.72; 95% CI, 1.31 to 2.24). Similar results were
found using baseline or time–dependent urine sodium
measurements. Why dietary sodium restriction would
increase the risk of kidney failure in patients with
more proteinuria is unclear. It is possible that the
observed lower dietary sodium intake resulted from
decreased intake in sicker patients. Those with proteinuria may have been more susceptible to episodes
of AKI with a lower sodium intake. This association
contradicted findings by Vegter et al. (29) in the
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Ramipril Efficacy in Nephropathy Trial of patients
who were highly proteinuric (median daily protein
excretion 3 g/d), where increased time–averaged urine
sodium excretion was associated with an increased risk
for developing ESRD.
A randomized, double–blinded, placebo, controlled trial was performed to assess dietary sodium
intake on BP and proteinuria in 20 elderly participants
with stages 3 or 4 CKD (30). Changes in ambulatory
BP, 24-hour urine protein and albumin excretion, fluid
status measured by a body composition monitor, renin
and aldosterone levels, and arterial stiffness measured
by pulse wave velocity (aPWV) and augmentation
index were assessed on high- and low-sodium diets.
Participants were counseled on a low-sodium diet of
60–80 mmol/d. The high-sodium diet consisted of the
same diet plus 120 mmol/d sodium tablets for a goal of
180–200 mmol/d. Placebo tablets were given to participants ingesting the low-sodium diet. The mean 24hour urine sodium excretion values were 75 and 168
mmol/L for low- and high-sodium diets, respectively.
The two diets lasted 6 weeks before participants
crossed over to the other diet after a 1-week washout
period. The low-sodium diet resulted in multiple
beneficial effects. BP decreased by a mean of 10/4
mmHg (95% CI, 5 to 15/1 to 6 mmHg), extracellular
fluid volume decreased (mean ¼0.8 L; 95% CI, 0.4 to
4.2), and albuminuria and proteinuria decreased by
medians of 148 and 342 mg/d, respectively (Table 2).
The large effect on BP may have been because these
patients may have been especially salt sensitive, with
a mean of 3.2 antihypertensive medications. Although
promising, this small trial was of short duration. A
possible detrimental effect was that plasma renin and
aldosterone increased with the low-sodium diet. The
change in the six participants on RAAS blockade was not
reported.
Taken together, the studies by McQuarrie et al.
(27) and Fan et al. (28) question what the optimal
sodium intake is for patients with CKD. These were
post hoc analyses that did not target specific sodium
intakes for outcomes. The sodium intake trial by
McMahon et al. (30) confirms the influence of sodium
restriction on lowering BP and proteinuria. Therefore,
making adjustments for these variables to assess an
independent effect of restricting sodium intake is
questionable. Perhaps, in the study by Fan et al. (28),
the worse outcomes with sodium restriction in patients
who were proteinuric resulted in either increased AKI
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
313
Table 2. Values during low- and high-salt periods
Reprinted with permission from McMahon EJ, Bauer JD, Hawley CM, Isbel NM, Stowasser M, Johnson DW, Campbell KL: A randomized trial of dietary sodium restriction in
CKD. J Am Soc Nephrol 24: 2096-2103, 2013.
events or excessive activation of the RAAS. In this
study, the optimal sodium intake for patients with
.1 g/d proteinuria seemed to be about 3 g sodium,
with worsening with more restriction. For those with
,1 g/d proteinuria, the optimal sodium intake seemed
to be ,3 g/d. Perhaps those with CKD who are not
hypertensive may not benefit from limiting sodium
intake. In practice, there will be a difference between
advising and following sodium restriction. For now,
aiming for a daily 2- to 3-g sodium intake seems
reasonable.
Bicarbonate
Acidosis stimulates ammonia, aldosterone, and
endothelin production to increase tubular acid excretion to ameliorate acidosis. However, these mediators
may also increase tubulointerstitial damage and CKD
progression. Low serum bicarbonate levels may represent a biomarker for those who will develop increased CKD progression.
Driver et al. (31) examined the association
between serum bicarbonate and rapid renal function
decline (.5% per year) as well as incident CKD
defined as ,60 ml/min per 1.73 m2 by CKD-EPI
cystatin C-creatinine formula. Participants from the
Multi-Ethnic Study of Atherosclerosis had a mean age
of 61 years old, mean bicarbonate level of 23 mEq/L,
and baseline eGFR of 84 ml/min per 1.73 m2 with
median ACR of 5.2 mg/g. The median follow-up was
3.9 years. After adjustment, each 1.8-mEq/L decline in serum bicarbonate concentration was associated with a slightly higher odds of rapid kidney
function decline (OR, 1.12; 95% CI, 1.06 to 1.20)
and incident CKD (incidence rate ratio, 1.12; 95% CI,
1.03 to 1.20), although the relationship for incident
disease seemed to dissipate at both lower and higher
values for serum bicarbonate. Limitations of this
study are that these findings are on the basis of a single
serum bicarbonate value. It is also not known which
patients had a concurrent primary respiratory acidbase disorder. In addition, bicarbonate was measured
from frozen samples, which can significantly reduce
serum bicarbonate levels.
Goldenstein et al. (32) also examined the association between serum bicarbonate and GFR decline and
incident CKD in an older cohort (mean age of 75.2 years
old) followed for 7 years. These participants had a mean
baseline eGFR of 84.2 ml/min per 1.73 m2 determined
by the CKD-EPI cystatin C-creatinine formula. A serum
bicarbonate ,23.0 mEq/L was associated with a 0.54
ml/min per year (95% CI, 20.97 to 20.12; P,0.01)
faster annual decline as well as an 81% increased chance
for incident CKD after adjustment compared with the
reference group with serum bicarbonate levels of 23–
28 mEq/L.
Small trials using sodium bicarbonate supplementation showed decreased CKD progression compared
with control groups (33,34). Sodium citrate also slowed
314
Figure 6. Similar changes in eGFR in stage 4 CKD
participants with serum bicarbonate ,22 mEq/L after one
year of sodium bicarbonate or fruits and vegetables. Box
plots of plasma cystatin C eGFR (cys GFR; left panel) and
plasma creatinine eGFR (cr GFR; right panel) at baseline and
1-year follow-up. The bottom and top of the boxes span the
25th and 75th percentiles of data points. The dark bar within
the box indicates the 50th percentile or median. The whiskers
indicate 1.5 times the interquartile range from the lower and
upper quartiles. F/U, follow-up; F1V, fruits and vegetables.
*P,0.05 versus respective baseline. Reprinted with permission from Goraya N, Simoni J, Jo CH, Wesson DE: A
comparison of treating metabolic acidosis in CKD stage 4
hypertensive kidney disease with fruits and vegetables or
sodium bicarbonate. Clin J Am Soc Nephrol 8: 371–381,
2013
the rate of decrease in eGFR in participants with stage
3b CKD (35). Although sodium bicarbonate may not
increase volume to the same degree as sodium
chloride, alkali supplementation with sodium could
result in worsening hypertension or volume. Fruits
and vegetables (F1V) represent an alternative source
of citrate alkali, which may help delay CKD progression without sodium but with high potassium
content. Goraya et al. (36) examined the use of F1V
in participants with stage 4 CKD with total CO2 levels
,22 mEq/L over 1 year. Participants were excluded if
they had edema, congestive heart failure (CHF), liver
failure, nephrotic syndrome, diabetes, or serum potassium levels .4.6 mEq/L (36). Alkali therapy as
1 mEq/kg per day sodium bicarbonate was administered to 35 individuals, and F1V, approximately
0.4 mEq/kg per day, were provided to reduce dietary
acid by one half in 36 individuals. Mean serum bicarbonate levels were higher at 1 year versus baseline in
both groups (19.561.5 to 21.261.3 in the sodium
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
bicarbonate group and 19.361.9 to 19.961.7 in the
F1V group). After 1 year, markers of tubulointerstitial damage (urinary N-acetyl-b-d-glucosaminidase
[NAG] and urinary TGF-b) were lower compared
with baseline in both groups. Decreases in systolic BP
(approximately 4.5 mmHg) and weight (approximately 4.7 kg) occurred in the F1V group, with no
change in the bicarbonate group. There were no
differences between the alkali treatment groups as
far as baseline or 1-year eGFR values by cystatin C or
creatinine. In both treatment groups, eGFR decreased
slightly over 1 year. The change in mean eGFR in the
sodium bicarbonate group was 21.763.4 to 20.363.2
ml/min per 1.73 m2 for eGFRcys and 23.063.5 to
21.463.3 ml/min per 1.73 m2 for estimated GFR from
serum creatinine (eGFRcr), whereas mean change in
the F1V group was 21.664.6 to 20.764.7 ml/min
per 1.73 m2 for eGFRcys and 22.864.9 to 21.965.1
ml/min per 1.73 m2 for eGFRcr (Figure 6). Plasma
potassium did not change from baseline in either
group.
The same group examined whether treatment with
alkali in those with stage 3 CKD who had serum
bicarbonate levels between 22 and 24 mEq/L (above
the threshold for treatment by Kidney Disease Outcomes
Quality Initiative guidelines) might slow CKD progression in a 2-year trial (37). Individuals were divided into
sodium bicarbonate (n¼36), F1V (n¼36), or usual care
(n¼36) groups. The participants had metabolic acidosis
by venous blood gas and moderate to severe albumin
excretion (mean .300 mg/g) and were treated with
RAAS blockers. Exclusion criteria were similar to those
used in the trial above. The sodium bicarbonate group
was treated with 0.3 mEq/kg per day bicarbonate to
reduce acid excretion by 50%. Compared with baseline,
serum bicarbonate increased in the alkali treatment
groups, whereas it decreased in the usual care group.
Urinary albumin decreased in all groups with RAAS
blockade, but there were greater decreases in both alkali
treatment groups. Markers of tubulointerstitial damage
and kidney angiotensin levels (urinary angiotensinogen)
decreased in the alkali groups but increased in the usual
care group. Systolic BP was lower in all groups, with
the largest decrease in the F1V group (approximately
35 mmHg), which corresponded to the largest weight loss
(approximately 4 kg). Declines in eGFR by creatinine and
cystatin C over 3 years were significantly less in both
alkali treatment groups compared with the usual care
group (eGFRcr from 42.667.6 to 28.867.3 ml/min per
315
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 7. Changes in eGFR in stage 3 CKD participants with
serum bicarbonate 22–24 mEq/L with ACR.300 mg/g after
3 years of sodium bicarbonate, fruits and vegetables or usual
care. Plasma creatinine–based eGFR (cys GFR; left panel)
and cystatin C eGFR (cr GFR; right panel) in the group
treated with fruits and vegetables (F1V) versus usual care.
*P,0.05 versus usual care at 2 years; 1P,0.05 versus usual
care at 3 years. Reprinted with permission from Goraya N,
Simoni J, Jo CH, Wesson DE: Treatment of metabolic
acidosis in patients with stage 3 chronic kidney disease with
fruits and vegetables or oral bicarbonate reduces urine
angiotensinogen and preserves glomerular filtration rate.
Kidney Int 86: 1031–1038, 2014
1.73 m2 in the usual care group, eGFRcr from 42.667.0
to 35.266.9 ml/min per 1.73 m2 in the sodium
bicarbonate group, and eGFRcr from 42.367.1 to
36.966.7 ml/min per 1.73 m2 in the F1V group)
(Figure 7). Plasma potassium was unchanged from
baseline in all three groups.
It is unclear what the optimal level of serum
bicarbonate should be for patients with CKD. Experimental studies in cardiac myocytes show that alkalosis
may modify proteins that regulate gene transcription
and decrease cell survival. Dobre et al. (38) examined
serum bicarbonate levels as a risk factor for renal
outcomes, cardiovascular events (CVEs), and mortality
in CKD. There were 3939 participants in the CRIC
Study with a mean eGFR of 44.8 ml/min per 1.73 m2
and a median bicarbonate of 24 mEq/L (interquartile
range 22–26 mEq/L) who were followed for 3.9 years
on average. After adjustment, every 1-mEq/L increase
in serum bicarbonate was associated with a 3% lower
risk of developing a renal end point (50% decrease in
eGFR or ESRD; HR, 0.97; 95% CI, 0.85 to 0.97;
P¼0.04), which increased to 9% in those with an
eGFR.45 ml/min per 1.73 m2 (HR, 0.91; 95% CI,
0.85 to 0.97). However, for every 1-mEq/L increase in
serum bicarbonate over 24 mEq/L, there was a 14%
higher adjusted risk of developing HF (adjusted HR,
1.14; 95% CI, 1.03 to 1.26). There were no increases in
atherosclerotic events or all-cause mortality. These
relationships were maintained after accounting for
diuretic use and exclusion of the small minority of
patients who were on alkali supplementation (n¼91).
Again, it was not known which participants had respiratory acidosis.
Alkali therapy, whether in the form of sodium
alkali or F1V, seems to decrease CKD progression
with moderate to severe CKD. This may benefit
those with only mildly decreased serum bicarbonate
levels, but the patients selected for therapy must be
chosen carefully. Although the optimal upper limits
are not clear, bicarbonate supplementation should
be titrated so as not to greatly exceed the level of
24 mEq/L.
Rate of Decline of GFR and GFR Variability
Clinical trials in nephrology have traditionally
used doubling of SCr, corresponding to a drop in GFR
of 57%, or ESRD as the end point to determine if
interventions slow CKD progression. Trials may
have to have follow-up for 5 years for there to be
a significant number of participants reaching these
end points. Using meta-analysis of 1.7 million participants from the Chronic Kidney Disease Prognosis
Consortium, Coresh et al. (39) explored whether a reduction in GFR,57% over a shorter timeframe would
predict CKD progression to ESRD and mortality. For
participants with an initial eGFR ,60 ml/min per 1.73
m2 who also suffered additional decrease of .30% in
eGFR over 2 years, there was a 5.4-fold increased odds
of ESRD (HR, 5.4; 95% CI, 4.5 to 6.4) after adjustment
(39). For those participants with initial eGFR .60 ml/min
per 1.73 m2, there was a 6.7-fold increase in odds of
ESRD if there was an additional .30% decrease in
eGFR (95% CI, 3.9 to 11.5). For the entire consortium,
the 10-year risks of ESRD and death were 64% and
50%, respectively, for a 30% decrease in eGFR over
2 years. The odds of ESRD were markedly increased if
the standard doubling of SCr, corresponding to the
57% decrease in eGFR, was used, but the prevalence of
this larger change was much lower (cumulative prevalence of 0.79% versus 6.9% with 257% versus
230% change in eGFR over 2 years, respectively)
(Figure 8).
316
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 8. 30% decrease in eGFR over 2 years robustly predicts increased ESRD risk. ESRD associated with percentage change
in eGFR during a 2-year baseline period. Values were trimmed at ,270% change (0.22% and 0.055% of the study population
for eGFR,60 [A] and $60 ml/min per 1.73 m2 [B], respectively) and .40% change (5.9% and 0.51% of the population for
eGFR,60 [A] and $60 ml/min per 1.73 m2 [B], respectively). In top panels, the diamonds indicate the reference point of 0%
change in eGFR. Reprinted with permission from Coresh J, Turin TC, Matsushita K, Sang Y, Ballew SH, Appel LJ, Arima H,
Chadban SJ, Cirillo M, Djurdjev O, Green JA, Heine GH, Inker LA, Irie F, Ishani A, Ix JH, Kovesdy CP, Marks A, Ohkubo T,
Shalev V, Shankar A, Wen CP, de Jong PE, Iseki K, Stengel B, Gansevoort RT, Levey AS; CKD Prognosis Consortium:
Decline in estimated glomerular filtration rate and subsequent risk of end-stage renal disease and mortality. JAMA 311: 2518–
2531, 2014
It is assumed that increased proteinuria is correlated with increased CKD progression. Turin et al. (40)
quantified the decrease in annual eGFR by baseline
eGFR level and proteinuria divided into normal, mild,
or heavy categories. There were 638,150 adults in the
Alberta Canada registry with more than three creatinine measurements over $1 year with either dipstick
proteinuria or ACR. Proteinuria was categorized as
normal (urine dipstick negative), mild (urine dipstick
trace or 11), or heavy (urine dipstick 21). In sensitivity
analyses, ACR was used with proteinuria categories
classified as normal (ACR,30 mg/g), mild (ACR¼30–
300 mg/g), or heavy (ACR.300 mg/g). The eGFR
decrease could not be adjusted for BP control, smoking,
317
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
or lipid status. Notably, in those with stage 4 CKD with
no or mild proteinuria, eGFR remained stable or actually
increased.
Changes smaller than a doubling of serum
creatinine (57% reduction in eGFR) predict
ESRD. The 10-year risks of ESRD and death
were 64% and 50%, respectively, for a 30%
decrease in eGFR over 2 years.
Volume Overload
Fluid overload may be a risk factor for progression
of CKD independent of hypertension, because this may
lead to vascular remodeling, diastolic dysfunction, and
inflammation. Chen et al. (41) had previously reported
that an enlarged left atrial diameter, a reflection of
volume overload and diastolic dysfunction, was associated with faster decline in eGFR. Tsai et al. (42) enrolled
472 Taiwanese patients with stages 4 or 5 CKD in
a prospective, observational study to discern whether
fluid overload was independently related to initiation of
RRT or rapid progression of CKD (.3 ml/min per 1.73 m2
per year). During a median follow-up of 17.3 months,
RRT was initiated in 15%, and 39.6% had rapid CKD
progression. Bioimpedance spectroscopy was used to
determine the degree of fluid overload as the difference
between measured and expected extracellular water.
Those in the highest tertile of fluid overload, corresponding to .1.6 L, had an increased HR of 3.16 (95%
CI, 1.33 to 7.50; P¼0.01) versus the lowest tertile
(,0.6 L). The highest tertile of relative hydration status
(fluid overload/extracellular water), corresponding to
.11%, similarly showed a .3-fold increased adjusted
HR (HR, 3.08; 95% CI, 1.33 to 7.16; P¼0.01) compared
with the lowest tertile. The adjusted ORs for rapid
progression in the highest tertiles of fluid overload and
relative hydration status were both .4 compared with the
lowest tertile (adjusted OR, 4.68; 95% CI, 2.30 to 9.52
and adjusted OR, 4.15; 95% CI, 2.06 to 8.36, respectively). Adjustments included systolic BP, cardiovascular
disease (CVD), and diabetes. The study had limitations,
because there was only baseline rather than time-varying
measurements of fluid overload. Rapid progression may
have been the cause rather than the result of fluid
overload. Baseline characteristics associated with more
fluid overload are related to CKD progression, thereby
making adjustments complex. However, subsets of patients
in the highest fluid overload tertile without CVD, diabetes,
or serum albumin ,3.5 g/dl also had an increased adjusted
HR for RRT compared with the lowest tertile (HR, 3.53;
95% CI, 1.43 to 8.69; HR, 6.38; 95% CI, 1.44 to 28.32;
and HR, 2.77; 95% CI, 1.0 to 7.01 without CVD, diabetes,
or serum albumin ,3.5 g/dl, respectively). The bioimpedance device has been validated for the general population
and in ESRD but not in CKD.
Tsai et al. (43) hypothesized that fluid overload
may be even more important than diabetes for rapid
progression and initiation of RRT in stages 4 and 5 CKD.
Using this same cohort, with 44% of participants having
diabetes and fluid overload (defined as relative hydration
status .7%; 90th percentile in normal individuals), there
were nearly 3- and .4-fold adjusted risks for initiating
RRT in patients without and with diabetes, respectively,
compared with those who had a hydration status ,7%
(43). The adjusted OR for rapid progression was also
higher. These results require verification in larger studies,
but they are certainly intriguing.
Vascular Disease
Atherosclerotic vascular disease detected outside of
the kidney may predict CKD progression. The anklebrachial index (ABI) is a marker of generalized atherosclerosis. Foster et al. (44) examined if low ABI ,0.9 was
associated with the outcomes of rapid eGFR decline
defined as decline .3 ml/min per 1.73 m2 per year,
incident stage 3 CKD, and incident microalbuminuria.
There were 2592 participants in the Framingham Offspring
Cohort followed over an average of 9.5 years. Rapid eGFR
decline and incident CKD stage 3 occurred in 9% and
11.1% of the cohort, respectively. A low ABI (,0.9) was
associated with 3.6-fold increased adjusted odds of rapid
eGFR decline. Low ABI was not associated with incident
CKD and incident albuminuria in fully adjusted models.
Hung et al. (45) examined the relationship between
stroke and subsequent ESRD. In a retrospective cohort
study of nationwide claims data of 442,355 Taiwanese
patients with stroke, 1.8% developed ESRD in a mean
follow-up of 4 years. The standardized incident ratio
(observed ESRD/expected ESRD) was used as a measure
of relative risk for ESRD. The relative risk for ESRD in
patients with stroke was 2.78 (95% CI, 2.72 to 2.84). The
relative risk for ESRD in young patients ages 25–44 years
old with stroke was markedly increased to 22.73 (95%
CI, 20.39 to 25.20). When patients with diabetes, hypertension, or prior CKD by claims data were excluded, the
relative risks for the entire cohort and those ages 25–44 years
318
old were slightly attenuated (2.27; 95% CI, 2.22 to 2.33 and
17.72; 95% CI, 15.66 to 19.91, respectively). Those who
suffered ischemic stroke had a higher standardized incident
ratio. The younger age stroke group had a higher prevalence
of small vessel disease than the older groups. Hung et al.
(45) postulated that stroke in younger patients could be from
vasculitis, which could cause renal disease as well, explaining the higher ESRD risk. Participant smoking history,
alcohol use, and socioeconomic status adjustments could not
be performed, because these data were not captured.
Pulsatility and Pulse Pressure
Vascular stiffness may cause kidney function decline through damage to small arteries, with pulsatile
stress leading to glomerular hypertension. Madero et al.
(46) explored the relationship between arterial stiffness
and kidney damage in 2129 elderly participants (mean
age 74 years old; mean eGFRcys of 79 ml/min per
1.73 m2). There was long follow-up (median follow-up
8.9 years) in this cohort of the Health, Aging, and Body
Composition Study (46). Arterial rigidity was measured by
aPWV and pulse pressure (PP). In multivariate analysis,
aPWV was not associated with rapid decline of eGFR
(.3 ml/min per 1.73 m2), but it was associated with a 42%
increased chance of incident CKD (95% CI, 1.12 to 1.81),
although this required doubling of aPWV. Increase in PP
by 10 mmHg was associated with a 9% increased chance
(95% CI, 1.02 to 1.16) of rapid decline as well as a 6%
increased chance (95% CI, 1.01 to 1.12) of incident CKD.
PP may have more accurately predicted renal outcomes
than aPWV, because pulsatility is influenced by left
ventricular contractility, pattern of left ventricular ejection,
and aortic valve competence as well as aortic stiffness.
Retinopathy
Retinal abnormalities may be associated with renal
vascular changes and could be a marker for disease
progression. In 1852 participants from the CRIC Study,
retinal images were compared with renal outcomes (47).
No link was established between retinopathy grade,
retinal arteriole diameter or retinal vein diameter, and
renal outcomes of ESRD or eGFR decline. Only the
highest quartile of retinal arteriole to vein diameter was
associated with an increased risk of ESRD compared
with the first quartile.
Preexisting CVD
CVD may increase CKD progression by altering
renal hemodynamics with cardiorenal syndrome, increasing
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
susceptibility to AKI by hospitalization and intervention for
coronary events, or generalized atherosclerosis with renovascular disease. Left ventricular hypertrophy is used as
a surrogate end point for cardiovascular morbidity in
patients with CKD. Peterson et al. (48) found no association
of left ventricular hypertrophy for the outcome of doubling
for SCr or ESRD in 578 participants in the AASK
Cohort, although there was an association with
CVEs causing hospitalization or death.
Sud et al. (49) performed a retrospective cohort
study examining the relationship between CVEs (HF,
myocardial infarction, and stroke) after initial nephrology
consultation and the outcomes of ESRD and all-cause
mortality before ESRD. This cohort had 2964 participants
with CKD with a mean eGFR of 35 ml/min per 1.73 m2
and a median ACR of 97 mg/g seen in an Ontario
nephrology clinic. Data for ESRD and CVEs were
obtained from electronic medical records and registry
data. Having a CVE after initial nephrology consultation was associated with an adjusted .5-fold increased
risk of subsequent ESRD (HR, 5.33; 95% CI, 3.73 to
7.58) and a .4-fold increase in all-cause mortality
before ESRD (HR, 4.15; 95% CI, 3.30 to 5.23). Of
those who died before ESRD, there was an almost
6-fold increase in cardiovascular-related deaths compared with noncardiovascular-related deaths.
Hypoxemia
Sleep disturbances and nocturnal hypoxemia could
potentially contribute to CKD progression through systemic inflammation, oxidative stress, and RAAS activation with resulting tubulointerstitial injury. In patients with
CKD, there may be few sleep–related symptoms in those
with sleep apnea (50). Ahmed et al. (51) had previously
linked nocturnal hypoxemia with accelerated loss of
kidney function (.4 ml/min per 1.73 m2 per year) in an
obese cohort (BMI¼32.8 kg/m) with relatively preserved
kidney function (eGFR¼70.8 ml/min per 1.73 m2). It
is difficult to determine whether CKD progression was
attributable to obesity or sleep apnea. Sakaguchi et al. (52)
examined the role of nocturnal hypoxemia and GFR loss
in 161 Japanese patients who were with not obese and had
stage 3 or 4 CKD. The mean BMI was 21.8 kg/m2 (95%
CI, 20.1 to 23.1) in this elderly cohort, and the eGFR loss
was measured over only 1 year. The mean eGFR of
the cohort was 31 ml/min per 1.73 m2. The oxygen
desaturation index (ODI) was the frequency of a .4%
drop from the baseline oxygen saturation level. Nocturnal
hypoxemia was defined as none if ODI was ,5 times per
319
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
hour, mild if ODI was 5–15 times per hour, and moderate
to severe if ODI was .15 times per hour. A portable
monitor also measured apnea and hypopnea. The moderate
to severe nocturnal hypoxemia group had a faster eGFR
decline of 28.59 ml/min per 1.73 m2 per year (22 to 15.2)
compared with no and mild degrees of hypoxemia. The
rates of eGFR loss were 22.13 ml/min per 1.73 m2 per
year (21.06 to 23.21) and 23.02 ml/min per 1.73 m2
per year (21.31 to 24.74) in the no and mild hypoxemia
groups, respectively. It is unclear if the relationship
between hypoxemia and loss of GFR is maintained for
the nonobese in other populations. Although the patients
were not obese by BMI, visceral adiposity was not
measured, and therefore, a contribution from adiposity
cannot be ruled out.
Hyperuricemia/Allopurinol
The possible contribution of hyperuricemia to
progression was reviewed in the last NephSAP CKD
and progression issue. Proposed mechanisms for uric
acid–induced kidney damage include hypertension with
RAAS activation, renal afferent arteriolopathy, increased
glomerular hypertension, and fibrosis. Allopurinol could
be protective by reducing not only serum uric acid but
also, oxidative stress by inhibition of xanthine oxidase. A
meta-analysis assessing the role of allopurinol and CKD
progression included eight heterogeneous trials with 476
participants (53). Median follow-up was only 11 months.
The trials had varying entry criteria for GFR, indications
(diabetic nephropathy and IgA nephropathy), and allopurinol doses (100–300 mg/d). In five trials that reported
GFR, there was a higher GFR in allopurinol versus
control groups of 3.1 ml/min per 1.73 m2, but this was not
statistically significant (95% CI, 20.9 to 7.1 ml/min per
1.73 m2; P¼0.10). In three trials that reported change in
SCr, there was a difference of a decrease of 0.4 mg/dl in
the allopurinol-treated group, but this finding was not
statistically significant (95% CI, 20.8 to 0.0; P¼0.22).
Although serum uric acid was reduced, there was no
change in proteinuria or BP in those studies that reported
these results. With the potentially severe toxicities with
this medication, there is not enough evidence to use
allopurinol to slow CKD progression in asymptomatic
patients with hyperuricemia and CKD at this time. There
have been no trials that have reported the use of febuxostat
on CKD progression, although one has completed
recruitment (54).
Pentoxifylline in Diabetes
Figure 9. Pentoxifylline decreases eGFR in open label trial
of type 2 diabetic participants. Evolution of the mean eGFR
at randomization (basal) and during follow-up in the control
and pentoxifylline (PTF) groups. Difference of the mean
eGFR between groups shows statistical significance after
24 months of follow-up. Vertical bars represent the SDs.
P values are for the comparison of the PTF group with the
control group. Reprinted with permission from NavarroGonzález JF, Mora-Fernández C, Muros de Fuentes M,
Chahin J, Méndez ML, Gallego E, Macía M, del Castillo N,
Rivero A, Getino MA, García P, Jarque A, García J: Effect of
pentoxifylline on renal function and urinary albumin excretion in patients with diabetic kidney disease: The PREDIAN
trial. J Am Soc Nephrol 26: 220–229, 2015.
Recent treatments for diabetic kidney disease have
been disappointing. Pentoxifylline (PTF) has been used
for peripheral vascular disease and has anti-inflammatory,
antiproliferative, and antifibrotic actions. Goicoechea
et al. (55) previously performed a 1-year-long trial using
PTF, which showed not only a decrease in inflammatory
measurements but also, a benefit on slowing progression
(approximately 6.8 ml/min per 1.73 m2 between groups.
Navarro-González et al. (56) performed a larger 2-year,
open-label, controlled trial using PTF. The trial included
169 Whites with type 2 diabetes on RAAS blockers. The
BP control, glucose control, and kidney parameters were
well matched at baseline (eGFR: 37.6 versus 37.1 ml/min
per 1.73 m2 by MDRD and urine albumin excretion: 1000
versus 1100 mg/d in the control versus PTF groups,
respectively). The eGFR with PTF had slower progression, because eGFR decreased by 2.160.4 compared with
6.560.4 ml/min per 1.73 m2 in the control group at the
end of 2 years (Figure 9). Urine albumin decreased by
14.9% (95% CI, 220.4% to 29.4%) with PTF, whereas it
increased by 5.7% (95% CI, 20.3% to 11.1%) in the
control group. ESRD was infrequent in both groups,
occurring in 2 of 82 patients with PTF treatment compared
320
with 3 of 87 patients in the control group. There was no
difference in hospitalization or CVEs between the two
groups, but there were more side effects (transient digestive symptoms in 21.9% assigned to the PTF group
versus 10.3% in control group). Urinary TNF-a also
decreased with PTF. These preliminary results of this
limited, single–center, open–label trial are exciting. This
enthusiasm should be balanced by experience with prior
trials aimed at slowing diabetic nephropathy that showed
early promise but were not effective in larger studies.
Microbiome
The role of the gut microbiome will be discussed
in more detail in the cardiovascular section. Briefly, the
gut microbiota is altered with CKD. Increased fermentation by certain harmful bacteria leads to increased
toxic metabolites, such as p-cresol and indoxyl sulfate.
Short–chain fatty acids produced by certain bacteria
may protect against kidney injury. A disrupted gut
barrier leads to increased endotoxin translocation,
which also contributes to the increased inflammatory
state in CKD. There have been prior trials with prebiotics, nondigested food products that promote the
growth of bacteria that are beneficial, and probiotics,
which are reviewed by Ramezani and Raj (57). However, there was no demonstration of a decrease in
progression. The phase 3 trial that evaluated the orally
administered spherical carbon adsorbent AST-120
(note there is no abbreviation for AST-120) that binds
gut indoxyl sulfate was reported in the last NephSAP
CKD and progression issue. This trial showed no
difference in CKD progression with treatment. Interventions aimed at restoring the gut microbiome in CKD
will be an area of increased future research.
Prediction Models
The ability to accurately predict who will have
rapidly progressive CKD would help providers focus
on which patients need closer monitoring and earlier
nephrology referral. In 2011, Tangri et al. (58) constructed prediction models for RRT that were developed
and validated in Canadian patients with stages 3–5 CKD.
The eight-variable model by Tangri et al. (58) used age,
sex, eGFR, albuminuria, serum calcium, serum phosphate, serum bicarbonate, and serum albumin to predict
dialysis or preemptive kidney transplant in individuals
with stages 3–5 CKDs. The ROC curve C statistics were
0.92 and 0.84 for the prediction of progression to ESRD
in their development and validation cohorts, respectively.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
The net classification improvement for outcome was
superior to the other models with fewer variables by
Tangri et al. (58). A meta-analysis of prediction models
for patients with CKD determined the models by Tangri
et al. (58) above to be the most clinically useful for
predicting CKD progression (59).
Elley et al. (60) aimed to develop a prediction
model specifically for individuals with type 2 diabetes
and less advanced CKD. This group used the New
Zealand Diabetes Cohort Study, which included 25,736
individuals with median eGFR of 77 ml/min per 1.73 m2.
The most complex model by Elley et al. (60) included
sex, ethnicity in New Zealand, age, diabetes duration,
albuminuria, SCr, systolic BP, hemoglobin A1C, smoking status, and history of CVD. This 10-variable model
performed well in the derivation and validation cohorts
for predicting the outcomes of ESRD or death with ESRD
as a contributing cause (ROC curve C statistics: 0.89 and
0.92, respectively). This 10-variable model for individuals with type 2 diabetes and higher eGFR values outperformed the models by Tangri et al. (58) in
predicting fatal and nonfatal ESRD outcomes. It is
unclear if the model by Tangri et al. (58) was used as
a comparator in the study. Death was not one of the
outcomes tested for in the models by Tangri et al.
(58). Elley et al. (60) felt that inclusion of ethnicity in
New Zealand increased the accuracy of the model.
For adults .65 years old, Drawz et al. (61) created
a model for predicting ESRD at 1 year in those with
baseline eGFR values ,30 ml/min per 1.73 m2. This was
developed in 1866 veteran participants. The model used
age, systolic BP, eGFR, potassium, serum albumin, and
CHF. Interestingly, proteinuria was not helpful for
improving prediction of ESRD in this model. In the
validation cohort, the C statistic was 0.82 compared to
0.78 using the model by Tangri et al. (58).
Rucci et al. (62) examined predictive factors that
determined annual decline in eGFR before ESRD in the
Prevention of Renal Insufficiency Progression Project.
This study included 2265 participants in Italy who had
four or more SCr measurements in 1 year. In general, this
was an elderly cohort with mean age of 71 years old, and
mean eGFR by the CKD-EPI equation was 28.8 ml/min
per 1.73 m2. As expected, those with proteinuria (urine
dipstick .20 mg/L or urine albumin .20 mg/L) had a
greater annual rate of decline. Those with proteinuria and
eGFR.33 ml/min per 1.73 m2 had the fastest progression
of 23.66 ml/min per 1.73 m2 per year. Individuals who
were proteinuric with eGFR,33 ml/min per 1.73 m2
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
and serum phosphorus .4.3 mg/dl had a decline of
22.83 ml/min per 1.73 m2 per year. If proteinuria was
absent and age was ,67 years old, individuals with
diabetes had a progression of 22.98 ml/min per 1.73 m2
per year.
The inclusion of ethnicity and CVD may improve
the performance of future prediction models for progression to ESRD. However, vascular disease, defined
as cardiovascular and peripheral vascular, was not predictive in the models by Tangri et al. (58).
Conclusions
Blacks have an increased incidence of CKD.
Those with two APOL1 risk variants were shown to
have increased risk of CKD progression. Black
participants without diabetes and with two high–risk
APOL1 alleles in the AASK Study had a higher risk of
doubling their SCr or developing ESRD. Blacks with or
without diabetes and with two APOL1 risk alleles had
faster CKD progression and higher chance of ESRD or
.50% decrease in measured GFR. Additional genes,
inflammation, infection, or environmental factors are
required to cause progressive CKD in individuals with
high–risk APOL1 variants.
Adiposity is linked to CKD progression and most
likely caused by associated hypertension, insulin resistance, and inflammation. Obesity is traditionally quantified by BMI, but this may not be the optimal measure for
risk in patients with CKD. Studies showed that central
adiposity as measured by WHR or visceral adiposity as
measured by liver ultrasound or magnetic resonance
imaging may prove more accurate in determining the risk
of CKD progression. One large cohort study revealed that
low BMIs may have attendant risks for progressive CKD
as well. Nephrologists may counsel on alterations in food
content, exercise, and finally, bariatric surgery for the
severely obese in attempts to slow CKD progression,
although there are no large therapeutic trials that validate
these measures.
Studies that assessed dietary interventions to slow
CKD progression showed mixed results. Fluid intake did
not seem to influence GFR in a large Australian cohort.
Dietary patterns did not seem to alter incidence of ESRD
in the REGARDS cohort, although plant-based diets
were associated with improved mortality for patients
with CKD, whereas Southern diets were associated with
increased mortality. Because the competing risk of death
was 6-fold higher than ESRD, a beneficial effect may
have been masked. In the ONTARGET cohort of
321
individuals with diabetes, the healthiest tertile diet had
a 27% lower risk of incident CKD. A pilot study using
a low-protein diet supplemented with a-keto analogs of
essential amino acids showed some initial promise in
slowing eGFR loss in those with rapidly progressive
stage 3, stage 4, or nondialysis stage 5 CKD and those
with refractory proteinuria. In pregnant women with
CKD, this diet seemed to decrease the likelihood of
small for gestational age babies but did not show a
significant improvement in longer–term infant growth or
kidney maternal outcomes.
Two cohort studies with varying sodium intakes did
not seem to show an improvement in CKD progression.
A post hoc analysis of dietary sodium intake in participants in the MDRD Study showed that urinary excretion
of ,3 g/d was associated with protection against kidney
failure in those with ,1 g/d proteinuria but increased risk
with the same restriction for those with .1 g/d proteinuria.
A placebo, controlled, cross–over trial of short duration
confirmed the benefits of low-sodium intake of 60–80
mmol/d on reducing BP and proteinuria, although clinical
results could not be assessed. Perhaps overadjustments for
BP and albumin excretion in observational studies of
sodium intake attenuate associations with improved kidney outcomes. However, there does not seem to be a
benefit in restricting sodium intake to ,2–3 g/d in these
observational studies. As mentioned previously, counseling or a goal for sodium restriction does not necessarily
equate to actual intake for patients.
Lower serum bicarbonate may be a biomarker of
progressive CKD. In addition, in carefully selected
patients without hyperkalemia, a diet high in F1V that
provides alkali as potassium citrate seemed to stabilize
GFR as well as sodium bicarbonate in trials of participants with acidotic stage 4 CKD as well as stage 3 CKD
with normal serum bicarbonate but severely increased
albuminuria. Optimal serum bicarbonate levels are unknown. Participants in the CRIC cohort who had higher
serum bicarbonate over 24 mEq/L had an increased
association with CHF.
Excessive volume overload may be a risk factor
independent of hypertension, diabetes, and CVD for CKD
progression, because it can lead to vascular remodeling
and inflammation. A Taiwanese cohort study of stages 4
and 5 CKD showed that those with volume overload
measured by baseline bioimpedance spectroscopy had an
increased risk for RRT and rapid progression. Tsai et al.
(43) felt that fluid overload was more important than
diabetes in CKD progression in a separate analysis. This
322
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
association is intriguing, although confirmation in other
studies will be required.
The presence of vascular disease is associated
with CKD progression. Atherosclerosis detected by a low
ABI and stroke was associated with markedly increased
odds for RRT in observational studies. Increased PP was
associated with small increased odds of a rapid eGFR
decline and incident CKD. aPWV was associated with
a 42% increase in incident CKD but required a doubling
of this value. Retinal abnormalities in the CRIC Study
were not associated with ESRD or eGFR decline. A
CVE in those with CKD referred to nephrology predicted
ESRD.
It is unclear if sleep disorders play a role in CKD
progression because of the difficulty in separating the
independent effects of sleep disorders and obesity. In
a Japanese cohort with moderate CKD and normal
BMI, nocturnal hypoxemia was associated with more
rapid GFR loss.
End points for CKD progression have traditionally been selected as a doubling of SCr or ESRD.
Data from the large Chronic Kidney Disease Prognosis
Consortium suggest that a .30% decline in eGFR over 2
years is strongly associated with ESRD. This end point
may help evaluate the efficacy of therapies that would
normally require a larger number of patients with longer
time to complete. One potential intervention is the use of
PTF, which has anti-inflammatory, antiproliferative, and
antifibrotic actions that could be beneficial in slowing
CKD progression. One open-label trial reported promising results in reducing eGFR loss in patients with type
2 diabetes with a mean GFR of 37 ml/min per 1.73 m2
who also had severe proteinuria. However, there were
too few ESRD events to assess this end point. Larger
placebo, controlled trials will hopefully confirm these
results. Future interventions to restore the normal gut
microbiome altered by CKD may hopefully slow CKD
progression as well as reduce mortality.
References
1. Caramori ML, Parks A, Mauer M: Renal lesions predict progression of
diabetic nephropathy in type 1 diabetes. J Am Soc Nephrol 24: 1175–
1181, 2013 PubMed
2. Kramer HJ, Nguyen QD, Curhan G, Hsu CY: Renal insufficiency in the
absence of albuminuria and retinopathy among adults with type 2
diabetes mellitus. JAMA 289: 3273–3277, 2003 PubMed
3. Bhensdadia NM, Hunt KJ, Lopes-Virella MF, Michael Tucker J,
Mataria MR, Alge JL, Neely BA, Janech MG, Arthur JM; Veterans
Affairs Diabetes Trial (VADT) study group: Urine haptoglobin levels
predict early renal functional decline in patients with type 2 diabetes.
Kidney Int 83: 1136–1143, 2013 PubMed
4. Parsa A, Kao WH, Xie D, Astor BC, Li M, Hsu CY, Feldman HI, Parekh
RS, Kusek JW, Greene TH, Fink JC, Anderson AH, Choi MJ, Wright JT
Jr., Lash JP, Freedman BI, Ojo A, Winkler CA, Raj DS, Kopp JB, He J,
Jensvold NG, Tao K, Lipkowitz MS, Appel LJ; AASK Study Investigators;
CRIC Study Investigators: APOL1 risk variants, race, and progression of
chronic kidney disease. N Engl J Med 369: 2183–2196, 2013 PubMed
5. Stenvinkel P, Zoccali C, Ikizler TA: Obesity in CKD–what should
nephrologists know? J Am Soc Nephrol 24: 1727–1736, 2013 PubMed
6. de Vries APRP, Ruggenenti P, Ruan XZ, Praga M, Cruzado JM, Bajema
IM, D’Agati VD, Lamb HJ, Pongrac Barlovic D, Hojs R, Abbate M,
Rodriquez R, Mogensen CE, Porrini E; ERA-EDTA Working Group
Diabesity: Fatty kidney: Emerging role of ectopic lipid in obesity-related
renal disease. Lancet Diabetes Endocrinol 2: 417–426, 2014 PubMed
7. Stefan N, Artunc F, Heyne N, Machann J, Schleicher ED, Häring HU:
Obesity and renal disease: Not all fat is created equal and not all obesity
is harmful to the kidneys [published online ahead of print April 20,
2014]. Nephrol Dial Transplant PubMed
8. Blüher M: The distinction of metabolically ‘healthy’ from ‘unhealthy’
obese individuals. Curr Opin Lipidol 21: 38–43, 2010 PubMed
9. Obermayr RPTC, Temml C, Knechtelsdorfer M, Gutjahr G, Kletzmayr
J, Heiss S, Ponholzer A, Madersbacher S, Oberbauer R, Klauser-Braun
R: Predictors of new-onset decline in kidney function in a general
middle-european population. Nephrol Dial Transplant 23: 1265–1273,
2008 PubMed
10. Grubbs V, Lin F, Vittinghoff E, Shlipak MG, Peralta CA, Bansal N,
Jacobs DR, Siscovick DS, Lewis CE, Bibbins-Domingo K: Body mass
index and early kidney function decline in young adults: A longitudinal
analysis of the CARDIA (Coronary Artery Risk Development in Young
Adults) study. Am J Kidney Dis 63: 590–597, 2014 PubMed
11. Chang A, Van Horn L, Jacobs DR Jr., Liu K, Muntner P, Newsome B,
Shoham DA, Durazo-Arvizu R, Bibbins-Domingo K, Reis J, Kramer H:
Lifestyle-related factors, obesity, and incident microalbuminuria: The
CARDIA (Coronary Artery Risk Development in Young Adults) study.
Am J Kidney Dis 62: 267–275, 2013 PubMed
12. Agarwal R, Bills JE, Light RP: Diagnosing obesity by body mass index
in chronic kidney disease: An explanation for the “obesity paradox?”
Hypertension 56: 893–900, 2010 PubMed
13. Kwakernaak AJ, Zelle DM, Bakker SJ, Navis G: Central body fat
distribution associates with unfavorable renal hemodynamics independent of body mass index. J Am Soc Nephrol 24: 987–994, 2013 PubMed
14. Targher G, Mantovani A, Pichiri I, Mingolla L, Cavalieri V, Mantovani
W, Pancheri S, Trombetta M, Zoppini G, Chonchol M, Byrne CD, Bonora
E: Nonalcoholic fatty liver disease is independently associated with an
increased incidence of chronic kidney disease in patients with type 1
diabetes. Diabetes Care 37: 1729–1736, 2014 PubMed
15. Lu JL, Kalantar-Zadeh K, Ma JZ, Quarles LD, Kovesdy CP: Association of body mass index with outcomes in patients with CKD. J Am Soc
Nephrol 25: 2088–2096, 2014 PubMed
16. Abou-Mrad RM, Abu-Alfa AK, Ziyadeh FN: Effects of weight reduction
regimens and bariatric surgery on chronic kidney disease in obese patients.
Am J Physiol Renal Physiol 305: F613–F617, 2013 PubMed
17. Navaneethan SD, Yehnert H: Bariatric surgery and progression of
chronic kidney disease. Surg Obes Relat Dis 5: 662–665, 2009 PubMed
18. Clark WF, Sontrop JM, Macnab JJ, Suri RS, Moist L, Salvadori M, Garg
AX: Urine volume and change in estimated GFR in a community-based
cohort study. Clin J Am Soc Nephrol 6: 2634–2641, 2011 PubMed
19. Palmer SC, Wong G, Iff S, Yang J, Jayaswal V, Craig JC, Rochtchina E,
Mitchell P, Wang JJ, Strippoli GFM: Fluid intake and all-cause
mortality, cardiovascular mortality and kidney function: A populationbased longitudinal cohort study. Nephrol Dial Transplant 29: 1377–1384,
2014 PubMed
20. Gutiérrez OM, Muntner P, Rizk DV, McClellan WM, Warnock DG,
Newby PK, Judd SE: Dietary patterns and risk of death and progression
to ESRD in individuals with CKD: A cohort study. Am J Kidney Dis 64:
204–213, 2014 PubMed
323
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
21. Tirosh A, Golan R, Harman-Boehm I, Henkin Y, Schwarzfuchs D,
Rudich A, Kovsan J, Fiedler GM, Blüher M, Stumvoll M, Thiery J,
Stampfer MJ, Shai I: Renal function following three distinct weight loss
dietary strategies during 2 years of a randomized controlled trial.
Diabetes Care 36: 2225–2232, 2013 PubMed
22. Juraschek SP, Appel LJ, Anderson CA, Miller ER 3rd: Effect of a highprotein diet on kidney function in healthy adults: Results from the
OmniHeart trial. Am J Kidney Dis 61: 547–554, 2013 PubMed
23. Dunkler D, Dehghan M, Teo KK, Heinze G, Gao P, Kohl M, Clase CM,
Mann JF, Yusuf S, Oberbauer R; ONTARGET Investigators: Diet and
kidney disease in high-risk individuals with type 2 diabetes mellitus.
JAMA Intern Med 173: 1682–1692, 2013 PubMed
24. Piccoli GB, Ferraresi M, Deagostini MC, Vigotti FN, Consiglio V,
Scognamiglio S, Moro I, Clari R, Fassio F, Biolcati M, Porpiglia F:
Vegetarian low-protein diets supplemented with keto analogues: A
niche for the few or an option for many? Nephrol Dial Transplant 28:
2295–2305, 2013 PubMed
25. Piccoli GB, Leone F, Attini R, Parisi S, Fassio F, Deagostini MC,
Ferraresi M, Clari R, Ghiotto S, Biolcati M, Giuffrida D, Rolfo A,
Todros T: Association of low-protein supplemented diets with fetal
growth in pregnant women with CKD. Clin J Am Soc Nephrol 9: 864–
873, 2014 PubMed
26. Al-Solaiman Y, Jesri A, Zhao Y, Morrow JD, Egan BM: Low-Sodium
DASH reduces oxidative stress and improves vascular function in saltsensitive humans. J Hum Hypertens 23: 826–835, 2009 PubMed
27. McQuarrie EP, Traynor JP, Taylor AH, Freel EM, Fox JG, Jardine AG,
Mark PB: Association between urinary sodium, creatinine, albumin, and
long-term survival in chronic kidney disease. Hypertension 64: 111–
117, 2014 PubMed
28. Fan L, Tighiouart H, Levey AS, Beck GJ, Sarnak MJ: Urinary sodium
excretion and kidney failure in nondiabetic chronic kidney disease.
Kidney Int 86: 582–588, 2014 PubMed
29. Vegter S, Perna A, Postma MJ, Navis G, Remuzzi G, Ruggenenti P:
Sodium intake, ACE inhibition, and progression to ESRD. J Am Soc
Nephrol 23: 165–173, 2012 PubMed
30. McMahon EJ, Bauer JD, Hawley CM, Isbel NM, Stowasser M, Johnson
DW, Campbell KL: A randomized trial of dietary sodium restriction in
CKD. J Am Soc Nephrol 24: 2096–2103, 2013 PubMed
31. Driver TH, Shlipak MG, Katz R, Goldenstein L, Sarnak MJ,
Hoofnagle AN, Siscovick DS, Kestenbaum B, de Boer IH, Ix JH:
Low serum bicarbonate and kidney function decline: The MultiEthnic Study of Atherosclerosis (MESA). Am J Kidney Dis 64: 534–541,
2014 PubMed
32. Goldenstein L, Driver TH, Fried LF, Rifkin DE, Patel KV, Yenchek
RH, Harris TB, Kritchevsky SB, Newman AB, Sarnak MJ, Shlipak MG,
Ix JH; Health ABC Study Investigators: Serum bicarbonate concentrations and kidney disease progression in community-living elders: The
Health, Aging, and Body Composition (Health ABC) Study. Am J
Kidney Dis 64: 542–549, 2014 PubMed
33. de Brito-Ashurst I, Varagunam M, Raftery MJ, Yaqoob MM: Bicarbonate supplementation slows progression of CKD and improves
nutritional status. J Am Soc Nephrol 20: 2075–2084, 2009 PubMed
34. Mahajan A, Simoni J, Sheather SJ, Broglio KR, Rajab MH, Wesson
DE: Daily oral sodium bicarbonate preserves glomerular filtration rate
by slowing its decline in early hypertensive nephropathy. Kidney Int 78:
303–309, 2010 PubMed
35. Phisitkul S, Khanna A, Simoni J, Broglio K, Sheather S, Rajab MH,
Wesson DE: Amelioration of metabolic acidosis in patients with low
GFR reduced kidney endothelin production and kidney injury, and
better preserved GFR. Kidney Int 77: 617–623, 2010 PubMed
36. Goraya N, Simoni J, Jo CH, Wesson DE: A comparison of treating
metabolic acidosis in CKD stage 4 hypertensive kidney disease with
fruits and vegetables or sodium bicarbonate. Clin J Am Soc Nephrol 8:
371–381, 2013 PubMed
37. Goraya N, Simoni J, Jo CH, Wesson DE: Treatment of metabolic acidosis
in patients with stage 3 chronic kidney disease with fruits and vegetables or
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
oral bicarbonate reduces urine angiotensinogen and preserves glomerular
filtration rate. Kidney Int 86: 1031–1038, 2014 PubMed
Dobre M, Yang W, Chen J, Drawz P, Hamm LL, Horwitz E, Hostetter
T, Jaar B, Lora CM, Nessel L, Ojo A, Scialla J, Steigerwalt S, Teal V,
Wolf M, Rahman M; CRIC Investigators: Association of serum
bicarbonate with risk of renal and cardiovascular outcomes in CKD:
A report from the Chronic Renal Insufficiency Cohort (CRIC) study.
Am J Kidney Dis 62: 670–678, 2013 PubMed
Coresh J, Turin TC, Matsushita K, Sang Y, Ballew SH, Appel LJ,
Arima H, Chadban SJ, Cirillo M, Djurdjev O, Green JA, Heine GH,
Inker LA, Irie F, Ishani A, Ix JH, Kovesdy CP, Marks A, Ohkubo T,
Shalev V, Shankar A, Wen CP, de Jong PE, Iseki K, Stengel B,
Gansevoort RT, Levey AS; CKD Prognosis Consortium: Decline in
estimated glomerular filtration rate and subsequent risk of end-stage
renal disease and mortality. JAMA 311: 2518–2531, 2014 PubMed
Turin TC, James M, Ravani P, Tonelli M, Manns BJ, Quinn R, Jun M,
Klarenbach S, Hemmelgarn BR: Proteinuria and rate of change in
kidney function in a community-based population. J Am Soc Nephrol
24: 1661–1667, 2013 PubMed
Chen SCSH, Su HM, Hung CC, Chang JM, Liu WC, Tsai JC, Lin MY,
Hwang SJ, Chen HC: Echocardiographic parameters are independently
associated with rate of renal function decline and progression to dialysis
in patients with chronic kidney disease. Clin J Am Soc Nephrol 6: 2750–
2758, 2011 PubMed
Tsai YCTJ, Tsai JC, Chen SC, Chiu YW, Hwang SJ, Hung CC, Chen
TH, Kuo MC, Chen HC: Association of fluid overload with kidney
disease progression in advanced CKD: A prospective cohort study. Am
J Kidney Dis 63: 68–75, 2014 PubMed
Tsai YCTJ, Tsai JC, Chiu YW, Kuo HT, Chen SC, Hwang SJ, Chen
TH, Kuo MC, Chen HC: Is fluid overload more important than diabetes
in renal progression in late chronic kidney disease? PLoS One 8:
e82566, 2013 PubMed
Foster MC, Ghuman N, Hwang SJ, Murabito JM, Fox CS: Low anklebrachial index and the development of rapid estimated GFR decline and
CKD. Am J Kidney Dis 61: 204–210, 2013 PubMed
Hung P-H, Huang Y-T, Hsiao C-Y, Sung P-S, Guo H-R, Tsai K-J:
Young stroke patients are at high risk for subsequent end-stage renal
disease: A population-based observational study. Nephrol Dial Transplant 29: 873–878, 2014 PubMed
Madero M, Peralta C, Katz R, Canada R, Fried L, Najjar S, Shlipak M,
Simonsick E, Lakatta E, Patel K, Rifkin D, Hawkins M, Newman A,
Sarnak M; Health ABC Study: Association of arterial rigidity with
incident kidney disease and kidney function decline: The Health ABC
study. Clin J Am Soc Nephrol 8: 424–433, 2013 PubMed
Grunwald JE, Pistilli M, Ying GS, Daniel E, Maguire MG, Xie D,
Whittock-Martin R, Parker Ostroff C, Lo JC, Townsend RR, Gadegbeku CA, Lash JP, Fink JC, Rahman M, Feldman HI, Kusek JW;
Chronic Renal Insufficiency Cohort Study Investigators: Retinopathy
and progression of CKD: The CRIC study. Clin J Am Soc Nephrol 9:
1217–1224, 2014 PubMed
Peterson GE, de Backer T, Contreras G, Wang X, Kendrick C, Greene T,
Appel LJ, Randall OS, Lea J, Smogorzewski M, Vagaonescu T, Phillips
RA; African American Study of Kidney Disease Investigators: Relationship of left ventricular hypertrophy and diastolic function with cardiovascular and renal outcomes in African Americans with hypertensive
chronic kidney disease. Hypertension 62: 518–525, 2013 PubMed
Sud M, Tangri N, Pintilie M, Levey AS, Naimark D: Risk of end-stage
renal disease and death after cardiovascular events in chronic kidney
disease. Circulation 130: 458–465, 2014 PubMed
Hanly PJ, Ahmed SB: Sleep apnea and the kidney: Is sleep apnea
a risk factor for chronic kidney disease? Chest 146: 1114–1122,
2014 PubMed
Ahmed SB, Ronksley PE, Hemmelgarn BR, Tsai WH, Manns BJ,
Tonelli M, Klarenbach SW, Chin R, Clement FM, Hanly PJ:
Nocturnal hypoxia and loss of kidney function. PLoS One 6:
e19029, 2011 PubMed
324
52. Sakaguchi Y, Hatta T, Hayashi T, Shoji T, Suzuki A, Tomida K, Okada
N, Rakugi H, Isaka Y, Tsubakihara Y: Association of nocturnal
hypoxemia with progression of CKD. Clin J Am Soc Nephrol 8:
1502–1507, 2013 PubMed
53. Bose B, Badve SV, Hiremath SS, Boudville N, Brown FG, Cass A, de
Zoysa JR, Fassett RG, Faull R, Harris DC, Hawley CM, Kanellis J,
Palmer SC, Perkovic V, Pascoe EM, Rangan GK, Walker RJ, Walters
G, Johnson DW: Effects of uric acid-lowering therapy on renal
outcomes: A systematic review and meta-analysis. Nephrol Dial
Transplant 29: 406–413, 2014 PubMed
54. Hosoya T, Kimura K, Itoh S, Inaba M, Uchida S, Tomino Y, Makino H,
Matsuo S, Yamamoto T, Ohno I, Shibagaki Y, Iimuro S, Imai N,
Kuwabara M, Hayakawa H: The effect of febuxostat to prevent a further
reduction in renal function of patients with hyperuricemia who have
never had gout and are complicated by chronic kidney disease stage 3:
Study protocol for a multicenter randomized controlled study. Trials 15:
26, 2014 PubMed
55. Goicoechea M, García de Vinuesa S, Quiroga B, Verdalles U,
Barraca D, Yuste C, Panizo N, Verde E, Muñoz MA, Luño J:
Effects of pentoxifylline on inflammatory parameters in chronic
kidney disease patients: A randomized trial. J Nephrol 25: 969–975,
2012 PubMed
56. Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, Chahin
J, Méndez ML, Gallego E, Macía M, del Castillo N, Rivero A, Getino
MA, García P, Jarque A, García J: Effect of pentoxifylline on renal
function and urinary albumin excretion in patients with diabetic
kidney disease: The PREDIAN trial. J Am Soc Nephrol 26: 220–229,
2015 PubMed
57. Ramezani A, Raj DS: The gut microbiome, kidney disease, and targeted
interventions. J Am Soc Nephrol 25: 657–670, 2014 PubMed
58. Tangri N, Stevens LA, Griffith J, Tighiouart H, Djurdjev O, Naimark
D, Levin A, Levey AS: A predictive model for progression of
chronic kidney disease to kidney failure. JAMA 305: 1553–1559,
2011 PubMed
59. Tangri N, Kitsios GD, Inker LA, Griffith J, Naimark DM, Walker S,
Rigatto C, Uhlig K, Kent DM, Levey AS: Risk prediction models for
patients with chronic kidney disease: A systematic review. Ann Intern
Med 158: 596–603, 2013 PubMed
60. Elley CR, Robinson T, Moyes SA, Kenealy T, Collins J, Robinson E,
Orr-Walker B, Drury PL: Derivation and validation of a renal risk
score for people with type 2 diabetes. Diabetes Care 36: 3113–3120,
2013 PubMed
61. Drawz PE, Goswami P, Azem R, Babineau DC, Rahman M: A simple
tool to predict end-stage renal disease within 1 year in elderly adults
with advanced chronic kidney disease. J Am Geriatr Soc 61: 762–768,
2013 PubMed
62. Rucci P, Mandreoli M, Gibertoni D, Zuccalà A, Fantini MP, Lenzi J,
Santoro A; Prevention of Renal Insufficiency Progression (PIRP)
Project: A clinical stratification tool for chronic kidney disease progression rate based on classification tree analysis. Nephrol Dial Transplant 29: 603–610, 2014 PubMed
Diabetic Kidney Disease and Kidney Biopsy
The classic course of diabetic nephropathy is
development of increasing albuminuria from microalbuminuria to overt proteinuria and then, decline in
glomerular filtration, leading to ESRD. It has now been
recognized that many individuals with diabetes develop
kidney disease with a low eGFR but without overt
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
proteinuria. Recent studies have found that this heterogeneity in kidney disease is also reflected in a corresponding heterogeneity in kidney biopsies, particularly
in those without overt albuminuria.
Ekinci et al. (1) invited patients attending the
Diabetes Clinics at Austin Health with type 2 diabetes,
an eGFR ,60 ml/min per 1.73 m2, and an albuminuria
,200 mg/min (approximately 300 mg/d) to participate
in a study that included a kidney biopsy; 14 participants
(10 with normoalbuminuria and 4 with microalbuminuria, although 2 with normoalbuminuria had microalbuminuria after renin-angiotensin-aldosterone system
blocking agents were held) were biopsied. The biopsy
results from these research biopsies were compared
with biopsies from individuals with diabetes and overt
proteinuria who underwent biopsies for clinical indications (n¼17). Typical histologic changes consistent
with diabetic nephropathy were seen in all patients
with macroalbuminuria, five of six (83%) patients with
microalbuminuria, and only three of eight patients with
normoalbuminuria (mean eGFR ¼31 ml/min per
1.73 m2). In three of eight patients with normoalbuminuria, predominantly vascular or interstitial changes
were seen. The mean mesangial area was greater,
with greater degrees of albuminuria (Figure 10).
Most kidney biopsies in individuals with diabetes are done because of atypical clinical factors that
suggest a nondiabetic process. Sharma et al. (2)
reviewed all consecutive native renal biopsies from
2011 that had been reviewed at the Columbia Renal
Pathology Laboratory. Of 2642 biopsies during this
time period, almost one quarter (23.5%) were performed in individuals with diabetes. Diabetic nephropathy alone was found in 37% of specimens, diabetic
nephropathy plus nondiabetic kidney disease was
found in 27% of specimens, and nondiabetic kidney
disease alone was found in 36% of biopsies. Although
the proportion with nondiabetic kidney disease is high,
it is important to remember that this is a selected
population that underwent biopsy for clinical reasons.
The study also evaluated clinical predictors of finding
diabetic kidney disease or nondiabetic kidney disease.
The main predictor was duration of diabetes. A longer
duration of diabetes was associated with a greater
likelihood of finding diabetic nephropathy and a lower
likelihood of finding nondiabetic kidney disease; each
additional 1 year of diabetes decreased the odds of
nondiabetic kidney disease by 5%. Using receiver
operating characteristic curves, a duration of diabetes
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 10. Increase in mean mesangial area with greater
levels of albuminuria in type 2 diabetes. The figure shows
mean mesangial area (micrometer2) across normal controls
(normal C), patients with type 2 diabetes and normoalbuminuria (normo), patients with type 2 diabetes and microalbuminuria (micro), and patients with type 2 diabetes and
macroalbuminuria (macro). P¼0.02 (one-way ANOVA for
mean mesangial area across four groups). Reprinted with
permission from Ekinci EI, Jerums G, Skene A, Crammer P,
Power D, Cheong KY, Panagiotopoulos S, McNeil K, Baker
ST, Fioretto P, Macisaac RJ: Renal structure in normoalbuminuric and albuminuric patients with type 2 diabetes and
impaired renal function. Diabetes Care 36: 3620–3626,
2013.
$12 years was the best predictor of finding diabetic
nephropathy alone (57.5% sensitivity, 73.3% specificity, 56.0% positive predictive value, and 74.5% negative predictive value).
References
1. Ekinci EI, Jerums G, Skene A, Crammer P, Power D, Cheong KY,
Panagiotopoulos S, McNeil K, Baker ST, Fioretto P, Macisaac RJ: Renal
structure in normoalbuminuric and albuminuric patients with type 2
diabetes and impaired renal function. Diabetes Care 36: 3620–3626,
2013 PubMed
2. Sharma SG, Bomback AS, Radhakrishnan J, Herlitz LC, Stokes MB,
Markowitz GS, D’Agati VD: The modern spectrum of renal biopsy
findings in patients with diabetes. Clin J Am Soc Nephrol 8: 1718–1724,
2013 PubMed
Renin-Angiotensin-Aldosterone System Blockers
It is not uncommon that physicians are uncomfortable treating individuals with advanced kidney
disease with an angiotensin–converting enzyme inhibitor
(ACEI) or angiotensin receptor blocker (ARB). However,
325
the benefits and harms of continuing ACEI or ARB use
have not been directly studied.
Using the National Health Insurance Research
Database in Taiwan, Hsu et al. (1) evaluated the
association between use of ACEIs or ARBs and risk
of ESRD in adult individuals with stage 5 CKD who
had hypertension and were treated with erythropoiesisstimulating agents. Hsu et al. (1) identified 28,497
individuals with stage 5 CKD, approximately one half
of which were on an ACEI, an ARB, or both. The mean
age of the population was 65 years old. Users of ACEIs
or ARBs were younger and more likely to have diabetes
or cardiovascular disease. The majority of users and
nonusers progressed to ESRD (70.7%), whereas 20%
died before reaching ESRD. Users of ACEIs or ARBs
were less likely to progress to dialysis (hazard ratio
[HR], 0.94; 95% confidence interval [95% CI], 0.91 to
0.97) or the composite end point of dialysis or
mortality (HR, 0.94; 95% CI, 0.92 to 0.97). Although
the benefit was small, it is encouraging that the
outcomes are not worse in those taking ACEIs or
ARBs, which would counter the practitioner tendency
to stop these medications in advanced CKD. Although
analyses of outcomes of users versus nonusers of
medications are potentially biased, it is important to
note that there are also no randomized studies that
suggest that we should stop ACEI or ARB use in
patients with advanced kidney disease. The only
randomized study in individuals with advanced kidney disease (SCr 3.1–5.0 mg/dl) and proteinuria
showed that starting an ACEI or ARB slowed progression (doubling of SCr, ESRD, or death) (2). Thus,
the weight of evidence supports continuing ACEIs or
ARBs in patients with advanced kidney disease who
are tolerating the medications, especially if they are
proteinuric.
Treatment with an ACEI or ARB decreases the
progression of kidney disease in patients with proteinuria (3–6). The percentage reduction in proteinuria
with these agents is a marker of response and correlates
with benefit; residual proteinuria after use of ACEIs
and ARBs predicts progression (7).
Imai et al. (8), in a secondary analysis of the
Olmesartan Reducing Incidence of End Stage Renal
Disease in Diabetic Nephropathy (ORIENT) Study,
evaluated the association of baseline proteinuria
level, percentage reduction in proteinuria, and residual
proteinuria with doubling of SCr, ESRD, or death.
326
The ORIENT Study was a double-blind study of
olmesartan versus placebo added onto standard of
care in patients with type 2 diabetes and an albumin to
creatinine ratio (ACR) .300 mg/g. Standard of care
could include an ACEI, and therefore, the olmesartan
group was a mix of patients on dual therapy (73%)
and monotherapy (27%), which was compared with
a placebo group that was predominantly on an ACEI
(9). The mean age was 59 years old, mean creatinine
was 1.62 mg/dl, and median urine ACR was 1.7 g/g.
For baseline proteinuria, compared with individuals
with albuminuria ,1 g/g, the risk of progression was
three times higher in individuals with an ACR of 1–3
g/g and nine times higher in those with an ACR.9 g/g.
Percentage reduction in proteinuria and residual proteinuria (proteinuria at 24 weeks) were both associated with progression, with the lowest risk being for
those individuals who had residual proteinuria ,1 g
and ,30% reduction. However, the title of the paper
labeling albuminuria as a therapeutic target implies
that the percentage change was a response to treatment. The association of proteinuria and change in
proteinuria with progression was seen in both placebo
and olmesartan groups. The study overall was negative
with regards to progression (HR, 0.97; 95% CI, 0.76 to
1.24) (9). Therefore, the association of albuminuria and
change in albuminuria with outcomes indicates that it is
a risk marker, and the study does not inform us
regarding a benefit of adding olmesartan
Because 73% of the individuals in the ORIENT
Study were on an ACEI at baseline, the study was
primarily a study of combination ARB plus ACEI versus
monotherapy with ACEI alone. As reviewed in the prior
CKD NephSAP issue, combination therapy has been
shown to decrease proteinuria but has not been shown to
improve outcomes.
Makani et al. (10) performed a meta-analysis of
studies of dual blockade of the renin-angiotensin
system. Studies were included if they reported data
on long-term efficacy ($1 year) or safety ($4 weeks)
with a sample size of at least 50 individuals. Analysis
was stratified by trials of heart failure (HF) versus nonHF studies; few studies were in patients with kidney
disease. The studies included seven efficacy studies
with 56,824 subjects, and five were HF studies. Dual
therapy had no benefit on mortality overall. There
seemed to be a difference in mortality outcomes when
stratified by HF versus non-HF studies (P for interaction
¼0.02). Combining the two non-HF studies (the
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Ongoing Telmisartan Alone and in Combination with
Ramipril Global Endpoint Trial and Aliskiren Trial
in Type 2 Diabetes Using Cardiorenal Endpoints
(ALTITUDE)), dual therapy was associated with a small
increase in mortality in those without HF (HR, 1.07; 95%
CI, 1.0 to 1.14; P¼0.04). In contrast, there was not an
increased risk in those without HF (HR, 0.96; 95% CI,
0.88 to 1.05). Dual therapy led to a decrease in
admissions for HF overall (HR, 0.82; 95% CI, 0.74 to
0.92), in those with HF (HR, 0.77; 95% CI, 0.68 to
0.88), and in those without HF (HR, 0.91; 95% CI, 0.82
to 1.01). In studies with and without HF, dual therapy
was associated with an increased risk of hyperkalemia,
hypotension, and withdrawal caused by drug–related
adverse events. Dual therapy was also associated with
an increased risk of renal failure indicated as SCr .2.0
mg/dl or a doubling of SCr (HR, 1.41; 95% CI, 1.09 to
1.84), which was predominantly caused by an increased
risk in the studies with HF (HR, 2.19; 95% CI, 1.82 to
2.65).
Since publication of the meta-analysis, VA
NEPHRON-D: Diabetes in Nephropathy Study was
published (11). VA NEPHRON-D compared dual
therapy with losartan and lisinopril with monotherapy
with losartan (plus placebo) in individuals with type 2
diabetes, ACR.300 mg/g, and eGFR¼30–89.9 ml/min
per 1.73 m2. The primary outcome was time for first
event of all-cause mortality, ESRD, or progression of
CKD (absolute reduction of eGFR by at least 30 ml/min
per 1.73 m2 if baseline eGFR was between 60 and
89.9 ml/min per 1.73 m2 at enrollment and halving of
eGFR if baseline eGFR was ,60 ml/min per 1.73 m2).
The study was stopped early by the data monitoring
safety committee; 1448 individuals were randomized,
mean age was 65 years old, mean eGFR was 54 ml/min
per 1.73 m2, and median urine ACR was 845 mg/g.
There was no significant benefit on the primary end
point (HR, 0.88; 95% CI, 0.70 to 1.12), renal progression (change in eGFR or ESRD; HR, 0.78; 95% CI,
0.58 to 1.05), or all-cause mortality (HR, 1.04; 95% CI,
0.73 to 1.49) (Figure 11). As anticipated, combination
therapy decreased the ACR (baseline to 1 year: 786 to
517 mg/g in the combination group versus 829 to 701
mg/g in the monotherapy group). There was a significantly increased risk of hyperkalemia (serum potassium
.6 mEq/L that required an ER visit, admission, or
dialysis; HR, 2.8; 95% CI, 1.8 to 4.3) and AKI (HR,
1.7; 95% CI, 1.3 to 2.2). AKI events were considered
serious adverse events that required or occurred during
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
327
Figure 11. Dual therapy versus monotherapy on primary end point, renal end point, and mortality in the VA NEPHRON-D Study (A)
Primary endpoint (time for first event of all-cause mortality, ESRD, or progression of CKD (absolute reduction of eGFR by at least
30ml/min per 1.73 m2 if baseline eGFR was between 60 and 89.9 ml/min per 1.73 m2 at enrollment and halving of eGFR if baseline
eGFR was, 60 ml/min per 1.73 m2); (B) secondary endpoint ESRD or progression of CKD; (C) all-cause mortality. CI, confidence
interval. Reprinted with permission from Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, Leehey DJ,
McCullough PA, O’Connor T, Palevsky PM, Reilly RF, Seliger SL, Warren SR, Watnick S, Peduzzi P, Guarino P; VA NEPHRON-D
Investigators: Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 369: 1892–1903, 2013.
328
admission, but outpatient changes in creatinine were
not considered AKI events.
Combination ACEI plus ARB decreases
albuminuria but does not significantly decrease progression of kidney disease and
is associated with an increased risk of AKI
and hyperkalemia.
The addition of a mineralocorticoid receptor blocker
to an ACEI or an ARB is another approach for dual
therapy. The addition of spironolactone to full-dose
ACEI lowers proteinuria more than dual ACEI plus
ARB therapy (12,13). However, it is associated with
a much higher risk of hyperkalemia, especially in
individuals with reduced eGFR. Van Buren et al.
(14) reported on hyperkalemia in a randomized study
comparing the addition of placebo, losartan (100mg), or
spironolactone (25mg) to lisinopril (80 mg) in patients
with diabetes and proteinuria. Full details of the study
results have been previously published (12). The study
enrolled 81 individuals with a mean creatinine clearance
of 64.5 ml/min and mean albuminuria of 1 g determined
from 24-hour urine collections. Compared with placebo,
spironolactone decreased albuminuria by 34%, and losartan decreased albuminuria by 17%. A potassium level
.6.0 mEq/L occurred in 7.4%, 38.5%, and 51.9% in the
placebo, losartan, and spironolactone groups, respectively. In addition to study medications, higher baseline
potassium levels (.4.5 mEq/L) and lower 24-hour
creatinine clearances were predictors of hyperkalemia.
Esteghamati et al. (13) performed an 18-month
open–label study of spironolactone added to an ARB in
136 individuals with diabetes and ACR.30 mg/g. At
baseline, individuals were being treated with combination therapy with enalapril (30–40 mg/d) and losartan
(50–100 mg/d) for at least 1 year. Then, participants
were randomized to either a group that continued their
ACEI and ARB combination or one that underwent
enalapril washout with spironolactone substitution at
25 mg daily. The mean age was 58 years old, mean
eGFR was approximately 70 ml/min per 1.73 m2, and
the majority had microalbuminuria (72%). Spironolactone led to a greater reduction in BP (18% difference
in systolic BP and 16% difference in diastolic BP)
and albuminuria (8.4%) at 18 months. The change in
SCr over time was similar in the two groups. The risk
of discontinuation because of hyperkalemia was low.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
However, because the study enrolled individuals
who had been tolerating dual ACEI and ARB therapy
for over 1 year and did not have advanced kidney
disease, the study may have enrolled a group at lower
risk for hyperkalemia (mean serum K and the cutoff
for hyperkalemia were not defined).
At this time, hyperkalemia would be the limiting
factor to studying whether a mineralocorticoid receptor antagonist decreases the risk of progression of
CKD. Whether one of two new potassium–lowering
medications currently under development, patiromer
(15) and sodium zirconium cyclosilicate (16), would
allow continuation of spironolactone as GFR declines, enabling an outcomes study to be conducted,
deserves additional consideration. However, these
medications have not been approved by the Food and
Drug Administration at the time that this issue was
composed.
References
1. Hsu TW, Liu JS, Hung SC, Kuo KL, Chang YK, Chen YC, Hsu CC,
Tarng DC: Renoprotective effect of renin-angiotensin-aldosterone
system blockade in patients with predialysis advanced chronic kidney
disease, hypertension, and anemia. JAMA Intern Med 174: 347–354,
2014 PubMed
2. Hou FF, Zhang X, Zhang GH, Xie D, Chen PY, Zhang WR, Jiang JP,
Liang M, Wang GB, Liu ZR, Geng RW: Efficacy and safety of
benazepril for advanced chronic renal insufficiency. N Engl J Med
354: 131–140, 2006 PubMed
3. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving
HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S; RENAAL Study
Investigators: Effects of losartan on renal and cardiovascular outcomes
in patients with type 2 diabetes and nephropathy. N Engl J Med 345:
861–869, 2001 PubMed
4. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz
E, Atkins RC, Rohde R, Raz I; Collaborative Study Group: Renoprotective effect of the angiotensin-receptor antagonist irbesartan in
patients with nephropathy due to type 2 diabetes. N Engl J Med 345:
851–860, 2001 PubMed
5. Lewis EJ, Hunsicker LG, Bain RP, Rohde RD; The Collaborative
Study Group: The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. N Engl J Med 329: 1456–1462,
1993 PubMed
6. Maschio G, Alberti D, Janin G, Locatelli F, Mann JF, Motolese M,
Ponticelli C, Ritz E, Zucchelli P; The Angiotensin-ConvertingEnzyme Inhibition in Progressive Renal Insufficiency Study Group:
Effect of the angiotensin-converting-enzyme inhibitor benazepril on
the progression of chronic renal insufficiency. N Engl J Med 334:
939–945, 1996 PubMed
7. de Zeeuw D, Remuzzi G, Parving HH, Keane WF, Zhang Z, Shahinfar S,
Snapinn S, Cooper ME, Mitch WE, Brenner BM: Proteinuria, a target for
renoprotection in patients with type 2 diabetic nephropathy: Lessons from
RENAAL. Kidney Int 65: 2309–2320, 2004 PubMed
8. Imai E, Haneda M, Chan JCN, Yamasaki T, Kobayashi F, Ito S,
Makino H: Reduction and residual proteinuria are therapeutic targets
in type 2 diabetes with overt nephropathy: A post hoc analysis
(ORIENT-proteinuria). Nephrol Dial Transplant 28: 2526–2534,
2013 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
9. Imai E, Chan JC, Ito S, Yamasaki T, Kobayashi F, Haneda M, Makino
H; ORIENT study investigators: Effects of olmesartan on renal and
cardiovascular outcomes in type 2 diabetes with overt nephropathy: A
multicentre, randomised, placebo-controlled study. Diabetologia 54:
2978–2986, 2011 PubMed
10. Makani H, Bangalore S, Desouza KA, Shah A, Messerli FH:
Efficacy and safety of dual blockade of the renin-angiotensin
system: Meta-analysis of randomised trials. BMJ 346: f360,
2013 PubMed
11. Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth
W, Leehey DJ, McCullough PA, O’Connor T, Palevsky PM, Reilly
RF, Seliger SL, Warren SR, Watnick S, Peduzzi P, Guarino P;
VA NEPHRON-D Investigators: Combined angiotensin inhibition
for the treatment of diabetic nephropathy. N Engl J Med 369:
1892–1903, 2013 PubMed
12. Mehdi UF, Adams-Huet B, Raskin P, Vega GL, Toto RD: Addition of
angiotensin receptor blockade or mineralocorticoid antagonism to
maximal angiotensin-converting enzyme inhibition in diabetic nephropathy. J Am Soc Nephrol 20: 2641–2650, 2009 PubMed
13. Esteghamati A, Noshad S, Jarrah S, Mousavizadeh M, Khoee SH,
Nakhjavani M: Long-term effects of addition of mineralocorticoid
receptor antagonist to angiotensin II receptor blocker in patients with
diabetic nephropathy: A randomized clinical trial. Nephrol Dial Transplant 28: 2823–2833, 2013 PubMed
14. Van Buren PN, Adams-Huet B, Nguyen M, Molina C, Toto RD:
Potassium handling with dual renin-angiotensin system inhibition in diabetic nephropathy. Clin J Am Soc Nephrol 9: 295–301,
2014 PubMed
15. Weir MR, Bakris GL, Bushinsky DA, Mayo MR, Garza D, Stasiv
Y, Wittes J, Christ-Schmidt H, Berman L, Pitt B; OPAL-HK
Investigators: Patiromer in patients with kidney disease and hyperkalemia receiving RAAS inhibitors. N Engl J Med 372: 211–221,
2015 PubMed
16. Packham DK, Rasmussen HS, Lavin PT, El-Shahawy MA, Roger SD,
Block G, Qunibi W, Pergola P, Singh B: Sodium zirconium cyclosilicate in hyperkalemia. N Engl J Med 372: 222–231, 2015 PubMed
329
images of plaque on the basis of the emission and
reflection of near-infrared light (3–5). Using the
Massachusetts General Hospital Optical Coherence
Tomography Multicenter Registry, Kato et al. (6)
evaluated the plaque characteristics in patients with
and without CKD. Kato et al. (6) analyzed 61 plaques
in 37 patients with CKD and 402 plaques in 250
patients without CKD. CKD was defined as an
eGFR,60 ml/min per 1.73 m2, and the mean eGFR
in individuals with CKD was 48 ml/min per 1.73 m2.
The plaques in individuals with CKD had a higher
prevalence of calcification, cholesterol crystal, and
disruption, with a trend toward a greater prevalence
of thrombus (Figure 12). These findings may indicate
more vulnerable plaques (7).
Nakano et al. (8) evaluated the relationship of
CKD with neovascularization and intraplaque hemorrhage in coronary atherosclerosis in autopsies of elderly
residents of Hisayama, Japan. The Hisayama study is
a population-based study with autopsy verification of
death in 75% of those who died. For the current analysis,
Nakano et al. (8) randomly selected 126 subjects from
844 consecutive autopsies. In individuals with an
eGFR,45 ml/min per 1.73 m2, there was a statistically
Cardiovascular Disease
Since the last issue of CKD NephSAP, there have
been many articles on cardiovascular disease (CVD) in
CKD. We cannot cover all of the interesting and relevant
articles that address CVD and have primarily focused this
section on subtopics that are most applicable to clinical
practice or could become applicable in the near future.
Coronary Plaque Characteristics in CKD
In the last issue of NephSAP, we reviewed studies
that showed that the characteristics of coronary plaques
differ between those with and without CKD. One
particular factor that may increase the risk of a cardiovascular event (CVE) is that individuals with CKD are
more likely to have thin-cap fibroatheromas (1), which
increases the likelihood of plaque rupture (2). Recent
studies have further refined the information regarding
coronary plaque.
Intracoronary optical coherence tomography is an
intravascular technique that produces high-resolution
Figure 12. Qualitative optical coherence tomography findings. Compared with non-CKD plaques, plaques with CKD
had a higher prevalence of calcification, cholesterol crystals,
and disruption. Thrombus tended to be more frequently
observed in the CKD group. TCFA, thin-cap fibroatheroma.
Reprinted with permission from Kato K, Yonetsu T, Jia H,
Abtahian F, Vergallo R, Hu S, Tian J, Kim SJ, Lee H,
McNulty I, Lee S, Uemura S, Jang Y, Park SJ, Mizuno K, Yu
B, Jang IK: Nonculprit coronary plaque characteristics of
chronic kidney disease. Circ Cardiovasc Imaging 6: 448–
456, 2013.
330
greater degree of immunohistochemical identification of
vascular endothelial growth factor and a higher number
of new vessels; in those with an eGFR,30 ml/min per
1.73 m2, there was also greater staining for oxidized
LDL. The odds of intraplaque hemorrhage increased
with lower eGFR; in individuals with an eGFR,30 ml/
min per 1.73 m2, the adjusted odds ratio for intraplaque
hemorrhage was 6.22 (95% confidence interval [95%
CI], 1.10 to 35.04) compared with that in those with an
eGFR$60 ml/min per 1.73 m2. These findings are also
consistent with more vulnerable plaques in individuals
with CKD.
Association of Kidney Disease with CVD in Type
2 Diabetes
Prior issues of the CKD NephSAP have discussed
that individuals with CKD have a higher risk of CVD
and mortality. One reason is the higher prevalence of
diabetes among individuals with CKD. Recent studies
have evaluated the association of CKD with cardiovascular risk and mortality within individuals with type 2
diabetes.
Wang et al. (9) evaluated the association of eGFR
with the development of coronary heart disease (CHD)
and stroke in 28,391 individuals with type 2 diabetes
receiving care in the Louisiana State University Health
Care Services Division and compared whether the associations differed in black and white patients. Individuals
with a prior history of CHD and stroke were excluded.
There was no interaction of sex and eGFR with risk of
CHD or stroke (i.e., the relationship of eGFR with events
was similar in men and women). There was a statistical
interaction of race with eGFR regarding the development
of stroke, although the relationships seem similar with an
increasing risk as GFR decreases in both racial groups. In
white individuals, the rates of stroke per 1000 personyears were 11.9, 15.9, 18.2, 26.9, and 39.3 for
eGFRs$90, 75–89, 60–74, 30–59, and 15–29 ml/min
per 1.73 m2, respectively. In blacks, the stroke rates were
10.9, 12.6, 15.4, 21.4, and 24.8 events per 1000 personyears for eGFR$90, 75–89, 60–74, 30–59, and 15–29 ml/
min per 1.73 m2, respectively.
In type 1 diabetes, two separate cohort studies have
found that kidney disease explains much of the increased
mortality rate seen in type 1 diabetes (10,11). In the
absence of kidney disease, the age– and sex–adjusted
mortality rates in individuals with type 1 diabetes were
similar to those of the general population. Afkarian et al.
(12) evaluated whether this was also true in type 2 diabetes
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
using the third National Health and Nutrition Examination
Survey III (NHANES III). Of 15,762 individuals participating in NHANES III, 9.5% had type 2 diabetes; among
those with diabetes, 42.3% had CKD, with the majority
defined by the presence of albuminuria. Both diabetes
and kidney disease were risk factors for mortality. The
presence of both diabetes and kidney disease showed an
additive interaction for the risk of mortality. In individuals
with diabetes but without kidney disease, there was a small
increased risk of mortality compared with the general
population. However, most of the excess mortality risk
was in those with both diabetes and kidney disease
(Figure 13).
Interaction of Age and CKD on Mortality and
Coronary Event
Recent studies have found that the cardiovascular
risk associated with CKD varies across age groups.
Choi et al. (13) analyzed the association of eGFR with
1-year mortality within age strata in individuals with
myocardial infarction (MI) in the Korean Acute Myocardial Infarction Registry. Choi et al. (13) divided
patients into four age groups: ,55, 55–64, 65–74 and
$75 years old. There were 11,268 participants, and
Figure 13. Increased ten-year mortality in type 2 diabetes
with both albuminuria and impaired eGFR. Absolute differences in mortality risk were estimated using linear regression
and adjusted for age, sex, and race. Standardized 10-year all–
cause cumulative incidences were estimated for the mean
levels of the covariates in the study population. The dashed line
indicates mortality in persons without diabetes or kidney
disease (the reference group). The numbers above bars indicate
excess mortality above the reference group. Error bars indicate
95% confidence intervals. Reprinted with permission from
Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR,
Himmelfarb J, de Boer IH: Kidney disease and increased
mortality risk in type 2 diabetes. J Am Soc Nephrol 24: 302–
308, 2013.
331
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 3. Cox proportional analysis for mortality by age
Reprinted with permission from Choi JS, Kim MJ, Kang YU, Kim CS, Bae EH, Ma SK, Ahn YK, Jeong MH, Kim YJ, Cho MC, Kim CJ, Kim SW; Korea Acute Myocardial Infarction
Registry Investigators: Association of age and CKD with prognosis of myocardial infarction. Clin J Am Soc Nephrol 8: 939–944, 2013.
26% had an eGFR,60 ml/min per 1.73 m2. Mortality
increased as age increased and eGFR decreased.
However, although the absolute mortality rate was
higher in older age with low GFR, the relative mortality
was lower in the older group (Table 3). As an example,
18% of individuals ages 55–74 years old with an eGFR
of 30–44 ml/min per 1.73 m2 had died by 1 year versus
28.6% of those ages $75 years old. The relative risk of
death compared with the same age group with an
eGFR$60 ml/min per 1.73 m2 was 9.93 for those ages
55–64 years old but only 3.36 for those $75 years old.
The difference in relative risk reflects the higher overall
mortality in older individuals as well as competing risks
of other diseases on mortality.
From a prevention and care of an individual patient
point of view, it is more important to use absolute risk
rather than relative risk when considering interventions.
An example is the lipid treatment guidelines that base
treatment for primary prevention on a 10-year absolute
risk of CHD events of .10% (10 per 1000 person-years).
Tonelli et al. (14) analyzed the association of age and
CKD with risk of hospitalization for MI or death caused
by CHD in the Alberta Kidney Disease Network Database
(n¼1,268,538). Not surprisingly, these investigators found
that the risk of CHD events was greater in those with CKD
versus without CKD (Table 4). None of the groups under
age 40 years old had a risk of CHD events .10 per 1000
person-years. In those ages 40–49 years old, the only
group with the risk .10 per 1000 person-years was that
with an eGFR,60 ml/min per 1.73 m2 and an albumin to
creatinine ratio (ACR) .30 mg/g. However, the risk is not
.10 per 1000 person years if one excludes individuals
with diabetes or a prior MI. Participants with CKD over
the age 50 years old with an elevated ACR or a low eGFR
had a high risk of CHD events. This supports the current
Kidney Disease: Improving Global Outcomes (KDIGO)
lipid guidelines to treat all individuals over age of 50 years
old with nondialysis CKD with a statin and treat those
over age 40 years old if they have a 10-year risk .10%
(e.g., with diabetes).
To illustrate where one might draw different
conclusions on the relationship of age and risk in CKD
using absolute and relative risks, we have calculated
the relative risk (incident rate CKD group/incident rate
no CKD group) (Table 4). Overall, one can see that there
is a higher risk of CHD events in those with both lower
eGFR and higher ACR in all age groups. However, from
Table 4, you can see that the highest absolute risk is in
individuals over age 50 years old, but the highest
relative risk is in those younger than age 40 years old.
Lipid Guidelines
In November of 2013, KDIGO published a clinical practice guideline for lipid management in CKD
(15). Also, in November of 2013, the American Heart
Association/American College of Cardiology (AHA/
ACC) updated guideline was released (16). There is
a paradigm shift in the recommendations in these
guidelines. Both move away from LDL-based treatment, and now, treatment recommendations are on
the basis of cardiovascular risk assessment instead of LDL
levels and goals. A summary of the KDIGO recommendations for nondialysis, nontransplant CKD is shown in
Table 5. A major change is that the guidelines recommend
treating all individuals with CKD ages 50 years old and
over with a statin, regardless of LDL levels. Individuals
332
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 4. Rate of coronary death or nonfatal myocardial infarction by age and CKD stage
Primary Cohort
Age ,40 yr
No CKD
Any CKD
eGFR¼15–59.9, ACR.30
eGFR¼15–59.9, ACR,30
eGFR$60, ACR.30
Age 40–49 yr
No CKD
Any CKD
eGFR¼15–59.9, ACR.30
eGFR¼15–59.9, ACR,30
eGFR$60, ACR.30
Age $50 yr
No CKD
Any CKD
eGFR¼15–59.9, ACR.30
eGFR¼15–59.9, ACR,30
eGFR$60, ACR.30
Rate per 1000
person-years (95%
Confidence Interval)
0.2 (0.2 to 0.2)
0.5 (0.4 to 0.6)
3.2 (1.6 to 6.4)
—
0.4 (0.3 to 0.5)
1.1
3.2
10.3
2.6
2.9
(1.1
(2.8
(7.6
(1.7
(2.6
to
to
to
to
to
1.2)
3.5)
14.2)
3.8)
3.3)
5.0
17.3
33.2
16.2
12.8
(4.8 to 5.1)
(16.9 to 17.1)
(31.8 to 34.6)
(15.7 to 16.7)
(12.3 to 13.4)
Subgroup without Diabetes or
Prior Myocardial Infarction
Relative Risk
Rate per 1000
person-years (95%
Confidence Interval)
Relative Risk
1.0 (reference)
2.5
16
—
2
0.2 (0.1 to 0.2)
0.3 (0.2 to 0.4)
—
—
0.3 (0.2 to 0.4)
1.0 (reference)
1.5
—
—
1.5
1.0 (reference)
2.9
9.3
2.3
2.6
1.0
2.1
5.2
1.9
2.0
1.0 (reference)
2.1
5.2
1.9
2.0
1.0 (reference)
3.46
6.6
3.24
2.6
4.2
13.4
25.9
13.3
9.3
(0.9
(1.8
(3.1
(1.2
(1.7
to
to
to
to
to
1.1)
2.4)
8.7)
3.1)
2.3)
(4.1 to 4.3)
(13.0 to 13.8)
(24.3 to 27.5)
(12.8 to 13.9)
(8.7 to 9.8)
1.0 (reference)
3.1
6.2
3.2
2.2
Events that were so small that a reliable estimate could not be calculated are indicated by —. ACR, albumin to creatinine ratio. Modified with permission from Tonelli M,
Muntner P, Lloyd A, Manns B, Klarenbach S, Pannu N, James M, Hemmelgarn B; Alberta Kidney Disease Network: Impact of age on the association between CKD and the risk of
future coronary events. Am J Kidney Dis 64: 375–382, 2014.
age ,50 years old should be treated if they have CVD or
are at high risk for future events.
The treatment recommendations are on the basis
of the CKD subgroup analyses of large statin studies in
mainly non-CKD populations and the Study of Heart
and Renal Protection (SHARP) in CKD. These studies
used a fixed dose of statins in individuals with CVD or
who were at risk for CVD. Because the prior studies did
not adjust statin doses on the basis of target, the new
recommendations reflect the study design. The recommendations also reflect the high risk of CVD in
individuals with CKD, especially in older individuals.
The recommendations to move away from LDL
targets in the AHA/ACC guidelines are controversial
(in light of the recently presented Improved Reduction of Outcomes: Vytorin Efficacy International Trial
(IMPROVE-IT). The results of the IMPROVE-IT were
presented at the AHA meeting and are unpublished.
From the presentation, the study found that the addition of ezetimibe to simvastatin in patients with acute
coronary syndrome (ACS) reduced LDL further (54 versus
69 mg/dl) and reduced CVE (primary end point) by 6%.
The findings suggest that lowering LDL further in highrisk individuals is beneficial, but the negative findings in
other studies that included the addition of a fibrate or niacin
to a statin indicate that it may matter how LDL is lowered.
An argument could be made that the age cutoff in
the KDIGO guidelines for treatment should be age 40
years old, because the SHARP Study enrolled individuals over the age of 40 years old(17); therefore, using
this cutoff would put the guideline more in line with the
trial evidence, and using age 40 years old would put
the KDIGO guidelines more in line with the AHA/ACC
guidelines. One difficulty that may arise with use of the
KDIGO guidelines is that there are currently no risk
calculators for individuals under the age of 40 years old
with or without CKD. There is also a lack of treatment
studies in younger individuals. This leaves us with clinical
judgment for risk in individuals under the age of 40 years
old (e.g., nephrotic-range proteinuria, multiple risk factors,
and length of time with CKD) as well as potential benefits
(e.g., life expectancy and transplant candidacy).
Overall, the guidelines would recommend that
most individuals with nondialysis CKD should receive
333
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 5. Kidney Disease: Improving Global Outcomes Lipid Guideline Recommendations for nondialysis,
nontransplant CKD
Assessment of lipid status
In adults and children with newly identified CKD, evaluation with a lipid profile recommended
In adults with CKD, follow-up measurement of lipid levels is not required for the majority of patients
In children with CKD, annual follow-up of fasting lipid levels suggested
Pharmacologic treatment
In adults ages $50 years old with CKD and eGFR$60 ml/min per 1.73 m2, treatment with a statin recommended
In adults ages $50 years old with eGFR,60 ml/min per 1.73 m2 not treated with chronic dialysis or kidney transplantation,
treatment with a statin or statin/ezetimibe combination recommended
In adults ages 18–49 years old with CKD not treated with chronic dialysis or kidney transplantation, treatment with a statin
suggested for those with one or more of the following
Known coronary disease (myocardial infarction or coronary revascularization)
Diabetes mellitus
Prior ischemic stroke
Estimated 10-year incidence of coronary death or nonfatal myocardial infarction .10%
In children with CKD, suggest not initiating statins or statin/ezetimibe combination
Triglyceride lowering
In adults and children with CKD and hypertriglyceridemia, therapeutic lifestyle changes should be advised
Modified with permission from KDIGO Lipid Work Group: KDIGO Clinical Practice Guideline for Lipid Management in Chronic Kidney Disease. Kidney Int Suppl 3: 259–305,
2013.
lipid treatment with moderate–intensity statin treatment
but that, in general, we no longer need to follow lipid
levels or target a particular LDL level.
Cardiac Biomarkers
Cardiac biomarkers are often elevated in CKD, even
without the presence of acute coronary syndrome (ACS)
or congestive heart failure (CHF), and this can affect their
clinical use. A recent systematic review evaluated the
clinical use of troponin in patients with CKD and
suspected ACS (18). It identified 23 relevant studies.
However, there was significant heterogeneity among
studies, and many of the studies were low quality; thus,
no definitive conclusion could be drawn regarding the
clinical use of troponin in ACS. A second systematic
review addressed cardiac troponin’s prognostic value in
patients with CKD in the absence of suspected ACS
(19). The review identified 98 studies of patients with
dialysis and nondialysis CKD that evaluated the use of
troponin for risk stratification. In both groups, elevated
troponin levels were associated with a 2- to 4-fold
increased risk of all-cause mortality and major adverse
cardiovascular events (CVE).
In asymptomatic patients with CKD, an elevated
troponin may indicate chronic structural heart disease
rather than ACS, and this observation may explain the
prognostic significance of the elevated echocardiographic findings in the Chronic Renal Insufficiency
Cohort (CRIC) Study (20). Among 2735 participants,
84% had detectable cardiac troponin T by a highsensitive assay. Dividing the participants into five groups
of undetectable troponin levels and quartiles of detectable
levels, the prevalence of left ventricular hypertrophy
(LVH), low ejection fraction (,45%), and diastolic
dysfunction increased across the groups, although the
relationship was strongest for LVH. With adjustment for
demographics, cause of kidney disease, BP, and other
cardiovascular risk factors, the relationship remained
significant for LVH. The odds ratios for LVH compared
with those with undetectable troponin were 1.26, 1.35,
1.91, and 2.43 for quartiles 1–4, respectively. When
evaluated as a screening test for the diagnosis of LVH,
only troponin T had an area under the curve (ROC) of
0.64, which is inadequate for a diagnostic test.
Although not a cardiac biomarker, an analogous
issue of altered clinical use of a biomarker in CKD is
the use of D-dimer to rule out pulmonary embolus (PE).
In evaluation for pulmonary embolism, the first step is to
estimate the pretest probability of a PE with a score, such
as the Wells score or the modified Geneva score.
Individuals with intermediate to high pretest probability
should undergo imaging. In individuals with low pretest
probability, a D-dimer can be drawn. If the level is ,500
mg/L, additional testing is not necessary. This can save
a patient from a test with iodinated radiographic contrast.
However, there are issues with a one size fits all cutoff
334
for D-dimer; D-dimer levels rise with age, and recent
studies have found that using an age-based cutoff led to
a larger proportion of patients in whom a PE could be
ruled out with a low risk of a false-negative result (,1%)
(21). Lindner et al. (22) subsequently proposed that
there should be renal function–adjusted D-dimer cutoffs.
Lindner et al. (22) evaluated the data on individuals
who presented to the emergency room and had data by
computed tomography angiogram (CTA), D-dimer level,
and kidney function. Overall, 12% had a D-dimer level
,500 mg/L; 13% of those with an eGFR.60 ml/min
per 1.73 m2 had a D-dimer ,500 mg/L, whereas only 6%
of individuals with an eGFR of 30–60 ml/min per
1.73 m2 and no individuals with an eGFR,30 ml/min
per 1.73 m2 had a D-dimer level ,500 mg/L. By ROC
curve analysis, Lindner et al. (22) found that the suggested
cutoff levels for D-dimer would be ,594 mg/L for an
eGFR of 30–60 ml/min per 1.73 m2 and ,1738 mg/L for
an eGFR ,30 ml/min per 1.73 m2. There are a number of
limitations that would need to be addressed before this
could be used in daily practice. The number of individuals
with kidney disease was relatively small, especially for
eGFRs,30 ml/min per 1.73 m2. In addition, in this
analysis, all participants had undergone CTA, which
would have excluded a number of individuals at low risk
of PE. In fact, the prevalence of D-dimer ,500 mg/L was
one half that in a prospective study evaluating age-based
cutoffs for dimer (28%), indicating that they had reviewed
a higher-risk group (21). However, it does highlight the
need for future studies to address this issue, which might
help avoid unnecessary contrast administration to patients
with CKD.
Inflammatory Markers and the Microbiome
CKD is associated with higher levels of inflammatory markers and markers of oxidative stress, which
may be one of the mediators that increases the risk of
CVD in CKD (23). In NHANES III, Stack et al. (24)
found that higher levels of fibrinogen, an acute-phase
reactant, were associated with an increased risk for
mortality. The relationship with mortality was similar
between those with and without kidney disease. The
hazard ratios (HRs) per 1-mmol/L increase were 1.06
(95% CI, 1.02 to 1.10), 1.05 (95% CI, 1.10 to 1.09), and
1.07 (95% CI, 1.03 to 1.11) for eGFRs,60, 60–89, and
$90 ml/min per 1.73 m2, respectively. However, individuals with CKD had higher levels of fibrinogen, and
fibrinogen may be an additional CKD risk factor.
Another acute-phase reactant that is associated with
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
cardiovascular risk in CKD is ceruloplasmin. Kennedy
et al. (25) evaluated ceruloplasmin levels with major
adverse CVEs in 654 individuals with CKD who were
undergoing elective cardiovascular evaluation by either
coronary angiography or CTA. A ceruloplasmin level
.25.5 mg/dl was associated with an increased risk of
CVE (adjusted HR, 1.69; 95% CI, 1.18 to 2.42).
As inflammatory levels rise, there is also an increase
in immunoregulatory cytokines that balance the response.
Elevated immunoregulatory cytokine levels may reflect
an ongoing inflammatory state. What may be important in
predicting disease severity is the balance between
proinflammatory and anti-inflammatory responses
(26,27). Yilmaz et al. (28) evaluated the association
of serum IL-10, a counter-regulatory cytokine, with
CVD in 403 patients with stages 1–5 CKD (27). With
worsening CKD stage, levels of C-reactive protein,
pentraxin-3, IL-6, and IL-10 increased, although the
IL-6 to IL-10 ratio decreased. Higher IL-10 levels
were associated with an increased risk of CVEs,
which persisted after adjustment for demographics,
other cardiovascular risk factors, and comorbidities.
IL-6 and pentraxin-3 were also associated with the
risk of CVE. Whether the findings reflect harmful
effects of IL-10, an issue of balance between proinflammatory and counterinflammatory responses, or
that IL-10 levels reflect the overall inflammatory
state is unknown and will require controlled treatment trials to sort out.
Although there are not currently approved interventions that decrease inflammation and cardiovascular
risk (except perhaps for statins), inflammation is a potentially remediable risk factor. An emerging approach
would be to target the microbiome. The human gut
microbiome is comprised of up to 100 trillion organisms
that live in symbiosis with us (29). The predominant
phyla are Bacteroidetes and Firmicutes, and others are
present in smaller proportion (30). The microbiome has
a number of physiologic beneficial effects (Table 6) (29).
CKD alters the microbiome. Vaziri et al. (31) analyzed
the microbial population in stool samples from 24
patients with ESRD and 12 controls. Microbial DNA
was extracted and analyzed by phylogenetic microarray,
classifying the bacteria into 190 operational taxonomic
units (OTUs). The pattern of OTUs differed between
patients with ESRD and controls. To determine whether
the change in the microbiome could be caused by ESRD
per se rather than differences in diet and medications,
Vaziri et al. (31) examined the microbiome in rats after
335
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 6. Physiologic effect of gut microbiota
(1) Integrity and function of gastrointestinal tract
Restoration of tight junction protein structure
Induction of epithelial heat–shock proteins
Upregulation of mucin genes
Competition with pathogenic bacteria for binding to
intestinal epithelial cells
Secretion of antimicrobial peptides
Suppression of intestinal inflammation
(2) Immunologic effects
Maturation of intestinal immune system
Reduction of allergic response to food and environmental
antigens
Promotion of immunomodulation and cell differentiation
(3) Metabolic effects
Breakdown of indigestible plant polysaccharides and
resistant starch
Facilitated absorption of complex carbohydrates
Synthesis of vitamins (K and B groups)
Synthesis of amino acids (threonine and lysine)
Biotransformation of conjugated bile acids
Degradation of dietary oxalates
Modified with permission from Sabatino A, Regolisti G, Brusasco I, Cabassi A,
Morabito S, Fiaccadori E: Alterations of intestinal barrier and microbiota in chronic
kidney disease. Nephrol Dial Transplant 30: 924–933, 2015.
5/6th nephrectomy and control rats. Changes in microbiome OTUs were also seen in the rats after nephrectomy, indicating that uremia may lead to alterations of
the microbiome.
In addition to uremia affecting the microbiome,
the microbiome could affect the health of the individual. There can be an imbalance in the microbiome
with an increase in pathogenic bacteria and a decrease
in commensal bacteria (30,32). This can lead to
disruption in the epithelial barrier and an increase in
inflammation. A number of the uremic toxins, such as
p-cresol, are produced by microbiomes that, because
of decreased renal clearance, accumulate in renal
failure (30,32). These changes could promote atherosclerosis. One gut-derived toxin is trimethylamine-Noxide (TMAO), which is derived from gut microbiota
metabolism of dietary phosphatidylcholine, choline,
or L-carnitine (33). Tang et al. (33) evaluated the association of TMAO levels in 3687 individuals undergoing
cardiac catheterization at the Cleveland Clinic, and 521 of
those individuals had CKD. Individuals with CKD had
higher TMAO levels (median of 7.9 versus 3.4 mM),
and TMAO levels correlated with eGFR (r¼20.48).
In CKD, the highest quartile was associated with increased risk of mortality at 5 years after controlling for
cardiovascular risk factors, C-reactive protein, and kidney
function by eGFR or cystatin C (HR, 1.45; 95% CI, 1.05
to 2.02).
The negative changes in the microbiome with
CKD might be amenable to dietary interventions. Patel
et al. (34) found that, in healthy subjects, individuals
who ate a vegetarian diet have a nearly 60% lower
generation of p-cresol and indoxyl sulfate than individuals who eat an unrestricted diet with meat. The
decreased production of these compounds was associated with a higher fiber intake and lower protein intake.
The microbiome may also be affected by interventions
with dietary supplements (Table 7 shows terms describing
the types of supplements) (35).
Guida et al. (36) performed a 30-day randomized
study of a synbiotic supplement (Probinul-neutro) versus
placebo in 30 patients with stages 3 or 4 CKD. Treatment with the synbiotic decreased p-cresol levels by
40%, whereas there was a nonstatistically significant
increase in p-cresol levels in the control group. The main
side effects were gastrointestinal, although all subjects
completed the study. In patients on hemodialysis, treatment with probiotics (fiber) has been shown to decrease
their levels of p-cresol and indoxyl sulfate (35,37). Future
studies should be done to determine whether these
interventions can decrease the risk of CVD.
Urine Biomarkers
Urinary biomarkers of acute tubular injury were
developed for AKI. Recent studies have found that they
predict mortality in the general population, even after
controlling for eGFR. In an analysis from the Health,
Aging and Body Composition (Health ABC) Study (a
cohort study of older individuals), Sarnak et al. (38)
evaluated the association of urine kidney injury molecule
1 (KIM-1), IL-18, and microalbuminuria with mortality
and CVD in 3010 individuals; 17% of the participants
had an eGFR,60 ml/min per 1.73 m2, and 19% had
a urine ACR .30 mg/g. Urinary KIM-1/creatinine was
modestly associated with urine ACR (r¼0.17) and
weakly associated with eGFR (r¼20.08). Urinary
IL-18/creatinine was weakly positively associated with
eGFR (r¼0.10) and modestly associated with urine ACR
(r¼0.17). An eGFR,60 ml/min per 1.73 m2 was significantly associated with higher KIM-1 and urine albumin
levels but not with IL-18 levels. The patterns of association with mortality differed among the markers (Figure
14). Urine KIM-1 levels were associated with mortality.
After adjustment for eGFR and albuminuria as well, the
336
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 7. Types of supplements affecting microbiome
Term
Definition
Examples
Prebiotic
Nondigestible food ingredients that stimulate growth
and/or activity of bacteria
Living organisms that, on ingestion, can improve
the health of the organism
Combines prebiotics with probiotics
Inulin (nondigestible fiber) present in foods, such
as Jerusalem artichokes and chicory
Bifidus regularis in some yogurts
Probiotic
Synbiotic
Some yogurts and supplements
Modified with permission from National Kidney Foundation: 2012 Update. Am J Kidney Dis 60: 850–886, 2012.
relationship was attenuated, but the fourth quartile remained significantly associated with mortality (HR,
1.33; 95% CI, 1.12 to 1.57). KIM-1 was not associated
with CVD, and IL-18 was not associated with either
outcome. In contrast, urine albuminuria remained significantly associated with both mortality and CVD. In
a separate Health ABC analysis, Driver et al. (39) found
that urinary KIM-1 was associated with risk for heart
failure, but the association with albuminuria was also
stronger; IL-18 was not associated with risk of heart
failure.
Carlsson et al. (40) evaluated the association of
urinary KIM-1 with total and cardiovascular mortality
in 529 men participating in the Uppsala Longitudinal
Study of Adult Men. KIM-1/creatinine was associated
with a higher risk of cardiovascular and all-cause mortality. Comparing individuals with normal values of all three
markers (eGFR, albuminuria, and KIM-1), individuals
with one abnormal value had a 2-fold greater risk of
cardiovascular mortality. The greatest risk was if all three
were abnormal (low eGFR, high urine ACR, and high
KIM-1), where the risk of all-cause mortality was 3-fold
higher and the risk of cardiovascular mortality .6-fold
higher, even after adjustment for other cardiovascular risk
factors.
Another urinary biomarker is urinary liver–type
fatty acid–binding protein (L-FABP), which is an intracellular carrier protein of free fatty acids. It is produced by the liver and kidney (proximal tubule);
circulating L-FABP is filtered at the glomerulus and
taken up by the proximal tubules (41,42). Araki et al.
(43) studied the association of L-FABP with the subsequent development of a renal and CVD composite
(hemodialysis, MI, angina, stroke, cerebral hemorrhage, and peripheral vascular disease) in individuals
with type 2 diabetes. Individuals at baseline had a SCr
#1.0 mg/dl and either normoalbuminuria (n¼422) or
microalbuminuria (n¼196). After a median of 12 years,
higher tertiles of L-FABP were associated with a higher
risk of the composite end point (HR, 1.64 and 1.93
for the second and third tertiles, respectively). The
highest two tertiles had a 75% higher risk of developing
CVD.
Figure 14. Mortality and cardiovascular disease (CVD) by
urinary injury markers. (A) Mortality rates by quartiles of
kidney injury marker 1 (KIM-1), IL-18, and urinary albumin
to creatinine ratio (UACR). (B) CVD by quartiles of KIM-1,
IL-18, and UACR. CVD was defined by a coronary heart
disease event and/or stroke. Q, quartile. Reprinted with
permission from Sarnak MJ, Katz R, Newman A, Harris T,
Peralta CA, Devarajan P, Bennett MR, Fried L, Ix JH,
Satterfield S, Simonsick EM, Parikh CR, Shlipak MG; Health
ABC Study: Association of urinary injury biomarkers with
mortality and cardiovascular events. J Am Soc Nephrol 25:
1545–1553, 2014.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Echocardiographic Changes in CKD
Echocardiographic changes, such as LVH, diastolic
dysfunction, and increased left atrial volume, are common
in CKD and associated with an increased risk of CVEs
and development of HF (44–46). Peterson et al. (46)
evaluated the relationship of baseline echocardiographic
findings with outcomes in the black Study of Kidney
Disease and Hypertension Trial. The average left ventricular mass index (LVMI) was 70.6 g/m2 in patients with
CVE versus 59.2 g/m2 in patients without CVE; for HF
events, LVMI was 75.6 g/m2 in patients with HF events
(hospitalization for CHF or death caused by CHF) versus
59.7 g/m2 in patients without events. The presence of
LVH was associated with higher average systolic BP,
greater prevalence of albuminuria, and greater prevalence
of baseline CVD. After adjustment for other covariates,
LVMI remained a significant predictor of CVEs and HF
events. In addition, measures of diastolic dysfunction were
predictors of HF. An E/A ratio #0.75 was associated with
a 2-fold higher risk of HF compared with E/A ratios
between 0.75 and 1.5 (adjusted HR, 2.05; 95% CI, 1.03 to
4.06).
What has been less well studied in CKD are the
longitudinal changes in echocardiographic findings
over time. Cai et al. (47) performed a longitudinal
study in 300 individuals with stages 3–5 CKD. Of 300
individuals, 278 had baseline and 1-year echocardiograms. Over the 1-year period, LVMI increased (105632
to 112634 g/m2), left ventricular volume index increased
(42.8611.1 to 43.9611.8 ml/m2), and multiple parameters of diastolic dysfunction worsened. There was also
a small but statistically significant decrease in left
ventricular ejection fraction (LVEF; from 70% to
69%). The echocardiographic changes were greater in
CKD stages 4 or 5 than in stages 3A or 3B.
Bansal et al. (48) evaluated change in echocardiographic findings before and after the transition to ESRD
in the CRIC Study. For this analysis, Bansal et al. (48)
analyzed 190 individuals who had an echocardiogram at
advanced CKD and after ESRD was established, excluding those who received a preemptive transplant.
At the first echocardiogram, the mean eGFR was 16.9
ml/min per 1.73 m2, and median proteinuria was 2.75 g
per day. After transitioning to ESRD, there was a decrease in BP and body mass index, an increase in
hemoglobin, and an increase in the prevalence of
reported CVD. The LVMI did not change, but LVEF
decreased. The lack of change in LVMI contrasts with
337
the work by Cai et al. (46). The reasons for the difference
could reflect the wider eGFR range in the study by Cai
et al. (47) or perhaps, better volume and BP control after
transition to ESRD. Additional studies would need to
determine whether the decline in ejection fraction seen in
both studies could be prevented with interventions, such
as anti–renin-angiotensin-aldosterone system treatment,
and whether this could decrease the risk of mortality.
Percutaneous Cardiovascular Intervention and
Drug-Eluting Stents
Over the past 25 years, there have been major
changes in the care of patients with acute MI. Nauta
et al. (49) evaluated time trends in care received and
outcomes in 12,087 adult patients who were admitted
with an ST elevation MI or non-ST elevation MI to the
intensive care unit of the Erasmus University Medical
Center between June of 1985 and December of 2008.
Patients were divided into three groups according to the
year of hospitalization: 1985–1990, 1990–2000, and
2000–2008. Of the patients in the study, 29% had no
kidney disease, 46% had stage 2 CKD, 21% had stage
3 CKD, and 4% had stage 4 or 5 CKD. The use
of reperfusion therapy (thrombolytics or percutaneous
coronary intervention [PCI]) increased over time during
all CKD stages but remained lower in those with
advanced CKD compared with those with normal
kidney function (Figure 15). Not surprisingly, mortality
was higher in individuals with CKD, with the risk for
30-day mortality 4 times higher in those with stage 3
CKD and 8.5 times higher in those with stage 4 or 5 CKD
compared with the normal population. Over 20 years,
there was improved 30-day mortality after MI in all CKD
stages, although there was no significant change in 5-year
mortality. The increased use of indicated medications for
CKD is encouraging as is the decline in 30-day mortality;
however, the results still reveal that such medications are
underused and that additional interventions are vital for
better longer–term outcomes.
The use of a drug-eluting stent (DES) rather than
a bare metal stent (BMS) might decrease adverse
outcomes after PCI. Wang et al. (50) performed
a meta-analysis of DESs versus BMSs in individuals
with eGFRs,60 ml/min per 1.73 m2. These studies
were primarily observational, because there are few
randomized studies of PCI that include individuals with
CKD, and some were post hoc subgroup analyses of
trials. Altogether, Wang et al. (50) compiled 26 studies
with 66,840 individuals. DESs were associated with
338
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Figure 15. Treatment of patients with cardiovascular treatments after myocardial infarction over time (A) Use of PCI; (B) Use of
aspirin; (C) Use of statin. ACE, angiotensin-converting enzyme; ARB, angiotensin II receptor blocker; ASA, aspirin; ICCU, intensive
coronary care unit; PCI, percutaneous coronary intervention; ThT, thrombolytic therapy. Reprinted with permission from Nauta ST,
van Domburg RT, Nuis RJ, Akkerhuis M, Deckers JW: Decline in 20-year mortality after myocardial infarction in patients with chronic
kidney disease: Evolution from the prethrombolysis to the percutaneous coronary intervention era. Kidney Int 84: 353–358, 2013.
a lower mortality (odds ratio, 0.77; 95% CI, 0.65 to
0.90). It is notable that this result was not seen in the
subgroup analysis of randomized trials. There was
a significantly decreased risk of revascularization (odds
ratio, 0.61; 95% CI, 0.50 to 0.74) that was seen in
randomized and observational studies.
Since the meta-analysis was published, there has
been a randomized study that focused on CKD. The
Randomized Comparison of Xience V and Multilink
Vision Coronary Stents in the Same Multivessel Patient
with Chronic Kidney Disease Study compared an everolimus-eluting stent with a BMS in individuals with
a creatinine clearance between 30 and 59 ml/min with
two significant lesions in two major epicardial vessels
(51). The stents were randomized by coronary vessel (so
each patient received both). The primary outcome was
ischemia–driven target vessel revascularization as detected
by myocardial scintigraphy at 12 months (a positive stress
test indicating ischemia in area supplied by vessel); 215
individuals were randomized. The incidence of ischemia–
driven target level revascularization was lower for the
DES group (11.4% versus 2.4%; P,0.001). The benefit
seemed greater with more advanced CKD (moderate
CKD, 7.3% versus 2.6% and stages 4 and 5 CKD, 24.2%
versus 3.1% for BMS and DES, respectively). However,
because the study randomized vessels rather than patients, mortality cannot be assessed.
The use of a DES improved revascularization of blood vessels compared with a BMS
in patients with stage 3 CKD.
Atrial Fibrillation
CKD is a risk factor for the development of atrial
fibrillation (AF) (52,53). Warfarin use in CKD is
associated with a decreased risk of thromboembolic
events but also, an increased risk of bleeding. What is
not clear is the balance between risk and benefit.
Bonde et al. (54) used nationwide registries to identify
Danish individuals who had nonvalvular AF, and this
information was linked to laboratory data and medication data. Of 154,259 individuals with nonvalvular AF,
11,128 had nondialysis CKD, and 1728 had ESRD.
In those with CKD and a CHA2DS2-VASc score $2
(high risk), which includes a score for prior stroke or
thromboembolism, individuals who were not treated
with warfarin had a higher risk of stroke than individuals who were treated with warfarin. Net clinical benefit
was evaluated using a composite that combined efficacy
and safety (stroke and fatal bleeding) plus cardiovascular death, all-cause mortality, and fatal bleeding. In
high–risk nondialysis CKD, treatment with warfarin
was associated with decreased risk in all three outcomes: fatal stroke/fatal bleeding (HR, 0.71; 95% CI,
0.57 to 0.88), cardiovascular death (HR, 0.80; 95% CI,
0.74 to 0.88), and all-cause mortality (HR, 0.64; 95%
CI, 0.60 to 0.69).
Using a nationwide Swedish registry of subjects
admitted to a coronary care unit with symptoms
suggestive of ACS, Carrero et al. (55) analyzed the
association of warfarin treatment with cardiovascular
outcomes and bleeding in patients with AF. There were
24,317 individuals included in the analysis: 41.7% had
stage 3 CKD, 8.1% had stage 4 CKD, and 2.0% had
339
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
stage 5 CKD. Most individuals had a CHAD2 score
$2. Warfarin was prescribed in 22.0% of individuals
with no CKD, 22.4% of individuals with stage 3
CKD, 18.9% of individuals with stage 4 CKD, and
13.8% of individuals with stage 5 CKD. The risk of
the composite end point of mortality, stroke, and MI
increased across all CKD stages. Within each CKD
stratum, those treated with warfarin had an approximately 25% lower risk. Overall, the risk of bleeding
was greater in those with CKD. However, within CKD
strata, warfarin use was not associated with increased
bleeding risk, indicating that individuals with CKD
have a higher bleeding risk overall and not just with
anticoagulants.
These two studies were not randomized, and
therefore, the characteristics of patients who were
treated are likely to differ significantly from those of
patients who were not treated, which could lead to
bias. Overall, however, it suggests that treatment with
anticoagulation is warranted in high-risk patients with
CKD.
Cardioverter Defibrillator
Individuals with CKD have an increased risk of
sudden cardiac death (SCD). Implanted cardioverter
defibrillators (ICDs) decrease SCD in the non-CKD
population, raising the question of whether they would
be beneficial in patients with CKD.
Hess et al. (56) studied the association of CKD
with mortality after placement of an ICD for primary
prevention using the National Cardiovascular Data
Registry’s ICD Registry. ICDs were placed in individuals with a history of MI and LVEF#30% or a history
of HF with an LVEF#35%. Among 47,282 patients,
21,226 had CKD. CKD was associated with a higher
mortality risk (13.5%, 26.2%, 44.7%, and 49.0% at
3 years for no CKD, stages 3 or 4/5 CKD not on dialysis,
and ESRD, respectively). The very high mortality,
especially for individuals with an eGFR,30 ml/min
per 1.73 m2, raises the question of whether one can
change the natural history of the disease in those with
advanced CKD.
Pun et al. (57) performed a patient–level metaanalysis of randomized studies of ICD placement for
primary prevention of SCD. There were seven studies
with 2867 patients; 1040 had an eGFR,60 ml/min
per 1.73 m2. Because these were randomized studies,
the majority of patients with CKD had stage 3 CKD
(only 3.6% had an eGFR,30 ml/min per 1.73 m2).
Individuals with CKD were older and had more
comorbidities. There was a significant interaction
between eGFR and benefit of ICD. The survival benefit
with ICDs steadily declined with lower eGFR, although
it was not possible to determine where the threshold
should be (where benefit disappears), because there
were few individuals with advanced CKD. This metaanalysis, however, supports the conduct of a randomized intervention study in advanced CKD, because
there would now be equipoise (i.e., ethical to randomize to no ICD for primary prevention in advanced
CKD).
References
1. Wada M, Ueda Y, Higo T, Matsuo K, Nishio M, Hirata A, Asai M,
Nemoto T, Kashiyama T, Murakami A, Kashiwase K, Kodama K:
Chronic kidney disease and coronary artery vulnerable plaques. Clin J
Am Soc Nephrol 6: 2792–2798, 2011 PubMed
2. Schaar JA, Muller JE, Falk E, Virmani R, Fuster V, Serruys PW, Colombo
A, Stefanadis C, Ward Casscells S, Moreno PR, Maseri A, van der Steen
AF: Terminology for high-risk and vulnerable coronary artery plaques.
Report of a meeting on the vulnerable plaque, June 17 and 18, 2003,
Santorini, Greece. Eur Heart J 25: 1077–1082, 2004 PubMed
3. Jang IK, Bouma BE, Kang DH, Park SJ, Park SW, Seung KB, Choi
KB, Shishkov M, Schlendorf K, Pomerantsev E, Houser SL, Aretz
HT, Tearney GJ: Visualization of coronary atherosclerotic plaques
in patients using optical coherence tomography: Comparison
with intravascular ultrasound. J Am Coll Cardiol 39: 604–609,
2002 PubMed
4. Jegere S, Narbute I, Erglis A: Use of intravascular imaging in managing
coronary artery disease. World J Cardiol 6: 393–404, 2014 PubMed
5. Bezerra HG, Costa MA, Guagliumi G, Rollins AM, Simon DI: Intracoronary optical coherence tomography: A comprehensive review
clinical and research applications. JACC Cardiovasc Interv 2: 1035–
1046, 2009 PubMed
6. Kato K, Yonetsu T, Jia H, Abtahian F, Vergallo R, Hu S, Tian J,
Kim SJ, Lee H, McNulty I, Lee S, Uemura S, Jang Y, Park SJ,
Mizuno K, Yu B, Jang IK: Nonculprit coronary plaque characteristics of chronic kidney disease. Circ Cardiovasc Imaging 6: 448–
456, 2013 PubMed
7. Falk E, Shah PK, Fuster V: Coronary plaque disruption. Circulation 92:
657–671, 1995 PubMed
8. Nakano T, Ninomiya T, Sumiyoshi S, Onimaru M, Fujii H, Itabe
H, Nakashima Y, Sueishi K, Tsuruya K, Oda Y, Kitazono T,
Kiyohara Y: Chronic kidney disease is associated with neovascularization and intraplaque hemorrhage in coronary atherosclerosis
in elders: Results from the Hisayama Study. Kidney Int 84: 373–
380, 2013 PubMed
9. Wang Y, Katzmarzyk PT, Horswell R, Zhao W, Johnson J, Hu G:
Kidney function and the risk of cardiovascular disease in patients with
type 2 diabetes. Kidney Int 85: 1192–1199, 2014 PubMed
10. Groop PH, Thomas MC, Moran JL, Wadèn J, Thorn LM, Mäkinen VP,
Rosengård-Bärlund M, Saraheimo M, Hietala K, Heikkilä O, Forsblom
C; FinnDiane Study Group: The presence and severity of chronic kidney
disease predicts all-cause mortality in type 1 diabetes. Diabetes 58:
1651–1658, 2009 PubMed
11. Orchard TJ, Secrest AM, Miller RG, Costacou T: In the absence of
renal disease, 20 year mortality risk in type 1 diabetes is comparable to
that of the general population: A report from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetologia 53: 2312–2319,
2010 PubMed
340
12. Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR,
Himmelfarb J, de Boer IH: Kidney disease and increased mortality risk
in type 2 diabetes. J Am Soc Nephrol 24: 302–308, 2013 PubMed
13. Choi JS, Kim MJ, Kang YU, Kim CS, Bae EH, Ma SK, Ahn YK, Jeong
MH, Kim YJ, Cho MC, Kim CJ, Kim SW; Korea Acute Myocardial
Infarction Registry Investigators: Association of age and CKD with
prognosis of myocardial infarction. Clin J Am Soc Nephrol 8: 939–944,
2013 PubMed
14. Tonelli M, Muntner P, Lloyd A, Manns B, Klarenbach S, Pannu N,
James M, Hemmelgarn B; Alberta Kidney Disease Network: Impact of
age on the association between CKD and the risk of future coronary
events. Am J Kidney Dis 64: 375–382, 2014 PubMed
15. Kidney Disease: Improving Global Outcomes (KDIGO) Lipid Work
Group: KDIGO Clinical Practice Guideline for Lipid Management in
Chronic Kidney Disease. Kidney Int Suppl 3: 259–305, 2013
16. Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB,
Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM,
McBride P, Schwartz JS, Shero ST, Smith SC Jr., Watson K, Wilson
PW, Eddleman KM, Jarrett NM, LaBresh K, Nevo L, Wnek J, Anderson
JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Curtis LH, DeMets
D, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen
WK, Smith SC Jr., Tomaselli GF; American College of Cardiology/
American Heart Association Task Force on Practice Guidelines: 2013
ACC/AHA guideline on the treatment of blood cholesterol to reduce
atherosclerotic cardiovascular risk in adults: A report of the American
College of Cardiology/American Heart Association Task Force on
Practice Guidelines. Circulation 129[Suppl 2]: S1–S45, 2014 PubMed
17. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson
C, Wanner C, Krane V, Cass A, Craig J, Neal B, Jiang L, Hooi LS,
Levin A, Agodoa L, Gaziano M, Kasiske B, Walker R, Massy ZA,
Feldt-Rasmussen B, Krairittichai U, Ophascharoensuk V, Fellström
B, Holdaas H, Tesar V, Wiecek A, Grobbee D, de Zeeuw D,
Grönhagen-Riska C, Dasgupta T, Lewis D, Herrington W, Mafham M,
Majoni W, Wallendszus K, Grimm R, Pedersen T, Tobert J, Armitage J,
Baxter A, Bray C, Chen Y, Chen Z, Hill M, Knott C, Parish S, Simpson
D, Sleight P, Young A, Collins R; SHARP Investigators: The effects
of lowering LDL cholesterol with simvastatin plus ezetimibe in
patients with chronic kidney disease (Study of Heart and Renal
Protection): A randomised placebo-controlled trial. Lancet 377:
2181–2192, 2011 PubMed
18. Stacy SR, Suarez-Cuervo C, Berger Z, Wilson LM, Yeh HC, Bass EB,
Michos ED: Role of troponin in patients with chronic kidney disease
and suspected acute coronary syndrome: A systematic review. Ann
Intern Med 161: 502–512, 2014 PubMed
19. Michos ED, Wilson LM, Yeh HC, Berger Z, Suarez-Cuervo C, Stacy SR,
Bass EB: Prognostic value of cardiac troponin in patients with chronic
kidney disease without suspected acute coronary syndrome: A systematic
review and meta-analysis. Ann Intern Med 161: 491–501, 2014 PubMed
20. Mishra RK, Li Y, DeFilippi C, Fischer MJ, Yang W, Keane M, Chen J,
He J, Kallem R, Horwitz EJ, Rafey M, Raj DS, Go AS, Shlipak MG;
CRIC Study Investigators: Association of cardiac troponin T with left
ventricular structure and function in CKD. Am J Kidney Dis 61: 701–
709, 2013 PubMed
21. Righini M, Van Es J, Den Exter PL, Roy PM, Verschuren F, Ghuysen
A, Rutschmann OT, Sanchez O, Jaffrelot M, Trinh-Duc A, Le Gall C,
Moustafa F, Principe A, Van Houten AA, Ten Wolde M, Douma RA,
Hazelaar G, Erkens PM, Van Kralingen KW, Grootenboers MJ, Durian
MF, Cheung YW, Meyer G, Bounameaux H, Huisman MV, Kamphuisen
PW, Le Gal G: Age-adjusted D-dimer cutoff levels to rule out pulmonary embolism: The ADJUST-PE study. JAMA 311: 1117–1124,
2014 PubMed
22. Lindner G, Funk G-C, Pfortmueller CA, Leichtle AB, Fiedler GM,
Schwarz C, Exadaktylos AK, Puig S: D-dimer to rule out pulmonary
embolism in renal insufficiency. Am J Med 127: 343–347, 2014 PubMed
23. Stenvinkel P, Ketteler M, Johnson RJ, Lindholm B, Pecoits-Filho R,
Riella M, Heimbürger O, Cederholm T, Girndt M: IL-10, IL-6, and
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
TNF-alpha: Central factors in the altered cytokine network of
uremia–the good, the bad, and the ugly. Kidney Int 67: 1216–1233,
2005 PubMed
Stack AG, Donigiewicz U, Abdalla AA, Weiland A, Casserly LF,
Cronin CJ, Nguyen HT, Hannigan A: Plasma fibrinogen associates
independently with total and cardiovascular mortality among subjects
with normal and reduced kidney function in the general population.
QJM 107: 701–713, 2014 PubMed
Kennedy DJ, Fan Y, Wu Y, Pepoy M, Hazen SL, Tang WH: Plasma
ceruloplasmin, a regulator of nitric oxide activity, and incident
cardiovascular risk in patients with CKD. Clin J Am Soc Nephrol 9:
462–467, 2014
Bone RC: Immunologic dissonance: A continuing evolution in our
understanding of the systemic inflammatory response syndrome (SIRS)
and the multiple organ dysfunction syndrome (MODS). Ann Intern Med
125: 680–687, 1996 PubMed
Gigante A, Gasperini ML, Afeltra A, Barbano B, Margiotta D, Cianci R,
De Francesco I, Amoroso A: Cytokines expression in SLE nephritis.
Eur Rev Med Pharmacol Sci 15: 15–24, 2011 PubMed
Yilmaz MI, Solak Y, Saglam M, Cayci T, Acikel C, Unal HU, Eyileten
T, Oguz Y, Sari S, Carrero JJ, Stenvinkel P, Covic A, Kanbay M: The
relationship between IL-10 levels and cardiovascular events in patients
with CKD. Clin J Am Soc Nephrol 9: 1207–1216, 2014 PubMed
Sabatino A, Regolisti G, Brusasco I, Cabassi A, Morabito S, Fiaccadori
E: Alterations of intestinal barrier and microbiota in chronic kidney
disease. Nephrol Dial Transplant 30: 924–933, 2015 PubMed
Ramezani A, Raj DS: The gut microbiome, kidney disease, and targeted
interventions. J Am Soc Nephrol 25: 657–670, 2014 PubMed
Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, DeSantis TZ, Ni Z,
Nguyen TH, Andersen GL: Chronic kidney disease alters intestinal
microbial flora. Kidney Int 83: 308–315, 2013 PubMed
Mafra D, Lobo JC, Barros AF, Koppe L, Vaziri ND, Fouque D: Role of
altered intestinal microbiota in systemic inflammation and cardiovascular
disease in chronic kidney disease. Future Microbiol 9: 399–410,
2014 PubMed
Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li
XS, Levison BS, Hazen SL: Gut microbiota-dependent trimethylamine
N-oxide (TMAO) pathway contributes to both development of renal
insufficiency and mortality risk in chronic kidney disease. Circ Res 116:
448–455, 2015 PubMed
Patel KP, Luo FJ, Plummer NS, Hostetter TH, Meyer TW: The
production of p-cresol sulfate and indoxyl sulfate in vegetarians versus
omnivores. Clin J Am Soc Nephrol 7: 982–988, 2012 PubMed
Vitetta L, Gobe G: Uremia and chronic kidney disease: The role of the
gut microflora and therapies with pro- and prebiotics. Mol Nutr Food
Res 57: 824–832, 2013 PubMed
Guida B, Germanò R, Trio R, Russo D, Memoli B, Grumetto L, Barbato
F, Cataldi M: Effect of short-term synbiotic treatment on plasma
p-cresol levels in patients with chronic renal failure: A randomized
clinical trial. Nutr Metab Cardiovasc Dis 24: 1043–1049, 2014 PubMed
Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW: Effect of
increasing dietary fiber on plasma levels of colon-derived solutes in
hemodialysis patients. Clin J Am Soc Nephrol 9: 1603–1610, 2014 PubMed
Sarnak MJ, Katz R, Newman A, Harris T, Peralta CA, Devarajan P,
Bennett MR, Fried L, Ix JH, Satterfield S, Simonsick EM, Parikh CR,
Shlipak MG; Health ABC Study: Association of urinary injury
biomarkers with mortality and cardiovascular events. J Am Soc Nephrol
25: 1545–1553, 2014 PubMed
Driver TH, Katz R, Ix JH, Magnani JW, Peralta CA, Parikh CR, Fried L,
Newman AB, Kritchevsky SB, Sarnak MJ, Shlipak MG; Health ABC
Study: Urinary kidney injury molecule 1 (KIM-1) and interleukin 18
(IL-18) as risk markers for heart failure in older adults: The Health,
Aging, and Body Composition (Health ABC) Study. Am J Kidney Dis
64: 49–56, 2014 PubMed
Carlsson AC, Larsson A, Helmersson-Karlqvist J, Lind L, Ingelsson E,
Larsson TE, Bottai M, Sundström J, Ärnlöv J: Urinary kidney injury
341
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
molecule-1 and the risk of cardiovascular mortality in elderly men. Clin
J Am Soc Nephrol 9: 1393–1401, 2014 PubMed
Oyama Y, Takeda T, Hama H, Tanuma A, Iino N, Sato K, Kaseda R,
Ma M, Yamamoto T, Fujii H, Kazama JJ, Odani S, Terada Y, Mizuta K,
Gejyo F, Saito A: Evidence for megalin-mediated proximal tubular
uptake of L-FABP, a carrier of potentially nephrotoxic molecules. Lab
Invest 85: 522–531, 2005
Kamijo-Ikemori A, Sugaya T, Kimura K: Urinary fatty acid binding
protein in renal disease. Clin Chim Acta 374: 1–7, 2006 PubMed
Araki S, Haneda M, Koya D, Sugaya T, Isshiki K, Kume S,
Kashiwagi A, Uzu T, Maegawa H: Predictive effects of urinary
liver-type fatty acid-binding protein for deteriorating renal function
and incidence of cardiovascular disease in type 2 diabetic patients
without advanced nephropathy. Diabetes Care 36: 1248–1253,
2013 PubMed
Hee L, Nguyen T, Whatmough M, Descallar J, Chen J, Kapila S, French
JK, Thomas L: Left atrial volume and adverse cardiovascular outcomes
in unselected patients with and without CKD. Clin J Am Soc Nephrol 9:
1369–1376, 2014 PubMed
Park M, Hsu CY, Li Y, Mishra RK, Keane M, Rosas SE, Dries D, Xie
D, Chen J, He J, Anderson A, Go AS, Shlipak MG; Chronic Renal
Insufficiency Cohort (CRIC) Study Group: Associations between
kidney function and subclinical cardiac abnormalities in CKD. J Am
Soc Nephrol 23: 1725–1734, 2012 PubMed
Peterson GE, de Backer T, Contreras G, Wang X, Kendrick C, Greene T,
Appel LJ, Randall OS, Lea J, Smogorzewski M, Vagaonescu T, Phillips
RA; African American Study of Kidney Disease Investigators: Relationship of left ventricular hypertrophy and diastolic function with cardiovascular and renal outcomes in African Americans with hypertensive
chronic kidney disease. Hypertension 62: 518–525, 2013 PubMed
Cai QZ, Lu XZ, Lu Y, Wang AY: Longitudinal changes of cardiac
structure and function in CKD (CASCADE study). J Am Soc Nephrol
25: 1599–1608, 2014 PubMed
Bansal N, Keane M, Delafontaine P, Dries D, Foster E, Gadegbeku
CA, Go AS, Hamm LL, Kusek JW, Ojo AO, Rahman M, Tao K,
Wright JT, Xie D, Hsu CY; CRIC Study Investigators: A longitudinal study of left ventricular function and structure from CKD
to ESRD: The CRIC study. Clin J Am Soc Nephrol 8: 355–362,
2013 PubMed
Nauta ST, van Domburg RT, Nuis RJ, Akkerhuis M, Deckers JW:
Decline in 20-year mortality after myocardial infarction in patients
with chronic kidney disease: Evolution from the prethrombolysis to
the percutaneous coronary intervention era. Kidney Int 84: 353–358,
2013 PubMed
Wang ZJ, Harjai KJ, Shenoy C, Gao F, Shi DM, Liu YY, Zhao YX, Zhou YJ:
Drug-eluting stents versus bare-metal stents in patients with decreased GFR:
A meta-analysis. Am J Kidney Dis 62: 711–721, 2013 PubMed
Tomai F, Ribichini F, De Luca L, Petrolini A, Ghini AS, Weltert L,
Spaccarotella C, Proietti I, Trani C, Nudi F, Pighi M, Vassanelli C:
Randomized Comparison of Xience V and Multi-Link Vision Coronary
Stents in the Same Multivessel Patient With Chronic Kidney Disease
(RENAL-DES) Study. Circulation 129: 1104–1112, 2014 PubMed
Myrvang H: Cardiovascular disease: CKD increases the risk of atrial
fibrillation. Nat Rev Nephrol 7: 426, 2011 PubMed
Xu D, Murakoshi N, Sairenchi T, Irie F, Igarashi M, Nogami A,
Tomizawa T, Yamaguchi I, Yamagishi K, Iso H, Ota H, Aonuma K:
Anemia and reduced kidney function as risk factors for new onset of
atrial fibrillation (from the Ibaraki prefectural health study). Am J
Cardiol 115: 328–333, 2015 PubMed
Bonde AN, Lip GY, Kamper AL, Hansen PR, Lamberts M, Hommel K,
Hansen ML, Gislason GH, Torp-Pedersen C, Olesen JB: Net clinical
benefit of antithrombotic therapy in patients with atrial fibrillation and
chronic kidney disease: A nationwide observational cohort study. J Am
Coll Cardiol 64: 2471–2482, 2014 PubMed
Carrero JJ, Evans M, Szummer K, Spaak J, Lindhagen L, Edfors R,
Stenvinkel P, Jacobson SH, Jernberg T: Warfarin, kidney dysfunction,
and outcomes following acute myocardial infarction in patients with
atrial fibrillation. JAMA 311: 919–928, 2014 PubMed
56. Hess PL, Hellkamp AS, Peterson ED, Sanders GD, Al-Khalidi HR,
Curtis LH, Hammill BG, Pun PH, Curtis JP, Anstrom KJ, Hammill
SC, Al-Khatib SM: Survival after primary prevention implantable cardioverter-defibrillator placement among patients with
chronic kidney disease. Circ Arrhythm Electrophysiol 7: 793–799,
2014 PubMed
57. Pun PH, Al-Khatib SM, Han JY, Edwards R, Bardy GH, Bigger JT,
Buxton AE, Moss AJ, Lee KL, Steinman R, Dorian P, Hallstrom A,
Cappato R, Kadish AH, Kudenchuk PJ, Mark DB, Hess PL, Inoue LY,
Sanders GD: Implantable cardioverter-defibrillators for primary prevention of sudden cardiac death in CKD: A meta-analysis of patient-level data
from 3 randomized trials. Am J Kidney Dis 64: 32–39, 2014 PubMed
Exercise in CKD
Individuals with CKD have decreased physical
activity, which may contribute to the high risk of
cardiovascular disease as well as frailty.
Chen et al. (1), in a cohort study of 6363 patients
with stages 3–5 CKD at the China Medical University
Hospital, found that 65.7%, 73.1%, and 77.5% of
individuals with stages 3, 4, and 5 CKD, respectively,
never exercised. In individuals who did exercise, the most
common form of exercise was walking. Overall, 21% of
the cohort walked for exercise. Walking was associated
with a decreased risk of mortality (hazard ratio, 0.67; 95%
confidence interval, 0.53 to 0.84) and progression to
ESRD (hazard ratio, 0.79; 95% confidence interval, 0.73
to 0.85), even after adjusting for eGFR, age, body mass
index, and comorbidity. Robinson-Cohen et al. (2) also
found that higher physical activity was associated with
a slower change in kidney function. Robinson-Cohen
et al. (2) evaluated the association of physical activity as
assessed by the Four-Week Physical Activity History
Questionnaire with a change in kidney function in 256
individuals with stage 3 or 4 CKD who were participating
in the Seattle Kidney Study. The overall change in eGFR
was 27.6% per year. The percentage change in kidney
function was slower in those with greater physical activity:
29.65%, 28.25%, 26.85%, and 26.2% for 0, 1–59,
60–150, and $150 minutes leisure time physical activity minutes per week, respectively. In adjusted analysis,
each 60-minute higher level of weekly physical activity
was associated with a 0.5% slower decline in eGFR
annually (2). However, observational studies of exercise
are potentially confounded, because sicker people are
less likely to exercise. In addition, it may not be exercise
per se that leads to a slower decline, but rather other
factors that associate with exercise (e.g., individuals who
exercise may be less likely to smoke).
342
Heiwe and Jacobson (3) performed a meta-analysis
of randomized studies of exercise training in CKD.
Heiwe and Jacobson (3) found 41 studies with 928
participants. Most of the studies were in individuals on
dialysis or after transplantation. Confining the outcomes
to those with nondialysis, nontransplant CKD, exercise
increased aerobic capacity, lowered systolic BP, and
increased thigh muscle mass and muscle strength.
Although the importance of exercise is generally
appreciated, how to increase patient participation in
exercise is not clear and has not been adequately studied.
Watson et al. (4) performed a randomized study of an
8-week progressive resistance exercise program versus
usual care in individuals ages $40 years old with stage 3b
or 4 CKD who were receiving care at the Leicester
General Hospital Nephrology Outpatient Clinic. Exclusion criteria included myocardial infarction within the past
6 months, an unstable chronic condition, physical impairment preventing ability to undertake intervention, hemoglobin A1C $9%, insufficient command of English, or
inability to provide informed consent. Similar to the
findings of the meta-analysis by Heiwe and Jacobson
(3), the resistance program increased muscle mass, lower
extremity muscle strength, and exercise capacity. What is
notable from the study is how small a proportion of
individuals was eligible and willing to participate in the
study. Over a 16-month period, Watson et al. (4) screened
2349 individuals, and 403 were found to be eligible and
were approached for recruitment (17%). Of 403 eligible,
only 38 consented to participate (9.4% of those eligible
and 1.6% of those screened in clinic).
A randomized study in patients with CKD in Brazil
highlights that how exercise is delivered might influence
the effectiveness of the intervention. Baria et al. (5)
randomized 27 sedentary men with stage 3 or 4 CKD and
a body mass index .25 kg/m2 (mean ¼30.4 kg/m2) to an
aerobic exercise group (n¼19) or a control group (n¼9).
Those in the exercise group could participate in either
a center-based program (n¼10) or a home-based program
(n¼9) for 12 weeks. An exercise stress test was done
before randomization to ensure that individuals did not
have active coronary artery disease and determine the
ventilatory threshold (VT), the point where metabolism
switches from aerobic to anaerobic metabolism (6,7). At
this point, the minute ventilation increases faster than the
VO2 (oxygen consumption) to compensate for the
metabolic acidosis. The prescribed exercise program in
the study was on the basis of VT and heart rate at VT and
used to guide exercise intensity. In both groups, there was
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
an improvement in VO2 peak and the sit to stand test. In
the center-based group, visceral fat and waist circumference decreased, but these increased in the control group.
In addition, leg lean mass increased in the in-center group.
In the home-based group, there was no change in body
composition. In both groups, there was an improvement
in the 6-minute walk and sit to stand tests, but there were
no changes in the control group. Exercise was effective,
and the in-center approach was more effective than the
home-based program.
An effective home–based program would be more
feasible to widely implement than in-center programs.
One reason that the home program seemed less effective
might be lower adherence. A non–CKD meta-analysis of
studies of home exercise programs to prevent falls
showed that the components of the home program
seemed to affect adherence in older adults (8). Programs
with greater adherence included home visit support or
telephone support and used a physiotherapist rather than
a trained instructor to deliver the program. A factor that
might relate to why the participants chose to participate
was that higher adherence was found for programs that
included balance exercises, which the participants might
be able to more directly relate to fall prevention. The
factors that might affect patient acceptance and adherence
to exercise studies in CKD have not, to our knowledge,
been previously studied. This would be an important area
of future research.
References
1. Chen IR, Wang SM, Liang CC, Kuo HL, Chang CT, Liu JH, Lin HH,
Wang IK, Yang YF, Chou CY, Huang CC: Association of walking with
survival and RRT among patients with CKD stages 3-5. Clin J Am Soc
Nephrol 9: 1183–1189, 2014 PubMed
2. Robinson-Cohen C, Littman AJ, Duncan GE, Weiss NS, Sachs MC,
Ruzinski J, Kundzins J, Rock D, de Boer IH, Ikizler TA, Himmelfarb
J, Kestenbaum BR: Physical activity and change in estimated
GFR among persons with CKD. J Am Soc Nephrol 25: 399–406,
2014 PubMed
3. Heiwe S, Jacobson SH: Exercise training in adults with CKD: A
systematic review and meta-analysis. Am J Kidney Dis 64: 383–393,
2014 PubMed
4. Watson EL, Greening NJ, Viana JL, Aulakh J, Bodicoat DH, Barratt J,
Feehally J, Smith AC: Progressive resistance exercise training in CKD: A
feasibility study [published online ahead of print December 17, 2014].
Am J Kidney Dis doi:10.1053/j.ajkd.2014.10.019 PubMed
5. Baria F, Kamimura MA, Aoike DT, Ammirati A, Rocha ML, de Mello
MT, Cuppari L: Randomized controlled trial to evaluate the impact of
aerobic exercise on visceral fat in overweight chronic kidney disease
patients. Nephrol Dial Transplant 29: 857–864, 2014 PubMed
6. Kaminsky DA, Knyazhitskiy A, Sadeghi A, Irvin CG: Assessing
maximal exercise capacity: Peak work or peak oxygen consumption?
Respir Care 59: 90–96, 2014 PubMed
7. Loe H, Steinshamn S, Wisløff U: Cardio-respiratory reference data in
4631 healthy men and women 20-90 years: The HUNT 3 fitness study.
PLoS One 9: e113884, 2014 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
8. Simek EM, McPhate L, Haines TP: Adherence to and efficacy of home
exercise programs to prevent falls: A systematic review and metaanalysis of the impact of exercise program characteristics. Prev Med
55: 262–275, 2012 PubMed
Depression in CKD
As reviewed in the last issue of NephSAP, depression is common in patients with CKD and associated
with decreased quality of life and worse outcomes.
A recent meta-analysis evaluated the prevalence
of depression in CKD. The meta-analysis included 216
studies of 55,982 individuals with various levels of
CKD (1). The largest group was individuals undergoing
dialysis. When questionnaires were used to diagnoses
depressive symptoms, the prevalence was 39.3% in
patients on dialysis versus 26.5% in patients with CKD
stages 1–5 not on dialysis and 26.6% in patients with
transplants. The prevalence in patients on dialysis was
lower if interview-based assessments of depression were
used. In fact, the rates of depression were similar across
all stages of CKD (22.8% for patients on dialysis, 21.4%
for patients with stages 1–5 CKD, and 25.7% for patients
with transplants) when using interview-based assessment, which is the gold standard for diagnosis of
depression. The high prevalence of depressive symptoms
using questionnaire-based measures may be because of
the overlap between somatic symptoms of depression
and common symptoms of renal failure (e.g., fatigue and
poor sleep). These findings suggest that depression
instruments might be more useful as a screening test,
but this may be mainly an issue for patients on dialysis, because there was a greater disparity between the
interview- and questionnaire-based measures in this
population. It is notable that, even with the gold
standard clinical interview, approximately one fourth
of patients with CKD are clinically depressed.
One of the commonly used instruments for
depressive symptoms is the Patient Health Questionnaire-9 (PHQ-9), which is composed of nine questions regarding symptoms that the patient has experienced during the past 2 weeks. Questions are graded
on a zero (not at all) to three (nearly every day)
scale, and the scores across questions are summed.
Scores of 15 or greater are consistent with moderately
severe or severe depression. Yu et al. (2) evaluated the
PHQ-9 in 3886 adults with diabetes enrolled in
primary care in Group Health, a health maintenance
organization in Washington and Idaho. In this study,
Yu et al. (2) classified major and minor depressive
343
symptoms on the basis of the number of symptoms
rather than the score (five or more symptoms .50%
of the time for major and two to four symptoms
for minor). The mean scores using the traditional
scoring method were 17.263.8 and 9.662.2 for major
and minor depressive symptoms, respectively. Participants with major depressive symptoms tended to be
younger, black, and women, were more likely to
smoke, and had lower socioeconomic status (lower
education and income). Hemoglobin A1C was higher
in individuals with depressive symptoms (7.7%, 7.9%,
and 8.1% for no, minor, and major depressive symptoms, respectively). Adherence to aspects of diabetes
self-care (e.g., glucose testing, diet, and exercise) was
lower in individuals with major depressive symptoms.
More significantly, the risk of ESRD was higher in
those with depressive symptoms (incident rates: 2.78,
4.30, and 6.65 per 1000 patient-years for those with
no, mild, and severe depressive symptoms, respectively). The association of severe but not mild depressive symptoms with ESRD risk persisted after
adjustment.
Depressive symptoms have also been shown to
be associated with an increased risk of mortality, but
most of the studies have been performed in patients
on dialysis (3). In addition to depressive symptoms,
health–related quality of life is associated with adverse
outcomes. Porter et al. (4) analyzed the association of
quality of life with the 36 Item Short Form Health
Survey (SF-36) in the black Study of Kidney Disease
and Hypertension Trial and cohort with outcomes. At
baseline, the mean age was 55 years old, and mean
eGFR was 48 ml/min per 1.73 m2. The median mental
health composite score was 50, and the median
physical composite score was 47. Both scores were
associated with an increased risk of cardiovascular
event/cardiovascular death, which persisted after adjustment for other factors.
What is not clear from these studies is the
mechanism of association between worse depressive
symptoms or quality of life and adverse events. Furthermore, it is not known whether treatment of depression
would decrease adverse events or patient-reported outcomes. In the study by Yu et al. (2) discussed above, 52%
of individuals with severe depression were prescribed
antidepressant medications. Although this may be relatively low, it suggests that we need additional studies to
determine the best approach to deliver care for individuals
with depression.
344
References
1. Palmer S, Vecchio M, Craig JC, Tonelli M, Johnson DW, Nicolucci A,
Pellegrini F, Saglimbene V, Logroscino G, Fishbane S, Strippoli GF:
Prevalence of depression in chronic kidney disease: Systematic review and
meta-analysis of observational studies. Kidney Int 84: 179–191, 2013 PubMed
2. Yu MK, Weiss NS, Ding X, Katon WJ, Zhou XH, Young BA:
Associations between depressive symptoms and incident ESRD in
a diabetic cohort. Clin J Am Soc Nephrol 9: 920–928, 2014 PubMed
3. Palmer SC, Vecchio M, Craig JC, Tonelli M, Johnson DW, Nicolucci
A, Pellegrini F, Saglimbene V, Logroscino G, Hedayati SS, Strippoli
GF: Association between depression and death in people with CKD:
A meta-analysis of cohort studies. Am J Kidney Dis 62: 493–505,
2013 PubMed
4. Porter A, Fischer MJ, Wang X, Brooks D, Bruce M, Charleston J,
Cleveland WH, Dowie D, Faulkner M, Gassman J, Hiremath L, Kendrick
C, Kusek JW, Norris KC, Thornley-Brown D, Greene T, Lash JP; AASK
Study Group: Quality of life and outcomes in African Americans with
CKD. J Am Soc Nephrol 25: 1849–1855, 2014 PubMed
CKD and Safety
The multiple complications of CKD and coexistent comorbid conditions often lead to complex medication regimens. This can increase the risk of adverse
events from side effects, drug interactions, and medication errors.
Farag et al. (1) evaluated outpatient antibiotic
dosing in patients over age 65 years old with stages 4
and 5 CKD in southwest Ontario. The study from 2003
to 2010 was divided into monthly intervals. Ambulatory
laboratories began reporting eGFR in January of 2006.
The average number of patients was 667 per month.
For antibiotics that require adjustment for the degree of
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
kidney function, the prescribed dose was too high in two
thirds of prescriptions (970 of 1464). This was true for
the time periods before and after automated eGFR
reporting. In addition, nitrofurantoin, contraindicated in
individuals with low levels of kidney function, was
prescribed 169 times. This indicated a need for greater
attention to drug dosing in kidney disease and that
automatic reporting, by itself, will not be sufficient.
The Safe Kidney Care Study is an observational
cohort study in adults with nondialysis CKD and an
eGFR,60 ml/min per 1.73 m2 designed to track the
frequency of disease–specific adverse safety events. The
events included patient-reported events (class 1) and
abnormal laboratory results (class 2) (2). Ginsberg et al.
(2) fount that, of 267 participants in the cohort, 185
(69%) had at least one safety event, and 102 (38%) had
more than one event. The most common adverse safety
event was hypoglycemia, occurring in 34% of subjects
overall, with 58% in those on a medication that lowers
glucose. Hypoglycemia was followed by severe falling
or dizziness (24%). This study did not define the severity
of the hypoglycemic episodes. With regard to abnormal
laboratory results that might relate to medications, the
most common was hyperkalemia (serum potassium .5.5
mEq/L), with 19% of participants taking a medication
that could raise potassium levels. Individuals with diabetes were more likely to have more than one event
(e.g., hypoglycemia and falling).
Table 8. Glycemic targets in Kidney Disease Outcomes Quality Initiative and American Diabetes Association guidelines
Kidney Disease Outcomes Quality Initiative
(2.1) We recommend a target hemoglobin A1C of approximately 7.0% to prevent or delay progression of microvascular
complications of diabetes, including CKD
(2.2) We recommend not treating to a target ,7% in patients at risk of hypoglycemia
(2.3) We suggest that the target hemoglobin A1C be extended above 7% in individuals with comorbidities or limited life
expectancy and risk of hypoglycemia
American Diabetes Association (targets 1 and 3 most applicable to patients with CKD)
(1) Lowering hemoglobin A1C to approximately 7% or less has been shown to reduce microvascular complications of
diabetes, and if implemented soon after the diagnosis of diabetes, it is associated with long-term reduction in
macrovascular disease; therefore, a reasonable hemoglobin A1C goal for many nonpregnant adults is ,7%
(2) Providers might reasonably suggest more stringent hemoglobin A1C goals (such as ,6.5%) for selected individual
patients if this can be achieved without significant hypoglycemia or other adverse effects of treatment; appropriate
patients might include those with short duration of diabetes, type 2 diabetes treated with lifestyle or metformin only,
long life expectancy, or no significant cardiovascular disease
(3) Less stringent hemoglobin A1C goals (such as ,8%) may be appropriate for patients with a history of severe
hypoglycemia, limited life expectancy, advanced microvascular or macrovascular complications, extensive comorbid
conditions, or long-standing diabetes, in whom the general goal is difficult to attain, despite diabetes self-management
education, appropriate glucose monitoring, and effective doses of multiple glucose–lowering agents, including insulin
Modified with permission from National Kidney Foundation: 2012 Update. Am J Kidney Dis 60: 850–886, 2012 and American Diabetes Association: Standards of medical care in
diabetes, 2015. Diabetes Care 38[Suppl]: S1–S93, 2015.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
345
Figure 16. Patient and disease factors used to determine optimal hemoglobin A1C targets. Characteristics and predicaments
toward the left justify more stringent efforts to lower hemoglobin A1C targets; those toward the right suggest less stringent
efforts. Reprinted with permission from American Diabetes Association: Standards of medical care in diabetes, 2015. Diabetes
Care 38(Suppl): S1–S93, 2015.
Yun et al. (3) evaluated the incidence of severe
hypoglycemia, defined as an episode requiring the assistance of another person, in a cohort of 1217 individuals
with type 2 diabetes and preserved kidney function
(eGFR$60 ml/min per 1.73 m2). Over a median 10 years
of follow-up, there were 140 episodes in 111 patients.
Severe hypoglycemia was more common in individuals
with overt albuminuria, even after accounting for age,
duration of diabetes, mean hemoglobin A1C, insulin and
sulfonylureas, and eGFR (3). The risk was 2.5 times
higher compared with that in those without albuminuria.
Individuals with microalbuminuria did not have an increased risk. Other independent risk factors included older
age and longer duration of diabetes. Because eGFR was
not lower, decreased clearance of medications would not
explain the finding, and the mechanism is not clear.
However, the results of studies by Ginsberg et al. (2)
and Yun et al. (3) reinforce the recommendation that tight
glycemic control in the older, diabetic patient with advanced microvascular complications is not advisable.
The current recommendations for hemoglobin A1C targets from Kidney Disease Outcomes Quality Initiative (4)
and the American Diabetes Association (ADA) (5) are
shown in Table 8. A useful figure that can be a guide for
when to be less stringent in hemoglobin A1C goals is
depicted in Figure 16 from the 2015 ADA Practice
Guidelines (5).
Complex regimens can also affect medication
adherence. The Chronic Kidney Disease in Children
Study is a prospective cohort study in North America of
children with CKD (6). In this study of 558 children, the
median age was 11 years old, and median GFR was 44
ml/min per 1.73 m2. Not unexpectedly, more medications
were required with worse levels of GFR. Self-reported
346
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
nonadherence with medications was associated with
older age, White race, and increased dosing frequency. Nonadherence was 70% more likely for
medications dose greater than two times per day compared with that one time per day. A larger number of
comorbidities was not associated with nonadherence
or the number of different medication classes prescribed. This suggests that, if the medications can be
taken all together, the number of medications taken is
not as important as a regimen that requires medications to be spaced apart or taken at multiple times
during the day.
Other common medication issues include the
use of nonsteroidal anti–inflammatory drugs and the
Table 9. National Kidney Foundation list of 37 unique
herbs potentially harmful in the setting of CKD (http://
www.kidney.org/atoz/herbalsupp) as listed by Grubbs
et al.
Potentially Toxic
to the Kidneys
Artemisia absinthium
(wormwood plant)a
Autumn crocus
Chuifong tuokuwan
(black pearl)
Horse chestnut
Periwinkle
Sassafrasb
Tung shueh
Vandelia cordifolia
Potentially
Harmful in
CKD
Known to Be
Unsafe for All
People
Alfalfaa
Chapparal
Aloea
Bayberrya
Comfrey
Ephedra (ma
huang)a
Lobelia
Mandrake
Pennyroyal
Pokeroota
Sassafrasb
Sennaa,b
Yohimbea
Blue cohosh
Brooma
Buckthorna
Capsicuma
Cascaraa
Coltsfoot
Dandeliona
Gingera
Ginsenga
Horsetaila
Licoricea
Mate
Nettlea
Noni juicea
Panaxa
Rhubarba
Sennaa,b
Vervain
Modified with permission from Grubbs V, Plantinga LC, Tuot DS, Hedgeman E,
Saran R, Saydah S, Rolka D, Powe NR; Centers for Disease Control and Prevention
CKD Surveillance Team: Americans’ use of dietary supplements that are potentially
harmful in CKD. Am J Kidney Dis 61: 739–747, 2013.
a
Identified in dietary supplements reportedly taken in the study population.
b
Herb listed in more than one category but only counted one time as a unique herb.
relatively frequent use of herbal supplements that may be
harmful in patients with CKD. Some of these herbs are
nephrotoxic, whereas others can interfere with medications or aggravate other CKD complications or risk
factors. Grubbs et al. (7) evaluated the use of dietary
supplements that might be harmful in CKD in the
National Health and Nutrition Examination Survey.
Grubbs et al. (7) used the list compiled by the National
Kidney Foundation of 37 supplements/herbs that are
harmful in CKD (Table 9). Over 50% of participants
took some form of herbal supplement. Approximately
8.5% of individuals without CKD versus 6% of those
with CKD used a supplement that was potentially
harmful. Although the proportion using a harmful supplement was slightly lower in those with CKD, individuals with CKD were more likely to be taking the
supplement for .3 years, increasing the risk of cumulative harm. The most commonly used potentially harmful supplement was ginseng or supplements that contained ginseng. Individuals who used supplements were
more likely to be non-Hispanic white, have higher education levels, have higher income, and have a higher
number of health care visits. This suggests that more
attention needs to be paid to supplement use among
patients with CKD. The higher number of visits among
those who did use supplements indicates that there are
opportunities for education to decrease risk.
The pharmacokinetics and safety of a number of
diabetic medications are changed by CKD. The half-life
of insulin and a number of sulfonylureas are increased
in CKD, increasing the risk of hypoglycemia. Metformin, typically the first-choice medication for diabetes, is contraindicated in advanced CKD. The use
of metformin in CKD was discussed in one of the
medication safety recommendations from the 2012
Kidney Disease: Improving Global Outcomes Guideline 4.4.6: “We recommend that metformin be continued in people with GFR $45 ml/min per 1.73 m2
(GFR categories G1–G3a); its use should be reviewed
in those with GFR 30–44 ml/min per 1.73 m2 (GFR
category G3b); and it should be discontinued in people
with GFR ,30 ml/min per 1.73 m2 (GFR categories
G4–G5)” (4). After lower GFR levels are reached,
other agents are needed, although they may be associated with an increased risk of hypoglycemia.
There are a number of new diabetic medications that patients with CKD may be using. One
class is the dipeptidyl peptidase-4 (DPP-4) inhibitors.
347
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Table 10. Dose adjustment of Food and Drug Administration–approved dipeptidyl peptidase 4 inhibitors in CKD
Medication
Creatinine Clearance
.50 ml/min
Creatinine Clearance
¼30–50 ml/min
Sitagliptin (mg/d)
Saxagliptin (mg/d)
Linagliptin (mg/d)
Alogliptin (mg/d)
100
2.5–5
5
25a
50 (1/2 dose)
2.5 (1/2 dose)
5 (no adjustment)
12.5a (1/2 dose)
Creatinine Clearance
,30 ml/min
25
2.5
5
6.25
(1/4 dose)
(1/2 dose)
(no adjustment)
(1/4 dose)
Modified with permission from Ioannidis I: Diabetes treatment in patients with renal disease: Is the landscape clear enough? World J Diabetes 5: 651–658, 2014.
a
Alogliptin creatinine clearance categories are $60 and 30 to ,60.
The inhibition of DPP-4 blocks breakdown of
glucagon-like peptide-1, which enhances insulin secretion in response to a meal, reduces gastric emptying time, and reduces appetite (8). Except for
linagliptin, the DPP-4 inhibitors must be dose adjusted
in kidney disease (Table 10) (9). A recent metaanalysis evaluated the safety and efficacy of DPP-4
inhibitors in CKD in 10 studies (4). Compared with
placebo, DPP-4 medications lowered hemoglobin
A1C, on average, by 0.5%. In two studies that compared a DPP-4 inhibitor with glipizide, the degree
of hemoglobin A1C lowering was similar. DPP-4
inhibitors had a lower risk of hypoglycemia than
glipizide and a similar rate to placebo (Figure 17)
(10). Thus, these medications may be safer alternatives
in kidney disease; however, we do not have large,
long–term safety studies.
Individuals with CKD are at increased risk
for hypoglycemia, with at least one episode
occurring in more than one half of patients
with CKD. This observation should reinforce
the need to adjust hypoglycemic agents
and avoid targeting a hemoglobin A1C of
,7% in individuals at risk for hypoglycemia.
References
1. Farag A, Garg AX, Li L, Jain AK: Dosing errors in prescribed
antibiotics for older persons with CKD: A retrospective time series
analysis. Am J Kidney Dis 63: 422–428, 2014 PubMed
2. Ginsberg JS, Zhan M, Diamantidis CJ, Woods C, Chen J, Fink JC:
Patient-reported and actionable safety events in CKD. J Am Soc Nephrol
25: 1564–1573, 2014 PubMed
3. Yun JS, Ko SH, Ko SH, Song KH, Ahn YB, Yoon KH, Park YM, Ko
SH: Presence of macroalbuminuria predicts severe hypoglycemia in
Figure 17. Meta-analysis for the risk of hypoglycemia. (A) DPP-4 inhibitor vs. placebo or no treatment. (B) DPP-4 inhibitor
versus glipizide 95% CI, 95% confidence interval; DPP4i, dipeptidyl pepdidase-4; M-H. Reprinted with permission from Cheng
D, Fei Y, Liu Y, Li J, Chen Y, Wang X, Wang N: Efficacy and safety of dipeptidyl pepdidase-4 in type 2 diabetes mellitus
patients with moderate to severe renal impairment: A systematic review and meta-analysis. PLoS One 9: e111543, 2014.
348
4.
5.
6.
7.
8.
9.
10.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
patients with type 2 diabetes: A 10-year follow-up study. Diabetes Care
36: 1283–1289, 2013 PubMed
National Kidney Foundation: 2012 Update. Am J Kidney Dis 60: 850–
886, 2012 PubMed
American Diabetes Association: Standards of medical care in diabetes,
2015. Diabetes Care 38[Suppl]: S1–S93, 2015 PubMed
Blydt-Hansen TD, Pierce CB, Cai Y, Samsonov D, Massengill S,
Moxey-Mims M, Warady BA, Furth SL: Medication treatment complexity and adherence in children with CKD. Clin J Am Soc Nephrol 9:
247–254, 2014 PubMed
Grubbs V, Plantinga LC, Tuot DS, Hedgeman E, Saran R, Saydah S, Rolka
D, Powe NR; Centers for Disease Control and Prevention CKD Surveillance Team: Americans’ use of dietary supplements that are potentially
harmful in CKD. Am J Kidney Dis 61: 739–747, 2013 PubMed
Garber AJ: Incretin therapy–present and future. Rev Diabet Stud 8: 307–
322, 2011 PubMed
Ioannidis I: Diabetes treatment in patients with renal disease: Is the
landscape clear enough? World J Diabetes 5: 651–658, 2014 PubMed
Cheng D, Fei Y, Liu Y, Li J, Chen Y, Wang X, Wang N: Efficacy and
safety of dipeptidyl peptidase-4 inhibitors in type 2 diabetes mellitus
patients with moderate to severe renal impairment: A systematic review
and meta-analysis. PLoS One 9: e111543, 2014 PubMed
Systems of Care and Effect on CKD
There has been increasing emphasis on the understanding the effect of the education programs and care
delivery on the recognition and progression of CKD. With
the financial costs associated with CKD, assessing the
effectiveness of the interventions to slow the progression
of kidney disease has been the focus of several studies.
Multidisciplinary Clinics
A multidisciplinary care (MDC) approach to CKD
management has been promoted on the basis of the
potential to affect the decline of renal function. Chen et al.
(1) studied the effect of MDC versus usual care in
a prospective cohort study involving 1056 Taiwanese
subjects over a 3-year period. Primary end points were
progression to ESRD (initiation of dialysis) and mortality.
The rate of renal functional decline, BP control, anemia
management, and metabolic bone disease parameters were
studied as well. The rates of decline for CKD stages 4 and
5 were slower in the MDC group (25.1% versus 27.3%
ml/min). The MDC group spent more time with RRT
planning/education, which was reflected by the greater
percentage of participants who opted for peritoneal dialysis, fewer individuals beginning dialysis with a temporary catheter, and a 51% decrease in mortality but an
anticipated increase in dialysis initiation of 68%. The latter
finding may be attributed to lower mortality, because there
was no difference in the combined end point of death
or dialysis initiation (adjusted hazard ratio, 1.02; 95%
confidence interval, 0.70 to 1.50). There was an increased
use of medications for reduction of proteinuria, phosphate
binding, and anemia treatment with erythropoiesisstimulating agents in the MDC group. The predialysis
education and team management approach helped delay
progression of renal disease and facilitated RRT preparation.
The addition of nephrology nurses or nurse practitioners to usual care has not consistently shown positive
results as reviewed in the last NephSAP CKD edition.
The Multifactorial Approach and Superior Treatment
Efficacy in Renal Patients with the Aid of Nurse Practitioners Study involved 788 individuals with moderate to
severe CKD randomized to additional care by a nurse
practitioner (intervention group) or usual care by a physician group (control) (2). Over a median follow-up of 5–7
years, there was a 20% reduction in the composite renal
end point of death, ESRD, and a 50% increase in the SCr
(99.6 versus 118.7 events per 100 person-years). The
intervention group had a 0.45-ml/min less decline in GFR
per year and better achievement of clinical targets, such
as BP control, thereby showing the value of a team
approach to CKD.
Although prior trials showed inconsistent
results, in a recent randomized trial comparing additional care by a nurse practitioner
with usual care, there was a 20% reduction
in progression of kidney disease.
Preparation for RRT is one of the key steps in
management of CKD. The ability to influence the choice
of RRT modality through a combination of education,
MDC, and patient engagement was highlighted in a prospective study by Rayner et al. (3). Changes in the incidence and treatment modality were followed in 10,522
patients with CKD and 8509 patients with diabetes but
no CKD between 2003 and 2006. Patient engagement
strategies, such as education, financial incentives, and
personalized reports after each clinical visit, and the
incorporation of practice changes of automated GFR
reporting and multidisciplinary pre–RRT care were used
on the cohort of patients. After 2 years, the incident rate
of RRT declined from 23.9% to 19.6%%. A 63% increase
in the combined end points of transplantation, choice of
peritoneal dialysis, and arteriovenous fistula use was noted
by 2012. Of those who opted not to pursue dialysis, 52%
were able to make the arrangements to die at home.
The lack of having a multidisciplinary team
approach can result in gaps in care. Identifying these
issues can be turned into opportunities for collaboration.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Reviewing the records of 196 adolescents with CKD,
ESRD, or transplantation who were transitioning from
pediatric to adult care providers, Hooper et al. (4)
determined that only 58% had the recommended cardiovascular risk assessment. Recommended therapy
for modifiable risk factors was documented just 57%
of the time. The identification of the gap in care for
these adolescents in terms of adequate cardiovascular
risk management shows an opportunity for developing
collaborative teams to help transition them from pediatric practitioners to adult nephrologists. Ovbiagele et al.
(5) reviewed the records of 236,662 patients with
ischemic stroke and CKD hospitalized between 2009
and 2012. Those with severe renal dysfunction or failure
were less likely to receive the therapy-based guidelines,
such as anticoagulation, smoking cessation intervention,
or lipid reduction. Inpatient mortality was higher in
patients with CKD.
Economics
Using National Health and Nutrition Examination
Survey data and Medicare claims, Honeycutt et al. (6)
analyzed medical costs associated with different stages of
CKD. Medicare costs ranged from $1700 per year for
stage 2 CKD and $3500 per year for stage 3 CKD to
$12,700 per year for stage 4 CKD. The financial burdens
of CKD and ESRD are significant and make the case for
interventions that slow progression of CKD. The cost of
care and lack of coverage influence the outcomes and
management decisions. The effect of broadened Medicaid
coverage was examined in 408,535 adults ages 20–65
years old with variable levels of Medicaid coverage (7).
With every 10% increase in the population covered by
Medicaid, there was a 1.8% decrease in ESRD incidence.
There were also concomitant reductions in the access gap
to transplantation and peritoneal dialysis (Figure 18).
Electronic Health Records
The use of electronic health records has the ability
to influence practice patterns and trigger management
strategies for aspects of CKD. The influence of eGFR
reporting on decisions in dialysis initiation, referral
patterns to nephrology, recognition of CKD stage, and
effects on medication prescription has been studied
recently. A retrospective review of patients with CKD
followed in a rural primary care practice showed the
effect of eGFR reporting on recognition of CKD (8). Of
those with CKD on the basis of laboratory values, 52%
had no documentation of a CKD diagnosis. Patients
349
Figure 18. Effects of insurance status on various aspects of
renal replacement therapy preparation. Rates of access to
care among privately insured, Medicaid, and uninsured nonelderly adults between 2001 and 2008. AVF, arteriovenous
fistula. Reprinted with permission from Kurella-Tamura M,
Goldstein BA, Hall YN, Mitani AA, Winkelmayer WC: State
medicaid coverage, ESRD incidence, and access to care. J Am
Soc Nephrol 25: 1321–1329, 2014.
formally diagnosed with CKD by primary care practitioners had more antihypertensive medication changes
and increased comanagement with nephrologists. Some
data suggest greater recognition of CKD but not
necessarily with changes in management. In a survey of
nephrologists regarding their likelihood of initiating
dialysis in four different clinical scenarios, Brimble
et al. (9) examined the effect that eGFR had on decision
making. For each scenario, the respondent was asked to
rank his or her likelihood of recommending dialysis
initiation on a modified eight-point Likert scale ranging
from one (definitely would not) to eight (definitely would).
Dialysis initiation recommendations increased by 0.55
points when eGFR was reported. A subgroup analysis
showed that physicians in practice for .13 years were
more influenced (P¼0.03). eGFR reporting was also
shown to effect the timing of referrals to nephrology. A
study of 25,000 patients who initiated RRT between
January 1, 1999 and December 31, 2010 from the
Australia and New Zealand Dialysis Transplant Registry
showed an absolute reduction of 3.7% (P,0.001) in late
referrals comparing the pre–GFR reporting period with its
post-GFR counterpart (10). However, this small reduction
in late referrals did not extend to improvements in
preparation for RRT, such as arteriovenous fistula creation. Another example of where eGFR reporting facilitated
recognition and documentation of CKD but did not
350
translate to meaningful changes in outcomes is found in
the study of two Veterans Administration cohorts before
and after introduction of laboratory–automated eGFR
reporting (11). Wang et al. (11) detected an increased
recognition and documentation of CKD stage, but there no
change in nephrology referral rate.
Wei et al. (12) looked at the effect of reporting
eGFR on prescribing of nonsteroidal anti–inflammatory
drugs (NSAIDs). NSAID prescriptions decreased from
39,459 to 35,415 after implementation of eGFR reporting. Those patients who stopped NSAIDs experienced
improvements in eGFR. In patients with stage 4 CKD,
eGFR increased slightly from 23.9 to 27.1 ml/min per
1.73 m2; however, the increase was from 12.4 to 26.4 ml/min
per 1.73 m2 among patients with stage 5 CKD.
There are several guidelines for CKD management, but incorporation into routine practice has been
difficult to achieve. In a prospective study of an
academic primary care clinic, the implementation of
a checklist for laboratory testing and interventions for
each stage of CKD was evaluated (13). The patients
assigned to primary care providers using the checklist
were more routinely tested for albuminuria, hyperparathyroidism by parathyroid hormone, and phosphorus.
The same group displayed superior rates of diabetes
control, anti–renin-angiotensin-aldosterone system drug
use, appropriate immunizations, and avoidance of
NSAIDs.
CKD Education
The importance of education regarding CKD and
RRT options has been established. However, the ability
to provide this during a routine physician visit is limited.
Verhave et al. (14) assessed awareness of CKD and
cardiovascular risk factors among 20,004 patients who
had free access to medical care in Quebec. Self-awareness
of CKD, diabetes, hypertension, or hyperlipidemia was
low, despite the regular physician checkups. Thus, there is
a need for interventions to increase awareness of the
disease processes and subsequently, improve achievement
of guideline targets. Attempts to incorporate a teaching
tool that could be used during a routine nephrology visit
was assessed by Wright Nunes et al. (15). A one–page
education worksheet was used during routine visits for
patients with stages 1–5 CKD. Of 155 patients who
received this teaching tool, there were higher rates of CKD
awareness and acknowledgment of kidney function level
and stage. Kurella Tamura et al. (16) compared 595
patients with ESRD who had participated in the National
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Kidney Foundation Kidney Early Evaluation Program
(KEEP) with a group of matched patients not in the
KEEP. Those who had gone through the KEEP had
significantly higher rates of pre–ESRD nephrology care,
choice of peritoneal dialysis as renal replacement modality, preemptive transplant listing, and transplantation over
participants not in the KEEP (76.0% versus 69.3%, 10.3%
versus 6.4%, 24.1% versus 17.2%, and 9.7% versus 6.4%
in KEEP versus non-KEEP, respectively). There was no
statistically significant difference in mortality.
References
1. Chen Y-R, Yang Y, Wang S-C, Chiu P-F, Chou W-Y, Lin C-Y, Chang
J-M, Chen T-W, Ferng S-H, Lin C-L: Effectiveness of multidisciplinary
care for chronic kidney disease in Taiwan: A 3-year prospective cohort
study. Nephrol Dial Transplant 28: 671–682, 2013 PubMed
2. Peeters MJ, van Zuilen AD, van den Brand JA, Bots ML, van Buren M,
Ten Dam MA, Kaasjager KA, Ligtenberg G, Sijpkens YW, Sluiter HE,
van de Ven PJ, Vervoort G, Vleming LJ, Blankestijn PJ, Wetzels JF:
Nurse practitioner care improves renal outcome in patients with CKD. J
Am Soc Nephrol 25: 390–398, 2014 PubMed
3. Rayner HC, Baharani J, Dasgupta I, Suresh V, Temple RM, Thomas
ME, Smith SA: Does community-wide chronic kidney disease management improve patient outcomes? Nephrol Dial Transplant 29: 644–
649, 2014 PubMed
4. Hooper DK, Williams JC, Carle AC, Amaral S, Chand DH, Ferris ME,
Patel HP, Licht C, Barletta GM, Zitterman V, Mitsnefes M, Patel UD:
The quality of cardiovascular disease care for adolescents with kidney
disease: A Midwest Pediatric Nephrology Consortium study. Pediatr
Nephrol 28: 939–949, 2013 PubMed
5. Ovbiagele B, Schwamm LH, Smith EE, Grau-Sepulveda MV, Saver
JL, Bhatt DL, Hernandez AF, Peterson ED, Fonarow GC: Patterns of
care quality and prognosis among hospitalized ischemic stroke
patients with chronic kidney disease. J Am Heart Assoc 3: e000905,
2014 PubMed
6. Honeycutt AA, Segel JE, Zhuo X, Hoerger TJ, Imai K, Williams D:
Medical costs of CKD in the Medicare population. J Am Soc Nephrol
24: 1478–1483, 2013 PubMed
7. Kurella-Tamura M, Goldstein BA, Hall YN, Mitani AA, Winkelmayer
WC: State medicaid coverage, ESRD incidence, and access to care. J
Am Soc Nephrol 25: 1321–1329, 2014 PubMed
8. Rao MK, Morris CD, O’Malley JP, Davis MM, Mori M, Anderson S:
Documentation and management of CKD in rural primary care. Clin J
Am Soc Nephrol 8: 739–748, 2013 PubMed
9. Brimble KS, Mehrotra R, Tonelli M, Hawley CM, Castledine C,
McDonald SP, Levidiotis V, Gangji AS, Treleaven DJ, Margetts PJ,
Walsh M: Estimated GFR reporting influences recommendations for
dialysis initiation. J Am Soc Nephrol 24: 1737–1742, 2013 PubMed
10. Foote C, Clayton PA, Johnson DW, Jardine M, Snelling P, Cass A:
Impact of estimated GFR reporting on late referral rates and practice
patterns for end-stage kidney disease patients: A multilevel logistic
regression analysis using the Australia and New Zealand Dialysis
and Transplant Registry (ANZDATA). Am J Kidney Dis 64: 359–
366, 2014 PubMed
11. Wang V, Maciejewski ML, Hammill BG, Hall RK, Van Scoyoc L, Garg
AX, Jain AK, Patel UD: Recognition of CKD after the introduction of
automated reporting of estimated GFR in the Veterans Health Administration. Clin J Am Soc Nephrol 9: 29–36, 2014 PubMed
12. Wei L, MacDonald TM, Jennings C, Sheng X, Flynn RW, Murphy MJ:
Estimated GFR reporting is associated with decreased nonsteroidal antiinflammatory drug prescribing and increased renal function. Kidney Int
84: 174–178, 2013 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
13. Mendu ML, Schneider LI, Aizer AA, Singh K, Leaf DE, Lee TH,
Waikar SS: Implementation of a CKD checklist for primary care
providers. Clin J Am Soc Nephrol 9: 1526–1535, 2014 PubMed
14. Verhave JC, Troyanov S, Mongeau F, Fradette L, Bouchard J, Awadalla
P, Madore F: Prevalence, awareness, and management of CKD and
cardiovascular risk factors in publicly funded health care. Clin J Am Soc
Nephrol 9: 713–719, 2014 PubMed
15. Wright Nunes J, Greene JH, Wallston K, Eden S, Shintani A, Elasy T,
Rothman RL, Ikizler TA, Cavanaugh KL: Pilot study of a physiciandelivered education tool to increase patient knowledge about CKD. Am
J Kidney Dis 62: 23–32, 2013 PubMed
16. Kurella Tamura M, Li S, Chen SC, Cavanaugh KL, Whaley-Connell
AT, McCullough PA, Mehrotra RL: Educational programs improve the
preparation for dialysis and survival of patients with chronic kidney
disease. Kidney Int 85: 686–692, 2014 PubMed
Disparities
Risk for development of CKD, rate of progression of kidney disease, and outcomes can be influenced
by different factors, such as sex, race, socioeconomic
status, and health literacy. Having a better understanding of these factors will guide more targeted management strategies.
Race and Sex
The effect of race on incidence, rate of progression, and mortality continues to be an area of focus.
Derose et al. (1) analyzed a retrospective cohort from the
Kaiser Permanente Database from 2003 to 2009 and
documented racial differences in projected kidney failure
and mortality. Black patients had more extreme rates of
eGFR decline. The first percentile of 136,923 black
patients decreased by 223.6 ml/min per year compared
with the first percentile of 350,917 Hispanic patients
(220.9-ml/min per year decline), the first percentile
of 526,498 white patients (220.1-ml/min per year decline), and the first percentile of 105,476 Asian patients
(217.6-ml/min per year decline). However, these greater
rates of decline in black patients did not result in greater
rates of mortality. The highest rate of mortality was
among white patients. The difference in survival, despite
slower rates of GFR decline, was noted again in the
historical cohort of 518,406 white men and 52,402 black
men in the United Stated veterans studies by Kovesdy
et al. (2). Over a median follow-up of 4.7 years, 172,093
patients died. Black race was associated with lower crude
mortality rates, especially in the setting of more advanced
CKD (hazard ratio, 0.95; 95% confidence interval, 0.94 to
0.97); the effect persisted with full adjustment (hazard
ratio, 0.72; 95% confidence interval, 0.70 to 0.73). The
difference in mortality may be attributable to the different
etiologies of CKD. The varying rates of GFR decline
were further illustrated in the longitudinal study of 3348
351
black and white adults who were enrolled in the Coronary
Artery Risk Development in Young Adults Study (3).
Renal functional assessment by cystatin C–based eGFR
was completed during routine study visits. Among blacks,
there was a more rapid decline in GFR at earlier ages.
During period 1 of the study (10–15 years postbaseline
examination), the rate of decline in GFR was 0.13 ml/min
faster. In period 2 (.15 years postbaseline), the rate of
decline was 0.32 ml/min faster. Some of the difference in
GFR decline could be related to creatinine differences.
The identification of different genes associated
with renal disorders has been implicated in the racial
differences in prevalence and progression of CKD.
Case-control studies have noted that blacks with variants
of apolipoprotein L1 (APOL1) are more likely to have
FSGS, HIV-associated nephropathy, and ESRD. The
importance of APOL1 variants in terms of susceptibility
was examined in blacks with nondiabetic nephropathy
(4). Both related and unrelated blacks with nondiabetic
nephropathy were assessed for the interaction between
JC virus, BK polyomavirus, human herpesvirus 6, or
cytomegalovirus and APOL1 risk alleles. Given the rare
incidence of human herpesvirus 6 and cytomegalovirus
within the study, conclusions about the interaction of
these viruses with APOL1 could not be made; however,
BK virus did not cause nephropathy. Blacks at risk for
APOL1-associated nephropathy who had concomitant
JC viruria had a lower risk of kidney disease.
The effect of APOL1 G1 and G2 risk alleles on the
development of CKD and progression to ESRD was
assessed in 3607 blacks in the Atherosclerosis Risk in
Communities (ARIC) Study (5). Carrying two risk alleles
was associated with a 1.49-fold increased risk of CKD
and a 1.58-fold increased risk of ESRD compared with
those with zero or one allele. The association of APOL1
risk variants on progression of CKD in black patients
compared with white patients was assessed in the black
Study of Kidney Disease and Hypertension (AASK) Trial
and the Chronic Renal Insufficiency Cohort (CRIC) Study
(6) as previously reviewed in this issue of NephSAP
in the section on Genetics. The effect of having two risk
alleles of APOL1 (high risk) versus zero or one copy
(low risk) was analyzed using data from 693 black patients enrolled in the AASK Trial and 2955 black and
white patients enrolled in the CRIC Study, in which 45%
of the population had diabetes. In the AASK Trial, the
primary outcome of ESRD or doubling of the SCr was
achieved in 58.1% of the APOL1 high–risk group and
36.6% of the APOL1 low–risk group. In the CRIC Study,
352
the APOL1 high–risk group had a more rapid decline in
GFR and higher risk of worse renal outcomes.
The presence of albuminuria is a known risk factor
for renal disease, and it is a marker of vascular injury.
The effect of albumin to creatinine ratio (ACR) and
eGFR on risk of stroke, heart failure, and coronary artery
disease was assessed in the ARIC Study observational
cohort of 11,060 white and black individuals who were
52–75 years old (7). An eGFR,70 ml/min per 1.73 m2
and the log ACR were linearly associated with risk of
cardiovascular disease (CVD). In this study, associations
of eGFR and ACR with CVD were generally similar
among subgroups of age, sex, and race. However, it
remains unclear if albuminuria has a different effect on
CVD on the basis of sex or race. A meta-analysis of 46
cohorts from Europe, North and South America, Asia,
and Australia focused on the interaction of sex on GFR
and albuminuria with all-cause mortality, cardiovascular
mortality, and ESRD (8). Risks of all-cause mortality and
cardiovascular mortality were higher in men at all levels
of eGFR and ACR. The slope of the risk relation was
steeper in women. There was no difference between
sexes in terms of ESRD risk.
There were a few studies addressing the role of
albuminuria as a risk factor for racial differences in the
outcomes. Extrapolating on the increased rate of incident
stroke in black individuals with excess urinary albumin
excretion, Gutiérrez et al. (9) looked at the risk of incident
coronary heart disease (CHD) related to albuminuria in
a prospective cohort study of subjects enrolled in the
Reasons for Geographic and Racial Difference in Stroke
Study between 2003 and 2007. Among those who, at
baseline, had no CHD (n¼23,373), rates of incident events
occurred at a 1.5-fold greater rate than in black participants.
Higher rates of albuminuria were associated with greater
risk of incident CHD among black participants. Those with
recurrent CHD had similar degrees of albuminuria. The
prevalence of diabetes and ESRD was greater in blacks
and Hispanics. Using the National Health and Nutrition
Examination Survey (NHANES) data, a survey of 2310
patients with diabetes over the age of 20 years old was
performed to assess the effect of albuminuria (10). The
prevalence of early CKD was greater among Hispanics and
blacks and associated with urinary albumin excretion and
C–reactive protein levels.
Access to Care
Access to care, specifically education, has an effect on modality choice of RRT, pursuit of preemptive
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
transplantation, and rate of progression of CKD. Whether
this can be generalized to different racial groups, economic groups, or geographic areas was addressed by
a few studies. On the basis of 2009–2011 data from The
Netherlands Predialysis Patient Record Study of patients
with CKD within a universal health care system, blacks
have faster disease progression than whites (11). At
baseline, black patients were younger, more likely diabetic, had greater proteinuria, and had greater eGFRs.
There was more rapid decline in renal function of 0.18 ml/min
per month in blacks compared with white patients. van
den Beukel et al. (11) drew the conclusion that the
health care system factors have a less influential role in
explaining black and white differences in progression
to ESRD. Vart et al. (12) studied the effect of education
in two cohorts: one from the United States (NHANES)
and one from The Netherlands (Prevention of Renal
and Vascular Endstage Disease Study). In the United
States, where access to care is income dependent, low
income was a predictor of CKD. In The Netherlands,
lower education levels had an association with CKD.
Racial variations in pre-ESRD care were noted in the
analysis of the US Renal Data System data regarding
404,622 non–Hispanic white and black patients starting
dialysis between 2005 and 2010 (13). Yan et al. (13)
consistently noted patterns, in which black patients received less pre–ESRD care, defined as having an arteriovenous access at the start of dialysis, nephrology care
at 6–12 months, and use of erythropoietin-stimulating
agents. Larger metropolitan and small rural areas had
lower rates of pre-ESRD care. The importance of education and health literacy is difficult to assess given that
a lack of access to care has a more overwhelming effect.
References
1. Derose SF, Rutkowski MP, Crooks PW, Shi JM, Wang JQ, KalantarZadeh K, Kovesdy CP, Levin NW, Jacobsen SJ: Racial differences in
estimated GFR decline, ESRD, and mortality in an integrated health
system. Am J Kidney Dis 62: 236–244, 2013 PubMed
2. Kovesdy CP, Quarles LD, Lott EH, Lu JL, Ma JZ, Molnar MZ,
Kalantar-Zadeh K: Survival advantage in black versus white men with
CKD: Effect of estimated GFR and case mix. Am J Kidney Dis 62: 228–
235, 2013 PubMed
3. Peralta CA, Vittinghoff E, Bansal N, Jacobs D Jr., Muntner P,
Kestenbaum B, Lewis C, Siscovick D, Kramer H, Shlipak M,
Bibbins-Domingo K: Trajectories of kidney function decline in young
black and white adults with preserved GFR: Results from the Coronary
Artery Risk Development in Young Adults (CARDIA) study. Am J
Kidney Dis 62: 261–266, 2013 PubMed
4. Divers J, Núñez M, High KP, Murea M, Rocco MV, Ma L, Bowden
DW, Hicks PJ, Spainhour M, Ornelles DA, Kleiboeker SB, Duncan K,
Langefeld CD, Turner J, Freedman BI: JC polyoma virus interacts with
APOL1 in African Americans with nondiabetic nephropathy. Kidney Int
84: 1207–1213, 2013 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
5. Foster MC, Coresh J, Fornage M, Astor BC, Grams M, Franceschini N,
Boerwinkle E, Parekh RS, Kao WH: APOL1 variants associate with
increased risk of CKD among African Americans. J Am Soc Nephrol 24:
1484–1491, 2013 PubMed
6. Parsa A, Kao WH, Xie D, Astor BC, Li M, Hsu CY, Feldman HI,
Parekh RS, Kusek JW, Greene TH, Fink JC, Anderson AH, Choi MJ,
Wright JT Jr., Lash JP, Freedman BI, Ojo A, Winkler CA, Raj DS,
Kopp JB, He J, Jensvold NG, Tao K, Lipkowitz MS, Appel LJ; AASK
Study Investigators; CRIC Study Investigators: APOL1 risk variants,
race, and progression of chronic kidney disease. N Engl J Med 369:
2183–2196, 2013 PubMed
7. Hui X, Matsushita K, Sang Y, Ballew SH, Fülöp T, Coresh J: CKD and
cardiovascular disease in the Atherosclerosis Risk in Communities
(ARIC) study: Interactions with age, sex, and race. Am J Kidney Dis 62:
691–702, 2013 PubMed
8. Nitsch D, Grams M, Sang Y, Black C, Cirillo M, Djurdjev O, Iseki K,
Jassal SK, Kimm H, Kronenberg F, Oien CM, Levey AS, Levin A,
Woodward M, Hemmelgarn BR; Chronic Kidney Disease Prognosis
Consortium: Associations of estimated glomerular filtration rate and
albuminuria with mortality and renal failure by sex: A meta-analysis.
BMJ 346: f324, 2013 PubMed
9. Gutiérrez OM, Khodneva YA, Muntner P, Rizk DV, McClellan
WM, Cushman M, Warnock DG, Safford MM; REGARDS Investigators: Association between urinary albumin excretion and coronary heart disease in black vs white adults. JAMA 310: 706–714,
2013 PubMed
10. Sinha SK, Shaheen M, Rajavashisth TB, Pan D, Norris KC, Nicholas
SB: Association of race/ethnicity, inflammation, and albuminuria in
patients with diabetes and early chronic kidney disease. Diabetes Care
37: 1060–1068, 2014 PubMed
11. van den Beukel TO, de Goeij MC, Dekker FW, Siegert CE,
Halbesma N; PREPARE Study Group: Differences in progression
to ESRD between black and white patients receiving predialysis
care in a universal health care system. Clin J Am Soc Nephrol 8:
1540–1547, 2013 PubMed
12. Vart P, Gansevoort RT, Coresh J, Reijneveld SA, Bültmann
U: Socioeconomic measures and CKD in the United States and The
Netherlands. Clin J Am Soc Nephrol 8: 1685–1693, 2013 PubMed
13. Yan G, Cheung AK, Ma JZ, Yu AJ, Greene T, Oliver MN, Yu W,
Norris KC: The associations between race and geographic area and
quality-of-care indicators in patients approaching ESRD. Clin J Am Soc
Nephrol 8: 610–618, 2013 PubMed
Geriatric Nephrology Issues
Many of the relevant topic areas have been
covered by the geriatrics NephSAP issue. Here,
we highlight some more recent studies. We refer the
reader to the geriatrics NephSAP for a more complete
overview.
Physical Function and Frailty
Individuals with CKD have decreased physical
function, which increases the risk of developing
disability. This section reviews some recent studies
related to function.
In a cross-sectional study of 120 patients with
stages 2–5 CKD in Japan with a mean age of 66.5
years, the correlation of eGFR with grip and knee
strength, single–leg stance time, and gait speed was
353
approximately 0.45 for all measures, and these correlations persisted in adjusted models (1).
One concern with using creatinine–based eGFR
measures in studies of physical function is that creatinine itself is a function of muscle mass. Decreases in
muscle mass might mask the association of reduced
kidney function with decline in physical function. Liu
et al. (2) evaluated the association of cystatin C–based
eGFR and creatinine-based eGFR with the development of mobility impairment (inability to walk 0.5 mile
and/or climb a flight of stairs) in 1226 individuals over
age 60 years old participating in the Framingham
Offspring Study. CKD as defined by cystatin C was
associated with the development of mobility impairment (adjusted hazard ratio, 1.55; 95% confidence
interval, 1.05 to 2.31), but CKD defined by creatinine
was not (adjusted hazard ratio, 1.03; 95% confidence
interval, 0.64 to 1.62).
Dalrymple et al. (3) evaluated eGFR by cystatin C
and creatinine with prevalent and incident frailty in
the Cardiovascular Health Study, a community-based
study of adults ages $65 years old. Frailty is a state of
decreased physiologic function. In research studies,
frailty is commonly operationalized as the presence of
three of five criteria: (1) shrinking (unintentional
weight loss $10 lb or 5% in the past year), (2) poor
endurance and energy as assessed by two questions
from the Centers for Epidemiologic Studies Depression
Scale, (3) weakness (grip strength in the lowest 20%
adjusted for sex and body mass index), (4) slowness
(lowest gait speed adjusted for sex and height), and (5)
low physical activity (lowest quintile for each sex). The
prevalence of frailty increased with lower eGFR by
cystatin C and was greater in women than in men.
Individuals with an eGFR,45 ml/min per 1.73 m2 had
a 2-fold higher risk for incident frailty (assessed 4 years
later). In contrast, CKD as assessed by creatinine was
not associated with a higher prevalence or incidence of
frailty.
In the general population, lower physical performance is associated with increased risk of disability,
nursing home placement, and mortality (4). Roshanravan
et al. (5) analyzed the association of physical performance
measures with mortality in individuals with CKD participating in the Seattle Kidney Study and the Baltimore
University of Maryland Study. There were 385 participants with mean eGFR of 41619 ml/min per 1.73 m2.
At baseline, gait speed, timed up and go test, and the
6-minute walk test were 30%–39% lower than predicted.
354
Similar to the findings for prevalence of frailty by
Dalrymple et al. (3), the decrements in physical function
were worse in women. Handgrip strength was not lower
than normal older adult controls. The median follow-up
was 3 years; 13% died. In unadjusted analyses, lower
performance in all four measures was associated with
a greater risk of mortality. However, with adjustment,
handgrip strength was no longer associated with
mortality.
Another way to look at mobility is the life-space
approach that considers mobility in the context of
a person’s environment (6). A person’s life space is the
area through which one routinely and purposefully moves
(Figure 19). Assessment by this concept may allow earlier
detection of problems before decrements in instrumental
activities of daily living or activities of daily living
recognize that chronic conditions may lead to greater
isolation and inactivity. Functional assessment can be
evaluated by a questionnaire using the different levels
multiplied by frequency over a 4-week period (scores
range from 0 to 120) (6). Bowling et al. (7) applied this
concept to individuals with CKD in the University of
Alabama Study of Aging, a population–based longitudinal
study of mobility among community–dwelling older
adults. Bowling et al. (7) evaluated whether kidney
function was associated with the trajectories of life-space
levels over time. Of 390 participants, 49.7% had an
eGFR$60 ml/min per 1.73 m2, 30% had an eGFR
between 45 and 59 ml/min per 1.73 m2, and 20.2% had
an eGFR,45 ml/min per 1.73 m2. At baseline, the mean
life–space scores were lower (more limited) with lower
eGFRs, and those with lower eGFRs had a greater decline
over time. More information is needed on the implications
Figure 19. Functional mobility status by questionnaire of life
space area. Life-space assessment is on the basis of a distribution range, which is divided among six different categories.
Reprinted with permission from Parker M, Baker PS, Allman
RM: A life-space approach to functional assessment of
mobility in the elderly. J Gerontol Soc Work 35: 35–55, 2002.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
of the decline in life space and whether interventions
would improve function and independence.
Cognitive Impairment
In a prior issue of NephSAP, we reviewed studies
that found that lower eGFR and higher albuminuria
were associated with cognitive impairment, with albuminuria being the stronger marker in CKD. Recent
studies have further refined this risk (8–12).
CKD is associated with an increased risk of
cognitive impairment. Potential causes of cognitive
impairment include small vessel vascular disease, higher
levels of inflammatory markers, and other potential risk
factors for neurotoxicity (13). These issues were recently
discussed in a review by Bugnicourt et al. (13). A recent
study lends support to the association of small vessel
vascular disease with cognitive impairment in CKD. Yaffe
et al. (11) evaluated the association of retinopathy with
cognitive impairment in the Chronic Renal Insufficiency
Cohort (CRIC) Study; 30% of participants in the CRIC
Study had retinopathy. Those with retinopathy were older,
were less likely to be white, had higher systolic BP, and
were more likely to have diabetes, a history of coronary
artery disease, stroke, and proteinuria. In unadjusted
analyses, individuals with retinopathy were more likely to
have cognitive impairment. After adjustment, participants
with retinopathy had worse scores in trials A and B of
testing, measures of attention and executive function, and
the Boston Naming Test, a measure of language function.
In addition, the scores on these three tests were worse with
more severe retinopathy.
Davey et al. (9) evaluated the association of
kidney function with longitudinal change in cognitive
function in 590 participants in the Main-Syracuse
Longitudinal Study, a community-based study of cardiovascular risk factors and cognition. Individuals
completed a complete battery of cognitive tests covering visual-spatial organization and memory, scanning,
tracking (including executive function), verbal memory, working memory, and abstract reasoning. Baseline
eGFR was not associated with change in cognitive
function; however, a greater decline in kidney function
(.3 ml/min per 1.73 m2) was associated with a greater
decline in verbal memory and abstract reasoning.
Individuals with diabetes have an increased risk for
cognitive impairment. Barzilay et al. (8) evaluated the
association of albuminuria with cognitive decline in
individuals with diabetes and preserved eGFR in the
Action to Control Cardiovascular Risk in Diabetes
355
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Memory in Diabetes Follow-Up Study (ACCORD MIND).
The ACCORD MIND was an ancillary study that
aimed to evaluate whether the interventions studied in
the Action to Control Cardiovascular Risk in Diabetes
Study slowed the deterioration of cognitive function over
40 months of follow-up. The primary test of cognitive
function was the Digit Symbol Substitution Test (measure
of psychomotor speed and performance); other cognitive
tests were the Rey Auditory Verbal Learning Test (verbal
memory) and the modified Stroop test (executive function). Albuminuria was measured at baseline and three
follow–up visits. Individuals with albumin to creatinine
ratios (ACRs) ,30 mg/g on all visits were classified as no
albuminuria (n¼1689), individuals with an ACR$30 mg/g
on all three visits were classified as persistent albuminuria (n¼469), individuals without albuminuria at baseline
but with an ACR.30 mg/g on one follow–up measurement were classified as progressors (n¼429), and individuals who had albuminuria at baseline but not at the
follow-up visits were classified as remitters (n¼370).
The mean eGFR in all of the groups was approximately
90 ml/min per 1.73 m2. After adjustment, both persistent
albuminuria and progression were associated with a $5%
decline in the Dantes Subject Standardized Test. None of
the albuminuria groups were associated to a .5% decline
in verbal memory or executive function.
Palliative Care
Individuals .65 years of age have accounted for
the largest proportion of the incident ESRD population
recently, and decisions regarding whether to start dialysis
or continue therapy are being discussed with greater
frequency. Having an understanding about the outcomes
of older patients on dialysis with multiple comorbidities
and the perceptions of individuals on dialysis are key for
practitioners to help guide decision making.
Tonge et al. (14) performed a systematic review
of 26 studies involving 711 patients to identify common
themes of attitudes toward dialysis therapy. The majority
of patients studied were on hemodialysis (n¼544), and
a small proportion of patients were treated with conservative management (n¼86). Common themes of pervasive suffering (loss of freedom and independence),
personal vulnerability (imminence of death and medical
abandonment), relational responsibility (burden to family),
and negotiating existential tensions (preparing for death)
were noted among patients. Patients were experiencing
both physical and psychosocial stress, but because of
concerns of upsetting their families or their physicians,
they did not pursue palliative care. Caregivers often
would like treatment to focus on relief of pain but were
unsure how to go about discontinuation of dialysis
support.
This highlights that palliative care options need
to be part of routine CKD management. Additional
support for palliative care comes from data that show
that older patients with CKD and other comorbidities do
not have improvement in morbidity or mortality from
dialysis compared with those treated conservatively.
The review by Luckett et al. (15) raised the importance
of advance care planning for older individuals on
dialysis who may become unwell and unable to speak
for themselves. Because cognitive decline is an issue
among older patients on dialysis, the need for providers
and family members to determine whether to continue
dialysis treatments is becoming an increasing and
regular issue. The review highlighted different interventions to achieve better advance care planning, including
involvement of families in decision making, ensuring
that the patient’s wishes will be carried out, education
for the patient and family regarding conservative therapy, and separating decisions about RRT from other
life–sustaining measures.
One of the limitations to offering appropriate end
of life care counseling is the lack of comfort among
providers. Combs et al. (16) surveyed 104 of the
nephrology fellowship programs in the United States
in 2013 regarding the perceptions of end-of-life care.
Compared with a similar survey of fellowship programs,
there was an increase in the sense of importance of the
ability to provide end-of-life care (95% in 2013 versus
54% in 2003). Fellows surveyed felt that training in this
area was still lacking and that they were unprepared to
provide end-of-life care counseling.
References
1. Hiraki K, Yasuda T, Hotta C, Izawa KP, Morio Y, Watanabe S,
Sakurada T, Shibagaki Y, Kimura K: Decreased physical function in
pre-dialysis patients with chronic kidney disease. Clin Exp Nephrol 17:
225–231, 2013 PubMed
2. Liu CK, Lyass A, Massaro JM, D’Agostino RB Sr., Fox CS, Murabito
JM: Chronic kidney disease defined by cystatin C predicts mobility
disability and changes in gait speed: The Framingham Offspring Study.
J Gerontol A Biol Sci Med Sci 69: 301–307, 2014 PubMed
3. Dalrymple LS, Katz R, Rifkin DE, Siscovick D, Newman AB, Fried LF,
Sarnak MJ, Odden MC, Shlipak MG: Kidney function and prevalent and
incident frailty. Clin J Am Soc Nephrol 8: 2091–2099, 2013 PubMed
4. Guralnik JM, Simonsick EM, Ferrucci L, Glynn RJ, Berkman LF,
Blazer DG, Scherr PA, Wallace RB: A short physical performance
battery assessing lower extremity function: Association with selfreported disability and prediction of mortality and nursing home
admission. J Gerontol 49: M85–M94, 1994 PubMed
356
5. Roshanravan B, Robinson-Cohen C, Patel KV, Ayers E, Littman AJ, de
Boer IH, Ikizler TA, Himmelfarb J, Katzel LI, Kestenbaum B, Seliger S:
Association between physical performance and all-cause mortality in
CKD. J Am Soc Nephrol 24: 822–830, 2013 PubMed
6. Parker M, Baker PS, Allman RM: A life-space approach to functional
assessment of mobility in the elderly. J Gerontol Soc Work 35: 35–55, 2002
7. Bowling CB, Muntner P, Sawyer P, Sanders PW, Kutner N, Kennedy R,
Allman RM: Community mobility among older adults with reduced kidney
function: A study of life-space. Am J Kidney Dis 63: 429–436, 2014 PubMed
8. Barzilay JI, Lovato JF, Murray AM, Williamson J, Ismail-Beigi F, Karl
D, Papademetriou V, Launer LJ: Albuminuria and cognitive decline in
people with diabetes and normal renal function. Clin J Am Soc Nephrol
8: 1907–1914, 2013 PubMed
9. Davey A, Elias MF, Robbins MA, Seliger SL, Dore GA: Decline in
renal functioning is associated with longitudinal decline in global
cognitive functioning, abstract reasoning and verbal memory. Nephrol
Dial Transplant 28: 1810–1819, 2013 PubMed
10. Seidel UK, Gronewold J, Volsek M, Todica O, Kribben A, Bruck H,
Hermann DM: The prevalence, severity, and association with HbA1c
and fibrinogen of cognitive impairment in chronic kidney disease.
Kidney Int 85: 693–702, 2014 PubMed
11. Yaffe K, Ackerson L, Hoang TD, Go AS, Maguire MG, Ying GS,
Daniel E, Bazzano LA, Coleman M, Cohen DL, Kusek JW, Ojo A,
Seliger S, Xie D, Grunwald JE; CRIC Study Investigators: Retinopathy
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
12.
13.
14.
15.
16.
and cognitive impairment in adults with CKD. Am J Kidney Dis 61:
219–227, 2013 PubMed
Yaffe K, Kurella-Tamura M, Ackerson L, Hoang TD, Anderson AH,
Duckworth M, Go AS, Krousel-Wood M, Kusek JW, Lash JP, Ojo A,
Robinson N, Sehgal AR, Sondheimer JH, Steigerwalt S, Townsend RR;
CRIC Study Investigators: Higher levels of cystatin C are associated
with worse cognitive function in older adults with chronic kidney
disease: The chronic renal insufficiency cohort cognitive study. J Am
Geriatr Soc 62: 1623–1629, 2014 PubMed
Bugnicourt JM, Godefroy O, Chillon JM, Choukroun G, Massy ZA:
Cognitive disorders and dementia in CKD: The neglected kidney-brain
axis. J Am Soc Nephrol 24: 353–363, 2013 PubMed
Tong A, Cheung KL, Nair SS, Kurella Tamura M, Craig JC,
Winkelmayer WC: Thematic synthesis of qualitative studies on patient
and caregiver perspectives on end-of-life care in CKD. Am J Kidney Dis
63: 913–927, 2014 PubMed
Luckett T, Sellars M, Tieman J, Pollock CA, Silvester W, Butow PN,
Detering KM, Brennan F, Clayton JM: Advance care planning for adults
with CKD: A systematic integrative review. Am J Kidney Dis 63: 761–
770, 2014 PubMed
Combs SA, Culp S, Matlock DD, Kutner JS, Holley JL, Moss AH:
Update on end-of-life care training during nephrology fellowship: A
cross-sectional national survey of fellows. Am J Kidney Dis 65: 233–
239, 2015 PubMed
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Chronic Kidney Disease and Progression
Claiming Credits and Evaluation Process
Accreditation Statement
The American Society of Nephrology (ASN) is accredited by the Accreditation Council for Continuing Medical Education to
provide continuing medical education for physicians.
AMA Credit Designation Statement
The ASN designates this enduring material for a maximum of 8.0 AMA PRA Category 1 Credits™. Physicians should claim
only the credit commensurate with the extent of their participation in the activity.
Original Release Date: October 2015
CME Credit Termination Date: September 30, 2017
Examination Available Online: On or before Thursday, October 15, 2015
Estimated Time for Completion: 8 hours
Answers with Explanations
Provided with a passing score after the first and/or after the second attempt
October 2017: posted on the ASN website when the issue is archived.
•
•
Method of Participation
Read the syllabus that is supplemented by original articles in the reference lists.
Complete the online self-assessment examination.
Each participant is allowed two attempts to pass the examination (.75% correct) for CME credit.
Upon completion, review your score and incorrect answers and print your certificate.
Answers and explanations are provided with a passing score or after the second attempt.
•
•
•
•
•
Activity Evaluation and CME Credit Instructions
Go to www.asn-online.org/cme, and enter your ASN login on the right.
Click the ASN CME Center.
Locate the activity name and click the corresponding ENTER ACTIVITY button.
Read all front matter information.
On the left-hand side, click and complete the Demographics & General Evaluations.
Complete and pass the examination for CME credit.
Upon completion, click Claim Your Credits, check the Attestation Statement box, and enter the number of CME
credits commensurate with the extent of your participation in the activity.
If you need a certificate, Print Your Certificate on the left.
•
•
•
•
•
•
•
•
For your complete ASN transcript, click the ASN CME Center banner, and click View/Print Transcript on the left.
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 enrolled in the ABIM–MOC program
Enroll for MOC via the ABIM website (www.abim.org).
Enter your (ABIM) Candidate Number and Date of Birth prior to completing the examination.
Take the self-assessment examination within the timeframe specified in this issue of NephSAP.
Below your score select “Click here to post to ABIM.”
•
•
•
•
•
MOC points will be applied to only those ABIM candidates who have enrolled in the MOC program. It is your responsibility to
complete the ABIM MOC enrollment process.
358
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
NephSAP, Volume 14, Number 4, October 2015—Chronic Kidney Disease and Progression
3. You are seeing a 65-year-old woman in follow-up
for advanced kidney disease. Her disease has
progressed slowly. Her weight has been stable,
and her appetite is preserved. Medications are
lisinopril, amlodipine, furosemide, metoprolol,
calcitriol, darbepoetin, and atorvastatin. Her BP
is 139/85 mmHg, and her pulse is 65 per minute.
She has no edema. Laboratory studies show sodium
139 mEq/L, potassium 4.2 mEq/L, total CO2
23 mmol/L, BUN 65 mg/dl, SCr 4.5 mg/dl (eGFR
10 ml/min per 1.73 m2), calcium 9.5 mg/dl,
phosphorus 4.5 mg/dl, parathyroid hormone
(PTH) 115 pg/ml (normal 10–65 pg/ml) unchanged from 3 months ago, and Hb 10.0 g/dl.
Which ONE of the following is the MOST
appropriate management?
A. Discontinue lisinopril
B. Increase darbepoetin
C. Increase calcitriol
D. No change in management
1. A 51-year-old woman with stage 4 CKD secondary
to polycystic kidney disease is evaluated during
a routine clinic visit. She has hypertension that is
controlled on lisinopril and amlodipine. She does
not smoke. The physical examination is notable
for a body mass index (BMI) of 32 kg/m2, a BP of
132/70 mmHg, and enlarged kidneys. The remainder of the examination is normal. Laboratory
studies show normal serum electrolytes, eGFR
29 ml/min per 1.73 m2, hemoglobin (Hb) 12 g/dl,
total cholesterol 186 mg/dl, HDL-cholesterol
35 mg/dl, LDL-cholesterol 95 mg/dl, and triglycerides 300 mg/dl.
Which ONE of the following is the MOST
appropriate management?
A. Start fenofibrate
B. Start pravastatin
C. No change in management
D. You should use a risk calculator to determine whether she should receive pharmacologic treatment for her lipid values
4. You are asked to evaluate a 65-year-old man with
diabetes mellitus in consultation for management
of CKD. His medications are losartan 100 mg
daily, lisinopril 20 mg daily, sodium bicarbonate
650 mg two times per day, and furosemide 40 mg
daily. His BP is 145/80 mmHg. He has trace edema.
Laboratory studies show potassium 4.9 mEq/L,
total CO2 23 mmol/L, and SCr 2.5 mg/dl.
Continuation of the current medication
regimen places him at increased risk for which
ONE of the following complications?
A. Congestive heart failure
B. Mortality
C. AKI
D. ESRD
2. A 65-year-old man with type 2 diabetes mellitus
and stage 3b CKD is evaluated in follow-up. His
medications include losartan 100 mg daily,
furosemide 20 mg daily, simvastatin 20 mg
daily, metoprolol succinate 50 mg daily, amlodipine 5 mg daily, and aspirin 81 mg daily. His
BP is 129/79 mmHg. The remainder of the
examination is normal. Laboratory studies show
sodium 138 mEq/L, potassium 4.9 mEq/L, total
CO2 22 mmol/L, BUN 40 mg/dl, serum creatinine
(SCr) 1.9 mg/dl (eGFR 42 ml/min per 1.73 m2),
LDL-cholesterol 105 mg/dl, and HDL-cholesterol
35 mg/dl. The urine albumin-to-creatinine ratio
(UACR) is 800 mg/g.
Which ONE of the following is the MOST
appropriate management to delay progression
of CKD?
A. Add lisinopril
B. Double the losartan dose
C. Add ezetimibe
D. Double the furosemide dose
E. No change in management
5. A 45-year-old man with stage 4 CKD secondary
to IgA nephropathy is evaluated during a routine
clinic visit. He smokes one pack of cigarettes daily.
He has hypertension but does not have diabetes.
There is no family history of kidney disease or heart
disease. His medications include valsartan, fish oil,
amlodipine, and furosemide. The physical examination is notable for a BP of 140/70 mmHg. The
359
360
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
remainder of the examination is normal. Laboratory
studies show eGFR 29 ml/min per 1.73 m2, HDLcholesterol 35 mg/dl, LDL-cholesterol 125 mg/dl,
and triglycerides 300 mg/dl.
According to the most recent Kidney Disease
Improving Global Outcomes lipid guidelines
(November of 2013), which ONE of the
following is the recommended treatment
strategy?
A. He should start a fibrate
B. He should start a statin
C. He should start a bile acid sequestrant
D. You should use a risk calculator to determine whether he should be treated
6. Which ONE of the following regarding blockade
of the renin-angiotensin-aldosterone system is
correct?
A. Combination angiotensin-converting enzyme
inhibitors (ACEI) and angiotensin receptor
blocker therapy reduces proteinuria more
than the addition of spironolactone to an
ACEI
B. Combination of spironolactone and lisinopril
has a lower risk of hyperkalemia than
losartan and lisinopril
C. Because combination ACEI and angiotensin receptor blocker therapy decreases
proteinuria, it will decrease progression
of kidney disease (doubling SCr, ESRD, or
death)
D. Combination therapy is associated with
an increased risk of mortality in heart
failure
E. A high normal serum potassium level
(4.5–4.9 mEq/L) is a risk factor for hyperkalemia on renin-angiotensin blockade
7. You are seeing a 75-year-old man with type 2
diabetes and hypertension. His current medications
include metformin and glyburide. His wife reports
that he recently had an episode of severe hypoglycemia requiring the intervention of emergency
services. His BP is 140/70 mmHg. The physical
examination is otherwise normal. His SCr is
0.8 mg/dl (Chronic Kidney Disease Epidemiology
Collaboration [CKD-EPI] eGFR¼87 ml/min per
1.73 m2). The UACR is 200 mg/g. The HbA1c is
7.2%.
Which ONE of the following places him at
increased risk for serious hypoglycemia?
A. His age
B. His clearance of glyburide
C. His level of albuminuria
D. His use of metformin
8. A 17-year-old young man with CKD caused by
FSGS is evaluated in follow-up. His current
medications include lisinopril (40 mg daily),
bumetanide (1 mg two times per day), clonidine
(0.1 mg three times per day), iron sulfate (325 mg
two times per day), sodium bicarbonate (650 mg
three times per day), and tacrolimus (2 mg two
times per day). He reports difficulty taking all of
his medications, and he misses doses on a regular
basis.
Which ONE of the following would be the
MOST effective intervention to improve
medication adherence for this young man?
A. Change three times per day–dosed medications to two times per day
B. Decrease the number of classes of medications taken per day
C. Discontinue iron sulfate
D. Provision of an educational intervention
9. A 65-year-old woman with type 2 diabetes
mellitus is evaluated for ongoing follow-up of
advanced CKD. She has recently moved to the
area to be near her daughter and grandchildren.
She has coronary artery disease, hypertension,
and atrial fibrillation. She does not have symptoms of uremia and remains active. Her medications are losartan 100 mg daily, diltiazem
(extended release formulation) 120 mg daily,
atorvastatin 20 mg daily, warfarin 4 mg daily,
bumetanide 1 mg two times per day, glipizide
5 mg two times per day, and sitagliptin 50 mg
one time per day. Her BP is 130/70 mmHg,
and her pulse is irregularly irregular, with an
average rate of 85 per minute. There is trace lower
extremity edema. Laboratory studies show sodium
139 mEq/L, potassium 4.2 mEq/L, chloride
102 mEq/L, total CO2 23 mmol/L, BUN 60 mg/dl,
SCr 5.0 mg/dl (Modification of Diet in Renal
Disease eGFR 9 ml/min per 1.73 m2), international normalized ratio of 2.0, and HbA1c of
7.2%. The UACR is 350 mg/g.
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Which ONE of the following is the MOST
appropriate treatment intervention for this
patient?
A. Change glipizide to glyburide
B. Decrease the dose of sitagliptin
C. Change warfarin to aspirin
D. Discontinue the losartan
10. A 55-year-old woman with stage 3b CKD caused
by autosomal dominant polycystic kidney disease
is hospitalized for evaluation of right–sided chest
pain and shortness of breath. On physical examination, she is in mild distress from the chest
discomfort. Her respirations are nonlabored with
a rate of 16 per minute. The oxygen saturation on
ambient air is 96%. The BP is 140/80 mmHg, and
the pulse is 106 per minute. The chest examination shows tenderness on palpation of the right
anterior chest wall. The lung fields show faint
end–expiratory wheezes bilaterally. There is no
leg edema or calf tenderness. Her eGFR is stable at
29 ml/min per 1.73 m2. An arterial blood gas on
ambient air shows pH 7.44, PaCO2 of 38 mmHg,
and PaO2 of 88 mmHg. The D-dimer level is
0.7 mg/ml (reference range ,0.5 mg/ml). A chest
radiograph is normal. The Wells score for pulmonary embolism (PE) is 1, consistent with a low
probability for PE.
Which ONE of the following regarding the
interpretation of this woman’s D-dimer level is
correct?
A. The D-dimer level is diagnostic of a thrombus (deep venous thrombosis or pulmonary
embolus)
B. The elevated D-dimer level is atypical of
patients with CKD
C. The specificity of D-dimer is lower in CKD
D. The positive predictive value of this woman’s D-dimer level is equivalent to that of
persons with a normal GFR
11. A 60-year-old man with stage 3b CKD secondary
to membranous glomerulopathy is hospitalized for
evaluation of atypical chest pain. He noted onset
of intermittent sharp left–sided chest pain that was
not consistently associated with exertion on the
day of admission. On physical examination, he is
in mild respiratory distress, with a respiratory rate
of 18 per minute. The BP is 150/84 mmHg, and
the pulse is regular, with a rate of 84 per minute.
361
The jugular venous pressure is 10 cm H2O. The
lungs show basilar crackles. There is 21 leg
edema. His eGFR is 35 ml/min per 1.73 m2. The
highly sensitive serum troponin T level on admission is 0.06 mg/L (reference value #0.014 mg/L),
and a follow-up level the following morning is
unchanged. The electrocardiogram shows a right
bundle branch block and no ST-T wave changes,
and it is unchanged from his electrocardiogram
performed 7 months ago.
Which ONE of the following is correct
regarding the significance of the elevated
serum troponin T levels in this patient?
A. The increased highly sensitive serum troponin T level on admission is highly specific
for acute myocardial infarction (MI)
B. The lack of a dynamic change in the serum
troponin level excludes acute MI
C. The increased levels are most strongly
associated with left ventricular hypertrophy on echocardiography
D. The increased levels are not of clinical
significance at his level of GFR
12. A 65-year-old man with stage 3 CKD is admitted
for management of acute coronary syndrome. After
a rapid prehydration protocol to decrease the risk
of contrast nephropathy, he undergoes coronary
angiography. This reveals a 90% stenosis in the
left circumflex artery.
Which ONE of the following interventions
has the greatest likelihood of decreasing the
need for repeat coronary interventions at
1 year?
A. Coronary angioplasty with placement of
a bare metal stent
B. Coronary angioplasty with placement of
a drug-eluting stent
C. Coronary angioplasty without placement
of a stent
D. Coronary angioplasty without a stent plus
clopidogrel
13. A 70-year-old woman with stage 5 CKD is diagnosed with new–onset atrial fibrillation. She is
preparing for but has not yet started dialysis. She
has hypertension and type 2 diabetes mellitus.
Her CHA2DS2-VASC score is 4. Her eGFR is
14 ml/min per 1.73 m2. Her heart rate is controlled
with diltiazem.
362
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
Which ONE of the following is the MOST
appropriate treatment?
A. Aspirin 325 mg daily
B. Warfarin to a target international normalized ratio of 2–3
C. Rivaroxaban 20 mg daily
D. Clopidogrel 75 mg daily
E. No additional medication
14. A 70-year-old woman with CKD secondary to
diabetes mellitus is evaluated in follow-up. She has
a history of coronary artery disease and hypertension. She is concerned because her laboratory
reports show that her HbA1c is above the reference range. Her current medications for diabetes
include linagliptin (5 mg daily) and insulin glargine
(25 units at bedtime). Laboratory values show an
SCr of 1.6 (eGFR 37 ml/min per 1.73 m2), a UACR
of 100 mg/g, and a HbA1c of 7.2%.
Which ONE of the following is the MOST
appropriate management?
A. Add glipizide
B. Increase insulin glargine to 30 units daily
C. Add insulin aspartate before meals
D. Increase linagliptin to 10 mg daily
E. Make no changes to the current regimen
15. A 67-year-old man has an eGFR of 25 ml/min per
1.73 m2 and a BMI of 32 kg/m2. He has hypertension and type 2 diabetes mellitus. His BP is
130/80 mmHg on losartan 100 mg daily, furosemide 40 mg daily, and metoprolol succinate 100 mg
daily. The UACR is 400 mg/g. He has been
advised to lose weight in preparation for transplantation. He asks about the effect of obesity on
CKD progression.
Which ONE of the following statements is
MOST accurate regarding CKD progression
and obesity in this patient?
A. Lowering the BMI to 20–25 kg/m2 would
decrease his CKD progression
B. This patient’s BMI is not associated with
faster progression to dialysis compared with
other BMI categories
C. The patient’s BMI is associated with the
highest rate of CKD progression
D. The patient should undergo bariatric surgery to decrease CKD progression
16. A 55-year-old woman with diabetes mellitus is
referred for evaluation of CKD. Her eGFR has
ranged from 48 to 53 ml/min per 1.73 m2, and the
UACR has been persistently elevated in the 40 to
80 mg/g range over a period of 6 months.
The LDL-cholesterol is 136 mg/dl. Therapy with
losartan was initiated 6 months ago, and subsequent home BP monitoring shows average
BPs of approximately 132/78 mmHg. On physical
examination, her BP is 138/80 mmHg. The
remainder of the examination is normal.
According to the American College of
Physicians guidelines for CKD screening and
management, which ONE of the following is
the MOST appropriate management?
A. Serial measurements of UACR
B. Initiate statin therapy
C. Add a thiazide diuretic
D. Begin a low-protein diet
E. No change in management
17. Which ONE of the following is correct regarding the effect of dietary sodium restriction
and CKD progression on the basis of recent
prospective trials and studies?
A. It slowed CKD progression
B. It hastened CKD progression
C. It increased serum renin and aldosterone
levels
D. It had no effect on BP
18. A 64-year-old woman with stage 4 CKD is evaluated in follow-up. Medications are low-dose
aspirin, pravastatin, metoprolol, and bumetanide.
Her eGFR is 29 ml/min per 1.73 m2, and it
decreased from 32 ml/min per 1.73 m2 last year.
Her physical examination shows a BP of 138/88
mmHg. She has no edema. Other laboratory studies
show serum potassium of 4.4 mEq/L, total CO2
of 18 mmol/L, and a normal anion gap. She has
not tolerated sodium bicarbonate or sodium citrate
because of gastrointestinal side effects.
Which ONE of the following is the MOST
likely to correct this patient’s metabolic
acidosis?
A. An endothelin antagonist
B. A diet enriched in fruits and vegetables
C. Lisinopril
D. Spironolactone
19. A 47-year-old African-American man with stage
4 CKD caused by diabetic kidney disease is
evaluated in follow-up. He has a family history
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
of diabetes, hypertension, and CKD. The BP is
134/82 mmHg, and there is no edema. Laboratory
tests reveal an eGFR of 17 ml/min per 1.73 m2,
and it decreased from 21 ml/min per 1.73 m2 last
year. He recently read a report about APOL1 and
the risk of CKD on the internet and asks for more
information about the effect of the risk allele on
CKD progression.
Which ONE of the following would you tell
him regarding APOL1 risk status and CKD?
A. APOL1 risk status does not affect the
risk of CKD progression in patients with
diabetes
B. APOL1 high–risk compared with low–risk
status is associated with a 4-fold increase
in the composite risk of ESRD plus doubling of SCr
C. The majority of individuals with two APOL1
risk alleles will develop CKD
D. Individuals with two APOL1 risk alleles
are at risk for developing CKD
20. A 48-year-old man with type 2 diabetes mellitus
and stage 3a CKD is evaluated for intermittent
right upper quadrant abdominal pain. An abdominal ultrasound shows fatty infiltration of the
liver and a single stage I Bosniak right–sided kidney cyst. He does not smoke and has never consumed excess alcohol. On physical examination,
the BP is 130/78 mmHg, and there is no edema.
The BMI is 26.9 kg/m2. He has a waist-to-hip
ratio of 0.8.
Which ONE of the following findings in this
patient has been shown to be associated with
an increased incidence of CKD?
A. His waist-to-hip ratio
B. Nonalcoholic fatty liver disease
C. His BMI
D. The renal cyst
21. A 22-year-old man is evaluated in the emergency
department for a several week history of progressive fatigue. He had recently moved from
Costa Rica, where he was an agricultural worker
in a sugar cane field, frequently working in a hot
and humid environment. He does not take prescribed or over the counter medications and has
not used illicit drugs. There is no family history
of kidney disease. His BP is 145/78 mmHg, and
his pulse is 76 per minute. The remainder of the
363
examination is normal. His laboratory studies
show an SCr of 1.8 mg/dl. The urine protein-tocreatinine ratio is 500 mg/g.
Which ONE of the following would you MOST
likely expect to find in this patient?
A. Hypokalemia
B. Hypertriglyceridemia
C. Hypouricemia
D. Hematuria
22. You are asked to participate in a focus group to
develop a clinical trial that studies the efficacy of
screening for CKD. Information is given to you
about current United States position statements
as well as worldwide screening and intervention
programs.
Which ONE of the following statements is
correct regarding CKD epidemiology and
CKD screening/intervention programs?
A. Death attributed to CKD worldwide has
remained relatively constant for the past 2
decades
B. Diabetes is no longer the leading cause of
ESRD worldwide
C. Implementation of a CKD screening and
intervention program in England has been
associated with an increase in planned
initiation of dialysis
D. It is projected that ESRD incidence in
developing countries will decrease relative
to developed countries
23. A 56-year-old man has type 2 diabetes, stage 3b
CKD, hypertension, and hyperlipidemia. His medications are valsartan, chlorthalidone, metformin,
atorvastatin, and low-dose aspirin. He has claudication and was recently prescribed pentoxifylline
by his primary care provider, because cilostazol
caused diarrhea. His BP is 130/76 mmHg, and he
has no edema. His eGFR is 39 ml/min per 1.73 m2
compared with an eGFR of 43 ml/min per 1.73 m2
3 years ago. The UACR is 280 mg/g. Other
laboratory studies are HbA1c 7%, uric acid
7.4 mg/dl, calcium 9.5 mg/dl, phosphorous
4.5 mg/dl, intact PTH 50 pg/ml, total cholesterol
190 mg/dl, and LDL-cholesterol 90 mg/dl.
Which ONE of the following is the MOST
appropriate management?
A. Discontinue metformin and start sitagliptin
B. Discontinue pentoxifylline
364
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
C. Discontinue atorvastatin
D. Start paricalcitol
E. No change in management
24. A 57-year-old man with type 2 diabetes mellitus
is referred for evaluation and management of
stage 4 CKD. He has hypertension and hyperlipidemia. He is on glipizide 2.5 mg daily, furosemide 40 mg daily, linagliptin, enalapril,
amlodipine, metoprolol tartrate, and atorvastatin.
He does not have chest pain, shortness of breath,
or joint pain. On physical examination, the BP is
138/78 mmHg, and the pulse is 62 per minute.
The jugular venous pressure is 10 cm, and there is
11 leg edema. Laboratory studies show an eGFR
of 19 ml/min per 1.73 m2, which is decreased
from 24 ml/min per 1.73 m2 last year. The
potassium is 5.6 mEq/L, the total CO2 is 24 mEq/L,
the uric acid is 7 mg/dl, the Hb is 10.6 g/dl, the
HbA1c is 7%, and the UACR is 220 mg/g.
Which ONE of the following is the MOST
appropriate management?
A. Discontinue enalapril
B. Increase the dose of glipizide
C. Increase the dose of furosemide
D. Start sodium bicarbonate
25. A 53-year-old man is seen for ongoing management of hypertension. His BP is 144/88 mmHg.
The BMI is 24 kg/m2. The remainder of the
physical examination is normal. The eGFR by the
CKD-EPI creatinine equation is 59 ml/min per
1.73 m2. His UACR is 56 mg/g.
Which ONE of the following statements is
MOST accurate regarding risk prediction by
various eGFR estimating methods for this
patient?
A. Cystatin C–based eGFR reclassification to
a category with lower eGFR is associated
with an increased risk of death
B. Cystatin C–based eGFR reclassification to
a category with lower eGFR improves risk
prediction for ESRD but not mortality
C. Associations of albuminuria .30 mg/g and
hypertension with lower measured GFR are
best approximated by CKD-EPI cystatin
C–based but not creatinine–based eGFR
estimations
D. Measured GFR is accurately predicted by
the CKD-EPI creatinine equation
26. A 51-year-old man returns for follow-up after an
initial evaluation of stage 3 CKD one year ago. His
initial evaluation was significant for the presence
of moderate hypertensive retinopathy and left
ventricular hypertrophy. Three months ago, he
suffered an MI and underwent percutaneous
coronary intervention requiring the placement of
two drug–eluting stents. His eGFR decreased
from 42 to 38 ml/min per 1.73 m2 after the
procedure. On physical examination, the BP is
150/50 mmHg. Cardiac auscultation reveals
an S4 gallop. Funduscopic examination shows
unchanged moderate hypertensive retinopathy
with cotton wool spots and hard exudates bilaterally. The remainder of the examination is
normal.
Which ONE of the following clinical findings
in this patient has been found to be MOST
strongly associated with risk for progression to
ESRD?
A. Hypertensive retinopathy
B. Recent MI
C. Increased pulse pressure
D. Left ventricular hypertrophy
27. Which ONE of the following is correct about
CKD progression in African Americans?
A. Higher 24-hour ambulatory systolic BP levels
are not associated with increased risk of
progression
B. CKD progresses more rapidly but is not
associated with a higher level of mortality
compared with white individuals
C. The presence of two APOL1 alleles has
a greater role in the progression of diabetic
compared with nondiabetic CKD
D. The degree of albuminuria has little effect
on the risk of CKD progression
28. Which ONE of the following outcomes has
been shown in recent trials of individuals
treated in CKD clinics that use physician
extenders?
A. A decreased rate in the decline of GFR
B. No difference in the use of erythropoiesisstimulating agents
C. No difference in the use of phosphate
binders
D. A decreased rate of initiation of dialysis
Nephrology Self-Assessment Program - Vol 14, No 4, October 2015
29. Which ONE of the following is the MOST likely
reason that older adults with stage 5 CKD
are often not provided or offered conservative
management?
A. Lack of provider comfort or training in
discussing end-of-life options
B. Providers’ unwillingness to have discussions
regarding end-of-life care
C. Improved outcomes for older individuals
on dialysis
D. Frequent use of limited trials of dialysis
30. A clinic associated with a local hospital laboratory is about to incorporate a new electronic
medical record with automatic eGFR reporting. A recent quality improvement project showed
that an excessive number of patients were not
365
meeting targets for Hb and intact PTH. Stakeholders expect automatic eGFR reporting to improve the number of patients in the target range for
these laboratory parameters. You are asked to
provide an opinion of the effects of automatic
eGFR reporting as an initial intervention.
Which ONE of the following is an expected
outcome from implementation of automated
eGFR reporting?
A. No effect on the decision to initiate dialysis
B. Increased recognition and documentation
of CKD
C. Increased number of patients with Hb and
intact PTH levels in the target range
D. An improvement in the timely placement
of dialysis access
Download