OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF NEPHROLOGY
VOLUME 7 | ISSUE 1 | JULY 2017
www.kisupplements.org
KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis,
Evaluation, Prevention, and Treatment of Chronic Kidney
Disease–Mineral and Bone Disorder (CKD-MBD)
KISU_v7_i1_COVER.indd 1KISU_v7_i1_COVER.indd 1 31-05-2017 13:23:0531-05-2017 13:23:05
KDIGO 2017 CLINICAL PRACTICE GUIDELINE UPDATE
FOR THE DIAGNOSIS, EVALUATION, PREVENTION, AND
TREATMENT OF CHRONIC KIDNEY DISEASEMINERAL AND
BONE DISORDER (CKD-MBD)
Kidney International Supplements (2017) 7, 1–59 1
KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis,
Evaluation, Prevention, and Treatment of Chronic Kidney DiseaseMineral
and Bone Disorder (CKD-MBD)
3 Tables and supplementary material
6 KDIGO Executive Committee
7 Reference keys
8 CKD nomenclature
9 Conversion factors
10 Abbreviations and acronyms
11 Notice
12 Foreword
13 Work Group membership
14 Abstract
15 Summary of KDIGO CKD-MBD recommendations
19 Summary and comparison of 2017 updated and 2009 KDIGO CKD-MBD
recommendations
22 Chapter 3.2: Diagnosis of CKD-MBD: bone
25 Chapter 4.1: Treatment of CKD-MBD targeted at lowering high serum
phosphate and maintaining serum calcium
33 Chapter 4.2: Treatment of abnormal PTH levels in CKD-MBD
38 Chapter 4.3: Treatment of bone with bisphosphonates, other osteoporosis
medications, and growth hormone
39 Chapter 5: Evaluation and treatment of kidney transplant bone disease
41 Methodological approach to the 2017 KDIGO CKD-MBD guideline update
49 Biographic and disclosure information
55 Acknowledgments
56 References
www.kisupplements.org contents
VOL 7 | ISSUE 1 | JULY 2017
2 Kidney International Supplements (2017) 7, 1–59
TABLES
24 Table 1. Utility of KDOQI and KDIGO PTH thresholds for diagnostic decision making
42 Table 2. Research questions
45 Table 3. Question-specic eligibility criteria
46 Table 4. GRADE system for grading quality of evidence for an outcome
47 Table 5. Final grade for overall quality of evidence
47 Table 6. Balance of benets and harms
47 Table 7. Implications of the strength of a recommendation
47 Table 8. Determinants of strength of recommendation
SUPPLEMENTARY MATERIAL
Appendix A. PubMed search strategy
Appendix B. Summary of search and review process
Table S1. Summary table of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5: study characteristics
Table S2. Summary table of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5: study population characteristics
Table S3. Summary table of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5: results
Table S4. Summary table of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5: quality
Table S5. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5
Table S6. Evidence prole of randomized controlled trials examining the treatment of CKD-MBD with
bisphosphonates in CKD G3aG5
Table S7. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5: study characteristics
Table S8. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5: study population characteristics
Table S9. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5: results
Table S10. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5: quality
Table S11. Evidence matrix of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5
Table S12. Evidence prole of studies evaluating the ability of bone mineral density results to predict fracture or
renal osteodystrophy among patients with CKD G3aG5
Table S13. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D: study characteristics
Table S14. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D: study population characteristics
Table S15. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D: results
Table S16. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D: quality
Table S17. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D
www.kisupplements.org contents
Kidney International Supplements (2017) 7, 1–59 3
Table S18. Evidence prole of randomized controlled trials examining the treatment of CKD-MBD with varying
dialysate calcium concentration levels in CKD G5D
Table S19. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders: study characteristics
Table S20. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders: study population characteristics
Table S21. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders: results
Table S22. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders: quality
Table S23. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders
Table S24. Evidence prole of randomized controlled trials examining the treatment of CKD-MBD with calcium-
containing phosphate binders versus calcium-free phosphate binders
Table S25. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate: study characteristics
Table S26. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate: study population characteristics
Table S27. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate: results
Table S28. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate: quality
Table S29. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate
Table S30. Evidence prole of randomized controlled trials examining the treatment of CKD-MBD with dietary
phosphate
Table S31. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: study
characteristics
Table S32. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: study
population characteristics
Table S33. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: results
Table S34. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: quality
Table S35. Evidence matrix of randomized controlled trials examining the treatment of PTH in CKD-MBD
Table S36. Evidence prole of randomized controlled trials examining the treatment of PTH in CKD-MBD
Table S37. Summary table of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis: study characteristics
Table S38. Summary table of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis: study population characteristics
Table S39. Summary table of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis: results
Table S40. Summary table of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis: quality
Table S41. Evidence matrix of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis
Table S42. Evidence prole of randomized controlled trials examining the treatment of high levels of PTH with
calcitriol or activated vitamin D analogs in CKD G3aG5 not on dialysis
Table S43. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD
G5D: study characteristics
Table S44. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD
G5D: study population characteristics
contents www.kisupplements.org
4
Kidney International Supplements (2017) 7, 1–59
Table S45. Summary table of randomized controlled trials examining the treatment of high levels of PTH in
CKD G5D: results
Table S46. Summary table of randomized controlled trials examining the treatment of high levels of PTH in
CKD G5D: quality
Table S47. Evidence matrix of randomized controlled trials examining the treatment of high levels of PTH in
CKD G5D
Table S48. Evidence prole of randomized controlled trials examining the treatment of high levels of PTH in
CKD G5D
Table S49. Summary table of studies evaluating different concentrations of serum phosphate or calcium among
patients with CKD G3aG5 or G5D: study characteristics
Table S50. Summary table of studies evaluating different concentrations of serum phosphate or calcium among
patients with CKD G3aG5 or G5D: study population characteristics
Table S51. Summary table of studies evaluating different concentrations of serum phosphate among patients with
CKD G3aG5 or G5D: results
Table S52. Summary table of studies evaluating different concentrations of serum calcium among patients with
CKD G3aG5 or G5D: results
Table S53. Summary table of studies evaluating different concentrations of serum phosphate or calcium among
patients with CKD G3aG5 or G5D: quality
Table S54. Evidence matrix of studies evaluating different concentrations of serum phosphate or calcium among
patients with CKD G3aG5 or G5D
Table S55. Evidence prole of studies evaluating different concentrations of serum phosphate or calcium among
patients with CKD G3aG5 or G5D
Supplementary material is linked to the online version of the paper at www.kisupplements.org.
www.kisupplements.org contents
Kidney International Supplements (2017) 7, 1–59 5
KDIGO EXECUTIVE COMMITTEE
Garabed Eknoyan, MD
Norbert Lameire, MD, PhD
Founding KDIGO Co-chairs
Bertram L. Kasiske, MD
Immediate Past Co-chair
David C. Wheeler, MD, FRCP
KDIGO Co-chair
Wolfgang C. Winkelmayer, MD, MPH, ScD
KDIGO Co-chair
Ali K. Abu-Alfa, MD
Olivier Devuyst, MD, PhD
Jürgen Floege, MD
John S. Gill, MD, MS
Kunitoshi Iseki, MD
Andrew S. Levey, MD
Zhi-Hong Liu, MD
Ziad A. Massy, MD, PhD
Roberto Pecoits-Filho, MD, PhD
Brian J.G. Pereira, MBBS, MD, MBA
Paul E. Stevens, MB, FRCP
Marcello A. Ton elli, MD, SM, FRCPC
Angela Yee-Moon Wang, MD, PhD, FRCP
Angela C. Webster, MBBS, MM (Clin Ep), PhD
KDIGO Staff
John Davis, Chief Executive Ofcer
Danielle Green, Managing Director
Michael Cheung, Chief ScienticOfcer
Tanya Green, Communications Director
Melissa McMahan, Programs Director
www.kisupplements.org
6
Kidney International Supplements (2017) 7, 1–59
Reference keys
NOMENCLATURE AND DESCRIPTION FOR RATING GUIDELINE
RECOMMENDATIONS
Within each recommendation, the strength of recommendation is indicated as Level 1, Level 2,ornot graded, and the quality of the
supporting evidence is shown as A, B, C,orD.
Grade
*
Implications
Patients Clinicians Policy
Level 1
We recommend
Most people in your situation would
want the recommended course of
action, and only a small proportion
would not.
Most patients should receive the
recommended course of action.
The recommendation can be
evaluated as a candidate for
developing a policy or a performance
measure.
Level 2
We suggest
The majority of people in your
situation would want the
recommended course of action, but
many would not.
Different choices will be appropriate
for different patients. Each patient
needs help to arrive at a management
decision consistent with her or his
values and preferences.
The recommendation is likely to
require substantial debate and
involvement of stakeholders before
policy can be determined.
*The additional category not graded is used, typi cally, to provide guidance based on common sense or when the topic does not allow adequate application of evidence.
The most common examples include recommendations regarding monitoring intervals, counseling, and referral to other clinical specialists. The ungraded recommendations
are generally written as simple declarative statements, but are not meant to be interpreted as being stronger recommendations than Level 1 or 2 recommendations.
Grade Quality of evidence Meaning
A High We are condent that the true effect lies close to that of the estimate of the effect.
B Moderate The true effect is likely to be close to the estimate of the effect, but there is a possibility
that it is substantially different.
C Low The true effect may be substantially different from the estimate of the effect.
D Very low The estimate of effect is very uncertain, and often will be far from the truth.
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 7
CURRENT CHRONIC KIDNEY DISEASE (CKD) NOMENCLATURE USED BY KDIGO
CKD is dened as abnormalities of kidney structure or function, present for > 3 months, with implications for health. CKD is
classied
based on cause, GFR category (G1G5), and albuminuria category (A1A3), abbreviated as CGA.
Prognosis of CKD by GFR and albuminuria category
Prognosis of CKD by GFR
and albuminuria categories:
KDIGO 2012
Persistent albuminuria categories,
description and range
A1
A2
Normal to
mildly
increased
Moderately
increased
<30 mg/g
<3 mg/mmol
30–300 mg/g
3–30 mg/mmol
GFR categories (ml/min/1.73 m
2
),
description and range
G1
Normal or high
≥90
G2
Mildly decreased
60–89
G3a
Mildly to moderately
decreased
45–59
G3b
Moderately to
severely decreased
30–44
G4
Severely decreased
15–29
G5
Kidney failure
<15
green, low risk (if no other markers of kidney disease, no CKD); yellow, moderately increased risk;
orange, high risk; red, very high risk.
web 4C/FPO
www.kisupplements.org
8
Kidney International Supplements (2017) 7, 1–59
CONVERSION FACTORS OF CONVENTIONAL UNITS TO SI UNITS
Conventional unit Conversion factor SI unit
Calcium, total mg/dl 0.2495 mmol/l
Calcium, ionized mg/dl 0.25 mmol/l
Creatinine mg/dl 88.4
m
mol/l
Parathyroid hormone pg/ml 0.106 pmol/l
Phosphate (inorganic) mg/dl 0.3229 mmol/l
Note: conventional unit conversion factor ¼ SI unit.
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 9
Abbreviations and acronyms
1,25(OH)
2
D 1,25-dihydroxyvitamin D
25(OH)D 25-hydroxyvitamin D
AUC area under the curve
bALP bone-specic alkaline phosphatase
BMD bone mineral density
CAC coronary ar tery calcication
CI condence interval
CT computed tomography
CV coefcient of variation
DXA dual-energ y X-ray absorptiometry
eGFR estimated glomerular ltration rate
ERT evidence review team
FGF broblast growth factor
FRAX fracture risk assessment tool
GFR glomerular ltration rate
GI gastrointestinal
GRADE Grading of Recommendations Assessment,
Development, and Evaluation
HD hemodialysis
HPT hyperparathyroidism
HR hazard ratio
iPTH intact parathyroid hormone
ISCD International Society of Clinical
Densitometry
ITT intention-to-treat
IU international unit
KDIGO Kidney Disease: Improving Global
Outcomes
KDOQI Kidney Disease Outcomes Quality
Initiative
LVH left ventricular hypertrophy
LVMI left ventricular mass index
MRI magnetic resonance imaging
OR odds ratio
P1NP amino-terminal propeptide of type 1
procollagen
PTH parathyroid hormone
RCT randomized controlled trial
ROC receiver operating characteristic
SD standard deviation
SHPT secondary hyperparathyroidism
VDR vitamin D receptor
www.kisupplements.org
10
Kidney International Supplements (2017) 7, 1–59
Notice
SECTION I: USE OF THE CLINICAL PRACTICE GUIDELINE
This Clinical Practice Guideline Update is based upon systematic literature searches last conducted in Septemb er 2015 sup-
plemented with additional evidence through February 2017. It is designed to assist decision making. It is not intended to dene
a standard of care, and should not be interpreted as prescribing an exclusive course of management. Variations in practice will
inevitably and appropriately occur when clinicians consider the needs of individual patients, available resources, and limitations
unique to an institution or type of practice. Health care professionals using these recommendations should decide how to apply
them to their own clinical practice.
SECTION II: DISCLOSURE
Kidney Disease: Improving Global Outcomes (KDIGO) makes every effort to avoid any actual or reasonably perceived conicts
of interest that may arise from an outside relationship or a personal, professional, or business interest of a member of the Work
Group. All members of the Work Group are required to complete, sign, and submit a disclosure and attestation form showing
all such relationships that might be perceived as or are actual conicts of interest. This document is updated annually, and
information is adjusted accordingly. All reported information is published in its entirety at the end of this document in the
Work Group members Biographic and Disclosure section, and is kept on le at KDIGO.
Copyright Ó 2017, KDIGO. Published by Elsevier on behalf of the International Society of Nephrology. This is an open
access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Single copies may be made for personal use as allowed by national copyright laws. Special rates are available fo r educational
institutions that wish to make photocopies for nonprot educational use. No part of this publication may be reproduced,
amended, or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any
information storage and retrieval system, without explicit permission in wr iting from KDIGO. Details on how to seek
permission for reproduction or translation, and further information about KDIGOs permissions policies can be obtained by
contacting Danielle Green, Managing Director, at danielle.g[email protected].
To the fullest extent of the law, neither KDIGO, Kidney International Supplements, nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or
otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein.
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 11
Foreword
Kidney International Supplements (2017) 7, 1–59; http://dx.doi.org/10.1016/j.kisu.2017.04.001
With the growing awareness that chronic kidney disease is an
international health problem, Kidney Disease: Improving
Global Outcomes (KDIGO) was established in 2003 with its
stated mission to improve the care and outcomes of kidney
disease patients worldwide through promoting coordination,
collaboration, and integration of initiatives to develop and
implement clinical practice guidelines.
When the KDIGO Clinical Practice Guideline for the
Diagnosis, Evaluation, Prevention, and Treatment of Chronic
Kidney DiseaseMineral and Bone Disorder (CKD-MBD)
was originally published in 2009, the Work Group acknowl-
edged the lack of high-quality evidence on which to base
recommendations. The Guideline included specic research
recommendations to encourage investigators to help ll the
gaps and bolster the evidence base.
Multiple randomized controlled trials and prospective cohort
studies have been published since the 2009 Guideline, and
therefore KDIGO recognizes the need to reexamine the currency
of all of its guidelines on a periodic basis. Accordingly, KDIGO
convened a Controversies Conference in 2013, titled CKD-
MBD: Back to the Future, whose objective was to determine
whether sufcient new data had emerged to support a reassess-
ment of the 2009 CKD-MBD Clinical Practice Guideline and, if
so, to determine the scope of the potential revisions.
Although most of the recommendations were still
considered to be cur rent, the conference identied a total of
12 recommendations for reevaluation based on new data. In
addition, the conference prepared a table of additional topic
questions to be considered by the guideline update Work
Group. The conference noted that, in spite of the completion
of several key clinical trials since the 2009 publication of the
CKD-MBD guideline, large ga ps of knowledge still remained,
as demonstrated by the relatively small number of recom-
mendation statements identi ed for reevaluation. Interested
readers should refer to the conference publicat ion for further
details regarding its processes and deliberations.
1
Therefore, KDIGO commissioned an update to the CKD-
MBD guideline and formed a Work Group, led by Drs.
Markus Ketteler and Mary Leonard. The Work Group
convened in June 2015 to review and appraise the evidence
accumulated since the 2009 Guideline. The topics addressed
for revision are listed in Table 2 and included issues prompted
by EVOLVE post hoc analyses, which were published after the
2013 Controversies Conference. Though 8 years have passed
since the 2009 CKD-MBD guideline, evidence in many areas
is still lacking, which has resulted in many of the opinion-
based recommendation statements from the original guide-
line document remaining unchanged .
In keeping with the standard KDIGO policy of maintain-
ing transparency during the guideline development process
and attesting to its rigor, we conducted an open public review
of the draft CKD-MBD guideline update, and all feedback
received was reviewed and considered by the Work Group
before nalizing this guideline document for publication. The
comments and suggestions greatly assisted us in shaping a
nal document that we felt would be as valuable as possible to
the entire nephrology community.
We wish to thank the Work Group co-chairs, Drs. Markus
Ketteler and Mary Leonard, along with all of the Work Group
members, who volunteered countless hours of their time to
develop this guideline. We also thank Dr. Karen Robinson and
her Evidence Review Team at Johns Hopkins University, the
KDIGO staff, and many others for their support that made
this project possible.
David C. Wheeler, MD, FRCP
Wolfgang C. Winkelmayer, MD, MPH, ScD
KDIGO Co-chairs
foreword www.kisupplements.org
12
Kidney International Supplements (2017) 7, 1–59
Work Group membership
WORK GROUP CO-CHAIRS
Markus Ketteler, MD, FERA
Klinikum Coburg
Coburg, Germany
Mary B. Leonard, MD, MSCE
Stanford University School of Medicine
Stanford, CA, USA
WORK GROUP
Geoffrey A. Block, MD
Denver Nephrology
Denver, CO, USA
Pieter Evenepoel, MD, PhD, FERA
University Hospitals Leuven
Leuven, Belgium
Masafumi Fukagawa, MD, PhD, FASN
Tokai University School of Medicine
Isehara, Japan
Charles A. Herzog, MD, FACC, FAHA
Hennepin County Medical Center
Minneapolis, MN, USA
Linda McCann, RD, CSR
Eagle, ID, USA
Sharon M. Moe, MD
Indiana University School of Medicine
Roudebush Veterans Affairs Medical Center
Indianapolis, IN, USA
Rukshana Shroff, MD, FRCPCH, PhD
Great Ormond Street Hospital for Children
NHS Foundation Trust,
London, UK
Marcello A. Tonelli, MD, SM, FRCPC
University of Calgary
Calgary, Canada
Nigel D. Toussaint MBBS, FRACP, PhD
The Royal Melbourne Hospital
University of Melbourne
Melbourne, Australia
Marc G. Vervloet, MD, PhD, FERA
VU University Medical Center Amsterdam
Amsterdam, The Netherlands
EVIDENCE REVIEW TEAM
Johns Hopkins University
Baltimore, MD, USA
Karen A. Robinson, PhD, Associate Professor of Medicine and Project Director
Casey M. Rebholz, PhD, MPH, MS, Co-investigator
Lisa M. Wilson, ScM, Project Manager
Ermias Jirru, MD, MPH, Research Assistant
Marisa Chi Liu, MD, MPH, Research Assistant
Jessica Gayleard, BS, Research Assistant
Allen Zhang, BS, Research Assistant
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 13
Abstract
The Kidney Disease: Improving Global Outcomes (KDIGO) 2017 Clinical Practice Guideline
Update for the Diagnosis, Evaluation, Prevention, and Treatment of chronic kidney disease
mineral and bone disorder (CKD-MBD) represents a selective update of the prior guideline
published in 2009. This update, along with the 2009 publication, is intended to assist the
practitioner caring for adults and children with CKD, those on chronic dialysis therapy, or in-
dividuals with a kidney transplant. Specically, the topic areas for which updated recommen-
dations are issued include diagnosis of bone abnormalities in CKD-MBD; treatment of CKD-
MBD by targeting phosphate lowering and calcium maintenance, treatment of abnormalities
in parathyroid hormone in CKD-MBD; treatment of bone abnormalities by antiresorptives and
other osteoporosis therapies; and evaluation and treatment of kidney transplant bone disease.
Development of this guideline update followed an explicit process of ev idence review and
appraisal. Treatment approaches and guideline recommendations are based on systematic reviews
of relevant trials, and appraisal of the quality of the evidence and the strength of recommen-
dations followed the GRADE (Grading of Recommendations Assessment, Development,
and Evaluation) approach. Limitations of the ev idence are discussed, with areas of future research
also presented.
Keywords: bone abnormalities; bone mineral density; calcium; chronic kidne y disease; CKD-
MBD; dialysis; guideline ; hyperparathyroidism; hyperphosphatemia; KDIGO; kidney trans-
plantation; mineral and bone disorder; parathyroid hormone; phosphate; phosphorus; systematic
review
CITATION
In citing this document, the following format should be used: Kidney Disease: Improving
Global Outcomes (KDIGO) CKD-MBD Update Work Group. KDIGO 2017 Clinical Practice
Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic
Kidney DiseaseMineral and Bone Disorder (CKD-MBD). Kidney Int Suppl. 2017;7:159.
www.kisupplements.org
14
Kidney International Supplements (2017) 7, 1–59
Summary of KDIGO CKD-MBD recommendations
*
Updated recommendations are denoted in boxes
Chapter 3.1: Diagnosis of CKD-MBD: biochemical abnormalities
3.1.1: We recommend monit oring serum levels of calcium, phosphate, PTH, and alkaline phosphatase activity beginning in
CKD G3a (1C). In children, we suggest such monitoring beginning in CKD G2 (2D).
3.1.2: In patients with CKD G3a G5D, it is reasonable to base the frequency of monitoring serum calcium,
phosphate, and PTH on the presence and magnitude of abnormalities, and the rate of progression of CKD
(Not Graded ).
Reasonable monitoring intervals would be:
In CKD G3aG3b: for serum calcium and phosphate, every 612 months; and for PTH, based on baseline level and
CKD progression.
In CKD G4: for serum calcium and phosphate, every 3 6 months; and for PTH, every 612 months.
In CKD G5, including G5D: for serum calcium and phosphate, every 13 months; and for PTH, every 36
months.
In CKD G4G5D: for alkal ine phosphatase activity, every 12 months, or more frequently in the presence of
elevated PTH (see Chapter 3.2).
In CKD patients receiving treatments for CKD-MBD, or in whom biochemical abnormalities are identied, it is
reasonable to increase the frequency of measurements to monitor for trends and treatment efcacy and side effects
(Not Graded).
3.1.3: In patients with CKD G3aG5D, we suggest that 25(OH)D (calcidiol) levels might be measured, and repeated testing
determined by baseline values and therapeutic interventions (2C). We suggest that vitamin D deciency and
insufciency be corrected using treatment strategies recommended for the general population (2C).
3.1.4: In patients with CKD G3aG5D, we recommend that therapeutic decisions be based on trends rather than on a
single laboratory value, taking into account all available CKD-MBD assessments (1C).
3.1.5: In patients with CKD G3aG5D, we suggest that individual values of serum calcium and phosph ate, evaluated
together, be used to gui de clinical practice rather than the mathematical construct of calcium-phosphate product
(Ca 3 P) (2D).
3.1.6: In reports of laboratory tests for patients with CKD G3aG5D, we recommend that clinical laboratories inform
clinicians of the actual assay method in use and report any change in methods, sample source (plasma or serum), or
handling specications to facilitate the appropriate interpretation of biochemistry data (1B).
Chapter 3.2: Diagnosis of CKD-MBD: bone
3.2.1: In patients with CKD G3aG5D with evidence of CKD-MBD and/or risk factors for osteoporosis, we suggest
BMD testing to assess fracture risk if results will impact treatment decisions (2B).
3.2.2: In patients with CKD G3aG5D, it is reasonable to per form a bone biopsy if knowledge of the type of renal
osteodystrophy will impact treatment decisions (Not Graded).
3.2.3: In patients with CKD G3aG5D, we suggest that measurements of serum PTH or bone-specic alkalin e
phosphatase can be used to evaluate bone dis ease because markedly high or low values predict underlying
bone turnover (2B).
3.2.4: In patients with CKD G3aG5D, we suggest not to routinely measure bone-derived turnover markers of collagen
synthesis (such as procollagen type I C-terminal propeptide) and breakdown (such as type I collagen cross-linked
telopeptide, cross-laps, pyridinoline, or deoxypyridinoline) (2C).
*
The 2009 Guideline Chapters 1 and 2 provide the Introduction and Methodological Approach, respectively, and therefore guideline recommendations
begin in Chapter 3.1.
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 15
3.2.5: We recommend that infants with CKD G2G5D have their length measured at least quarterly, while children with
CKD G2G5D should be asses sed for linear growth at least annually (1B).
Chapter 3.3: Diagnosis of CKD-MBD: vascular calcication
3.3.1: In patients with CKD G3aG5D, we suggest that a lateral abdominal radiograph can be used to detect the presence or
absence of vascular calcication, and an echocardiogram can be used to detect the presence or absence of valvular
calcication, as reasonable alternatives to computed tomography-based imaging (2C).
3.3.2: We suggest that patients with CKD G3aG5D with known vascular or valvular calcication be considered at
highest cardiovascular risk (2A). It is reasonable to use this information to guide the management of CKD-MBD
(Not Graded).
Chapter 4.1: Treatment of CKD-MBD targeted at lowering high serum phosphate and maintaining
serum calcium
4.1.1: In patients with CKD G3aG5D, treatments of CKD-MBD should be based on serial assessments of phosphate,
calcium, and PTH levels, considered together (Not Graded).
4.1.2: In patients with CKD G3aG5D, we suggest lowering elevated phosphate levels toward the normal range (2C).
4.1.3: In adult patients with CKD G3aG5D, we suggest avoiding hypercalcemia (2C). In children with CKD G3aG5D,
we suggest maintaining serum calcium in the age-appropriate normal range (2C).
4.1.4: In patients with CKD G5D, we suggest using a dialysate calcium concentration between 1.25 and 1.50 mmol/l
(2.5 and 3.0 mEq/l) (2C).
4.1.5: In patients with CKD G3a-G5D, decisions about phosphate-lowering treatment should be based on progressively
or persistently elevated serum phosphate (Not Graded).
4.1.6: In adult patients with CKD G3aG5D receiving phosphate-lowering treatment, we suggest restricting the dose of
calcium-based phosphate binders (2B). In children with CKD G3aG5D, it is reasonable to base the choice of
phosphate-lowering treatment on serum calcium levels (Not Graded).
4.1.7: In patients with CKD G3a-G5D, we recommend avoiding the long-term use of alumin um-containing phosphate
binders and, in patients with CKD G5D, avoiding dialysate aluminum contamination to prevent aluminum
intoxication (1C).
4.1.8: In patients with CKD G3aG5D, we suggest limiting dietary pho sphate intake in the treatment of hyper-
phosphatemia alone or in combination with other trea tments (2D). It is reasonable to consider phosphate source
(e.g., animal, vegetable, additives) in making dietary recommendations (Not Graded ).
4.1.9: In patients with CKD G5D , we suggest increasing dialytic phosphat e removal in the treatment of persistent
hyperphosphatemia (2C).
Chapter 4.2: Treatment of abnormal PTH levels in CKD-MBD
4.2.1: In patients with CKD G3aG5 not on dialysis, the optimal PTH level is not known. However, we suggest that
patients with levels of intact PTH progressively rising or persistently above the upper normal limit for the assay
be evaluate d for modiable factors, including hyperphosphatemia, hypocalcemia, high phosphate intake, and
vitamin D deciency (2C).
4.2.2: In adult patients with CKD G3aG5 not on dialysis, we suggest that calcitriol and vitamin D analogs not be
routinely used (2C). It is reasonable to reserve the use of calcitriol and vitamin D analogs for patients with CKD
G4G5 with severe and progres sive hyperparathyroidism (Not Graded).
In children, calcitriol and vitamin D analogs may be considered to maintain serum calcium levels in the
age-appropriate normal range (Not Graded).
summary of recommendation statements www.kisupplements.org
16
Kidney International Supplements (2017) 7, 1–59
4.2.3: In patients with CKD G5D, we suggest maintaining iPTH levels in the range of approximately 2 to 9 times the upper
normal limit for the assay (2C).
We suggest that marked changes in PTH levels in either direction within this range prompt an initiation or change in
therapy to avoid progression to levels outside of this range (2C).
4.2.4: In patients with CKD G5D requiring PTH-lowering therapy, we suggest calcimimetics, calcitriol, or vitamin D
analogs, or a combination of calcimimetics with calcitriol or vitamin D analogs (2B).
4.2.5: In patients with CKD G3aG5D with severe hyperparathyroidism (HPT) who fail to respond to medical or phar-
macological therapy, we suggest parathyroidectomy (2B).
Chapter 4.3: Treatment of bone with bisphosphonates, other osteoporosis medications, and
growth hormone
4.3.1: In patients with CKD G1G2 with osteoporosis and/or high risk of fract ure, as identied by World Health Orga-
nization criteria, we recommend management as for the general population (1A).
4.3.2: In patients with CKD G3aG3b with PTH in the normal range and osteoporosis and/or high risk of fracture, as
identied by World Health Organization criteria, we suggest treatment as for the general population (2B).
4.3.3: In patients with CKD G3aG5D with biochemical abnormalities of CKD-MBD and low BMD and/or fragility
fractures, we suggest that treatment choices take into account the magnitude and reversibility of the biochemical
abnormalities and the progression of CKD, with consideration of a bone biopsy (2D).
4.3.4: In children and adolescents with CKD G2G5D and related heig ht decits, we recommend treatment with re-
combinant human growth hormone when additional growt h is desired, after rst addressing malnutrition and
biochemical abnormalities of CKD-MBD (1A).
Chapter 5: Evaluation and treatment of kidney transplant bone disease
5.1: In patients in the immediate postkidney transplant period, we recommend measuring serum calcium and phosphate
at least weekly, until stable (1B).
5.2: In patients after the immediate postkidney transplant period, it is reasonable to base the frequency of monitoring
serum calcium, phosphate, and PTH on the presence and magnitude of abnor malities, and the rate of progression of
CKD (Not Graded).
Reasonable monitoring intervals would be:
In CKD G1TG3bT, for serum calcium and phosphate, every 612 months; and for PTH, once, with subsequent
intervals depending on ba seline level and CKD progression.
In CKD G4T, for serum calcium and phosphate, every 36 months; and for PTH, every 6 12 months.
In CKD G5T, for serum calcium and phosphate, every 13 months; and for PTH, every 3 6 months.
In CKD G3aTG5T, measurement of alkaline phosphatases annually, or more frequently in the presence of elevated
PTH (see Chapter 3.2).
In CKD patients receiving treatments for CKD-MBD, or in whom biochemical abnormalities are identied, it is
reasonable to increase the freq uency of measurements to monitor for efcacy and side effects (Not Graded).
It is reasonable to manage these abnormalities as for patients with CKD G3aG5 (see Chapters 4.1 and 4.2)
(Not Graded).
5.3: In patients with CKD G1TG5T, we suggest that 25(OH)D (calcidiol) levels might be measured, and repeated testing
determined by baseline values and interventions (2C).
5.4: In patients with CKD G1TG5T, we suggest that vitamin D deciency and insufciency be corrected using treatment
strategies recommended for the general population (2C).
www.kisupplements.org summary of recommendation statements
Kidney International Supplements (2017) 7, 1–59 17
5.5: In patients with CKD G1TG5T with risk factors for osteoporosis, we suggest that BMD testing be used to assess
fracture risk if results will alter therapy (2C).
5.6: In patients in the rst 12 months after kidney transplant with an estimated glomerular ltration rate greater than
approximately 30 ml/min/1.73 m
2
and low BMD, we suggest that treatment with vitamin D, calcitriol/alfacalcidol,
and/or antiresorptive agents be considered (2D).
We suggest that treatment choices be inuenced by the presence of CKD-MBD, as indicated by abnormal levels
of calcium, phosphate, PTH, alkaline phosphatases, and 25(OH)D (2C).
It is reasonable to consider a bone biopsy to guide treatment (Not Graded).
There are insufcient data to guide treatment after the rst 12 months.
5.7: In patients with CKD G4TG5T with known low BMD, we suggest management as for patients with CKD G4G5 not
on dialysis, as detailed in Chapters 4.1 and 4.2 (2C).
The 2017 updated recommendations resulted in renumbering of several adjacent guideline statements. Specically, 2009
Recommendation 4.1.6 now becomes 2017 Recommendation 4.1.7; 2009 Reco mmendation 4.1.8 now becomes 2017
Recommendation 4.1.9; 2009 Recommendation 4.3.5 now becomes 2017 Recommendation 4.3.4; and 2009 Recommen-
dation 5.8 now becomes 2017 Recommendation 5.7.
summary of recommendation statements www.kisupplements.org
18
Kidney International Supplements (2017) 7, 1–59
Summary and comparison of 2017 updated and 2009
KDIGO CKD-MBD recommendations
2017 revised KDIGO CKD-MBD
recommendations 2009 KDIGO CKD-MBD recommendations Brief rationale for updating
3.2.1. In patients with CKD G3aG5D with
evidence of CKD-MBD and/or risk factors for
osteoporosis, we suggest BMD testing to
assess fracture risk if results will impact
treatment decisions (2B).
3.2.2. In patients with CKD G3aG5D with evidence
of CKD-MBD, we suggest that BMD testing not be
performed routinely, because BMD does not
predict fracture risk as it does in the general
population, and BMD does not predict the type of
renal osteodystrophy (2B).
Multiple new prospective studies have
documented that lower DXA BMD predicts
incident fractures in patients with CKD G3a
G5D. The order of these rst 2
recommendations was changed because a
DXA BMD result might impact the decision to
perform a bone biopsy.
3.2.2. In patients with CKD G3aG5D, it is
reasonable to perform a bone biopsy if
knowledge of the type of renal osteodystrophy
will impact treatment decisions (Not Graded).
3.2.1. In patients with CKD G3aG5D, it is
reasonable to perform a bone biopsy in various
settings including, but not limited to: unexplained
fractures, persistent bone pain, unexplained
hypercalcemia, unexplained hypophosphatemia,
possible aluminum toxicity, and prior to therapy
with bisphosphonates in patients with CKD-MBD
(Not Graded).
The primary motivation for this revision was
the growing experience with osteoporosis
medications in patients with CKD, low BMD,
and a high risk of fracture. The inability to
perform a bone biopsy may not justify
withholding antiresorptive therapy from
patients at high risk of fracture.
4.1.1. In patients with CKD G3aG5D,
treatments of CKD-MBD should be based on
serial assessments of phosphate, calcium, and
PTH levels, considered together (Not Graded).
This new recommendation was provided in
order to emphasize the complexity and
interaction of CKD-MBD laboratory parameters.
4.1.2. In patients with CKD G3aG5D, we
suggest lowering elevated phosphate levels
toward the normal range (2C).
4.1.1. In patients with CKD G3aG5, we suggest
maintaining serum phosphate in the normal
range (2C). In patients with CKD G5D, we suggest
lowering elevated phosphate levels toward the
normal range (2C).
There is an absence of data supporting that
efforts to maintain phosphate in the normal
range are of benet to CKD G3aG4 patients,
including some safety concerns. Treatment
should aim at overt hyperphosphatemia.
4.1.3. In adult patients with CKD G3aG5D, we
suggest avoiding hypercalcemia (2C).
In children with CKD G3aG5D, we suggest
maintaining serum calcium in the age-
appropriate normal range (2C).
4.1.2. In patients with CKD G3aG5D, we suggest
maintaining serum calcium in the normal range (2D).
Mild and asymptomatic hypocalcemia (e.g., in
the context of calcimimetic treatment) can be
tolerated in order to avoid inappropriate
calcium loading in adults.
4.1.4. In patients with CKD G5D, we suggest
using a dialysate calcium concentration
between 1.25 and 1.50 mmol/l (2.5 and 3.0
mEq/l) (2C).
4.1.3. In patients with CKD G5D, we suggest using
a dialysate calcium concentration between 1.25
and 1.50 mmol/l (2.5 and 3.0 mEq/l) (2D).
Additional studies of better quality are
available; however, these do not allow for
discrimination of benets and harms between
calcium dialysate concentrations of 1.25 and
1.50 mmol/l (2.5 and 3.0 mEq/l). Hence, the
wording is unchanged, but the evidence grade
is upgraded from 2D to 2C.
4.1.5. In patients with CKD G3aG5D, decisions
about phosphate-lowering treatment should
be based on progressively or persistently
elevated serum phosphate (Not Graded).
4.1.4. In patients with CKD G3aG5 (2D) and G5D
(2B), we suggest using phosphate-binding agents
in the treatment of hyperphosphatemia. It is
reasonable that the choice of phosphate binder
takes into account CKD stage, presence of other
components of CKD-MBD, concomitant therapies,
and side effect prole (Not Graded).
Emphasizes the perception that early
preventive phosphate-lowering treatment is
currently not supported by data (see
Recommendation 4.1.2).
The broader term phosphate-lowering
treatment is used instead of phosphate
binding agents since all possible approaches
(i.e., binders, diet, dialysis) can be effective.
(Continued on next page)
www.kisupplements.org
Kidney International Supplements (2017) 7, 1–59 19
2017 revised KDIGO CKD-MBD
recommendations 2009 KDIGO CKD-MBD recommendations Brief rationale for updating
4.1.6. In adult patients with CKD G3aG5D
receiving phosphate-lowering treatment, we
suggest restricting the dose of calcium-based
phosphate binder (2B). In children with CKD
G3aG5D, it is reasonable to base the choice of
phosphate-lowering treatment on serum
calcium levels (Not Graded).
4.1.5. In patients with CKD G3aG5D and
hyperphosphatemia, we recommend restricting
the dose of calcium-based phosphate binders
and/or the dose of calcitriol or vitamin D analog in
the presence of persistent or recurrent
hypercalcemia (1B).
New evidence from 3 RCTs supports a more
general recommendation to restrict calcium-
based phosphate binders in
hyperphosphatemic patients across all
severities of CKD.
In patients with CKD G3aG5D and
hyperphosphatemia, we suggest restricting the
dose of calcium-based phosphate binders in the
presence of arterial calcication (2C) and/or
adynamic bone disease (2C) and/or if serum PTH
levels are persistently low (2C).
4.1.8. In patients with CKD G3aG5D, we
suggest limiting dietary phosphate intake in
the treatment of hyperphosphatemia alone or
in combination with other treatments (2D). It is
reasonable to consider phosphate source (e.g.,
animal, vegetable, additives) in making dietary
recommendations (Not Graded).
4.1.7. In patients with CKD G3aG5D, we suggest
limiting dietary phosphate intake in the treatment
of hyperphosphatemia alone or in combination
with other treatments (2D).
New data on phosphate sources were deemed
to be included as an additional qualier to the
previous recommendation.
4.2.1. In patients with CKD G3aG5 not on
dialysis, the optimal PTH level is not known.
However, we suggest that patients with levels
of intact PTH progressively rising or
persistently above the upper normal limit for
the assay be evaluated for modiable factors,
including hyperphosphatemia, hypocalcemia,
high phosphate intake, and vitamin D
deciency (2C).
4.2.1. In patients with CKD G3aG5 not on dialysis,
the optimal PTH level is not known. However, we
suggest that patients with levels of intact PTH
above the upper normal limit of the assay are rst
evaluated for hyperphosphatemia, hypocalcemia,
and vitamin D deciency (2C ).
It is reasonable to correct these abnormalities with
any or all of the following: reducing dietary
phosphate intake and administering phosphate
binders, calcium supplements, and/or native
vitamin D (Not Graded).
The Work Group felt that modest increases in
PTH may represent an appropriate adaptive
response to declining kidney function and has
revised this statement to include persistently
above the upper normal PTH level as well as
progressively rising PTH levels, rather than
above the upper normal limit. That is,
treatment should not be based on a single
elevated value.
4.2.2. In adult patients with CKD G3a
G5 not on
dialysis, we suggest that calcitriol and vitamin D
analogs not be routinely used. (2C)Itis
reasonable to reserve the use of calcitriol and
vitamin D analogs for patients with CKD G4G5
with severe and progressive
hyperparathyroidism (Not Graded).
4.2.2. In patients with CKD G3aG5 not on dialysis,
in whom serum PTH is progressively rising and
remains persistently above the upper limit of
normal for the assay despite correction of
modiable factors, we suggest treatment with
calcitriol or vitamin D analogs (2C).
Recent RCTs of vitamin D analogs failed to
demonstrate improvements in clinically
relevant outcomes but demonstrated
increased risk of hypercalcemia.
In children, calcitriol and vitamin D analogs
may be considered to maintain serum calcium
levels in the age-appropriate normal range
(Not Graded).
4.2.4. In patients with CKD G5D requiring PTH-
lowering therapy, we suggest calcimimetics,
calcitriol, or vitamin D analogs, or a
combination of calcimimetics with calcitriol or
vitamin D analogs (2B).
4.2.4. In patients with CKD G5D and elevated or
rising PTH, we suggest calcitriol, or vitamin D
analogs, or calcimimetics, or a combination of
calcimimetics and calcitriol or vitamin D analogs
be used to lower PTH (2B).
It is reasonable that the initial drug selection for
the treatment of elevated PTH be based on
serum calcium and phosphate levels and other
aspects of CKD-MBD (Not Graded).
It is reasonable that calcium or non-calcium-based
phosphate binder dosage be adjusted so that
treatments to control PTH do not compromise
levels of phosphate and calcium (Not Graded).
We recommend that, in patients with hyper-
calcemia, calcitriol or another vitamin D sterol
be reduced or stopped (1B).
This recommendation originally had not been
suggested for updating by the KDIGO
Controversies Conference in 2013. However,
due to a subsequent series of secondary and
post hoc publications of the EVOLVE trial, the
Work Group decided to reevaluate
Recommendation 4.2.4 as well. Although
EVOLVE did not meet its primary endpoint, the
majority of the Work Group members were
reluctant to exclude potential benets of
calcimimetics for G5D patients based on
subsequent prespecied analyses. The Work
Group, however, decided not to prioritize any
PTH-lowering treatment at this time because
calcimimetics, calcitriol, or vitamin D analogs
are all acceptable rst-line options in G5D
patients.
summary and comparison of 2017 updated and 2009 KDIGO CKD-MBD recommendations www.kisupplements.org
20
Kidney International Supplements (2017) 7, 1–59
2017 revised KDIGO CKD-MBD
recommendations 2009 KDIGO CKD-MBD recommendations Brief rationale for updating
We suggest that, in patients with hyper-
phosphatemia, calcitriol or another vitamin D
sterol be reduced or stopped (2D).
We suggest that, in patients with hypocalcemia,
calcimimetics be reduced or stopped depend-
ing on severity, concomitant medications, and
clinical signs and symptoms (2D).
We suggest that, if the intact PTH levels fall
below 2 times the upper limit of normal for the
assay, calcitriol, vitamin D analogs, and/or cal-
cimimetics be reduced or stopped (2C).
4.3.3. In patients with CKD G3aG5D with
biochemical abnormalities of CKD-MBD and
low BMD and/or fragility fractures, we suggest
that treatment choices take into account the
magnitude and reversibility of the biochemical
abnormalities and the progression of CKD,
with consideration of a bone biopsy (2D).
4.3.3. In patients with CKD G3aG3b with
biochemical abnormalities of CKD-MBD and low
BMD and/or fragility fractures, we suggest that
treatment choices take into account the
magnitude and reversibility of the biochemical
abnormalities and the progression of CKD, with
consideration of a bone biopsy (2D).
Recommendation 3.2.2 now addresses the
indications for a bone biopsy prior to
antiresorptive and other osteoporosis
therapies. Therefore, 2009 Recommendation
4.3.4 has been removed and 2017
Recommendation 4.3.3 is broadened from CKD
G3aG3b to CKD G3aG5D.
4.3.4. In patients with CKD G4G5D having
biochemical abnormalities of CKD-MBD, and low
BMD and/or fragility fractures, we suggest
additional investigation with bone biopsy prior to
therapy with antiresorptive agents (2C).
5.5. In patients with G1TG5T with risk factors
for osteoporosis, we suggest that BMD testing
be used to assess fracture risk if results will
alter therapy (2C).
5.5. In patients with an estimated glomerular
ltration rate greater than approximately 30 ml/
min/1.73 m
2
, we suggest measuring BMD in the
rst 3 months after kidney transplant if they
receive corticosteroids, or have risk factors for
osteoporosis as in the general population (2D).
2009 Recommendations 5.5 and 5.7 were
combined to yield 2017 Recommendation 5.5.
5.7. In patients with CKD G4TG5T, we suggest
that BMD testing not be performed routinely,
because BMD does not predict fracture risk as it
does in the general population and BMD does
not predict the type of kidney transplant bone
disease (2B).
5.6. In patients in the rst 12 months after
kidney transplant with an estimated
glomerular ltration rate greater than
approximately 30 ml/min/1.73 m
2
and low
BMD, we suggest that treatment with vitamin
D, calcitriol/alfacalcidol, and/or antiresorptive
agents be considered (2D).
We suggest that treatment choices be
inuenced by the presence of CKD-MBD, as
indicated by abnormal levels of calcium,
phosphate, PTH, alkaline phosphatases, and
25(OH)D (2C).
It is reasonable to consider a bone biopsy to
guide treatment (Not Graded).
There are insufcient data to guide treatment
after the rst 12 months.
5.6. In patients in the rst 12 months after kidney
transplant with an estimated glomerular ltration
rate greater than approximately 30 ml/min/1.73
m
2
and low BMD, we suggest that treatment with
vitamin D, calcitriol/alfacalcidol, or
bisphosphonates be considered (2D).
We suggest that treatment choices be inu-
enced by the presence of CKD-MBD, as indi-
cated by abnormal levels of calcium, phosphate,
PTH, alkaline phosphatases, and 25(OH)D (2C).
It is reasonable to consider a bone biopsy to
guide treatment, specically before the use of
bisphosphonates due to the high incidence of
adynamic bone disease (Not Graded).
There are insufcient data to guide treatment after
the rst 12 months.
The second bullet is revised, consistent with
the new bone biopsy recommendation (i.e.,
2017 Recommendation 3.2.2).
25(OH)D, 25-hydroxyvitamin D; BMD, bone mineral density; CKD, chronic kidney disease; CKD-MBD, chronic kidney diseasemineral bone disorder; DXA, dual-energy x-ray
absorptiometry; PTH, parathyroid hormone; RCT, randomized controlled trial.
Changes to above summarized recommendations resulted in renumbering of several adjacent guideline statements. Specically, 2009 Recommendation 4.1.6 now becomes
2017 Recommendation 4.1.7; 2009 Recommendation 4.1.8 now becomes 2017 Recommendation 4.1.9; 2009 Recommendation 4.3.5 now becomes 2017 Recommendation
4.3.4; and 2009 Recommendation 5.8 now becomes 2017 Recommendation 5.7.
www.kisupplements.org summary and comparison of 2017 updated and 2009 KDIGO CKD-MBD recommendations
Kidney International Supplements (2017) 7, 1–59 21
Chapter 3.2: Diagnosis of CKD-MBD: bone
3.2.1: In patients with CKD G3a G5D with evidence of
CKD-MBD and/or risk factors for osteoporosis, we
suggest BMD testing to assess fracture risk if results
will impact treatment decisions (2B).
Rationale
It is well established that patients with CKD G3aG5D have
increased fracture rates compared with the general popula-
tion,
24
and moreover, incident hip fractures are associated
with substantial morbidity and mortality.
59
At the time of
the 2009 KDIGO CKD-MBD guideline, pu blications
addressing the ability of dual-energy X-ray absorptiometry
(DXA) measures of bone mineral density (BMD) to estimate
fracture risk in CKD were limited to cross-sectional studies
comparing BMD in CKD patien ts with and without a prev-
alent fracture. The results were variable across studies and
across skeletal sites. In light of the lack of evidence that DXA
BMD predicted fractures in CKD patients as it does in the
general population, and the inability of DXA to indicate the
histological type of bone disease, the 2009 Guideline recom-
mended that BMD testing not be performed routinely in
patients with CKD G3a to G5D with CKD-MBD. Further-
more, the lack of clinical trials in patients with low BMD and
CKD also limited the enthusiasm for measuring BMD in the
rst place.
The current evidence-based review identied 4 prospective
cohort studies of DXA BMD and incident fractures in adults
with CKD G3a to G5D (Supplementary Tables S7S12).
These studies demonst rated that DXA BMD predicted frac-
tures across the spectrum from CKD G3a to G5D
(Supplementary Tables S7S12).
1013
In the earlies t study,
DXA BMD was measured annually in 485 hemodialysis (HD)
patients (mean age: 60 years) in a single center in Japan.
10
In
adjusted Cox proportional analyses, lower baseline femoral
neck and total hip BMD predicted a greater risk of fracture;
for example, the hazard ratio (HR) was 0.65 (95% condence
interval [CI]: 0.470.90) for each standard deviation (SD)
higher femoral neck BMD. In receiver operating character istic
(ROC) analyses stratied according to parathyroid hormone
(PTH) below or above the median value of 204 pg/ml (21.6
pmol/l), the area under the curve (AUC) for femoral neck
BMD was 0.717 in the lower stratum and 0.512 in the higher
stratum. Of note, higher serum bone-specic alkaline phos-
phate levels also predicted incident fractures.
In the second study, Yenchek et al. assessed whether DXA
total hip and femoral neck BMD were associated with incident
nonspine fragility fractures in participants with estimated
glomerular ltration rate (eGFR) < 60 ml/min/1.73 m
2
and
without CKD in the Health, Aging and Body Composition
Study, a prospective study of community-living individuals, 70
to 79 years of age at enrollment.
13
A total of 587 (21%) of the
2754 participants had CKD, and among those, 83% and 13%
had CKD G3a and G3b, respectively. In adjusted analyses, the
fracture HR for each SD lower femoral neck BMD was 2.14
(95% CI: 1.802.55) in participants w ithout CKD, and 2.69
(95% CI: 1.963.69) in those with CKD. Similar results were
obser ved for total hip BMD. When limited to hip fractures, the
adjusted femoral neck BMD HRs were 5.82 (95% CI: 3.27
10.35) among those with CKD and 3.08 (95% CI: 2.294.14)
among those without CKD. Interaction terms demonstrated
that the association of BMD with fracture did not differ in those
with versus without CKD. However, the association of femoral
neck BMD with fract ure was signicantly less pronounced (test
for interaction, P ¼ 0.04) among those with PTH > 65 pg/ml
(6.9 pmol/l; HR: 1.56, 95% CI: 0.902.70) compared with those
with a PTH # 65 pg/ml (6.9 pmol/l; HR: 2.41, 95% CI: 2.04
2.85) in all participants combined. This is noteworthy in light
of the similar pattern observed in dialysis patients, as descr ibed
above.
10
West et al. repor ted the results of a prospective cohort
study of 131 predialysis participants, mean age 62 years, fol-
lowed up over a 2-year interval.
12
At baseline, the proportions
with CKD G3a to G3b, G4, and G5 were 34%, 40%, and 26%,
respectively. DXA BMD was measured in the total hip, lum-
bar spine, and ultradistal and one-third radius at baseline and
2 years. Low BMD at all sites, and a greater annualized per-
centage decrease in BMD predicted fracture. For example, in
multivariate models, each SD lower total hip BMD was
associated with an odds ratio (OR) of fracture of 1.75 (95%
CI: 1.302.20). The ROC AUC ranged from 0.62 in the spine
to 0.74 in the ultradistal radius in adjusted models.
Most recently, Naylor, et al.
11
assessed the ability of the
Fracture Risk Assessment Tool (FRAX) to predict a major
osteoporotic fracture in 2107 adu lts $ 40 years of age in the
Canadian Multicenter Osteoporosis Study, including 320 with
an eGFR # 60 ml/min/1.73 m
2
. Of these, 72% and 24% had
CKD G3a and G3b, respectively. FRAX with BMD, FRAX
without BMD, and the femoral neck T-score all predicted
fractures (AUC: 0.65 to 0.71); the AUC was highest for femoral
neck T-score with inclusion of fall history. Importantly, the
AUCs did not differ between those with and without CKD.
There is growing evidence that DXA BMD predicts frac-
tures in healthy children and adolescents, and those with
chronic disease.
14,15
However, no studies have examined the
associations among DXA BMD and fractures in children and
adolescents with CKD. In light of the lack of evidence that the
chapter 3.2 www.kisupplements.org
22
Kidney International Supplements (2017) 7, 1–59
ability of DXA BMD to predict fracture in children with CKD
is different than in adults, no specic recommendations are
provided for children. However, it should be noted that
children and adolescents with CKD frequently exhibit sub-
stantial growth failure. Given that DXA measures of areal
BMD (g/cm
2
) underestimate volumetric BMD (g/cm
3
)in
children with short stature,
16
DXA results should be adjusted
for bone size, consistent with the 2013 International Society
of Clinical Densitometry (ISCD) Pediatric Ofcial Posi-
tions.
17
Prediction equations to adjust DXA results for height
Z-score are now available,
16
and the impact on DXA BMD
Z-scores in children with CKD is substantial.
18
Finally, a single-
center study in 171 children with CKD G2 to G5D reported
that lower cor tical volumetric BMD in the tibia, as measured
by peripheral quantitative computed tomography (CT), pre-
dicted fractures over a 1-year interval (Supplementary
Tables S7S12).
19
The HR per unit lower cortical BMD
Z-score was 1.75 (95% CI: 1.152.67; P < 0.01).
The evidence-based review also evaluated clinical trials of
the effects of osteoporosis medications on BMD in CKD G3a
to G5D (Supplementary Tables S1S6). Prior analyses of large
randomized clinical trials (RCTs) evaluating medications for
the treatment of postmenopausal osteoporosis (risedronate,
alendronate, teriparatide, and raloxifene) were described in
the 2009 Guideline. These trials specically excluded patients
with an elevated serum creatinine, hyperparathyroidism, or
abnormal alkaline phosphate levels (i.e., CKD-MBD).
2023
However, post hoc analyses found that these drugs had
similar efcacy on improving BMD and reducing fracture
incidence in individuals with moderately reduced eGFR,
compared with those with mildly decreased or normal eGFR.
Three new trials were identied. The denosumab study was
also a post hoc analysis of an RCT in women with post-
menopausal os teoporosis and normal PTH levels.
24
The
analysis demonstrated efcacy of denosumab in decreasing
fracture r isk and increasing BMD in 2817 women with CKD
G3a to G3b and 73 with CKD G4. Here, the risk of hypo-
calcemia associated w ith denosumab in advanced CKD re-
quires mentioning. The remaining 2 new trials on
alendronate
25
and raloxifene
26
were small studies (<60 par-
ticipants) that did not exclude patients with evidence of CKD-
MBD. These studies did not show consistent benecial effects
on DXA BMD. Generally, a major limitation is the lack of
data on fracture prevention by such therapeutic interventions
in advanced CKD (especially in CKD G5G5D).
In summary, the aforementioned 4 prospective studies
evaluating BMD testing in adults with CKD represent a
substantial advance since the orig inal guideline from 2009.
Despite the fact that they were conducted across a spectrum
of CKD severity, the nding that hip BMD predicted fractures
was consistent across studies, and 2 studies demonstrated
associations comparable to those seen in the absence of
CKD.
11,13
Based on these insights, if a low or declining BMD
will lead to additional interventions to reduce falls or use
osteoporosis medications, then BMD assessment is
reasonable.
Research recommendations
RCTs are needed to determine whether interventions based
on DXA BMD are associated with lower fracture rates, and
whether the effects vary based on clinical variables such as
the baseline PTH level, underlying cause of kidney disease,
and CKD GFR category.
Prospective studies are needed to determine whether
alternative imaging techniques, such as quantitative CT,
improve fracture prediction in CKD.
Prospective studies are needed in child ren and adolescents
to determine whether DXA predicts fractures in children
and to determine whether the ISCD recommendations to
measure whole-body and spine BMD in children are the
appropriate sites in the context of CKD.
17
Hip and radius
BMD pediatric reference data are now available and predict
incident fractures in healthy children and adolescents.
27,28
3.2.2: In patients with CKD G3aG5D, it is reasonable to
perform a bone biopsy if knowledge of the type of
renal osteodystrophy will impac t trea tment decisions
(Not Graded).
Rationale
Renal osteodystrophy is dened as abnormal bone histology
and is 1 component of the bone abnormalities of CKD-
MBD.
29
Bone biopsy is the gold standard for the diagnosis
and classication for renal osteodystrophy. As detailed in the
2009 KDIGO CKD-MBD Guideline,
30
DXA BMD does not
distinguish among types of renal osteodystrophy, and the
diagnostic utility of biochemical markers is limited by poor
sensitivity and specicity. Differences in PTH assays (e.g.,
intact vs. whole PTH) and reference ranges have contributed
to differences across studies. Unfortunately, cross-sectional
studies have provided conicting information on the use of
biomarkers to predict underlying bone histology. This is not
surprising given the short half-lives of most of the circulating
biomarkers, and the long (36 months) bone remodeling
(turnover) cycle.
KDIGO recently led an international consortium to
conduct a cross-sectional retrospective diagnostic study of
biomarkers (all run in a single laboratory) and bone biopsies
in 492 dialysis patients.
31
The objective was to determine the
predictive value of PTH (determined by both intact PTH
[iPTH] and whole PTH assays), bone-specic alkaline phos-
phatase (bALP), and amino-terminal propeptide of type 1
procollagen (P1NP) as markers of bone turnover. Altho ugh
iPTH, whole PTH, and bALP levels were associated with bone
turnover, no biomarker singly or in combination was suf-
ciently robust to diagnose low, normal, and high bone turn-
over in an individual patient. The conclusion was in support
of the 2009 KDIGO Guideline to use trends in PTH rather
than absolute target values when making decisions as to
whether to start or stop treatments to lower PTH. Table 1
provides the sensitivity, specicit y, and positive and negative
predictive value of PTH in helping clinicians determine
therapies, demonstrating the challenges clinicians face. Thus,
www.kisupplements.org chapter 3.2
Kidney International Supplements (2017) 7, 1–59 23
the Work Group encourages the continued use of trends in
PTH to guide therapy, and when trends in PTH are incon-
sistent, a bone biopsy should be considered.
A bone biopsy should also be considered in patients with
unexplained fractures, refractory hypercalcemia, suspicion of
osteomalacia, an atypical response to standard therapies for
elevated PTH, or progressive decreases in BMD despite
standard therapy. The goal of a bone biopsy would be to: (i)
rule out atypical or unexpected bone pathology ; (ii) deter-
mine whether the patient has high- or low-turnover disease,
which may alter the dose of medications to treat renal
osteodystrophy (e.g., initiate or discontinue calcimimetics,
calcitriol, or vitamin D analogs); or (iii) identify a minerali-
zation defect that would alter treatment (e.g., stop intake of
aluminum, or aggressively treat hypophosphatemia or
vitamin D deciency).
The 2009 Guideline recommended a bone biopsy prior to
antiresorptive therapy in patients with CKD G4 to G5D and
evidence of biochemical abnormalities of CKD-MBD, low
BMD, and/or fragility fractures. The rationale was that low
BMD may be due to CKD-MBD (e.g., high PTH) and that
lowering PTH is a safer and more appropriate therapy than an
antiresorptive. In addition, there was concern that
bisphosphonates would induce low-turnover bone disease.
This was based on a single cross-sectional study in 13 patients
with CKD G2 to G4 that were refer red for bone biopsy after a
variable duration of bisphosphonate therapy.
32
To date,
studies in patients with CKD have not deniti vely demon-
strated that bisphosphonates cause adynamic bone disease.
Furthermore, the concerns in patients with CKD are only
theoretical, as it is w ell established that antiresorptive medi-
cations suppress bone formation rates, even in the absence of
kidney disease. For examp le, in an RCT of zoledronic acid for
the treatment of postmenopausal osteoporosis, bALP levels
were 59% lower in the zoledronic acid group compared with
the placebo group at 12 months.
33
Despite these limitations, in weighing the risk-benet ratio
of bisphosphonate treatment, the 2009 KDIGO Guideline
suggested a biopsy prior to therapy. Since 2009, an additional
antiresorptive treatment (denosumab) has proven to be
effective in CKD G3a to G3b and G4, as discussed in
Recommendation 3.2.1. The g rowing experience with osteo-
porosis medications in patients with CKD increases the
comfort of treating patients with low BMD and a high risk of
fracture with antiresorptive therapy, although denitive trials
are lacking. Furthermore, additional data clearly support that
the incidence of fracture is markedly increased in patients
with CKD, and thus the inability to perform a bone biopsy
may not justify withholding antiresorptive therapy to patients
at high risk of fracture. Thus, the Work Group voted to
remove the requirement of bone biopsy prior to the use of
antiresorptive ther apy for osteoporosis because the use of
these drugs must be individualized in patients with CKD.
However, it is still prudent that these drugs be used with
caution and that the underlying renal osteodystrophy be
addressed rst. With regard to efcacy, one may speculate that
antiresorptive therapies confer less benet in the absence of
activated osteoclasts, as is the case in adynamic bone disease.
Moreover, additional side effects such as acute kidney injury
may also merit consideration in CKD G3a to G5.
In summary, bone biopsy is the gold standard for the
assessment of renal osteodystrophy and should be considered
in patients in whom the etiology of clinical symptoms and
biochemical abnormalities is in question, and the results may
lead to changes in therapy. With this statement, the Work
Group is well aware that experience concerning performance
and evaluation of bone biopsies is limited in many centers.
34
With this in mind, in addition to the growing evidence that
antiresorptive therapies are effective in patients with CKD
G3a to G3b and G4, and the lack of robust evidence that these
medications induce adynamic bone disease, the guideline no
longer suggests that a bone biopsy be performed prior to
initiation of these medications.
Research recommendation
Prospective studies of circulating biomarkers are needed to
determine whether they can predict changes in bone
histology.
Table 1 | Utility of KDOQI and KDIGO PTH thresholds for diagnostic decision making
KDOQI
*
KDIGO
D
Sens Spec PPV NPV Sens Spec PPV NPV
Differentiating low-turnover from nonlow-turnover bone disease,
or When do I stop therapy?
69% 61% 72% 58% 66% 65% 73% 57%
Differentiating high-turnover from nonhigh-turnover bone disease,
or When do I start therapy?
58% 78% 35% 90% 37% 86% 35% 87%
iPTH, intact parathyroid hormone; KDIGO, Kidney Disease: Improving Global Outcomes; KDOQI, Kidney Disease Outcomes Quality Initiative; NPV, negative predictive value;
PPV, positive predictive value; PTH, parathyroid hormone; Sens, sensitivity; Spec, specicity.
*Using serum iPTH < 150 pg/ml (16 pmol/l) for lower and > 300 pg/ml (32 pmol/l) for upper threshold.
þ
Using serum iPTH < 130 pg/ml (14 pmol/l) for lower and > 585 pg/ml (62 pmol/l) for upper threshold (2X and 9X of upper limit of normal for assay).
Reproduced with permission from Sprague SM, Bellorin-Font E, Jorgetti V, et al. Diagnostic accuracy of bone turnover markers and bone histology in patients with CKD treated
by dialysis. Am J Kidney Dis. 2016;67:559 566.
chapter 3.2 www.kisupplements.org
24
Kidney International Supplements (2017) 7, 1–59
Chapter 4.1: Treatment of CKD-MBD targeted at
lowering high serum phosphate and maintaining
serum calcium
4.1.1: In patients with CKD G3aG5D, treatments of CKD-
MBD should be based on serial assessments of
phosphate, calcium, and PTH levels, considered
together (Not Graded).
Rationale
The previous Recommendation 4.1.1 from the 2009 KDIGO
CKD-MBD guideline gave treatment directions concerning
serum phosphate levels in different GFR categories of CKD.
The accumulated evidence on this issue to date is now
depicted in Supplementary Tables S49 S51, S53S55. Results
of this evidence review can be summar ized as follows: most
studies showed increasing risk of all-cause mortality with
increasing levels of serum phosphate in a consistent and
direct fashion, with moderate risk of bias and low quality of
evidence, thus not essentially different from the study results
before 2009. For GFR decline and cardiovascular event rate,
results were considered less conclusive.
Serum phosphate, calcium, and PTH concentrations are all
routinely measured in CKD patients, and clinical decisions
are often made bas ed on these values. However, the results of
these tests are inuenced by food intake, adherence to and the
timing of drug intake and dietary modications, differences
in assay methods and their intra-assay coefcient of variation
(CV), and also by the interval from the last dialysis session in
CKD G5D patients. Furthermore, it has recently been sug-
gested that these mar kers undergo signicant diurn al changes
even in CKD patients.
35,36
Accordingly, the decision should be
based not on a single result, but rather on the trends of serial
results, which stands very much in accordance to 2009
Recommendation 3.1.4. In addition, recent post hoc analyses
of large dialysis cohorts suggest that the prognostic implica-
tions of individual biochemical components of CKD-MBD
largely depend on their context with regard to constellations
of the full array of MBD bioma rkers.
37
This analysis identied
a wide range of CKD-MBD phenotypes, based on phosphate,
calcium, and PTH measurements categorized into mutually
exclusive categories of low, medium, and high levels using
previous Kidney Disease Outcomes Quality Initiative
(KDOQI)/KDIGO guideline targets, further illustrating
important potential interactions between components of
CKD-MBD in terms of risk prediction for death or cardio-
vascular events. This analysis, however, did not provide
guidance for treatment, because it is unknown whether
switching from risk classes parallels changes in incidence of
complications or mortality over time. Of note, biomarkers
such as bALP and 25(OH)vitamin D were also still considered
valuable, but as no new evidence has been published on their
account, recommendations remained unchanged from the
previous guideline (2009 Recommendations 3.1.3, 3.2.3).
Finally, therapeutic maneuvers aimed at improving 1
parameter often have unin tentional effects on other parame-
ters, as exemplied by the recent EVOLVE trial.
38
The guideline
Work Group considered it reasonable to take the context of
therapeutic interventions into account when assessing values of
phosphate, calcium, and PTH, and felt that it was important to
emphasize the interdependency of these biochemical param-
eters for clinical therapeutic decision making.
Based on these assumptions, it was also decided to split
previous 2009 Recommendation 4.1.1 into 2 new Recom-
mendations, 4.1.1 (diagnostic recommendation based on
accumulated observational evidence) and 4.1.2 (therapeutic
recommendation based mostly on RCTs).
Research recommendations
Prospective cohort studies or RCTs are needed to evaluate
whether changes in CKD-MBD risk marker patterns over time
associate with changes in risk (e.g., multiple interventions).
Prospective cohort studies or RCTs are needed to examine
whether biochemical abnormalities of CKD-MBD must be
weighed differently when induced by pharmacotherapy
compared with baseline values (e.g., past experience with
hemoglobin as risk predictor vs. active treatment to targets
by erythropoiesis-stimulating agents).
Investigations contributing to the understanding of the
usefulness of broblast g rowth factor 23 (FGF23) as a
complementary marker for treatment indications (e.g.,
phosphate-lowering therapies to halt CKD progression)
and direct treatment target (e.g., regression of left ventric-
ular hypertrophy [LVH]) should be undertaken.
4.1.2: In patients with CKD G3aG5D, we suggest lowering
elevated phosphate levels toward the normal range
(2C).
Rationale
As outlined above, since publication of the 2009 KDIGO
CKD-MBD Guideline, additional high-quality evidence now
links higher concentrations of phosphate with mortality
www.kisupplements.org chapter 4.1
Kidney International Supplements (2017) 7, 1–59 25
among patients with CKD G3a to G5 or after trans-
plantation
3948
(Supplementary Tables S49S51, S53S55),
although some studies did not conrm this association.
49,50
However, trial data demonstrating that treatments that lower
serum ph osphate will improve patient-centered outcomes are
still lacking, and therefore the strength of this recommendation
remains weak (2C). The rationale of interventions, therefore, is
still only based on epidemiological evidence as described above
and biological plausibility pointing to possible phosphorus
toxicity as recently summarized.
51
Three recent historical
cohort analyses from DOPPS, ArMORR, and COSMOS were
not eligible for this evidence-based review ; however, it is
noteworthy that these analyses suggeste d that those dialysis
patients who had been prescribed phosphate-binder therapy
showed improved survival.
5254
It is important to note that
phosphate-binder prescription was associate d with better
nutritional status. Indeed, correction for markers of nutritional
status in the DOPPS study did mitigate the strength of the
association, yet a statistically signicant benet persisted. In
addition, propensity scoring attempting to correct for selection
bias and subgroup analysis applied by Isakova et al.
53
in the
ArMORR cohort suggested robustness of the benecial nd-
ings for those treated with phosphate binders. However, re-
sidual confounding still cannot be completely ruled out, and
due to the nature of the observational data, these studies did
not affect the current recommendation.
Methods to prevent the development of hyper-
phosphatemia essentially includ e dietary modication, the
use of phosphate-lowering therapy, and intensied dialysis
schedules for those with CKD G5D. In the 2009 KDIGO
Guideline it was suggeste d to maintain serum phosphate in
the normal range in the predialysis setting and lower serum
phosphate toward the normal range in patients on dialysis.
Interestingly, in the prospective observational COSMOS
study cohort of HD patients (Supplementary Tables S49S51,
S53S55), the best patient survival was observed with serum
phosphate close to 4.4 mg/dl (1.42 mmol/l).
55
The previous recommendation suggested that clinicians
maintain serum phosphate in the normal range fo r patients
with CKD G3a to G3b and G4. The Work Group reevaluated
the evidence underlying this assu mption. The majorit y of
studies (Supplementary Table S49) found ph osphate to be
consistently associated with excess mortality at levels above
and below the limits of normal, but not in the normal
range.
4043,47,48,56,57
This nding is in line with the previously
found U-shaped relation of phosphate with mortality risk in
dialysis patients.
58
However, a recent trial comparing placebo
with active phosphate-binder therapy in predialysis patients
(CKD G3bG4) with a mean baseline phosphate concentration
of 4.2 mg/dl (1.36 mmol/l), found a minimal decline in serum
phosphate, no effect on FGF23, and increases in coronary
calcication scores for the active treatment group
59
calling
into question the efcacy and safety of phosphate binding in
this population, with normal phosphate concentration prior to
initiation of binder treatment (Supplementary Tables S1924).
In this analysis, all phosphate binders were analyz ed
collectively, and the study was underpowered to detect differ-
ences between phosphate binders. Although the data suggested
that the observed increase in coronary artery calcication
(CAC) was mainly driven by the group treated with calcium-
containing phosphate binders, those treated with calcium-
free binders had no advantage over placebo in terms of pro-
gression of CAC. In addition, a well-executed mineral balance
study in predialysis patients using calcium-containing phos-
phate binders demonstrated the absence of any effect on
phosphate balance (while showing in the short term a positive
calcium balance).
60
The second principal option to control phosphate in pre-
dialysis patients is dietary restriction, as will be addressed in
Recommendation 4.1.8. However, in both the NHANES and
MDRD cohorts that examined the general population and
advanced CKD, respectively, dietary intake or intervention to
reduce dietary phosphate intake as assessed by either ur inary
excretion or dietary recall had only minimal effects on serum
phosphate.
61,62
It is unknown whether this minimal decline in
serum phosphate concentrations or the more robust lower
phosphate intake translates into benecial clinical outcome. A
subsequent analysis of the MDRD study found no impact of
low phosphate intake as compared with hig her intake on car-
diovascular disease or all-cause mortalit y.
63
It needs to be noted
that in this study baseline phosphate levels were normal on
average, so results are possibly not applicable to CKD patients
with progressively or persistently elevated serum phosphate
(see rationale for Recommendation 4.1.5).
Taken together, the key insights from these data were: (i)
the association between serum phosphate and cli nical
outcome is not monotonic; (ii) there is a lack of demon-
strated efcacy of phosphate binders for lowering serum
phosphate in patients with CKD G3a to G4; (iii) the safety of
phosphate binders in this population is unproven; and (iv)
there is an absence of data showing that dietary phosphate
restriction improves clinical outcomes. Consequently, the
Work Group has abandoned the previous suggestion to
maintain phosphate in the normal range, instead suggesting
that treatment be focused on patients with hyper-
phosphatemia. The Work Group recognizes that preventing,
rather than treating, hyperphosphatemia may be of value in
patients with CKD G3a to G5D, but acknowledges that current
data are inadequate to support the safety or efcacy of such an
approach and encourages research in this specic area.
Only 2 RCTs have examined phosphate-lowering therapy in
children with CKD or on dialysis;
64,65
due to the low number
of patients and short follow-up, both studies did not meet
literature inclusion criteria set a priori together with the
evidence review team (ERT). The rst RCT examined
biochemical endpoints only and showed equivalent phosphate
control with calcium acetate and sevelamer hydrochloride in
an 8-week cross-over trial.
65
In the second, 29 children were
randomized to different combinations of phosphate binders
and vitamin D analogs; bone biopsies suggested that the
sevelamer group had reduced bone formation versus baseline
at 8-month follow-up, but numbers were too small for
chapter 4.1 www.kisupplements.org
26
Kidney International Supplements (2017) 7, 1–59
comparison versus the calcium carbonatetreated group.
64
Several studies in children on dialysis have shown an associ-
ation between high phosphate levels and increased vessel
thickness,
6668
vessel stiffness
68,69
and CAC.
67,68,70,71
In young
adults on dialysis, the CAC score was shown to double within
20 months, and progression was associated with higher serum
phosphate levels.
71
Research recommendations
RCTs for controlling hyperphosphatemia in patients with
CKD G3a to G5D, with appropriate follow-up and power,
should be conducted to assess various phosphate-lowering
therapy strategies for reducing the incidence of patient-
level endpoints (e.g., CKD progression) in children and
adults.
RCTs of low and high dietary phosphate intake in patients
in CKD G3a to G5 should be conducted to test the hy-
pothesis that high dietary phosphate intake increases car-
diovascular risk either directly or indirectly through
induction of FGF23.
If the feasibility of a placebo-controlled trial is threatened
due to perceived lack of equipoise (i.e., unethical to not
lower elevated serum phosphate levels, despite the lack of
high-quality data), a prospective trial comparing 2 different
phosphate targets in patients with CKD G3 a to G5D is
encouraged.
RCTs should be conducted in normophosphatemic CKD
patients in order to test the hypothesis that active
compensatory mechanisms to counter balance increased
phosphate intake (such as increases in FGF23 and PTH) are
associated with poorer clinical outcome, des pite compara-
ble serum phosphate concentration. Interventions could
include dietary phosphate restriction, phosphate-binder
therapy, novel compounds to limit phosphate uptake, or a
combination thereof.
4.1.3: In adult patients with CKD G3aG5D , we suggest
avoiding hypercalcemia (2C). In children with CKD
G3aG5D, we suggest maintaining serum calcium in
the age-appropriate normal range (2C).
Rationale
As is the case for phosphate, novel epidemiological evidence
linking higher calcium concentrations to increased
mortality in adults with CKD has accumulated since the
2009 KDIGO CKD-MBD guideline (Supplementary
Tables S49S50, S52S55).
4244,47,50,60,7274
Moreover, and in
addition to previous observations,
48
novel studies link higher
concentrations of serum calcium to nonfatal cardiovascular
events.
75,76
This consistency justies the change of this
recommendation from 2D to 2C, although the overall evi-
dence base remains limited due to the lack of prospective
controlled trial data.
Hypocalcemia is a classical feature of untreated CKD, in
part secondary to diminished gastrointestinal (GI) uptake of
calcium due to vitamin D deciency.
77
Hypocalcemia
contributes to the pathogenesis of secondary hyperparathy-
roidism (SHPT) and renal osteodystrophy. Therefore, the
previous recommendation suggested maintaining serum cal-
cium in the normal range, including the correction of hy-
pocalcemia. A more recent retrospective observational
analysis of a large dialysis cohort conrmed the association
between hypocalcemia and mortality risk.
50
Two other recent
obser vations, however, raised doubt within the KDIGO
guideline Work Group about the generalizabilit y of the sug-
gestion to correct hypocalcemia. The rst is the potential
harm for some adults associated with a positive calcium
balance (while serum calcium levels do not necessarily reect
calcium balance).
78,79
The second observation is that the
prevalence of hypocalcemia may have increased after the
introduction of calcimimetics (cinacalcet) in patients on
dialysis.
38,80,81
The clinical implications of this increased
incidence of low calcium due to the therapeutic institution of
a calcimimetic is uncertain, but may be less harmful. With
regard to the intention-to-treat (ITT) population of the
EVOLVE trial, no negative signals were associated with the
persistently low serum calcium levels in the cinacalcet arm of
the trial. Retaining the 2009 KDIGO Guideline on this issue
would support the concept that patients developing hypo-
calcemia during calcimimetic treatment require aggressive
calcium treatment. Given the unproven benets of this
treatment and the potential for harm, the Work Group
emphasizes an individua lized approach to the treatment of
hypocalcemia rather than recommending the correction of
hypocalcemia for all patients. However, signicant or symp-
tomatic hypocalcemia should still be addressed. Symptomatic
or severe hypocalcemia may benet from correction to
prevent adverse consequences such as bone disease, hyper-
parathyroidism, and QTc interval prolongation.
Childhood and adolescence are critical periods for bone
mass accrual: in healthy children the calcium content of the
skeleton increases from w25 g at birth to w1000 g in adults,
and w25% of total skeletal mass is laid down during the 2-
year interval of peak height velocity.
82
The mean calcium
accretion rate in healthy pubertal boys and girls peaks at 359
and 284 mg/d, respectively.
83
The updated evidence review
identied a prospective cohort study in 170 children
and adolescents with CKD G2 to G5D (Supplementary
Table S49S50, S52 S55) that showed that lower serum cal-
cium levels were independently associated with lower cortical
volumetric BMD Z-scores.
19
Over 1 year of follow-up in 89
children, a change in the cortical BMD Z-score positively
correlated with baseline calcium (P ¼ 0.008) and increase in
calcium (P ¼ 0.002) levels, particularly in growing children.
During the 1-year follow-up, 6.5% of children sustained a
fracture. Notably, a lower cortical BMD Z-score predicted
future fractures: the HR for fracture was 1.75 (95% CI: 1.15
2.67; P ¼ 0.009) per SD decrease in baseline BMD.
19
Thus, the Work Group recognizes the higher calcium re-
quirements of the growing skeleton and suggests that serum
calcium levels are maintained in the age-appropriate normal
range in children and adolescents.
www.kisupplements.org chapter 4.1
Kidney International Supplements (2017) 7, 1–59 27
Research recommendations
Calcium balance study in dialysis patients should be pur-
sued at baseline versus after start of calcimimetic treatment
(with and without calcium supplementation, adaptations in
dialysate calcium concentrations, and/or concomitant active
vitamin D analog treatment).
RCTs in children and adolescents w ith CKD should be
conducted to determine whether calcium-based phosphate
binders, as compared with calcium-free phosphate binders,
promote bone accrual (as measured by bone density and
structure, and fractures), and to determine the impact of
phosphate binders on arterial calcication in the context of
the high calcium requirement of growing bones.
4.1.4: In patients with CKD G5D, we suggest using a dial-
ysate calcium concentration between 1.25 and 1.50
mmol/l (2.5 and 3.0 mEq/l) ( 2C).
Rationale
Based on the available evidence, the 2009 Work Group
considered that a dialysate calcium concentration of 1.25
mmol/l (2.5 mEq/l) would yield neutral calcium balance, but
this statement was subsequently challenged by kinetic
modeling studies.
84
Two relevant new RCTs are available concerning this topic
(Supplementary Tables S13S18).
85,86
In the study by Spa-
sovski et al.,
86
the effects of 2 different dialysate calcium
concentrations were examined in patients with adynamic
bone disease, and the lower dialysate calcium (1.25 mmol/l
[2.5 mEq/l]) was found to improve bone and mineral pa-
rameters compared with the higher concentration of 1.75
mmol/l (3.5 mEq/l). Their data conrmed the results of
previous papers and also support individualization of dialy-
sate calcium concentrations as recommended previously by
the Work Group. The comparator in this study, however, was
a high dialysate calcium concentration of 1.75 mmol/l
(3.5 mEq/l), leaving open the possibility that lower levels
of dialysate calcium (>1.25 mmol/l [2.5 mEq/l] but
<1.75 mmol/l [3.5 mEq/l]) would be equally benecial.
Ok et al. randomized 425 HD patients with iPTH levels
< 300 pg/ml (32 pmol/l) and baseline dialysate calcium
concentrations between 1.5 and 1.75 mmol/l (3.03.5 mEq/l)
to concentrations of either 1.25 mmol/l (2.5 mEq/l) or
1.75 mmol/l (3.5 mEq/l).
85
Lowering dialysate calcium levels
slowed the progression of CAC and improved biopsy-proven
bone turnover (low bone turnover decreased from 85.0% to
41.8%) in this cohort of patients on HD. In this trial, the
comparative effects of a 1.5 mmol/l (3.0 mEq/l) calcium
concentration were not addressed.
Retrospective observational data by Brunelli et al.
87
sug-
gested safety concerns (i.e., heart failure events, hypotension)
associated with the default use of dialysate calcium
concentrations < 1.25 mmol/l (2.5 mEq/l). Conversely, at the
high end of dialysate calcium concentration (1.75 mmol/l
[3.5 mEq/l]), Kim et al.
88
found increased risk for all-cause
mortality and cardiovascular or infection-related
hospitalization in incident HD patients for hig h dialysate cal-
cium. However, observational studies, in general, may not be
sufcient to warrant changes to treatment recommendations.
Patients with mild hypocalcemia might potentially even
have a positive calcium mass transfer when dialyzed against a
concentration of 1.25 mmol/l (2.5 mEq/l), but no such
metabolic balance studies exist. Taken together, the Work
Group felt that this recommendation remains valid as written
in 2009 and that there is no new evidence justifying a change
in the wording. However, additional studies of better quality
are now available, and as such the evidence grade has been
upgraded from 2D to 2C.
Research recommendation
Calcium balance studies should be performed with non
calcium-containing versus calcium-containing phosphate
binders, and vitamin D sterols versus cinacalcet in different
calcium dialysate settings. These studies should include
children and adolescents and assess calcium balance in the
context of skeletal calcium accrual.
4.1.5: In patients with CKD G3aG5D, decisions about
phosphate-lowering treatment should be based on
progressively or persistently elevated serum phos-
phate (Not Graded).
Rationale
With regard to 2017 Recommendation 4.1.5 (formerly 2009
Recommendation 4.1.4), the previous 2009 KDIGO CKD-
MBD guideline commented that available phosphate
binders are all effective in the treatment of hyper-
phosphatemia, and that there is evidence that calciu m-free
binders may favor halting progression of vascular calcica-
tions compared with calcium-containing binders.
30
Concerns
about calcium balance and uncertainties about phosphate
lowering in CKD patients not on dialysis, coupled with
additional hard endpoint RCTs and a systematic review
(comparing effects on mortality for calcium-free vs. calcium-
containing phosphate binders), resulted in the decision to
reevaluate this recommendation.
Based on new pathophysiological insights into phosphate
regulation and the roles of FGF23 and (soluble) Klotho in
early CKD, clinical studies had been initiated investigating
phosphate-lowering therapies in CKD patients in whom
hyperphosphatemia had not yet developed. Here, the
concept of early phosphate retention, possibly represented
by increases in FGF23 serum or plasma concentrations, was
the focus of scientic attention. The most notable RCT
was performed by Block et al.
59
In this study, predialysis
patients (CKD G3bG4) with mean baseline serum phos-
phate concentrations of 4.2 mg/dl (1.36 mmol/l) were
exposed to 3 different phosphate binders (sevelamer,
lanthanum, or calcium acetate) versus matching placebos, in
order to explore effects on serum phosphate levels, urinary
phosphate excretion, serum FGF23 levels, vascular calcica-
tion, bone densit y, etc., w ith a 9-month follow-up
chapter 4.1 www.kisupplements.org
28
Kidney International Supplements (2017) 7, 1–59
(Supplementary Tables S19S24). While there was a small
decrease in serum phosphate concentrations (for those allo-
cated to active treatment) and a 22% decrease in urinary
phosphate excretion (sugge sting adherence to therapy), no
differences in changes in FGF23 levels were observed versus
placebo, as already discussed in Recommendation 4.1.2. In
contrast to the authors expectations, progression of coronary
and aortic calcication was obser ved with active phosphate-
binder treatment, while there was no progression in the
placebo arm. Subgroup analysis suggested that this negative
effect was accounted for by calcium acetate treatment, but
neither calcium-free binders were superior to placebo with
regard to this surrogate endpoint.
This study was further supported by another meta bolic
study in a small group of patients with CKD G3b to G4, in
whom the addition of 3 500 mg calcium carbonate to meals
containing 1 g of calcium and 1.5 g of phosphorus per day did
not affect baseline neutral phosphate balance, but caused a
signicant ly positive calcium balance,
60
at least in the short
term. Due to its low number of patients and short duration , this
study did not ful ll the predened inclusion criteria for full
evidence review. Nevertheless, in the Work Groups opinion,
this well-performed metabolic study may present a plausible
and relevant safety signal, and thus should be mentioned here.
Both Block et al.
59
and Hill et al.
60
studied subjects with
essentially normal phosphate concentrations at baseline.
Thus, there may be 2 key messages from these studies. First,
normophosphatemia may not be an indication to start
phosphate-lowering treatments. Second, the concept that not
all phosphate binders are interchangeable must be noted.
Whether disproportional elevations in FGF23 serum con-
centrations may become a signal in order to start phosphate-
lowering therapies in early CKD will need to be investigated
in appropriate trial settings.
Considering these insights, especially regarding CKD pa-
tients not on dialysis, and as already suggested in the rationale
of Recommendation 4.1.2, the Work Group felt that the
updated guideline should clarify that phosphate-lowering
therapies may only be indicated in the event of progressive
or persistent hyperphosphatemia, and not to prevent
hyperphosphatemia. When thinking about risk-benet ratios,
even calcium-free binders may poss ess a potential for harm
(e.g., due to side effects such as GI distress and binding of
essential nutrients). The broader term phosphate-lowering
therapies instead of phosphate-binding agents was intro-
duced, because all possible approaches (i.e., binders, diet, and
dialysis) can be effective and because phosphate transport
inhibitors may expand the therapeutic armamentarium in the
not-so-distant future.
There have been no additional data since 2008 with regard
to safe phosphate level thresholds or hard endpoints (i.e.,
mortality, cardiovascular events, and progression of CKD)
from RCTs treating patients toward different phosphate (or
FGF23) targets. The previous qualiers (presence of other
components of CKD -MBD, concomitant therapies, side effect
prole) were deleted because the Work Group thought that
their consideration was self-evident. Diurnal variation of
serum phosphate concentrations was discussed as another
pathophysiologically relevant aspect of evaluation. While it was
felt that these variations in daily phosphate levels do affect the
accuracy of evaluations, the notion of variable timing for blood
sampling was considered unfeasible in clinical routine practice
and therefore not included in the guideline text.
Research recommendations
Prospective clinical trials studying the value of levels of
FGF23 (and possibly soluble Klotho) as indicators for
establishing phosphate-lowering therapies should be un-
dertaken; desirable endpoints should include: CKD pro-
gression, cardiovascular calcication, cardiovascular events,
and mortality.
See research recommendations following Recommendation
4.1.2.
4.1.6: In adult patients with CKD G3aG5D receiving
phosphate-lowering treatment, we suggest restrict-
ing the dose of calcium-based phosphate binders
(2B). In children with CKD G3aG5D, it is reason-
able to base the choice of phosphate-lowering
treatment on serum calcium levels (Not Graded).
Rationale
The Work Group thought that the new available data and the
changes applied to 2009 Recommendation 4.1.4 (now
Recommendation 4.1.5) suggested a need to revise the 2009
Recommendation 4.1.5 (now Recommendation 4.1.6). The
balance study by Hill et al.
60
supported results reported by
Spiegel and Brady
79
in normophosphatemic adults with CKD
G3b to G4, which suggested potential harms of liberal cal-
cium exposure in such cohorts, but due to their study designs
were not eligible for full evidence review by the ERT. The RCT
by Block et al.
59
in a much larger, similar cohort and 2
additional RCTs in hyperphosphatemic CKD patients have
added hard endpoint data when prospectively comparing the
calcium-free binders, mostly sevelamer, with calcium-
containing binders in predialysis or dialysis adult patients,
respectively (Supplementary Tables S19S24)
59,89,90
These
results were also supported by results from recent systematic
reviews;
9194
however, because the evidence review team
(ERT) had considered all includ ed studies separately and
individually during this updat e process, the se meta-analyses
did not have addit ional bearing on the decision making by
the Work Group.
Overall, the Work Group determined that there is new
evidence suggesting that excess exposure to calcium through
diet, medications, or dialysate may be harmful across all GFR
categories of CKD, regardless of whether other cand idate
markers of risk such as hypercalcemia, arterial calcication,
adynamic bone disease, or low PTH levels are also present.
Therefore, these previous qualiers in the 2009 KDIGO
recommendation were deleted, acknowledging that they may
still be valid in high-risk scenarios.
www.kisupplements.org chapter 4.1
Kidney International Supplements (2017) 7, 1–59 29
Di Iorio et al. reported RCTs in both predialysis and dialysis
patients showing signicant survival benets over a 3-year
interval for patients treated with sevelamer versus calcium-
containing binders (Supplementary Tables S19S24).
89,90
Both studies w ere analyzed by the ERT and were
graded as relevant RCTs with a moderate risk of bias
(Supplementary Tables S23 and S24), leading to a 2B
recommendation. Overall, the ndings from all identied
studies seemed to show either a potential for bene toran
absence of harm associated with calcium-free phosphate-
binding agents to treat hyperphosphatemia compared with
calcium-based agents (Supplementary Tables S20 and S21).
The wording in Recommendation 4.1.6 of restricting the
dose of calcium-based phosphate binders was retained from
previous 2009 Recommendation 4.1.5; however, the qualier
that the recommendation applies to patients with persistent
or recurrent hypercalcemia was removed. Given the fact of 2
reasonably large RCTs demonstrating mortality risks associ-
ated w ith calcium-containing binder treatment, it was debated
within the Work Group whether the recommendation should
be stronger, possibly using avoid instead of restrict. How-
ever, some members of the Work Group felt that available
evidence does not conclusively demonstrate that calcium-free
agents are superior to calcium-based agents. In addition,
none of the studies provided sufcient dose threshold infor-
mation about calcium exposure, nor did they give information
on the safety of moderately dosed calcium-containing binders
in combination therapies. Finally, because KDIGO guidelines
are intended for a global audience and calcium-free agents are
not available or affordable in all jurisdictions, recommending
against the use of calcium-based binders would imply that no
treatment is preferable to using calcium-based agents. Despite
the understandable clinical desire to have numeric targets and
limits, the Work Group could not make an explicit recom-
mendation about a maximum dose of calcium-based binders,
preferring to leave this to the judgment of individual physicians
while acknowledging the potential existence of a safe upper
limit of calcium dose. Of note, 2 short-term studies in stable
CKD patients not on dialysis found that positive calcium
balance may occur with total intakes as low as 800 and
1000 mg/d, respectively.
60,79
Such short-terms results are
informative but not conclusive, and decisions must be indi-
vidualized for each patient.
The recent availability of iron-containing phosphate binders
was discussed within the Work Group but did not affect the
recommendations given the absence of data on long-term pa-
tient-centered outcomes in the published phase 3 trials.
9597
All of the above studies were limited to adults. Importantly,
concerns regarding the adverse effects of excess exposure to
calcium through diet, medications, or dialysate may not be
generalizable to children. Skeletal growth and development are
characterized by rapid calcium accrual,
83
as described in
Recommendation 4.1.3. Furthermore, recent studies demon-
strated that bone accrual continues into the third decade of
life in healthy individuals, well beyond cessation of linear
growth.
98,99
Of relevance to adolescents with CKD, bone accrual
between ages 18 and 24 was especially pronounced among those
with late puberty.
100
Therefore, studies of calcium- and non
calcium-containing binders and other therapies that impact
calcium balance should consider the needs of the developing
skeleton. The observation that serum calcium levels were
positively associated with increases in BMD in children with
CKD, and that this association was signicantly more pro-
nounced with greater linear growth velocity,
19
illustrates the
unique needs of the growing skeleton (see Recommendation
4.1.3). Lastly, a recent prospective cohort study in 537 children
with predialysis CKD demonstrated that phosphate-binder
treatment (calcium-based in 82%) was associated with
decreased risk of incident fractures (HR: 0.37, 95% CI: 0.15
0.91), independent of age, sex, eGFR, and PTH levels.
101
Although this study did not meet the criteria for inclusion in
the evidence review, it highlights the need for additional studies
in children. There is a lack of data suggesting adverse effects of
excess exposure to calcium through diet, medications or dial-
ysate in children. The Work Group concluded that there was
insufcient evidence to change this recommendation in chil-
dren, who may be uniquely vulnerable to calcium restriction.
Research recommendations
Calcium and phosphate balance studies should be con-
ducted using different calcium-based binder doses and
combinations with calcium-free binders in hyper-
phosphatemic patients across all GFR categories of CKD.
RCTs assessing the effect of iron-based phosphate binders on
patient-centered and surrogate outcomes across all GFR cat-
egories of CKD should be undertaken; comparators should be
placebo, calcium-based binders, or other calcium-free binders.
RCTs using phosphate transport inhibitors (e.g., nicotin-
amide,
102
tenapanor
103
)asadd-on treatments in patients
with resistant hyperphosphatemia should be investigated.
Prospective clinical and balance studies should examine the
role of magnesium as a phosphate binder, with regard to
patient-centered outcomes, calcication, and cardiovascular
event rates.
RCTs in children and adolescents with CKD should be
conducted to determine whether calcium-based phosphate
binders, as compared with calcium-free phosphate binders,
promote bone accrual (as measured by bone density and
structure, and fractures), and to determine the impact of
phosphate binders on arterial calcication in the context of
the high calcium requirement of growing bones.
4.1.8: In patients with CKD G3aG5D, we suggest limiting
dietary phosphate intake in the treatment of hyper-
phosphatemia alone or in combination with other
treatments (2D). It is reasonable to consider phos-
phate source (e.g., animal, vegetable, additives) in
making dietary recommendations (Not Graded).
Rationale
There was no general controversy toward the 2009 KDIGO
CKD-MBD Guideline on dietary phosphate restriction as an
chapter 4.1 www.kisupplements.org
30
Kidney International Supplements (2017) 7, 1–59
important standard of practice to lower elevated phosphate
levels, but previous 2009 recommendatio n 4.1.7 (now 4.1.8)
on limiting dietary phosphate intake was considered vague,
especially with regard to new evidence on different phosphate
and phosphoprotein sources. Within this guideline update,
predened criteria on study duration and cohort size pro-
hibited inclusion of some study reports for full evidence
review. Nevertheless, the Work Group felt that some of these
reports presented safety signals demanding a brief discussion.
As summar ized in Supplementary Tables S25S30 , only 2
studies on this topic in dialysis patients fullled the evidence
review criteria.
104,105
Both studies investigated the impact of
intensied versus routine dietary counseling on serum
phosphate levels after a follow-up of 6 months. In both
studies, the intensied counseling groups more successfully
reached the laboratory targets; however, no hard endpoints
were documented. Accordingly, the quality of evidence
for outcome was rated as very low. Similarly, a recent
Cochrane review concluded that there is low-quality evidence
that dietary interventions positively affect CKD-MBD
biomarkers.
106
The daily phosphate intake for a typical American diet
varies with age and gender. A majority of young to middle-
aged men take in more than 1600 mg/d, whereas women in
the same age groups take in about 1000 mg/d.
107
On a
global scale, there are quite signicant differences in diet
compositions to be considered. Estimates of dietary phos-
phate from food composition tables likely underestimate the
phosphate content because they may mostly reect the
natural phosphate content of foods that are highest (e.g.,
dairy products, meats, poultry, sh, and grains). There are
actually 3 major sources of phosphates: natural phosphates
(as cellular and protein constituents) contained in raw or
unprocessed foods, phosphates added to foods during pro-
cessing, and phosphates in dietary supplements/medications.
Russo et al.
108
assessed the effect of dietary phosphate
restriction on the progression of CAC. This study was not
designed to compare the efcacy of phosphate binders against
dietary phosphate restriction. However, they found that di-
etary phosphate restriction alone did not lead to a decrease in
urinary phosphate excretion, nor did it prevent progression of
CAC. However, the urine data cast doubt on compliance with
the diet, and there was no control group on a normal or high-
phosphate diet.
Aggressive dietary phosphate restriction is difcult because
it has the potential to compromise adequate intake of other
nutrients, especially protein. Zeller et al.
109
showed that the
restriction of dietary protein and phosphate could be achieved
with maintenance of good nutrition status with intense
counseling. They demonstrated that dietary protein/phos-
phate restriction resulted in a signicant reduction in urinary
phosphate excretion when compared with a control diet.
Dietary supplements and over-the-counter or prescription
medications are hidden sources of phosphate. They may
contain phosphate salts within their inactive ing redients. The
data on the amount of phosphate in oral medications and
vitamin/mineral supplements is limited, but they have the
potential to contribute signicantly to the ph osphate load
considering the number of medications CKD patients are
required to take.
110,111
Another consideration for modication of dietary phos-
phate and control of serum phosphate is the bioavailability
of phosphorus in different foods based on the formorganic
versus inorganic sources of phosphate. Animal- and plant-
based foods contain the organic form of phosphate. Food
additives contain inorganic phosphate. About 40% to 60% of
animal-based phosphate is absorbed, depending on the GI
vitamin D receptor activations. Plant phosphate, mostly
associated with phytates, is less absorbable (generally 20%
50%) in the human GI tract. It be hooves the dietitian and
other interdisciplinary staff to include education about the
best food choices as they relate to absorbable phosphate.
Additionally, it is important for patients to be guided toward
fresh and homemade foods rather than processed foods in
order to avoid additives.
Organic phosphate in such plant foods as seeds and
legumes is less bioavailable because of limited GI absorption
of phytate-based phosphorus. In this context, Moe et al.
35
recently demonstrated that a vegetable-based diet showed
signicantly lower phosphate absor ption versus a meat-based
diet with similar phosphate content. Inorganic phosphate is
more readily absorbed, and its presence in additive-laden
processed, preserved, or enhanced foods or soft drinks is
likely to be underreported in nutrient databases. Hence, the
phosphate burden from food additives is disproport ionately
high relative to natural sources that are derived from organic
(animal and vegetable) food proteins, and these additives are
almost completely absorbed in the GI tract. For example,
Benini et al. showed that foods that contain phosphate
additives have a phosphorus content nearly 70% higher than
those that do not contain additives.
112
Sherman and Mehta
also demonstrated that phosphate contents between unpro-
cessed and processed meat or poultry may differ by more than
60%, and thus the absorbable phosphate may even be 2 to 3
times higher per weight in processed food.
113
In contrast, many of the foods that are traditionally labeled
as high phosphorus may be more acceptable with the
knowledge that the phosphorus is absorbed more slowly and
not as efciently. For example, beans and nuts have always
been listed as very high in phosphorus; however, considering
their lower absorption rate, they may be acceptable as protein
sources, if they are not too high in other nutrients such as
potassium.
The amount of phosphorus contributed by food intake is
increasing with current and new processing practices that
utilize phosphorus-containing ingredients, including popular
foods such as restructured meats (formed, pressed, rolled, and
shaped for ease of preparation and ingestion), processed and
spreadable cheeses, instant products (puddings, sauces),
frozen breaded products, and soft drinks.
114
Phosphate ad-
ditives are also widely used in fast foods and convenience
foods that are fully or partially pre-made or instant.
110
www.kisupplements.org chapter 4.1
Kidney International Supplements (2017) 7, 1–59 31
Various types of nutrition education have had mixed
results for control of serum phosphate. Intense education
focusing on phosphate intake has been useful to reduce
retention in some studies.
105,115
A simple education tool on
how to read food labels and look for PHOS (the study
acronym) was successful in helping dialysis patients reduce
their phosphate intake. A magnifying glass was provided to
help patients read labels,
116
as well as instructions available to
guide better choices in fast-food restaurants. Other studies
have had less favorable results.
117
Taken together, these insights led the Work Group to the
decision to not change the principal recommendation, but to
add a qualier statement suggesting that phosphate sources
should be better substantiated and patient education should
focus on best choices. Finally, it must be emphasized that
efforts to restr ict dietary phosphate must not compromise
adequate protein intake.
Research recommendations
RCTs comparing low, medium, and high phosphate intake on
phosphate metabolism and homeostasis, including responses
concerning FGF23, PTH, calcication, and CKD progression,
in patients in CKD G3b to G4, should be performed.
In such study designs, the role of the phosphate quality
should be studied: vegetable versus meat versus additive
sources.
Kinetic and balance studies on the uptake of phosphate
additives in dialysis patients should be per formed.
Prospective trials identifying the most effective phosphate-
lowering approach (benet-risk-cost ratio) should be per-
formed across all CKD GFR categoriessuch as how best
to combine phosphate binders, phosphate transport in-
hibitors and diet (plus dialysis treatment in CKD G5D)
with appropriate patient-centered and surrogate endpoints
in mind (e.g., calcication, FGF23 levels, and LVH) .
chapter 4.1 www.kisupplements.org
32
Kidney International Supplements (2017) 7, 1–59
Chapter 4.2: Treatment of abnormal PTH levels in
CKD-MBD
4.2.1: In patients with CKD G3aG5 not on dialysis, the
optimal PTH level is not known. However, we suggest
that patients with levels of intact PTH progressively
rising or persistently above the upper normal limit
for the assay be evaluated for modiable factors,
including hyperphosphatemia, hypocalcemia, high
phosphate intake, and vitamin D deciency (2C).
Rationale
The pathogenesis of SHPT is complex and driven by several
factors, including vitamin D deciency, hypocalcemia, and
hyperphosphatemia. Elevated FGF23 concentrations exacer-
bate SHPT through further reductions in 1,25(OH)
2
vitamin
D (calcitriol) levels. Calcitriol deciency results in decreased
intestinal absorption of calcium and may lead to hypocalce-
mia, a major stimulus for PTH secretion. This leads to
parathyroid cell proliferation, contributing to SHPT. The
incidence and severity of SHPT increases as kidney function
declines and can lead to signi cant abnormalities in bone
mineralization and turnover.
The 2009 KDIGO CKD-MBD Guideline recommended
addressing modiable risk factors for all patients with a PTH
level above the upper limit of normal for the assay used.
30
Unfortunately, there is still an absence of RCTs that dene
an optimal PTH level for patients with CKD G3a to G5, or
clinical endpoints of hospitalization, fracture, or mortality.
The Work Group felt that modest increases in PTH may
represent an appropriate adaptive response to declining kid-
ney function, due to its phosphaturic effects and increasing
bone resistance to PTH,
118
and have revised this statement to
include persistently above the upper normal PTH level as
well as progressively rising PTH levels, rather than simply
above the upper normal limit as in the 2009 KDIGO
Guideline. Thus, treatment should not be based on a single
elevated value.
Although the optimal PTH is not known, the Work Group
felt that rising PTH levels in CKD G3a-G5 warrant exami-
nation of modiable factors , such as vitamin D insufci ency
or deciency, hypocalcemia, and hyperphosphatemia. In the
interval since the 2009 KDIGO Guideline, 1 eligible RCT
examined the impact of cholecalciferol supplementation
(Supplementary Table S31) and 3 examined the impact of
phosphate binders on PTH levels in the nondialysis
CKD population. Oksa et al.
119
reported an RCT of a
high (20,000 international units [IU]/wk) versus low
(5,000 IU/wk) dose of cholecalciferol supplementation in 87
adults with CKD G2 to G4 (Supplementary Tables S31S36).
Serum 25(OH) v itamin D levels increased signicantly in
both groups and were signicantly greater in the high-dose
arm at the completion of the 12-month intervention. PTH
levels decreased signicantly in both groups; however, the
PTH levels did not differ signicantly between groups at the
completion of the study. In this context, Recommendation
3.1.3 on native vitamin D supplementation remains valid
from the previous 2009 guideline publication.
Three recent RCTs in the nondialysis CKD population
evaluated phosphate binders and their effects on surrogate
endpoints, such as vascular calcication, arterial compliance,
left ventricular mass, and BMD, as well as calcium, phosphate,
and PTH levels. Two RCTs compared sevelamer with placebo
(Supplementary Tables S31 S36), the rst in 109 nondiabetic
CKD G3a to G3b patients
120
and the second in 117 CKD
patients with a mean eGFR of 36 17 ml/min/1.73 m
2
.
121
The studies were conducted over 36 weeks and 24 months,
respectively, and neither study demonstrated signicant dif-
ferences in PTH levels between sevelamer and placebo groups.
Another RCT involving 148 CKD patients (eGFR: 2045 ml/
min/1.73 m
2
) compared placebo w ith 3 different phosph ate
binders (calcium-based, lanthanum, and sevelamer) over a
9-month per iod and reported that PTH levels remained stable
in those on active therapy (combined phos phate-binder
groups) but increased by 21% in the placebo group
(P ¼ 0.002)
59
(Supplementary Table S33).
In the updated recommendation, an additional modiable
risk factor, high phosphate intake, was added because of the
increasing recognition that excess phosphate intake does not
always result in hyper phosphatemia, especially in early CKD,
and that high phosphate intake may promote SHPT. While
dietary phosphate, whether from food or additives, is modi-
able, better methods for assessment of dietary phosphate
intake are required.
4.2.2: In adult patients with CKD G3aG5 not on dialysis,
we suggest that calcitriol and vitamin D analogs not
be routinely used (2C). It is reasonable to reserve the
use of calcitriol and vitamin D analogs for patients
with CKD G4G5 with severe and progressive
hyperparathyroidism (Not Graded).
In children, calcitriol and vitamin D analogs may be
considered to maintain serum calcium levels in the
age-appropriate normal range (Not Graded).
www.kisupplements.org chapter 4.2
Kidney International Supplements (2017) 7, 1–59 33
Rationale
Prevention and treatment of SHPT is important because
imbalances in min eral metabolism are associated with CKD-
MBD and higher PTH levels are associated with increased
morbidity and mor tality in CKD patients. Calcitriol and other
vitamin D analogs have been the mainstay of treatment of
SHPT in individuals with CKD for many decades. The 2009
KDIGO CKD-MBD Guideline summarized multiple studies
demonstrating that administratio n of calcitriol or vitamin D
analogs (such as paricalcitol, doxercalciferol, and alfacalcidol)
resulted in suppression of PTH levels.
30
However, there was a
notable lack of trials demonstrating improvements in patient-
centered outcomes.
Multiple well-conducted RCTs cited in the 2009 guideline
reported benets of calcitriol or vitamin D analogs in treating
SHPT in patients with CKD G3a to G5; 2 primarily involved
biochemical endpoints,
122,123
and 2 evaluated bone histo-
morphometry.
124,125
Despite the lack of hard clinical end-
points, these data led to the original recommendation to treat
elevated PTH with calcitriol or vitamin D analogs early in
CKD to prevent parathyroid hyperplasia and its skeletal
consequences (2C). Although benets were predominantly
related to suppression of SHPT, adverse effects of hypercal-
cemia were noted to be of concern in the 2009 KDIGO CKD-
MBD Guideline.
30
The effects of vitamin D therapy on biochemical endpoints
in CKD have been previously documented, especially with
regard to reduced PTH levels. Numerous previous studies
have reported signicant reductions of PTH levels with cal-
citriol or vitamin D analogs in CKD G3a to G3b and G4 when
compared with placebo
123,125,126
and recent RCTs have also
demonstrated that vitamin D treatment effectively lowers
PTH levels in CKD G3a to G5.
127,128
Additional RCTs of calcitriol or vitamin D analog therapy
have been published since the 2009 KDIGO CKD-MBD
Guideline (Supplementary Tables S37S42). Two, in partic-
ular, demonstrated a signicantly increased risk of hypercal-
cemia in patients treated with paricalcitol, compared with
placebo, in the absence of benecial effects on surrogate
cardiac endpoints, as detailed below.
127,128
These results,
combined with the opinion that moderate PTH elevati ons
may represent an appropriate adaptive response, led the Work
Group to conclude that the risk-benet ratio of treating
moderate PTH elevations was no longer favorable and that
the use of calcitriol or vitamin D analogs should be reserved
for only severe and progressive SHPT.
The 2 recent RCTs were designed to detect potential benets
of calcitriol or vitamin D analogs on cardiac structure and
function, as measured by magnetic resonance imaging (MRI),
in adults with CKD (Supplementary Tables S37S42). The
rationale for these studies is that calcitriol and vitamin D an-
alogs act through the vitamin D receptor (VDR) to exert their
benets to inhibit PTH secretion, and the VDR is also present
in many tissues and organs including vascular smooth muscle,
endothelial cells, and the heart. The key evidence for changes in
Recommendation 4.2.2 predominantly came from these trials.
The rst study was a double-blind RCT by Thadhani et al.
(the PRIMO study), where participants with CKD G3a to G4,
mild to moderate LVH, and PTH levels between 50 and 300
pg/ml (5.332 pmol/l) were assigned to placebo (n ¼ 112) or
paricalcitol (n ¼ 115) to test the primary hypothesis that
paricalcitol will reduce left ventricular mass index (LVMI)
over a 48-week interval.
128
Paricalcitol was administered at a
dose of 2
m
g/d, with protocol-specied dose reduction to
1
m
g/d, if the serum calcium was > 11 mg/dl (2.75 mmol/l).
Baseline PTH levels were approximately 1.5 times the upper
limit of normal. The ITT analysis revealed that paricalcitol did
not reduce LVMI, nor did it modify diastolic function. Of
subjects on paricalcitol, the mean serum calcium increased by
0.32 mg/dl (0.08 mmol/l) (95% CI: 0.190.45 mg/dl; 0.05
0.11 mmol/l) versus a decrease by 0.25 mg/dl (0.06 mmol/l)
(95% CI: 0.37 to 0.12 mg/dl; 0.09 to 0.03 mmol/l) in
the placebo group. Hypercalcemia was dened as 2 consec-
utive measurements of serum calcium > 10.5 mg/dl (> 2.63
mmol/l), and the number of patients requiring dose
reductions from 2
m
g/d to 1
m
g/d and episodes of
hypercalcemia were more common in the paricalcitol group
(22.6%) compared with the placebo (0.9%) group.
In the second key study, a double-blind RCT by Wang et al.
(the OPERA study), subjects with CKD G3a to G5, LVH, and
PTH $ 55 pg/ml (5.83 pmol/l) were randomly assigned to
receive paricalcitol (n ¼ 30) or placebo (n ¼ 30).
127
The
primary endpoint was change in LVMI over 52 weeks. Base-
line PTH levels were approximately twice the upper limit of
normal. Change in LVMI did not differ signicantly between
groups, nor did secondary outcomes such as measures of
systolic and diastolic function. The median (interquartile
range) changes in serum calcium were 0.08 mmol/l (0.32 mg/
dl) (95% CI: 0.020.16 mmol/l; 0.080.64 mg/dl) and 0.01
mmol/l (0.04 mg/dl) (95% CI: 0.06 to 0.05 mmol/l; 0.24 to
0.2 mg/dl) in the paricalcitol and placebo arms, respectively.
Hypercalcemia, dened as any serum calcium > 2.55 mmol/l
(> 10.2 mg/dl), occurred in 43.3% and 3.3% of participants
in the paricalcitol and placebo arms, respectively. Of note,
70% of those who were hypercalcemic received concomitant
calcium-based ph osphate binders. Generally the hypercalce-
mia was mild and could be corrected by stopping the binder
without changing the paricalcitol dose.
Recent meta-analyses were largely conrmatory and sup-
ported the hypercalcemia risk association with calcitriol and
vitamin D analogs.
129,130
The evidence review identied 2 RCTs comparing par-
icalcitol with calcitriol (Supplementary Tables S37S42);
neither demonstrated differences in the incidence of hypercal-
cemia.
131,132
Coyne et al.
131
compared calcitriol (0.25
m
g/d)
with paricalcitol (1
m
g/d) in 110 patients with CKD G3a to G3b
and G4 and PTH > 120 pg/ml (12.7 pmol/l). The change in
PTH was comparable in the 2 arms (a decline of 52% vs. 46%)
over the 6-month trial, and the incidence of hypercalcemia was
very low in both groups (only 3 with paricalcitol and 1 with
calcitr iol). Further details regarding changes in biochemical
parameters are provided in Supplementary Tables S37S42.
chapter 4.2 www.kisupplements.org
34
Kidney International Supplements (2017) 7, 1–59
An alternative to calcitriol and its analogs is nutritional
vitamin D supplementation (cholecalciferol and ergo-
calciferol), which can also suppress PTH (especially in CKD
G3aG3b) and decrease hypercalcemia because the nor mal
homeostatic loops that suppress the CYP27B remain intact.
However, no studies of sufcient duration were identied in
this evidence review, and thus this therapy remains unproven.
Several studies have assessed the effect of PTH-lowering
comparing nutritional vitamin D supple ments and calcitriol
or vitamin D analogs.
133,134
However, these studies were not
identied in this evidence review because of their short
duration.
The use of extended-release calcifediol, a novel vitamin D
prohormone, to correct low serum 25(OH) vitamin D levels
and lower PTH has also been recently studied. This agent
reduces the catabolism of both 25(OH) vitamin D and
1,25(OH)
2
vitamin D and increases levels of both. An RCT of
429 patients with CKD G3a to G3b and G4 published after our
guideline systematic rev iew reported at least a 10% reduction of
intact PTH levels in 72% of participants after 12 months, with
no signicant impact on calcium, phosphate, or FGF23
levels.
135
No patient-level outcomes were reported, and thus
this study did not impact the current recommendation.
All of the above studies were conducted in adults. A recent
Cochrane review examined vita min D therapy for bone dis-
ease in children with CKD G2 to G5 on dialysis.
136
Bone
disease, as assessed by changes in PTH levels, was improved
by all vitamin D preparations regardless of route or
frequency of administration. The prospective cohort study
demonstrated that high PTH levels were independently
associated with reduced cortical BMD Z-scores at baseline
(P ¼ 0.002) and 1-year follow-up (P < 0.001).
19
High PTH
levels are associated with CAC in children on dialysis.
67,68
The
Cochrane review has not shown any signicant difference in
hypercalcemia risk with vitamin D preparations compared
with placebo, but 1 study showed a signicantly greater risk of
hypercalcemia with i.v. calcitriol administration.
136
No dif-
ference in growth rates was detected between different
vitamin D analogs or use of oral or i.v. vitamin D treat-
ments.
136
As noted in Recommendation 4.1.3, the Work
Group recommended that serum calcium should be main-
tained within age-appropriate reference range in children, and
given the association of high PTH levels with reduced bone
mineralization and increased vascular calcication, child ren
are likely to require calcitriol or other active vitamin D analog
therapy.
In summar y, the PRIMO and OPERA studies failed to
demonstrate improvements in clinically relevant outcomes
but demonstrated increased r isk of hypercalcemia. Accord-
ingly, the guideline no longer recommends routine use of
calcitriol or its analogs in CKD G3a to G5. This was not a
uniform consensus among the Work Group. It should be
noted that the participants in the PRIMO and OPERA trials
only had moderately increased PTH levels, thus therapy with
calcitriol and vitamin D analogs may be considered in those
with progressive and severe SHPT.
There are still no RCTs demonstrating benecial effects of
calcitriol or vitamin D analogs on patient-level outcomes,
such as cardiac events or mortality, and the optimal level of
PTH in CKD G3a to G5 is not known. Furthermore, therapy
with these agents may have additional harmful effects related
to increases in serum phosphate and FGF23 levels. If initiated
for severe and progressive SHPT, calcitr iol or vitamin D an-
alogs should be started with low doses, independent of the
initial PTH concentration, and then titrated based on the
PTH response. Hypercalcemia should be avoided.
Research recommendation
Multicenter RCTs should be conducted in children and
adults to determine the benets or harms of calcitriol or
vitamin D analogs in patients with CKD G3a to G5;
patient-level outcomes including falls, fractures, sarcopenia,
muscle strength, physical function, progression to end-stage
kidney disease, cardiovascular events, hospitalizations, and
mortality should be assessed. Additional important patient-
level outcomes to include are bone pain, pruritus, and
health-related quality of life. Studies should also include
patients with more severe SHPT and should determine the
impact of reducing PTH to different target levels, such as
the normal range versus higher levels.
4.2.4: In patients with CKD G5D requiring PTH-lowering
therapy, we suggest calcimimetics, calcitriol, or
vitamin D analogs, or a combination of calcimimetics
with calcitriol or vitamin D analogs (2B).
Rationale
New data published since the 2013 KDIGO Madrid Contro-
versies Conference prompted the Work Group to reappraise
the use of PTH-lowering therapies in patients with CKD G5D.
As shown in Supplementary Table S43, the ERT identied 2
new trials evaluating treatment with cinacalcet versus placebo
and 1 new trial evaluating calcitriol versus a vitamin D analog.
One open-label clinical trial was conducted evaluating the
effect of cinacalcet on bone histomorphometry.
145
There are
still no new trials of calcitriol or vitamin D analogs that
demonstrated clear benets in patient-level outcomes.
The Work Group discussed the EVOLVE trial at length.
EVOLVE evaluated the effect of cinacalcet versus placebo on
patient-level outcomes in 3883 HD patients using a composite
endpoint of all-cause mortality, nonfatal myocardial infarc-
tion, hospitalization for unstable angina, congestive heart
failure, and per ipheral vascular events. Secondary endpoints
included individual components of the primary endpoint,
clinical fracture, stroke, parathyroidectomy, and cardiovas-
cular events and cardiovascular death.
38
The results of EVOLVE have proven controversial. The
unadjusted primary composite endpoint showed a nonsig-
nicant reduction (HR: 0.93; P ¼ 0.112) with cinacalcet use.
However, analyses adjusted for imbalances in baseline char-
acteristics demonstrated a nominally signicant reduction in
the primary composite endpoint (HR: 0.88; P ¼ 0.008), as did
www.kisupplements.org chapter 4.2
Kidney International Supplements (2017) 7, 1–59 35
sensitivity analyses accounting for patient nonadherence to
randomized study medication (HR: 0.77; 95% CI: 0.70
0.92)
137
or when patients were censored at the time of kidney
transplant, parathy roidectomy, or the use of commercial
cinacalcet (HR: 0.84; P # 0.001).
38
Further challenging the
interpretation of the nonsignicant reduction in risk seen
with the unadjusted primary endpoint was a signicant
treatment-age interaction (P ¼ 0.03),
38
leading to speculation
that cinacalcet may be effective predominantly in older dial-
ysis patients. Approximately one-third of the EVOLVE par-
ticipants were under the age of 55, and prespecied analyses
that evaluated subjects above or below age 65 demonstrated a
signicant reduction in risk associated with use of cinacalcet
for both the primary endpoint (HR: 0.74; P # 0.001) and all-
cause mortality (HR: 0.73; P # 0.001) for those aged
above 65.
138
The Work Group also considered additional prespecied
and post hoc analyses from EVOLVE. These included a
demonstrated signicant reduction in the r isk of severe un-
remitting SHPT (dened by the persistence of markedly
elevated PTH concentrations [2 consecutive PTH values over
1000 pg/ml (106 pmol/l)] together with hy percalcemia
[serum calcium > 10.5 mg/dl (2.63 mmol/l)] or para-
thyroidectomy). Cinacalcet appeared to consistently reduce
the risk of this endpoint regardless of baseline PTH (HR: 0.31,
P # 0.001 for those with baseline PTH 300600 pg/ml [3264
pmol/l]; HR: 0.49, P # 0.001 for those with baseline PTH
600900 pg/ml [64 95 pmol/l]; HR: 0.41, P < 0.001 for those
with PTH > 900 pg/ml [95 pmol/l]).
139
Cinacalcet had no
effect on the risk of clinical fractures in unadjusted analyses
(HR: 0.93; P ¼ 0.111) and showed a nominally signicant
reduction in risk of fracture when adjusted for age (HR: 0.88;
P ¼ 0.007).
140
Thus, EVOLVE did not meet its primary endpoint that
cinacalcet reduces the risk of death or clinically imp ortant
vascular events in CKD G5D patients. However, the results of
secondary analyses suggest that cinacalcet may be benecial in
this population or a subset. There was a lack of uniform
consensus among the Work Group members in their inter-
pretation of these data with regard to establishing cinacalcet as
the recommended rst-line therapy for patients with CKD
G5D requiring PTH-lowering therapy. While some felt that
only the primary analysis should be used to interpret the
outcome, others w ere equally conv inced that the secondary
analyses strongly suggested a benet of treatment with cina-
calcet on important patient-level outcomes.
Despite these differences in interpretation, there was
agreement among Work Group membe rs that the higher cost
of cinacalcet was also a relevant consideration given its un-
certain clinical bene ts. There was al so agreement that the
documented associat ion between good clinical outcomes and
the extent of FGF23 reduction with cinacalcet warrants
further study.
141
No trials demonstrated the benets of combination ther-
apy (cinacalcet plus another agent) on clinically relevant
outcomes. However, several additional RCTs were identied
that studied the effect of combination therapy on putative
surrogate outcomes (summarized in Supplementary
Tables S43S48). Two trials evaluated the use of cinacalcet
with low-dose active vitamin D versus standard therapy.
Urena-Torres et al.
142
demonstrated improved PTH-lowerin g
efcacy in subjects treated with cinacalcet or low-dose active
vitamin D, while Raggi et al.
143
found that cinacalcet with
low-dose vitamin D attenuated the progression of coronary
artery calcium accumulation when assessed using calcium
volume scores (P ¼ 0.009) although not when using the more
common Agatston score (P ¼ 0.07). Two open-label trials of
cinacalcet were considered important in reaching consensus
for Recommendation 4.2.4. The PARADIGM trial compared a
cinacalcet-based treatment strate gy with an active vitamin D
based strategy in 312 HD patients and demonstrated similar
reductions in PTH in both treatment arms.
144
The BONA-
FIDE trial evaluated bone histomorphometry in 77 paired
bone biopsy samples in cinacalcet-treated subjects with
proven high-turnover bone disease and demonstrated re-
ductions in bone formation rates and substantial increase in
the number of subjects with normal bone histology (from 0 at
baseline to 20 after 612 months of therapy).
145
Two subjects
developed adynamic bone disease, both of whom had PTH
values < 150 pg/ml (16 pmol/l), and 1 patient developed
osteomalacia coincident with hypophosphatemia. Despite
being a prospective interventional trial, the BONAFIDE trial
did not fulll our literature inclusion criteria, because there
was no control group and only longitudinal assessments were
available, and thus is not listed in the Supplementary Tables.
It was recognized by the Work Group that newer, i.v.
calcimimetic agents have undergone clinical trial investigation
and were published after our guideline systematic review.
However, while data on safety and efcacy were generated, no
patient-level outcomes were reported. Therefore, these trials
did not impact the current recommendation.
146,147
In summary, the Work Group was divided as to whether
the EVOLVE data are sufcient to recommend cinacalcet as
rst-line therapy for all patients with SHPT and CKD G5D
requiring PTH lowering. One viewpoint is that the primary
endpoint of the EVOLVE study was negative. The alternative
viewpoint is that secondary analyses found effects on patient-
level endpoints, while there are no positive data on mortality
or patient-centered endpoints from trials with calcitriol or
other vitamin D analogs. Given the lack of uniform consensus
among the Work Group and the higher acquisition cost of
cinacalcet, it was decided to modify the 2009 recommenda-
tion to list all acceptable treatment options in alphabetical
order. The individual choice should continue to be guided by
considerations about concomitant therapies and the present
calcium and phosphate levels. In addition, the choice of
dialysate calcium concentrations will impact on serum PTH
levels. Finally, it should be pointed out that para-
thyroidectomy remains a valid treatment option especially in
cases when PTH-lowering therapies fail, as advocated in
Recommendation 4.2.5 from the 2009 KDIGO CKD-MBD
guideline.
chapter 4.2 www.kisupplements.org
36
Kidney International Supplements (2017) 7, 1–59
To date, studies of cinacalcet in children are limited to case
reports,
148
case series,
149,150
a single-center experience (with
28 patients with CKD G4G5),
151
and an open-label study of
a single dose in 12 children on dialysis.
152
In recognition of
the unique calcium demands of the growing skeleton, PTH-
lowering therapies should be used with caution in children
to avoid hypocalcemia. Future studies are needed in children
before pediatric-specic recommendations can be issued.
Research recommendations
The Work Group expli citly endorses the presence of clinical
equipoise and the need to conduct placebo-controlled trials
with calcimimetics versus standard therapy for the
treatment of SHPT in patients with CKD G5D with
emphasis on those at greatest risk (e.g., older, with
cardiovascular disease).
Prospective RCTs aiming at patient-centered surrogate
outcomes (primary endpoints: mortality, cardiovascular
events; secondary endpoints: FGF23, LVH progression,
calcication) should be performed with the new parenteral
calcimimetic compound (e.g., etelcalcitide).
Given the disparate effects of calcimimetic and active
vitamin D therapies on FGF23 and data suggesting a clinical
benet from FGF23 reduction, RCTs evaluating the specic
reduction of FGF23 as a therapeutic endpoint should be
undertaken.
www.kisupplements.org chapter 4.2
Kidney International Supplements (2017) 7, 1–59 37
Chapter 4.3: Treatment of bone with
bisphosphonates, other osteoporosis medications,
and growth hormone
4.3.3: In patients with CKD G3aG5D with biochemical
abnormalities of CKD-MBD and low BMD and/or
fragility fractures, we suggest that treatment choices
take into account the magnitude and reversibility of
the biochemical abnormalities and the progression
of CKD, with consideration of a bone biopsy (2D).
Rationale
Recommendation 3.2.2 now addresses the indications for a
bone biopsy prior to antiresorptive and other osteoporosis
therapies. Therefore, the original Recommendation 4.3.4
from the 2009 KDIGO CKD-MBD Guideline has been
removed, and Recommendation 4.3.3 has broadened from
CKD G3a to G3b to CKD G3a to G5D. Nevertheless, when
such treatment choices are considered, their specic side ef-
fects must also be taken into account (e.g., antiresorptives will
exacerbate low bone turnover, denosumab may induce sig-
nicant hypocalcemia), and the risk of their administration
must be weighed against the accuracy of the diagnosis of the
underlying bone phenotype.
chapter 4.3 www.kisupplements.org
38
Kidney International Supplements (2017) 7, 1–59
Chapter 5: Evaluation and treatment of kidney
transplant bone disease
5.5: In patients with CKD G1TG5T with risk factors for
osteoporosis, we suggest that BMD testing be used to
assess fracture risk if results will alter therapy (2C).
Rationale
Fracture risk is 4-fold higher in patients with end-stage kid-
ney disease
153
compared with the general population and
increases further in the early post-transplant period .
154
A
2002 study examined the risk of hip fracture in kidney
transplant recipients and estimated the fracture rate at 3.3
events per 1000 person-years, a 34% higher risk compared
with patients receiving dialysis who were waitlisted for
transplantation.
3
Bone disease in transplant recipients is
complex and heterogeneous.
155
Essentially, transplant bone
disease is the composite of preexistin g damage to the bone
acquired during the period of renal insufciency and damage
to the bone starting in the period of transplantation. In
contrast to older studies,
156
recent cohort studies showed
minimal BMD losses in the early post-transplant period,
which seem to be restricted to sites rich in cortical bone such
as the distal radius.
157
A low cumulative steroid exposure
along with persistent hyperparathyroidism most likely ac-
counts for this shift.
157
The widespread implementation of
steroid minimization protocols may explain the favorable
trend in fracture risk in kidney transplant recipients observed
over the last 2 decades.
158160
The 2009 KDIGO CKD-MBD Guideline
30
recommended
BMD testing in the rst 3 months following transplantation
in patient s with an eGFR greater than 30 ml/min/1.73 m
2
if
they receive corticosteroids or have risk factors for osteopo-
rosis (Recommendation 5.5), but recommended that DXA
BMD not be performed in those with CKD G4T to G5T
(Recommendation 5.7). As detailed in the new aforemen-
tioned Recommendation 3.2.1, there is growing evidence that
DXA BMD predicts fractures across the spectrum of CKD
severity, including 4 prospective cohort studies in patients
with CKD G3a to G5D
1013
(Supplementary Tables S7S12).
To date, there are no prospective studies addressing the ability
of DXA to predict fractures in transplant recipients. However,
a retrospective cohort study conducted in 238 kidney trans-
plant recipients with CKD G1T to G5T examined the asso-
ciations of DXA BMD with fracture events.
161
Lumbar spine
and total-hip BMD results were expressed as T-scores and
categorized as normal (T-score $ 1), osteopenic (T-
score < 1 and > 2.5), or osteoporotic ( T-score # 2.5).
A total of 46 incident fractures were recorded in 53 patients.
In a multivariate Cox analysis of DXA BMD results in the
total hip, osteopenia (HR: 2.7, 95% CI: 1.64.6) and osteo-
porosis (HR: 3.5, 95% CI: 1.86.4) were associated with
signicantly increased risk of fracture compared with normal
BMD, independent of age, sex, and diabetes. Multivariate
models were not provided for the lumbar spine BMD T-score
results; however, unadjusted analyses suggested that spine
BMD provided less fracture prediction compared with total-
hip BMD. Although this DXA study in kidney transplant
recipients was not eligible for the evidence-based review due
to its retrospective design, the Work Group concluded that
the ndings were consistent with the other studi es in CKD
G3a to G5D described above.
In summary, there is growing evidence that DXA BMD
predicts fractures in patients with CKD across the spectrum,
with limited data suggesting these ndings extend to trans-
plant recipients. The revised guideline statement recommends
BMD testing in transplant recipients, as in those with CKD
G3a to G5D, if the results will impact treatment decisions.
Research recommendations
The research recommendations outlined for Recommen-
dation 3.2.1 should be expand ed to include studies in
kidney transplant recipients.
Prospective studi es in patients with CKD G1T to G5T
should be performed to determine the value of BMD and
bone biomarkers as predictors of fractures.
5.6: In patients in the rst 12 months after kidney trans-
plant with an estimated glomerular ltration rate
greater than approximately 30 ml/min/1.73 m
2
and low
BMD, we suggest that treatment with vitamin D, cal-
citriol/alfacalcidol, and/or antiresorptive agents be
considered (2D).
We suggest that treatment choices be inuenced by
the presence of CKD-MBD, as indicated by abnormal
levels of calcium, phosphate, PTH, alkaline phos-
phatases, and 25(OH)D (2C).
It is reasonable to consider a bone biopsy to guide
treatment (Not Graded).
There are insufcient data to guide treatment after the
rst 12 months.
Rationale
The rationale for revised guideline Recommendation 3.2.2
now addresses the indications for a bone biopsy prior to
antiresorptive and other osteoporosis therapies. Therefore,
www.kisupplements.org chapter 5
Kidney International Supplements (2017) 7, 1–59 39
the second bullet statement above concerning bone biopsies
has been modied.
Cinacalcet is not approved for the treatment of hyper-
parathyroidism in kidney transplant recipients; however, it is
clinically used, especially in patients with signicant hyper-
calcemia. While efciently correcting hypercalcemia, cina-
calcet so far has failed to show a benecial impact on bone
mineralization in the transplant population.
162
Denosumab
was recently shown to effectively increase BMD in de novo
kidney transplant recipients.
163
However, baseline BMD on
average was not very low in this trial, and there was no
progressive BMD loss in the control group. Fur thermore, an
increased rate of urinary tract infections was observed. In
terms of safety and efcacy, an RCT comparing denosumab
and bisphosphonates in eligible patients at risk may be a
reasonable future study approach. As pointed out in the
rationale for Recommendation 5.5, however, the urgency for
correcting bone mineralization in transplant recipients may
diminish due to the wide application of steroid minimization
schemes.
159
chapter 5 www.kisupplements.org
40
Kidney International Supplements (2017) 7, 1–59
Methodological approach to the 2017 KDIGO
CKD-MBD guideline update
Purpose
In 2009, KDIGO developed a clinical practice guideline on
the diagnosis, evaluation, prevention, and treatment of
CKD-MBD.
30
Because of the limited evidence, many of the
recommendations were deliberately vague.
In October of 2013, KDIGO held a Controversies
Conference to determine whether there was sufcient new
evidence to support updating any of the recommendations.
Based on the discussions at the conference, the participants
opted for a selective update of the guideline.
1
The purpose of this chapter is to describe the methods
used to conduct the evidence review and to develop and
update the guideline recommendations.
Overview of the Process
The process of updatin g the guideline consisted of the
following steps:
Convening of a Controversies Conference to determine
whether sufcient new data exist to support a reassessment
of the guideline
Appointing a Work Group and an ERT
Rening the research questions
Developing the search strategy, inclusion/exclusion criteria,
and data extraction tables
Drafting the evidence matrices and evidence proles
Revising the recommendations
Grading the quality of the evidence
Grading the strength of the recommendation
Controversies Conference
In October 2013, KDIGO held a Controversies Conference
entitled, CKD-MBD: Back to the Future, in Madrid, Spain.
1
The purpose of the conference was to determine whether there
was sufcient new evidence to support updating any of the rec-
ommendations from the 2009 KDIGO guideline on the diag-
nosis, evaluation, prevention, and treatment of CKD-MBD.
Seventy-four experts in adult, pediatric, and transplant
nephrology, endocrinology, cardiology, bone histomorphometry
pathology, and epidemiology attended the conference.
Four topic areas were considered: (i) vascular calcication;
(ii) bone quality; (iii) calcium and phosphate; and (iv)
vitamin D and PTH. Each participant was assigned to 1 of the
4 topics based on their area of expertise. Participants identi-
ed new studies in their topic area and answered a set of
questions to determine which recommendations required
reevaluation.
1
The result was a list of recommendations to be
addressed in a s elected update (i.e ., t o use speci cmethods
to update only those parts of the guideline in need of
update).Therewasapublicreviewofthescopeofworkfor
the guidelin e.
Appointment of guideline Work Group and evidence
review team
The KDIGO co-chairs appointed 2 chairs for the Guideline
Work Group, who then assembled the Work Group to be
responsible for the development of the guideline. The Work
Group comprised domain experts, including individuals with
expertise in adult and pediatric nephrology, bone disease,
cardiology, and nutrition. The Johns Hopkins University in
Baltimore, MD, was contracted as the ERT to provide
expertise in guideline development methodology and sys-
tematic evidence review. KDIGO support staff provided
administrative assistance and facilitated communication.
The ERT consisted of methodologists with expertise in
nephrology and internal medicine, and research associates
and assistants. The ERT and the Work Group worked closely
throughout the project. In January 2015, the ERT and the
Work Group Co-Chairs held a 2-day meeting in Baltimore,
MD, to discuss the guideline development and systematic
review processes and to rene the key questions.
The ERT performed systematic reviews for each of the
questions conducting literature searches, abstract and full-text
screening, data extracti on, risk of bias assessment, and syn-
thesis. The ERT provided suggestions and edits on the
wording of recommendations, and on the use of specic
grades for the strength of the recommendations and the
quality of evidence. The Work Group took on the primary
role of writing the recommendations and rationale, and
retained nal responsibility for the content of the recom-
mendations and the accompanying narrative.
Renement of the research questions
The rst task was to dene the overall topics and goals for the
guideline. Using the recomm endations identied during the
Controversies Conference, the ERT drafted research ques-
tions and identied the population , interventions, compari-
son, and outcomes (PICO elements) for each research
question.
The ERT recruited a technical exper t panel to review the
research questions. The technical expert panel included in-
ternal and external clinicians and researchers in nephrology
and CKD. During a conference call, the technical expert panel
provided feedback on the research questions.
The Work Group Co-Chairs and the ERT rened the
research questions at the 2-day meeting in Baltimore, MD.
www.kisupplements.org methodological approach
Kidney International Supplements (2017) 7, 1–59 41
Table 2 | Research questions
Section
2009
rec. no. Research question Key outcomes Additional outcomes
Bone quality 3.2.1 In patients with CKD G3aG5D, what is the
effect on bone quality of bisphosphonates,
teriparatide, denosumab, and raloxifene?
TMV (as measured by bone
biopsy)
BMD/bone mineral content
Fracture
4.3.4 In patients with CKD G4G5D, what is the
effect on bone quality of bisphosphonates,
teriparatide, denosumab, and raloxifene?
TMV (as measured by bone
biopsy)
BMD/bone mineral content
Fracture
3.2.2 (a) In patients with CKD G3aG5D, how well do
BMD results predict fractures?
(b) In patients with CKD G3a G5D, how well do
BMD results predict renal osteodystrophy?
(a) Fracture
(b) TMV
5.5 In patients with CKD G1G3b and transplant
recipients, how well do BMD results predict
fractures?
Fracture
5.7 In patients with CKD G4G5 and transplant
recipients, how well do BMD results predict
fractures?
Fracture
Calcium and
phosphate
4.1.1 In patients with G3aG5 or G5D, what is the
evidence for benet or harm in maintaining
serum phosphate in the normal range
compared with other targets of serum
phosphate in terms of biochemical outcomes,
other surrogate outcomes, and patient-
centered outcomes?
Mortality
GFR decline
Cardiovascular and cerebrovas-
cular events
Phosphate
Bone histology, BMD
Vascular and valvular calcication
imaging
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.1.3 In patients with CKD G5D, what is the evidence
for benet or harm in using a dialysate calcium
concentration between 1.25 and 1.50 mmol/l
(2.5 and 3.0 mEq/l) compared with other
concentrations of dialysate calcium in terms of
biochemical outcomes, other surrogate
outcomes, and patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
Calcium
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.1.2 In patients with CKD G3aG5D, what is the
evidence for benet or harm in maintaining
serum calcium in the normal range compared
with other targets of serum calcium in terms of
biochemical outcomes, other surrogate
outcomes, and patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
Calcium
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
methodological approach www.kisupplements.org
42
Kidney International Supplements (2017) 7, 1–59
Section
2009
rec. no. Research question Key outcomes Additional outcomes
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.1.4 In patients with CKD G3aG5 or G5D with
hyperphosphatemia, what is the evidence for
benet or harm in using calcium-containing
phosphate-binding agents to treat
hyperphosphatemia compared with calcium-
free phosphate-binding agents in terms of
biochemical outcomes, other surrogate
outcomes, and patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
Phosphate
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.1.7 In patients with CKD G3aG5D with
hyperphosphatemia, what is the evidence for
benet or harm in limiting dietary phosphate
intake compared with a standard diet in terms
of biochemical outcomes, other surrogate
outcomes, and patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
Vascular and valvular calcication
Phosphate
Bone histology, BMD
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
Vitamin D
and PTH
4.2.1 In patients with CKD G3aG5 not on dialysis
with levels of intact PTH above the upper
normal limit of the assay, what is the evidence
for benet or harm in reducing dietary
phosphate intake or treating with phosphate-
binding agents, calcium supplements, or
native vitamin D in terms of biochemical
outcomes, other surrogate outcomes, and
patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
GFR decline
Calcium
Phosphate
Parathyroid hormone
25-hydroxyvitamin D [25(OH)D]
1,25-dihydroxyvitamin D
[1,25(OH)
2
D]
Alkaline phosphatases
Bone-specic alkaline
phosphatase
Bicarbonate
FGF23
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.2.2 In patients with CKD G3aG5 not on dialysis, in
whom serum PTH is progressively rising and
remains persistently above the upper limit of
normal for the assay despite correction of
modiable factors, what is the evidence for
LVH
Hypercalcemia
Mortality
Cardiovascular and cerebrovas-
cular events
Calcium
Phosphate
Parathyroid hormone
25-hydroxyvitamin D [25(OH)D]
(Continued on next page)
Table 2 | (Continued)
www.kisupplements.org methodological approach
Kidney International Supplements (2017) 7, 1–59 43
During this meeting decisions were also made abou t out-
comes, including those considered most important for deci-
sion making that would be graded (key outcomes). The
nalized research questions and outcomes are presented in
Table 2.
Search strategy
The ERT searched MEDLIN E and the Cochrane Central
Register of Controlled Trials (CENTRAL) for the date range
of December 2006 through September 2015. The December
2006 date provided the recommended 1-year overlap with the
end of the previous search.
164
The search yield was also
supplemented by articles provided by the Work Group
members through February 2017.
The search strategy included MeSH and text terms for
CKD and the interventions and markers of interest
(Supplementary Appendix A) and was limited to the Eng lish
language. The ERT also rev iewed the list of references that
were suggested during the Controversies Conference.
All studi es that had been included in the prior guideline
were rereviewed to ensure that they met the eligibility criteria.
Inclusion and exclusion criteria. With in put from the Work
Group, the ERT dened the eli gibility criteria a priori. The
eligibility criteria for all studies were: (i) original data pub-
lished in English, (ii) followed up at least 10 patie nts with
CKD for at least 6 months, and (iii) addressed 1 of the
research questions. The minimum mean duration of follow-
up of 6 months was chosen on the basis of clinical
Section
2009
rec. no. Research question Key outcomes Additional outcomes
benet or harm in treating with calcitriol or
vitamin D analogs compared with placebo or
active control in terms of biochemical
outcomes, other surrogate outcomes, and
patient-centered outcomes?
1,25-dihydroxyvitamin D
[1,25(OH)
2
D]
Alkaline phosphatases
Bone-specic alkaline
phosphatase
Bicarbonate
FGF23
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
4.2.4 In patients with CKD G5D, what is the evidence
for benet or harm in treating with calcitriol,
vitamin D analogs, calcimimetics or
combination thereof compared with placebo
or active control in terms of biochemical
outcomes, other surrogate outcomes, and
patient-centered outcomes?
Mortality
Cardiovascular and cerebrovas-
cular events
Fracture
Vascular and valvular calcication
imaging
Calcium
Phosphate
Parathyroid hormone
25-hydroxyvitamin D [25(OH)D]
1,25-dihydroxyvitamin D
[1,25(OH)
2
D]
Alkaline phosphatases
Bone-specic alkaline
phosphatase
Bicarbonate
FGF23
Bone histology, BMD
Vascular and valvular calcication
imaging
Measures of GFR
Hospitalizations
Quality of life
Kidney or kidney graft failure
Fracture
Parathyroidectomy
Clinical adverse events
Growth, skeletal deformities,
bone accrual
Calciphylaxis/CUA
BMD, bone mineral density; CKD, chronic kidney disease; CUA, calcic uremic arteriolopathy; GFR, glomerular ltration rate; FGF23, broblast growth factor 23; LVH, left
ventricular hypertrophy; PTH, parathyroid hormone; rec. no., recommendation number; TMV, bone turnover mineralization volume.
Table 2 | (Continued) Research questions
methodological approach www.kisupplements.org
44
Kidney International Supplements (2017) 7, 1–59
reasoning, accounting for the hypothetical mechanisms of
action. For treatments of interest, the proposed effects on
patient-centered outcomes require long-term exposure and
typically would not be evident before several months of
follow-up. The question-specic eligibility criteria are pro-
vided in Table 3, and the overall search yield for the guideline
systematic review is summarized in Supplementary Appendix
B.
Two reviewers independently screened titles and abstracts
and full-text articles for inclusion. Differences regarding in-
clusion were resolved through consensus adjudication.
Any study not meeting the inclusion criteria could be cited
in the narrative but was not considered part of the body of
evidence for a particular recommendation.
Data extraction
The ER Tmodied the online supplementary tables from the prior
guideline. One reviewer abstracted data directly into the modied
tables, and a second reviewer conrmed the data abstraction.
The ER T abstracted data on general study characteristics,
participant characteristics, interventions and co-interventions,
and outcome measures, including measures of variability .
Two reviewers independently assessed individual study
quality using the Cochrane Collaborations tool
165
for
assessing risk of bias for RCTs and using the Quality in
Prognosis Studies tool
166
for observational studies.
The Work Group critically reviewed draft tables, and tables
were revised as appropriate.
Evidence matrices and evidence proles
The ERT created evidence matrices for each of the key out-
comes for each research question. For each key outcome, the
matrix lists the individual studies, their sample size, follow-up
duration, and the individual study quality. The ERT also
drafted evidence proles to display the total number and
overall quality of the studies addressing each key outcome for
each research question.
Revising recommendations
In June 2015, the Work Group and the ERT convened a 3-day
meeting in Baltimore, MD, to review the summary tables,
evidence proles, and evidence matrices; to decide whether and
how the recommendations should be rev ised; and to determine
a grade that described the quality of the overall evidence and a
grade for the strength of each recommendation.
Grading
A structured approachmodeled after Grading of Recom-
mendations Assessment, Development, and Evaluation
(GRADE)
167172
and facilitated by the use of evidence proles
and evidence matriceswas used to determine a grade that
described the quali ty of the overall evidence and a grade for
the strength of a recommendation. For each topic, the
Table 3 | Question-specic eligibility criteria
2009 recommendation no. Exposure or intervention Eligibility criteria
3.2.1, 4.3.4 Bisphosphonates, teriparatide,
denosumab, or raloxifene
RCT with at least 10 participants per arm for the outcome of bone quality and
at least 25 participants per arm for all other outcomes
Evaluates bone quality, bone mineral density, or fracture
3.2.2, 5.5, 5.7 Predictive value of BMD results
RCT with at least 25 participants per arm or a prospective cohort study with
50 participants
Evaluates fractures or renal osteodystrophy
4.1.1, 4.1.2 Serum phosphate or serum calcium
levels
RCT with at least 25 participants per arm or a prospective cohort study with
50 participants
Evaluates mortality, GFR decline, cardiovascular or cerebrovascular events,
phosphate levels, calcium levels, bone histology, bone mineral density, bone
volume, vascular and valvular calcication imaging, hospitalizations, quality
of life, kidney or kidney graft failure, fractures, parathyroidectomy, clinical
adverse events, growth, skeletal deformities, bone accrual, or calciphylaxis or
calcic uremic arteriolopathy
4.1.3 Dialysate calcium concentrations
RCT with at least 25 participants per arm
Evaluates mortality, cardiovascular or cerebrovascular events, calcium levels,
bone histology, bone mineral density, bone volume, vascular and valvular
calcication imaging, measures of GFR, hospitalizations, quality of life, kidney
or kidney graft failure, fractures, parathyroidectomy, clinical adverse events,
growth, skeletal deformities, bone accrual, or calciphylaxis or calcic uremic
arteriolopathy
4.1.4, 4.1.7, 4.2.1, 4.2.2, 4.2.4 Dietary phosphate intake,
phosphate-binding agents, calcium
supplements, native vitamin D,
vitamin D analogs, calcitriol, or
calcimimetics
RCT with at least 25 participants per arm
Evaluates mortality, cardiovascular or cerebrovascular events, GFR decline,
calcium levels, phosphate levels, parathyroid hormone levels, 25-
hydroxyvitamin D or 1,25-dihydroxyvitamin D levels, alkaline phosphatases,
bone-specic alkaline phosphatase, bicarbonate, FGF23, bone histology, bone
mineral density, bone volume, vascular and valvular calcication imaging,
measures of GFR, hospitalizations, quality of life, kidney or kidney graft failure,
fractures, parathyroidectomy, clinical adverse events, growth, skeletal de-
formities, bone accrual, calciphylaxis or calcic uremic arteriolopathy for any of
the above interventions, or LVH or hypercalcemia for vitamin D analogs
BMD, bone mineral density; FGF23, broblast growth factor 23; GFR, glomerular ltration rate; LVH, left ventricular hypertrophy; RCT, randomized controlled trial.
www.kisupplements.org methodological approach
Kidney International Supplements (2017) 7, 1–59 45
discussion on g rading of the quality of evidence was led by the
ERT, and the discussion regarding the strength of the rec-
ommendations was led by the Work Group Chairs.
Grading the quality of evidence for each outcome
The quality of a body of evidence refers to the extent to
which our condence in an estimate of effect is sufcient to
support a particular recommendation. Following GRADE, the
quality of a body of evidence pertaining to a particular
outcome of interest is initially categorized on the basis of
study design. For questions of interventions, the initial quality
grade is high if the body of evidence consists of RCTs, low
if it consists of observational studies, or very low if it
consists of studies of other study designs. For questions of
interventions, the Work Group graded only RCTs. The g rade
for the quality of evidence for each interventionoutcome
pair was then decreased if there were serious limitations to the
methodological quality of the aggregate of studies; if there
were important inconsistencies in the results across studies; if
there was uncertainty about the directness of evidence
including a limite d applicability of ndings to the population
of interest; if the data were imprecise or sparse; or if there was
thought to be a high likelihood of bias. The nal grade for the
quality of evidence for an inter ventionoutcome pair could be
1 of the following 4 grades: high,”“moderate,”“low, or
very low (Table 4).
Grading the overall quality of evidence
The quality of the overall body of evidence was then deter-
mined on the basis of the quality grades for all outcomes of
interest, taking into account explicit judgments about the
relative importance of each outcome. The resulting 4 nal
categories for the quality of overall ev idence were A, B, C, and D
(Table 5). This grade for overall evidence is indicated behind
the strength of recommendations. The summary of the overall
quality of evidence across all outcomes proved to be very
complex. Thus, as an interim step, the evidence proles
recorded the quality of evidence for each of 3 outcome cate-
gories: patient-centered outcomes, other bone and vascular
surrogate outcomes, and laboratory outcomes. The overall
quality of evidence was determined by the Work Group and is
based on an overall assessment of the evidence. It reects that,
for most interventions and tests, there is no high-quality evi-
dence for net benet in terms of patient-centered outcomes.
Assessment of the net health benet across all important
clinical outcomes
Net health benet was determined on the basis of the antic-
ipated balance of benets and harm across all clinically
important outcomes. The assessment of net health benetby
the Work Group and ERT is summarized in one of the
following statements: (i) There is net benet from interven-
tion when be nets outweigh harm; (ii) there is no net benet;
(iii) there are trade-offs between benets and harm when
harm does not altogether offset benets but requires
consideration in decision making; or (iv) uncertaint y remains
regarding net benet(Table 6).
Grading the recommendations
The strength of a recommendation indica tes the extent to
which one can be condent that adherence to the recom-
mendation will do more good than harm. The strength of a
recommendation is graded as Level 1 or Level 2.
173
Table 7
shows the nomenclature for grading the strength of a
recommendation and the implications of each level for pa-
tients, clinicians, and policy makers. Recommendations can
be fo r or against doing something. Table 8 shows that the
strength of a recommendation is determined not just by the
Table 4 | GRADE system for grading quality of evidence for an outcome
Step 1: starting grade for quality of
evidence based on study design Step 2: reduce grade Step 3: raise grade
Final grade for quality of
evidence for an outcome
a
High for randomized controlled trials
Moderate for quasi-randomized trial
Low for observational study
Very low for any other evidence
Study quality
1 level if serious limitations
2 levels in very serious limitations
Consistency
1 level if important inconsistency
Directness
1 level if some uncertainty
2 levels if major uncertainty
Other
1 level if sparse or imprecise data
1 level if high probability of
reporting bias
Strength of association
þ1 level is strong,
b
no plausible
confounders, consistent and direct
evidence
þ2 levels if very strong,
c
no major threats
to validity and direct evidence
Other
þ1 level if evidence of a dose-response
gradient
þ1 level if all residual confounders would
have reduced the observed effect
High
Moderate
Low
Very low
GRADE, grading of recommendations assessment, development, and evaluation; RR, relative risk.
a
The highest possible grade is high and the lowest possible grade is very low.
b
Strong evidence of association is dened as signicant RR of > 2(< 0.5) based on consistent evidence from two or more observational studies, with no plausible
confounders.
c
Very strong evidence of association is dened as signicant RR of > 5(< 0.2) based on direct evidence with no major threats to validity.
Modied with permission from Uhlig K, Macleod A, Craig J, et al. Grading evidence and recommendations for clinical practice guidelines in nephrology. A position statement
from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006;70:20582065.
171
methodological approach www.kisupplements.org
46
Kidney International Supplements (2017) 7, 1–59
quality of evidence, but also by other, often complex judg-
ments regarding the size of the net medical benet, values and
preferences, and costs.
Ungraded statements
The Work Group felt that havi ng a category that allows it to
issue general advice would be useful. For this purpose, the
Work Group chose the categor y of a recommendation that
was not graded. Typically, this type of ungraded statement
met the following criter ia: it provides guidance on the basis of
common sense; it provides reminders of the obvious; and it is
not sufciently specic to allow an application of evidence to
the issue, and therefore it is not based on a systematic review.
Common examples include recommendations regarding the
frequency of testing, referral to specialists, and routine
medical care. The ERT and Work Group strove to minimize
the use of ungraded recommendations.
Limitations of approach
Although the literature searches were intended to be
comprehensive, they were not exhaustive. MEDLINE and
Cochrane CENTRAL were the only databases searched, and
the search was limited to Eng lish language publications. Hand
searches of journals were not performed, and review articles
and textbook chapters were not systematically searched.
However, Work Group me mbers did identify additional or
new studies for consideration.
Nonrandomized studies were not systematically reviewed
for studies of interventions. The ERT and Work Group re-
sources were devoted to review of randomized trials, as these
Table 5 | Final grade for overall quality of evidence
Grade Quality of evidence Meaning
A High We are condent that the true effect lies
close to that of the estimate of the effect.
B Moderate The true effect is likely to be close to the
estimate of the effect, but there is a
possibility that it is substantially different.
C Low The true effect may be substantially
different from the estimate of the effect.
D Very low The estimate of effect is very uncertain,
and often will be far from the truth.
Table 6 | Balance of benets and harms
When there was evidence to determine the balance of medical benets
and harm of an intervention to a patient, conclusions were categorized
as follows:
Net benets The intervention clearly does more good than
harm.
Trade-offs There are important trade-offs between the
benets and harm.
Uncertain trade-offs It is not clear whether the intervention does
more good than harm.
No net benets The intervention clearly does not do more
good than harm.
Table 7 | Implications of the strength of a recommendation
Grade
Implications
Patients Clinicians Policy
Level 1:
We recommend
Most people in your situation would
want the recommended course of
action, and only a small proportion
would not.
Most patients should receive the
recommended course of action.
The recommendation can be evaluated
as a candidate for developing a policy
or a performance measure.
Level 2:
We suggest
The majority of people in your situation
would want the recommended course
of action, but many would not.
Different choices will be appropriate
for different patients. Each patient
needs help to arrive at a management
decision consistent with her or his
values and preferences.
The recommendation is likely to require
substantial debate and involvement of
stakeholders before policy can be
determined.
Table 8 | Determinants of strength of recommendation
Factor Comment
Balance between desirable
and undesirable effects
The larger the difference between the
desirable and undesirable effects, the more
likely a strong recommendation is
warranted. The narrower the gradient, the
more likely a weak recommendation is
warranted.
Quality of the evidence The higher the quality of evidence, the
more likely a strong recommendation is
warranted.
Values and preferences The more variability in values and
preferences, or the more uncertainty in
values and preferences, the more likely a
weak recommendation is warranted.
Costs (resource allocation) The higher the costs of an intervention
that is, the more resources consumedthe
less likely a strong recommendation is
warranted.
www.kisupplements.org methodological approach
Kidney International Supplements (2017) 7, 1–59 47
were deemed most likely to provide data to support treatment
recommendations with higher-quality evidence.
Evidence for patient-relevant clinical outcomes was low.
Usually, low-quality evidence required a substantial use of
expert judgment in deriving a recommendation from the
evidence reviewed.
Formulation and vetting of recommendations
Recommendations were drafted to be clear and actionable,
and the wording also considered the ability of concepts to be
translated accurately into other languages. The nal wording
of recommendations and corresponding grades for the
strength of the recommendations and the quality of evidence
were voted upon by the Work Group and required a majority
to be accepted.
The process of peer review included an external review by
the public to ensure widespread input from numerous
stakeholders, including patients, experts, and industry and
national organizations.
Format for chapters
Each chapter contains one or more specic recommenda-
tions. Within each recommendation, the strength of the
recommendation is indicated as level 1 o r level 2, and the
quali ty of the overall suppor tin g evidence is sh own as A, B,
C, or D. The recommendations are followed by a section
that describes the body of evidence and rationale for the
recommendations. In relevant sections, research recom-
mendations suggest future research to resolve current
uncertainties.
methodological approach www.kisupplements.org
48
Kidney International Supplements (2017) 7, 1–59
Biographic and disclosure information
web 4C=FPOweb 4C=FPO
Markus Ketteler, MD, FERA (Work
Group co-chair), is professor of
medicine and currently serves as Divi-
sion Chief of Nephrology at the Klini-
kum Coburg in Coburg, Germany. In
2016, he was additionally appointed as
chief medical ofcer at this institution.
He is also chairman of the Medical
Board of a large German not-for-prot
dialysis provider (KfH Kuratorium für
Dialyse und Nierentransplantation
e.V.). His research focus is aimed at the understanding of the
pathomechanisms involved in extraosseous calcications, and of
phosphate and vitamin D metabolism in CKD. He has authored
more than 190 peer-reviewed publications including in The
Lancet, Journal of the American Society of Nephrology (JASN),
Journal of Clinical Investigation,andKidney International.Dr.
Ketteler has acted as local, national, or European Principal
Investigator in several clinical multicenter trials in the CKD-
MBD eld (e.g., SVCARB, CALMAG, IMPACT-SHPT, PA-CL-
05A and -05B, and NOPHOS). He serves on the editorial boards
of nephrology journals such as JASN and Nephrology Dialysis
Transplantation (theme editor), acted as a KDIGO Work Group
member on the CKD-MBD Guideline from 2006 to 2009, and
co-leads the German Calciphylaxis Registry. Dr. Ketteler is
currently council member and chairman of the administrative
ofce of the European Renal AssociationEuropean Dialysis and
Transplant Association (ERA-EDTA) (20122018). He acted as
co-chair of the KDIGO Controversies Conference on CKD-
MBD, which took place in Madrid in October 2013. In 2017,
he was elected into the board of directors of the Kidney Health
Initiative, representing the ERA-EDTA for 3 years.
Consultant: Amgen, Fresenius Medical Care, Pzer, Sanit,
Sano, Vifor Fresenius Medical Care Renal Pharma
Speaker: Amgen, BMS, Medice, Pzer, Sano , Vifor Fresenius
Medical Care Renal Pharma
Mary B. Leonard, MD, MSCE (Work
Group co-chair), is the Arline and
Pete Harman Professor and chair of
the Department of Pediatrics at Stan-
ford University School of Medicine.
She is also the Adalyn Jay Physician-in-
Chief of Lucile Packard Childrens
Hospital, and the director of the
Stanford Child Health Research Insti-
tute. Dr. Leonard received her MD
degree from Stanford University and
subsequently completed her pediatrics internship, residency,
and nephrology fellowship at the Child rens Hospital of
Philadelphia. After completing a masters degree in clinical
epidemiolog y at the University of Pennsylvania in 1997, she
joined the faculty in the Departments of Pediatrics and
Biostatistics and Epidemiology. She has maintained contin-
uous National Institutes of Health funding for over 20 years,
and her multidisciplinary research program is primarily
focused on the impact of childhood chronic diseases on
growth, skeletal development, nutrition, and physical function,
with an emphasis on the detrimental effects of CKD. She co-
chaired the ISCD Pediatric Position Development Conferences
in 2007 and 2013. Dr. Leonard has served as an Associate
Editor for Journal of the American Society of Nephrology and
Journal of Bone and Mineral Research. She has published over
150 peer-reviewed manuscripts and is a member of the
American Society of Clinical Investigation, American Pediatric
Society, and the Society for Pediatric Research.
Dr. Leonard declared no competing interests.
web 4C=FPOweb 4C=FPO
Geoffrey A. Block, MD, is a clinical
nephrologist and director of research
at Denver Nephrology. He received his
MD at the University of Cincinnati and
completed his fellowship in nephrology
at the University of Michigan, where he
was trained under the mentorship of
Dr. Friedrich Port at the United States
Renal Data System. Dr. Block started a
clinical research department at Denver Nephrology in 1998, and
his primary research focus has been on clinical outcomes asso-
ciated with CKD-MBD. He has published observational reports
on the risks associated with disorders of mineral metabolism and
has designed, conducted, and published numerous RCTs using a
variety of interventions related to mineral metabolism and
complications of CKD. He has served as a Work Group member
on the 2009 KDIGO CKD-MBD guideline, 2 technical expert
panels for Centers for Medicare and Medicaid Services related to
bone and mineral disorders, and as a member of the steering
committee for the EVOLVE trial. He serves as a reviewer for
Clinical Journal of the American Society of Nephrology, Journal of
the American Society of Nephrology, American Journal of Kidney
Diseases, and Kidney International and was associate editor of
Nephron-Clinical Practice.
Consultant: Ardelyx, Amgen, AstraZeneca, Celgene, Keryx,
Kirin, Ono Pharmaceutical, OPKO
Grant/research support: Keryx*
Speaker: OPKO
www.kisupplements.org biographic and disclosure information
Kidney International Supplements (2017) 7, 1–59 49
Development of educational materials: Amgen, Keryx,
OPKO
Stock/stock options: Ardelyx
Other: medical director, DaVita
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Pieter Evenepoel, MD, PhD, FERA,
is head of the dialysis unit, division of
nephrology, at the University Hospi-
tals Leuven. Dr. Evenepoel completed
his medical training at the Catholic
University of Leuven, Belgium, in
1992, where he also received his PhD
for research on protein assimilation
and fermentation in 1997. In 2000, he
joined the University Hospitals
Leuven, where he gained his certi-
cation as Specialist in Internal Medicine and Nephrology. Dr.
Evenepoel has maintained an active research interest in
mainly clinical aspects of CKD-MBD, as exemplied by
numerous original articles, review papers, and commentaries in
this eld. He is currently a board member of the European
Renal AssociationEuropean Dialysis and Transplant Associa-
tion (ERA-EDTA) working group on CKD-MBD. His research
interests span areas including uremic toxins, nutrition, and
anticoagulation, and he has authored over 200 publications. He
serves presently as associate editor of Nephrology Dialysis
Transplantation and editorial board member for Kidney Inter-
national. He is also an Ordinary Council Member of the ERA-
EDTA.
Consultant: Amgen, Vifor Fresenius Medical Care Renal
Pharma
Grant/research support: Amgen, TECOmedical
Speaker: Amgen, Vifor Fresenius Medical Care Renal Pharma
Miscellaneous travel and meeting expens es unrelated to
above: Amgen, Shire
web 4C=FPOweb 4C=FPO
Masafumi Fukagawa, MD, PhD,
FASN, received his MD in 1983 from
the University of Tokyo School of
Medicine,Tokyo,Japan.Followinga
subspecialty training and PhD pro-
gram in Tokyo, he was a research
fellow at Vanderbilt University School
of Medicine, Nas h ville, TN, until 1995.
From 2000 to 2009 , he was associate
professor and director of the Division
of Nephrology and Kidney Center at
K obe U niv ersity School of Medicine, Kobe, Japan, and he later
moved to Tokai University School of Medicine, Isehara, Japan,
as professor of medicine and the director of the Division of
Nephrology, Endocrinology, and Metabolism. He was interna-
tional associate editor of Clinical Journal of the American Society
of Nephrology (CJASN) (20052010) and currently serves as
associate editor for Jo urnal of Bone and Mineral Metabolism.He
is an editorial board member for Kidney International, American
Journal of Kidney Diseases, CJASN, Nephrology Dialysis Trans-
plantation,andClinical and Experimental Nephrology. In addi-
tion, he served as a Work Group member for the 2009 KDIGO
CKD-MBD guideline and also chaired the committee for the
new version of the Japanese clinical guideline on CKD-MBD by
the Japanese Society for Dialysis Therapy.
Consultant: Kyowa Hakko Kirin, Ono Pharmaceutical, Torii
Grant/research support: Bayer Japan*, Kyowa Hakko Kirin*
Speaker: Bayer Japan, Kyowa Hakko Kirin, Tor ii
Manuscript preparation: Bayer Japan, Kyowa Hakko Kirin
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Charles A. Herzog, MD, FACC,
FAHA, is professor of medicine at
University of Minnesota and a cardi-
ologist at Hennepin County Medical
Center (HCMC) for 32 years. He
founded the program in interventional
cardiology at HCMC and served as
cardiac catheterization laboratory di-
rector from 1985 to 1991, and cardiac
ultrasound laboratory director from 1997 to 2012. He was di-
rector of the United States Renal Data System Cardiovascular
Special Studies Center from 1999 to 2014. Dr. Herzog partici-
pated in the development of the National Kidney Foundations
K/DOQI Clinical Practice Guidelines for Cardiovascular Dis-
ease in Dialysis Patients and KDIGO Clinical Practice Guideline
on Acute Kidney Injury. He also co-chaired the 2010 KDIGO
Controversies Conference, Cardiovascular Disease in CKD:
What is it and What Can We Do About It? and is a co-chair of
the ongoing KDIGO Kidney, Heart, and Vascular Conference
Series. Dr. Herzog was an executive committee member of the
EVOLVE trial, and presently he is chairing the renal committee
of the ISCHEMIA-CKD trial and is co-principal investigator
of the WED-HED (Wearable Cardioverter Debrillator in He-
modialysis Patients) study. He has over 220 published papers
and has served on the editorial boards of the American
Heart Journal, Journal of Nephrology, Clinical Journal of the
American Society of Nephrology, and liaison editor for Nephrology
Dialysis Transplantation. His special interests include cardiac
disease and CKD, and echocardiography.
Consultant: AbbVie, Fibrogen, Relypsa, Sanit, ZS Pharma
Employment: Hennepin County Medical Center, Chronic
Disease Research Group
Grants/research support: Amgen*, Zoll*
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Linda McCann, RD, CSR, has been a
nephrology dietitian for over 43 years,
focused on quality patient care, pro-
fessional management, and electronic
applications for kidney disease and
nutrition. She is currently a
nephrology nutrition consultant and
speaker. Ms. McCann was a member
of the or iginal KDOQI and KDIGO
biographic and disclosure information www.kisupplements.org
50
Kidney International Supplements (2017) 7, 1–59
advisory committees as well as the KDOQI and KDIGO (original
and update) Work Groups that developed and published clinical
practice guidelines and recommendati ons for bone and
mineral abnormalities in CKD. She is the author of the
popular National Kidney Foundation (NKF) publication,
Pocket Guide to Nutrition Assessment of the Patient with Kidney
Disease, currently in its fth edition. She has authored book
chapters and published multiple pee r-reviewed papers in the
area of nephrology nutrition. She has a long history of
mentoring other professionals, driving clinical excellence, and
is a dedicated patient advocate. Ms. McCann has also received
numerous awards including: Recognized Renal Dietitian
(Council on Renal Nutrition), Multiple Outstanding Service
Awards (NKF), Award of Distinguished Service (NKF
Northern California), Champion of Hope (NKF Northe rn
California), Joel D. Kopple Award (NKF National), San Jose
Business Journal 100 Women of Inuence 2012, and the
American Association of Kidney Patients Medal of Excellence.
Consultant: Amgen, Relypsa, Sano
Speaker: Amgen, Relypsa, Sa no
Development of educational presentations: Amgen, Relypsa,
Sano
web 4C=FPOweb 4C=FPO
Sharon M. Moe, MD, is the director
of the Division of Nephrology and
Stuart A. Kleit Professor of Medi-
cine for the Indiana University
School of Medicine. She received
her medical degree from t he Uni-
versity of IllinoisCollege of Medi-
cine at Chicago in 1989 an d
completed her residency in Internal
Medicine at Loyola University
Medical Center in Maywood, IL.
Her research and clinical fellowships were completed in
nephrology at the University of Chicago in Illinois. Dr.
Moe has been a faculty member at In diana University since
1992 and is currently division director for nephrology in
the Departm ent of Medicine at Indiana University School
of Medicine and section chief f or nephrology at the Rou-
debush VA Medical Center. She has also served as the
associate dean for Research Support in the Indiana Uni-
versity School of Medicine and the vice-chair for research
in the Department of Medicine.
Dr. Moe is the principal investigator for several ongoing
basic and clinical research studies in the eld of CKD-MBD,
including studies on vascular calcication, mineral meta-
bolism, and bone metabolism in kidney disease. Her research
is funded by the Veterans Affairs Department, the National
Institutes of Health, foundations, and pharmaceutical com-
panies. She has authored over 200 scientic manuscripts,
teaching manuscripts, and textbook chapters. Dr. Moe served
on the National Kidney Foundations (NKF) KDOQI Bone
and Mineral Metabolism Clinical Practice Guideline Work
Group in 2003 and was co-chair of the international KDIGO
CKD-MBD guideline released in 2009.
Dr. Moes key honors include: election to the American
Society for Clinical Research in 2005; the NKF Garabed
Eknoyan Award for exceptional contributions to key initia-
tives such as KDOQI in 2009; councilor to the American
Heart Association Kidney Council (20022004); International
Society of Nephrology (20052007); councilor for the
American Society of Nephrology (20082015) and president
of the American Society of Nephrology (2013-2014); and
election to the Association of American Physicians in 2017.
Consultant: Lilly, Ultragenyx
Grants/research support: NIH, Veterans Administration
Speaker: Sano, University of Kansas
Stock/stock options: Lilly
web 4C=FPOweb 4C=FPO
Rukshana Shroff, MD, FRCPCH,
PhD, is a consultant in pediatric
nephrology at Great Ormond Street
Hospital for Children in London, UK,
and holds an academic position
(reader) in nephrology at University
College London. Her research focuses
on cardiovascular disease in child-
hood CKD, including laborat ory
work, clinical research studies, and clinical trials. She is the
principal investigator on a multicenter study comparing long-
term outcomes of conventional hemodialysis and hemodia-
ltration in children. She currently holds a prestigious senior
fellowship from the National Institute for Health Research to
continue research into mineral dysregulation in CKD. She has
published more than 130 original articles, reviews, and book
chapters in the elds of nephrology and dialysis. Dr. Shroff
has also ser ved on 2 guideline committees for the National
Institute for Health and Care Excellence. She is on the council
for the European Society for Pediatric Nephrology. She is
presently an associate editor for Pediatric Nephrology and
serves on the editorial board of Clinical Journal of the Amer-
ican Society of Nephrology.
Consultant: AstraZeneca
Grant/research support: Fresenius Medical Care*
Speaker: Amgen, Fresenius Medical Care
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Marcello A. Tonelli, MD, SM,
FRCPC, is senior associate dean
(clinical research) at the Cumming
School of Medicine and associate vice
president (health research) at the
University of Calgary. He received an
MD from the University of Western
Ontario, specialist certication in
nephrology (FRCPC) at Dalhousie
University, an SM in epidemiology
from Harvard University, and an MSc
in health policy from Imperial College London. He is a
nephrologist and professor at the University of Calgary.
www.kisupplements.org biographic and disclosure information
Kidney International Supplements (2017) 7, 1–59 51
Dr. Tonell i is the past president of the Canadian Society of
Nephrology, a past councilor of the International Society
of Nephrology, and the chair of the International Societ y of
Nephrology Research Committee. He is a fellow of the Ca-
nadian Academy of Health Sciences, and a member of the
American Society for Clinical Investigation. Dr. Tonelli is the
chair of the Canadian Task Force for Preventive Health Care, a
national panel of experts that makes recommendations about
preventive health services to Canadas 36,000 family
physicians.
Dr. Tonelli was the recipient of the 2013 United States
National Kidney Foundation Medal for Distinguished Service
and also the 2013 Kidney Foundation of Canada Medal for
Research Excellence.
Dr. Tonelli declared no competing interests.
web 4C=FPOweb 4C=FPO
Nigel D. Toussaint, MBBS, FRACP,
PhD, is the deputy director of
nephrology at Melbourne He al th
and a clinic al a ssociate p rofessor
within the De par tment of Medicine
at the University of Melbourne ,
Australia. Dr. Toussaint completed
his nephrology training in Mel-
bourne in 2005 and completed his
PhD studies at Monash Universit y in
2009, undertaking clinical research
in the a rea of vascular calcicat ion and cardiovascular r isk
in patients w ith CKD. He has also completed a graduate
diploma in clinical epidemiology (Monash University,
2007), and a National Health and Medical Research Council
National Institute of Clinic al Studies Fellowship (2011) in
the area of implementation research. He i s a member of the
Editorial Board for Nephrology and is cur rently a member of
council for the Australian and New Zealand Societ y of
Nephrology.
Dr. Toussaint practices nephrology at The Royal Mel-
bourne Hospital, where he is also the lead physician for
clinical research within the Nephrology Department. His
current research involves clinical epidemiology and clinical
trials, as well as translational research, in the areas of CKD-
MBD biomarkers, vascular calcication, and renal osteodys-
trophy. He has been an awardee of the Royal Australasian
College of Physicians Jacquot Research Establishment Award
and is a member of the Scientic Committee for the Aus-
tralasian Kidney Trials Network and the steering committee
for the Australia and New Zealand Dialysis and Transplant
Registry.
Advisory board: Amgen, Sano, Shire
Consultant: Amgen, Sano, Shire
Grants/research support: Amgen*, Shire*
Speaker: Amgen, Sano, Shire
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Marc G. Vervloet, MD, PhD, FERA,
is a nephrologist, associate professor of
nephrology, and director of the
nephrology research program and se-
nior consultant at the Intensive Care
Medicine and Vascular Medicine, VU
U niversity Medical Center , Amsterdam,
Netherlands. He is program leader at the
Institute of Cardiovascular Rese arch VU
(ICaR-VU), secretary of the CKD-MBD
working group of the Eur opean Renal
AssociationE ur opean Dialysis and Transplant Association, and a
member of the scientic committee of the Dutch Kidney Foun-
dation. Dr . Vervloet obtained his medical degree in 1989, and
graduated as internist in 1997 and as nephrologist in 1999. His
current resear ch interests encompass research on the deranged
bone and metabolism in CKD . Dr. Vervloet currently heads the
hemodialysis unit at his hospital, where he performs instructional
duties to medical students, residents, and nephrology fellows, and
guides several PhD students. He has gained numerous research
grants, mainly covering topics related to CKD-MBD, in particular
on the role of FGF23 and Klotho, and the clinical use of calci-
mimetic therapy . In addition, his laboratory research comprises
severalanimalmodelsofCKDandvitaminDdeciency, exam-
ining mainly cardiovascular endpoints as assessed by imaging
and functional testing in vivo.
Advisory board: Amgen, Fresenius Medical Care
Consultant: Amgen, Fresenius Medical Care, Otsuk a
Grants/research support: AbbVie*, Fresenius Medical Care*,
Sano*, Shire*
Speaker: Amgen, Baxter
*Monies paid to institution.
KDIGO Chairs
web 4C=FPOweb 4C=FPO
David C. Wheeler, MD, FRCP, is
professor of kidney medicine at Uni-
versity College London, UK, and
honorary consultant nep hrologist at
the Royal Free London NHS Foun-
dation Trust. He is a clinician scientist
with an interest in the complications
of CKD, specically those that in-
crease the burden of cardiovascular
disease and/or accelerate progression
of kidney failure. He has participated
in the design, roll-out, and monitoring of several large-scale
clinical trials . He was a member of the steering committee
of the Study of Heart and Renal Protection (SHARP) and the
EValuation Of Cinacalcet HCl Therapy to Lower CardioVas-
cular Events (EVOLVE). He currently sits on the steering
committee of Canaglifozin and Renal Events in Diabetes with
Established Nephropathy Clinical Evaluation (CREDENCE),
acting as UK principal investigator for this study. He is clinical
lead for Division 2 of the North Thame s Clinical Research
Network and heads a team of eight clinical trial nurses and
biographic and disclosure information www.kisupplements.org
52
Kidney International Supplements (2017) 7, 1–59
practitioners at the Centre for Nephrology, Royal Free
Hospital in London. He is past president of the UK Renal
Association and past chair of the UK Renal Registry. His
other responsibilities include serving as associate editor
of Nephrology Dialysis Transplantation and member of the
editorial board of Journal of the American Society of
Nephrology.
Consultant: Akebia, Alberta Innovates Health Solutions,
Amgen, AstraZeneca, Bio Nano Consulting, Boehringer
Ingelheim, Bristol-Myers Squibb, Fresenius, GSK, Janssen*,
Otsuka, UCB Celltech, Vifor
Speaker: Amgen, Fresenius Medical Care, Janssen, Vifor
Fresenius Medical Care Renal Pharma, ZS Pharma
*Monies paid to institution.
web 4C=FPOweb 4C=FPO
Wolfgang C. Winkelmayer, MD,
MPH, ScD, is the Gordon A. Cain
Chair of Nephrology and professor of
medicine at Baylor College of Medi-
cine in Houston, TX. Dr. Winkel-
mayer received his medical degree
(1990) from the University of Vienna,
Austria, and later earned a Master of
Public Health degree in health care
management (1999) and a Doctor of
Science degree in health policy (2001)
from Harvard University. He then spent 8 years on the faculty
of Brigham and Womens Hospital and Harvard Medical
School, where he established himself as a prolic investigator
and leader in the discipline of comparative-effectiveness
research as it pe rtains to patients with kidney disease. From
2009 to 2014, he was the director of clinical research in the
Division of Nephrology at Stan ford University School of
Medicine, Palo Alto, CA. He assumed his current position as
chief of nephrology at Baylor College of Medicine in
September 2014. His main areas of research interest include
comparative effectiveness and safety research of treatment
strategies in anemia as well as of various interventions for
cardiovascular disease in patient s with kidney disease. His
clinical passion lies in providing quality kidney care to the
predominantly disadvantaged and un(der)insured population
in the public safety net health system of Harris County, TX.
Dr. Winkelmayer has authored over 270 peer-reviewed pub-
lications, and he has a particular interest in medical pub-
lishing. He currently serves as an associate editor for The
Journal of the American Medical Association, was a co-editor of
the American Journal of Kidney Diseases from 2007 to 2016,
and has been appointed to several other editorial boards of
leading nephrology and epidemiology journals. He also vol-
unteers his time toward important initiatives of the American
Society of Nephrology (e.g., public policy board). He joined
KDIGO volunteer leadership as an executive committee
member in 2015 and has served as its co-chair since 2016.
Advisory board: Akebia, AMAG Pharmaceuticals, Amgen,
AstraZeneca, Bayer, Daichi Sankyo, Medtronic, Relypsa,
Vifor Fresensius Medical Care Renal Pharma
Consultant: Akebia, AMAG Pharmaceuticals, Amgen,
AstraZeneca, Bayer, Daichi Sankyo, Medtronic, Relypsa,
Vifor Fresensius Medical Care Renal Pharma
Evidence review team
Karen A. Robinson, PhD, is associate professor of medicine,
epidemiolog y and health policy & management, and director
of the Evidence-Based Practice Center at Johns Hopkins
University, serving as the project director. She provided
methodological expertise in the conduct of the systematic
review and guideline development processes, and oversaw and
participated in all aspects of the project, including topic
renement, abstract and full-text screening, data extraction,
study assessment, evidence grading, and recommendation
formulation.
Dr. Robinson declared no competing interests.
Casey M. Rebholz, PhD, MPH, MS, is assistant professor
of epidemiology at Johns Hopkins Bloomberg School of
Public Health and Core Faculty at the Welch Center for
Prevention, Epidemiology, and Clinical Research. She guided
the team through all phases of the project, includin g rening
the questions, conducting literature searches, screening ab-
stracts and full-text articles, abstracting data, drafting and
nalizing the evidence tables, and synthesizing the results.
Dr. Rebholz declared no competing interests.
Lisa M. Wilso n, ScM, is a research associate at the
Johns Hopkins University Bloomberg School of Public
Health. As the project coordinator for the evidence review
team, Ms. Wilson managed and participated in all phases
of the project, including conducting literature se arches,
screening abstracts and full-text ar ticles, abstracting data,
drafting and nalizing the ev idence tables, and synthesizing
the results.
Ms. Wilson declared no competing interests.
Ermias Jirru, MD, MPH, is currently an internal medicine
resident at Mount Sinai St. Lukes and Mount Sinai West. He
completed his MPH at the Johns Hopkins University
Bloomberg School of Public Health. For this project, he
participated in screening abstracts and full-text articles, and
abstracting data.
Dr. Jirru declared no competing interests.
Marisa Chi Liu, MD, MPH, is currently a resident
physician at University of California, Irvine, in obstetrics and
gynecology. She completed her MPH at Johns Hopkins Uni-
versity Bloomberg School of Public Health and graduated
from the University of Vermont Colleg e of Medicine. For this
project, she participated in screening abstracts and articles,
and abstracting data.
Dr. Liu declared no competing interests.
Jessica Gayleard, BS, is a research assistant within the
Johns Hopkins University Evidence-Based Practice Center.
She participated in all phases of the project, including con-
ducting literature searches, screening abstracts and full-text
www.kisupplements.org biographic and disclosure information
Kidney International Supplements (2017) 7, 1–59 53
articles, abstracting data, and drafting and nalizing the evi-
dence tables.
Ms. Gayleard declared no competing interests.
Allen Zhang, BS, is a research data analyst at the Johns
Hopkins University Evidence-Based Practice Center. He has a
degree in microbiology from the Virginia Polytechnic
Institute and University. Mr. Zhang participated in the sys-
tematic review development, including search string creation,
screening, data cleaning, data manage ment, and writing. In
addition, he supplied the necessary statistical analysis,
including meta-analyses and meta-regression calculations.
Mr. Zhang declared no competing interests.
biographic and disclosure information www.kisupplements.org
54
Kidney International Supplements (2017) 7, 1–59
Acknowledgments
A special debt of gratitude is owed to the KDIGO co-chairs,
David Wheeler and Wolfgang Winkelmayer, for their invalu-
able guidance throughout the development of this guideline.
In particular, we thank Karen Robinson and her ERT mem-
bers for their substantial contribution to the rigorous
assessment of the available evidence. We are also especially
grateful to the Work Group members for their expertise
throughout the entire process of literature review, data
extraction, meeting participation, the critical writing and
editing of the statements and rationale, which made the
publication of this guideline possible. The generous gift of
their time and dedication is greatly appreciated. Finally, and
on behalf of the Work Group, we gratefully acknowledge the
careful assessment of the draft guideline by external reviewers.
The Work Group considered all of the valuable comments
made, and where appropriate, suggested changes were
incorporated into the nal publication. The following in-
dividuals provided feedback during the public review of the
draft guideline:
Patricia Abreu, Adama Lengani, Kamal Ahmed, Bülent
Altun, Luis Felipe Alva, Rui Alves, Pablo Amair, Alessandro
Amore, Andrea Angioi, Mustafa Arici, Mariano Arriola,
Rommel Bataclan, Ezequiel Bellorin-Font, Deborah Benner,
Mohammed Benyahia, Patrick Biggar, Charles Bishop, Boris
Bogov, Jordi Bover, Laura Brereton, Philippe Brunet, Rafael
Burgos-Calderon, Stephen Carrithers, Sue Cary, Rolando
Claure-Del Granado, Adrian Covic, Mario Cozzolino,
Andrew Crannage, John Cunningham, Pierre Delanaye,
Nida Dinçel, Tilman B. Drüeke, Nordin Eezsafryna Azalin,
Grahame J. Elder, Madgy ElSharkawy, Joyce Ezaki-Yamaguchi,
Toshiro Fujita, Alvaro Garc ia, Carlo Francisco Gochui co,
Heong Keong Goh, Hai An Ha Phan, Takayuki Hamano,
Ditte Hansen, Li Hao, Eero Honkanen, Alastair Hutchison ,
Atul Ingale, Joachim H. Ix, Faical Jarraya, Chandra Mauli
Jha, Kamyar Kalantar-Zadeh, Arif Khwaja, Csaba P. Kovesdy,
Holly Kramer, Craig B. Langman, Kevin V. Lemley, Edgar V.
Lerma, Nathan W. Levin, Maria Jesus Lloret, José António
Lopes, Franklin W. Maddux, Francesca Mallamaci, Sandro
Mazzaferro, Peter A. McCullough, Donald A. Molony,
Sameh Morgan, Eugen Mota, Ricardo Mouzo, Lavinia
Neg rea, Armando Negri , Michal Nowicki , Tom Nusbic kel,
Basma Osman, Susan M. Ott, Antonino Paglialunga, Saime
Paydas, Adriana Peñalba, Gerson Marques Pereir a Junior,
Eduardo Perez, Ligia Petrica, Friedrich K. Port, Pradeep
Kumar Rai, Dwarakanathan Ranganathan, Nicolas Roberto
Robles, Cibele Rodrigues, Hector Rodriguez, Guillermo
Rosa Diez, Ibrahim Saig, Deepak Sharma, Laura Sola, David
M. Spiegel, Kyriaki Stamatelou, Ekamol Tantisattamo,
Mihály Tapolyai, Francesca Tentori, Katrin Uhlig, Harun Ur
Rashid, Pablo Ureña Torres, Keth Vuthy, A ngela Yee-Moon
Wang , Talia Weinstein, Jane Wheeler, Janie Xiong, and
Xueqing Y u.
Participation in the review does not necessarily consti-
tute endorsement of the content of this report by the
above individuals, or the organization or institution they
represent.
Markus Ketteler, MD, FERA
Mary B. Leonard, MD, MSCE
Work Group Co-chairs
www.kisupplements.org acknowledgments
Kidney International Supplements (2017) 7, 1–59 55
References
1. Ketteler M, Elder GJ, Evenepoel P, et al. Revisiting KDIGO clinical practice
guideline on chronic kidney disease-mineral and bone disorder: a
commentary from a Kidney Disease: Improving Global Outcomes
controversies conference. Kidney Int. 2015;87:502528.
2. Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture
among patients with end-stage renal disease. Kidney Int. 2000;58:396399.
3. Ball AM, Gillen DL, Sherrard D, et al. Risk of hip fracture among dialysis
and renal transplant recipients. JAMA. 2002;288:30143018.
4. Nickolas TL, McMahon DJ, Shane E. Relationship between moderate to
severe kidney disease and hip fracture in the United States. J Am Soc
Nephrol. 2006;17:3223 3232.
5. Beaubrun AC, Kilpatrick RD, Freburger JK, et al. Temporal trends in
fracture rates and postdischarge outcomes among hemodialysis patients.
J Am Soc Nephrol. 2013;24:14611469.
6. Lin ZZ, Wang JJ, Chung CR, et al. Epidemiology and mortality of hip
fracture among patients on dialysis: Taiwan National Cohort Study. Bone.
2014;64:235239.
7. Maravic M, Ostertag A, Tor res PU, et al. Incidence and risk factors for hip
fractures in dialysis patients. Osteoporos Int. 2014;25:159165.
8. Mittalhenkle A, Gillen DL, Stehman-Breen CO. Increased risk of mortality
associated with hip fracture in the dialysis population. Am J Kidney Dis.
2004;44:672679.
9. Tentor i F, McCullough K, Kilpatrick RD, et al. High rates of death and
hospitalization follow bone fracture among hemodialysis patients. Kidney
Int. 2014;85:166173.
10. Iimori S, Mori Y, Akita W, et al. Diagnostic usefulness of bone mineral
density and biochemical markers of bone turnover in predicting fracture
in CKD stage 5D patientsa single-center cohort study. Nephrol Dial
Transplant. 2012;27:345351.
11. Naylor KL, Garg AX, Zou G, et al. Comparison of fracture risk prediction
among indiv iduals with reduced and normal kidney function. Clin J Am Soc
Nephrol. 2015;10:646653.
12. West SL, Lok CE, Langsetmo L, et al. Bone mineral density predicts
fractures in chronic kidney disease. J Bone Miner Res. 2015;30:913
919.
13. Yenchek RH, Ix JH, Shlipak MG, et al. Bone mineral density and fracture
risk in older individuals with CKD. Clin J Am Soc Nephrol. 2012;7:
11301136.
14. Bianchi ML, Leonard MB, Bechtold S, et al. Bone health in children and
adolescents with chronic diseases that may affect the skeleton: the 2013
ISCD Pediatric Ofcial Positions. J Clin Densitom. 2014;17:281294.
15. Bishop N, Arundel P, Clark E, et al. Fracture prediction and the de nition of
osteoporosis in children and adolescents: the ISCD 2013 Pediatric Ofcial
Positions. J Clin Densitom. 2014;17:275280.
16. Zemel BS, Leonard MB, Kelly A, et al. Height adjustment in assessing dual
energy x-ray absorptiometry measurements of bone mass and density in
children. J Clin Endocrinol Metab. 2010;95:12651273.
17. Crabtree NJ, Arabi A, Bachrach LK, et al. Dual-energy X-ray absorptiometry
interpretation and reporting in children and adolescents: the revised 2013
ISCD Pediatric Ofcial Positions. J Clin Densitom. 2014;17:225242.
18. Grifn LM, Kalkwarf HJ, Zemel BS, et al. Assessment of dual-energy X-ray
absorptiometry measures of bone health in pediatric chronic kidney disease.
Pediatr Nephrol. 2012;27:1139 1148.
19. Denburg MR, Tsampalieros AK, de Boer IH, et al. Mineral metabolism and
cortical volumetric bone mineral density in childhood chronic kidney
disease. J Clin Endocrinol Metab. 2013;98:19301938.
20. Ishani A, Blackwell T, Jamal SA, et al. The effect of raloxifene treatment in
postmenopausal women with CKD. J Am Soc Nephrol. 2008;19:14301438.
21. Jamal SA, Bauer DC, Ensrud KE, et al. Alendronate treatment in women
with normal to severely impaired renal function: an analysis of the
fracture intervention trial. J Bone Miner Res. 2007;22:503508.
22. Miller PD, Roux C, Boonen S, et al. Safety and efcacy of risedronate in
patients with age-related reduced renal function as estimated by the
Cockcroft and Gault method: a pooled analysis of nine clinical trials.
J Bone Miner Res. 2005;20:21052115.
23.
Miller PD, Schwartz EN, Chen P, et al. Teriparatide in postmenopausal
women with osteoporosis and mild or moderate renal impairment.
Osteoporos Int. 2007;18:5968.
24. Jamal SA, Ljunggren O, Stehman-Breen C, et al. Effects of denosumab on
fracture and bone mineral density by level of kidney function. J Bone
Miner Res. 2011;26:18291835.
25. Toussaint ND, Lau KK, Strauss BJ, et al. Effect of alendronate on vascular
calcication in CKD stages 3 and 4: a pilot randomized controlled trial.
Am J Kidney Dis. 2010;56:5768.
26. Haghverdi F, Farbodara T, Mortaji S, et al. Effect of raloxifene on
parathyroid hormone in osteopenic and osteoporotic postmenopausal
women with chronic kidney disease stage 5. Iran J Kidney Dis. 2014;8:
461466.
27. Wren TA, Kalkwarf HJ, Zemel BS, et al. Longitudinal tracking of dual-
energy X-ray absorptiometry bone measures over 6 years in children and
adolescents: persistence of low bone mass to maturity. J Pediatr. 2014;164:
12801285.
28. Zemel BS, Kalkwarf HJ, Gilsanz V, et al. Revised reference curves for bone
mineral content and areal bone mineral density according to age and sex for
black and non-black children: results of the bone mineral density in
childhood study. J Clin Endocrinol Metab. 2011;96:31603169.
29. Moe S, Drueke T, Cunningham J, et al. Denition, evaluation, and
classication of renal osteodystrophy: a position statement from Kidney
Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006;69:
19451953.
30. Kidney Disease: Improving Global Outcomes (KDIGO) CKDMBD
Work Group. KDIGO clinical practice guideline for the diagnosis,
evaluation, prevention, and treatment of chronic kidney disease
mineral and bone disorder (CKDMBD). Kidney Int. 2009;76(Suppl
113):S1S130.
31. Sprague SM, Bellorin-Font E, Jorgetti V, et al. Diagnostic accuracy of bon e
turnover markers and bone histology in patients with CKD treated by
dialysis. Am J Kidney Dis. 2016;67:559566.
32. Amerling R, Harbord NB, Pullman J, et al. Bisphosphonate use in chronic
kidney disease: association with adynamic bone disease in a bone histology
series. Blood Purif. 2010;29:293299.
33. Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for
treatment of postmenopausal osteoporosis. N Engl J Med. 2007;356:
18091822.
34. Evenepoel P, DHaese P, Bacchetta J, et al. Bone biopsy practice patterns
across Europe: the European renal osteodystrophy (EU-ROD) initiative-a
position paper [e-pub ahead of print]. Nephrol Dial Transplant. http://dx.
doi.org/10.1093/ndt/gfw468.
35. Moe SM, Zidehsarai MP, Chambers MA, et al. Vegetarian compared with
meat dietary protein source and phosphorus homeostasis in chronic
kidney disease. Clin J Am Soc Nephrol. 2011;6:257264.
36. Viaene L, Meijers B, Vanrenterghem Y, et al. Day time rhythm and
treatment-related uctuations of serum phosphorus concentration in
dialysis patients. Am J Nephrol. 2012;35:242248.
37. Block GA, Kilpatrick RD, Lowe KA, et al. CKD-mineral and bone disorder
and risk of death and cardiovascular hospitalization in patients on
hemodialysis. Clin J Am Soc Nephrol. 2013;8:21322140.
38. Chertow GM, Block GA, Correa-Rotter R, et al. Effect of cinacalcet on
cardiovascular disease in patients undergoing dialysis. N Engl J Med.
2012;367:24822494.
39. Chartsrisak K, Vipattawat K, Assanatham M, et al. Mineral metabolism and
outcomes in chronic kidney disease stage 2-4 patients. BMC Nephrol.
2013;14:14.
40. Connolly GM, Cunningham R, McNamee PT, et al. Elevated serum
phosphate predicts mortality in renal transplant recipients.
Transplantation. 2009;87:10401044.
41. Eddington H, Hoeeld R, Sinha S, et al. Serum phosphate and mortality in
patients with ch ronic kidney disease. Clin J Am Soc Nephrol. 2010;5:
22512257.
references www.kisupplements.org
56
Kidney International Supplements (2017) 7, 1–59
42. Fouque D, Roth H, Pelletier S, et al. Control of mineral metabolism and
bone disease in haemodialysis patients: which optimal targets? Nephrol
Dial Transplant. 2013;28:360367.
43. Fukagawa M, Kido R, Komaba H, et al. Abnormal mineral metabolism and
mortality in hemodialysis patients with secondary hyperparathyroidism:
evidence from marginal structural models used to adjust for time-
dependent confounding. Am J Kidney Dis. 2014;63:979987.
44. Lacson E Jr, Wang W, Hakim RM, et al. Associates of mortality and
hospitalization in hemodialysis: potentially actionable laboratory variables
and vascular access. Am J Kidney Dis. 2009;53:7990.
45. McGovern AP, de Lusignan S, van Vlymen J, et al. Serum phosphate as a
risk factor for cardiovascular events in people w ith and without chronic
kidney disease: a large community based cohort study. PLoS One. 2013;8:
e74996.
46. Moore J, Tomson CR, Tessa Savage M, et al. Serum phosphate and calcium
concentrations are associated with reduced patient surv ival following
kidney transplantation. Clin Transplant. 2011;25:406416.
47. Nakai S, Akiba T, Kazama J, et al. Effects of serum calcium, phosphorous,
and intact parathyroid hormone levels on survival in chronic
hemodialysis patients in Japan. Ther Apher Dial. 2008;12:4954.
48. Tentori F, Blayney MJ, Albert JM, et al. Mortalit y risk for dialysis patients
with different levels of serum calcium, phosphorus, and PTH: the
Dialysis Outcomes and Practice Patterns Study (DOPPS). Am J Kidney
Dis. 2008;52:519530.
49. Fliser D, Kollerits B, Neyer U, et al. Fibroblast growth factor 23 (FGF23)
predicts progression of chronic kidney disease: the Mild to Moderate
Kidney Disease (MMKD) study. J Am Soc Nephrol. 2007;18:26002608.
50. Floege J, Kim J, Ireland E, et al. Serum iPTH, calcium and phosphate, and
the risk of mortalit y in a European haemodialysis population. Nephrol Dial
Transplant. 2011;26:19481955.
51. Gross P, Six I, Kamel S, et al. Vascular toxicity of phosphate in chronic
kidney disease: beyond vascular calcication. Circ J. 2014;78:23392346.
52. Cannata-Andia JB, Fernandez-Martin JL, Locatelli F, et al. Use of
phosphate-binding agents is associated with a lower risk of mortality.
Kidney Int. 2013;84:9981008.
53. Isakova T, Gutierrez OM, Chang Y, et al. Phosphorus binders and survival
on hemodialysis.
J Am Soc Nephrol. 2009;20:388396.
54. Lopes AA, Tong L, Thumma J, et al. Phosphate binder use and mortality
among hemodialysis patients in the Dialysis Outcomes and Practice
Patterns Study (DOPPS): evaluation of possible confounding by
nutritional status. Am J Kidney Dis. 2012;60:90101.
55. Fernandez-Martin JL, Martinez-Camblor P, Dionisi MP, et al. Improvement
of mineral and bone metabolism markers is associated with better survival
in haemo dialysis patients: the COSMOS study. Nephrol Dial Transplant.
2015;30:15421551.
56. Scialla JJ, Astor BC, Isakova T, et al. Mineral metabolites and CKD
progression in African Americans. J Am Soc Nephrol. 2013;24:125135.
57. Silva AP, Fragoso A, Pinho A, et al. Phosphorus as an early marker of
morbidity and mortality in type 2 chronic kidney disease diabetic
patients. J Diabetes Complications. 2013;27:328332.
58. Kalantar-Zadeh K, Kuwae N, Regidor DL, et al. Surv ival predictability of
time-varying indicators of bone disease in maintenance hemodialysis
patients. Kidney Int. 2006;70:771780.
59. Block GA, Wheeler DC, Persky MS, et al. Effects of phosphate binders in
moderate CKD. J Am Soc Nephrol. 2012;23:14071415.
60. Hill KM, Martin BR, Wastney ME, et al. Oral calcium carbonate affects
calcium but not phosphorus balance in stage 3-4 chronic kidney disease.
Kidney Int. 2013;83:959966.
61. de Boer IH, Rue TC, Kestenbaum B. Serum phosphorus concentrations in
the third National Health and Nutrition Examination Survey (NHANES
III). Am J Kidney Dis. 2009;53:399407.
62. Newsome B, Ix JH, Tighiouart H, et al. Effect of protein restriction on
serum and urine phosphate in the modication of diet in renal disease
(MDRD) study. Am J Kidney Dis. 2013;61:10451046.
63. Selamet U, Tighiouart H, Sarnak MJ, et al. Relationship of dietary
phosphate intake with risk of end-stage renal disease and mortality in
chronic kidney disease stages 3-5: The Modication of Diet in Renal
Disease Study. Kidney Int. 2016;89:176184.
64. Salusky IB, Goodman WG, Sahney S, et al. Sevelamer controls parathyroid
hormone-induced bone disease as efciently as calcium carbonate without
increasing serum calcium levels during therapy with active vitamin D
sterols. J Am Soc Nephrol. 2005;16:25012508.
65. Pieper AK, Haffner D, Hoppe B, et al. A randomized crossover trial
comparing sevelamer with calcium acetate in children with CKD. Am J
Kidney Dis. 2006;47:625635.
66. Litwin M, Wuhl E, Jourdan C, et al. Evolution of large-vessel arteriopathy in
paediatric patients with chronic kidney disease. Nephrol Dial Transplant.
2008;23:25522557.
67. Oh J, Wunsch R, Turzer M, et al. Advanced coronary and carotid
arteriopathy in young adults with childhood-onset chronic renal failure.
Circulation. 2002;106:100105.
68. Shroff RC, Donald AE, Hiorns MP, et al. Mineral metabolism and vascular
damage in children on dialysis. J Am Soc Nephrol. 2007;18:29963003.
69. Mitsnefes MM, Kimball TR, Kartal J, et al. Cardiac and vascular adaptation
in pediatric patients with chronic kidney disease: role of calcium-
phosphorus metab olism. J Am Soc Nephrol. 2005;16:27962803.
70. Civilibal M, Caliskan S, Adaletli I, et al. Coronar y artery calcications in
children with end-stage renal disease. Pediatr Nephrol. 2006;21:14261433.
71. Goodman WG, Goldin J, Kuizon BD, et al. Coronary-artery calcication in
young adults with end-stage renal disease who are undergoing dialysis.
N Engl J Med. 2000;342:14781483 .
72. Coen G, Pierantozzi A, Spizzichino D, et al. Risk factors of one year
increment of coronary calcications and survival in hemodialysis patients.
BMC Nephrol. 2010;11:10.
73. Fein PA, Asadi S, Singh P, et al. Relationship between alkaline phosphatase
and all-cause mortality in peritoneal dialysis patients. Adv Perit Dial.
2013;29:6163.
74. Markaki A, Kyriazis J, Stylianou K, et al. The role of serum magnesium and
calcium on the association between adiponectin levels and all-cause
mortality in end-stage renal disease patients. PLoS One. 2012;7:e52350.
75. Gallieni M, Caputo F, Filippini A, et al. Prevalence and progression of
cardiovascular calcications in peritoneal dialysis patients: A prospective
study. Bone. 2012;51:332337
.
76. Nakano C, Hamano T, Fujii N, et al. Combined use of vitamin D status and
FGF23 for risk stratication of renal outcome. Clin J Am Soc Nephrol.
2012;7:810819.
77. Rutherford WE, Blondin J, Hruska K, et al. Effect of 25-
hydroxycholecalciferol on calcium absorption in chronic renal disease.
Kidney Int. 1975;8:320324.
78. Raggi P, Bommer J, Chertow GM. Valvular calcication in hemodialysis
patients randomized to calcium-based phosphorus binders or sevelamer.
J Heart Valve Dis. 2004;13:134141.
79. Spiegel DM, Brady K. Calcium balance in normal individuals and in
patients with chronic kidney disease on low- and high-calcium diets.
Kidney Int. 2012;81:11161122.
80. Chonchol M, Locatelli F, Abboud HE, et al. A randomized, double-blind,
placebo-controlled study to assess the efcacy and safety of cinacalcet
HCl in participants with CKD not receiving dialysis. Am J Kidney Dis.
2009;53:197207.
81. St Peter WL, Li Q, Liu J, et al. Cinacalcet use patterns and effect on
laboratory values and other medications in a large dialysis organization,
2004 through 2006. Clin J Am Soc Nephrol. 2009;4:354360.
82. Baxter-Jones AD, Faulkner RA, Forwood MR, et al. Bone mineral accrual
from 8 to 30 years of age: an estimation of peak bone mass. J Bone Miner
Res. 2011;26:17291739.
83. Bailey DA, Martin AD, McKay HA, et al. Calcium accretion in girls and
boys during puberty: a longitudinal analysis. J Bone Miner Res. 2000;15:
22452250.
84. Gotch FA, Kotanko P, Thijssen S, et al. The KDIGO guideline for dialysate
calcium will result in an increased incidence of calcium accumulation in
hemodialysis patients. Kidney Int. 2010;78:343350.
85. Ok E, Asci G, Bayraktaroglu S, et al. Reduction of dialysate calcium level
reduces progression of coronary artery calcication and improves low
bone turnover in patients on hemodialysis. J Am Soc Nephrol. 2016;27:
24752486.
86. Spasovski G, Gelev S, Masin-Spasovska J, et al. Improvement of bone and
mineral parameters related to adynamic bone disease by diminishing
dialysate calcium. Bone. 2007;41:698703.
87. Brunelli SM, Sibbel S, Do TP, et al. Facility dialysate calcium practices and
clinical outcomes among patients receiving hemodialysis: a retrospective
observational study. Am J Kidney Dis. 2015;66:655665.
88. Kim HW, Kim SH, Kim YO, et al. Impact of dialysate calcium concentration
on clinical outcomes in incident hemodialysis patients. Medicine
(Baltimore). 2015;94:e1694.
www.kisupplements.org references
Kidney International Supplements (2017) 7, 1–59 57
89. Di Iorio B, Bellasi A, Russo D. Mortality in kidney disease patients treated
with phosphate binders: a randomized study. Clin J Am Soc Nephrol. 2012;7:
487493.
90. Di Iorio B, Molony D, Bell C, et al. Sevelamer versus calcium carbonate in
incident hemodialysis patients: results of an open-label 24-month
randomized clinical trial. Am J Kidney Dis. 2013;62:771778.
91. Jamal SA, Vandermeer B, Raggi P, et al. Effect of calcium-based versus non-
calcium-based phosphate binders on mortality in patients with chronic
kidney disease: an updated systematic review and meta-analysis. Lancet.
2013;382:12681277.
92. Palmer SC, Gardner S, Tonelli M, et al. Phosphate-binding agents in adults
With CKD: a network meta-analysis of randomized trials. Am J Kidney Dis.
2016;68:691702.
93. Patel L, Bernard LM, Elder GJ. Sevelamer versus calcium-based binders for
treatment of hyperphosphatemia in CKD: a meta-analysis of randomized
controlled trials. Clin J Am Soc Nephrol. 2016;11:232244.
94. Sekercioglu N, Thabane L, Diaz Martinez JP, et al. Comparative
effectiveness of phosphate binders in patients with chronic kidney
disease: a systematic review and network meta-analysis. PLoS One.
2016;11:e0156891.
95. Floege J, Covic AC, Ketteler M, et al. A phase III study of the efcacy and
safety of a novel iron-based phosphate binder in dialysis patients. Kidney
Int. 2014;86:638647.
96. Van Buren PN, Lewis JB, Dwyer JP, et al. The phosphate binder ferric citrate
and mineral metabolism and inammatory markers in maintenance dialysis
patients: results from prespecied analyses of a randomized clinical trial.
Am J Kidney Dis. 2015;66:479488.
97. Lewis JB, Sika M, Koury MJ, et al. Ferric citrate controls phosphorus and
delivers iron in patients on dialysis. J Am Soc Nephrol. 2015;26:493503.
98. Ohlsson C, Darelid A, Nilsson M, et al. Cortical consolidation due to
increased mineralization and endosteal contraction in young adult men: a
ve-year longitudinal study. J Clin Endocrinol Metab. 2011;96:22622269.
99. Berger C, Goltzman D, Langsetmo L, et al. Peak bone mass from
longitudinal data: implications for the prevalence, pathophysiology, and
diagnosis of osteoporosis. J Bone Miner Res
. 2010;25:19481957.
100. Darelid A, Ohlsson C, Nilsson M, et al. Catch up in bone acquisition in
young adult men with late normal puberty. J Bone Miner Res. 2012;27:
21982207.
101. Denburg MR, Kumar J, Jemielita T, et al. Fracture burden and risk factors in
childhood CKD: results from the CKiD cohort study. J Am Soc Nephrol.
2016;27:543550.
102. El Borolossy R, El Wakeel LM, El Hakim I, et al. Efcacy and safety
of nicotinamide in the management of hyperphosphatemia in
pediatric patients on regular hemodialysis. Pediatr Neph rol. 2016;31:
289296 .
103. Block GA, Rosenbaum DP, Leonsson-Zachrisson M, et al. Effect of
tenapanor on serum phosphate in patients receiving hemodialysis. JAm
Soc Nephrol. 2017;28:19331942.
104. Karavetian M, de Vries N, Elzein H, et al. Effect of behavioral stage-based
nutrition education on management of osteodystrophy among
hemodialysis patients. Lebanon. Patient Educ Couns. 2015;98:11161122.
105. Lou LM, Caverni A, Gimeno JA, et al. Dietary intervention focused on
phosphate intake in hemodialysis patients with hyperphosphoremia. Clin
Nephrol. 2012;77:476483.
106. Liu Z, Su G, Guo X, et al. Dietary interventions for mineral and bone
disorder in people with chronic kidney disease. Cochrane Database Syst
Rev. 2015:CD010350.
107. Calvo MS, Park YK. Changing phosphorus content of the U.S. diet:
potential for adverse effects on bone. J Nutr. 1996;126:1168S1180S.
108. Russo D, Miranda I, Ruocco C, et al. The progression of coronary artery
calcication in predialysis patients on calcium carbonate or sevelamer.
Kidney Int. 2007;72:12551261.
109. Zeller K, Whittaker E, Sullivan L, et al. Effect of restricting dietary protein
on the progression of renal failure in patients with insulin-dependent
diabetes mellitus. N Engl J Med. 1991;324:7884.
110. Anderson JJB, Sell ML, Garner S, Calvo MS. Phosphorus. In: Bowman BA,
Russell RM, eds. Present Knowledge of Nutrition. Washington, DC: ILSI
Press; 2001
.
111. Sherman RA, Ravella S, Kapoian T. The phosphate content of
prescription medication: a new consideration. Ther Innov Regul Sci.
2015;49:886889.
112. Benini O, DAlessandro C, Gianfaldoni D, et al. Extra-phosphate load from
food additives in commonly eaten foods: a real and insidious danger for
renal patients. J Ren Nutr. 2011;21:303308.
113. Sherman RA, Mehta O. Phosphorus and potassium content of enhanced
meat and poultry products: implications for patients who receive dialysis.
Clin J Am Soc Nephrol. 2009;4:13701373.
114. Calvo MS. Dietary considerations to prevent loss of bone and renal
function. Nutrition. 2000;16:564566.
115. Caldeira D, Amaral T, David C, et al. Educational strategies to reduce serum
phosphorus in hyperphosphatemic patients with chronic kidney disease:
systematic review with meta-analysis. J Ren Nutr. 2011;21:285294.
116. Sullivan C, Sayre SS, Leon JB, et al. Effect of food additives on
hyperphosphatemia among patients with end-stage renal disease: a
randomized controlled trial. JAMA. 2009;301:629635.
117. Durose CL, Holdsworth M, Watson V, et al. Knowledge of dietary
restrictions and the medical consequences of noncompliance by patients
on hemodialysis are not predictive of dietar y compliance. J Am Diet
Assoc. 2004;104:3541.
118. Evenepoel P, Bover J, Urena Torres P. Parathyroid hormone metabolism and
signaling in health and chronic kidney disease. Kidney Int. 2016;90:
11841190.
119. Oksa A, Spustova V, Krivosikova Z, et al. Effects of long-term cholecalciferol
supplementation on mineral metabolism and calciotropic hormones in
chronic kidney disease. Kidney Blood Press Res. 2008;31:322329.
120. Chue CD, Townend JN, Moody WE, et al. Cardiovascular effects of
sevelamer in stage 3 CKD. J Am Soc Nephrol. 2013;24:842852.
121. Lemos MM, Watanabe R, Carvalho AB, et al. Effect of rosuvastatin and
sevelamer on the progression of coronary artery calcication in chronic
kidney disease: a pilot study. Clin Nephrol. 2013;80:18.
122. Coyne D, Acharya M, Qiu P, et al. Paricalcitol capsule for the treatment of
secondary hyperparathyroidism in stages 3 and 4 CKD.
Am J Kidney Dis.
2006;47:263276.
123. Coburn JW, Maung HM, Elangovan L, et al. Doxercalciferol safely
suppresses PTH levels in patients with secondary hyperparathyroidism
associated with chronic kidney disease stages 3 and 4. Am J Kidney Dis.
2004;43:877890.
124. Nordal KP, Dahl E. Low dose calcitriol versus placebo in patients with
predialysis chronic renal failure. J Clin Endocrinol Metab. 1988;67:929936.
125. Hamdy NA, Kanis JA, Beneton MN, et al. Effect of alfacalcidol on natural
course of renal bone disease in mild to moderate renal failure. BMJ.
1995;310:358363.
126. Fishbane S, Chittineni H, Packman M, et al. Oral paricalcitol in the
treatment of patients with CKD and proteinuria: a randomized trial. Am
J Kidney Dis. 2009;54:647652.
127. Wang AY, Fang F, Chan J, et al. Effect of paricalcitol on left ventricular mass
and function in CKDthe OPERA trial. J Am Soc Nephrol. 2014;25:175186.
128. Thadhani R, Appelbaum E, Pritchett Y, et al. Vitamin D therapy and cardiac
structure and function in patients with chronic kidney disease: the PRIMO
randomized controlled trial. JAMA. 2012;307:674684.
129. Li XH, Feng L, Yang ZH, et al. The effect of active vitamin d on
cardiovascular outcomes in predialysis chronic kidney diseases: a
systematic review and meta-analysis. Nephrology (Carlton). 2015;20:
706714.
130. Xu L, Wan X, Huang Z, et al. Impact of vitamin D on chronic kidney
diseases in non-dialysis patients: a meta-analysis of randomized
controlled trials. PLoS One. 2013;8:e61387.
131. Coyne DW, Goldberg S, Faber M, et al. A randomized multicenter trial of
paricalcitol versus calcitriol for secondary hyperparathyroidism in stages
3-4 CKD. Clin J Am Soc Nephrol. 2014;9:16201626.
132. Riccio E, Sabbatini M, Bruzzese D, et al. Effect of paricalcitol vs calcitriol on
hemoglobin levels in chronic kidney disease patients: a randomized trial.
PLoS One. 2015;10:e0118174.
133. Kovesdy CP, Lu JL, Malakauskas SM, et al. Paricalcitol versus ergocalciferol
for secondary hyperparathyroidism in CKD stages 3 and 4: a randomized
controlled trial. Am J Kidney Dis. 2012;59:5866.
134. Moe SM, Saifullah A, LaClair RE, et al. A randomized trial of cholecalciferol
versus doxercalciferol for lowering parathyroid hormone in chronic kidney
disease. Clin J Am Soc Nephrol. 2010;5:299
306.
135. Sprague SM, Crawford PW, Melnick JZ, et al. Use of extended-release
calcifediol to treat secondary hyperparathyroidism in stages 3 and 4
chronic kidney disease. Am J Nephrol. 2016;44:316325.
136. Hahn D, Hodson EM, Craig JC. Interventions for metabolic bone disease in
children with chronic kidney disease. Cochrane Database Syst Rev. 2015:
CD008327.
137. Kubo Y, Sterling LR, Parfrey PS, et al. Assessing the treatment effect in a
randomized controlled trial with extensive non-adherence: the EVOLVE
trial. Pharm Stat. 2015;14:242251.
references www.kisupplements.org
58
Kidney International Supplements (2017) 7, 1–59
138. Parfey PS, Drüeke TB, Block GA, et al. The effects of cinacalcet in older and
younger patients on hemodialysis: the Evaluation of Cinacalcet HCI
Therapy to Lower Cardiovascular Events (EVOLVE) trial. Clin J Am Soc
Nephrol. 2015;10:791799.
139. Parfrey PS, Chertow GM, Block GA, et al. The clinical course of treated
hyperparathyroidism among patie nts receiving hemodialysis and the effect
of cinacalcet: the EVOLVE trial. J Clin Endocrinol Metab. 2013;98:
48344844.
140. Moe SM, Abdalla S, Chertow GM, et al. Effects of cinacalcet on fracture
events in patients receiving hemodialysis: the EVOLVE trial. J Am Soc
Nephrol. 2015;26:1466 1475.
141. Moe SM, Chertow GM, Parfrey PS, et al. Cinacalcet, broblast growth
factor-23, and cardiovascular disease in hemodialysis: the evaluation of
cinacalcet HCl therapy to lower cardiovascular events (EVOLVE) trial.
Circulation. 2015;132:2739.
142. Urena-Torres P, Bridges I, Christiano C, et al. Efcacy of cinacalcet with
low-dose vitamin D in incident haemodialysis subjects with secondary
hyperparathyroidism. Nephrol Dial Transplant. 2013;28:12411254.
143. Raggi P, Chertow GM, Torres PU, et al. The ADVANCE study: a randomized
study to evaluate the effects of cinacalcet plus low-dose vitamin D on
vascular calcication in patients on hemodialysis. Nephrol Dial Transplant.
2011;26:13271339.
144. Wetmore JB, Gurevich K, Sprague S, et al. A randomized trial of
cinacalcet versus vitamin D analogs as monotherapy in secondary
hyperparathyroidism (PARADIGM). J Clin Am Soc Nephrol. 2015;10:
10311040.
145. Behets GJ, Spasovski G, Sterling LR, et al. Bone histomorphometry before
and after long-term treatment with cinacalcet in dialysis patients with
secondary hyperparathyroidism. Kidney Int. 2015;87:846856.
146. Block GA, Bushinsky DA, Cheng S, et al. Effect of etelcalcetide vs cinacalcet
on serum parathyroid hormone in patients receiving hemodialysis with
secondary hyperparathyroidism: a randomized clinical trial. JAMA.
2017;317:156164.
147. Block GA, Bushinsky DA, Cunningham J, et al. Effect of etelcalcetide vs
placebo on serum parathyroid hormone in patients receiving
hemodialysis with secondary hyperparathyroidism: two randomized
clinical trials. JAMA. 2017;317:146155.
148. Nichols KR, Knoderer CA, Johnston B, et al. Cinacalcet administration by
gastrostomy tube in a child receiving peritoneal dialysis. J Pediatr
Pharmacol Ther. 2014;19:202
205.
149. Dotis J, Printza N, Ghogha C, et al. Short- and middle-term continuous use
of cinacalcet in children on peritoneal dialysis. J Pediatr Endocrinol Metab.
2013;26:3943.
150. Platt C, Inward C, McGraw M, et al. Middle-term use of Cinacalcet in
paediatric dialysis patients. Pediatr Nephrol. 2010;25:143148.
151. Alharthi AA, Kamal NM, Abukhatwah MW, et al. Cinacalcet in pediatric
and adolescent chronic kidney disease: a single-center experience.
Medicine (Baltimore). 2015;94:e401.
152. Padhi D, Langman CB, Fathallah-Shaykh S, et al. An open-label study to
evaluate a single-dose of cinacalcet in pediatric dialysis subjects. Pediatr
Nephrol. 2012;27:1953 1959.
153. Jadoul M, Albert JM, Akiba T, et al. Incidence and risk factors for hip or
other bone fractures among hemodialysis patients in the Dialysis
Outcomes and Practice Patterns Study. Kidney Int. 2006;70:13581366.
154. Vautour LM, Melton LJ 3rd, Clarke BL, et al. Long-term fracture risk following
renal transplantation: a population-based study. Osteoporos Int. 200 4;15:16 0167.
155. Evenepoel P. Recovery versus persistence of disordered mineral metabolism
in kidney transplant recipients. Semin Nephrol. 2013;33:191203 .
156. Heaf JG. Bone disease after renal transplantation. Transplantation. 2003;75:
315325.
157. Iyer SP, Nikkel LE, Nishiyama KK, et al. Kidney transplantation with early
corticosteroid withdrawal: paradoxical effects at the central and peripheral
skeleton. J Am Soc Nephrol. 2014;25:13311341.
158. Naylor KL, Jamal SA, Zou G, et al. Fracture incidence in adult kidney
transplant recipients. Transplantation. 2016;100:167175.
159. Nikkel LE, Mohan S, Zhang A, et al. Reduced fracture risk with early
corticosteroid withdrawal after kidney transplant. Am J Transplant.
2012;12:649659.
160. Sukumaran Nair S, Lenihan CR, Montez-Rath ME, et al. Temporal
trends in the incidence, treatment and outcomes of hip fracture after
rst kidney transplan tation in the United States. Am J Transplant.
2014;14:943
951.
161. Akaberi S, Simonsen O, Lindergard B, et al. Can DXA predict fractures in
renal transplant patients? Am J Transplant. 2008;8:26472651.
162. Evenepoel P, Cooper K, Holdaas H, et al. A randomized study evaluating
cinacalcet to treat hypercalcemia in renal transplant recipients with
persistent hyperparathyroidism. Am J Transplant. 2014;14:25452555.
163. Bonani M, Frey D, Brockmann J, et al. Effect of twice-yearly denosumab on
prevention of bone mineral density loss in de novo kidney transplant
recipients: a randomized controlled trial. Am J Transplant. 2016;16:
18821891.
164. Tsertsvadze A, Maglione M, Chou R, et al. Updating comparative
effectiveness reviews: current efforts in AHRQs Effective Health Care
Program. J Clin Epidemiol. 2011;64:12081215.
165. Higgins JPT, Green S. Cochrane handbook for systematic reviews of
interventions, version 5.1.0. The Cochrane Collaboration; 2011.
166. Hayden JA, van der Windt DA, Cartwright JL, et al. Assessing bias in studies
of prognostic factors. Ann Intern Med. 2013;158:280286.
167. Guyatt GH, Thorlund K, Oxman AD, et al. GRADE guidelines: 13.
Preparing summary of ndings tables and evidence proles-continuous
outcomes. J Clin Epidemiol. 2013;66:173183.
168. Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. Introduction-
GRADE evidence proles and summary of ndings tables. J Clin
Epidemiol. 2011;64:383394.
169. Guyatt GH, Oxman AD, Santesso N, et al. GRADE guidelines: 12. Preparing
summary of ndings tables-binary outcomes. J Clin Epidemiol. 2013;66:
158172.
170. Guyatt G, Oxman AD, Sultan S, et al. GRADE guidelines: 11. Making an
overall rating of condence in effect estimates for a single outcome and
for all outcomes. J Clin Epidemiol. 2013;66:151157.
171. Uhlig K, Macleod A, Craig J, et al. Grading evidence and recommendations
for clinical practice guidelines in nephrology. A position statement from
Kidney Disease: Improving Global Outcomes (KDIGO).
Kidney Int.
2006;70:20582065.
172. Atkins D, Best D, Briss PA, et al. Grading quality of evidence and strength of
recommendations. BMJ. 2004;328:1490.
173. Guyatt GH, Oxman AD, Kunz R, et al. Going from evidence to
recommendations. BMJ. 2008;336:10491051.
www.kisupplements.org references
Kidney International Supplements (2017) 7, 1–59 59