Sei sulla pagina 1di 27

VOL 55, NO 5, MAY 2010

KDOQI
COMMENTARY

KDOQI US Commentary on the 2009 KDIGO Clinical Practice Guideline


for the Diagnosis, Evaluation, and Treatment of CKDMineral and Bone
Disorder (CKD-MBD)
Katrin Uhlig, MD, MS,1 Jeffrey S. Berns, MD,2 Bryan Kestenbaum, MD, MS,3 Raj Kumar, MBBS,4
Mary B. Leonard, MD,5 Kevin J. Martin, MB, BCh,6 Stuart M. Sprague, DO,7 and
Stanley Goldfarb, MD8

This commentary provides a US perspective on the 2009 KDIGO (Kidney Disease: Improving
Global Outcomes) Clinical Practice Guideline for the Diagnosis, Evaluation, Prevention, and
Treatment of Chronic Kidney DiseaseMineral and Bone Disorder (CKDMBD). KDIGO is an
independent international organization with the primary mission of the promotion, coordination,
collaboration, and integration of initiatives to develop and implement clinical practice guidelines for
the care of patients with kidney disease. The National Kidney Foundations Kidney Disease
Outcomes Quality Initiative (KDOQI), recognizing that international guidelines need to be adapted
for each country, convened a group of experts to comment on the application and implementation of
the KDIGO guideline for patients with CKD in the United States. This commentary puts the KDIGO
guideline into the context of the supporting evidence and the setting of care delivered in the United
States and summarizes important differences between prior KDOQI guidelines and the newer
KDIGO guideline. It also considers the potential impact of a new bundled payment system for
dialysis clinics.
The KDIGO guideline addresses the evaluation and treatment of abnormalities of CKD-MBD in
adults and children with CKD stages 3-5 on long-term dialysis therapy or with a kidney transplant.
Tests considered are those that relate to laboratory, bone, and cardiovascular abnormality
detection and monitoring. Treatments considered are interventions to treat hyperphosphatemia,
hyperparathyroidism, and bone disease in patients with CKD stages 3-5D and 1-5T. Limitations of
the evidence are discussed. The lack of definitive clinical outcome trials explains why most
recommendations are not of level 1 but of level 2 strength, which means weak or discretionary
recommendations. Suggestions for future research highlight priority areas.
Am J Kidney Dis 55:773-799. 2010 by the National Kidney Foundation, Inc.

From 1Tufts Medical Center, Tufts University School of Originally published online as doi:10.1053/j.ajkd.2010.
Medicine, Boston MA; 2University of Pennsylvania School 02.340 on April 5, 2010.
of Medicine, Philadelphia, PA; 3University of Washington Reprint requests to Kerry Willis, PhD, National Kidney
Kidney Research Institute, Seattle, WA; 4Mayo Clinic Col- Foundation, 30 E 33rd St, New York, NY 10016. E-mail:
lege of Medicine, Rochester, MN; 5The Childrens Hospital kerryw@kidney.org
of Philadelphia, University of Pennsylvania School of Medi- Address correspondence to Katrin Uhlig, MD, MS, Tufts
cine, Philadelphia, PA; 6Division of Nephrology, Saint Louis Medical Center, Box 391, 800 Washington St, Boston MA
University, St. Louis, MO; 7North Shore University Health 02445.
System University of Chicago Pritzker School of Medicine, 2010 by the National Kidney Foundation, Inc.
Chicago, IL; and 8University of Pennsylvania School of 0272-6386/10/5505-0002$36.00/0
Medicine, Philadelphia, PA. doi:10.1053/j.ajkd.2010.02.340

American Journal of Kidney Diseases, Vol 55, No 5 (May), 2010: pp 773-799 773
774 Uhlig et al

INDEX WORDS: Clinical practice guideline; guideline adaptation; chronic kidney disease; mineral and
bone disease; renal osteodystrophy; Kidney Disease Outcomes Quality Initiative (KDOQI); Kidney
Disease: Improving Global Outcomes (KDIGO).

K DIGO (Kidney Disease: Improving Global


Outcomes) is an international initiative
with a key mission of developing clinical prac-
Box 1. Definitions of CKD-MBD and of
Renal Osteodystrophy

tice guidelines in the area of chronic kidney Definition of CKD-MBD


disease (CKD). KDIGO recently published an A systemic disorder of mineral and bone metabolism
evidence-based clinical practice guideline for the due to CKD manifested by either one or a
prevention, diagnosis, evaluation, and treatment combination of the following:
Abnormalities of calcium, phosphorus, PTH, or
of metabolic bone disease in individuals with vitamin D metabolism
CKD.1 Because an international guideline needs Abnormalities in bone turnover, mineralization,
to be adapted for the United States, the National volume, linear growth, or strength
Kidney Foundations Kidney Disease Outcomes Vascular or other soft-tissue calcification
Quality Initiative (KDOQI) convened a multidis- Definition of renal osteodystrophy
ciplinary group to comment on the applicability
and implementation of the KDIGO guideline for Renal osteodystrophy is an alteration of bone
morphology in patients with CKD.
patients with CKD in the United States. This It is one measure of the skeletal component of the
commentary presents the KDIGO guideline rec- systemic disorder of CKD-MBD that is quantifiable by
ommendation and statements, provides a suc- histomorphometry of bone biopsy.
cinct discussion and annotation of the supporting Abbreviations: CKD, chronic kidney disease; CKD-MBD,
rationale, and comments on their applicability in chronic kidney diseasemineral and bone disorder; PTH,
the context of practice in the United States. parathyroid hormone.
Adapted from Moe et al3 with permission of Nature
KDIGO was established in 2003 as an indepen-
Publishing Group.
dent nonprofit foundation governed by an interna-
tional board of directors, with its stated mission
to improve the care and outcomes of kidney bone structure and function is a common finding
disease patients worldwide through promoting in patients with CKD requiring dialysis (stage
coordination, collaboration, and integration of 5D) and many patients with CKD stages 3-5. In
initiatives.2 Mineral abnormalities and renal os- addition, extraskeletal calcification is a feature,
teodystrophy in CKD and, more recently, linkage at least in part, of deranged mineral and bone
of these with extraosseous calcification have metabolism of CKD and may even be exacer-
been areas of intense interest and controversy. In bated by some of the therapies used to correct
2005, KDIGO sponsored a controversies confer- mineral and bone changes in CKD. Interactions
ence, Definition, Evaluation and Classification among abnormal mineral metabolism, abnormal
of Renal Osteodystrophy. The resulting 2006 bone, and extraskeletal calcification may contrib-
KDIGO position statement proposed a definition ute to the morbidity and mortality of patients
for CKDmineral and bone disorder (CKD- with CKD. Hence, this guideline is needed to
MBD) and for renal osteodystrophy, shown in help define best practices based on current dis-
Box 1.3 ease concepts and best available research evi-
Both initial bone formation during growth dence.
(bone modeling) and changes in bone structure
and function during adulthood (bone remodel- KDIGO GUIDELINE PROCESS
ing) are severely disrupted in patients with CKD. A KDIGO Work Group of international ex-
This results from disturbances in mineral metab- perts and an Evidence Review Team defined
olism, and a number of abnormalities in levels of pertinent questions related to the clinical manage-
hormones and cytokines that regulate blood lev- ment of CKD-MBD and developed study inclu-
els of calcium, phosphorus, and various other sion criteria. Target populations for the KDIGO
ionic species, as well as bone, directly. Abnormal guideline and this commentary are adults and
KDOQI Commentary 775

Table 1. Strength of a Recommendation

Implications for

Grade for Strength Patients Clinicians Policy Makers

Level 1 Most people in your situation Most patients should receive The recommendation can be
Strong would want the the recommended course evaluated as a candidate
We recommend . . . should recommended course of of action for developing a policy or
action and only a small a performance measure
proportion would not

Level 2 Most people in your situation Different choices will be The recommendation is
Weak or discretionary would want the appropriate for different likely to require substantial
We suggest . . . might recommended course of patients. Each patient debate and involvement of
action, but many would needs help to arrive at a stakeholders before policy
not management decision can be determined
consistent with her or his
values and preferences
Note: In addition to graded recommendations, the KDIGO Work Group could also issue statements that were not graded
(see text).
Abbreviation: KDIGO, Kidney Disease: Improving Global Outcomes.
Based in part on Guyatt et al.6

children with CKD stages 3-5, those on long- Grading of Recommendations Assessment, De-
term dialysis therapy, and kidney transplant re- velopment and Evaluation (GRADE) ap-
cipients. The target audience is practitioners car- proach.4,5 The strength of each recommenda-
ing for these patients. tion is rated as level 1, which means strong, or
For treatment questions, outcomes of interest level 2, which means weak or discretionary.
were grouped into 3 categories: outcomes with Wording corresponding to a level 1 recommen-
direct importance to patients (eg, mortality, car- dation is We recommend . . . should and
diovascular disease events, hospitalizations, frac- implies that most patients should receive the
ture, and quality of life), intermediate outcomes course of action. Wording for a level 2 recom-
(eg, vascular calcification, bone mineral density mendation is We suggest . . . might, which
[BMD], and bone biopsy), and biochemical out- implies that different choices will be appropri-
comes (eg, serum calcium, phosphorus, alkaline ate for different patients, with the suggested
phosphatase, and parathyroid hormone [PTH] course of action being a reasonable choice in
levels). Clinical outcomes were considered to be many patients. Usually, level 1 but not level 2
of critical or high importance, whereas intermedi- recommendations are candidates for develop-
ate and laboratory outcomes were considered to ing a performance measure.6 In addition, each
be of moderate importance. Thus, the work group statement is assigned a grade for the quality of
acknowledged that these intermediate and bio- the supporting evidence: A (high), B (moder-
chemical outcomes currently are not sufficiently ate), C (low), or D (very low). Furthermore,
validated surrogates for hard clinical events. for topics that cannot be subjected to system-
The KDIGO Work Group agreed a priori to atic evidence review, the work group could
include only randomized controlled trials (RCTs) issue statements that are not graded. Typically,
of 6 months duration with a sample size of at these provide guidance based on common
least 50 patients in systematic reviews. Excep- sense; for example, reminders of the obvious
tions were made for studies with bone biopsy or recommendations that are not specific
outcomes (minimum sample size, 20 per study). enough to allow direct application of evidence.
Studies of smaller sample size involving children Table 1 lists implications of the guideline
were tabulated in overview tables. grades and describes how the strength of the
The grading approach followed in the recommendations should be interpreted by
KDIGO bone guideline is adopted from the guideline users. With the evolution in grading,
776 Uhlig et al

grades between KDOQI and KDIGO recom- nal sequential number assigned to each state-
mendations are not directly comparable. ment. After each set of recommendations, we
provide a commentary regarding their rationale
KDOQI PROCESS FOR ADAPTATION OF THE and a statement regarding their applicability to
KDIGO BONE GUIDELINE the United States. Table 2 shows a summary of
KDIGO recommendations on evaluation for
Certain organizational, legislative, and cul- CKD-MBD in patients with CKD stages 3-5D,
tural issues may affect the applicability of evi- and Table 3 shows this for patients with CKD
dence to a specific context.7 Variability in values stages 1-5T. Because the KDIGO guideline builds
and judgments also may legitimately impact on on more recent evidence, its recommendations
interpretation of evidence and its translation into should replace those published previously by the
recommendations.6 This highlights the need for a KDOQI.8 Table 4 juxtaposes pertinent KDIGO
process of adapting an existing guideline, in this and KDOQI recommendations. Our commentary
case, the global KDIGO guideline, to the US provides additional information guiding clinical
setting. A large amount of evidence reviewed for practice in the United States and should be used
the KDIGO guidelines was generated in the in conjunction with careful reading of the KDIGO
United States or settings similar to the United clinical practice guideline.
States with regard to the epidemiologic character-
istics of CKD-MBD, resources, and health care
delivery systems. In addition, 6 members of the CONSIDERATIONS FOR IMPLEMENTATION:
KDIGO guidelines work group were US based. POTENTIAL IMPACT OF A NEW BUNDLED
Therefore, it can be expected that many recom- PAYMENT SYSTEM FOR DIALYSIS CLINICS
mendations generally are applicable to the United Implementation of the guideline recommenda-
States. However, this commentary provides an tions in outpatient dialysis patients is likely to be
opportunity for additional summary and reflec- affected greatly by the introduction of new pay-
tion regarding their appropriateness for imple- ment policies created through the Medicare Im-
mentation in the US health care system. KDOQI provements for Patients and Providers Act of
convened a multidisciplinary group to comment 2008 (MIPPA). Beginning in 2011, the Centers
on the application of the KDIGO CKD-MBD for Medicare & Medicaid Services (CMS) plans
clinical practice guidelines in the United States. to implement an updated prospective payment
After the authors approved a commentary draft, system including an expanded bundle. In a draft
the KDOQI Chair and Vice Chairs for Guidelines regulation published in the Federal Register on
and Commentary, Research, Education, and Pub- September 29, 2009, the CMS proposed that the
lic Policy, as well as the National Kidney Foun- expanded bundle include all drugs and biologics
dations Scientific Advisory Board, reviewed the formerly reimbursed under either Medicare Part
commentary and their recommendations were B or Part D that are used to treat patients with
incorporated into the final article. end-stage renal disease (ESRD), regardless of
Explicit cost considerations of the recommen- the administration route.10,11 The health care
dations warrant detailed analysis and are beyond reform legislation passed by the US House of
the scope of this report; however, the potential Representatives (HR 3962) includes a similar
impact of a newly proposed bundled payment requirement. The proposal would make the dialy-
system for dialysis clinics is considered in the
sis unit responsible for provision of the follow-
first section of this commentary. In the following
ing items under the bundle:
section the original KDIGO guideline recommen-
dations for CKD-MBD are presented with the Services included in the composite rate
strength that originally was assigned to them. as of 2010
KDIGO guideline chapters 3 and 4 relate to the Injectable biologics used to treat anemia;
evaluation and treatment of CKD-MBD in pa- erythropoiesis-stimulating agents and any
tients with CKD stages 3-5 and 5D, and chapter oral form of such agents
5, to CKD stages 1-5T. For ease of referencing Other injectable medications that are
with the KDIGO guideline, we retained the origi- furnished to ESRD beneficiaries and paid
KDOQI Commentary
Table 2. KDIGO Recommendations on Evaluation for CKD-MBD in CKD Stages 3-5D

Biochemical Componentsa Bone Blood Vessels

CKD Stage (GFR Bone-specific


[mL/min/1.73 m2]) Ca, P PTH ALP 25(OH)D ALP Bone Biopsy BMD Calcification Testing

Stage 3 (30-59) Once (1C)b; then Once (1C)b; then Once (1C)b
every 6-12 mo based on level
(NG) and CKD Once (2C); then In various settingsc
Can be used to No routine testing
progression based on and before No recommendation
evaluate in the presence
(NG) level and treatment with for routine
bone of CKD-MBD
Stage 4 (15-29) Every 3-6 mo (NG) Every 6-12 mo treatments bisphosphonates screening
disease (2B) (2B)
(NG) Every 12 mod (2C) (NG)
Stage 5 (15 or Every 1-3 mo (NG) Every 3-6 mo (NG)
dialysis) (NG)
Note: Number and letters in parentheses refer to strength of recommendation and quality of evidence (see Table 1 for grades).
Abbreviations and definitions: 25(OH)D, 25-hydroxyvitamin D (calcidiol); ALP, alkaline phosphatase; BMD, bone mineral density; Ca, calcium; CKD, chronic kidney disease;
CKD-MBD, chronic kidney disease-mineral and bone disorder; D, dialysis (when referring to CKD stage); GFR, glomerular filtration rate; KDIGO, Kidney Disease: Improving
Global Outcomes; NG, statement not graded; P, phosphorus; PTH, parathyroid hormone.
a
Base the frequency of laboratory measurements on the presence and magnitude of abnormalities and rate of CKD progression. Increase frequency intervals as needed to
monitor for trends, treatment efficacy, and side effects (NG).
b
In children, monitoring of Ca, P, and ALP levels is suggested beginning in CKD stage 2 (2D).
c
Various settings include unexplained fractures, persistent bone pain, unexplained hypercalcemia, unexplained hypophosphatemia, and possible aluminum toxicity.
d
More frequently in presence of increased PTH levels.

777
778
Table 3. KDIGO Recommendations on Evaluation for CKD-MBD in CKD Stages 1-5T

Biochemical Componentsa Bone


CKD Stage
(GFR [mL/min/1.73 m2]) Ca, P PTH ALP 25(OH)D Bone Biopsy BMD

Immediate posttransplant At least every wk


until stable (1B)

Stage 1T (90) Once and then based Consider to guide In first 3 mo post-
Every 6-12 mo
Stage 2T (60-89) on level and CKD treatment, transplant if
(NG)
Stage 3T (30-59) progression (NG) specifically before patient receives
Once and then based
treatment with corticosteroids or
Every 12 mo
b on level and
bisphosphonates has risk factors for
(NG) treatments (2C)
(NG) osteoporosis (2D)
Stage 4T (15-29) Every 3-6 mo (NG) Every 6-12 mo (NG) No routine testing
Stage 5T (15) Every 1-3 mo (NG) Every 3-6 mo (NG) (2B)
Note: Number and letters in parentheses refer to strength of recommendation and quality of evidence (see Table 1 for grades).
Abbreviations and definitions: 25(OH)D, 25-hydroxyvitamin D (calcidiol); ALP, alkaline phosphatase; BMD, bone mineral density; Ca, calcium; CKD, chronic kidney disease;
CKD-MBD, chronic kidney diseasemineral and bone disorder; GFR, glomerular filtration rate; KDIGO, Kidney Disease: Improving Global Outcomes; NG, statement not
graded; P, phosphorus; PTH, parathyroid hormone; T, transplant (when referring to CKD stage).
a
Base the frequency of laboratory measurements on the presence and magnitude of abnormalities and rate of CKD progression. Increase frequency intervals as needed to
monitor for trends and treatment efficacy and side effects (NG).
b
More frequently in presence of increased PTH levels.

Uhlig et al
KDOQI Commentary 779

Table 4. Comparison of Key KDIGO 2009 and KDOQI 2003 Recommendations on Testing and Treatment Targets

KDIGO 2009 Grade KDOQI 2003 Grade Comment

CKD Stages 3-5 and 5D


In adults, recommend monitoring serum calcium, 1C Same, except no recommendation for E Suggestion for using ALP as
phosphorus, PTH, and ALP levels beginning ALP [KDOQI guideline 1.1] adjunct test
in CKD stage 3 [KDIGO recommendation
3.1.1]

In children, suggest monitoring serum calcium, 2D Same, but also recommendation for O
phosphorus, PTH, and ALP levels beginning total CO2 [1.1P]
in CKD stage 2 [3.1.1]

In CKD stages 3-5D, suggest measuring 2C In CKD stages 3-4, measure E Suggestion for expanded testing
25(OH)D levels [3.1.3] 25(OH)D, if PTH is above target of 25(OH)D
range for stage of CKD [7.1]

Suggest that vitamin D deficiency and 2C If serum 25(OH)D level 30 ng/mL O Suggestion for expanded
insufficiency be corrected using treatment (75 nmol/L), supplementation treatment with vitamin
strategies recommended for the general with vitamin D2 (ergocalciferol) D2 or D3 given suggestions for
population [3.1.3] should be initiated [7.2] expanded testing

Suggest using individual serum calcium and 2D Ca P 55 mg2/dL2 recommended E Suggestion against product as
phosphorus values, rather than the in CKD stages 3-5 [6.5] mostly driven by phosphorus
mathematical construct of Ca P [3.1.5] and not more informative than
both components individually

Recommend clinical laboratories report assay 1B Evidence and recommendations for Reflects evolution in PTH assays
methods used and any change in methods, adults based mostly on PTH
sample source, and handling specifications. measured using
For appropriate interpretation of biochemistry second-generation Allegro
data in CKD stages 3-5D, clinicians need to assay from Nichols, which is not
understand assay characteristics and currently available [background
limitations [paraphrased from 3.1.6] to guideline 1]
Rationale recommends use of Evidence and recommendation in
second-generation assay for PTH [3.1.6] children mostly based on use of
first-generation immunometric
PTH assay [1.1P]

In patients with CKD stages 3-5D, suggest that 2B Bone-specific ALP not specifically Suggestion for testing
measurements of serum PTH or bone-specific recommended bone-specific ALP in certain
ALP can be used to evaluate bone disease Unexplained increases in bone ALP O individuals
because markedly high or low values predict activity together with increases
underlying bone turnover [3.2.3] in PTH is indication for
considering bone biopsy in CKD
stage 5D [2.2.b]

In CKD stages 3-5D, reasonable to perform bone NG Bone biopsy should be considered in O Expanded indication for bone
biopsy in various settings and before therapy CKD stage 5D in various biopsy including before
with bisphosphonates in patients with CKD- settings [2.2] treatment with
MBD [3.2.1] bisphosphonates

In CKD stages 3-5D with biochemical 2B DXA should be used in patients with O Suggestion for restricted testing
abnormalities of CKD-MBD, suggest that BMD fractures and those with known of BMD
testing not be routinely performed [3.2.2] risks for osteoporosis [2.4]

No recommendation given for routine screening Same


for vascular calcification [rationale for 3.3]

(Continued)
780 Uhlig et al

Table 4 (Contd). Comparison of Key KDIGO 2009 and KDOQI 2003 Recommendations on Testing and Treatment Targets

KDIGO Grade KDOQI Grade Comment

In CKD stages 3-5, suggest maintaining serum 2C In CKD stages 3-4, maintain serum Suggestion for same target range
phosphorus in the reference range [4.1.1] phosphorus at [3.1]: in CKD stages 3-4, lower in
2.7 mg/dL E CKD stage 5
4.6 mg/dL O

In CKD stage 5D, suggest lowering increased 2C In CKD stages 5 and 5D, maintain E No prescriptive phosphorus
phosphorus levels toward the reference range serum phosphorus at 3.5-5.5 target range. Suggestion to
[4.1.1] mg/dL [3.2] lower toward reference permits
greater flexibility based on
assessment of risk-benefit and
patient preferences

In CKD stages 3-5D, suggest maintaining serum 2D In CKD stages 3-4, same for E Suggestion not specific for
calcium in reference range [4.1.2] corrected total calcium [6.1] corrected total calcium
In CKD stages 5 and 5D, same for O
corrected total calcium, but
preference for aiming toward
lower end of reference range
(8.4-9.5 mg/dL) [6.2]

In CKD stages 3-5 not on dialysis therapy, 2C PTH target ranges: [1.4] No prescriptive PTH target range.
suggest evaluating patients with PTH levels CKD stage 3, 35-70 pg/mL O Suggestion for lower PTH
above the upper reference limit of the assay CKD stage 4, 70-110 pg/mL O threshold in CKD stages 4-5 to
for hyperphosphatemia, hypocalcemia, and CKD stage 5, 150-300 pg/mL E prompt evaluation, correction
vitamin D deficiency [4.2.1] of modifiable factors and
possibly treatment

In CKD stage 5D, suggest maintaining PTH level 2C In CKD stage 5D, PTH target range is E No prescriptive PTH target range.
in the range of 2-9 times the upper reference 150-300 pg/mL [1.4] Suggestion for wider target
limit for the assay [4.2.3] range, corresponding to PTH
Suggest that marked changes in PTH within this 2C of 130-600 pg/mL
range prompt initiation or change in therapy
[4.2.3]

CKD Stages 1-5T


In patients in the immediate postkidney 1B During the first week after kidney O
transplant period, recommend measuring transplant, serum phosphorus
serum calcium and phosphorus at least level should be measured daily
weekly until stable [5.1] [16.2]

In patients after the immediate postkidney NG Also recommendation for monitoring O Suggestion for using ALP as an
transplant period, monitor serum calcium, total CO2 [16.1] adjunct test
phosphorus, PTH, and ALP levels. Reasonable No recommendation for ALP
to base frequencies on the presence and
magnitude of abnormalities and rate of CKD
progression [paraphrased from 5.2]

Suggest that 25(OH)D levels might be measured 2C No recommendation Suggestion for expanded testing
and repeated testing determined by baseline of 25(OH)D
values and interventions [5.3]

Reasonable to manage abnormalities of CKD- NG Similar [16.5] O


MBD as for patients with CKD stages 3-5 [5.2]

Suggest that vitamin D deficiency and 2C No recommendation Suggestion for expanded


insufficiency be corrected [5.4] treatment with vitamin D2 or D3
(Continued)
KDOQI Commentary 781

Table 4 (Contd). Comparison of Key KDIGO 2009 and KDOQI 2003 Recommendations on Testing and Treatment Targets

KDIGO Grade KDOQI Grade Comment

In patients with eGFR 30 mL/min/1.73 m2 and 2D Kidney transplant recipients should O Suggestion for more restricted
treatment with corticosteroids or risk factors have BMD measured using DXA testing of BMD
for osteoporosis, suggest measuring BMD in [16.4]
the first 3 mo after kidney transplant [5.5]
In CKD stages 4-5T, suggest that BMD testing 2B
not be routinely performed [5.7]

In patients in first 12 mo after kidney transplant 2D If BMD T score is 2 or less at or after O Expanded indication for bone
with eGFR 30 mL/min/1.73 m2 and low transplant, consider treatment biopsy including before
BMD, suggest consideration of treatment with with parenteral bisphosphonates treatment with
vitamin D, calcitriol/alfacalcidiol, or [16.4b] bisphosphonates
bisphosphonates
Consider a bone biopsy, specifically before use NG No recommendation
of bisphosphonates [5.6]

Note: Recommendations may be paraphrased, abbreviated, or summarized from KDIGO CKD-MBD,1 KDOQI bone,8 and
KDOQI nutrition in children9 guidelines. For verbatim KDIGO recommendations, see text.
Abbreviations and definitions: 25(OH)D, 25-hydroxyvitamin D (calcidiol); ALP, alkaline phosphatase; BMD, bone mineral
density; Ca P, calcium-phosphate product; CKD, chronic kidney disease; CKD-MBD, chronic kidney diseasemineral and
bone disorder; D, dialysis (when referring to CKD stage); DXA, dual-energy x-ray absorptiometry; E, graded as evidence in
KDOQI guideline; eGFR, estimated glomerular filtration rate; KDIGO, Kidney Disease: Improving Global Outcomes; KDOQI,
Kidney Disease Outcomes Quality Initiative; NG, statement not graded; O, graded as opinion in KDOQI guideline; P,
pediatric (ie, guideline from KDOQI nutrition in children guideline51); PTH, parathyroid hormone.

for separately under Part B and any oral responsible for ensuring patients access to the
equivalent to such medications prescribed medications. Many clinics are not set
Laboratory tests and other items and up to dispense these medications that patients
services furnished to beneficiaries for the now receive through a pharmacy. Also, there is
treatment of ESRD real concern that clinics could discourage physi-
Currently, this definition has been interpreted cians from prescribing certain brands of medica-
broadly by the CMS to include all medications tions that are more costly, particularly if the
given in the dialysis unit, as well as any labora- CMS do not adequately reimburse for the cost of
tory test prescribed by a physician providing these drugs in the bundle. Additionally, it is
dialysis regardless of the site of service and even unclear how the cost for new drugs would be
if not entirely related to ESRD care. factored.
This proposal aims to reduce total Medicare With regard to the KDIGO CKD-MBD guide-
payments for dialysis services by 2% both during line, the additional medications that dialysis clin-
the 4-year phase-in period and after the bundled ics would be paid for as part of the proposed
payment system is fully implemented. Facilities bundled payment include phosphate binders, oral
will have the opportunity to opt out of the phase-in or intravenous vitamin D analogues, and calcimi-
and be paid under the new bundled system start- metics. Additionally, laboratory tests including,
ing in 2011. for example, vitamin D or bone-specific alkaline
Therefore, access to medications by patients phosphatase levels, would be included in the
receiving hemodialysis treatments in outpatient expanded bundle if ordered by a dialysis pro-
centers may change. Because Medicare Part D vider or sent from the dialysis unit. It is expected
most likely will no longer cover medications that the CMS will reimburse $14 per treatment
now considered by the CMS to be a part of renal on average above and beyond the current compos-
dialysis services, patients may no longer have ite rate for the new items going into the bundle
access to the same medications they do now. that were previously covered under Part D. With
Dialysis clinics would receive payment from fixed bundle reimbursement, providers will have
Medicare for dialysis-related medications and be to trade off costs for different treatments, for
782 Uhlig et al

example, treatment with erythropoietin in one riority over total calcium alone.18 It considered
patient versus treatment with a calcimimetic in measurement of ionized calcium to be more
another, or treatment with calcium-based versus specific, but presently not practical or cost-
noncalcium-based phosphorus binders. Given effective. These recommendations are applicable
the overall low quality of evidence for clinical to the United States.
benefit from treatments for CKD-MBD and the 3.1.2 In patients with CKD stages 3-5D, it is reasonable
corresponding weak guideline recommenda- to base the frequency of monitoring serum cal-
tions, it is likely that the cost of a drug will cium, phosphorus, and PTH on the presence and
directly impact on decision making and access to magnitude of abnormalities and rate of progression
more expensive drugs will be restricted. More of CKD (not graded).
Reasonable monitoring intervals would be:
global inclusion of laboratory tests also has the In CKD stage 3: For serum calcium and phospho-
potential to impact on the frequency of testing, as rus, every 6-12 months; and for PTH, based on
well as selection of tests related to MBD care. baseline level and CKD progression
Currently, the US nephrology community is In CKD stage 4: For serum calcium and phospho-
rus, every 3-6 months; and for PTH, every 6-12
responding to the proposed rule (eg, see editori-
months
als12-16 in the February 2010 issue of the Ameri- In CKD stage 5, including 5D: For serum calcium
can Journal of Kidney Diseases). A final rule by and phosphorus, every 1-3 months; and for PTH,
the CMS is expected later in 2010. How practice every 3-6 months
patterns and outcomes related to CKD-MBD In CKD stages 4-5D: For alkaline phosphatase
care may be influenced by the proposed changes activity, every 12 months or more frequently in the
presence of increased PTH levels (see chapter 3.2)
in the payment system in the United States will
require careful scrutiny by the CMS, providers, These statements provide guidance for testing
and researchers. intervals. The frequency of repeated testing needs
to take into account whether and what abnormali-
COMMENTARY ON KDIGO BONE GUIDELINE ties were identified, their severity and duration,
and level of kidney function and rate of kidney
Recommendations in Chapter 3.1: Diagnosis of disease progression. The KDIGO Work Group
CKD-MBD: Biochemical Abnormalities recommends using total alkaline phosphatase
3.1.1 We recommend monitoring serum levels of cal- activity as an adjunct test. It may provide supple-
cium, phosphorus, PTH, and alkaline phosphatase mental information in the assessment of bone
beginning in CKD stage 3 (1C). In children, we turnover, particularly in the setting of increased
suggest such monitoring beginning in CKD stage 2 PTH levels and in the assessment of response to
(2D).
therapy for increased PTH levels if liver disease
In adults, changes in levels of biochemical is not likely to be the cause of increased total
markers of CKD-MBD may begin in CKD stage alkaline phosphatase levels.19,20 Bone-specific
3; however, the rate of change and severity of alkaline phosphatase derives more specifically
abnormalities are highly variable among pa- from bone and can be used when the clinical
tients. The strong recommendation indicates that situation is more ambiguous; however, the test is
assessment of CKD-MBD should begin in stage not readily available. High total alkaline phospha-
3. In children, PTH level increases occur as early tase levels have been associated with higher
as CKD stage 2,17 and the Work Group thus mortality,21-23 but it is not known whether thera-
made a weak recommendation suggesting assess- pies aimed at decreasing these levels improve
ment of calcium, phosphorus, PTH, and alkaline patient outcomes. The KDOQI bone guideline
phosphatase in children starting in CKD stage 2. also discussed that concomitant consideration of
The rationale for this recommendation ad- alkaline phosphatase levels can increase the pre-
dresses the issue of whether calcium should be dictive power of PTH levels,24,25 although data
measured as total calcium, ionized calcium, or were insufficient to determine the sensitivity and
total calcium corrected for measured albumin. specificity of alkaline phosphatase levels for re-
The work group did not recommend that cor- nal osteodystrophy alone or together with PTH
rected calcium measurement be abandoned at levels.8 The value of alkaline phosphatase for
present, although recent data did not show supe- clinical decision making remains to be proved,
KDOQI Commentary 783

especially in the present era with different PTH and insufficiency. This represents a change
assays and a lower prevalence of osteomalacia. from the KDOQI guidelines, in which the
These statements are applicable to the United recommendation to measure 25(OH)D was lim-
States. The practitioner can individualize as ited to patients with CKD stages 3-4 and
needed. elevated PTH level.
3.1.2 In patients with CKD receiving treatments for These recommendations are applicable to the
(continued) CKD-MBD or in whom biochemical abnor- United States. Ultimately, the practitioner in the
malities are identified, it is reasonable to United States needs to individualize the decision
increase the frequency of measurements to
monitor for trends and treatment efficacy and
for whether, when, and how often to measure
side effects (not graded). vitamin D and below what threshold and to what
target range to treat. A reasonable approach is to
There are no data to directly support a specific periodically measure 25-hydroxyvitamin D in
testing frequency. This statement provides the patients with CKD and initiate treatment if the
necessary flexibility for more frequent measure- level is low. Recommendations for vitamin D
ment when levels are changing rapidly and to repletion in the general population specify a
monitor the effects of treatments, including poten-
cholecalciferol dose of 1,000-2,000 IU/d be-
tial adverse effects. This statement is applicable
cause lower doses minimally impact on 25-
to the United States.
hydroxyvitamin D level.38-40 However, a more
3.1.3 In patients with CKD stages 3-5D, we suggest that aggressive dosing regimen may be used in pa-
calcidiol (25[OH]D) might be measured, with
repeated testing determined by baseline values and
tients with CKD. The KDOQI bone guideline
therapeutic interventions (2C). We suggest that provided a recommendation for supplementation
vitamin D deficiency and insufficiency be cor- in patients with CKD stages 3-4 with ergocalcif-
rected using treatment strategies recommended for erol, 50,000 IU, orally dosed weekly or monthly
the general population (2C).
based on serum 25-hydroxyvitamin D level.8
The serum vitamin D level that represents This has not been tested in patients with CKD
sufficiency is the subject of an ongoing debate stage 5, 5D, or T. Monitoring calcium, phospho-
and is complicated by variability in measure- rus, vitamin D, and PTH levels can guide subse-
ments of vitamin D compounds.26-28 Substrate quent dose adjustments.
vitamin D deficiency has been variably defined 3.1.4 In patients with CKD stages 3-5D, we recommend
as 25(OH)D level 10-20 ng/mL, and insuffi- that therapeutic decisions be based on trends,
ciency has been variably defined as 25(OH)D rather than a single laboratory value, taking into
level higher than the limits for deficiency but account all available CKD-MBD assessments (1C).
32-35 ng/mL.29,30 There are no data about
This statement recommends assessing all pa-
whether vitamin D levels should vary between
rameters of CKD-MBD together and each param-
those with or without CKD. In patients with
CKD, vitamin D deficiency or insufficiency may eter over time, rather than one value in isolation.
be a cause of increased PTH levels. Observa- Thus, the practitioner needs to review patterns
tional studies show an association between and temporal trends to make clinical decisions.
low vitamin D levels and adverse clinical out- Furthermore, the practitioner needs to know what
comes,24,30-33 and link treatment with vitamin D or assays are used. PTH and phosphorus levels are
its analogues to improvements in surrogate or clini- subject to diurnal variation and vitamin D levels
cal outcomes.23,34-37 However, data from clinical are subject to seasonal variation. Testing for PTH
trials with vitamin D for important clinical out- and phosphorus should be performed at similar
comes are lacking at this time. The potential times during the day and week. To eliminate
risks of vitamin D repletion are minimal; there- between-assay variability, the same assay should
fore, although the benefits of this have not been be used for monitoring changes over time. This
proved, the work group believed that measure- recommendation is applicable to the United
ment and treatment of deficiency might be ben- States. This recommendation has significant im-
eficial, making weak recommendations for plications for dialysis provider performance mea-
measuring vitamin D and correcting deficiency sures that typically focus on laboratory values at
784 Uhlig et al

a single point in time and do not consider trends better test performance. To assist interpretation
over time. of values obtained using different PTH assays,
3.1.5 In patients with CKD stages 3-5D, we suggest that the reader is referred to the article by Souber-
individual values of serum calcium and phospho- bielle et al,46 which suggests conversion factors
rus, evaluated together, be used to guide clinical for many currently used PTH assays using the
practices rather than the mathematical construct of Allegro iPTH assay as a reference. Although this
calcium-phosphorus product (2D).
provides a practical tool to reduce some of the
Despite epidemiologic data linking increased interassay discrepancies in the absence of a true
calcium-phosphorus product with poorer patient reference standard, it has not been externally
outcomes,41,42 the work group suggests using the validated and does not overcome the problem of
individual components rather than the mathemati- variable assay reactivity with PTH fragments.
cal construct of calcium-phosphorus product, For measurement of 25-hydroxyvitamin D,
which is driven largely by serum phosphorus. immunoassays have reasonably good precision
The product generally does not provide addi- and many clinical laboratories now routinely
tional clinical information beyond that provided measure 25-hydroxyvitamin D using liquid chro-
by its components.43-45 Thus, the KDIGO Work matography-mass spectroscopy, which has excel-
Group advised against relying on the product in lent precision.26-28,50 This recommendation is
clinical decision making. This recommendation applicable to the United States. Readers are re-
is applicable to the United States. ferred to Section 3.1.6 of the KDIGO guidelines
3.1.6 In reports of laboratory tests for patients with CKD for discussion of issues related to measurement
stages 3-5D, we recommend that clinical laborato- of calcium, phosphorus, PTH, vitamin D, and
ries inform clinicians of the actual assay method in alkaline phosphatase.
use and report any change in methods, sample
source (plasma or serum), and handling specifica-
tions to facilitate appropriate interpretation of Recommendations in Chapter 3.2: Diagnosis of
biochemistry data (1B). CKD-MBD: Bone
This recommendation is addressed to clinical 3.2.1 In patients with CKD stages 3-5D, it is reasonable
pathologists performing the measurement of the to perform a bone biopsy in various settings,
laboratory components of CKD-MBD. Given the including, but not limited to, unexplained frac-
variability within and across assays, especially tures, persistent bone pain, unexplained hypercal-
cemia, unexplained hypophosphatemia, possible
for PTH and vitamin D compounds, clinical aluminum toxicity, and before therapy with
laboratories should assist clinicians in interpreta- bisphosphonates in patients with CKD-MBD (not
tion of test results by reporting assay characteris- graded).
tics and methods used. Clinicians need to stan-
dardize within their outpatient clinical practices Renal osteodystrophy is a complex disorder.
and dialysis units the methods of sample collec- Biochemical laboratory and imaging tests do not
tion, processing, and assays used. adequately predict the underlying bone histol-
Analytic problems with PTH measurement ogy. Thus, although bone biopsy is invasive and
include: (1) poor standardization among differ- cannot be performed easily in all patients, it is
ent PTH assays, (2) high biological variation the gold standard for the diagnosis of renal
within individuals, and (3) uncertainty about the osteodystrophy. Bone biopsy should be consid-
role of unmeasured PTH fragments.27,46 The ered in patients for whom the cause of clinical
widely used second-generation intact PTH symptoms and biochemical abnormalities is not
(iPTH) assays measure not only full-length ac- certain and for whom the effect of treatment on
tive PTH, but also different types and amounts of bone needs to be assessed. As detailed in the
circulating fragments.47-49 Despite these limita- KDOQI bone and mineral guidelines, aluminum
tions, the work group favored the continued use bone disease also requires bone biopsy for diag-
of second-generation iPTH assays in clinical nosis. However, this is less common in the cur-
practice rather than the more recently introduced rent era. The KDIGO position statement sug-
bioactive or biointact third-generation PTH gested that tissue from bone biopsies in patients
assays, which have not yet been shown to have with CKD should be characterized by determin-
KDOQI Commentary 785

ing bone turnover, mineralization, and volume tion, and BMD does not predict the type of renal
(TMV).3 osteodystrophy (2B).
The statement that bone biopsy is reasonable In the general population, low BMD measured
before initiating treatment with bisphosphonates using dual-energy x-ray absorptiometry (DXA)
applies to only those with evidence of CKD- predicts fracture and mortality; however, evi-
MBD. Although there is a large number of el- dence for its ability to predict fractures or other
derly with CKD stage 3 and low BMD in the clinical outcomes in patients with CKD stages
United States, this statement applies to only 4-5 is limited.51 In patients with CKD stage 5D,
those who also have CKD-MBD, which in prac- evidence linking low BMD and fracture risk is
tical terms means increased PTH or phosphate weak, inconsistent, and varies by site.52 Volumet-
level. Bone biopsy is the most accurate test for ric BMD, measured predominantly using quanti-
the diagnosis of adynamic bone disease, and the
tative computed tomography (CT), correlated
presence of adynamic bone disease is a contrain-
with fractures in 1 small study of hemodialysis
dication to bisphosphonate treatment.
patients.53 Also, determination of BMD, whether
The preferred site for a bone biopsy is the iliac
measured using DXA or quantitative CT, does
crest, and the biopsy is undertaken with a trocar
not distinguish among types of renal osteodystro-
or a drill. Assessment of turnover requires double
phy. Patients with CKD-MBD and osteoporosis
labeling with tetracycline according to a proto-
cannot be assumed to benefit from therapies such
col, and reading of the biopsy specimen requires
as bisphosphonates. Thus, the work group issued
particular expertise and resource. The procedu-
this suggestion against routine BMD testing in
ral risk is low; however, discomfort and pain
at the biopsy site are common. Although biopsy is individuals with laboratory evidence of CKD-
the gold standard test for renal osteodys- MBD, ie, individuals with CKD and increased
trophy, the natural history of renal bone disease phosphorus or PTH levels. This recommendation
shows great variability and different types of is applicable to the United States.
osteodystrophy have at best only modest relation- 3.2.3 In patients with CKD stages 3-5D, we suggest that
ships with clinical outcomes. Future studies need serum PTH or bone-specific alkaline phosphatase
to evaluate the merit of the TMV classification measurement can be used to evaluate bone disease
because markedly high or low values predict
for informing clinical treatment decisions and underlying bone turnover (2B).
ultimately improving bone and other clinical
outcomes. Circulating PTH or bone-specific alkaline
In the United States, bone biopsies currently phosphatase levels correlate with some histomor-
are not widely undertaken for the evaluation of phometric measurements in bone biopsy speci-
renal osteodystrophy. In some centers, this ser- mens. However, the positive predictive value for
vice is provided by a specialist in bone health, both tests is only modest for detection of high
such as an endocrinologist or nephrologist. Wide and low bone turnover states, especially for de-
implementation of this statement would require a tecting adynamic bone.19,20,54-56 Nevertheless,
greater pool of individuals with proficiency in because bone biopsy is not feasible in most
the performance and interpretation of bone biop- patients, the work group issued a weak recom-
sies. In addition to addressing the barriers to mendation suggesting measurement of these se-
getting a bone biopsy, future research is needed rum markers because they may be useful to
to evaluate the usefulness of bone biopsy results estimate bone turnover, especially when values
for the selection of therapies. Regarding the use are very abnormal. This recommendation is appli-
of bisphosphonates in patients with CKD stage 3 cable to the United States.
with CKD-MBD, see the commentary on recom-
3.2.4 In patients with CKD stages 3-5D, we suggest not
mendation 4.3.3. to routinely measure bone-derived turnover mark-
3.2.2 In patients with CKD stages 3-5D with evidence of ers of collagen synthesis (such as procollagen type
CKD-MBD, we suggest that BMD testing not be I C-terminal pro-peptide) and breakdown (such as
performed routinely because BMD does not pre- type I collagen cross-linked telopeptide, cross-
dict fracture risk as it does in the general popula- laps, pyridinoline, or deoxypyridinoline) (2C).
786 Uhlig et al

The markers mentioned in this recommenda- tion as reasonable alternatives to CT-based imag-
tion do not predict clinical outcomes or bone ing (2C).
histologic states any better than do circulating Extraosseous calcification is one of the compo-
PTH or bone-specific alkaline phosphatase lev- nents of CKD-MBD (Box 1). The prevalence
els. Therefore, the work group issued a weak and severity of extraosseous calcification, includ-
recommendation against their routine measure- ing calcification of arteries and cardiac valves,
ment. Although they currently are not suffi- increase as kidney function decreases. Calcifica-
ciently validated to be recommended for wide tion is more severe and follows an accelerated
use, some of these markers appear promising for course in people with CKD compared with
monitoring the treatment of osteoporosis in pa- healthy people.57,58 CT-based tests, such as elec-
tients with earlier stages of CKD. This recommen- tron beam (EBCT) or multislice CT (MSCT),
dation is applicable to the United States. can measure coronary artery and valvular calcifi-
3.2.5 We recommend that infants with CKD stages 2-5D cations, but other more widely available tests
have length measured at least quarterly, whereas also can measure calcifications in other vessels,
children with CKD stages 2-5D should be assessed for example, lateral abdominal x-ray and echocar-
for linear growth at least annually (1B).
diography (valvular calcification). In the general
Children with CKD stages 2-5D commonly have population, the magnitude of coronary artery
defects in linear growth. The text accompanying calcification imaged using either EBCT or MSCT
this KDIGO recommendation provides additional is a strong predictor of cardiovascular event risk.
age-specific monitoring intervals, which are at least In patients with CKD, the presence and severity
monthly in infants (ie, children aged 1 year), at of cardiovascular calcification also predict cardio-
least quarterly in children younger than 2 years, vascular morbidity and mortality.58
and at least annually in older children and adoles- Ongoing investigation centers on the question
cents. Linear height should be plotted accurately on whether calcification in patients with CKD is
the appropriate growth chart for either height, veloc- located in the intima and thus is similar to that
ity, or ideally both. These intervals and age groups found in non-CKD patients, for whom it corre-
are not entirely consistent with those provided in lates with calcified atherosclerotic plaque, or in
the KDOQI guideline on nutrition in children with the media as an expression of arteriosclerosis,
CKD.9 The latter KDOQI guideline recommends which possibly is related to CKD-MBD. In addi-
more frequent monitoring, as often as monthly in tion to the uncertainty regarding the pathologic
infants with CKD stage 5D and every 6 months in correlate of calcification in patients with CKD, it
older children. However, taken together, the KDIGO has not been shown that measurement of calcifi-
and KDOQI recommendations emphasize that cations using any technique has clinical utility
growth is a sensitive indicator of bone health in for stratification into distinct risk groups, which
children. Children with CKD therefore require more then might derive benefit from modification of
frequent monitoring than healthy children. Beyond their treatment. Thus, most of the work group
the minimum frequency recommended, frequency believed that indiscriminate screening for calcifi-
needs to be individualized based on the degree of cation in patients with CKD-MBD could not be
abnormal height and velocity observed. Delays in recommended.59 This weak recommendation in-
growth should prompt evaluation for causes of dicates the work groups suggestion that if a
growth failure. This recommendation is applicable practitioner still wants to test for calcification in
in the United States. a patient with CKD, lateral abdominal radiogra-
phy and echocardiography can be used as alterna-
Recommendations in Chapter 3.3: Diagnosis of tives to the more costly CT-based imaging.
CKD-MBD: Vascular Calcication In the United States, screening of asymptom-
atic patients with CKD for calcification is not
3.3.1 In patients with CKD stages 3-5D, we suggest that
recommended. The clinical utility of testing spe-
a lateral abdominal radiograph can be used to
detect the presence or absence of vascular calcifi- cific patients for calcification also is not clear. If
cation, and an echocardiogram can be used to a practitioner still wants to perform untargeted
detect the presence or absence of valvular calcifica- testing for calcification, using lateral abdominal
KDOQI Commentary 787

radiography and echocardiography provides as across studies and ranges from 5.0 to 7.0 mg/
much or as little useful information as the more dL.23,42,45,60-62 In patients with CKD stages 3-5,
costly tests using EBCT or MSCT. the risk relationship between phosphorus level
3.3.2 We suggest that patients with CKD stages 3-5D and poor outcome is not found consistently;
with known vascular/valvular calcification be con- however, in some studies, high-normal levels are
sidered at highest cardiovascular risk (2A). It is associated with increased risk, as also seen in
reasonable to use this information to guide manage- individuals without CKD.63,64
ment of CKD-MBD (not graded). Laboratory experimental data show that hyper-
The work group made a discretionary recom- phosphatemia may directly cause or exacerbate
mendation that a patient who is known to have other aspects of CKD-MBD, including second-
vascular or valvular calcification might be consid- ary hyperparathyroidism (HPT), decreased se-
ered to be at highest cardiovascular risk and an rum calcitriol levels, abnormal bone remodeling,
ungraded statement about incorporation of infor- and soft-tissue calcification. However, it has not
mation about calcification into the selection of been examined in placebo-controlled RCTs
CKD-MBD treatments. This recommendation whether treating hyperphosphatemia to specific
rests on epidemiologic data showing higher mor- treatment goals improves clinical outcomes of
tality in those with some or more severe calcifica- patients with CKD. Thus, the work group made a
tion (see online Supplementary Tables 12 and 13 discretionary recommendation for treating hyper-
for chapter 3.3 of the KDIGO guideline). How- phosphatemia, acknowledging that despite the
ever, as discussed, how information about calci- robust risk relationships, observational data do
fication improves the precision of predicting rela- not prove a causal relationship and laboratory
tive or absolute risk in an individual patient is experimental data may not be directly applicable
unclear. Furthermore, it has not been shown that to patients. For CKD stages 3-5, the work group
modification of treatment strategies based on issued a discretionary recommendation suggest-
calcification tests can achieve better patient out- ing to maintain phosphorus levels in the refer-
comes. Recommendation 4.1.5 provides a discre- ence range, and for CKD stage 5D, it issued a
tionary recommendation suggesting that the discretionary recommendation suggesting to de-
amount of calcium-based phosphate binders crease phosphorus levels toward the reference
might be restricted in the presence of arterial range.
calcification. The rationale for this is discussed Treatments for patients with hyperphos-
under recommendation 4.1.5. The work group phatemia include phosphate binders, limiting
found no prospective clinical studies addressing dietary phosphate intake, and/or increasing the
the effects of vitamin D, vitamin D analogues, frequency or duration of dialysis. Use of phos-
and calcimimetics on vascular calcification. phate-restricted diets in combination with oral
phosphate binders has become well established
Recommendations in Chapter 4.1: Treatment of in the management of patients with CKD stages
CKD-MBD Targeted at Lowering High Serum 3-5 and 5D. However, use of phosphate binders
Phosphorus and Maintaining Serum Calcium is associated with side effects, most commonly
4.1.1 In patients with CKD stages 3-5, we suggest of gastrointestinal origin. In dialysis patients,
maintaining serum phosphorus levels in the refer- phosphate binders make up 50% of the high pill
ence range (2C). In patients with CKD stage 5D, burden.65 Thus, in some patients, treatment to
we suggest decreasing increased phosphorus lev- achieve a serum phosphorus level within the
els toward the reference range (2C).
reference range may not be possible, the number
Many patients with CKD stages 4-5D have of pills necessary may be too large, or the degree
high serum phosphorus levels. Observational data of dietary restriction may impact on quality of
for patients with CKD stage 5D show an associa- life. Furthermore, in many patients, mobilization
tion of higher serum phosphorous levels with of phosphorus from the skeleton (or perhaps
mortality and cardiovascular events.41,60 In dialy- other tissues) may contribute to hyperphos-
sis patients, the positive relationship of hyper- phatemia, and this cannot be treated using di-
phosphatemia with mortality is robust, but the etary phosphate binders. This discretionary rec-
threshold above which risk is increased varies ommendation allows clinicians to discuss the
788 Uhlig et al

potential benefits and harms of drug therapy with dialysis patients. Others have argued that in
their patients and individualize decision making many patients, hemodialysate calcium concentra-
based on differing clinical circumstances and tion may need to be lower to achieve a neutral
patient preferences. calcium mass balance.69 A higher dialysate cal-
This recommendation is applicable to the cium concentration may be needed in patients on
United States. nocturnal hemodialysis therapy.70,71 The ratio-
4.1.2 In patients with CKD stages 3-5D, we suggest nale accompanying the KDIGO recommenda-
maintaining serum calcium levels in the reference tion emphasizes the need to individualize dialy-
range (2D). sate calcium concentration for both hemodialysis
The threshold above which calcium level be- and peritoneal dialysis patients. Thus, the US
comes significantly associated with increased practitioner needs to use judgment.
relative risk for all-cause mortality varies among Selecting dialysate calcium concentration re-
studies from 9.5 to 11.4 mg/dL.23,45,60-62 It is quires consideration of the patients calcium lev-
unclear at what level of low serum calcium the els and other laboratory components of CKD-
risk increases. It also is unknown whether treat- MBD; concomitant therapies with phosphate
ment-related hypocalcemia, for example, from binders, calcitriol, vitamin D analogues, or calci-
calcimimetics, confers a risk similar to that with mimetics; and treatment goals. However, in the
identical calcium levels that is not related to absence of robust clinical data regarding the
treatment with this class of drugs. This weak optimal dialysate calcium concentration, the prac-
recommendation suggests using the laboratory titioner in the outpatient dialysis setting also
reference range as the treatment target. A cal- needs to weigh safety concerns in prescribing
cium level outside the reference range requires individualized dialysate calcium concentrations.
evaluation for treatment effects or other causes. 4.1.4 In patients with CKD stages 3-5 (2D) and 5D (2B),
This recommendation is applicable to the United we suggest using phosphate-binding agents in the
States. treatment of hyperphosphatemia. It is reasonable
that the choice of phosphate binder takes into
4.1.3 In patients with CKD stage 5D, we suggest using a
account CKD stage, presence of other components
dialysate calcium concentration of 1.25-1.50
of CKD-MBD, concomitant therapies, and side-
mmol/L (2.5-3.0 mEq/L) (2D).
effect profile (not graded).
Studies that have measured calcium in spent
These weak recommendations suggest using
dialysate to determine net calcium flux with
phosphate binders for the treatment of hyperphos-
hemodialysis have found near-neutral calcium
flux in patients with a dialysate calcium concen- phatemia in patients with CKD. The recommen-
tration of 2.5 mEq/L, although there was variabil- dations are weak because there are no placebo-
ity among patients.66-68 Although maintenance controlled randomized trials that show that
of neutral calcium balance probably is desirable decreasing hyperphosphatemia with a phosphate
in most adult dialysis patients (with slightly binder decreases patient mortality or morbidity.
negative calcium balance perhaps being prefer- Phosphate binders are effective in decreasing
able in patients with extensive vascular calcifica- phosphorus levels. A body of RCT evidence
tion), overall calcium balance is influenced by focuses on the comparative effectiveness of non
diet, vitamin D level, use of vitamin D or its calcium-containing phosphate binders versus cal-
analogues, dialysate calcium concentration, and cium-containing binders.72 There is no proven
other factors. It is not possible to assess overall superiority of any one drug or class for clinical
calcium balance in routine clinical care. It also outcomes. Commonly used phosphate binders
generally is not feasible or likely to be safe to use and their potential advantages and disadvantages
widely varying and individualized dialysate cal- are listed in Table 5, which is reproduced from
cium concentrations within a dialysis unit. The the KDIGO guideline.1
weak recommendation based on a majority vote In children with CKD, calcium-based phos-
of the work group suggests calcium dialysate phate binders have been effective for decreasing
concentration of 2.5-3.0 mEq/L (1.25-1.50 phosphate levels.79 Evidence comparing newer
mmol/L) for both hemodialysis and peritoneal noncalcium-containing binders with calcium-
KDOQI Commentary
Table 5. Phosphate-Binding Compounds

Content
Binder Source Rx Forms (mineral/metal/element) Potential Advantages Potential Disadvantages

Aluminum hydroxide No Liquid, tablet, capsule Aluminum content varies from Very effective phosphate-binding capacity; variety Potential for aluminum toxicity; altered bone
100 to 200 mg/tablet of forms mineralization, dementia; GI side effects
Calcium acetate Yes/No Capsule, tablet Contains 25% elemental Effective phosphate binding, potential for Potential for hypercalcemia-associated
Ca2 (169 mg elemental enhanced phosphate-binding capability over risks, including extraskeletal calcification
Ca2 per 667-mg capsule) CaCO3, potentially less calcium absorption and PTH suppression; more costly than
CaCO3; GI side effects
Calcium carbonate No Liquid, tablet, chewable, Contains 40% elemental Effective, inexpensive, readily available Potential for hypercalcemia-associated
capsule, gum Ca2 (200 mg elemental risks, including extraskeletal calcification
Ca2 per 500 mg CaCO3) and PTH suppression; GI side effects
Calcium citrate No Tablet, liquid, capsule Contains 22% elemental Not recommended in CKD Enhancement of aluminum absorption; GI
Ca2 side effects
Calcium ketoglutarate Similar to other calcium salts, costly, GI side
effects, potentially less hypercalcemic
than calcium carbonate or acetate, not
well studied
Calcium gluconate Tablet, powder Similar to other calcium salts, not well
studied
Ferric citrate GI side effects, not well studied
Magnesium/calcium No Tablet 28% elemental Mg2 (85 mg) Effective; potential for lower calcium load than GI side effects, potential for
carbonate per total MgCO3 and 25% pure calcium-based binders hypermagnesemia, not well studied
elemental Ca2 (100 mg)
per total CaCO3
Magnesium Yes Tablet Lack of availability worldwide; assumed to
carbonate/calcium have similar effects of its components
acetate
Sevelamer-HCl Yes Caplet None Effective; no calcium/metal; not absorbed; Cost; potential for decreased bicarbonate
potential for reduced coronary/aortic levels; may require calcium supplement
calcification compared with calcium-based in presence of hypocalcemia; GI side
binders in some studies; reduces plasma effects
concentration of LDL-C
Sevelamer carbonate Yes Caplet, powder None Effective; no calcium/metal; not absorbed; Cost; may require calcium supplement in
assumed to have similar advantages as presence of hypocalcemia; GI side
sevelamer HCl; potentially improved acid-base effects
balance
Lanthanum carbonate Yes Wafer, chewable Contains 250, 500, or 1,000 Effective; no calcium; chewable Cost, potential for accumulation of
mg elemental lanthanum lanthanum due to GI absorption,
per wafer although long-term clinical
consequences unknown; GI side effects

Abbreviations: CaCO3, calcium carbonate; CKD, chronic kidney disease; CKD-MBD, chronic kidney diseasemineral and bone disease; GI, gastrointestinal; KDIGO, Kidney
Disease: Improving Global Outcomes; LDL-C, low-density lipoprotein cholesterol; PTH, parathyroid hormone; Rx, prescription.

789
Reproduced from the KDIGO CKD-MBD guideline1 with permission of Nature Publishing Group.
790 Uhlig et al

containing binders is limited and insufficient to recommendations do not provide a specific


support specific recommendations. amount for the upper limit of a safe amount of
This recommendation is applicable to the calcium intake because there are no trial data to
United States. It allows practitioners to initiate support one. The KDOQI guidelines provided an
phosphate-binder treatment based on their and opinion-based suggestion to limit daily calcium
their patients judgments for phosphorus targets. intake from phosphate binders to 1,500 mg/d for
It also provides flexibility to choose a binder elemental calcium and 2,000 mg/d for total in-
based on its profile of effects and side effects and take of elemental calcium including dietary cal-
allows combining binders to minimize side ef- cium regardless of the presence of calcification.8
fects from high doses of one agent. Higher calcium intake may be considered when
4.1.5 In patients with CKD stages 3-5D and hyperphos-
serum calcium level is low or PTH level is high.
phatemia, we recommend restricting the dose of However, dialysate calcium and administration
calcium-based phosphate binder and/or the dose of vitamin D analogues also may contribute to a
of calcitriol or vitamin D analogue in the presence net positive calcium balance.
of persistent or recurrent hypercalcemia (1B).
In patients with CKD stages 3-5D and hyperphos- 4.1.6 In patients with CKD stages 3-5D, we recommend
phatemia, we suggest restricting the dose of cal- avoiding long-term use of aluminum-containing
cium-based phosphate binders in the presence of phosphate binders, and in patients with CKD stage
arterial calcification (2C) and/or adynamic bone 5, avoiding dialysate aluminum contamination to
disease (2C) and/or if serum PTH levels are prevent aluminum intoxication (1C).
persistently low (2C).
This recommendation reflects the opinion of
Hypercalcemia is a recognized side effect of the work group that because numerous alterna-
calcium-containing phosphate binders and vita- tive phosphate binders are available and there is
min D analogues. Severe or persistent hypercal- no ability to predict a safe aluminum dose, long-
cemia necessitates the reduction or cessation of term use of aluminum-based phosphate binders
either or both drugs. A weak recommendation should be avoided. This recommendation is appli-
suggests exercising restraint in prescribing high cable to the United States and reflects current
doses of calcium-based phosphorus binders in practice in the United States. Short-term treat-
the presence of arterial calcification. In aggre- ment with aluminum-containing phosphate bind-
gate, studies comparing calcium carbonate or ers may be used when clinically appropriate.
calcium acetate against the noncalcium-based 4.1.7 In patients with CKD stages 3-5D, we suggest
binder sevelamer are inconclusive in showing limiting dietary phosphate intake in the treatment
superiority for clinical outcomes.72 However, of hyperphosphatemia alone or in combination
with other treatments (2D).
some studies showed less progression of calcifi-
cation in sevelamer-treated patients, although Data are insufficient to strongly endorse di-
this effect was inconsistent across studies.72 Tri- etary phosphate restriction as the primary inter-
als of lanthanum versus calcium-containing bind- vention for the management of CKD-MBD. How-
ers have not examined clinical outcomes. ever, dietary phosphorus restriction may help
A dose reduction of calcium-based phosphate keep phosphorus levels normal in patients with
binders also is suggested in the presence of CKD stages 3-5 and serve as an adjunct to
adynamic bone disease or suppressed PTH level. phosphate binders and dialytic phosphorus re-
Data from trials comparing noncalcium binders moval in dialysis patients. This requires attention
with calcium-based binders are inconclusive re- to maintaining adequate protein intake. In the
garding effects on bone histologic states, and United States, a significant portion of phospho-
clinical bone fracture has not been examined as rus intake may derive from phosphate salts used
an outcome. as additives and preservatives, especially in pro-
Any discretionary recommendation depends cessed and fast foods.73-75 Thus, dietary counsel-
on the work groups judgments to a large degree ing to avoid food with high phosphorus content
and the suggested course of action allows indi- while ensuring adequate protein intake may help
vidualization of therapy. Following these sugges- with management of increased serum phospho-
tions is discretionary for the US practitioner. The rus concentrations, particularly in long-term di-
KDOQI Commentary 791

alysis patients. In the United States, most practi- phosphate intake and administering phosphate
tioners will rely on dialysis unit dieticians to binders, calcium supplements, and/or native vita-
min D (not graded).
counsel patients regarding dietary phosphorus
and protein intake. In other settings for patients As the recommendation states, the optimal
with CKD not on dialysis therapy, such dietary PTH level for patients with CKD stages 3-5 who
counseling often is more difficult to obtain. are not on dialysis therapy is not known. The
4.1.8 In patients with CKD stage 5D, we suggest recommendation suggests evaluating those with
increasing dialytic phosphate removal in the treat- PTH levels higher than the upper limit of the
ment of persistent hyperphosphatemia (2C). reference range for potentially modifiable fac-
How modification of dialysis prescription can tors, such as hyperphosphatemia, hypocalcemia,
improve phosphorus removal is an area of inter- and vitamin D deficiency, that may have led to
est. Despite the absence of evidence supporting secondary HPT. Treatment of these factors may
specific phosphorus target levels in patients on decrease PTH levels into the reference range or
dialysis therapy, better dialytic control of serum prevent further increase. The suggested course of
phosphorus levels has the potential to reduce the action is discretionary for US practitioners.
need for phosphate binders and allow more lib- 4.2.2 In patients with CKD stages 3-5 not on dialysis
eral dietary phosphorus intake. One study com- therapy in whom serum PTH levels are progres-
sively increasing and remain persistently higher
paring nocturnal prolonged-duration hemodialy-
than the upper reference limit for the assay despite
sis 6 times weekly with standard thrice-weekly correction of modifiable factors, we suggest treat-
hemodialysis found lower phosphorus levels and ment with calcitriol or vitamin D analogues (2C).
a lower amount of required oral phosphate
binder.76 If PTH levels progressively increase and re-
Control of hyperphosphatemia may be fac- main higher than the reference range, treatment
tored in as a treatment goal when choosing a with calcitriol or vitamin D analogues is sug-
modality or prescription for a hemodialysis pa- gested. In the absence of placebo-controlled tri-
tient. However, thrice-weekly hemodialysis, typi- als showing clinical benefit from treatment of
cally 3.5-4 hours per session in a dialysis center, HPT, this weak recommendation is based on the
is the most common prescription in the United decrease in PTH levels in response to active
States, and any deviation from this delivery model vitamin D compounds.77 The suggested course
encounters logistic, administrative, and financial of action is discretionary for US practitioners.
challenges. Because dialysis dose and intensity Caution should be exercised to avoid hypercalce-
affect not only serum phosphate levels, it will mia and increases in serum phosphorus levels.
require studies of clinical outcomes comparing The work group did not recommend use of
conventional with more extended or more fre- calcimimetics in patients with stages 3-5 CKD
quent dialysis to support the need for changing because of insufficient data for efficacy and
the status quo. There is no evidence that there are safety.
clinically meaningful differences in phosphorus 4.2.3 In patients with CKD stage 5D, we suggest
removal among different dialysis membranes or maintaining iPTH levels in the range of approxi-
mately 2-9 times the upper reference limit for the
dialyzers in current routine use that would enable
assay (2C).
increasing phosphorus removal. We suggest that marked changes in PTH levels in
either direction within this range prompt an initia-
Recommendations in Chapter 4.2: Treatment of tion or change in therapy to avoid progression to
Abnormal PTH Levels in CKD-MBD levels outside of this range (2C).

4.2.1 In patients with CKD stages 3-5 not on dialysis The suggested PTH level range for patients
therapy, the optimal PTH level is not known. with CKD stage 5D is not supported by high-
However, we suggest that patients with iPTH quality evidence. It takes into account wide inter-
levels higher than the upper reference limit of the assay variability of values obtained with many of
assay first are evaluated for hyperphosphatemia,
hypocalcemia, and vitamin D deficiency (2C). the commercial iPTH assays in use, likely be-
It is reasonable to correct these abnormalities with cause of variable reactivity with accumulating
any or all of the following: decreasing dietary PTH fragments. Thus, it is important to know the
792 Uhlig et al

characteristics of the particular iPTH assay in severity, concomitant medications, and clinical
use. See also discussion of recommendation 3.1.6. signs and symptoms (2D).
We suggest that if iPTH levels decrease to less
The KDOQI guidelines suggested a PTH level
than 2 times the upper reference limit for the assay,
range of 150-300 pg/mL for patients with CKD calcitriol, vitamin D analogues, and/or calcimimet-
stage 5D using a Nichols iPTH assay that is no ics be reduced or stopped (2C).
longer available; iPTH levels within this range
also are not uniformly predictive of bone histo- These medical treatments are effective for
logic states, especially when considered alone. decreasing PTH levels. Selection of an agent
The point above which PTH level becomes sig- needs to consider the trends of calcium and
nificantly associated with increased all-cause mor- phosphorus levels along with those of PTH, their
tality varies among studies from 400 to 600 degree of abnormality, and concomitant therapy
pg/mL. The PTH level range suggested in the with phosphorus binders. If serum calcium level
KDIGO guideline corresponds to approximately is low, vitamin D sterols can be the mainstay. If
130-600 pg/mL, taking into account the different serum calcium level is increased, a calcimimetic
iPTH assays in use commercially.48 To date, no can be used. There are no data supporting the
RCT has examined whether treatment to achieve clinical superiority of any vitamin D analogues
a specific PTH target improves clinical out- available in the United States compared with
comes. calcitriol or placebo.77
It is important to recognize that treatments In children with CKD stages 3-5D, trial data
aimed at affecting PTH levels also invariably were limited to comparisons of calcitriol with
influence calcium and phosphorus levels and placebo, calcitriol in different frequency or
levels of other hormones, making it difficult to through a different route, and paricalcitol with
assess the therapeutic benefit of interventions placebo. The evidence was insufficient to make
based on PTH level changes. The recommenda- specific recommendations in children. There were
tion suggests that marked changes in PTH levels no studies evaluating calcimimetics in children.
within this PTH range should trigger a response These recommendations are applicable in the
to avoid a future level outside the range. This United States.
recommendation gives flexibility to US practitio-
4.2.5 In patients with CKD stages 3-5D with severe HPT
ners in using and adjusting treatments that are who fail to respond to medical/pharmacologic
effective in decreasing PTH levels despite lack therapy, we suggest parathyroidectomy (2B).
of proof for clinical benefit from a specific PTH
range. The number of parathyroidectomies in the
United States has decreased in the past 10-15
4.2.4 In patients with CKD stage 5D and increased or
years given the effectiveness of drugs for medi-
increasing PTH levels, we suggest that calcitriol,
vitamin D analogues, calcimimetics, or a combina- cal treatment of HPT and lack of evidence
tion of calcimimetics and calcitriol or vitamin D showing clear superiority of parathyroidec-
analogues be used to decrease PTH levels (2B). tomy on meaningful clinical outcomes. How-
It is reasonable that the initial drug selection for ever, severe HPT may be resistant to medical
the treatment of increased PTH level be based on
serum calcium and phosphorus levels and other
therapy. Subtotal or total parathyroidectomy
aspects of CKD-MBD (not graded). with autotransplant performed by an expert
It is reasonable that calcium- or noncalcium- surgeon effectively decreases PTH, calcium,
based phosphate binder dosage be adjusted so that and phosphorus levels. There is a lack of RCTs
treatments to control PTH levels do not compro-
directly comparing medical with surgical
mise phosphorus and calcium levels (not graded).
We recommend that in patients with hypercalce- therapy for HPT. Nevertheless, it should be
mia, calcitriol or other vitamin D sterol be reduced remembered that surgical treatment is an op-
or stopped (1B). tion in patients with acceptable surgical risk in
We suggest that in patients with hyperphos- whom medical therapy has failed either be-
phatemia, calcitriol or other vitamin D sterol be
reduced or stopped (2D).
cause of lack of response of PTH or side
We suggest that in patients with hypocalcemia, effects from medical therapy. This recommen-
calcimimetics be reduced or stopped depending on dation is applicable to the United States.
KDOQI Commentary 793

Recommendations in Chapter 4.3: Treatment of ifene had similar efficacy in those with moder-
Bone With Bisphosphonates, Other Osteoporosis ately decreased eGFR as in those with a mildly
Medications, and Growth Hormone decreased or normal eGFR, resulting in im-
4.3.1 In patients with CKD stages 1-2 with osteoporosis
proved BMD and reduced fractures. This recom-
and/or high risk of fracture, identified using World mendation is applicable to the United States. It
Health Organization (WHO) criteria, we recom- may apply to a large group of older patients with
mend management as for the general population eGFRs in the upper range of CKD stage 3 (ie,
(1A). GFR, 45-60 mL/min/1.73 m2) who do not have
Because CKD-MBD usually is not present in laboratory evidence of CKD-MBD.
patients with CKD stages 1-2, these individuals 4.3.3 In patients with CKD stage 3 with biochemical
should be treated for osteoporosis or high frac- abnormalities of CKD-MBD and low BMD and/or
ture risk as the general population. The narrative fragility fractures, we suggest that treatment choices
for this recommendation contains a link to a take into account the magnitude and reversibility
of biochemical abnormalities and progression of
web-based tool (FRAX; www.shef.ac.uk/FRAX/ CKD, with consideration of a bone biopsy (2D).
index.htm), which provides a calculator to deter-
mine risk of fracture within the subsequent de- The narrative to this recommendation de-
cade based on race/ethnicity. Given the high scribes that in CKD stage 3, some patients have
prevalence of early stages of CKD in elderly already developed abnormalities of CKD-MBD,
patients who are likely to have osteoporosis, this in particular, secondary HPT. As kidney disease
recommendation calls attention to the need to progresses, bone disease changes from idio-
evaluate fracture risk in this population and treat pathic osteoporosis to renal osteodystrophy. De-
accordingly. This recommendation is applicable spite wide variability among patients regarding
in the United States. this transition, biochemical manifestations of
4.3.2 In patients with CKD stage 3 with PTH levels in the CKD-MBD initially appear at an approximate
reference range and osteoporosis and/or high risk of GFR of 40-50 mL/min/1.73 m2. The pathogene-
fracture identified using WHO criteria, we suggest sis of bone disease in patients with CKD-MBD is
treatment as for the general population (2B). different from that in patients with postmeno-
Although there are no trials of antiosteoporo- pausal osteoporosis. Therefore, extrapolating ef-
sis therapies specifically in individuals with des- fects from patients with osteoporosis that ex-
ignated CKD, there is some evidence from large cluded individuals with abnormal PTH values to
trials with bisphosphonates, teriparatide, and patients with CKD stages 3-5D may not be valid.
raloxifene in women with postmenopausal osteo- Along with concerns about the applicability of
porosis. Given the trial inclusion criteria, these treatment effects, there is increasing concern
individuals did not have evidence of CKD-MBD about long-term safety with drugs that are cleared
because they excluded participants with manifest by the kidneys.82 The work group suggests that
kidney disease, as well as those with increased secondary HPT be addressed first, as outlined in
PTH or abnormal calcium or phosphorus levels. recommendation 4.2.1.
Their vitamin D status at baseline is unknown. In patients in whom HPT has been corrected,
However, because the trials used a serum creati- GFR is stable and risk of a fracture outweighs the
nine cutoff level to determine eligibility and potential long-term risk of inducing irreversible
included a large number of elderly women, they low bone turnover, therapy with bisphosphonates
unknowingly included a substantial group of may be considered. However, because bisphos-
individuals with decreased estimated glomerular phonates are likely to prevent fractures only in
filtration rate (eGFR) corresponding to CKD patients who have increased bone resorption, the
stage 3 and even some individuals with eGFR work group suggests a bone biopsy when fea-
corresponding to CKD stage 4.78-81 Despite some sible. If therapy with bisphosphonates is given,
uncertainty stemming from the method of estimat- lower dose and shorter treatment duration should
ing GFR and loss of precision in post hoc sub- be considered. The suggested approach is appli-
group analyses, it appears that treatments with cable in the United States, although as discussed,
risedronate, alendronate, teriparatide, or ralox- bone biopsy availability may be limited.
794 Uhlig et al

4.3.4 In patients with CKD stages 4-5D with biochemi- tuations in serum calcium and phosphorus levels
cal abnormalities of CKD-MBD and low BMD may be seen and thus frequent monitoring is
and/or fragility fractures, we suggest additional
investigation with bone biopsy before therapy with
needed. Hypophosphatemia occurs in a large
antiresorptive agents (2C). proportion of patients immediately after trans-
plant, and serum calcium levels tend to increase
In individuals with CKD stages 4-5D and after transplant; these changes usually stabilize
biochemical evidence of CKD-MBD, trial data after about 6 months.84 PTH levels decrease
for the efficacy and safety of antiresorptive agents significantly during the first 3 months after trans-
are lacking. The work group thus could not plant, but typically stabilize at increased values
recommend routine use of these agents. A bone after 1 year. This recommendation is applicable
biopsy is suggested before therapy with bisphos- to the United States.
phonates, teriparatides, or raloxifene. This sug-
5.2 In patients after the immediate postkidney trans-
gestion is applicable in the United States.
plant period, it is reasonable to base the frequency of
4.3.5 In children and adolescents with CKD stages 2-5D monitoring serum calcium, phosphorus, and PTH on
and related height deficits, we recommend treat- the presence and magnitude of abnormalities and
ment with recombinant human growth hormone rate of progression of CKD (not graded). Reasonable
when additional growth is desired, after first monitoring intervals would be:
addressing malnutrition and biochemical abnor- In CKD stages 1-3T, for serum calcium and phospho-
malities of CKD-MBD (1A). rus, every 6-12 months, and PTH, once, with subse-
quent intervals dependent on baseline level and
In children with CKD, linear growth abnor- CKD progression
malities are common and can be corrected using In CKD stage 4T, for serum calcium and phospho-
recombinant human growth hormone.83 Deci- rus, every 3-6 months, and for PTH, every 6-12
sions to start growth hormone therapy should be months
In CKD stage 5T, for serum calcium and phospho-
based on height velocity and potential for linear rus, every 1-3 months, and for PTH, every 3-6
growth (bone age and maturational stage), as months
well as height deficits. Treatment entails daily In CKD stages 3-5T, measurement of alkaline phos-
subcutaneous injection. Adverse events may in- phatases annually or more frequently in the presence
clude impaired glucose tolerance. The current pedi- of increased PTH levels (see chapter 3.2)
In patients with CKD receiving treatments for CKD-
atric KDOQI guideline for nutrition in children
MBD or in whom abnormalities are identified, it is
with CKD9 specifies that Recombinant human reasonable to increase the frequency of measurements
growth hormone therapy should be considered in to monitor for efficacy and side effects (not graded).
children with CKD stages 2-5D, short stature [height It is reasonable to manage these abnormalities as for
SDS 1.88 (height-for-age 3rd percentile)], patients with CKD stages 3-5 (not graded) (see chapter
and potential for linear growth if growth failure 4.1 and 4.2)
(height velocity-for-age SDS 1.88) persist be- After the immediate posttransplant period,
yond 3 months despite treatment of nutritional transplant function usually stabilizes. The above
deficiency and metabolic abnormalities. In the ungraded statements provide guidance for the
United States, preferences of the patient and his or frequency of monitoring for laboratory abnor-
her guardian about the value placed on accelerated malities at that time. They are extrapolated from
linear growth versus the burden of daily injections those provided for nontransplant patients with
and the potential for harm should be elicited. corresponding CKD stage (see recommendation
3.1.2). Data directly supporting the utility of
RECOMMENDATIONS IN CHAPTER 5: these measurements are limited in nontransplant
EVALUATION AND TREATMENT OF KIDNEY CKD patients and even more limited in kidney
TRANSPLANT BONE DISEASE transplant patients. However, CKD-MBD is com-
mon after kidney transplant. Furthermore, kid-
5.1 In patients in the immediate postkidney transplant ney transplant patients with normal or mildly
period, we recommend measuring serum calcium decreased eGFRs are still considered to have
and phosphorus at least weekly until stable (1B).
CKD and may have residual bone disease from
During the immediate posttransplant period pretransplant CKD-MBD. Thus, in addition to
with usually rapidly changing GFRs, wide fluc- suggesting monitoring serum calcium, phospho-
KDOQI Commentary 795

rus, and PTH levels in patients with CKD stage be reserved for high-risk populations, including
3T (as for CKD stage 3), this is also suggested those receiving significant doses of corticoste-
for CKD stages 1-2T. As for nontransplant pa- roids or those with risk factors for osteoporosis
tients with CKD-MBD, the frequency of measure- in the general population (see recommendation
ments may be increased to monitor for trends and 3.2). In addition, DXA screening is suggested
treatment efficacy and side effects (see identical only in individuals with a well-functioning trans-
statement under 3.1.2.). These frequencies pro- plant, in other words, CKD stages 1-3T. Those
vide a reasonable framework and apply to the with more advanced CKD will be more likely to
United States. have abnormal bone quality from CKD-MBD,
The statement to follow the same principles which is likely to compromise the ability of
for treatment of biochemical abnormalities of BMD to predict fractures. This discretionary
CKD-MBD in kidney transplant patients as out- recommendation is applicable in the United
lined for patients with CKD stages 3-5 is reason- States.
able. However, CKD-MBD in kidney transplant 5.6 In patients in the first 12 months postkidney
patients is an even more heterogeneous disease transplant with eGFR greater than approximately 30
than in nontransplant patients. It is the conse- mL/min/1.73 m2 and low BMD, we suggest that
quence of many different factors, including pre- treatment with vitamin D, calcitriol/alphacalcidiol,
transplant CKD-MBD, effects of immunosup- or bisphosphonates be considered (2D).
We suggest that treatment choices be influenced by
pressive drugs, level of kidney function recovery, the presence of CKD-MBD, indicated by abnormal
and risk factors for osteoporosis. levels of calcium, phosphorus, PTH, alkaline phos-
5.3 In patients with CKD stages 1-5T, we suggest that phatases, and 25(OH)D (2C)
It is reasonable to consider a bone biopsy to guide
25(OH) vitamin D (calcidiol) levels might be mea-
sured, with repeated testing determined by baseline treatment, specifically before the use of bisphospho-
values and interventions (2C). nates because of the high incidence of adynamic
bone disease (not graded)
This weak recommendation also is extrapo-
There are insufficient data to guide treatment
lated from the recommendation for nontrans-
after the first 12 months.
plant patients with CKD (recommendation 3.1.3).
As discussed, bone disease in kidney trans-
It is supported by finding low vitamin D levels in
plant patients is heterogeneous, with variable
some patients after kidney transplant. Avoidance
pathologic states resulting from an overlap of
of sunlight and wider use of sunscreen may
risk factors related to CKD, transplant, and osteo-
contribute to low vitamin D levels in transplant
porosis. Thus, treatment data from the general
patients. This discretionary recommendation is
population without CKD, patients with CKD
applicable in the United States.
without a kidney transplant, or other solid-organ
5.4 In patients with CKD stages 1-5T, we suggest that transplant patients without CKD-MBD cannot
vitamin D deficiency and insufficiency be corrected be directly extrapolated. There are no RCTs in
using treatment strategies recommended for the
general population (2C). kidney transplant patients examining bone-
specific therapies on clinical outcomes, such as
This weak recommendation is extrapolated fractures or cardiovascular disease events. Al-
from the recommendation for nontransplant pa- though loss of bone density occurs after kidney
tients with CKD. See also commentary on recom- transplant,85 particularly in the first year, and
mendation 3.1.3. This discretionary recommenda- some trials have examined BMD as an out-
tion is applicable in the United States. come, this has not been validated as a surro-
5.5 In patients with eGFR greater than approximately 30 gate outcome for fractures. The overall num-
mL/min/1.73 m2, we suggest measuring BMD in the ber of studies of kidney transplant patients and
first 3 months postkidney transplant if they receive number of patients treated with vitamin D,
corticosteroids or have risk factors for osteoporosis, calcitriol/alphacalcidiol, or bisphosphonates are
as in the general population (2D).
low, both within and beyond the first 12 months
It is unclear whether low BMD in kidney after transplant. Thus, treatment recommenda-
transplant patients corresponds to fracture risk. tions are discretionary. Evaluation and selec-
Thus, the work group believed that DXA should tion of any treatments should consider the
796 Uhlig et al

constellation of abnormalities in calcium, phos- seems prudent that treatment with bone-specific
phorus, PTH, and vitamin D levels. Bisphos- therapies other than those aiming at correcting
phonates should not be used if there are abnor- abnormalities of calcium, phosphorus, PTH, and
malities in calcium, phosphate, vitamin D, or vitamin D levels would be guided by a bone
PTH levels. biopsy (see recommendation 5.6).
In children with CKD stages 1-5T, there is
only one trial comparing placebo versus alfa- RESEARCH RECOMMENDATIONS
cacidiol versus calcitonin versus alendronate.86 The KDIGO guideline document summarizes
The evidence was deemed to be insufficient to our current knowledge regarding the manage-
support specific recommendations for treatments ment of CKD-MBD and highlights areas in need
for bone disease in pediatric kidney transplant of future research. Each guideline chapter con-
recipients. tains research recommendations at the end, and
Given the complexity of MBD in kidney trans- chapter 6 contains a short list of research ques-
plant recipients, it is reasonable to consider a tions deemed to have high priority for advancing
bone biopsy to guide bone-specific treatment. the field. Research comparing therapeutic strate-
This would be particularly important before us- gies for CKD-MBD admittedly is difficult to do
ing bisphosphonates because these agents have because phosphate-lowering drugs, vitamin D
better efficacy in high bone turnover and may and its analogues, calcimimetics, and dialysis
lead to adynamic bone disease. These recommen- calcium concentrations variably impact on the
dations and statements are applicable in the components of MBD. However, key questions
United States. are to what target phosphorus levels should be
decreased in patients with CKD stages 3-5 and
5.7 In patients with CKD stages 4-5T, we suggest that 5D and what the optimal treatment strategy is for
BMD testing not be performed routinely because this. Similarly, a key question is to what PTH
BMD does not predict fracture risk as it does in the target we should treat and with what strategy. In
general population and BMD does not predict type
of kidney transplant bone disease (2B).
a population with a great burden of mortality and
morbidity, treatment trials have to examine clini-
The uncertainty surrounding the value of BMD cal end points. Future treatment trials also should
for predicting underlying bone disease, fracture, examine surrogate outcomes along with clinical
or other clinical outcomes in kidney transplant outcomes because reliance on surrogate end
patients increases with more advanced stages points requires validation by showing concor-
of CKD because there is a higher likelihood of dance with clinical outcomes in trials of similar
more severe underlying bone abnormalities of agents or strategies in addition to robust risk
CKD-MBD. This recommendation is applicable relationships in observational studies.87
to the United States.
CONCLUSION
5.8 In patients with CKD stages 4-5T with known low The KDIGO guideline is based on the newly
BMD, we suggest management as for patients with created disease concept of CKD-MBD. KDIGO
CKD stages 4-5 not on dialysis therapy, as detailed
in chapter 4.1 and 4.2.(2C)
used an approach to guideline development that
required stringent mapping of recommendations
Although routine testing for BMD in patients to the available evidence. It exposed the uncer-
with CKD stages 4-5T is discouraged, some tainty surrounding a number of practice areas, as
patients may still undergo testing that shows low well as the lack of definitive evidence for clinical
BMD. This discretionary recommendation sug- benefit from a range of currently widely used and
gests that these individuals be referred to as advocated treatments. This prohibits the issuance
having low BMD rather than osteoporosis and of specific and directive recommendations, but
that they be evaluated and managed for hyper- allows for more flexibility in setting treatment
phosphatemia and HPT as patients with CKD goals and weighing benefits and harms in spe-
without a kidney transplant (as described in cific situations. Target ranges proposed by the
chapter 4.1 and 4.2 in the KDIGO guideline). It 2003 KDOQI guideline for phosphorus and PTH
KDOQI Commentary 797

levels in patients with CKD stage 5D are not 10. Centers for Medicare & Medicaid Services. End-
supported by high-quality evidence and should stage renal disease prospective payment system. Proposed
rule and notice. Fed Regist. 2009;74(187):49923-50102.
not be used for performance measurement.
11. Weiner DE, Watnick SG. The 2009 proposed rule for
ACKNOWLEDGEMENTS prospective ESRD payment: historical perspectives and pub-
lic policiesbundle up! Am J Kidney Dis. 2010;55(2):217-
Guideline recommendations included in this article origi- 222.
nally were published in Kidney International and were 12. Nissenson AR, Mayne TJ, Krishnan M. The 2009
reproduced with permission from KDIGO.1 We thank Amy proposed rule for prospective ESRD payment: perspectives
Earley for help with reference management. from a large dialysis organization. Am J Kidney Dis. 2010;
Financial Disclosure: Dr Berns has received honorarium 55(2):223226.
or grants from Affymax, Amgen, Wyeth, and Glaxo Smith 13. Moran J. The 2009 proposed rule for prospective
Kline and is a Board Member for Litholink. Dr Kestenbaum ESRD payment: perspectives from a medium-sized dialysis
is a consultant for Shire and Genzyme and has received organization. Am J Kidney Dis. 2010;55(2):227228.
grant/research support from Amgen. Dr Kumar has received 14. Sadler JH. The 2009 proposed rule for prospective
support from Genzyme, Abbott, Bayer, and Deltanoid. Dr ESRD payment: perspectives from a not-for-profit small
Martin is a consultant for Abbott, Shire, Genzyme, and dialysis organization. Am J Kidney Dis. 2010;55(2):229
Amgen and is on the Advisory Board for Cytochroma. Dr 230.
Sprague serves on the Advisory Board for Shire, Genzyme, 15. Bhat JG, Bhat P. The 2009 proposed rule for prospec-
and Ineos and has received support from Mitsubishi Re- tive ESRD payment: perspectives from a for-profit small
search. Dr Goldfarb is a speaker for Fresenius Health Care
dialysis organization. Am J Kidney Dis. 2010;55(2):231
and GE Healthcare. Drs Leonard and Uhlig have no financial
233.
relationships with any commercial entity producing health
16. Kristensen C, Wish J. The 2009 proposed rule for
carerelated products and/or services. Dr Uhlig served as the
prospective ESRD payment: perspectives from the forum of
Director of the Evidence Review Team for the KDIGO
ESRD Networks. Am J Kidney Dis. 2010;55(2):234236.
CKD-MBD guideline.
17. Norman ME, Mazur AT, Borden S, et al. Early diag-
REFERENCES nosis of juvenile renal osteodystrophy. J Pediatr. 1980;97(2):
226-232.
1. Kidney Disease: Improving Global Outcomes (KDIGO)
18. Gauci C, Moranne O, Fouqueray B, et al. Pitfalls of
CKD-MBD Work Group. KDIGO Clinical Practice Guide-
measuring total blood calcium in patients with CKD. J Am
line for the Diagnosis, Evaluation, Prevention, and Treat-
Soc Nephrol. 2008;19(8):15921598.
ment of Chronic Kidney Disease-Mineral and Bone Disor-
19. Urena P, Hruby M, Ferreira A, et al. Plasma total
der (CKD-MBD). Kidney Int Suppl. 2009;113:S1-130.
versus bone alkaline phosphatase as markers of bone turn-
2. Eknoyan G, Lameire N, Barsoum R, et al. The burden
over in hemodialysis patients. J Am Soc Nephrol. 1996;7(3):
of kidney disease: improving global outcomes. Kidney Int.
506-512.
2004;66(4):1310-1314.
20. Coen G, Ballanti P, Bonucci E, et al. Bone markers in
3. Moe S, Drueke T, Cunningham J, et al. Definition,
evaluation, and classification of renal osteodystrophy: a the diagnosis of low turnover osteodystrophy in haemodialy-
position statement from Kidney Disease: Improving Global sis patients. Nephrol Dial Transplant. 1998;13(9):2294-
Outcomes (KDIGO). Kidney Int. 2006;69(11):1945-1953. 2302.
4. Atkins D, Best D, Briss PA, et al. Grading quality of 21. Blayney MJ, Pisoni RL, Bragg-Gresham JL, et al.
evidence and strength of recommendations. BMJ. 2004; High alkaline phosphatase levels in hemodialysis patients
328(7454):1490-1497. are associated with higher risk of hospitalization and death.
5. Uhlig K, MacLeod A, Craig J, et al. Grading evidence Kidney Int. 2008;74(5):655-663.
and recommendations for clinical practice guidelines in 22. Regidor DL, Kovesdy CP, Mehrotra R, et al. Serum
nephrology. A position statement from Kidney Disease: alkaline phosphatase predicts mortality among maintenance
Improving Global Outcomes (KDIGO). Kidney Int. 2006; hemodialysis patients. J Am Soc Nephrol. 2008;19(11):2193
70(12):2058-2065. 2203.
6. Guyatt GH, Oxman AD, Kunz R, et al. Going from 23. Kalantar-Zadeh K, Kuwae N, Regidor DL, et al.
evidence to recommendations. BMJ. 2008;336(7652):1049- Survival predictability of time-varying indicators of bone
1051. disease in maintenance hemodialysis patients. Kidney Int.
7. Fervers B, Remy-Stockinger M, Graham ID, et al. 2006;70(4):771-780.
Guideline adaptation: an appealing alternative to de novo 24. Levin A, Bakris GL, Molitch M, et al. Prevalence of
guideline development. Ann Intern Med. 2008;148(7):563- abnormal serum vitamin D, PTH, calcium, and phosphorus
564; author reply 564-565. in patients with chronic kidney disease: results of the study
8. National Kidney Foundation. K/DOQI Clinical Prac- to evaluate early kidney disease. Kidney Int. 2007;71(1):
tice Guidelines for Bone Metabolism and Disease in Chronic 31-38.
Kidney Disease. Am J Kidney Dis. 2003;42(suppl 3):S1-202. 25. Urena P, Ferreira A, Kung VT, et al. Serum pyridino-
9. National Kidney Foundation. KDOQI Clinical Prac- line as a specific marker of collagen breakdown and bone
tice Guideline for Nutrition in Children With CKD: 2008 metabolism in hemodialysis patients. J Bone Miner Res.
update. Am J Kidney Dis. 2009;53(suppl 2):S1-124. 11995;0(6):932-939.
798 Uhlig et al

26. Binkley N, Krueger D, Lensmeyer G. 25-Hydroxyvi- 45. Tentori F, Blayney M, Albert JM, et al. Mortality risk
tamin D measurement, 2009: a review for clinicians. J Clin for dialysis patients with different levels of serum calcium,
Densitometry. 2009;12(4):417-427. phosphorus, and parathyroid hormone: the Dialysis Out-
27. Zidehsarai MP, Moe SM. Review article: chronic comes and Practice Patterns Study (DOPPS). Am J Kidney
kidney disease-mineral bone disorder: have we got the Dis. 2008;52(3):519-530.
assays right? Nephrology. 2009;14(4):374-382. 46. Souberbielle JC, Roth H, Fouque DP. Parathyroid
28. Jones G, Horst R, Carter G, et al. Contemporary hormone measurement in CKD. Kidney Int. 2010;77(2):93-
diagnosis and treatment of vitamin D-related disorders. 100.
J Bone Miner Res. 2007;22(suppl 2):V11-15. 47. Gao P, DAmour P. Evolution of the parathyroid
29. Holick MF, Chen TC. Vitamin D deficiency: a world- hormone (PTH) assayimportance of circulating PTH im-
wide problem with health consequences. Am J Clin Nutr. munoheterogeneity and of its regulation. Clin Lab. 2005;
2008;87(4):1080S-1086S. 51(1-2):21-29.
30. Holick MF. Vitamin D for health and in chronic 48. Souberbielle JC, Boutten A, Carlier MC, et al. Inter-
kidney disease. Semin Dial. 2005;18(4):266-275. method variability in PTH measurement: implication for the
31. Giovannucci E. Vitamin D status and cancer inci- care of CKD patients. Kidney Int. 2006;70(2):345-350.
dence and mortality. Adv Exp Med Biol. 2008;624:31-42. 49. Joly D, Drueke TB, Alberti C, et al. Variation in
32. Wolf M, Shah A, Gutierrez O, et al. Vitamin D levels serum and plasma PTH levels in second-generation assays in
and early mortality among incident hemodialysis patients. hemodialysis patients: a cross-sectional study. Am J Kidney
Kidney Int. 2007;72(8):1004-1013. Dis. 2008;51(6):987-995.
33. Lappe JM, Travers-Gustafson D, Davies KM, et al. 50. Cavalier E, Rozet E, Gadisseur R, et al. Measurement
Vitamin D and calcium supplementation reduces cancer risk: uncertainty of 25-OH vitamin D determination with differ-
results of a randomized trial. Am J Clin Nutr. 2007;85(6): ent commercially available kits: impact on the clinical cut
1586-1591. offs [published online ahead of print September 9, 2009].
34. Al-Badr W, Martin KJ. Vitamin D and kidney dis- Osteoporos Int. doi 10.1007/s00198-009-1052-5.
ease. Clin J Am Soc Nephrol. 2008;3(5):1555-1560. 51. Elder GJ, Mackun K. 25-Hydroxyvitamin D defi-
35. Fishbane S, Chittineni H, Packman M. et al. Oral
ciency and diabetes predict reduced BMD in patients with
paricalcitol in the treatment of patients with CKD and
chronic kidney disease. J Bone Miner Res. 2006;21(11):1778-
proteinuria: a randomized trial. Am J Kidney Dis. 2009;54(4):
1784.
647-652.
52. Jamal SA, Hayden JA, Beyene J. Low bone mineral
36. Agarwal R, Acharya M, Tian J, et al. Antiproteinuric
density and fractures in long-term hemodialysis patients: a
effect of oral paricalcitol in chronic kidney disease. Kidney
meta-analysis. Am J Kidney Dis. 2007;49(5):6746-6781.
Int. 2005;68(6):2823-2828.
53. Jamal SA, Gilbert J, Gordon C, et al. Cortical pQCT
37. Teng M, Wolf M, Lowrie E, et al. Survival of patients
measures are associated with fractures in dialysis patients.
undergoing hemodialysis with paricalcitol or calcitriol
J Bone Miner Res. 2006;21(4):543-548.
therapy. N Engl J Med. 2003;349(5):446-456.
54. Carmen SM, Auxiliadora BM, Selgas R, et al.
38. Vieth R. Critique of the considerations for establish-
ing the tolerable upper intake level for vitamin D: critical Parathormone secretion in peritoneal dialysis patients with
need for revision upwards. J Nutr. 2006;136(4):1117-1122. adynamic bone disease. Am J Kidney Dis. 2000;36(5):953-
39. Heaney RP, Davies KM, Chen TC, et al. Human 961.
serum 25-hydroxycholecalciferol response to extended oral 55. Bervoets AR, Spasovski GB, Behets GJ, et al. Useful
dosing with cholecalciferol. Am J Clin Nutr. 2003;77(1):204- biochemical markers for diagnosing renal osteodystrophy in
210. predialysis end-stage renal failure patients. Am J Kidney Dis.
40. Trang HM, Cole DE, Rubin LA, et al. Evidence that 2003;41(5):997-1007.
vitamin D3 increases serum 25-hydroxyvitamin D more 56. Gal-Moscovici A, Popovtzer MM. New worldwide
efficiently than does vitamin D2. Am J Clin Nutr. 1998;68(4): trends in presentation of renal osteodystrophy and its relation-
854-858. ship to parathyroid hormone levels. Clin Nephrol. 2005;63(4):
41. Block GA, Hulbert-Shearon TE, Levin NW, et al. 284-289.
Association of serum phosphorus and calcium x phosphate 57. Goodman WG, Goldin J, Kuizon BD, et al. Coronary-
product with mortality risk in chronic hemodialysis patients: artery calcification in young adults with end-stage renal
a national study. Am J Kidney Dis. 1998;31(4):607-617. disease who are undergoing dialysis. N Engl J Med. 2000;
42. Noordzij M, Korevaar JC, Boeschoten EW, et al. The 342(20):1478-1483.
Kidney Disease Outcomes Quality Initiative (KDOQI) 58. Raggi P, Boulay A, Chasan-Taber S, et al. Cardiac
Guideline for Bone Metabolism and Disease in CKD: asso- calcification in adult hemodialysis patients. A link between
ciation with mortality in dialysis patients. Am J Kidney Dis. end-stage renal disease and cardiovascular disease? J Am
2005;46(5):925-932. Coll Cardiol. 2002;39(4):695-701.
43. Ketteler M, Brandenburg V, Jahnen-Dechent W. Do 59. Uhlig K. There is no practical utility in routinely
not be misguided by guidelines: the calcium x phosphate screening dialysis patients for vascular calcification. Semin
product can be a Trojan horse. Nephrol Dial Transplant. Dial., in press.
2005;20:673-677. 60. Block GA, Klassen PS, Lazarus JM, et al. Mineral
44. ONeill WC. The fallacy of the calcium-phosphorus metabolism, mortality, and morbidity in maintenance hemo-
product. Kidney Int. 2007;72(7):792-796. dialysis. J Am Soc Nephrol. 2004;15(8):2208-2218.
KDOQI Commentary 799

61. Young EW, Albert JM, Satayathum S, et al. Predictors poultry, and fish products as protein sources. Am J Kidney
and consequences of altered mineral metabolism: the Dialy- Dis. 2009;54(1):18-23.
sis Outcomes and Practice Patterns Study. Kidney Int. 2005; 75. Sherman RA, Mehta O. Phosphorus and potassium
67(3):1179-1187. content of enhanced meat and poultry products: implications
62. Kimata N, Albert JM, Akiba T, et al. Association of for patients who receive dialysis. Clin J Am Soc Nephrol.
mineral metabolism factors with all-cause and cardiovascu- 2009;4(8):1370-1373.
lar mortality in hemodialysis patients: the Japan Dialysis 76. Culleton BF, Walsh M, Klarenbach SW, et al. Effect
Outcomes and Practice Patterns Study. Hemodial Int. 2007; of frequent nocturnal hemodialysis vs conventional hemodi-
11(3):340-348. alysis on left ventricular mass and quality of life: a random-
63. Kestenbaum B, Sampson JN, Rudser KD, et al. Serum ized controlled trial. JAMA. 2007;298(11):1291-1299.
phosphate levels and mortality risk among people with chronic 77. Palmer SC, McGregor DO, Craig JC, et al. Vitamin D
kidney disease. J Am Soc Nephrol. 2005;16(2):520-528. compounds for people with chronic kidney disease not
64. Menon V, Greene T, Pereira AA, et al. Relationship of requiring dialysis. Cochrane Database Syst Rev. 2009;(4):
phosphorus and calcium-phosphorus product with mortality CD008175.
in CKD. Am J Kidney Dis. 2005;46(3):455-463. 78. Neer RM, Arnaud CD, Zanchetta JR, et al. Effect of
65. Chiu YW, Teitelbaum I, Misra M, et al. Pill burden, parathyroid hormone (1-34) on fractures and bone mineral
adherence, hyperphosphatemia, and quality of life in mainte- density in postmenopausal women with osteoporosis. N Engl
nance dialysis patients. Clin J Am Soc Nephrol. 2009;4(6): J Med. 2001;344(19):1434-1441.
1089-1096. 79. Miller PD, Roux C, Boonen S, et al. Safety and
66. Hou SH, Zhao J, Ellman CF, et al. Calcium and efficacy of risedronate in patients with age-related reduced
phosphorus fluxes during hemodialysis with low calcium renal function as estimated by the Cockcroft and Gault
dialysate. Am J Kidney Dis. 1991;18(2):217-224.
method: a pooled analysis of nine clinical trials. J Bone
67. Argiles A, Kerr PG, Canaud B, et al. Calcium kinetics
Miner Res. 2005;20:2105-2115.
and the long-term effects of lowering dialysate calcium
80. Jamal SA, Bauer DC, Ensrud KE, et al. Alendronate
concentration. Kidney Int. 1993;43(3):630-640.
treatment in women with normal to severely impaired renal
68. Sigrist M, McIntyre CW. Calcium exposure and re-
function: an analysis of the fracture intervention trial. J Bone
moval in chronic hemodialysis patients. J Ren Nutr. 2006;
Miner Res. 2007;22(4):503-508.
16(1):41-46.
81. Ishani A, Blackwell T, Jamal SA, et al. The effect of
69. Gotch F, Levin NW, Kotanko P. Calcium balance in
raloxifene treament in postmenopausal women with CKD.
dialysis is best managed by adjusting dialysate calcium guided
by kinetic modeling of the interrelationship between calcium J Am Soc Nephrol. 2008;19:1430-1438.
intake, dose of vitamin D analogues and the dialysate calcium 82. Cunningham J. Bisphosphonates in the renal patient.
concentration. Blood Purif. 2010;29(2):163-176. Nephrol Dial Transplant. 2007;22(6):1505-1507.
70. Al-Hejaili F, Kortas C, Leitch R, et al. Nocturnal but 83. Vimalachandra D, Hodson EM, Willis NS, et al.
not short hours quotidian hemodialysis requires an elevated Growth hormone for children with chronic kidney disease.
dialysate calcium concentration. J Am Soc Nephrol. 2003; Cochrane Database Syst Rev. 2006;3:CD003264.
14(9):2322-2328. 84. Sprague SM, Belozeroff V, Danese MD, et al. Abnor-
71. Toussaint ND, Polkinghorne KR, Kerr PG, et al. mal bone and mineral metabolism in kidney transplant
Comparison between different dialysate calcium concentra- patientsa review. Am J Nephrol. 2008;28(2):246-253.
tions in nocturnal hemodialysis. Hemodial Int. 2007;11(2): 85. Julian BA, Laskow DA, Dubovsky J, et al. Rapid loss
217-224. of vertebral mineral density after renal transplantation. N Engl
72. Navaneethan SD, Palmer SC, Craig JC, et al. Benefits J Med. 1991;325(8):544-550.
and harms of phosphate binders in CKD: a systematic 86. El-Husseini AA, El-Agroudy AE, El-Sayed MF, et al.
review of randomized controlled trials. Am J Kidney Dis. Treatment of osteopenia and osteoporosis in renal transplant
2009;54(4):619-637. children and adolescents. Pediatr Transplant. 2004;8(4):357-
73. Kalantar-Zadeh K, Gutekunst L, Mehrotra R, et al. 361.
Understanding sources of dietary phosphorus in the treat- 87. Bucher HC, Guyatt GH, Cook DJ, et al. Users guides
ment of patients with chronic kidney disease. Clin J Am Soc to the medical literature: XIX. Applying clinical trial results.
Nephrol. 2010;5(3):519-530. A. How to use an article measuring the effect of an interven-
74. Sherman RA, Mehta O. Dietary phosphorus restric- tion on surrogate end points. Evidence-Based Medicine
tion in dialysis patients: potential impact of processed meat, Working Group. JAMA. 1999;282(8):771-778.

Potrebbero piacerti anche