Вы находитесь на странице: 1из 8

In Practice

Long-term Management of CKD–Mineral and Bone Disorder


Kevin J. Martin, MB, BCh, and Esther A. González, MD

Chronic kidney disease–mineral and bone disorder (CKD-MBD) is the term used to describe the abnormali-
ties of bone and mineral metabolism that occur in the setting of kidney disease. The spectrum of these
abnormalities is wide, ranging from severe high-turnover bone disease on one end to marked low bone
turnover bone disease on the other. Similarly, some patients have severe vascular calcifications while others
do not, and the values for biochemistry determinations, including calcium, phosphorus, and parathyroid
hormone, also may vary widely among patients. This variability may be influenced by such things as the
chronicity of the particular kidney disease, effects of therapies such as corticosteroids on modifying the course
of kidney disease, and comorbid conditions, such as diabetes, heart disease, age, and osteoporosis. The
heterogeneity of CKD-MBD makes strict protocol-driven therapeutic approaches difficult; accordingly, consid-
erable individualized therapy is required. Using a case history, we explore several of the variables and
difficulties involved in patient management.
Am J Kidney Dis. 60(2):308-315. © 2012 by the National Kidney Foundation, Inc.

INDEX WORDS: Chronic kidney disease–mineral and bone disorder (CKD-MBD); hyperparathyroidism; renal
osteodystrophy.

CASE PRESENTATION list. Cardiac stress echocardiography was negative for ischemia,
but left ventricular hypertrophy and modest calcification of the
A 56-year-old white woman with a history of lupus nephritis mitral valve were noted. Calcium carbonate therapy was stopped
class 4, which had been treated with prednisone and intravenous and sevelamer was begun as a phosphate binder to treat phospho-
cyclophosphamide with an apparent good response, returned to the rus values ranging from 5.4-6.2 mg/mL (1.74-2.0 mmol/L). iPTH
nephrology clinic after a 6-year absence. During that time, she had levels progressively increased, peaking at 562 pg/mL (562 ng/L);
remained asymptomatic, discontinued all medications, and did not treatment with paricalcitol, 2 ␮g per dialysis treatment, was begun.
seek medical care. Evaluation showed the following values: serum PTH level remained at 364-486 pg/mL (364-486 ng/L) for the next
creatinine, 4.1 mg/dL (362.44 ␮mol/L; estimated glomerular filtra- year. After approximately 3 years on dialysis therapy, the patient
tion rate [eGFR], 12 mL/min/1.73 m2 [0.20 mL/s/1.73 m2]), which began to notice vague pains in her hip and knee joints and legs and
previously was 1.2 mg/dL (106.1 ␮mol/L); calcium, 8.9 mg/dL back. Pain was aggravated by exercise, and overall, she felt that
(2.22 mmol/L); phosphorus, 4.8 mg/dL (1.55 mmol/L); 25- her health was declining. These symptoms gradually became
hydroxyvitamin D, 14 ng/mL (34.94 nmol/L); intact parathyroid worse during the ensuing months. Review of laboratory values for
hormone (iPTH), 432 pg/mL (432 ng/L); and hemoglobin, 9.4 the past year showed calcium values in the low-normal range,
g/dL (94 g/L). Ultrasound showed kidney size to be 9.1 and 8.7 cm. phosphorus values of 5.1-6.2 mg/dL (1.6-2.0 mmol/L), and iPTH
Urinary protein-creatinine ratio was 2.4 g/g. Urine sediment was values of 372-463 pg/mL (372-463 ng/L). Alkaline phosphatase
unremarkable. Anti–double-stranded DNA was negative and C3 level was 192 U/L, having increased from 139 U/L 1 year earlier.
and C4 levels were normal. Blood pressure was 152/82 mm Hg, Dual-energy x-ray absorptiometry showed a T score of ⫺2.3 at the
and there was mild peripheral edema. Therapy was initiated with hip. Bone biopsy showed severe osteitis fibrosa. Paricalcitol therapy
furosemide; lisinopril; ergocalciferol, 50,000 units every 2 weeks; was intensified, and iPTH values stabilized near 200 pg/mL (200
darbepoetin; and calcium carbonate, 500 mg, with meals. After 3 ng/L) during the next 6 months, with a marked decrease in skeletal
months of treatment, 25-hydroxyvitamin D level increased to 26 symptoms occurring after 3 months of intensified paricalcitol
ng/mL (64.90 nmol/L), and iPTH level decreased to 296 pg/mL therapy.
(296 ng/L). During the next 6 months, serum creatinine level
slowly increased, and hemodialysis therapy was initiated when INTRODUCTION
serum creatinine level reached 8.7 mg/dL (769.1 ␮mol/L; eGFR, 5
mL/min/1.73 m2 [0.08 mL/s/1.73 m2]) through an arteriovenous Our view of the consequences of abnormal bone
fistula. While receiving hemodialysis, the patient remained clini- and mineral metabolism in the setting of chronic
cally well during the next 2 years and was placed on the transplant kidney disease (CKD) has evolved from focusing
entirely on the skeleton to a broader perspective in
which not only are bone abnormalities observed and
From the Division of Nephrology, Saint Louis University, St require therapy, but these abnormalities also seem to
Louis, MO.
Received July 7, 2011. Accepted in revised form January 4, be related to cardiovascular disease (specifically vas-
2012. Originally published online April 23, 2012. cular calcification) and are implicated in increased
Address correspondence to Kevin J. Martin, MB, BCh, Division mortality risk. Accordingly, the term renal osteodystro-
of Nephrology, Saint Louis University Medical Center, Division of phy can be considered to represent the skeletal abnor-
Nephrology (9-FDT), 3635 Vista Ave, St Louis, MO 63110. E-mail: malities, whereas the broader term CKD–mineral and
martinkj@slu.edu
© 2012 by the National Kidney Foundation, Inc. bone disorder (CKD-MBD) spans the full spectrum of
0272-6386/$36.00 the consequences of abnormal bone and mineral me-
doi:10.1053/j.ajkd.2012.01.027 tabolism in people with CKD.1 Secondary hyperpara-

308 Am J Kidney Dis. 2012;60(2):308-315


Long-term Management of CKD-MBD

thyroidism is a major part of the MBD spectrum and Box 1. Principles of Therapy of Hyperparathyroidism in ESRD
has been one focus of research to understand the Consider calcium balance
factors that generate and maintain hyperparathyroid-
● Control hyperphosphatemia/phosphate retention
ism.2 ● Correct hypocalcemia (if present)
To date, it is well established that phosphate reten- ● Consider vitamin D sterols
tion and abnormalities in vitamin D metabolism result
Consider use of calcimimetic therapy or parathyroidec-
from CKD and, either directly or by inducing changes tomy in select patients
in serum calcium levels, stimulate the growth and
Abbreviation: ESRD, end-stage renal disease.
activity of the parathyroid glands. The resultant high
levels of PTH in blood can affect the bone, causing abnormalities in vitamin D metabolism, and, finally,
osteitis fibrosa (high-turnover bone disease), manifest consider the use of a calcimimetic agent or parathyroid-
with demineralization of bone, predisposition to frac- ectomy to directly control PTH secretion at the level
tures, and not infrequently, bone pain. In addition, the of the parathyroid gland. These measures are under-
consequences of high PTH levels may extend to taken in the context of minimizing the calcium burden
nonskeletal tissue, contributing to abnormalities in to the patient to prevent excessive calcium loading,
multiple systems throughout the body, most notably which may aggravate the progression of vascular
cardiovascular disease, with manifestations including calcification.
left ventricular hypertrophy, vascular calcification, Just as there is a wide spectrum of clinical and
and hypertension. biochemical abnormalities in patients with CKD, there
Abnormalities in vitamin D metabolism occur early is also a wide spectrum of therapeutic approaches to
in the course of CKD, with 1,25-dihydroxyvitamin D the patient with CDK-MBD, as illustrated in Fig 1.
levels having been shown to decrease progressively, Thus, some clinicians favor a vitamin D–based ap-
thereby promoting increases in PTH levels. One rea- proach with calcimimetic as add-on therapy, whereas
son for the decrease in 1,25-dihydroxyvitamin D others favor the use of a low-dose vitamin D sterol as
levels is phosphate retention, which may directly background for calcimimetic-based therapy. The de-
suppress the activity of 1-hydroxylase or may act tails of individual patients likely determine the appro-
indirectly by increasing levels of fibroblast growth priate approach, as well as issues of treatment adher-
factor 23 (FGF-23), which in turn will decrease 1␣- ence, cost, and comorbid conditions. There presently
hydroxylase activity and increase 24-hydroxylase ac- are no definitive long-term data to compare these
tivity.3,4 Compensatory efforts to maintain the concen- approaches. These principles have been derived mostly
tration of 1,25-dihydroxyvitamin D are compromised from observations in patients with end-stage renal
because 25-hydroxyvitamin D levels have been shown disease (ESRD), but to some degree may be applied to
to be reduced in many people with CKD, possibly the earlier stages of CKD when the abnormalities in
contributing to the inability of the damaged kidney to bone and mineral homeostasis begin. Calcimimetic
augment 1,25-dihydroxyvitamin D production when therapy is not approved for use in patients with CKD
needed, even in the presence of hyperparathyroid- not on dialysis therapy and therefore is not recom-
ism.5 The accumulation of N-terminally truncated mended for such patients at this time.
PTH fragments also may decrease 1␣-hydroxylase In the patient under discussion, the opportunity
activity.6 Ultimately, when kidney disease is ad- to begin therapy early in the course of CKD was not
vanced, 1,25-dihydroxyvitamin D production may be possible because of loss of follow-up. However, in
limited most by the marked decrease in renal parenchy- general, therapy should be undertaken at this time if
mal mass. possible. Because clinically measurable hyperphos-
phatemia does not occur until GFR is ⬍20 mL/min/
PRINCIPLES OF THERAPY 1.73 m2 (⬍0.33 mL/s/1.73 m2), the use of phos-
As the pathophysiology of secondary hyperparathy- phate binders is not part of routine clinical practice.
roidism has been elucidated over the past, our under- However, phosphate binders perhaps should be
standing of this pathophysiology has provided a ratio- considered in light of extensive experimental obser-
nal framework for therapy in which the abnormalities vations in animals, in which dietary phosphorus
noted to contribute to hyperparathyroidism are di- restriction in proportion to the decrease in GFR was
rectly targeted with therapeutic efforts. As listed in shown to be effective in preventing the develop-
Box 1, the principles of therapy for hyperparathyroid- ment of secondary hyperparathyroidism.7,8 It is
ism are to control phosphorus levels and prevent possible that in the future, measurements of FGF-23
phosphate retention, correct hypocalcemia if it is might be useful in early CKD because these values
present because this is the major driver of PTH may increase even before elevations in PTH levels
secretion, supply vitamin D sterols to counteract the are seen.9 However, current practice guidelines

Am J Kidney Dis. 2012;60(2):308-315 309


Martin and González

range, with the opinion of the guideline develop-


Vitamin D Sterol ment team that calcium levels preferably should be
in the lower half of this range. It also was recom-
Calcimimec mended that phosphate values be controlled to
3.5-5.5 mg/dL (1.1-1.8 mmol/L), although the up-
per level of this range exceeded the typical labora-
tory normal range. The more recent KDIGO clinical
practice guideline sought to limit recommendations
Non-calcium based binders to those with convincing data to support them.
Accordingly, these guidelines broaden the range for
Calcium based binders iPTH to 2-9 times the upper limit of normal and
continue to suggest that serum calcium levels be
maintained within the normal range, but advocated
Figure 1. The spectrum of therapies for chronic kidney trying to move phosphorus values toward the nor-
disease–mineral and bone disorder (CKD-MBD). This reflects
the strategies of principally using vitamin D sterols together mal range, if achievable. Of note, these are all level
with non– calcium-based phosphate binders and possibly a 2, or weak, clinical guidelines and cautiously worded
low-dose calcimimetic compared with an alternate strategy of as suggestions rather than recommendations. The
using calcimimetic therapy together with calcium-based phos-
phate binders and low-dose vitamin D sterols. most notable change between the KDOQI and
KDIGO guidelines is the difference in iPTH target
recommend measurement of PTH in patients with level. In the patient under consideration, these
GFR ⬍60 mL/min/1.73 m2 (⬍1.0 mL/s/1.73 m2) laboratory values were mostly within the recom-
and, if values are elevated, suggest assessing the mended ranges, except to note that values for iPTH
level of vitamin D nutrition with measurement of exceeded the KDOQI recommendations, but were
25-hydroxyvitamin D. within the KDIGO target.
When the patient returned to the nephrology clinic, One major limitation with regard to PTH target
25-hydroxyvitamin D levels were low and hyperpara- levels is the actual measurement of PTH, reflecting a
thyroidism was established. Administration of ergocal- lack of PTH standards for assays, as well as the fact
ciferol, as currently is recommended, resulted in only that many of the commercially available iPTH assays
a modest increment of 25-hydroxyvitamin D levels, show varying degrees of cross-reactivity to N-
although iPTH levels decreased somewhat. This result terminally truncated PTH peptides that circulate and
is similar to previous findings in a larger group of may accumulate in patients with advanced kidney
patients.10,11 Use of oral active vitamin sterols might disease, particularly patients treated with dialysis.14,15
have been considered in this patient, but eGFR was The variable cross-reactivity of PTH assays to these
low and decreasing progressively and they were not fragments that can accumulate in CKD renders the
prescribed. recommendation as a multiple of the upper limit of
In applying these principles of therapy to patients normal as conceptually problematic (in the opinion of
with advanced CKD, including patients with ESRD the authors) because of the amplification of the contri-
being treated with dialysis, clinical practice guide-
lines for therapeutic biochemical targets, initially Table 1. Mineral Metabolism Treatment Goals: KDOQI and
introduced by the National Kidney Foundation’s KDIGO Targets in CKD Stage 5
KDOQI (Kidney Disease Outcomes Quality Initia- Parameter KDOQI Goal KDIGO Goal
tive) guidelines and later modified and updated by
the KDIGO (Kidney Disease: Improving Global iPTH (pg/mL) 150-300 2-9 ⫻ upper limit
Outcomes) guidelines, can serve as a useful re- of normal
source. The later iteration of the CKD-MBD guide- Ca (mg/dL) 8.4-10.2a 8.4-10.2
lines, published in 2009, was far more transparent P (mg/dL) 3.5-5.5 Toward normal
regarding the data limitations inherent in this Ca ⫻ P (mg2/dL2) ⬍55 Not specified
field.12,13 Both the KDOQI and KDIGO clinical Note: Conversion factors for units: calcium in mg/dL to mmol/L,
guidelines for therapy are summarized in Table 1. ⫻0.2495; phosphorus in mg/dL to mmol/L, ⫻0.3229. No conver-
Initial recommendations by KDOQI were to target sion necessary for iPTH in pg/mL and ng/L.
iPTH levels in dialysis patients of 150-300 pg/mL Abbreviations: Ca, calcium; Ca ⫻ P, calcium-phosphorus prod-
uct; CKD, chronic kidney disease; iPTH, intact parathyroid hor-
(150-300 ng/L) because the prevailing opinion at
mone; KDIGO, Kidney Disease: Improving Global Outcomes;
the time was that this range was associated with KDOQI, Kidney Disease Outcomes Quality Initiative; P, phospho-
relatively normal bone turnover. Levels of serum rus.
calcium were recommended to be in the normal a
Lower half of range preferable.

310 Am J Kidney Dis. 2012;60(2):308-315


Long-term Management of CKD-MBD

bution of these PTH peptides to measured values strued as indicating cause and effect. It is important
caused by the accumulation of these peptides in the to note that such studies should be considered with
absence of kidney function. Thus, an assay that mea- the realization that there is considerable potential
sures PTH fragments will result in markedly higher for residual confounding and limitations of the
values as GFR decreases compared with results of an artificial constraints induced by the statistical mod-
assay that does not recognize these fragments. Thus, eling in these types of studies. Certain patient
the range recommended by KDIGO includes the wid- groups may require slightly higher PTH values, so
est range possible considering all the PTH assays that that any recommendations with regard to the target
are available. However, such a range would include range for PTH should be considered “soft” and
values indicative of moderate to severe hyperparathy- other factors should be considered according to
roidism for many of the assays that are in use at the clinical circumstances. Recommendations of appro-
present time. KDIGO guidelines also suggest evalua- priate PTH values in the earlier stages of CKD are
tion of changes in PTH levels over time as an additive entirely opinion based at this time.
consideration that may be clinically useful. The issue
is complicated further because iPTH values are not a CONTROL OF HYPERPHOSPHATEMIA
precise index of the underlying bone histology. These Physiologically, it is intuitive that it should be
issues are reflected in the uncertainty of an appropri- important to control hyperphosphatemia, reflecting
ate range for PTH measurements in patients with potential consequences for bone and mineral metabo-
ESRD. lism and for the cardiovascular system, as shown in
In the patient discussed in the clinical vignette, it Fig 2. Thus, hyperphosphatemia can decrease cal-
was unclear whether the bone pain might represent citriol production, cause resistance to the actions of
hyperparathyroidism or could reflect microfractures calcitriol and PTH, and lead directly to increased PTH
as a result of osteoporosis from prior corticosteroid secretion and stimulate parathyroid growth. In addi-
therapy and/or postmenopausal status or be due to tion, hyperphosphatemia is associated with an in-
another cause altogether. Although the PTH values creased risk of metastatic calcification, including cal-
might be considered within the KDIGO target, the cification in the vasculature,19-21 and has been shown
increased alkaline phosphatase level was suggestive to be associated, as an independent risk factor, with
of high-turnover bone. For this reason, a bone biopsy increased mortality risk.16-18,22
was performed, showed severe hyperparathyroid bone For these reasons, it is reasonable to try to
disease, and indicated that intensified therapy for control hyperphosphatemia in patients on dialysis
hyperparathyroidism was required. Such therapy was therapy. Phosphorus control is accomplished best
successful in decreasing PTH values to the lower part by addressing both intake and clearance. Accord-
of recommended ranges and achieved marked im- ingly, a multifaceted approach could encompass
provement in symptoms. Bone biopsy is not done dietary phosphorus restriction, use of phosphate
often in clinical practice, but is useful in a case such as binders to complex phosphate in the intestine to
this, for which the diagnosis may be unclear and bone prevent its absorption, adequate hemodialysis or
biopsy can be very useful in guiding the approach to peritoneal dialysis to facilitate phosphorus re-
therapy. moval, and efforts to control secondary hyperpara-
It is important to emphasize that the recom- thyroidism to try to minimize phosphate efflux from
mended values for PTH initially were focused on bone as a contributor to hyperphosphatemia. The
the effects of PTH on bone; however, it now is quantitative contribution of each of these ap-
recognized that measurements of PTH alone are not proaches is not well understood at the present time,
a precise predictor of the state of bone turnover, and there are insufficient data to show that these
particularly in patients receiving dialysis. More efforts will have a favorable influence on mortality.
recent studies have looked to find the relationship Phosphate binders are an essential part of the therapy
between PTH values and patient outcome, with and have evolved over many years, from the use of
large retrospective epidemiologic studies con- aluminum compounds to the use of calcium-contain-
ducted in the United States, Europe, and South ing phosphate binders, such as calcium carbonate or
America all showing that the lowest mortality, most calcium acetate, to magnesium carbonate, sevelamer
of which is cardiovascular in nature, is seen with hydrochloride, and later, to sevelamer carbonate and
iPTH values within the range recommended by the lanthanum carbonate, and in the future, to other phos-
initial KDOQI group, that is, iPTH values of 150- phate binders that are currently in development, as
300 pg/mL (150-300 ng/L).16-18 Such epidemio- listed in Table 2. The aluminum-based phosphate
logic observations indicate only an association of binders largely have been abandoned in the United
PTH level with outcome and should not be con- States because of the risk of aluminum toxicity, but

Am J Kidney Dis. 2012;60(2):308-315 311


Martin and González

↑ Pi
↓ Ca++ ↓ Calcitriol
PTH Resistance Calcitriol Resistance
Figure 2. The consequences of hyper-
phosphatemia. Elevations in serum phos-
↑ PTH Secretion phate levels can affect end-organ response
to parathyroid hormone (PTH) and calcitriol,
can directly stimulate PTH secretion and
↑ Parathyroid Cell Growth parathyroid growth, and are associated with
increased fibroblast growth factor 23 (FGF-
23) levels and increased risk of vascular
Increased Risk of calcification and death. Abbreviations: Ca,
Metastatic Calcification ↑ Mortality ↑ FGF-23 calcium; Pi, inorganic phosphorus.

are still used throughout the world. If aluminum- patients treated with calcium acetate compared with
based phosphate binders are used, it is reasonable to sevelamer acetate.27 Nonetheless, caution regarding
have continued surveillance of serum aluminum the use of high calcium intake is advised, especially in
levels. the presence of vascular calcification. Accordingly, if
Calcium-containing phosphate binders are widely calcium-containing phosphate binders are used, ac-
used, but in certain settings may be associated with cording to the KDOQI recommendations, the amount
the development and maintenance of hypercalcemia. should be limited to no more than 1.5 g of elemental
Several, but not all, trials comparing calcium- versus calcium per day. No specific limit was suggested by
non–calcium-containing binders suggest that in- KDIGO, but this would appear to be a reasonable
creased calcium absorption may contribute to progres- limit unless it is desirable to promote a positive
sion of vascular calcification, as suggested with the calcium balance. In the patient under discussion,
Treat-to-Goal and other studies.23,24 This issue re- because of mitral valve calcification noted on pretrans-
mains somewhat controversial.25,26 Thus, the CARE plant cardiac evaluation, calcium carbonate was re-
2 (Calcium Acetate Renagel Evaluation-2) Study com- duced and sevelamer was prescribed. Lanthanum car-
pared prevalent hemodialysis patients randomly as- bonate also might have been considered to limit the
signed to calcium acetate or sevelamer hydrochloride calcium load.
treatment, with both groups receiving atorvastatin to In theory, it seems intuitive that adding to the
lower cholesterol levels. Although there was a high substrate that precipitates in the vasculature (calcium
dropout rate, no difference in progression of vascular and phosphorus) would be undesirable because the
calcification was observed.26 Similarly, the BRiC more vascular calcification that is present, the worse
(Phosphate Binder Impact on Bone Remodeling and the survival probability for the patient.28 To further
Coronary Calcification) study did not show a differ- explore this theory, several studies have compared
ence in rate of progression of vascular calcification in survival in individuals prescribed calcium-containing

Table 2. Phosphate Binders

Type Cation Content Advantages Disadvantages

CaCO3 200 mg Ca in 500 mg CaCO3 Inexpensive; widely available 1 Ca


CaAc 169 mg Ca in 667 mg CaAc Inexpensive; widely available 1 Ca; pill burden
Ca citrate 200 mg Ca in 950 mg Ca citrate — 1 Ca; 1Al absorption
Sevelamer HCl None No Ca; 2 LDL GI side effects; cost; pill burden
Sevelamer carbonate None No Ca; 2 LDL GI side effects; cost; pill burden
Mg/CaCO3 114 mg Mg in 400 MgCO3; 80 2 Ca load Mg accumulation; not widely available
mg Ca in 200 mg CaCO3
MgCO3/CaAc 60 mg Mg in 235 MgCO3; 110 2 Ca load Mg accumulation; not widely available
mg Ca in 435 mg CaAc
La2(CO3)3 250, 500, 1,000 mg La/tablet Potent Chewable; GI side effects
Al(OH)3 100-200 mg Al/tablet Potent Al toxicity
Abbreviations: Al, aluminum; Al(OH)3, aluminum hydroxide; Ca, calcium; CaAc, calcium acetate; CaCO3, calcium carbonate; GI,
gastrointestinal; HCl, hydrochloride; La2(CO3)3, lanthanum carbonate; LDL, low-density lipoprotein cholesterol; Mg, magnesium;
MgCO3, magnesium carbonate.

312 Am J Kidney Dis. 2012;60(2):308-315


Long-term Management of CKD-MBD

or non–calcium-containing phosphate binders. The gesting that the effects of paricalcitol on increasing
Dialysis Clinical Outcomes Revisited (DCOR) Study, phosphate absorption are rather small.36 In contrast,
conducted in 2,103 prevalent dialysis patients, ran- in a crossover study in which paricalcitol was com-
domly assigned participants to either sevelamer or pared with doxercalciferol, there were more episodes
calcium-based phosphate binders, with primary study of hyperphosphatemia with doxercalciferol than with
end points of all-cause mortality and cardiovascular paricalcitol, suggesting that there are potential differ-
death. Overall, the study was negative and was com- ences between these vitamin D sterols that may be-
plicated by a nearly 50% dropout rate, although a come relevant to their use. In patients with CKD not
nonprespecified subgroup analysis showed a benefit on dialysis therapy, the oral forms of these active
of uncertain methodologic validity in patients 65 vitamin D sterols may be used, but there are no
years and older,29 the KDIGO CKD-MBD work group definitive studies of the relative efficacy or toxicity. A
states that this subgroup analysis “could be, at best, substantial body of evidence in experimental animals
considered hypothesis generating and should be inter- confirms that there are important differences between
preted with extreme caution.”13(pS55) These results are the various vitamin D sterols, particularly dramatic in
contrasted with those in the RIND (Reversible Isch- terms of vascular calcification, as shown in the work
emic Neurological Deficit) Study, which examined of Mizobuchi et al,37 in which calcitriol and doxercal-
this issue in 127 incident dialysis patients. In RIND, ciferol appear to be associated with severe vascular
although the numbers of patients were considerably calcification, whereas paricalcitol was not. Compa-
smaller, use of the non–calcium-containing phosphate rable data in patients are not available at the present
binder was associated with decreased progression of time.
vascular calcification over time.23 Follow-up of these Because 25-hydroxyvitamin D levels are low in
patients (albeit uncontrolled) over the subsequent sev- many patients with advanced kidney disease, an impor-
eral years showed an apparent survival benefit for tant clinical question is whether this needs to be
patients treated with sevelamer,30 raising the issue supplemented, especially in patients who are receiv-
that the DCOR results potentially may not fully reflect ing active vitamin D sterols. In this regard, one should
the benefits of the non–calcium-containing binder consider the observations of Wolf et al,38 who found
because of the use of prevalent patients, and calling to that mortality in the first 90 days of dialysis therapy
attention that perhaps this question should be exam- appears to stratify by 25-hydroxyvitamin D levels,
ined earlier in the course of CKD. and if these patients were given an active vitamin D
sterol, this stratification was abolished. These observa-
VITAMIN D STEROLS tions suggest that administration of an active vitamin
Several active vitamin D sterols are available for D sterol may be sufficient to achieve the apparent
use in patients to control hyperparathyroidism; in the mortality benefit shown with this therapy. One also
United States, these agents include calcitriol, parical- should consider results of studies in 1␣-hydroxylase
citol, and doxercalciferol. Use of vitamin D sterols is knockout animals, in which calcitriol administration
an important part of the therapy for hyperparathyroid- has been shown to have beneficial effects on cardiac
ism, not only because of the efficacy in controlling hypertrophy that conceivably may be relevant to this
PTH secretion, but also because of observations that issue and suggesting that the circulating hormonal
use of vitamin D sterols appears to be associated with form of active vitamin D may have important effects
a survival advantage, first noticed in patients on on the cardiovascular system.39 However, cholecalcif-
hemodialysis therapy and later in patients with erol supplementation may decrease the amount of
CKD.31-35 The vitamin D analogue paricalcitol was active vitamin D sterol required and also may de-
introduced and developed to try to obtain more selec- crease the amount of erythropoietic-stimulating agent
tive action on the parathyroid gland while minimizing required.40
the effects of active vitamin D sterols to cause hyper- If hyperparathyroidism cannot be controlled with
calcemia or hyperphosphatemia (the main manifesta- adequate control of hyperphosphatemia, correction of
tions of vitamin D toxicity) and was supported in hypocalcemia, and the use of vitamin D sterols, an
detailed preclinical studies. The vitamin D2 prohor- additional approach in patients with ESRD is to di-
mone doxercalciferol, because it also was based on rectly target PTH secretion from the parathyroid gland
the D2 structure, also appeared to have the potential by the use of a calcimimetic agent or to consider
for decreased hypercalcemia and is widely used. Di- parathyroidectomy in selected patients. The calcimi-
rect head-to-head randomized studies in patients are metic cinacalcet was introduced for this purpose and
limited at the present time. In one observational study, by targeting the calcium receptor, can decrease PTH
we noted that withdrawal of paricalcitol therapy does levels effectively, significantly decrease serum cal-
not significantly change serum phosphate values, sug- cium levels, and achieve a slight decrease in serum

Am J Kidney Dis. 2012;60(2):308-315 313


Martin and González

phosphate values if taken in conjunction with the data and many therapeutic challenges to achieving
other therapeutic measures.41,42 Recent data for 360 effective and sustained control of MBD. Given the
patients in the ADVANCE (Action in Diabetes and limited overall success with addressing disease in
Cardiovascular Disease: Preterax and Diamicron patients with kidney failure, at which time much of
Modified Release Controlled Evaluation) trial suggest the milieu promoting MBD has been present for years
that this approach is associated with a trend toward or even decades, it becomes important to try to ad-
decreasing the progression of vascular and valvular dress these problems earlier in the course of CKD.2,50
calcification and was significant for aortic valvular Possibly, if addressed early, improved and more con-
calcification.43 Because this agent is administered stant control of MBD during kidney failure might be
orally, one has to ensure patient adherence to the facilitated. Further ongoing research and future devel-
regimen, which may be somewhat problematic, as opments of new therapeutic agents may facilitate our
shown by the studies of Gincherman et al.44 If medi- efforts on this important clinical problem.
cal therapy cannot achieve sustained control of hyper-
parathyroidism, consideration should be given to para- ACKNOWLEDGEMENTS
thyroidectomy. This usually is reserved for patients Support: None.
with severe hyperparathyroidism for whom medical Financial Disclosure: Dr Martin has served as a consultant for
Abbott, Cytochroma, Genzyme, and KAI Pharmaceuticals and as a
therapy cannot be tolerated.
speaker for Abbott and Genzyme. Dr González declares that she
A recent consideration for the therapeutic approach has no relevant financial interests.
to this important clinical problem relates to FGF-23
levels. Thus, vitamin D therapy might be expected to REFERENCES
directly increase FGF-23 levels. However, because 1. Moe SM, Drueke T, Lameire N, Eknoyan G. Chronic kidney
FGF-23 comes from bone, reductions in bone cell disease-mineral-bone disorder: a new paradigm. Adv Chronic
activity might be expected to decrease FGF-23 levels. Kidney Dis. 2007;14(1):3-12.
2. Martin KJ, Gonzalez EA. Metabolic bone disease in chronic
This issue may become important because epidemio- kidney disease. J Am Soc Nephrol. 2007;18(3):875-885.
logic studies indicate that high FGF-23 levels appear 3. Perwad F, Zhang MY, Tenenhouse HS, Portale AA. Fibro-
to be associated with increased mortality for patients blast growth factor 23 impairs phosphorus and vitamin D metabo-
on hemodialysis therapy.45,46 This observation also lism in vivo and suppresses 25-hydroxyvitamin D-1alpha-hydroxy-
appears to extend to patients with earlier stages of lase expression in vitro. Am J Physiol. 2007;293(5):F1577-F1583.
4. Shimada T, Hasegawa H, Yamazaki Y, et al. FGF-23 is a
CKD.47 Although the exact mechanism for this mortal- potent regulator of vitamin D metabolism and phosphate homeosta-
ity association is not known, recent studies show sis. J Bone Miner Res. 2004;19(3):429-435.
important direct effects of FGF-23 on the heart, par- 5. Nykjaer A, Dragun D, Walther D, et al. An endocytic
ticularly in causing left ventricular hypertrophy.48 At pathway essential for renal uptake and activation of the steroid
the very least, FGF-23 appears to be an important 25-(OH) vitamin D3. Cell. 1999;96(4):507-515.
6. Usatii M, Rousseau L, Demers C, et al. Parathyroid hormone
biomarker for adverse outcomes, and it may be caus- fragments inhibit active hormone and hypocalcemia-induced
ative for inducing left ventricular hypertrophy. It may 1,25(OH)2D synthesis. Kidney Int. 2007;72(11):1330-1335.
become useful in the future to consider what happens 7. Slatopolsky E, Caglar S, Gradowska L, Canterbury J, Reiss
to FGF-23 during our therapeutic efforts and perhaps E, Bricker NS. On the prevention of secondary hyperparathyroid-
ism in experimental chronic renal disease using “proportional
develop approaches to modify its production or ac-
reduction” of dietary phosphorus intake. Kidney Int. 1972;
tion. Calcimimetic therapy would be expected to 2(3):147-151.
decrease FGF-23 by decreasing PTH levels, whereas 8. Slatopolsky E, Caglar S, Pennell JP, et al. On the pathogene-
vitamin D sterols would be expected to increase sis of hyperparathyroidism in chronic experimental renal insuffi-
FGF-23 production by a direct effect on gene transcrip- ciency in the dog. J Clin Invest. 1971;50(3):492-499.
9. Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor
tion. However, in one clinical trial that compared 23 is elevated before parathyroid hormone and phosphate in
cinacalcet plus fixed-dose active vitamin D sterol chronic kidney disease. Kidney Int. 2011;79(12):1370-1378.
therapy versus active vitamin D sterol therapy alone 10. Al-Aly Z, Qazi RA, Gonzalez EA, Zeringue A, Martin KJ.
to control hyperparathyroidism, there appeared to be a Changes in serum 25-hydroxyvitamin D and plasma intact PTH
greater decrease in FGF-23 levels in the cinacalcet levels following treatment with ergocalciferol in patients with
CKD. Am J Kidney Dis. 2007;50(1):59-68.
arm.49 Further studies are required of the conse- 11. Zisman AL, Hristova M, Ho LT, Sprague SM. Impact of
quences of changes in FGF-23 levels with the current ergocalciferol treatment of vitamin D deficiency on serum parathy-
approach to treatment of abnormalities in CKD- roid hormone concentrations in chronic kidney disease. Am J
MBD. Nephrol. 2007;27(1):36-43.
Much of our approach to the treatment of mineral 12. National Kidney Foundation. K/DOQI Clinical Practice
Guidelines for Bone Metabolism and Disease in Chronic Kidney
and bone disorders in CKD has been in patients who Disease. Am J Kidney Dis. 2003;42(4)(suppl 3):S1-S201.
are treated with dialysis, and despite decades of clini- 13. Kidney Disease: Improving Global Outcomes (KDIGO)
cal experience, there remains a paucity of high-quality CKD-MBD Work Group. KDIGO clinical practice guideline for

314 Am J Kidney Dis. 2012;60(2):308-315


Long-term Management of CKD-MBD

the diagnosis, evaluation, prevention, and treatment of chronic 31. Kovesdy CP, Ahmadzadeh S, Anderson JE, Kalantar-Zadeh
kidney disease-mineral and bone disorder (CKD-MBD). Kidney K. Association of activated vitamin D treatment and mortality in
Int. 2009;76(Suppl 113):S1-S130. chronic kidney disease. Arch Intern Med. 2008;168(4):397-403.
14. Martin KJ, Gonzalez EA. Parathyroid hormone assay: prob- 32. Shoben AB, Rudser KD, de Boer IH, Young B, Kestenbaum
lems and opportunities. Pediatr Nephrol. 2007;22(10):1651-1654. B. Association of oral calcitriol with improved survival in nondia-
15. Souberbielle JC, Boutten A, Carlier MC, et al. Inter-method lyzed CKD. J Am Soc Nephrol. 2008;19(8):1613-1619.
variability in PTH measurement: implication for the care of CKD 33. Teng M, Wolf M, Lowrie E, Ofsthun N, Lazarus JM,
patients. Kidney Int. 2006;70(2):345-350. Thadhani R. Survival of patients undergoing hemodialysis with
16. Floege J, Kim J, Ireland E, et al. Serum iPTH, calcium and paricalcitol or calcitriol therapy. N Engl J Med. 2003;349(5):446-
phosphate, and the risk of mortality in a European haemodialysis 456.
population. Nephrol Dial Transplant. 2011;26(6):1948-1955. 34. Teng M, Wolf M, Ofsthun MN, et al. Activated injectable
17. Kalantar-Zadeh K, Kuwae N, Regidor DL, et al. Survival vitamin D and hemodialysis survival: a historical cohort study.
predictability of time-varying indicators of bone disease in mainte- J Am Soc Nephrol. 2005;16(4):1115-1125.
nance hemodialysis patients. Kidney Int. 2006;70(4):771-780. 35. Tentori F, Albert JM, Young EW, et al. The survival advan-
tage for haemodialysis patients taking vitamin D is questioned:
18. Naves-Diaz M, Passlick-Deetjen J, Guinsburg A, et al.
findings from the Dialysis Outcomes and Practice Patterns Study.
Calcium, phosphorus, PTH and death rates in a large sample of
Nephrol Dial Transplant. 2009;24(3):963-972.
dialysis patients from Latin America. The CORES Study. Nephrol
36. Martin KJ. When is vitamin D contraindicated in dialysis
Dial Transplant. 2011;26(6):1938-1947.
patients? Semin Dial. 2009;22(3):247-249.
19. Kendrick J, Chonchol M. The role of phosphorus in the 37. Mizobuchi M, Finch JL, Martin DR, Slatopolsky E. Differ-
development and progression of vascular calcification. Am J Kid- ential effects of vitamin D receptor activators on vascular calcifica-
ney Dis. 2011;58(5):826-834. tion in uremic rats. Kidney Int. 2007;72(6):709-715.
20. Roman-Garcia P, Carrillo-Lopez N, Fernandez-Martin JL, 38. Wolf M, Shah A, Gutierrez O, et al. Vitamin D levels and
Naves-Diaz M, Ruiz-Torres MP, Cannata-Andia JB. High phospho- early mortality among incident hemodialysis patients. Kidney Int.
rus diet induces vascular calcification, a related decrease in bone 2007;72(8):1004-1013.
mass and changes in the aortic gene expression. Bone. 2010;46(1): 39. Zhou C, Lu F, Cao K, Xu D, Goltzman D, Miao D.
121-128. Calcium-independent and 1,25(OH)2D3-dependent regulation of
21. Srivaths PR, Goldstein SL, Silverstein DM, Krishnamurthy the renin-angiotensin system in 1alpha-hydroxylase knockout mice.
R, Brewer ED. Elevated FGF 23 and phosphorus are associated Kidney Int. 2008;74(2):170-179.
with coronary calcification in hemodialysis patients. Pediatr Neph- 40. Matias PJ, Jorge C, Ferreira C, et al. Cholecalciferol supple-
rol. 2011;26(6):945-951. mentation in hemodialysis patients: effects on mineral metabolism,
22. Block GA, Hulbert-Shearon TE, Levin NW, Port FK. Asso- inflammation, and cardiac dimension parameters. Clin J Am Soc
ciation of serum phosphorus and calcium x phosphate product with Nephrol. 2010;5(5):905-911.
mortality risk in chronic hemodialysis patients: a national study. 41. Block GA, Martin KJ, de Francisco AL, et al. Cinacalcet for
Am J Kidney Dis. 1998;31(4):607-617. secondary hyperparathyroidism in patients receiving hemodialy-
23. Block GA, Spiegel DM, Ehrlich J, et al. Effects of sevelamer sis. N Engl J Med. 2004;350(15):1516-1525.
and calcium on coronary artery calcification in patients new to 42. Moe SM, Chertow GM, Coburn JW, et al. Achieving
hemodialysis. Kidney Int. 2005;68(4):1815-1824. NKF-K/DOQI bone metabolism and disease treatment goals with
24. Chertow GM, Burke SK, Raggi P. Sevelamer attenuates the cinacalcet HCl. Kidney Int. 2005;67(2):760-771.
progression of coronary and aortic calcification in hemodialysis 43. Raggi P, Chertow GM, Torres PU, et al. The ADVANCE
patients. Kidney Int. 2002;62(1):245-252. study: a randomized study to evaluate the effects of cinacalcet plus
25. Navaneethan SD, Palmer SC, Craig JC, Elder GJ, Strippoli low-dose vitamin D on vascular calcification in patients on hemo-
GF. Benefits and harms of phosphate binders in CKD: a systematic dialysis. Nephrol Dial Transplant. 2011;26(4):1327-1339.
review of randomized controlled trials. Am J Kidney Dis. 2009; 44. Gincherman Y, Moloney K, McKee C, Coyne DW. Assess-
54(4):619-637. ment of adherence to cinacalcet by prescription refill rates in
26. Qunibi W, Moustafa M, Muenz LR, et al. A 1-year random- hemodialysis patients. Hemodial Int. 2010;14(1):68-72.
45. Gutierrez OM, Mannstadt M, Isakova T, et al. Fibroblast
ized trial of calcium acetate versus sevelamer on progression of
growth factor 23 and mortality among patients undergoing hemodi-
coronary artery calcification in hemodialysis patients with compa-
alysis. N Engl J Med. 2008;359(6):584-592.
rable lipid control: the Calcium Acetate Renagel Evaluation-2
46. Jean G, Terrat JC, Vanel T, et al. High levels of serum fibroblast
(CARE-2) Study. Am J Kidney Dis. 2008;51(6):952-965.
growth factor (FGF)-23 are associated with increased mortality in long
27. Barreto DV, Barreto Fde C, de Carvalho AB, et al. Phos-
haemodialysis patients. Nephrol Dial Transplant. 2009;24(9):2792-2796.
phate binder impact on bone remodeling and coronary calcifica- 47. Isakova T, Xie H, Yang W, et al. Fibroblast growth factor 23
tion—results from the BRiC Study. Nephron Clin Pract. 2008; and risks of mortality and end-stage renal disease in patients with
110(4):c273-c283. chronic kidney disease. Jama. 2011;305(23):2432-2439.
28. Blacher J, Guerin AP, Pannier B, Marchais SJ, London GM. 48. Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left
Arterial calcifications, arterial stiffness, and cardiovascular risk in ventricular hypertrophy. J Clin Invest. 2011;121(11):4393-4408.
end-stage renal disease. Hypertension. 2001;38(4):938-942. 49. Wetmore JB, Liu S, Krebill R, Menard R, Quarles LD.
29. Suki WN, Zabaneh R, Cangiano JL, et al. Effects of Effects of cinacalcet and concurrent low-dose vitamin D on
sevelamer and calcium-based phosphate binders on mortality in FGF-23 levels in ESRD. Clin J Am Soc Nephrol. 2010;5(1):110-
hemodialysis patients. Kidney Int. 2007;72(9):1130-1137. 116.
30. Block GA, Raggi P, Bellasi A, Kooienga L, Spiegel DM. 50. Martin KJ, Gonzalez EA. Prevention and control of
Mortality effect of coronary calcification and phosphate binder phosphate retention/hyperphosphatemia in CKD-MBD: what is
choice in incident hemodialysis patients. Kidney Int. 2007;71(5): normal, when to start, and how to treat? Clin J Am Soc Nephrol.
438-441. 2011;6(2):440-446.

Am J Kidney Dis. 2012;60(2):308-315 315

Вам также может понравиться