6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Osteoporosis in Patients with Chronic Kidney Diseases: A Systemic Review

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Chronic kidney disease (CKD) is associated with the development of mineral bone disorder (MBD), osteoporosis, and fragility fractures. Among CKD patients, adynamic bone disease or low bone turnover is the most common type of renal osteodystrophy. The consequences of CKD-MBD include increased fracture risk, greater morbidity, and mortality. Thus, the goal is to prevent the occurrences of fractures by means of alleviating CKD-induced MBD and treating subsequent osteoporosis. Changes in mineral and humoral metabolism as well as bone structure develop early in the course of CKD. CKD-MBD includes abnormalities of calcium, phosphorus, PTH, and/or vitamin D; abnormalities in bone turnover, mineralization, volume, linear growth, or strength; and/or vascular or other soft tissue calcification. In patients with CKD-MBD, using either DXA or FRAX to screen fracture risk should be considered. Biomarkers such as bALP and iPTH may assist to assess bone turnover. Before initiating an antiresorptive or anabolic agent to treat osteoporosis in CKD patients, lifestyle modifications, such as exercise, calcium, and vitamin D supplementation, smoking cessation, and avoidance of excessive alcohol intake are important. Managing hyperphosphatemia and SHPT are also crucial. Understanding the complex pathogenesis of CKD-MBD is crucial in improving one’s short- and long-term outcomes. Treatment strategies for CKD-associated osteoporosis should be patient-centered to determine the type of renal osteodystrophy. This review focuses on the mechanism, evaluation and management of patients with CKD-MBD. However, further studies are needed to explore more details regarding the underlying pathophysiology and to assess the safety and efficacy of agents for treating CKD-MBD.

          Related collections

          Most cited references181

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          The 2011 Report on Dietary Reference Intakes for Calcium and Vitamin D from the Institute of Medicine: What Clinicians Need to Know

          A new public health report on dietary intake requirements for calcium and vitamin D from the Institute of Medicine (IOM) (1), released on November 30, 2010, updates the IOM report of 1997 (2). The three central questions addressed by the report, in light of the growing body of research on these nutrients over the past 10–15 yr, are: 1) which health outcomes are influenced by vitamin D and/or calcium intake?; 2) how much calcium and vitamin D are needed to achieve desirable health outcomes?; and 3) how much is too much? This article, authored by the IOM Committee, describes the Committee’s process for meeting this charge, reviews the evidence base for both skeletal and extraskeletal outcomes examined by the Committee, summarizes the new Dietary Reference Intakes (DRIs) for these nutrients (Table 1), highlights challenges and uncertainties in the process, and summarizes future research priorities. Although this article provides a readily accessible overview of the Committee’s work, we encourage readers to review the full report (1) at www.iom.edu/vitamind for a fuller understanding of the process and the pertinent evidence base. The IOM Committee and Its Charge The IOM, at the request of agencies of the U.S. and Canadian governments, assembled a committee to update the DRIs for calcium and vitamin D based upon a rigorous and comprehensive review of the scientific data. The IOM Committee included 14 scientists with a broad range of expertise, assisted by experienced IOM staff members. The DRI process involves identification of health outcomes, or “indicators,” that are consistently and causally linked to the nutrient of interest, determination of the Estimated Average Requirement (EAR; corresponding to the median intake needs of the population), and calculation of the level of intake that would “cover” (meet) the requirements of at least 97.5% of the population [defined as the Recommended Dietary Allowance (RDA), corresponding to 2 sd above the median needs]. The DRI process also specifies the tolerable upper intake level (UL; the highest daily intake of the nutrient that is likely to pose no risk). When the evidence base is insufficient for development of the EAR/RDA, an adequate intake (AI) level may be estimated instead. Importantly, the DRIs are developed for “normal healthy persons” in the North American population (not intended for individuals with specific disease states) and are provided separately for several age and gender life-stage groups (Table 1). Other charges for the Committee were to assess the dietary intakes of calcium and vitamin D in the U.S. and Canadian populations, as well as to identify research needs. The IOM Committee further specified that, due to the unique feature of cutaneous synthesis of vitamin D and the variation in synthesis due to seasonality of solar exposure, skin pigmentation, individual genetic factors, sunscreen use, latitude, outdoor activity, and other factors, as well as concerns about sun exposure and skin cancer, the DRIs for vitamin D would be based on an assumption of minimal or no sun exposure. The Process: Health Outcomes Considered and Key Challenges The IOM Committee’s work extended from March 2009 to November 2010, including eight in-person meetings in Washington, D.C., an open public workshop, two open sessions to receive input from other scientists, and the maintenance of a public web site for stakeholder input. The Committee conducted an extensive and comprehensive review of the existing evidence on calcium and vitamin D in relation to diverse health outcomes. Two key systematic reviews used by the Committee included reports conducted by the Agency for Healthcare Research and Quality (AHRQ) in 2007 (3) and 2009 (4), providing evidence-based reviews of the research on calcium and vitamin D in relation to both skeletal and extraskeletal chronic disease outcomes. The Committee conducted its own literature review in addition to consideration of the AHRQ reports. The Committee, using a risk assessment framework, considered a wide range of chronic disease and other indicators to assess nutrient adequacy for calcium and vitamin D. Indicators that were considered and reviewed in detail in the report included: bone and skeletal health (including bone mineral content and density, fracture risk, and rickets/osteomalacia), calcium absorption and balance, measures such as serum 25-hydroxyvitamin D (25OHD) and PTH, cancer and site-specific neoplasms, cardiovascular disease, hypertension, diabetes, metabolic syndrome, falls and physical performance, autoimmune disorders, infectious diseases, neuropsychological functioning (including autism, cognition, and depression), and disorders of pregnancy. After careful consideration of the evidence, the Committee concluded that bone health was the only outcome that satisfied criteria for use as an “indicator” whereby causality was established and the available evidence on dose-response was sufficient to support its use for DRI development. In addition, serum 25OHD levels were considered to be the most useful marker of vitamin D exposure, incorporating endogenous synthesis from solar exposure, dietary intake from foods, fortified products, and/or supplements, and other factors. For cancer, cardiovascular disease, diabetes, falls, physical performance, autoimmune disorders, and other extraskeletal chronic disease outcomes, the evidence was deemed to be inconsistent, inconclusive as to causality, and insufficient to serve as a basis for DRI development. Importantly, randomized trial evidence was sparse, and few clinical trials of calcium and/or vitamin D had been done with these extraskeletal outcomes as the primary prespecified outcomes. The AHRQ systematic review of 2009 (4) had also concluded that the evidence for an association between these nutrients and extraskeletal outcomes was inconsistent and inconclusive. Key challenges included the strong interrelationship between calcium and vitamin D and the difficulty separating their effects in many studies, the small number of relevant randomized clinical trials allowing for assessment of dose-response relationships, the complexity that vitamin D is obtained not only from diet but also synthesized endogenously, and the potential for confounding in observational studies. Although measures of 25OHD were considered to be a useful marker of exposure, the Committee was cognizant of its limitations as a biomarker of effect. Correlation does not prove causation in observational studies, underscoring the need for caution in interpretation of study findings. Specific factors relevant to vitamin D are sources of potential confounding, such as obesity (due to sequestration in adipose tissue), physical activity (correlated with time outdoors and solar exposure), race/skin pigmentation, and nutritional status including supplementation practices. Reverse causation bias is also a threat to study validity if poor health curtails outdoor activities and sunlight exposure or adversely affects diet. These potential biases must be carefully considered in the interpretation of observational studies. In this regard, it should be noted that many micronutrient interventions that seemed promising in observational studies (e.g. β-carotene, vitamins C and E, folic acid, and selenium) did not withstand rigorous testing in clinical trials, and many even suggested hazards with high levels of supplementation (5,6). Bone Health: Dietary Reference Intakes and Updates since 1997 The DRIs shown in Table 1 are based on dietary requirements using bone health as an indicator. DRIs for each nutrient were predicated on intakes meeting requirements for the other nutrient. For both calcium and vitamin D, available evidence allowed for estimation of EARs and RDAs for all life-stage groups except infants (for whom AIs are provided). At the time of the 1997 report on calcium and vitamin D, evidence was insufficient for estimation of EARs and RDAs; thus, AIs were estimated for all life-stage groups. For calcium, the 2011 DRIs are based largely on the calcium content of human breast milk for infants, calcium balance studies for ages 1–50 yr, and observational and clinical trial evidence after age 50. For vitamin D, the 2011 DRIs are based primarily on the integration of bone health outcomes with evidence concerning 25OHD levels, which suggest that levels of 16 ng/ml (40 nmol/liter) meet the needs of approximately half the population (median population requirement, or EAR), and levels of at least 20 ng/ml (50 nmol/liter) meet the needs of at least 97.5% of the population (akin to the RDA). Intakes of vitamin D required to achieve these 25OHD concentrations are shown in Table 1, based on a simulation of available data across ages under conditions of minimal sun exposure. The AI in infancy is estimated to be 400 IU/d. After age 1, the RDA is estimated to be 600 IU/d for all life-stage groups except men and women aged 71 and older (for whom the RDA is 800 IU/d). The Committee did not find compelling evidence that 25OHD levels or dietary intakes above these levels were associated with greater benefit for bone health or other outcomes. The assumption of minimal or no sun exposure for estimation of these intake levels provided further safety for individuals with lower endogenous synthesis of vitamin D. The specific studies contributing to these estimates are reviewed in detail in the full report (1). The 2011 DRIs are based on much more information and higher-quality studies than were available when the reference values for these nutrients were first set in 1997, allowing for the estimation of EARs and RDAs rather than AIs. Because the old and new DRIs reflect different calculations, the figures are not directly comparable. In 1997, the AIs for vitamin D were 200 IU/d through age 50, 400 IU/d for ages 51–70, and 600 IU/d for ages 71 and older. The 2011 DRIs for vitamin D specify RDAs, with levels of 400 IU/d for infants, 600 IU/d for children and adults through age 70, and 800 IU/d for ages 71 and older. However, the 2011 DRIs for vitamin D are nonetheless lower than those proposed by some in the current literature based on higher 25OHD levels that the Committee did not find justified by the evidence. Cancer, Cardiovascular Disease, Diabetes, Infections, Autoimmune Disorders, and Other Extraskeletal Outcomes The IOM Committee concluded that the evidence that vitamin D or calcium reduced risk of nonskeletal chronic disease outcomes was inconsistent, inconclusive, and did not meet criteria for establishing cause-and-effect relationships. Randomized trial evidence was sparse, and few trials assessed these outcomes as primary prespecified endpoints. Moreover, emerging evidence suggested a curvilinear or U-shaped curve for several outcomes related to vitamin D, including cardiovascular disease, vascular calcification, falls, frailty, pancreatic cancer, and all-cause mortality (7,8,9,10,11), with the lowest risk at moderate levels and increased risk at both low and high levels of 25OHD. These studies are reviewed in detail in the report (1). Although future research may elucidate clear benefits and possibly even different requirement levels for vitamin D in relation to these nonskeletal outcomes, existing data cannot support such conclusions. Tolerable Upper Intake Levels The indicators considered in the determination of ULs included hypercalcemia, hypercalciuria, vascular and soft tissue calcification, nephrolithiasis, and, for vitamin D, emerging evidence of a U-shaped relationship for all-cause mortality, cardiovascular disease, selected cancers, falls, and fractures. The 1997 ULs for calcium were 2500 mg/d for all ages above 1 yr, whereas the ULs for calcium now range from 1000–3000 mg/d, depending on life-stage group (Table 1). For vitamin D, the ULs are now 4000 IU/d for ages 9 and older but are lower for infants and young children (Table 1). The 1997 ULs for vitamin D were 2000 IU/d for most age groups. The starting point for the current UL for vitamin D was 10,000 IU/d, because lower intakes have not been linked to hypercalcemia or acute toxicity. However, given that toxicity is not the appropriate basis for a UL that is intended to reflect long-term chronic intake and to be used for public health purposes, this value was corrected for uncertainty based on chronic disease outcomes and all-cause mortality, as well as emerging concerns about risks at serum 25OHD levels above 50 ng/ml (125 nmol/liter). Thus, the Committee followed an approach to maximize public health protection. The UL is not intended as a target intake; rather, the risk for harm begins to increase once intakes surpass this level. Serum 25OHD Levels and Screening Guidelines regarding the use of serum markers of vitamin D status for medical management of individual patients and for screening were beyond the scope of the Committee’s charge, and evidence-based consensus guidelines are not available. However, these issues should be addressed by appropriate federal agencies and professional organizations in light of the findings in this report. As noted above, the Committee recognized that serum 25OHD is a useful integrated marker of vitamin D exposure, incorporating endogenous synthesis from solar exposure, dietary intake from foods, fortified products, and/or supplements, and other factors. However, the Committee also recognized that observational studies of correlations between 25OHD and clinical outcomes are subject to confounding and do not prove causation. Concerns about inaccurate or imprecise serum 25OHD measurements are being overcome by methodological advances, frequent quality assessments, and accurate calibration tools. In contrast, serum 1,25-dihydroxyvitamin D represents a more complex endocrine parameter, regulated by calcium and PTH status and affected by kidney function. After a careful review of available literature, the Committee concluded that serum 25OHD levels of 16 ng/ml (40 nmol/liter) cover the requirements of approximately half the population, and levels of 20 ng/ml (50 nmol/liter) cover the requirements of at least 97.5% of the population. These levels will be useful to clinicians as they consider management of patients under their care. For upper levels of serum 25OHD, sparse data are available, particularly regarding long-term effects of chronically high concentrations, and a margin of safety for public health recommendations is prudent. Thus, serum 25OHD levels above 50 ng/ml (125 nmol/liter) should raise concerns among clinicians about potential adverse effects. Dietary Intake Assessments Major food sources of calcium include dairy products, selected low-oxalate vegetables, legumes, nuts, and fortified foods; for vitamin D, primary sources are fortified dairy products, fortified foods, and fatty fish. Based on national government surveys in the United States and Canada, it appears that most groups have adequate intake of calcium (as defined by intakes above the EAR), with the exception of girls aged 9–18 who have high requirements. The data underscore the need to increase calcium intake among girls in mid-to-late childhood and adolescence; in contrast, among postmenopausal women, high calcium intake from supplements may be concerning. Regarding vitamin D, average intake from foods tends to be less than 400 IU/d, but mean 25OHD levels have been above 20 ng/ml (50 nmol/liter) in representative samples. Thus, based on these data and a level of 20 ng/ml (50 nmol/liter) identified as meeting the needs of at least 97.5% of the population across all life-stage groups, it appears that the majority of the North American population currently is meeting its needs for vitamin D. Nonetheless, subgroups of individuals, particularly those with poor nutrition, those living at northerly latitudes or in institutions, or those with dark skin pigmentation may be at increased risk of not meeting their needs, especially if their 25OHD levels are below 16 ng/ml (40 nmol/liter), the level identified as the average requirement as discussed above. Uncertainties and Future Research Needs The Committee identified a large number of uncertainties surrounding the DRI values, as well as extensive research needs. A particular priority is rigorous, large-scale, randomized clinical trials to test the effects of vitamin D on skeletal and nonskeletal outcomes, as well as to identify threshold effects and possible adverse effects where present. Elucidating the biology of the diverse effects of vitamin D, as well as effects of sun exposure, adiposity, body composition, race/ethnicity, and genetic factors on these associations, is also of great importance. Conclusions The available scientific evidence supports a key role for calcium and vitamin D in skeletal health, providing a sound basis for DRIs. The evidence, however, is not yet compelling that either nutrient confers benefits for, or is causally related to, extraskeletal health outcomes. Moreover, existing evidence suggests that nearly all individuals meet their needs at intake levels (RDAs) provided in this report and, for vitamin D, at 25OHD levels of at least 20 ng/ml (50 nmol/liter) even under conditions of minimal sun exposure. Furthermore, higher levels have not been shown consistently to confer greater benefits, challenging the concept that “more is better.” The Committee finds that the prevalence of vitamin D inadequacy in the North American population has been overestimated by some groups due to the use of inappropriate cut-points that greatly exceed the levels identified in this report. Serum concentrations of 25OHD above 30 ng/ml (75 nmol/liter) are not consistently associated with increased benefit, and risks have been identified for some outcomes at 25OHD levels above 50 ng/ml (125 nmol/liter). Additional research, including large-scale, randomized clinical trials, is needed. In the meantime, however, we believe that there is an urgent clinical and public health need for consensus cut-points for serum 25OHD inadequacy to avoid problems of both undertreatment and overtreatment.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease.

            Fibroblast growth factor 23 (FGF23) regulates phosphorus metabolism and is a strong predictor of mortality in dialysis patients. FGF23 is thought to be an early biomarker of disordered phosphorus metabolism in the initial stages of chronic kidney disease (CKD). We measured FGF23 in baseline samples from 3879 patients in the Chronic Renal Insufficiency Cohort study, which is a diverse cohort of patients with CKD stage 2-4. Mean serum phosphate and median parathyroid hormone (PTH) levels were in the normal range, but median FGF23 was markedly greater than in healthy populations, and increased significantly with decreasing estimated glomerular filtration rate (eGFR). High levels of FGF23, defined as being above 100 RU/ml, were more common than secondary hyperparathyroidism and hyperphosphatemia in all strata of eGFR. The threshold of eGFR at which the slope of FGF23 increased was significantly higher than the corresponding threshold for PTH based on non-overlapping 95% confidence intervals. Thus, increased FGF23 is a common manifestation of CKD that develops earlier than increased phosphate or PTH. Hence, FGF23 measurements may be a sensitive early biomarker of disordered phosphorus metabolism in patients with CKD and normal serum phosphate levels.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD)

              (2017)
              KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD) 3 Tables and supplementary material 6 KDIGO Executive Committee 7 Reference keys 8 CKD nomenclature 9 Conversion factors 10 Abbreviations and acronyms 11 Notice 12 Foreword 13 Work Group membership 14 Abstract 15 Summary of KDIGO CKD-MBD recommendations 19 Summary and comparison of 2017 updated and 2009 KDIGO CKD-MBD recommendations 22 Chapter 3.2: Diagnosis of CKD-MBD: bone 25 Chapter 4.1: Treatment of CKD-MBD targeted at lowering high serum phosphate and maintaining serum calcium 33 Chapter 4.2: Treatment of abnormal PTH levels in CKD-MBD 38 Chapter 4.3: Treatment of bone with bisphosphonates, other osteoporosis medications, and growth hormone 39 Chapter 5: Evaluation and treatment of kidney transplant bone disease 41 Methodological approach to the 2017 KDIGO CKD-MBD guideline update 49 Biographic and disclosure information 55 Acknowledgments 56 References Tables 24 Table 1. Utility of KDOQI and KDIGO PTH thresholds for diagnostic decision making 42 Table 2. Research questions 45 Table 3. Question-specific eligibility criteria 46 Table 4. GRADE system for grading quality of evidence for an outcome 47 Table 5. Final grade for overall quality of evidence 47 Table 6. Balance of benefits and harms 47 Table 7. Implications of the strength of a recommendation 47 Table 8. Determinants of strength of recommendation Supplementary Material Appendix A. PubMed search strategy Appendix B. Summary of search and review process Table S1. Summary table of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5: study characteristics Table S2. Summary table of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5: study population characteristics Table S3. Summary table of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5: results Table S4. Summary table of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5: quality Table S5. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5 Table S6. Evidence profile of randomized controlled trials examining the treatment of CKD-MBD with bisphosphonates in CKD G3a–G5 Table S7. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5: study characteristics Table S8. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5: study population characteristics Table S9. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5: results Table S10. Summary table of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5: quality Table S11. Evidence matrix of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5 Table S12. Evidence profile of studies evaluating the ability of bone mineral density results to predict fracture or renal osteodystrophy among patients with CKD G3a–G5 Table S13. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D: study characteristics Table S14. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D: study population characteristics Table S15. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D: results Table S16. Summary table of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D: quality Table S17. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D Table S18. Evidence profile of randomized controlled trials examining the treatment of CKD-MBD with varying dialysate calcium concentration levels in CKD G5D Table S19. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders: study characteristics Table S20. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders: study population characteristics Table S21. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders: results Table S22. Summary table of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders: quality Table S23. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders Table S24. Evidence profile of randomized controlled trials examining the treatment of CKD-MBD with calcium-containing phosphate binders versus calcium-free phosphate binders Table S25. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate: study characteristics Table S26. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate: study population characteristics Table S27. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate: results Table S28. Summary table of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate: quality Table S29. Evidence matrix of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate Table S30. Evidence profile of randomized controlled trials examining the treatment of CKD-MBD with dietary phosphate Table S31. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: study characteristics Table S32. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: study population characteristics Table S33. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: results Table S34. Summary table of randomized controlled trials examining the treatment of PTH in CKD-MBD: quality Table S35. Evidence matrix of randomized controlled trials examining the treatment of PTH in CKD-MBD Table S36. Evidence profile of randomized controlled trials examining the treatment of PTH in CKD-MBD Table S37. Summary table of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis: study characteristics Table S38. Summary table of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis: study population characteristics Table S39. Summary table of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis: results Table S40. Summary table of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis: quality Table S41. Evidence matrix of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis Table S42. Evidence profile of randomized controlled trials examining the treatment of high levels of PTH with calcitriol or activated vitamin D analogs in CKD G3a–G5 not on dialysis Table S43. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D: study characteristics Table S44. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D: study population characteristics Table S45. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D: results Table S46. Summary table of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D: quality Table S47. Evidence matrix of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D Table S48. Evidence profile of randomized controlled trials examining the treatment of high levels of PTH in CKD G5D Table S49. Summary table of studies evaluating different concentrations of serum phosphate or calcium among patients with CKD G3a–G5 or G5D: study characteristics Table S50. Summary table of studies evaluating different concentrations of serum phosphate or calcium among patients with CKD G3a–G5 or G5D: study population characteristics Table S51. Summary table of studies evaluating different concentrations of serum phosphate among patients with CKD G3a–G5 or G5D: results Table S52. Summary table of studies evaluating different concentrations of serum calcium among patients with CKD G3a–G5 or G5D: results Table S53. Summary table of studies evaluating different concentrations of serum phosphate or calcium among patients with CKD G3a–G5 or G5D: quality Table S54. Evidence matrix of studies evaluating different concentrations of serum phosphate or calcium among patients with CKD G3a–G5 or G5D Table S55. Evidence profile of studies evaluating different concentrations of serum phosphate or calcium among patients with CKD G3a–G5 or G5D Supplementary material is linked to the online version of the paper at www.kisupplements.org. KDIGO Executive Committee Garabed Eknoyan, MDNorbert Lameire, MD, PhDFounding KDIGO Co-chairs Bertram L. Kasiske, MDImmediate Past Co-chair David C. Wheeler, MD, FRCPKDIGO Co-chair Wolfgang C. Winkelmayer, MD, MPH, ScDKDIGO Co-chair Ali K. Abu-Alfa, MDOlivier Devuyst, MD, PhDJürgen Floege, MDJohn S. Gill, MD, MSKunitoshi Iseki, MDAndrew S. Levey, MDZhi-Hong Liu, MD Ziad A. Massy, MD, PhDRoberto Pecoits-Filho, MD, PhDBrian J.G. Pereira, MBBS, MD, MBAPaul E. Stevens, MB, FRCPMarcello A. Tonelli, MD, SM, FRCPCAngela Yee-Moon Wang, MD, PhD, FRCPAngela C. Webster, MBBS, MM (Clin Ep), PhD KDIGO Staff John Davis, Chief Executive OfficerDanielle Green, Managing DirectorMichael Cheung, Chief Scientific OfficerTanya Green, Communications DirectorMelissa McMahan, Programs Director Reference keys Nomenclature and Description for Rating Guideline Recommendations Within each recommendation, the strength of recommendation is indicated as Level 1, Level 2, or not graded, and the quality of the supporting evidence is shown as A, B, C, or D. Grade∗ Implications Patients Clinicians Policy Level 1 “We recommend” Most people in your situation would want the recommended course of action, and only a small proportion would not. Most patients should receive the recommended course of action. The recommendation can be evaluated as a candidate for developing a policy or a performance measure. Level 2 “We suggest” The majority of people in your situation would want the recommended course of action, but many would not. Different choices will be appropriate for different patients. Each patient needs help to arrive at a management decision consistent with her or his values and preferences. The recommendation is likely to require substantial debate and involvement of stakeholders before policy can be determined. ∗ The additional category “not graded” is used, typically, to provide guidance based on common sense or when the topic does not allow adequate application of evidence. The most common examples include recommendations regarding monitoring intervals, counseling, and referral to other clinical specialists. The ungraded recommendations are generally written as simple declarative statements, but are not meant to be interpreted as being stronger recommendations than Level 1 or 2 recommendations. Grade Quality of evidence Meaning A High We are confident that the true effect lies close to that of the estimate of the effect. B Moderate The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different. C Low The true effect may be substantially different from the estimate of the effect. D Very low The estimate of effect is very uncertain, and often will be far from the truth. Current Chronic Kidney Disease (CKD) Nomenclature Used by KDIGO CKD is defined as abnormalities of kidney structure or function, present for > 3 months, with implications for health. CKD is classified based on cause, GFR category (G1–G5), and albuminuria category (A1–A3), abbreviated as CGA. Prognosis of CKD by GFR and albuminuria category Conversion Factors of Conventional Units to SI Units Conventional unit Conversion factor SI unit Calcium, total mg/dl 0.2495 mmol/l Calcium, ionized mg/dl 0.25 mmol/l Creatinine mg/dl 88.4 μmol/l Parathyroid hormone pg/ml 0.106 pmol/l Phosphate (inorganic) mg/dl 0.3229 mmol/l Note: conventional unit × conversion factor = SI unit. Abbreviations and acronyms 1,25(OH)2D 1,25-dihydroxyvitamin D 25(OH)D 25-hydroxyvitamin D AUC area under the curve bALP bone-specific alkaline phosphatase BMD bone mineral density CAC coronary artery calcification CI confidence interval CT computed tomography CV coefficient of variation DXA dual-energy X-ray absorptiometry eGFR estimated glomerular filtration rate ERT evidence review team FGF fibroblast growth factor FRAX fracture risk assessment tool GFR glomerular filtration rate GI gastrointestinal GRADE Grading of Recommendations Assessment, Development, and Evaluation HD hemodialysis HPT hyperparathyroidism HR hazard ratio iPTH intact parathyroid hormone ISCD International Society of Clinical Densitometry ITT intention-to-treat IU international unit KDIGO Kidney Disease: Improving Global Outcomes KDOQI Kidney Disease Outcomes Quality Initiative LVH left ventricular hypertrophy LVMI left ventricular mass index MRI magnetic resonance imaging OR odds ratio P1NP amino-terminal propeptide of type 1 procollagen PTH parathyroid hormone RCT randomized controlled trial ROC receiver operating characteristic SD standard deviation SHPT secondary hyperparathyroidism VDR vitamin D receptor Notice Section I: Use of the Clinical Practice Guideline This Clinical Practice Guideline Update is based upon systematic literature searches last conducted in September 2015 supplemented with additional evidence through February 2017. It is designed to assist decision making. It is not intended to define a standard of care, and should not be interpreted as prescribing an exclusive course of management. Variations in practice will inevitably and appropriately occur when clinicians consider the needs of individual patients, available resources, and limitations unique to an institution or type of practice. Health care professionals using these recommendations should decide how to apply them to their own clinical practice. Section II: Disclosure Kidney Disease: Improving Global Outcomes (KDIGO) makes every effort to avoid any actual or reasonably perceived conflicts of interest that may arise from an outside relationship or a personal, professional, or business interest of a member of the Work Group. All members of the Work Group are required to complete, sign, and submit a disclosure and attestation form showing all such relationships that might be perceived as or are actual conflicts of interest. This document is updated annually, and information is adjusted accordingly. All reported information is published in its entirety at the end of this document in the Work Group members’ Biographic and Disclosure section, and is kept on file at KDIGO. Copyright © 2017, KDIGO. Published by Elsevier on behalf of the International Society of Nephrology. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Single copies may be made for personal use as allowed by national copyright laws. Special rates are available for educational institutions that wish to make photocopies for nonprofit educational use. No part of this publication may be reproduced, amended, or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without explicit permission in writing from KDIGO. Details on how to seek permission for reproduction or translation, and further information about KDIGO’s permissions policies can be obtained by contacting Danielle Green, Managing Director, at danielle.green@kdigo.org. To the fullest extent of the law, neither KDIGO, Kidney International Supplements, nor the authors, contributors, or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein. Foreword With the growing awareness that chronic kidney disease is an international health problem, Kidney Disease: Improving Global Outcomes (KDIGO) was established in 2003 with its stated mission to “improve the care and outcomes of kidney disease patients worldwide through promoting coordination, collaboration, and integration of initiatives to develop and implement clinical practice guidelines.” When the KDIGO Clinical Practice Guideline for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD) was originally published in 2009, the Work Group acknowledged the lack of high-quality evidence on which to base recommendations. The Guideline included specific research recommendations to encourage investigators to help fill the gaps and bolster the evidence base. Multiple randomized controlled trials and prospective cohort studies have been published since the 2009 Guideline, and therefore KDIGO recognizes the need to reexamine the currency of all of its guidelines on a periodic basis. Accordingly, KDIGO convened a Controversies Conference in 2013, titled “CKD-MBD: Back to the Future,” whose objective was to determine whether sufficient new data had emerged to support a reassessment of the 2009 CKD-MBD Clinical Practice Guideline and, if so, to determine the scope of the potential revisions. Although most of the recommendations were still considered to be current, the conference identified a total of 12 recommendations for reevaluation based on new data. In addition, the conference prepared a table of additional topic questions to be considered by the guideline update Work Group. The conference noted that, in spite of the completion of several key clinical trials since the 2009 publication of the CKD-MBD guideline, large gaps of knowledge still remained, as demonstrated by the relatively small number of recommendation statements identified for reevaluation. Interested readers should refer to the conference publication for further details regarding its processes and deliberations. 1 Therefore, KDIGO commissioned an update to the CKD-MBD guideline and formed a Work Group, led by Drs. Markus Ketteler and Mary Leonard. The Work Group convened in June 2015 to review and appraise the evidence accumulated since the 2009 Guideline. The topics addressed for revision are listed in Table 2 and included issues prompted by EVOLVE post hoc analyses, which were published after the 2013 Controversies Conference. Though 8 years have passed since the 2009 CKD-MBD guideline, evidence in many areas is still lacking, which has resulted in many of the “opinion-based” recommendation statements from the original guideline document remaining unchanged. Table 2 Research questions Section 2009 rec. no. Research question Key outcomes Additional outcomes Bone quality 3.2.1 In patients with CKD G3a–G5D, what is the effect on bone quality of bisphosphonates, teriparatide, denosumab, and raloxifene? • TMV (as measured by bone biopsy) • BMD/bone mineral content • Fracture 

 4.3.4 In patients with CKD G4–G5D, what is the effect on bone quality of bisphosphonates, teriparatide, denosumab, and raloxifene? • TMV (as measured by bone biopsy) • BMD/bone mineral content • Fracture 

 3.2.2 (a) In patients with CKD G3a–G5D, how well do BMD results predict fractures?(b) In patients with CKD G3a–G5D, how well do BMD results predict renal osteodystrophy? (a) Fracture (b) TMV 

 5.5 In patients with CKD G1–G3b and transplant recipients, how well do BMD results predict fractures? • Fracture 

 5.7 In patients with CKD G4–G5 and transplant recipients, how well do BMD results predict fractures? • Fracture 

 Calcium and phosphate 4.1.1 In patients with G3a–G5 or G5D, what is the evidence for benefit or harm in maintaining serum phosphate in the normal range compared with other targets of serum phosphate in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • GFR decline • Cardiovascular and cerebrovascular events • Phosphate • Bone histology, BMD • Vascular and valvular calcification imaging • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.1.3 In patients with CKD G5D, what is the evidence for benefit or harm in using a dialysate calcium concentration between 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l) compared with other concentrations of dialysate calcium in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • Calcium • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.1.2 In patients with CKD G3a–G5D, what is the evidence for benefit or harm in maintaining serum calcium in the normal range compared with other targets of serum calcium in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • Calcium • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.1.4 In patients with CKD G3a–G5 or G5D with hyperphosphatemia, what is the evidence for benefit or harm in using calcium-containing phosphate-binding agents to treat hyperphosphatemia compared with calcium-free phosphate-binding agents in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • Phosphate • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.1.7 In patients with CKD G3a–G5D with hyperphosphatemia, what is the evidence for benefit or harm in limiting dietary phosphate intake compared with a standard diet in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • Vascular and valvular calcification • Phosphate • Bone histology, BMD • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 Vitamin D and PTH 4.2.1 In patients with CKD G3a–G5 not on dialysis with levels of intact PTH above the upper normal limit of the assay, what is the evidence for benefit or harm in reducing dietary phosphate intake or treating with phosphate-binding agents, calcium supplements, or native vitamin D in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • GFR decline • Calcium • Phosphate • Parathyroid hormone • 25-hydroxyvitamin D [25(OH)D] • 1,25-dihydroxyvitamin D [1,25(OH)2D] • Alkaline phosphatases • Bone-specific alkaline phosphatase • Bicarbonate • FGF23 • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.2.2 In patients with CKD G3a–G5 not on dialysis, in whom serum PTH is progressively rising and remains persistently above the upper limit of normal for the assay despite correction of modifiable factors, what is the evidence for benefit or harm in treating with calcitriol or vitamin D analogs compared with placebo or active control in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • LVH • Hypercalcemia • Mortality • Cardiovascular and cerebrovascular events • Calcium • Phosphate • Parathyroid hormone • 25-hydroxyvitamin D [25(OH)D] • 1,25-dihydroxyvitamin D [1,25(OH)2D] • Alkaline phosphatases • Bone-specific alkaline phosphatase • Bicarbonate • FGF23 • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA 

 4.2.4 In patients with CKD G5D, what is the evidence for benefit or harm in treating with calcitriol, vitamin D analogs, calcimimetics or combination thereof compared with placebo or active control in terms of biochemical outcomes, other surrogate outcomes, and patient-centered outcomes? • Mortality • Cardiovascular and cerebrovascular events • Fracture • Vascular and valvular calcification imaging • Calcium • Phosphate • Parathyroid hormone • 25-hydroxyvitamin D [25(OH)D] • 1,25-dihydroxyvitamin D [1,25(OH)2D] • Alkaline phosphatases • Bone-specific alkaline phosphatase • Bicarbonate • FGF23 • Bone histology, BMD • Vascular and valvular calcification imaging • Measures of GFR • Hospitalizations • Quality of life • Kidney or kidney graft failure • Fracture • Parathyroidectomy • Clinical adverse events • Growth, skeletal deformities, bone accrual • Calciphylaxis/CUA BMD, bone mineral density; CKD, chronic kidney disease; CUA, calcific uremic arteriolopathy; GFR, glomerular filtration rate; FGF23, fibroblast growth factor 23; LVH, left ventricular hypertrophy; PTH, parathyroid hormone; rec. no., recommendation number; TMV, bone turnover mineralization volume. In keeping with the standard KDIGO policy of maintaining transparency during the guideline development process and attesting to its rigor, we conducted an open public review of the draft CKD-MBD guideline update, and all feedback received was reviewed and considered by the Work Group before finalizing this guideline document for publication. The comments and suggestions greatly assisted us in shaping a final document that we felt would be as valuable as possible to the entire nephrology community. We wish to thank the Work Group co-chairs, Drs. Markus Ketteler and Mary Leonard, along with all of the Work Group members, who volunteered countless hours of their time to develop this guideline. We also thank Dr. Karen Robinson and her Evidence Review Team at Johns Hopkins University, the KDIGO staff, and many others for their support that made this project possible. David C. Wheeler, MD, FRCP Wolfgang C. Winkelmayer, MD, MPH, ScD KDIGO Co-chairs Work Group membership Work Group Co-chairs Markus Ketteler, MD, FERAKlinikum CoburgCoburg, Germany Mary B. Leonard, MD, MSCEStanford University School of MedicineStanford, CA, USA Work Group Geoffrey A. Block, MDDenver NephrologyDenver, CO, USAPieter Evenepoel, MD, PhD, FERAUniversity Hospitals LeuvenLeuven, BelgiumMasafumi Fukagawa, MD, PhD, FASNTokai University School of MedicineIsehara, JapanCharles A. Herzog, MD, FACC, FAHAHennepin County Medical CenterMinneapolis, MN, USALinda McCann, RD, CSREagle, ID, USASharon M. Moe, MDIndiana University School of MedicineRoudebush Veterans Affairs Medical CenterIndianapolis, IN, USA Rukshana Shroff, MD, FRCPCH, PhDGreat Ormond Street Hospital for ChildrenNHS Foundation Trust,London, UKMarcello A. Tonelli, MD, SM, FRCPCUniversity of CalgaryCalgary, CanadaNigel D. Toussaint MBBS, FRACP, PhDThe Royal Melbourne HospitalUniversity of MelbourneMelbourne, AustraliaMarc G. Vervloet, MD, PhD, FERAVU University Medical Center AmsterdamAmsterdam, The Netherlands Evidence Review Team Johns Hopkins University Baltimore, MD, USA Karen A. Robinson, PhD, Associate Professor of Medicine and Project DirectorCasey M. Rebholz, PhD, MPH, MS, Co-investigatorLisa M. Wilson, ScM, Project ManagerErmias Jirru, MD, MPH, Research AssistantMarisa Chi Liu, MD, MPH, Research AssistantJessica Gayleard, BS, Research AssistantAllen Zhang, BS, Research Assistant Abstract The Kidney Disease: Improving Global Outcomes (KDIGO) 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of chronic kidney disease–mineral and bone disorder (CKD-MBD) represents a selective update of the prior guideline published in 2009. This update, along with the 2009 publication, is intended to assist the practitioner caring for adults and children with CKD, those on chronic dialysis therapy, or individuals with a kidney transplant. Specifically, the topic areas for which updated recommendations are issued include diagnosis of bone abnormalities in CKD-MBD; treatment of CKD-MBD by targeting phosphate lowering and calcium maintenance, treatment of abnormalities in parathyroid hormone in CKD-MBD; treatment of bone abnormalities by antiresorptives and other osteoporosis therapies; and evaluation and treatment of kidney transplant bone disease. Development of this guideline update followed an explicit process of evidence review and appraisal. Treatment approaches and guideline recommendations are based on systematic reviews of relevant trials, and appraisal of the quality of the evidence and the strength of recommendations followed the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) approach. Limitations of the evidence are discussed, with areas of future research also presented. Keywords: bone abnormalities; bone mineral density; calcium; chronic kidney disease; CKD-MBD; dialysis; guideline; hyperparathyroidism; hyperphosphatemia; KDIGO; kidney transplantation; mineral and bone disorder; parathyroid hormone; phosphate; phosphorus; systematic review CITATION In citing this document, the following format should be used: Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Update Work Group. KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD). Kidney Int Suppl. 2017;7:1–59. Summary of KDIGO CKD-MBD recommendations∗ Updated recommendations are denoted in boxes Chapter 3.1: Diagnosis of CKD-MBD: biochemical abnormalities 3.1.1: We recommend monitoring serum levels of calcium, phosphate, PTH, and alkaline phosphatase activity beginning in CKD G3a (1C). In children, we suggest such monitoring beginning in CKD G2 (2D). 3.1.2: In patients with CKD G3a–G5D, it is reasonable to base the frequency of monitoring serum calcium, phosphate, and PTH on the presence and magnitude of abnormalities, and the rate of progression of CKD (Not Graded). Reasonable monitoring intervals would be: • In CKD G3a–G3b: for serum calcium and phosphate, every 6–12 months; and for PTH, based on baseline level and CKD progression. • In CKD G4: for serum calcium and phosphate, every 3–6 months; and for PTH, every 6–12 months. • In CKD G5, including G5D: for serum calcium and phosphate, every 1–3 months; and for PTH, every 3–6 months. • In CKD G4–G5D: for alkaline phosphatase activity, every 12 months, or more frequently in the presence of elevated PTH (see Chapter 3.2). In CKD patients receiving treatments for CKD-MBD, or in whom biochemical abnormalities are identified, it is reasonable to increase the frequency of measurements to monitor for trends and treatment efficacy and side effects (Not Graded). 3.1.3: In patients with CKD G3a–G5D, we suggest that 25(OH)D (calcidiol) levels might be measured, and repeated testing determined by baseline values and therapeutic interventions (2C). We suggest that vitamin D deficiency and insufficiency be corrected using treatment strategies recommended for the general population (2C). 3.1.4: In patients with CKD G3a–G5D, we recommend that therapeutic decisions be based on trends rather than on a single laboratory value, taking into account all available CKD-MBD assessments (1C). 3.1.5: In patients with CKD G3a–G5D, we suggest that individual values of serum calcium and phosphate, evaluated together, be used to guide clinical practice rather than the mathematical construct of calcium-phosphate product (Ca × P) (2D). 3.1.6: In reports of laboratory tests for patients with CKD G3a–G5D, we recommend that clinical laboratories inform clinicians of the actual assay method in use and report any change in methods, sample source (plasma or serum), or handling specifications to facilitate the appropriate interpretation of biochemistry data (1B). Chapter 3.2: Diagnosis of CKD-MBD: bone 3.2.1: In patients with CKD G3a–G5D with evidence of CKD-MBD and/or risk factors for osteoporosis, we suggest BMD testing to assess fracture risk if results will impact treatment decisions (2B). 3.2.2: In patients with CKD G3a–G5D, it is reasonable to perform a bone biopsy if knowledge of the type of renal osteodystrophy will impact treatment decisions (Not Graded). 3.2.3: In patients with CKD G3a–G5D, we suggest that measurements of serum PTH or bone-specific alkaline phosphatase can be used to evaluate bone disease because markedly high or low values predict underlying bone turnover (2B). 3.2.4: In patients with CKD G3a–G5D, we suggest not to routinely measure bone-derived turnover markers of collagen synthesis (such as procollagen type I C-terminal propeptide) and breakdown (such as type I collagen cross-linked telopeptide, cross-laps, pyridinoline, or deoxypyridinoline) (2C). 3.2.5: We recommend that infants with CKD G2–G5D have their length measured at least quarterly, while children with CKD G2–G5D should be assessed for linear growth at least annually (1B). Chapter 3.3: Diagnosis of CKD-MBD: vascular calcification 3.3.1: In patients with CKD G3a–G5D, we suggest that a lateral abdominal radiograph can be used to detect the presence or absence of vascular calcification, and an echocardiogram can be used to detect the presence or absence of valvular calcification, as reasonable alternatives to computed tomography-based imaging (2C). 3.3.2: We suggest that patients with CKD G3a–G5D with known vascular or valvular calcification be considered at highest cardiovascular risk (2A). It is reasonable to use this information to guide the management of CKD-MBD (Not Graded). Chapter 4.1: Treatment of CKD-MBD targeted at lowering high serum phosphate and maintaining serum calcium 4.1.1: In patients with CKD G3a–G5D, treatments of CKD-MBD should be based on serial assessments of phosphate, calcium, and PTH levels, considered together (Not Graded). 4.1.2: In patients with CKD G3a–G5D, we suggest lowering elevated phosphate levels toward the normal range (2C). 4.1.3: In adult patients with CKD G3a–G5D, we suggest avoiding hypercalcemia (2C). In children with CKD G3a–G5D, we suggest maintaining serum calcium in the age-appropriate normal range (2C). 4.1.4: In patients with CKD G5D, we suggest using a dialysate calcium concentration between 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l) (2C). 4.1.5: In patients with CKD G3a-G5D, decisions about phosphate-lowering treatment should be based on progressively or persistently elevated serum phosphate (Not Graded). 4.1.6: In adult patients with CKD G3a–G5D receiving phosphate-lowering treatment, we suggest restricting the dose of calcium-based phosphate binders (2B). In children with CKD G3a–G5D, it is reasonable to base the choice of phosphate-lowering treatment on serum calcium levels (Not Graded). 4.1.7: In patients with CKD G3a-G5D, we recommend avoiding the long-term use of aluminum-containing phosphate binders and, in patients with CKD G5D, avoiding dialysate aluminum contamination to prevent aluminum intoxication (1C). 4.1.8: In patients with CKD G3a–G5D, we suggest limiting dietary phosphate intake in the treatment of hyperphosphatemia alone or in combination with other treatments (2D). It is reasonable to consider phosphate source (e.g., animal, vegetable, additives) in making dietary recommendations (Not Graded). 4.1.9: In patients with CKD G5D, we suggest increasing dialytic phosphate removal in the treatment of persistent hyperphosphatemia (2C). Chapter 4.2: Treatment of abnormal PTH levels in CKD-MBD 4.2.1: In patients with CKD G3a–G5 not on dialysis, the optimal PTH level is not known. However, we suggest that patients with levels of intact PTH progressively rising or persistently above the upper normal limit for the assay be evaluated for modifiable factors, including hyperphosphatemia, hypocalcemia, high phosphate intake, and vitamin D deficiency (2C). 4.2.2: In adult patients with CKD G3a–G5 not on dialysis, we suggest that calcitriol and vitamin D analogs not be routinely used (2C). It is reasonable to reserve the use of calcitriol and vitamin D analogs for patients with CKD G4–G5 with severe and progressive hyperparathyroidism (Not Graded). In children, calcitriol and vitamin D analogs may be considered to maintain serum calcium levels in the age-appropriate normal range (Not Graded). 4.2.3: In patients with CKD G5D, we suggest maintaining iPTH levels in the range of approximately 2 to 9 times the upper normal limit for the assay (2C). We suggest that marked changes in PTH levels in either direction within this range prompt an initiation or change in therapy to avoid progression to levels outside of this range (2C). 4.2.4: In patients with CKD G5D requiring PTH-lowering therapy, we suggest calcimimetics, calcitriol, or vitamin D analogs, or a combination of calcimimetics with calcitriol or vitamin D analogs (2B). 4.2.5: In patients with CKD G3a–G5D with severe hyperparathyroidism (HPT) who fail to respond to medical or pharmacological therapy, we suggest parathyroidectomy (2B). Chapter 4.3: Treatment of bone with bisphosphonates, other osteoporosis medications, and growth hormone 4.3.1: In patients with CKD G1–G2 with osteoporosis and/or high risk of fracture, as identified by World Health Organization criteria, we recommend management as for the general population (1A). 4.3.2: In patients with CKD G3a–G3b with PTH in the normal range and osteoporosis and/or high risk of fracture, as identified by World Health Organization criteria, we suggest treatment as for the general population (2B). 4.3.3: In patients with CKD G3a–G5D with biochemical abnormalities of CKD-MBD and low BMD and/or fragility fractures, we suggest that treatment choices take into account the magnitude and reversibility of the biochemical abnormalities and the progression of CKD, with consideration of a bone biopsy (2D). 4.3.4: In children and adolescents with CKD G2–G5D and related height deficits, we recommend treatment with recombinant human growth hormone when additional growth is desired, after first addressing malnutrition and biochemical abnormalities of CKD-MBD (1A). Chapter 5: Evaluation and treatment of kidney transplant bone disease 5.1: In patients in the immediate post–kidney transplant period, we recommend measuring serum calcium and phosphate at least weekly, until stable (1B). 5.2: In patients after the immediate post–kidney transplant period, it is reasonable to base the frequency of monitoring serum calcium, phosphate, and PTH on the presence and magnitude of abnormalities, and the rate of progression of CKD (Not Graded). Reasonable monitoring intervals would be: • In CKD G1T–G3bT, for serum calcium and phosphate, every 6–12 months; and for PTH, once, with subsequent intervals depending on baseline level and CKD progression. • In CKD G4T, for serum calcium and phosphate, every 3–6 months; and for PTH, every 6–12 months. • In CKD G5T, for serum calcium and phosphate, every 1–3 months; and for PTH, every 3–6 months. • In CKD G3aT–G5T, measurement of alkaline phosphatases annually, or more frequently in the presence of elevated PTH (see Chapter 3.2). In CKD patients receiving treatments for CKD-MBD, or in whom biochemical abnormalities are identified, it is reasonable to increase the frequency of measurements to monitor for efficacy and side effects (Not Graded). It is reasonable to manage these abnormalities as for patients with CKD G3a–G5 (see Chapters 4.1 and 4.2) (Not Graded). 5.3: In patients with CKD G1T–G5T, we suggest that 25(OH)D (calcidiol) levels might be measured, and repeated testing determined by baseline values and interventions (2C). 5.4: In patients with CKD G1T–G5T, we suggest that vitamin D deficiency and insufficiency be corrected using treatment strategies recommended for the general population (2C). 5.5: In patients with CKD G1T–G5T with risk factors for osteoporosis, we suggest that BMD testing be used to assess fracture risk if results will alter therapy (2C). 5.6: In patients in the first 12 months after kidney transplant with an estimated glomerular filtration rate greater than approximately 30 ml/min/1.73 m 2 and low BMD, we suggest that treatment with vitamin D, calcitriol/alfacalcidol, and/or antiresorptive agents be considered (2D). • We suggest that treatment choices be influenced by the presence of CKD-MBD, as indicated by abnormal levels of calcium, phosphate, PTH, alkaline phosphatases, and 25(OH)D (2C). • It is reasonable to consider a bone biopsy to guide treatment (Not Graded). There are insufficient data to guide treatment after the first 12 months. 5.7: In patients with CKD G4T–G5T with known low BMD, we suggest management as for patients with CKD G4–G5 not on dialysis, as detailed in Chapters 4.1 and 4.2 (2C). The 2017 updated recommendations resulted in renumbering of several adjacent guideline statements. Specifically, 2009 Recommendation 4.1.6 now becomes 2017 Recommendation 4.1.7; 2009 Recommendation 4.1.8 now becomes 2017 Recommendation 4.1.9; 2009 Recommendation 4.3.5 now becomes 2017 Recommendation 4.3.4; and 2009 Recommendation 5.8 now becomes 2017 Recommendation 5.7. Summary and comparison of 2017 updated and 2009 KDIGO CKD-MBD recommendations 2017 revised KDIGO CKD-MBD recommendations 2009 KDIGO CKD-MBD recommendations Brief rationale for updating 3.2.1. In patients with CKD G3a–G5D with evidence of CKD-MBD and/or risk factors for osteoporosis, we suggest BMD testing to assess fracture risk if results will impact treatment decisions (2B). 3.2.2. In patients with CKD G3a–G5D with evidence of CKD-MBD, we suggest that BMD testing not be performed routinely, because BMD does not predict fracture risk as it does in the general population, and BMD does not predict the type of renal osteodystrophy (2B). Multiple new prospective studies have documented that lower DXA BMD predicts incident fractures in patients with CKD G3a–G5D. The order of these first 2 recommendations was changed because a DXA BMD result might impact the decision to perform a bone biopsy. 

 3.2.2. In patients with CKD G3a–G5D, it is reasonable to perform a bone biopsy if knowledge of the type of renal osteodystrophy will impact treatment decisions (Not Graded). 3.2.1. In patients with CKD G3a–G5D, it is reasonable to perform a bone biopsy in various settings including, but not limited to: unexplained fractures, persistent bone pain, unexplained hypercalcemia, unexplained hypophosphatemia, possible aluminum toxicity, and prior to therapy with bisphosphonates in patients with CKD-MBD (Not Graded). The primary motivation for this revision was the growing experience with osteoporosis medications in patients with CKD, low BMD, and a high risk of fracture. The inability to perform a bone biopsy may not justify withholding antiresorptive therapy from patients at high risk of fracture. 

 4.1.1. In patients with CKD G3a–G5D, treatments of CKD-MBD should be based on serial assessments of phosphate, calcium, and PTH levels, considered together (Not Graded). This new recommendation was provided in order to emphasize the complexity and interaction of CKD-MBD laboratory parameters. 

 4.1.2. In patients with CKD G3a–G5D, we suggest lowering elevated phosphate levels toward the normal range (2C). 4.1.1. In patients with CKD G3a–G5, we suggest maintaining serum phosphate in the normal range (2C). In patients with CKD G5D, we suggest lowering elevated phosphate levels toward the normal range (2C). There is an absence of data supporting that efforts to maintain phosphate in the normal range are of benefit to CKD G3a–G4 patients, including some safety concerns. Treatment should aim at overt hyperphosphatemia. 

 4.1.3. In adult patients with CKD G3a–G5D, we suggest avoiding hypercalcemia (2C).In children with CKD G3a–G5D, we suggest maintaining serum calcium in the age-appropriate normal range (2C). 4.1.2. In patients with CKD G3a–G5D, we suggest maintaining serum calcium in the normal range (2D). Mild and asymptomatic hypocalcemia (e.g., in the context of calcimimetic treatment) can be tolerated in order to avoid inappropriate calcium loading in adults. 

 4.1.4. In patients with CKD G5D, we suggest using a dialysate calcium concentration between 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l) (2C). 4.1.3. In patients with CKD G5D, we suggest using a dialysate calcium concentration between 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l) (2D). Additional studies of better quality are available; however, these do not allow for discrimination of benefits and harms between calcium dialysate concentrations of 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l). Hence, the wording is unchanged, but the evidence grade is upgraded from 2D to 2C. 

 4.1.5. In patients with CKD G3a–G5D, decisions about phosphate-lowering treatment should be based on progressively or persistently elevated serum phosphate (Not Graded). 4.1.4. In patients with CKD G3a–G5 (2D) and G5D (2B), we suggest using phosphate-binding agents in the treatment of hyperphosphatemia. It is reasonable that the choice of phosphate binder takes into account CKD stage, presence of other components of CKD-MBD, concomitant therapies, and side effect profile (Not Graded). Emphasizes the perception that early “preventive” phosphate-lowering treatment is currently not supported by data (see Recommendation 4.1.2).The broader term “phosphate-lowering” treatment is used instead of phosphate binding agents since all possible approaches (i.e., binders, diet, dialysis) can be effective. 4.1.6. In adult patients with CKD G3a–G5D receiving phosphate-lowering treatment, we suggest restricting the dose of calcium-based phosphate binder (2B). In children with CKD G3a–G5D, it is reasonable to base the choice of phosphate-lowering treatment on serum calcium levels (Not Graded). 4.1.5. In patients with CKD G3a–G5D and hyperphosphatemia, we recommend restricting the dose of calcium-based phosphate binders and/or the dose of calcitriol or vitamin D analog in the presence of persistent or recurrent hypercalcemia (1B). New evidence from 3 RCTs supports a more general recommendation to restrict calcium-based phosphate binders in hyperphosphatemic patients across all severities of CKD. In patients with CKD G3a–G5D and hyperphosphatemia, we suggest restricting the dose of calcium-based phosphate binders in the presence of arterial calcification (2C) and/or adynamic bone disease (2C) and/or if serum PTH levels are persistently low (2C). 

 4.1.8. In patients with CKD G3a–G5D, we suggest limiting dietary phosphate intake in the treatment of hyperphosphatemia alone or in combination with other treatments (2D). It is reasonable to consider phosphate source (e.g., animal, vegetable, additives) in making dietary recommendations (Not Graded). 4.1.7. In patients with CKD G3a–G5D, we suggest limiting dietary phosphate intake in the treatment of hyperphosphatemia alone or in combination with other treatments (2D). New data on phosphate sources were deemed to be included as an additional qualifier to the previous recommendation. 

 4.2.1. In patients with CKD G3a–G5 not on dialysis, the optimal PTH level is not known. However, we suggest that patients with levels of intact PTH progressively rising or persistently above the upper normal limit for the assay be evaluated for modifiable factors, including hyperphosphatemia, hypocalcemia, high phosphate intake, and vitamin D deficiency (2C). 4.2.1. In patients with CKD G3a–G5 not on dialysis, the optimal PTH level is not known. However, we suggest that patients with levels of intact PTH above the upper normal limit of the assay are first evaluated for hyperphosphatemia, hypocalcemia, and vitamin D deficiency (2C).It is reasonable to correct these abnormalities with any or all of the following: reducing dietary phosphate intake and administering phosphate binders, calcium supplements, and/or native vitamin D (Not Graded). The Work Group felt that modest increases in PTH may represent an appropriate adaptive response to declining kidney function and has revised this statement to include “persistently” above the upper normal PTH level as well as “progressively rising” PTH levels, rather than “above the upper normal limit.” That is, treatment should not be based on a single elevated value. 

 4.2.2. In adult patients with CKD G3a–G5 not on dialysis, we suggest that calcitriol and vitamin D analogs not be routinely used. (2C) It is reasonable to reserve the use of calcitriol and vitamin D analogs for patients with CKD G4–G5 with severe and progressive hyperparathyroidism (Not Graded). 4.2.2. In patients with CKD G3a–G5 not on dialysis, in whom serum PTH is progressively rising and remains persistently above the upper limit of normal for the assay despite correction of modifiable factors, we suggest treatment with calcitriol or vitamin D analogs (2C). Recent RCTs of vitamin D analogs failed to demonstrate improvements in clinically relevant outcomes but demonstrated increased risk of hypercalcemia. 

 In children, calcitriol and vitamin D analogs may be considered to maintain serum calcium levels in the age-appropriate normal range (Not Graded). 

 4.2.4. In patients with CKD G5D requiring PTH-lowering therapy, we suggest calcimimetics, calcitriol, or vitamin D analogs, or a combination of calcimimetics with calcitriol or vitamin D analogs (2B). 4.2.4. In patients with CKD G5D and elevated or rising PTH, we suggest calcitriol, or vitamin D analogs, or calcimimetics, or a combination of calcimimetics and calcitriol or vitamin D analogs be used to lower PTH (2B). • It is reasonable that the initial drug selection for the treatment of elevated PTH be based on serum calcium and phosphate levels and other aspects of CKD-MBD (Not Graded). • It is reasonable that calcium or non-calcium-based phosphate binder dosage be adjusted so that treatments to control PTH do not compromise levels of phosphate and calcium (Not Graded). • We recommend that, in patients with hypercalcemia, calcitriol or another vitamin D sterol be reduced or stopped (1B). • We suggest that, in patients with hyperphosphatemia, calcitriol or another vitamin D sterol be reduced or stopped (2D). • We suggest that, in patients with hypocalcemia, calcimimetics be reduced or stopped depending on severity, concomitant medications, and clinical signs and symptoms (2D). • We suggest that, if the intact PTH levels fall below 2 times the upper limit of normal for the assay, calcitriol, vitamin D analogs, and/or calcimimetics be reduced or stopped (2C). This recommendation originally had not been suggested for updating by the KDIGO Controversies Conference in 2013. However, due to a subsequent series of secondary and post hoc publications of the EVOLVE trial, the Work Group decided to reevaluate Recommendation 4.2.4 as well. Although EVOLVE did not meet its primary endpoint, the majority of the Work Group members were reluctant to exclude potential benefits of calcimimetics for G5D patients based on subsequent prespecified analyses. The Work Group, however, decided not to prioritize any PTH-lowering treatment at this time because calcimimetics, calcitriol, or vitamin D analogs are all acceptable first-line options in G5D patients. 

 4.3.3. In patients with CKD G3a–G5D with biochemical abnormalities of CKD-MBD and low BMD and/or fragility fractures, we suggest that treatment choices take into account the magnitude and reversibility of the biochemical abnormalities and the progression of CKD, with consideration of a bone biopsy (2D). 4.3.3. In patients with CKD G3a–G3b with biochemical abnormalities of CKD-MBD and low BMD and/or fragility fractures, we suggest that treatment choices take into account the magnitude and reversibility of the biochemical abnormalities and the progression of CKD, with consideration of a bone biopsy (2D). Recommendation 3.2.2 now addresses the indications for a bone biopsy prior to antiresorptive and other osteoporosis therapies. Therefore, 2009 Recommendation 4.3.4 has been removed and 2017 Recommendation 4.3.3 is broadened from CKD G3a–G3b to CKD G3a–G5D. 

 4.3.4. In patients with CKD G4–G5D having biochemical abnormalities of CKD-MBD, and low BMD and/or fragility fractures, we suggest additional investigation with bone biopsy prior to therapy with antiresorptive agents (2C). 

 5.5. In patients with G1T–G5T with risk factors for osteoporosis, we suggest that BMD testing be used to assess fracture risk if results will alter therapy (2C). 5.5. In patients with an estimated glomerular filtration rate greater than approximately 30 ml/min/1.73 m2, we suggest measuring BMD in the first 3 months after kidney transplant if they receive corticosteroids, or have risk factors for osteoporosis as in the general population (2D). 2009 Recommendations 5.5 and 5.7 were combined to yield 2017 Recommendation 5.5. 

 5.7. In patients with CKD G4T–G5T, we suggest that BMD testing not be performed routinely, because BMD does not predict fracture risk as it does in the general population and BMD does not predict the type of kidney transplant bone disease (2B). 

 5.6. In patients in the first 12 months after kidney transplant with an estimated glomerular filtration rate greater than approximately 30 ml/min/1.73 m2 and low BMD, we suggest that treatment with vitamin D, calcitriol/alfacalcidol, and/or antiresorptive agents be considered (2D). • We suggest that treatment choices be influenced by the presence of CKD-MBD, as indicated by abnormal levels of calcium, phosphate, PTH, alkaline phosphatases, and 25(OH)D (2C). • It is reasonable to consider a bone biopsy to guide treatment (Not Graded). There are insufficient data to guide treatment after the first 12 months. 5.6. In patients in the first 12 months after kidney transplant with an estimated glomerular filtration rate greater than approximately 30 ml/min/1.73 m2 and low BMD, we suggest that treatment with vitamin D, calcitriol/alfacalcidol, or bisphosphonates be considered (2D). • We suggest that treatment choices be influenced by the presence of CKD-MBD, as indicated by abnormal levels of calcium, phosphate, PTH, alkaline phosphatases, and 25(OH)D (2C). • It is reasonable to consider a bone biopsy to guide treatment, specifically before the use of bisphosphonates due to the high incidence of adynamic bone disease (Not Graded). There are insufficient data to guide treatment after the first 12 months. The second bullet is revised, consistent with the new bone biopsy recommendation (i.e., 2017 Recommendation 3.2.2). 25(OH)D, 25-hydroxyvitamin D; BMD, bone mineral density; CKD, chronic kidney disease; CKD-MBD, chronic kidney disease–mineral bone disorder; DXA, dual-energy x-ray absorptiometry; PTH, parathyroid hormone; RCT, randomized controlled trial. Changes to above summarized recommendations resulted in renumbering of several adjacent guideline statements. Specifically, 2009 Recommendation 4.1.6 now becomes 2017 Recommendation 4.1.7; 2009 Recommendation 4.1.8 now becomes 2017 Recommendation 4.1.9; 2009 Recommendation 4.3.5 now becomes 2017 Recommendation 4.3.4; and 2009 Recommendation 5.8 now becomes 2017 Recommendation 5.7. Chapter 3.2: Diagnosis of CKD-MBD: bone 3.2.1: In patients with CKD G3a–G5D with evidence of CKD-MBD and/or risk factors for osteoporosis, we suggest BMD testing to assess fracture risk if results will impact treatment decisions (2B). Rationale It is well established that patients with CKD G3a–G5D have increased fracture rates compared with the general population,2, 3, 4 and moreover, incident hip fractures are associated with substantial morbidity and mortality.5, 6, 7, 8, 9 At the time of the 2009 KDIGO CKD-MBD guideline, publications addressing the ability of dual-energy X-ray absorptiometry (DXA) measures of bone mineral density (BMD) to estimate fracture risk in CKD were limited to cross-sectional studies comparing BMD in CKD patients with and without a prevalent fracture. The results were variable across studies and across skeletal sites. In light of the lack of evidence that DXA BMD predicted fractures in CKD patients as it does in the general population, and the inability of DXA to indicate the histological type of bone disease, the 2009 Guideline recommended that BMD testing not be performed routinely in patients with CKD G3a to G5D with CKD-MBD. Furthermore, the lack of clinical trials in patients with low BMD and CKD also limited the enthusiasm for measuring BMD in the first place. The current evidence-based review identified 4 prospective cohort studies of DXA BMD and incident fractures in adults with CKD G3a to G5D (Supplementary Tables S7–S12). These studies demonstrated that DXA BMD predicted fractures across the spectrum from CKD G3a to G5D (Supplementary Tables S7–S12).10, 11, 12, 13 In the earliest study, DXA BMD was measured annually in 485 hemodialysis (HD) patients (mean age: 60 years) in a single center in Japan. 10 In adjusted Cox proportional analyses, lower baseline femoral neck and total hip BMD predicted a greater risk of fracture; for example, the hazard ratio (HR) was 0.65 (95% confidence interval [CI]: 0.47–0.90) for each standard deviation (SD) higher femoral neck BMD. In receiver operating characteristic (ROC) analyses stratified according to parathyroid hormone (PTH) below or above the median value of 204 pg/ml (21.6 pmol/l), the area under the curve (AUC) for femoral neck BMD was 0.717 in the lower stratum and 0.512 in the higher stratum. Of note, higher serum bone-specific alkaline phosphate levels also predicted incident fractures. In the second study, Yenchek et al. assessed whether DXA total hip and femoral neck BMD were associated with incident nonspine fragility fractures in participants with estimated glomerular filtration rate (eGFR)  65 pg/ml (6.9 pmol/l; HR: 1.56, 95% CI: 0.90–2.70) compared with those with a PTH ≤ 65 pg/ml (6.9 pmol/l; HR: 2.41, 95% CI: 2.04–2.85) in all participants combined. This is noteworthy in light of the similar pattern observed in dialysis patients, as described above. 10 West et al. reported the results of a prospective cohort study of 131 predialysis participants, mean age 62 years, followed up over a 2-year interval. 12 At baseline, the proportions with CKD G3a to G3b, G4, and G5 were 34%, 40%, and 26%, respectively. DXA BMD was measured in the total hip, lumbar spine, and ultradistal and one-third radius at baseline and 2 years. Low BMD at all sites, and a greater annualized percentage decrease in BMD predicted fracture. For example, in multivariate models, each SD lower total hip BMD was associated with an odds ratio (OR) of fracture of 1.75 (95% CI: 1.30–2.20). The ROC AUC ranged from 0.62 in the spine to 0.74 in the ultradistal radius in adjusted models. Most recently, Naylor, et al. 11 assessed the ability of the Fracture Risk Assessment Tool (FRAX) to predict a major osteoporotic fracture in 2107 adults ≥ 40 years of age in the Canadian Multicenter Osteoporosis Study, including 320 with an eGFR ≤ 60 ml/min/1.73 m2. Of these, 72% and 24% had CKD G3a and G3b, respectively. FRAX with BMD, FRAX without BMD, and the femoral neck T-score all predicted fractures (AUC: 0.65 to 0.71); the AUC was highest for femoral neck T-score with inclusion of fall history. Importantly, the AUCs did not differ between those with and without CKD. There is growing evidence that DXA BMD predicts fractures in healthy children and adolescents, and those with chronic disease.14, 15 However, no studies have examined the associations among DXA BMD and fractures in children and adolescents with CKD. In light of the lack of evidence that the ability of DXA BMD to predict fracture in children with CKD is different than in adults, no specific recommendations are provided for children. However, it should be noted that children and adolescents with CKD frequently exhibit substantial growth failure. Given that DXA measures of areal BMD (g/cm2) underestimate volumetric BMD (g/cm3) in children with short stature, 16 DXA results should be adjusted for bone size, consistent with the 2013 International Society of Clinical Densitometry (ISCD) Pediatric Official Positions. 17 Prediction equations to adjust DXA results for height Z-score are now available, 16 and the impact on DXA BMD Z-scores in children with CKD is substantial. 18 Finally, a single-center study in 171 children with CKD G2 to G5D reported that lower cortical volumetric BMD in the tibia, as measured by peripheral quantitative computed tomography (CT), predicted fractures over a 1-year interval (Supplementary Tables S7–S12). 19 The HR per unit lower cortical BMD Z-score was 1.75 (95% CI: 1.15–2.67; P  300 pg/ml (32 pmol/l) for upper threshold. + Using serum iPTH  585 pg/ml (62 pmol/l) for upper threshold (2X and 9X of upper limit of normal for assay). Reproduced with permission from Sprague SM, Bellorin-Font E, Jorgetti V, et al. Diagnostic accuracy of bone turnover markers and bone histology in patients with CKD treated by dialysis. Am J Kidney Dis. 2016;67:559–566. A bone biopsy should also be considered in patients with unexplained fractures, refractory hypercalcemia, suspicion of osteomalacia, an atypical response to standard therapies for elevated PTH, or progressive decreases in BMD despite standard therapy. The goal of a bone biopsy would be to: (i) rule out atypical or unexpected bone pathology; (ii) determine whether the patient has high- or low-turnover disease, which may alter the dose of medications to treat renal osteodystrophy (e.g., initiate or discontinue calcimimetics, calcitriol, or vitamin D analogs); or (iii) identify a mineralization defect that would alter treatment (e.g., stop intake of aluminum, or aggressively treat hypophosphatemia or vitamin D deficiency). The 2009 Guideline recommended a bone biopsy prior to antiresorptive therapy in patients with CKD G4 to G5D and evidence of biochemical abnormalities of CKD-MBD, low BMD, and/or fragility fractures. The rationale was that low BMD may be due to CKD-MBD (e.g., high PTH) and that lowering PTH is a safer and more appropriate therapy than an antiresorptive. In addition, there was concern that bisphosphonates would induce low-turnover bone disease. This was based on a single cross-sectional study in 13 patients with CKD G2 to G4 that were referred for bone biopsy after a variable duration of bisphosphonate therapy. 32 To date, studies in patients with CKD have not definitively demonstrated that bisphosphonates cause adynamic bone disease. Furthermore, the concerns in patients with CKD are only theoretical, as it is well established that antiresorptive medications suppress bone formation rates, even in the absence of kidney disease. For example, in an RCT of zoledronic acid for the treatment of postmenopausal osteoporosis, bALP levels were 59% lower in the zoledronic acid group compared with the placebo group at 12 months. 33 Despite these limitations, in weighing the risk-benefit ratio of bisphosphonate treatment, the 2009 KDIGO Guideline suggested a biopsy prior to therapy. Since 2009, an additional antiresorptive treatment (denosumab) has proven to be effective in CKD G3a to G3b and G4, as discussed in Recommendation 3.2.1. The growing experience with osteoporosis medications in patients with CKD increases the comfort of treating patients with low BMD and a high risk of fracture with antiresorptive therapy, although definitive trials are lacking. Furthermore, additional data clearly support that the incidence of fracture is markedly increased in patients with CKD, and thus the inability to perform a bone biopsy may not justify withholding antiresorptive therapy to patients at high risk of fracture. Thus, the Work Group voted to remove the requirement of bone biopsy prior to the use of antiresorptive therapy for osteoporosis because the use of these drugs must be individualized in patients with CKD. However, it is still prudent that these drugs be used with caution and that the underlying renal osteodystrophy be addressed first. With regard to efficacy, one may speculate that antiresorptive therapies confer less benefit in the absence of activated osteoclasts, as is the case in adynamic bone disease. Moreover, additional side effects such as acute kidney injury may also merit consideration in CKD G3a to G5. In summary, bone biopsy is the gold standard for the assessment of renal osteodystrophy and should be considered in patients in whom the etiology of clinical symptoms and biochemical abnormalities is in question, and the results may lead to changes in therapy. With this statement, the Work Group is well aware that experience concerning performance and evaluation of bone biopsies is limited in many centers. 34 With this in mind, in addition to the growing evidence that antiresorptive therapies are effective in patients with CKD G3a to G3b and G4, and the lack of robust evidence that these medications induce adynamic bone disease, the guideline no longer suggests that a bone biopsy be performed prior to initiation of these medications. Research recommendation • Prospective studies of circulating biomarkers are needed to determine whether they can predict changes in bone histology. Chapter 4.1: Treatment of CKD-MBD targeted at lowering high serum phosphate and maintaining serum calcium 4.1.1: In patients with CKD G3a–G5D, treatments of CKD-MBD should be based on serial assessments of phosphate, calcium, and PTH levels, considered together (Not Graded). Rationale The previous Recommendation 4.1.1 from the 2009 KDIGO CKD-MBD guideline gave treatment directions concerning serum phosphate levels in different GFR categories of CKD. The accumulated evidence on this issue to date is now depicted in Supplementary Tables S49–S51, S53–S55. Results of this evidence review can be summarized as follows: most studies showed increasing risk of all-cause mortality with increasing levels of serum phosphate in a consistent and direct fashion, with moderate risk of bias and low quality of evidence, thus not essentially different from the study results before 2009. For GFR decline and cardiovascular event rate, results were considered less conclusive. Serum phosphate, calcium, and PTH concentrations are all routinely measured in CKD patients, and clinical decisions are often made based on these values. However, the results of these tests are influenced by food intake, adherence to and the timing of drug intake and dietary modifications, differences in assay methods and their intra-assay coefficient of variation (CV), and also by the interval from the last dialysis session in CKD G5D patients. Furthermore, it has recently been suggested that these markers undergo significant diurnal changes even in CKD patients.35, 36 Accordingly, the decision should be based not on a single result, but rather on the trends of serial results, which stands very much in accordance to 2009 Recommendation 3.1.4. In addition, recent post hoc analyses of large dialysis cohorts suggest that the prognostic implications of individual biochemical components of CKD-MBD largely depend on their context with regard to constellations of the full array of MBD biomarkers. 37 This analysis identified a wide range of CKD-MBD phenotypes, based on phosphate, calcium, and PTH measurements categorized into mutually exclusive categories of low, medium, and high levels using previous Kidney Disease Outcomes Quality Initiative (KDOQI)/KDIGO guideline targets, further illustrating important potential interactions between components of CKD-MBD in terms of risk prediction for death or cardiovascular events. This analysis, however, did not provide guidance for treatment, because it is unknown whether switching from “risk classes” parallels changes in incidence of complications or mortality over time. Of note, biomarkers such as bALP and 25(OH)vitamin D were also still considered valuable, but as no new evidence has been published on their account, recommendations remained unchanged from the previous guideline (2009 Recommendations 3.1.3, 3.2.3). Finally, therapeutic maneuvers aimed at improving 1 parameter often have unintentional effects on other parameters, as exemplified by the recent EVOLVE trial. 38 The guideline Work Group considered it reasonable to take the context of therapeutic interventions into account when assessing values of phosphate, calcium, and PTH, and felt that it was important to emphasize the interdependency of these biochemical parameters for clinical therapeutic decision making. Based on these assumptions, it was also decided to split previous 2009 Recommendation 4.1.1 into 2 new Recommendations, 4.1.1 (diagnostic recommendation based on accumulated observational evidence) and 4.1.2 (therapeutic recommendation based mostly on RCTs). Research recommendations • Prospective cohort studies or RCTs are needed to evaluate whether changes in CKD-MBD risk marker patterns over time associate with changes in risk (e.g., multiple interventions). • Prospective cohort studies or RCTs are needed to examine whether biochemical abnormalities of CKD-MBD must be weighed differently when induced by pharmacotherapy compared with baseline values (e.g., past experience with hemoglobin as risk predictor vs. active treatment to targets by erythropoiesis-stimulating agents). • Investigations contributing to the understanding of the usefulness of fibroblast growth factor 23 (FGF23) as a complementary marker for treatment indications (e.g., phosphate-lowering therapies to halt CKD progression) and direct treatment target (e.g., regression of left ventricular hypertrophy [LVH]) should be undertaken. 4.1.2: In patients with CKD G3a–G5D, we suggest lowering elevated phosphate levels toward the normal range (2C). Rationale As outlined above, since publication of the 2009 KDIGO CKD-MBD Guideline, additional high-quality evidence now links higher concentrations of phosphate with mortality among patients with CKD G3a to G5 or after transplantation39, 40, 41, 42, 43, 44, 45, 46, 47, 48 (Supplementary Tables S49–S51, S53–S55), although some studies did not confirm this association.49, 50 However, trial data demonstrating that treatments that lower serum phosphate will improve patient-centered outcomes are still lacking, and therefore the strength of this recommendation remains weak (2C). The rationale of interventions, therefore, is still only based on epidemiological evidence as described above and biological plausibility pointing to possible phosphorus toxicity as recently summarized. 51 Three recent historical cohort analyses from DOPPS, ArMORR, and COSMOS were not eligible for this evidence-based review; however, it is noteworthy that these analyses suggested that those dialysis patients who had been prescribed phosphate-binder therapy showed improved survival.52, 53, 54 It is important to note that phosphate-binder prescription was associated with better nutritional status. Indeed, correction for markers of nutritional status in the DOPPS study did mitigate the strength of the association, yet a statistically significant benefit persisted. In addition, propensity scoring attempting to correct for selection bias and subgroup analysis applied by Isakova et al. 53 in the ArMORR cohort suggested robustness of the beneficial findings for those treated with phosphate binders. However, residual confounding still cannot be completely ruled out, and due to the nature of the observational data, these studies did not affect the current recommendation. Methods to prevent the development of hyperphosphatemia essentially include dietary modification, the use of phosphate-lowering therapy, and intensified dialysis schedules for those with CKD G5D. In the 2009 KDIGO Guideline it was suggested to maintain serum phosphate in the normal range in the predialysis setting and lower serum phosphate toward the normal range in patients on dialysis. Interestingly, in the prospective observational COSMOS study cohort of HD patients (Supplementary Tables S49–S51, S53–S55), the best patient survival was observed with serum phosphate close to 4.4 mg/dl (1.42 mmol/l). 55 The previous recommendation suggested that clinicians “maintain serum phosphate in the normal range” for patients with CKD G3a to G3b and G4. The Work Group reevaluated the evidence underlying this assumption. The majority of studies (Supplementary Table S49) found phosphate to be consistently associated with excess mortality at levels above and below the limits of normal, but not in the normal range.40, 41, 42, 43, 47, 48, 56, 57 This finding is in line with the previously found U-shaped relation of phosphate with mortality risk in dialysis patients. 58 However, a recent trial comparing placebo with active phosphate-binder therapy in predialysis patients (CKD G3b–G4) with a mean baseline phosphate concentration of 4.2 mg/dl (1.36 mmol/l), found a minimal decline in serum phosphate, no effect on FGF23, and increases in coronary calcification scores for the active treatment group 59 —calling into question the efficacy and safety of phosphate binding in this population, with normal phosphate concentration prior to initiation of binder treatment (Supplementary Tables S19–24). In this analysis, all phosphate binders were analyzed collectively, and the study was underpowered to detect differences between phosphate binders. Although the data suggested that the observed increase in coronary artery calcification (CAC) was mainly driven by the group treated with calcium-containing phosphate binders, those treated with calcium-free binders had no advantage over placebo in terms of progression of CAC. In addition, a well-executed mineral balance study in predialysis patients using calcium-containing phosphate binders demonstrated the absence of any effect on phosphate balance (while showing in the short term a positive calcium balance). 60 The second principal option to control phosphate in predialysis patients is dietary restriction, as will be addressed in Recommendation 4.1.8. However, in both the NHANES and MDRD cohorts that examined the general population and advanced CKD, respectively, dietary intake or intervention to reduce dietary phosphate intake as assessed by either urinary excretion or dietary recall had only minimal effects on serum phosphate.61, 62 It is unknown whether this minimal decline in serum phosphate concentrations or the more robust lower phosphate intake translates into beneficial clinical outcome. A subsequent analysis of the MDRD study found no impact of low phosphate intake as compared with higher intake on cardiovascular disease or all-cause mortality. 63 It needs to be noted that in this study baseline phosphate levels were normal on average, so results are possibly not applicable to CKD patients with progressively or persistently elevated serum phosphate (see rationale for Recommendation 4.1.5). Taken together, the key insights from these data were: (i) the association between serum phosphate and clinical outcome is not monotonic; (ii) there is a lack of demonstrated efficacy of phosphate binders for lowering serum phosphate in patients with CKD G3a to G4; (iii) the safety of phosphate binders in this population is unproven; and (iv) there is an absence of data showing that dietary phosphate restriction improves clinical outcomes. Consequently, the Work Group has abandoned the previous suggestion to maintain phosphate in the normal range, instead suggesting that treatment be focused on patients with hyperphosphatemia. The Work Group recognizes that preventing, rather than treating, hyperphosphatemia may be of value in patients with CKD G3a to G5D, but acknowledges that current data are inadequate to support the safety or efficacy of such an approach and encourages research in this specific area. Only 2 RCTs have examined phosphate-lowering therapy in children with CKD or on dialysis;64, 65 due to the low number of patients and short follow-up, both studies did not meet literature inclusion criteria set a priori together with the evidence review team (ERT). The first RCT examined biochemical endpoints only and showed equivalent phosphate control with calcium acetate and sevelamer hydrochloride in an 8-week cross-over trial. 65 In the second, 29 children were randomized to different combinations of phosphate binders and vitamin D analogs; bone biopsies suggested that the sevelamer group had reduced bone formation versus baseline at 8-month follow-up, but numbers were too small for comparison versus the calcium carbonate–treated group. 64 Several studies in children on dialysis have shown an association between high phosphate levels and increased vessel thickness,66, 67, 68 vessel stiffness68, 69 and CAC.67, 68, 70, 71 In young adults on dialysis, the CAC score was shown to double within 20 months, and progression was associated with higher serum phosphate levels. 71 Research recommendations • RCTs for controlling hyperphosphatemia in patients with CKD G3a to G5D, with appropriate follow-up and power, should be conducted to assess various phosphate-lowering therapy strategies for reducing the incidence of patient-level endpoints (e.g., CKD progression) in children and adults. • RCTs of low and high dietary phosphate intake in patients in CKD G3a to G5 should be conducted to test the hypothesis that high dietary phosphate intake increases cardiovascular risk either directly or indirectly through induction of FGF23. • If the feasibility of a placebo-controlled trial is threatened due to perceived lack of equipoise (i.e., “unethical” to not lower elevated serum phosphate levels, despite the lack of high-quality data), a prospective trial comparing 2 different phosphate targets in patients with CKD G3a to G5D is encouraged. • RCTs should be conducted in normophosphatemic CKD patients in order to test the hypothesis that active compensatory mechanisms to counterbalance increased phosphate intake (such as increases in FGF23 and PTH) are associated with poorer clinical outcome, despite comparable serum phosphate concentration. Interventions could include dietary phosphate restriction, phosphate-binder therapy, novel compounds to limit phosphate uptake, or a combination thereof. 4.1.3: In adult patients with CKD G3a–G5D, we suggest avoiding hypercalcemia (2C). In children with CKD G3a–G5D, we suggest maintaining serum calcium in the age-appropriate normal range (2C). Rationale As is the case for phosphate, novel epidemiological evidence linking higher calcium concentrations to increased mortality in adults with CKD has accumulated since the 2009 KDIGO CKD-MBD guideline (Supplementary Tables S49–S50, S52–S55).42, 43, 44, 47, 50, 60, 72, 73, 74 Moreover, and in addition to previous observations, 48 novel studies link higher concentrations of serum calcium to nonfatal cardiovascular events.75, 76 This consistency justifies the change of this recommendation from 2D to 2C, although the overall evidence base remains limited due to the lack of prospective controlled trial data. Hypocalcemia is a classical feature of untreated CKD, in part secondary to diminished gastrointestinal (GI) uptake of calcium due to vitamin D deficiency. 77 Hypocalcemia contributes to the pathogenesis of secondary hyperparathyroidism (SHPT) and renal osteodystrophy. Therefore, the previous recommendation suggested maintaining serum calcium in the normal range, including the correction of hypocalcemia. A more recent retrospective observational analysis of a large dialysis cohort confirmed the association between hypocalcemia and mortality risk. 50 Two other recent observations, however, raised doubt within the KDIGO guideline Work Group about the generalizability of the suggestion to correct hypocalcemia. The first is the potential harm for some adults associated with a positive calcium balance (while serum calcium levels do not necessarily reflect calcium balance).78, 79 The second observation is that the prevalence of hypocalcemia may have increased after the introduction of calcimimetics (cinacalcet) in patients on dialysis.38, 80, 81 The clinical implications of this increased incidence of low calcium due to the therapeutic institution of a calcimimetic is uncertain, but may be less harmful. With regard to the intention-to-treat (ITT) population of the EVOLVE trial, no negative signals were associated with the persistently low serum calcium levels in the cinacalcet arm of the trial. Retaining the 2009 KDIGO Guideline on this issue would support the concept that patients developing hypocalcemia during calcimimetic treatment require aggressive calcium treatment. Given the unproven benefits of this treatment and the potential for harm, the Work Group emphasizes an individualized approach to the treatment of hypocalcemia rather than recommending the correction of hypocalcemia for all patients. However, significant or symptomatic hypocalcemia should still be addressed. Symptomatic or severe hypocalcemia may benefit from correction to prevent adverse consequences such as bone disease, hyperparathyroidism, and QTc interval prolongation. Childhood and adolescence are critical periods for bone mass accrual: in healthy children the calcium content of the skeleton increases from ∼25 g at birth to ∼1000 g in adults, and ∼25% of total skeletal mass is laid down during the 2-year interval of peak height velocity. 82 The mean calcium accretion rate in healthy pubertal boys and girls peaks at 359 and 284 mg/d, respectively. 83 The updated evidence review identified a prospective cohort study in 170 children and adolescents with CKD G2 to G5D (Supplementary Table S49–S50, S52–S55) that showed that lower serum calcium levels were independently associated with lower cortical volumetric BMD Z-scores. 19 Over 1 year of follow-up in 89 children, a change in the cortical BMD Z-score positively correlated with baseline calcium (P = 0.008) and increase in calcium (P = 0.002) levels, particularly in growing children. During the 1-year follow-up, 6.5% of children sustained a fracture. Notably, a lower cortical BMD Z-score predicted future fractures: the HR for fracture was 1.75 (95% CI: 1.15–2.67; P = 0.009) per SD decrease in baseline BMD. 19 Thus, the Work Group recognizes the higher calcium requirements of the growing skeleton and suggests that serum calcium levels are maintained in the age-appropriate normal range in children and adolescents. Research recommendations • Calcium balance study in dialysis patients should be pursued at baseline versus after start of calcimimetic treatment (with and without calcium supplementation, adaptations in dialysate calcium concentrations, and/or concomitant active vitamin D analog treatment). • RCTs in children and adolescents with CKD should be conducted to determine whether calcium-based phosphate binders, as compared with calcium-free phosphate binders, promote bone accrual (as measured by bone density and structure, and fractures), and to determine the impact of phosphate binders on arterial calcification in the context of the high calcium requirement of growing bones. 4.1.4: In patients with CKD G5D, we suggest using a dialysate calcium concentration between 1.25 and 1.50 mmol/l (2.5 and 3.0 mEq/l) (2C). Rationale Based on the available evidence, the 2009 Work Group considered that a dialysate calcium concentration of 1.25 mmol/l (2.5 mEq/l) would yield neutral calcium balance, but this statement was subsequently challenged by kinetic modeling studies. 84 Two relevant new RCTs are available concerning this topic (Supplementary Tables S13–S18).85, 86 In the study by Spasovski et al., 86 the effects of 2 different dialysate calcium concentrations were examined in patients with adynamic bone disease, and the lower dialysate calcium (1.25 mmol/l [2.5 mEq/l]) was found to improve bone and mineral parameters compared with the higher concentration of 1.75 mmol/l (3.5 mEq/l). Their data confirmed the results of previous papers and also support individualization of dialysate calcium concentrations as recommended previously by the Work Group. The comparator in this study, however, was a high dialysate calcium concentration of 1.75 mmol/l (3.5 mEq/l), leaving open the possibility that lower levels of dialysate calcium (>1.25 mmol/l [2.5 mEq/l] but 11 mg/dl (2.75 mmol/l). Baseline PTH levels were approximately 1.5 times the upper limit of normal. The ITT analysis revealed that paricalcitol did not reduce LVMI, nor did it modify diastolic function. Of subjects on paricalcitol, the mean serum calcium increased by 0.32 mg/dl (0.08 mmol/l) (95% CI: 0.19–0.45 mg/dl; 0.05–0.11 mmol/l) versus a decrease by 0.25 mg/dl (0.06 mmol/l) (95% CI: −0.37 to −0.12 mg/dl; −0.09 to –0.03 mmol/l) in the placebo group. Hypercalcemia was defined as 2 consecutive measurements of serum calcium > 10.5 mg/dl (> 2.63 mmol/l), and the number of patients requiring dose reductions from 2 μg/d to 1 μg/d and episodes of hypercalcemia were more common in the paricalcitol group (22.6%) compared with the placebo (0.9%) group. In the second key study, a double-blind RCT by Wang et al. (the OPERA study), subjects with CKD G3a to G5, LVH, and PTH ≥ 55 pg/ml (5.83 pmol/l) were randomly assigned to receive paricalcitol (n = 30) or placebo (n = 30). 127 The primary endpoint was change in LVMI over 52 weeks. Baseline PTH levels were approximately twice the upper limit of normal. Change in LVMI did not differ significantly between groups, nor did secondary outcomes such as measures of systolic and diastolic function. The median (interquartile range) changes in serum calcium were 0.08 mmol/l (0.32 mg/dl) (95% CI: 0.02–0.16 mmol/l; 0.08–0.64 mg/dl) and 0.01 mmol/l (0.04 mg/dl) (95% CI: –0.06 to 0.05 mmol/l; –0.24 to 0.2 mg/dl) in the paricalcitol and placebo arms, respectively. Hypercalcemia, defined as any serum calcium > 2.55 mmol/l (> 10.2 mg/dl), occurred in 43.3% and 3.3% of participants in the paricalcitol and placebo arms, respectively. Of note, 70% of those who were hypercalcemic received concomitant calcium-based phosphate binders. Generally the hypercalcemia was mild and could be corrected by stopping the binder without changing the paricalcitol dose. Recent meta-analyses were largely confirmatory and supported the hypercalcemia risk association with calcitriol and vitamin D analogs.129, 130 The evidence review identified 2 RCTs comparing paricalcitol with calcitriol (Supplementary Tables S37–S42); neither demonstrated differences in the incidence of hypercalcemia.131, 132 Coyne et al. 131 compared calcitriol (0.25 μg/d) with paricalcitol (1 μg/d) in 110 patients with CKD G3a to G3b and G4 and PTH > 120 pg/ml (12.7 pmol/l). The change in PTH was comparable in the 2 arms (a decline of 52% vs. 46%) over the 6-month trial, and the incidence of hypercalcemia was very low in both groups (only 3 with paricalcitol and 1 with calcitriol). Further details regarding changes in biochemical parameters are provided in Supplementary Tables S37–S42. An alternative to calcitriol and its analogs is “nutritional” vitamin D supplementation (cholecalciferol and ergocalciferol), which can also suppress PTH (especially in CKD G3a–G3b) and decrease hypercalcemia because the normal homeostatic loops that suppress the CYP27B remain intact. However, no studies of sufficient duration were identified in this evidence review, and thus this therapy remains unproven. Several studies have assessed the effect of PTH-lowering comparing nutritional vitamin D supplements and calcitriol or vitamin D analogs.133, 134 However, these studies were not identified in this evidence review because of their short duration. The use of extended-release calcifediol, a novel vitamin D prohormone, to correct low serum 25(OH) vitamin D levels and lower PTH has also been recently studied. This agent reduces the catabolism of both 25(OH) vitamin D and 1,25(OH)2 vitamin D and increases levels of both. An RCT of 429 patients with CKD G3a to G3b and G4 published after our guideline systematic review reported at least a 10% reduction of intact PTH levels in 72% of participants after 12 months, with no significant impact on calcium, phosphate, or FGF23 levels. 135 No patient-level outcomes were reported, and thus this study did not impact the current recommendation. All of the above studies were conducted in adults. A recent Cochrane review examined vitamin D therapy for bone disease in children with CKD G2 to G5 on dialysis. 136 Bone disease, as assessed by changes in PTH levels, was improved by all vitamin D preparations regardless of route or frequency of administration. The prospective cohort study demonstrated that high PTH levels were independently associated with reduced cortical BMD Z-scores at baseline (P = 0.002) and 1-year follow-up (P  10.5 mg/dl (2.63 mmol/l)] or parathyroidectomy). Cinacalcet appeared to consistently reduce the risk of this endpoint regardless of baseline PTH (HR: 0.31, P ≤ 0.001 for those with baseline PTH 300–600 pg/ml [32–64 pmol/l]; HR: 0.49, P ≤ 0.001 for those with baseline PTH 600–900 pg/ml [64–95 pmol/l]; HR: 0.41, P  900 pg/ml [95 pmol/l]). 139 Cinacalcet had no effect on the risk of clinical fractures in unadjusted analyses (HR: 0.93; P = 0.111) and showed a nominally significant reduction in risk of fracture when adjusted for age (HR: 0.88; P = 0.007). 140 Thus, EVOLVE did not meet its primary endpoint that cinacalcet reduces the risk of death or clinically important vascular events in CKD G5D patients. However, the results of secondary analyses suggest that cinacalcet may be beneficial in this population or a subset. There was a lack of uniform consensus among the Work Group members in their interpretation of these data with regard to establishing cinacalcet as the recommended first-line therapy for patients with CKD G5D requiring PTH-lowering therapy. While some felt that only the primary analysis should be used to interpret the outcome, others were equally convinced that the secondary analyses strongly suggested a benefit of treatment with cinacalcet on important patient-level outcomes. Despite these differences in interpretation, there was agreement among Work Group members that the higher cost of cinacalcet was also a relevant consideration given its uncertain clinical benefits. There was also agreement that the documented association between good clinical outcomes and the extent of FGF23 reduction with cinacalcet warrants further study. 141 No trials demonstrated the benefits of combination therapy (cinacalcet plus another agent) on clinically relevant outcomes. However, several additional RCTs were identified that studied the effect of combination therapy on putative surrogate outcomes (summarized in Supplementary Tables S43–S48). Two trials evaluated the use of cinacalcet with low-dose active vitamin D versus standard therapy. Urena-Torres et al. 142 demonstrated improved PTH-lowering efficacy in subjects treated with cinacalcet or low-dose active vitamin D, while Raggi et al. 143 found that cinacalcet with low-dose vitamin D attenuated the progression of coronary artery calcium accumulation when assessed using calcium volume scores (P = 0.009) although not when using the more common Agatston score (P = 0.07). Two open-label trials of cinacalcet were considered important in reaching consensus for Recommendation 4.2.4. The PARADIGM trial compared a cinacalcet-based treatment strategy with an active vitamin D–based strategy in 312 HD patients and demonstrated similar reductions in PTH in both treatment arms. 144 The BONAFIDE trial evaluated bone histomorphometry in 77 paired bone biopsy samples in cinacalcet-treated subjects with proven high-turnover bone disease and demonstrated reductions in bone formation rates and substantial increase in the number of subjects with normal bone histology (from 0 at baseline to 20 after 6–12 months of therapy). 145 Two subjects developed adynamic bone disease, both of whom had PTH values   −2.5), or osteoporotic (T-score ≤ −2.5). A total of 46 incident fractures were recorded in 53 patients. In a multivariate Cox analysis of DXA BMD results in the total hip, osteopenia (HR: 2.7, 95% CI: 1.6–4.6) and osteoporosis (HR: 3.5, 95% CI: 1.8–6.4) were associated with significantly increased risk of fracture compared with normal BMD, independent of age, sex, and diabetes. Multivariate models were not provided for the lumbar spine BMD T-score results; however, unadjusted analyses suggested that spine BMD provided less fracture prediction compared with total-hip BMD. Although this DXA study in kidney transplant recipients was not eligible for the evidence-based review due to its retrospective design, the Work Group concluded that the findings were consistent with the other studies in CKD G3a to G5D described above. In summary, there is growing evidence that DXA BMD predicts fractures in patients with CKD across the spectrum, with limited data suggesting these findings extend to transplant recipients. The revised guideline statement recommends BMD testing in transplant recipients, as in those with CKD G3a to G5D, if the results will impact treatment decisions. Research recommendations • The research recommendations outlined for Recommendation 3.2.1 should be expanded to include studies in kidney transplant recipients. • Prospective studies in patients with CKD G1T to G5T should be performed to determine the value of BMD and bone biomarkers as predictors of fractures. 5.6: In patients in the first 12 months after kidney transplant with an estimated glomerular filtration rate greater than approximately 30 ml/min/1.73 m 2 and low BMD, we suggest that treatment with vitamin D, calcitriol/alfacalcidol, and/or antiresorptive agents be considered (2D). • We suggest that treatment choices be influenced by the presence of CKD-MBD, as indicated by abnormal levels of calcium, phosphate, PTH, alkaline phosphatases, and 25(OH)D (2C). • It is reasonable to consider a bone biopsy to guide treatment (Not Graded). There are insufficient data to guide treatment after the first 12 months. Rationale The rationale for revised guideline Recommendation 3.2.2 now addresses the indications for a bone biopsy prior to antiresorptive and other osteoporosis therapies. Therefore, the second bullet statement above concerning bone biopsies has been modified. Cinacalcet is not approved for the treatment of hyperparathyroidism in kidney transplant recipients; however, it is clinically used, especially in patients with significant hypercalcemia. While efficiently correcting hypercalcemia, cinacalcet so far has failed to show a beneficial impact on bone mineralization in the transplant population. 162 Denosumab was recently shown to effectively increase BMD in de novo kidney transplant recipients. 163 However, baseline BMD on average was not very low in this trial, and there was no progressive BMD loss in the control group. Furthermore, an increased rate of urinary tract infections was observed. In terms of safety and efficacy, an RCT comparing denosumab and bisphosphonates in eligible patients at risk may be a reasonable future study approach. As pointed out in the rationale for Recommendation 5.5, however, the urgency for correcting bone mineralization in transplant recipients may diminish due to the wide application of steroid minimization schemes. 159 Methodological approach to the 2017 KDIGO CKD-MBD guideline update Purpose In 2009, KDIGO developed a clinical practice guideline on the diagnosis, evaluation, prevention, and treatment of CKD-MBD. 30 Because of the limited evidence, many of the recommendations were deliberately vague. In October of 2013, KDIGO held a Controversies Conference to determine whether there was sufficient new evidence to support updating any of the recommendations. Based on the discussions at the conference, the participants opted for a “selective update” of the guideline. 1 The purpose of this chapter is to describe the methods used to conduct the evidence review and to develop and update the guideline recommendations. Overview of the Process The process of updating the guideline consisted of the following steps: • Convening of a Controversies Conference to determine whether sufficient new data exist to support a reassessment of the guideline • Appointing a Work Group and an ERT • Refining the research questions • Developing the search strategy, inclusion/exclusion criteria, and data extraction tables • Drafting the evidence matrices and evidence profiles • Revising the recommendations • Grading the quality of the evidence • Grading the strength of the recommendation Controversies Conference In October 2013, KDIGO held a Controversies Conference entitled, “CKD-MBD: Back to the Future,” in Madrid, Spain. 1 The purpose of the conference was to determine whether there was sufficient new evidence to support updating any of the recommendations from the 2009 KDIGO guideline on the diagnosis, evaluation, prevention, and treatment of CKD-MBD. Seventy-four experts in adult, pediatric, and transplant nephrology, endocrinology, cardiology, bone histomorphometry pathology, and epidemiology attended the conference. Four topic areas were considered: (i) vascular calcification; (ii) bone quality; (iii) calcium and phosphate; and (iv) vitamin D and PTH. Each participant was assigned to 1 of the 4 topics based on their area of expertise. Participants identified new studies in their topic area and answered a set of questions to determine which recommendations required reevaluation. 1 The result was a list of recommendations to be addressed in a selected update (i.e., to use specific methods to update only those parts of the guideline in need of update). There was a public review of the scope of work for the guideline. Appointment of guideline Work Group and evidence review team The KDIGO co-chairs appointed 2 chairs for the Guideline Work Group, who then assembled the Work Group to be responsible for the development of the guideline. The Work Group comprised domain experts, including individuals with expertise in adult and pediatric nephrology, bone disease, cardiology, and nutrition. The Johns Hopkins University in Baltimore, MD, was contracted as the ERT to provide expertise in guideline development methodology and systematic evidence review. KDIGO support staff provided administrative assistance and facilitated communication. The ERT consisted of methodologists with expertise in nephrology and internal medicine, and research associates and assistants. The ERT and the Work Group worked closely throughout the project. In January 2015, the ERT and the Work Group Co-Chairs held a 2-day meeting in Baltimore, MD, to discuss the guideline development and systematic review processes and to refine the key questions. The ERT performed systematic reviews for each of the questions conducting literature searches, abstract and full-text screening, data extraction, risk of bias assessment, and synthesis. The ERT provided suggestions and edits on the wording of recommendations, and on the use of specific grades for the strength of the recommendations and the quality of evidence. The Work Group took on the primary role of writing the recommendations and rationale, and retained final responsibility for the content of the recommendations and the accompanying narrative. Refinement of the research questions The first task was to define the overall topics and goals for the guideline. Using the recommendations identified during the Controversies Conference, the ERT drafted research questions and identified the population, interventions, comparison, and outcomes (PICO elements) for each research question. The ERT recruited a technical expert panel to review the research questions. The technical expert panel included internal and external clinicians and researchers in nephrology and CKD. During a conference call, the technical expert panel provided feedback on the research questions. The Work Group Co-Chairs and the ERT refined the research questions at the 2-day meeting in Baltimore, MD. During this meeting decisions were also made about outcomes, including those considered most important for decision making that would be graded (key outcomes). The finalized research questions and outcomes are presented in Table 2. Search strategy The ERT searched MEDLINE and the Cochrane Central Register of Controlled Trials (CENTRAL) for the date range of December 2006 through September 2015. The December 2006 date provided the recommended 1-year overlap with the end of the previous search. 164 The search yield was also supplemented by articles provided by the Work Group members through February 2017. The search strategy included MeSH and text terms for CKD and the interventions and markers of interest (Supplementary Appendix A) and was limited to the English language. The ERT also reviewed the list of references that were suggested during the Controversies Conference. All studies that had been included in the prior guideline were rereviewed to ensure that they met the eligibility criteria. Inclusion and exclusion criteria With input from the Work Group, the ERT defined the eligibility criteria a priori. The eligibility criteria for all studies were: (i) original data published in English, (ii) followed up at least 10 patients with CKD for at least 6 months, and (iii) addressed 1 of the research questions. The minimum mean duration of follow-up of 6 months was chosen on the basis of clinical reasoning, accounting for the hypothetical mechanisms of action. For treatments of interest, the proposed effects on patient-centered outcomes require long-term exposure and typically would not be evident before several months of follow-up. The question-specific eligibility criteria are provided in Table 3, and the overall search yield for the guideline systematic review is summarized in Supplementary Appendix B. Table 3 Question-specific eligibility criteria 2009 recommendation no. Exposure or intervention Eligibility criteria 3.2.1, 4.3.4 Bisphosphonates, teriparatide, denosumab, or raloxifene • RCT with at least 10 participants per arm for the outcome of bone quality and at least 25 participants per arm for all other outcomes • Evaluates bone quality, bone mineral density, or fracture 3.2.2, 5.5, 5.7 Predictive value of BMD results • RCT with at least 25 participants per arm or a prospective cohort study with 50 participants • Evaluates fractures or renal osteodystrophy 4.1.1, 4.1.2 Serum phosphate or serum calcium levels • RCT with at least 25 participants per arm or a prospective cohort study with 50 participants • Evaluates mortality, GFR decline, cardiovascular or cerebrovascular events, phosphate levels, calcium levels, bone histology, bone mineral density, bone volume, vascular and valvular calcification imaging, hospitalizations, quality of life, kidney or kidney graft failure, fractures, parathyroidectomy, clinical adverse events, growth, skeletal deformities, bone accrual, or calciphylaxis or calcific uremic arteriolopathy 4.1.3 Dialysate calcium concentrations • RCT with at least 25 participants per arm • Evaluates mortality, cardiovascular or cerebrovascular events, calcium levels, bone histology, bone mineral density, bone volume, vascular and valvular calcification imaging, measures of GFR, hospitalizations, quality of life, kidney or kidney graft failure, fractures, parathyroidectomy, clinical adverse events, growth, skeletal deformities, bone accrual, or calciphylaxis or calcific uremic arteriolopathy 4.1.4, 4.1.7, 4.2.1, 4.2.2, 4.2.4 Dietary phosphate intake, phosphate-binding agents, calcium supplements, native vitamin D, vitamin D analogs, calcitriol, or calcimimetics • RCT with at least 25 participants per arm • Evaluates mortality, cardiovascular or cerebrovascular events, GFR decline, calcium levels, phosphate levels, parathyroid hormone levels, 25-hydroxyvitamin D or 1,25-dihydroxyvitamin D levels, alkaline phosphatases, bone-specific alkaline phosphatase, bicarbonate, FGF23, bone histology, bone mineral density, bone volume, vascular and valvular calcification imaging, measures of GFR, hospitalizations, quality of life, kidney or kidney graft failure, fractures, parathyroidectomy, clinical adverse events, growth, skeletal deformities, bone accrual, calciphylaxis or calcific uremic arteriolopathy for any of the above interventions, or LVH or hypercalcemia for vitamin D analogs BMD, bone mineral density; FGF23, fibroblast growth factor 23; GFR, glomerular filtration rate; LVH, left ventricular hypertrophy; RCT, randomized controlled trial. Two reviewers independently screened titles and abstracts and full-text articles for inclusion. Differences regarding inclusion were resolved through consensus adjudication. Any study not meeting the inclusion criteria could be cited in the narrative but was not considered part of the body of evidence for a particular recommendation. Data extraction The ERT modified the online supplementary tables from the prior guideline. One reviewer abstracted data directly into the modified tables, and a second reviewer confirmed the data abstraction. The ERT abstracted data on general study characteristics, participant characteristics, interventions and co-interventions, and outcome measures, including measures of variability. Two reviewers independently assessed individual study quality using the Cochrane Collaboration’s tool 165 for assessing risk of bias for RCTs and using the Quality in Prognosis Studies tool 166 for observational studies. The Work Group critically reviewed draft tables, and tables were revised as appropriate. Evidence matrices and evidence profiles The ERT created evidence matrices for each of the key outcomes for each research question. For each key outcome, the matrix lists the individual studies, their sample size, follow-up duration, and the individual study quality. The ERT also drafted evidence profiles to display the total number and overall quality of the studies addressing each key outcome for each research question. Revising recommendations In June 2015, the Work Group and the ERT convened a 3-day meeting in Baltimore, MD, to review the summary tables, evidence profiles, and evidence matrices; to decide whether and how the recommendations should be revised; and to determine a grade that described the quality of the overall evidence and a grade for the strength of each recommendation. Grading A structured approach—modeled after Grading of Recommendations Assessment, Development, and Evaluation (GRADE)167, 168, 169, 170, 171, 172 and facilitated by the use of evidence profiles and evidence matrices—was used to determine a grade that described the quality of the overall evidence and a grade for the strength of a recommendation. For each topic, the discussion on grading of the quality of evidence was led by the ERT, and the discussion regarding the strength of the recommendations was led by the Work Group Chairs. Grading the quality of evidence for each outcome The ‘quality of a body of evidence’ refers to the extent to which our confidence in an estimate of effect is sufficient to support a particular recommendation. Following GRADE, the quality of a body of evidence pertaining to a particular outcome of interest is initially categorized on the basis of study design. For questions of interventions, the initial quality grade is “high” if the body of evidence consists of RCTs, “low” if it consists of observational studies, or “very low” if it consists of studies of other study designs. For questions of interventions, the Work Group graded only RCTs. The grade for the quality of evidence for each intervention–outcome pair was then decreased if there were serious limitations to the methodological quality of the aggregate of studies; if there were important inconsistencies in the results across studies; if there was uncertainty about the directness of evidence including a limited applicability of findings to the population of interest; if the data were imprecise or sparse; or if there was thought to be a high likelihood of bias. The final grade for the quality of evidence for an intervention–outcome pair could be 1 of the following 4 grades: “high,” “moderate,” “low,” or “very low” (Table 4). Table 4 GRADE system for grading quality of evidence for an outcome Step 1: starting grade for quality of evidence based on study design Step 2: reduce grade Step 3: raise grade Final grade for quality of evidence for an outcomea High for randomized controlled trialsModerate for quasi-randomized trialLow for observational studyVery low for any other evidence Study quality–1 level if serious limitations–2 levels in very serious limitationsConsistency–1 level if important inconsistencyDirectness–1 level if some uncertainty–2 levels if major uncertaintyOther–1 level if sparse or imprecise data–1 level if high probability of reporting bias Strength of association+1 level is strong,b no plausible confounders, consistent and direct evidence+2 levels if very strong,c no major threats to validity and direct evidenceOther+1 level if evidence of a dose-response gradient+1 level if all residual confounders would have reduced the observed effect HighModerateLowVery low GRADE, grading of recommendations assessment, development, and evaluation; RR, relative risk. a The highest possible grade is “high” and the lowest possible grade is “very low.” b Strong evidence of association is defined as “significant RR of > 2 ( 5 (< 0.2)” based on direct evidence with no major threats to validity. Modified with permission from Uhlig K, Macleod A, Craig J, et al. Grading evidence and recommendations for clinical practice guidelines in nephrology. A position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006;70:2058–2065. 171 Grading the overall quality of evidence The quality of the overall body of evidence was then determined on the basis of the quality grades for all outcomes of interest, taking into account explicit judgments about the relative importance of each outcome. The resulting 4 final categories for the quality of overall evidence were A, B, C, and D (Table 5). This grade for overall evidence is indicated behind the strength of recommendations. The summary of the overall quality of evidence across all outcomes proved to be very complex. Thus, as an interim step, the evidence profiles recorded the quality of evidence for each of 3 outcome categories: patient-centered outcomes, other bone and vascular surrogate outcomes, and laboratory outcomes. The overall quality of evidence was determined by the Work Group and is based on an overall assessment of the evidence. It reflects that, for most interventions and tests, there is no high-quality evidence for net benefit in terms of patient-centered outcomes. Table 5 Final grade for overall quality of evidence Grade Quality of evidence Meaning A High We are confident that the true effect lies close to that of the estimate of the effect. 

 B Moderate The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different. 

 C Low The true effect may be substantially different from the estimate of the effect. 

 D Very low The estimate of effect is very uncertain, and often will be far from the truth. Assessment of the net health benefit across all important clinical outcomes Net health benefit was determined on the basis of the anticipated balance of benefits and harm across all clinically important outcomes. The assessment of net health benefit by the Work Group and ERT is summarized in one of the following statements: (i) There is net benefit from intervention when benefits outweigh harm; (ii) there is no net benefit; (iii) there are trade-offs between benefits and harm when harm does not altogether offset benefits but requires consideration in decision making; or (iv) uncertainty remains regarding net benefit (Table 6). Table 6 Balance of benefits and harms When there was evidence to determine the balance of medical benefits and harm of an intervention to a patient, conclusions were categorized as follows: Net benefits The intervention clearly does more good than harm. 

 Trade-offs There are important trade-offs between the benefits and harm. 

 Uncertain trade-offs It is not clear whether the intervention does more good than harm. 

 No net benefits The intervention clearly does not do more good than harm. Grading the recommendations The “strength of a recommendation” indicates the extent to which one can be confident that adherence to the recommendation will do more good than harm. The strength of a recommendation is graded as Level 1 or Level 2. 173 Table 7 shows the nomenclature for grading the strength of a recommendation and the implications of each level for patients, clinicians, and policy makers. Recommendations can be for or against doing something. Table 8 shows that the strength of a recommendation is determined not just by the quality of evidence, but also by other, often complex judgments regarding the size of the net medical benefit, values and preferences, and costs. Table 7 Implications of the strength of a recommendation Grade Implications Patients Clinicians Policy Level 1:“We recommend” Most people in your situation would want the recommended course of action, and only a small proportion would not. Most patients should receive the recommended course of action. The recommendation can be evaluated as a candidate for developing a policy or a performance measure. 

 Level 2:“We suggest” The majority of people in your situation would want the recommended course of action, but many would not. Different choices will be appropriate for different patients. Each patient needs help to arrive at a management decision consistent with her or his values and preferences. The recommendation is likely to require substantial debate and involvement of stakeholders before policy can be determined. Table 8 Determinants of strength of recommendation Factor Comment Balance between desirable and undesirable effects The larger the difference between the desirable and undesirable effects, the more likely a strong recommendation is warranted. The narrower the gradient, the more likely a weak recommendation is warranted. 

 Quality of the evidence The higher the quality of evidence, the more likely a strong recommendation is warranted. 

 Values and preferences The more variability in values and preferences, or the more uncertainty in values and preferences, the more likely a weak recommendation is warranted. 

 Costs (resource allocation) The higher the costs of an intervention—that is, the more resources consumed—the less likely a strong recommendation is warranted. Ungraded statements The Work Group felt that having a category that allows it to issue general advice would be useful. For this purpose, the Work Group chose the category of a recommendation that was not graded. Typically, this type of ungraded statement met the following criteria: it provides guidance on the basis of common sense; it provides reminders of the obvious; and it is not sufficiently specific to allow an application of evidence to the issue, and therefore it is not based on a systematic review. Common examples include recommendations regarding the frequency of testing, referral to specialists, and routine medical care. The ERT and Work Group strove to minimize the use of ungraded recommendations. Limitations of approach Although the literature searches were intended to be comprehensive, they were not exhaustive. MEDLINE and Cochrane CENTRAL were the only databases searched, and the search was limited to English language publications. Hand searches of journals were not performed, and review articles and textbook chapters were not systematically searched. However, Work Group members did identify additional or new studies for consideration. Nonrandomized studies were not systematically reviewed for studies of interventions. The ERT and Work Group resources were devoted to review of randomized trials, as these were deemed most likely to provide data to support treatment recommendations with higher-quality evidence. Evidence for patient-relevant clinical outcomes was low. Usually, low-quality evidence required a substantial use of expert judgment in deriving a recommendation from the evidence reviewed. Formulation and vetting of recommendations Recommendations were drafted to be clear and actionable, and the wording also considered the ability of concepts to be translated accurately into other languages. The final wording of recommendations and corresponding grades for the strength of the recommendations and the quality of evidence were voted upon by the Work Group and required a majority to be accepted. The process of peer review included an external review by the public to ensure widespread input from numerous stakeholders, including patients, experts, and industry and national organizations. Format for chapters Each chapter contains one or more specific recommendations. Within each recommendation, the strength of the recommendation is indicated as level 1 or level 2, and the quality of the overall supporting evidence is shown as A, B, C, or D. The recommendations are followed by a section that describes the body of evidence and rationale for the recommendations. In relevant sections, research recommendations suggest future research to resolve current uncertainties. Biographic and disclosure information Markus Ketteler, MD, FERA (Work Group co-chair), is professor of medicine and currently serves as Division Chief of Nephrology at the Klinikum Coburg in Coburg, Germany. In 2016, he was additionally appointed as chief medical officer at this institution. He is also chairman of the Medical Board of a large German not-for-profit dialysis provider (KfH Kuratorium für Dialyse und Nierentransplantation e.V.). His research focus is aimed at the understanding of the pathomechanisms involved in extraosseous calcifications, and of phosphate and vitamin D metabolism in CKD. He has authored more than 190 peer-reviewed publications including in The Lancet, Journal of the American Society of Nephrology (JASN), Journal of Clinical Investigation, and Kidney International. Dr. Ketteler has acted as local, national, or European Principal Investigator in several clinical multicenter trials in the CKD-MBD field (e.g., SVCARB, CALMAG, IMPACT-SHPT, PA-CL-05A and -05B, and NOPHOS). He serves on the editorial boards of nephrology journals such as JASN and Nephrology Dialysis Transplantation (theme editor), acted as a KDIGO Work Group member on the CKD-MBD Guideline from 2006 to 2009, and co-leads the German Calciphylaxis Registry. Dr. Ketteler is currently council member and chairman of the administrative office of the European Renal Association–European Dialysis and Transplant Association (ERA-EDTA) (2012–2018). He acted as co-chair of the KDIGO Controversies Conference on CKD-MBD, which took place in Madrid in October 2013. In 2017, he was elected into the board of directors of the Kidney Health Initiative, representing the ERA-EDTA for 3 years. Consultant: Amgen, Fresenius Medical Care, Pfizer, Sanifit, Sanofi, Vifor Fresenius Medical Care Renal Pharma Speaker: Amgen, BMS, Medice, Pfizer, Sanofi, Vifor Fresenius Medical Care Renal Pharma Mary B. Leonard, MD, MSCE (Work Group co-chair), is the Arline and Pete Harman Professor and chair of the Department of Pediatrics at Stanford University School of Medicine. She is also the Adalyn Jay Physician-in-Chief of Lucile Packard Children’s Hospital, and the director of the Stanford Child Health Research Institute. Dr. Leonard received her MD degree from Stanford University and subsequently completed her pediatrics internship, residency, and nephrology fellowship at the Children’s Hospital of Philadelphia. After completing a master’s degree in clinical epidemiology at the University of Pennsylvania in 1997, she joined the faculty in the Departments of Pediatrics and Biostatistics and Epidemiology. She has maintained continuous National Institutes of Health funding for over 20 years, and her multidisciplinary research program is primarily focused on the impact of childhood chronic diseases on growth, skeletal development, nutrition, and physical function, with an emphasis on the detrimental effects of CKD. She co-chaired the ISCD Pediatric Position Development Conferences in 2007 and 2013. Dr. Leonard has served as an Associate Editor for Journal of the American Society of Nephrology and Journal of Bone and Mineral Research. She has published over 150 peer-reviewed manuscripts and is a member of the American Society of Clinical Investigation, American Pediatric Society, and the Society for Pediatric Research. Dr. Leonard declared no competing interests. Geoffrey A. Block, MD, is a clinical nephrologist and director of research at Denver Nephrology. He received his MD at the University of Cincinnati and completed his fellowship in nephrology at the University of Michigan, where he was trained under the mentorship of Dr. Friedrich Port at the United States Renal Data System. Dr. Block started a clinical research department at Denver Nephrology in 1998, and his primary research focus has been on clinical outcomes associated with CKD-MBD. He has published observational reports on the risks associated with disorders of mineral metabolism and has designed, conducted, and published numerous RCTs using a variety of interventions related to mineral metabolism and complications of CKD. He has served as a Work Group member on the 2009 KDIGO CKD-MBD guideline, 2 technical expert panels for Centers for Medicare and Medicaid Services related to bone and mineral disorders, and as a member of the steering committee for the EVOLVE trial. He serves as a reviewer for Clinical Journal of the American Society of Nephrology, Journal of the American Society of Nephrology, American Journal of Kidney Diseases, and Kidney International and was associate editor of Nephron-Clinical Practice. Consultant: Ardelyx, Amgen, AstraZeneca, Celgene, Keryx, Kirin, Ono Pharmaceutical, OPKO Grant/research support: Keryx* Speaker: OPKO Development of educational materials: Amgen, Keryx, OPKO Stock/stock options: Ardelyx Other: medical director, DaVita *Monies paid to institution. Pieter Evenepoel, MD, PhD, FERA, is head of the dialysis unit, division of nephrology, at the University Hospitals Leuven. Dr. Evenepoel completed his medical training at the Catholic University of Leuven, Belgium, in 1992, where he also received his PhD for research on protein assimilation and fermentation in 1997. In 2000, he joined the University Hospitals Leuven, where he gained his certification as Specialist in Internal Medicine and Nephrology. Dr. Evenepoel has maintained an active research interest in mainly clinical aspects of CKD-MBD, as exemplified by numerous original articles, review papers, and commentaries in this field. He is currently a board member of the European Renal Association–European Dialysis and Transplant Association (ERA-EDTA) working group on CKD-MBD. His research interests span areas including uremic toxins, nutrition, and anticoagulation, and he has authored over 200 publications. He serves presently as associate editor of Nephrology Dialysis Transplantation and editorial board member for Kidney International. He is also an Ordinary Council Member of the ERA-EDTA. Consultant: Amgen, Vifor Fresenius Medical Care Renal Pharma Grant/research support: Amgen, TECOmedical Speaker: Amgen, Vifor Fresenius Medical Care Renal Pharma Miscellaneous travel and meeting expenses unrelated to above: Amgen, Shire Masafumi Fukagawa, MD, PhD, FASN, received his MD in 1983 from the University of Tokyo School of Medicine, Tokyo, Japan. Following a subspecialty training and PhD program in Tokyo, he was a research fellow at Vanderbilt University School of Medicine, Nashville, TN, until 1995. From 2000 to 2009, he was associate professor and director of the Division of Nephrology and Kidney Center at Kobe University School of Medicine, Kobe, Japan, and he later moved to Tokai University School of Medicine, Isehara, Japan, as professor of medicine and the director of the Division of Nephrology, Endocrinology, and Metabolism. He was international associate editor of Clinical Journal of the American Society of Nephrology (CJASN) (2005–2010) and currently serves as associate editor for Journal of Bone and Mineral Metabolism. He is an editorial board member for Kidney International, American Journal of Kidney Diseases, CJASN, Nephrology Dialysis Transplantation, and Clinical and Experimental Nephrology. In addition, he served as a Work Group member for the 2009 KDIGO CKD-MBD guideline and also chaired the committee for the new version of the Japanese clinical guideline on CKD-MBD by the Japanese Society for Dialysis Therapy. Consultant: Kyowa Hakko Kirin, Ono Pharmaceutical, Torii Grant/research support: Bayer Japan*, Kyowa Hakko Kirin* Speaker: Bayer Japan, Kyowa Hakko Kirin, Torii Manuscript preparation: Bayer Japan, Kyowa Hakko Kirin *Monies paid to institution. Charles A. Herzog, MD, FACC, FAHA, is professor of medicine at University of Minnesota and a cardiologist at Hennepin County Medical Center (HCMC) for 32 years. He founded the program in interventional cardiology at HCMC and served as cardiac catheterization laboratory director from 1985 to 1991, and cardiac ultrasound laboratory director from 1997 to 2012. He was director of the United States Renal Data System Cardiovascular Special Studies Center from 1999 to 2014. Dr. Herzog participated in the development of the National Kidney Foundation’s K/DOQI Clinical Practice Guidelines for Cardiovascular Disease in Dialysis Patients and KDIGO Clinical Practice Guideline on Acute Kidney Injury. He also co-chaired the 2010 KDIGO Controversies Conference, “Cardiovascular Disease in CKD: What is it and What Can We Do About It?” and is a co-chair of the ongoing KDIGO Kidney, Heart, and Vascular Conference Series. Dr. Herzog was an executive committee member of the EVOLVE trial, and presently he is chairing the renal committee of the ISCHEMIA-CKD trial and is co-principal investigator of the WED-HED (Wearable Cardioverter Defibrillator in Hemodialysis Patients) study. He has over 220 published papers and has served on the editorial boards of the American Heart Journal, Journal of Nephrology, Clinical Journal of the American Society of Nephrology, and liaison editor for Nephrology Dialysis Transplantation. His special interests include cardiac disease and CKD, and echocardiography. Consultant: AbbVie, Fibrogen, Relypsa, Sanifit, ZS Pharma Employment: Hennepin County Medical Center, Chronic Disease Research Group Grants/research support: Amgen*, Zoll* *Monies paid to institution. Linda McCann, RD, CSR, has been a nephrology dietitian for over 43 years, focused on quality patient care, professional management, and electronic applications for kidney disease and nutrition. She is currently a nephrology nutrition consultant and speaker. Ms. McCann was a member of the original KDOQI and KDIGO advisory committees as well as the KDOQI and KDIGO (original and update) Work Groups that developed and published clinical practice guidelines and recommendations for bone and mineral abnormalities in CKD. She is the author of the popular National Kidney Foundation (NKF) publication, Pocket Guide to Nutrition Assessment of the Patient with Kidney Disease, currently in its fifth edition. She has authored book chapters and published multiple peer-reviewed papers in the area of nephrology nutrition. She has a long history of mentoring other professionals, driving clinical excellence, and is a dedicated patient advocate. Ms. McCann has also received numerous awards including: Recognized Renal Dietitian (Council on Renal Nutrition), Multiple Outstanding Service Awards (NKF), Award of Distinguished Service (NKF Northern California), Champion of Hope (NKF Northern California), Joel D. Kopple Award (NKF National), San Jose Business Journal 100 Women of Influence 2012, and the American Association of Kidney Patients Medal of Excellence. Consultant: Amgen, Relypsa, Sanofi Speaker: Amgen, Relypsa, Sanofi Development of educational presentations: Amgen, Relypsa, Sanofi Sharon M. Moe, MD, is the director of the Division of Nephrology and Stuart A. Kleit Professor of Medicine for the Indiana University School of Medicine. She received her medical degree from the University of Illinois–College of Medicine at Chicago in 1989 and completed her residency in Internal Medicine at Loyola University Medical Center in Maywood, IL. Her research and clinical fellowships were completed in nephrology at the University of Chicago in Illinois. Dr. Moe has been a faculty member at Indiana University since 1992 and is currently division director for nephrology in the Department of Medicine at Indiana University School of Medicine and section chief for nephrology at the Roudebush VA Medical Center. She has also served as the associate dean for Research Support in the Indiana University School of Medicine and the vice-chair for research in the Department of Medicine. Dr. Moe is the principal investigator for several ongoing basic and clinical research studies in the field of CKD-MBD, including studies on vascular calcification, mineral metabolism, and bone metabolism in kidney disease. Her research is funded by the Veterans Affairs Department, the National Institutes of Health, foundations, and pharmaceutical companies. She has authored over 200 scientific manuscripts, teaching manuscripts, and textbook chapters. Dr. Moe served on the National Kidney Foundation's (NKF) KDOQI Bone and Mineral Metabolism Clinical Practice Guideline Work Group in 2003 and was co-chair of the international KDIGO CKD-MBD guideline released in 2009. Dr. Moe’s key honors include: election to the American Society for Clinical Research in 2005; the NKF Garabed Eknoyan Award for exceptional contributions to key initiatives such as KDOQI in 2009; councilor to the American Heart Association Kidney Council (2002–2004); International Society of Nephrology (2005–2007); councilor for the American Society of Nephrology (2008–2015) and president of the American Society of Nephrology (2013-2014); and election to the Association of American Physicians in 2017. Consultant: Lilly, Ultragenyx Grants/research support: NIH, Veterans Administration Speaker: Sanofi, University of Kansas Stock/stock options: Lilly Rukshana Shroff, MD, FRCPCH, PhD, is a consultant in pediatric nephrology at Great Ormond Street Hospital for Children in London, UK, and holds an academic position (reader) in nephrology at University College London. Her research focuses on cardiovascular disease in childhood CKD, including laboratory work, clinical research studies, and clinical trials. She is the principal investigator on a multicenter study comparing long-term outcomes of conventional hemodialysis and hemodiafiltration in children. She currently holds a prestigious senior fellowship from the National Institute for Health Research to continue research into mineral dysregulation in CKD. She has published more than 130 original articles, reviews, and book chapters in the fields of nephrology and dialysis. Dr. Shroff has also served on 2 guideline committees for the National Institute for Health and Care Excellence. She is on the council for the European Society for Pediatric Nephrology. She is presently an associate editor for Pediatric Nephrology and serves on the editorial board of Clinical Journal of the American Society of Nephrology. Consultant: AstraZeneca Grant/research support: Fresenius Medical Care* Speaker: Amgen, Fresenius Medical Care *Monies paid to institution. Marcello A. Tonelli, MD, SM, FRCPC, is senior associate dean (clinical research) at the Cumming School of Medicine and associate vice president (health research) at the University of Calgary. He received an MD from the University of Western Ontario, specialist certification in nephrology (FRCPC) at Dalhousie University, an SM in epidemiology from Harvard University, and an MSc in health policy from Imperial College London. He is a nephrologist and professor at the University of Calgary. Dr. Tonelli is the past president of the Canadian Society of Nephrology, a past councilor of the International Society of Nephrology, and the chair of the International Society of Nephrology Research Committee. He is a fellow of the Canadian Academy of Health Sciences, and a member of the American Society for Clinical Investigation. Dr. Tonelli is the chair of the Canadian Task Force for Preventive Health Care, a national panel of experts that makes recommendations about preventive health services to Canada’s 36,000 family physicians. Dr. Tonelli was the recipient of the 2013 United States National Kidney Foundation Medal for Distinguished Service and also the 2013 Kidney Foundation of Canada Medal for Research Excellence. Dr. Tonelli declared no competing interests. Nigel D. Toussaint, MBBS, FRACP, PhD, is the deputy director of nephrology at Melbourne Health and a clinical associate professor within the Department of Medicine at the University of Melbourne, Australia. Dr. Toussaint completed his nephrology training in Melbourne in 2005 and completed his PhD studies at Monash University in 2009, undertaking clinical research in the area of vascular calcification and cardiovascular risk in patients with CKD. He has also completed a graduate diploma in clinical epidemiology (Monash University, 2007), and a National Health and Medical Research Council National Institute of Clinical Studies Fellowship (2011) in the area of implementation research. He is a member of the Editorial Board for Nephrology and is currently a member of council for the Australian and New Zealand Society of Nephrology. Dr. Toussaint practices nephrology at The Royal Melbourne Hospital, where he is also the lead physician for clinical research within the Nephrology Department. His current research involves clinical epidemiology and clinical trials, as well as translational research, in the areas of CKD-MBD biomarkers, vascular calcification, and renal osteodystrophy. He has been an awardee of the Royal Australasian College of Physicians Jacquot Research Establishment Award and is a member of the Scientific Committee for the Australasian Kidney Trials Network and the steering committee for the Australia and New Zealand Dialysis and Transplant Registry. Advisory board: Amgen, Sanofi, Shire Consultant: Amgen, Sanofi, Shire Grants/research support: Amgen*, Shire* Speaker: Amgen, Sanofi, Shire *Monies paid to institution. Marc G. Vervloet, MD, PhD, FERA, is a nephrologist, associate professor of nephrology, and director of the nephrology research program and senior consultant at the Intensive Care Medicine and Vascular Medicine, VU University Medical Center, Amsterdam, Netherlands. He is program leader at the Institute of Cardiovascular Research VU (ICaR-VU), secretary of the CKD-MBD working group of the European Renal Association–European Dialysis and Transplant Association, and a member of the scientific committee of the Dutch Kidney Foundation. Dr. Vervloet obtained his medical degree in 1989, and graduated as internist in 1997 and as nephrologist in 1999. His current research interests encompass research on the deranged bone and metabolism in CKD. Dr. Vervloet currently heads the hemodialysis unit at his hospital, where he performs instructional duties to medical students, residents, and nephrology fellows, and guides several PhD students. He has gained numerous research grants, mainly covering topics related to CKD-MBD, in particular on the role of FGF23 and Klotho, and the clinical use of calcimimetic therapy. In addition, his laboratory research comprises several animal models of CKD and vitamin D deficiency, examining mainly cardiovascular endpoints as assessed by imaging and functional testing in vivo. Advisory board: Amgen, Fresenius Medical Care Consultant: Amgen, Fresenius Medical Care, Otsuka Grants/research support: AbbVie*, Fresenius Medical Care*, Sanofi*, Shire* Speaker: Amgen, Baxter *Monies paid to institution. KDIGO Chairs David C. Wheeler, MD, FRCP, is professor of kidney medicine at University College London, UK, and honorary consultant nephrologist at the Royal Free London NHS Foundation Trust. He is a clinician scientist with an interest in the complications of CKD, specifically those that increase the burden of cardiovascular disease and/or accelerate progression of kidney failure. He has participated in the design, roll-out, and monitoring of several large-scale clinical trials. He was a member of the steering committee of the Study of Heart and Renal Protection (SHARP) and the EValuation Of Cinacalcet HCl Therapy to Lower CardioVascular Events (EVOLVE). He currently sits on the steering committee of Canaglifozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE), acting as UK principal investigator for this study. He is clinical lead for Division 2 of the North Thames Clinical Research Network and heads a team of eight clinical trial nurses and practitioners at the Centre for Nephrology, Royal Free Hospital in London. He is past president of the UK Renal Association and past chair of the UK Renal Registry. His other responsibilities include serving as associate editor of Nephrology Dialysis Transplantation and member of the editorial board of Journal of the American Society of Nephrology. Consultant: Akebia, Alberta Innovates Health Solutions, Amgen, AstraZeneca, Bio Nano Consulting, Boehringer Ingelheim, Bristol-Myers Squibb, Fresenius, GSK, Janssen*, Otsuka, UCB Celltech, Vifor Speaker: Amgen, Fresenius Medical Care, Janssen, Vifor Fresenius Medical Care Renal Pharma, ZS Pharma *Monies paid to institution. Wolfgang C. Winkelmayer, MD, MPH, ScD, is the Gordon A. Cain Chair of Nephrology and professor of medicine at Baylor College of Medicine in Houston, TX. Dr. Winkelmayer received his medical degree (1990) from the University of Vienna, Austria, and later earned a Master of Public Health degree in health care management (1999) and a Doctor of Science degree in health policy (2001) from Harvard University. He then spent 8 years on the faculty of Brigham and Women’s Hospital and Harvard Medical School, where he established himself as a prolific investigator and leader in the discipline of comparative-effectiveness research as it pertains to patients with kidney disease. From 2009 to 2014, he was the director of clinical research in the Division of Nephrology at Stanford University School of Medicine, Palo Alto, CA. He assumed his current position as chief of nephrology at Baylor College of Medicine in September 2014. His main areas of research interest include comparative effectiveness and safety research of treatment strategies in anemia as well as of various interventions for cardiovascular disease in patients with kidney disease. His clinical passion lies in providing quality kidney care to the predominantly disadvantaged and un(der)insured population in the public safety net health system of Harris County, TX. Dr. Winkelmayer has authored over 270 peer-reviewed publications, and he has a particular interest in medical publishing. He currently serves as an associate editor for The Journal of the American Medical Association, was a co-editor of the American Journal of Kidney Diseases from 2007 to 2016, and has been appointed to several other editorial boards of leading nephrology and epidemiology journals. He also volunteers his time toward important initiatives of the American Society of Nephrology (e.g., public policy board). He joined KDIGO volunteer leadership as an executive committee member in 2015 and has served as its co-chair since 2016. Advisory board: Akebia, AMAG Pharmaceuticals, Amgen, AstraZeneca, Bayer, Daichi Sankyo, Medtronic, Relypsa, Vifor Fresensius Medical Care Renal Pharma Consultant: Akebia, AMAG Pharmaceuticals, Amgen, AstraZeneca, Bayer, Daichi Sankyo, Medtronic, Relypsa, Vifor Fresensius Medical Care Renal Pharma Evidence review team Karen A. Robinson, PhD, is associate professor of medicine, epidemiology and health policy & management, and director of the Evidence-Based Practice Center at Johns Hopkins University, serving as the project director. She provided methodological expertise in the conduct of the systematic review and guideline development processes, and oversaw and participated in all aspects of the project, including topic refinement, abstract and full-text screening, data extraction, study assessment, evidence grading, and recommendation formulation. Dr. Robinson declared no competing interests. Casey M. Rebholz, PhD, MPH, MS, is assistant professor of epidemiology at Johns Hopkins Bloomberg School of Public Health and Core Faculty at the Welch Center for Prevention, Epidemiology, and Clinical Research. She guided the team through all phases of the project, including refining the questions, conducting literature searches, screening abstracts and full-text articles, abstracting data, drafting and finalizing the evidence tables, and synthesizing the results. Dr. Rebholz declared no competing interests. Lisa M. Wilson, ScM, is a research associate at the Johns Hopkins University Bloomberg School of Public Health. As the project coordinator for the evidence review team, Ms. Wilson managed and participated in all phases of the project, including conducting literature searches, screening abstracts and full-text articles, abstracting data, drafting and finalizing the evidence tables, and synthesizing the results. Ms. Wilson declared no competing interests. Ermias Jirru, MD, MPH, is currently an internal medicine resident at Mount Sinai St. Luke’s and Mount Sinai West. He completed his MPH at the Johns Hopkins University Bloomberg School of Public Health. For this project, he participated in screening abstracts and full-text articles, and abstracting data. Dr. Jirru declared no competing interests. Marisa Chi Liu, MD, MPH, is currently a resident physician at University of California, Irvine, in obstetrics and gynecology. She completed her MPH at Johns Hopkins University Bloomberg School of Public Health and graduated from the University of Vermont College of Medicine. For this project, she participated in screening abstracts and articles, and abstracting data. Dr. Liu declared no competing interests. Jessica Gayleard, BS, is a research assistant within the Johns Hopkins University Evidence-Based Practice Center. She participated in all phases of the project, including conducting literature searches, screening abstracts and full-text articles, abstracting data, and drafting and finalizing the evidence tables. Ms. Gayleard declared no competing interests. Allen Zhang, BS, is a research data analyst at the Johns Hopkins University Evidence-Based Practice Center. He has a degree in microbiology from the Virginia Polytechnic Institute and University. Mr. Zhang participated in the systematic review development, including search string creation, screening, data cleaning, data management, and writing. In addition, he supplied the necessary statistical analysis, including meta-analyses and meta-regression calculations. Mr. Zhang declared no competing interests. Acknowledgments A special debt of gratitude is owed to the KDIGO co-chairs, David Wheeler and Wolfgang Winkelmayer, for their invaluable guidance throughout the development of this guideline. In particular, we thank Karen Robinson and her ERT members for their substantial contribution to the rigorous assessment of the available evidence. We are also especially grateful to the Work Group members for their expertise throughout the entire process of literature review, data extraction, meeting participation, the critical writing and editing of the statements and rationale, which made the publication of this guideline possible. The generous gift of their time and dedication is greatly appreciated. Finally, and on behalf of the Work Group, we gratefully acknowledge the careful assessment of the draft guideline by external reviewers. The Work Group considered all of the valuable comments made, and where appropriate, suggested changes were incorporated into the final publication. The following individuals provided feedback during the public review of the draft guideline: Patricia Abreu, Adama Lengani, Kamal Ahmed, Bülent Altun, Luis Felipe Alva, Rui Alves, Pablo Amair, Alessandro Amore, Andrea Angioi, Mustafa Arici, Mariano Arriola, Rommel Bataclan, Ezequiel Bellorin-Font, Deborah Benner, Mohammed Benyahia, Patrick Biggar, Charles Bishop, Boris Bogov, Jordi Bover, Laura Brereton, Philippe Brunet, Rafael Burgos-Calderon, Stephen Carrithers, Sue Cary, Rolando Claure-Del Granado, Adrian Covic, Mario Cozzolino, Andrew Crannage, John Cunningham, Pierre Delanaye, Nida Dinçel, Tilman B. Drüeke, Nordin Eezsafryna Azalin, Grahame J. Elder, Madgy ElSharkawy, Joyce Ezaki-Yamaguchi, Toshiro Fujita, Alvaro Garcia, Carlo Francisco Gochuico, Heong Keong Goh, Hai An Ha Phan, Takayuki Hamano, Ditte Hansen, Li Hao, Eero Honkanen, Alastair Hutchison, Atul Ingale, Joachim H. Ix, Faical Jarraya, Chandra Mauli Jha, Kamyar Kalantar-Zadeh, Arif Khwaja, Csaba P. Kovesdy, Holly Kramer, Craig B. Langman, Kevin V. Lemley, Edgar V. Lerma, Nathan W. Levin, Maria Jesus Lloret, José António Lopes, Franklin W. Maddux, Francesca Mallamaci, Sandro Mazzaferro, Peter A. McCullough, Donald A. Molony, Sameh Morgan, Eugen Mota, Ricardo Mouzo, Lavinia Negrea, Armando Negri, Michal Nowicki, Tom Nusbickel, Basma Osman, Susan M. Ott, Antonino Paglialunga, Saime Paydas, Adriana Peñalba, Gerson Marques Pereira Junior, Eduardo Perez, Ligia Petrica, Friedrich K. Port, Pradeep Kumar Rai, Dwarakanathan Ranganathan, Nicolas Roberto Robles, Cibele Rodrigues, Hector Rodriguez, Guillermo Rosa Diez, Ibrahim Saig, Deepak Sharma, Laura Sola, David M. Spiegel, Kyriaki Stamatelou, Ekamol Tantisattamo, Mihály Tapolyai, Francesca Tentori, Katrin Uhlig, Harun Ur Rashid, Pablo Ureña Torres, Keth Vuthy, Angela Yee-Moon Wang, Talia Weinstein, Jane Wheeler, Janie Xiong, and Xueqing Yu. Participation in the review does not necessarily constitute endorsement of the content of this report by the above individuals, or the organization or institution they represent. Markus Ketteler, MD, FERA Mary B. Leonard, MD, MSCE Work Group Co-chairs
                Bookmark

                Author and article information

                Journal
                Int J Mol Sci
                Int J Mol Sci
                ijms
                International Journal of Molecular Sciences
                MDPI
                1422-0067
                18 September 2020
                September 2020
                : 21
                : 18
                : 6846
                Affiliations
                [1 ]Department of Rehabilitation Medicine, Ten-Chan General Hospital, Zhongli, Taoyuan 320, Taiwan; f927kimo@ 123456gmail.com
                [2 ]Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 320, Taiwan
                [3 ]Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan; gracealex168@ 123456gmail.com
                [4 ]Department of Mechanical Engineering, National Chiao-Tung University, Hsinchu 300, Taiwan
                [5 ]Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei 231, Taiwan
                [6 ]Department of Medicine, School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
                Author notes
                [* ]Correspondence: alexgfctw@ 123456yahoo.com.tw ; Tel.: +886-2662-89779
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0002-7649-0179
                https://orcid.org/0000-0002-4758-0351
                Article
                ijms-21-06846
                10.3390/ijms21186846
                7555655
                32961953
                3b0cef97-d240-4e0b-8865-00f928ae285a
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 15 August 2020
                : 16 September 2020
                Categories
                Review

                Molecular biology
                chronic kidney disease,osteoporosis,dialysis,fracture
                Molecular biology
                chronic kidney disease, osteoporosis, dialysis, fracture

                Comments

                Comment on this article