Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Ann Surg ; 276(3): e141-e176, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35848728

RESUMO

OBJECTIVE: To develop evidence-based recommendations for safe, effective, and appropriate treatment of secondary (SHPT) and tertiary (THPT) renal hyperparathyroidism. BACKGROUND: Hyperparathyroidism is common among patients with chronic kidney disease, end-stage kidney disease, and kidney transplant. The surgical management of SHPT and THPT is nuanced and requires a multidisciplinary approach. There are currently no clinical practice guidelines that address the surgical treatment of SHPT and THPT. METHODS: Medical literature was reviewed from January 1, 1985 to present January 1, 2021 by a panel of 10 experts in SHPT and THPT. Recommendations using the best available evidence was constructed. The American College of Physicians grading system was used to determine levels of evidence. Recommendations were discussed to consensus. The American Association of Endocrine Surgeons membership reviewed and commented on preliminary drafts of the content. RESULTS: These clinical guidelines present the epidemiology and pathophysiology of SHPT and THPT and provide recommendations for work-up and management of SHPT and THPT for all involved clinicians. It outlines the preoperative, intraoperative, and postoperative management of SHPT and THPT, as well as related definitions, operative techniques, morbidity, and outcomes. Specific topics include Pathogenesis and Epidemiology, Initial Evaluation, Imaging, Preoperative and Perioperative Care, Surgical Planning and Parathyroidectomy, Adjuncts and Approaches, Outcomes, and Reoperation. CONCLUSIONS: Evidence-based guidelines were created to assist clinicians in the optimal management of secondary and tertiary renal hyperparathyroidism.


Assuntos
Hiperparatireoidismo Secundário , Falência Renal Crônica , Cirurgiões , Humanos , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/cirurgia , Rim , Falência Renal Crônica/complicações , Falência Renal Crônica/cirurgia , Paratireoidectomia/métodos , Estados Unidos/epidemiologia
2.
Nephrol Dial Transplant ; 35(6): 946-954, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32259248

RESUMO

BACKGROUND: The iron-based phosphate binders, sucroferric oxyhydroxide (SFOH) and ferric citrate (FC), effectively lower serum phosphorus in clinical studies, but gastrointestinal iron absorption from these agents appears to differ. We compared iron uptake and tissue accumulation during treatment with SFOH or FC using experimental rat models. METHODS: Iron uptake was evaluated during an 8-h period following oral administration of SFOH, FC, ferrous sulphate (oral iron supplement) or control (methylcellulose vehicle) in rat models of anaemia, iron overload and inflammation. A 13-week study evaluated the effects of SFOH and FC on iron accumulation in different organs. RESULTS: In the pharmacokinetic experiments, there was a minimal increase in serum iron with SFOH versus control during the 8-h post-treatment period in the iron overload and inflammation rat models, whereas a moderate increase was observed in the anaemia model. Significantly greater increases (P < 0.05) in serum iron were observed with FC versus SFOH in the rat models of anaemia and inflammation. In the 13-week iron accumulation study, total liver iron content was significantly higher in rats receiving FC versus SFOH (P < 0.01), whereas liver iron content did not differ between rats in the SFOH and control groups. CONCLUSIONS: Iron uptake was higher from FC versus SFOH following a single dose in anaemia, iron overload and inflammation rat models and 13 weeks of treatment in normal rats. These observations likely relate to different physicochemical properties of SFOH and FC and suggest distinct mechanisms of iron absorption from these two phosphate binders.


Assuntos
Anemia/tratamento farmacológico , Compostos Férricos/administração & dosagem , Inflamação/tratamento farmacológico , Sobrecarga de Ferro/tratamento farmacológico , Ferro/farmacocinética , Sacarose/administração & dosagem , Administração Oral , Anemia/patologia , Animais , Combinação de Medicamentos , Feminino , Inflamação/patologia , Sobrecarga de Ferro/patologia , Cinética , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Distribuição Tecidual
3.
Semin Dial ; 30(4): 361-368, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28382631

RESUMO

Musculoskeletal manifestations in chronic kidney disease (CKD) are the result of a series of complex alterations in mineral metabolism, which has been defined as chronic kidney disease - mineral and bone-related disorder (CKD-MBD). Biochemical assessment and, at times, bone biopsy remains the mainstay of disease assessment, however, radiological imaging is an important adjunct in evaluating disease severity. This review aims to illustrate the radiological features of CKD-MBD, such as secondary hyperparathyroidism, osteomalacia, adynamic bone disease, osteopenia, and extra-skeletal calcifications.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico por imagem , Humanos , Radiologia
4.
Nephrol Dial Transplant ; 32(11): 1918-1926, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28339993

RESUMO

BACKGROUND: Sucroferric oxyhydroxide is a noncalcium, iron-based phosphate binder that demonstrated sustained serum phosphorus control, good tolerability and lower pill burden compared with sevelamer carbonate (sevelamer) in a Phase 3 study conducted in dialysis patients. This subanalysis examines the efficacy and tolerability of sucroferric oxyhydroxide and sevelamer in the peritoneal dialysis (PD) patient population. METHODS: The initial study (NCT01324128) and its extension (NCT01464190) were multicenter, Phase 3, open-label, randomized (2:1), active-controlled trials comparing sucroferric oxyhydroxide (1.0-3.0 g/day) with sevelamer (2.4-14.4 g/day) in dialysis patients over 52 weeks in total. RESULTS: In the overall study, 84/1055 (8.1%) patients received PD and were eligible for efficacy analysis (sucroferric oxyhydroxide, n = 56; sevelamer, n = 28). The two groups were broadly comparable to each other and to the overall study population. Serum phosphorus concentrations decreased comparably with both phosphate binders by week 12 (mean change from baseline - 0.6 mmol/L). Over 52 weeks, sucroferric oxyhydroxide effectively reduced serum phosphorus concentrations to a similar extent as sevelamer; 62.5% and 64.3% of patients, respectively, were below the Kidney Disease Outcomes Quality Initiative target range (≤1.78 mmol/L). This was achieved with a lower pill burden (3.4 ± 1.3 versus 8.1 ± 3.7 tablets/day) with sucroferric oxyhydroxide compared with sevelamer. Treatment adherence rates were 91.2% with sucroferric oxyhydroxide and 79.3% with sevelamer. The proportion of patients reporting at least one treatment-emergent adverse event was 86.0% with sucroferric oxyhydroxide and 93.1% with sevelamer. The most common adverse events with both treatments were gastrointestinal: diarrhea and discolored feces with sucroferric oxyhydroxide and nausea, vomiting and constipation with sevelamer. CONCLUSIONS: Sucroferric oxyhydroxide is noninferior to sevelamer for controlling serum phosphorus in patients undergoing PD, while providing a relatively low pill burden and a high rate of adherence.


Assuntos
Compostos Férricos/uso terapêutico , Hiperfosfatemia/tratamento farmacológico , Sacarose/uso terapêutico , Adulto , Idoso , Terapia Combinada , Combinação de Medicamentos , Feminino , Compostos Férricos/efeitos adversos , Humanos , Hiperfosfatemia/sangue , Hiperfosfatemia/etiologia , Masculino , Adesão à Medicação , Pessoa de Meia-Idade , Diálise Peritoneal , Fosfatos/sangue , Sacarose/efeitos adversos , Resultado do Tratamento
5.
Nephrol Dial Transplant ; 32(8): 1330-1338, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-27342579

RESUMO

BACKGROUND: Sucroferric oxyhydroxide is a non-calcium, iron-based phosphate binder indicated for the treatment of hyperphosphataemia in adult dialysis patients. This post hoc analysis of a randomized, 24-week Phase 3 study and its 28-week extension was performed to evaluate the long-term effect of sucroferric oxyhydroxide on iron parameters. METHODS: A total of 1059 patients were randomized to sucroferric oxyhydroxide 1.0-3.0 g/day (n = 710) or sevelamer carbonate ('sevelamer') 2.4-14.4 g/day (n = 349) for up to 52 weeks. The current analysis only included patients who completed 52 weeks of continuous treatment (n = 549). Changes in iron-related parameters and anti-anaemic product use during the study were measured. RESULTS: Some changes in iron-related parameters across both treatment groups were observed during the first 24 weeks of the study, and to a lesser extent with longer-term treatment. There were small, but significantly greater increases in mean transferrin saturation (TSAT) and haemoglobin levels with sucroferric oxyhydroxide versus sevelamer during the first 24 weeks (change in TSAT: +4.6% versus +0.6%, P = 0.003; change in haemoglobin: +1.6 g/L versus -1.1 g/L, P = 0.037). Mean serum ferritin concentrations also increased from Weeks 0 to 24 with sucroferric oxyhydroxide and sevelamer (+119 ng/mL and +56.2 ng/mL respectively; no statistically significant difference between groups). In both treatment groups, ferritin concentrations increased to a greater extent in the overall study population [>70% of whom received concomitant intravenous (IV) iron], compared with the subset of patients who did not receive IV iron therapy during the study. The pattern of anti-anaemic product use was similar in both treatment groups, with a trend towards higher use of IV iron and erythropoiesis-stimulating agents with sevelamer. CONCLUSIONS: Initial increases in some iron-related parameters were observed in both treatment groups but were more pronounced with sucroferric oxyhydroxide. These differences between treatment groups with respect to changes in iron parameters are likely due to minimal iron absorption from sucroferric oxyhydroxide.


Assuntos
Compostos Férricos/uso terapêutico , Hiperfosfatemia/tratamento farmacológico , Ferro/metabolismo , Diálise Renal/efeitos adversos , Sacarose/uso terapêutico , Combinação de Medicamentos , Feminino , Humanos , Hiperfosfatemia/etiologia , Hiperfosfatemia/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico
6.
Arthritis Rheumatol ; 69(2): 320-334, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27598995

RESUMO

OBJECTIVE: Granzyme A (GzmA) levels are elevated in the plasma and synovium of patients with rheumatoid arthritis (RA), suggesting involvement of this protease in the pathogenesis of the disease. GzmA contributes to sepsis by regulating the production of proinflammatory cytokines. The purpose of this study was to evaluate the contribution of GzmA to the pathogenesis of RA in vivo and to examine the possibility that GzmA acting via tumor necrosis factor (TNF) stimulates osteoclastogenesis. METHODS: Inflammatory arthritis induced by type II collagen was evaluated in wild-type, GzmA-deficient, and perforin-deficient mice. The osteoclastogenic potential of GzmA was examined in vitro using bone marrow cells and colony-forming unit-granulocyte-macrophage (CFU-GM) cells and in vivo using GzmA-deficient mice. RESULTS: Gene deletion of GzmA attenuated collagen-induced arthritis, including serum levels of proinflammatory cytokines, joint damage, and bone erosion in affected mice, suggesting that osteoclast activity is reduced in the absence of GzmA. Accordingly, GzmA-treated bone marrow cells produced multinucleated cells that fulfilled the criteria for mature osteoclasts: tartrate-resistant acid phosphatase (TRAP) activity, ß integrin expression, calcitonin receptor expression, and resorptive activity on dentin slices. GzmA appeared to act without accessory cells, and its activity was not affected by osteoprotegerin, suggesting a minor contribution of RANKL. It also induced the expression and secretion of TNF. Neutralization of TNF or stimulation of CFU-GM cells from TNF-/- mice prevented GzmA-induced osteoclastogenesis. GzmA-deficient mice had reduced osteoclastogenesis in vivo (fewer calcitonin receptor-positive multinucleated cells and fewer transcripts for cathepsin K, matrix metalloproteinase 9, and TRAP in joints) and reduced serum levels of C-terminal telopeptide of type I collagen. CONCLUSION: GzmA contributes to the joint destruction of RA partly by promoting osteoclast differentiation.


Assuntos
Artrite Experimental/enzimologia , Artrite Experimental/etiologia , Artrite Reumatoide/enzimologia , Artrite Reumatoide/etiologia , Granzimas/fisiologia , Osteogênese/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL
7.
Am J Kidney Dis ; 67(4): 559-66, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26321176

RESUMO

BACKGROUND: The management of chronic kidney disease-mineral and bone disorder requires the assessment of bone turnover, which most often is based on parathyroid hormone (PTH) concentration, the utility of which remains controversial. STUDY DESIGN: Cross-sectional retrospective diagnostic test study. SETTING & PARTICIPANTS: 492 dialysis patients from Brazil, Portugal, Turkey, and Venezuela with prior bone biopsy and stored (-20 °C) serum. INDEX TESTS: Samples were analyzed for PTH (intact [iPTH] and whole PTH), bone-specific alkaline phosphatase (bALP), and amino-terminal propeptide of type 1 procollagen (P1NP). REFERENCE TEST: Bone histomorphometric assessment of turnover (bone formation rate/bone surface [BFR/BS]) and receiver operating characteristic curves for discriminating diagnostic ability. RESULTS: The biomarkers iPTH and bALP or combinations thereof allowed discrimination of low from nonlow and high from nonhigh BFR/BS, with an area under the receiver operating characteristic curve > 0.70 but < 0.80. Using iPTH level, the best cutoff to discriminate low from nonlow BFR/BS was <103.8 pg/mL, and to discriminate high from nonhigh BFR/BS was >323.0 pg/mL. The best cutoff for bALP to discriminate low from nonlow BFR/BS was <33.1 U/L, and for high from nonhigh BFR/BS, 42.1U/L. Using the KDIGO practice guideline PTH values of greater than 2 but less than 9 times the upper limit of normal, sensitivity and specificity of iPTH level to discriminate low from nonlow turnover bone disease were 65.7% and 65.3%, and to discriminate high from nonhigh were 37.0% and 85.8%, respectively. LIMITATIONS: Cross-sectional design without consideration of therapy. Potential limited generalizability with samples from 4 countries. CONCLUSIONS: The serum biomarkers iPTH, whole PTH, and bALP were able to discriminate low from nonlow BFR/BS, whereas iPTH and bALP were able to discriminate high from nonhigh BFR/BS. Prospective studies are required to determine whether evaluating trends in biomarker concentrations could guide therapeutic decisions.


Assuntos
Remodelação Óssea/fisiologia , Osso e Ossos/patologia , Diálise Renal , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/fisiopatologia , Adulto , Fosfatase Alcalina/sangue , Biomarcadores/sangue , Colágeno Tipo I/sangue , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hormônio Paratireóideo/sangue , Valor Preditivo dos Testes , Insuficiência Renal Crônica/terapia , Reprodutibilidade dos Testes , Estudos Retrospectivos
8.
Nephrol Dial Transplant ; 30(6): 1037-46, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25691681

RESUMO

BACKGROUND: Hyperphosphatemia necessitates the use of phosphate binders in most dialysis patients. Long-term efficacy and tolerability of the iron-based phosphate binder, sucroferric oxyhydroxide (previously known as PA21), was compared with that of sevelamer carbonate (sevelamer) in an open-label Phase III extension study. METHODS: In the initial Phase III study, hemo- or peritoneal dialysis patients with hyperphosphatemia were randomized 2:1 to receive sucroferric oxyhydroxide 1.0-3.0 g/day (2-6 tablets/day; n = 710) or sevelamer 2.4-14.4 g/day (3-18 tablets/day; n = 349) for 24 weeks. Eligible patients could enter the 28-week extension study, continuing the same treatment and dose they were receiving at the end of the initial study. RESULTS: Overall, 644 patients were available for efficacy analysis (n = 384 sucroferric oxyhydroxide; n = 260 sevelamer). Serum phosphorus concentrations were maintained during the extension study. Mean ± standard deviation (SD) change in serum phosphorus concentrations from extension study baseline to Week 52 end point was 0.02 ± 0.52 mmol/L with sucroferric oxyhydroxide and 0.09 ± 0.58 mmol/L with sevelamer. Mean serum phosphorus concentrations remained within Kidney Disease Outcomes Quality Initiative target range (1.13-1.78 mmol/L) for both treatment groups. Mean (SD) daily tablet number over the 28-week extension study was lower for sucroferric oxyhydroxide (4.0 ± 1.5) versus sevelamer (10.1 ± 6.6). Patient adherence was 86.2% with sucroferric oxyhydroxide versus 76.9% with sevelamer. Mean serum ferritin concentrations increased over the extension study in both treatment groups, but transferrin saturation (TSAT), iron and hemoglobin concentrations were generally stable. Gastrointestinal-related adverse events were similar and occurred early with both treatments, but decreased over time. CONCLUSIONS: The serum phosphorus-lowering effect of sucroferric oxyhydroxide was maintained over 1 year and associated with a lower pill burden, compared with sevelamer. Sucroferric oxyhydroxide was generally well tolerated long-term and there was no evidence of iron accumulation.


Assuntos
Compostos Férricos/uso terapêutico , Hiperfosfatemia/tratamento farmacológico , Ferro/metabolismo , Fósforo/metabolismo , Diálise Renal/efeitos adversos , Sacarose/uso terapêutico , Combinação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Fatores de Tempo
9.
Kidney Int ; 86(3): 638-47, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24646861

RESUMO

Efficacy of PA21 (sucroferric oxyhydroxide), a novel calcium-free polynuclear iron(III)-oxyhydroxide phosphate binder, was compared with that of sevelamer carbonate in an open-label, randomized, active-controlled phase III study. Seven hundred and seven hemo- and peritoneal dialysis patients with hyperphosphatemia received PA21 1.0-3.0 g per day and 348 received sevelamer 4.8-14.4 g per day for an 8-week dose titration, followed by 4 weeks without dose change, and then 12 weeks maintenance. Serum phosphorus reductions at week 12 were -0.71 mmol/l (PA21) and -0.79 mmol/l (sevelamer), demonstrating non-inferiority of, on average, three tablets of PA21 vs. eight of sevelamer. Efficacy was maintained to week 24. Non-adherence was 15.1% (PA21) vs. 21.3% (sevelamer). The percentage of patients that reported at least one treatment-emergent adverse event was 83.2% with PA21 and 76.1% with sevelamer. A higher proportion of patients withdrew owing to treatment-emergent adverse events with PA21 (15.7%) vs. sevelamer (6.6%). Mild, transient diarrhea, discolored feces, and hyperphosphatemia were more frequent with PA21; nausea and constipation were more frequent with sevelamer. After 24 weeks, 99 hemodialysis patients on PA21 were re-randomized into a 3-week superiority analysis of PA21 maintenance dose in 50 patients vs. low dose (250 mg per day (ineffective control)) in 49 patients. The PA21 maintenance dose was superior to the low dose in maintaining serum phosphorus control. Thus, PA21 was effective in lowering serum phosphorus in dialysis patients, with similar efficacy to sevelamer carbonate, a lower pill burden, and better adherence.


Assuntos
Quelantes/uso terapêutico , Compostos Férricos/uso terapêutico , Hiperfosfatemia/tratamento farmacológico , Fósforo/sangue , Adulto , Idoso , Quelantes/efeitos adversos , Constipação Intestinal/induzido quimicamente , Diarreia/induzido quimicamente , Feminino , Compostos Férricos/efeitos adversos , Humanos , Hiperfosfatemia/sangue , Hiperfosfatemia/etiologia , Quimioterapia de Indução , Quimioterapia de Manutenção , Masculino , Adesão à Medicação , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Pacientes Desistentes do Tratamento , Diálise Peritoneal , Poliaminas/efeitos adversos , Poliaminas/uso terapêutico , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia , Sevelamer
10.
J Tissue Eng Regen Med ; 8(9): 747-56, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22888041

RESUMO

Endothelial colony-forming cells (ECFCs) isolated from peripheral blood are a highly promising cell source for a wide range of applications, including tissue engineering, in vivo vasculogenesis and anti-cancer therapeutics. Because of the potential for clinical translation, it is increasingly important to isolate and study ECFCs from patient cohorts that may benefit from such technologies. The primary objective of this investigation was to determine whether ECFCs could be obtained from patients with chronic kidney disease and diabetes (CKD-DM), using techniques that can be readily applied in the clinical setting. We also investigated the impact of autologous vs commercially available (i.e. allogeneic) human serum on ECFCs isolation. Surprisingly, the efficacy of ECFCs isolation from the CKD-DM group was comparable to a healthy control group when autologous serum was used. In contrast, substitution of allogeneic serum reduced ECFCs isolation in CKD-DM and control groups. In characterization studies, ECFCs were positive for several endothelial cell markers. ECFCs from the CKD-DM group were sensitive to inflammatory activation but their cellular proliferation was compromised. The concentrations of IL-4 and IL-8 were significantly increased in allogeneic serum, which induced a pro-inflammatory environment, including the release of IL-4, IL-6, IL-8 and MCP-1 into the conditioned media of cell cultures. Taken together, these data support further investigation into the use of autologous serum and cells for ECFC-based therapeutics and underscore the importance of the cytokine content in serum used for ECFCs isolation.


Assuntos
Separação Celular/métodos , Ensaio de Unidades Formadoras de Colônias , Citocinas/metabolismo , Células Endoteliais/citologia , Mediadores da Inflamação/metabolismo , Soro/citologia , Adulto , Doadores de Sangue , Estudos de Casos e Controles , Diabetes Mellitus/sangue , Feminino , Humanos , Masculino , Fenótipo , Insuficiência Renal Crônica/sangue , Adulto Jovem
11.
Adv Chronic Kidney Dis ; 20(5): 423-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23978548

RESUMO

Development of CKD-mineral and bone disorder (MBD) increases morbidity and mortality in men and women with CKD. The corresponding link among bone disease, fracture, and extraskeletal calcifications has been the subject of much focus. In the general population, the incidence of cardiovascular disease is higher in men than women, and this gender differences in degree of calcification and morbidity is maintained in kidney disease. Gender differences in phosphorus and fibroblast growth factor-23 (FGF-23) have been described. Increases in both have been linked with increasing likelihood of death in the CKD population as a whole; however, this link is not as well described when looking at women alone. The clinical significance of these differences, and the potential associated outcomes, are poorly understood. Traditional understanding of bone disease in women without kidney disease may not be fully applicable in women with CKD. Use of bone densitometry is limited in this population, and the traditional preventative interventions may not be fully transferrable to women with CKD.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Cálcio/metabolismo , Doenças Cardiovasculares/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fósforo/metabolismo , Insuficiência Renal Crônica/metabolismo , Densidade Óssea , Doenças Ósseas Metabólicas/etiologia , Calcinose/etiologia , Calcinose/metabolismo , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino , Insuficiência Renal Crônica/complicações , Fatores Sexuais
13.
Am J Kidney Dis ; 55(5): 773-99, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20363541

RESUMO

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


Assuntos
Doenças Ósseas Metabólicas/etiologia , Guias de Prática Clínica como Assunto , Insuficiência Renal Crônica/complicações , Doenças Ósseas Metabólicas/diagnóstico , Doenças Ósseas Metabólicas/prevenção & controle , Doenças Ósseas Metabólicas/terapia , Calcinose , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Saúde Global , Humanos , Hipercalcemia/etiologia , Hipercalcemia/terapia , Hiperfosfatemia/etiologia , Hiperfosfatemia/terapia , Medicare/economia , Monitorização Fisiológica/normas , Proteínas de Ligação a Fosfato/administração & dosagem , Mecanismo de Reembolso , Diálise Renal/economia , Insuficiência Renal Crônica/classificação , Insuficiência Renal Crônica/economia , Insuficiência Renal Crônica/terapia , Estados Unidos
14.
Curr Rheumatol Rep ; 11(3): 185-90, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19604462

RESUMO

Osteoporosis is a common bone disease characterized by low bone density, microarchitectural deterioration of bone tissue, and a consequent increase in fracture risk. Recent decades have seen major advances in the understanding of the pathophysiology, prevention, and treatment of this disorder. Chronic kidney disease (CKD) affects more than 20 million Americans and is associated with unique metabolic mineral disorders related to low bone density and increased fractures. Because many patients with low bone density may have CKD, and the combination of osteoporosis and CKD may further increase fracture risk with increased morbidity and mortality, appropriate identification is necessary for effective management. The clinical, laboratory, and imaging studies currently used to diagnose osteoporosis in the general population inadequately detect the complex bone and metabolic changes that occur with CKD. This review focuses on the pathophysiology of osteoporosis and CKD and discusses problems with current diagnostic tools and considerations for treatment regimens.


Assuntos
Nefropatias/fisiopatologia , Osteoporose/fisiopatologia , Biópsia , Densidade Óssea , Osso e Ossos/fisiopatologia , Doença Crônica , Humanos , Nefropatias/etiologia , Osteoporose/complicações
15.
Clin J Am Soc Nephrol ; 3 Suppl 3: S170-4, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18988703

RESUMO

Secondary hyperparathyroidism develops relatively early in chronic kidney disease as a consequence of impaired phosphate, calcium, and vitamin D homeostasis. The disease state in chronic kidney disease, which includes the histologic features of bone disease, defined as renal osteodystrophy, and the hormonal and biochemical disturbances, have recently been redefined as a disease syndrome and is referred to as "chronic kidney disease-mineral and bone disorder." As chronic kidney disease progresses, specific histologic disturbances in the bone develop, which may or may not be predictable from the biochemical and hormonal changes that are associated with chronic kidney disease. In addition, patients may have had underlying bone disease before developing kidney failure or may have been treated with agents that will alter the classical pathologic findings of the bones in chronic kidney disease and their relation to parathyroid hormone. Thus, in stage 5 chronic kidney disease, bone biopsy with quantitative histomorphometric analysis is considered the gold standard in the diagnosis of renal osteodystrophy. In contrast to stage 5 chronic kidney disease, there are very few data on the histologic changes in bone in earlier stages of chronic kidney disease. There also is no adequate information on the etiopathogenesis of bone disease in stages 3 and 4 chronic kidney disease. Thus, because biochemical data cannot predict bone pathology in stages 3 and 4 chronic kidney disease, bone biopsy should be used to define these bone changes and to allow appropriate therapeutic approaches.


Assuntos
Osso e Ossos/patologia , Nefropatias/patologia , Biópsia , Doenças Ósseas Metabólicas/etiologia , Doença Crônica , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Nefropatias/complicações , Osteogênese , Hormônio Paratireóideo/sangue
16.
Am J Nephrol ; 28(2): 246-53, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17989497

RESUMO

BACKGROUND/AIMS: Abnormal bone and mineral metabolism is common in patients with kidney failure and often persists after successful kidney transplant. METHODS: To better understand the natural history of this disease in transplant patients, we reviewed the literature by searching MEDLINE for English language articles published between January 1990 and October 2006 that contained Medical Subject Headings and key words related to secondary or persistent hyperparathyroidism and kidney transplant. RESULTS: Parathyroid hormone levels decreased significantly during the first 3 months after transplant but typically stabilized at elevated values after 1 year. Calcium tended to increase after transplant and then stabilize at the higher end of the normal range within 2 months. Phosphorus decreased rapidly to within or below normal levels after surgery and hypophosphatemia, if present, resolved within 2 months. Low levels of 1,25(OH)2 vitamin D typically did not reach normal values until almost 18 months after transplant. CONCLUSION: This review provides evidence demonstrating that abnormal bone and mineral metabolism exists in patients after kidney transplant and suggests the need for treatment of this condition. However, better observational and interventional research is needed before advocating such a treatment guideline.


Assuntos
Osso e Ossos/metabolismo , Hiperparatireoidismo Secundário/diagnóstico , Hiperparatireoidismo Secundário/etiologia , Transplante de Rim/efeitos adversos , Biomarcadores/metabolismo , Osso e Ossos/patologia , Cálcio/sangue , Progressão da Doença , Humanos , Rim/metabolismo , Nefropatias/cirurgia , Fósforo/sangue , Fatores de Tempo , Resultado do Tratamento , Vitamina D/sangue
17.
Adv Chronic Kidney Dis ; 14(1): 27-36, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200041

RESUMO

Chronic kidney disease (CKD) is accompanied by disturbances in calcium, phosphate, vitamin D, and parathyroid hormone (PTH) homeostasis that play an important role in the pathophysiology of renal bone disease. The increased cardiovascular morbidity and mortality observed among patients with CKD has recently been recognized to be associated with these disturbances in mineral metabolism. Thus, disturbances in mineral metabolism observed in renal failure results in a multisystem disorder, making the development of a standardized definition of these disorders a top priority. Therefore, the Board of Directors of Kidney Disease: Improving Global Outcomes proposed to define the broader category of mineral disorders associated with CKD as CKD-mineral and bone disorder (CKD-MBD). This newly proposed definition will include the disorders of mineral metabolism, bone histology (renal osteodystrophy), and the extraskeletal manifestations such as vascular calcification. This new definition and stratification of disease should result in improvement not only in the clinical management of patients but also will facilitate the interpretation and translation of clinical research. Renal osteodystrophy is now considered as 1 component of this disorder and will be defined as a morphologic alteration only, based on unification of the histomorphometric definitions that will include parameters of turnover, mineralization, and volume. An internationally accepted classification system will enable the consensus for bone biopsy evaluation as well as for the role of biomarkers. This article will focus on the newly proposed definitions of bone disease as part of CKD-MBD, based on the complex pathophysiologic process underlying bone disease in CKD stages 2 to 5.


Assuntos
Doenças Ósseas/patologia , Osso e Ossos/patologia , Nefropatias/metabolismo , Doenças Metabólicas/metabolismo , Minerais/metabolismo , Biomarcadores/análise , Doenças Ósseas/classificação , Doenças Ósseas/diagnóstico , Osso e Ossos/metabolismo , Doença Crônica , Diagnóstico por Imagem/métodos , Humanos , Nefropatias/diagnóstico , Nefropatias/patologia , Falência Renal Crônica/classificação , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Doenças Metabólicas/patologia , Modelos Biológicos
18.
Blood Purif ; 23(4): 287-97, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15942167

RESUMO

BACKGROUND: Dialysis-related amyloidosis (DRA) is a devastating and costly condition that affects patients with end stage kidney disease. A key feature of DRA is the formation of amyloid fibrils, consisting primarily of beta2-microglobulin. Except for kidney transplantation, conventional kidney replacement therapies, which are based on nonspecific mechanisms, do not adequately address beta2-microglobulin removal. An antihuman beta2-microglobulin single-chain variable region antibody fragment (scFv) was developed to confer specificity to beta2-microglobulin removal during hemodialysis. METHODS: The scFv was immobilized onto agarose and characterized for beta2m binding capacity, thermal stability at 37 degrees C, regeneration capacity, storage conditions, and sterility. RESULTS: The beta2-microglobulin binding capacity was 1.3 mg/ml scFv gel. The immunoadsorbent is thermally stable, can be regenerated, stored short-term in 20% ethanol, lyophilized for long-term storage, and withstand process conditions similar to that of a patient's hemodialysis therapy. CONCLUSIONS: The results support further investigation of immobilized scFvs as a novel tool to remove beta2-microglobulin from blood.


Assuntos
Amiloidose/prevenção & controle , Técnicas de Imunoadsorção , Falência Renal Crônica/terapia , Microglobulina beta-2/imunologia , Microglobulina beta-2/isolamento & purificação , Amiloidose/etiologia , Estabilidade de Medicamentos , Circulação Extracorpórea , Humanos , Região Variável de Imunoglobulina , Falência Renal Crônica/complicações , Diálise Renal/efeitos adversos , Sefarose , Esterilização/métodos
19.
Semin Nephrol ; 24(1): 82-90, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14730514

RESUMO

Kidney transplantation is the optimal form of renal replacement therapy for many with end-stage kidney disease. However, kidney transplantation comes with a unique set of medical complications, important among them is bone disease. Posttransplant bone disorders are manifestations of pathologic processes occurring posttransplant that are superimposed on preexisting disorders of bone and mineral metabolism secondary to kidney failure and/or diabetes mellitus. As a consequence of early rapid bone loss, which is seen commonly within the first 3 to 6 months of transplant, the fracture risk posttransplant increases and has been reported as high as 5% to 44%. Posttransplant fractures occur more commonly at peripheral than central sites. Patients with a history of diabetes mellitus are at particular risk for fracture. Parathyroid hormone (PTH) and osteocalcin levels generally decrease after transplantation. Alkaline phosphatase and urinary collagen cross-links are unpredictable. Bone histology varies. No single biomarker unequivocally distinguishes between the various bone disorders found on biopsy examination. Immunosuppression is a major cause of posttransplant bone disorders. Glucocorticoids lead to decreased bone formation whereas the calcineurin inhibitors appear to cause increased bone turnover. Evaluating and managing posttransplant bone disease is an integral part of posttransplant medical care.


Assuntos
Azatioprina/efeitos adversos , Doenças Ósseas/etiologia , Calcifediol/uso terapêutico , Ciclosporina/efeitos adversos , Fraturas Ósseas/etiologia , Imunossupressores/efeitos adversos , Transplante de Rim/efeitos adversos , Azatioprina/uso terapêutico , Densidade Óssea , Doenças Ósseas/metabolismo , Doenças Ósseas/prevenção & controle , Ciclosporina/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Humanos , Imunossupressores/uso terapêutico , Falência Renal Crônica/cirurgia , Hormônio Paratireóideo/metabolismo , Fatores de Risco
20.
Semin Nephrol ; 22(3): 268-75, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12012312

RESUMO

Renal failure is associated with many complex bone and mineral complications. The spectrum of diseases is wide, encompassing defects in bone turnover, remodeling, and mineralization. Disease is currently defined in terms of whether a high or low turnover lesion is present. Measurement of serum parathyroid hormone levels (PTH) remains an important aspect in the management of renal bone disease, however, is limited by its lack of sensitivity in many clinical settings. Multiple biochemical markers are also available both commercially and experimentally to assist in assessing the degree of bone formation or resorption. However, when definitive diagnosis is important, when the clinical setting is confusing or complex, or when parathyroidectomy is being considered, the use of percutaneous bone biopsy is an essential tool in the understanding of underlying bone pathology and in directing therapy intervention.


Assuntos
Osso e Ossos/patologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Biópsia/métodos , Humanos , Hormônio Paratireóideo/sangue , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA