Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Calcif Tissue Int ; 108(4): 539-550, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33219822

RESUMO

Chronic kidney diseases (CKD) are associated with mineral and bone diseases (MBD), including pain, bone loss, and fractures. Bone fragility related to CKD includes the risk factors observed in osteoporosis in addition to those related to CKD, resulting in a higher risk of mortality related to fractures. Unawareness of such complications led to a poor management of fractures and a lack of preventive approaches. The current guidelines of the Kidney Disease Improving Global Outcomes (KDIGO) recommend the assessment of bone mineral density if results will impact treatment decision. In addition to bone density, circulating biomarkers of mineral, serum bone turnover markers, and imaging techniques are currently available to evaluate the fracture risk. The purpose of this review is to provide an overview of the epidemiology and pathogenesis of CKD-associated bone loss. The contribution of the current tools and other techniques in development are discussed. We here propose a current view of how to better predict bone fragility and the therapeutic options in CKD.


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Fraturas Ósseas , Osteoporose , Insuficiência Renal Crônica , Densidade Óssea , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Humanos , Osteoporose/etiologia , Insuficiência Renal Crônica/complicações
2.
Calcif Tissue Int ; 108(4): 512-527, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33398414

RESUMO

Abnormal bone metabolism is an integral part of the chronic kidney disease-mineral bone disorder (CKD-MBD). For several reasons, the difficult bone compartment was neglected for some time, but there has been renewed interest as a result of the conception of bone as a new endocrine organ, the increasing recognition of the cross-talk between bone and vessels, and, especially, the very high risk of osteoporotic fractures (and associated mortality) demonstrated in patients with CKD. Therefore, it has been acknowledged in different guidelines that action is needed in respect of fracture risk assessment and the diagnosis and treatment of osteoporosis in the context of CKD and CKD-MBD, even beyond renal osteodystrophy. These updated guidelines clearly underline the need to improve a non-invasive approach to these bone disorders in order to guide treatment decisions aimed at not only controlling CKD-MBD but also decreasing the risk of fracture. In this report, we review the current role of the most often clinically used or promising biochemical circulating biomarkers such as parathyroid hormone, alkaline phosphatases, and other biochemical markers of bone activity as alternatives to some aspects of bone histomorphometry. We also mention the potential role of classic and new imaging techniques for CKD patients. Information on many aspects is still scarce and heterogeneous, but many of us consider that it is indeed time for action, recognizing our definitely limited ability to base certain treatment decisions only on our current non-comprehensive knowledge.


Assuntos
Doenças Ósseas , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Osteoporose , Fraturas por Osteoporose , Insuficiência Renal Crônica , Biomarcadores , Distúrbio Mineral e Ósseo na Doença Renal Crônica/diagnóstico , Humanos , Osteoporose/diagnóstico , Fraturas por Osteoporose/diagnóstico , Insuficiência Renal Crônica/complicações
3.
Nephrol Dial Transplant ; 36(8): 1511-1518, 2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34021358

RESUMO

BACKGROUND: Intradialytic hypotension (IDH), a common complication in haemodialysis (HD) patients, is associated with multiple risk factors including cardiac dysfunction and alterations of the peripheral autonomic nervous system. To what extent dysautonomia may contribute to the occurrence of IDH remains elusive. We sought to investigate the clinical utility of Sudocan®, a device that quantifies dysautonomia, in the prediction of IDH. METHODS: We conducted a prospective monocentric study in adult HD patients from July 2019 to February 2020. Dysautonomia was assessed by the measurements of hand and foot electrochemical skin conductance (ESC) using Sudocan®, before HD. The primary endpoint was the incidence of IDH (The National Kidney Foundation/Kidney-Dialysis Outcome Quality Initiative definition), according to the presence of a pathological hand and/or foot ESC value, during the 3-month study period. RESULTS: A total of 176 HD patients (64 ± 14 years old) were enrolled. Mean pre-dialysis HD hand and foot ESC was 45 ± 20 and 54 ± 22 µS, respectively. About 35% and 40% of patients had a pathological ESC at the hand and foot, respectively. IDH occurred in 46 patients. Logistic regression showed that pathologic pre-dialysis HD hand ESC was associated with an increased risk of IDH [odds ratio = 2.56, 95% CI (1.04-6.67), P = 0.04]. The cumulative risk incidence of IHD during the study was 5.65 [95% CI (2.04-15.71), P = 0.001] and 3.71 [95% CI (1.41-9.76), P = 0.008], with a pathological hand and foot ESC, respectively. CONCLUSIONS: A pathological hand ESC, as assessed by a non-invasive Sudoscan® test, is associated with an increased risk of IDH.


Assuntos
Hipotensão , Falência Renal Crônica , Adulto , Idoso , Humanos , Hipotensão/diagnóstico , Hipotensão/etiologia , Falência Renal Crônica/terapia , Pessoa de Meia-Idade , Estudos Prospectivos , Diálise Renal/efeitos adversos , Fatores de Risco
4.
Nephrol Dial Transplant ; 36(1): 160-169, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33068419

RESUMO

BACKGROUND: Optimal parathyroid hormone (PTH) control during non-dialysis chronic kidney disease (ND-CKD) might decrease the subsequent risk of parathyroid hyperplasia and uncontrolled secondary hyperparathyroidism (SHPT) on dialysis. However, the evidence for recommending PTH targets and therapeutic strategies is weak for ND-CKD. We evaluated the patient characteristics, treatment patterns and PTH control over the first year of haemodialysis (HD) by PTH prior to HD initiation. METHODS: We studied 5683 incident HD patients from 21 countries in Dialysis Outcomes and Practice Patterns Study Phases 4-6 (2009-18). We stratified by PTH measured immediately prior to HD initiation and reported the monthly prescription prevalence of active vitamin D and calcimimetics over the first year of HD and risk of PTH >600 pg/mL after 9-12 months on HD. RESULTS: The 16% of patients with PTH >600 pg/mL prior to HD initiation were more likely to be prescribed active vitamin D and calcimimetics during the first year of HD. The prevalence of PTH >600 pg/mL 9-12 months after start of HD was greater for patients who initiated HD with PTH >600 (29%) versus 150-300 (7%) pg/mL (adjusted risk difference: 19%; 95% confidence interval : 15%, 23%). The patients with sustained PTH >600 pg/mL after 9-12 months on HD were younger, more likely to be black, and had higher serum phosphorus and estimated glomerular filtration rates at HD initiation. CONCLUSIONS: Increased PTH before HD start predicted a higher PTH level 9-12 months later, despite greater use of active vitamin D and calcimimetics. More targeted PTH control during ND-CKD may influence outcomes during HD, raising the need for PTH target guidelines in these patients.


Assuntos
Biomarcadores/sangue , Hiperparatireoidismo Secundário/etiologia , Hormônio Paratireóideo/sangue , Fósforo/sangue , Diálise Renal/efeitos adversos , Idoso , Feminino , Humanos , Hiperparatireoidismo Secundário/sangue , Hiperparatireoidismo Secundário/patologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco
5.
Am J Nephrol ; 51(10): 833-838, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32911468

RESUMO

BACKGROUND: The incidence of skeletal fractures is high in dialysis patients. Current available tools are insufficient to predict bone fragility. We analyzed the microarchitecture in patients on dialysis therapy using bone biopsies and peripheral microcomputed tomography. METHODS: We analyzed 12 trans-iliac bone biopsies of patients with recent fractures. Bone microarchitecture was assessed in the bone cores by histology (2D-), microcomputed tomography (3D-µCT), and high-resolution peripheral quantitative computed tomography (HR-pQCT) at the tibia. RESULTS: Trabecular bone volume/tissue volume was similar in 2D histology and 3D-µCT (p = 0.40), while lower in HR-pQCT (p < 0.01). There was no correlation in trabecular microarchitectural indices between 2-histology and 3D-µCT, or HR-pQCT. The 3D-µCT cortical thickness (Ct.Th) were positively correlated with 2D (p < 0.05), but with HR-pQCT (p = 0.33). Ct.Th was lower in patients with ≥2 vertebral fractures than with one fracture. CONCLUSIONS: 3D-µCT is a reliable method for the measurement of cortical bone in bone biopsies. Prospective studies are awaited to address its value in discriminating fracture risk.


Assuntos
Osso Cortical/diagnóstico por imagem , Falência Renal Crônica/complicações , Fraturas por Osteoporose/epidemiologia , Diálise Renal/efeitos adversos , Microtomografia por Raio-X , Idoso , Idoso de 80 Anos ou mais , Biópsia , Osso Cortical/patologia , Feminino , Seguimentos , Humanos , Incidência , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade , Fraturas por Osteoporose/etiologia , Fraturas por Osteoporose/patologia , Estudos Prospectivos , Reprodutibilidade dos Testes , Medição de Risco/métodos
6.
Nephrol Dial Transplant ; 35(12): 2046-2053, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31326992

RESUMO

Ischaemic heart disease, sudden cardiac death and arrhythmias, heart failure, stroke and peripheral arterial disease make up >50% of the causes of death in advanced chronic kidney disease (CKD). Calcification of the vascular tree and heart valves is partially related to these complications and has received growing attention in the literature. However, the main focus of research has been on the pathophysiology and consequences of vascular calcification, with less attention being paid to valvular calcification (VC) and its impact on the survival of CKD patients. Although VC has long been seen as an age-related degenerative disorder with minimal functional impact, several studies proved that it carries an increased risk of death and clinical consequences different from those of vascular calcification. In dialysis patients, the annual incidence of aortic valve calcification is nearly 3.3% and the reported prevalence of aortic and mitral VC varies between 25% and 59%. Moreover, calcification of both valves occurs 10-20 years earlier in CKD patients compared with the general population. Therefore, the purpose of this review is to summarize the current knowledge on the pathophysiology and relevance of VC in CKD patients, and to highlight specific clinical consequences and potential therapeutic implications.


Assuntos
Estenose da Valva Aórtica/complicações , Valva Aórtica/patologia , Calcinose/complicações , Doenças das Valvas Cardíacas/etiologia , Insuficiência Renal Crônica/fisiopatologia , Calcificação Vascular/complicações , Doenças das Valvas Cardíacas/patologia , Humanos , Prognóstico
7.
Nephrol Dial Transplant ; 35(10): 1769-1778, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30859218

RESUMO

BACKGROUND: Secondary hyperparathyroidism (sHPT), a common complication of chronic kidney disease, is characterized by elevated serum parathyroid hormone (PTH). Etelcalcetide is an intravenous calcimimetic that increases sensitivity of the calcium-sensing receptor to calcium and decreases PTH secretion. This open-label extension (OLE) trial evaluated the long-term effects of etelcalcetide for sHPT treatment in patients receiving hemodialysis. METHODS: This 52-week, multicenter, single-arm OLE enrolled patients from three parent trials: two randomized, double-blind, placebo-controlled trials and one open-label, single-arm, 'switch' study from cinacalcet to etelcalcetide. The primary endpoint was to investigate the nature, frequency, severity and relation to treatment of all adverse events (AEs) reported throughout the trial. Secondary endpoints included the proportion of patients with >30% reduction from baseline in PTH and the percentage change from baseline in PTH, albumin-corrected calcium (Ca), phosphate (P) and the calcium-phosphate product (Ca × P).ClinicalTrials.gov identifier: NCT01785875; Amgen study: 20120231. RESULTS: Overall, 89.8% of the patients experienced one or more treatment-emergent AE. The most common were decreased blood Ca (43.3%), diarrhea (10.8%), vomiting (10.4%) and nausea (9.6%); symptomatic hypocalcemia occurred in 3.7% of the patients. Approximately 68% of patients achieved >30% reduction in PTH, and ∼56% achieved PTH ≤300 pg/mL. Mean percent changes from baseline ranged from -25.4% to -26.1% for PTH, -8.3% to -9.1% for Ca, -3.6% to -4.1% for P and -12.0% to -12.6% for Ca × P. CONCLUSIONS: Etelcalcetide effectively lowered PTH and its effect was sustained, while no new safety concerns emerged over a 1-year treatment period.


Assuntos
Hiperparatireoidismo Secundário/tratamento farmacológico , Peptídeos/administração & dosagem , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/complicações , Administração Intravenosa , Idoso , Cálcio/sangue , Método Duplo-Cego , Feminino , Humanos , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/patologia , Masculino , Hormônio Paratireóideo/sangue , Prognóstico , Insuficiência Renal Crônica/terapia
8.
Nephrol Dial Transplant ; 33(12): 2092-2100, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29733407

RESUMO

Recent improvements in our understanding of physiology have altered the way in which bone is perceived: no longer is it considered as simply the repository of divalent ions, but rather as a sophisticated endocrine organ with potential extraskeletal effects. Indeed, a number of pathologic conditions involving bone in different ways can now be reconsidered from a bone-centred perspective. For example, in metabolic bone diseases like osteoporosis (OP) and renal osteodystrophy (ROD), the association with a worse cardiovascular outcome can be tentatively explained by the possible derangements of three recently discovered bone hormones (osteocalcin, fibroblast growth factor 23 and sclerostin) and a bone-specific enzyme (alkaline phosphatase). Further, in recent years the close link between bone and inflammation has been better appreciated and a wide range of chronic inflammatory states (from rheumatoid arthritis to ageing) are being explored to discover the biochemical changes that ultimately lead to bone loss and OP. Also, it has been acknowledged that the concept of the bone-vascular axis may explain, for example, the relationship between bone metabolism and vessel wall diseases like atherosclerosis and arteriosclerosis, with potential involvement of a number of cytokines and metabolic pathways. A very important discovery in bone physiology is the bone marrow (BM) niche, the functional unit where stem cells interact, exchanging signals that impact on their fate as bone-forming cells or immune-competent haematopoietic elements. This new element of bone physiology has been recognized to be dysfunctional in diabetes (so-called diabetic mobilopathy), with possible clinical implications. In our opinion, ROD, the metabolic bone disease of renal patients, will in the future probably be identified as a cause of BM niche dysfunction. An integrated view of bone, which includes the BM niche, now seems necessary in order to understand the complex clinical entity of chronic kidney disease-mineral and bone disorders and its cardiovascular burden. Bone is thus becoming a recurrently considered paradigm for different inter-organ communications that needs to be considered in patients with complex diseases.


Assuntos
Doenças Ósseas Metabólicas/complicações , Medula Óssea/patologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Inflamação/complicações , Osteoporose/complicações , Insuficiência Renal Crônica/fisiopatologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Humanos
9.
Kidney Int ; 92(6): 1343-1355, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28964571

RESUMO

Mineral and bone disease is omnipresent in patients with chronic kidney disease (CKD) and leads to a diverse range of clinical manifestations, including bone pain and fractures. The accumulation of traditional clinical risk factors, in addition to those related to CKD, enhances the risk of comorbidity and mortality. Despite significant advances in understanding bone disease in CKD, most clinical and biochemical targets used in clinical practice remain controversial, resulting in an undermanagement of bone fragility. Vitamin D supplementation is widely used, but only a few studies have shown beneficial effects and a reduced risk of fracture and mortality. The achievement of serum levels of 25-hydroxyvitamin D is recommended for CKD patients to reduce a high parathyroid hormone level, which is associated with skeletal fractures. Optimal control of parathyroid hormone also improves bone mineralization and lowers circulating bone biomarkers such as alkaline phosphatase and cross-linked collagen type I peptide. The potential value of more recent biomarkers such as sclerostin and fibroblast growth factor 23, as surrogates for bone fragility, is an encouraging new direction in clinical research but is far from being firmly established. This article reviews the literature related to the pathophysiological role of various mineral and biochemical factors involved in renal osteodystrophy. To better understand bone fragility in CKD, new information related to the impact of disturbances of mineral metabolism on bone strength is urgently needed. The combined expertise of clinicians from various medical disciplines appears crucial for the most successful prevention of fractures in these patients.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/terapia , Fraturas Ósseas/prevenção & controle , Hormônio Paratireóideo/sangue , Insuficiência Renal Crônica/terapia , Vitamina D/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal , Biomarcadores/sangue , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/sangue , Proteínas Morfogenéticas Ósseas/sangue , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiopatologia , Calcificação Fisiológica/efeitos dos fármacos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/sangue , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fraturas Ósseas/sangue , Fraturas Ósseas/etiologia , Marcadores Genéticos , Humanos , Rim/metabolismo , Hormônio Paratireóideo/metabolismo , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Vitamina D/sangue
10.
Kidney Int ; 90(6): 1184-1190, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27653840

RESUMO

Circulating parathyroid hormone (PTH) shows a complex relationship with hard outcomes in subjects with chronic kidney disease (CKD). Moreover, intervention studies directly targeting PTH failed to yield unequivocal results. Disturbed PTH metabolism, posttranslational modifications of PTH, and end-organ hyporesponsiveness to PTH may explain the poor performance of PTH as an outcome biomarker and precise target of therapy in the setting of CKD, at least in the gray middle target zone. PTH fragments accumulate in CKD patients and may exert effects that are distinct from, if not opposite to biointact (1-84)PTH. Posttranslational modification of PTH and especially oxidation may alter the interaction of PTH with its receptor. Its clinical relevance, however, remains a matter of ongoing debate. Less controversial is the issue of end-organ hyporesponsiveness to PTH. This phenomenon, formally referred to as PTH resistance, has long been recognized in CKD, but factors and mechanisms contributing to it remain poorly defined. Subsequent evidence identified downregulation of the PTH receptor and competing downstream signals as underlying pathophysiologic mechanisms. End-organ hyporesponsiveness to PTH in CKD, along with important analytical and biological variability, renders defining the PTH target range in CKD challenging. Although this may still be accomplished at the population level, it may prove to be very difficult at the individual level. This is a disillusioning thought in an era of personalized medicine. Parallel to the search of a functional and readily available assay quantifying PTH signaling tone or sensitivity, additional biomarkers (or a panel of biomarkers) should be formally evaluated.


Assuntos
Hormônio Paratireóideo/metabolismo , Insuficiência Renal Crônica/metabolismo , Biomarcadores/metabolismo , Humanos
11.
Am J Kidney Dis ; 68(2): 247-255, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26724836

RESUMO

BACKGROUND: Recent randomized trials report that mortality is lower with high-convection-volume hemodiafiltration (HDF) than with hemodialysis (HD). STUDY DESIGN: We used data from the French national Renal Epidemiology and Information Network (REIN) registry to investigate trends in HDF use and its relationship with mortality in the total population of incident dialysis patients. SETTING & PARTICIPANTS: The study included those who initiated HD therapy from January 1, 2008, through December 31, 2011, and were dialyzed for more than 3 months; follow-up extended to the end of 2012. FACTOR: HDF use at the patient and facility level. OUTCOMES: All-cause and cardiovascular mortality, using Cox models to estimate HRs of HDF as time-dependent covariate at the patient level, with age as time scale and fully adjusted for comorbid conditions and laboratory data at baseline, catheter use, and facility type as time-dependent covariates. Analyses completed by Cox models for HRs of the facility-level exposure to HDF updated yearly. RESULTS: Of 28,407 HD patients, 5,526 used HDF for a median of 1.2 (IQR, 0.9-1.9) years; 2,254 of them used HDF exclusively. HRs for all-cause and cardiovascular mortality associated with HDF use were 0.84 (95% CI, 0.77-0.91) and 0.73 (95% CI, 0.61-0.88), respectively. In patients treated exclusively with HDF, these HRs were 0.77 (95% CI, 0.67-0.87) and 0.66 (95% CI, 0.50-0.86). At the facility level, increasing the percentage of patients using HDF from 0% to 100% was associated with HRs for all-cause and cardiovascular mortality of 0.87 (95% CI, 0.77-0.99) and 0.72 (95% CI, 0.54-0.96), respectively. LIMITATIONS: Observational study. CONCLUSIONS: Whether analyzed as a patient- or facility-level predictor, HDF treatment was associated with better survival.


Assuntos
Falência Renal Crônica/mortalidade , Falência Renal Crônica/terapia , Diálise Renal , Idoso , Idoso de 80 Anos ou mais , Feminino , França , Hemodiafiltração , Humanos , Serviços de Informação , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Sistema de Registros , Taxa de Sobrevida
12.
Nephrol Dial Transplant ; 31(8): 1211-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27005994

RESUMO

Calcific uraemic arteriolopathy (CUA), or calciphylaxis, is a rare disease predominantly occurring in comorbidity with dialysis. Due to the very low frequency of CUA, prospective studies on its management are lacking and even anecdotal reports on treatment remain scarce. Therefore, calciphylaxis is still a challenging disease with dismal prognosis urgently requiring adequate strategies for diagnosis and treatment.In an attempt to fill some of the current gaps in evidence on various, highly debated and controversial aspects of dialysis-associated calciphylaxis, 13 international experts joined the 1st Consensus Conference on CUA, held in Leuven, Belgium on 21 September 2015. The conference was supported by the European Calciphylaxis Network (EuCalNet), which is a task force of the ERA-EDTA scientific working group on Chronic Kidney Disease-Mineral and Bone Disorders (CKD-MBD). After an intense discussion, a 9-point Likert scale questionnaire regarding 20 items on calciphylaxis was anonymously answered by each participant. These 20 items addressed unsolved issues in terms of diagnosis and management of calciphylaxis. On the one hand, the analysis of the expert opinions identified areas of general consensus, which might be a valuable aid for physicians treating such a disease with less experience in the field. On the other hand, some topics such as the pertinence of skin biopsy and administration of certain treatments revealed divergent opinions. The aim of the present summary report is to provide some guidance for clinicians who face patients with calciphylaxis in the current setting of absence of evidence-based medicine.


Assuntos
Calciofilaxia/patologia , Calciofilaxia/prevenção & controle , Avaliação das Necessidades , Gerenciamento Clínico , Medicina Baseada em Evidências , Humanos
13.
Nephrol Dial Transplant ; 30(3): 345-51, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25712934

RESUMO

It is increasingly acknowledged that mineral and bone disorders (MBDs) contribute to the excessively high cardiovascular (CV) disease morbidity and mortality observed in patients with chronic kidney disease (CKD). There is ongoing debate as to whether screening for CV calcification, one of the hallmarks of CKD-MBD, should be implemented in clinical practice in patients with CKD. Issues to be considered in this controversy relate to prevalence, severity, relevance, and last but not least, modifiability and reversibility of vascular and valvular calcifications in the setting of CKD. The recent expansion of the armamentarium to treat CKD-MBD (calcium-free phosphate binders and calcimimetics) creates new opportunities. Mounting experimental and clinical evidence indicates that progression of CV calcification may indeed be attenuated. Whether this will translate into better outcomes remains to be proven. We acknowledge that hard outcome data so far are limited and, overall, yielded inconclusive results. Nevertheless, in an era in which personalized medicine has gained much popularity, we consider it reasonable, awaiting the results of additional studies, to screen for CV calcification in selected individuals. This policy may help to stratify CV risk and to guide therapy. We speculate that such an approach will ultimately improve outcomes and reduce health costs.


Assuntos
Doenças Ósseas/fisiopatologia , Calcinose/complicações , Doenças Cardiovasculares/etiologia , Minerais/metabolismo , Insuficiência Renal Crônica/fisiopatologia , Progressão da Doença , Humanos
14.
Rheumatology (Oxford) ; 53(3): 547-51, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24292346

RESUMO

OBJECTIVE: The objective of this study was to describe the efficacy of sodium thiosulphate (STS) in tumoral calcinosis (TC). METHODS: The methodology involved the reporting of four retrospective case reports of TC complicating end-stage renal disease (ESRD). RESULTS: We investigated STS treatment in four patients (two men; ages 46-70 years) with TC. ESRD was secondary to nephronophthisis (n = 1), membranoproliferative glomerulonephritis (n = 1), diabetic nephropathy (n = 1), and thrombotic microangiopathy (n = 1). TC developed 3-28 years after dialysis began and resulted in articular pain (n = 4) and stiffness (n = 1). It involved shoulders and hips and was diffuse in one patient. Several treatments were tried without success. STS 12.5-25 g was given intravenously after each dialysis session for 11-14 months. Pain and stiffness rapidly disappeared and TC showed partial or total regression. Side effects during infusion included increased blood pressure (n = 1), nausea (n = 1) and vomiting (n = 1). TC did not recur after treatment discontinuation with follow-up of 1.5-12 years. CONCLUSION: STS showed promising efficacy in this short series of TC. Further studies are warranted.


Assuntos
Calcinose/tratamento farmacológico , Calcinose/epidemiologia , Nefropatias/tratamento farmacológico , Nefropatias/epidemiologia , Tiossulfatos/uso terapêutico , Uremia/tratamento farmacológico , Uremia/epidemiologia , Idoso , Calcinose/etiologia , Comorbidade , Feminino , Humanos , Hipertensão/induzido quimicamente , Hipertensão/epidemiologia , Incidência , Nefropatias/etiologia , Falência Renal Crônica/complicações , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Náusea/epidemiologia , Diálise Renal , Estudos Retrospectivos , Tiossulfatos/efeitos adversos , Resultado do Tratamento , Vômito/induzido quimicamente , Vômito/epidemiologia
15.
Nephrol Dial Transplant ; 29(10): 1815-20, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24516228

RESUMO

The concept of chronic kidney disease-mineral bone disorder (CKD-MBD) does not appear to fulfil the requirements for a syndrome at first glance, but its definition has brought some clear-cut benefits for clinicians and patients, including wider and more complex diagnostic and therapeutic approaches to the management of this challenging set of issues. Admittedly, not all components of CKD-MBD are present in all patients at all times, but these are highly interrelated, involving mineral and bone laboratory abnormalities, clinical and histological bone disease and finally, cardiovascular disease. The presence of typical biological bone ossification processes in an ectopic anatomical location in CKD has helped to define the existence of an unprecedented bone-vascular relationship, extending its interest even to other medical specialities. For now, we believe that CKD-MBD does not reach full criteria to be defined as a syndrome. However, this novel concept has clearly influenced current clinical guidelines. The National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF/KDOQI™) guidelines in 2003 for instance recommended that calcium-based phosphate binders should be avoided to treat hyperphosphataemia in the presence of cardiovascular calcifications. In 2009, the KDIGO and other guidelines reinforced and extended this recommendation by stating that it is reasonable to choose oral phosphate binder therapy by taking into consideration other components of CKD-MBD. Similarly, it is also considered reasonable to use information on vascular/valvular calcification to guide the management of CKD-MBD. Our current assumption as a working group 'CKD-MBD' is that CKD-MBD has the potential to be defined a true syndrome, such as a constellation of concurrent signs and symptoms that suggest a common underlying mechanism for these components as opposed to the term disease. The term 'syndrome' also implies that in any patient at risk due to the presence of one or a few components of the entire syndrome, the screening for additional components is highly recommended. However, it has not currently been demonstrated that there is an additive predictive value, which can be derived from identifying individual components. Despite all we have learned about this putative syndrome, we have been left with only a hypothetical framework about how to treat patients. So while we agree that the concept of CKD-MBD has influenced, and continues to influence, our current clinical hypotheses, definitive proof of a benefit of interventions in CKD-MBD is still lacking and a global-multiple therapeutic approach to treat simultaneously several components of CKD-MBD should be tested by well-designed new randomized controlled trials.


Assuntos
Densidade Óssea , Doenças Ósseas Metabólicas/patologia , Calcinose/patologia , Hiperfosfatemia/patologia , Insuficiência Renal Crônica/patologia , Doenças Ósseas Metabólicas/metabolismo , Calcinose/metabolismo , Humanos , Hiperfosfatemia/metabolismo , Insuficiência Renal Crônica/metabolismo , Síndrome
18.
BMC Nephrol ; 15: 71, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24885942

RESUMO

BACKGROUND: High levels of circulating fibroblast growth factor 23 (FGF23) are associated with chronic kidney disease (CKD) progression and high mortality. In the Phosphate Reduction Evaluation of FGF23 in Early CKD Treatment (PREFECT) study, we assessed the effect of reducing intestinal phosphate absorption using lanthanum carbonate on FGF23 levels in normophosphatemic patients with CKD stage 3. METHODS: Thirty-five individuals were randomized to lanthanum carbonate 3000 mg/day (n=23) or placebo (n=12) for 12 weeks. Levels of intact FGF23 (iFGF23), C-terminal FGF23, serum and urinary phosphate and calcium, intact parathyroid hormone and 1,25-dihydroxyvitamin D were assessed. RESULTS: The median age was 65 years in the lanthanum group and 73 years in the placebo group; 58.8% and 41.7% were men, respectively. No significant difference was seen in mean iFGF23 between groups at week 12. There was, however, a transient reduction from baseline in iFGF23 in the lanthanum group at week 1, from 70.5 pg/ml to 51.9 pg/ml, which was not seen in the placebo group; this between-group difference in percentage change from baseline was significant in post hoc analyses (p=0.0102). Urinary phosphate decreased after 1 week of lanthanum treatment and remained low at week 12. CONCLUSIONS: Reducing intestinal phosphate absorption with lanthanum carbonate did not lead to sustained reductions in iFGF23 in patients with CKD stage 3, although phosphaturia decreased. This suggests that factors other than phosphate burden may be responsible for driving increases in circulating FGF23 in patients with CKD. TRIAL REGISTRATION: ClinicalTrials.gov NCT01128179, 20 May 2010.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , Lantânio/uso terapêutico , Fósforo/sangue , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Método Duplo-Cego , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino , Pessoa de Meia-Idade , Efeito Placebo , Valores de Referência , Insuficiência Renal Crônica/diagnóstico , Resultado do Tratamento
19.
Kidney Int ; 93(5): 1247, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29680023
20.
Kidney Int ; 93(5): 1248-1249, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29680024
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa