Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 69
1.
FASEB J ; 38(11): e23726, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38847773

Calcitriol and calcimimetics are used to treat hyperparathyroidism secondary to chronic kidney disease (CKD). Calcitriol administration and the subsequent increase in serum calcium concentration decrease parathyroid hormone (PTH) levels, which should reduce bone remodeling. We have previously reported that, when maintaining a given concentration of PTH, the addition of calcimimetics is associated with an increased bone cell activity. Whether calcitriol administration affects bone cell activity while PTH is maintained constant should be evaluated in an animal model of renal osteodystrophy. The aim of the present study was to compare in CKD PTH-clamped rats the bone effects of calcitriol and calcimimetic administration. The results show that the administration of calcitriol and calcimimetic at doses that induced a similar reduction in PTH secretion produced dissimilar effects on osteoblast activity in 5/6 nephrectomized (Nx) rats with secondary hyperparathyroidism and in Nx rats with clamped PTH. Remarkably, in both rat models, the administration of calcitriol decreased osteoblastic activity, whereas calcimimetic increased bone cell activity. In vitro, calcitriol supplementation inhibited nuclear translocation of ß-catenin and reduced proliferation, osteogenesis, and mineralization in mesenchymal stem cells differentiated into osteoblasts. In conclusion, besides the action of calcitriol and calcimimetics at parathyroid level, these treatments have specific effects on bone cells that are independent of the PTH level.


Calcimimetic Agents , Calcitriol , Osteoblasts , Parathyroid Hormone , Animals , Calcitriol/pharmacology , Rats , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Parathyroid Hormone/pharmacology , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , Rats, Wistar , Renal Insufficiency/drug therapy , Renal Insufficiency/metabolism , Osteogenesis/drug effects , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/complications , Cell Differentiation/drug effects , Calcium/metabolism
2.
Bone ; 185: 117126, 2024 Aug.
Article En | MEDLINE | ID: mdl-38777312

Chronic kidney disease-induced secondary hyperparathyroidism (CKD-SHPT) heightens fracture risk through impaired mineral homeostasis and elevated levels of uremic toxins (UTs), which in turn enhance bone remodeling. Etelcalcetide (Etel), a calcium-sensing receptor (CaSR) agonist, suppresses parathyroid hormone (PTH) in hyperparathyroidism to reduce excessive bone resorption, leading to increased bone mass. However, Etel's effect on bone quality, chemical composition, and strength is not well understood. To address these gaps, we established a CKD-SHPT rat model and administered Etel at a human-equivalent dose concurrently with disease induction. The effects on bone and mineral homeostasis were compared with a CKD-SHPT (vehicle-treated group) and a control group (rats without SHPT). Compared with vehicle-treated CKD-SHPT rats, Etel treatment improved renal function, reduced circulating UT levels, improved mineral homeostasis parameters, decreased PTH levels, and prevented mineralization defects. The upregulation of mineralization-promoting genes by Etel in CKD-SHPT rats might explain its ability to prevent mineralization defects. Etel preserved both trabecular and cortical bones with attendant suppression of osteoclast function, besides increasing mineralization. Etel maintained the number of viable osteocytes to the control level, which could also contribute to its beneficial effects on bone. CKD-SHPT rats displayed increased carbonate substitution of matrix and mineral, decreased crystallinity, mineral-to-matrix ratio, and collagen maturity, and these changes were mitigated by Etel. Further, Etel treatment prevented CKD-SHPT-induced deterioration in bone strength and mechanical behavior. Based on these findings, we conclude that in CKD-SHPT rats, Etel has multiscale beneficial effects on bone that involve remodeling suppression, mineralization gene upregulation, and preservation of osteocytes.


Bone and Bones , Calcimimetic Agents , Hyperparathyroidism, Secondary , Peptides , Rats, Sprague-Dawley , Renal Insufficiency, Chronic , Animals , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/pathology , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Peptides/pharmacology , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Rats , Parathyroid Hormone/pharmacology , Male , Calcification, Physiologic/drug effects , Bone Density/drug effects
3.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5005-5013, 2024 Jul.
Article En | MEDLINE | ID: mdl-38183449

Cinacalcet is a calcimimetic medicine that has been used to treat secondary hyperparathyroidism and parathyroid cancer. Various studies have proposed the positive role of calcium and its receptor in skin wound healing. Furthermore, Cinacalcet interacts with other skin repair-related mechanisms, including inflammation and nitric oxide pathways. The present study evaluated the effect of Cinacalcet on the random-pattern skin flap survival. Eighty-four Wistar male rats were used. Multiple doses of Cinacalcet (30, 3, 1, 0.3, and 0.05 mg/kg) were used in 3 different routes of administration before the surgery. Histopathological evaluations, quantitative assessment of IL-6, TNF-α, and nitric oxide (NO), and the expression of calcium-sensing receptor (CaSR) and E-cadherin were evaluated in the skin tissue. To assess the role of NO, a NO synthase inhibitor, N-nitro-L-arginine methyl ester hydrochloride (L-NAME), was used, and histopathological effects were investigated. Cinacalcet pretreatment at the IP chronic 1 mg/kg dose significantly increased the skin flap survival rate and enhanced the NO tissue level compared to the control. However, the administration of L-NAME abolished its protective effects. IP Chronic 1 mg/kg of Cinacalcet could also decline the levels of IL-6 and TNF-α and also increase the expression of CaSR and E-cadherin in the flap tissue compared with the control group. Chronic Cinacalcet at 1 mg/kg could improve skin flap survival, probably mediated by the CaSR, NO, and inflammation-related pathways.


Cadherins , Calcimimetic Agents , Cinacalcet , Interleukin-6 , Nitric Oxide , Rats, Wistar , Receptors, Calcium-Sensing , Skin , Animals , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Male , Nitric Oxide/metabolism , Calcimimetic Agents/pharmacology , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/antagonists & inhibitors , Interleukin-6/metabolism , Cadherins/metabolism , Skin/metabolism , Skin/drug effects , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism , Rats , Surgical Flaps/pathology , Wound Healing/drug effects , Signal Transduction/drug effects , Graft Survival/drug effects
4.
Eur J Pharmacol ; 956: 175936, 2023 Oct 05.
Article En | MEDLINE | ID: mdl-37541363

Upacicalcet (formerly SK-1403/AJT240) is a novel non-peptide calcimimetic agent that acts as a calcium-sensing receptor (CaSR) agonist for the treatment of secondary hyperparathyroidism (SHPT) in chronic kidney disease (CKD). We compared upacicalcet with other calcimimetics (etelcalcetide or cinacalcet) and examined its in vitro and in vivo characteristics in terms of its human CaSR agonistic activity, its efficacy in normal and CKD rats after a single administration, and its effect on gastric emptying in rats. Upacicalcet activated human CaSR depending on the extracellular calcium (Ca2+) concentration without exhibiting an agonistic action when the extracellular Ca2+ level was below the physiological level. On the other hand, etelcalcetide had an agonistic activity even in the absence of physiological levels of extracellular Ca2+. The intravenous administration of upacicalcet to normal and double-nephrectomized rats dose-dependently (0.03-3mg/kg and 0.3-30mg/kg, respectively) decreased the serum intact parathyroid hormone (iPTH) and serum Ca2+ levels; however, the effect of upacicalcet on the reduction in serum Ca2+ disappeared at extracellular Ca2+ levels below the physiologically range, even when administered at a dose higher (100-fold) than the effective dose. Furthermore, upacicalcet did not affect gastric emptying in normal rats when administered up to a dose of 10mg/kg (300-fold higher than the dose affecting serum iPTH levels), while the administration of cinacalcet significantly slowed gastric emptying by approximately 50%. These findings suggest that upacicalcet has potential as an alternative calcimimetic agent with good pharmacological properties and a lower risk of hypocalcemia and gastrointestinal complications.


Hyperparathyroidism, Secondary , Renal Insufficiency, Chronic , Humans , Rats , Animals , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Receptors, Calcium-Sensing/agonists , Parathyroid Hormone , Calcium , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Renal Dialysis/adverse effects
5.
Drugs ; 81(13): 1593-1596, 2021 Sep.
Article En | MEDLINE | ID: mdl-34390486

Upacicalcet (UPASITA®) is an intravenous calcimimetic agent being developed by Sanwa Kagaku Kenkyusho, under license from EA Pharma, for the treatment of secondary hyperparathyroidism (SHPT), a common and early complication of chronic kidney disease, in patients undergoing haemodialysis. By acting directly on parathyroid cell membrane calcium-sensing receptors, upacicalcet suppresses excessive parathyroid hormone (PTH) secretion, thereby lowering blood PTH levels. Upacicalcet received its first approval on 23 June 2021 for the treatment of SHPT in adults undergoing haemodialysis in Japan. It is administered intravenously three times per week into the venous side of the haemodialysis circuit at the time of blood return at the end of the haemodialysis session. The generally recommended starting dose of upacicalcet is 25 µg, with the dose adjusted within a 25-300 µg range based on PTH and serum calcium levels. This article summarizes the milestones in the development of upacicalcet leading to this first approval for the treatment of SHPT in patients undergoing haemodialysis.


Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Propionates/pharmacology , Propionates/therapeutic use , Calcimimetic Agents/pharmacokinetics , Drug Approval , Humans , Propionates/pharmacokinetics
6.
Brain Res ; 1765: 147508, 2021 08 15.
Article En | MEDLINE | ID: mdl-33930376

Cerebral vasospasm (CVS) causes mortality and morbidity in patients after subarachnoid hemorrhage (SAH). The mechanism and adequate treatment of CVS are still elusive. R-568 is a calcimimetic agent known to exert a vasodilating effect. However, there is no report on its vasodilator effect against SAH-induced vasospasm. In the present study, we investigated the therapeutic effect of R-568 on the SAH-induced CVS model in rats. Seventy-two adult male Sprague-Dawley rats were divided into 8 groups: sham surgery; SAH only; SAH + Vehicle, SAH + R-568; SAH + R-568 + Wortmannin (the PI3K inhibitor); SAH + Wortmannin; SAH + R-568 + Calhex-231 (a calcilytic agent); SAH + Calhex-231. SAH was induced by blood (0.3 mL) given by intracisternal injection. R-568 (20 µM) was administered intracisternal immediately prior to experimental SAH. Basilar arteries (BAs) were obtained to evaluate PI3K/Akt/eNOS pathway (immunoblotting) and morphological changes 48 h after SAH. Perimeters of BAs were decreased by 24.1% in the SAH group compared to the control group and the wall thickness was increased by 75.3%. With R-568 treatment, those percentages were 9.6% and 29.6%, respectively, indicating that vasospasm was considerably improved when compared with the SAH group (P < 0.001 in both). While p-PI3K/PI3K and p-Akt/Akt ratio and eNOS protein expression were markedly decreased in the SAH rats, treatment with R-568 resulted in a significant increase in these levels. The beneficial effects of R-568 were partially blocked in the presence of Calhex-231 and completely blocked in the presence of Wortmannin. Herein, we found that treatment with R-568 would attenuate SAH-induced CVS through the PI3K/Akt/eNOS pathway and demonstrate therapeutic promise in CVS treatment following SAH.


Phenethylamines/pharmacology , Propylamines/pharmacology , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/drug therapy , Animals , Calcimimetic Agents/pharmacology , Disease Models, Animal , Male , Nitric Oxide Synthase Type III/metabolism , Phenethylamines/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Propylamines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/metabolism
7.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118836, 2020 12.
Article En | MEDLINE | ID: mdl-32861746

The calcium-sensing receptor (CaSR) is a ubiquitously expressed multifunctional G protein-coupled receptor. Several studies reported that the CaSR plays an anti-inflammatory and anti-tumorigenic role in the intestine, and that it is down-regulated during colorectal carcinogenesis. We hypothesized that positive allosteric CaSR modulators (type II calcimimetics) selectively targeting the intestinal cells could be used for the treatment of intestinal pathologies. Therefore, the aim of this study was to determine the effect of pharmacological stimulation of CaSR on gene expression in vitro and on tumor growth in vivo. We stably transduced two colon cancer cell lines (HT29 and Caco2) with lentiviral vectors containing either the CaSR fused to GFP or GFP only. Using RNA sequencing, RT-qPCR experiments and ELISA, we determined that CaSR over-expression itself had generally little effect on gene expression in these cells. However, treatment with 1 µM of the calcimimetic NPS R-568 increased the expression of pro-inflammatory factors such as IL-23α and IL-8 and reduced the transcription of various differentiation markers in the cells over-expressing the CaSR. In vivo, neither the presence of the CaSR nor p.o. treatment of the animals with the calcimimetic cinacalcet affected tumor growth, tumor cell proliferation or tumor vascularization of murine HT29 xenografts. In summary, CaSR stimulation in CaSR over-expressing cells enhanced the expression of inflammatory markers in vitro, but was not able to repress colorectal cancer tumorigenicity in vivo. These findings suggest potential pro-inflammatory effects of the CaSR and type II calcimimetics in the intestine.


Calcimimetic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Receptors, Calcium-Sensing/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Caco-2 Cells , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Interleukin-23 Subunit p19/genetics , Interleukin-8/genetics , Mice , Phenethylamines/pharmacology , Propylamines/pharmacology
8.
Ther Apher Dial ; 24(3): 248-257, 2020 Jun.
Article En | MEDLINE | ID: mdl-31486206

Patients with chronic kidney disease often develop secondary hyperparathyroidism (SHPT), marked by high levels of circulating parathyroid hormone (PTH) and increased risk of morbidity and mortality. Patients with SHPT are treated with a therapeutic combination that commonly includes calcimimetics, which have recently become popular in clinical settings, and other agents such as vitamin D preparations. Calcimimetics are a drug class that reduces PTH levels by targeting the calcium-sensing receptor. Cinacalcet, a representative calcimimetic, is widely used; however, a high incidence of upper gastrointestinal (GI) tract-related adverse events (AEs) can result in insufficient dosage and poor long-term compliance. The newly approved evocalcet has equivalent efficacy to cinacalcet at a lower clinical dose, with improved bioavailability, fewer upper GI tract-related AEs, and fewer safety concerns. This review gives an overview of calcimimetic agents, with a special focus on evocalcet, and describes the clinical advantages of evocalcet in the treatment of dialysis patients with SHPT.


Hyperparathyroidism, Secondary , Naphthalenes/pharmacology , Pyrrolidines/pharmacology , Renal Insufficiency, Chronic , Calcimimetic Agents/pharmacology , Drug Discovery , Humans , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Receptors, Calcium-Sensing/antagonists & inhibitors , Renal Dialysis/methods , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Treatment Outcome
9.
Kidney Blood Press Res ; 44(6): 1327-1338, 2019.
Article En | MEDLINE | ID: mdl-31747666

OBJECTIVE: To assess the long-term effects including all-cause mortality, cardiovascular mortality, and fracture incidence, of cinacalcet on secondary hyperparathyroidism (SHPT) in patients on dialysis. METHODS: PubMed, Embase, and the Cochrane Central Register of Controlled Trials were searched from their inception to October 2018. Randomized controlled trials (RCTs) and cohort design prospective observational studies assessing cinacalcet for the treatment of SHPT in dialysis patients were included. Data extraction was independently completed by 2 authors who determined the methodological quality of the studies and extracted data in duplicate. Study-specific risk estimates were tested by using a fixed effects model. RESULTS: A total of 14 articles with 38,219 participants were included, of which 10 RCTs with 7,471 participants and 4 prospective observational studies with 30,748 participants fulfilled the eligibility criteria. Compared with no cinacalcet, cinacalcet administration reduced all-cause mortality (relative risk [RR] 0.91, 95% CI 0.89-0.94, p < 0.001) and cardiovascular mortality (RR 0.92, 95% CI 0.89-0.95, p < 0.001), but it did not significantly reduce the incidence of fractures (RR 0.93, 95% CI 0.87-1.00, p = 0.05). CONCLUSIONS: The results of this meta-analysis indicated that the treatment of SHPT with cinacalcet may in fact reduce all-cause mortality and cardiovascular mortality among patients receiving maintenance dialysis.


Cinacalcet/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Kidney Failure, Chronic/therapy , Survival Rate , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Cinacalcet/pharmacology , Fractures, Bone/prevention & control , Humans , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/mortality , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/mortality , Renal Dialysis/mortality
10.
Am J Physiol Renal Physiol ; 317(2): F240-F253, 2019 08 01.
Article En | MEDLINE | ID: mdl-31042050

To garner insights into the renal regulation of Ca2+ homeostasis, we performed an mRNA microarray on kidneys from mice treated with the Ca2+-sensing receptor (CaSR) agonist cinacalcet. This revealed decreased gene expression of Na+/H+ exchanger isoform 8 (NHE8) in response to CaSR activation. These results were confirmed by quantitative real-time PCR. Moreover, administration of vitamin D also decreased NHE8 mRNA expression. In contrast, renal NHE8 protein expression from the same samples was increased. To examine the role of NHE8 in transmembrane Ca2+ fluxes, we used the normal rat kidney (NRK) cell line. Cell surface biotinylation and confocal immunofluorescence microscopy demonstrated NHE8 apical expression. Functional experiments found 5-(N-ethyl-N-isopropyl)amiloride (EIPA)-inhibitable NHE activity in NRK cells at concentrations minimally attenuating NHE1 activity in AP-1 cells. To determine how NHE8 might regulate Ca2+ balance, we measured changes in intracellular Ca2+ uptake by live cell Ca2+ imaging with the fluorophore Fura-2 AM. Inhibition of NHE8 with EIPA or by removing extracellular Na+-enhanced Ca2+ influx into NRK cells. Ca2+ influx was mediated by a voltage-dependent Ca2+ channel rather than directly via NHE8. NRK cells express Cav1.3 and display verapamil-sensitive Ca2+ influx and NHE8 inhibition-augmented Ca2+ influx via a voltage-dependent Ca2+ channel. Finally, proximal tubules perused ex vivo demonstrated increased Ca2+ influx in the presence of luminal EIPA at a concentration that would inhibit NHE8. The results of the present study are consistent with NHE8 regulating Ca2+ uptake into the proximal tubule epithelium.


Calcium Signaling , Calcium/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , CHO Cells , Calcimimetic Agents/pharmacology , Calcium Channels/metabolism , Cinacalcet/pharmacology , Cricetulus , Epithelial Cells/drug effects , Homeostasis , Kidney Tubules, Proximal/drug effects , Mutation , Rats , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Sodium-Hydrogen Exchanger 1/genetics , Sodium-Hydrogen Exchanger 1/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics
11.
Am J Physiol Renal Physiol ; 316(5): F1006-F1015, 2019 05 01.
Article En | MEDLINE | ID: mdl-30838870

316: F1006-F1015, 2019. First published March 6, 2019; doi: 10.1152/ajprenal.00413.2018 .-Experimental studies have shown that pharmacological activation of calcium-sensing receptor (CaSR) attenuates renal fibrosis in some animal models beyond modification of bone and mineral homeostasis; however, its underlying mechanisms remain largely unknown. Since excessive collagen deposition is the key feature of fibrosis, the present study aimed to examine whether CaSR was involved in the regulation of collagen expression in rats with adenine diet-induced renal fibrosis and in profibrotic transforming growth factor (TGF)-ß1-treated renal proximal tubular epithelial cells (PTECs). The results showed that the CaSR agonist cinacalcet significantly attenuated renal collagen accumulation and tubular injury in adenine diet-fed rats. Additionally, the in vitro experiment showed that profibrotic TGF-ß1 significantly increased the expression of collagen and decreased CaSR expression at the mRNA and protein levels in a concentration- and time-dependent manner. Furthermore, the CaSR CRISPR activation plasmid and cinacalcet partially abrogated the upregulation of collagen induced by TGF-ß1 treatment. Blockade of CaSR by the CRISPR/Cas9 KO plasmid or the pharmacological antagonist Calhex231 further enhanced TGF-ß1-induced collagen expression. Mechanistic experiments found that Smad2 phosphorylation and Snail expression were markedly increased in PTECs treated with TGF-ß1, whereas the CaSR CRISPR activation plasmid and cinacalcet substantially suppressed this induction. In summary, this study provides evidence for a direct renal tubular epithelial protective effect of CaSR activation in renal fibrosis, possibly through suppression of collagen expression in PTECs.


Calcimimetic Agents/pharmacology , Cinacalcet/pharmacology , Collagen/metabolism , Epithelial Cells/drug effects , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/drug effects , Receptors, Calcium-Sensing/agonists , Adenine , Animals , Benzamides/pharmacology , CRISPR-Cas Systems , Cells, Cultured , Cyclohexylamines/pharmacology , Disease Models, Animal , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Phosphorylation , Rats, Wistar , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Smad2 Protein/metabolism , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology
12.
Pediatr Nephrol ; 34(3): 413-422, 2019 03.
Article En | MEDLINE | ID: mdl-29552709

The calcium-sensing receptor (CaSR) plays an important role in the homeostasis of serum ionized calcium by regulating parathyroid hormone (PTH) secretion and tubular calcium handling. Calcimimetics, which act by allosteric modulation of the CaSR, mimic hypercalcemia resulting in suppression of PTH release and increase in calciuria. Mostly used in children to treat secondary hyperparathyroidism associated with advanced renal failure, we have shown that calcimimetics can also be successfully used in children with bone and mineral disorders in which elevated PTH plays a detrimental role in skeletal pathophysiology in the face of normal kidney function. The current review briefly discusses the role of the CaSR and calcimimetics in calcium homeostasis, and then addresses the potential applications of calcimimetics in children with normal kidney function with disorders in which suppression of PTH is beneficial.


Bone Diseases, Metabolic/drug therapy , Calcimimetic Agents/pharmacology , Hyperparathyroidism/drug therapy , Parathyroid Hormone/antagonists & inhibitors , Receptors, Calcium-Sensing/metabolism , Allosteric Regulation/drug effects , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/urine , Calcimimetic Agents/therapeutic use , Calcium/blood , Calcium/metabolism , Calcium/urine , Child , Humans , Hyperparathyroidism/blood , Hyperparathyroidism/urine , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Renal Elimination/drug effects , Treatment Outcome , Vitamin D/metabolism
13.
Clin Exp Nephrol ; 23(2): 258-267, 2019 Feb.
Article En | MEDLINE | ID: mdl-30159688

BACKGROUND: This study investigated the pharmacokinetics, pharmacodynamics, and safety of multiple doses of evocalcet in Japanese secondary hyperparathyroidism (SHPT) patients receiving hemodialysis. METHODS: In this multicenter, open-label study, conducted between August 2013 and March 2014, 27 patients received multiple doses of 1 and 4 mg evocalcet for 14 days, followed by an extension period of multiple doses of 8 and 12 mg evocalcet for 7 days using an intra-patient dose escalation protocol. Pharmacodynamic parameters consisted of measurement of intact parathyroid hormone (PTH), serum-corrected calcium, serum phosphorus and intact fibroblast growth factor 23 concentrations. Safety was assessed by analysis of adverse events. RESULTS: Plasma evocalcet levels reached steady state 3 days after the first day of administration. Pharmacodynamic analyses showed that evocalcet effectively reduced intact PTH and serum-corrected calcium levels. Adverse events (AEs) occurred in 29.6 and 62.5% of patients receiving multiple doses of 1 or 4 mg, respectively. The AE 'blood calcium decreased' occurred in eight patients (33.0%) after multiple doses of 4 mg. All events were mild, except for one patient with a moderate AE (abnormal liver function) and one patient with a severe adverse drug reaction (blood calcium decreased). CONCLUSION: Multiple doses of evocalcet reduced intact PTH levels with a concomitant decrease in serum calcium levels. Evocalcet was well tolerated in SHPT patients receiving hemodialysis.


Calcimimetic Agents , Hyperparathyroidism, Secondary/drug therapy , Naphthalenes , Pyrrolidines , Renal Dialysis , Renal Insufficiency, Chronic/therapy , Adult , Aged , Calcimimetic Agents/administration & dosage , Calcimimetic Agents/adverse effects , Calcimimetic Agents/pharmacokinetics , Calcimimetic Agents/pharmacology , Calcium/blood , Drug Administration Schedule , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Humans , Hyperparathyroidism, Secondary/blood , Hyperparathyroidism, Secondary/diagnosis , Hyperparathyroidism, Secondary/etiology , Japan , Male , Middle Aged , Naphthalenes/administration & dosage , Naphthalenes/adverse effects , Naphthalenes/pharmacology , Parathyroid Hormone/blood , Phosphorus/blood , Pyrrolidines/administration & dosage , Pyrrolidines/adverse effects , Pyrrolidines/pharmacology , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/diagnosis , Treatment Outcome , Young Adult
14.
Ther Apher Dial ; 23(4): 309-318, 2019 Aug.
Article En | MEDLINE | ID: mdl-30411503

The classic pathogenesis of secondary hyperparathyroidism (SHPT) began with the trade-off hypothesis based on parathyroid hormone hypersecretion brought about by renal failure resulting from a physiological response to correct metabolic disorder of calcium, phosphorus, and vitamin D. In dialysis patients with failed renal function, physiological mineral balance control by parathyroid hormone through the kidney fails and hyperparathyroidism progresses. In this process, many significant genetic findings have been established. Abnormalities of Ca-sensing receptor and vitamin D receptor are associated with the pathogenesis of SHPT, and fibroblast growth factor 23 has also been shown to be involved in the pathogenesis. Vitamin D receptor activators (VDRAs) are widely used for treatment of SHPT. However, VDRAs have calcemic and phosphatemic effects that limit their use to a subset of patients, and calcimimetics have been developed as alternative drugs for SHPT. Hyperphosphatemia also affects progression of SHPT, and control of hyperphosphatemia is, therefore, thought to be fundamental for control of SHPT. Currently, a combination of a VDRA and a calcimimetic is recognized as the optimal strategy for SHPT, and for other outcomes such as reduced cardiovascular disease and improved survival. The latest findings on the pathogenesis and treatment of SHPT are summarized in this review.


Hyperparathyroidism, Secondary , Parathyroid Hormone/metabolism , Renal Dialysis , Renal Insufficiency , Bone Density Conservation Agents/pharmacology , Calcimimetic Agents/pharmacology , Calcium/metabolism , Humans , Hyperparathyroidism, Secondary/diagnosis , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/metabolism , Hyperparathyroidism, Secondary/therapy , Phosphorus/metabolism , Renal Dialysis/adverse effects , Renal Dialysis/methods , Renal Insufficiency/complications , Renal Insufficiency/therapy , Vitamin D/metabolism
15.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3585-3594, 2018 11.
Article En | MEDLINE | ID: mdl-30251678

Obesity is a major current public health problem worldwide due to the severe co-morbid conditions that this disease entails. The development of obesity-related cardiometabolic disorders is in direct association with adipose tissue inflammation that leads to its functional impairment. Activation of the Calcium-Sensing Receptor (CaSR) in adipose tissue contributes to inflammation and adipose dysfunction. Autophagy, a process of cell component degradation, is closely related to inflammation in many diseases, however, whether autophagy is associated with CaSR-induced inflammation remains unknown. Using LS14 and SW872 preadipose cell lines as well as primary human preadipocytes, we show that CaSR activation with the allosteric activator cinacalcet induces autophagosome formation. Cinacalcet-induced LC3II content elevation was precluded by knockdown of the CaSR and enhanced by CaSR overexpression, indicating a specific effect. Autophagy inhibition using 3-methyladenine prevented CaSR-induced TNFα production, indicating that autophagy contributes to CaSR-induced inflammation in human preadipocytes. Our results suggest that modulation of CaSR-induced autophagy is an attractive target in obese inflamed adipose tissue, to prevent the development of diseases triggered by adipose dysfunction. We describe a novel mechanism and possible new target to modulate and prevent adipose inflammation and hence the resulting disease-generating adipose tissue dysfunction.


Adipose Tissue/pathology , Autophagy , Inflammation/pathology , Receptors, Calcium-Sensing/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Calcimimetic Agents/pharmacology , Cell Line , Cinacalcet/pharmacology , Gene Knockdown Techniques , Humans , Inflammation/etiology , Obesity/complications , Obesity/metabolism , Primary Cell Culture , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics
17.
Ther Apher Dial ; 22(3): 242-245, 2018 Jun.
Article En | MEDLINE | ID: mdl-29767854

Anemia is a common complication of chronic kidney disease (CKD). There are various causes of renal anemia such as decreased production of erythropoietin, resistance to erythropoietin, shortened survival of red blood cells, and bone marrow fibrosis. Secondary hyperparathyroidism (SHPT) is a less recognized, but potentially significant cause of renal anemia in CKD patients. Parathyroid hormone (PTH) has been regarded as a uremic toxin that has multiple adverse effects, and its elevated levels have been associated with renal anemia in hemodialysis patients. Moreover, recent clinical studies have shown that the treatment of SHPT using either vitamin D receptor activators, calcimimetics, or parathyroidectomy leads to improvement of anemia, supporting the role of PTH in renal anemia. Emerging data have also indicated the involvement of bone-derived fibroblast growth factor 23 in renal anemia. This review summarizes recent insights into the role of PTH in renal anemia and discusses the importance of treating SHPT in improving the control of renal anemia in hemodialysis patients.


Anemia/therapy , Hyperparathyroidism, Secondary/therapy , Renal Insufficiency, Chronic/therapy , Anemia/etiology , Calcimimetic Agents/pharmacology , Humans , Hyperparathyroidism, Secondary/complications , Hyperparathyroidism, Secondary/etiology , Parathyroid Hormone/blood , Parathyroidectomy/methods , Receptors, Calcitriol/drug effects , Receptors, Calcitriol/metabolism , Renal Dialysis/methods , Renal Insufficiency, Chronic/complications
18.
Semin Nephrol ; 38(3): 251-266, 2018 05.
Article En | MEDLINE | ID: mdl-29753401

Cardiovascular disease has earned its place as one of the leading noncommunicable diseases that has become a modern-day global epidemic. The increasing incidence and prevalence of chronic kidney disease (CKD) has added to this enormous burden, given that CKD is now recognized as an established risk factor for accelerated cardiovascular disease. In fact, cardiovascular disease remains the leading cause of death in the CKD population, with significant prognostic implications. Alterations in vitamin D levels as renal function declines has been linked invariably to the development of cardiovascular disease beyond a mere epiphenomenon, and has become an important focus in recent years in our search for new therapies. Another compound, cinacalcet, which belongs to the calcimimetic class of agents, also has taken center stage over the past few years as a potential cardiovasculoprotective agent. However, given limited well-designed randomized trials to inform us, our clinical practice for the management of cardiovascular disease in CKD has not been adequately refined. This article considers the biological mechanisms, regulation, and current experimental, clinical, and trial data available to help guide the therapeutic use of vitamin D and calcimimetics in the setting of CKD and cardiovascular disease.


Calcimimetic Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cinacalcet/therapeutic use , Renal Insufficiency/complications , Vascular Calcification/prevention & control , Vitamin D/analogs & derivatives , Vitamin D/therapeutic use , Vitamins/therapeutic use , Animals , Calcimimetic Agents/pharmacology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Clinical Trials as Topic , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Vascular Calcification/etiology , Vascular Calcification/physiopathology , Ventricular Remodeling , Vitamin D/physiology , Vitamin D Deficiency/complications , Vitamins/physiology
19.
Br J Pharmacol ; 175(21): 4095-4108, 2018 11.
Article En | MEDLINE | ID: mdl-29714810

BACKGROUND AND PURPOSE: Strontium ranelate, a drug approved and until recently used for the treatment of osteoporosis, mediates its effects on bone at least in part via the calcium-sensing (CaS) receptor. However, it is not known whether bone-targeted CaS receptor positive allosteric modulators (PAMs; calcimimetics) represent an alternative (or adjunctive) therapy to strontium (Sr2+ o ). EXPERIMENTAL APPROACH: We assessed three structurally distinct calcimimetics [cinacalcet, AC-265347 and a benzothiazole tri-substituted urea (BTU-compound 13)], alone and in combination with extracellular calcium (Ca2+ o ) or Sr2+ o , in G protein-dependent signalling assays and trafficking experiments in HEK293 cells and their effects on cell differentiation, tartrate-resistant acid phosphatase (TRAP) activity and hydroxyapatite resorption assays in human blood-derived osteoclasts. KEY RESULTS: Sr2+ o activated CaS receptor-dependent signalling in HEK293 cells in a similar manner to Ca2+ o , and inhibited the maturation, TRAP expression and hydroxyapatite resorption capacity of human osteoclasts. Calcimimetics potentiated Ca2+ o - and Sr2+ o -mediated CaS receptor signalling in HEK293 cells with distinct biased profiles, and only cinacalcet chaperoned an endoplasmic reticulum-retained CaS mutant receptor to the cell surface in HEK293 cells, indicative of a conformational state different from that engendered by AC-265347 and BTU-compound 13. Intriguingly, only cinacalcet modulated human osteoclast function, reducing TRAP activity and profoundly inhibiting resorption. CONCLUSION AND IMPLICATIONS: Although AC-265347 and BTU-compound 13 potentiated Ca2+ o - and Sr2+ o -induced CaS receptor activation, they neither replicated nor potentiated the ability of Sr2+ o to inhibit human osteoclast function. In contrast, the FDA-approved calcimimetic, cinacalcet, inhibited osteoclast TRAP activity and hydroxyapatite resorption, which may contribute to its clinical effects on bone mineral density LINKED ARTICLES: This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.


Calcimimetic Agents/pharmacology , Cinacalcet/pharmacology , Osteoclasts/drug effects , Receptors, Calcium-Sensing/antagonists & inhibitors , Strontium/pharmacology , Allosteric Regulation/drug effects , Calcimimetic Agents/chemistry , Cell Differentiation/drug effects , Cells, Cultured , Cinacalcet/chemistry , HEK293 Cells , Humans , Molecular Structure , Osteoclasts/metabolism , Receptors, Calcium-Sensing/metabolism , Strontium/chemistry
20.
G Ital Nefrol ; 35(3)2018 May.
Article It | MEDLINE | ID: mdl-29786181

Bone mineral abnormalities (defined as Chronic Kidney Disease Mineral Bone Disorder; CKD-MBD) are prevalent and associated with a substantial risk burden and poor prognosis in CKD population. Several lines of evidence support the notion that a large proportion of patients receiving maintenance dialysis experience a suboptimal biochemical control of CKD-MBD. Although no study has ever demonstrated conclusively that CKD-MBD control is associated with improved survival, an expanding therapeutic armamentarium is available to correct bone mineral abnormalities. In this position paper of Lombardy Nephrologists, a summary of the state of art of CKD-MBD as well as a summary of the unmet clinical needs will be provided. Furthermore, this position paper will focus on the potential and drawbacks of a new injectable calcimimetic, etelcalcetide, a drug available in Italy since few months ago.


Calcimimetic Agents/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Peptides/therapeutic use , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/therapeutic use , Calcimimetic Agents/pharmacology , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Cinacalcet/therapeutic use , Clinical Trials as Topic , Drug Therapy, Combination , Health Services Needs and Demand , Humans , Hypercalcemia/etiology , Hypercalcemia/prevention & control , Hyperparathyroidism, Secondary/blood , Parathyroid Glands/pathology , Parathyroid Hormone/biosynthesis , Parathyroid Hormone/blood , Peptides/pharmacology , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Vitamin D/metabolism , Vitamin D/therapeutic use
...