Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Endocrinology ; 162(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33242090

ABSTRACT

LA-PTH is a long-acting parathyroid hormone (PTH) peptide analogue in preclinical development for hypoparathyroidism (HP). Like native PTH, LA-PTH contains a methionine at position 8 (Met8) that is predicted to be critical for function. We assessed the impact of Met oxidation on the functional properties of LA-PTH and control PTH ligands. Oxidation of PTH(1-34) resulted in marked (~20-fold) reductions in binding affinity on the PTH receptor-1 (PTHR1) in cell membranes, similarly diminished potency for 3',5'-cyclic AMP signaling in osteoblastic cell lines (SaOS-2 and UMR106), and impaired efficacy for raising blood calcium in mice. Surprisingly, oxidation of LA-PTH resulted in little or no change in these functional responses. The signaling potency of oxidized-LA-PTH was, however, reduced approximately 40-fold compared to LA-PTH in cells expressing a PTHR1 construct that lacks the N-terminal extracellular domain (ECD). Molecular modeling revealed that while Met8 of both LA-PTH and PTH(1-34) is situated within the orthosteric ligand-binding pocket of the receptor's transmembrane domain bundle (TMD), the Met8 sidechain position is shifted for the 2 ligands so that on Met8 oxidation of PTH(1-34), steric clashes occur that are not seen with oxidized LA-PTH. The findings suggest that LA-PTH and PTH(1-34) engage the receptor differently in the Met8-interaction environment of the TMD bundle, and that this interaction environment can be allosterically influenced by the ECD component of the ligand-receptor complex. The findings should be useful for the future development of novel PTH-based peptide therapeutics for diseases of bone and mineral ion metabolism.


Subject(s)
Hypoparathyroidism/drug therapy , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/agonists , Animals , Calcium/blood , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , HEK293 Cells , Humans , Methionine/metabolism , Mice , Models, Molecular , Norleucine , Oxidation-Reduction , Parathyroid Hormone/metabolism , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism
2.
Arch Osteoporos ; 15(1): 10, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31897759

ABSTRACT

The mutual effects of drugs used in osteoporosis and cardiovascular diseases are a point of interest. A literature review and meta-analysis were conducted to address the impact of PTH analogs and anti-Rank ligand on cardiovascular events and overall mortality in individuals with idiopathic osteoporosis; these treatments do not appear to have any effect. INTRODUCTION: Two meta-analyses have been conducted to explore the cardiovascular effects of bisphosphonates. There is no review for other osteoporosis treatments. A literature review and meta-analysis were conducted to address the impact of PTH analogs and anti-Rank ligand on cardiovascular events and overall mortality in individuals with idiopathic osteoporosis. METHODS: A systematic review was conducted in December 2017 in the PubMed, Embase, and Cochrane databases and updated on PubMed in July 2019, selecting trials with a treatment and a control group. We also conducted a search for abstracts of the French Rheumatology Society, American College of Rheumatology, and European League Against Rheumatism's annual meetings over the past 4 years. The main endpoint was the occurrence of cardiovascular events; the secondary was mortality (all causes). RESULTS: Of the 2782 reports initially found, 16 articles were used for the meta-analysis (6 for the anti-Rank ligand and 10 for the PTH analog group). After meta-analysis, there was no significant difference between the placebo group and the anti-Rank ligand group for overall mortality (p = 0.13), the combined endpoint (overall mortality, coronary artery disease, and stroke; p 0.77), and the individual risk of coronary artery disease (p 0.53), arrhythmia (p 0.95), and stroke (p 0.62). After meta-analysis, there was no significant difference between the placebo group and the PTH analogs group for overall mortality (p 0.77), the combined endpoint (p = 0.95), and the individual risk of coronary artery disease (p = 0.74), arrhythmia (p = 0.28), and stroke (p = 0.61). CONCLUSIONS: The anti-Rank ligand and PTH analogs have no impact on cardiovascular risk and overall mortality in idiopathic osteoporosis. To better answer the question whether these treatments can reduce the long-term cardiovascular risk, further comparative studies with longer duration are required.


Subject(s)
Cardiovascular Diseases/prevention & control , Osteoporosis, Postmenopausal/drug therapy , Parathyroid Hormone/analogs & derivatives , RANK Ligand/antagonists & inhibitors , Arrhythmias, Cardiac/mortality , Arrhythmias, Cardiac/prevention & control , Bone Density Conservation Agents/therapeutic use , Cardiovascular Diseases/mortality , Coronary Artery Disease/mortality , Coronary Artery Disease/prevention & control , Denosumab/therapeutic use , Diphosphonates/therapeutic use , Female , Humans , Risk Factors , Stroke/mortality , Stroke/prevention & control , Teriparatide/therapeutic use
3.
J Mol Endocrinol ; 62(2): R145-R154, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30389901

ABSTRACT

Bone-forming approaches to treat patients with severe osteoporosis are effective, but treatment options are limited, and there is an unmet clinical need for additional drugs. This review discusses two novel and advanced anabolic therapeutic concepts that have successfully completed phase 3 trials. Romosozumab is a monoclonal antibody that targets the Wnt inhibitor sclerostin. Two phase 3 trials (FRAME and ARCH) of romosozumab for the treatment of postmenopausal osteoporosis have been completed. Both trials successfully reached their primary endpoint by reducing vertebral fractures by 75% compared to placebo (FRAME trial) and 48% compared to alendronate (ARCH trial), respectively. Abaloparatide is a PTH-related protein (PTHrP) analog that has displayed bone anabolic activity. In the phase 3 ACTIVE trial, abaloparatide was compared to placebo and teriparatide for 18 months in postmenopausal women who had already experienced an osteoporotic fracture. Abaloparatide successfully reduced the rate of new vertebral fractures by 86% compared to placebo. Furthermore, abaloparatide achieved greater BMD increases at all measured sites compared to both placebo and teriparatide. Based on these results, abaloparatide was FDA approved in April 2017. This review discusses available data of both agents with regard to efficacy and safety as well as their possible future application.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Osteoporosis/drug therapy , Parathyroid Hormone/analogs & derivatives , Peptides/therapeutic use , Clinical Trials as Topic , Humans , Parathyroid Hormone-Related Protein/metabolism
4.
Osteoporos Int ; 30(1): 59-70, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30539271

ABSTRACT

Combination therapy with parathyroid hormone (PTH) analogs and antiresorptive agents may be more effective than monotherapy for the treatment of osteoporosis. This study aimed to estimate the effectiveness and safety of this combination therapy for osteoporosis. MEDLINE, EMBASE, and Cochrane Library were searched from inception to May 1, 2018, including randomized controlled trials (RCTs) with a duration of at least 6 months on adults with osteoporosis treated with combination therapy versus monotherapy. Outcomes included fractures, bone mineral density (BMD) changes, and adverse events. A meta-analysis was performed using a random-effect model, to estimate risk ratios (RRs) for fractures, and mean differences (MDs) for BMD changes. A total of 19 RCTs and 2177 patients were included. Compared with monotherapy, combination therapy had an advantage of 36% (RR, 0.64; 95% confidence interval (CI), 0.42-0.98) regarding fracture risk reduction. It also appears to improve lumbar spine BMD by 4.06% (95%CI = 2.60-5.53) and total hip BMD by 1.89% (95%CI = 1.25-2.53). No RCT reported an increased risk of serious adverse events. Among patients with osteoporosis, combination therapy was superior to monotherapy regarding improvement of the lumbar spine and total hip BMD, without risk of serious adverse events. Combination therapy also had an advantage over monotherapy on fracture risk reduction. However, owing to the limited sample size, additional larger studies are required to confirm this benefit.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Osteoporosis/drug therapy , Parathyroid Hormone/analogs & derivatives , Bone Density/drug effects , Drug Therapy, Combination , Hip Joint/physiopathology , Humans , Lumbar Vertebrae/physiopathology , Osteoporosis/physiopathology , Osteoporotic Fractures/prevention & control , Parathyroid Hormone/therapeutic use , Randomized Controlled Trials as Topic
5.
J Biol Chem ; 293(52): 20200-20213, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30377251

ABSTRACT

Osteoporosis can result from the loss of sex hormones and/or aging. Abaloparatide (ABL), an analog of parathyroid hormone-related protein (PTHrP(1-36)), is the second osteoanabolic therapy approved by the United States Food and Drug Administration after teriparatide (PTH(1-34)). All three peptides bind PTH/PTHrP receptor type 1 (PTHR1), but the effects of PTHrP(1-36) or ABL in the osteoblast remain unclear. We show that, in primary calvarial osteoblasts, PTH(1-34) promotes a more robust cAMP response than PTHrP(1-36) and ABL and causes a greater activation of protein kinase A (PKA) and cAMP response element-binding protein (CREB). All three peptides similarly inhibited sclerostin (Sost). Interestingly, the three peptides differentially modulated two other PKA target genes, c-Fos and receptor activator of NF-κB ligand (Rankl), and the latter both in vitro and in vivo Knockdown of salt-inducible kinases (SIKs) 2 and 3 and CREB-regulated transcription coactivator 3 (CRTC3), indicated that all three are part of the pathway that regulates osteoblastic Rankl expression. We also show that the peptides differentially regulate the nuclear localization of CRTC2 and CRTC3, and that this correlates with PKA activation. Moreover, inhibition of protein phosphatases 1 and 2A (PP1/PP2A) activity revealed that they play a major role in both PTH-induced Rankl expression and the effects of PTH(1-34) on CRTC3 localization. In summary, in the osteoblast, the effects of PTH(1-34), PTHrP(1-36), and ABL on Rankl are mediated by differential stimulation of cAMP/PKA signaling and by their downstream effects on SIK2 and -3, PP1/PP2A, and CRTC3.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Osteoblasts/metabolism , Parathyroid Hormone , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , RANK Ligand/biosynthesis , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Mice , Osteoblasts/cytology , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/pharmacology , Protein Phosphatase 1/genetics , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/genetics , RANK Ligand/genetics , Rats , Signal Transduction/genetics , Transcription Factors/genetics
6.
BMJ Open ; 8(1): e019291, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362267

ABSTRACT

INTRODUCTION: Fracture healing is a complex physiological process. Impaired healing will increase the need for care and cause serious complications. Thus, identifying strategies to accelerate the rate of healing, preventing delayed unions and non-unions, is essential. Parathyroid hormone (PTH) is a key systemic regulator of calcium and phosphate metabolism. It has been determined that intermittent administration of PTH and its analogue can exert anabolic effect on bone, increase bone mass and reduce bone loss, leading to an increase in bone formation. Owing to their anabolic effect, there is an increasing interest in its potential in promoting the process of fracture healing. However, in clinical studies, the results are in conflict. This objective of this study is to determine the role of PTH analogues for fracture healing in adults. METHODS AND ANALYSIS: MEDLINE, EMBASE and Cochrane databases will be searched to identify all randomised controlled trials (RCTs) and quasi-RCTs that compare the different effects between PTH analogues and any other treatments in adults with any type of fracture. The primary outcome is the functional recovery. And the secondary outcomes are fracture union and adverse events. The meta-analysis will be performed using a random effects model. Heterogeneity will be assessed by the P values and I² statistic. And subgroup analyses and sensitivity analyses will be used to explore the heterogeneity. Risk of bias will be assessed using the Cochrane tool and the quality of evidence will be assessed using the Grading of Recommendations Assessment, Development and Evaluation approach. ETHICS AND DISSEMINATION: Ethical approval is not required because this proposed systematic review and meta-analysis is based on published data, without including confidential personal data or data on interventions on patients. The findings of this study will be published in a peer-reviewed journaland presented at a relevant conference. PROSPERO REGISTRATION NUMBER: CRD42017062093.


Subject(s)
Bone Density/drug effects , Fracture Healing/drug effects , Fractures, Bone/therapy , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/therapeutic use , Adult , Humans , Randomized Controlled Trials as Topic , Systematic Reviews as Topic
7.
Br J Pharmacol ; 175(2): 262-271, 2018 01.
Article in English | MEDLINE | ID: mdl-28898923

ABSTRACT

BACKGROUND AND PURPOSE: Human parathyroid hormone (PTH) is critical for maintaining physiological calcium homeostasis and plays an important role in the formation and maintenance of the bone. Full-length PTH and a truncated peptide form are approved for treatment of hypoparathyroidism and osteoporosis respectively. Our initial goal was to develop an improved PTH therapy for osteoporosis, but clinical development was halted. The novel compound was then repurposed as an improved therapy for hypoparathyroidism. EXPERIMENTAL APPROACH: A longer-acting form of PTH was synthesised by altering the peptide to increase cell surface residence time of the bound ligand to its receptor. In vitro screening identified a compound, which was tested in an animal model of osteoporosis before entering human trials. This compound was subsequently tested in two independent animal models of hypoparathyroidism. KEY RESULTS: The peptide identified, LY627-2K, exhibited delayed internalization kinetics. In an ovariectomy-induced bone loss rat model, LY627-2K demonstrated improved vertebral bone mineral density and biomechanical properties at skeletal sites and a modest increase in serum calcium. In a Phase I clinical study, dose-dependent increases in serum calcium were reproduced. These observations prompted us to explore a second indication, hypoparathyroidism. In animal models of this disease, LY627-2K restored serum calcium, comparing favourably to treatment with wild-type PTH. CONCLUSIONS AND IMPLICATIONS: We summarize the repositioning of a therapeutic candidate with substantial preclinical and clinical data. Our results support its repurposing and continued development, from a common indication (osteoporosis) to a rare disease (hypoparathyroidism) by exploiting a shared molecular target. LINKED ARTICLES: This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.


Subject(s)
Drug Repositioning/methods , Hypoparathyroidism/drug therapy , Parathyroid Hormone/analogs & derivatives , Animals , Bone Density/drug effects , Calcium/blood , Female , Humans , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use , Rats
8.
Cell Signal ; 41: 46-55, 2018 01.
Article in English | MEDLINE | ID: mdl-28495495

ABSTRACT

It is increasingly apparent that ligand structure influences both the efficiency with which G protein-coupled receptors (GPCRs) engage their downstream effectors and the manner in which they are activated. Thus, 'biased' agonists, synthetic ligands whose intrinsic efficacy differs from the native ligand, afford a strategy for manipulating GPCR signaling in ways that promote beneficial signals while blocking potentially deleterious ones. Still, there are significant challenges in relating in vitro ligand efficacy, which is typically measured in heterologous expression systems, to the biological response in vivo, where the ligand is acting on natively expressed receptors and in the presence of the endogenous ligand. This is particularly true of arrestin pathway-selective 'biased' agonists. The type 1 parathyroid hormone receptor (PTH1R) is a case in point. Parathyroid hormone (PTH) is the principal physiological regulator of calcium homeostasis, and PTH1R expressed on cells of the osteoblast lineage are an established therapeutic target in osteoporosis. In vitro, PTH1R signaling is highly sensitive to ligand structure, and PTH analogs that affect the selectivity/kinetics of G protein coupling or that engage arrestin-dependent signaling mechanisms without activating heterotrimeric G proteins have been identified. In vivo, intermittent administration of conventional PTH analogs accelerates the rate of osteoblastic bone formation, largely through known cAMP-dependent mechanisms. Paradoxically, both intermittent and continuous administration of an arrestin pathway-selective PTH analog, which in vivo would be expected to antagonize endogenous PTH1R-cAMP signaling, also increases bone mass. Transcriptomic analysis of tissue from treated animals suggests that conventional and arrestin pathway-selective PTH1R ligands act in largely different ways, with the latter principally affecting pathways involved in the regulation of cell cycle, survival, and migration/cytoskeletal dynamics. Such multi-dimensional in vitro and in vivo analyses of ligand bias may provide insights into the physiological roles of non-canonical arrestin-mediated signaling pathways in vivo, and provide a conceptual framework for translating arrestin pathway-selective ligands into viable therapeutics.


Subject(s)
Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arrestins/genetics , Arrestins/metabolism , Drug Design , Humans , In Vitro Techniques , Ligands , Models, Animal , Osteogenesis/drug effects , Osteoporosis/drug therapy , Osteoporosis/metabolism , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/genetics
9.
Curr Osteoporos Rep ; 15(2): 110-119, 2017 04.
Article in English | MEDLINE | ID: mdl-28303448

ABSTRACT

PURPOSE OF REVIEW: The purpose is to review the efficacy and optimal use of parathyroid hormone and parathyroid hormone-related protein analogs in osteoporosis treatment. RECENT FINDINGS: The parathyroid hormone analog teriparatide, a potent stimulator of bone remodeling, increases hip and spine bone mineral density and reduces the risk of vertebral and non-vertebral fractures in postmenopausal osteoporotic women. The parathyroid hormone-related protein analog, abaloparatide, also reduces fracture incidence but has pharmacological effects that differ from teriparatide, particularly in cortical bone. These analogs provide maximal benefit when their use is followed by a potent antiresorptive medication. Moreover, studies have shown that the combination of teriparatide and the RANK-ligand inhibitor, denosumab, increase bone density and estimated strength more than monotherapy and more than any currently available regimen. Parathyroid hormone and parathyroid hormone-related protein analogs, whether as monotherapy, in combination with antiresorptive agents or in sequence with antiresorptive agents, will likely play an expanding role in osteoporosis management.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Denosumab/therapeutic use , Osteoporosis/drug therapy , Osteoporotic Fractures/prevention & control , Parathyroid Hormone-Related Protein/therapeutic use , Parathyroid Hormone/therapeutic use , Spinal Fractures/prevention & control , Teriparatide/therapeutic use , Bone Density , Drug Therapy, Combination , Humans , Osteoporosis, Postmenopausal/drug therapy , Parathyroid Hormone/analogs & derivatives
10.
J Bone Miner Res ; 31(7): 1405-12, 2016 07.
Article in English | MEDLINE | ID: mdl-26865415

ABSTRACT

Hypoparathyroidism is a disease of chronic hypocalcemia and hyperphosphatemia due to a deficiency of parathyroid hormone (PTH). PTH and analogs of the hormone are of interest as potential therapies. Accordingly, we examined the pharmacological properties of a long-acting PTH analog, [Ala(1,3,12,18,22) , Gln(10) ,Arg(11) ,Trp(14) ,Lys(26) ]-PTH(1-14)/PTHrP(15-36) (LA-PTH) in thyroparathyroidectomized (TPTX) rats, a model of HP, as well as in normal monkeys. In TPTX rats, a single intravenous administration of LA-PTH at a dose of 0.9 nmol/kg increased serum calcium (sCa) and decreased serum phosphate (sPi) to near-normal levels for longer than 48 hours, whereas PTH(1-34) and PTH(1-84), each injected at a dose 80-fold higher than that used for LA-PTH, increased sCa and decreased sPi only modestly and transiently (<6 hours). LA-PTH also exhibited enhanced and prolonged efficacy versus PTH(1-34) and PTH(1-84) for elevating sCa when administered subcutaneously (s.c.) into monkeys. Daily s.c. administration of LA-PTH (1.8 nmol/kg) into TPTX rats for 28 days elevated sCa to near normal levels without causing hypercalciuria or increasing bone resorption markers, a desirable goal in the treatment of hypoparathyroidism. The results are supportive of further study of long-acting PTH analogs as potential therapies for patients with hypoparathyroidism. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Hypoparathyroidism/drug therapy , Parathyroid Hormone , Parathyroidectomy , Animals , Biomarkers/blood , Bone Resorption/blood , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , HEK293 Cells , Humans , Hypercalciuria/blood , Hypoparathyroidism/blood , Macaca fascicularis , Male , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/pharmacokinetics , Parathyroid Hormone/pharmacology , Rats
11.
J Bone Miner Res ; 31(5): 975-84, 2016 05.
Article in English | MEDLINE | ID: mdl-26678919

ABSTRACT

Hypoparathyroidism (HP) arises most commonly from parathyroid (PT) gland damage associated with neck surgery, and is typically treated with oral calcium and active vitamin D. Such treatment effectively increases levels of serum calcium (sCa), but also brings risk of hypercalciuria and renal damage. There is thus considerable interest in using PTH or PTH analogs to treat HP. To facilitate study of this disease and the assessment of new treatment options, we developed two mouse models of acquired HP, and used them to assess efficacy of PTH(1-34) as well as a long-acting PTH analog (LA-PTH) in regulating blood calcium levels. In one model, we used PTHcre-iDTR mice in which the diphtheria toxin (DT) receptor (DTR) is selectively expressed in PT glands, such that systemic DT administration selectively ablates parathyroid cells. For the second model, we generated GFP-PT mice in which green fluorescent protein (GFP) is selectively expressed in PT cells, such that parathyroidectomy (PTX) is facilitated by green fluorescence of the PT glands. In the PTHcre-iDTR mice, DT injection (2 × 5 µg/kg, i.p.) resulted in moderate yet consistent reductions in serum PTH and sCa levels. The more severe hypoparathyroid phenotype was observed in GFP-PT mice following GFP-guided PTX surgery. In each model, a single subcutaneous injection of LA-PTH increased sCa levels more effectively and for a longer duration (>24 hours) than did a 10-fold higher dose of PTH(1-34), without causing excessive urinary calcium excretion. These new mouse models thus faithfully replicate two degrees of acquired HP, moderate and severe, and may be useful for assessing potential new modes of therapy. © 2015 American Society for Bone and Mineral Research.


Subject(s)
Diphtheria Toxin/toxicity , Hypoparathyroidism/drug therapy , Parathyroid Hormone , Animals , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypoparathyroidism/chemically induced , Hypoparathyroidism/genetics , Hypoparathyroidism/metabolism , Male , Mice , Mice, Transgenic , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/pharmacology
12.
Medicine (Baltimore) ; 94(38): e1156, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26402797

ABSTRACT

Parathyroid hormone (PTH) analogues increase bone strength primarily by stimulating bone formation, whereas antiresorptive drugs (bisphosphonates) reduce bone resorption. Therefore, some studies have been designed to test the hypothesis that the concurrent administration of the 2 agents would increase bone density more than the use of either one alone. This meta-analysis aimed to determine whether combining PTH analogues with bisphosphonates would be superior to PTH alone. Electronic databases were searched to identify relevant publications up to March, 2014. Randomized controlled trials (RCTs) comparing PTH analogues combined bisphosphonates with PTH for osteoporosis were analyzed. According to the Cochrane Handbook for systematic Reviews of Interventions 5.2, we identified eligible studies, evaluated the methodological quality, and abstracted relevant data. Totally 7 studies involving 641 patients were included for meta-analysis. The pooled data showed that there were no significant differences in the percent change of spine BMD (MD1-year = -0.97, 95% CI -2.81 to 0.86, P = 0.30; MD2-year =  - 0.57, 95% CI -5.01 to 6.14, P = 0.84), femoral neck BMD (MD1-year = 0.60, 95% CI -0.91 to 2.10, P = 0.44; MD2-year = -0.73, 95% CI -4.97 to 3.51, P = 0.74), the risk of vertebral fracture (risk ratio [RR] = 1.27; 95% CI 0.29-5.57; P = 0.75), and the risk of nonvertebral fracture (RR = 0.97; 95% CI 0.40-2.35; P = 0.95) between the 2 groups, whereas combination group improves the percent change of hip BMD at 1 year (MD = 1.16, 95% CI 0.56-1.76; P < 0.01) than PTH analogues group. Our results showed that there was no evidence for the superiority of combination therapy, although significant change was found for hip BMD at 1 year in combination group. Further large multicenter randomized controlled trials are still needed to investigate the efficacy of combination therapy.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Diphosphonates/therapeutic use , Osteoporosis/drug therapy , Parathyroid Hormone , Bone Density , Drug Therapy, Combination , Fractures, Bone/etiology , Fractures, Bone/prevention & control , Humans , Osteoporosis/complications , Osteoporosis/metabolism , Osteoporosis/physiopathology , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/metabolism , Randomized Controlled Trials as Topic , Treatment Outcome
13.
Drugs ; 75(10): 1049-58, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26056029

ABSTRACT

Bone remodeling, the fundamental process for bone renewal, is targeted by treatments of osteoporosis to correct the imbalance between bone resorption and bone formation and reduce the risk of fractures and associated clinical consequences. Currently available therapeutics affect bone resorption and bone formation in the same direction and either decrease (inhibitors of bone resorption) or increase (parathyroid hormone [PTH] peptides) bone remodeling. Studies of patients with rare bone diseases and genetically modified animal models demonstrated that bone resorption and bone formation may not necessarily be coupled, leading to identification of molecular targets in bone cells for the development of novel agents for the treatment of osteoporosis. Application of such agents to the treatment of women with low bone mass confirmed that bone resorption and bone formation can be modulated in different directions and so far two new classes of therapeutics for osteoporosis have been defined with distinct mechanisms of action. Such treatments, if combined with a favorable safety profile, will offer new therapeutic options and will improve the management of patients with osteoporosis.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/drug therapy , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Parathyroid Hormone/analogs & derivatives , Animals , Humans , Models, Biological , Parathyroid Hormone/pharmacology , Parathyroid Hormone/therapeutic use
14.
Br J Nutr ; 113(6): 909-22, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25744000

ABSTRACT

In the present study, we investigated whether high dietary Ca and exogenous parathyroid hormone 1-34 fragments (PTH 1-34) have synergistic effects on bone formation in adult mice, and explored the related mechanisms. Adult male mice were fed a normal diet, a high-Ca diet, a PTH-treated diet, or a high-Ca diet combined with subcutaneously injected PTH 1-34 (80 µg/kg per d) for 4 weeks. Bone mineral density, trabecular bone volume, osteoblast number, alkaline phosphatase (ALP)- and type I collagen-positive areas, and the expression levels of osteoblastic bone formation-related genes and proteins were increased significantly in mice fed the high-Ca diet, the PTH-treated diet, and, even more dramatically, the high-Ca diet combined with PTH. Osteoclast number and surface and the ratio of receptor activator for nuclear factor-κB ligand (RANKL):osteoprotegerin (OPG) were decreased in the high-Ca diet treatment group, increased in the PTH treatment group, but not in the combined treatment group. Furthermore, third-passage osteoblasts were treated with high Ca (5 mM), PTH 1-34 (10⁻8 M) or high Ca combined with PTH 1-34. Osteoblast viability and ALP activity were increased in either the high Ca-treated or PTH-treated cultures and, even more dramatically, in the cultures treated with high Ca plus PTH, with consistent up-regulation of the expression levels of osteoblast proliferation and differentiation-related genes and proteins. These results indicate that dietary Ca and PTH play synergistic roles in promoting osteoblastic bone formation by stimulating osteoblast proliferation and differentiation.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone Resorption/prevention & control , Calcium, Dietary/therapeutic use , Food-Drug Interactions , Osteoblasts/drug effects , Osteogenesis/drug effects , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/therapeutic use , Animals , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium, Dietary/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Combined Modality Therapy , Male , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoblasts/pathology , Parathyroid Hormone/pharmacology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, Parathyroid Hormone/agonists , Receptors, Parathyroid Hormone/genetics , Receptors, Parathyroid Hormone/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Up-Regulation/drug effects
15.
PLoS One ; 10(3): e0120402, 2015.
Article in English | MEDLINE | ID: mdl-25775025

ABSTRACT

Hypocalcemia and hypophosphatemia are common complications after parathyroidectomy (PTX). Sudden removal of high circulating levels of parathyroid hormone (PTH) causes decreased osteoclastic resorption resulting in a decreased bone remodeling space. These phenomena are likely due to an increased influx of calcium and phosphorus into bone. However, there are currently no data to support this hypothesis. In this study, we found that PTX significantly reduced levels of PTH, calcium and phosphate. Compared with preoperative levels, after 1 year, postoperative PTH, calcium and phosphate levels were 295.6 ± 173.7 pg/mL (P < 0.05), 86.62 ± 15.98 mg/dL (P < 0.05) and 5.56 ± 2.03 mg/dL (P < 0.05), respectively. We investigated continuous bovine PTH administration as well as withdrawal of bovine PTH stimulation in the mouse osteoblast precursor cell line MC3T3-E1. MC3T3-E1 cells were cultured with continuous bovine PTH treatment for 20 days or with transient bovine PTH treatment for 10 days. High doses of continuous bovine PTH exposure strongly reduced cell proliferation, alkaline phosphatase activity and the number of mineralized calcium nodules. However, withdrawal of bovine PTH (100 ng/mL) significantly increased the number of mineralized calcium nodules and caused a rapid decline in calcium and phosphorus content of culture medium. In conclusion, continuous exposure to bovine PTH inhibited osteoblast differentiation and reduced the formation of mineralized nodules. However, this inhibition was removed and mineralized nodule formation resumed with withdrawal of bovine PTH. According to the results of our clinical examinations and in vitro experiments, we hypothesize that the sudden removal of high levels of PTH may cause an increased influx of calcium and phosphorus into bone after PTX.


Subject(s)
Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/analogs & derivatives , Phosphorus/metabolism , Adult , Aged , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Calcium/metabolism , Cattle , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Male , Mice , Middle Aged , Parathyroid Hormone/pharmacology , Parathyroidectomy/adverse effects , Perioperative Period
16.
Expert Opin Investig Drugs ; 24(2): 145-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25316089

ABSTRACT

INTRODUCTION: Intermittent parathyroid hormone (PTH) administration, acting through multiple signaling pathways, exerts an osteoanabolic effect on the skeleton that surpasses the effect of other antiosteoporotic agents. However, its efficacy is limited by the coupling effect and relatively common adverse events. Thus, the development of more sophisticated PTH receptor analogs seems imperative. AREAS COVERED: In this review, the authors summarize the role of PTH signaling pathway in bone remodeling. The authors also summarize investigational analogs targeting this pathway, which may be potential treatments for osteoporosis. EXPERT OPINION: ß-arrestins are multifunctional cytoplasmic molecules that are decisive for regulating intracellular PTH signaling. Recently, in preclinical studies, arrestin analogs have achieved the anabolic bone effect of PTH without an accompanying increase in bone resorption. However, it is not yet known whether these analogs have adverse effects and there are no clinical data for their efficacy to date. On the other hand, several molecules derived either from PTH and PTH-related protein (PTHrP) molecules have been developed. Alternative routes of PTH 1 - 34 delivery (oral, transdermal), the PTH analog ostabolin and the N-terminal PTHrP analogs PTHrP 1 - 36 and abaloparatide, have recently been or are currently being tested in clinical trials and are more likely to become available for use in the near future.


Subject(s)
Osteoporosis/drug therapy , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/drug effects , Animals , Arrestins/metabolism , Bone Remodeling/drug effects , Bone Resorption/drug therapy , Drug Design , Humans , Osteoporosis/physiopathology , Parathyroid Hormone/administration & dosage , Parathyroid Hormone/metabolism , Parathyroid Hormone-Related Protein/pharmacology , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction/drug effects , beta-Arrestins
17.
Anticancer Drugs ; 25(1): 30-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24025564

ABSTRACT

Alopecia is a psychologically devastating complication of chemotherapy for which there is currently no effective therapy. PTH-CBD is a collagen-targeted parathyroid hormone analog that has shown promise as a therapy for alopecia disorders. This study compared the efficacy of prophylactic versus therapeutic administration of PTH-CBD in chemotherapy-induced alopecia using a mouse model that mimics the cyclic chemotherapy dosing used clinically. C57BL/6J mice were treated with a single subcutaneous injection of PTH-CBD (320 mcg/kg) or vehicle control before or after hair loss developing from three courses of cyclophosphamide chemotherapy (50-150 mg/kg/week). Mice receiving chemotherapy alone developed hair loss and depigmentation over 6-12 months. Mice pretreated with PTH-CBD did not develop these changes and maintained a normal-appearing coat. Mice treated with PTH-CBD after development of hair loss showed a partial recovery. Observations of hair loss were confirmed quantitatively by gray scale analysis. Histological examination showed that in mice receiving chemotherapy alone, there were small, dystrophic hair follicles mostly in the catagen phase. Mice receiving PTH-CBD before chemotherapy showed a mix of normal-appearing telogen and anagen hair follicles with no evidence of dystrophy. Mice receiving PTH-CBD therapy after chemotherapy showed intermediate histological features. PTH-CBD was effective in both the prevention and the treatment of chemotherapy-induced alopecia in mice, but pretreatment appears to result in a better cosmetic outcome. PTH-CBD shows promise as an agent in the prevention of this complication of chemotherapy and improving the quality of life for cancer patients.


Subject(s)
Alopecia/drug therapy , Antineoplastic Agents, Alkylating/adverse effects , Collagen/metabolism , Cyclophosphamide/adverse effects , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/therapeutic use , Alopecia/chemically induced , Alopecia/prevention & control , Animals , Bacterial Proteins/genetics , Collagenases/genetics , Hair Follicle/drug effects , Hair Follicle/growth & development , Humans , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Parathyroid Hormone/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
18.
Curr Osteoporos Rep ; 11(4): 400-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24078470

ABSTRACT

Osteoporotic fractures result in significant morbidity and mortality. Anabolic agents reverse the negative skeletal balance that characterizes osteoporosis by stimulating osteoblast-dependent bone formation to a greater degree than osteoclast-dependent bone resorption. Parathyroid hormone (PTH) and parathyroid hormone- related protein (PTHrP) are peptide hormones, which have anabolic actions when administered intermittently. The only FDA-approved anabolic bone agent for the treatment of osteoporosis in the United States is PTH 1-34, or teriparatide, administered by daily subcutaneous injections. However, PTH 1-84 is also available in Europe. Synthetic human PTHrP 1-36 and a PTHrP 1-34 analog, BA058, have also been shown to increase lumbar spine bone density. These agents and several other PTH and PTHrP analogs, including some which are not administered as injections, continue to be investigated as potential anabolic therapies for osteoporosis.


Subject(s)
Anabolic Agents/therapeutic use , Osteoporosis/drug therapy , Parathyroid Hormone-Related Protein/therapeutic use , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/therapeutic use , Anabolic Agents/pharmacology , Animals , Bone Resorption/drug therapy , Disease Models, Animal , Humans , Osteogenesis/drug effects , Parathyroid Hormone/pharmacology , Parathyroid Hormone-Related Protein/pharmacology , Teriparatide/pharmacology , Teriparatide/therapeutic use , Treatment Outcome
19.
Nephrol Dial Transplant ; 28(9): 2260-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23787544

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is associated with elevations in serum phosphate, calcium-phosphorus product and bone-specific alkaline phosphatase (BAP), with attendant risks of cardiovascular and bone disorders. Active vitamin D can suppress parathyroid hormone (PTH), but may raise serum calcium and phosphate concentrations. Paricalcitol, a selective vitamin D activator, suppressed PTH in CKD patients (stages 3 and 4) with secondary hyperparathyroidism (SHPT) with minimal changes in calcium and phosphate metabolism. METHODS: The VITAL study enrolled patients with CKD stages 2-4. We examined the effect and relationship of paricalcitol to calcium and phosphate metabolism and bone markers in a post hoc analysis of VITAL. The study comprised patients with diabetic nephropathy enrolled in a double-blind, placebo-controlled, randomized trial of paricalcitol (1 or 2 µg/day). Urinary and serum calcium and phosphate, serum BAP, and intact PTH (iPTH) concentrations were measured throughout the study. RESULTS: Baseline demographics and calcium, phosphate, PTH (49% with iPTH <70 pg/mL), and BAP concentrations were similar between groups. A transient, modest yet significant increase in phosphate was observed for paricalcitol 2 µg/day (+0.29 mg/dL; P < 0.001). Dose-dependent increases in serum and urinary calcium were observed; however, there were few cases of hypercalcemia: one in the 1-µg/day group (1.1%) and three in the 2-µg/day group (3.2%). Significant reductions in BAP were observed that persisted for 60 days after paricalcitol discontinuation (P < 0.001 for combined paricalcitol groups versus placebo). Paricalcitol dose-dependent reductions in iPTH were observed. Paricalcitol in CKD patients (±SHPT) was associated with modest increases in calcium and phosphate. CONCLUSION: Paricalcitol reduces BAP levels, which may be beneficial for reducing vascular calcification. TRIAL REGISTRATION: Trial is registered with ClinicalTrials.gov, number NCT00421733.


Subject(s)
Alkaline Phosphatase/metabolism , Biomarkers/metabolism , Bone and Bones/metabolism , Calcium/metabolism , Diabetic Nephropathies/metabolism , Ergocalciferols/pharmacology , Phosphates/metabolism , Aged , Bone Density Conservation Agents/pharmacology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Double-Blind Method , Female , Humans , Male , Middle Aged , Parathyroid Hormone/analogs & derivatives
20.
J Cell Sci ; 126(Pt 10): 2305-13, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525004

ABSTRACT

Cholesterol depletion reversibly abolishes carbachol-evoked Ca(2+) release from inositol (1,4,5)-trisphosphate (IP3)-sensitive stores, without affecting the distribution of IP3 receptors (IP3R) or endoplasmic reticulum, IP3 formation or responses to photolysis of caged IP3. Receptors that stimulate cAMP formation do not alone evoke Ca(2+) signals, but they potentiate those evoked by carbachol. We show that these potentiated signals are entirely unaffected by cholesterol depletion and that, within individual cells, different IP3-sensitive Ca(2+) stores are released by carbachol alone and by carbachol combined with receptors that stimulate cAMP formation. We suggest that muscarinic acetylcholine receptors in lipid rafts deliver IP3 at high concentration to associated IP3R, stimulating them to release Ca(2+). Muscarinic receptors outside rafts are less closely associated with IP3R and provide insufficient local IP3 to activate IP3R directly. These IP3R, probably type 2 IP3R within a discrete Ca(2+) store, are activated only when their sensitivity is increased by cAMP. Sensitization of IP3R by cAMP extends the effective range of signalling by phospholipase C, allowing muscarinic receptors that are otherwise ineffective to recruit additional IP3-sensitive Ca(2+) stores.


Subject(s)
Cyclic AMP/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Membrane Microdomains/metabolism , Receptor Cross-Talk , Bone Remodeling , Calcium/metabolism , Calcium Signaling , Carbachol/metabolism , Cholesterol , HEK293 Cells , Humans , Intracellular Space/metabolism , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Muscarinic/metabolism , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...