Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Osteoporos Int ; 31(1): 193-201, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31642977

RESUMO

Following 150 mg of oral ibandronate, Taiwanese females have greater serum and urine levels of this drug and bone resorption marker suppression than Caucasian women. These inter-ethnic differences seems to be partly explained by a 2.48-fold higher bioavailability of ibandronate in Taiwanese postmenopausal women. INTRODUCTION: Interethnic differences in the pharmacokinetics of oral ibandronate for osteoporosis are unknown. We compared the disposition of oral ibandronate between Caucasian and Taiwanese postmenopausal women. METHODS: Ibandronate 150 mg was administered to 35 Caucasian and 16 Taiwanese postmenopausal women in two separate phase 1 studies. Interethnic comparisons were performed to assess pharmacokinetic properties, including the area under the concentration-time curve (AUC), peak concentration (Cmax), elimination half-life, urinary drug recovery (Ae%), renal clearance (CLr), apparent total clearance (CL/F), and apparent volume of distribution (Vd/F). RESULTS: The mean AUC, Cmax, and Ae% were 2.41-, 1.69-, and 2.95-fold greater in the Taiwanese than in the Caucasian subjects, and the average CL/F and Vd/F were 2.48- and 2.46-fold smaller. There were no significant differences in mean CLr and half-life between both groups. As bisphosphonates are not biotransformed but are mainly excreted in the urine, the total body clearance is close to the CLr. These results suggested a larger bioavailability in the Taiwanese group which resulted in the differences in the CL/F and Vd/F. Multiple linear regression analysis demonstrated ethnicity influences of the pharmacokinetic properties after adjusting for the other variables. CONCLUSIONS: Bioavailability was largely responsible for the interethnic pharmacokinetic differences following oral administration of 150 mg ibandronate and seemed greater in the Taiwanese compared with the Caucasian subjects. Further dose-ranging studies are warranted to determine the optimal dosages of oral ibandronate in patients of Asian or Taiwanese ethnicity.


Assuntos
Conservadores da Densidade Óssea , Ácido Ibandrônico , Osteoporose Pós-Menopausa , Pós-Menopausa , Administração Oral , Idoso , Povo Asiático , Disponibilidade Biológica , Conservadores da Densidade Óssea/farmacocinética , Difosfonatos/uso terapêutico , Feminino , Humanos , Ácido Ibandrônico/farmacocinética , Pessoa de Meia-Idade , Osteoporose Pós-Menopausa/tratamento farmacológico , Fatores Raciais , População Branca
2.
J Pharmacokinet Pharmacodyn ; 47(5): 473-484, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32647957

RESUMO

To develop a framework for evaluating the resorption effects of Cathepsin K (CatK) inhibitors and to inform dose regimen selection, a pharmacokinetic/pharmacodynamic (PK/PD) model for odanacatib (ODN) was developed based upon data from Phase 1 studies. Pooled PK/PD data from 11 studies (N = 249) were fit reasonably to a population inhibitory sigmoid Emax model. Body weight on E0 (baseline uNTx/Cr, urinary N-terminal telopeptide normalized by creatinine) and age on Emax (fractional inhibition of the biomarker response) were significant covariates for biomarker response. Simulations of typical osteoporosis patients (by age, sex and weight) indicated minimal differences between sexes in concentration-uNTx/Cr relationship. There was no evidence that regimen (daily vs. weekly dosing) influenced the PK/PD relationship of resorption inhibition for odanacatib. PK/PD models based on data from odanacatib (ODN) Phase 1 studies demonstrated that uNTx/Cr was an appropriate bone resorption biomarker for assessment of the effects of a CatK inhibitor. The models also identified the determinants of response in the PK/PD relationship for ODN (body weight on E0 and age on Emax).


Assuntos
Compostos de Bifenilo/farmacocinética , Conservadores da Densidade Óssea/farmacocinética , Reabsorção Óssea/prevenção & controle , Catepsina K/antagonistas & inibidores , Adulto , Idoso , Biomarcadores/urina , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/efeitos adversos , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/efeitos adversos , Reabsorção Óssea/diagnóstico , Reabsorção Óssea/urina , Catepsina K/metabolismo , Creatinina/urina , Relação Dose-Resposta a Droga , Esquema de Medicação , Monitoramento de Medicamentos/métodos , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/urina , Pró-Colágeno/urina , Resultado do Tratamento , Adulto Jovem
3.
Drug Dev Ind Pharm ; 46(4): 557-565, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32126844

RESUMO

Raloxifene hydrochloride (RH) suffers from low oral bioavailability due to its low water-solubility and first-pass metabolism. Therefore, a novel phospholipid complex of RH (RHPC) and a matrix dispersion based on phospholipid complex (RHPC-MD) were successfully prepared and optimized. Several methods were used to validate the formation of RHPC and RHPC-MD, such as differential scanning calorimetry, X-ray diffraction, scanning electron microscopy, transmission electron microscopy, infrared spectroscopy, particle size, and zeta potential, meanwhile, their octanol-water partition coefficient, solubility, and dissolution in vitro were also evaluated. To investigate the absorption mechanism of RHPC in vivo, the RHPC was administered to the chylomicron flow blockage rat model. Interestingly, as we expected, a significant reduction in RHPC absorption (67%) (**p< .01) in presence of cycloheximide (CXI) inhibitor was observed, thus confirming the RHPC could be absorbed by lymphatic transport in vivo. Pharmacokinetic studies revealed that the relative oral bioavailability of RHPC as well as RHPC-MD was 223% and 329%, respectively, when comparing with the commercial RH tablets. These outcomes suggested that the current study provided an attractive formulation to enhance the oral bioavailability of RH and stimulated to further research the absorption mechanism of RHPC in vivo.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Fosfolipídeos/química , Cloridrato de Raloxifeno/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Administração Oral , Animais , Disponibilidade Biológica , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/farmacocinética , Varredura Diferencial de Calorimetria , Quilomícrons/biossíntese , Cicloeximida/administração & dosagem , Liberação Controlada de Fármacos , Feminino , Humanos , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Modelos Animais , Osteoporose Pós-Menopausa/tratamento farmacológico , Tamanho da Partícula , Cloridrato de Raloxifeno/química , Cloridrato de Raloxifeno/farmacocinética , Ratos , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/farmacocinética , Solubilidade , Comprimidos , Difração de Raios X
4.
Pharm Dev Technol ; 25(7): 899-907, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32324081

RESUMO

Calcitriol, as the biologically active form of vitamin D3, is essential for patients with renal osteopathy. The solubilization, stabilization, and content uniformity are key issues in its formulation development. In our previous study, the incomplete release of calcitriol was solved by using the hybrid lipid-based solid dispersion (SD) for calcitriol. However, good stability and content uniformity are still urgently needed. In this study, solid lipid with antioxidant properties and liquid lipid compatible with calcitriol were employed as hybrid lipid carrier (HLC) to establish a solid dispersion. Moreover, the content uniformity of tablets with hybrid lipid carrier based SDs (HLCTs) was further guaranteed due to the multi-dispersion of calcitriol in HLC, solidification, and blank granules. Additionally, the compression of the blank granules was adjusted by the water content. The mixing method of calcitriol-containing and blank granules was also optimized. The obtained HLCTs were evaluated for hardness, disintegration time, in vitro drug dissolution, content uniformity, and stability. Satisfactory HLCTs were developed successfully in this study with superior content uniformity and better stability than the commercial soft capsule (Rocaltrol®). It was proved to be a promising formulation for drugs with poor water-solubility, instability to oxygen and heat, and dose-related toxicity.


Assuntos
Conservadores da Densidade Óssea/síntese química , Calcitriol/síntese química , Portadores de Fármacos/síntese química , Composição de Medicamentos/métodos , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/farmacocinética , Calcitriol/administração & dosagem , Calcitriol/farmacocinética , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Liberação Controlada de Fármacos/fisiologia , Estabilidade de Medicamentos , Comprimidos
5.
AAPS PharmSciTech ; 21(2): 71, 2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-31953574

RESUMO

This study developed a thermosensitive hydrogel based on poly(2-ethyl-2-oxazoline)-poly(D,L-lactide)-poly(2-ethyl-2-oxazoline) (PPP) for the delivery of salmon calcitonin to improve the hypocalcemic effect. The tube inversion and rheological tests revealed that the copolymer solution underwent temperature-dependent sol-gel-sol transitions. Observation by scanning electron microscopy (SEM) showed that the hydrogel exhibited a porous three-dimensional network. The swelling test demonstrated that there was a maximum swelling ratio at low temperature (25°C) as compared with the high temperature (37°C). In vitro release revealed that the PPP hydrogel were capable of sustained release of salmon calcitonin (sCT). The in vivo biodegradability study indicated the good degradability of PPP hydrogel. More importantly, the in vivo retention time of the hydrogel in situ was significantly prolonged after subcutaneous injection of the PPP hydrogel compared to the F127 hydrogel. In vivo pharmacodynamics analysis showed that the hypocalcemic effect of both PPP and F127 hydrogel was significantly greater than that of sCT solution, and the mean serum Ca reduction effect could be maintained for 24 h of PPP hydrogel, indicating that PPP hydrogel could achieve a significant enhanced hypocalcemic effect. In conclusion, the PPP hydrogel has been shown to be prospective as a controlled release carrier for injection delivery of protein drugs.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Calcitonina/administração & dosagem , Hidrogéis/química , Animais , Conservadores da Densidade Óssea/farmacocinética , Calcitonina/farmacocinética , Cálcio/sangue , Preparações de Ação Retardada , Composição de Medicamentos , Masculino , Camundongos , Microscopia Eletrônica de Varredura , Oxazóis , Poliaminas , Poliésteres , Polímeros , Ratos , Ratos Sprague-Dawley , Reologia , Temperatura
6.
Acta Orthop ; 91(2): 126-132, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31680611

RESUMO

Background and purpose - Targeted delivery of drugs is important to achieve efficient local concentrations and reduce systemic side effects. We hypothesized that locally implanted synthetic hydroxyapatite (HA) particles can act as a recruiting moiety for systemically administered drugs, leading to targeted drug accretion.Methods - Synthetic HA particles were implanted ectopically in a muscle pouch in rats, and the binding of systemically circulating drugs such as zoledronic acid (ZA), tetracycline and 18F-fluoride (18F) was studied. The local biological effect was verified in an implant integration model in rats, wherein a hollow implant was filled with synthetic HA particles and the animals were given systemic ZA, 2-weeks post-implantation. The effect of HA particle size on drug binding and the possibility of reloading HA particles were also evaluated in the muscle pouch.Results - The systemically administered biomolecules (ZA, tetracycline and 18F) all sought the HA moiety placed in the muscle pouch. Statistically significant higher peri-implant bone volume and peak force were observed in the implant containing HA particles compared with the empty implant. After a single injection of ZA at 2 weeks, micro HA particles showed a tendency to accumulate more 14C-zoledronic acid (14C-ZA) than nano-HA particles in the muscle pouch. HA particles could be reloaded when ZA was given again at 4 weeks, showing increased 14C-ZA accretion by 73% in microparticles and 77% in nanoparticles.Interpretation - We describe a novel method of systemic drug loading resulting in targeted accretion in locally implanted particulate HA, thereby biologically activating the material.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Sistemas de Liberação de Medicamentos , Durapatita/metabolismo , Ácido Zoledrônico/administração & dosagem , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Conservadores da Densidade Óssea/farmacocinética , Materiais Revestidos Biocompatíveis , Portadores de Fármacos , Fluoretos/administração & dosagem , Fluoretos/farmacocinética , Implantes Experimentais , Masculino , Tamanho da Partícula , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ratos Sprague-Dawley , Tetraciclina/administração & dosagem , Tetraciclina/farmacocinética , Ácido Zoledrônico/farmacocinética
7.
Osteoporos Int ; 30(3): 675-683, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30357438

RESUMO

To demonstrate the clinical comparability between RGB-10 (a biosimilar teriparatide) and the originator, a comparative pharmacokinetic trial was conducted. The study was successful in establishing bioequivalence. Marketing authorisation for RGB-10 (Terrosa®) was granted by the European Medicines Agency in 2017. INTRODUCTION: Teriparatide, the first bone anabolic agent, is the biologically active fragment of human parathyroid hormone. The imminent patent expiry of the originator will open the door for biosimilars to enter the osteology market, thereby improving access to a highly effective, yet prohibitively expensive therapy. METHODS: Subsequent to establishing comparability on the quality and non-clinical levels between RGB-10, a biosimilar teriparatide, and its reference product (Forsteo®), a randomised, double-blind, 2-way cross-over comparative study (duration: four days) was conducted in 54 healthy women (ages: 18 to 55 years) to demonstrate the pharmacokinetic/pharmacodynamic (PK/PD) equivalence and comparable safety of these products. Extents of exposure (AUC0-tlast) and peak exposure (Cmax), as measured by means of ELISA, were evaluated as co-primary PK endpoints, and serum calcium levels, as measured using standard automated techniques, were assessed for PD effects. Safety was monitored throughout the study. RESULTS: The 94.12% CIs for the ratio of the test to the reference treatments, used due to the two-stage design (85.20-98.60% and 85.51-99.52% for AUC0-tlast and Cmax, respectively), fell within the 80.00-125.00% acceptance range. The calcium PD parameters were essentially identical with geometric mean ratios (GMRs) of 99.93% and 99.87% for AUC and Cmax, respectively. Analysis of the safety data did not reveal any differences between RGB-10 and its reference. CONCLUSION: Based on the high level of similarity in the preclinical data and the results of this clinical study, marketing authorisation for RGB-10 (Terrosa®) was granted by the European Medicines Agency (EMA) in 2017.


Assuntos
Medicamentos Biossimilares/farmacologia , Medicamentos Biossimilares/farmacocinética , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/farmacocinética , Osteoporose/tratamento farmacológico , Teriparatida/farmacologia , Teriparatida/farmacocinética , Adolescente , Adulto , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/uso terapêutico , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/uso terapêutico , Cálcio/sangue , Estudos Cross-Over , Método Duplo-Cego , Feminino , Meia-Vida , Humanos , Injeções Subcutâneas , Pessoa de Meia-Idade , Teriparatida/efeitos adversos , Teriparatida/uso terapêutico , Equivalência Terapêutica , Adulto Jovem
8.
Br J Clin Pharmacol ; 85(6): 1136-1146, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30690761

RESUMO

Mathematical modelling and simulation (M&S) of drug concentrations, pharmacologic effects and the (patho)physiologic systems within which they interact can be powerful tools for the preclinical, translational and clinical development of drugs. Indeed, the Prescription Drug User Fee Act (PDUFA VI), incorporated as part of the FDA Reauthorization Act of 2017 (FDARA), highlights the goal of advancing model-informed drug development (MIDD). MIDD can benefit development across many drug classes, including for metabolic bone diseases such as osteoporosis, cancer-related and numerous rare metabolic bone diseases; conditions characterized by significant morbidity and mortality. A drought looms in terms of the availability of new drugs to better treat these devastating diseases. This review provides an overview of several M&S approaches ranging from simple pharmacokinetic to integrated pharmacometric and systems pharmacology modelling. Examples are included to illustrate the use of these approaches during the development of several drugs for metabolic bone diseases such as bisphosphonates, denosumab, teriparatide and sclerostin inhibitors (romosozumab and blosozumab).


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Doenças Ósseas Metabólicas/tratamento farmacológico , Remodelação Óssea/efeitos dos fármacos , Desenvolvimento de Medicamentos/métodos , Modelos Biológicos , Biologia de Sistemas , Animais , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Doenças Ósseas Metabólicas/fisiopatologia , Simulação por Computador , Monitoramento de Medicamentos , Humanos , Segurança do Paciente , Medição de Risco
9.
Br J Clin Pharmacol ; 85(6): 1052-1062, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30650219

RESUMO

The biological effects of the bisphosphonates (BPs) as inhibitors of calcification and bone resorption were first described in the late 1960s. In the 50 years that have elapsed since then, the BPs have become the leading drugs for the treatment of skeletal disorders characterized by increased bone resorption, including Paget's disease of bone, bone metastases, multiple myeloma, osteoporosis and several childhood inherited disorders. The discovery and development of the BPs as a major class of drugs for the treatment of bone diseases is a paradigm for the successful journey from "bench to bedside and back again". Several of the leading BPs achieved "blockbuster" status as branded drugs. However, these BPs have now come to the end of their patent life, making them highly affordable. The opportunity for new clinical applications for BPs also exists in other areas of medicine such as ageing, cardiovascular disease and radiation protection. Their use as inexpensive generic medicines is therefore likely to continue for many years to come. Fifty years of research into the pharmacology of bisphosphonates have led to a fairly good understanding about how these drugs work and how they can be used safely in patients with metabolic bone diseases. However, while we seemingly know much about these drugs, a number of key aspects related to BP distribution and action remain incompletely understood. This review summarizes the existing knowledge of the (pre)clinical and translational pharmacology of BPs, and highlights areas in which understanding is lacking.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Doenças Ósseas Metabólicas/tratamento farmacológico , Remodelação Óssea/efeitos dos fármacos , Difosfonatos/uso terapêutico , Animais , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Doenças Ósseas Metabólicas/diagnóstico , Doenças Ósseas Metabólicas/epidemiologia , Doenças Ósseas Metabólicas/fisiopatologia , Difosfonatos/efeitos adversos , Difosfonatos/farmacocinética , Humanos , Fatores de Risco , Resultado do Tratamento
10.
Br J Clin Pharmacol ; 85(6): 1125-1135, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30536446

RESUMO

Potent antiresorptive drugs (bisphosphonate and denosumab) are often used to protect bone health in postmenopausal breast cancer patients. In addition, clinical trials have shown that these drugs increase disease-free survival, though the mechanism of adjuvant benefit is largely unknown. Here we review the bone health and adjuvant data for both classes of antiresorptive drugs and highlight differences in their pharmacology. Inhibition of bone resorption is vitally important to protect against osteoporotic fractures, and may also contribute to adjuvant survival benefits by making the bone microenvironment less amenable to breast cancer metastasis. After a course of therapy, stoppage of bisphosphonates yields a persistent antiresorptive effect, whereas discontinuation of denosumab causes a rebound increase in bone resorption markers and a loss of bone mineral density to baseline levels. Whether the potential adjuvant benefits of denosumab are also rapidly lost after drug discontinuation deserves further investigation.


Assuntos
Inibidores da Aromatase/efeitos adversos , Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Reabsorção Óssea/prevenção & controle , Neoplasias da Mama/tratamento farmacológico , Denosumab/uso terapêutico , Difosfonatos/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Neoplasias Ósseas/mortalidade , Neoplasias Ósseas/fisiopatologia , Neoplasias Ósseas/secundário , Reabsorção Óssea/mortalidade , Reabsorção Óssea/fisiopatologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Quimioterapia Adjuvante , Denosumab/efeitos adversos , Denosumab/farmacocinética , Difosfonatos/efeitos adversos , Difosfonatos/farmacocinética , Intervalo Livre de Doença , Feminino , Fraturas Ósseas/fisiopatologia , Fraturas Ósseas/prevenção & controle , Humanos , Pós-Menopausa , Medição de Risco , Fatores de Risco
11.
Br J Clin Pharmacol ; 85(6): 1072-1083, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30663085

RESUMO

Cathepsin K (CatK) is a cysteine protease abundantly expressed by osteoclasts and localized in the lysosomes and resorption lacunae of these cells. CatK is the principal enzyme responsible for the degradation of bone collagen. Odanacatib is a selective, reversible inhibitor of CatK at subnanomolar potency. The pharmacokinetics of odanacatib have been extensively studied and are similar in young healthy men, postmenopausal women and elderly men, and were qualitatively similar throughout Phase 1 development and in-patient studies. Following 3 weeks of 50 mg once weekly dosing the geometric mean area under the curve from 0 to 168 hours was 41.1 µM h, the concentration at 168 hours was 126 nM and the harmonic mean apparent terminal half-life was 84.8 hr. Odanacatib exposure increased in a less than dose proportional manner due to solubility limited absorption. It is estimated that approximately 70% of the absorbed dose of odanacatib is eliminated via metabolism, 20% is excreted as unchanged drug in the bile or faeces, and 10% is excreted as unchanged drug in the urine. The systemic clearance was low (approximately 13 mL/min). Odanacatib decreases the degradation of bone matrix proteins and reduces the efficiency of bone resorption with target engagement confirmed by a robust decrease in serum C-telopeptides of type 1 collagen (approximately 60%), urinary aminoterminal crosslinked telopeptides of type 1 collagen to creatinine ratio (approximately 50%) and total urine deoxypyridinoline/Cr (approximately 30%), with an increase in serum cross-linked carboxy-terminal telopeptide of type 1 collagen (approximately 55%). The 50-mg weekly dosing regimen evaluated in Phase 3 achieved near maximal reduction in bone resorption throughout the treatment period. The extensive clinical programme for odanacatib, together with more limited clinical experience with other CatK inhibitors (balicatib and ONO-5334), provides important insights into the clinical pharmacology of CatK inhibition and the potential role of CatK in bone turnover and mineral homeostasis. Key findings include the ability of this mechanism to: (i) provide sustained reductions in resorption markers, increases in bone mineral density, and demonstrated fracture risk reduction; (ii) be associated with relative formation-sparing effects such that sustained resorption reduction is achieved without accompanying meaningful reductions in bone formation; and (iii) lead to increases in osteoclast number as well as other osteoclast activity (including build-up of CatK enzyme), which may yield transient increases in resorption following treatment discontinuation and the potential for nonmonotonic responses at subtherapeutic doses.


Assuntos
Compostos de Bifenilo/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/uso terapêutico , Osteoporose/tratamento farmacológico , Animais , Compostos de Bifenilo/efeitos adversos , Compostos de Bifenilo/farmacocinética , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Osso e Ossos/enzimologia , Osso e Ossos/fisiopatologia , Catepsina K/metabolismo , Inibidores de Cisteína Proteinase/efeitos adversos , Inibidores de Cisteína Proteinase/farmacocinética , Feminino , Humanos , Masculino , Osteoporose/enzimologia , Osteoporose/patologia , Transdução de Sinais , Pesquisa Translacional Biomédica , Resultado do Tratamento
12.
Br J Clin Pharmacol ; 85(6): 1095-1102, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30308088

RESUMO

Pharmacologists have been interested in vitamin D since its metabolism was elucidated in the early 1970s. Despite the synthesis of thousands of vitamin D analogues in the hope of separating its calcemic and anti-proliferative properties, few molecules have reached the market for use in the treatment of clinical conditions from psoriasis to chronic kidney disease. This review discusses vitamin D drugs, recently developed or still under development, for use in various diseases, but in particular bone disease. In the process we explore the mechanisms postulated to explain the action of these vitamin D analogues including action through the vitamin D receptor, action through other receptors e.g. FAM57B2 and dual action on transcriptional processes.


Assuntos
24,25-Di-Hidroxivitamina D 3/uso terapêutico , Conservadores da Densidade Óssea/uso terapêutico , Doenças Ósseas Metabólicas/tratamento farmacológico , Remodelação Óssea/efeitos dos fármacos , Calcifediol/uso terapêutico , Calcitriol/uso terapêutico , Vitaminas/uso terapêutico , 24,25-Di-Hidroxivitamina D 3/efeitos adversos , 24,25-Di-Hidroxivitamina D 3/farmacocinética , Animais , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacocinética , Doenças Ósseas Metabólicas/diagnóstico , Doenças Ósseas Metabólicas/fisiopatologia , Calcifediol/efeitos adversos , Calcifediol/farmacocinética , Calcitriol/efeitos adversos , Calcitriol/farmacocinética , Humanos , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Resultado do Tratamento , Vitaminas/efeitos adversos , Vitaminas/farmacocinética
13.
Drug Dev Ind Pharm ; 45(5): 787-804, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30672348

RESUMO

Hydrogels forming in-situ have gained great attention in the area of bone tissue engineering recently, they were also showed to be a good and less invasive alternative to surgically applied ones. The primal focus of this study was to prepare chitosan-glycerol phosphate thermosensitive hydrogel formed in-situ and loaded with risedronate (bone resorption inhibitor) in an easy way with no requirement of complicated processes or large number of equipment. Then we investigated its effectiveness for bone regeneration. In-situ forming hydrogels were prepared using chitosan cross-linked with glycerol phosphate and loaded with risedronate and nano-hydroxyapatite as bone cement. The prepared hydrogels were characterized by analyzing their gelation time at 37 °C, % porosity, swelling index, in-vitro degradation, rheological properties, and in-vitro drug release. Results showed that the in-situ hydrogels prepared using 2.5% (w/v) chitosan cross-linked with 50% (w/v) glycerol phosphate in the ratio (9:1, v/v) reinforced with 20 mg/mL and nano-hydroxyapatite possessed the most sustained drug release profile. This optimized formulation was further evaluated using DSC and FTIR studies, in addition to their morphological properties using scanning electron microscopy. The effect on Saos-2 cell line viability was evaluated also using MTT assay on the optimized hydrogel formulation in addition to their action on cell proliferation using fluorescence microscope. Moreover, calcium deposition on the hydrogel and alkaline phosphatase activity were evaluated. Risedronate-nano-hydroxyapatite loaded hydrogels significantly enhanced the Saos-2 cell proliferation in addition to enhanced alkaline phosphatase activity and calcium deposition. Such results suggest that risedronate-nano-hydroxyapatite loaded hydrogels present great biocompatibility for bone regeneration. Proliferation of cells, as well as deposition of mineral on the hydrogel, was an evidence of the biocompatible nature of the hydrogel. This hydrogel formed in-situ present a good less invasive alternative for bone tissue engineering.


Assuntos
Conservadores da Densidade Óssea/administração & dosagem , Regeneração Óssea/efeitos dos fármacos , Durapatita/química , Osteogênese/efeitos dos fármacos , Ácido Risedrônico/administração & dosagem , Cimentos Ósseos/química , Conservadores da Densidade Óssea/farmacocinética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Liberação Controlada de Fármacos , Glicerofosfatos/química , Humanos , Hidrogéis/química , Teste de Materiais , Nanopartículas/química , Porosidade , Ácido Risedrônico/farmacocinética , Temperatura , Engenharia Tecidual/métodos
14.
AAPS PharmSciTech ; 20(1): 24, 2019 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-30604153

RESUMO

Tamoxifen is an antiestrogen drug that is widely used in the adjuvant chemotherapy of estrogen receptor-α (ERα)-positive breast cancer. Chemotherapy could suppress immune function in breast cancer patients, which may cause invasive fungal infections (IFIs). Triazoles (voriconazole, fluconazole, and itraconazole) were commonly used for IFI. The physiologically based pharmacokinetic (PBPK) models were developed to investigate the influence of different triazoles on tamoxifen pharmacokinetics in this paper. To investigate the influence of different triazoles (voriconazole, fluconazole, itraconazole) on tamoxifen pharmacokinetics. Adjusted physicochemical data and pharmacokinetic parameters of voriconazole, fluconazole, itraconazole, and tamoxifen were obtained from published literatures. PBPK models were built and verified in healthy subjects using GastroPlus™. Voriconazole, itraconazole, and tamoxifen were administered orally. Fluconazole was administered intravenously. Simulated plasma concentration-time curves of the voriconazole, fluconazole, itraconazole, and tamoxifen showed good agreement with the observed profiles, respectively. The DDI simulations showed that the pharmacokinetic parameters of tamoxifen were increased by various degrees when coadministered with different triazoles. In healthy subjects, the area under the plasma concentration-time curve from 0 to t h (AUC0-t) of tamoxifen was increased by 41%, 5%, and1% when coadministrated with voriconazole, fluconazole, and itraconazole, respectively. The PBPK models adequately characterized the pharmacokinetics of tamoxifen and triazoles. Among the three triazoles, voriconazole exhibited the greatest effect on tamoxifen pharmacokinetics. In clinical practice, an effective dosage adjustment of tamoxifen may need to be considered and TDM for tamoxifen is advisable to guide dosing and optimize therapy when coadministered with voriconazole.


Assuntos
Antifúngicos/farmacocinética , Conservadores da Densidade Óssea/farmacocinética , Modelos Biológicos , Tamoxifeno/farmacocinética , Triazóis/farmacocinética , Interações Medicamentosas/fisiologia , Feminino , Fluconazol/farmacocinética , Previsões , Humanos , Itraconazol/farmacologia , Pessoa de Meia-Idade , Voriconazol/farmacocinética
15.
Osteoporos Int ; 29(9): 2139-2146, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29947866

RESUMO

This work examines the skeletal accumulation of fluorescently tagged zoledronate in an animal model of chronic kidney disease. The results show higher accumulation in 24-h post-dose animals with lower kidney function due to greater amounts of binding at individual surfaces. INTRODUCTION: Chronic kidney disease (CKD) patients suffer from increased rates of skeletal-related mortality from changes driven by biochemical abnormalities. Bisphosphonates are commonly used in reducing fracture risk in a variety of diseases, yet their use is not recommended in advanced stages of CKD. This study aimed to characterize the accumulation of a single dose of fluorescently tagged zoledronate (FAM-ZOL) in the setting of reduced kidney function. METHODS: At 25 weeks of age, FAM-ZOL was administered to normal and CKD rats. Twenty-four hours later, multiple bones were collected and assessed using bulk fluorescence imaging, two-photon imaging, and dynamic histomorphometry. RESULTS: CKD animals had significantly higher levels of FAM-ZOL accumulation in the proximal tibia, radius, and ulna, but not in lumbar vertebral body or mandible, based on multiple measurement modalities. Although a majority of trabecular bone surfaces were covered with FAM-ZOL in both normal and CKD animals, the latter had significantly higher levels of fluorescence per unit bone surface in the proximal tibia. CONCLUSIONS: These results provide new data regarding how reduced kidney function affects drug accumulation in rat bone.


Assuntos
Conservadores da Densidade Óssea/farmacocinética , Osso e Ossos/metabolismo , Insuficiência Renal Crônica/metabolismo , Ácido Zoledrônico/farmacocinética , Animais , Osso e Ossos/diagnóstico por imagem , Modelos Animais de Doenças , Masculino , Imagem Óptica/métodos , Ratos Endogâmicos , Tíbia/diagnóstico por imagem , Tíbia/metabolismo
16.
Osteoporos Int ; 28(12): 3475-3487, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28956091

RESUMO

Strontium ranelate treatment is known to prevent fractures. Here, we showed that strontium ranelate treatment enhances bone healing and affects bone cellular activities differently in intact and healing bone compartments: Bone formation was increased only in healing compartment, while resorption was reduced in healing and normal bone compartments. INTRODUCTION: Systemic administration of strontium ranelate (SrRan) accelerates the healing of bone defects; however, controversy about its action on bone formation remains. We hypothesize that SrRan could affect bone formation differently in normal mature bone or in the bone healing process. METHODS: Proximal tibia bone defects were created in 6-month-old female rats, which orally received SrRan (625 mg/kg/day, 5/7 days) or vehicle (control groups) for 4, 8, or 12 weeks. Bone samples were analyzed by micro-computed tomography and histomorphometry in various regions, i.e., metaphyseal 2nd spongiosa, a region close to the defect, within the healing defect and in cortical defect bridging region. Additionally, we evaluated the quality of the new bone formed by quantitative backscattered electron imaging and by red picosirius histology. RESULTS: Healing of the bone defect was characterized by a rapid onset of bone formation without cartilage formation. Cortical defect bridging was detected earlier compared with healing of trabecular defect. In the healing zone, SrRan stimulated bone formation early and laterly decreased bone resorption improving the healing of the cortical and trabecular compartment without deleterious effects on bone quality. By contrast, in the metaphyseal compartment, SrRan only decreased bone resorption from week 8 without any change in bone formation, leading to little progressive increase of the metaphyseal trabecular bone volume. CONCLUSIONS: SrRan affects bone formation differently in normal mature bone or in the bone healing process. Despite this selective action, this led to similar increased bone volume in both compartments without deleterious effects on the newly bone-formed quality.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Osteogênese/efeitos dos fármacos , Tiofenos/farmacologia , Tíbia/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Animais , Conservadores da Densidade Óssea/farmacocinética , Remodelação Óssea/efeitos dos fármacos , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/metabolismo , Osso Esponjoso/fisiopatologia , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Osteogênese/fisiologia , Ratos Sprague-Dawley , Tiofenos/farmacocinética , Tíbia/lesões , Tíbia/metabolismo , Tíbia/fisiopatologia , Cicatrização/fisiologia , Microtomografia por Raio-X
17.
Pediatr Nephrol ; 32(7): 1221-1232, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28332096

RESUMO

BACKGROUND: Elevated intact parathyroid hormone (iPTH) levels can contribute to morbidity and mortality in children with chronic kidney disease (CKD). We evaluated the pharmacokinetics, efficacy, and safety of oral paricalcitol in reducing iPTH levels in children with stages 3-5 CKD. METHODS: Children aged 10-16 years with stages 3-5 CKD were enrolled in two phase 3 studies. The stage 3/4 CKD study characterized paricalcitol pharmacokinetics and compared the efficacy and safety of paricalcitol with placebo followed by an open-label period. The stage 5 CKD study evaluated the efficacy and safety of paricalcitol (no comparator) in children with stage 5 CKD undergoing dialysis. RESULTS: In the stage 3/4 CKD study, mean peak plasma concentration and area under the time curve from zero to infinity were 0.13 ng/mL and 2.87 ng•h/((or ng×h/))mL, respectively, for 12 children who received 3 µg paricalcitol. Thirty-six children were randomized to paricalcitol or placebo; 27.8% of the paricalcitol group achieved two consecutive iPTH reductions of ≥30% from baseline versus none of the placebo group (P = 0.045). Adverse events were higher in children who received placebo than in those administered paricalcitol during the double-blind treatment (88.9 vs. 38.9%; P = 0.005). In the stage 5 CKD study, eight children (61.5%) had two consecutive iPTH reductions of ≥30% from baseline, and five (38.5%) had two consecutive iPTH values of between 150 and 300 pg/mL. Clinically meaningful hypercalcemia occurred in 21% of children. CONCLUSIONS: Oral paricalcitol in children aged 10-16 years with stages 3-5 CKD reduced iPTH levels and the treatment was well tolerated. Results support an initiating dose of 1 µg paricalcitol 3 times weekly in children aged 10-16 years.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Ergocalciferóis/uso terapêutico , Hiperparatireoidismo Secundário/terapia , Falência Renal Crônica/terapia , Hormônio Paratireóideo/sangue , Diálise Renal , Adolescente , Conservadores da Densidade Óssea/farmacocinética , Cálcio/sangue , Criança , Método Duplo-Cego , Ergocalciferóis/farmacocinética , Feminino , Humanos , Hipercalcemia/sangue , Hipercalcemia/induzido quimicamente , Hiperparatireoidismo Secundário/sangue , Hiperparatireoidismo Secundário/etiologia , Hiperfosfatemia/sangue , Hiperfosfatemia/induzido quimicamente , Falência Renal Crônica/sangue , Falência Renal Crônica/complicações , Masculino , Fósforo/sangue , Resultado do Tratamento
18.
J Pharmacokinet Pharmacodyn ; 44(6): 599-609, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29081020

RESUMO

A hazard model of fracture was developed using individual patient data (IPD) from the NHANES (2005-2008) database and summary-level data from an aggregate dataset (AD). The AD was built by performing a comprehensive and systematic literature search of clinical studies published from 1995 to 2015, recording fracture rate and bone mineral density (BMD) for both treatment and placebo arms. The search resulted in a metadata set comprised of 21 studies investigating the effects of various bisphosphonates, teriparatide, denosumab, and raloxifene in 65,254 patients over a cumulative 56.75 years of study. The IPD was used to augment an AD in a model-based meta-analysis (MBMA) hierarchical modeling approach. The resulting model predicts the probability of fracture events in patients with osteoporosis. The object of model building using this approach was to promote understanding of the impact of therapeutic drug effects on the probability of fracture together with, or independent of their effects on BMD. Candidate models were evaluated by deviance information criteria and posterior predictive check. The model with covariates for lumbar spine BMD with interaction with a drug effect on BMD, and patient body mass index, years post-menopause, fracture measure method (clinical or radiological) and an additional drug effect outperformed those models without interaction and without additional drug effects. The model quantitatively supports the widely held notion that changes in bone microarchitecture, which cannot be measured by areal BMD elicited by therapy contribute in a significant way to a reduction in fracture. Furthermore, this model can be used to simulate fracture risk in a clinical cohort similar to those contained in the MBMA.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Difosfonatos/uso terapêutico , Modelos Biológicos , Osteoporose Pós-Menopausa/tratamento farmacológico , Fraturas por Osteoporose/tratamento farmacológico , Idoso , Densidade Óssea/fisiologia , Conservadores da Densidade Óssea/farmacocinética , Difosfonatos/farmacocinética , Feminino , Humanos , Pessoa de Meia-Idade , Inquéritos Nutricionais/tendências , Osteoporose/diagnóstico , Osteoporose/tratamento farmacológico , Osteoporose/epidemiologia , Osteoporose Pós-Menopausa/diagnóstico , Osteoporose Pós-Menopausa/epidemiologia , Fraturas por Osteoporose/diagnóstico , Fraturas por Osteoporose/epidemiologia , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento
19.
Br J Clin Pharmacol ; 81(2): 362-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26447647

RESUMO

AIMS: Denosumab is a fully human monoclonal immunoglobulin G2 antibody that inhibits bone resorption and increases bone mass and strength. The present clinical study assessed serum and seminal fluid pharmacokinetics following a single denosumab dose in healthy men, and evaluated whether denosumab in seminal fluid poses any risk to a fetus in the event of unprotected sexual intercourse with a pregnant partner. METHODS: An open-label, single-dose study in 12 healthy men was conducted over a 106-day period. Subjects received a single subcutaneous dose of 60-mg denosumab on day 1. Serum and seminal fluid samples were collected at specified time points to assess denosumab pharmacokinetics. Adverse events were recorded. RESULTS: Denosumab was measurable at low concentrations in seminal fluid (~2% of serum concentrations). The mean [standard deviation (SD)] maximum observed drug concentration (Cmax ) was 6170 (2070) ng ml(-1) (serum) and 100 (81.9) ng ml(-1) (seminal fluid). The median time to Cmax (tmax ) was 8 days (serum) and 21 days (seminal fluid). The mean (SD) area under the plasma concentration-time curve (AUC) from time zero to the time of the last quantifiable concentration (AUClast ) was 333 000 (122 000) day•ng ml(-1) (serum) and 5220 (4880) day•ng ml(-1) (seminal fluid). The mean (SD) Cmax and AUC ratios between seminal fluid and serum were 0.0217 (0.0154) and 0.0170 (0.0148), respectively. Using conservative assumptions for ejaculate volume (6 ml), vaginal absorption (100%) and placental transfer (100%), the measured mean denosumab seminal fluid Cmax would result in fetal exposure that was more than 110 times below the preclinically derived 'no effect level' for denosumab. CONCLUSIONS: These results indicate a negligible risk to a fetus exposed to denosumab via seminal fluid transfer to a pregnant partner.


Assuntos
Conservadores da Densidade Óssea/farmacocinética , Denosumab/farmacocinética , Troca Materno-Fetal , Sêmen/metabolismo , Vagina/metabolismo , Adulto , Idoso , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/sangue , Denosumab/administração & dosagem , Denosumab/sangue , Feminino , Voluntários Saudáveis , Humanos , Injeções Subcutâneas , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Gravidez , Risco
20.
Biol Pharm Bull ; 39(3): 323-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26934925

RESUMO

Bisphosphonates are antiosteoporotic agents prescribed for patients with osteoporosis. Drug package inserts for bisphosphonate supplements indicate that their bioavailability is reduced by high levels of metal cations (Ca(2+), Mg(2+), etc.). However, standards for these cations in water used for taking risedronate have not been defined. Here, we examined the effect of calcium and magnesium in mineral waters on the bioavailability of the third-generation bisphosphonate, risedronate, following oral administration in rats. As risedronate is unchanged and eliminated renally, risedronate absorption was estimated from the amount excreted in the urine. Risedronate was dissolved in mineral water samples and administered orally at 0.35 mg/kg. Urine samples were collected for 24 h after dosing. Risedronate was extracted from urine using ion-pair solid-phase cartridges and quantified by HPLC with UV detection (262 nm). Cumulative recovery of risedronate was calculated from the amount excreted in the urine. The 24-h recovery of risedronate from evian® (0.32±0.02% [mean±standard deviation (S.D.)], n=4) and Contrex(®) (0.22±0.05%) mineral waters was significantly lower than that from tap water (0.47±0.04%, p<0.01). Absorption of risedronate in calcium chloride and magnesium chloride aqueous solutions of the same hardness (822 mg/L) was 54% (0.27±0.04%) and 12% (0.51±0.08%) lower, respectively, compared with ultrapure water; suggesting that absorption of risedronate declines as the calcium concentration of mineral waters increases. Consumption of mineral waters containing high levels of calcium (80 mg/L or above), such as evian® and Contrex(®), is therefore not recommended when taking risedronate.


Assuntos
Conservadores da Densidade Óssea/farmacocinética , Cálcio/farmacologia , Magnésio/farmacologia , Águas Minerais , Ácido Risedrônico/farmacocinética , Administração Oral , Animais , Conservadores da Densidade Óssea/urina , Água Potável , Interações Alimento-Droga , Absorção Intestinal , Masculino , Ratos Wistar , Ácido Risedrônico/urina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA