Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pharm Res ; 40(4): 961-975, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36959411

RESUMO

INTRODUCTION: Although the eye is directly accessible on the surface of the human body, drug delivery can be extremely challenging due to the presence of multiple protective barriers in eye tissues. Researchers have developed complex formulation strategies to overcome these barriers to ophthalmic drug delivery. Current development strategies rely heavily on in vitro experiments and animal testing to predict human pharmacokinetics (PK) and pharmacodynamics (PD). OBJECTIVE: The primary objective of the study was to develop a high-fidelity PK/PD model of the anterior eye for topical application of ophthalmic drug products. METHODS: Here, we present a physiologically-based in silico approach to predicting PK and PD in rabbits after topical administration of ophthalmic products. A first-principles based approach was used to describe timolol dissolution, transport, and distribution, including consideration of ionized transport, following topical instillation of a timolol suspension. RESULTS: Using literature transport and response parameters, the computational model described well the concentration-time and response-time profiles in rabbit. Comparison of validated rabbit model results and extrapolated human model results demonstrate observable differences in the distribution of timolol at multiple time points. CONCLUSION: This modeling framework provides a tool for model-based prediction of PK in eye tissues and PD after topical ophthalmic drug administration to the eyes.


Assuntos
Olho , Timolol , Animais , Humanos , Coelhos , Timolol/farmacocinética , Soluções Oftálmicas/farmacocinética , Córnea , Administração Tópica
2.
Clin Infect Dis ; 71(12): 3232-3236, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-32435791

RESUMO

Translation of in vitro antiviral activity to the in vivo setting is crucial to identify potentially effective dosing regimens of hydroxychloroquine. In vitro 50%/90% maximal effective concentration values for hydroxychloroquine should be compared to the in vivo free extracellular tissue concentration, which is similar to the free plasma hydroxychloroquine concentration.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Hidroxicloroquina , Antivirais/uso terapêutico , Humanos , SARS-CoV-2
3.
Mol Pharm ; 15(12): 5454-5467, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30372084

RESUMO

The goal of this project was to explore and to statistically evaluate the responsible gastrointestinal (GI) factors that are significant factors in explaining the systemic exposure of ibuprofen, between and within human subjects. In a previous study, we determined the solution and total concentrations of ibuprofen as a function of time in aspirated GI fluids, after oral administration of an 800 mg IR tablet (reference standard) of ibuprofen to 20 healthy volunteers in fasted state conditions. In addition, we determined luminal pH and motility pressure recordings that were simultaneously monitored along the GI tract. Blood samples were taken to determine ibuprofen plasma levels. In this work, an in-depth statistical and pharmacokinetic analysis was performed to explain which underlying GI variables are determining the systemic concentrations of ibuprofen between (inter-) and within (intra-) subjects. In addition, the obtained plasma profiles were deconvoluted to link the fraction absorbed with the fraction dissolved. Multiple linear regressions were performed to explain and quantitatively express the impact of underlying GI physiology on systemic exposure of the drug (in terms of plasma Cmax/AUC and plasma Tmax). The exploratory analysis of the correlation between plasma Cmax/AUC and the time to the first phase III contractions postdose (TMMC-III) explains ∼40% of the variability in plasma Cmax for all fasted state subjects. We have experimentally shown that the in vivo intestinal dissolution of ibuprofen is dependent upon physiological variables like, in this case, pH and postdose phase III contractions. For the first time, this work presents a thorough statistical analysis explaining how the GI behavior of an ionized drug can explain the systemic exposure of the drug based on the individual profiles of participating subjects. This creates a scientifically based and rational framework that emphasizes the importance of including pH and motility in a predictive in vivo dissolution methodology to forecast the in vivo performance of a drug product. Moreover, as no extensive first-pass metabolism is considered for ibuprofen, this study demonstrates how intraluminal drug behavior is reflecting the systemic exposure of a drug.


Assuntos
Liberação Controlada de Fármacos , Jejum/fisiologia , Absorção Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Ibuprofeno/farmacocinética , Administração Oral , Adulto , Área Sob a Curva , Disponibilidade Biológica , Variação Biológica Individual , Variação Biológica da População/fisiologia , Conjuntos de Dados como Assunto , Feminino , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Ibuprofeno/administração & dosagem , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Solubilidade , Comprimidos , Adulto Jovem
4.
Mol Pharm ; 15(12): 5468-5478, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30417648

RESUMO

Exploring the intraluminal behavior of an oral drug product in the human gastrointestinal (GI) tract remains challenging. Many in vivo techniques are available to investigate the impact of GI physiology on oral drug behavior in fasting state conditions. However, little is known about the intraluminal behavior of a drug in postprandial conditions. In a previous report, we described the mean solution and total concentrations of ibuprofen after oral administration of an immediate-release (IR) tablet in fed state conditions. In parallel, blood samples were taken to assess systemic concentrations. The purpose of this work was to statistically evaluate the impact of GI physiology (e.g., pH, contractile events) within and between individuals (intra and intersubject variability) for a total of 17 healthy subjects. In addition, a pharmacokinetic (PK) analysis was performed by noncompartmental analysis, and PK parameters were correlated with underlying physiological factors (pH, time to phase III contractions postdose) and study parameters (e.g., ingested amount of calories, coadministered water). Moreover, individual plasma profiles were deconvoluted to assess the fraction absorbed as a function of time, demonstrating the link between intraluminal and systemic behavior of the drug. The results demonstrated that the in vivo dissolution of ibuprofen depends on the present gastric pH and motility events at the time of administration. Both intraluminal factors were responsible for explaining 63% of plasma Cmax variability among all individuals. For the first time, an in-depth analysis was performed on a large data set derived from an aspiration/motility study, quantifying the impact of physiology on systemic behavior of an orally administered drug product in fed state conditions. The data obtained from this study will help us to develop an in vitro biorelevant dissolution approach and optimize in silico tools in order to predict the in vivo performance of orally administered drug products, especially in fed state conditions.


Assuntos
Liberação Controlada de Fármacos , Absorção Gástrica/fisiologia , Ibuprofeno/farmacocinética , Período Pós-Prandial/fisiologia , Estômago/fisiologia , Administração Oral , Adulto , Área Sob a Curva , Disponibilidade Biológica , Variação Biológica Individual , Variação Biológica da População/fisiologia , Simulação por Computador , Conjuntos de Dados como Assunto , Feminino , Interações Alimento-Droga/fisiologia , Esvaziamento Gástrico/fisiologia , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Ibuprofeno/administração & dosagem , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Solubilidade , Comprimidos , Adulto Jovem
5.
Pharm Res ; 35(9): 174, 2018 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-29987398

RESUMO

PURPOSE: The mechanisms underlying doxorubicin cytotoxicity and cardiotoxicity were broadly explored but remain incompletely understood. A multiscale physiologically-based pharmacokinetic (PBPK) model was developed to assess doxorubicin dispositions at levels of system, tissue interstitial, cell, and cellular organelles. This model was adopted to explore the mechanisms-of-action/toxicity of doxorubicin in humans. METHODS: The PBPK model was developed by analyzing data from mice and the model was verified by scaling up to predict doxorubicin multiscale dispositions in rats and humans. The multiscale dispositions of doxorubicin in human heart and tumors were explicitly simulated to elucidate the potential mechanisms of its cytotoxicity and cardiotoxicity. RESULTS: The developed PBPK model was able to adequately describe doxorubicin dispositions in mice, rats and humans. In humans, prolonged infusion, a dosing regimen with less cardiotoxicity, was predicted with substantially reduced free doxorubicin concentrations at human heart interstitium, which were lower than the concentrations associated with oxidative stress. However, prolonged infusion did not reduce doxorubicin-DNA adduct at tumor nucleus, consistent with clinical observations that prolonged infusion did not compromise anti-tumor effect, indicating that one primary anti-tumor mechanism was DNA torsion. CONCLUSIONS: A multiscale PBPK model for doxorubicin was developed and further applied to explore its cytotoxic and cardiotoxic mechanisms.


Assuntos
Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/toxicidade , Cardiotoxicidade/etiologia , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidade , Animais , Simulação por Computador , Citotoxinas/farmacocinética , Citotoxinas/toxicidade , Humanos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Ratos , Distribuição Tecidual
6.
Mol Pharm ; 14(12): 4281-4294, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-28737409

RESUMO

In this study, we determined the pH and buffer capacity of human gastrointestinal (GI) fluids (aspirated from the stomach, duodenum, proximal jejunum, and mid/distal jejunum) as a function of time, from 37 healthy subjects after oral administration of an 800 mg immediate-release tablet of ibuprofen (reference listed drug; RLD) under typical prescribed bioequivalence (BE) study protocol conditions in both fasted and fed states (simulated by ingestion of a liquid meal). Simultaneously, motility was continuously monitored using water-perfused manometry. The time to appearance of phase III contractions (i.e., housekeeper wave) was monitored following administration of the ibuprofen tablet. Our results clearly demonstrated the dynamic change in pH as a function of time and, most significantly, the extremely low buffer capacity along the GI tract. The buffer capacity on average was 2.26 µmol/mL/ΔpH in fasted state (range: 0.26 and 6.32 µmol/mL/ΔpH) and 2.66 µmol/mL/ΔpH in fed state (range: 0.78 and 5.98 µmol/mL/ΔpH) throughout the entire upper GI tract (stomach, duodenum, and proximal and mid/distal jejunum). The implication of this very low buffer capacity of the human GI tract is profound for the oral delivery of both acidic and basic active pharmaceutical ingredients (APIs). An in vivo predictive dissolution method would require not only a bicarbonate buffer but also, more significantly, a low buffer capacity of dissolution media to reflect in vivo dissolution conditions.


Assuntos
Líquidos Corporais/química , Motilidade Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Ibuprofeno/farmacocinética , Absorção Intestinal/fisiologia , Absorção Fisiológica , Administração Oral , Adulto , Líquidos Corporais/fisiologia , Soluções Tampão , Liberação Controlada de Fármacos , Voluntários Saudáveis , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/fisiologia , Manometria , Pessoa de Meia-Idade , Solubilidade , Comprimidos , Equivalência Terapêutica , Fatores de Tempo , Adulto Jovem
7.
Drug Metab Dispos ; 44(7): 1123-38, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27098743

RESUMO

We used the intestinal segregated flow model (SFM) versus the traditional model (TM), nested within physiologically based pharmacokinetic (PBPK) models, to describe the biliary and urinary excretion of morphine 3ß-glucuronide (MG) after intravenous and intraduodenal dosing of morphine in rats in vivo. The SFM model describes a partial (5%-30%) intestinal blood flow perfusing the transporter- and enzyme-rich enterocyte region, whereas the TM describes 100% flow perfusing the intestine as a whole. For the SFM, drugs entering from the circulation are expected to be metabolized to lesser extents by the intestine due to the segregated flow, reflecting the phenomenon of shunting and route-dependent intestinal metabolism. The poor permeability of MG crossing the liver or intestinal basolateral membranes mandates that most of MG that is excreted into bile is hepatically formed, whereas MG that is excreted into urine originates from both intestine and liver metabolism, since MG is effluxed back to blood. The ratio of MG amounts in urine/bile [Formula: see text] for intraduodenal/intravenous dosing is expected to exceed unity for the SFM but approximates unity for the TM. Compartmental analysis of morphine and MG data, without consideration of the permeability of MG and where MG is formed, suggests the ratio to be 1 and failed to describe the kinetics of MG. The observed intraduodenal/intravenous ratio of [Formula: see text] (2.55 at 4 hours) was better predicted by the SFM-PBPK (2.59 at 4 hours) and not the TM-PBPK (1.0), supporting the view that the SFM is superior for the description of intestinal-liver metabolism of morphine to MG. The SFM-PBPK model predicts an appreciable contribution of the intestine to first pass M metabolism.


Assuntos
Duodeno/irrigação sanguínea , Duodeno/metabolismo , Circulação Hepática , Fígado/irrigação sanguínea , Fígado/metabolismo , Modelos Biológicos , Derivados da Morfina/farmacocinética , Morfina/farmacocinética , Circulação Esplâncnica , Administração Intravenosa , Administração Oral , Animais , Permeabilidade da Membrana Celular , Eliminação Hepatobiliar , Inativação Metabólica , Masculino , Morfina/administração & dosagem , Morfina/sangue , Morfina/urina , Derivados da Morfina/sangue , Derivados da Morfina/urina , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Eliminação Renal
8.
Gastroenterology ; 146(4): 1048-59, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24365583

RESUMO

BACKGROUND & AIMS: Little is known about the effects of the vitamin D receptor (VDR) on hepatic activity of human cholesterol 7α-hydroxylase (CYP7A1) and cholesterol metabolism. We studied these processes in mice in vivo and mouse and human hepatocytes. METHODS: Farnesoid X receptor (Fxr)(-/-), small heterodimer partner (Shp)(-/-), and C57BL/6 (wild-type control) mice were fed normal or Western diets for 3 weeks and were then given intraperitoneal injections of vehicle (corn oil) or 1α,25-dihydroxyvitamin D3 (1,25[OH]2D3; 4 doses, 2.5 µg/kg, every other day). Plasma and tissue samples were collected and levels of Vdr, Shp, Cyp7a1, Cyp24a1, and rodent fibroblast growth factor (Fgf) 15 expression, as well as levels of cholesterol, were measured. We studied the regulation of Shp by Vdr using reporter and mobility shift assays in transfected human embryonic kidney 293 cells, quantitative polymerase chain reaction with mouse tissues and mouse and human hepatocytes, and chromatin immunoprecipitation assays with mouse liver. RESULTS: We first confirmed the presence of Vdr mRNA and protein expression in livers of mice. In mice fed normal diets and given injections of 1,25(OH)2D3, liver and plasma concentrations of 1,25(OH)2D3 increased and decreased in unison. Changes in hepatic Cyp7a1 messenger RNA (mRNA) correlated with those of Cyp24a1 (a Vdr target gene) and inversely with Shp mRNA, but not ileal Fgf15 mRNA. Similarly, incubation with 1,25(OH)2D3 increased levels of Cyp24a1/CYP24A1 and Cyp7a1/CYP7A1 mRNA in mouse and human hepatocytes, and reduced levels of Shp mRNA in mouse hepatocytes. In Fxr(-/-) and wild-type mice with hypercholesterolemia, injection of 1,25(OH)2D3 consistently reduced levels of plasma and liver cholesterol and Shp mRNA, and increased hepatic Cyp7a1 mRNA and protein; these changes were not observed in Shp(-/-) mice given 1,25(OH)2D3 and fed Western diets. Truncation of the human small heterodimer partner (SHP) promoter and deletion analyses revealed VDR-dependent inhibition of SHP, and mobility shift assays showed direct binding of VDR to enhancer regions of SHP. In addition, chromatin immunoprecipitation analysis of livers from mice showed that injection of 1,25(OH)2D3 increased recruitment of Vdr and rodent retinoid X receptor to the Shp promoter. CONCLUSIONS: Activation of the VDR represses hepatic SHP to increase levels of mouse and human CYP7A1 and reduce cholesterol.


Assuntos
Calcitriol/farmacologia , Colesterol 7-alfa-Hidroxilase/metabolismo , Colesterol/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Receptores de Calcitriol/agonistas , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Sítios de Ligação , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Hepatócitos/enzimologia , Humanos , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Íleo/efeitos dos fármacos , Íleo/enzimologia , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores de Calcitriol/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Esteroide Hidroxilases/metabolismo , Fatores de Tempo , Transfecção , Vitamina D3 24-Hidroxilase
9.
Pharm Res ; 32(3): 1128-40, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25319098

RESUMO

PURPOSE: Since the vitamin D receptor (VDR) was found to up-regulate cerebral P-glycoprotein expression in vitro and in mice, we extend our findings to rats by assessing the effect of rat Vdr activation on brain efflux of quinidine, a P-gp substrate that is eliminated primarily by cytochrome P450 3a. METHODS: We treated rats with vehicle or the active VDR ligand, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] (4.8 or 6.4 nmol/kg i.p. every 2nd day × 4) and examined P-gp expression and cerebral quinidine disposition via microdialysis in control and treatment studies conducted longitudinally in the same rat. RESULTS: The 6.4 nmol/kg 1,25(OH)2D3 dose increased cerebral P-gp expression 1.75-fold whereas hepatic Cyp3a remained unchanged. Although there was no change in systemic clearance elicited by 1,25(OH)2D3, brain extracellular fluid quinidine concentrations were lower in treated rats. We noted that insertion of indwelling catheters increased plasma protein binding of quinidine and serial sampling decreased the blood:plasma concentration ratio, factors that alter distribution ratios in microdialysis studies. After appropriate correction, KECF/P,uu and KECF/B,uu, or ratios of quinidine unbound concentrations in brain extracellular fluid to plasma or blood at steady-state, were more than halved. CONCLUSION: We demonstrate that VDR activation increases cerebral P-gp expression and delimits brain penetration of P-gp substrates.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Calcitriol/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Microdiálise , Quinidina/metabolismo , Receptores de Calcitriol/agonistas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Estado de Consciência , Citocromo P-450 CYP3A/metabolismo , Relação Dose-Resposta a Droga , Masculino , Microssomos Hepáticos/enzimologia , Ligação Proteica , Quinidina/sangue , Ratos Sprague-Dawley , Receptores de Calcitriol/metabolismo , Regulação para Cima
10.
Clin Cancer Res ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38875108

RESUMO

On November 15, 2023, the U.S. Food and Drug Administration (FDA) granted traditional approval to repotrectinib (Augtyro®, Bristol Myers Squibb Corporation), for the treatment of adult patients with locally advanced or metastatic ROS1-positive non-small cell lung cancer (NSCLC). The approval was based on TRIDENT-1, a single arm trial with multiple cohorts of patients with ROS1 fusion-positive (hereafter "ROS1-positive") NSCLC, (NCT03093116), who were either treatment naïve or had received prior ROS1 TKI and/or platinum-based chemotherapy. The primary efficacy outcome measure is objective response rate (ORR) assessed by blinded independent central review (BICR) using response evaluation criteria in solid tumors (RECIST) version 1.1. ORR was assessed in 71 patients who were ROS1 TKI naïve and 56 patients who had received a prior ROS1 TKI. Among 71 patients who were ROS1 TKI naïve, the ORR was 79% (95% CI 68, 88); median duration of response was 34.1 months (95% CI 26, NE). In patients who had received a prior ROS1 TKI and no prior chemotherapy, the ORR was 38% (95% CI 25, 52). The median duration of response was 14.8 months (95% CI 7.6, NE) BICR-assessed responses were observed in CNS metastases in patients in both cohorts, and in patients who developed resistance mutations following prior TKI therapy. The most common (> 20%) adverse reactions were dizziness, dysgeusia, peripheral neuropathy, constipation, dyspnea, ataxia, fatigue, cognitive disorders, and muscular weakness. A unique feature of this ROS1 TKI approval is the inclusion of robust evidence of efficacy in patients with ROS1-positive NSCLC who had progressed on prior ROS1 TKIs.

11.
J Clin Oncol ; 42(10): 1193-1201, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38381994

RESUMO

PURPOSE: The US Food and Drug Administration (FDA) approved elacestrant for the treatment of postmenopausal women or adult men with estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-), estrogen receptor 1 (ESR1)-mutated advanced or metastatic breast cancer with disease progression after at least one line of endocrine therapy (ET). PATIENTS AND METHODS: Approval was based on EMERALD (Study RAD1901-308), a randomized, open-label, active-controlled, multicenter trial in 478 patients with ER+, HER2- advanced or metastatic breast cancer, including 228 patients with ESR1 mutations. Patients were randomly assigned (1:1) to receive either elacestrant 345 mg orally once daily (n = 239) or investigator's choice of ET (n = 239). RESULTS: In the ESR1-mut subgroup, EMERALD demonstrated a statistically significant improvement in progression-free survival (PFS) by blinded independent central review assessment (n = 228; hazard ratio [HR], 0.55 [95% CI, 0.39 to 0.77]; P value = .0005). Although the overall survival (OS) end point was not met, there was no trend toward a potential OS detriment (HR, 0.90 [95% CI, 0.63 to 1.30]) in the ESR1-mut subgroup. PFS also reached statistical significance in the intention-to-treat population (ITT, N = 478; HR, 0.70 [95% CI, 0.55 to 0.88]; P value = .0018). However, improvement in PFS in the ITT population was primarily attributed to results from patients in the ESR1-mut subgroup. More patients who received elacestrant experienced nausea, vomiting, and dyslipidemia. CONCLUSION: The approval of elacestrant in ER+, HER2- advanced or metastatic breast cancer was restricted to patients with ESR1 mutations. Benefit-risk assessment in the ESR1-mut subgroup was favorable on the basis of a statistically significant improvement in PFS in the context of an acceptable safety profile including no evidence of a potential detriment in OS. By contrast, the benefit-risk assessment in patients without ESR1 mutations was not favorable. Elacestrant is the first oral estrogen receptor antagonist to receive FDA approval for patients with ESR1 mutations.


Assuntos
Neoplasias da Mama , Tetra-Hidronaftalenos , Adulto , Estados Unidos , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/genética , United States Food and Drug Administration , Receptor ErbB-2/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
12.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 30(6): 1204-8, 2013 Dec.
Artigo em Zh | MEDLINE | ID: mdl-24645597

RESUMO

Assessing dry weight accurately is crucial in providing effective and safe haemodialysis. Biases towards dry weight assessment may bring a series of dialysis complications. This study introduces an online detection technique of relative blood volume (RBV) based on ultrasound, which analyzes the correlation between changes in blood density and sound speed. By measuring the attenuation in sound velocity, this method was employed to calculate RBV, and then to evaluate the dry weight of patients on dialysis. TDC-GP2 time measurement chip and MSP430 Single-chip Microcontroller (SCM) were used in the system to measure the ultrasonic travel time. In the clinical trials, RBV values range between 71.3% and 108.1%, showing consistent result with Fresenius 4008S blood volume monitor (BVM). This detection method possesses several advantages, such as real time, convenient, reproducible, non-invasive, and etc.


Assuntos
Volume Sanguíneo , Diálise Renal , Ultrassom , Peso Corporal , Humanos , Monitorização Fisiológica
13.
CPT Pharmacometrics Syst Pharmacol ; 12(5): 585-597, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36530026

RESUMO

This report summarizes the proceedings for day 2 sessions 1 and 3 of the 2-day public workshop entitled "Regulatory Utility of Mechanistic Modeling to Support Alternative Bioequivalence Approaches," a jointly sponsored workshop by the US Food and Drug Administration (FDA) and the Center for Research on Complex Generics (CRCG). The aims of this workshop were: (1) to discuss how mechanistic modeling, including physiologically-based pharmacokinetic (PBPK) modeling and simulation, can support product development, and regulatory submissions; (2) to share the current state of mechanistic modeling for bioequivalence (BE) assessment through case studies; (3) to establish a consensus on best practices for using PBPK modeling for BE assessment to help drive further investment by the generic drug industry into mechanistic modeling and simulation; and (4) to introduce the concept of a Model Master File to improve model-sharing. The theme of day 2 covered PBPK absorption model for oral products as an alternative BE approach and a tool for supporting risk assessment and biowaiver (session 1), oral PBPK for evaluating the impact of food on BE (session 2), successful cases, and challenges for oral PBPK (session 3). This report summarizes the topics of the presentations of day 2 sessions 1 and session 3 from FDA, academia, and pharmaceutical industry, including the current status of oral PBPK, case examples as well as the challenges and opportunities in this area. In addition, panel discussions on the utility of oral PBPK in both new drugs and generic drugs from regulatory and industry perspective are also summarized.


Assuntos
Modelos Biológicos , Relatório de Pesquisa , Humanos , Equivalência Terapêutica , Simulação por Computador
14.
Clin Cancer Res ; 29(20): 4027-4031, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37289037

RESUMO

On September 30, 2022, the FDA granted accelerated approval to futibatinib for the treatment of adult patients with previously treated, unresectable, locally advanced or metastatic intrahepatic cholangiocarcinoma (iCCA) with FGFR2 fusions or other rearrangements. Approval was based on Study TAS-120-101, a multicenter open-label, single-arm trial. Patients received futibatinib 20-mg orally once daily. The major efficacy outcome measures were overall response rate (ORR) and duration of response (DoR) as determined by an independent review committee (IRC) according to RECIST v1.1. ORR was 42% (95% confidence interval, 32%-52%). Median DoR was 9.7 months. Adverse reactions occurring in ≥30% patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, and abdominal pain. The most common laboratory abnormalities (≥50%) were increased phosphate, increased creatinine, decreased hemoglobin, and increased glucose. Ocular toxicity (including dry eye, keratitis, and retinal epithelial detachment) and hyperphosphatemia are important risks of futibatinib, which are listed under Warnings and Precautions. This article summarizes the FDA's thought process and data supporting the approval of futibatinib.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Pirazóis , Pirróis , Adulto , Humanos , Pirimidinas/efeitos adversos , Colangiocarcinoma/tratamento farmacológico , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos , Aprovação de Drogas , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
15.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 37(1): 11-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22349387

RESUMO

OBJECTIVE: Hepatic stellate cells (HSCs) are the principal cells responsible for the development of hepatic fibrosis and cirrhosis. During the fibrotic process, HSCs undergo proliferation and transdifferentiation from a quiescent to myofibroblast-like phenotype. The fate of myofibroblast like HSCs includes apoptosis or reversion back to a quiescent phenotype. The mechanisms involved in the apoptotic process of HSCs have yet to be determined. The purpose of the present study is to determine the effects of extracellular signal-regulated kinases (ERKs) phosphorylation on the apoptosis of HSCs induced by staurosporine. METHODS: We used Western blot and flow cytometry to detect the expression level of ERK and cell apoptosis status in four rat hepatic stellate cell lines (CFSC-8B, -2G, -3H and-5H). RESULTS: Each hepatic stellate cell line had a distinct morphology consistent with their expression level of α-SMA and that CFSC-8B cells had the highest α-SMA expression. Although all four cell types expressed similar levels of ERK1/2, phosphorylation levels were significantly higher in CFSC- 8B and CFSC-2G than in CFSC-3H and CFSC-5H cells. When CFSC-8B cells (high ERK1/2 phosphorylation) and CFSC-5H cells (low ERK1/2 phosphorylation) were employed to examine staurosporine-induced apoptosis, CFSC-8B cells were significantly more sensitive. Staurosporine further increased ERK1/2 phosphorylation in both cell lines. CONCLUSION: ERK1/2 phosphorylation in HSCs determines the sensitivity of HSCs to staurosporine-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/efeitos dos fármacos , Estaurosporina/farmacologia , Animais , Linhagem Celular , Citometria de Fluxo , Células Estreladas do Fígado/enzimologia , Fosforilação , Ratos
16.
Clin Pharmacol Ther ; 111(3): 624-634, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34656075

RESUMO

Remdesivir (RDV) is the first drug approved by the US Food and Drug Administration (FDA) for the treatment of coronavirus disease 2019 (COVID-19) in certain patients requiring hospitalization. As a nucleoside analogue prodrug, RDV undergoes intracellular multistep activation to form its pharmacologically active species, GS-443902, which is not detectable in the plasma. A question arises that whether the observed plasma exposure of RDV and its metabolites would correlate with or be informative about the exposure of GS-443902 in tissues. A whole body physiologically-based pharmacokinetic (PBPK) modeling and simulation approach was utilized to elucidate the disposition mechanism of RDV and its metabolites in the lungs and liver and explore the relationship between plasma and tissue pharmacokinetics (PK) of RDV and its metabolites in healthy subjects. In addition, the potential alteration of plasma and tissue PK of RDV and its metabolites in patients with organ dysfunction was explored. Our simulation results indicated that intracellular exposure of GS-443902 was decreased in the liver and increased in the lungs in subjects with hepatic impairment relative to the subjects with normal liver function. In subjects with severe renal impairment, the exposure of GS-443902 in the liver was slightly increased, whereas the lung exposure of GS-443902 was not impacted. These predictions along with the organ impairment study results may be used to support decision making regarding the RDV dosage adjustment in these patient subgroups. The modeling exercise illustrated the potential of whole body PBPK modeling to aid in decision making for nucleotide analogue prodrugs, particularly when the active metabolite exposure in the target tissues is not available.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Modelos Biológicos , Insuficiência de Múltiplos Órgãos/metabolismo , Monofosfato de Adenosina/sangue , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/farmacocinética , Monofosfato de Adenosina/urina , Adulto , Alanina/sangue , Alanina/metabolismo , Alanina/farmacocinética , Alanina/urina , Humanos , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Distribuição Tecidual
17.
Clin Pharmacol Ther ; 111(3): 572-578, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34807992

RESUMO

Leveraging limited clinical and nonclinical data through modeling approaches facilitates new drug development and regulatory decision making amid the coronavirus disease 2019 (COVID-19) pandemic. Model-informed drug development (MIDD) is an essential tool to integrate those data and generate evidence to (i) provide support for effectiveness in repurposed or new compounds to combat COVID-19 and dose selection when clinical data are lacking; (ii) assess efficacy under practical situations such as dose reduction to overcome supply issues or emergence of resistant variant strains; (iii) demonstrate applicability of MIDD for full extrapolation to adolescents and sometimes to young pediatric patients; and (iv) evaluate the appropriateness for prolonging a dosing interval to reduce the frequency of hospital visits during the pandemic. Ongoing research activities of MIDD reflect our continuous effort and commitment in bridging knowledge gaps that leads to the availability of effective treatments through innovation. Case examples are presented to illustrate how MIDD has been used in various stages of drug development and has the potential to inform regulatory decision making.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , COVID-19 , Desenvolvimento de Medicamentos/métodos , Modelos Biológicos , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/farmacologia , COVID-19/epidemiologia , Aprovação de Drogas , Reposicionamento de Medicamentos , Humanos , Farmacologia Clínica/métodos , SARS-CoV-2/imunologia
18.
J Asian Nat Prod Res ; 13(3): 205-14, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21409681

RESUMO

The pharmacokinetics, tissue distribution, and excretion of buagafuran (BF, 4-butyl-α-agarofuran), a promising antianxiety drug isolated from Gharu-wood (Aquilaria agallocha Roxb), were investigated in rats. BF plasma concentration was determined in rats after oral and intravenous doses by GC-TOF-MS. BF showed nonlinear pharmacokinetics after oral and intravenous administration of 4, 16, and 64 mg/kg. The AUC(0-∞) and C(max) did not increase proportionally with doses, indicating the saturation in absorption kinetics of BF in rats after oral dosage. BF absorption was extremely poor with an absolute bioavailability below 9.5%. After oral administration of (3)H-BF (4 mg/kg) to rats, radioactivity was well distributed to the tissues examined. The highest radioactivity was found in gastrointestinal tract, followed by liver and kidney. Radioactivity in brain, as a target organ, was about 20-40% of that in plasma at all time points. Total mean percent recovery of radioactive dose was about 80% in rats (51.2% in urine; 28.7% in feces). Bile elimination was also the major excretion route of BF, and 45.4% of the radioactive dose was recovered in bile.


Assuntos
Ansiolíticos/farmacocinética , Sesquiterpenos/farmacocinética , Administração Oral , Animais , Ansiolíticos/sangue , Ansiolíticos/química , Ansiolíticos/metabolismo , Ansiolíticos/urina , Fezes/química , Feminino , Masculino , Estrutura Molecular , Ratos , Ratos Wistar , Sesquiterpenos/sangue , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Sesquiterpenos/urina , Distribuição Tecidual
19.
Methods Mol Biol ; 2342: 595-629, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34272707

RESUMO

This chapter provides regulatory perspectives on how to translate in vitro drug metabolism findings into in vivo drug-drug interaction (DDI) predictions and how this affects the decision of conducting in vivo DDI evaluation. The chapter delineates rationale and analyses that have supported the recommendations in the U.S. Food and Drug Administration (FDA) DDI guidances in terms of in vitro-in vivo extrapolation of cytochrome P450 (CYP) inhibition-mediated DDI potential for investigational new drugs and their metabolites as substrates or inhibitors. The chapter also describes the framework and considerations to assess UDP-glucuronosyltransferase (UGT) inhibition-mediated DDI potential for drugs as substrates or inhibitors. The limitations of decision criteria and further improvements needed are also discussed. Case examples are provided throughout the chapter to illustrate how decision criteria have been utilized to evaluate in vivo DDI potential from in vitro data.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/metabolismo , Legislação de Medicamentos/organização & administração , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/química , Interações Medicamentosas , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/química , Humanos , Cinética , Guias de Prática Clínica como Assunto , Estados Unidos , United States Food and Drug Administration/legislação & jurisprudência
20.
J Clin Pharmacol ; 61(6): 782-788, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33460193

RESUMO

The key parameters necessary to predict drug-drug interactions (DDIs) are intrinsic clearance (CLint ) and fractional contribution of the metabolizing enzyme toward total metabolism (fm ). Herein, we summarize the accumulated knowledge from 53 approved new drug applications submitted to the Office of Clinical Pharmacology, US Food and Drug Administration, from 2016 to 2018 that contained physiologically based pharmacokinetic (PBPK) models to understand how in vitro data are used in PBPK models to assess drug metabolism and predict DDIs. For evaluation of CLint and fm , 29 and 20 new drug applications were included for evaluation, respectively. For CLint , 86.2% of the PBPK models used modified values based on in vivo data with modifications ranging from -82.5% to 2752.5%. For fm , 45.0% of the models used modified values with modifications ranging from -28% to 178.6%. When values for CLint were used from in vitro testing without modification, the model resulted in up to a 14.3-fold overprediction of the area under the concentration-time curve of the substrate. When values for fm from in vitro testing were used directly, the model resulted in up to a 2.9-fold underprediction of its DDI magnitude with an inducer, and up to a 1.7-fold overprediction of its DDI magnitude with an inhibitor. Our analyses suggested that the in vitro system usually provides a reasonable estimation of fm when the drug metabolism by a given CYP pathway is more than 70% of the total clearance. In vitro experiments provide important information about basic PK properties of new drugs and can serve as a starting point for building a PBPK model. However, key PBPK parameters such as CLint and fm still need to be optimized based on in vivo data.


Assuntos
Interações Medicamentosas/fisiologia , Técnicas In Vitro/estatística & dados numéricos , Modelos Biológicos , United States Food and Drug Administration/estatística & dados numéricos , Área Sob a Curva , Simulação por Computador , Aprovação de Drogas/estatística & dados numéricos , Humanos , Técnicas In Vitro/normas , Taxa de Depuração Metabólica , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA