Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Hepatol ; 80(2): 322-334, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37972659

RESUMO

BACKGROUND & AIMS: There is a knowledge gap in understanding mechanisms of resistance to fibroblast growth factor receptor (FGFR) inhibitors (FGFRi) and a need for novel therapeutic strategies to overcome it. We investigated mechanisms of acquired resistance to FGFRi in patients with FGFR2-fusion-positive cholangiocarcinoma (CCA). METHODS: A retrospective analysis of patients who received FGFRi therapy and underwent tumor and/or cell-free DNA analysis, before and after treatment, was performed. Longitudinal circulating tumor DNA samples from a cohort of patients in the phase I trial of futibatinib (NCT02052778) were assessed. FGFR2-BICC1 fusion cell lines were developed and secondary acquired resistance mutations in the mitogen-activated protein kinase (MAPK) pathway were introduced to assess their effect on sensitivity to FGFRi in vitro. RESULTS: On retrospective analysis of 17 patients with repeat sequencing following FGFRi treatment, new FGFR2 mutations were detected in 11 (64.7%) and new alterations in MAPK pathway genes in nine (52.9%) patients, with seven (41.2%) patients developing new alterations in both the FGFR2 and MAPK pathways. In serially collected plasma samples, a patient treated with an irreversible FGFRi tested positive for previously undetected BRAF V600E, NRAS Q61K, NRAS G12C, NRAS G13D and KRAS G12K mutations upon progression. Introduction of a FGFR2-BICC1 fusion into biliary tract cells in vitro sensitized the cells to FGFRi, while concomitant KRAS G12D or BRAF V600E conferred resistance. MEK inhibition was synergistic with FGFRi in vitro. In an in vivo animal model, the combination had antitumor activity in FGFR2 fusions but was not able to overcome KRAS-mediated FGFRi resistance. CONCLUSIONS: These findings suggest convergent genomic evolution in the MAPK pathway may be a potential mechanism of acquired resistance to FGFRi. CLINICAL TRIAL NUMBER: NCT02052778. IMPACT AND IMPLICATIONS: We evaluated tumors and plasma from patients who previously received inhibitors of fibroblast growth factor receptor (FGFR), an important receptor that plays a role in cancer cell growth, especially in tumors with abnormalities in this gene, such as FGFR fusions, where the FGFR gene is fused to another gene, leading to activation of cancer cell growth. We found that patients treated with FGFR inhibitors may develop mutations in other genes such as KRAS, and this can confer resistance to FGFR inhibitors. These findings have several implications for patients with FGFR2 fusion-positive tumors and provide mechanistic insight into emerging MAPK pathway alterations which may serve as a therapeutic vulnerability in the setting of acquired resistance to FGFRi.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Animais , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/uso terapêutico , Estudos Retrospectivos , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Mutação , Ductos Biliares Intra-Hepáticos/patologia , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Inibidores de Proteínas Quinases/efeitos adversos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
2.
N Engl J Med ; 388(3): 228-239, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36652354

RESUMO

BACKGROUND: Alterations in fibroblast growth factor receptor 2 (FGFR2) have emerged as promising drug targets for intrahepatic cholangiocarcinoma, a rare cancer with a poor prognosis. Futibatinib, a next-generation, covalently binding FGFR1-4 inhibitor, has been shown to have both antitumor activity in patients with FGFR-altered tumors and strong preclinical activity against acquired resistance mutations associated with ATP-competitive FGFR inhibitors. METHODS: In this multinational, open-label, single-group, phase 2 study, we enrolled patients with unresectable or metastatic FGFR2 fusion-positive or FGFR2 rearrangement-positive intrahepatic cholangiocarcinoma and disease progression after one or more previous lines of systemic therapy (excluding FGFR inhibitors). The patients received oral futibatinib at a dose of 20 mg once daily in a continuous regimen. The primary end point was objective response (partial or complete response), as assessed by independent central review. Secondary end points included the response duration, progression-free and overall survival, safety, and patient-reported outcomes. RESULTS: Between April 16, 2018, and November 29, 2019, a total of 103 patients were enrolled and received futibatinib. A total of 43 of 103 patients (42%; 95% confidence interval, 32 to 52) had a response, and the median duration of response was 9.7 months. Responses were consistent across patient subgroups, including patients with heavily pretreated disease, older adults, and patients who had co-occurring TP53 mutations. At a median follow-up of 17.1 months, the median progression-free survival was 9.0 months and overall survival was 21.7 months. Common treatment-related grade 3 adverse events were hyperphosphatemia (in 30% of the patients), an increased aspartate aminotransferase level (in 7%), stomatitis (in 6%), and fatigue (in 6%). Treatment-related adverse events led to permanent discontinuation of futibatinib in 2% of the patients. No treatment-related deaths occurred. Quality of life was maintained throughout treatment. CONCLUSIONS: In previously treated patients with FGFR2 fusion or rearrangement-positive intrahepatic cholangiocarcinoma, the use of futibatinib, a covalent FGFR inhibitor, led to measurable clinical benefit. (Funded by Taiho Oncology and Taiho Pharmaceutical; FOENIX-CCA2 ClinicalTrials.gov number, NCT02052778.).


Assuntos
Antineoplásicos , Neoplasias dos Ductos Biliares , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma , Inibidores de Proteínas Quinases , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Idoso , Humanos , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Qualidade de Vida , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Antineoplásicos/administração & dosagem
3.
Future Oncol ; 18(30): 3377-3387, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36039910

RESUMO

PTEN acts as a potent tumor suppressor within the PI3K/AKT/mTOR pathway. Germline mutations in the PTEN gene are a hallmark of PTEN hamartoma tumor syndrome, which includes Cowden syndrome, where they appear to elevate lifetime risk of cancer. Targeted AKT directed therapy has been proposed as an effective approach in cancer patients having germline PTEN mutations. The mechanism of action, safety and dosing regimen for the novel allosteric AKT inhibitor TAS-117 have been explored in a phase I study in Japan in which activity was observed against certain tumor types. Here we describe the study protocol of an international, two-part phase II study evaluating the safety, tolerability, pharmacokinetics, pharmacodynamics and antitumor activity of TAS-117 in patients with advanced solid tumors harboring germline PTEN-inactivating mutations.


Signaling paths control growth and activities inside cells. Overactivity in these paths can encourage many types of cancers to develop. Tumor suppressor proteins can inhibit cell signals that promote cancer. This protection can be lost if there are errors in any gene coding for a tumor suppressor protein. We are carrying out a clinical study to test TAS-117, a potential new oral medicine, in people who have solid tumors and whose cells have changes in their genes that inactivate a tumor suppressor protein called PTEN. TAS-117 targets part of a signaling path that may be overactive due to loss of PTEN activity. In early research, TAS-117 has shown promising activity against certain tumor types. Our trial will explore if TAS-117 can provide a new treatment for rare forms of cancer where genetic changes have led to a loss of PTEN activity. Clinical Trial Registration: NCT04770246 (ClinicalTrials.gov).


Assuntos
Síndrome do Hamartoma Múltiplo , Neoplasias , Humanos , Ensaios Clínicos Fase II como Assunto , Células Germinativas/metabolismo , Mutação em Linhagem Germinativa , Síndrome do Hamartoma Múltiplo/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , PTEN Fosfo-Hidrolase/genética
4.
NPJ Breast Cancer ; 7(1): 57, 2021 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-34016993

RESUMO

The METRIC study (NCT#0199733) explored a novel antibody-drug conjugate, glembatumumab vedotin (GV), targeting gpNMB that is overexpressed in ~40% of patients with triple-negative breast cancer (TNBC) and associated with poor prognosis. The study was a randomized, open-label, phase 2b study that evaluated progression-free survival (PFS) of GV compared with capecitabine in gpNMB-overexpressing TNBC. Patients who had previously received anthracycline and taxane-based therapy were randomized 2:1 to receive, GV (1.88 mg/kg IV q21 days) or capecitabine (2500 mg/m2 PO daily d1-14 q21 days). The primary endpoint was RECIST 1.1 PFS per independent, blinded central review. In all, 327 patients were randomized to GV (213 treated) or capecitabine (92 treated). Median PFS was 2.9 months for GV vs. 2.8 months for capecitabine. The most common grade ≥3 toxicities for GV were neutropenia, rash, and leukopenia, and for capecitabine were fatigue, diarrhea, and palmar-plantar erythrodysesthesia. The study did not meet the primary endpoint of improved PFS over capecitabine or demonstrate a relative risk/benefit improvement over capecitabine.

5.
Curr Drug Discov Technol ; 17(1): 2-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30251606

RESUMO

Cell-based assays are an important part of the drug discovery process and clinical research. One of the main hurdles is to design sufficiently robust assays with adequate signal to noise parameters while maintaining the inherent physiology of the cells and not interfering with the pharmacology of target being investigated. A plethora of assays that assess cell viability (or cell heath in general) are commercially available and can be classified under different categories according to their concepts and principle of reactions. The assays are valuable tools, however, suffer from a large number of limitations. Some of these limitations can be procedural or operational, but others can be critical as those related to a poor concept or the lack of proof of concept of an assay, e.g. those relying on differential permeability of dyes in-and-out of viable versus compromised cell membranes. While the assays can differentiate between dead and live cells, most, if not all, of them can just assess the relative performance of cells rather than providing a clear distinction between healthy and dying cells. The possible impact of relatively high molecular weight dyes, used in most of the assay, on cell viability has not been addressed. More innovative assays are needed, and until better alternatives are developed, setup of current cell-based studies and data interpretation should be made with the limitations in mind. Negative and positive control should be considered whenever feasible. Also, researchers should use more than one orthogonal method for better assessment of cell health.


Assuntos
Bioensaio/métodos , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Bioensaio/economia , Bioensaio/instrumentação , Descoberta de Drogas/economia , Descoberta de Drogas/instrumentação , Avaliação Pré-Clínica de Medicamentos/economia , Avaliação Pré-Clínica de Medicamentos/instrumentação , Ensaios de Triagem em Larga Escala/economia , Ensaios de Triagem em Larga Escala/instrumentação , Humanos
6.
Ann N Y Acad Sci ; 1346(1): 63-70, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25758153

RESUMO

An incredibly high failure rate in the pharmaceutical industry has positioned personalized medicine with its prerequisite drug-diagnostic codevelopment, commonly known as companion diagnostics (CDx), in the frontline as an potential rescuer. This hopefulness is potentiated by the recent major advances and competitiveness in molecular diagnostics, making laboratory tests widely accessible at affordable prices. If executed correctly, biomarkers and CDx can potentially help the drug industry by enhancing the probability of success and possibly accelerating time to market; help the diagnostics industry develop tests utilizing precious, clinically annotated human samples; and, more importantly, benefit patients by supporting accurate diagnosis and selection of the most efficacious and least toxic therapies. However, this spectacular road is not yet paved, and it faces an enormous number of challenges. This paper will list these challenges and highlight some critical problems with representative examples of imminent but still overlooked preanalytical and analytical variables that can defeat the whole purpose of biomarkers and CDx and mislead drug developers and clinicians. The paper will provide some suggestions for mitigation.


Assuntos
Biomarcadores/análise , Técnicas de Laboratório Clínico/métodos , Medicina de Precisão , Manejo de Espécimes , Técnicas de Laboratório Clínico/normas , Descoberta de Drogas/economia , Descoberta de Drogas/organização & administração , Indústria Farmacêutica/economia , Indústria Farmacêutica/organização & administração , Eficiência Organizacional , Humanos , Técnicas de Diagnóstico Molecular/normas , Medicina de Precisão/métodos , Medicina de Precisão/normas , Preservação Biológica/normas , Controle de Qualidade , Reprodutibilidade dos Testes , Manejo de Espécimes/métodos , Manejo de Espécimes/normas
7.
Thromb Res ; 134(4): 909-13, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25179520

RESUMO

INTRODUCTION: Edoxaban is an oral, once-daily direct factor Xa (FXa) inhibitor. Although rapidly cleared, strategies to reverse edoxaban-mediated effects on anticoagulation are needed in cases of excessive bleeding or emergency. This study evaluated the effect of two prohemostatic agents, recombinant factor VIIa (rFVIIa) and factor VIII inhibitor bypass activity (FEIBA), on the anticoagulatory effects of supratherapeutic concentrations of edoxaban in human whole blood ex vivo. MATERIALS AND METHODS: Blood samples were collected from six healthy volunteers. Edoxaban (500 or 1000 ng/mL), alone or followed by rFVIIa (0.8 or 1.8µg/mL) or FEIBA (0.75 or 1.5 U/mL), was added to an aliquot of each sample. Biomarkers, including prothrombin time (PT), activated partial thromboplastin time (aPTT), extrinsic FXa activity (anti-FXa), intrinsic factor X activity, and D-dimer, were assessed at 0.25, 0.5, 1, 2, and 4 hours after adding rFVIIa or FEIBA. RESULTS: Decreases in measures of PT (p<0.0001), aPTT (p<0.0001), and anti-FXa (p<0.0001) were observed when rFVIIa or FEIBA was added to edoxaban-containing blood samples. Intrinsic FX activity was increased up to 20% and 31% of normal in the presence of edoxaban by rFVIIa and FEIBA, respectively. The impact of these agents on the anticoagulant effects of edoxaban were observed within 15 minutes and remained relatively unchanged at each timepoint thereafter. CONCLUSIONS: The findings of this ex vivo study suggest that rFVIIa and FEIBA rapidly reversed edoxaban-mediated anticoagulation effects based on PT and aPTT, but had minimal effect based on intrinsic FX activity. No dose response was observed for rFVIIa or FEIBA.


Assuntos
Anticoagulantes/farmacologia , Fatores de Coagulação Sanguínea/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Hemostáticos/farmacologia , Piridinas/farmacologia , Tiazóis/farmacologia , Humanos , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Proteínas Recombinantes/farmacologia
8.
J Clin Pharmacol ; 54(8): 910-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24706516

RESUMO

Edoxaban is an oral, once-daily, direct factor Xa (FXa) inhibitor that has been evaluated for the prevention of stroke in patients with atrial fibrillation (AF) and for the treatment and prevention of venous thromboembolism (VTE) recurrence. Pharmacokinetic (PK) and pharmacodynamic (PD) modeling and logistic regression analyses were used to explore the clinical data from phase 1 and 2 studies to determine the relationship among PK exposure, PD response, and bleeding risk. Population PK/PD modeling was performed using plasma edoxaban concentration data from combined phase 1 and 2 studies and using intrinsic FXa data from a phase 2 AF study. A 2-compartment PK model adequately described the combined PK data, and a dynamic binding model characterized the dynamics of the intrinsic factor X activity (iFXa) data. Logistic regression analysis then identified the relationship between the iFXa and the observed bleeding risk. The more protracted suppression of FXa over the dosing interval for a 30-mg twice-daily dose compared with a 60-mg once-daily dose offers an explanation for the significantly higher bleeding rate at the former dose.


Assuntos
Inibidores do Fator Xa/farmacologia , Modelos Biológicos , Piridinas/farmacologia , Tiazóis/farmacologia , Adulto , Idoso , Fator Xa/metabolismo , Inibidores do Fator Xa/sangue , Inibidores do Fator Xa/farmacocinética , Feminino , Hemorragia/tratamento farmacológico , Hemorragia/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Piridinas/sangue , Piridinas/farmacocinética , Tiazóis/sangue , Tiazóis/farmacocinética
9.
Expert Rev Mol Diagn ; 14(1): 27-35, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24308333

RESUMO

Low productivity in the pharmaceutical industry positions personalized medicine and companion diagnostics as an attractive approach. The oncology community is enthusiastic and optimistic that advances in molecular diagnostics have the potential to materialize the dream of personalized cancer therapy with the aim to: help the drug industry enhance the probability of success and, probably, accelerate time to market; help the diagnostics industry develop diagnostic tests utilizing precious human samples; and support accurate diagnosis and selection of the most efficacious and least toxic therapies. However, this spectacular road is not yet paved and remains facing the following biggest challenges: the need for better understanding of biology and limitations of preclinical models; pre-analytical variables that are likely to alter results; analytical discrepancies; and the wide gap between clinical/pharmaceutical and diagnostic understandings. This article explores these challenges, their possible impacts, and provides some suggestions for mitigation.


Assuntos
Neoplasias/diagnóstico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Descoberta de Drogas , Humanos , Técnicas de Diagnóstico Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Medicina de Precisão , Melhoria de Qualidade
10.
Clin Cancer Res ; 19(11): 3078-87, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23591447

RESUMO

PURPOSE: HER3 is a key dimerization partner for other HER family members, and its expression is associated with poor prognosis. This first-in-human study of U3-1287 (NCT00730470), a fully human anti-HER3 monoclonal antibody, evaluated its safety, tolerability, and pharmacokinetics in patients with advanced solid tumor. EXPERIMENTAL DESIGN: The study was conducted in 2 parts: part 1--sequential cohorts received escalating doses (0.3-20 mg/kg) of U3-1287 every 2 weeks, starting 3 weeks after the first dose; part 2--additional patients received 9, 14, or 20 mg/kg U3-1287 every 2 weeks, based on observed tolerability and pharmacokinetics from part 1. Recommended phase II dose, adverse event rates, pharmacokinetics, and tumor response were determined. RESULTS: Fifty-seven patients (part 1: 26; part 2: 31) received U3-1287. As no dose-limiting toxicities were reported, the maximum-tolerated dose was not reached. The maximum-administered dose was 20 mg/kg every 2 weeks. The most frequent adverse events related to U3-1287 were fatigue (21.1%), diarrhea (12.3%), nausea (10.5%), decreased appetite (7.0%), and dysgeusia (5.3%). No patient developed anti-U3-1287 antibodies. In these heavily pretreated patients, stable disease was maintained 9 weeks or more in 19.2% in part 1 and 10 weeks or more in 25.8% in part 2. CONCLUSION: U3-1287 treatment was well tolerated, and some evidence of disease stabilization was observed. Pharmacokinetic data support U3-1287 dosing of 9 to 20 mg/kg every 2 to 3 weeks. Combination studies of U3-1287 are ongoing.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Biomarcadores/metabolismo , Anticorpos Amplamente Neutralizantes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/metabolismo , Prognóstico , Receptor ErbB-3/antagonistas & inibidores , Resultado do Tratamento
11.
Invest New Drugs ; 31(4): 1016-22, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23417696

RESUMO

BACKGROUND: Tivantinib is a selective, small-molecule inhibitor of the MET receptor tyrosine kinase. Preclinical and phase 1 data suggested a possible role for MET in the pathophysiology of germ cell tumors (GCTs) and a potential clinical benefit from tivantinib in patients with these tumors. METHODS: Men (≥ 16 years) with relapsed or refractory, histologically confirmed, non-central nervous system GCTs received oral tivantinib 360 mg twice daily in 28-day cycles until progressive disease or unacceptable toxicity. The primary endpoint was objective response rate in the first 4 cycles, with study termination for <2 responses among the first 21 patients. Secondary endpoints included 12-week progression-free survival (PFS), overall survival (OS), and safety. RESULTS: Twenty-seven patients were enrolled in 9 months (median age, 32 years). Most patients had tumors with nonseminoma histology (n = 25), and primary tumor sites were testis (n = 24) and mediastinum (n = 3). Among 25 evaluable patients, no objective responses were observed; accrual was halted when the 21st patient became evaluable. Best response was stable disease (n = 5). Median PFS was 1 month, the 12-week PFS rate was 21 %, and median OS was 6 months. Grade 3 or 4 adverse events considered related to study drug included grade 3 pneumonia and grade 3 syncope (n = 1, each). CONCLUSIONS: Tivantinib was well tolerated but did not demonstrate single-agent activity in patients with relapsed/refractory GCTs. Rapid accrual to this phase 2 trial was achieved in this rare patient population through multicenter collaboration.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Pirrolidinonas/uso terapêutico , Quinolinas/uso terapêutico , Adolescente , Adulto , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Demografia , Intervalo Livre de Doença , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias Embrionárias de Células Germinativas/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirrolidinonas/efeitos adversos , Pirrolidinonas/farmacocinética , Pirrolidinonas/farmacologia , Quinolinas/efeitos adversos , Quinolinas/farmacocinética , Quinolinas/farmacologia , Recidiva , Resultado do Tratamento , Adulto Jovem
12.
Thromb Haemost ; 108(1): 166-75, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22628060

RESUMO

Edoxaban is an oral direct factor (F)Xa inhibitor in advanced stages of clinical development. The primary objective of the present study was to assess the pharmacodynamics (PD) and safety of enoxaparin 1 mg/kg followed 12 hours (h) post-dose by edoxaban 60 mg, which is the regimen being used in the phase III study of edoxaban for the treatment of venous thromboembolism (Hokusai-VTE). This was a phase I, open-label, randomised, four-period, four-treatment cross-over study. Treatments were edoxaban alone (EDOX), enoxaparin alone (ENOX), edoxaban plus enoxaparin (EDOX+ENOX), and enoxaparin followed by edoxaban 12 h later (ENOX12-EDOX). Serial blood samples were collected for PD (thrombin generation, anti-FXa) and pharmacokinetic (PK) variables (edoxaban and its principal metabolite M4 by LC-MS/MS, and anti-FIIa as a surrogate of enoxaparin). The highest effect on thrombin AUC (endogenous thrombin potential, or ETP), thrombin (peak), thrombin generation lag time, and velocity index was observed for EDOX+ENOX, followed by ENOX, ENOX12-EDOX, and EDOX. The greatest effect on anti-FXa activity was observed for EDOX+ENOX, followed by ENOX12-EDOX. As expected, neither edoxaban nor enoxaparin significantly altered the PK of the other drug. There were no serious adverse events during the study. It is concluded that a 60-mg dose of edoxaban can be safely administered 12 h following enoxaparin 1 mg/kg.


Assuntos
Enoxaparina/administração & dosagem , Piridinas/administração & dosagem , Tiazóis/administração & dosagem , Tromboembolia Venosa/tratamento farmacológico , Adulto , Testes de Coagulação Sanguínea , Cromatografia Líquida , Interações Medicamentosas , Quimioterapia Combinada , Enoxaparina/efeitos adversos , Enoxaparina/farmacocinética , Inibidores do Fator Xa , Feminino , Humanos , Masculino , Espectrometria de Massas , Piridinas/efeitos adversos , Piridinas/farmacocinética , Tiazóis/efeitos adversos , Tiazóis/farmacocinética , Trombina/biossíntese , Tromboembolia Venosa/sangue
13.
Cancer ; 118(21): 5403-13, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22570147

RESUMO

BACKGROUND: Efatutazone (CS-7017), a novel peroxisome proliferator-activated receptor gamma (PPARγ) agonist, exerts anticancer activity in preclinical models. The authors conducted a phase 1 study to determine the recommended phase 2 dose, safety, tolerability, and pharmacokinetics of efatutazone. METHODS: Patients with advanced solid malignancies and no curative therapeutic options were enrolled to receive a given dose of efatutazone, administered orally (PO) twice daily for 6 weeks, in a 3 + 3 intercohort dose-escalation trial. After the third patient, patients with diabetes mellitus were excluded. Efatutazone dosing continued until disease progression or unacceptable toxicity, with measurement of efatutazone pharmacokinetics and plasma adiponectin levels. RESULTS: Thirty-one patients received efatutazone at doses ranging from 0.10 to 1.15 mg PO twice daily. Dose escalation stopped when maximal impact on PPARγ-related biomarkers had been reached before any protocol-defined maximum-tolerated dose level. On the basis of a population pharmacokinetic/pharmacodynamic analysis, the recommended phase 2 dose was 0.5 mg PO twice daily. A majority of patients experienced peripheral edema (53.3%), often requiring diuretics. Three episodes of dose-limiting toxicities, related to fluid retention, were noted in the 0.10-, 0.25-, and 1.15-mg cohorts. Of 31 treated patients, 27 were evaluable for response. A sustained partial response (PR; 690 days on therapy) was observed in a patient with myxoid liposarcoma. Ten additional patients had stable disease (SD) for ≥60 days. Exposures were approximately dose proportional, and adiponectin levels increased after 4 weeks of treatment at all dose levels. Immunohistochemistry of archived specimens demonstrated that PPARγ and retinoid X receptor expression levels were significantly greater in patients with SD for ≥60 days or PR (P = .0079), suggesting a predictive biomarker. CONCLUSIONS: Efatutazone demonstrates acceptable tolerability with evidence of disease control in patients with advanced malignancies.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , PPAR gama/agonistas , Tiazolidinedionas/administração & dosagem , Administração Oral , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/patologia , Tiazolidinedionas/efeitos adversos , Tiazolidinedionas/farmacocinética
14.
Biomark Med ; 5(2): 211-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21473726

RESUMO

Despite improvements achieved in laboratory medicine over the years, discrepant results from different clinical laboratories are still a source of imminent risk to patients and the pharmaceutical industry. The problem is aggravated by a disconnect between the in vitro diagnostic community and clinicians or drug developers. This is the sequel to a previous report where data from proficiency testing, originally established as a tool for good laboratory practice, were used to highlight the size of the problem, and its possible impact on patients and pharmaceutical trials. Results from three laboratory tests commonly used as tools to monitor patients undergoing therapy with heparin or antithrombin agents, activated partial thromboplastin time, anti-factor Xa and thrombin time, were investigated. Unfortunately, data show that, owing to an absence of standardization approaches or a reasonable technique to harmonize results from different laboratories, results for the same sample, if analyzed by different laboratories or even by one laboratory employing different platforms or reagents, can be extremely different and misleading.


Assuntos
Antitrombinas/uso terapêutico , Técnicas de Laboratório Clínico/normas , Heparina/uso terapêutico , Fator Xa/metabolismo , Humanos , Tempo de Tromboplastina Parcial , Grupo Associado , Tempo de Trombina
15.
Biomark Med ; 3(3): 231-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20477475

RESUMO

AIMS: Despite improvements achieved in laboratory medicine over the years, discrepant results from different clinical laboratories are still a source of imminent risk to patients and the pharmaceutical industry. The problem is aggravated by a disconnection between laboratorians and clinicians or drug developers. MATERIALS & METHODS: In this report, results from proficiency testing, originally established as a tool for good laboratory practice, were used to highlight the size of the problem and its impact on patients and pharmaceutical trials. Results from three laboratory tests, believed to be standardized and commonly requested as tools in the management of patients, and decision-making biomarkers in drug development were used: prothrombin time expressed as international normalization ratio, digoxin and low-density lipoprotein-cholesterol. RESULTS: Unfortunately, data demonstrate that results for the same sample or batch of samples, if analyzed by different laboratories or even by one laboratory that employs different platforms or reagents, can be extremely different. CONCLUSION: In the absence of an effective laboratory-to-laboratory, method-to-method or platform-to-platform standardization, or at lowest harmonization of test results, laboratory results could be misleading to clinicians and the drug industry. Drug developers need to understand the current challenge before any attempt to use biomarker data to make decisions, compile data for a biomarker from different studies or use biomarkers to bridge between different drug candidates belonging to a particular class of compounds.

16.
Transplantation ; 77(8): 1147-54, 2004 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15114076

RESUMO

BACKGROUND: Culturing human islets in Memphis serum-free media (M-SFM) is associated with excellent postculture recovery, in vitro function, and in vivo survival. The authors investigate the possibility of preserving islet function for extended periods (6 months) in culture and describe the in vitro and in vivo functional outcomes associated with these extended culture times. METHODS: Human islets isolated from three cadaveric donor organs were cultured in M-SFM for 1, 3, or 6 months before transplantation under the kidney capsule of nonobese diabetic (NOD)-severe combined immunodeficiency (SCID) mice. In vitro function was measured by static incubation at the time of transplantation. In vivo function was assessed by measuring human insulin and C-peptide production, and by the ability of 6-month cultured islets to cure streptozotocin-induced diabetes in this mouse model. RESULTS: Islet recovery ratios after 1 month in culture ranged from 85% to 88% and declined to 28% to 53% after 6 months of culture (P <0.01). Insulin stimulation indices did not differ among the fresh or the 6-month cultured preparations. All preparations cultured for 1 to 3 months functioned in the NOD-SCID mice. After 6 months of culture, two of the three preparations demonstrated in vivo function and were able to cure streptozotocin-induced diabetes. CONCLUSIONS: These data demonstrate that human islets can be cultured in M-SFM for extended periods and still retain in vitro and in vivo function and the ability to cure experimental diabetes. The ability to maintain islets in culture for prolonged periods is an important step toward the development of islet tissue repositories and distribution centers.


Assuntos
Ilhotas Pancreáticas/fisiologia , Preservação de Tecido/métodos , Animais , Peptídeo C/biossíntese , Meios de Cultura Livres de Soro , Técnicas de Cultura , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/terapia , Sobrevivência de Enxerto , Humanos , Insulina/biossíntese , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Tempo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA