Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Br J Clin Pharmacol ; 90(2): 568-581, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37872122

RESUMO

AIMS: Mirvetuximab soravtansine is a first-in-class antibody-drug conjugate recently approved for the treatment of folate receptor-α positive ovarian cancer. The aim of this study was to develop a population pharmacokinetic model to describe the concentration-time profiles of mirvetuximab soravtansine, the payload (DM4) and a metabolite (S-methyl-DM4). METHODS: Mirvetuximab soravtansine was administered intravenously from 0.15 to 7 mg/kg to 543 patients with predominantly platinum-resistant ovarian cancer in 3 clinical studies, and the plasma drug concentrations were analysed using a nonlinear mixed-effects modelling approach. Stepwise covariate modelling was performed to identify covariates. RESULTS: We developed a semi-mechanistic population pharmacokinetic model that included linear and nonlinear routes for the elimination of mirvetuximab soravtansine and a target compartment for the formation and disposition of the payload and metabolite in tumour cells. The clearance and volume of the central compartment were 0.0153 L/h and 2.63 L for mirvetuximab soravtansine, 8.83 L/h and 3.67 L for DM4, and 2.04 L/h and 6.3 L for S-methyl-DM4, respectively. Body weight, serum albumin and age were identified as statistically significant covariates. Exposures in patients with renal or hepatic impairment and who used concomitant cytochrome P450 (CYP) 3A4 inhibitors were estimated. CONCLUSION: There is no need for dose adjustment due to covariate effects for mirvetuximab soravtansine administered at the recommended dose of 6 mg/kg based on adjusted ideal body weight. Dose adjustment is not required for patients with mild or moderate renal impairment, mild hepatic impairment, or when concomitant weak and moderate CYP3A4 inhibitors are used.


Assuntos
Anticorpos Monoclonais Humanizados , Imunoconjugados , Maitansina , Neoplasias Ovarianas , Humanos , Feminino , Resistencia a Medicamentos Antineoplásicos , Neoplasias Ovarianas/tratamento farmacológico , Imunoconjugados/efeitos adversos , Ácido Fólico/farmacologia , Ácido Fólico/uso terapêutico , Maitansina/análogos & derivados
2.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35511740

RESUMO

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Assuntos
Imunoconjugados , Proteínas ADAM , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Imunoconjugados/química , Proteínas de Membrana/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Clin Cancer Res ; 23(17): 5015-5023, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490463

RESUMO

Purpose: To evaluate the safety, MTD, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of TAK-117 (MLN1117/INK1117), an investigational PI3Kα-selective inhibitor, in patients with advanced solid tumors.Experimental Design: Seventy-one patients received oral TAK-117 once daily [100-300 mg (n = 24)] or 3 days per week [Monday-Wednesday-Friday (MWF), 200-1,200 mg (n = 27); Monday-Tuesday-Wednesday (MTuW), 200-900 mg (n = 20)], in 21-day cycles. Dose escalation proceeded via a 3 + 3 design.Results: TAK-117 once-daily dosing was associated with dose-limiting grade ≥3 alanine/aspartate aminotransferase (ALT/AST) elevations, resulting in a narrow range of tolerable doses (100-150 mg once daily). With MWF/MTuW dosing, no dose-limiting ALT/AST elevations occurred until the MTD of 900 mg; total weekly dose was 2.6-fold that of 150 mg once daily. Drug-related grade ≥3 adverse events occurred in 25%/22%/35% (including hyperglycemia in 0%/7%/15%) of once-daily/MWF/MTuW patients. TAK-117 (100-1,200 mg) exhibited moderately fast oral absorption, a generally dose proportional increase in exposure, and plasma half-life of approximately 11 hours. Total weekly exposures with 900 mg MWF/MTuW dosing were approximately 4 times greater than with 150 mg once daily. Skin pS6 expression was suppressed at ≥200 mg. There were 3/1/0 partial responses (once daily/MWF/MTuW) and 5/7/5 patients had stable disease lasting ≥3 months (all PIK3CA mutated).Conclusions: Intermittent dosing of TAK-117 had an acceptable safety profile and enabled higher doses and total weekly exposures versus once-daily dosing. Although the potential for TAK-117 as single-agent therapy appears limited, further evaluation in combination approaches for advanced solid tumors is warranted. Clin Cancer Res; 23(17); 5015-23. ©2017 AACR.


Assuntos
Benzoxazóis/administração & dosagem , Classe I de Fosfatidilinositol 3-Quinases/genética , Imidazóis/administração & dosagem , Morfolinas/administração & dosagem , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Piridinas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Benzoxazóis/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Humanos , Imidazóis/efeitos adversos , Masculino , Pessoa de Meia-Idade , Morfolinas/efeitos adversos , Estadiamento de Neoplasias , Neoplasias/genética , Neoplasias/patologia , Inibidores de Proteínas Quinases/efeitos adversos , Piridinas/efeitos adversos
4.
Cancer Chemother Pharmacol ; 79(2): 315-326, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28097385

RESUMO

PURPOSE: This first-in-human report examined the recommended Phase 2 dose and schedule of litronesib, a selective allosteric kinesin Eg5 inhibitor. METHODS: Two concurrent dose-escalation studies investigated litronesib across the dose range of 0.125-16 mg/m2/day, evaluating the following schedules of administration on a 21-day cycle: Days 1, 2, 3; Days 1, 5, 9; Days 1, 8; Days 1, 5; or Days 1, 4, with or without pegfilgrastim. Best overall response was defined per Response Evaluation Criteria in Solid Tumors (RECIST Version 1.0). Pharmacokinetic (PK) evaluations were performed. Exploratory PK/pharmacodynamic analyses investigated the relationship between litronesib plasma exposure and changes in phosphohistone H3 (pHH3) levels. RESULTS: One hundred and seventeen patients with advanced malignancies were enrolled. Neutropenia was the primary dose-limiting toxicity. Prophylactic pegfilgrastim reduced neutropenia frequency and severity, allowing administration of higher litronesib doses, but increases in the incidences of mucositis and stomatitis were observed. Among 86 response-evaluable patients, 2 patients (2%) achieved partial response, both on the Days 1, 2, 3 regimen (5 and 6 mg/m2/day with pegfilgrastim), and 17 patients (20%) maintained stable disease for ≥6 cycles. Dose-dependent increases in litronesib plasma exposure were observed, with minor intra- and inter-cycle accumulation, along with exposure-dependent increases in pHH3 expression in tumor and skin biopsies. CONCLUSIONS: On the basis of the results of these studies, two regimens were selected for Phase 2 exploration: 6 mg/m2/day on Days 1, 2, 3 plus pegfilgrastim and 8 mg/m2/day on Days 1, 5, 9 plus pegfilgrastim, both on a 21-day cycle.


Assuntos
Cinesinas/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Sulfonamidas/administração & dosagem , Tiadiazóis/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Esquema de Medicação , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Polietilenoglicóis , Proteínas Recombinantes/administração & dosagem , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Tiadiazóis/efeitos adversos , Tiadiazóis/farmacocinética
5.
Mol Cancer Ther ; 14(11): 2463-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26304237

RESUMO

Intervention of cancer cell mitosis by antitubulin drugs is among the most effective cancer chemotherapies. However, antitubulin drugs have dose-limiting side effects due to important functions of microtubules in resting normal cells and are often rendered ineffective by rapid emergence of resistance. Antimitotic agents with different mechanisms of action and improved safety profiles are needed as new treatment options. Mitosis-specific kinesin Eg5 represents an attractive anticancer target for discovering such new antimitotic agents, because Eg5 is essential only in mitotic progression and has no roles in resting, nondividing cells. Here, we show that a novel selective Eg5 inhibitor, LY2523355, has broad target-mediated anticancer activity in vitro and in vivo. LY2523355 arrests cancer cells at mitosis and causes rapid cell death that requires sustained spindle-assembly checkpoint (SAC) activation with a required threshold concentration. In vivo efficacy of LY2523355 is highly dose/schedule-dependent, achieving complete remission in a number of xenograft tumor models, including patient-derived xenograft (PDX) tumor models. We further establish that histone-H3 phosphorylation of tumor and proliferating skin cells is a promising pharmacodynamic biomarker for in vivo anticancer activity of LY2523355.


Assuntos
Apoptose/efeitos dos fármacos , Cinesinas/antagonistas & inibidores , Mitose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Sulfonamidas/farmacologia , Tiadiazóis/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células HCT116 , Células HT29 , Células HeLa , Humanos , Immunoblotting , Cinesinas/metabolismo , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/patologia , Fatores de Tempo , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Drug Discov Today ; 18(11-12): 510-22, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22944609

RESUMO

Targeting multiple hallmarks of cancer with drug combinations may provide unique opportunities for cancer therapeutics; however, phenotypic quantification is necessary to understand in vivo mechanisms of action of each drug alone or in combination. Immunohistochemistry (IHC) can quantify phenotypic changes, but traditional methods are not amenable for high-throughput drug discovery. In this article, we describe a high-content method to quantify changes in tumor angiogenesis, vascular normalization, hypoxia, tumor cell proliferation, and apoptosis using IHC. This method to quantify tumor model phenotypes can be useful for cancer drug discovery by increasing the understanding of: (i) tumor models used in efficacy studies, (ii) changes occurring during the growth of the tumor, and (iii) novel mechanisms of actions of cancer therapeutics.


Assuntos
Antineoplásicos , Descoberta de Drogas , Imuno-Histoquímica , Neoplasias/metabolismo , Animais , Antineoplásicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
7.
Invest New Drugs ; 31(1): 136-44, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22492020

RESUMO

PURPOSE: This phase I study aims at assessing the safety and tolerability of LY2603618, a selective inhibitor of Checkpoint Kinase 1, in combination with pemetrexed and determining the maximum tolerable dose and the pharmacokinetic parameters. EXPERIMENTAL DESIGN: This was an open-label, multicenter, dose-escalation study in patients with advanced solid tumors. Increasing doses of LY2603618 (40-195 mg/m(2)) were combined with 500 mg/m(2) of pemetrexed. LY2603618 was administered on Days 1 and 9 and pemetrexed on Day 8 in a 28-day cycle. For all subsequent 21-day cycles, pemetrexed was administered on Day 1 and LY2603618 on Day 2. Antitumor activity was evaluated as per Response Evaluation Criteria in Solid Tumors 1.0. RESULTS: A total of 31 patients were enrolled into six cohorts (three at 40 mg/m(2) over 4.5-hour infusion, 1-hour infusion in subsequent cohorts: three each at 40 mg/m(2), 70 mg/m(2), and 195 mg/m(2); 13 at 105 mg/m(2); six at 150 mg/m(2)). Four patients experienced a dose-limiting toxicity: diarrhea (105 mg/m(2)); reversible infusion-related reaction (150 mg/m(2)); thrombocytopenia (195 mg/m(2)); and fatigue (195 mg/m(2)). The maximum tolerated dose was defined as 150 mg/m(2). The pharmacokinetic data demonstrated that the exposure of LY2603618 increased in a dose-dependent manner, displayed a suitable half-life for maintaining required human exposures while minimizing the intra- and inter-cycle accumulation, and was unaffected by the pemetrexed administration. The pharmacokinetic-defined biologically efficacious dose was achieved at doses ≥105 mg/m(2). CONCLUSION: LY2603618 administered approximately 24 h after pemetrexed showed acceptable safety and pharmacokinetic profiles.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Quinase 1 do Ponto de Checagem , Feminino , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Guanina/análogos & derivados , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/sangue , Pemetrexede , Compostos de Fenilureia/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Quinases , Pirazinas/administração & dosagem
8.
Semin Oncol ; 31(2): 198-205, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15112150

RESUMO

Lymphoma and myeloma represent an increasingly important cause of morbidity and mortality in the older patient group. Studies to date, while limited, suggest that there may be differences in the underlying biology of the tumor cell of some lymphoid malignancies in younger versus older patients. Significant new data will be required to document and elucidate the differences and use them to tailor therapy to discrete disease entities. Increasing numbers of clinical trials are addressing the issues of treatment in the elderly, particularly in the aggressive non-Hodgkin's lymphomas. However, much work remains in improving the participation of older patient groups in current clinical trials and in overcoming inherent treatment biases related to concerns of toxicity simply as a function of age, both of which limit application of existing successful therapy to older patient groups.


Assuntos
Linfoma , Mieloma Múltiplo , Idoso , Envelhecimento/fisiologia , Citogenética , Humanos , Linfoma/epidemiologia , Linfoma/patologia , Linfoma/terapia , Mieloma Múltiplo/epidemiologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Estadiamento de Neoplasias , Prognóstico
9.
Nutrition ; 18(2): 139-46, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11844645

RESUMO

OBJECTIVE: Renal transplantation is associated with an increased risk of atherosclerotic cardiovascular disease and marked racial and ethnic disparities in graft and patient survival. We characterized differences in racial and ethnic susceptibility to weight gain, diabetes, and alterations in circulating lipid levels and isolated independent predictors of those changes in a diverse population of kidney transplant recipients. METHODS: The data for this analysis were drawn from a prospectively collected database of 506 renal transplant recipients obtained between 1983 and 1998. Univariate and multivariate analyses characterized differences in outcomes and predictors of cardiovascular risk by race and ethnicity. RESULTS: In all recipients, coronary artery disease was the most common cause of death, and African-American recipients had the shortest graft survival and the highest percentage of deaths. At 1 y post-transplantation, 39% of African-American recipients were obese (body mass index > 30), and the odds ratios for post-transplant diabetes were 3.5 and 5 times greater in non-white and obese recipients, respectively. CONCLUSIONS: Multiple regression analysis confirmed the predominant independent effect of African American race or ethnicity on weight gain; however, hypercholesterolemia was independent of race or ethnicity and predicted by cyclosporine treatment and post-transplant diabetes. Therefore, kidney transplantation represents a state of accelerated atherogenic risk induced in part by the metabolic effects of immunosuppressive medications and compounded by marked racial and ethnic disparities in weight gain and diabetes risk.


Assuntos
Doenças Cardiovasculares/etiologia , Diabetes Mellitus/epidemiologia , Transplante de Rim/fisiologia , Obesidade/epidemiologia , Aumento de Peso/fisiologia , Adulto , População Negra , Doenças Cardiovasculares/mortalidade , Colesterol/sangue , Estudos de Coortes , Diabetes Mellitus/etnologia , Diabetes Mellitus/genética , Feminino , Rejeição de Enxerto/epidemiologia , Rejeição de Enxerto/etnologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/genética , Humanos , Imunossupressores/uso terapêutico , Transplante de Rim/efeitos adversos , Transplante de Rim/mortalidade , Masculino , Obesidade/etnologia , Obesidade/genética , Estudos Prospectivos , Análise de Regressão , Fatores de Risco , Análise de Sobrevida , Triglicerídeos/sangue , Aumento de Peso/genética , População Branca
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA