Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 21(11): 2233-2239, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20444843

RESUMEN

BACKGROUND: Erlotinib and pemetrexed are approved single agents for second-line treatment of non-small-cell lung cancer (NSCLC) and, in combination, have shown synergistic antitumor activity in NSCLC cell lines. We investigated the safety, pharmacokinetics and preliminary efficacy of combined erlotinib-pemetrexed in patients with refractory advanced NSCLC. PATIENTS AND METHODS: A nonrandomized, open-label, phase IB study was performed in patients with advanced NSCLC whose disease had progressed on or following first-line chemotherapy with a platinum-containing regimen or for whom the erlotinib-pemetrexed combination was considered appropriate. Patients received i.v. pemetrexed 500-700 mg/m² every 3 weeks and oral erlotinib 100-150 mg/day. RESULTS: Twenty patients were recruited. The most common adverse events (AEs) were rash, diarrhea and fatigue. Serious AEs occurred in eight patients (three treatment related) and there were eight deaths (none treatment related). Dose-limiting toxic effects were not experienced up to erlotinib 150 mg/day plus pemetrexed 600 mg/m². Concurrent administration did not affect pharmacokinetic parameters. Two patients achieved partial responses and nine had stable disease. CONCLUSIONS: Erlotinib-pemetrexed combination is well tolerated at doses equal to the licensed single-agent doses (150 mg/day and 500 mg/m², respectively). The good tolerability profile and promising efficacy indicate that this combination warrants further investigation for patients with advanced NSCLC.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Grandes/tratamiento farmacológico , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/secundario , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/secundario , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundario , Progresión de la Enfermedad , Receptores ErbB/genética , Clorhidrato de Erlotinib , Femenino , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mutación/genética , Estadificación de Neoplasias , Pemetrexed , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Quinazolinas/administración & dosificación , Tasa de Supervivencia , Factores de Tiempo , Distribución Tisular , Resultado del Tratamiento , Proteínas ras/genética
2.
Ann Oncol ; 20(9): 1565-1575, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19487488

RESUMEN

BACKGROUND: This phase Ib study evaluated the safety, pharmacokinetics, and activity of enzastaurin either 500 mg once daily (QD) or 250 mg twice daily (b.i.d.) in combination with pemetrexed. PATIENTS AND METHODS: Pemetrexed 500 mg/m(2) with folic acid and vitamin B(12) was given on day 1 every 21 days with enzastaurin 500 mg orally QD starting on day 5 of cycle 1 after a loading dose of 400 mg thrice daily on day 4. To evaluate whether a b.i.d. regimen results in higher enzastaurin exposures, the study was amended. After amendment, in cycle 1, patients received 500 mg enzastaurin QD on days 1-15 without initial loading dose and 250 mg b.i.d. on days 16-30; in subsequent cycles, patients received pemetrexed on day 1 every 21 days with enzastaurin b.i.d. RESULTS: Sixty-eight patients (42 preamendment and 26 postamendment) were assessed. Pemetrexed toxicity and pharmacokinetics did not appear to be altered by enzastaurin. Enzastaurin average steady-state plasma concentration (C(av,ss)) decreased by approximately 25% in the presence of pemetrexed. Enzastaurin C(av,ss) were approximately 40% higher in the b.i.d. versus QD regimen. Three patients (4.4%) with thyroid cancer of follicular/papillary type had partial response as defined by RECIST. CONCLUSIONS: Pemetrexed plus enzastaurin is well tolerated with preliminary evidence of anticancer activity, particularly in thyroid cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Glutamatos/administración & dosificación , Glutamatos/efectos adversos , Glutamatos/farmacocinética , Guanina/administración & dosificación , Guanina/efectos adversos , Guanina/análogos & derivados , Guanina/farmacocinética , Humanos , Indoles/administración & dosificación , Indoles/efectos adversos , Indoles/farmacocinética , Masculino , Persona de Mediana Edad , Pemetrexed
3.
J Natl Cancer Inst ; 84(23): 1816-20, 1992 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-1331485

RESUMEN

BACKGROUND: Topotecan [(S)-9-dimethylaminomethyl(10-hydroxy-camptothecin), NSC 609699, SK&F 104864A], a semisynthetic analogue of the natural product camptothecin, is a cell cycle-specific drug that exerts antineoplastic activity through inhibition of topoisomerase I. Currently, topotecan is undergoing phase I and early phase II clinical trials. The dose-limiting toxicity for topotecan is myelosuppression. PURPOSE: Our purpose was to determine plasma concentrations and exposure times necessary for optimal clinical activity and tumor types that may be responsive in phase II clinical studies of topotecan. METHODS: A soft-agar cloning system assay was used to determine the in vitro effects of topotecan against cells from biopsy specimens of colorectal, breast, lung, ovarian, renal cell, and gastric cancers and cancers of unknown primary origin. We studied 141 freshly explanted tumor specimens, using 1-hour exposure to topotecan, and 80 were studied using continuous exposure. A decrease in tumor colony formation resulting from drug exposure was considered an in vitro response if survival of colonies was up to 50% of that in controls. RESULTS: With 1-hour exposure, in vitro responses were seen in 10% and 25% of assessable tumor specimens at final topotecan concentrations of 1.0 and 10.0 micrograms/mL, respectively. With continuous exposures at concentrations of 0.1 and 1.0 micrograms/mL, in vitro response rates were 34% and 76%, respectively. Specific activity was seen against colorectal, breast, non-small-cell lung, ovarian, and renal cell cancers, with responses observed in 27%, 25%, 32%, 39%, and 83%, respectively, of assessable tumor specimens after continuous exposure to 0.1 micrograms/mL topotecan. A subset of tumor specimens resistant to doxorubicin or fluorouracil was sensitive to topotecan, and the difference in sensitivity was statistically significant. In addition, some of the tumor specimens resistant to cyclophosphamide and etoposide were also sensitive to topotecan. CONCLUSIONS: Topotecan appears to be active in vitro against a variety of human tumors, including a subgroup resistant in vitro to standard antineoplastic agents. If plasma levels of 0.1 micrograms/mL can be achieved for prolonged periods of time in ongoing clinical trials, topotecan should have substantial clinical activity. IMPLICATIONS: Further clinical development of topotecan is warranted.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Inhibidores de Topoisomerasa I , Ensayo de Tumor de Célula Madre , Camptotecina/farmacología , Distribución de Chi-Cuadrado , Células Clonales , Humanos , Topotecan , Células Tumorales Cultivadas/efectos de los fármacos
4.
Cancer Res ; 53(16): 3747-51, 1993 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8339286

RESUMEN

Four cell lines from human breast cancer (HTB 19, HTB 133, IZB B, and MCF 7M) were investigated for in vitro growth before and after incubation with the ether lipid 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine (ET-18-OCH3) for 4 h and at increasing concentrations (25, 50, 75, and 100 micrograms/ml), as well as with and without cryopreservation. When measured with the human tumor cloning assay in agar or methyl-cellulose the surviving fraction showed an inverse correlation with the concentrations of ET-18-OCH3. After a 4-h exposure with 75 micrograms/ml ET-18-OCH3 at a cell density of 2 x 10(5)/ml the number of colonies of HTB 19 decreased from 75 +/- 10/10(3) cells (100%) to 1 +/- 0/10(3) cells (1%), and after subsequent cryopreservation no remaining colonies were found. In order to simulate the situation in autologous bone marrow transplantation we contaminated normal human bone marrow cells with malignant HTB 19 breast cancer cells at a ratio of 100:1. After incubation with 75 micrograms/ml ET-18-OCH3 for 4 h and subsequent cryopreservation there was a considerable reduction of HTB 19 colonies whereas the majority of normal human hematopoietic progenitors recovered. We conclude that ET-18-OCH3, in combination with cryopreservation, can remove breast cancer cells from bone marrow by up to 1 log and may be useful for purging bone marrow for autologous bone marrow transplantation not only in acute leukemia and non-Hodgkin's lymphoma but also in breast cancer.


Asunto(s)
Antineoplásicos , Purgación de la Médula Ósea , Trasplante de Médula Ósea , Neoplasias de la Mama/patología , Criopreservación , Éteres Fosfolípidos , Neoplasias de la Mama/cirugía , Femenino , Humanos , Trasplante Autólogo , Células Tumorales Cultivadas
5.
Cancer Res ; 46(11): 5567-70, 1986 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3463415

RESUMEN

alpha-Transforming growth factors (TGFs) are low-molecular-weight polypeptides (Mr 5000-7000) which are secreted by a variety of human cancer cells in vitro. Their presence has also been reported in the urine of patients with malignancies, human tumor extracts, and in the conditioned medium of primary human tumor cell cultures. There is evidence that alpha-TGFs bind to membrane receptors of the secreting cells, thus stimulating cell growth in a positive feedback manner (autocrine secretion). We have used a synthetic linear alpha-TGF to study the biological activity of affinity-purified polyclonal sheep antibodies against the carboxyterminal part (17 amino acids) of synthetic rat alpha-TGF. The antigen was found to bind to epidermal growth factor receptors of target cells and to stimulate soft agarose colony formation of normal fibroblasts. Although the antibodies recognized the linear alpha-TGF molecule, they did not inhibit the binding to epidermal growth factor receptors. The antibodies also failed to inhibit alpha-TGF-stimulated colony formation of normal rat fibroblasts. In addition, essentially no cytotoxic activity of the antibodies was found against 41 fresh human tumor specimens in a human tumor cloning assay. Antibodies against the complete alpha-TGF molecule should be used in further attempts to interfere with the autocrine secretion of transforming growth factors.


Asunto(s)
Receptores ErbB/metabolismo , Péptidos/farmacología , Animales , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Células Cultivadas , Humanos , Fragmentos de Péptidos/síntesis química , Péptidos/inmunología , Péptidos/metabolismo , Neoplasias Hipofisarias/patología , Radioinmunoensayo , Ratas , Factores de Crecimiento Transformadores
6.
Cancer Res ; 48(17): 5023-8, 1988 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-3165707

RESUMEN

alpha Transforming growth factors (alpha-TGFs) are polypeptides that stimulate anchorage-independent growth of various nontransformed cells in vitro and are believed to be involved in autocrine stimulation of tumor cells. alpha-TGF activity is secreted by a variety of human cancers leading to the possibility that it may serve as a tumor marker. alpha-TGF activity was measured in 130 effusions from patients with various types of cancer with a radioimmunoassay using sheep antibodies against the C-terminal 17 amino acids of linear rat alpha-TGF. Forty-two % of the effusions contained immunoreactive alpha transforming growth factor (Ir-alpha-TGF) activity, including 13 of 34 (38%) breast cancer, 12 of 24 (50%) lung cancer, and 13 of 31 (42%) ovarian cancer specimens. Concentrations ranged from 1.56 to 50 ng/ml. Only 3 of 17 control effusions from noncancer patients had low levels of activity, all less than 2 ng/ml. The presence of Ir-alpha-TGF activity correlated with patients' performance status (PS) and tumor burden. It was present in 18 of 67 (27%) effusions of patients with PS less than or equal to 2 and in 23 of 33 (70%) with PS 3 or 4 (P less than 0.0001). Only 2 of 43 (4%) patients with one site of metastatic disease had detectable Ir-alpha-TGF (mean, 0.23 ng/ml); 18 of 37 (48%) with two sites (mean, 5.22 ng/ml, P less than 0.0001); and 33 of 34 (97%) with greater than two sites (mean, 5.93 ng/ml, P = 0.002). It was present in a larger percentage of effusions from breast cancer patients with estrogen- and progesterone receptor-negative tumors. Univariate analysis revealed that detectable Ir-alpha-TGF activity, PS 3 or 4, and the number of sites of disease correlated with a shorter survival. Only Ir-alpha-TGF and PS 3 or 4 retained significance in a multivariate analysis. In conclusion, Ir-alpha-TGF is frequently detectable in effusions from cancer patients, it correlates with other known adverse prognostic factors, and its presence predicts for a poor survival. Further studies of alpha-TGF activity in more readily accessible body fluids such as serum or urine are warranted.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias/análisis , Péptidos/análisis , Líquido Ascítico/análisis , Humanos , Metástasis de la Neoplasia , Neoplasias/mortalidad , Péptidos/inmunología , Derrame Pleural/metabolismo , Pronóstico , Factores de Crecimiento Transformadores
7.
Cancer Res ; 48(18): 5353-7, 1988 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-2842047

RESUMEN

Carbetimer (carbethimer, N-137, NED-137, carboxyimamidate) is a low molecular weight polyelectrolyte with antitumor activity in a variety of tumor models. This phase I trial evaluated a single dose of carbetimer infused over 1-2 h every 28 days. Forty-three patients received 71 courses of the drug at doses ranging from 180 to 8500 mg/m2. The dose-limiting toxicity was hypercalcemia (serum calcium greater than 12.5 mg/dl) noted in two of three patients at a dose of 8500 mg/m2. Serum calcium levels between 10.5 and 12.5 mg/dl were noted in an additional three patients treated at doses greater than or equal to 1600 mg/m2. Calcium balance studies in three patients treated at 6500 mg/m2 revealed an increase in urinary cyclic AMP and phosphate excretion after treatment accompanied by a mild elevation of serum parathyroid hormone. Immunological studies in these patients revealed a statistically significant increase in the percentage of peripheral T-helper cells. An increase in the T-helper/suppressor cell ratio was observed in two of the three patients studied. Interleukin 2 production by phytohemagglutinin-stimulated peripheral mononuclear cells was increased in two of three patients. One patient with a renal cell carcinoma showed a mixed response. The recommended dose for phase II trials as assessed from this study is 6500 mg/m2 as a single dose every 28 days.


Asunto(s)
Neoplasias/tratamiento farmacológico , Polímeros/uso terapéutico , Adulto , Anciano , Anticuerpos Monoclonales , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Calcio/sangre , Calcio/orina , AMP Cíclico/orina , Evaluación de Medicamentos , Femenino , Humanos , Interleucina-2/biosíntesis , Masculino , Persona de Mediana Edad , Fosfatos/orina , Polímeros/efectos adversos , Albúmina Sérica/análisis
8.
Biochim Biophys Acta ; 1221(2): 115-24, 1994 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-8148388

RESUMEN

The formation of the unusual amino-acid hypusine in eIF-5A (eukaryotic initiation factor 5A) is associated with cellular proliferation. We used a panel of compounds, including mimosine, to probe the relationship between the exit from the G1 phase of the cell cycle, i.e., the onset of DNA replication, and the formation of hypusine by the enzyme deoxyhypusyl hydroxylase (DOHH). These two parameters displayed the same dose dependency and structure-activity relationship. Only compounds that inhibited DOHH also suppressed proliferation. This effect was observed: (i) in spontaneously proliferating, virally transformed, and mitogen-stimulated cells; (ii) for both anchorage-dependent and anchorage-independent proliferation; and (iii) with normal and malignant cell lines. DOHH reactivation occurred rapidly after inhibitor withdrawal and correlated with synchronized entry into S. The changes in the expression of specific genes during the G1-to-S transition mimicked the physiological pattern. These findings suggest that hypusine formation in eIF-5A which occurs in a specific, invariant sequence motif acquired early in evolution, may be involved in the G1-to-S transition in the eukaryotic cells tested.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fase G1/efectos de los fármacos , Lisina/análogos & derivados , Oxigenasas de Función Mixta/antagonistas & inhibidores , Proteínas de Unión al ARN , Fase S/efectos de los fármacos , Alanina/análogos & derivados , Alanina/farmacología , Secuencia de Aminoácidos , Animales , División Celular/efectos de los fármacos , Línea Celular , Línea Celular Transformada , Humanos , Lisina/análisis , Lisina/biosíntesis , Mimosina/farmacología , Datos de Secuencia Molecular , Factores de Iniciación de Péptidos/química , Pironas/farmacología , Alineación de Secuencia , Relación Estructura-Actividad , Factor 5A Eucariótico de Iniciación de Traducción
9.
J Clin Oncol ; 17(10): 3009-16, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10506594

RESUMEN

PURPOSE: Multitargeted antifolate (MTA; LY231514) has broad preclinical antitumor activity and inhibits a variety of intracellular enzymes involved in the folate pathways. This study was designed to (1) determine the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetics of MTA combined with cisplatin; (2) determine a recommended dose for phase II studies; and (3) collect anecdotal information on the antitumor activity of MTA combined with cisplatin. PATIENTS AND METHODS: Patients with solid tumors received MTA intravenously over 10 minutes and cisplatin over 2 hours once every 21 days. In cohort 1, both agents were administered on day 1 starting with MTA 300 mg/m(2) and cisplatin 60 mg/m(2). In cohort 2, MTA (500 or 600 mg/m(2)) was administered on day 1, followed by cisplatin (75 mg/m(2)) on day 2. RESULTS: In cohort 1, 40 assessable patients received 159 courses of treatment. The MTD was MTA 600 mg/m(2)/cisplatin 100 mg/m(2). DLTs were reversible leukopenia/neutropenia and delayed fatigue. Hydration before cisplatin therapy did not influence MTA pharmacokinetics. Eleven objective remissions included one complete response in a patient with relapsed squamous cell head and neck carcinoma, and partial responses in four of ten patients with epithelial pleural mesothelioma. In cohort 2, 11 assessable patients received 23 courses of treatment. The MTD was MTA 600 mg/m(2) and cisplatin 75 mg/m(2). DLTs were neutropenic sepsis, diarrhea, and skin toxicity. Two patients died of treatment-related complications during the study. Two patients had objective remissions (one mesothelioma patient, one colon cancer patient). CONCLUSION: The combination of MTA and cisplatin is clinically active, and administering both agents on day 1 is superior to a split schedule. Further development of this combination for mesothelioma is warranted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antagonistas del Ácido Fólico/farmacocinética , Glutamatos/farmacocinética , Guanina/análogos & derivados , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas del Ácido Fólico/administración & dosificación , Antagonistas del Ácido Fólico/efectos adversos , Glutamatos/administración & dosificación , Glutamatos/efectos adversos , Guanina/administración & dosificación , Guanina/efectos adversos , Guanina/farmacocinética , Humanos , Infusiones Intravenosas , Masculino , Mesotelioma/tratamiento farmacológico , Persona de Mediana Edad , Pemetrexed , Resultado del Tratamiento
10.
J Clin Oncol ; 18(14): 2772-9, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10894878

RESUMEN

PURPOSE: To investigate the side effects, determine the maximum-tolerated dose (MTD), and study the pharmacokinetics of S-1, an oral fluoropyrimidine-based antineoplastic agent consisting of the fluorouracil (5-FU) prodrug tegafur combined with two modulators, 5-chloro-2,4-dihydroxypyridine and potassium oxonate. PATIENTS AND METHODS: Patients with advanced solid tumors received S-1 bid for 28 days, followed by 1 week of rest. 5-FU pharmacokinetics were investigated after a single initial dose of S-1 during the first 24 hours and weekly thereafter. RESULTS: Twenty-eight patients received S-1 at the four consecutive dose levels of 25, 45, 35, and 40 mg/m(2). The MTD was initially found at 45 mg/m(2), with diarrhea as the dose-limiting toxicity (DLT). Diarrhea was also the DLT at the dose of 40 mg/m(2), which was the MTD for patients exposed to extensive prior chemotherapy. Other toxicities were generally mild. Two patients had a reduction of more than 50% in tumor dimension. Plasma pharmacokinetics of 5-FU were linear; at the highest S-1 dose level, 5-FU plasma peak concentrations reached 1 to 2 micromol/L, and the half-life of 5-FU was 3 to 4 hours. A statistically significant relationship was observed between the severity of diarrhea and pharmacokinetic parameters of 5-FU. CONCLUSION: The recommended dose of S-1 in chemotherapy-naive or minimally chemotherapy-exposed patients is 40 mg/m(2) bid on 28 consecutive days, every 5 weeks. In heavily pretreated patients, the recommended dose is 35 mg/m(2) bid. Phase II trials are warranted in tumors known to be responsive to 5-FU treatment.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Ácido Oxónico/uso terapéutico , Piridinas/uso terapéutico , Tegafur/uso terapéutico , Administración Oral , Adulto , Anciano , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Combinación de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ácido Oxónico/administración & dosificación , Ácido Oxónico/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacocinética , Tegafur/administración & dosificación , Tegafur/farmacocinética
11.
J Clin Oncol ; 19(4): 1167-75, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11181683

RESUMEN

PURPOSE: A single-agent dose-escalating phase I and pharmacokinetic study on the farnesyl transferase inhibitor SCH 66336 was performed to determine the safety profile, maximum-tolerated dose, and recommended dose for phase II studies. Plasma and urine pharmacokinetics were determined. PATIENTS AND METHODS: SCH 66336 was given orally bid without interruption to patients with histologically or cytologically confirmed solid tumors. Routine antiemetics were not prescribed. RESULTS: Twenty-four patients were enrolled onto the study. Dose levels studied were 25, 50, 100, 200, 400, and 300 mg bid. Pharmacokinetic sampling was performed on days 1 and 15. At 400 mg bid, the dose-limiting toxicity (DLT) consisted of grade 4 vomiting, grade 4 neutropenia and thrombocytopenia, and the combination of grade 3 anorexia and diarrhea with reversible grade 3 plasma creatinine elevation. After dose reduction, at 300 mg bid, the DLTs consisted of grade 4 neutropenia, grade 3 neurocortical toxicity, and the combination of grade 3 fatigue with grade 2 nausea and diarrhea. The recommended dose for phase II studies is 200 mg bid, which was found feasible for prolonged periods of time. Pharmacokinetic analysis showed a greater than dose-proportional increase in drug exposure and peak plasma concentrations, with increased parameters at day 15 compared with day 1, indicating some accumulation on multiple dosing. Plasma half-life ranged from 4 to 11 hours and seemed to increase with increasing doses. Steady-state plasma concentrations were attained at days 7 through 14. A large volume of distribution at steady-state indicated extensive distribution outside the plasma compartment. CONCLUSION: SCH 66336 can be administered safely using a continuous oral bid dosing regimen. The recommended dose for phase II studies using this regimen is 200 mg bid.


Asunto(s)
Antineoplásicos/uso terapéutico , Piperidinas/uso terapéutico , Piridinas/uso terapéutico , Administración Oral , Adulto , Anciano , Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Farnesiltransferasa , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/farmacocinética , Piridinas/farmacocinética
12.
Clin Cancer Res ; 6(5): 1736-43, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10815892

RESUMEN

A single-agent dose-escalating Phase I and pharmacological study of the polyamine synthesis inhibitor SAM 486A was performed. A dosing regimen of four weekly infusions followed by 2 weeks off therapy was studied. Fifty patients were entered into the study. Dose levels studied were 1.25, 2.5, 5, 8, 16, 32, 48, 70, 110, 170, 270, and 325 mg/m2/week. Pharmacokinetic sampling was done on day 1, and trough samples were taken weekly during the first treatment cycle. Pharmacodynamic sampling was done on days 1 and 22. At 325 mg/m2/week, dose-limiting toxicity was seen (one patient each with grade 4 febrile neutropenia, grade 3 neurotoxicity, and grade 3 hypotension with syncope and T-wave inversions on electrocardiogram). The recommended dose for further testing was set at 270 mg/m2/week. Infusion time was increased from 10 to 180 min due to facial paresthesias and flushing and somnolence. Drug exposure increased linearly with dose. Mean +/- SD t1,2 at 70-325 mg/m2 doses was 61.4+/-26.2 h, with a large volume of distribution at steady state. In peripheral blood leukocytes, a clear relationship between dose and inhibitory effect on S-adenosylmethionine decarboxylase or changes in intracellular polyamine pools was not recorded. SAM 486A can be administered safely using a dosing regimen of four weekly infusions followed by 2 weeks off therapy. The recommended dose for Phase II studies using this regimen is 270 mg/m2/week.


Asunto(s)
Amidinas/uso terapéutico , Antineoplásicos/uso terapéutico , Indanos/uso terapéutico , Neoplasias/tratamiento farmacológico , Poliaminas/antagonistas & inhibidores , Adulto , Anciano , Amidinas/efectos adversos , Amidinas/farmacocinética , Anorexia/inducido químicamente , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Área Bajo la Curva , Diarrea/inducido químicamente , Relación Dosis-Respuesta a Droga , Fatiga/inducido químicamente , Femenino , Enfermedades Hematológicas/inducido químicamente , Humanos , Indanos/efectos adversos , Indanos/farmacocinética , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Neoplasias/patología , Poliaminas/metabolismo , Resultado del Tratamiento , Vómitos/inducido químicamente
13.
J Immunother (1991) ; 11(3): 155-8, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1515420

RESUMEN

Interleukin-1 alpha (IL-1 alpha) is a low-molecular-weight cytokine that regulates proliferation and differentiation of lymphatic and myeloid cells. It also has pleiotropic activity on a variety of other target cells and acts as an important mediator of inflammation and septic shock. Recombinant human IL-1 alpha (rhIL-1 alpha) is undergoing clinical evaluation of its potential as an anticancer agent. We have studied the growth modulating effects of rhIL-1 alpha on a variety of freshly explanted human tumor specimens using an in vitro soft agar cloning system. Final concentrations of 0.01-100 ng/ml were used in continuous incubation experiments. Of 139 specimens tested, 56 (40%) were evaluable for determination of tumor growth modulating activity. The most common tumor types examined included breast, nonsmall cell lung, ovarian, colorectal cancer, and melanoma. Stimulation of tumor colony-forming units (colony formation greater than or equal to 1.5 x controls) was observed in only 1/56 (2%) tumors. No evidence was found for increased size of individual colonies after incubation with rhIL-1 alpha. At a concentration of 100 ng/ml, colony formation of 9/56 (16%) tumor specimens was significantly inhibited (colony formation less than or equal to 0.5 x controls). We conclude that rhIL-1 alpha is not a major modulator of tumor colony formation in vitro. However, some antitumor effects may be observed at high concentrations.


Asunto(s)
Interleucina-1/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Humanos , Proteínas Recombinantes/farmacología , Ensayo de Tumor de Célula Madre
14.
FEBS Lett ; 319(1-2): 133-7, 1993 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8454045

RESUMEN

Multidrug-resistant KB-V1 cells carry amplified mdrl gene sequences located in an extrachromosomal compartment (on episomes). Since episomes do not contain centromeric or telomeric sequences it is unclear whether they are able to bind to nuclear matrix proteins that may regulate episomal gene expression. Using high salt treatments followed by in situ hybridization and dot blot analyses we found evidence for direct binding of episomal DNA to nuclear matrix proteins. This binding could only be reversed after incubation with trypsin or proteinase K as determined by contour-clamped homogeneous electric field (CHEF) electrophoresis. Our findings are consistent with the concept that circular extrachromosomal DNA may not only reintegrate into nuclear DNA but may also be subject to functional control by regulatory proteins within the nuclear matrix.


Asunto(s)
ADN/metabolismo , Resistencia a Medicamentos/genética , Proteínas Nucleares/metabolismo , Plásmidos , Línea Celular , Electroforesis , Endopeptidasa K , Hibridación in Situ , Microscopía Fluorescente , Matriz Nuclear/química , Serina Endopeptidasas/farmacología , Cloruro de Sodio/administración & dosificación , Cloruro de Sodio/farmacología , Tripsina/farmacología
15.
FEBS Lett ; 366(2-3): 92-8, 1995 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-7789538

RESUMEN

S phase entry, i.e. start of DNA replication, is a crucial step in proliferation. Inhibition of S phase entry correlates with inhibition of hypusine formation, an event affecting only the eukaryotic initiation factor 5A (eIF-5A). Its hypusine-containing sequence was postulated to authorize polysomal utilization of specific transcripts for proteins necessary to enable DNA replication. Using mimosine to reversibly suppress the hypusine-forming deoxyhypusyl hydroxylase (E.C. 1.14.99.29) in cells while differentially displaying their polysomal versus non-polysomal mRNA populations, we report the detection and classification of several mRNA species that indeed disappear from and reappear at polysomes in concert with inhibition and disinhibition, respectively, of hypusine formation. Based on initial sequence data, two translationally controlled enzymes, both critical for proliferation, are identified as candicate products of such mRNAs, methionine adenosyltransferase (E.C. 2.5.1.6) and cytochrome-c oxidase (EC 1.9.3.1) subunit I. The existence of such putative hypusine-dependent messenger nucleic acids (hymns) provides the basis for a proposal on their molecular function in onset of multiplication.


Asunto(s)
Replicación del ADN , Complejo IV de Transporte de Electrones/metabolismo , Fase G1 , Lisina/análogos & derivados , Metionina Adenosiltransferasa/metabolismo , Factores de Iniciación de Péptidos/fisiología , Polirribosomas/química , ARN Mensajero/genética , Proteínas de Unión al ARN , Fase S , Secuencia de Bases , Línea Celular , Complejo IV de Transporte de Electrones/genética , Humanos , Linfocitos , Lisina/biosíntesis , Metionina Adenosiltransferasa/genética , Mimosina/farmacología , Oxigenasas de Función Mixta/antagonistas & inhibidores , Datos de Secuencia Molecular , ARN Mensajero/aislamiento & purificación , Alineación de Secuencia , Factor 5A Eucariótico de Iniciación de Traducción
16.
Curr Pharm Des ; 10(16): 1923-36, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15180529

RESUMEN

As our understanding of tumorigenesis increases, interference with the various signaling pathways of tumor cells has become an attractive approach to arresting tumor cell growth and overcoming chemoresistance. Among many intracellular signaling proteins, protein kinase C (PKC) isoenzymes have been identified as possible targets to render tumor cells more susceptible to apoptosis and growth arrest. We review the known biology of the alpha-isoenzyme of PKC in different cancers to provide a rational approach for developing targeted therapies using PKC modulators, including aprinocarsen, an antisense oligonucleotide (ASO) against PKC-alpha.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Tionucleótidos/uso terapéutico , Animales , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias/enzimología , Oligodesoxirribonucleótidos Antisentido/farmacología , Proteína Quinasa C-alfa , Tionucleótidos/farmacología
17.
Eur J Cancer ; 30A(11): 1688-94, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7833145

RESUMEN

Epidermal growth factor (EGF) is a mitogenic peptide that binds to surface membrane receptors (EGFR) of breast cancer cells. After binding, secondary transmitter molecules are activated by tyrosine phosphorylation of the intracellular receptor domaine. The activity of the EGF/EGFR system can be modulated by a variety of chemically unrelated compounds including cytostatic agents. The purpose of our present study was to determine the effects of mitotic inhibitors on EGF receptor binding on human breast cancer cells. We found that MDA-231 and MDA-468 cells bind substantially more [125I]EGF after preincubation with docetaxel, vinblastine and vincristine. This effect was concentration- and time-dependent, reaching a maximum at 3000 ng/ml and 48 h incubation for docetaxel, and 100 ng/ml and 48 h incubation for vinca alcaloids. Subsequent experiments showed an increase in the rate of EGF binding as well as maximal binding capacity. Scatchard analysis of binding experiments under equilibrium conditions indicated that this was due to an increase in the number of apparent EGF binding sites. Modulation of EGF receptor binding by docetaxel, vinblastine, and vincristine was not detectable when isolated membranes were used, indicating that intact cytoplasmatic mechanisms are required for the upregulation of EGF receptors.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Paclitaxel/análogos & derivados , Taxoides , Vinblastina/farmacología , Vincristina/farmacología , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Docetaxel , Relación Dosis-Respuesta a Droga , Receptores ErbB/efectos de los fármacos , Femenino , Humanos , Cinética , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Paclitaxel/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
18.
Eur J Cancer ; 29A(3): 423-5, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8398345

RESUMEN

SR 4233 (3-amino-1,2,4-benzotriazine 1,4-dioxide) is a novel bioreductive agent selectively toxic to hypoxic cells. It is active as a radiation sensitiser in vitro. Using a human tumour cloning system we have studied the effects of SR 4233 against freshly explanted human tumour specimens under hypoxic and non-hypoxic culture conditions. For hypoxic conditions, final concentrations of SR 4233 of 10.0-500 mumol/l were used in short-term (1 h) exposure experiments. Final concentrations in non-hypoxic experiments ranged from 10 to 1350 mumol/l. 25 tumour specimens were tested under each culture condition. Of those, 14 (56%) were evaluable. The most common tumour types recruited included ovarian, non-small cell lung, and breast cancer. A moderate concentration-dependent increase in the frequency of inhibited tumour specimens under non-hypoxic conditions was observed with zero out of 10 sensitive specimens at 10 mumol/l as compared with five out of 14 (36%) sensitive specimens at 500 mumol/l (P < 0.02). However, when hypoxic conditions were used SR 4233 had a profound antitumour activity, (two out of 14 specimens sensitive at 10 mumol/l compared with 10 out of 10 specimens sensitive at 500 mumol/l, P < 0.00005). We conclude that SR 4233 is active against tumour colony-forming units in vitro and that its antitumour activity is greatly increased against hypoxic tumour cells.


Asunto(s)
Antineoplásicos/farmacología , Triazinas/farmacología , Ensayo de Tumor de Célula Madre , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Oxígeno , Fármacos Sensibilizantes a Radiaciones/farmacología , Tirapazamina
19.
Eur J Cancer ; 35(6): 1009-13, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10533486

RESUMEN

E91 (17 beta-[N-[N'-(2-chloroethyl)-N'-nitrosocarbamoyl]-L-alanyl]-5 alpha-dihydrotestosterone) (CNC-ala-DHT) is a newly synthesised alkylating compound consisting of N-[N'-(2-chloroethyl)-N'-nitrosocarbamoyl]-L-alanine (CNC-ala) as the alkylating moiety and of 5 alpha-dihydrotestosterone (DHT) as a steroid carrier molecule. We studied the antitumour activity of E91 (final concentrations: 0.1, 1, 10 and 30 mumol/l) against freshly explanted human tumours, using an in vitro soft agar cloning system. A total of 54 tumour samples was evaluated using 1 h-exposure and 51 tumour specimens were studied using a continuous exposure for 21-28 days. In addition, the compound's activity was compared with other clinically used anticancer agents. After short-term exposure, 49 of 53 evaluable specimens (92%) had adequate colony formation, as compared with 49 of 50 (98%) after long-term exposure. After short-term exposure, E91 exhibited only marginal antitumour activity. However, in long-term exposure experiments, E91 had marked and concentration-dependent antitumour activity (P < 0.001). At concentrations of > 10 mumol/l, E91 was as active as the other clinically used antineoplastic agents and at 30 mumol/l, E91 was significantly more active than 5-fluorouracil (P = 0.041). E91 showed activity against a wide spectrum of tumour types. The highest activity was observed against colorectal carcinomas (3/4 tumour specimens inhibited at 30 mumol/l). Sensitivity was also high remarkable in breast cancer specimens with 3/6 specimens inhibited at 30 mumol/l. In vitro myelotoxicity was less than that of doxorubicin. At 30 mumol/l, E91 induced a reduction of colony forming units-granulocyte macrophage (CFU-GM) to only 53% of control and of CFU-GEMM to 20% of control. We conclude that because of broad activity and reduced myelotoxicity further clinical development of E91 appears warranted.


Asunto(s)
Antineoplásicos/uso terapéutico , Dihidrotestosterona/análogos & derivados , Células Madre Hematopoyéticas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Compuestos de Mostaza Nitrogenada/uso terapéutico , Compuestos de Nitrosourea/uso terapéutico , Testosterona/análogos & derivados , División Celular/efectos de los fármacos , Dihidrotestosterona/uso terapéutico , Humanos , Neoplasias/patología , Testosterona/uso terapéutico , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
20.
Eur J Cancer ; 37(13): 1642-7, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11527690

RESUMEN

A phase II study was performed to evaluate 1-(2-tetrahydrofuryl)-5-fluorouracil and uracil (UFT) and leucovorin as first-line chemotherapy in European patients with advanced gastric cancer. From 38 patients, 25 were evaluable for response and 36 for toxicity. Patients received UFT at 300 mg/m(2)/day for 28 days, every 35 days and leucovorin at 90 mg/day on an identical schedule. Overall response rate was 10.5% (95% confidence interval (CI): 3.7-22.5%) in intent-to-treat analysis and 16% (95% CI: 5.7-33%) in evaluable patients. Grade 3-4 common toxicity criteria (CTC) toxicities were diarrhoea (28%; 10/36), nausea (11%; 4/36), vomiting (8%; 3/36) and asthenia (11%; 4/36). 23 patients in 44% (42/96) of the courses had to skip days of treatment due to toxicity or to non-compliance. In conclusion, UFT+leucovorin has a definitive, but low, efficacy in advanced gastric cancer patients. Toxicities were mainly gastrointestinal and treatment needs to be withheld if grade 2 diarrhoea occurs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Femenino , Humanos , Leucovorina/administración & dosificación , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Análisis de Supervivencia , Tegafur/administración & dosificación , Resultado del Tratamiento , Uracilo/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA