Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Br J Cancer ; 106(7): 1249-58, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22395661

RESUMEN

Erythropoiesis-stimulating agents (ESAs) increase red blood cell (RBC) production in bone marrow by activating the erythropoietin receptor (EpoR) on erythrocytic-progenitor cells. Erythropoiesis-stimulating agents are approved in the United States and Europe for treating anaemia in cancer patients receiving chemotherapy based on randomised, placebo-controlled trials showing that ESAs reduce RBC transfusions. Erythropoiesis-stimulating agent-safety issues include thromboembolic events and concerns regarding whether ESAs increase disease progression and/or mortality in cancer patients. Several trials have reported an association between ESA use and increased disease progression and/or mortality, whereas other trials in the same tumour types have not provided similar findings. This review thoroughly examines available evidence regarding whether ESAs affect disease progression. Both clinical-trial data on ESAs and disease progression, and preclinical data on how ESAs could affect tumour growth are summarised. Preclinical topics include (i) whether tumour cells express EpoR and could be directly stimulated to grow by ESA exposure and (ii) whether endothelial cells express EpoR and could be stimulated by ESA exposure to undergo angiogenesis and indirectly promote tumour growth. Although assessment and definition of disease progression vary across studies, the current clinical data suggest that ESAs may have little effect on disease progression in chemotherapy patients, and preclinical data indicate a direct or indirect effect of ESAs on tumour growth is not strongly supported.


Asunto(s)
Hematínicos/efectos adversos , Neoplasias/tratamiento farmacológico , Anemia/complicaciones , Anemia/tratamiento farmacológico , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Hematínicos/metabolismo , Hematínicos/uso terapéutico , Humanos , Metaanálisis como Asunto , Neoplasias/irrigación sanguínea , Neoplasias/complicaciones , Neoplasias/metabolismo , Receptores de Eritropoyetina/metabolismo
2.
Br J Cancer ; 99(1): 1-5, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18506149

RESUMEN

The clinical experience recently reported with epidermal growth factor receptor (EGFR)-targeting drugs confirms the synergistic interactions observed between these compounds and conventional cytotoxic agents, which were previously established at the preclinical stage. There are, however, examples of major gaps between the bench and the bedside. Particularly demonstrative is the failure of the tyrosine kinase inhibitors (TKIs) (gefitinib and erlotinib) combined with chemotherapy in pretreated nonsmall cell lung cancer patients. These discrepancies can be due to several factors such as the methodology used to evaluate TKI plus cytotoxic agent combinations in preclinical models and the insufficient consideration given to the importance of the drug sequences for the tested combinations. Recent advances in understanding the biologic basis of acquired resistance to these agents have great potential to improve their clinical effectiveness. The purpose of this review is to critically examine the experimental conditions of the preclinical background for anti-EGFR drug-cytotoxic agent combinations and to attempt to explain the gap between clinical observations and preclinical data.


Asunto(s)
Antineoplásicos/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Inhibidores de Proteínas Quinasas/farmacología
3.
Oncology ; 61 Suppl 2: 31-6, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11694785

RESUMEN

Preclinical and phase I/II studies of Herceptin demonstrated a dose-related, non-linear pharmacokinetic profile. The results of a dose-finding study supported a regimen comprising an initial intravenous (i.v.) dose of 4 mg/kg with subsequent weekly doses of 2 mg/kg. However, pharmacokinetic and safety data suggested that increased dose and reduced frequency of Herceptin administration are feasible. In addition, evidence shows that Herceptin plus paclitaxel has additive antitumor efficacy in vitro, and this regimen produces significant clinical benefits. These observations form the rationale for conducting a phase I/II trial of Herceptin (8 mg/kg initial dose, 6 mg/kg maintenance dose) plus paclitaxel, both given 3-weekly. Preliminary results are promising, with serum trough Herceptin concentrations being similar and AUC being greater than those observed with weekly Herceptin plus 3-weekly paclitaxel. Two further trials are proposed: 3-weekly Herceptin as first-line monotherapy of metastatic breast cancer; and 3-weekly Herceptin with the oral 5-fluorouracil derivative, Xeloda (capecitabine). Subcutaneous (s.c.) administration of Herceptin would further simplify administration and studies are also underway to clinically evaluate s.c. administration of Herceptin in combination with paclitaxel. With the recent development of oral taxanes, it is predicted that combinations including an oral taxane, Xeloda and either 3-weekly or possibly s.c. Herceptin may become future therapies for breast cancer patients.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Músculos Abdominales , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Neoplasias de la Mama/patología , Capecitabina , Cisplatino/administración & dosificación , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Desoxicitidina/administración & dosificación , Esquema de Medicación , Antagonismo de Drogas , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Estudios de Factibilidad , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/antagonistas & inhibidores , Semivida , Haplorrinos , Humanos , Infusiones Intravenosas , Inyecciones Subcutáneas , Metotrexato/administración & dosificación , Metástasis de la Neoplasia , Paclitaxel/administración & dosificación , Seguridad , Trastuzumab , Resultado del Tratamiento , Vinblastina/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Natl Cancer Inst ; 81(15): 1133-43, 1989 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-2664191

RESUMEN

4-Ipomeanol (IPO) is the first agent to undergo preclinical development at the National Cancer Institute (NCI) based principally on a specific biochemical-biological rationale for clinical investigation as an antineoplastic agent targeted against lung cancer. This disease-specific development of IPO was initially stimulated by observations that the compound was activated by metabolism, preferentially within the mammalian lung, specifically within bronchiolar Clara cells, and that its predominant toxicity was to the lung in most species. IPO is inactive or only minimally active against most conventional antitumor test systems. However, some human lung cancer cell lines, as well as a variety of fresh human lung tumor biopsy specimens, have been shown to be capable of mediating the in situ biotransformation of IPO to a potentially cytotoxic intermediate. In this report, the biochemistry, metabolism, preclinical pharmacology, and toxicology of IPO are reviewed and the clinical development plans for this unique and challenging new agent are presented.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Terpenos/farmacología , Animales , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Humanos , Terpenos/toxicidad
5.
J Clin Oncol ; 6(5): 906-18, 1988 May.
Artículo en Inglés | MEDLINE | ID: mdl-3284976

RESUMEN

N-methylformamide (NMF), a polar solvent, is currently being evaluated by the National Cancer Institute (NCI) as an antineoplastic agent because of its activity against colon, mammary, and lung tumor xenografts. Results from preclinical studies suggest that it has radiosensitizing, chemosensitizing, and differentiating activity. Its mechanism of action remains unknown, but may involve cellular depletion of glutathione, cell membrane changes, or modulation of proto-oncogene expression. Preclinical toxicology studies conducted in mice, rats, and beagle dogs showed reversible hepatotoxicity to be dose-limiting. Clinically, NMF is administered both orally and by intravenous (IV) injection. The bioavailability with oral administration is 90% to 95%. The highest reported plasma concentration of NMF is approximately 4 mmol/L in a patient who received a dose of 2,000 mg/m2 of IV NMF. Biphasic elimination with IV NMF is seen on both the daily for five days and weekly for 3 weeks schedule. Approximately 5% to 7% of the total administered IV dose is excreted in the urine. In phase I studies, dose-limiting toxicities included reversible hepatotoxicity, a generalized malaise syndrome, and nausea and vomiting. One partial response has been reported in the 111 patients treated on phase II trials in colorectal, head and neck, and renal carcinomas. Suggestions for the future development of this drug are presented.


Asunto(s)
Antineoplásicos/farmacología , Formamidas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Formamidas/farmacocinética , Formamidas/toxicidad , Humanos , Proto-Oncogenes Mas , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Fármacos Sensibilizantes a Radiaciones/toxicidad
6.
Cancer Treat Rep ; 70(11): 1311-9, 1986 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3533251

RESUMEN

Gallium nitrate is the anhydrate salt of the naturally occurring heavy metal. It has demonstrated antitumor activity in a variety of murine tumor models, including Walker carcinosarcoma 256, fibrosarcoma M-89, leukemia K-1964, adenocarcinoma 755, mammary carcinoma YMC, reticulum cell sarcoma A-RCS, lymphoma P1798, and osteosarcoma 124F. Preclinical studies performed in rats, rabbits, dogs, and monkeys showed the dose-limiting toxicity to be renal. The hepatic, pulmonary, gastrointestinal, hematologic, and integumentary systems were also involved. The major route of elimination is the kidneys, with 35%-71% of the infused dose excreted within 24 hours. Three phase I studies suggested the following phase II doses: 700-750 mg/m2 by short infusion, once every 2-3 weeks; 300 mg/m2/day by short infusion for 3 consecutive days, to be repeated every 2 weeks; and 300 mg/m2/day by continuous infusion for 7 consecutive days, to be repeated every 3-5 weeks. The major organ toxicity reported was renal; however, this can be adequately controlled either by hydration and osmotic diuresis or by use of continuous schedule. (Either maneuver appears to allow delivery of the recommended phase II dose with a less than 30% risk of change in serum creatinine.) In limited phase II evaluation, the drug has shown antitumor activity in patients with either refractory lymphomas or small cell lung carcinoma, with total objective response rates of 28% and 11%, respectively. In addition, it has been effective in the treatment of patients with cancer-related hypercalcemia by having an inhibitory effect on calcium reabsorption from bone. Single-agent phase II studies are planned in all major tumor types. Some are already ongoing in patients with genitourinary malignancies (renal, bladder, prostate, testicular), small cell lung carcinoma, and multiple myeloma. Metabolic studies are in progress at Memorial Sloan-Kettering Cancer Center to further elucidate the mechanism or mechanisms of the hypocalcemic effects.


Asunto(s)
Antineoplásicos/uso terapéutico , Galio/uso terapéutico , Animales , Carcinoma 256 de Walker/tratamiento farmacológico , Carcinoma 256 de Walker/enzimología , Ensayos Clínicos como Asunto , Esquema de Medicación , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Galio/efectos adversos , Galio/metabolismo , Galio/toxicidad , Humanos , Hipocalcemia/inducido químicamente , Enfermedades Renales/inducido químicamente , Cinética , Linfoma/tratamiento farmacológico , Distribución Tisular
10.
Cancer Treat Rep ; 70(3): 383-9, 1986 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-3082512

RESUMEN

The s-triazine derivatives have shown preclinical antitumor activity against several histologic types. The most widely used compound of this class in the clinic, hexamethylmelamine, has been largely restricted to oral use because of its low solubility and lack of stability in solutions suitable for parenteral administration. New analogs were sought which were soluble and stable and retained antitumor activity. Pentamethylmelamine (PMM), the monodemethylated derivative, showed these promising characteristics. Preclinical toxicology studies of PMM in mice, dogs, and monkeys showed toxic manifestations that involved the hematopoietic, lymphatic, renal, male reproductive, gastrointestinal, and nervous systems; these changes were both infusion-rate- and dose-dependent. Clinical phase I trials of PMM were performed using a variety of infusion durations and frequency schedules. The dose-limiting toxic effect common to all of these trials was protracted nausea and vomiting. In addition, some studies reported dose-limiting central nervous system manifestations in the form of agitation, drowsiness, somnolence, and even coma. Mild to moderate hematologic changes were noted. Because of the severity and frequency of the gastrointestinal and central nervous system toxic effects observed in the completed trials, no new clinical trials of PMM sponsored by the National Cancer Institute are planned. However, the interest in finding a clinically useful parenteral triazine continues.


Asunto(s)
Altretamina/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Triazinas/uso terapéutico , Altretamina/análogos & derivados , Altretamina/metabolismo , Altretamina/toxicidad , Animales , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Ensayos Clínicos como Asunto , Perros , Relación Dosis-Respuesta a Droga , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Haplorrinos , Humanos , Cinética , Ratones , Relación Estructura-Actividad
11.
Invest New Drugs ; 4(3): 279-84, 1986.
Artículo en Inglés | MEDLINE | ID: mdl-3546184

RESUMEN

A new class of marine compounds, the didemnins, with potent antitumor activity has been identified. They share the novel structure of a cyclic depsipeptide. Among three structurally related compounds, didemnin B is by far the most potent in its in vitro cytotoxicity and in vivo antitumor activity (0.001 microgram/ml inhibits the growth of L1210 leukemia cells by 50%). It also demonstrates good antitumor activity against B16 melanoma and moderate activity against M5076 sarcoma and P388 leukemia. The compound also has good antiviral and potent immunosuppressive properties. Although the precise mechanism of action for the cytotoxicity remains unknown, the agent inhibits protein synthesis more than DNA synthesis and the inhibition of protein synthesis is closely correlated with inhibition of L1210 cell growth. Toxicology studies in CD2F1 mice, Fischer 344 rats and beagle dogs reveal that major target organs are the lymphatics, gastrointestinal tract, liver and kidney. Phase I trials are currently in progress under the auspices of the National Cancer Institute.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Depsipéptidos , Animales , Antibióticos Antineoplásicos/toxicidad , Antivirales/farmacología , Ciclo Celular/efectos de los fármacos , Fenómenos Químicos , Química , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inmunosupresores/farmacología , Masculino , Neoplasias Experimentales/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Péptidos Cíclicos/toxicidad , Ensayo de Tumor de Célula Madre
12.
Cancer Treat Rev ; 12 Suppl A: 43-9, 1985 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3910221

RESUMEN

Carboplatin has been developed for clinical trials as a less nephrotoxic, less emetogenic analog of cisplatin. In preclinical tumor models it was less potent than the parent compound on a molar basis, but reduced toxicity allowed comparable antitumor doses to be given. In phase I studies its dose-limiting toxicities were reversible myelosuppression, especially thrombocytopenia. Leucopenia and anemia occurred to a lesser degree. Other reported toxicities included nausea, vomiting, malaise, myalgia, arthralgia, ototoxicity, hypomagnesemia, and proteinuria. Nausea and vomiting occurred frequently, but was much less severe than that observed with cisplatin. The incidence of serum creatinine elevations was low. The increase was usually reversible and occurred only in association with administration of aminoglycosides, or abnormal pretreatment renal function. Recommended phase II doses by schedule are: bolus every 4 weeks, 400-500 mg/m2 (560 mg/m2 in children); 24 hour continuous infusion every 4 weeks, 320-400 mg/m2; weekly bolus for 4 consecutive weeks with 2 weeks rest, 100-125 mg/m2 (175 mg/m2 in children); bolus for 5 consecutive days every 4 weeks, 77-95 mg/m2. Objective responses were observed during these phase I studies in adult patients (head and neck, breast, renal carcinomas) and children (osteosarcoma, brain stem lesions). In addition to phase II evaluations in all major tumor types, plans for phase III studies in selected tumors are underway.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/efectos adversos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Carboplatino , Cisplatino/efectos adversos , Perros , Esquema de Medicación , Evaluación de Medicamentos , Evaluación Preclínica de Medicamentos , Humanos , Cinética , Ratones , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/metabolismo
15.
Invest New Drugs ; 3(1): 71-5, 1985.
Artículo en Inglés | MEDLINE | ID: mdl-3157662

RESUMEN

Trimetrexate, a 2,4-diaminoquinazoline derivative, is a new antifol recently introduced into clinical trials. It differs from methotrexate principally in its transport (not carrier-mediated), and its intracellular retention (not polyglutamylated). Trimetrexate is active against tumors which are methotrexate-resistant on the basis of impaired transport, and has a broader range of antitumor activity in preclinical models. Animal studies predict toxicity principally to the central nervous system, gastrointestinal tract and bone marrow.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Quinazolinas/farmacología , Animales , Disponibilidad Biológica , Transporte Biológico , Células Cultivadas , Ensayos Clínicos como Asunto , ADN/biosíntesis , Perros , Evaluación Preclínica de Medicamentos , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/toxicidad , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Nucleósidos/metabolismo , Quinazolinas/metabolismo , Quinazolinas/toxicidad , Trimetrexato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA