Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Chemother ; 19(5): 546-53, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18073154

RESUMEN

The aim of this study was to investigate the antiproliferative effects of C-1311 (Symadex), a member of the imidazoacridinone family, in human colorectal cancer cells. In the in vitro screen, C-1311 led to the most prominent growth inhibition in HT29, HCT116, and COLO205 cell lines when compared to oxaliplatin, CPT-11, DFUR, 5-FU and capecitabine. The GI(50)values for C-1311 ranged from 0.12 to 0.83 microM and the TGI concentrations (resulting in total growth inhibition) were 6- to 13-fold lower than those of other agents. In the hollow fiber assay in vivo, C-1311 caused 77% growth inhibition of HT29 in the intraperitoneal site as compared to paclitaxel (17% growth inhibition). In the subcutaneous site, C-1311 produced 57% growth inhibition while paclitaxel showed no cell growth inhibition effects. This unique cytotoxicity profile of C-1311 warrants further investigation and supports its clinical development in colon cancer patients. Symadex (C-1311) is currently in phase 2 clinical trials.


Asunto(s)
Aminoacridinas/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Aminoacridinas/química , Animales , Antineoplásicos/química , Camptotecina/análogos & derivados , Camptotecina/farmacología , Capecitabina , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos Fase II como Asunto , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacología , Células HT29 , Humanos , Técnicas In Vitro , Irinotecán , Ratones , Ratones Desnudos , Compuestos Organoplatinos/farmacología , Oxaliplatino , Paclitaxel/farmacología
2.
J Natl Cancer Inst ; 82(16): 1321-6, 1990 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-2143234

RESUMEN

Since our original proposal 4 years ago, considerable support has evolved for the concept of pharmacologically guided dose escalation in phase I clinical trials with new anticancer drugs. The original focus has been broadened to develop additional links between preclinical testing and phase I clinical trials. Recent experiences with very lengthy phase I trials for at least eight drugs have provided particular impetus for this project. The original pharmacodynamic hypothesis for the proposal was equal toxicity at equal plasma levels. Specifically, two facets of the concept were that (a) dose-limiting toxicity correlates with, and in turn is predicted by, drug concentrations in plasma and (b) that the quantitative relationship between toxicity and drug exposure, as measured by plasma drug concentration times time (C x T), holds across species. If true, this hypothesis would suggest that dose escalations in humans could be safely based on measurement of drug levels in plasma, rather than on empirical escalation schemes. In addition to the collection of a larger retrospective data base to validate this hypothesis, practical results have already been achieved. In two studies sponsored by the National Cancer Institute (NCI), the escalation pattern was prospectively modified on the basis of measurements of drug levels in plasma. In addition, for three NCI-sponsored drugs, more careful matching of schedules between clinical and preclinical testing produced entry doses that were up to 25 times higher than doses used in standard procedures. Consequently, the phase I trials for each drug were completed with a savings of 12-24 months. As a result of work in both the United States and Europe, a substantial collection of data now demonstrates that coordination with preclinical pharmacology and toxicology studies can save both time and resources in early clinical trials without a loss of safety.


Asunto(s)
Antineoplásicos/administración & dosificación , Evaluación de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales , Animales , Antineoplásicos/sangre , Antineoplásicos/toxicidad , Esquema de Medicación , Humanos , Metaanálisis como Asunto , Ratones , Especificidad de la Especie , Factores de Tiempo
3.
J Natl Cancer Inst ; 86(4): 273-80, 1994 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-8158681

RESUMEN

BACKGROUND: 9-Methoxypyrazoloacridine (PZA) is an anticancer agent that shows selectivity of action for carcinomas over leukemias. It also has nearly equal potency against cycling and quiescent or hypoxic and normoxic target cells. Phase I trials of PZA in humans are nearing completion. PURPOSE: This study was conducted to determine (a) if PZA is directly inhibitory to hematopoietic cells and, if it is, to characterize the inhibition pharmacodynamically, (b) whether species-specific differences in direct toxicity could explain differences in myelosuppression in mice, dogs, and humans, and (c) whether in vitro data correlate with in vivo myelosuppression data. METHODS: In vitro clonogenic assays of hematopoietic progenitors of myeloid and erythroid lineages from human, canine, and murine femoral marrow were used to measure the direct toxicity of PZA. Results from these assays were compared on an area-under-the-curve (AUC) basis to clinical myelosuppression data. RESULTS: On the basis of maximum tolerated concentrations, canine hematopoietic progenitors are most susceptible to PZA, followed by human and then murine progenitors. We found no difference in susceptibility to PZA toxicity between the human progenitors of myeloid and erythroid lineages. Both concentration and duration of exposure contribute to the in vitro toxicity of PZA. In contrast to antimetabolites, the in vitro toxicity of PZA could be minimized at a given AUC by lowering drug concentration and prolonging the period of exposure. On an AUC basis, the in vitro data are consistent with limited in vivo myelosuppression data from preclinical models and correlate with neutropenia data from a phase I trial. CONCLUSIONS: PZA directly inhibits hematopoietic progenitors, an action that is responsible for the myelosuppression observed in humans. Human marrow appears able to compensate for the loss of up to 35% of its myeloid progenitors, in that peripheral neutrophil counts remain unchanged at that level of loss. Although in vivo studies show that prolonged infusion reduces myelosuppression at a given total dose in both rodent and canine models, pharmacokinetic differences make it unlikely that this approach will benefit human patients. IMPLICATIONS: The in vitro data quantitatively predict the AUCs at maximum tolerated dose in preclinical models and human patients. Thus, in vitro clonogenic assays of myelotoxic agents can provide data that make both preclinical toxicology testing and clinical trial planning and interpretation more efficient and accurate.


Asunto(s)
Acridinas/toxicidad , Antineoplásicos/toxicidad , Médula Ósea/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Pirazoles/toxicidad , Animales , Células de la Médula Ósea , Células Cultivadas , Perros , Humanos , Recuento de Leucocitos/efectos de los fármacos , Ratones
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.
Cancer Res ; 50(13): 4038-43, 1990 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-2354454

RESUMEN

The effects of L-buthionine sulfoximine (L-BSO) and L-phenylalanine mustard (L-PAM), alone and in combination, on human and murine marrow were explored using in vitro clonogenic assays to establish whether enhanced myelotoxicity might limit the clinical utility of this potent chemotherapeutic combination. One-h exposure to L-PAM produced significant concentration-dependent colony inhibition, with 70% inhibitory concentration (IC70) values ranging from 4.5 to 7.2 microM for all hematopoietic progenitors assayed. The combination of L-PAM plus 4500 microM L-BSO for 1 h did not effectively alter the IC70 values derived for L-PAM alone. In studies where marrow cells were pretreated with L-BSO for 4 h and then L-PAM for 1 additional h, the IC70 values were decreased in both murine and human marrow progenitors compared to the L-PAM control, suggesting modest potentiation of myelotoxicity. The potentiation is not so significant as to preclude human studies with this combination. One- to 5-h exposure of marrow cells from both species to 4500 microM L-BSO was only mildly myelotoxic, producing colony reductions of 22-49%. However, continuous exposure to L-BSO produced concentration-dependent colony inhibition, with IC70 values of 70, 84, and 43 microM for murine colony-forming units-granulocyte/macrophage, blast-forming units-erythroid, and colony-forming unit-erythroid, respectively.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Melfalán/farmacología , Metionina Sulfoximina/análogos & derivados , Animales , Butionina Sulfoximina , Ensayo de Unidades Formadoras de Colonias , Sinergismo Farmacológico , Femenino , Humanos , Melfalán/toxicidad , Metionina Sulfoximina/farmacología , Metionina Sulfoximina/toxicidad , Ratones , Factores de Tiempo
6.
Cancer Res ; 49(19): 5385-91, 1989 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-2766304

RESUMEN

Intravenous doses of buthionine sulfoximine (BSO, NSC 326231), an inhibitor of glutathione synthesis, were eliminated rapidly from mouse plasma in a biexponential manner. The initial phase of the plasma concentration versus time curve had a half-life of 4.9 min and accounted for 94% of the total area under the curve. The half-life of the terminal phase of the curve was 36.7 min and the area accounted for only 6% of the total area under the curve. Plasma clearance of BSO was 28.1 ml/min/kg and the steady state volume of distribution was 280 ml/kg. The oral bioavailability of BSO, based on plasma BSO levels, was extremely low. However, comparable glutathione depletion was apparent after i.v. and p.o. doses of BSO, suggesting a rapid tissue uptake and/or metabolism of BSO. Therefore, due to the rapid elimination of BSO from mouse plasma, plasma drug levels do not directly correlate with BSO-induced tissue glutathione depletion. Administration of multiple i.v. doses of BSO to male and female mice resulted in a marked 88% depletion of liver glutathione at doses of 400-1600 mg/kg/dose. Toxicity of i.v. administered BSO was limited to a transient depression of peripheral WBC levels in female mice given six doses of 1600 mg/kg. Multiple i.v. doses of BSO of up to 800 mg/kg/dose (every 4 h for a total of six doses) did not alter the toxicity of i.v. administered melphalan. However, multiple doses of 1600 mg/kg/dose of BSO did potentiate histopathological evidence of melphalan-induced bone marrow toxicity in 30% of the mice and, additionally, the combination of BSO and melphalan produced renal tubular necrosis in 80% of the male mice. The potentiation of melphalan induced toxicity did not appear to be related to GSH depletion, since: quantitatively similar amount of GSH depletion occurred at lower dose of BSO without any increase in melphalan toxicity.


Asunto(s)
Melfalán/administración & dosificación , Metionina Sulfoximina/análogos & derivados , Administración Oral , Animales , Butionina Sulfoximina , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Glutatión/sangre , Semivida , Inyecciones Intravenosas , Hígado/metabolismo , Metionina Sulfoximina/administración & dosificación , Metionina Sulfoximina/sangre , Metionina Sulfoximina/farmacocinética , Metionina Sulfoximina/farmacología , Metionina Sulfoximina/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Premedicación
7.
Exp Hematol ; 18(7): 832-6, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2379548

RESUMEN

Three nucleoside analogues, 2',3'-dideoxyadenosine (ddA), 2',3'-dideoxyinosine (ddI), and 2',3'-dideoxycytosine (ddC), were evaluated for their potential myelotoxic effects to normal human hematopoietic progenitor cells. The myeloid (granulocyte-monocyte colony-forming units, CFU-gm) and erythroid (erythroid burst-forming units, BFU-e: and erythroid colony-forming units, CFU-e) committed progenitor cells were exposed to the agents for a 1-h period prior to culture in a microcapillary assay or continuously exposed during the entire culture period. Both ddA and ddI (100 microM) were mildly toxic (less than 50% colony inhibition) to human CFU-gm, BFU-e, and CFU-e following either 1-h or continuous exposures. Marrow progenitor sensitivities to ddA and ddI were indistinguishable. Colony inhibition ranged from 47% to 67% for 1-h ddC exposure (100 microM), values that were comparable to ddA and ddI. Continuous exposure to ddC was highly myelotoxic to human hematopoietic progenitors, with concentrations of 10 and 100 microM suppressing colony formation by 79%-92% and 93%-97%, respectively. These results demonstrate that 1-h and continuous exposures to ddA and ddI were similarly myelotoxic to human hematopoietic cells, whereas a 1-h exposure to ddC was equivalent to ddA and ddI, yet continuous ddC exposure was extremely toxic to marrow cell progenitors.


Asunto(s)
Médula Ósea/efectos de los fármacos , Didesoxinucleósidos/toxicidad , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Precursoras Eritroides/efectos de los fármacos , Humanos , Técnicas In Vitro
8.
Cancer Chemother Pharmacol ; 34(2): 103-8, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8194161

RESUMEN

We studied the toxicity of a new experimental anticancer drug, cyclopentenyl cytosine (CPE-C), to human and murine hematopoietic progenitor cells in vitro. Due to CPE-C's in vivo myelotoxicity, it was important to characterize its potential adverse effects on human marrow cells during preclinical development of the drug. Marrow cells were exposed to CPE-C for either 1 h prior to addition in clonal assays or continuously during their culture period. The inhibitory effects of CPE-C on myeloid (CFU-gm) and erythroid (CFU-e, BFU-e) colony formation were concentration- and time-dependent, with continuous CPE-C exposure being significantly more inhibitory than 1-h exposure. The results of both exposure experiments were combined to investigate colony inhibition as a function of overall drug exposure (concentration x time, AUC) and data analyzed by the nonlinear Emax equation. Human and murine CFU-gm had similar AUC-response curves and IAUC70 values (i.e., AUC at 70% colony inhibition) of 40.8 and 41.9 microM h, respectively. In contrast, murine CFU-e and BFU-e were more sensitive to CPE-C, having lower IAUC70 values (both, 21.1 microM h) than human CFU-e and BFU-e (107.8 and 33.0 microM h, respectively). This difference was most prominent with the late erythroid progenitor, CFU-e, in that the human cells were 5 times more resistant to inhibition by CPE-C. CPE-C was myelotoxic in vitro to human and murine marrow cells and toxicity correlated with overall drug exposure.


Asunto(s)
Antineoplásicos/toxicidad , Médula Ósea/efectos de los fármacos , Citidina/análogos & derivados , Animales , Células de la Médula Ósea , Carmustina/toxicidad , Recuento de Células/efectos de los fármacos , Células Clonales/citología , Células Clonales/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Citidina/toxicidad , Relación Dosis-Respuesta a Droga , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones , Factores de Tiempo
9.
Cancer Chemother Pharmacol ; 28(1): 33-8, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-2040031

RESUMEN

The acute toxicities of the cellular differentiating agent hexamethylene bisacetamide (HMBA) in humans and animals include CNS toxicity (agitation, somnolence, seizures, hallucinations) and an anion-gap metabolic acidosis. N-Acetyl-1,6-diaminohexane (NADAH), the first metabolite of HMBA, is as active as the parent compound in causing differentiation of leukemic cells in vitro, whereas 6-acetamidohexanoic acid (6AcHA), which is formed by the oxidation of NADAH in the presence of monoamine oxidase (MAO) and aldehyde dehydrogenase, is inactive. To test whether the inhibition of MAO blocks the production of an inactive and possibly toxic HMBA metabolite (6AcHA) or increases the amount of active compounds (HMBA + NADAH) in vivo, we investigated the effect of the MAO inhibitor isocarboxazid on the metabolism and toxicity of HMBA in beagle dogs. Two groups of dogs, composed of one male and one female dog per group, were used in the study. One group received isocarboxazid (3.3 mg/kg p.o. q8h x 9) beginning at 24 h before the initiation of a 48-h i.v. infusion of HMBA (40 mg kg-1 h-1), whereas the other received placebo in an identical fashion prior to the start of an identical HMBA infusion. The mean plasma steady-state concentration (css) of HMBA was 0.91 mM in dogs given HMBA and isocarboxazid as opposed to 0.78 mM in those given HMBA and placebo. As measured spectrophotometrically, plasma MAO activity was inhibited by 86% +/- 3% in dogs receiving isocarboxazid. Gas chromatography/mass spectrometry detected 6AcHA in the plasma of animals that were given placebo but not in the plasma of dogs that received isocarboxazid. Gas chromatographic analysis of urine samples revealed that the total amount of 6AcHA and of NADAH excreted in urine was 8 times less and 3 times greater, respectively, in isocarboxazid-treated dogs than in animals that received HMBA and placebo. One dog was excitable after the initial two doses of isocarboxazid and developed seizures at the end of the HMBA infusion. Another dog was agitated during treatment with HMBA and isocarboxazid. No CNS toxicity occurred in animals that were treated with HMBA and placebo. We conclude that isocarboxazid inhibits the production of 6AcHA in vivo, thus supporting the involvement of MAO in HMBA metabolism. Because the combination of HMBA and isocarboxazid produces CNS toxicity, 6AcHA is probably not the neurotoxic agent in dogs.


Asunto(s)
Acetamidas/metabolismo , Isocarboxazida/farmacología , Acetamidas/farmacocinética , Acetamidas/toxicidad , Administración Oral , Aminocaproatos , Ácido Aminocaproico/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Cromatografía de Gases , Perros , Interacciones Farmacológicas , Femenino , Cromatografía de Gases y Espectrometría de Masas , Infusiones Intravenosas , Masculino , Monoaminooxidasa/sangre
10.
Cancer Chemother Pharmacol ; 39(1-2): 143-9, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8995512

RESUMEN

Bizelesin is a potent synthetic derivative of the anticancer agent CC-1065 that preferentially alkylates and binds the minor grove of DNA. Preclinical animal studies have found bizelesin to be more toxic to beagle dogs than to rodents and that myelosuppression was the dose-limiting toxicity. This toxicity was dose- and time-dependent in all species. Due to the significant difference in the in vivo myelotoxicity between species, it was important to determine which one most closely resembles humans on a pharmacodynamic basis. Therefore, hematopoietic clonal assays were utilized to evaluate the effects of bizelesin on granulocyte-macrophage (CFU-gm) colony formation. Marrow cells were exposed in vitro to bizelesin (0.001-1000 nM) for 1 or 8 h and then assayed for colony formation. There was a 3-log difference in drug concentration at which 100% colony inhibition occurred (1 or 8 h) for murine CFU-gm versus human or canine CFU-gm. The IC70 value after an 8-h bizelesin exposure for human CFU-gm (0.006 +/- 0.002 nM) was 2220-times lower than for murine CFU-gm (13.32 +/- 8.31 nM). At any given concentration, an 8 h drug exposure resulted in greater colony inhibition than a 1 h exposure for all species (P < 0.05). Increasing exposure time from 1 to 8 h increased toxicity to human and canine CFU-gm much more than to murine CFU-gm. The clinically formulated drug solution was a more potent inhibitor of human colony formation than drug dissolved in DMSO. The IC70 value after a 1-h exposure was 1.7 times lower for human CFU-gm with formulated bizelesin (0.106 +/- 0.105 nM) than bulk drug in DMSO (0.184 +/- 0.044 nM). The results of these in vitro clonal assays were qualitatively consistent with those seen in whole animal studies, suggesting that bizelesin will be a potent myelosuppressive agent in the clinic. Since the dose-limiting toxicity in preclinical models is myelosuppression and the in vitro sensitivity of human and canine CFU-gm is similar, the canine maximum tolerated dose (MTD) is better than the murine MTD to determine a safe starting dose for phase I clinical trials.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/toxicidad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Indoles/farmacología , Indoles/toxicidad , Urea/análogos & derivados , Animales , Médula Ósea/efectos de los fármacos , Perros , Duocarmicinas , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Especificidad de la Especie , Células Madre/efectos de los fármacos , Urea/farmacología , Urea/toxicidad
11.
Food Chem Toxicol ; 21(3): 305-11, 1983 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-6683227

RESUMEN

Diets containing 25,000 (2.5%) or 50,000 ppm (5.0%) agar, guar gum, gum arabic, locust-bean gum or tara gum were fed to groups of 50 male and 50 female F344 rats and B6C3F1 mice for 103 wk. Separate groups of 50 rats and 50 mice of each sex served as controls for each study. There were no significant differences in survival between any of the dosed groups of rats or mice and their respective control groups. Depressions in body-weight gain greater than 10% for dosed groups relative to their respective control groups were observed for male (low dose only) and female mice fed diets containing agar, female mice fed diets containing guar gum (high dose only), male mice fed diets containing locust-bean gum (high dose only) and male and female mice fed diets containing tara gum (high dose only). Depressions in body-weight gain greater than 5% were observed for female rats fed diets containing agar, guar gum or gum arabic. There were no histopathological effects associated with the administration of the test materials. Under the conditions of these bioassays, none of the five polysaccharides was carcinogenic for F344 rats or B6C3F1 mice of either sex.


Asunto(s)
Agar/toxicidad , Galactanos/toxicidad , Goma Arábiga/toxicidad , Mananos/toxicidad , Polisacáridos/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos , Neoplasias Experimentales/inducido químicamente , Gomas de Plantas , Ratas , Ratas Endogámicas F344
12.
Br J Cancer ; 97(1): 58-64, 2007 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-17551498

RESUMEN

Xanafide, a DNA-intercalating agent and topoisomerase II inhibitor, has previously demonstrated comparable cytotoxicity to the parent drug amonafide (NSC 308847). The current study was conducted to investigate further the anti-proliferative effects of xanafide in human breast cancer cell lines, in vitro and in vivo. The in vitro activity of xanafide against MCF-7, MDA-MB-231, SKBR-3 and T47D cell lines was compared to that of paclitaxel, docetaxel, gemcitabine, vinorelbine and doxorubicin. In MCF-7, xanafide demonstrated comparable total growth inhibition (TGI) concentrations to the taxanes and lower TGI values than gemcitabine, vinorelbine and doxorubicin. MCF-7 (oestrogen receptor (ER)+/p53 wild-type) was the most sensitive cell line to xanafide. MDA-MB-231 and SKBR-3 exhibited similar sensitivity to xanafide. T47 D (ER+/p53 mutated), showed no response to this agent. The in vivo activity of xanafide was further compared to that of docetaxel in MCF-7 and MDA-MB-231 cell lines using the hollow fibre assay. Xanafide was slightly more potent than docetaxel, at its highest dose in MCF-7 cell line, whereas docetaxel was more effective than xanafide in MDA-MB-231 cell line. Our results show that there is no relationship between sensitivity of these cell lines to xanafide and cellular levels of both isoforms of topoisomerase II and suggest that ER and p53 status and their crosstalk may predict the responsiveness or resistance of breast cancer patients to xanafide.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Imidas/uso terapéutico , Isoquinolinas/uso terapéutico , Naftalimidas/uso terapéutico , Adenina , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Desnudos , Organofosfonatos
13.
Biochem Genet ; 13(7-8): 447-56, 1975 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-810135

RESUMEN

The specific activity (k'1) and concentration of red blood cell catalase from four inbred strains of mice (BALB/c, C57BL, C57BL/6, and NBL) were measured to determine the mechanisms responsible for interstrain variations in enzyme activity. The specific activities of RBC catalase in NBL and the C57BL sublines are equal (2.5 x 10(7) M-1 sec-1), while that of BALB/c (4.0 x 10(7) M-1 sec-1) is 67% greater. The relative concentration of catalase is approximately 30% lower in NBL erythrocytes compared to the other three strains. The activity of BALB/c RBC catalase is due to a high k'1 coupled with a high intracellular concentration; RBC catalase activity in the C57BL sublines is the result of a low k'1 and high concentration. A low k'1 and a low concentration are responsible for the low catalase activity levels found in NBL erythrocytes.


Asunto(s)
Catalasa/sangre , Eritrocitos/enzimología , Biología Molecular , Animales , Catalasa/inmunología , Inmunodifusión , Ratones , Ratones Endogámicos , Pruebas de Precipitina , Especificidad de la Especie
14.
Cancer Treat Rep ; 70(1): 65-72, 1986 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3943115

RESUMEN

The evolution of the Division of Cancer Treatment's preclinical toxicology protocols over the last decade, a description of the current protocol, and the progress of this protocol regarding quantitative and qualitative relationships to current clinical findings are presented in this report. Data are reviewed for seven experimental antineoplastic drugs. Preliminarily, the following conclusions can be made from preclinical and clinical toxicology experience with these first seven drugs. The 1/10 MELD10 establishes a safe human starting dose. Six of seven drugs could have started at 1/10 MELD10 with no adverse effects. Toxicity data from the beagle dog did not effectively predict whether the 1/10 MELD10 dose level represents a hazard to humans. Clinical dose escalation procedures can not be efficiently predicted from currently acquired preclinical information. Toxicity studies in dogs were effective in disclosing the human dose-limiting toxic effects and approximated the maximally tolerated dose for six of seven drugs. In the exceptional case, the dose level predicted for human toxicity was grossly underestimated in both experimental species. We conclude that the preclinical toxicity data from the beagle dog are valuable in predicting the potential risk to humans, even though the safety of the entry-level doses is occasionally underpredicted.


Asunto(s)
Antineoplásicos/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Evaluación de Medicamentos/métodos , Animales , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Humanos , Dosificación Letal Mediana , Ratones , Especificidad de la Especie
15.
Biochem Genet ; 14(1-2): 59-66, 1976 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1259704

RESUMEN

Starch gel electrophoresis of kidney catalase in inbred strains C3H and C57BL/6, their F1 hybrid, and first and second backcross generations demonstrated that single-component (type A) v. multiple-component (type B) electrophoretic patterns are controlled by a single locus. The type A electrophoretic pattern is dominant. Twenty-five inbred strains of mice were classified according to their kidney catalase electrophoretic pattern. The data indicate that the segregating genetic factor determines a specific substance in the type A kidney which affects the electrophoretic mobility of catalase. A comparison of the F1 hybrid enzyme with a 1:1 mixture of C3H and C57BL/6 enzyme showed that the alteration of electrophoretic mobility is the result of posttranslational modification of the catalase molecule. An association of kidney catalase electrophoretic pattern and the H-2kappa haplotype indicates that the locus controlling the electrophoretic pattern is most likely located on chromosome 17 in close proximity to the H-2 complex.


Asunto(s)
Catalasa/análisis , Isoenzimas/análisis , Riñón/enzimología , Animales , Cruzamientos Genéticos , Electroforesis en Gel de Almidón , Ratones , Ratones Endogámicos
16.
Invest New Drugs ; 9(2): 149-57, 1991 May.
Artículo en Inglés | MEDLINE | ID: mdl-1874599

RESUMEN

The in vitro myelotoxic potentials of three investigational antitumor agents, Fostriecin, Hepsulfam and pyrazine diazohydroxide (PZDH), were evaluated utilizing clonogenic assays. Human and murine marrow cells were exposed to each drug for 1 hr prior to culture in microcapillary (human) or Petri dish (murine) assays. Fostriecin (0.22-220 microM), Hepsulfam (0.34-340 microM) and PZDH (0.68-680 microM) inhibited myeloid (CFU-gm), erythroid (BFU-e, CFU-e) and megakaryocytic (CFU-meg) colony formation in a concentration-dependent manner. CFU-e from both species were more sensitive to Fostriecin than the other progenitors and murine cells more sensitive overall to Fostriecin than their human counterparts. Murine CFU-e were also more sensitive to Hepsulfam than human CFU-e, with CFU-gm and BFU-e being similarly affected in both species. Human BFU-e were greatly inhibited by PZDH, whereas murine BFU-e were relatively resistant to its toxic effects. Fostriecin was the most toxic of the three antitumor agents, with PZDH the least toxic.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Antineoplásicos/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Alquenos/toxicidad , Animales , Médula Ósea/efectos de los fármacos , Células de la Médula Ósea , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias/métodos , Drogas en Investigación/toxicidad , Humanos , Ratones , Polienos , Pirazinas/toxicidad , Pironas , Ácidos Sulfónicos/toxicidad
17.
Br J Haematol ; 80(4): 437-45, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1316140

RESUMEN

The myelotoxicities of three antiretroviral agents, 3'-azido-3'-deoxythymidine (AZT), carbovir (CBV) and 2',3'-didehydro-2',3'-dideoxythymidine (d4T), were evaluated in vitro with normal human and murine haematopoietic progenitor cells. These studies demonstrated that continuous AZT exposure was more inhibitory to human and murine colony formation than 1 h exposure, with murine and human progenitors similarly inhibited by continuous AZT exposure. These in vitro results on AZT's myelotoxicity correlate with both human and murine in vivo studies. CBV was only moderately toxic to human and murine cells following either 1 h or continuous exposure, with human and murine progenitors similarly suppressed by continuous CBV exposure. 1 h d4T exposure was less toxic to both human and murine marrow cells than continuous exposure and both species were equivalently inhibited when continuously exposed to d4T. In general, CBV was the least toxic agent to human and murine haematopoietic cells and AZT the most toxic. The study establishes CBV and d4T as less myelotoxic agents to human and murine haematopoietic progenitor cells in vitro than AZT which therefore could be considered as alternatives to AZT for the treatment of HIV infection.


Asunto(s)
Didesoxinucleósidos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Zidovudina/farmacología , Animales , Antivirales/farmacología , Células de la Médula Ósea , División Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos , Estavudina
18.
Cancer Treat Rep ; 70(12): 1415-21, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3791254

RESUMEN

Flavone acetic acid is a novel structure which exhibits an interesting spectrum of antitumor activity in preclinical studies. It has little antitumor activity in the leukemias and pronounced antitumor activity in solid tumors. Preclinical therapeutic, toxicologic, and pharmacokinetic studies are summarized and considered together to introduce the concept of a therapeutic window of effective plasma concentrations and effective exposure times in attempts to maximize therapeutic effects and minimize toxic effects. Plasma concentrations, predicted to fall from 600 to 100 micrograms/ml over 10 hours resulting from 267 mg/kg ip bolus injections in mice are curative to sc implanted colon 38. Doses of 356 mg/kg and higher cause acute lethality in many mice. Iv doses cause acute lethality in mice more frequently than ip doses, which suggests a peak toxic effect. However, iv infusions in mice, which also can be curative to colon 38, can also result in a lethal effect, although more delayed, even though the predicted plasma concentrations are much below the peak plasma concentrations that appear to be necessary for acute lethality. Plasma concentrations, 100 to 600 micrograms/ml predicted to result from single doses that are therapeutic and not acutely lethal in mice, if maintained by infusion in dogs for 28 hours or longer result in delayed lethality. We conclude that relatively high plasma concentrations (greater than 100 micrograms/ml) are needed for therapeutic activity with this antitumor agent and that lethality can result from two distinctly different causes. An acute lethality can result from an excessively high peak plasma concentration (greater than 600 micrograms/ml). A delayed lethality can result from a too-long exposure (greater than 24 hrs) at therapeutically effective plasma concentrations (100-600 micrograms/ml). We also note that unexpected kinetic differences exist among the mouse, dog, and man. Whereas usually with antitumor agents plasma clearances are proportional to body surface area, and hence faster in small species, quite the opposite is true with flavone acetic acid. Mice exhibit a slower plasma clearance relative to dogs and man.


Asunto(s)
Antineoplásicos/toxicidad , Flavonoides/toxicidad , Neoplasias Experimentales/metabolismo , Animales , Antineoplásicos/metabolismo , Química Farmacéutica , Cromatografía Líquida de Alta Presión , Perros , Relación Dosis-Respuesta a Droga , Flavonoides/metabolismo , Cinética , Ratones
19.
Ann Oncol ; 9(4): 357-64, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9636825

RESUMEN

Several clinical oncology units are studying the roles of in vitro hematotoxicology in phase I evaluations. At the same time, the European Center for the Validation of Alternative Methods (ECVAM) is supporting a validation study of the CFU-GM assay. It is important that these activities be coordinated so that high performance, optimized technical protocols are used for prospective and retrospective clinical evaluations. The EROTC, the NCI and ECVAM could provide support for these coordinated efforts. There is an opportunity for medical oncologists involved in early clinical trials to participate in the evaluation of in vitro tests and their clinical application . Fundamental to acceptance of these assays by oncologists and regulatory scientists, they must predict clinical outcome for myelosuppressive agents and then improve phase I design and performance. These achievements would justify more aggressive dose escalation schemes using guidance from in vitro studies without compromising patient safety. Success in predicting neutropenia might also stimulate the research required to understand how to predict other hematologic toxicities, such as a thrombocytopenia. The complexity of a validation study in hematotoxicology is that it seeks to predict the level of exposure that causes neutropenia, in contrast to other validation studies that have sought to classify a xenobiotic as toxic or not. It may be that the clinical relevance of a new assay is not just a yes-no answer. This important distinction came from the realization that the xenobiotic tolerance in other organ systems of the body must be the same or greater than marrow in order for myelosuppression to be a clinical consequence of exposure. Pharmacological principles of system exposure and toxicity that are integrated into the prediction model provide the links to clinical oncology. It is also important to anticipate future applications of in vitro hematotoxicology. If the maximum tolerated level of drug exposure for human hematopoietic cells can be predicted, then in vitro hematotoxicology could play an important role in new drug discovery. One concept involves screening for compounds that show efficacy at the IC level that predicts maximum tolerated exposure levels in the human. 'Therapeutic index based' drug discovery has been applied to the tallimustine family with some success.


Asunto(s)
Antineoplásicos/efectos adversos , Células de la Médula Ósea/efectos de los fármacos , Enfermedades Hematológicas/inducido químicamente , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Área Bajo la Curva , Células de la Médula Ósea/patología , Ensayos Clínicos Fase I como Asunto , Modelos Animales de Enfermedad , Perros , Humanos , Técnicas In Vitro , Ratones , Neutropenia/inducido químicamente , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Especificidad de la Especie , Células Tumorales Cultivadas
20.
Int J Cell Cloning ; 10(2): 87-93, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1545153

RESUMEN

Four nucleoside analogues, 2',3'-dideoxyinosine (ddI), 2',3'-dideoxyadenosine (ddA), 2',3'-dideoxycytosine (ddC) and 5-fluoro-2',3'-dideoxycytosine (5-F-ddC), were evaluated for their potential in vitro myelotoxic effects on normal murine hematopoietic progenitor cells. Myeloid granulocyte-macrophage colony-forming units (CFU-gm), erythroid burst-forming units (BFU-e) and colony-forming units (CFU-e) and megakaryocytic (CFU-meg) progenitors were exposed to the agents for 1 h prior to culture in Petri dish assays or continuously throughout the entire culture period. At 10 microM, both ddA and ddI were moderately toxic (2-36% colony inhibition) to murine CFU-gm, BFU-e, CFU-e and CFU-meg following either 1 h or continuous exposure. Colony inhibition for the progenitors ranged from 2-31% at 10 microM for 1 h ddC or 5-F-ddC exposure. Continuous exposure to ddC was highly myelotoxic to murine hematopoietic progenitors with 100 microM suppressing colony formation 82-89%. At the same concentration and exposure time, 5-F-ddC inhibited colony formation 56-67%. Our results demonstrate that 1 h and continuous exposures to ddA and ddI were similarly myelotoxic to murine hematopoietic cells regardless of exposure time. In contrast, continuous ddC or 5-F-ddC exposure was more toxic to murine progenitors than 1 h exposure to these agents.


Asunto(s)
Didesoxinucleósidos/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Células de la Médula Ósea , Ensayo de Unidades Formadoras de Colonias , Femenino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA