Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Cancer Res ; 49(23): 6680-6, 1989 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-2510929

RESUMEN

The murine embryonic limb at day 14 of gestation suppresses tumor formation by melanoma cells. Conditioned media of embryonic limbs have been found cytotoxic for B16 melanoma cells. The cytotoxicity is due to the catabolism of polyamines in the limb bud extracts by an amine oxidase in the serum supplement of the culture medium. However, a polyamine oxidase activity, similar to that in adult rat liver, is also detectable in homogenates of embryonic limbs. Thus, the embryonic limb contains the necessary components to produce polyamine-derived cytotoxic metabolites, which are present at the time programmed cell death occurs. This leads to the hypothesis that injected melanoma cells are killed incidentally by the mechanism that mediates programmed cell death.


Asunto(s)
Extremidades/embriología , Melanoma Experimental/patología , Poliaminas/metabolismo , Amina Oxidasa (conteniendo Cobre)/metabolismo , Animales , Supervivencia Celular , Citotoxinas/fisiología , Ratones , Técnicas de Cultivo de Órganos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Factores de Crecimiento Transformadores/fisiología , Células Tumorales Cultivadas , Poliamino Oxidasa
3.
Cancer Res ; 52(18): 4858-65, 1992 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1516043

RESUMEN

Although regeneration-competent newts like Notophthalmus viridescens have been reported to be resistant to carcinogenesis, we have been able to induce transplantable epidermal squamous cell carcinomas with 10-20% incidence by implanting 20-methylcholanthrene s.c. into the scapular region, a tissue that cannot regenerate. As soon as 1 week after exposure to this carcinogen, malignant cells were present in the treated skin, and after 4 weeks, macroscopic tumors of infiltrating squamous carcinoma cells positive for Type IV collagenase and/or rasHa p21 had dissolved areas of the epidermal basement membrane and colonized the dermis. Analysis of Ki-67 expression revealed that these tumors grow via a high growth fraction rather than a short cell cycle time. Morphological and immunohistochemical analyses showed that these tumors caricature the biology of the renewing epidermis: the presence of basal-like cells; differentiating cells; apoptotic cells; and keratinized horn pearls with an exaggerated or overabundant stem cell compartment as compared to the differentiated cell compartment. Immunochemical analyses indicated that the squamous carcinomas arose from the epidermis rather than the mucous glands. Thus, the principle that malignant tumors caricature the process of tissue renewal originally established in rodent tumors appears to be valid down the phylogenetic tree at least to regeneration-competent amphibia. Such a broad conservation indicates that the caricature principle also holds in human tumors.


Asunto(s)
Carcinoma de Células Escamosas/patología , Células Madre Neoplásicas/patología , Salamandridae/fisiología , Neoplasias Cutáneas/patología , Animales , Carcinoma de Células Escamosas/inducido químicamente , Queratinocitos/citología , Metilcolantreno , Filogenia , Regeneración , Neoplasias Cutáneas/inducido químicamente
4.
Clin Cancer Res ; 6(6): 2474-81, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10873102

RESUMEN

Dose intensity, defined as dose administered per unit time, has emerged as a potentially important measurement of anticancer drug exposure and determinant of efficacy. There are several strategies for increasing dose intensity, one being a protracted daily dosing strategy without major dose reduction for toxicity. This strategy involves continued therapy during periods of recovery from reversible toxicity, and it inherently challenges our understanding that renewing tissues cannot repopulate (recover) in the continued presence of cytotoxic drug. We have tested this idea directly in a murine preclinical trial. Specifically, we have tested whether acutely myelotoxic doses of gemcitabine (i.p. injection, 6.0 mg/m2/day), acetyldinaline [CI-994; GOE 5549; PD 123 654; 4-acetylamino-N-(2'-aminophenyl)-benzamide, 150 mg/m2/day p.o.], and/or melphalan (i.p. injection, 7.2 mg/m2/day) can be tolerated for 28 consecutive days and whether suppressed bone marrow function recovers despite this protracted daily therapy. The three drugs all caused acute neutropenia and suppression of medullary hematopoiesis. Damage to progenitor populations exposed to acetyldinaline and gemcitabine was not as severe as that caused by melphalan, in which case absolute neutrophil count, mature progenitors (colony-forming unit granulocyte/macrophage), and immature progenitors (colony-forming unit-S) progressively declined to severely depressed levels. Marrow recovery was observed during continued daily treatment with acetyldinaline and gemcitabine but not melphalan, and marrow function completely recovered after finishing the 28-day course. Pharmacology studies proved that protracted therapy causes little, if any, change in cellular drug tolerance or systemic exposure.


Asunto(s)
Antineoplásicos/administración & dosificación , Desoxicitidina/administración & dosificación , Melfalán/administración & dosificación , Fenilendiaminas/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Antineoplásicos/efectos adversos , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Benzamidas , Recuento de Células Sanguíneas , Plaquetas/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Femenino , Melfalán/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Neutropenia/inducido químicamente , Neutrófilos/efectos de los fármacos , Fenilendiaminas/efectos adversos , Distribución Aleatoria , Células Madre/efectos de los fármacos , Factores de Tiempo , Gemcitabina
5.
Clin Cancer Res ; 7(11): 3336-42, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11705845

RESUMEN

PURPOSE: XK469 (2-[4-(7-chloro-2-quinoxalinyloxy) phenoxy]propionic acid), a synthetic quinoxaline phenoxypropionic acid derivative, has broad activity against murine tumors and is entering Phase I clinical development as a topoisomerase IIbeta inhibitor. This study investigated the underlying molecular mechanism of XK469's effects on the cell cycle. EXPERIMENTAL DESIGN: Growth inhibition, cell cycle arrest, induction of p53 and p21 mRNA and protein, and cdc2 phosphorylation and kinase activity were studied in treated cells from the H460 lung cancer line and p21 and p53 knockout cells of the HCT 116 colon cancer line. RESULTS: XK469 arrested H460 cells at G(2)-M, which was associated with cdc2 phosphorylation and decreased cdc2 kinase activity. Moreover, XK469 stabilized p53 and subsequently increased p21(WAF1/CIP1). Furthermore, HCT116 p21(-/-) cells were less sensitive than wild-type cells to XK469-induced growth inhibition, but p53(+/+) and p53(-/-) cells were equally sensitive despite the absence of p21 induction in the p53(-/-) cells. CONCLUSIONS: When considered with published data, our study suggests a complex mechanism of XK469-mediated anticancer activity involving multiple pathways, including p53-dependent and -independent G(2)-M arrest via inactivation of cdc2-cyclin B1 kinase activity.


Asunto(s)
Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Quinoxalinas/farmacología , Proteína p53 Supresora de Tumor/fisiología , Ciclo Celular/fisiología , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ciclinas/fisiología , Relación Dosis-Respuesta a Droga , Fase G2/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Mutación , Neoplasias/patología , Neoplasias/prevención & control , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
6.
Exp Hematol ; 29(3): 286-94, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11274755

RESUMEN

OBJECTIVE: The protracted administration of near-conventional daily doses of chemotherapeutic agents is a strategy to increase dose intensity and, potentially, efficacy as well. However, protracted therapy carries the risk of damage to stem cells in proliferative tissues that are not targeted by intermittent schedules. Therefore, we have investigated the effects produced by the protracted administration of two anticancer drugs on hematopoietic stem cell function. MATERIALS AND METHODS: We used the competitive repopulating assay to assess stem cell damage caused by protracted daily drug treatment of mice. RESULTS: Treatment with acetyldinaline for 10 consecutive days mediated a modest effect on the short-term repopulating cells (STRCs) but spared the long-term repopulating cells (LTRCs). Gemcitabine for 10 days led to a modest decline in both the STRCs and LTRCs. Extending treatment with gemcitabine for 28 days resulted in more severe repopulating cell (RC) damage, which was much worse than in acetyldinaline-treated mice. As expected, melphalan for 10 or 28 days mediated a marked reduction in all of the RCs of treated mice. The analysis of the RCs from mice that were allowed a 1-year recovery period after completing the 28-day treatment with either acetyldinaline or gemcitabine showed normal levels of neutrophils and bone marrow (BM) progenitors. However, a reduction in the RCs was observed in both groups, with larger reductions in gemcitabine-treated mice. CONCLUSIONS: Our data show that protracted treatment with gemcitabine, but not acetyldinaline, of mice caused severe permanent damage to the stem cell components. Therefore, although 28-day therapy with acetyldinaline or gemcitabine appeared to be well tolerated at the level of peripheral blood and bone marrow progenitors, gemcitabine produces permanent stem cell damage when used in long-term administration regimens that should perhaps only be explored clinically with stem cell support available.


Asunto(s)
Antineoplásicos/toxicidad , Desoxicitidina/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Melfalán/toxicidad , Fenilendiaminas/toxicidad , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Benzamidas , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Ensayo de Unidades Formadoras de Colonias , Cruzamientos Genéticos , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Hematopoyesis/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Melfalán/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenilendiaminas/administración & dosificación , Factores de Tiempo , Gemcitabina
7.
Int J Dev Biol ; 37(1): 75-83, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8507572

RESUMEN

Programmed cell death (apoptosis) is the ubiquitous biological phenomenon of intentional cell death that eliminates redundant cells, changes phenotypic composition during histogenesis, provides form during morphogenesis and balances mitosis in renewing tissues. This form of cell death is controlled by a genetic program(s) that kills the targeted cell without causing subsequent inflammation. Malignant cells implanted into the appropriate regulatory field in the embryo will lose their malignant phenotype yet retain the capacity for proliferation and differentiation. This embryonic regulation of cancer requires simultaneous contact with specific structures on the surfaces of normal cells and exposure to soluble, extracellular signals. During studies to identify such soluble factors in the blastocyst, extracellular hydrogen peroxide was discovered in the blastocele fluid. Current evidence indicates that this hydrogen peroxide causes apoptosis of inner cell mass cells destined to develop into trophectoderm--the first apoptotic event during mammalian development which likely prevents the formation of ectopic trophectoderm in the soon-to-appear germ layers (histogenesis). The evidence also suggests that the hydrogen peroxide is generated during the oxidation of extracellular polyamines by a family of enzymes called amine oxidases. The components of this mechanism are also present in the mammalian epidermis, where they are proposed to control the survival of basal cell progeny and hence epidermal homeostasis (essentially controlling the production of tissue mass). This mechanism causes not only apoptosis in vivo, but also the unwanted and artefactual cell death in vitro known as the crisis of spontaneous transformation. These data suggest a novel link between polyamines and apoptosis, a link that has practical as well as theoretical implications.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Amina Oxidasa (conteniendo Cobre) , Apoptosis/fisiología , Inhibidores de Crecimiento/química , Peróxido de Hidrógeno/metabolismo , Poliaminas/metabolismo , Teratoma/metabolismo , Animales , Apoptosis/efectos de los fármacos , Blastocisto/metabolismo , Embrión de Mamíferos/fisiología , Ratones , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Teratoma/fisiopatología , Células Tumorales Cultivadas
8.
Environ Health Perspect ; 106 Suppl 2: 541-57, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9599702

RESUMEN

Hematopoietic tissues are the targets of numerous xenobiotics. Clinical hematotoxicity is either a decrease or an increase in peripheral blood cell counts in one or more cell lineages--a cytopenia or a cytosis, respectively--that carries a risk of an adverse clinical event. The purpose of in vitro hematotoxicology is the prediction of these adverse hematologic effects from the effects of the toxicants on human hematopoietic targets under controlled experimental conditions in the laboratory. Building on its important foundations in experimental hematology and the wealth of hematotoxicology data found in experimental oncology, this field of alternative toxicology has developed rapidly during the past decade. Although the colony-forming unit-granulocyte/monocyte neutrophil progenitor is most frequently evaluated, other defined progenitors and stem cells as well as cell types found in the marrow stroma can be evaluated in vitro. End points have been proposed for predicting toxicant exposure levels at the maximum tolerated dose and the no observable adverse effect level for the neutrophil lineage, and several clinical prediction models for neutropenia have developed to the point that they are ready for prospective evaluation and validation in both preclinical species and humans. Known predictive end points are the key to successful comparisons across species or across chemical structures when in vitro dose-response curves are nonparallel. Analytical chemistry support is critical for accurate interpretation of in vitro data and for relating the in vitro pharmacodynamics to the in vivo pharmacokinetics. In contrast to acute neutropenia, anemia and acute thrombocytopenia, as well as adverse effects from chronic toxicant exposure, are much more difficult to predict from in vitro data. Pharmacologic principles critical for clinical predictions from in vitro data very likely will apply to toxicities to other proliferative tissues, such as mucositis.


Asunto(s)
Alternativas a las Pruebas en Animales , Hematología/métodos , Pruebas de Toxicidad/métodos , Xenobióticos/toxicidad , Anemia/inducido químicamente , Animales , Relación Dosis-Respuesta a Droga , Hematología/tendencias , Humanos , Técnicas In Vitro , Neutropenia/inducido químicamente , Valor Predictivo de las Pruebas , Trombocitopenia/inducido químicamente , Xenobióticos/farmacocinética
9.
Environ Health Perspect ; 106 Suppl 2: 427-39, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9599689

RESUMEN

In the past decade in vitro tests have been developed that represent a range of anatomic structure from perfused whole organs to subcellular fractions. To assess the use of in vitro tests for toxicity testing, we describe and evaluate the current status of organotypic cultures for the major target organs of toxic agents. This includes liver, kidney, neural tissue, the hematopoietic system, the immune system, reproductive organs, and the endocrine system. The second part of this report reviews the application of in vitro culture systems to organ specific toxicity and evaluates the application of these systems both in industry for safety assessment and in government for regulatory purposes. Members of the working group (WG) felt that access to high-quality human material is essential for better use of in vitro organ and tissue cultures in the risk assessment process. Therefore, research should focus on improving culture techniques that will allow better preservation of human material. The WG felt that it is also important to develop and make available relevant reference compounds for toxicity assessment in each organ system, to organize and make available via the Internet complete in vivo toxicity data, including human data, containing dose, end points, and toxicokinetics. The WG also recommended that research should be supported to identify and to validate biological end points for target organ toxicity to be used in alternative toxicity testing strategies.


Asunto(s)
Alternativas a las Pruebas en Animales , Bienestar del Animal , Técnicas de Cultivo/métodos , Técnicas de Cultivo de Órganos/métodos , Pruebas de Toxicidad/métodos , Animales , Humanos , Salud Pública , Reproducibilidad de los Resultados , Investigación/tendencias
10.
Toxicol Sci ; 75(2): 355-67, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12883091

RESUMEN

In a previous study of prevalidation, a standard operating procedure (SOP) for two independent in vitro tests (human and mouse) had been developed, to evaluate the potential hematotoxicity of xenobiotics from their direct and the adverse effects on granulocyte-macrophages (CFU-GM). A predictive model to calculate the human maximum tolerated dose (MTD) was set up, by adjusting a mouse-derived MTD for the differential interspecies sensitivity. In this paper, we describe an international blind trial designed to apply this model to the clinical neutropenia, by testing 20 drugs, including 14 antineoplastics (Cytosar-U, 5-Fluorouracil, Myleran, Thioguanine, Fludarabine, Bleomycin, Methotrexate, Gemcitabine, Carmustine, Etoposide, Teniposide, Cytoxan, Taxol, Adriamycin); two antivirals (Retrovir, Zovirax,); three drugs for other therapeutic indications (Cyclosporin, Thorazine, Indocin); and one pesticide (Lindane). The results confirmed that the SOP developed generates reproducible IC90 values with both human and murine GM-CFU. For 10 drugs (Adriamycin, Bleomycin, Etoposide, Fludarabine, 5-Fluorouracil, Myleran, Taxol, Teniposide, Thioguanine, and Thorazine), IC90 values were found within the range of the actual drug doses tested (defined as the actual IC90). For the other 10 drugs (Carmustine, Cyclosporin, Cytosar-U, Cytoxan, Gemcitabine, Indocin, Lindane, Methotrexate, Retrovir, and Zovirax) extrapolation on the regression curve out of the range of the actual doses tested was required to derive IC90 values (extrapolated IC90). The model correctly predicted the human MTD for 10 drugs out of 10 that had "actual IC90 values" and 7 drugs out of 10 for those having only an extrapolated IC90. Two of the incorrect predictions (Gemcitabine and Zovirax) were within 6-fold of the correct MTD, instead of the 4-fold range required by the model, whereas the prediction with Cytosar-U was approximately 10-fold in error. A possible explanation for the failure in the prediction of these three drugs, which are pyrimidine analogs, is discussed. We concluded that our model correctly predicted the human MTD for 20 drugs out of 23, since the other three drugs (Topotecan, PZA, and Flavopiridol) were tested in the prevalidation study. The high percentage of predicitivity (87%), as well as the reproducibility of the SOP testing, confirm that the model can be considered scientifically validated in this study, suggesting promising applications to other areas of research in developing validated hematotoxicological in vitro methods.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Dosis Máxima Tolerada , Neutropenia/inducido químicamente , Valor Predictivo de las Pruebas , Xenobióticos/toxicidad , Enfermedad Aguda , Animales , Células de la Médula Ósea/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Europa (Continente) , Sangre Fetal/citología , Humanos , Cooperación Internacional , Masculino , Ratones , Ratones Endogámicos C57BL , Neutropenia/patología , Reproducibilidad de los Resultados , Método Simple Ciego , Estados Unidos , Xenobióticos/clasificación
11.
Toxicol Sci ; 59(1): 17-36, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134541

RESUMEN

Advances in the technology of human cell and tissue culture and the increasing availability of human tissue for laboratory studies have led to the increased use of in vitro human tissue models in toxicology and pharmacodynamics studies and in quantitative modeling of metabolism, pharmacokinetic behavior, and transport. In recognition of the potential importance of such models in toxicological risk assessment, the Society of Toxicology sponsored a workshop to evaluate the current status of human cell and tissue models and to develop consensus recommendations on the use of such models to improve the scientific basis of risk assessment. This report summarizes the evaluation by invited experts and workshop attendees of the current status of such models for prediction of human metabolism and identification of drug-drug interactions, prediction of human toxicities, and quantitative modeling of pharmacokinetic and pharmaco-toxicodynamic behavior. Consensus recommendations for the application and improvement of current models are presented.


Asunto(s)
Técnicas de Cultivo de Célula , Técnicas de Cultivo , Modelos Biológicos , Medición de Riesgo/métodos , Xenobióticos/farmacocinética , Xenobióticos/toxicidad , Humanos
12.
Urology ; 54(6A Suppl): 22-9, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10606281

RESUMEN

Paclitaxel and docetaxel have emerged in the last two decades as effective antitumor agents in a variety of malignancies. Paclitaxel is a semisynthetic taxane isolated from bark of the Pacific yew tree. Docetaxel is a semisynthetic taxane derived from the needles of the European yew (Taxus baccata). These compounds bind to tubulin, leading to microtubule stabilization, mitotic arrest and, subsequently, cell death. Plasma clearance of paclitaxel exhibits nonlinear kinetics, which results in a disproportionate change in plasma concentration and area under the curve (AUC) with dose alterations. In contrast, docetaxel has a linear disposition over the dose ranges used clinically, so its concentration changes linearly with changes in the dosage. Premedicating with corticosteroids and histamine H1 and H2 receptor antagonists is advocated prior to paclitaxel administration; prior to docetaxel administration, premedication with corticosteroids is suggested. The taxanes are metabolized in the liver by the cytochrome P-450 enzymes and are eliminated in the bile. The known metabolites are either inactive or less potent than the parent compounds. The toxic effects associated with paclitaxel therapy are mainly neutropenia, peripheral neuropathy, and, rarely, cardiotoxicity. Docetaxel toxicity produces mainly myelosuppression and a cumulative dose fluid retention syndrome. Paclitaxel demonstrates sequence-dependent interactions with cisplatin, cyclophosphamide, and doxorubicin. Docetaxel has shown increased myelosuppression with preceding ifosfamide in a preliminary study. The future holds increasing indications for taxanes in newer combination regimens; consideration of their pharmacologic characteristics is an important aspect of designing and applying new taxane-based treatment regimens.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/farmacocinética , Hidrocarburos Aromáticos con Puentes/farmacología , Hidrocarburos Aromáticos con Puentes/farmacocinética , Neoplasias de la Próstata/tratamiento farmacológico , Taxoides , Docetaxel , Interacciones Farmacológicas , Humanos , Masculino , Mitosis/efectos de los fármacos , Estructura Molecular , Paclitaxel/análogos & derivados , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Neoplasias de la Próstata/patología
13.
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
14.
Cancer Chemother Pharmacol ; 39(5): 467-72, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9054963

RESUMEN

PURPOSE: 20(S)-Camptothecin (CAM), topotecan (TPT, active ingredient in Hycamtin) and 9-amino-20(S)-camptothecin (9AC) are topoisomerase I inhibitors that cause similar dose-limiting toxicities to rapidly renewing tissues, such as hematopoietic tissues, in humans, mice, and dogs. However, dose-limiting toxicity occurs at tenfold lower doses in humans than in mice. The purpose of the current study was to determine whether hematopoietic progenitors of the myeloid lineage from humans, mice, and dogs exhibit the differential sensitivity to these compounds that is evident in vivo. METHODS: Drug-induced inhibition of in vitro colony formation by a myeloid progenitor in human, murine, and canine marrow colony-forming unit-granulocyte/macrophage (CFU-GM) provided the basis for interspecies comparisons at concentrations which inhibited colony formation by 50% (IC50) and 90% (IC90). RESULTS: Murine IC90 values were 2.6-, 2.3-, 10-, 21-, 5.9-, and 11-fold higher than human values for CAM lactone (NSC-94600) and sodium salt (NSC-100880), TPT (NSC-609699), and racemic (NSC-629971), semisynthetic and synthetic preparations (NSC-603071) of 9AC, respectively. In contrast, canine IC90 values were the same as, or lower than, the human IC90 values for all six compounds. CONCLUSIONS: The greater susceptibility of humans and dogs to the myelotoxicity of camptothecins, compared to mice, was evident in vitro at the cellular level. Differential sensitivity between murine and human myeloid progenitors explains why the curative doses of TPT and 9AC in mice with human tumor xenografts are not achievable in patients. Realizing the curative potential of these compounds in humans will require the development of therapies to increase drug tolerance of human CFU-GM at least to a level equal to that of murine CFU-GM. Because these interspecies differences are complicated by species-specific effects of plasma proteins on drug stability, not all in vitro assay conditions will yield results which can contribute to the development of such therapies.


Asunto(s)
Camptotecina/análogos & derivados , Camptotecina/toxicidad , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/toxicidad , Células de la Médula Ósea , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Perros , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/citología , Humanos , Interleucina-3/farmacología , Ratones , Proteínas Recombinantes/farmacología , Inhibidores de Topoisomerasa II , Topotecan
15.
Toxicol In Vitro ; 15(6): 729-40, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11698175

RESUMEN

This report describes an international prevalidation study conducted to optimise the Standard Operating Procedure (SOP) for detecting myelosuppressive agents by CFU-GM assay and to study a model for predicting (by means of this in vitro hematopoietic assay) the acute xenobiotic exposure levels that cause maximum tolerated decreases in absolute neutrophil counts (ANC). In the first phase of the study (Protocol Refinement), two SOPs were assessed, by using two cell culture media (Test A, containing GM-CSF; and Test B, containing G-CSF, GM-CSF, IL-3, IL-6 and SCF), and the two tests were applied to cells from both human (bone marrow and umbilical cord blood) and mouse (bone marrow) CFU-GM. In the second phase (Protocol Transfer), the SOPs were transferred to four laboratories to verify the linearity of the assay response and its interlaboratory reproducibility. After a further phase (Protocol Performance), dedicated to a training set of six anticancer drugs (adriamycin, flavopindol, morpholino-doxorubicin, pyrazoloacridine, taxol and topotecan), a model for predicting neutropenia was verified. Results showed that the assay is linear under SOP conditions, and that the in vitro endpoints used by the clinical prediction model of neutropenia are highly reproducible within and between laboratories. Valid tests represented 95% of all tests attempted. The 90% inhibitory concentration values (IC(90)) from Test A and Test B accurately predicted the human maximum tolerated dose (MTD) for five of six and for four of six myelosuppressive anticancer drugs, respectively, that were selected as prototype xenobiotics. As expected, both tests failed to accurately predict the human MTD of a drug that is a likely protoxicant. It is concluded that Test A offers significant cost advantages compared to Test B, without any loss of performance or predictive accuracy. On the basis of these results, we proposed a formal Phase II validation study using the Test A SOP for 16-18 additional xenobiotics that represent the spectrum of haematotoxic potential.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias , Granulocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Neutropenia/inducido químicamente , Xenobióticos/toxicidad , Enfermedad Aguda , Animales , Células de la Médula Ósea , Recuento de Células , Perros , Sangre Fetal , Granulocitos/patología , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Biológicos , Neutropenia/patología , Valor Predictivo de las Pruebas , Ratas , Ratas Endogámicas F344 , Reproducibilidad de los Resultados
16.
In Vivo ; 5(5): 493-500, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1768800

RESUMEN

Programmed cell death can also be caused by extracellular, soluble factors, some of which are not physiologic. In vivo in the murine blastocyst, apoptosis eliminates redundant cells of unneeded phenotypes (pre-trophectoderm) from the inner cell mass. Although only a few cells die and many survive, all of the cells are exposed to the extracellular H2O2 in blastocele fluid. The specific control of life versus death is due to the developmental expression of glutathione-dependent protective mechanisms in the surviving cells. Based on recent results, a model is proposed in which apoptosis and necrosis reflect different temporal points along a single cell death program.


Asunto(s)
Blastocisto/citología , Muerte Celular , Citotoxinas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Radicales Libres , Peróxido de Hidrógeno/farmacología , Modelos Biológicos , Poliaminas/farmacología
18.
Cytotechnology ; 10(2): 93-124, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1369214

RESUMEN

It has been known for several decades that cultured murine cells undergo a defined series of changes, i.e., an in vitro evolution, which includes crisis, spontaneous transformation ('immortalization'), aneuploidy, and spontaneous neoplastic transformation. These changes have been shown to be caused by the in vitro environment rather than an inherent instability of the murine phenotype or genotype. Serum amine oxidases were recently identified as a predominant cause of crisis. These enzymes generate hydrogen peroxide from polyamine substrates that enter the extracellular milieu. This finding implicates free-radical toxicity as the underlying cause of in vitro evolution. We propose an oxyradical hypothesis to explain each of the stages of in vitro evolution and discuss its significance for cytotechnology and long-term cultivation of mammalian cell types.


Asunto(s)
Aneuploidia , Transformación Celular Neoplásica/inducido químicamente , Células Cultivadas/citología , Medios de Cultivo/toxicidad , Radicales Libres , Oxidorreductasas/toxicidad , Animales , Sangre , Muerte Celular , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Senescencia Celular , Medio de Cultivo Libre de Suero , Genotipo , Ratones , Modelos Biológicos , Oncogenes , Especificidad de Órganos , Oxidación-Reducción , Oxidorreductasas/sangre , Oxígeno/metabolismo , Fenotipo , Ratas , Células Madre/citología , Factores de Tiempo
19.
Cytotechnology ; 27(1-3): 149-64, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19002789

RESUMEN

DNA topoisomerase I is a nuclear enzyme which catalyzes the conversion of the DNA topology by introducing single-strand breaks into the DNA molecule. This enzyme represents a novel and distinct molecule target for cancer therapy by antitopoisomerase drugs belonging to the campthotecin series of antineoplastics. As many tumors can acquire resistance to drug treatment and become refractary to the chemotherapy it is very important to investigate the mechanisms involved in such a drug resistance for circumventing the phenomenon. This article describes the role of topoisomerase I in cell functions and the methods used to assess its in vitro catalytic activity. It reviews the mechanisms of cytotoxicity of the most specific antitopoisomerase I drugs by considering also the phenomenon of drug resistance. Some factors useful to drive the future perspectives in the development of new topoisomerase I inhibitors are also evidenced and discussed.

20.
Biochem Biophys Res Commun ; 119(3): 1015-21, 1984 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-6712660

RESUMEN

Electrofocusing of mouse small intestine mucosal cytosol yielded two prominent peaks of glutathione S-transferase activity, at pH 8.7 and pH 9.0. These intestinal transferases were indistinguishable from corresponding liver transferases in immunological reactivities and in several other properties. Dietary t-butyl-4-hydroxyanisole increased intestine cytosol protein immunologically identical to the liver transferase with pI of 8.7 and also elevated markedly and preferentially the intestinal transferase activity peak at pH 8.7. Thus, the predominant transferases of mouse intestine appear identical to two of the liver isozymes, and t-butyl-4-hydroxyanisole enhances intestinal transferase activity mainly by increasing the quantity of the isozyme(s) of pI 8.7.


Asunto(s)
Anisoles/farmacología , Hidroxianisol Butilado/farmacología , Glutatión Transferasa/biosíntesis , Mucosa Intestinal/enzimología , Intestino Delgado/enzimología , Isoenzimas/biosíntesis , Animales , Complejo Antígeno-Anticuerpo , Citosol/enzimología , Inducción Enzimática , Femenino , Sueros Inmunes , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA