Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin Cancer Res ; 7(11): 3336-42, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705845

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Quinoxalines/pharmacology , Tumor Suppressor Protein p53/physiology , Cell Cycle/physiology , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/physiology , Dose-Response Relationship, Drug , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitosis/drug effects , Mutation , Neoplasms/pathology , Neoplasms/prevention & control , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
2.
J Chromatogr B Biomed Sci Appl ; 759(1): 175-8, 2001 Aug 05.
Article in English | MEDLINE | ID: mdl-11499623

ABSTRACT

A simple and reliable HPLC method was developed for the estimation of a new anti-cancer agent that belongs to the thioxanthone class, SR271425 in mouse plasma. SR271425, it's metabolites and internal standard (SR233377) were separated from plasma by liquid-liquid extraction using dichloromethane after quenching the plasma proteins with acetonitrile. Chromatography was performed on a reversed-phase C18 column using methanol-10 mM phosphate buffer, pH 3.5 (45:55) as mobile phase at a flow-rate of 0.8 ml/min for first 10 min and 1.4 ml/min for the next 15 min with UV-Vis detection at 264 nm and SR233377 as internal standard. The retention times of SR271425 and internal standard were 18.6 and 14.8 min, respectively. The limit of detection was 40 ng/ml and the limit of quantification was 78 ng/ml. This method was also able to detect the three metabolites of SR271425. The intra- and inter-day relative standard deviations were less than 13% at all concentrations. This analytical method was precise and reproducible for pharmacokinetics and metabolism studies of the drug in mice. SR271425 is proceeding to phase I clinical trials in 2001.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Thioxanthenes/pharmacokinetics , Animals , Antineoplastic Agents/blood , Mice , Reference Standards , Reproducibility of Results , Spectrophotometry, Ultraviolet , Thioxanthenes/blood
3.
Exp Hematol ; 29(3): 286-94, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11274755

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/toxicity , Deoxycytidine/toxicity , Hematopoietic Stem Cells/drug effects , Melphalan/toxicity , Phenylenediamines/toxicity , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Benzamides , Bone Marrow/drug effects , Bone Marrow/pathology , Colony-Forming Units Assay , Crosses, Genetic , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematopoiesis/drug effects , Injections, Intraperitoneal , Male , Melphalan/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phenylenediamines/administration & dosage , Time Factors , Gemcitabine
4.
Toxicol Sci ; 59(1): 17-36, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134541

ABSTRACT

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.


Subject(s)
Cell Culture Techniques , Culture Techniques , Models, Biological , Risk Assessment/methods , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity , Humans
5.
Clin Cancer Res ; 6(6): 2474-81, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873102

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Deoxycytidine/administration & dosage , Melphalan/administration & dosage , Phenylenediamines/administration & dosage , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Benzamides , Blood Cell Count , Blood Platelets/drug effects , Bone Marrow/drug effects , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Female , Melphalan/adverse effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neutropenia/chemically induced , Neutrophils/drug effects , Phenylenediamines/adverse effects , Random Allocation , Stem Cells/drug effects , Time Factors , Gemcitabine
6.
Urology ; 54(6A Suppl): 22-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606281

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Bridged-Ring Compounds/pharmacology , Bridged-Ring Compounds/pharmacokinetics , Prostatic Neoplasms/drug therapy , Taxoids , Docetaxel , Drug Interactions , Humans , Male , Mitosis/drug effects , Molecular Structure , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Prostatic Neoplasms/pathology
7.
Ann Oncol ; 9(4): 357-64, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9636825

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow Cells/drug effects , Hematologic Diseases/chemically induced , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Area Under Curve , Bone Marrow Cells/pathology , Clinical Trials, Phase I as Topic , Disease Models, Animal , Dogs , Humans , In Vitro Techniques , Mice , Neutropenia/chemically induced , Predictive Value of Tests , Reproducibility of Results , Species Specificity , Tumor Cells, Cultured
8.
Environ Health Perspect ; 106 Suppl 2: 541-57, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9599702

ABSTRACT

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.


Subject(s)
Animal Testing Alternatives , Hematology/methods , Toxicity Tests/methods , Xenobiotics/toxicity , Anemia/chemically induced , Animals , Dose-Response Relationship, Drug , Hematology/trends , Humans , In Vitro Techniques , Neutropenia/chemically induced , Predictive Value of Tests , Thrombocytopenia/chemically induced , Xenobiotics/pharmacokinetics
9.
Cytotechnology ; 27(1-3): 149-64, 1998 Sep.
Article in English | MEDLINE | ID: mdl-19002789

ABSTRACT

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.

10.
Cancer Chemother Pharmacol ; 39(5): 467-72, 1997.
Article in English | MEDLINE | ID: mdl-9054963

ABSTRACT

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.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/toxicity , Hematopoietic Stem Cells/drug effects , Animals , Antineoplastic Agents, Phytogenic/toxicity , Bone Marrow Cells , Cells, Cultured , Colony-Forming Units Assay , Dogs , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Humans , Interleukin-3/pharmacology , Mice , Recombinant Proteins/pharmacology , Topoisomerase II Inhibitors , Topotecan
11.
Differentiation ; 60(2): 67-74, 1996 May.
Article in English | MEDLINE | ID: mdl-8641547

ABSTRACT

The effect of the mutagenic carcinogen 4-nitroquinoline-N-oxide (4NQO) on limb regeneration was studied in adult Triturus cristatus newts. Delayed limb regeneration was observed when a 10- to 15-micrograms crystal of 4NQO was implanted at the late dedifferentiation or late bud stage. Additionally, 4NQO administration at these stages caused developmental deformities in skeletal elements of the subsequent regenerates. In contrast, 4NQO implanted at the wound healing stage did not affect skeletal morphogenesis. These critical stages of limb regeneration affected by carcinogen exposure (but not other manipulations) represent mechanisms distinct from disruption of basement membrane deposition. Secondary regeneration, initiated by reamputation of the 4NQO-treated abnormal regenerates 4-5 mm proximal to the site of carcinogen implantation, produced normal regenerates, setting a limit on the diffusion distance of 4NQO in the limb. Distal reamputation through regenerates at the level of the abnormality resulted in regeneration of the original bone deformity, suggesting that the teratogenic effect of 4NQO is mediated via heritable mutational events in blastema cells. These results extend a previously published conclusion that 4-NQO is mutagenic in non-regenerative tissue to include regenerative tissue as well. However, in spite of mutagenic activity, squamous carcinomas were not induced by 4NQO exposure at any stage of regeneration. Therefore, the resistance of regenerative tissue to 4'-NQO carcinogenesis is not due to resistance to mutations, but rather to other mechanisms perhaps similar to those which regulate malignant cells in murine embryos.


Subject(s)
4-Nitroquinoline-1-oxide/toxicity , Carcinogens/toxicity , Carcinoma, Squamous Cell/chemically induced , Extremities/physiology , Mutagens/toxicity , Regeneration/drug effects , Animals , Extremities/embryology , Female , Male , Salamandridae
12.
Invest New Drugs ; 13(4): 303-14, 1996.
Article in English | MEDLINE | ID: mdl-8824348

ABSTRACT

Human colony forming units (CFUs) from both malignant and hematopoietic tissues can be assayed in vitro in microcapillary cultures, an alternative cloning system to the Petri dish methodology. For technical reasons, microcapillary culture may be ideally suited for new drug screening by therapeutic index. To achieve the high output required by screening programs, automated quantitation of CFUs is required. Toward this end, this paper reports the development of a prototype CapScan, an image analysis system that uses a novel axial laser illumination system to detect tumor cell colonies and, with technical modifications, CFU-granulocyte-macrophage (CFU-GM) colonies in microcapillary cultures. As currently configured, the CapScan can quantify colonies grown in a rack of 18 microcapillary cultures in 30 minutes or less. The sensitivity and detection specificity of tumor cell colonies is >90% with a coefficient of variance of 5-40%, dependent upon colony number. Over a range of colony numbers, CapScan and manual colony counts showed a linear correlation > -0.9, and yielded identical results in assays of doxorubicin inhibition of clonogenic P388 cells. As an additional advantage, the growth kinetics of individual colonies can also be monitored with the CapScan, making distinctions between cytotoxic and cytostatic drugs possible; colonies of freshly isolated human tumor cells can also be quantified. Thus, a microcapillary-based human tumor cloning assay that tests for resistance and/or sensitivity to chemotherapeutic agents may be useful in drug development programs and may also facilitate the development of chemotherapy for individual patient tumors, especially when tumor availability is limited.


Subject(s)
Drug Screening Assays, Antitumor , Hematopoietic Stem Cells/cytology , Image Processing, Computer-Assisted/methods , Neoplasms/pathology , Animals , Bone Marrow/drug effects , Bone Marrow Cells , Cytological Techniques , Granulocytes/cytology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia P388/drug therapy , Leukemia P388/pathology , Macrophages/cytology , Male , Mice , Mice, Inbred Strains , Microscopy/methods , Neoplasms/drug therapy , Tumor Cells, Cultured
13.
Cancer Chemother Pharmacol ; 39(1-2): 143-9, 1996.
Article in English | MEDLINE | ID: mdl-8995512

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/toxicity , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Indoles/pharmacology , Indoles/toxicity , Urea/analogs & derivatives , Animals , Bone Marrow/drug effects , Dogs , Duocarmycins , Female , Humans , Male , Mice , Mice, Inbred BALB C , Species Specificity , Stem Cells/drug effects , Urea/pharmacology , Urea/toxicity
14.
J Natl Cancer Inst ; 86(4): 273-80, 1994 Feb 16.
Article in English | MEDLINE | ID: mdl-8158681

ABSTRACT

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.


Subject(s)
Acridines/toxicity , Antineoplastic Agents/toxicity , Bone Marrow/drug effects , Erythroid Precursor Cells/drug effects , Pyrazoles/toxicity , Animals , Bone Marrow Cells , Cells, Cultured , Dogs , Humans , Leukocyte Count/drug effects , Mice
15.
Br J Haematol ; 84(2): 197-203, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8398818

ABSTRACT

The regulation of megakaryocytopoiesis and platelet production has not yet been clearly elucidated. Several cytokines have been shown to be capable of producing megakaryocyte colonies from bone marrow [i.e. Interleukin (IL)-3, granulocyte-macrophage (GM)-colony-stimulating factor (CSF), erythropoietin (Epo)]. In addition, other activities have been reported to stimulate megakaryocyte precursors, yet a megakaryocyte-CSF (Meg-CSF) has not been purified to homogeneity and IL-3, GM-CSF and/or Epo often contaminate purification attempts which could account for the activities. A Meg-CSF has been isolated from the urine of patients with aplastic anaemia and purified by sequential ultrafiltration, cation exchange, G-50 chromatography, preparative PAGE, chromatofocusing and cation exchange HPLC. The activity of this material is 2-4 x 10(4) CFU-Meg/mg as measured in a murine marrow, serum-containing assay. This activity also stimulates CFU-Meg in the absence of adherent accessory cells and in serum-free cultures, indicative of the direct stimulation on CFU-Meg. Immunoassays, colony forming assays, and proliferation assays demonstrate that purified Meg-CSF has no GM-CSF, IL-3, M-CSF, G-CSF or IL-1 alpha, -3, -6, -9 and -11. In confirmation of these results, neutralizing antibody to IL-6 also did not abrogate Meg-CSF activity. Therefore the previously-reported megakaryocyte colony-stimulating activity in purified aplastic anaemia patient urine is due to a unique cytokine: Meg-CSF.


Subject(s)
Megakaryocytes/cytology , Proteins/pharmacology , Anemia, Aplastic/urine , Animals , Colony-Forming Units Assay , Colony-Stimulating Factors/pharmacology , Erythropoietin/pharmacology , GPI-Linked Proteins , Humans , Interleukins/pharmacology , Megakaryocytes/drug effects , Membrane Glycoproteins , Mesothelin , Mice , Proteins/metabolism
16.
Int J Dev Biol ; 37(1): 75-83, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8507572

ABSTRACT

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)


Subject(s)
Amine Oxidase (Copper-Containing) , Apoptosis/physiology , Growth Inhibitors/chemistry , Hydrogen Peroxide/metabolism , Polyamines/metabolism , Teratoma/metabolism , Animals , Apoptosis/drug effects , Blastocyst/metabolism , Embryo, Mammalian/physiology , Mice , Oxidation-Reduction , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Teratoma/physiopathology , Tumor Cells, Cultured
17.
Toxicol Pathol ; 21(2): 241-50, 1993.
Article in English | MEDLINE | ID: mdl-8210945

ABSTRACT

Myelosuppression is the dose-limiting side effect for most anti-cancer and many anti-human immunodeficiency virus agents, which can be quantitated with optimized colony-forming assays (granulocyte-macrophage, late erythroid, and megakaryocytic [for murine only] colony-forming units and early erythroid burst-forming units (BFUs)). When applied to new drug development, the assays are used for therapeutic index-based screening (e.g., less myelosuppressive analogues, structure-toxicity studies, new drug leads), interpreting efficacy data from xenotransplant models, and selecting the most accurate animal model for human hematopoietic toxicity. However, other types of assays may be required to identify the mechanism underlying myelosuppression. In clinical trial planning, in vitro colony-forming assays can be used to elucidate schedule dependency of myelotoxicity (which in turn provides clues about mechanism of action), to plan cytokine support, and to estimate dose-escalation effects. The inhibition of colony formation can be measured relative to a compound with known clinical myelotoxicity and schedule dependency to provide some idea of the toxicity expected at particular doses, and the degree of heterogeneity between individuals, during clinical trials. The predictive accuracy of the in vitro data has been proven by validation studies with alkylating agents.


Subject(s)
Bone Marrow/drug effects , Animals , Cells, Cultured , Clinical Trials as Topic , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Hematopoietic Stem Cells/drug effects , Humans , Mice
18.
Int J Cell Cloning ; 10(6): 359-68, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1334113

ABSTRACT

Because of the bias toward successful cloning of human tumor cells from more advanced malignancies, alternative approaches to clinical correlations of drug resistance are needed to determine the validity of the human tumor clonogenic assay (HTCA) as a clinically useful test. Capitalizing on the prevalence of clinical drug resistance among these advanced malignancies, we have taken an independent approach to testing the validity of HTCAs based upon pharmacologic principles rather than tumor response. A database of results from drug sensitivity/resistance testing in 1,777 HTCAs has been examined retrospectively for specimens exhibiting either the MDR1 or Topo-II pleiotropic drug resistance phenotype. Twenty specimens were identified as MDR1 based upon test results showing resistance to adriamycin, vinca alkaloid, and etoposide. Test results with mitomycin-c confirmed the MDR1 phenotype in eight out of nine of these specimens. Seven out of eight of the confirmed MDR1 samples were resistant to either cis-platinum or alkylating agents or to both. There was no significant difference in the 5-fluorouracil resistance of these MDR1 specimens and the database as a whole, demonstrating the specific nature of this drug resistance phenotype in vitro. One specimen, a squamous carcinoma of the lung, was mitomycin-c sensitive, even though it exhibited all the other drug resistances characteristic of the MDR1 phenotype. Six specimens with the Topo-II phenotype were identified based upon resistance to adriamycin and etoposide with sensitivity to vinca alkaloids. Surprisingly, the Topo-II phenotype showed a strong association with increased cis-platinum resistance and a weaker one with decreased 5-fluorouracil resistance. Thus, 26/30 (87%) of analyzable specimens showed some form of clinically characterized multidrug resistance, illustrating how easily one can obtain 90% accuracy in predicting clinical drug resistance with HTCAs that are heavily biased by a disproportionate number of successful cloning assays with advanced malignancies. The data analysis also shows that prediction of adriamycin resistance based on lack of Topoisomerase II expression will not be very accurate, in contrast to a previous claim. Until cell culture technology can facilitate frequent successes in the cloning of early detected, drug-sensitive lesions, this bias will remain in HTCA databases, and studies comparing HTCA results with clinical response will continue to be uninformative. However, the in vitro identification of pleiotropic drug resistance phenotypes exactly analogous to those previously observed in patients provides pharmacologic validation at least for the prediction of drug resistance as measured by current HTCAs using suprapharmacologic drug concentrations.


Subject(s)
Drug Resistance , Tumor Stem Cell Assay/methods , Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II , Databases, Bibliographic , Phenotype , Retrospective Studies
19.
Cancer Res ; 52(18): 4858-65, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1516043

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/pathology , Neoplastic Stem Cells/pathology , Salamandridae/physiology , Skin Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/chemically induced , Keratinocytes/cytology , Methylcholanthrene , Phylogeny , Regeneration , Skin Neoplasms/chemically induced
20.
Cytotechnology ; 10(2): 93-124, 1992.
Article in English | MEDLINE | ID: mdl-1369214

ABSTRACT

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.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/chemically induced , Cells, Cultured/cytology , Culture Media/toxicity , Free Radicals , Oxidoreductases/toxicity , Animals , Blood , Cell Death , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cellular Senescence , Culture Media, Serum-Free , Genotype , Mice , Models, Biological , Oncogenes , Organ Specificity , Oxidation-Reduction , Oxidoreductases/blood , Oxygen/metabolism , Phenotype , Rats , Stem Cells/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...