Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 10(19): 6677-85, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15475458

ABSTRACT

PURPOSE: The hollow fiber assay is used successfully as a routine in vivo screening model to quantitatively define anticancer activity by the National Cancer Institute. This study investigates whether the hollow fiber assay can be used as a short-term in vivo model to demonstrate specific pharmacodynamic end points, namely microtubule and cell cycle disruption. EXPERIMENTAL DESIGN: The growth of A549 cells was characterized within hollow fibers over 5 days in vivo at both subcutaneous (s.c.) and intraperitoneal (i.p.) sites. Drugs were administered on day 4 (i.p.). RESULTS: At 24 hours, cells were retrieved from fibers at both i.p. and s.c. sites of paclitaxel-treated (20 mg/kg) and combretastatin A1 phosphate-treated (150 mg/kg) mice. Cell cycle analysis after paclitaxel treatment revealed a mean G(2)-M phase population of 48.04% (i.p.) and 25.76% (s.c.) compared with vehicle group mice (6.78 and 5.56%, respectively; P = <0.001 and 0.005, respectively). Tumor cells retrieved from combretastatin A1 phosphate-treated mice had a mean G2-M phase population of 36.3% (i.p.) and 29.36% (s.c.) compared with cells retrieved from vehicle group mice (5.58 and 5.49%, respectively; P = <0.001). Using fluorescence and laser-confocal microscopy, paclitaxel was revealed to induce the formation of spindle asters and tubulin polymerization. Combretastatin A1 phosphate was shown to hold cells in mitosis. Changes in nuclear morphology were also observed. CONCLUSION: These data demonstrate that the hollow fiber assay can be used as a short-term in vivo model for studying the pharmacodynamic effects of both standard and novel compounds on microtubules. Evidence has also been provided to support the routine use of the in vivo hollow fiber assay for demonstrating the mechanism of action of a drug.


Subject(s)
Drug Screening Assays, Antitumor/methods , Microtubules/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/instrumentation , Female , Flow Cytometry , Humans , Immunohistochemistry , Membranes, Artificial , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/drug effects , Neoplasm Transplantation/methods , Paclitaxel/pharmacology , Polyvinyls , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Stilbenes/pharmacology , Transplantation, Heterologous , Tubulin/analysis , Tubulin/metabolism
2.
J Nat Prod ; 67(9): 1445-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15387639

ABSTRACT

Bioassay-guided fractionation was applied to the cytotoxic chloroform fraction of the red alga Polysiphonia lanosa. The major compounds of the most active fraction were identified using GLC-MS analysis as lanosol (1), methyl, ethyl, and n-propyl ethers of lanosol (1a, 1b, and 1c, respectively), and aldehyde of lanosol (2), although 1b appears to be an artifact arising during the fractionation procedure. These compounds and other known bromophenols were synthesized in addition to four novel isomers (3, 3a-c). The cytotoxic activities of all the synthetic compounds were determined against DLD-1 cells using the MTT assay. Compounds with IC(50) < 20 micromol were also tested against HCT-116 cells. Compound 3c (2,5-dibromo-3,4-dihydroxybenzyl n-propyl ether) was the most active compound against both cell lines (IC(50) = 1.72 and 0.80 micromol, respectively), and its effect on the cell cycle was studied using flow cytometry.


Subject(s)
Antineoplastic Agents/isolation & purification , Hydrocarbons, Brominated/isolation & purification , Phenols/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , England , Hydrocarbons, Brominated/chemistry , Hydrocarbons, Brominated/pharmacology , Inhibitory Concentration 50 , Isomerism , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phenols/chemistry , Phenols/pharmacology , Rhodophyta , Tumor Cells, Cultured
3.
Biochem Pharmacol ; 67(8): 1587-99, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15041476

ABSTRACT

Methionine dependence is a feature unique to cancer cells, exhibited as inability to grow in a methionine-depleted environment supplemented with homocysteine, the immediate metabolic precursor of methionine. This study explores the effect of methionine depletion and homocysteine supplementation on the viability, sulfur amino acid metabolism and cell-cycle kinetics of normal and cancer cells, as well as their ability to recover from the treatments. An array of cells including hepatomas (HTC, Phi-1), prostate adenocarcinomas (PC-3) and transformed (3T3) and normal (HS-27) fibroblasts, has been used aiming to evaluate the importance of tissue specificity. All cell lines proliferated well in methionine-complete media (M+H-), whilst only the normal fibroblasts HS-27 grew in methionine-depleted homocysteine-supplemented media (M-H+). None of the tested cell lines were able to grow in media without methionine or homocysteine (M-H-). HTC was the only cell line that did not recover from the M-H+ treatment whilst PC-3 did not recover from the M-H- treatment. Methionine and homocysteine depletion (M-H+ and M-H-) were found to induce arrest at different phases of the cell cycle, depending on the cell line: the methionine-dependent HTC, PC-3 and 3T3 arrested at the S and G2/M phase, whilst Phi-1 and the methionine-independent HS-27 accumulated in the G1 phase. The cell-cycle kinetics showed that the observed blockades were reversible. The information resulting from these studies is important for not only the behavior of cancer cells, but also for appreciating the potential of developing cancer therapies based on methionine-depletion strategies.


Subject(s)
Homocysteine/pharmacology , Methionine/pharmacology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Humans , Mice , Rats , Sulfur/chemistry
4.
Clin Cancer Res ; 10(4): 1446-53, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14977848

ABSTRACT

PURPOSE: Combretastatin A4 phosphate (CA4P) and its structural analog, combretastatin A1 phosphate (CA1P), are soluble prodrugs capable of interacting with tubulin and causing rapid vascular shutdown within tumors. CA4P has completed Phase I clinical trials, but recent preclinical studies have shown that CA1P displays a greater antitumor effect than the combretastatin A4 (CA4) analog at equal doses. The aim of this study, therefore, is to compare pharmacokinetics and metabolism of the two compounds to determine whether pharmacokinetics plays a role in their differential activity. EXPERIMENTAL DESIGN: NMRI mice bearing MAC29 tumors received injection with either CA4P or CA1P at a therapeutic dose of 150 mg x kg(-1), and profiles of both compounds and their metabolites analyzed by a sensitive and specific liquid chromatography/mass spectroscopy method. RESULTS: The metabolic profile of both compounds is complex, with up to 14 metabolites being detected for combretastatin A1 (CA1) in the plasma. Many of these metabolites have been identified by liquid chromatography/mass spectroscopy. Initial studies, however, focused on the active components CA4 and CA1, where plasma and tumor areas under the curve were 18.4 and 60.1 microg x h x ml(-1) for CA4, and 10.4 and 13.1 microg x h x ml(-1) for CA1, respectively. In vitro metabolic comparisons of the two compounds strongly suggest that CA1 is metabolized to a more reactive species than the CA4. CONCLUSIONS: Although in vitro studies suggest that variable rates of tumor-specific prodrug dephosphorylation may explain these differences in pharmacokinetics profiles, the improved antitumor activity and altered pharmacokinetic profile of CA1 may be due to the formation of a more reactive metabolite.


Subject(s)
Bibenzyls/pharmacology , Stilbenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Area Under Curve , Calibration , Cell Line, Tumor , Chromatography , Chromatography, High Pressure Liquid , Coloring Agents/pharmacology , Female , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Mice , Models, Chemical , Phosphorylation , Prodrugs , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors
5.
Mol Cancer Ther ; 3(12): 1565-75, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15634650

ABSTRACT

Phortress is a novel, potent, and selective experimental antitumor agent. Its mechanism of action involves induction of CYP1A1-catalyzed biotransformation of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203) to generate electrophilic species, which covalently bind to DNA, exacting lethal damage to sensitive tumor cells, in vitro and in vivo. Herein, we investigate the effects of DNA adduct formation on cellular DNA integrity and progression through cell cycle and examine whether a relevant pharmacodynamic end point may be exploited to probe the clinical mechanism of action of Phortress and predict tumor response. Single cell gel electrophoresis (SCGE) was applied to quantify DNA damage and cell cycle analyses conducted upon 5F 203 treatment of benzothiazole-sensitive MCF-7 and inherently resistant MDA-MB-435 breast carcinoma cells. Following treatment of xenograft-bearing mice and mice possessing hollow fiber implants containing MCF-7 or MDA-MB-435 cells with Phortress (20 mg/kg, i.p., 24 hours), tumor cells and xenografts were recovered for analyses by SCGE. Dose- and time-dependent DNA single and double strand breaks occurred exclusively in sensitive cells following treatment with 5F 203 in vitro (10 nmol/L-10 micromol/L; 24-72 hours). In vivo, Phortress-sensitive and Phortress-resistant tumor cells were distinct; moreover, DNA damage in xenografts, following treatment of mice with Phortress, could be determined. Interrogation of the mechanism of action of 5F 203 in silico by self-organizing map-based cluster analyses revealed modulation of phosphatases and kinases associated with cell cycle regulation, corroborating observations of selective cell cycle perturbation by 5F 203 in sensitive cells. By conducting SCGE, tumor sensitivity to Phortress, an agent currently undergoing clinical evaluation, may be determined.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , DNA Adducts/drug effects , Drug Resistance, Neoplasm , Thiazoles/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Cycle/drug effects , Comet Assay , Computational Biology , Cytochrome P-450 CYP1A1/metabolism , DNA Damage/drug effects , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Membranes, Artificial , Mice , Mice, Nude , Time Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...