Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 9(12): 1352-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478472

ABSTRACT

Mononuclear and multinuclear platinum complexes are known to induce distinct types of DNA lesions and exhibit different profiles of antitumor activity, in relation to p53 mutational status. In this study, we investigated the cellular effects of exposure to two platinum compounds (cisplatin and the multinuclear platinum complex BBR 3464), in the osteosarcoma cell line, U2-OS, carrying the wild-type p53 gene and capable of undergoing apoptosis or cell cycle arrest in response to diverse genotoxic stresses. In spite of the ability of both compounds to up-regulate p53 at cytotoxic concentrations, exposure to BBR 3464 resulted in cell cycle arrest but only cisplatin was capable of inducing significant levels of apoptosis and phosphorylation at the Ser15 residue of p53. The cisplatin-induced protein phosphorylation, not detectable in cells treated with BBR 3464, was associated with RPA phosphorylation, a specific up-regulation of Bax and down-regulation of p21(WAF1). Cells treated with BBR 3464 displayed a different cellular response with evidence of cytostasis associated with a high induction of p21(WAF1). The regulation of p21(WAF1) after cisplatin or BBR 3464 exposure required a p53 signal, as documented using stable transfectants expressing a dominant-negative form of p53 (175(his)). Taken together, these results indicate that cellular response to different genotoxic lesions (i.e. apoptosis or growth arrest) is associated with a specific recognition of DNA damage and a different p53-mediated signaling pathway. Multinuclear platinum complexes could be regarded as useful tools for investigating the p53-mediated process of cell cycle arrest in response to DNA damage.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Damage/drug effects , Neoplasms/drug therapy , Platinum Compounds/pharmacology , Tumor Suppressor Protein p53/drug effects , Amino Acid Sequence/drug effects , Amino Acid Sequence/physiology , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cisplatin/pharmacology , Cytotoxins/pharmacology , DNA Damage/physiology , Humans , Neoplasms/metabolism , Neoplasms/physiopathology , Phosphorylation/drug effects , Serine/drug effects , Serine/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Eur J Cancer ; 37(17): 2247-56, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677115

ABSTRACT

Based on the role of p53 in the control of apoptosis following DNA damage, the status of the TP53 gene has been implicated as a major determinant of tumour responsiveness to cytotoxic therapies. In spite of the high frequency of TP53 mutations, small-cell lung cancer (SCLC) is recognised as one of the most chemoresponsive solid tumours. Since the relevance of the TP53 gene status in the modulation of tumour responsiveness is dependent on the molecular/biological context, in the present study, we have examined the relationship between chemosensitivity and susceptibility to apoptosis of a TP53-mutant human SCLC cell line. The cell line, in spite of TP53 mutation, retained an efficient response to genotoxic stress as documented by cells ability to modulate the p53 protein, arrest in the G1 and G2 phases of the cell cycle and its marked susceptibility to apoptosis following treatment with DNA damaging agents. Exposure to DNA-damaging agents caused an increase of c-Myc, a DNA damage-responsive transcription factor. An analysis of damage-induced apoptosis in the presence of an anti-Fas/CD95 inhibitory antibody indicated that Fas/CD95 was not required for the apoptotic response. The results support an implication of c-myc in sensitising cells to apoptosis, since inhibition of c-Myc expression with an antisense oligodeoxynucleotide (AS-ODN) almost abolished the drug-induced apoptotic response. In conclusion, the present results support a role for c-myc in the induction of apoptosis by genotoxic stress in the absence of a functional p53 and provide new insights into the mechanisms that may influence apoptosis in TP53-mutant cells. Elucidation of this pathway and of the possible cooperation with p53-dependent mechanisms may provide a basis for therapeutic intervention.


Subject(s)
Apoptosis/genetics , Carcinoma, Small Cell/genetics , DNA Damage/genetics , Genes, p53 , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Small Cell/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA, Neoplasm/genetics , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Doxorubicin/pharmacology , Gamma Rays , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Mutation , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
3.
Histol Histopathol ; 16(3): 719-26, 2001 07.
Article in English | MEDLINE | ID: mdl-11510961

ABSTRACT

The effects of electromagnetic fields on several processes related to cell physiology and proliferation are currently being investigated. Although the results are still not conclusive and even conflicting, there seems to be a fairly good agreement on the early effects of electromagnetic fields on the generation of free radicals and on Ca++-intracellular concentration and transport. To evaluate the long-lasting consequences of these precocious events, we examined the effects of short- and long-term magnetic field exposure on structural organization (cytokeratin or actin detection), proliferation (bromodeoxyuridine incorporation and propidium iodide staining), colony forming ability and viability (trypan blue exclusion test) of highly proliferating MCF-7 cells (from human breast carcinoma) and on slowly proliferating normal human fibroblasts (from healthy donors). Cells were exposed to either 20 or 500 microT sinusoidally oscillating (50Hz) magnetic fields for different lengths of time (1 to 4 days). Short (1 day)- and long (4 days)-time exposure to the two intensities did not affect MCF-7 growth and viability, colony number and size, or cellular distribution along the cell cycle; neither were the cell morphology and the intracellular distribution and amount of cytokeratin modified. Similarly, no modifications in the actin distribution and proliferative potential were observed in normal human fibroblasts. These findings suggest that under our experimental conditions, continuous exposure to magnetic fields does not result in any appreciable effect in both normal and tumor cells in vitro.


Subject(s)
Cell Division , Magnetics/adverse effects , Actins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival , DNA, Neoplasm/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Infant , Keratins/metabolism , Ki-67 Antigen/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay
4.
Br J Cancer ; 80(12): 1912-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10471039

ABSTRACT

Multinuclear platinum compounds were rationally designed to bind to DNA in a different manner from that of cisplatin and its mononuclear analogues. A triplatinum compound of the series (BBR 3464) was selected for preclinical development, since, in spite of its charged nature, it was very potent as cytotoxic agent and effective against cisplatin-resistant tumour cells. Anti-tumour efficacy studies were performed in a panel of human tumour xenografts refractory or poorly responsive to cisplatin. The novel platinum compound exhibited efficacy in all tested tumours and an impressive efficacy (including complete tumour regressions) was displayed in two lung carcinoma models, CaLu-3 and POCS. Surprisingly, BBR 3464 showed a superior activity against p53-mutant tumours as compared to those carrying the wild-type gene. The involvement of p53 in tumour response was investigated in an osteosarcoma cell line, SAOS, which is null for p53 and is highly sensitive to BBR 3464, and in the same cells following introduction of the wild-type p53 gene. Thus the pattern of cellular response was investigated in a panel of human tumour cells with a different p53 gene status. The results showed that the transfer of functional p53 resulted in a marked (tenfold) reduction of cellular chemosensitivity to the multinuclear platinum complex but in a moderate sensitization to cisplatin. In addition, in contrast to cisplatin, the triplatinum complex was very effective as an inducer of apoptosis in a lung carcinoma cell line carrying mutant p53. The peculiar pattern of anti-tumour activity of the triplatinum complex and its ability to induce p53-independent cell death may have relevant pharmacological implications, since p53, a critical protein involved in DNA repair and induction of apoptosis by conventional DNA-damaging agents, is defective in several human tumours. We suggest that the peculiar DNA binding properties of the triplatinum complex may contribute to the striking profile of anti-tumour efficacy. Taken together, the available information supports that anti-tumour activity of the novel compound is mediated by a mechanism different from that of conventional platinum complexes, and compounds of this series could represent a new class of promising anti-tumour agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Genes, p53 , Mutation , Neoplasms/drug therapy , Organoplatinum Compounds/therapeutic use , Animals , Cell Cycle/drug effects , Female , Humans , Male , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Neoplasms/genetics , Neoplasms/pathology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/toxicity , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/pathology , Polymorphism, Single-Stranded Conformational , Transplantation, Heterologous
5.
Mol Pharmacol ; 55(3): 528-34, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10051537

ABSTRACT

Multinuclear platinum compounds have been designed to circumvent the cellular resistance to conventional platinum-based drugs. In an attempt to examine the cellular basis of the preclinical antitumor efficacy of a novel multinuclear platinum compound (BBR 3464) in the treatment of cisplatin-resistant tumors, we have performed a comparative study of cisplatin and BBR 3464 in a human osteosarcoma cell line (U2-OS) and in an in vitro selected cisplatin-resistant subline (U2-OS/Pt). A marked increase of cytotoxic potency of BBR 3464 in comparison with cisplatin in U2-OS cells and a complete lack of cross-resistance in U2-OS/Pt cells were found. A detailed analysis of the cisplatin-resistant phenotype indicated that it was associated with reduced cisplatin accumulation, reduced interstrand cross-link (ICL) formation and DNA platination, microsatellite instability, and reduced expression of the DNA mismatch repair protein PMS2. Despite BBR 3464 charge and molecular size, in U2-OS and U2-OS/Pt cells, BBR 3464 accumulation and DNA-bound platinum were much higher than those observed for cisplatin. In contrast, the frequency of ICLs after exposure to BBR 3464 was very low. The time course of ICL formation after drug removal revealed a low persistence of these types of DNA lesions induced by BBR 3464, in contrast to an increase of DNA lesions induced by cisplatin, suggesting that components of the DNA repair pathway handle the two types of DNA lesions differently. The cellular response of HCT116 mismatch repair-deficient cells was consistent with a lack of influence of mismatch repair status on BBR 3464 cytotoxicity. Because BBR 3464 produces high levels of lesions different from ICLs, likely including intra-strand cross-links and monoadducts, the ability of the triplatinum complex to overcome cisplatin resistance appears to be related to a different mechanism of DNA interaction (formation of different types of drug-induced DNA lesions) as compared with conventional mononuclear complexes rather than the ability to overcome specific cellular alterations.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Organoplatinum Compounds/pharmacology , Apoptosis , Carboplatin/pharmacology , Cisplatin/pharmacokinetics , DNA Adducts/metabolism , DNA Damage , DNA Ligases/biosynthesis , DNA Ligases/physiology , DNA Repair , Drug Resistance, Neoplasm , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Tumor Cells, Cultured
6.
J Inorg Biochem ; 77(1-2): 59-64, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10626355

ABSTRACT

Multinuclear platinum compounds have been designed to circumvent the cellular resistance to conventional mononuclear platinum-based drugs. In this study we performed a comparative study of cisplatin and of the triplatinum complex BBR 3464 in a human osteosarcoma cell system (U2-OS) including an in vitro selected cisplatin-resistant subline (U2-OS/Pt). BBR 3464 was extremely potent in comparison with cisplatin in U2-OS cells and completely overcame resistance of U2-OS/Pt cells. In both cell lines, BBR 3464 accumulation and DNA-bound platinum were higher than those observed for cisplatin. On the contrary, a low frequency of interstrand cross-links after exposure to BBR 3464 was found. Differently from the increase of DNA lesions induced by cisplatin, kinetics studies indicated a low persistence of interstrand cross-link formation for BBR 3464. Western blot analysis of DNA mismatch repair proteins revealed a marked decrease of expression of PMS2 in U2-OS/Pt cells, which also exhibited microsatellite instability. Studies on DNA mismatch repair deficient and proficient colon carcinoma cells were consistent with a lack of influence of the DNA mismatch repair status on BBR 3464 cytotoxicity. In conclusion, the cytotoxic potency and the ability of the triplatinum complex to overcome cisplatin resistance appear to be related to a different mechanism of DNA interaction (formation of different types of drug-induced DNA lesions) as compared to conventional mononuclear complexes.


Subject(s)
Adenosine Triphosphatases , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Cisplatin/pharmacology , DNA Repair Enzymes , DNA-Binding Proteins , Drug Resistance, Neoplasm , Organoplatinum Compounds/pharmacology , Osteosarcoma/drug therapy , Adaptor Proteins, Signal Transducing , Base Pair Mismatch/drug effects , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Carcinoma/drug therapy , Carcinoma/genetics , Carrier Proteins , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Cross-Linking Reagents/pharmacology , DNA Polymerase beta/drug effects , DNA Polymerase beta/metabolism , DNA Repair/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Nuclear Proteins , Osteosarcoma/genetics , Osteosarcoma/metabolism , Platinum/pharmacokinetics , Proteins/drug effects , Proteins/genetics , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured
7.
Biochem Pharmacol ; 57(10): 1133-9, 1999 May 15.
Article in English | MEDLINE | ID: mdl-11230800

ABSTRACT

The amino sugar is recognized to be a critical determinant of the activity of anthracycline monosaccharides related to doxorubicin and daunorubicin. In an attempt to improve the pharmacological properties of such agents, novel anthracycline disaccharides have been designed in which the amino sugar, daunosamine, is separated from the aglycone by another carbohydrate moiety. In the present study, we examined the influence of the orientation of the second sugar residue on drug biochemical and biological properties in a series of closely related analogs. This structure-activity relationship study showed that the substitution of the daunosamine for the disaccharide moiety dramatically reduced the cytotoxic potency of the drug in the 4-methoxy series (daunorubicin analogs). In contrast, in the 4-demethoxy series (idarubicin analogs), the C-4 axial, but not the equatorial, configuration conferred a cytotoxic potency and antitumor activity comparable to that of doxorubicin. The configuration also influenced the drug's ability to stimulate topoisomerase II alpha-mediated DNA cleavage. Indeed, the glycosides with the equatorial orientation were ineffective as topoisomerase II poisons, whereas the compounds with axial orientation were active, although the daunorubicin analog exhibited a lower activity than the idarubicin analog. It is conceivable that the axial orientation allows an optimal interaction of the drug with the DNA-enzyme complex only in the absence of the methoxy group. Our results are consistent with a critical role of the sugar moiety in drug interaction with the target enzyme in the ternary complex.


Subject(s)
Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Disaccharides/pharmacology , Idarubicin/pharmacology , Animals , Anthracyclines/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , DNA/drug effects , DNA/metabolism , Disaccharides/chemistry , Disease Models, Animal , Humans , Idarubicin/chemistry , Idarubicin/therapeutic use , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Molecular Conformation , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 4(11): 2833-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829750

ABSTRACT

On the basis of a structure-activity study of a new series of anthracycline disaccharides, we recently identified a doxorubicin analogue (MEN 10755) with a promising antitumor activity. In the present study, to better support the pharmacological interest of MEN 10755, we extended the preclinical evaluation of antitumor efficacy to a large panel of 16 human tumor xenografts, which originated from different clinicopathological types. Tumors with typical multidrug-resistant phenotype were excluded because MEN 10755 was found unable to overcome resistance mediated by transport systems. In the doxorubicin-responsive series, MEN 10755 exhibited a higher activity in three of five tumors, as documented by a more marked tumor growth inhibition and an increased value of log-cell kill. In the series of doxorubicin-resistant tumors, MEN 10755 was found effective in 6 of 11 tumors (1 breast, 3 lung, and 2 prostate carcinomas). The overall response rates were 31% and 69% for doxorubicin and MEN 10755, respectively. The improvement in drug efficacy was also supported by a substantial increase in the long-term survivor rate of animals implanted with responsive tumors. Most of the tumors refractory to doxorubicin and responsive to MEN 10755 were characterized by overexpression of the antiapoptotic protein Bcl-2. In one of these tumors (MX-1 breast carcinoma), we examined the ability of MEN 10755 to induce phosphorylation of Bcl-2 after a single treatment with therapeutic doses. The results indicated that, unlike doxorubicin, MEN 10755 induced protein phosphorylation. A similar modification was produced by Taxol, which is known to be very effective against the tumor. The correlation between drug efficacy and Bcl-2 phosphorylation may underly a peculiar feature related to improvement of efficacy of the disaccharide analogue. In conclusion, the present study supports some favorable features of the novel doxorubicin analogue in terms of both efficacy and tolerability with comparison to doxorubicin, although the improvement is somewhat tumor- and schedule-dependent.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Disaccharides/therapeutic use , Doxorubicin/analogs & derivatives , Animals , Blotting, Western , Carcinoma/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
9.
Free Radic Biol Med ; 24(6): 913-23, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9607601

ABSTRACT

The stable nitroxide 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TEMPOL) is widely used as a probe in biophysical studies and as an antioxidant in several experimental models. The potential cytotoxic effects of TEMPOL were tested on a panel of human and rodent cell lines, and the nitroxide proved to be significantly more effective in inhibiting the growth of neoplastic than nonneoplastic cell lines after a 96-h exposure. More detailed studies on MCF-7/WT cells indicate that at least 24 h are necessary for TEMPOL to induce irreversible cell damage, which seems to be related to the reactivity of the nitroxyl group. This observation, together with the antagonistic effect of N-acetylcysteine, suggests an involvement of free radical-mediated processes. Cell cycle studies indicate a biphasic effect of TEMPOL, with a short-term accumulation of the cells in the G1 phase and a later increase in G2/M phase; the pattern of DNA fragmentation observed in TEMPOL-treated cells points to an apoptotic mode of cell death. In conclusion, our data suggest that, while the possible cytotoxic effects of TEMPOL should not be overlooked when using this compound as a biophysical probe or antioxidant, these same properties could be exploited as a novel approach to cancer chemotherapy, especially in tumor cells exhibiting unfavorable characteristics, such as a multidrug-resistant phenotype or loss of hormone receptors.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclic N-Oxides/pharmacology , Growth Inhibitors/pharmacology , Acetylcysteine/pharmacology , Animals , CHO Cells , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Cricetinae , DNA Fragmentation/drug effects , Drug Screening Assays, Antitumor , Electron Spin Resonance Spectroscopy , Female , Humans , Rats , Spin Labels , Tumor Cells, Cultured
10.
J Natl Cancer Inst ; 89(16): 1217-23, 1997 Aug 20.
Article in English | MEDLINE | ID: mdl-9274917

ABSTRACT

BACKGROUND: Although doxorubicin remains one of the most effective agents for the treatment of solid tumors, there is an intensive effort to synthesize doxorubicin analogues (compounds with similar chemical structures) that may have improved antitumor properties. We have synthesized a novel doxorubicin disaccharide analogue (MEN 10755) and have characterized some of its relevant biochemical, biologic, and pharmacologic properties. METHODS: The antitumor activity of this compound (MEN 10755) was studied in a panel of human tumor xenografts, including xenografts of A2780 ovarian tumor cells, MX-1 breast carcinoma cells, and POVD small-cell lung cancer cells. MEN 10755 was compared with doxorubicin according to the optimal dose and schedule for each drug. The drug's cytotoxic effects, induction of DNA damage, and intracellular accumulation were studied in A2780 cells. DNA cleavage mediated by the enzyme topoisomerase II was investigated in vitro by incubating fragments of simian virus 40 DNA with the purified enzyme at various drug concentrations and analyzing the DNA cleavage-intensity patterns. Drug-induced apoptosis (programmed cell death) in tumors was determined with the use of MX-1 and POVD tumor-bearing athymic Swiss nude mice. RESULTS: MEN 10755 was more effective than doxorubicin as a topoisomerase II poison and stimulated DNA fragmentation at lower intracellular concentrations. In addition, MEN 10755 exhibited striking antitumor activity in the treatment of human tumor xenografts, including those of the doxorubicin-resistant breast carcinoma cell line MX-1. CONCLUSIONS: The high antitumor activity of MEN 10755 in human tumor xenografts, including doxorubicin-resistant xenografts, and its unique pharmacologic and biologic properties make this disaccharide analogue a promising candidate for clinical evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Topoisomerases, Type II/drug effects , DNA, Neoplasm/drug effects , Doxorubicin/analogs & derivatives , Neoplasms, Experimental/drug therapy , Animals , Breast Neoplasms/drug therapy , Carcinoma, Small Cell/drug therapy , DNA Damage , Disaccharides/chemical synthesis , Doxorubicin/chemical synthesis , Female , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Ovarian Neoplasms/drug therapy , Time Factors , Transplantation, Heterologous
11.
Clin Cancer Res ; 3(6): 955-61, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9815771

ABSTRACT

Preclinical and clinical studies have documented the pharmacological interest in camptothecin derivatives in the treatment of resistant tumors. In particular, topotecan, a water-soluble derivative, exhibited promising activity in pretreated ovarian carcinoma. The present study investigated the pattern of tumor response in two human ovarian carcinoma xenografts and in their cisplatin-resistant sublines characterized by different mechanisms of drug resistance. In IGROV-1/Pt1 cells, cisplatin resistance has been ascribed to a reduced susceptibility to apoptosis as a consequence of p53 mutation and inactivation of its function. In the A2780 cisplatin-resistant subline, which retained the wild-type p53 gene status, the development of resistance has been possibly related to increased cell ability to repair drug-induced DNA damage. The in vivo results of the present study showed that topotecan could overcome the resistance in A2780/CP but not in IGROV-1/Pt1 tumor xenografts. The pattern of tumor response following in vivo topotecan treatment correlated with drug ability to induce apoptosis but not with its in vitro antiproliferative activity. The antitumor efficacy of topotecan in the four tumors reflected a different cell response to drug-induced DNA damage, as suggested by different perturbations of cell cycle progression. Indeed, only in the subline refractory to topotecan in vivo, IGROV-1/Pt1, did we observe a persistent arrest of the cells in the S-phase, resulting in a cytostatic and not a cytotoxic effect, since a low level of apoptosis was induced by the drug. In conclusion, the current results support that determination of drug-induced apoptosis is a useful predictor of tumor response to topotecan in ovarian carcinomas and suggest that p53 gene status may be a critical determinant of cell response to topoisomerase inhibitors.


Subject(s)
Apoptosis/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Topotecan/toxicity , Topotecan/therapeutic use , Animals , Cell Survival , Cisplatin/therapeutic use , Cisplatin/toxicity , Clone Cells , DNA Damage , DNA Repair , Drug Resistance, Neoplasm , Exons , Female , Genes, p53 , Humans , Mice , Mice, Nude , Mutation , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Apoptosis ; 2(6): 540-8, 1997.
Article in English | MEDLINE | ID: mdl-14646525

ABSTRACT

Since apoptosis is the primary mode of cell death induced by cisplatin, the role of apoptosis and apoptosis-related gene products in cisplatin resistance was investigated in four human cisplatin-resistant cell lines of different tumour type. A common feature of the resistant sublines was a reduced susceptibility to drug-induced apoptosis compared to parental sensitive lines. Loss of wild-type p53 function was not a general event associated with the development of drug resistance. An increased bcl-2 expression was found in resistant cells characterized by mutant p53 (A431/Pt and IGROV-1/Pt), whereas in osteosarcoma (U2-OS/Pt) and in ovarian carcinoma (A2780/CP) cells with wild-type p53, bcl-2 levels were markedly reduced. U2-OS/Pt cells had a 16-fold increase in the level of Bcl-xL protein. Stable transfection of U2-OS cells with bcl-xL cDNA conferred a low level of drug resistance to cisplatin, suggesting that overexpression of this gene contributes to the cisplatin-resistant phenotype of this osteosarcoma cell system. In conclusion, these observations suggest a variable contribution of apoptosis-related genes to cisplatin resistance depending on the biological background of the cell system and presumably reflecting different pathways of apoptosis.

13.
Mol Pharmacol ; 50(3): 603-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8794900

ABSTRACT

In an attempt to better understand the role of the cyclohexene ring (ring A) in the biochemical and pharmacological properties of anthracyclines related to doxorubicin and daunorubicin, we investigated the effects of introduction of a fluorine atom at position 8 of idarubicin (4-demethoxydaunorubicin) on drug molecular conformation and biochemical and pharmacological activities. The study showed that the stereochemistry of the substituent at position 8 influenced the "half-chair" conformation, so that in the (8R)-fluoroepimer the A ring retained the alpha half-chair conformation, which is the most stable for natural compounds (i.e., daunorubicin and doxorubicin), and the (8S)-fluoroepimers preferred the beta half-chair conformation. The (8R)-fluoroepimer was more effective than the (8S)-fluoroepimer and idarubicin in stimulating topoisomerase II-mediated DNA cleavage. Similarly, the epimer with the alpha conformation was markedly more potent than the (8S)-epimer as a cytotoxic agent in a variety of human tumor cell lines and was more effective as an antitumor agent in the treatment of an ovarian carcinoma xenograft. In addition, 8-fluoro derivatives were able to overcome the resistance to doxorubicin in a number of human tumor cell lines expressing different mechanisms of resistance. In conclusion, these findings provide evidence that drug interactions involving the external (nonintercalating) moiety of the anthracycline chromophore play an important role in determining pharmacological properties, including drug ability to induce DNA cleavage, and therefore their antitumor efficacy.


Subject(s)
Anthracyclines/chemistry , Anthracyclines/toxicity , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/toxicity , Ovarian Neoplasms/drug therapy , Animals , Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Cell Line , Cell Survival/drug effects , DNA Topoisomerases, Type II/metabolism , Daunorubicin/analogs & derivatives , Daunorubicin/therapeutic use , Daunorubicin/toxicity , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Doxorubicin/toxicity , Female , Fluorine , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous , Tumor Cells, Cultured
14.
Anticancer Drugs ; 7(5): 604-12, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8862730

ABSTRACT

The aim of the present study was to potentiate the cytotoxic effects of melphalan through pharmacological and physical modulators. The combination of the cytotoxic agent with ethacrynic acid, a glutathione-S-transferase pi (GST pi) inhibitor, or topotecan, a topoisomerase I inhibitor, or mild hyperthermia was investigated. The selected cell lines exhibited variable levels of expression of GST pi, DNA topoisomerase I and heat-shock proteins. Mild hyperthermia (42 degrees C) alone potentiated melphalan cytotoxicity, especially in the two cell lines exhibiting low basal levels of HSP70 expression. The combination of the GST inhibitor with melphalan resulted in a potentiation of drug cytotoxicity only in JR8 cells, one of the two cell lines which expressed high levels of GST pi mRNA and which were the less responsive to ethacrinic acid alone. A synergistic interaction between topotecan and melphalan was observed only in the cell lines expressing low levels of topoisomerase I even if all cell lines exhibited a comparable sensitivity to this agent. The results support an involvement of GST and DNA topoisomerase in cell defense and response to the alkylating agent. However, the variable potentiation of the cytotoxic effects of melphalan achieved in different cell systems suggests that factors other than the level of expression of the modulation target are responsible of such potentiation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Camptothecin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Ethacrynic Acid/pharmacology , Melphalan/pharmacology , Topoisomerase I Inhibitors , Blotting, Northern , Camptothecin/pharmacology , Drug Interactions , Humans , Melanoma , Temperature , Topotecan , Tumor Cells, Cultured/drug effects
15.
Int J Cancer ; 66(3): 342-6, 1996 May 03.
Article in English | MEDLINE | ID: mdl-8621255

ABSTRACT

The effect of topotecan, a topoisomerase I inhibitor, on ionizing radiation-induced cytotoxicity was studied in 2 human tumor cell lines characterized by a different expression of the target enzyme. The cytotoxicity of topotecan alone or in combination with radiation was assessed in exponentially growing non-small-cell lung cancer (H460) and glioblastoma (GBM) cells using the colony-forming assay. An isobologram method was used to evaluate the treatment interaction. An apparent supra-additive effect in cell killing following drug-radiation-combined treatment was observed only in GBM cells exposed to topotecan for 24 hr. In the case of H460 cells, interaction varied from a strong infra-additive effect at low radiation doses to a slight supra-additive effect when cells were exposed to radiation doses greater than 3 Gy. Northern blot analysis indicated that topoisomerase I expression in H460 cells was 8-fold higher than that of GBM cells. Although the H460 cell line exhibited an increased sensitivity to topotecan, only in the GBM cell line (which expressed a lower level of topoisomerase I) did the drug potentiate the radiation cytotoxicity. The observation that the radiosensitization by topotecan was related to topoisomerase I level is consistent with a putative role of the enzyme in processes involved in the repair of radiation damage. It is conceivable that the modulation of enzyme function results in an effective reduction of cellular capability for repair of radiation damage only if the enzyme is not over-expressed. Although a precise role of topoisomerase I in the cellular response to ionizing radiations (in particular, in DNA repair) remains to be documented, such results suggest the potential interest of topoisomerase I inhibitors in combination with radiation therapy for tumors expressing low topoisomerase I levels.


Subject(s)
Antineoplastic Agents/toxicity , Camptothecin/analogs & derivatives , Cell Survival/drug effects , Cell Survival/radiation effects , Blotting, Northern , Brain Neoplasms , Camptothecin/toxicity , Cell Line , DNA Topoisomerases, Type I/biosynthesis , Dose-Response Relationship, Radiation , Gene Expression , Glioblastoma , Humans , Lung Neoplasms , Radiation, Ionizing , Topoisomerase I Inhibitors , Topotecan , Tumor Cells, Cultured
16.
Cancer Res ; 56(3): 556-62, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8564971

ABSTRACT

P53 status may be a determinant of chemosensitivity of tumor cells; however, its involvement in cellular resistance to cisplatin remains uncertain. To investigate the relationships between p53 and the development of resistance to cisplatin, the p53 gene status was studied in ovarian carcinoma cell systems which included two cisplatin-resistant variants (IGROV-1/Pt 0.5 and IGROV-1/Pt 1) selected in vitro after prolonged drug exposure of the cisplatin-sensitive parental IGROV-1 cell line. IGROV-1/Pt 0.5 and IGROV-1/Pt 1 cell lines exhibited a degree of resistance of approximately 6 and 14, respectively, following 96-h exposure to the drug and were cross-resistant to other DNA-damaging agents (ionizing radiation and melphalan). Resistance to cisplatin paralleled a reduced cell susceptibility to cisplatin-induced apoptosis. DNA single-strand conformation polymorphism analysis of exons 5-9 demonstrated the presence of two mutants alleles at exon 8 in the two resistant cell lines, in contrast to the parental IGROV-1 cell line which exhibited the wild-type p53 gene. Direct DNA sequencing revealed that the mutations consist of two nucleotide changes in the DNA-binding domain at codons 270 (T/A) and 282 (C/T). The consecutive levels of p53 protein were lower in IGROV-1 than in IGROV-1/Pt cells. Following exposure to ionizing radiation or cisplatin, accumulation of the p53 protein was markedly enhanced only in the sensitive cells. Concomitantly, the expression of WAF-1 protein was strongly induced in the parental IGROV-1 cells, whereas WAF-1 protein remained undetectable in the IGROV-1/Pt 1 subline after DNA-damaging treatment. Consistent with this finding is the observation that ionizing radiation caused a different pattern of cell cycle perturbation in sensitive and resistant cells. Northern blot analysis demonstrated a marked reduction in bax mRNA levels in IGROV-1/Pt 1 cisplatin-resistant cells. Cotransfection assays with wild-type or mutant p53 expression plasmids and a reporter gene plasmid that utilized the bax gene promoter to drive transcription of chloramphenicol acetyltransferase were consistent with the role of p53 in regulation of bax expression in these cells. Taken together, these observations support a role for mutations of the p53 gene in the development of cisplatin resistance in ovarian cancer as a consequence of loss of the ability of p53 to transactivate bax, an apoptosis-inducing gene.


Subject(s)
Cisplatin/pharmacology , Genes, p53 , Mutation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Apoptosis/drug effects , Base Sequence , Blotting, Northern , Blotting, Western , Cell Cycle/physiology , Chloramphenicol O-Acetyltransferase/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , DNA Damage , Drug Resistance, Neoplasm , Female , Gene Expression , Genes, Reporter , Humans , Molecular Sequence Data , Ovarian Neoplasms/metabolism , Polymorphism, Single-Stranded Conformational , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Transcriptional Activation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/analysis , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...