Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
J Pharmacol Exp Ther ; 348(3): 360-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24345465

ABSTRACT

Since response to platinum-based therapy in non-small-cell lung cancer (NSCLC) is poor, the present study was designed to rationally identify novel drug combinations in cell models including the A549 cell line and the cisplatin-resistant subline A549/Pt, characterized by reduced sensitivity to cisplatin-induced apoptosis and by upregulation of efflux transporters of the ATP binding cassette (ABC) superfamily. Given the molecular features of these cells, we focused on compounds triggering apoptosis through different mechanisms, such as the mitochondria-targeting drug arsenic trioxide and the phenanthridine analog sanguinarine, which induce apoptosis through the extrinsic pathway. Sanguinarine, not recognized by ABC transporters, could overcome cisplatin resistance and, when used in combination with arsenic trioxide, was synergistic in A549 and A549/Pt cells. The arsenic trioxide/sanguinarine cotreatment upregulated genes implicated in apoptosis activation through the extrinsic pathway. Drug combination experiments indicated that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment improved arsenic trioxide/sanguinarine efficacy, a feature associated with a striking apoptosis induction, particularly in the cisplatin-resistant variant. Thus, a synergistic interaction between sanguinarine and arsenic trioxide could be obtained independent of relative cell sensitivity to arsenic trioxide, and an enhanced apoptosis induction could be achieved in combination with TRAIL through modulation of the extrinsic apoptotic pathway. Antitumor activity studies supported the interest of drug combinations including TRAIL in NSCLC, indicating that drug-resistant NSCLC cells can efficiently be killed by the combination of proapoptotic agents. Our results suggest that the molecular changes occurring in treated cells may be exploited to rationally hit surviving cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Benzophenanthridines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Isoquinolines/pharmacology , Lung Neoplasms/drug therapy , Oxides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Arsenic Trioxide , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , Drug Resistance, Neoplasm , Drug Synergism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
2.
Mini Rev Med Chem ; 9(9): 1102-12, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19689406

ABSTRACT

ATP binding cassette transporters are implicated in multidrug resistant phenotypes of tumor cells and may be cancer stem cell markers. Inhibitors of drug efflux pumps represent an emerging group of potentially useful agents for the improvement of antitumor therapy. Here we provide an overview of drug transporter functions and modulation.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Neoplasm/drug effects , Animals , Humans
3.
Mini Rev Med Chem ; 8(9): 856-68, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18691143

ABSTRACT

Small molecules targeting p53 represent an emerging group of potentially useful agents for the improvement of antitumor therapy. These modulators include agents that activate wild-type p53 or reactivate mutant p53 and inhibitors of p53 functions. Preclinical evidences support the interest of combination strategies with conventional antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Chemistry, Pharmaceutical/methods , Drug Design , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemical synthesis , Apoptosis , Drug Screening Assays, Antitumor , Humans , Models, Biological , Models, Chemical , Neoplasms/metabolism , Nuclear Proteins/chemistry , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/chemistry
4.
Curr Med Chem ; 13(27): 3291-305, 2006.
Article in English | MEDLINE | ID: mdl-17168852

ABSTRACT

The camptothecins are among the most promising antitumor agents endowed with a unique mechanism of action, because they act through inhibition of DNA topoisomerase I, an enzyme involved in regulating critical cellular functions including DNA replication, transcription and recombination. On the basis of the pharmacological interest of camptothecins in cancer chemotherapy, medicinal chemistry has played a crucial role in the development of novel analogs, and recently some compounds have emerged as promising agents for clinical evaluation. A major limitation to the clinical efficacy of camptothecin-containing therapies is represented by drug resistance. As with other cytotoxic drugs, clinical resistance to camptothecins may be a multifactorial phenomenon likely involving pharmacological and tumor-related factors. An additional problem in understanding clinically relevant resistance mechanisms is the observation that preclinical cell/tumor models may be not adequately predictive of clinical resistance. Here, we review the mechanisms of cell sensitivity/resistance to camptothecins and current approaches to overcome specific mechanisms, either by chemical modifications or by combination with modulating agents. In particular, the realization that most camptothecins are substrates for ATP binding cassette transporters has stimulated efforts in molecular design of novel non-cross-resistant analogs. Finally, a better understanding of the mechanism of cell response at a cellular level could help in defining new strategies to overcome resistance as well as chemical features required for efficacy.


Subject(s)
Camptothecin/pharmacokinetics , Drug Resistance, Neoplasm , Neoplasms/pathology , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Camptothecin/analogs & derivatives , Drug Design , Humans , Neoplasms/drug therapy
5.
Biochem Pharmacol ; 71(3): 231-8, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16303117

ABSTRACT

The expression of gamma-glutamyltransferase (GGT), a cell surface enzyme involved in cellular glutathione homeostasis, is often significantly increased in human tumors, and its role in tumor progression, invasion and drug resistance has been repeatedly suggested. As GGT participates in the metabolism of cellular glutathione, its activity has been mostly regarded as a factor in reconsitution of cellular antioxidant/antitoxic defences. On this basis, an involvement of GGT expression in resistance of cancer cells to cytotoxic drugs (in particular, cisplatin and other electrophilic agents) has been envisaged. Mechanistic aspects of GGT involvement in antitumor pharmacology deserve however further investigations. Recent evidence points to a more complex role of GGT in modulation of redox equilibria, with effects acting both intracellularly and in the extracellular microenvironment. Indications exist that the protective effects of GGT may be independent of intracellular glutathione, and derive rather from processes taking place at extracellular level and involving reactions of electrophilic drugs with thiol metabolites originating from GGT-mediated cleavage of extracellular glutathione. Although expression of GGT cannot be regarded as a general mechanism of resistance, the involvement of this enzyme in modulation of redox metabolism is expected to have impact in cellular response to several cytotoxic agents. The present commentary is a survey of data concerning the role of GGT in tumor cell biology and the mechanisms of its potential involvement in tumor drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Glutathione/metabolism , Neoplasms/metabolism , gamma-Glutamyltransferase/biosynthesis , Animals , Humans , Tumor Cells, Cultured
6.
J Natl Cancer Inst ; 77(2): 453-7, 1986 Aug.
Article in English | MEDLINE | ID: mdl-3461206

ABSTRACT

The ability of the synthetic cationic polypeptide poly-L-lysine (PLL) to induce differentiation of Friend murine erythroleukemia cells was examined with the use of polymers of different molecular weights. Like other membrane-interacting agents (polar solvents), all the polymers tested were effective in inducing cell differentiation. This effect was found to be molecular weight dependent, as already reported for other membrane-related actions of these polymers. Since anthracyclines also exert direct effects on the cell membrane, the activity of the daunorubicin-PLL conjugate was also examined. The covalent linking of the drug to the polyamino acid somewhat reduced the differentiation-inducing activity in this system. Indeed, free daunorubicin was found to inhibit the maturational process. Although the inducing effect was observed when the free PLL or daunorubicin-linked PLL was added alone, polymers enhanced differentiation produced by suboptimal concentrations of dimethyl sulfoxide (DMSO). Since PLL was inactive as an initiator of the maturation of a DMSO-resistant line, it is likely that some events (presumably membrane-related effects) involved in the multistep stimulation process are common to polar-planar solvents and the polycationic polymer.


Subject(s)
Daunorubicin/pharmacology , Leukemia, Erythroblastic, Acute/pathology , Polylysine/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dimethyl Sulfoxide/pharmacology , Friend murine leukemia virus , Mice , Molecular Weight
7.
J Natl Cancer Inst ; 84(22): 1710-6, 1992 Nov 18.
Article in English | MEDLINE | ID: mdl-1331483

ABSTRACT

BACKGROUND: Small-cell lung cancer (SCLC) is a highly chemosensitive tumor, but the recurrent disease that is common after initial response is often unresponsive to further chemotherapy. Although the mechanisms of drug resistance in SCLC have not been established, studies suggest that alterations of the nuclear enzyme DNA topoisomerase II may reduce the sensitivity of the cell to drug action. This enzyme is recognized as a primary target for cytotoxic activity of important antitumor agents. PURPOSE: In this study, we attempted to determine if altered forms of DNA topoisomerase II are responsible for reduced drug sensitivity. METHODS: We characterized a rearrangement of the topoisomerase II p170 gene (also known as TOP2) in a relatively chemoresistant SCLC cell line, NCI-H69, and compared topoisomerase II expression and activity in this line with those in the chemosensitive NCI-H187 cell line. Fragments of complementary DNA from the topoisomerase II gene were generated by polymerase chain reaction. Immunodetection was accomplished by using the monoclonal antibody 7E6 against the human topoisomerase II p170 isoform. Using DNA probes corresponding to different complementary DNA regions, we showed that the rearrangement was localized at the 3' terminus of one allele of the topoisomerase II gene. RESULTS: In addition to the normal 6.2-kilobase (kb) topoisomerase II messenger RNA (mRNA), the NCI-H69 line expressed a 7.4-kb topoisomerase II transcript, presumably encoded by the rearranged allele. Moreover, this transcript, although longer than the normal mRNA, lacked a substantial portion of the 3'-terminal p170 gene coding sequence. Topoisomerase II activity in nuclear extracts, as determined by the P4 phage DNA-unknotting assay, was more easily detected and measured at lower NaCl concentrations in NCI-H69 than in NCI-H187 cells. CONCLUSION: These results are consistent with the hypothesis that the chemoresistant NCI-H69 cell line may express, in addition to the normal enzyme, an altered topoisomerase II enzyme possibly encoded by the 7.4-kb mRNA, which in turn may be transcribed from the rearranged gene allele. IMPLICATION: These observations emphasize the role of topoisomerase II in determining drug sensitivity and suggest that such gene rearrangements may contribute to resistance of SCLC cells to topoisomerase II inhibitors.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Topoisomerases, Type II/genetics , Lung Neoplasms/genetics , Alleles , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/genetics , Gene Expression , Gene Rearrangement , Humans , RNA, Messenger/genetics , Restriction Mapping , Tumor Cells, Cultured
8.
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
9.
Med Chem ; 2(3): 227-37, 2006 May.
Article in English | MEDLINE | ID: mdl-16948468

ABSTRACT

The aim of this study was to investigate cellular response to several ruthenium(III), chromium(III) and rhodium(III) compounds carrying bidentate beta-diketonato ligands: [(acac)--acetylacetonate ligand, (tfac)--trifluoroacetylacetonate ligand]. Cell sensitivity studies were performed on several cell lines (A2780, cisplatin-sensitive and -resistant U2-OS and U2-OS/Pt, HeLa, B16) using growth-inhibition assay. Effect of intracellular GSH depletion on cell sensitivity to the agents was analyzed in A2780 cells. Flow cytometry was used to assess apoptosis by Annexin-V-FITC/PI staining, and to analyze induction of caspase-3 activity. Possible DNA binding/damaging affinity was investigated, by inductively coupled mass spectrometry, and by 14C-thymidine / 3H-uridine incorporation assay. Cell sensitivity studies showed that the pattern of sensitivity to Ru(tfac)3 complex of the two cisplatin-sensitive/-resistant osteosarcoma cell lines, U2-OS and U2-OS/Pt, was similar to that of A2780 cells (72 h exposure), with the IC50 being around 40 microM. The growth-inhibitory effect of Ru(acac)3 ranged over 100 microM, while Cr(III) and Rh(III) complexes were completely devoid of antitumor action in vitro. Ru(tfac)3 exhibited strong potential for apoptosis induction on A2780 cells (up to 40%) and caused cell cycle arrest in the S phase as well as decrease of the percent of G1 and G2 cells. Ru(acac)3-induced apoptosis was slightly higher than 10%, whereas activation of caspase-3 in HeLa cells was moderate. DNA binding study revealed that only Cr(acac)3 was capable of binding DNA, while Cr(III) and Ru(III) compounds possess potential to inhibit DNA/RNA synthesis. In conclusion, only Ru(III) complexes showed potential for antitumor action.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Chromium/pharmacology , Rhodium/pharmacology , Ruthenium/pharmacology , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , Flow Cytometry , Humans , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/drug effects
10.
Cancer Res ; 46(11): 5499-503, 1986 Nov.
Article in English | MEDLINE | ID: mdl-3463414

ABSTRACT

Single-strand DNA breaks induced by chromophore-modified anthracyclines related to doxorubicin (including 11-deoxydaunorubicin, 4-demethoxydaunorubicin, 4-demethoxy-11-deoxy-4'-epi-daunorubicin, 4-demethyl-6-O-methyldoxorubicin) in cultured P388 leukemia cells were determined by the filter alkaline elution method. The tested analogues differed markedly in their cytotoxic potency. In the range of cytotoxic concentrations, 11-deoxydaunorubicin produced single-strand DNA break frequency of the same order of magnitude as that produced by doxorubicin, while other derivatives caused much more marked damage on DNA than doxorubicin. Since DNA breaks were found to be protein associated, the type of DNA damage produced by all tested derivatives presumably resulted by action of DNA topoisomerases II, as proposed for doxorubicin and other intercalating agents. Although the "potent" (with respect to DNA damage) derivatives, except 4-demethyl-6-O-methyldoxorubicin, showed an increased cellular drug accumulation as compared to doxorubicin, this did not account for the marked differences in ability to damage DNA. 4-Demethyl-6-O-methyldoxorubicin was the most efficient derivative, producing DNA breaks in a lower range of cellular drug content. A striking biphasic dose-response curve was observed for the 4-demethoxy derivatives, suggesting a complex mechanism of interaction among drug, DNA, and enzyme. A lack of correlation was noted among DNA binding affinity, induction of strand breaks, and cytotoxic activity of these chromophore-modified derivatives. From these observations, it is suggested that multiple actions of anthracyclines at the DNA level are responsible for their cytotoxic activity, which is not simply related to inhibition of a specific DNA-dependent enzyme and/or function.


Subject(s)
DNA Damage , DNA, Neoplasm/drug effects , Animals , Antibiotics, Antineoplastic , Cell Survival/drug effects , Cells, Cultured , Cross-Linking Reagents , DNA/metabolism , Daunorubicin/analogs & derivatives , Daunorubicin/metabolism , Daunorubicin/toxicity , Leukemia P388 , Mice , Naphthacenes/metabolism , Naphthacenes/toxicity , Structure-Activity Relationship
11.
Cancer Res ; 55(2): 312-7, 1995 Jan 15.
Article in English | MEDLINE | ID: mdl-7812964

ABSTRACT

Antitumor drugs stimulate topoisomerase II-mediated DNA cleavage in a DNA sequence-specific manner. The drug sequence specificity is often very similar among antitumor agents of the same chemical class. In this work, we demonstrate, however, that 3'-epidaunorubicin has a markedly different sequence specificity as compared with the parent drugs daunorubicin and doxorubicin. The analogue stimulates distinct cleavage intensity patterns in agarose and sequencing gels with two different DNA substrates, although its cleaving activity was lower than that of daunorubicin. A statistical analysis of 44 sites specifically stimulated by the analogue showed that a major difference between the analogue and parent drugs was at position -2, where a guanine is highly preferred by the analogue, whereas parent drugs prefer a thymine and exclude instead a guanine. Interestingly, an analogue with no substituents at the 3'-C of the sugar was able to stimulate DNA cleavage at sites stimulated by parent drugs as well as at those stimulated by 3'-epidaunorubicin. In contrast, the presence of a 2'-OH or a 3'-epi-OH in the sugar moiety and the removal of the OH at 9-C of the A ring did not alter the drug site selectivity, in agreement with several other modifications studied previously. DNA binding affinities of studied agents were not related to drug sequence specificity. The data demonstrate a critical role of the 3' position for optimal anthracycline interactions in the ternary complex. The findings, for the first time, establish a clear relationship between a specific drug substituent and base sequence selectivity and indicate putative DNA- and enzyme-interacting domains of the anthracycline molecule.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , DNA Topoisomerases, Type II/drug effects , DNA/drug effects , Antibiotics, Antineoplastic/metabolism , Base Sequence , DNA/metabolism , DNA Topoisomerases, Type II/chemistry , Daunorubicin/chemistry , Daunorubicin/metabolism , Daunorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Idarubicin/chemistry , Idarubicin/metabolism , Idarubicin/pharmacology , Molecular Sequence Data , Structure-Activity Relationship
12.
Cancer Res ; 47(14): 3752-6, 1987 Jul 15.
Article in English | MEDLINE | ID: mdl-3474061

ABSTRACT

DNA single-strand and double-strand breaks produced by doxorubicin and two anthracycline derivatives (4-demethoxy-daunorubicin and 4'-deoxy-4'-iododoxorubicin) were measured in doxorubicin-sensitive and -resistant P388 leukemia cell lines, using filter elution methods, and compared with cellular drug accumulation to account for major differences in their cytotoxic activities and cross-resistance. The increased cytotoxic potency of the two derivatives reflects at least in part the enhanced drug accumulation by cells that results from their increased lipophilicity. However, the level of protein-linked DNA breaks was not directly related to cellular accumulation of drug analogues. It is possible that enhanced cytotoxicity may also be the consequence of the greatly enhanced ability of analogues to cause DNA strand breaks. The resistant line showed only a modest degree of resistance to both anthracycline derivatives compared with the high degree of resistance to doxorubicin. Although for all the anthracyclines tested drug accumulation was reduced in the resistant line, this did not correlate with the degree of resistance. A differential sensitivity of resistant and parental cell lines to DNA cleavage activity was consistently found for all three drugs tested. However, in contrast to a lack of effect of doxorubicin, the derivatives caused appreciable DNA strand breakage in resistant cells. The enhanced ability of these analogues to break DNA in resistant cells is consistent with the slight cross-resistance with doxorubicin. DNA double-strand breaks produced in isolated nuclei from these cells paralleled the pattern found in whole cells, thus indicating that a nuclear alteration, presumably involving DNA topoisomerases, is associated with anthracycline resistance. Our findings strongly support the hypothesis that anthracycline resistance in these cell variants may be mediated by multiple mechanisms, involving alterations of plasma membrane and changes of nuclear enzymatic activities responsible for DNA strand breaks.


Subject(s)
DNA/drug effects , Leukemia P388/genetics , Leukemia, Experimental/genetics , Animals , Antibiotics, Antineoplastic , Cell Line , Cell Nucleus/drug effects , Daunorubicin/analogs & derivatives , Daunorubicin/therapeutic use , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Idarubicin , Leukemia P388/drug therapy , Mice , Naphthacenes/therapeutic use , Nucleic Acid Conformation/drug effects
13.
Cancer Res ; 52(7): 1666-74, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1312895

ABSTRACT

Patterns of drug sensitivities in relation to topoisomerase II gene expression and activity were studied in eight human lung cancer cell lines not selected in vitro for drug resistance. The cytotoxicities of doxorubicin, etoposide, teniposide, cisplatin, camptothecin, and 5-fluorouracil were measured and, remarkably, these unselected cell lines were shown to have a common pattern of multidrug sensitivity, i.e., a multidrug sensitivity phenotype. In fact, drug sensitivities were significantly correlated with each other in the studied cell lines, the correlation being best for the topoisomerase II-targeted agents and cisplatin, less strong with camptothecin, and weak with 5-fluorouracil. Almost 1-log range difference of topoisomerase II gene expression was found in these cell lines, and this was not explained by the cell-doubling time or cell cycle distribution. The level of topoisomerase II gene expression was positively and highly correlated with the cell sensitivity to epipodophyllotoxins, doxorubicin, and cisplatin in seven cell lines. Although weaker, an association was also observed between topoisomerase II gene expression and camptothecin cytotoxicity, while no association was observed with 5-fluorouracil. However, a non-small cell lung cancer cell line with neuroendocrine properties had very low levels of expression of the topoisomerase II gene, despite being highly sensitive to all drugs tested. The levels of topoisomerase I gene expression were not found to be correlated with the cytotoxicity of any drug tested. A specific enzymatic activity assay and a teniposide-stimulated DNA cleavage assay showed that the extent of active topoisomerase II present in nuclear extracts paralleled the level of topoisomerase II gene expression. Furthermore, in addition to the normal transcript, an abnormally sized topoisomerase II message and a rearrangement of the topoisomerase II gene were detected in a poorly sensitive small cell lung cancer cell line. Therefore, low levels of topoisomerase II gene expression, and possibly mutations, may predict a reduced sensitivity of unselected human lung cancer cell lines to several drugs, including agents with a cellular target other than topoisomerase II. It is hypothesized that topoisomerase II might be involved in a common pathway of cell death induced by drugs in tumor cell lines which present a multidrug sensitivity phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , DNA Topoisomerases, Type II/genetics , Adenocarcinoma , Blotting, Northern , Blotting, Southern , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung , Carcinoma, Small Cell , Cell Line , Cisplatin/pharmacology , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Fluorouracil/pharmacology , Humans , Lung Neoplasms , Phenotype , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Restriction Mapping , Teniposide/pharmacology
14.
Cancer Res ; 49(8): 2022-7, 1989 Apr 15.
Article in English | MEDLINE | ID: mdl-2702645

ABSTRACT

Formation and persistence of DNA single- and double-strand breaks stimulated by doxorubicin, 9-deoxydoxorubicin, or 4-demethyl-6-deoxydoxorubicin in murine leukemia P388 cells were compared in relation to drug DNA affinity, cellular pharmacokinetics, and cytotoxicity. Although cellular uptake and retention and DNA affinity of the anthracycline derivatives were similar to those of the parent drug, cytotoxic potency was quite different, 9-deoxydoxorubicin being much less cytotoxic than doxorubicin, and 4-demethyl-6-deoxydoxorubicin the most effective agent. After 1-h exposure of cells to cytotoxic drug levels, the extent of DNA strand breaks produced by 4-demethyl-6-deoxydoxorubicin was greater than that produced by doxorubicin, whereas 9-deoxydoxorubicin induced very few DNA breaks. As for the parent drug, proteolytic treatment of cell lysates on the filter was needed to detect DNA cleavage produced by the analogues. A linear increase of DNA breaks was observed for 2 h following 4-demethyl-6-deoxydoxorubicin or doxorubicin addition; by contrast, DNA break levels reached a plateau after 45 min of exposure to 9-deoxydoxorubicin. DNA lesions produced by the derivatives persisted, and doxorubicin-induced DNA breaks even increased after drug removal, indicating an absence of DNA break resealing under our conditions. These observations indicate that modifications of the chromophore moiety of the anthracycline may enhance both drug cytotoxicity and specificity of drug-target interactions, and thus provide further strong evidence that the anthracycline effect on DNA integrity is a critical aspect of the mechanism of drug action.


Subject(s)
DNA Damage , DNA/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Leukemia P388/pathology , Leukemia, Experimental/pathology , Animals , Cell Survival/drug effects , DNA, Single-Stranded/drug effects , Doxorubicin/pharmacokinetics , Mice , Structure-Activity Relationship
15.
Cancer Res ; 57(9): 1710-6, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9135013

ABSTRACT

In contrast to other topoisomerase II poisons, the microbial terpenoid clerocidin was shown to stimulate irreversible topoisomerase II-mediated DNA cleavage. To establish the structural determinants for drug activity, in this study we have investigated intensity patterns and sequence specificity of clerocidin-stimulated DNA cleavage using 5'-end 32P-labeled DNA fragments. At a majority of the sites, clerocidin-stimulated cleavage did not revert upon NaCl addition; nevertheless, at some sites, cleavage completely reverted. Statistical analyses showed that drug-preferred bases were different in the two cases: guanine and cytosine were highly preferred at position -1 at irreversible and reversible sites, respectively. These results demonstrated that cleavage irreversibility was site selective and required a guanine at the 3' end of the cut. Further experiments revealed that some irreversible sites showed an abnormal electrophoretic mobility in sequencing gels with respect to cleaved bands generated by 4-(9-acridinylamino)methanesulfon-m-anisidide, suggesting a chemical alteration of the DNA strand. Interestingly, the ability to stimulate irreversible cleavage progressively decreased over time when clerocidin was stored in ethanol. Under these conditions, nuclear magnetic resonance measurements demonstrated that the drug underwent structural modifications that involved the C-12-C-15 side chain. Thus, the results indicate that a specific moiety of clerocidin may react with the DNA (guanine at -1) in the ternary complex, resulting in cleavage irreversibility and in altered DNA mobility in sequencing gels.


Subject(s)
DNA Topoisomerases, Type II/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Survival/drug effects , Cytotoxins , DNA Damage , DNA, Neoplasm/chemistry , DNA, Viral/chemistry , Diterpenes/chemistry , Diterpenes/pharmacology , Ethanol , Humans , Magnetic Resonance Spectroscopy , Tumor Cells, Cultured
16.
Cancer Res ; 56(8): 1855-62, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620504

ABSTRACT

DNA cleavage stimulated by different topoisomerase II inhibitors shows in vitro a characteristic sequence specificity. Since chromatin structure and genome organization are expected to influence drug-enzyme interactions and repair of drug-induced DNA lesions, we investigated topoisomerase II DNA cleavage sites stimulated by teniposide (VM-26), 4-demethoxy-3'-deamino-3'-hydroxy-4'-epi-doxorubicin (dh-EPI, a doxorubicin derivative), 4'-(9-acridinylamino)-methanesulfon-m-anisidide, and amonafide in the histone gene locus and satellite III DNA of Drosophila cells with Southern blottings and genomic sequencing by primer extension. VM-26 stimulated cleavage in the satellite III DNA, whereas the other studied drugs did not. All four drugs stimulated cleavage in the histone gene cluster, but they yielded drug-specific cleavage intensity patterns. Cleavage sites by dh-EPI and VM-26 were sequenced in the histone H2A gene promoter and were shown to be distinct. DNA cleavage analysis in cloned DNA fragments with Drosophila topoisomerase II showed that drugs stimulated the same sites in vivo and in vitro. Strand cuts were in vivo staggered by 4 bases, and base sequences at major dh-EPI and VM-26 sites completely agreed with known in vitro drug sequence specificities. Moreover, DNA cleavage reverted faster in the satellite III than in the histone repeats. While stimulating similar levels of DNA breakage in bulk genomic DNA, dh-EPI and VM-26 markedly differed for cleavage extent and reversibility in specific chromatin loci. The results demonstrate a high heterogeneity in the localization, extent, and reversibility of drug-stimulated DNA cleavage in the chromatin of living cells.


Subject(s)
Amsacrine/pharmacology , Chromatin/metabolism , DNA Topoisomerases, Type II/metabolism , DNA/metabolism , Enzyme Inhibitors/pharmacology , Teniposide/pharmacology , Animals , Base Sequence , Binding Sites , Cell Line , Chromatin/drug effects , DNA/drug effects , DNA Primers , DNA, Satellite/genetics , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Drosophila melanogaster , Genes, Insect , Histones/genetics , Kinetics , Molecular Sequence Data , Promoter Regions, Genetic , Substrate Specificity , Topoisomerase II Inhibitors
17.
Cancer Res ; 36(6): 1962-6, 1976 Jun.
Article in English | MEDLINE | ID: mdl-773533

ABSTRACT

The effects of 4'-epi-daunorubicin, 4'-epi-adriamycin, and the corresponding beta anomers on the in vitro activity of Escherichia coli DNA polymerase I and RNA polymerase were determined and compared with the effects of the parent compounds. The observed effects parallel the cytotoxic activities, assayed by inhibition of mouse embryo fibroblast proliferation, and the inhibitory activities on DNA synthesis in cultured cells. The data indicate that the inverted configuration at position 1 of the amino sugar results in a markedly reduced biological activity. This conclusion is also substantiated by the data obtained with the beta anomer of adriamycin. A preliminary investigation on the binding properties of these derivatives suggests that the inverted configuration at C-1' produces a significant decrease in the binding to DNA. In contrast, epimerization at position 4' did not produce any significant change in activity. The relationship between biological and biochemical activity and DNA binding properties of the tested compounds are discussed with particularly reference to antitumor activity.


Subject(s)
Daunorubicin/analogs & derivatives , Doxorubicin/analogs & derivatives , Cell Division/drug effects , Cells, Cultured , DNA/metabolism , DNA Nucleotidyltransferases/metabolism , DNA-Directed RNA Polymerases/metabolism , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Escherichia coli/enzymology , Fibroblasts/metabolism , Stereoisomerism , Structure-Activity Relationship
18.
Cancer Res ; 35(3): 754-60, 1975 Mar.
Article in English | MEDLINE | ID: mdl-163690

ABSTRACT

The effects of the anthracycline antiboties, daunomycin and adriamycin, on the DNA-directed activities of DNA polymerases from murine sarcoma virus, rat liver (high-molecular-weight species), Escherichia coli, and Micrococcus luteus were determined. Under all conditions tested, these compounds had greater inhibitory effect against the viral polymerase than against cellular polymerase. The inhibition of murine sarcoma virus DNA polymerase by daunomycin was competitive with respect to DNA. For viral DNA polymerase it was concluded that the inhibition was predominatly caused by the interaction of duanomycin with the primer-template DNA. Also, an appreciable reversal of the daunomycin-induced inhibition of this polymerase by an increase in Mg-2+ concentration is consistent with the conclusion derived by competition experiments. In contrast, the inhibition of both rat liver and M. luteus DNA polymerases was essentially noncompetitive with DNA. Also, bacterial enzymes wer e less sensitive to inhibition by these drugs than the virion polymerase. The strong and preferential inhibiton of viral DNA polymerase is discussed in relation to a differential sensitivity of normal as compared to tumor cells observed in some cell lines.


Subject(s)
DNA Nucleotidyltransferases/antagonists & inhibitors , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Animals , Binding, Competitive , DNA/metabolism , Escherichia coli/enzymology , Gammaretrovirus/enzymology , In Vitro Techniques , Liver/enzymology , Magnesium/pharmacology , Micrococcus/enzymology , Rats , Sarcoma/microbiology , Templates, Genetic
19.
Cancer Res ; 50(15): 4501-3, 1990 Aug 01.
Article in English | MEDLINE | ID: mdl-2369729

ABSTRACT

The efficacy of an originally developed photodynamic approach, using topical administration of tetraphenylporphinesulfonate as the photosensitizer, was evaluated in a series of 292 basal cell carcinoma lesions (less than 2-mm thick) in 50 treated patients. The lack of indication for conventional therapies was the main selection criterion. The photosensitizing agent (2% solution) was topically applied at 0.1 ml/cm2, followed by light irradiation with a dye laser emitting at 645 nm (120 or 150 J/cm2). After initial treatment, all lesions responded, with 273 (93.5%) complete responses. Recurrences were observed in 29 (10.6%). A second application of photoradiation was performed in 15 persistent lesions and 11 relapsed lesions, producing 19/26 complete responses. Our results suggest that this technique can be considered a promising alternative treatment modality in selected cases of superficial basal cell carcinomas.


Subject(s)
Carcinoma, Basal Cell/drug therapy , Photochemotherapy , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Skin Neoplasms/drug therapy , Aged , Biopsy , Carcinoma, Basal Cell/pathology , Follow-Up Studies , Humans , Skin Neoplasms/pathology
20.
Cancer Res ; 59(5): 1036-40, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10070960

ABSTRACT

Clinically available taxanes represent one of the most promising class of antitumor agents, despite several problems with their solubility and toxicity. In an attempt to improve the pharmacological profile of taxanes, a new series of analogues was synthesized from 14beta-hydroxy-10-deacetylbaccatin III and tested in a panel of human tumor cell lines. On the basis of the pattern of cytotoxicity and lack of cross-resistance in tumor cell lines expressing the typical multidrug-resistant phenotype, a compound (IDN5109) was selected for preclinical development. A comparative efficacy study of IDN5109 and paclitaxel was performed using a large panel of human tumor xenografts, characterized by intrinsic (seven tumors) or acquired (four tumors) resistance to cisplatin or doxorubicin, including four ovarian, one breast, one cervical, three lung, one colon, and one prostatic carcinoma. Drugs were delivered i.v. according to the same schedule (four times every 4th day). IDN5109 achieved a very high level of activity (percentage tumor weight inhibition >70%; log10 cell kill >1) in all but one of the tested tumors. Compared to paclitaxel, IDN5109 exhibited a significantly superior activity in six tumors (including the four tumors that were resistant to paclitaxel) and a comparable activity against the other five paclitaxel-responsive tumors. Additional advantages of IDN5109 over paclitaxel were also suggested by its toxicity profile. IDN5109 was not only less toxic (maximal tolerated doses were 90 and 54 mg/kg for IDN5109 and paclitaxel, respectively), but it also appeared to be endowed with a reduced neurotoxic potential and an improved profile of tolerability compared to the parent drug. Furthermore, the best antitumor efficacy was often already reached with doses lower than the maximal tolerated dose, suggesting an improved therapeutic index for the new drug. In conclusion, the results support the preclinical interest of IDN5109 in terms of the toxicity profile and of the efficacy with particular reference to the ability to overcome multiple mechanisms of drug resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/toxicity , Animals , Antineoplastic Agents, Phytogenic/toxicity , Breast Neoplasms/drug therapy , Cell Division/drug effects , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Drug Resistance, Multiple , Female , Humans , Male , Mice , Mice, Nude , Paclitaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL