Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Med Chem ; 49(26): 7740-53, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17181156

ABSTRACT

The biological activity of indenoisoquinoline topoisomerase I inhibitors is significantly enhanced by nitration of the isoquinoline ring. In the present study, nitrated analogues were synthesized with the indenone ring substituted with methoxy groups to further explore a previously identified structure-activity relationship between the nitrated isoquinoline ring and a methylenedioxy-substituted indenone ring. The results indicate that a single methoxy group at the 9-position of an indenoisoquinoline affords superior biological activity. Hypothetical binding models have been developed to rationalize these results, and they indicate that pi-stacking between the indenoisoquinolines and the DNA base pairs, as visualized by electrostatic complementarity, is important for the intercalation and biological activity of the indenoisoquinoline analogues. Collectively, the analysis of methoxy groups on the indenone ring also illustrates a strict steric requirement for substituents extending toward the nonscissile DNA backbone and emphasizes a need for planarity to afford potent biological activity.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indenes/pharmacology , Isoquinolines/pharmacology , Nitrates/chemistry , Topoisomerase I Inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , DNA Topoisomerases, Type I/metabolism , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indenes/chemical synthesis , Indenes/chemistry , Isoquinolines/chemical synthesis , Isoquinolines/chemistry , Models, Molecular , Structure-Activity Relationship
2.
J Med Chem ; 49(21): 6283-9, 2006 Oct 19.
Article in English | MEDLINE | ID: mdl-17034134

ABSTRACT

In connection with an ongoing investigation of indenoisoquinoline topoisomerase I (Top1) inhibitors as potential therapeutic agents, the pharmacophore possessing di(methoxy) and methylenedioxy substituents was held constant, and new derivatives were synthesized with nitrogen heterocycles appended to the lactam side chain. Compounds were evaluated for Top1 inhibition and for cytotoxicity in the National Cancer Institute's human cancer cell screen. Some of the more potent derivatives were also screened for in vivo activity in a hollow fiber assay. The results of these studies indicate that lactam substituents possessing nitrogen heterocycles can provide highly cytotoxic compounds with potent Top1 inhibition. Molecular modeling of these compounds in complex with DNA and Top1 suggests that some of the lactam substituents are capable of interacting with the DNA base pairs above and below the site of intercalation and/or with Top1 amino acid residues, resulting in increased biological activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , DNA Topoisomerases, Type I/chemistry , Indenes/chemical synthesis , Isoquinolines/chemical synthesis , Lactams/chemical synthesis , Topoisomerase I Inhibitors , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Indenes/chemistry , Isoquinolines/chemistry , Lactams/chemistry , Models, Molecular , Morpholines/chemical synthesis , Morpholines/chemistry , Piperazines/chemical synthesis , Piperazines/chemistry , Piperidines/chemical synthesis , Piperidines/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
3.
J Med Chem ; 49(17): 5129-40, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16913702

ABSTRACT

The indenoisoquinolines represent a class of non-camptothecin topoisomerase I (Top1) inhibitors that exert cytotoxicity by trapping the covalent complex formed between DNA and Top1 during relaxation of DNA supercoils. As an ongoing evaluation of Top1 inhibition and anticancer activity, indenoisoquinolines were linked via their lactam side chains to provide polyamines end-capped with intercalating motifs. The resulting bisindenoisoquinolines were evaluated for cytotoxicity in the National Cancer Institute's human cancer cell screen and for Top1 inhibition. Preliminary findings suggested that the 2-3-2 and 3-3-3 linkers, referring to the number of carbons between nitrogen atoms, were optimal for both potent Top1 inhibition and cytotoxicity. Using optimized linkers, bisindenoisoquinolines were synthesized with nitro and methoxy substituents on the aromatic rings. The biological results for substituted compounds revealed a disagreement between the structure-activity relationships of monomeric indenoisoquinolines and bisindenoisoquinolines as Top1 inhibitors, but cytotoxicity was maintained for both series of compounds.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Indenes/chemical synthesis , Indenes/pharmacology , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Topoisomerase I Inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Indenes/chemistry , Isoquinolines/chemistry , Models, Molecular , Molecular Structure , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 16(7): 1846-9, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16442283

ABSTRACT

A method has been developed that relies on a two-step, one-pot condensation between phthalide and 2-carboxybenzaldehydes to provide benz[d]indeno[1,2-b]pyran-5,11-diones in a multi-gram fashion. Treatment of these compounds with a primary amine allows rapid access to various N-substituted indenoisoquinolines, whose in vitro anticancer activity and topoisomerase I inhibition have been evaluated.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Pyrans/chemical synthesis , Topoisomerase I Inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyrans/chemistry , Pyrans/pharmacology
5.
Cancer Chemother Pharmacol ; 57(2): 135-44, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16151810

ABSTRACT

The effect of 7-alkyl substitutions on growth inhibition in seven Camptothecin (CPT) ring systems with various groups at the ten position was evaluated in three human breast cancer cell lines that model (1) hormone-sensitive (MCF-7/wt), (2) hormone insensitive (MDA-MB-231), or (3) alkylator-resistant (MCF-7/4-hc) forms of disease. To assess the impact of persistence of cleavage complexes on antiproliferative activity, a post-exposure recovery period in drug-free medium was incorporated into the growth inhibition assay. This modification produced on average a twofold reduction in the growth inhibition endpoint (the IC50), suggesting a greater apoptotic response. The results further revealed a three log range in potency from a mean IC50 of 2 nM (7-butyl-10,11-methylenedioxy-CPT) to 2.5 microM (7-bromomethyl-10-hydryoxy-CPT). Increasing 7-alkyl chain length in six of the ten-substituted CPTs enhanced potency, which was directly correlated with persistence of topoisomerase I-induced DNA cleavage complexes in 10-hydroxy, 10-methoxy, and 10,11-methylenedioxy substituted CPTs. Modeling of the binding mode of 7-butyl-10-amino-CPT revealed a direct hydrogen bond contact for the 10-amino to the side chain of Glu-356 of Core Subdomain I of top1 in addition to known contacts found for other camptothecins. More important, residues 350-356 and 425-431 of Core Subdomain I may provide induced fit stabilization to the lipophilic alkyl moiety at the seven position.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Camptothecin/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Tumor Cells, Cultured
6.
Cancer Chemother Pharmacol ; 58(1): 62-72, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16331501

ABSTRACT

PURPOSE: The fluorinated benzothiazole analogue 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203, NSC 703786) exhibits selective and potent anticancer activity, and its lysylamide prodrug (Phortress, NSC 710305) recently entered Phase I clinical trials in the United Kingdom. Only cancer cells sensitive to the anti-proliferative effects of 5F 203 deplete this drug candidate from nutrient media. 5F 203 induces cell cycle arrest, cytochrome P450 1A1 (CYP 1A1) mRNA and protein expression, and is metabolized into reactive electrophilic species that can covalently bind to DNA and form adducts in sensitive (i.e., MCF-7) but not in resistant (i.e., MDA-MB-435) breast cancer cells. METHODS: In this present study, we investigated additional anticancer effects of 5F 203 in MCF-7 cells. In addition, we sought to determine if cells deficient in the xeroderma pigmentosum D gene, a gene critical in DNA repair, would show greater sensitivity to the cytotoxic effects of 5F 203 than those complemented with XPD. RESULTS: Alkaline Elution revealed that 5F 203 induced single-strand breaks and DNA-protein cross-links in sensitive MCF-7 cells. In contrast, we detected no double-strand breaks or protein-associated strand breaks typically associated with topoisomerase I (top1) or topoisomerase II (top2) inhibition. In addition, 5F 203 was unable to trap top1- or top2-DNA cleavage complexes in MCF-7 cells. 5F 203 induced cell cycle arrest in MCF-7 cells following DNA damage after brief exposures. Cells deficient in the nucleotide excision repair xeroderma pigmentosum group D (XPD) gene displayed sensitivity to 5F 203 while cells complemented with XPD displayed resistance to 5F 203. CONCLUSION: These data suggest that the anti-cancer activity of 5F 203 depends upon targets other than top1 or top2 and on the ability of this benzothiazole to form single-strand breaks and DNA-protein cross-links in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor/drug effects , DNA Damage , Thiazoles/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor/metabolism , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Humans
7.
Cancer Res ; 65(11): 4844-51, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930305

ABSTRACT

FdUMP[10], a 10mer of 5-fluoro-2'-deoxyuridine 5'-monophosphate (FdUMP), the thymidylate synthase inhibitory metabolite of 5-fluorouracil (FU), is most closely correlated with the DNA topoisomerase I (Top1) inhibitor camptothecin in the National Cancer Institute COMPARE analysis, but not with FU. FdUMP[10] exhibits more potent antiproliferative activity than FdUMP or 5-fluoro-2'-deoxyuridine (FdU) and is markedly more active than FU. Camptothecin-resistant P388/CPT45 cells lacking Top1 are cross-resistant to FdUMP[10] as well as to FdUMP, FdU, and the thymidylate synthase inhibitor raltitrexed (Tomudex). FdUMP[10] induces DNA single-strand breaks and cellular Top1-DNA complexes. Such complexes are also observed in response to FdUMP, FdU, raltitrexed, and FU. The FdUMP[10]-induced Top1-DNA complexes are not inhibited by the caspase inhibitor z-VAD-fmk and form independently of apoptotic DNA fragmentation, indicating that they do not correspond to apoptotic Top1-DNA complexes. In biochemical assay, Top1 is directly trapped at uracil and FdU misincorporation sites. We propose that FdUMP[10] damages DNA by trapping Top1 at uracil and FdU misincorporation sites resulting from thymidylate synthase inhibition and thymine depletion.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Fluorodeoxyuridylate/analogs & derivatives , Fluorodeoxyuridylate/pharmacology , Animals , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Death/drug effects , DNA Damage , DNA, Neoplasm/drug effects , DNA, Single-Stranded/metabolism , Drug Screening Assays, Antitumor , Fluorouracil/metabolism , Fluorouracil/pharmacology , Leukemia P388/drug therapy , Leukemia P388/enzymology , Mice , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidine/deficiency , Thymidine/metabolism , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Topoisomerase I Inhibitors
8.
Cancer Res ; 65(12): 5337-43, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958581

ABSTRACT

Aminoflavone (5-amino-2,3-fluorophenyl)-6,8-difluoro-7-methyl-4H-1-benzopyran-4-one) (NSC 686288) is a candidate for possible advancement to phase I clinical trial. Aminoflavone has a unique activity profile in the NCI 60 cell lines (COMPARE analysis; http://www.dtp.nci.nih.gov/docs/dtp_search.html), and exhibits potent cellular and animal antitumor activity. To elucidate the mechanism of action of aminoflavone, we studied DNA damage in MCF-7 cells. Aminoflavone induced DNA-protein cross-links (DPC) and DNA single-strand breaks (SSB). Aminoflavone induced high levels of DPC and much lower level of SSB than camptothecin, which induces equal levels of DPC and SSB due to the trapping topoisomerase I-DNA complexes. Accordingly, neither topoisomerase I nor topoisomerase II were detectable in the aminoflavone-induced DPC. Aminoflavone also induced dose- and time-dependent histone H2AX phosphorylation (gamma-H2AX). Gamma-H2AX foci occurred with DPC formation, and like DPC, persisted after aminoflavone removal. Aphidicolin prevented gamma-H2AX formation, suggesting that gamma-H2AX foci correspond to replication-associated DNA double-strand breaks. Accordingly, no gamma-H2AX foci were found in proliferating cell nuclear antigen-negative or in mitotic cells. Bromodeoxyuridine incorporation and fluorescence-activated cell sorting analyses showed DNA synthesis inhibition uniformly throughout the S phase after exposure to aminoflavone. Aminoflavone also induced RPA2 and p53 phosphorylation, and induced p21(Waf1/Cip1) and MDM2, demonstrating S-phase checkpoint activation. These studies suggest that aminoflavone produces replication-dependent DNA lesions and S-phase checkpoint activation following DPC formation. Gamma-H2AX may be a useful clinical marker for monitoring the efficacy of aminoflavone in tumor therapies.


Subject(s)
Breast Neoplasms/drug therapy , DNA Damage , DNA, Neoplasm/metabolism , Flavonoids/pharmacology , Histones/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cross-Linking Reagents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , DNA Replication/drug effects , DNA, Neoplasm/drug effects , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/biosynthesis , Phosphorylation/drug effects , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-mdm2 , Replication Protein A , S Phase/drug effects , S Phase/genetics , Tumor Suppressor Protein p53/metabolism
9.
Mol Pharmacol ; 67(2): 523-30, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15531731

ABSTRACT

To overcome camptothecin's (CPT) lactone instability, reversibility of the drug-target interaction, and drug resistance, attempts to synthesize compounds that are CPT-like in their specificity and potency yet display a unique profile have been underway. In this pursuit, we have identified one of the idenoisoquinoline derivatives, MJ-III-65 (NSC 706744; 6-[3-(2-hydroxyethyl)amino-1-propyl]-5,6-dihydro-2,3-dimethoxy-8,9-methylenedioxy-5,11-dioxo-11H-indeno[1,2-c]isoquinoline) with both similarities and differences from CPT. MJ-III-65 traps topoisomerase I (Top1) reversibly like CPT but with different DNA sequence preferences. Consistent with Top1 poisoning, protein-linked DNA breaks were detected in cells treated with MJ-III-65 at nanomolar concentrations. These MJ-III-65-induced protein-linked DNA breaks were resistant to reversal after an hour of drug removal, compared with CPT, which completely reversed. Studies in human cells in culture found MJ-III-65 to be cytotoxic. Furthermore, limited cross-resistance was observed in camptothecin-resistant cell lines. MJ-III-65 also exhibits antitumor activity in mouse tumor xenografts.


Subject(s)
Cell Proliferation/drug effects , Growth Inhibitors/pharmacology , Indenes/pharmacology , Isoquinolines/pharmacology , Topoisomerase I Inhibitors , Animals , Cell Line , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Growth Inhibitors/poisoning , Humans , Indenes/poisoning , Insecta , Isoquinolines/poisoning , Mice , Mice, Nude , Xenograft Model Antitumor Assays/methods
10.
J Org Chem ; 69(22): 7495-501, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15497974

ABSTRACT

The indenoisoquinolines are a novel class of cytotoxic non-camptothecin topoisomerase I inhibitors. A potential DNA-threading agent was designed by attaching different amine side chains on the lactam nitrogen as well as on the C11 position of the indenoisoquinoline ring system. It was hypothesized that substituents on the lactam nitrogen could protrude out toward the DNA major groove while those on the C11 project out toward the DNA minor groove in the ternary "cleavage complex." Compound 4 was synthesized in order to test this DNA-threading scenario. It was found unexpectedly that an alkenyl substituent on the C11 position was autoxidatively cleaved under basic conditions to afford a ketone. A possible mechanism for this unusual oxidative cleavage was proposed on the basis of the studies of a 9-fluoredene model compound. The proposed mechanism was further supported by computational studies. Although the designed compound 4 showed potent cytotoxicities in various cancer cell lines, it was less potent than its nonthreading counterparts and was not a topoisomerase I inhibitor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fluorenes/chemical synthesis , Isoquinolines/chemical synthesis , Topoisomerase I Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin , Cell Line, Tumor , DNA/drug effects , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorenes/chemistry , Fluorenes/pharmacology , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Models, Molecular , Oxidation-Reduction , Structure-Activity Relationship
11.
J Med Chem ; 47(23): 5651-61, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15509164

ABSTRACT

The indenoisoquinolines are a class of cytotoxic topoisomerase I inhibitors that offer certain advantages over the camptothecins, including the greater stabilities of the compounds themselves, as well as the greater stabilities of their drug-enzyme-DNA cleavage complexes. To investigate the possible biological roles of the di(methoxy) and methylenedioxy substituents present on the aromatic rings of the previously synthesized indenoisoquinoline topoisomerase I inhibitors, a series of compounds lacking these substituents was synthesized and tested for both cytotoxicity in cancer cell cultures and for enzyme inhibitory activity. The results indicate that the aromatic substituents make a small, but consistently observable contribution to the biological activity. Molecular models derived for the binding of the unsubstituted indenoisoquinolines in ternary complex with DNA and topoisomerase I indicate that the substituents on the lactam nitrogen project out of the major groove, and the carbonyl group is directed out of the minor groove, where it is involved in a hydrogen bonding interaction with the side chain guanidine group of Arg364. The DNA cleavage patterns observed in the presence of topoisomerase I and various indenoisoquinolines were similar, although significant differences were detected. There were also variations in the DNA cleavage pattern seen with camptothecin vs the indenoisoquinolines, which indicates that these two classes of topoisomerase I inhibitors are likely to target the cancer cell genome differently, resulting in different spectra of anticancer activity. The most cytotoxic of the presently synthesized indenoisoquinolines has a 4-amino-n-butyl group on the lactam nitrogen.


Subject(s)
Antineoplastic Agents/chemical synthesis , Indenes/chemical synthesis , Isoquinolines/chemical synthesis , Topoisomerase I Inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA/chemistry , DNA Topoisomerases, Type I/chemistry , Drug Screening Assays, Antitumor , Electrophoresis, Polyacrylamide Gel , Humans , Indenes/chemistry , Indenes/pharmacology , Isoquinolines/chemistry , Isoquinolines/pharmacology , Models, Molecular
12.
Bioorg Med Chem ; 12(19): 5147-60, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15351398

ABSTRACT

The cytotoxic indenoisoquinolines are a novel class of noncamptothecin topoisomerase I inhibitors having certain features that compare favorably with the camptothecins. A new strategy was adopted to attach aminoalkenyl substituents at C-11 of the indenoisoquinoline ring system, which, according to molecular modeling, would orient the side chains toward the DNA minor groove. All of the newly synthesized compounds were more cytotoxic than the parent indenoisoquinoline NSC 314622. Despite an imperfect correlation between cytotoxicities and topoisomerase I inhibition results, the hypothetical structural model of the cleavage complex presented here provides a conceptual framework to explain the structure-activity relationships.


Subject(s)
Isoquinolines/chemical synthesis , Topoisomerase I Inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Isoquinolines/pharmacology , Models, Molecular , Structure-Activity Relationship
13.
Bioorg Med Chem ; 12(19): 5225-35, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15351405

ABSTRACT

Human topoisomerase I (Top1) plays a pivotal role in cell replication and transcription, and therefore is an important anti-cancer target. Homocamptothecin is a lead compound for inhibiting Top1, and is composed of five conjugated planar rings (A-E). The homocamptothecin E-ring beta-hydroxylactone opens slowly to a carboxylate at pH>7.0. We analyzed, which form of homocamptothecin was biochemically relevant in the following ways: (1) the homocamptothecin carboxylate was tested for activity in vitro and found to be inactive; (2) homocamptothecin was incubated with Top1 and dsDNA, and we found that the homocamptothecin beta-hydroxylactone form was stabilized; (3) the homocamptothecin E-ring beta-hydroxylactone was modified to prevent opening, and the derivatives were either inactive or had low activity. These results indicated that the homocamptothecin beta-hydroxylactone was the active form, and that an E-ring carbonyl oxygen and adjacent unsubstituted/unprotonated ring atom were required for full activity. Homocamptothecin and derivatives were docked into a Top1/DNA active site model, in which the +1 deoxyguanosine was rotated out of the helix, in order to compare the interaction energies between the ligands and the Top1/DNA active site with the in vitro activities of the ligands. It was found that the ligand interaction energies and in vitro activities were correlated, while the orientations of the ligands in the Top1/DNA active site explained the importance of the E-ring beta-hydroxylactone independently of E-ring opening. An essential component of this Top1/DNA active site model is the rotated +1 deoxyguanosine, and in vitro experiments and molecular modeling studies supported rotation of the +1 deoxyguanosine out of the helix. These results allow for the rational design of more potent Top1 inhibitors through engineered interactions with as yet unutilized Top1 active-site residues including: Glu356, Asn430, and Lys751.


Subject(s)
Camptothecin/analogs & derivatives , DNA Topoisomerases, Type I/chemistry , Deoxyguanosine/chemistry , Models, Molecular , Binding Sites , DNA , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Nucleic Acid Conformation , Protein Binding , Static Electricity , Thermodynamics , Topoisomerase I Inhibitors
14.
J Biol Chem ; 279(48): 50499-504, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15448130

ABSTRACT

Topoisomerase I (Top1), an abundant nuclear enzyme expressed throughout the cell cycle, relaxes DNA supercoiling by forming transient covalent DNA cleavage complexes. We show here that staurosporine, a ubiquitous inducer of apoptosis in mammalian cells, stabilizes cellular Top1 cleavage complexes. These complexes are formed indirectly as staurosporine cannot induce Top1 cleavage complexes in normal DNA with recombinant Top1 or nuclear extract from normal cells. In treated cells, staurosporine produces oxidative DNA lesions and generates reactive oxygen species (ROS). Quenching of these ROS by the antioxidant N-acetyl-l-cysteine or inhibition of the mitochondrial dependent production of ROS by the caspase inhibitor benzyloxycarbonyl-VAD prevents staurosporine-induced Top1 cleavage complexes. Down-regulation of Top1 by small interfering RNA decreases staurosporine-induced apoptotic DNA fragmentation. We propose that Top1 cleavage complexes resulting from oxidative DNA lesions generated by ROS in staurosporine-treated cells contribute to the full apoptotic response.


Subject(s)
Apoptosis/physiology , DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Enzyme Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Staurosporine/pharmacology , Apoptosis/drug effects , DNA/drug effects , DNA Fragmentation/drug effects , Humans , Tumor Cells, Cultured
15.
J Biol Chem ; 279(50): 51851-61, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15448168

ABSTRACT

DNA topoisomerase I (topo I) is involved in the regulation of DNA supercoiling, gene transcription, recombination, and DNA repair. The anticancer agent camptothecin specifically targets topo I. The mechanisms responsible for the regulation of topo I in cells, however, are not known. This study demonstrates that c-Abl-dependent phosphorylation up-regulates topo I activity. The c-Abl SH3 domain bound directly to the N-terminal region of topo I. The results demonstrate that c-Abl phosphorylated topo I at Tyr268 in core subdomain II. c-Abl-mediated phosphorylation of topo I Tyr268 in vitro and in cells conferred activation of the topo I isomerase function. Moreover, activation of c-Abl by treatment of cells with ionizing radiation was associated with c-Abl-dependent phosphorylation of topo I and induction of topo I activity. The functional significance of the c-Abl/topo I interaction is supported by the findings that (i) mutant topo I(Y268F) exhibited loss of c-Abl-induced topo I activity, and (ii) c-Abl-/- cells were deficient in the accumulation of protein-linked DNA breaks. In addition, loss of topo I phosphorylation in c-Abl-deficient cells conferred resistance to camptothecin-induced apoptosis. These findings collectively support a model in which c-Abl-mediated phosphorylation of topo I is functionally important to topo I activity and sensitivity to topo I poisons.


Subject(s)
Camptothecin/pharmacology , DNA Topoisomerases, Type I/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Nucleus/enzymology , Cells, Cultured , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/genetics , Drug Resistance , Enzyme Activation/radiation effects , Humans , Mice , Models, Biological , Phosphorylation , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Topoisomerase I Inhibitors , src Homology Domains
16.
Biochem Pharmacol ; 68(5): 857-66, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15294448

ABSTRACT

Cisplatin, cis-[PtCl2(NH3)2], is commonly utilized in various combination chemotherapy protocols for the treatment of both ovarian and breast cancer while the corresponding trans isomer is therapeutically inactive. This work describes efforts to elucidate the cellular mechanism of action of a novel trans-platinum compound, trans-(dichloroamminethiazole)platinum(II) (ATZ), which demonstrates antiproliferative and cytotoxic effects against both MCF-7 human breast and A2780 human ovarian carcinoma cells in culture. A2780 cells were approximately twofold more sensitive to ATZ than MCF-7 cells in both cell growth and clonogenic survival assays. Dye exclusion studies revealed a 50-70% loss in cell viability within the first 12 h of drug treatment in both cell lines. This initial wave of cell death was succeeded by a prolonged interval of growth arrest during which a small fraction of apoptotic cells was detected. Binding of ATZ to DNA, as estimated by atomic absorption spectroscopy, was similar for the two cell lines and was almost completely reversed 24 h after drug removal. ATZ also induced DNA strand breakage as well as DNA-protein crosslinking during the initial 12 h period when the bulk of cell death was evident. However, neither the extent of DNA strand breakage nor that of DNA protein crosslinking was sufficient to explain the different drug sensitivity in the two cell lines. At 24 and 48 h after exposure of MCF-7 cells to high concentrations of ATZ, the formation of DNA-topoisomerase I complexes is detected, coincident with a high degree of apoptosis. These studies suggest that ATZ has the capacity to interfere with topoisomerase I in the tumor cell, a function not evident in cis-platinum-based drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , DNA Damage/drug effects , DNA, Neoplasm/drug effects , Organoplatinum Compounds/pharmacology , Animals , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Cross-Linking Reagents/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA, Neoplasm/metabolism , Female , Humans , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
17.
Mol Cancer Ther ; 3(7): 849-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15252146

ABSTRACT

We identified five structurally related dimethane sulfonates with putative selective cytotoxicity in renal cancer cell lines. These compounds have a hydrophobic moiety linked to a predicted alkylating group. A COMPARE analysis with the National Cancer Institute Anticancer Drug Screen standard agent database found significant correlations between the IC50 of the test compounds and the IC50 of alkylating agents (e.g., r = 0.68, P < 0.00001 for chlorambucil). In this report, we examined whether these compounds had activities similar to those of conventional alkylating agents. In cytotoxicity studies, chlorambucil-resistant Walker rat carcinoma cells were 4- to 11-fold cross-resistant to the test compounds compared with 14-fold resistant to chlorambucil. To determine effects on cell cycle progression, renal cell carcinoma (RCC) line 109 was labeled with bromodeoxyuridine prior to drug treatment. Complete cell cycle arrest occurred in cells treated with an IC90 dose of NSC 268965. p53 protein levels increased as much as 5.7-fold in RCC line 109 and as much as 20.4-fold in breast cancer line MCF-7 following an 18-hour drug exposure. Finally, DNA-protein cross-links were found following a 6-hour pretreatment with all compounds. Thus, the dimethane sulfonate analogues have properties expected of some alkylating agents but, unlike conventional alkylating agents, appear to possess activity against RCC.


Subject(s)
Alkylating Agents/chemistry , Alkylating Agents/toxicity , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Mesylates/chemistry , Mesylates/toxicity , Alkylating Agents/therapeutic use , Animals , Bromodeoxyuridine/analysis , Busulfan/analogs & derivatives , Carmustine/analogs & derivatives , Cell Cycle/drug effects , DNA Damage , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Humans , Inhibitory Concentration 50 , Mesylates/therapeutic use , Rats , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , Yeasts/drug effects
18.
J Biol Chem ; 279(32): 33968-75, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15178684

ABSTRACT

Topoisomerase I is an essential enzyme that relaxes DNA supercoiling by forming covalent DNA cleavage complexes, which are normally transient. Topoisomerase I-DNA complexes can be trapped by anticancer drugs (camptothecins) as well as by endogenous and exogenous DNA lesions. We show here that arsenic trioxide (a potent inducer of apoptosis that induces the intracellular accumulation of reactive oxygen species and targets mitochondria) induces cellular topoisomerase I cleavage complexes. Bcl-2 overexpression and quenching of reactive oxygen species, which prevent arsenic trioxide-induced apoptosis, also prevent the formation of topoisomerase I-DNA complexes, whereas enhancement of reactive oxygen species accumulation promotes these complexes. The caspase inhibitor, benzyloxycarbonyl-VAD partially prevents arsenic trioxide-induced topoisomerase I-DNA complexes and apoptosis, suggesting that activated caspases further maintain intracellular levels of reactive oxygen species that induce the formation of topoisomerase I-DNA complexes. Down-regulation of topoisomerase I expression decreases arsenic trioxide-induced apoptotic DNA fragmentation. Thus, we propose that arsenic trioxide induces topoisomerase I-DNA complexes that participate in chromatin fragmentation and programmed cell death during apoptosis.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Oxides/pharmacology , Arsenic Trioxide , Blotting, Western , Buthionine Sulfoximine/pharmacology , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , DNA Fragmentation , DNA Topoisomerases, Type I/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Gene Expression , Gene Silencing , Humans , Leukemia , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Reactive Oxygen Species/metabolism , Recombinant Proteins , Tumor Cells, Cultured
19.
Bioorg Med Chem Lett ; 14(14): 3659-63, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15203138

ABSTRACT

Indenoisoquinolines and dihydroindenoisoquinolines have been synthesized possessing a nitro-substituted isoquinoline ring in an effort to explore the effects of electron-withdrawing substituents on biological activity. The in vitro anticancer activities of these molecules have been tested in the National Cancer Institute's screen of 55 cell lines. The compounds have also been tested for topoisomerase I (top1) inhibition. The results indicate that these substances are a potent class of top1 inhibitors with sub-micromolar cytotoxicity mean graph midpoints (MGM) and top1 inhibition equal to camptothecin.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Isoquinolines/chemical synthesis , Topoisomerase I Inhibitors , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Humans , Indenes/chemical synthesis , Indenes/pharmacology , Inhibitory Concentration 50 , Isoquinolines/pharmacology , Structure-Activity Relationship
20.
J Pharmacol Exp Ther ; 310(2): 836-42, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15075385

ABSTRACT

One activity potentially limiting the efficacy of camptothecin anticancer agents is their cellular efflux by the ATP-binding cassette half-transporter, ABCG2. Homocamptothecins are novel anticancer drugs that inhibit topoisomerase 1 with a greater potency than camptothecins. Homocamptothecins differ from camptothecins by their E-ring, which is seven-membered instead of the six-membered ring of camptothecins. We report herein that, like camptothecins, homocamptothecin and its difluoro derivative BN80915 are substrates for ABCG2. However, the resistance of three selected cell lines overexpressing wild-type or mutant ABCG2 to homocamptothecin or BN80915 was less than resistance to SN-38 (7-ethyl-10-hydroxycamptothecin), indicating that both the seven-membered E-ring present in homocamptothecin and the A- and B-ring modifications present in SN-38 are involved in substrate recognition by ABCG2. HEK-293 cells transfected with vectors encoding wild-type or mutant ABCG2 were found to be less resistant to both homocamptothecins than to SN-38. However, transfectants overexpressing mutant ABCG2 had relative resistance values for homocamptothecin and BN80915 4- to 14-fold higher than cells expressing wild-type ABCG2, suggesting that the gain of function resulting from mutation at amino acid 482, although not affecting SN-38, extends to the homocamptothecins. Resistance was reversed by the ABCG2 inhibitor fumitremorgin C. BN80915 was 17-fold more potent than SN-38 in wild-type ABCG2-transfected cells, suggesting that BN80915 has the potential to overcome ABCG2-related resistance to SN-38, the active metabolite of CPT-11 (irinotecan).


Subject(s)
ATP-Binding Cassette Transporters/physiology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Resistance, Neoplasm , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Camptothecin/chemistry , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Irinotecan , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Topoisomerase I Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...