Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters










Publication year range
1.
Lung Cancer ; 40(3): 325-32, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12781432

ABSTRACT

Among numerous clinical regimens of combination chemotherapy, synergy has been observed to be particularly marked with combinations containing cisplatin (CDDP). However, the clinical use of CDDP has sometimes been limited by acquired resistance. The new-generation platinum drug, ZD0473, was synthesized with the aim of hindering the reaction of the drug with thiols, by the introduction of a 2-methylpyridine ligand. This enables the drug to exert antitumor activity against cisplatin-resistant cancer cells with elevated glutathione and/or metallothionein levels. The drug was also shown experimentally to overcome cisplatin resistance due to impaired drug accumulation, and enhanced DNA repair/tolerance to platinum-DNA adducts. We investigated the effects of combinations of ZD0473 with other anticancer drugs on the growth of a human small-cell lung cancer cell line (SBC-3). Six novel anticancer drugs were tested: docetaxel (TXT), paclitaxel (TXL), vinorelbine (VNB), irinotecan (CPT-11), gemcitabine (GEM) and pemetrexed (MTA). The growth inhibitory effect of the drugs was measured by MTT assay and the effects of the combination regimens were evaluated by the combination index analysis method developed by Chou and Talalay. Synergy was demonstrated for the combination regimens of ZD0473-GEM and ZD0473-TXL, while an additive effect was observed with combinations containing TXT, VNB, CPT-11 or MTA. In the case of the ZD0473-GEM combination, synergy was observed over a wide range of inhibition levels at dose ratios of 50:1, 100:1 and 250:1. The level of synergy was equivalent to that observed for combinations of CDDP-etoposide, CDDP-GEM and nedaplatin-CPT-11. The results suggest that the combination of ZD0473 with GEM merits further investigation in small cell lung cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Small Cell/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Lung Neoplasms/pathology , Organoplatinum Compounds/pharmacology , Drug Screening Assays, Antitumor , Humans , Tumor Cells, Cultured , Gemcitabine
2.
Lung Cancer ; 34(3): 451-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714543

ABSTRACT

Vinorelbine (Navelbine, KW-2307), a semisynthetic vinca alkaloid, is a potent inhibitor of mitotic microtubule polymerization. The aims of this study were to demonstrate radiosensitization produced by vinorelbine in human non-small cell lung cancer (NSCLC) PC-9 cells and to elucidate the cellular mechanism of radiosensitization. A clonogenic assay demonstrated that PC-9 cells were sensitized to radiation by vinorelbine with a maximal sensitizer enhancement ratio at a 10% cell survival level of 1.35 after 24-h exposure to vinorelbine at 20 nM. After 24-h exposure to vinorelbine at 20 nM, the approximately 67% of the cells that had accumulated in the G2/M-phase were cultured in the absence of vinorelbine and then irradiated at a dose of 8 Gy. Flow cytometric analyses showed prolonged G2/M accumulation concomitant with continuous polyploidization, and induction of apoptosis was observed in the cells subjected to the combination of vinorelbine-pretreatment and radiation. Polyploidization and induction of apoptosis were confirmed by morphological examination and a DNA fragmentation assay, respectively. We concluded that vinorelbine at a minimally toxic concentration moderately sensitizes human NSCLC cells to radiation by causing accumulation of cells in the G2/M-phase of the cell cycle. Prolonged G2/M accumulation concomitant with continuous polyploidization and increased susceptibility to induction of apoptosis may be associated with the cellular mechanism of radiosensitization produced by vinorelbine.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Vinblastine/analogs & derivatives , Vinblastine/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Flow Cytometry , Humans , Lung Neoplasms/pathology , Tumor Cells, Cultured , Vinorelbine
3.
Cancer ; 91(8): 1494-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11301397

ABSTRACT

BACKGROUND: Oncoprotein 18 (op18) was first isolated as a molecule overexpressed in several malignant cells, suggesting a function of op18 in malignant processes, such as differentiation in hematologic malignancies, op18 also was found to enhance microtubule deassembly in the cells. Antimitotic agents that bind to tubulin have been used for chemotherapy to treat solid tumors, such as lung carcinoma. Vinca alkaloids, such as vindesine and vincristine, have commonly been used for chemotherapy of nonsmall cell lung carcinoma. The authors examined the role of op18 in the sensitivity of human lung carcinoma cells to antimitotic agents. METHODS: Expression of op18 mRNA was detected in all 17 lung carcinoma cell lines tested by Northern blotting. Oncoprotein 18 cDNA was transfected to SBC-3 human lung carcinoma cells, and the stable transfectants, SBC-3/op1-3, were isolated. The sensitivity of these transfectants against antimitotic agents were examined by the MTT assay in vitro. Cell cycle distribution of the transfectants on DNA histogram was analyzed by flow cytometry. RESULTS: Oncoprotein 18-transfected cells showed higher sensitivity to vindesine and vincristine, but not to taxanes. Vindesine-exposure increased the G2/M population of the cell cycle in the Mock transfectants, but not in SBC-3/op1, suggesting that the cell cycle dynamics were altered by op18 expression in SBC-3/op1. CONCLUSION: Oncoprotein 18 expression is associated with lung carcinoma cell sensitivity to vindesine and may be able to serve as a surrogate marker for the chemosensitivity to Vinca alkaloids in human lung carcinomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/pathology , Lung Neoplasms/pathology , Microtubule Proteins , Phosphoproteins/biosynthesis , Vindesine/pharmacology , Blotting, Northern , DNA, Complementary , Drug Screening Assays, Antitumor , Humans , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Phosphoproteins/pharmacology , RNA, Messenger/analysis , Stathmin , Transfection , Tumor Cells, Cultured , Vincristine/pharmacology
4.
Clin Cancer Res ; 7(1): 202-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11205910

ABSTRACT

Among the numerous clinical regimens used in combination chemotherapy, synergy is particularly marked in combinations containing cisplatin (CDDP). However, the clinical use of CDDP is sometimes limited due to its nephrotoxicity. Nedaplatin (NDP) is a second-generation platinum complex with reduced nephrotoxicity that may substitute for CDDP or even surpass it for use in combination with other drugs. We investigated the effects of combinations of NDP and other anticancer drugs on the growth of human small cell lung cancer cells (SBC-3) and non-small cell lung cancer cells (PC-14) using a three-dimensional analysis model. Among the combinations tested, the combination of NDP and irinotecan (CPT-11) showed the most marked synergistic interaction, and the synergism has also been observed against PC-14 cells. With regard to treatment schedule, a remarkable synergistic interaction was produced by concurrent exposure to NDP and CPT-11. On the other hand, sequential exposure to the two drugs led only to additivity. To analyze the interaction between the drugs, the effect of NDP on the 7-ethyl-1-hydroxy-CPT (the active form of CPT-11)-induced inhibitory effect on DNA topoisomerase I was examined. The topoisomerase I-inhibitory effect of 7-ethyl-1-hydroxy-CPT was enhanced 10-fold in the presence of NDP at microgram/milliliter concentrations. These biochemical interactions might be responsible for the synergistic interaction between NDP and CPT-11. These results suggest that the combination of NDP with CPT-11 may be clinically useful for the chemotherapy of lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Enzyme Inhibitors/pharmacology , Organoplatinum Compounds/pharmacology , Thiazolidinediones , Topoisomerase I Inhibitors , Tumor Cells, Cultured/drug effects , Antineoplastic Agents/metabolism , Antineoplastic Combined Chemotherapy Protocols , Camptothecin/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Cell Division/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Synergism , Enzyme Inhibitors/metabolism , Humans , Irinotecan , Lung Neoplasms , Organoplatinum Compounds/metabolism , Thiazoles/metabolism
5.
Cancer Chemother Pharmacol ; 43(5): 353-63, 1999.
Article in English | MEDLINE | ID: mdl-10100589

ABSTRACT

PURPOSE: KRN5500 is a new derivative of spicamycin produced by Streptomyces alanosinicus and is known to have a wide range of antitumor activities against human cancer cell lines. Because of its unique structure, this compound seems to have a different mode of action from other antitumor drugs and nonoverlapping toxicities. Therefore, KRN5500 is expected to be a suitable candidate for combination chemotherapy. METHODS: We investigated the effects of combinations of KRN5500 and other anticancer drugs on the growth of a human non-small-cell lung cancer cell line, PC14, using a revised three-dimensional model. RESULTS: Synergism was observed when KRN5500 and cisplatin were combined at concentrations in the ranges 0.005 to 0.25 microg/ml and 0.025 to 0.25 microg/ml, respectively. In combination with carboplatin, an analog of cisplatin, and etoposide, a marked synergistic interaction was also found. CONCLUSION: These results suggest the usefulness of combinations of KRN5500 with cisplatin, carboplatin or etoposide for chemotherapy for non-small-cell lung cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Antibiotics, Antineoplastic/administration & dosage , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cell Division/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Etoposide/administration & dosage , Etoposide/pharmacology , Humans , Models, Biological , Models, Statistical , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacology , Tumor Cells, Cultured
6.
Cancer Chemother Pharmacol ; 43(5): 394-401, 1999.
Article in English | MEDLINE | ID: mdl-10100595

ABSTRACT

PURPOSE: TAS-103 [6-((2-(dimethylamino) ethyl)amino)-3-hydroxy-7H-iindeno(2,1-c)quinolin-7-one dihydrochloride] is a newly synthesized dual inhibitor of topoisomerase I and II. Since anticancer drugs are used in combination with other drugs for effective chemotherapy, we investigated the cytotoxic effect of TAS-103 in combination with other conventional anticancer agents, such as cisplatin, vindesine, doxorubicin, 5-fluorouracil, and the antitopoisomerase inhibitors SN-38 and etoposide in vitro. METHODS: Inhibition of the growth of the human small-cell lung cancer cell line SBC-3 was evaluated using the tetrazolium dye (MTT) assay. Drug interactions were evaluated by isobologram analysis and the determination of combination indices supplemented by a three-dimensional model. RESULTS: Simultaneous use of TAS-103 and cisplatin had a supraadditive effect, but combinations of TAS-103 with other drugs had an additive or marginally subadditive effect. Three-dimensional model analysis added more information about the synergistic concentration ranges of two drugs (cisplatin 200-400 nM and TAS-103 7 10 nM). Sequential use of TAS-103 and cisplatin had only an additive effect. CONCLUSION: These results suggest that the concomitant use of TAS-103 and cisplatin has a greater cytotoxic effect on cancer cells than single drug use, and may provide a beneficial effect in the treatment of small-cell lung cancer.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Small Cell/pathology , Enzyme Inhibitors/pharmacology , Indenes/pharmacology , Lung Neoplasms/pathology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Carcinoma, Small Cell/enzymology , Cell Survival/drug effects , Drug Synergism , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Models, Biological , Models, Statistical , Tumor Cells, Cultured
7.
Invest New Drugs ; 17(4): 335-41, 1999.
Article in English | MEDLINE | ID: mdl-10759401

ABSTRACT

7-hydroxystaurosporine (UCN-01) is a new anticancer agent which exerts an inhibitory effect on cell cycle check points and is currently under phase I clinical trials in US and Japan. Preliminary clinical data indicated that UCN-01 remained in plasma at high concentrations for long periods of time. This unavoidable high plasma drug exposure is likely to lead to hematological toxicities in patients. In the present study, cultured human peripheral blood lymphocytes (PBLs) were used to evaluate the possible hematological toxicities of UCN-01 treatment. UCN-01 induces apoptosis, and the induction of apoptosis-related surface markers were also examined to investigate the involvement of these molecules in UCN-01-induced apoptosis in PBLs. In vitro viability of PBLs was decreased by high dose of UCN-01 (25 microM, 3-day exposure). This effect of UCN-01 was significantly suppressed by the presence of human serum, suggesting that some specific inhibitory factor(s) in human serum may antagonize the lympholytic effect of UCN-01. The percentage of annexin V-positive PI-negative cells increased with exposure to UCN-01 in a time- and dose-dependent manner; by up to 30.3% after exposure to 25 microM UCN-01 for 3 days. At the same time, the expression of both interleukin-2 receptor (IL-2R, CD25) and Fas (CD95), analyzed by flow cytometry, was induced. Con A-stimulated PBLs were more sensitive to UCN-01-induced apoptosis than non-stimulated lymphocytes and UCN-01 increased the sFas-L released into culture medium from con A-stimulated PBLs. Therefore, lymphocyte depletion mediated by activation-induced apoptosis is likely to occur in patients treated with UCN-01 at high doses.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media , Humans , Receptors, Interleukin-2/drug effects , Receptors, Interleukin-2/metabolism , Staurosporine/analogs & derivatives , Time Factors , fas Receptor/biosynthesis
8.
Int J Cancer ; 71(3): 311-9, 1997 May 02.
Article in English | MEDLINE | ID: mdl-9139860

ABSTRACT

Although the combination of cisplatin and etoposide has been used as standard therapy for small-cell lung cancer, it is difficult to demonstrate combination effects between cisplatin and etoposide in vitro. We therefore adopted a 3-dimensional (3-D) model to analyze the combination effects of anticancer drugs, and compared the results of analysis by the new 3-D model with those obtained from traditional 2-D models for the cisplatin-etoposide combination. In this study, using a human small-cell lung-cancer cell line (SBC-3), 3-D model analysis clearly identified a relationship depending on the concentrations of both drugs, and demonstrated that peak synergy occurred at the higher concentrations of cisplatin and etoposide. Antagonistic interactions were noted with a nadir at low concentrations of etoposide and cisplatin. In contrast, 2-D models such as combination index and isobologam analysis fail to characterize the complex interactions between cisplatin and etoposide, since their joint effects are concentration-dependent. Combination index (CI) plots show that synergy is evident only for molar ratios of cisplatin: etoposide of 2:1 to 1:5. On isobologram analysis, synergy could be detected when great inhibitory effects on cell growth were present (high endpoint), but not with small inhibitory effects (lower endpoints). Thus, either synergy or antagonism may occur, but depend on the selection of variables, such as the molar ratios or the endpoints chosen for the experiments. This could explain the inconsistency in the in vitro combination effects reported to date. The 3-D model, which compensates for the above deficiencies of 2-D models, can facilitate rigorous analysis of drug interactions over the entire clinical dose range, using microcomputers and sophisticated graphics programs. This direct and pragmatic method offers investigators a practical new tool with which to analyze drug combinations for cancer chemotherapy.


Subject(s)
Antineoplastic Agents/toxicity , Cell Division/drug effects , Cisplatin/toxicity , Etoposide/toxicity , Models, Theoretical , Carcinoma, Small Cell , Cisplatin/antagonists & inhibitors , Drug Synergism , Etoposide/antagonists & inhibitors , Humans , Lung Neoplasms , Probability , Tumor Cells, Cultured
9.
Semin Oncol ; 24(2 Suppl 7): S7-8-S7-16, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9194474

ABSTRACT

Gemcitabine (2',2'-difluorodeoxycytidine) is a deoxycytidine analog with significant antitumor activity against ovarian cancer and non-small cell lung cancer. It is a suitable candidate for combination chemotherapy in non-small cell lung cancer for three reasons: it is active as a single agent, it has no overlapping toxicities with other chemotherapeutic agents, and it has different mechanisms of action compared with other anticancer drugs. We therefore investigated the combination effects between gemcitabine and other anticancer drugs on the growth of the non-small cell lung cancer cell line, PC-14. Combination effects were analyzed by means of a three-dimensional model, which directly elucidates the shape of the dose-response surface over the entire clinical dose range, identifies regions of statistically significant synergy and antagonism, and quantifies these effects. The three-dimensional analysis clearly demonstrates a relationship, the nature of which depends on the concentration of both drugs. A synergistic effect was observed when gemcitabine and cisplatin were combined at concentrations of 0.0005 to 0.001 microg/mL and 0.025 to 0.25 microg/mL, respectively. In combination with vindesine, a remarkable synergistic interaction also was found when combined at concentrations of 0.00005 to 0.0005 microg/mL gemcitabine and 0.001 to 0.01 microg/mL vindesine. These results suggest the usefulness of combination therapy with gemcitabine/cisplatin and gemcitabine/vindesine. However, because these combination effects depend on drug concentrations, this parameter must be carefully considered when using such drug combinations in clinical applications.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cell Division/drug effects , Deoxycytidine/analogs & derivatives , Models, Theoretical , Ribonucleotide Reductases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Drug Synergism , Humans , Lung Neoplasms/drug therapy , Tumor Cells, Cultured , Gemcitabine
10.
Br J Cancer ; 76(5): 571-81, 1997.
Article in English | MEDLINE | ID: mdl-9303354

ABSTRACT

Drug resistance is one of the problems severely limiting chemotherapy in cancer patients. Thus, it is very important to develop new drugs that are effective against drug-resistant tumour cells. The novel anti-tumour agent NK109 has been developed from benzo[c]phenanthridine derivatives by Nippon Kayaku (Tokyo, Japan). We have confirmed that NK109 shows anti-tumour effects against a number of human tumour cell lines by inhibiting DNA topoisomerase II activity through the stabilization of the cleavable complex. Further, its efficacy against several drug-resistant tumour cell lines was also shown. NK109 showed potent anti-tumour activity against doxorubicin-resistant human tumour cell lines that have a typical multidrug resistance phenotype caused by P-glycoprotein. NK109 was not pumped extracellularly by P-glycoprotein and, consequently, NK109 accumulated in resistant cells. Cisplatin-resistant human tumour cell lines, which demonstrated decreased cisplatin accumulation, were sensitive to NK109. NK109 non-cross-resistance was confirmed using xenografts of tumour cells that were resistant to cisplatin in SCID mice. Furthermore, etoposide-resistant cells, with decreased topoisomerase II activity, were markedly sensitive to NK109 when compared with their parent cells, suggesting the possibility that the cytotoxic mechanism of NK109 differs from that of etoposide. In conclusion, NK109 is a very promising new anti-tumour drug for clinical use, because the efficacy of NK109 is not susceptible to several known molecular alterations that are associated with drug resistance. A clinical study of this compound is now in progress in Japan.


Subject(s)
Antineoplastic Agents/toxicity , Phenanthridines/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzophenanthridines , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacokinetics , Cisplatin/toxicity , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Etoposide/toxicity , Female , Humans , Lung Neoplasms/drug therapy , Membrane Glycoproteins/metabolism , Mice , Mice, SCID , Phenanthridines/pharmacokinetics , Phenanthridines/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured
11.
Jpn J Cancer Res ; 87(10): 1086-91, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8957068

ABSTRACT

2,3-(Methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]phenanthr idinium hydrogensulfate dihydrate, called NK109, is a benzo[c]phenanthridine derivative, which inhibits DNA topoisomerase II activity by stabilizing the DNA-enzyme-drug complex, and shows strong growth-inhibitory effects on several human cancer cells. In the present study, NK109 treatment induced DNA fragmentation and a rise in the level of cytoplasmic nucleosomes, which are markers of apoptosis, in human small-cell lung carcinoma SBC-3 cells. These effects were inhibited by zinc ions and enhanced by cycloheximide or actinomycin D. Dose-dependent single- and double-strand DNA breaks were observed, using alkaline and neutral elution assays, in SBC-3 cells treated with more than 0.2 microM NK109 for 4 h. Treatment with NK109 caused more DNA single- and double-strand breaks than treatment with an equimolar amount of VP-16. These results suggest that NK109 induces DNA strand breaks and apoptosis. In addition, it appears that this process does not require protein or RNA synthesis, but involves a specific endonuclease which is inhibited by zinc ions.


Subject(s)
DNA Damage , DNA Fragmentation , DNA, Neoplasm/drug effects , DNA/drug effects , Topoisomerase II Inhibitors , DNA/genetics , DNA, Neoplasm/genetics , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/genetics , HL-60 Cells , Humans , Nucleosomes , Tumor Cells, Cultured
12.
Int J Cancer ; 67(5): 702-8, 1996 Sep 04.
Article in English | MEDLINE | ID: mdl-8782662

ABSTRACT

We examined the effects of the introduction of H-ras oncogene into murine cell line NIH3T3 on growth inhibition by topoisomerase-I (topo-I) inhibitors. The H-ras-transformed cells (pT22-3) showed approximately 12-fold increased sensitivity to a novel topo-I inhibitor, NB-506 [6-N-formylamino-12,13-dihydro-1,11-dihydroxy-13-(beta-D-glucopyranosyl) -5H-indolo(2,3-a)pyrrolo(3,4-c) carbazole-5,7(6H)-dione], compared with the parental NIH3T3 cells. pT22-3 also showed increased sensitivity to other topo-I inhibitors such as camptothecin (approx. 3.0-fold) and CPT-11 (irinotecan, approx. 3.0-fold). Transformation of NIH3T3 by another oncogene (erbB2) did not affect their sensitivity to these topo-I inhibitors. pT22-3 had approximately 32-fold higher topo-I activity than NIH3T3, but the same topo-I content. In a cell-free system, topo-I activity was increased 2-fold by addition of the H-ras protein precipitated from pT22-3 cells. Topo I in the nuclear extract of pT22-3 was eluted easily by low concentrations of NaCl compared with that of NIH3T3, suggesting a qualitative change in pT22-3 topo 1. Increased phosphorylation of topo I was observed in pT22-3. Furthermore, NB-506 decreased the amount of the GTP-bound form of the H-ras product in pT22-3 cells. These results suggest that the high growth-inhibitory effect of a topo-I inhibitor, NB-506, on H-ras-transformed NIH3T3 cells is due to the H-ras-mediated signal-transduction pathway.


Subject(s)
3T3 Cells/drug effects , Enzyme Inhibitors/pharmacology , Genes, ras , Topoisomerase I Inhibitors , 3T3 Cells/metabolism , Animals , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Transformed , DNA Topoisomerases, Type I/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Immunoblotting , Irinotecan , Mice , Phosphorylation , ras Proteins/metabolism
13.
Cancer Res ; 56(4): 789-93, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8631015

ABSTRACT

Topoisomerase I-targeting anticancer agents such as 7-ethyl-10-[4-(1-piperidyl)-1-piperidyl]carbonyloxy-camptothecin (CPT-11) and 6-N-formylamino-12,13-dihydro-1,11-dihydroxy-13-(beta-D- glucopyranosyl)-5H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-5,7(6H)-di one (NB-506) have been developed and show strong antitumor activity against various cancers. We examined the interaction of these drugs and cisplatin (CDDP), and biochemical mechanisms of synergism between them. Interaction of drugs in human small cell lung cancer cells, SBC-3, was analyzed using the isobologram method. Combinations of CDDP with NB-506, CPT-11, and an active metabolite of CPT-11, 7-ethyl-10-hydroxy-CPT (SN-38), showed synergistic effects. Formation of DNA interstrand cross-links (ICLs) on the cells was analyzed using an alkaline elution assay and increased ICLs were observed by simultaneous exposure to CDDP (1.5 microM) and NB-506 (10 nM) compared with that in response to CDDP alone. DNA repair after ICL formation induced by 3-h exposure to CDDP (1.5 microM) was reduced by NB-506 (10 nM) exposure. On the other hand, a higher concentration of CDDP (150 microM) enhanced the topoisomerase I inhibitory activity of NB-506 and SN-38 determined by relaxation of supercoiled Escherichia coli DNA. These biological interactions might result in synergistic interactions between CDDP and NB-506 or SN-38. Topoisomerase I inhibitors and CDDP may be a key regimen for cancer chemotherapy and merit further examination.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Antineoplastic Agents/toxicity , Camptothecin/analogs & derivatives , Carbazoles/toxicity , Cisplatin/toxicity , Enzyme Inhibitors/toxicity , Glucosides/toxicity , Topoisomerase I Inhibitors , Camptothecin/toxicity , Carcinoma, Small Cell , Cell Division/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Survival/drug effects , DNA Repair/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Irinotecan , Lung Neoplasms , Tumor Cells, Cultured
14.
Cancer Res ; 55(13): 2806-13, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7796407

ABSTRACT

The novel anticancer glucosyl derivative of indolo-carbazole (NB-506), an inhibitor of DNA topoisomerase I, exhibited strong in vitro cytotoxicity against various human cancer cell lines. In order to elucidate its cytotoxic mechanisms, we established nine NB-506-resistant sublines with different resistance ratios from human small cell lung cancer cells (SBC-3/P) by stepwise and brief exposure (24 h) to NB-506. Among them, SBC-3/NB#9 was 454 times more resistant to NB-506 than the parent cell line. The SBC-3/NB#9 cells showed cross-resistance only to topoisomerase I inhibitors, such as 11,7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecia and 7-ethyl-10-hydroxy-camptothecin, and not to other anticancer drugs, such as vincristine, vinblastine, Adriamycin, etoposide, and teniposide. These results indicate that the difference on the effect of topoisomerase I was considered to be related to a resistance mechanism. The topoisomerase I activities of nuclear extracts eluted from SBC-3/NB#9 cells was only one-tenth of the parent cell activity. A Western blotting study indicated that this lower activity was due to a lower amount of DNA topoisomerase I. Furthermore, we found correlations between topoisomerase I activity and sensitivity to NB-506 in sublines with different degrees of resistance. Accumulation of 3H-labeled NB-506 by SBC-3/NB#9 cells was only one-fifth of that by the parent cells, whereas intracellular accumulation of 3H-labeled camptothecin by both cell lines did not differ. The reduction of accumulation was specific to NB-506, and this result may explain why the resistance ratio for NB-506 was higher than those for 11,7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxycamptothecin and 7-ethyl-10-hydroxy-camptothecin.


Subject(s)
Antineoplastic Agents , Carbazoles/pharmacology , Glucosides/pharmacology , Topoisomerase I Inhibitors , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Carcinoma, Small Cell , Cell Nucleus/enzymology , Drug Resistance , Humans , In Vitro Techniques , Irinotecan , Tumor Cells, Cultured
15.
Anticancer Res ; 15(2): 393-8, 1995.
Article in English | MEDLINE | ID: mdl-7763011

ABSTRACT

We determined the in vitro cytotoxicities of promising new platinum complexes, an inhibitor of topoisomerase I, and novel anthracycline derivatives, and examined using clonogenic assay whether these compounds are cross-resistant in CDDP-resistant sublines derived from human non-small cell lung cancer (NSCLC) cell lines. Cytotoxicities were evaluated by means of the relative antitumor activity (RAA = peak plasma concentration/IC50 values), which we reported previously as a model for predicting antitumor activity. Of the CDDP analogues tested, including carboplatin (CBDCA), [(glycolate-0,0') diammineplatinum (II) (254-S)], ormaplatin [tetrachloro-(d,1-trans)- 1,2-diaminocyclohexaneplatinum (IV) (OP)] and oxaliplatin [oxalato (trans-1-1,2-diaminocyclohexane) platinum (II) (1-OHP)], no agent showed superior antitumor activity compared to CDDP. ME2303 [7-O-(2,6-dideoxy-2-fluoro-alpha-L-talopyranosyl) adriamycinone-14-O -pimelate], a new anthracycline derivative, showed higher antitumor activity than adriamycin (ADM). The CDDPresistant sublines, PC-9/CDDP and PC-14/CDDP, were 18.3- and 7.7-fold more resistant to CDDP compared to the respective parent cell lines. The CDDP analogues were all cross-resistant to CDDP and the order of relative resistance values was CBDCA > 254-S > 1-OHP > OP for PC9/CDDP and 254-S > CBDCA > 1-OHP > OP for PC-14/CDDP. Although OP showed cross-resistance to CDDP, OP was the most active against the CDDP-resistant sublines with relative resistance values of 3.8 and 1.6 for PC-9/CDDP and PC-14/CDDP, respectively. ME2303 and CPT-11, [7-ethyl-10-(4-[1-piperidino]-1-piperidino) carbonyloxycamptothecin], an inhibitor of topoisomerase I, was active against CDDP-resistant human NSCLC cell lines. These results suggest that OP, ME2303 and CPT-11 could be active in patients with, NSCLC clinically resistant to CDDP.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Carboplatin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Doxorubicin/analogs & derivatives , Lung Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Doxorubicin/pharmacology , Drug Design , Drug Resistance, Multiple , Etoposide/pharmacology , Irinotecan , Lung Neoplasms/pathology , Oxaliplatin , Structure-Activity Relationship , Tumor Stem Cell Assay
16.
Jpn J Cancer Res ; 86(1): 124-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7737904

ABSTRACT

KW-2189, a novel antitumor antibiotic belonging to the duocarmycins, possesses marked DNA-binding activity upon activation by carboxyl esterase to its active form, DU-86. Three duocarmycins, KW-2189, DU-86 and duocarmycin SA, were active against the cisplatin (CDDP)-resistant human non-small cell lung cancer cell lines PC-9/CDDP and PC-14/CDDP, and the multidrug-resistant human small cell lung cancer cell line H69/VP. However, HAC2/0.1, a CDDP-resistant human ovarian cancer cell line which is also resistant to CPT-11 because of decreased intracellular activation of CPT-11, was about 12.8-fold more resistant to KW-2189. HAC2/0.1 was not resistant to other duocarmycins as compared to its parental cell line, HAC2. There was no difference between HAC2 and HAC2/0.1 with regard to the intracellular accumulation of KW-2189. Addition of 130 mU/ml of carboxyl esterase to the culture medium did not influence the sensitivity of HAC2 cells to KW-2189. However, the sensitivity of HAC2/0.1 cells to KW-2189 was enhanced to the level of HAC2. These results suggest that HAC2/0.1 is less potent than HAC2 in activating KW-2189. The carboxyl esterase activity of whole-cell and microsomal extracts from HAC2/0.1 was approximately 60% of that from HAC2. The cell-free experiment revealed that KW-2189 bound to DNA more efficiently in the presence of HAC2 than HAC2/0.1 cell extract. It was concluded that decreased intracellular carboxyl esterase activity in HAC2/0.1 cells caused decreased intracellular conversion of KW-2189 to its active form, thus producing resistance to KW-2189. The decreased conversion of CPT-11 to SN-38 in HAC2/0.1 cells might be explained by decreased carboxyl esterase activity.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carboxylic Ester Hydrolases/metabolism , Drug Resistance , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carboxylic Ester Hydrolases/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/metabolism , Cisplatin/therapeutic use , DNA/metabolism , Drug Resistance, Multiple , Duocarmycins , Female , Humans , Irinotecan , Lung Neoplasms/metabolism , Microsomes/enzymology , Ovarian Neoplasms/metabolism , Pyrrolidinones/metabolism , Pyrrolidinones/therapeutic use , Tumor Cells, Cultured
17.
Jpn J Cancer Res ; 85(10): 1050-6, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7961107

ABSTRACT

The effects of cisplatin (CDDP) and etoposide (ETP) in combination were evaluated in vitro and in vivo using small cell lung cancer cell lines. The combination effects in vitro were investigated using isobologram analysis. Used together, CDDP and ETP showed a synergistic effect against cell growth on only 1 cell line (SBC-3), additive effects on 6 (SBC-2, SBC-5, Lu130, Lu134AH, Lu135T and H69) and an antagonistic effect on 1 (SBC-1). In the in vivo experiment, nude mice were inoculated with SBC-1, SBC-3 and SBC-5 cells. Two or 5 mg/kg CDDP and 10 or 30 mg/kg ETP were administered intraperitoneally alone and simultaneously in combination to nude mice. The in vivo effects of the combination were determined by comparing the observed growth ratio in mice treated with the combination with the expected value of this ratio calculated based on the assumption that the effects of the drugs were simply additive. According to this definition, synergistic effects were observed against all 3 tumors. Thus, the in vivo and in vitro effects differed. The toxicity of the combination therapy, which was analyzed by estimating the body weight change of mice, was no higher than that of CDDP or ETP alone. These results suggest that the excellent clinical effects of CDDP and ETP combination therapy may be attributable not to drug interaction at the cellular level but to the feasibility of combined use of them at full doses without overlapping side effects.


Subject(s)
Carcinoma, Small Cell/drug therapy , Cisplatin/administration & dosage , Etoposide/administration & dosage , Lung Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Body Weight/drug effects , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Cancer Chemother Pharmacol ; 34 Suppl: S112-7, 1994.
Article in English | MEDLINE | ID: mdl-8070019

ABSTRACT

The camptothecin derivative 7-ethyl-10-[4-(1-piperidino)-1-piperidino]-carbonyloxy camptothecin (CPT-11) has attracted the attention of clinicians because of its high antitumor activity against refractory solid cancers. We established two CPT-11-resistant cell lines, a non-small-cell lung-cancer cell line (PC-7/CPT-11) from the parental PC-7 line and an ovarian cancer cell line (HAC-2/CPT-11) from the parental HAC-2 line. The mechanisms of resistance to CPT-11 in PC-7/CPT-11 cells were reduced conversion of CPT-11 to its active metabolite SN-38 and point mutation topoisomerase I. Those in HAC-2/CPT-11 cells were reduction of topoisomerase I activity and decreased sensitivity of topoisomerase to topoisomerase I inhibitors. No point mutation of the topoisomerase was observed in HAC-2/CPT-11 cells. We conducted two phase I trials using CPT-11 in combination with other anticancer agents. One was a phase I trial of CPT-11 and cisplatin given with a fixed dose of vindesine to patients with advanced non-small-cell lung-cancer and the other was a phase I study on a topoisomerase-targeting combination of CPT-11 and etoposide (VP-16) in patients with various malignant solid tumors. The results of the first trial indicated that the recommended dose of CPT-11 for phase II studies was 80 mg/m2 combined with 3 mg/m2 vindesine on days 1 and 8 and 60 mg/m2 cisplatin on day 1. In the second trial, the recommended dose of CPT-11/VP-16 given with recombinant granulocyte colony-stimulating factor (on days 4-17) was found to be 60/60 mg/m2. In both trials, diarrhea and granulocytopenia were considered to be dose-limiting toxicities.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Camptothecin/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Base Sequence , CHO Cells , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Camptothecin/toxicity , Cell Line , Cell Survival/drug effects , Clinical Trials, Phase I as Topic , Cricetinae , DNA Topoisomerases, Type I/genetics , Drug Resistance/genetics , Etoposide/administration & dosage , Female , Humans , Irinotecan , Lung Neoplasms , Molecular Sequence Data , Ovarian Neoplasms , Point Mutation , Topoisomerase I Inhibitors , Tumor Cells, Cultured
19.
Biochem Biophys Res Commun ; 188(2): 571-7, 1992 Oct 30.
Article in English | MEDLINE | ID: mdl-1332703

ABSTRACT

CPT-11, a recently developed topoisomerase I (Topo I) inhibitor, attracts the attention not only of basic researchers but also of clinicians because of its high antitumor activity. The CPT-11 resistant human lung cancer cell line, PC-7/CPT, showed 10-fold resistance compared to parental cell line, PC-7. The total activity of Topo I in the resistant cell line was one fourth that of the parental sensitive cell line. The Topo I from the resistant cells was also 5-fold more resistant to the inhibitory effect of CPT-11 than that of the parental cells. We speculated that the alteration of the Topo I gene may be responsible for the change in topoisomerase activity of the CPT-11 resistant cell line. Therefore, we analyzed the mutation of Topo I gene using the method of single strand conformation polymorphism of polymerase chain reaction and the reverse transcriptase. We divided Topo I cDNA into ten fragments which overlapped each other and covered whole coding sequences of the Topo I cDNA. We observed mobility shift of two fragments in the PC-7/CPT, suggesting the presence of some mutations in these fragments. We performed the direct-sequencing of these portions by the dideoxy chain termination method and observed an altered sequence having a G to A base change in PC-7/CPT. This base substitution results in replacement of the conserved threonine at 729 position with alanine. These results suggest that the point mutation of Topo I gene is related to the decreases of Topo I activity and the sensitivity to Topo I inhibitor in PC-7/CPT cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , DNA Topoisomerases, Type I/genetics , Drug Resistance/genetics , Point Mutation , Base Sequence , Camptothecin/pharmacology , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Humans , Irinotecan , Lung Neoplasms , Molecular Sequence Data , Oligodeoxyribonucleotides , Polymerase Chain Reaction/methods , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Tumor Cells, Cultured
20.
Cancer Res ; 50(18): 5919-24, 1990 Sep 15.
Article in English | MEDLINE | ID: mdl-2168285

ABSTRACT

Camptothecin-11 (CPT-11) is a new derivative of camptothecin, a plant alkaloid antitumor agent, and a good candidate for clinical trials because of higher antitumor activity, less toxicity, and high aqueous solubility. CPT-11 is known to be altered into an active form, SN-38, by esterase in in vivo. CPT-11-resistant cells (PC-7/CPT) established from a human non-small cell lung cancer cell (PC-7) by stepwise, continuous treatment with CPT-11 exhibit about a 10-fold increase in resistance to the drug. CPT-11-resistant cells show a moderate cross-resistance to camptothecin (x8.6) and SN-38 (x8.6), and weak cross-resistance to Adriamycin (x2.2) and 5-fluorouracil (x2.4). The comparative studies between the parent (PC-7) and resistant (PC-7/CPT) cell lines with respect to their growth characterization shows a longer cell doubling time (45.8 versus 35.5 h), a lower cloning efficiency (3.2 versus 7.1%), and a lower population of S-phase cells (26.4 versus 36.0%) in the CPT-11-resistant cells. This observation may partly explain the resistance to CPT-11, a drug whose activity is cell cycle specific. Accumulation of CPT-11 is nearly the same in both cell lines. However, the intracellular concentration of SN-38 formed in the parent cells was 2-fold greater than in the CPT-11-resistant cells. This alteration may affect to some extent to the resistance. As assayed by relaxation of supercoiled plasmid DNA, the total activity of DNA topoisomerase I from the CPT-11-resistant cells was shown to be reduced to one-fourth its level in sensitive cells. The reduced activity was caused by a reduction of amount of DNA topoisomerase I. Furthermore, the enzyme from the resistant cells was shown to be 5-fold more resistant to CPT-11 than the enzyme from the parent cells. Thus, decreased total activity of topoisomerase I may play an important role in cellular resistance to CPT-11, and it appears that this decreased activity is due to a resistant form of topoisomerase I in CPT-11 resistant cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Camptothecin/metabolism , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Nucleus/enzymology , DNA Topoisomerases, Type I/analysis , Drug Resistance , Humans , Irinotecan , Lung Neoplasms/enzymology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...