Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Oncogene ; 25(17): 2509-19, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16518418

ABSTRACT

Until recently, the use of Se-methylselenocysteine (MSC) as selective modulator of the antitumor activity and selectivity of anticancer drugs including irinotecan, a topoisomerase I poison, had not been evaluated. Therapeutic synergy between MSC and irinotecan was demonstrated by our laboratory in mice bearing human squamous cell carcinoma of the head and neck tumors. In FaDu xenografts, a poorly differentiated tumor-expressing mutant p53, the cure rate was increased from 30% with irinotecan alone to 100% with the combination of irinotecan and MSC. Cellular exposure to cytotoxic concentration of SN-38, the active metabolite of irinotecan (0.1 microM) alone and in combination with noncytotoxic concentration of MSC (10 microM) did not result in additional enhancement of chk2 phosphorylation and downregulation of specific DNA replication-associated proteins, cdc6, MCM2, cdc25A, nor increase in PARP cleavage, caspase activation and the 30-300 kb DNA fragmentation induced by SN-38 treatment. MSC did not alter significantly markers associated with apoptosis, nor potentiate irinotecan-induced apoptosis. These results indicate that apoptosis is unlikely to be one of the main mechanism associated with the observed in vivo therapeutic synergy. In contrast, significant downregulation of cyclooxygenase-2 (COX-2) expression and activity was observed in the cells exposed to SN-38 in combination with MSC compared to SN-38 alone. Moreover, the inhibition of PGE(2) production was also observed in the cells treated with the combination as compared with SN-38 alone. Analysis of tumor tissues at 24 h after treatment with synergistic modality of irinotecan and MSC revealed significant downregulation of COX-2, inducible nitric oxide synthase (iNOS) and hypoxia-induced factor-1alpha expression (HIF 1alpha). Moreover, decreased microvessel density was observed after irinotecan treatment with the addition of MSC. These results suggest that observed therapeutic synergy correlates with the inhibition of neoangiogenesis through the downregulation of COX-2, iNOS and HIF-1alpha expression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cyclooxygenase 2/metabolism , Head and Neck Neoplasms/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/prevention & control , Nitric Oxide Synthase Type II/metabolism , Animals , Apoptosis/drug effects , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/genetics , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Down-Regulation , Female , Head and Neck Neoplasms/genetics , Humans , Irinotecan , Mice , Mice, Nude , Organoselenium Compounds/administration & dosage , Selenocysteine/analogs & derivatives , Transplantation, Heterologous
2.
Cancer Metastasis Rev ; 20(1-2): 109-15, 2001.
Article in English | MEDLINE | ID: mdl-11831638

ABSTRACT

The common clinical problem in the successful treatment of cancer is the resistance of cancer cells to chemotherapeutic drugs. Chemotherapy kills drug-sensitive cells, but leaves behind a higher proportion of drug-resistant cells. The resistance can be due to altered drug accumulation, retention, metabolism and distribution, or to reduced drug-target interaction. More recently, cell cycle progression, DNA mismatch repair (MMR) and cell death have been shown to play an important role in the regulation of cell resistance to anticancer drugs. Chkl regulation pathways, DNA MMR and p73, as well as altered apoptotic cell death involved in the cell resistance toward DNA damaging agents will be reviewed in this article.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Protein Kinase Inhibitors , Protein Kinases , Checkpoint Kinase 1 , Humans
3.
Mol Pharmacol ; 57(3): 453-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10692484

ABSTRACT

Cellular topoisomerase I is an important target in cancer chemotherapy. A novel karenitecin, BNP1350, is a topoisomerase I-targeting anticancer agent with significant antitumor activity against human head and neck carcinoma A253 cells in vitro. As a basis for future clinical trials of BNP1350 in human head and neck carcinoma, in vitro studies were carried out to investigate its effect on DNA damage and cell cycle checkpoint response. The treatment of A253 cells with BNP1350 caused biphasic profiles of DNA fragmentation displayed from 0 to 48 h after 2-h exposure. Pulsed-field gel electrophoresis demonstrated that the first wave of DNA damage was mainly megabase DNA fragmentation, but the second wave of DNA damage was 50- to 300-kb DNA fragmentation in addition to megabase DNA damage. The cell cycle checkpoint response was characterized after exposure to 0.07 and 0.7 microM concentrations of BNP1350, the IC(50) and IC(90) values, respectively. After exposure to a low concentration of BNP1350 (IC(50)), A253 cells accumulated primarily in G(2) phase. In contrast, treatment with a high concentration of BNP1350 (IC(90)) resulted in S phase accumulation. The concentration-associated cell cycle perturbation by BNP1350 was correlated with different profiles of cell cycle-regulatory protein expression. When treated with the low concentration of BNP1350, cyclin B/cdc2 protein expression was up-regulated, whereas with the high concentration, no significant change was observed at 24 and 48 h. In addition, increased phosphorylation of a G(2) checkpoint kinase chk1 was observed when cells were treated with a low concentration of BNP1350, whereas only slight inhibition of chk1 activity was found in the cells treated with the higher concentration. Altered chk1 phosphorylation after DNA damage appears to be associated with specific phases of cell cycle arrest induced by BNP1350. Because A253 cells do not express the p53 protein, the drug-induced alterations of the G(2) checkpoint kinase chk1 are not p53-dependent.


Subject(s)
Camptothecin/analogs & derivatives , Cell Cycle/drug effects , Protein Kinases/metabolism , Topoisomerase I Inhibitors , CDC2 Protein Kinase/analysis , Camptothecin/pharmacology , Cell Cycle Proteins/biosynthesis , Cell Division/drug effects , Checkpoint Kinase 1 , Cyclin B/analysis , DNA Fragmentation/drug effects , DNA Topoisomerases, Type I/metabolism , Electrophoresis, Gel, Pulsed-Field , Head and Neck Neoplasms , Humans , Phosphorylation , Protein Kinases/drug effects , Tumor Cells, Cultured
4.
Int J Cancer ; 83(3): 341-8, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10495426

ABSTRACT

Time-dependent ladder-type DNA fragmentation and morphological alterations consistent with apoptosis were observed among A253 human head and neck squamous cell carcinoma (HNSCC) cells in nude mice from 15 to 18 days after transplantation, without any drug treatment. No evidence of ladder-type DNA fragmentation was detected in A253 cells in vitro or in normal nude mouse tissues (skin and muscle). Our aim was to explore molecular factors associated with such spontaneous apoptosis. Bcl-2 protein expression decreased, while bax protein expression increased from day 9 after transplantation. Moreover, altered expression of bcl-2 and bax was accompanied by the increased proteolytic cleavage of poly(ADP-ribose) polymerase (PARP). Time-dependent dephosphorylation of Rb, followed by proteolytic cleavage, was also observed from day 9 after transplantation. The data indicate that the caspase-3 activation and cleavage of Rb protein may represent important steps in the regulation pathway of bax-mediated spontaneous apoptosis. Interestingly, the time-dependent activation of spontaneous apoptosis was almost simultaneous with the induction of differentiation and increased expression of several differentiation-associated regulatory proteins. An increased expression of cyclin D1 and cyclin-dependent kinase-5 (cdk5) was observed from day 9 after transplantation, whereas only slight alteration of cdk4 expression was found. The time-dependent activation of cyclin D1 and cdk5 preceded both the induction of ladder-type DNA fragmentation and increased keratin pearl formation. Furthermore, MCM3 was cleaved early in spontaneous apoptosis and differentiation. Our observations suggest the involvement of cyclin D1-cdk5 overexpression and MCM3 cleavage in bax-mediated spontaneous apoptosis and differentiation in A253 xenografts. P53 and WAF1 proteins were not expressed in the xenografts, indicating that the changes in the regulatory proteins during apoptosis and differentiation were not p53 or WAF1 dependent.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Cyclin D1/physiology , Cyclin-Dependent Kinases/physiology , Head and Neck Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Animals , Caspase 1/physiology , Cell Differentiation , Cyclin-Dependent Kinase 5 , DNA Fragmentation , DNA-Binding Proteins , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Minichromosome Maintenance Complex Component 3 , Neoplasm Transplantation , Nuclear Proteins , Transplantation, Heterologous , Tumor Cells, Cultured , bcl-2-Associated X Protein
5.
Oncol Res ; 11(2): 91-9, 1999.
Article in English | MEDLINE | ID: mdl-10489165

ABSTRACT

Human head and neck squamous cell carcinoma A253 cells, which do not express p53 and p21 proteins, were engineered to stably express about 50-fold higher level of Bax protein (A253/Bax) than the mock-transfected (A253/vec) or parental cells. Using these cell lines, studies were carried out to evaluate the role of Bax in response to anticancer drugs and to study the associated mechanisms. A253/Bax cells exhibited a significant increase in in vitro sensitivity to various anticancer drugs, including tomudex (9.5-fold), SN-38 (13.8-fold), doxorubicin (7.9-fold), taxol (3.1-fold), 5-FU (2.7-fold), and 5-FU/LV (4.5-fold). Increased level of drug-induced apoptosis was observed in A253/Bax cells in a drug concentration-dependent manner. In untreated A253/Bax cells, Bax was expressed in a monomeric state. Treatment with tomudex induced the formation of Bax dimer in a drug concentration-dependent manner. Dimerization of Bax occurred only in mitochondria, while the cytosolic Bax was retained in the monomeric state. Low level of Bax dimerization was also detected in parental A253 cells following tomudex exposure. In addition, Bax dimer formation was associated with mitochondrial cytochrome c release and activation of caspases in A253/Bax cells. The data suggest that Bax overexpression increases drug response by enhancing drug-induced apoptosis. Furthermore, dimerization of mitochondrial Bax and downstream mechanisms are associated with drug-induced apoptotic cell death and increased drug sensitivity.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Cytochrome c Group/metabolism , Dimerization , Dose-Response Relationship, Drug , Enzyme Activation , Head and Neck Neoplasms/genetics , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Transfection/genetics , Tumor Cells, Cultured , bcl-2-Associated X Protein
6.
Exp Cell Res ; 247(1): 189-99, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10047461

ABSTRACT

Tomudex (ZD1694) is a specific antifolate-based thymidylate synthase inhibitor active in a variety of solid tumor malignancies. Studies were carried out in vitro to evaluate downstream molecular alterations induced as a consequence of the potent and sustained inhibition of thymidylate synthase by Tomudex. Twenty-four hours following the initial 2-h treatment with Tomudex, human A253 head and neck squamous carcinoma cells, not expressing p53 and p21(WAF1), were accumulated with DNA content characteristic of early S phase of the cell cycle with a concomitant reduction of cells in G1 and G2/M phases. The changes in cyclin and cdk protein expression and their kinase activities were examined in control and drug-treated A253 cells. Tomudex treatment resulted in the decrease in p27(kip1) expression, with an increase in cyclin E and cdk2 protein expression and kinase activities 24 h after a 2-h exposure. Although cyclin A protein expression was markedly increased, cyclin A kinase activity was only slightly increased. Cyclin D1, cyclin B, cdk4, and cdc2 protein expression and kinase activities remain constant. Lack of activation of cyclin A- and B-cdc2 was associated with a reduced proportion of cells in G2/M phases. Increased cyclin E-cdk2 protein expression was accompanied by the inhibition of DNA synthesis, with a decrease in E2F-1 expression. These results propose that cyclin E-cdk2 kinase can negatively regulate DNA replication. The studies with dThyd rescue from cyclin E-cdk2 protein overexpression and growth inhibition by Tomudex indicate that increased cyclin E-cdk2 protein expression is associated with effective inhibition of thymidylate synthase and resultant dNTP pool imbalance. Provision of dThyd more than 24 h after exposure to Tomudex allowed cells to replicate DNA for a single cycle back to G1, but did not prevent the profound growth-inhibitory effect manifested in the following 5 days. Tomudex treatment resulted in a time-dependent induction of the megabase DNA fragments, followed by secondary 50- to 300-kb DNA fragmentation. The 50- to 300-kb DNA fragmentation may be derived from the inhibition of DNA synthesis associated with cyclin E-cdk2 activation. These results suggest that the megabase DNA fragmentation is induced as a consequence of inhibition of thymidylate synthase by Tomudex and kilobase DNA fragmentation may correlate with the reduction of p27(kip1) expression and the increase in cyclin E and cdk2 kinase activities. Activation of cyclin E and cdk2 kinases allows cells to transit from G1 to S phase accompanied by the inhibition of DNA synthesis. The changes in cell cycle regulatory proteins associated with growth inhibition and DNA damage by Tomudex are not p53 dependent.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cell Cycle/drug effects , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins , DNA/antagonists & inhibitors , DNA/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Cyclin A/chemistry , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/biosynthesis , DNA Fragmentation/drug effects , DNA, Neoplasm/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Humans , Macromolecular Substances , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Retinoblastoma-Binding Protein 1 , Thymidine/physiology , Transcription Factor DP1 , Transcription Factors/biosynthesis , Tumor Cells, Cultured
7.
Anticancer Res ; 18(5A): 3499-505, 1998.
Article in English | MEDLINE | ID: mdl-9858931

ABSTRACT

BACKGROUND: SN-38 is the active metabolite of the topoisomerase-I (topo-I) inhibitor Irinotecan (CPT-11). Generally, topo-I inhibitors stabilize the complex between topo-I and DNA which collide with moving DNA replication forks, eventually leading to double stranded DNA damage. Therefore, topo-I inhibitors are regarded as S-phase specific. The present study investigated S-phase dependent and independent effects of SN-38. MATERIALS AND METHODS: Effects of exposure of A2780 cells to SN-38 (2 hours) were studied by assessing DNA/protein crosslinks, DNA damage and cytogenetic aberrations. RESULTS: A close correlation (r2 = 0.97) was established between drug-induced DNA/protein crosslinks and double stranded DNA breaks. Cytogenetic analysis revealed near maximum clastogenic effects already evident immediately following 2 hours drug exposure. However, qualitatively, chromatid breaks at 24 hours were different from those at 0 hours, in that at 24 hours they were associated with radial chromosome configurations and sister chromatid exchanges. CONCLUSION: The data corroborate that the S-phase dependent mechanism of action of topo-I inhibitors is also applicable to SN-38. The cytogenetic data indicate two distinct interactions of SN-38 with DNA: immediate induction of chromatid breaks independent from DNA synthesis, and induction of chromatid breaks associated with radial chromosome configurations dependent on DNA synthesis.


Subject(s)
Camptothecin/analogs & derivatives , Chromatids/drug effects , DNA Damage , DNA, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Topoisomerase I Inhibitors , Camptothecin/pharmacology , Chromosome Aberrations , DNA Replication , Dose-Response Relationship, Drug , Humans , Irinotecan , S Phase , Sister Chromatid Exchange , Tumor Cells, Cultured/drug effects
8.
Biochim Biophys Acta ; 1401(3): 265-76, 1998 Mar 05.
Article in English | MEDLINE | ID: mdl-9540817

ABSTRACT

The newly synthesized calcium channel blocker, Ro44-5912, significantly potentiates doxorubicin (Dox)-induced cytotoxicity at non-cytotoxic concentrations in Dox-resistant human ovarian cell line, A2780/DX5, overexpressing P170-glycoprotein (Pgp). Induction of DNA single- and double-strand breaks (ssbs and dsbs) was measured using alkaline elution and constant-field gel electrophoresis (CFGE) assays. The results indicate that potentiation of the cytotoxicity of Dox by Ro44-5912 was accompanied by significant increases in both, Dox-induced DNA ssbs and dsbs in the resistant cells. Pulsed-field gel electrophoresis (PFGE) analysis showed that Dox induced DNA fragments in the 50-800 kilobase (kb) and 0.8-5.7 megabase (Mb) ranges. The majority of the newly synthesized DNA fragments were in the 50-800 kb range. Ro44-5912 treatment resulted in significant potentiation of DNA fragmentation in the 50-800 kb range with a minor increase in 0.8-5.7 Mb DNA fragments, suggesting that the modulator functions by potentiating nascent DNA fragmentation in the resistant cells. Exposure to Dox with Ro44-5912 was associated with a prolonged blockage of cells in the S-phase. In contrast, exposure to Dox alone resulted in temporary blockage of cells in G2/M phase (approximately 24 h) followed by restoration of cell proliferation and normal DNA histograms at 48 h after 2 h drug exposure. Incorporation of BrdUrd by flow cytometric analysis was inhibited by Dox in the presence of Ro44-5912, showing that there is a block of DNA replication. An increased damage in newly synthesized DNA could concur with a blocked DNA replication. Moreover, slowing progression through the S-phase in cells exposed to Dox in combination with Ro44-5912 is accompanied by increased sensitivity of Dox poisons, indicating a correlation of specific S-phase perturbation with the reversal of Dox resistance by Ro44-5912 in cells expressing Pgp. The results suggest that drug-induced augmentation of nascent DNA fragmentation and specific cell-cycle perturbation are potentially important molecular determinants for reversal of multidrug resistance in addition to restoration of intracellular drug retention.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , DNA Damage , DNA, Neoplasm/drug effects , Drug Resistance, Multiple/physiology , Antibiotics, Antineoplastic/pharmacology , Calcium Channel Blockers/pharmacology , DNA Replication , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , G2 Phase/drug effects , Humans , Ovarian Neoplasms , Tumor Cells, Cultured , Verapamil/analogs & derivatives , Verapamil/pharmacology
9.
Anticancer Res ; 17(5A): 3707-11, 1997.
Article in English | MEDLINE | ID: mdl-9413228

ABSTRACT

The prognostic value of topoisomerase I (Topo I) catalytic activity and expression of the multidrug resistance (MDR) marker P-glycoprotein (Pgp) and multidrug resistance protein (MRP) for in vitro sensitivity to Topo I interactive agents were evaluated. The efficacy of short term (2 h) and long term (24 h) exposures of camptothecin (CPT), two CPT derivatives (SN-22, SN-38) and the indolocarbazole compound NB-506, was determined against human ovarian carcinoma (A2780 and A2780 DX5), human fibrosarcoma (HT1080 and IIT1080/DR4) and human ileocecal carcinoma (HCT-8). For each cell line the Topo I protein levels and catalytic activity were determined and correlated with drug-induced cytotoxicity. In general, the Topo I protein levels correlated with Topo I catalytic activity. Drug-induced cytotoxicity increased significantly with prolongation of the exposure time. With the 2 h exposure, the multidrug resistant A2780 DX5 cell line (Pgp+, MRP-) was moderately resistant to all four drugs compared to its parental cell line. In case of CPT and SN-22 but not for SN-38 and NB-506, this resistance was no longer detectable following 24 h drug exposure. No resistance was detectable for the multidrug resistant HT1080/DR4 (Pgp-, MRP+) cell line when compared to its parental cell line. With short term exposures a strong trend was observed toward increased cytotoxicity with increased Topo I catalytic activity, especially if this correlation was studied between derivative cell lines (A2780 vs. A2780 DX5 and HT1080 vs. HT1080/DR4). This correlation weakened when all 5 cell lines and both exposure conditions were considered. Thus, although overall the correlation between Topo I catalytic activity and sensitivity to Topo I interactive drugs between different cell lines is weak, this correlation may be stronger when comparing derivative cell lines. For CPT and SN-22 but not for SN-38 and NB-506, the moderate resistance levels observed in the Pgp-expressing cell line could be negated by prolongation of exposure duration. MRP expression did not effect drug efficacy. The data demonstrate that the importance of Topo I catalytic activity as single prognostic factor for drug response to Topo I interactive agents is weak and that additional mechanisms affecting drug response have to be taken into consideration.


Subject(s)
Camptothecin/pharmacology , DNA Topoisomerases, Type I/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Carbazoles/pharmacology , Catalysis , Enzyme Inhibitors/pharmacology , Glucosides/pharmacology , Humans , Irinotecan , Topoisomerase I Inhibitors , Tumor Cells, Cultured
10.
Mol Pharmacol ; 51(4): 630-6, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9106628

ABSTRACT

In a previous study, we found that treatment of HCT-8 cells with ZD1694, a specific antifolate-based thymidylate synthase inhibitor, resulted in DNA fragmentation. In this study, we have demonstrated the dose- and time-dependent induction of DNA fragmentation accompanied by elevation of p53 and WAF1 protein expression by ZD1694. WAF1 mRNA showed a time-dependent increase, whereas p53 mRNA was not found to be significantly overexpressed. The initial increase in WAF1 mRNA was detected at 4 hr, but increased WAF1 protein expression was detected 8-24 hr after a 2-hr exposure. The amount of total and hypophosphorylated pRb seems to be rising greatly after ZD1694 exposure. The effects of ZD1694 on the expression of E2F1 and formation of the E2F1-Rb complex were investigated after a 2-hr drug exposure (IC90). The results showed a time-dependent decrease in E2F1 mRNA and protein expression; an increase in the abundance of the E2F-Rb complex could be demonstrated beginning 4 hr after drug exposure by a gel shift assay. Kinetic analysis showed increased availability of hypophosphorylated pRb for inhibition of E2F, which could indirectly result from WAF1-induced inhibition cyclin-dependent kinase activity. Whereas thymidylate synthase inhibition by ZD1694 was rapid in onset and maintained for at least 24 hr after drug treatment, drug-induced cellular growth inhibition was significant 24 hr after drug exposure. The increased abundance of hypophosphorylated pRb and binding to transcription factor E2F-1 is consistent with ZD1694-induced cell growth inhibition in HCT-8 cells. Therefore, the observed effect on downstream events after effective inhibition of thymidylate synthase may offer the critical determinants of response to ZD1694.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cyclins/biosynthesis , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Quinazolines/pharmacology , Retinoblastoma Protein/metabolism , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/metabolism , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , DNA, Neoplasm/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression , Humans , Ileal Neoplasms/metabolism , Phosphorylation , RNA, Messenger/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Cells, Cultured
11.
Biochem Pharmacol ; 53(6): 801-9, 1997 Mar 21.
Article in English | MEDLINE | ID: mdl-9113101

ABSTRACT

Intracellular glutathione (GSH) concentrations have been implicated recently as a regulatory determinant of multidrug resistance protein (MRP)-mediated drug efflux. Inhibition of glutathione reductase (GR) activity of N,N-bis(2-chloroethyl)-N-nitrosourea (BCNU) has been employed extensively to investigate the role of GSH redox cycle in cellular function. The present study examined the effect of BCNU on the MRP-mediated efflux of doxorubicin in the multidrug-resistant human fibrosarcoma cell line HT1080/DR4 overexpressing MRP. No significant difference in GR activity between HT1080 (parental) and multidrug-resistant HT1080/DR4 cells was detected (38.6 +/- 2.2 and 37.8 +/- 5.28 nmol/min/10(6) cells, respectively). Exposure of HT1080 and HT1080/DR4 cells to 100-500 microM BCNU decreased GR activity concentration dependently with subsequent reduction in cellular GSH pools in both cell lines. Inhibition of GSH biosynthesis by D,L-buthionine-(S,R)-sulfoximine (D,L-BSO), a specific inhibitor of gamma-glutamylcysteine synthetase, significantly reduced MRP-mediated drug efflux and potentiated the cytotoxicity of doxorubicin in MRP-expressing HT1080/DR4 cells (dose modifying factor 20.8). While equally effective inhibition of GR activity by BCNU was observed in parental and resistant cells, a significant increase in intracellular retention of doxorubicin was only achieved in MRP-expressing HT1080/DR4 cells. Furthermore, inhibition of MRP function following treatment with BCNU or D,L-BSO was directly related to the degree of GSH depletion in MRP-expressing tumor cells [r = 0.94 (P < 0.001) and 0.99 (P < 0.001), respectively]. Based on northern blot analysis of MRP mRNA levels, exposure of HT1080/DR4 cells to BCNU did not produce down-regulation of MRP gene expression. The results reported herein indicate that derivatives of nitrosourea with carbamoylating properties are potent inhibitors of MRP function. Depletion of intracellular GSH pools by inhibition of the GSH redox cycle or GSH de novo biosynthesis significantly inhibited MRP-mediated doxorubicin transport and restored intracellular drug concentrations in vitro.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Glutathione Reductase/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Buthionine Sulfoximine/pharmacology , Drug Resistance, Multiple , Glutathione/analysis , Glutathione/metabolism , Glutathione Reductase/metabolism , Humans , Multidrug Resistance-Associated Proteins , RNA, Messenger/analysis , Tumor Cells, Cultured
12.
J Clin Oncol ; 15(1): 389-400, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8996166

ABSTRACT

PURPOSE AND METHODS: Although fluoropyrimidines, in particular, fluorouracil (5-FU) and fluorodeoxyuridine (FdUrd), are active alone and in combination with other agents in a variety of human malignancies, therapeutic selectivity, resistance, and efficacy have been a major limitation in cancer therapy. Preclinical and clinical results in advanced and adjuvant colorectal cancers confirmed that the therapeutic efficacy of fluoropyrimidines, with thymidylate synthase (TS) as a primary target, can be improved significantly with leucovorin (LV) modulation. With the recognition that TS is an important therapeutic target, direct and specific inhibitors have been developed and are under intensive preclinical and clinical evaluation, primarily in patients with colorectal cancer, with demonstrable activity. The direct TS inhibitors have been shown to be potent, with a high level of specificity under therapeutic conditions for TS. This includes ZD1694, AG337, and LY231514. To date, although the therapeutic activity of both direct and indirect inhibitors of TS is similar, differences in the magnitude and profile of toxicity have been observed. A phase III comparative evaluation of a direct inhibitor of TS (ZD1694) with an indirect inhibitor (5-FU/LV) has been completed and showed similar activity but reduced toxicity in favor of ZD1694. RESULTS: Recognition that greater than 95% of the injected dose of 5-FU is rapidly inactivated by dihydropyrimidine dehydrogenase (DPD) to therapeutically inactive products, but with toxicity to normal tissues, led to the development of inhibitors of this enzyme with the aim to modify the therapeutic index of 5-FU. Several inhibitors in combination with 5-FU are under preclinical and clinical evaluation, including uracil and 5-chloro-2,4-dihydroxy pyridine, as modulators of 5-FU derived from its prodrug tegafur and 5-ethynyluracil as a modulator of 5-FU. CONCLUSION: In this review, an update of the present status of direct and indirect inhibitors of TS is discussed, as well as the future prospect for new drugs alone and in combination.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Thymidylate Synthase/antagonists & inhibitors , Antimetabolites, Antineoplastic/metabolism , Colorectal Neoplasms/genetics , DNA Damage , DNA, Neoplasm/drug effects , Drug Resistance , Fluorouracil/metabolism , Fluorouracil/therapeutic use , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/therapeutic use , Forecasting , Humans , Prodrugs/metabolism , Prodrugs/therapeutic use , Quinazolines/therapeutic use , Thiophenes/therapeutic use
13.
Adv Enzyme Regul ; 37: 95-109, 1997.
Article in English | MEDLINE | ID: mdl-9381988

ABSTRACT

The rTS proteins have now been shown to be expressed in a variety of cell lines, with expression of rTS beta being found elevated in three cell lines which are resistant to TS inhibitors (3, 4) (Figure 1). In one of these cell lines (K562 B1A), the cells were selected for resistance to MTX, which has a primary site of action on DHFR, but was found to be cross-resistant to FUdR (4). The other two cell lines were selected for resistance to either 5-fluorouracil (H630-1) or a combination of ZD1694 and FU. In each case, elevation of rTS beta appears to be a selected response to thymidylate stress. In HCT-8 and HCT-8/DF2 cells, treatment of cells for a short period of time (2 hr) resulted in the elevation of rTS beta levels, again suggestive that expression of rTS beta is a response to thymidylate stress. rTS beta appears to be regulated with cell growth, its levels increasing at mid-log and at late-log/saturation phase in H630 and H630-1 cells (Fig. 2), and increasing with late-log in several other cell lines as well (Fig. 3). The increase in rTS beta is suggestive of a cellular function associated with a state where growth is no longer desirable, reminiscent of the starvation-sensing protein homolog RSPA in E. coli (22). While this relationship would not explain the spike in rTS beta levels in mid-log H630 and H630-1 cells, it does make sense if the rTS proteins (particularly rTS beta) are involved in down-regulating thymidylate biosynthesis. The potential mechanism of this down-regulation may be speculated to be the catabolism of some precursor for thymidylate biosynthesis or some direct effect upon TS through modulation by some other ligand, either a metabolite or another protein. Studies on the expression of rTS proteins in clinical specimens indicate that rTS beta is expressed at high levels in kidney and kidney tumor (Dolnick, unpublished results). Given the physiologic role of the kidney, high level expression of rTS in this organ is consistent with a role in a catabolic pathway. Since down-regulation of TS activity is expected to increase sensitivity to TS inhibitors, a role for rTS beta in directly down-regulating TS activity in the biochemical sense would seem unlikely. However, the manner of biochemical TS down-regulation may make a difference. In the TS- Cl/Cl cell line, there are two mutations in TS which likely reduce affinity for N-5,10-methylene tetrahydrofolates (23). This cell line is highly resistant to MTX, yet is still tumorigenic in vivo (24), and supplying the cells with high levels of exogenous folate can restore TS function (23). Thus in TS- Cl/Cl cells, the TS phenotype is conditionally dependent upon the presence of high levels of exogenous folate. This suggests that a role of rTS proteins as conditional down-regulators of TS, perhaps through modulating folate binding, may be possible. Two cell lines (K562 B1A and H630-1) that overproduce rTS beta have altered sensitivity to TS inhibitors that differ depending upon the nature of the inhibitor. The K562 B1A cell line was found to be approximately 2000-fold resistant to ZD1694 and BW1843U89 (120 hr exposures), but only three-fold resistant to AG331. The H630-1 cell line is approximately 30-fold resistant to BW1843U89 (120 hr exposure) and 40-fold resistant to ZD1694 (120 hr exposure), but only eight-fold resistant to AG331. Since K562 B1A cells overproduce rTS beta (2), but have no significant alterations in FPGS activity, the possibility that rTS may affect folate binding remains a hypothesis worth examining. The recent discovery that TS is a phosphoprotein and that it is nuclear as well as cytoplasmic (21) raises the possibility that the phosphorylation state of TS may regulate one of its cellular functions, and that the subcellular localization of this enzyme is regulated as well. Since rTS proteins have HSP with proteins that participate in kinase/phosphatase reactions, this also seems to be an avenue worthy of future investigation. (ABSTRACT TRUNCATED


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Proteins/genetics , Thymidylate Synthase/antagonists & inhibitors , Blotting, Western , Cell Division , Electrophoresis, Polyacrylamide Gel , Floxuridine/pharmacology , Folic Acid Antagonists/pharmacology , Humans , Hydro-Lyases , Indoles/pharmacology , Isoindoles , Protein Biosynthesis , Quinazolines/pharmacology , Racemases and Epimerases/chemistry , Racemases and Epimerases/metabolism , Thiophenes/pharmacology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Tumor Cells, Cultured
14.
Oncol Res ; 9(9): 485-94, 1997.
Article in English | MEDLINE | ID: mdl-9495454

ABSTRACT

Membrane protein-associated alterations in cellular drug accumulation have been recently implicated in resistance to topoisomerase I (TOP-I)-interactive drugs. The present study investigated the cellular determinants of resistance to the indolocarbazole compound NB-506 [6-N-formylamino-12,13-dihydro-1,11-dihydroxy-13(beta-D-glucopyranosyl)- 5H-indolo[2,3-alpha]pyrrolo[3,4-c]carbazole-5,7(6H)-dione], a structurally novel TOP-I-interactive drug, in parental and multidrug-resistant tumor cells expressing either the P-170 glycoprotein (Pgp170) or multidrug resistance protein (MRP). MRP-expressing 250-fold doxorubicin-resistant human fibrosarcoma HT1080/DR4 tumor cells were drug sensitive to NB-506 and camptothecin (CPT) (resistance factor: 0.7 and 0.8, respectively) with no alterations of TOP-I parameters including DNA relaxation, expression of TOP-I protein and mRNA. In contrast, doxorubicin-resistant human ovarian A2780/Dx5 tumor cells [pgp170 phenotype] were 6.2-fold resistant to NB-506, whereas resistance to CPT was 2.6-fold. HPLC analysis of cellular NB-506 accumulation showed no significant differences between A2780 and A2780/Dx5 cells (peak intracellular concentrations after 120-min exposure to 10 microM NB-506: 400+/-85.0 and 352+/-95.1 nmol NB-506/mg protein, respectively). However, resistant A2780/Dx5 cells expressed a lower amount of TOP-I mRNA and 29% protein levels of TOP-I compared to parental A2780 cells, resulting in decreased TOP-I catalytic activity (3.17+/-0.02 vs. 1.16+/-0.15 rel.U/microg nuclear protein) and reduced induction of NB-506-mediated cleavable complex formation in A2780/Dx5 cells. Furthermore, the lower induction of NB-506-induced protein-linked DNA breaks (PLDB) in A2780/Dx5 cells correlated with significantly decreased DNA 12.2-440 kb size fragmentation in these cells. The present study demonstrates that expression of MRP and Pgp170 does not confer resistance to NB-506. Resistance to indolocarbazole substance NB-506 in A2780/Dx5 cells was only related to downregulation of TOP-I associated with lower induction of cleavable complex formation and DNA fragmentation. The data reported herein may indicate that the new indolocarbazole compound NB-506 has potent antitumor efficacy in membrane-associated multidrug resistance.


Subject(s)
Carbazoles/pharmacology , Drug Resistance, Multiple , Enzyme Inhibitors/pharmacology , Fibrosarcoma/drug therapy , Glucosides/pharmacology , Ovarian Neoplasms/drug therapy , Topoisomerase I Inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Blotting, Northern , Camptothecin/pharmacology , Cell Count/drug effects , Chromatography, High Pressure Liquid , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Fibrosarcoma/metabolism , Humans , Multidrug Resistance-Associated Proteins , Ovarian Neoplasms/metabolism , RNA, Messenger/metabolism , Tumor Cells, Cultured
15.
Cytometry ; 25(1): 14-20, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8875050

ABSTRACT

Multidrug resistance (MDR) is often related to expression of P-glycoprotein (Pgp) or Multidrug Resistance Protein (MRP). Pgp-mediated MDR can be evaluated by determining cellular retention of fluorescent substrates by flow cytometry. This study determined if agents used to evaluate Pgp function also can be used to evaluate MRP function. Cellular retention of doxorubicin (Dox), Rhodamine-123 (Rh-123), and 3,3'-diethyloxacarbocyanine iodide (DiOC2(3)) were studied in MRP-expressing cell lines (HL60/Adr and HT1080/DR4), whereas a Pgp expressing cell line (A2780/Dx5) served as a positive control. Overexpression of Pgp correlated inversely with retention of Dox, Rh-123, and DiOC2(3); however, under identical experimental conditions (1 h reincubation in drug-free medium), no retention difference of the three agents was detected between parental and MRP-expressing resistant cells. Upon extending the reincubation time to 4 h, an efflux of Rh-123 and Dox in the resistant lines became apparent and even more pronounced after 24h; however, still no efflux was detectable for DiOC2(3). Incubation of the cells with a modulator of MDR, PAK-104P, negated the observed drug efflux in Pgp and MRP expressing cells, which correlated with increased sensitivity of the MDR lines to doxorubicin. Thus both Dox and Rh-123 can be used to evaluate MRP-function, but DiOC2(3) can not.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carbocyanines/metabolism , Flow Cytometry/methods , Fluorescent Dyes/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Biological Transport , Cyclic P-Oxides/pharmacology , Doxorubicin/metabolism , Drug Resistance, Multiple , Female , Humans , Neoplasm Proteins/antagonists & inhibitors , Nicotinic Acids/pharmacology , Rhodamine 123 , Rhodamines/metabolism , Tumor Cells, Cultured
16.
Biochem Pharmacol ; 50(3): 391-8, 1995 Jul 31.
Article in English | MEDLINE | ID: mdl-7646540

ABSTRACT

N-[5-[N-(3,4-Dihydro-2-methyl-4-oxoquinazolin-6-ylmethyl)-N- methylamino]-2-thenoyl]-L-glutamic acid (ZD1694) is a folate-based thymidylate synthase (TS; EC 2.1.1.45) inhibitor. Metabolism to higher chain length polyglutamates is essential for its optimal cytotoxic effect. A ZD1694-resistant (300-fold) human ileocecal carcinoma cell line (HCT-8/DW2) was developed, and its mechanism of resistance was evaluated. TS activities in situ and TS protein levels in the HCT-8 parental line and HCT-8/DW2 were similar (168 +/- 47 vs 137 +/- 25 pmol/hr/10(6) cells and 2.05 +/- 0.28 vs 2.07 +/- 0.19 pmol/mg protein, respectively). The IC50 values of ZD1694 for TS inhibition in cell-free extracts were similar in both lines, but the IC50 of ZD1694 for TS inhibition in situ in HCT-8/DW2 cells was 27- and 268-fold higher than that in HCT-8 cells at 0 and 24 hr, respectively, after a 2-hr drug exposure. Folylpolyglutamate synthetase (FPGS; EC6.3.2.17) activity was significantly lower in resistant HCT-8/DW2 cells as compared with parental HCT-8 cells (88 +/- 40 vs 1065 +/- 438 pmol/hr/mg protein when ZD1694 was used as substrate). The combined endogenous pool of methylenetetrahydrofolate and tetrahydrofolate in HCT-8/DW2 cells was also decreased. In addition, HCT-8/DW2 cells accumulated lower levels of methotrexate (MTX) in a 2-hr period, although the initial velocity of MTX transport was similar to that in parental HCT-8 cells. The lower level of FPGS activity and the lower level of (anti)folate accumulation in HCT-8/DW2 correlated with drug resistance and with the higher IC50 of ZD1694 for in situ TS inhibition. In addition, drug resistance was also correlated with the rapid recovery of in situ TS activity after drug treatment. In brief, in this highly ZD1694-resistant HCT-8 cell line, resistance is associated with decreased FPGS activity, which, in turn, affects the metabolism of ZD1694 and consequently the extent and duration of in situ TS inhibition by the drug.


Subject(s)
Folic Acid Antagonists/pharmacology , Peptide Synthases/antagonists & inhibitors , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Cell Division/drug effects , Cell-Free System , Dose-Response Relationship, Drug , Drug Resistance , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Methotrexate/pharmacology , Pemetrexed , Pteroylpolyglutamic Acids/metabolism , Time Factors , Tumor Cells, Cultured/drug effects
17.
Oncol Res ; 7(2): 73-81, 1995.
Article in English | MEDLINE | ID: mdl-7579730

ABSTRACT

The patterns of DNA fragmentation were evaluated following a brief exposure (2 h) of the human ileocecal adenocarcinoma cell line, HCT-8, to several specific thymidylate synthase inhibitors, a quinazoline (ZD1694) and benz[cd]indole-containing molecule (AG-331). The magnitude and size of DNA fragmentation induced by the two agents were assessed by alkaline elution for DNA single-strand breaks (ssbs), and by pulsed- and constant-field gel electrophoresis for DNA double-strand breaks (dsbs). Both agents induced dose-dependent DNA dsbs. While AG-331 induced ssbs and dsbs only in nascent DNA, ZD1694 affected both genomic and nascent DNA. The fragments of newly synthesized and genomic DNA, estimated by pulsed-field gel electrophoresis assay, were associated with the bands in the range of 0.05 to 1.1 and 1.1 to 5.7 megabases, respectively. 5-fluoro-2'-deoxyuridine (FdUrd), like ZD1694, produced both mature and nascent DNA fragmentation, whereas only nascent DNA breakage induced by 5-fluorouracil (FUra) was detected, similar to AG-331. The induction of both mature and nascent DNA fragmentation by ZD1694 and FdUrd appears to correlate with the higher, but similar, potency of these agents. Aphidicolin, a DNA polymerase inhibitor, protects from DNA dsbs and cytotoxicity by ZD1694 and AG-331. These observations suggest that replicative DNA synthesis is an important factor in ZD1694- and AG-331-induced DNA fragmentation and, subsequently, cell growth arrest. The results indicate that although the new antimetabolites investigated herein were developed and extensively evaluated as specific and potent thymidylate synthase inhibitors, DNA damage appears to be an important additional determinant of drug effect.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA Damage , DNA, Neoplasm/metabolism , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Quinazolines/pharmacology , Thiophenes/pharmacology , Aphidicolin/pharmacology , Cell Division/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Floxuridine/pharmacology , Fluorouracil/pharmacology , Genome , Humans , Thymidylate Synthase/antagonists & inhibitors , Tumor Cells, Cultured/drug effects
18.
Biochem Pharmacol ; 48(5): 997-1002, 1994 Aug 30.
Article in English | MEDLINE | ID: mdl-8093112

ABSTRACT

Heterogeneity in the response of the HCT-8 (human ileocecal adenocarcinoma) tumor cell line to a new thymidylate synthase inhibitor, ICI D1694, was investigated in terms of induction of DNA single-strand breaks and cytotoxicity, applying the single cell alkaline gel (SCG) electrophoresis assay and the individual colony formation assay (iCFA), respectively. ICI D1694 induced maximal total DNA single-strand breaks 24 hr after a 2-hr drug exposure with incomplete repair by 72 hr. The level of DNA damage was concentration dependent and paralleled cellular growth inhibition in vitro. The proportion of cells with DNA damage and the extent of DNA single-strand breaks increased with drug concentration. At 1 microM ICI D1694 (IC95), a significant level of DNA damage was detected in 58% of the cells; however, 25% of the cells had little or no damage. Using the iCFA system, it was observed that with 1 microM ICI D1694, only 2.6% of the seeded cells maintained a colony growth rate similar to that of the control colonies, and 22% of the cells were growing significantly more slowly. In conclusion, the SCG assay and the iCFA identified subpopulations of cells that were unaffected by ICI D1694. Although these cells represented only a small proportion of the total cell population, this phenomenon of heterogeneity in response to ICI D1694 might limit its therapeutic efficacy.


Subject(s)
DNA Damage , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Cell Division/drug effects , Cell Survival/drug effects , DNA/drug effects , DNA Repair , Humans , Tumor Cells, Cultured
19.
Biochem Pharmacol ; 48(2): 327-34, 1994 Jul 19.
Article in English | MEDLINE | ID: mdl-8053928

ABSTRACT

5-Fluoro-2'-deoxyuridine (FdUrd), a potent inhibitor of thymidylate synthase, induces extensive bulk DNA damage at drug concentrations that produce significant in vitro growth inhibition of human ileocecal carcinoma (HCT-8) cells. Constant- and pulsed-field gel electrophoresis (CFGE and PFGE), to detect size distribution of DNA double-strand breaks and repair kinetics, in parallel with northern and western blot analyses, to quantitate c-myc gene and protein expression, were utilized to analyze drug effects. At 24-hr post in vitro drug treatment, when maximum bulk DNA damage was detected, FdUrd produced a broad range of high molecular weight DNA fragments, clustering between 0.1 and 5.7 megabases in size, and resulted in a decrease in the level of c-myc transcripts and protein with no significant effect on the level of v-myc and H-ras. These effects preceded the observed cellular growth inhibition. Addition of the reduced folate leucovorin potentiated the effects induced by FdUrd, indicating that thymidylate synthase inhibition is an important initial step in drug effect followed by DNA fragmentation and suppression of c-myc expression. Changes in the integrity of the genetic materials and regulatory genes occurred prior to the observed cell growth inhibition by FdUrd, suggesting that these molecular alterations by FdUrd may be associated with subsequent FdUrd-induced cell growth inhibition.


Subject(s)
DNA/biosynthesis , Floxuridine/pharmacology , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-myc/genetics , Cell Division/drug effects , DNA Damage , Down-Regulation , Humans , Proto-Oncogene Proteins c-myc/biosynthesis , Tumor Cells, Cultured
20.
Adv Enzyme Regul ; 34: 57-70, 1994.
Article in English | MEDLINE | ID: mdl-7942285

ABSTRACT

The cytotoxicity and molecular effects of antifolate thymidylate synthase inhibitor, ICI-D1694, against human ileocecal carcinoma, were evaluated. The drug concentration for 50% inhibition of cell growth by ICI-D1694 is 73 nM and 3 nM following 2 hr and 72 hr exposure, respectively. The drug induces high level of DNA single strand breaks in a time dependent manner, but subsequent to maximum inhibition of thymidylate synthase. Drug effects can be reversed by thymidine and leucovorin at > 1 microM concentrations. Leucovorin action is primarily at the cell membrane level, competing with the transport and activation of ICI-D1694. Thymidine, however, exerts its competitive effect primarily at the level of thymidylate synthase.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma/metabolism , Intestinal Neoplasms/metabolism , Quinazolines/pharmacology , Thiophenes/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Antimetabolites, Antineoplastic/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Evaluation Studies as Topic , Humans , Leucovorin/metabolism , Quinazolines/toxicity , Thiophenes/toxicity , Thymidine/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...