Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 210
Filter
1.
Br J Cancer ; 110(7): 1733-43, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24619073

ABSTRACT

BACKGROUND: Identification and development of drugs that can effectively modulate the therapeutic efficacy and toxicity of chemotherapy remain an unmet challenge. We evaluated the effects of Se-methylselenocysteine (MSC) on the toxicity and antitumour activity of cyclophosphamide, cisplatin, oxaliplatin, and irinotecan in animal models. METHODS: Cyclophosphamide, cisplatin, and oxaliplatin were administered by a single i.v. injection and irinotecan by i.v. weekly × 4 schedules. For the combination, MSC was administered daily via the oral route for 7 days in mice and daily for 14 days in rats before and concurrent with drug administration. RESULTS: Se-methylselenocysteine significantly protected against organ-specific toxicity induced by lethal doses of cyclophosphamide, cisplatin, oxaliplatin, and irinotecan. These include diarrhoea, stomatitis, alopecia, bladder, kidney, and bone marrow toxicities. Protection from lethal toxicity by MSC was associated with enhanced antitumour activity in rats bearing advanced Ward colorectal carcinoma and in nude mice bearing human squamous cell carcinoma of the head and neck, FaDu, and A253 xenografts. CONCLUSIONS: Se-methylselenocysteine offers selective protection against organ-specific toxicity induced by clinically active agents and enhances further antitumour activity, resulting in improved therapeutic index. These data provided the rationale for the need to clinically evaluate MSC as selective modulator of the antitumour activity and selectivity of anticancer drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytoprotection , Neoplasms/drug therapy , Selenocysteine/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Female , Humans , Irinotecan , Mice , Mice, Nude , Neoplasms/pathology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Rats , Rats, Inbred F344 , Selenocysteine/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
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
3.
Cancer Chemother Pharmacol ; 56(5): 447-54, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15947933

ABSTRACT

Two studies of irinotecan (CPT-11) followed 24 h later by an antimetabolite were conducted. The objectives of the studies were: (1) to determine whether the increase in S-phase in tumor cells seen 24 h after CPT-11 administration in animal studies is seen in advanced solid tumors in patients, (2) to determine the dose of CPT-11 required to produce this effect, (3) to compare two methods (immunohistochemistry, IHC, for cyclin A, and DNA flow cytometry, FC) for evaluating S-phase in tumor biopsies from patients, and (4) to establish the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of CPT-11, given 24 h before gemcitabine (GEM, 1000 mg/m(2)). In one study CPT-11 was followed 24 h later by 5-fluorouracil (5-FU), 400 mg/m(2) per week for 4 weeks every 6 weeks. Tumor biopsies were obtained before and 24 h after CPT-11 administration before administration of 5-FU and assayed for S-phase by IHC for cyclin A and by FC. The starting dose of CPT-11 was 80 mg/m(2) per week with subsequent exploration of 40 and 60 mg/m(2) per week to establish the dose-effect relationship of the increase in tumor cells in S-phase. In the second study, CPT-11 was given 24 h before GEM 1000 mg/m(2) per week for 2 weeks every 3 weeks. Doses of 20-80 mg/m(2) were explored to establish the MTD and DLT and to study tumor cell S-phase in selected patients. CPT-11 80 mg/m(2) produced a mean increase in S-phase by IHC for cyclin A of 137%. Lesser increases were seen with 40 and 60 mg/m(2). CPT-11 followed 24 h later by 5-FU 400 mg/m(2) per week for 4 weeks was well tolerated. In the study of CPT-11 followed by GEM 1000 mg/m(2), 60 mg/m(2) of CPT-11 was the MTD.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Gastrointestinal Neoplasms/drug therapy , Respiratory Tract Neoplasms/drug therapy , S Phase/drug effects , Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biopsy , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cyclin A/analysis , Cyclin A/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Gastrointestinal Neoplasms/pathology , Humans , Irinotecan , Male , Maximum Tolerated Dose , Respiratory Tract Neoplasms/pathology , Gemcitabine
4.
Ann Oncol ; 14(9): 1442-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12954586

ABSTRACT

BACKGROUND: To define the maximum-tolerated dose (MTD) and to evaluate the dose-limiting toxicities (DLTs) of the combination of capecitabine and irinotecan in patients with metastatic colorectal cancer. PATIENTS AND METHODS: Thirty-seven patients with measurable metastatic colorectal cancer with no prior chemotherapy for metastatic disease were treated at three dose levels (DLs). For the first two dose levels, irinotecan (70 mg/m(2)) was administered once a week for 6 weeks in combination with 2 weeks of capecitabine at 1000 mg/m(2) (DL1) or 1250 mg/m(2) (DL2) twice daily, starting on days 1 and 22. In the last dose escalation step, the dose of irinotecan was increased to 80 mg/m(2) (DL3). One cycle lasted 7 weeks. RESULTS: In the subsequent phase I trial, 96 cycles of capecitabine and irinotecan were administered. At DL3, three out of six patients experienced DLTs (diarrhea, neutropenia, asthenia). In order to confirm the safety of the recommended dose, DL2 was extended to 15 patients. Five patients (33%) showed DLTs at this dose level, which was considered too high to embark on further clinical studies. Subsequently, the starting dose (DL1) was extended to a total of 16 patients, with diarrhea being the main toxicity. The overall response rate was 38% [95% confidence interval (CI) 21% to 58%], with a median response duration of 8.7 months (95% CI 6.4-11.5 months). CONCLUSIONS: The recommended doses for further studies are irinotecan 70 mg/m(2) and capecitabine 1000 mg/m(2). The combination of capecitabine and irinotecan appears to have significant therapeutic efficacy with manageable toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/secondary , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Camptothecin/adverse effects , Capecitabine , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Dose-Response Relationship, Drug , Female , Fluorouracil/analogs & derivatives , Humans , Irinotecan , Male , Maximum Tolerated Dose , Middle Aged
5.
Oncogene ; 20(38): 5249-57, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11536038

ABSTRACT

Promotion of apoptosis may potentiate the sensitivity of tumor cells to chemotherapeutic agents, thus improving the efficacy of cancer treatment. The transfection of the proapoptotic bax gene, which results in the overexpression of bax protein, augments the growth inhibition of A253 cells by BNP1350. Increased drug response was associated with the induction of DNA fragmentation in the size of 30-200 Kb, generating a cleaved fragment of 18 kDa from full-length 21 kDa bax and the cleavage of PARP. A253/vec cells treated with 0.07 microM(IC50) of BNP1350 accumulated in G2 phase at 24 h after drug removal. In contrast, A253/Bax cells treated with an equimolar concentration of BNP1350 primarily displayed a G1 phase accumulation with a concurrent decrease in G2 phase. Certain cell cycle regulatory protein expression and activities were altered following drug exposure in both cell lines under similar conditions. Cdk2- and cdc2-associated H1 kinase activities were markedly increased in the A253/Bax cell line with marginal increased activity in the A253/vec cell line. A chk1 activity assay was performed with GST-cdc25C (200-256) or GST-cdc25C(S216A) (200-256) fusion proteins as the substrate. Increased chk1 activity was observed in the A253/vec cell line, with little change in the A253/Bax cell line, when exposed to equimolar concentrations of BNP1350 (0.07 microM). A Western blot of immunoprecipitated chk1 indicated that increased chk1 phosphorylation following DNA damage induced by BNP1350 was accompanied by the observed G2 accumulation in the A253/vec cell line, while only a slight increase in chk1 phosphorylation was seen in the A253/Bax cell line. A decreased expression of cdc25C was observed in the BNP1350-treated A253/Bax cells, but not in the A253/vec cell line. Following exposure to BNP1350, increased binding of 14-3-3 proteins to chk1 occurred in both cell lines, with more being observed in the A253/vec cell line. The data have shown that inhibition of the chk1 pathway accompanied by the abrogation of G2 arrest is involved in sensitizing A253 cells to BNP1350 by bax gene transfer. These findings suggest that bax gene transfer sensitizes A253 cells to BNP1350 through apoptosis promoting and G2/M DNA damage checkpoint regulatory pathways.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Lung Neoplasms/enzymology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Topoisomerase I Inhibitors , cdc25 Phosphatases/metabolism , Apoptosis , CDC2 Protein Kinase/metabolism , Cell Line , Checkpoint Kinase 1 , Cyclin B/metabolism , DNA Fragmentation , Dose-Response Relationship, Drug , G2 Phase , Humans , Inhibitory Concentration 50 , Lung Neoplasms/genetics , Phosphorylation , Protein Binding , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein
6.
J Clin Oncol ; 19(5): 1501-18, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230497

ABSTRACT

PURPOSE AND METHODS: For more than three decades, the therapeutic options for patients with advanced colorectal cancer have almost exclusively been based on fluoropyrimidines. With the recognition that topoisomerase-I (TOP-I) is an important therapeutic target in cancer therapy, irinotecan, a semisynthetic TOP-I-interactive camptothecin derivative, has been clinically established in the treatment of colorectal cancer. RESULTS: Irinotecan was investigated as second-line chemotherapy after prior treatment with fluorouracil (FU)-based regimens in two large randomized phase III trials comparing irinotecan with either best supportive care or an infusional FU/leucovorin (LV) regimen. The outcomes of these trials established irinotecan as the standard therapy in the second-line treatment of colorectal cancer. The therapeutic value of irinotecan in the first-line treatment of metastatic colorectal cancer was investigated in two large randomized phase III trials comparing the combination of irinotecan and FU/LV with FU/LV alone. Both trials demonstrated significant superior efficacy for the combination of irinotecan and FU/LV in terms of response rate, median time to disease progression, and median survival time. Consequently, the combination of irinotecan and FU/LV has been approved as first-line chemotherapy for patients with metastatic colorectal cancer and constitutes the reference therapy against which other treatment options must be tested in the future. CONCLUSION: In this review, the clinical rationale and update of the present clinical status of irinotecan in the treatment of colorectal cancer and future prospects of irinotecan-based combinations are discussed.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/administration & dosage , Camptothecin/pharmacology , Clinical Trials as Topic , Fluorouracil/administration & dosage , Humans , Irinotecan , Leucovorin/administration & dosage , Survival Analysis , Treatment Outcome
7.
Clin Colorectal Cancer ; 1(2): 121-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-12445371

ABSTRACT

Colorectal cancer is one of the most common malignancies in Western nations. Treatment with 5-fluorouracil and leucovorin resulting in inhibition of DNA synthesis after inhibition of thymidylate synthase is the most common strategy for adjuvant and systemic chemotherapy in this disease. However, the majority of patients still fail to respond to this combination; therefore, new chemotherapeutic strategies have been developed. This process has proceeded rapidly due to a more proper translation of preclinical results to the clinic. Knowledge of proximal parameters (drug activation, drug-target interaction) and distal parameters (cell growth inhibitory parameters and cell death parameters) in drug response should offer the opportunity to discriminate between sensitivity and resistance to chemotherapy and therapeutic selectivity. This clinical commentary presents a review of a symposium held at the Vrije Universiteit Medical Center, Amsterdam, The Netherlands, which focused on both the preclinical and clinical aspects relating to the treatment of colorectal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Biomarkers, Tumor , Chemotherapy, Adjuvant , Colorectal Neoplasms/metabolism , Humans , Prognosis
8.
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
9.
Cancer Chemother Pharmacol ; 46(5): 351-6, 2000.
Article in English | MEDLINE | ID: mdl-11127938

ABSTRACT

PURPOSE: To assess the pharmacokinetics of Ftorafur (tegafur, FT), 5-fluorouracil (5-FU), and uracil in 31 cancer patients who were enrolled in phase I studies of oral uracil and FT (UFT). The correlation between pharmacokinetic parameters and toxic effects of UFT was evaluated. METHODS: Uracil and FT were orally administered in a 4:1 molar ratio at FT doses of 200-400 mg/m2 per day. Patients also received leucovorin at 150 mg/day. Daily doses were divided into three doses and administered at 8-h intervals for 28 consecutive days. Plasma FT concentrations were measured by high-performance liquid chromatography, and plasma 5-FU and uracil concentrations were determined using gas chromatography-mass spectrometry. National Institutes of Health Common Toxicity Criteria were used for assessment of toxicity. RESULTS: The concentrations of FT, 5-FU, and uracil showed wide interpatient variations. Maximum plasma concentrations (Cp(max)) of all three compounds were achieved in 0.3 to 4.0 h. At the various study doses, the terminal half-life (t 1/2beta) of FT ranged from 3.9 to 5.9 h, the area under the concentration-versus-time curve (AUC0-6h) ranged from 16,220 to 52,446 (ng/ml)h, the total clearance (ClT) ranged from 100 to 175 ml/min, and the steady-state volume of distribution (Vd(ss)) ranged from 18.3 to 28.7 l. The 5-FU generated from FT had an apparent distribution half-life (t 1/2alpha) and an apparent elimination half-life (t 1/2beta) of 0.3-1.3 h and 4.9-7.0 h, respectively. The AUC0-6h of 5-FU ranged from 120 to 325 (ng/ml)h. Uracil had a t 1/2alpha of 0.2-0.5 h and the level quickly returned to the endogenous level. The AUC0-6h for uracil ranged from 605 to 3764 (ng/ml)h, the ClT ranged from 3225 to 7748 ml/min, and the Vd(ss) ranged from 341 to 1354 l. The Cp(max) and AUC0-6h of both FT and uracil were significantly correlated with FT doses (P-values of 0.0244 and 0.0112) and with uracil doses (P-values of 0.0346 and 0.0083), respectively. In addition to interpatient variations, intrapatient variations were also observed in six patients who had pharmacology studies done on days 1 and 26+/-2 at the same study dose. We found that the repeated treatment with UFT caused cumulative increases in the values of Cp(max), Ctrough, and AUC0-6h of FT and 5-FU. The major toxic effects observed were diarrhea and nausea and vomiting. The occurrence of these toxic effects correlated significantly with the Cp(max) and AUC0-6h of 5-FU. CONCLUSIONS: The pharmacology studies showed that FT and uracil were readily absorbed orally and that FT was rapidly converted to 5-FU. The preliminary findings suggest that determination of plasma levels of 5-FU after oral administration of UFT may help predict subsequent toxic effects.


Subject(s)
Tegafur/adverse effects , Tegafur/pharmacokinetics , Uracil/adverse effects , Uracil/pharmacokinetics , Tegafur/blood , Uracil/blood
10.
Cytometry ; 41(4): 252-60, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11084610

ABSTRACT

BACKGROUND: Cellular heterogeneity in drug response has important clinical implications, and is believed to develop over many generations during clonal evolution in human tumors. The purpose of this study was to determine the level of heterogeneity exhibited by sister cells soon after their birth. METHODS: Human ileocecal carcinoma cells (HCT-8) were followed up to 11 days in vitro after a 2-h exposure to 1 microM raltitrexed (IC(95)) in a time-lapse video system. RESULTS: Over five experiments, 414 cells were followed after exposure to raltitrexed. Immediate sterility occurred in 74% of treated cells. Only 6% of cells could produce more than two generations of offspring, and heterogeneity in drug response was seen. Comparing sister cells < 24 h old, the more proliferative sibling produced up to 73 times more offspring, with a median ratio of 9.0 (control median = 1.19). Offspring of prolific drug-treated cells had a decreased probability of division (68% compared with 92%) and an increased average interdivision time (19.0 h compared with 15.1 h). CONCLUSIONS: Short-term exposure to raltitrexed resulted in increased interdivision times and production of sterile offspring extending seven generations. Cellular heterogeneity (difference in proliferation potential comparing drug-treated sister cells) was evident without a period of clonal evolution.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Ileal Neoplasms/drug therapy , Ileal Neoplasms/pathology , Microscopy, Video/methods , Quinazolines/toxicity , Thiophenes/toxicity , Aged , Cell Division/drug effects , Cell Lineage/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Humans , Male , Probability , Tumor Cells, Cultured
11.
Invest New Drugs ; 18(4): 365-71, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11081572

ABSTRACT

Eniluracil (5-ethynyluracil, GW 776, 776C85) is being developed as a novel modulator of 5-fluorouracil (5-FU) for the treatment of cancer. Eniluracil is an effective mechanism-based inactivator of dihydropyrimidine dehydrogenase (DPD), the first enzyme in the catabolic pathway of 5-FU. By temporarily eliminating this prevalent enzyme, eniluracil provides predictable dosing of 5-FU and enables oral administration of 5-FU to replace intravenous bolus and continuously infused dosing. New DPD is synthesized with a half-life of 2.6 days. It also eliminates the formation of problematic 5-FU catabolites. Most importantly, in laboratory animals, eniluracil increases the therapeutic index and absolute efficacy of 5-FU. Accompanying reports in this journal indicate that eniluracil has promising clinical potential.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Enzyme Inhibitors/pharmacology , Fluorouracil/administration & dosage , Uracil/analogs & derivatives , Uracil/pharmacology , Administration, Oral , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Dihydrouracil Dehydrogenase (NADP) , Drug Synergism , Fluorouracil/pharmacokinetics , Humans , Oxidoreductases/antagonists & inhibitors
12.
Cancer Res ; 60(14): 3717-21, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919639

ABSTRACT

The basis for current clinical trials in the treatment of colorectal cancer with the combination of irinotecan (CPT-11) and 5-fluorouracil (FUra) with or without leucovorin (LV) is their proven activity as single agents, their different mechanisms of action, and lack of CPT-11 cross-resistance to previous FUra/LV treatment. The role of drug dose and administration sequence in this combination was studied in vivo using a rat colon tumor model (Ward colon carcinoma); we administered CPT-11 and FUra by i.v. push once a week for four consecutive weeks (weekly x 4), a clinically relevant schedule. The maximum tolerated doses (MTDs) of CPT-11 and FUra administered as single agents were 100 mg/kg/week for both agents. Three different combination administration sequences were evaluated: (a) CPT-11 administered simultaneously with FUra (sequence I); (b) FUra administered 24 h before CPT-11 (sequence II); and (c) CPT-11 administered 24 h before FUra (sequence III). When combining the two drugs at 50% of their respective MTD, the antitumor efficacy was sequence dependent with 62, 38, and 95% complete tumor regression rate for sequences I, II, and III, respectively. For sequences I and II, dose escalation to 75% of the MTD for each drug was paralleled by reversible host toxicity with no significant increase in the antitumor activity of the combination. With sequence III, however, the combination was lethal in 100% of treated animals when the doses of both drugs were at 75% of the MTD or higher. With the sequential combination of CPT-11 followed 24 h later by FUra (sequence III), the high complete tumor regression rate (cure) could be maintained, even when the dose of CPT-11 was reduced to 12.5% of the MTD as long as the doses of FUra was kept at 50 -75 % of the MTD. The data demonstrate that the antitumor activity and toxicity of combining CPT-11 with FUra is highly sequence dependent and that a sequence of CPT-11 preceding FUra is superior with a significant increase in the therapeutic index over the other sequences tested. In addition, the data also demonstrate that toxicity associated with high dose of CPT-11 can be eliminated without loss of the antitumor efficacy by reducing the dose of CPT-11 to at least 50% of its MTD, whereas the dose of FUra is kept at 50-75 % of its MTD.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Fluorouracil/administration & dosage , Animals , Camptothecin/administration & dosage , Colorectal Neoplasms/chemically induced , Dose-Response Relationship, Drug , Female , Irinotecan , Kinetics , Maximum Tolerated Dose , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Time Factors
13.
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
14.
Biochem Pharmacol ; 59(8): 953-60, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10692560

ABSTRACT

We have shown previously that (R)-5-fluoro-5,6-dihydrouracil (FUraH(2)) attenuates the antitumor activity of 5-fluorouracil (FUra) in rats bearing advanced colorectal carcinoma. Presently, we found that alpha-fluoro-beta-alanine (FBAL), the predominant catabolite of FUra that is formed rapidly via FUraH(2), also decreased the antitumor activity and potentiated the toxicity of FUra. In rats treated with Eniluracil (5-ethynyluracil, GW776), excess FBAL, in a 9:1 ratio to FUra, produced similar effects when administered 1 hr before, simultaneously with, or 2 hr after FUra. FBAL also decreased the antitumor activity of FUra in Eniluracil-treated mice bearing MOPC-315 myeloma at a 9:1 ratio with FUra, but not at a 2:1 ratio. FBAL did not affect the antitumor activity of FUra in mice bearing Colon 38 tumors. We also evaluated the effect of thymidylate synthase (TS) and thymidine kinase (TK) from tumor extracts after FUra +/- Eniluracil +/- FBAL treatment. The activity of TK was similar among the three groups at both 18 and 120 hr. There was also no difference in TS inhibition ( approximately 35%) at 18 hr. However, significantly more TS inhibition was observed in the Eniluracil/FUra group than in the FUra-alone group at 120 hr. FBAL did not alter the effect of Eniluracil/FUra in TS inhibition. Neither FUraH(2) nor FBAL affected the IC(50) of FUra in culture. Thus, the effect of FBAL did not result from direct competition with FUra uptake or immediate anabolism. Either another downstream catabolite that is not formed in cell culture is the active agent, or the effect requires the complexity of a living organism or an established tumor.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , beta-Alanine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/drug therapy , Drug Interactions , Female , Fluorouracil/pharmacology , Mice , Multiple Myeloma/drug therapy , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tumor Cells, Cultured , beta-Alanine/pharmacology
15.
Clin Cancer Res ; 6(2): 718-24, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10690558

ABSTRACT

Overexpression of the Bax protein in human head and neck squamous cell carcinoma A253 cells was reported to result in an increased sensitivity to various chemotherapeutic agents in vitro (Guo et al., Oncol. Res., 11: 91-99, 1999). In the present study, the relationship between Bax expression and response to chemotherapy was further investigated in vitro and in vivo model systems. For in vitro study, A253, A253/Vec (pcDNA3 vector transfectant), and A253/Bax (pcDNA3/Bax transfectant, expressing 50-fold higher Bax protein than A253 and A253/Vec) cells were exposed to various concentrations of raltitrexed (a specific thymidylate synthase inhibitor) and SN-38 (a topoisomerase I inhibitor) for 2 h, and cell growth inhibition was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide clonogenic assay. Compared to A253/Vec, A253/Bax cells exhibited 9.5- and 13.5-fold increases in sensitivity to raltitrexed and SN-38, respectively. For in vivo study, A253/Vec and A253/Bax tumor xenografts were established by s.c. injection of tumor cells into nude mice. The antitumor activity and toxicity of raltitrexed (i.v. push daily for 5 days) and irinotecan (a prodrug of SN-38; i.v. push daily for 3 days) were evaluated. The maximum tolerated doses of raltitrexed and irinotecan were 30 and 100 mg/kg/day, respectively. At the maximum tolerated doses, minimal antitumor activity was observed with raltitrexed, although irinotecan was more active than raltitrexed against A253 or A253/Vec tumors. In contrast, both raltitrexed and irinotecan were significantly more active against A253/Bax xenografts than against A253/Vec xenografts; the yield for complete tumor regression (cure) was 40% and 100% with raltitrexed and irinotecan, respectively, with no significant toxicity. Furthermore, the observed increase of antitumor activity in A253/Bax tumors was associated with an enhanced induction of apoptosis in vivo. The in vivo results demonstrated a proof of the principal concept that selecting up-regulation of the proapoptosis gene Bax can provide the basis for a greater therapeutic efficacy to a variety of chemotherapeutic agents with different structures and mechanisms of action.


Subject(s)
Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Animals , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/toxicity , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Camptothecin/therapeutic use , Camptothecin/toxicity , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/therapeutic use , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Irinotecan , Mice , Mice, Nude , Proto-Oncogene Proteins/biosynthesis , Quinazolines/toxicity , Thiophenes/toxicity , Thymidylate Synthase/antagonists & inhibitors , Topoisomerase I Inhibitors , Transplantation, Heterologous , Tumor Cells, Cultured , bcl-2-Associated X Protein
16.
Cancer Chemother Pharmacol ; 45(1): 78-84, 2000.
Article in English | MEDLINE | ID: mdl-10647506

ABSTRACT

UNLABELLED: TAS-103 is a recently developed dual inhibitor of topoisomerase-I (topo-I) and topoisomerase-II (topoII). TAS-103 has documented cytotoxicity in vitro and antitumor activity against a variety of mouse, rat, and human xenografts in vivo. PURPOSE: To determine TAS-103 activity against (multi)drug resistant cells in vitro and to delineate its mechanism of action. METHODS: TAS-103 was evaluated for activity against three human multidrug-resistant cell lines representing resistance mediated by P-glycoprotein (Pgp)-, multidrug resistance protein (MRP), and lung resistance protein (LRP) as well as one camptothecin-resistant cell line associated with a mutated topo-I enzyme. Drug sensitivity following short (2 h), intermediate (6-8 h) and long term (24 h) exposures were compared. The mechanism of action was studied by evaluating inhibition of topoisomerase-I and -II specific DNA relaxation assays, drug-induced DNA/protein cross-link formation, and competitive DNA intercalation with ethidium bromide. RESULTS: Increasing the exposure time only modestly potentiated TAS-103 cytotoxicity (3-5 fold) demonstrating a lack of strong exposure duration dependency. TAS-103 cytotoxicity was not affected by the presence of any of the drug resistance mechanisms studied. TAS-103 inhibits topo-I and -II activity in DNA relaxation assays, but in our assay system TAS-103 was found to have only a weak ability to induce DNA-protein crosslinks. DNA migration patterns in agarose gel electrophoresis indicate that TAS-103 can interact directly with DNA. Also its ability to displace ethidium bromide which has intercalated into the DNA provides an indication on the nature of drug-DNA interaction. CONCLUSIONS: TAS-103 cytotoxicity is not affected by the presence of Pgp, MRP, LRP or mutations in the CAM binding region of the topo-I enzyme and its growth-inhibitory effect appears to be weakly dependent on exposure duration. The presented evidence suggest that the inhibitory effects of TAS-103 on topo-I and -II may in part be related to its DNA binding rather than primarily through stabilization of topo-I or -II intermediates with DNA through specific binding to the enzymes.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indenes/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Camptothecin/pharmacology , DNA/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Tumor Cells, Cultured
17.
Br J Cancer ; 81(8): 1304-10, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10604726

ABSTRACT

Recent data suggest that expression of the membrane P170-glycoprotein (P-gp) may confer resistance to the topoisomerase-I-interactive agent topotecan. The present study describes the cellular effects of a new dihydropyridine analogue, PAK-200S, on P-gp-mediated resistance to topotecan in human breast and ovarian tumour cells. PAK-200S at a non-cytotoxic concentration of 2.0 microM completely reversed resistance to topotecan in P-gp-expressing MCF-7/adr (breast) and A2780/Dx5 (ovarian) tumour cells, respectively, with no effects on parental cells. Cellular pharmacokinetic studies by reversed-phase high-performance liquid chromatography analysis showed significantly lower cellular drug concentrations of the pharmacologically active closed-ring lactone of topotecan in multidrug-resistant cells than in parental cells. PAK-200S was effective in restoring the cellular lactone concentrations of topotecan in resistant MCF-7/adr cells to levels comparable to those obtained in parental cells. Furthermore, exposure of MCF-7/adr cells to topotecan in the presence of PAK-200S significantly increased the induction of protein-linked DNA breaks. PAK-200S did not alter nuclear topoisomerase I-mediated ex vivo pBR322 DNA plasmid unwinding activity and topoisomerase-I protein expression. These results suggest that reversal of P-gp-mediated resistance to topotecan by PAK-200S was related to the restoration of cellular drug concentrations of the active lactone form of topotecan rather than a direct effect on topoisomerase-I function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/pharmacology , Dihydropyridines/pharmacology , Neoplasms/pathology , Topotecan/pharmacology , Antineoplastic Agents/pharmacokinetics , Blotting, Western , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Neoplasms/enzymology , Neoplasms/genetics , Phenotype , Topotecan/pharmacokinetics , Tumor Cells, Cultured
18.
Semin Oncol ; 26(6): 612-20, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606254

ABSTRACT

For years, 5-fluorouracil (5-FU) was the only chemotherapeutic agent for the treatment of patients with advanced colorectal cancer. Based on laboratory data, modulation of 5-FU by leucovorin (LV) was proven to be an active alternative. In addition, a number of 5-FU prodrugs and antifolate antimetabolites became available for preclinical and clinical evaluation. With the 5-FU prodrugs, the overall aim was to improve the therapeutic efficacy and selectivity of 5-FU and to provide an oral form of therapy. In preclinical systems, several of the 5-FU prodrugs, eg, capecitabine, uracil/ ftorafur (UFT)/LV, and S- , are active and offer significant therapeutic advantages over 5-FU/LV. A direct and specific new thymidylate synthase (TS) inhibitor, Tomudex (raltitrexed, ZDI694; Zeneca Pharmaceuticals, Macclesfield, UK), is active in several preclinical and clinical settings. The major focus of this report will be on the preclinical development of selected fluoropyrimidine prodrugs.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Fluorouracil/therapeutic use , Folic Acid Antagonists/therapeutic use , Prodrugs/therapeutic use , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Animals , Antimetabolites, Antineoplastic/pharmacology , Capecitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Dihydrouracil Dehydrogenase (NADP) , Drug Combinations , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Fluorouracil/pharmacology , Folic Acid Antagonists/pharmacology , Humans , Oxidoreductases/antagonists & inhibitors , Prodrugs/pharmacology , Quinazolines/pharmacology , Tegafur/pharmacology , Tegafur/therapeutic use , Thiophenes/pharmacology , Thymidine Phosphorylase/antagonists & inhibitors , Thymidylate Synthase/antagonists & inhibitors , Uracil/analogs & derivatives , Uracil/pharmacology , Uracil/therapeutic use
19.
Eur J Cancer ; 35(6): 984-93, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10533483

ABSTRACT

We developed a combination protocol for inhibitors of thymidylate synthase (TS) and DNA topoisomerase I (Topo I) that can exert highly lethal effects in vitro against HCT-8 human colorectal cancer cells. The specific schedule was constructed so that a TS inhibitor could induce not only primary DNA damage but also cellular conditions optimal for the efficient action of a Topo I inhibitor. The initial drug treatment consisted of a brief exposure to a quinazoline-based antifolate, ZD1694. After an interval of approximately one cell-doubling time, cells were exposed for 8-24 h to BNP1100, a Karenitecin-class 7-thiomethyl-camptothecin, in the presence of 1-10 microM thymidine; the latter acted as a crucial factor to promote the collision of moving replication forks with the drug-stabilised DNA-Topo I cleavable complexes even under continuous TS inhibition. Clonogenic analyses confirmed that these mechanistically distinct drugs at clinically achievable concentrations worked in a highly synergistic manner, with a maximum effect abolishing the viability of virtually all cancer cells (> 99.9%). The pretreatment with ZD1694 increased the amount of DNA-bound Topo I by up to 4-fold and the DNA-damaging capability of BNP1100 by up to 15-fold. The possibility of at least four DNA-damaging pathways is proposed which might have resulted from the individual actions of TS and Topo I inhibitors as well as their concerted actions. Taken together, the present findings provided a logically permissible explanation as to why TS and Topo I inhibitors in concerted interactions induced a highly lethal effect which was more than a simple additive effect. Since these drugs are effective specifically on actively proliferating cancer cells, but not on non-cycling G0/G1 cells, this mechanism-based protocol may warrant consideration for clinical verification.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Thymidylate Synthase/antagonists & inhibitors , Topoisomerase I Inhibitors , Camptothecin/therapeutic use , Cell Division , Cell Survival , Colonic Neoplasms/pathology , DNA Damage , Drug Synergism , Humans , Tumor Cells, Cultured
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...