417 results on '"Rustum, Y."'
Search Results
102. Current Landscape and the Potential Role of Hypoxia-Inducible Factors and Selenium in Clear Cell Renal Cell Carcinoma Treatment.
- Author
-
Garje R, An JJ, Sanchez K, Greco A, Stolwijk J, Devor E, Rustum Y, and Zakharia Y
- Subjects
- Angiogenesis Inhibitors pharmacology, Carcinogenesis metabolism, Carcinogenesis pathology, Carcinoma, Renal Cell pathology, Humans, Kidney Neoplasms pathology, Carcinoma, Renal Cell metabolism, Hypoxia-Inducible Factor 1 metabolism, Kidney Neoplasms metabolism, Selenium metabolism
- Abstract
In the last two decades, the discovery of various pathways involved in renal cell carcinoma (RCC) has led to the development of biologically-driven targeted therapies. Hypoxia-inducible factors (HIFs), angiogenic growth factors, von Hippel⁻Lindau ( VHL ) gene mutations, and oncogenic microRNAs (miRNAs) play essential roles in the pathogenesis and drug resistance of clear cell renal cell carcinoma. These insights have led to the development of vascular endothelial growth factor (VEGF) inhibitors, Mechanistic target of rapamycin (mTOR) inhibitors, and immunotherapeutic agents, which have significantly improved the outcomes of patients with advanced RCC. HIF inhibitors will be a valuable asset in the growing therapeutic armamentarium of RCC. Various histone deacetylase (HDAC) inhibitors, selenium, and agents like PT2385 and PT2977 are being explored in various clinical trials as potential HIF inhibitors, to ameliorate the outcomes of RCC patients. In this article, we will review the current treatment options and highlight the potential role of selenium in the modulation of drug resistance biomarkers expressed in clear cell RCC (ccRCC) tumors.
- Published
- 2018
- Full Text
- View/download PDF
103. Effects of selenomethionine on acute toxicities from concurrent chemoradiation for inoperable stage III non-small cell lung cancer.
- Author
-
Mix M, Ramnath N, Gomez J, de Groot C, Rajan S, Dibaj S, Tan W, Rustum Y, Jameson MB, and Singh AK
- Abstract
Aim: To prospectively determine the safety and tolerability of oral L-selenomethionine (SLM) with concurrent chemoradiation (CCRT) for Stage III non-small cell lung cancer (NSCLC) and estimate if the incidence and/or severity of adverse events could be reduced by its use., Methods: Sixteen patients with stage III NSCLC were accrued to this single arm, phase II study. CCRT consisted of radiation given at 2 Gy per fraction for 30-33 fractions, 5 d per week with concurrent weekly IV paclitaxel 50 mg/m(2) followed by carboplatin dosed at an area under the time-concentration curve of 2. SLM was dosed in a loading phase at 4800 μg twice daily for one week prior to CCRT followed by once daily dosing during treatment., Results: No selenium-related toxicity was observed. Analysis revealed grade 3 or higher esophagitis in 3 of 16 patients (19%), pneumonitis in 0, leukopenia in 2 (12.5%), and anemia in 1 (6%); the latter two were significantly reduced when compared to the protocol-stated expected rate of 35% (P = 0.045 for leukopenia, and P < 0.01 for anemia). Median overall survival was 14.9 mo and median failure-free survival was 9 mo (95%CI: 3.3-21.5)., Conclusion: There may be some protective benefit of selenium in the setting of CCRT for inoperable NSCLC. The data suggests decreased rates of myelosuppression when compared to similarly-treated historical and contemporary controls. Further evaluation of selenium in this setting may be warranted.
- Published
- 2015
- Full Text
- View/download PDF
104. Randomized phase II trial of selenomethionine as a modulator of efficacy and toxicity of chemoradiation in squamous cell carcinoma of the head and neck.
- Author
-
Mix M, Singh AK, Tills M, Dibaj S, Groman A, Jaggernauth W, Rustum Y, and Jameson MB
- Abstract
Aim: To investigate whether selenomethionine (SLM) reduces mucositis incidence in patients with head and neck squamous cell cancer (HNSCC) undergoing concurrent chemoradiation (CRT)., Methods: In this multi-institutional, randomized, double-blind phase II trial, patients with Stage III or IV HNSCC received SLM 3600 μg/m(2) or placebo twice daily for 7 d prior to CRT, once daily during CRT, and daily for 3 wk following CRT. CRT consisted of 70 Gy at 2 Gy per fraction with cisplatin 100 mg/m(2) IV on days 1, 22, and 43., Results: Eighteen patients were randomized, 10 received SLM, and there were no differences in baseline factors. There was no difference in mucositis or patient-reported side effects between groups. There was no difference in overall or relapse-free survival at 12 mo., Conclusion: Addition of SLM to CRT for HNSCC was well-tolerated but did not lower the incidence of severe mucositis or improve quality of life or survival outcomes.
- Published
- 2015
- Full Text
- View/download PDF
105. Investigation of hydrophobically derivatized hyperbranched polyglycerol with PEGylated shell as a nanocarrier for systemic delivery of chemotherapeutics.
- Author
-
Misri R, Wong NK, Shenoi RA, Lum CM, Chafeeva I, Toth K, Rustum Y, Kizhakkedathu JN, and Khan MK
- Subjects
- Animals, Docetaxel, Drug Carriers administration & dosage, Drug Carriers chemistry, Female, Glycerol administration & dosage, Glycerol chemistry, HT29 Cells, Humans, Hydrophobic and Hydrophilic Interactions, Mice, Nanoparticles chemistry, Neoplasms pathology, Polymers administration & dosage, Polymers chemistry, Taxoids chemistry, Tissue Distribution drug effects, Xenograft Model Antitumor Assays, Drug Delivery Systems, Nanoparticles administration & dosage, Neoplasms drug therapy, Taxoids administration & dosage
- Abstract
We report the synthesis and characterization of a polymeric nanoparticle (NP) based on hyperbranched polyglycerol (HPG) containing a hydrophobic core and a hydrophilic shell, and assessed its suitability to be developed as a systemic anticancer drug carrier. HPG NP displayed low toxicity to primary cell cultures and were well-tolerated in mice after intravenous administration. When tested in mice tumor xenograft models, HPG NP accumulated significantly in the tumors with low accumulation in the liver and the spleen. In vitro studies demonstrated that HPG NP was capable of hydrophobically binding docetaxel and releasing it in a controlled manner. The HPG NP formulation of docetaxel conferred a preferential protective effect on primary non-cancerous cells while effectively killing cancer cells, indicating great potential for widening its therapeutic index. Taken together, these data indicate that HPG NP is a highly promising nanocarrier platform for systemic delivery of anticancer drugs., From the Clinical Editor: The use of polyethylene glycol on nano-carriers as "stealth" to deliver intravenous drugs is well known. Here, the authors developed polymeric nanoparticle (NP) with hyperbranched polyglycerol (HPG) and tested its efficacy in delivering docetaxel. The results showed that this formulation could preferentially killed cancer cells with a high therapeutic index. It seems that this platform could have a great potential in cancer therapy., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
106. The Chk1-Cdc25C regulation is involved in sensitizing A253 cells to a novel topoisomerase I inhibitor BNP1350 by bax gene transfer.
- Author
-
Yin M, Hapke G, Guo B, Azrak RG, Frank C, and Rustum YM
- Subjects
- 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, Cell Cycle Proteins metabolism, Gene Expression Regulation, Enzymologic, Gene Transfer Techniques, Lung Neoplasms enzymology, Protein Kinases metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2, Topoisomerase I Inhibitors, cdc25 Phosphatases metabolism
- 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.
- Published
- 2001
- Full Text
- View/download PDF
107. Chemotherapeutic strategies for treatment of colorectal cancer: present and future developments.
- Author
-
Backus HH, Rustum YM, van Groeningen CJ, and Peters GJ
- Subjects
- Biomarkers, Tumor, Chemotherapy, Adjuvant, Colorectal Neoplasms metabolism, Humans, Prognosis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy
- 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.
- Published
- 2001
- Full Text
- View/download PDF
108. Irinotecan in the treatment of colorectal cancer: clinical overview.
- Author
-
Vanhoefer U, Harstrick A, Achterrath W, Cao S, Seeber S, and Rustum YM
- Subjects
- 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, Antineoplastic Agents, Phytogenic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Camptothecin analogs & derivatives, Camptothecin therapeutic use, Colorectal Neoplasms drug therapy
- 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.
- Published
- 2001
- Full Text
- View/download PDF
109. Targeting molecular signals in chk1 pathways as a new approach for overcoming drug resistance.
- Author
-
Hapke G, Yin MB, and Rustum YM
- Subjects
- Checkpoint Kinase 1, Humans, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Enzyme Inhibitors pharmacology, Neoplasms enzymology, Protein Kinase Inhibitors, Protein Kinases
- 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.
- Published
- 2001
- Full Text
- View/download PDF
110. Time-lapse video reveals immediate heterogeneity and heritable damage among human ileocecal carcinoma HCT-8 cells treated with raltitrexed (ZD1694).
- Author
-
Slocum HK, Parsons JC, Winslow EO, Broderick L, Minderman H, Tóth K, Greco WR, and Rustum YM
- Subjects
- Aged, Cell Division drug effects, Cell Lineage drug effects, Clone Cells cytology, Clone Cells drug effects, Humans, Male, Probability, Tumor Cells, Cultured, Antimetabolites, Antineoplastic toxicity, Ileal Neoplasms drug therapy, Ileal Neoplasms pathology, Microscopy, Video methods, Quinazolines toxicity, Thiophenes toxicity
- 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., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
111. Preclinical development of eniluracil: enhancing the therapeutic index and dosing convenience of 5-fluorouracil.
- Author
-
Paff MT, Baccanari DP, Davis ST, Cao S, Tansik RL, Rustum YM, and Spector T
- Subjects
- Administration, Oral, Animals, Antimetabolites, Antineoplastic pharmacokinetics, Dihydrouracil Dehydrogenase (NADP), Drug Synergism, Fluorouracil pharmacokinetics, Humans, Oxidoreductases antagonists & inhibitors, Antimetabolites, Antineoplastic administration & dosage, Enzyme Inhibitors pharmacology, Fluorouracil administration & dosage, Uracil analogs & derivatives, Uracil pharmacology
- 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.
- Published
- 2000
- Full Text
- View/download PDF
112. Synergistic antitumor activity of irinotecan in combination with 5-fluorouracil in rats bearing advanced colorectal cancer: role of drug sequence and dose.
- Author
-
Cao S and Rustum YM
- Subjects
- 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, 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
- 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.
- Published
- 2000
113. Characterization of protein kinase chk1 essential for the cell cycle checkpoint after exposure of human head and neck carcinoma A253 cells to a novel topoisomerase I inhibitor BNP1350.
- Author
-
Yin MB, Guo B, Vanhoefer U, Azrak RG, Minderman H, Frank C, Wrzosek C, Slocum HK, and Rustum YM
- Subjects
- 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, Camptothecin analogs & derivatives, Cell Cycle drug effects, Protein Kinases metabolism, Topoisomerase I Inhibitors
- 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.
- Published
- 2000
- Full Text
- View/download PDF
114. Overexpression of Bax enhances antitumor activity of chemotherapeutic agents in human head and neck squamous cell carcinoma.
- Author
-
Guo B, Cao S, Tóth K, Azrak RG, and Rustum YM
- Subjects
- 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, Camptothecin analogs & derivatives, Carcinoma, Squamous Cell drug therapy, Head and Neck Neoplasms drug therapy, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2, Quinazolines therapeutic use, Thiophenes therapeutic use
- 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.
- Published
- 2000
115. Mechanism of action of the dual topoisomerase-I and -II inhibitor TAS-103 and activity against (multi)drug resistant cells.
- Author
-
Minderman H, Wrzosek C, Cao S, Utsugi T, Kobunai T, Yamada Y, and Rustum YM
- Subjects
- Camptothecin pharmacology, DNA metabolism, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Humans, Tumor Cells, Cultured, Aminoquinolines pharmacology, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Indenes pharmacology, Topoisomerase I Inhibitors, Topoisomerase II Inhibitors
- 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.
- Published
- 2000
- Full Text
- View/download PDF
116. Oral uracil and Ftorafur plus leucovorin: pharmacokinetics and toxicity in patients with metastatic cancer.
- Author
-
Ho DH, Covington W, Brown N, Lin SN, Pazdur R, Huo YY, Creaven PJ, Rustum YM, Meropol NJ, Lassere Y, Kuritani J, and Hayakawa T
- Subjects
- Tegafur blood, Uracil blood, Tegafur adverse effects, Tegafur pharmacokinetics, Uracil adverse effects, Uracil pharmacokinetics
- 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.
- Published
- 2000
- Full Text
- View/download PDF
117. New drugs in therapy of colorectal cancer: preclinical studies.
- Author
-
Rustum YM and Cao S
- Subjects
- 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, 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
- 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.
- Published
- 1999
118. Involvement of cyclin D1-cdk5 overexpression and MCM3 cleavage in bax-associated spontaneous apoptosis and differentiation in an A253 human head and neck carcinoma xenograft model.
- Author
-
Yin MB, Tóth K, Cao S, Guo B, Frank C, Slocum HK, and Rustum YM
- Subjects
- 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, Apoptosis, Cell Cycle Proteins metabolism, Cyclin D1 physiology, Cyclin-Dependent Kinases physiology, Head and Neck Neoplasms pathology, Proto-Oncogene Proteins physiology, Proto-Oncogene Proteins c-bcl-2
- 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., (Copyright 1999 Wiley-Liss, Inc.)
- Published
- 1999
- Full Text
- View/download PDF
119. Antitumor activity of ZD1694 (tomudex) against human head and neck cancer in nude mouse models: role of dosing schedule and plasma thymidine.
- Author
-
Cao S, McGuire JJ, and Rustum YM
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents toxicity, Disease Models, Animal, Drug Administration Schedule, Female, Floxuridine therapeutic use, Floxuridine toxicity, Fluorouracil therapeutic use, Fluorouracil toxicity, Head and Neck Neoplasms blood, Humans, Mice, Mice, Nude, Neoplasm Transplantation, Quinazolines administration & dosage, Quinazolines toxicity, Thiophenes administration & dosage, Thiophenes toxicity, Thymidine pharmacology, Thymidine physiology, Transplantation, Heterologous, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, Head and Neck Neoplasms drug therapy, Quinazolines therapeutic use, Thiophenes therapeutic use, Thymidine blood
- Abstract
We studied the antitumor activity and toxicity of ZD1694 (tomudex), a specific inhibitor of thymidylate synthase (TS), in nude mice bearing human head and neck squamous cell carcinoma A253 and FaDu xenografts. Mice were treated by single i.v. push (i.v. x 1), i.v. push once a week for 3 weeks (weekly x 3), and i.v. push once a day for 5 days (daily x 5), and the maximum tolerated doses (MTDs) of ZD1694 were 300 mg/kg, 60 mg/kg/week, and 30 mg/kg/day, respectively. ZD1694 was moderately active against both A253 and FaDu xenografts. Antitumor activity was schedule-dependent in both tumors: weekly x 3 > or = i.v. x 1 >> daily x 5. In contrast, the rank order of toxicity was daily x 5 >> weekly x 3 > or = i.v. x 1. ZD1694 at the MTD produced 20% complete tumor regression and 20% partial tumor regression (PR) with i.v. x 1 and weekly x 3 schedules and 12-day tumor growth delay with daily x 5 schedule against FaDu xenografts. No complete tumor regression was achieved with ZD1694 with any schedule against A253; a 20% PR, 40% PR, and 10-day tumor growth delay were observed with i.v. x 1, weekly x 3, and daily x 5 schedules, respectively. The data indicate that ZD1694 was slightly more effective against FaDu than against A253. Of interest and potential clinical importance was the observation that ZD1694 was still active at doses lower than the MTD (> or =1/3 MTD), which showed a high therapeutic index and wide safety margin. Study of ZD1694 compared with 5-fluorouracil and 5-fluoro-2'-deoxyuridine at the MTD revealed that the antitumor activity of ZD1694 was comparable with or superior to 5-fluorouracil and 5-fluoro-2'-deoxyuridine against both A253 and FaDu xenografts, with less toxicity. High plasma thymidine in mouse relative to human (approximately 1.3 microM and <0.1 microM, respectively) may complicate the study of antitumor activity and toxicity of TS inhibitors with human tumor xenografts grown in the mouse. To test this hypothesis, we preadministered methoxypolyethyleneglycol-conjugated thymidine phosphorylase (MPEG-TPase; 2500 units/kg/dose) to reduce mouse plasma thymidine, then treated with various doses of ZD1694 using the daily x 5 or i.v. x 1 schedules in the A253 tumor model. MPEG-TPase significantly increased the toxicity of ZD1694; the MTD of ZD1694 plus MPEG-TPase was reduced 3- and 10-fold compared with ZD1694 alone for i.v x 1 and daily x 5 schedules, respectively. However, preadministration of MPEG-TPase did not potentiate the antitumor activity of ZD1694 with either schedule. The data indicate that the study of TS inhibitors in rodent models may not be suitable for predicting a safe dose for clinical study. However, rodent models, particularly human tumor xenografts, are still useful models for evaluation of antitumor activity and schedule selection for TS inhibitors.
- Published
- 1999
120. Clinical implications of 5-FU modulation.
- Author
-
Rustum YM
- Subjects
- Antimetabolites, Antineoplastic pharmacokinetics, Breast Neoplasms genetics, Breast Neoplasms metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, DNA, Neoplasm biosynthesis, DNA, Neoplasm drug effects, Drug Combinations, Drug Synergism, Female, Fluorouracil pharmacokinetics, Humans, Thymidylate Synthase metabolism, Antimetabolites, Antineoplastic therapeutic use, Breast Neoplasms drug therapy, Colorectal Neoplasms drug therapy, Fluorouracil therapeutic use, Leucovorin therapeutic use
- Abstract
In recent years, due to the advent of sensitive instrumentation and methodologies, it has been possible to identify parameters that predict the quality of response of individual patients to treatments for specific selected diseases, e.g., colon carcinoma and breast carcinoma. Ongoing studies are attempting to identify sensitive patients in order to select treatment regimens suitable for the individual patient. The critical question that remains is whether the basis for drug resistance is due in part to insufficient delivery of drugs to target tumor cells or to the resistance of target tumor cells by various mechanisms, including, in the case of 5-fluorouracil (5-FU), drug transport, metabolism, expression of the target enzyme thymidylate synthase (dTMPS), depletion of folate cofactors, and/or level of competing substrate deoxyuridine monophosphate. Also in recent years, attempts have been made to delineate mechanisms of resistance to the fluoropyrimidines. On the basis of such studies, it may be possible to develop approaches aimed at the selective modulation of the therapeutic efficacy of these agents in tumor tissues with varying degrees of sensitivity of fluoropyrimidines, e.g., patients with advanced colorectal cancer. One such approach is the use of calcium folinate to modulate the therapeutic efficacy of 5-FU.
- Published
- 1999
121. Roswell Park Cancer Institute.
- Author
-
Rustum Y
- Subjects
- History, 20th Century, Hospitals, Teaching history, Medical Oncology education, Molecular Biology history, Neoplasms genetics, Neoplasms immunology, New York, Academies and Institutes history, Medical Oncology history, Neoplasms history
- Published
- 1999
122. Characterisation of a synergistic interaction between a thymidylate synthase inhibitor, ZD1694, and a novel lipophilic topoisomerase I inhibitor karenitecin, BNP1100: mechanisms and clinical implications.
- Author
-
Matsui S, Endo W, Wrzosek C, Haridas K, Seetharamulu P, Hausheer FH, and Rustum YM
- Subjects
- Camptothecin therapeutic use, Cell Division, Cell Survival, Colonic Neoplasms pathology, DNA Damage, Drug Synergism, Humans, Tumor Cells, Cultured, 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
- 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.
- Published
- 1999
- Full Text
- View/download PDF
123. Phase I study of a weekly schedule of irinotecan, high-dose leucovorin, and infusional fluorouracil as first-line chemotherapy in patients with advanced colorectal cancer.
- Author
-
Vanhoefer U, Harstrick A, Köhne CH, Achterrath W, Rustum YM, Seeber S, and Wilke H
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Colorectal Neoplasms pathology, Diarrhea chemically induced, Drug Administration Schedule, Female, Fluorouracil administration & dosage, Humans, Irinotecan, Leucovorin administration & dosage, Male, Middle Aged, Neoplasm Metastasis, Remission Induction, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy
- Abstract
Purpose: To determine the maximum-tolerated dose (MTD) of a weekly schedule of irinotecan (CPT-11), leucovorin (LV), and a 24-hour infusion of fluorouracil (5-FU24h) as first-line chemotherapy in advanced colorectal cancer and to assess preliminary data on the antitumor activity., Patients and Methods: Twenty-six patients with measurable metastatic colorectal cancer were entered onto this phase I study. In the first six dose levels, fixed doses of CPT-11 (80 mg/m2) and LV (500 mg/m2) in combination with escalated doses of 5-FU24h ranging from 1.8 to 2.6 g/m2 were administered on a weekly-times-four (dose levels 1 to 4) or weekly-times-six (dose levels 5 to 6) schedule. The dose of CPT-11 was then increased to 100 mg/m2 (dose level 7)., Results: Seventy-nine cycles of 5-FU24h/LV with CPT-11 were administered in an outpatient setting. No dose-limiting toxicities were observed during the first cycle at dose levels 1 to 6, but diarrhea of grade 4 (National Cancer Institute common toxicity criteria) was observed in three patients after multiple treatment cycles. Other nonhematologic and hematologic side effects, specifically alopecia and neutropenia, did not exceed grade 2. With the escalation of CPT-11 to 100 mg/m2 (dose level 7), diarrhea of grade 3 or higher was observed in four of six patients during the first cycle; thus, the MTD was achieved. Sixteen of 25 response-assessable patients (64%; 95% confidence interval, 45% to 83%) achieved an objective response., Conclusion: The recommended doses for further studies are CPT-11 80 mg/m2, LV 500 mg/m2, and 5-FU24h 2.6 g/m2 given on a weekly-times-six schedule followed by a 1-week rest period. The addition of CPT-11 to 5-FU24h/LV seems to improve the therapeutic efficacy in terms of tumor response with manageable toxicity.
- Published
- 1999
- Full Text
- View/download PDF
124. Cyclin E-cdk2 activation is associated with cell cycle arrest and inhibition of DNA replication induced by the thymidylate synthase inhibitor Tomudex.
- Author
-
Yin MB, Guo B, Panadero A, Frank C, Wrzosek C, Slocum HK, and Rustum YM
- Subjects
- 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, CDC2-CDC28 Kinases, Carrier Proteins, Cell Cycle drug effects, Cell Cycle Proteins, Cyclin E metabolism, Cyclin-Dependent Kinases metabolism, DNA antagonists & inhibitors, DNA biosynthesis, DNA-Binding Proteins, Protein Serine-Threonine Kinases metabolism, Quinazolines pharmacology, Thiophenes pharmacology, Thymidylate Synthase antagonists & inhibitors
- 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., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
125. Persistent induction of apoptosis and suppression of mitosis as the basis for curative therapy with S-1, an oral 5-fluorouracil prodrug in a colorectal tumor model.
- Author
-
Cao S, Lu K, Tóth K, Slocum HK, Shirasaka T, and Rustum YM
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents toxicity, Colorectal Neoplasms enzymology, Disease Models, Animal, Drug Combinations, Female, Fluorouracil therapeutic use, Neoplasm Transplantation, Oxonic Acid pharmacokinetics, Prodrugs pharmacokinetics, Pyridines pharmacokinetics, Rats, Rats, Inbred F344, Tegafur pharmacokinetics, Thymidylate Synthase antagonists & inhibitors, Thymidylate Synthase metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Apoptosis, Colorectal Neoplasms drug therapy, Mitosis drug effects, Oxonic Acid therapeutic use, Prodrugs therapeutic use, Pyridines therapeutic use, Tegafur therapeutic use
- Abstract
In an effort to improve the therapeutic selectivity of 5-fluorouracil (FUra) against colorectal cancer, S-1, a combination agent including a prodrug of FUra with two modulators, was recently developed by Taiho Pharmaceuticals Co. S-1 is a combination of tegafur (FT), 5-chloro-2,4-hydroxypyridine, and potassium oxonate in the molar ratio of 1.0:0.4:1.0, with the latter two components as inhibitors of dihydropyrimidine dehydrogenase and phosphoribosylpyrophosphate transferase, respectively. In this study, the therapeutic selectivity and efficacy of S-1 (oral) was compared with FT (oral) and FUra (i.v. infusion) in rats bearing advanced colorectal cancer by using clinically relevant schedules. The maximum tolerated doses (MTDs) of S-1, FT, and FUra were 31.5, 200, and 25 mg/kg/d for 7 days and 22.5, 150, and 12.5 mg/kg/d for 28 days, respectively. The therapeutic index of S-1 was 4- to 5-fold higher than that of either FT or FUra. S-1 achieved 100% complete tumor regression (CR) at its MTD in both 7-day and 28-day schedules. Furthermore, the high incidences of stomatitis, alopecia, and diarrhea observed with FUra and FT, were not observed with S-1. In an attempt to understand the basis for the observed superior therapeutic selectivity with S-1, we studied pharmacokinetic analysis of FUra, drug-induced apoptosis, suppression of mitosis, and inhibition of thymidylate synthase (TS) after S-1, FUra, or FT administration. The peak plasma FUra concentrations derived from FUra or S-1 (FT) at comparable MTDs were similar, but the plasma level of FUra was higher with S-1 than with FUra. Induction of high and sustained apoptosis was achieved with S-1. Although the initial level of apoptosis induced by FUra was comparable to S-1, it was not sustained. The sustained level of apoptosis appears to correlate with tumor growth inhibition. Mitotic figures were more greatly suppressed with S-1 treatment than with FUra. Studies on TS inhibition indicated that, although both S-1 and FUra caused a 4- to 6-fold induction of total TS protein, single oral administration of S-1 was superior to 24-h infusion of FUra in suppressing free TS. The data are consistent with the observation that the therapeutic efficacy of S-1 (100% cure) over FUra is associated with high and sustained levels of drug-induced apoptosis, greater suppression of mitosis, and inhibition of free TS in tumor tissues.
- Published
- 1999
126. Phase I and pharmacokinetic study of weekly 5-fluorouracil administered with granulocyte-macrophage colony-stimulating factor and high-dose leucovorin: a potential role for growth factor as mucosal protectant.
- Author
-
Meropol NJ, Rustum YM, Creaven PJ, Blumenson LE, and Frank C
- Subjects
- Adult, Aged, Antidotes administration & dosage, Antidotes pharmacokinetics, Antimetabolites, Antineoplastic administration & dosage, Antimetabolites, Antineoplastic adverse effects, Antimetabolites, Antineoplastic pharmacokinetics, Bone Marrow Diseases chemically induced, Bone Marrow Diseases prevention & control, Cohort Studies, Diarrhea chemically induced, Drug Administration Schedule, Female, Fluorouracil administration & dosage, Fluorouracil adverse effects, Fluorouracil pharmacokinetics, Granulocyte-Macrophage Colony-Stimulating Factor administration & dosage, Granulocyte-Macrophage Colony-Stimulating Factor pharmacokinetics, Humans, Infusions, Intravenous, Injections, Intravenous, Injections, Subcutaneous, Intestinal Mucosa drug effects, Intestinal Mucosa pathology, Leucovorin administration & dosage, Leucovorin pharmacokinetics, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms drug therapy, Peripheral Nervous System Diseases chemically induced, Severity of Illness Index, Stomatitis chemically induced, Treatment Outcome, Venous Thrombosis chemically induced, Antidotes therapeutic use, Antimetabolites, Antineoplastic therapeutic use, Diarrhea prevention & control, Fluorouracil therapeutic use, Granulocyte-Macrophage Colony-Stimulating Factor therapeutic use, Leucovorin therapeutic use
- Abstract
Diarrhea is dose-limiting with weekly 5-fluorouracil (5-FU) plus high-dose leucovorin (LV). Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been associated with a decrease in chemotherapy-associated mucosal toxicity. This study was conducted to determine the maximum tolerated dose (MTD) of weekly 5-FU when administered with GM-CSF and high-dose LV. Patients were treated with intravenous LV 500 mg/m2 plus 5-FU weekly for six doses followed by a 2-week rest. GM-CSF 250 mg/m2 was administered subcutaneously 5 days each week. Cohorts were treated with 5-FU at 600, 700, and 800 mg/m2 weekly. Twenty-nine patients were treated. The MTD of 5-FU in this schedule was 700 mg/m2/week, with diarrhea dose-limiting. 5-FU delivered dose intensity at the MTD was 424 +/- 23.7 mg/m2/week, including rest periods. 5-FU and LV pharmacokinetics were not altered by concurrent treatment with GM-CSF. In a weekly schedule with high-dose LV and GM-CSF, the MTD of 5-FU and 5-FU delivered dose intensity were higher than previously reported with 5-FU and LV administered without GM-CSF.
- Published
- 1999
127. Dimerization of mitochondrial Bax is associated with increased drug response in Bax-transfected A253 cells.
- Author
-
Guo B, Yin MB, Tóth K, Cao S, Azrak RG, and Rustum YM
- Subjects
- 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, Apoptosis drug effects, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms metabolism, Neoplasm Proteins metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2
- 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.
- Published
- 1999
128. Image analysis for quantitation of solid tumor drug sensitivity.
- Author
-
Gibbs JF, Slocum HK, Cao S, and Rustum YM
- Subjects
- Adenocarcinoma metabolism, Animals, Antimetabolites, Antineoplastic pharmacology, Colonic Neoplasms metabolism, Dose-Response Relationship, Drug, Fluorouracil pharmacology, Microscopy, Fluorescence, Osmolar Concentration, Pyrimidines pharmacokinetics, Rats, Single-Blind Method, Drug Resistance, Neoplasm, Image Processing, Computer-Assisted
- Abstract
Background: A method of assessing chemosensitivity of tissue has been described by Rotman et al. The aim of this study was to use image analysis to provide a more rapid and quantitative means of assessing drug effect on tissue proliferative capacity., Method: Fluoropyrimidine sensitive Ward rat colon adenocarcinoma tumor was implanted onto collagen impregnated cellulose fibers suspended on metal grids at an air-fluid interface and kept in a 95% air/5% CO2 incubator at 37 degrees C. The fluorescent microscopic image captured by a silicon intensified target (low light detecting) camera and linked to an image processing unit was measured for fluorescent brightness and tumor image area. Blinded 5-Fluorouracil (5-FU) drug treatment was begun 8 days after tumor explantation on the collagen-cellulose matrix. Tumor image area and fluorescent brightness were measured at 24 h pretreatment, 48 h posttreatment, and at 48 h post drug removal., Results: Nontreated tumor cultures demonstrated an increase in area and fluorescent brightness with time following tumor implantation on the collagen gel. Dose responsiveness was seen with increasing concentrations of 5-FU. At the highest clinically achievable concentration of 5-FU (500 microM), there was a 39% decrease in area compared with the nontreated group, 113%. Linear dose responsiveness was not demonstrated between 50 and 150microM 5-FU., Conclusions: Fluoropyrimidine activity was demonstrated with the implemented image analysis system. The in vitro tumor sensitivity to FU using collagen gel was consistent with responsiveness of tumors in vivo borne by rats.
- Published
- 1999
129. Modeling of the time-dependency of in vitro drug cytotoxicity and resistance.
- Author
-
Levasseur LM, Slocum HK, Rustum YM, and Greco WR
- Subjects
- Antineoplastic Agents administration & dosage, Cell Division drug effects, Dose-Response Relationship, Drug, Statistics as Topic, Time Factors, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Models, Biological
- Abstract
For potential clinical extrapolation of in vitro findings, it is of interest to relate the measured effect of an anticancer agent to concentration and exposure time. The Hill model (A. V. Hill, J. Physiol., 40: iv-vii, 1910) is commonly used to describe pharmacodynamic (PD) effects, including drug-induced growth inhibition of cancer cells in vitro. The IC(X)n x T = k relationship, in which IC(X) is the concentration of agent required to reduce cell growth by X%, T is the exposure time, and n and k are estimable parameters, was first applied to bacterial disinfectant action and then was successfully used to model anticancer drug potency as a function of exposure time (D. J. Adams, Cancer Res., 49: 6615-6620, 1989). Our goal was to create a new global PD modeling paradigm to facilitate the quantitative assessment of the growth-inhibitory effect of anticancer agents as a function of concentration and exposure time. Wild-type human ovarian A2780 and ileocecal HCT-8 carcinoma cells and sublines that were resistant to cisplatin (A2780/CP3), doxorubicin (A2780/DX5B), and raltitrexed (RTX) (HCT-8/DW2) were exposed to various anticancer agents, cisplatin, doxorubicin, paclitaxel, trimetrexate, RTX, methotrexate, and AG2034, for periods ranging from 1 to 96 h. Cell growth inhibition was measured with the sulforhodamine B protein dye assay. Patterns of time-dependency of drug potency, slope of the concentration-effect curves, and relative degree of resistance were characterized. Empirical mathematical expressions were built into a global concentration-time-effect model. The global PD model was then fit to the concentration-time-effect data with iteratively reweighted nonlinear regression. Under specific treatment conditions, the examination of the slope and the shape of the concentration-effect curves revealed a large heterogeneity in drug response, e.g., shallow concentration-effect curve or double or triple Hill "roller coaster" concentration-effect curve. These patterns, which were observed at intermediate exposure times in parental and resistant cells for paclitaxel and trimetrexate or only in resistant HCT-8/DW2 cells for RTX, methotrexate, and AG2034, revealed mechanistic insights for the former cases but possible methodological artifacts for the latter cases. The comprehensive PD modeling of the cytotoxic effect of anticancer agents showed that it was possible to modulate drug effect, response heterogeneity, and drug resistance by altering the time of exposure to the agents. This approach will be useful for: (a) describing complex concentration-time-effect surfaces; (b) refining biological interpretations of data; (c) providing insights on mechanisms of drug action and resistance; and (d) generating leads for clinical use of anticancer drugs.
- Published
- 1998
130. Modulation of platinum-induced toxicities and therapeutic index: mechanistic insights and first- and second-generation protecting agents.
- Author
-
Hausheer FH, Kanter P, Cao S, Haridas K, Seetharamulu P, Reddy D, Petluru P, Zhao M, Murali D, Saxe JD, Yao S, Martinez N, Zukowski A, and Rustum YM
- Subjects
- Amifostine therapeutic use, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Cisplatin adverse effects, Cisplatin chemistry, Cisplatin pharmacology, Drug Interactions, Humans, Kidney Diseases chemically induced, Kidney Diseases prevention & control, Mesna pharmacology, Mesna therapeutic use, Platinum Compounds chemistry, Platinum Compounds pharmacology, Protective Agents pharmacology, Sulfhydryl Compounds chemistry, Amifostine pharmacology, Antineoplastic Agents adverse effects, Mesna analogs & derivatives, Platinum Compounds adverse effects, Sulfhydryl Compounds pharmacology
- Abstract
Platinum-type drugs have proven to be valuable in the treatment of a variety of solid tumors, beginning with the commercial approval of cisplatin 18 years ago. There are several clinically important toxicities commonly associated with the administration of these drugs. Despite the extensive use of cisplatin and carboplatin, the fundamental chemical transformations and mechanisms that underlie their antitumor and toxic effects have not been fully characterized. Several first-generation protective thiols have been clinically studied in an attempt to reduce the toxicity of platinum-type drugs; while some of these agents appear to protect against certain toxicities, nearly all platinum-protecting drugs have their own intrinsic toxicities, which can be additive to the toxicity of platinum-type drugs. Tumor protection by platinum-protecting drugs is an additional untoward effect that is associated with certain types of agents and must be addressed with care. Recent advances in theoretical and laboratory methods and the use of supercomputers have extended our understanding of the possible major mechanisms underlying platinum drug antitumor activity and toxicity; we present strong evidence that there are two classes of chemical species of platinum drug. One class appears to predominantly account for the antitumor activity, and the other class of chemical species produces many of the toxic effects of platinum drugs. We have discovered a new nontoxic, second-generation platinum-protecting agent, known as BNP7787, which appears to selectively inactivate and eliminate toxic platinum species. BNP7787 has recently entered phase I clinical testing in cancer patients.
- Published
- 1998
131. Topoisomerase-I inhibitor SN-38 can induce DNA damage and chromosomal aberrations independent from DNA synthesis.
- Author
-
Voigt W, Matsui S, Yin MB, Burhans WC, Minderman H, and Rustum YM
- Subjects
- Camptothecin pharmacology, Chromosome Aberrations, DNA Replication, Dose-Response Relationship, Drug, Humans, Irinotecan, S Phase, Sister Chromatid Exchange, Tumor Cells, Cultured drug effects, Camptothecin analogs & derivatives, Chromatids drug effects, DNA Damage, DNA, Neoplasm drug effects, Enzyme Inhibitors pharmacology, Topoisomerase I Inhibitors
- 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.
- Published
- 1998
132. Interleukin 15 offers selective protection from irinotecan-induced intestinal toxicity in a preclinical animal model.
- Author
-
Cao S, Black JD, Troutt AB, and Rustum YM
- Subjects
- Animals, Antineoplastic Agents, Phytogenic pharmacology, Camptothecin pharmacology, Camptothecin toxicity, Colon drug effects, Colon pathology, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Disease Models, Animal, Drug Interactions, Duodenum drug effects, Duodenum pathology, Female, Intestinal Mucosa drug effects, Intestinal Mucosa pathology, Irinotecan, Paraffin Embedding, Rats, Rats, Inbred F344, Antineoplastic Agents, Phytogenic toxicity, Camptothecin analogs & derivatives, Diarrhea chemically induced, Diarrhea prevention & control, Interleukin-15 therapeutic use
- Abstract
Irinotecan (CPT-11) is a chemotherapeutic agent that is active in the treatment of a variety of solid tumor malignancies. Diarrhea represents the most common dose-limiting toxicity that is independent of the schedule of administration. A rat model with dose-limiting toxicity profiles that are similar to those observed in patients treated with CPT-11 was developed and used to evaluate the role of interleukin 15 (IL-15) in the modulation of the therapeutic selectivity of CPT-11 in normal rats and rats bearing advanced colorectal cancer. The maximum tolerated dose and lethal dose (LD) of CPT-11 by i.v. push daily x 3 were 150 and 200 mg/kg/day, respectively. CPT-11 at the LD induced a 93-100% incidence of severe diarrhea and an 86-100% incidence of lethality in treated animals. IL-15, a cytokine with multiple mechanisms of action, was used at a 100 or 400 microg/kg/dose with different schedules of administration (3, 8, and 11 doses, i.p.) to protect against CPT-11-induced toxicity. IL-15 offered complete and sustained selective protection against CPT-11-induced delayed diarrhea and lethality. IL-15 also moderately potentiated the antitumor activity of CPT-11 in rats bearing advanced colorectal cancer. Morphological examination of rat intestinal tissues after treatment with LD of CPT-11 revealed dramatic protection of duodenal and colonic tissue architecture by IL-15. CPT-11 alone produced serious damage to duodenal villi and colonic crypts. The results clearly demonstrated the ability of IL-15 to provide significant protection from CPT-11-induced intestinal toxicity with maintenance of antitumor activity, resulting in an increase in the therapeutic index of CPT-11. The clinical relevance of the results obtained in this model system needs to be confirmed.
- Published
- 1998
133. Super in vitro synergy between inhibitors of dihydrofolate reductase and inhibitors of other folate-requiring enzymes: the critical role of polyglutamylation.
- Author
-
Faessel HM, Slocum HK, Jackson RC, Boritzki TJ, Rustum YM, Nair MG, and Greco WR
- Subjects
- Antineoplastic Combined Chemotherapy Protocols metabolism, Drug Synergism, Folic Acid Antagonists metabolism, Glutamates metabolism, Glutamates pharmacology, Humans, Methotrexate metabolism, Methotrexate pharmacology, Pyrimidines metabolism, Pyrimidines pharmacology, Thymidylate Synthase antagonists & inhibitors, Trimetrexate metabolism, Trimetrexate pharmacology, Tumor Cells, Cultured drug effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, Folic Acid Antagonists pharmacology, Polyglutamic Acid metabolism, Tetrahydrofolate Dehydrogenase drug effects
- Abstract
The combined action among polyglutamylatable and nonpolyglutamylatable antifolates, directed against dihydrofolate reductase (DHFR), glycinamide ribonucleotide formyltransferase (GARFT), 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (AICARFT), and thymidylate synthase (TS), in human ileocecal HCT-8 cells was examined in a 96-well plate growth inhibition assay (96-h continuous drug exposure). An interaction parameter, alpha, was estimated for each of 95 experiments by fitting a seven-parameter model to data with weighted nonlinear regression. In a representative experiment, raising the folic acid concentration in the medium dramatically increased the Loewe synergy for the combination of trimetrexate (TMTX) and the GARFT inhibitor AG2034 (from a mean alpha +/- SE of 1.50 +/- 0.25 at 2.3 microM folic acid to 146 +/- 20 at 78 microM folic acid). Enhancements were also found for combinations of TMTX with the GARFT inhibitors AG2032, Lometrexol, and LY309887, the AICARFT inhibitor AG2009, and the TS inhibitors LY231514 and Tomudex but not with the GARFT inhibitor LL95509 or with the TS inhibitors AG337, ZD9331, and BW1843U89. Replacing TMTX with methotrexate in two-drug mixtures decreased the intensity of Loewe synergy. Examination of isobolograms at different effect levels revealed informative reproducible changes in isobol patterns. No two-drug combinations among inhibitors of GARFT, AICARFT, and TS exhibited Loewe synergy at either 2.3 or 78 microM folic acid. Thus, the ideal requirement for the folic acid-enhanced synergy is that a nonpolyglutamylatable DHFR inhibitor be combined with a polyglutamylatable inhibitor of another folate-requiring enzyme. A hypothesis to explain this general phenomenon involves the critical role of folylpoly-gamma-glutamate synthetase and the effect of the DHFR inhibitor in decreasing the protection by folic acid of cells to the other antifolates.
- Published
- 1998
134. Cell cycle: molecular targets for diagnosis and therapy: tumor suppressor genes and cell cycle progression in cancer.
- Author
-
Giordano A, Rustum YM, and Wenner CE
- Subjects
- Animals, Humans, Neoplasms genetics, Cell Cycle genetics, Genes, Tumor Suppressor, Neoplasms pathology
- Abstract
A significant portion of published literature is dedicated to describing the cloning and the characterization of proteins involved in the progression of the cell cycle, which govern cell growth both in cancer and normal ontogenesis. With this abundance of information, the cascading pathways of molecular events that occur in the cell cycle are proving to be exceedingly complicated. The purpose of this conference was to attract the leading clinical and basic science investigators in the growth control field with a final goal to determine how this current wealth of knowledge can be used to impact upon patient care and management by the design of novel adjuvant therapeutics specifically targeted at tumor cells and the identification of molecular diagnostic and/or prognostic markers in an efficient and cost effective manner.
- Published
- 1998
- Full Text
- View/download PDF
135. Interleukin 15 protects against toxicity and potentiates antitumor activity of 5-fluorouracil alone and in combination with leucovorin in rats bearing colorectal cancer.
- Author
-
Cao S, Troutt AB, and Rustum YM
- Subjects
- Animals, Diarrhea chemically induced, Diarrhea prevention & control, Female, Fluorouracil adverse effects, Interleukin-2 administration & dosage, Leucovorin adverse effects, Rats, Rats, Inbred F344, Stomatitis chemically induced, Stomatitis prevention & control, Weight Loss drug effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Fluorouracil administration & dosage, Interleukin-15 administration & dosage, Leucovorin administration & dosage
- Abstract
5-Fluorouracil (FUra) modulated by leucovorin (LV) is active in the treatment of colorectal cancer. Diarrhea and stomatitis are the most common dose-limiting toxicities. We have developed a model system in rats bearing a transplantable colon carcinoma sensitive to FUra therapy with dose-limiting toxicity profiles similar to what is observed in patients treated with either daily or weekly schedules of FUra plus LV. Interleukin 15 (IL-15), a cytokine that shares many biological activities with IL-2, was used at different doses (25, 100, and 400 microg/kg) and schedules (three doses before a single dose of FUra, FUra/LV daily x 5, or before each week of FUra/LV weekly x 4, or three doses before a single dose of FUra or FUra/LV daily x 5, then twice daily x 5 for a total of 11 doses) to evaluate its role in the modulation of the therapeutic selectivity of FUra alone and modulated by LV. IL-15 induced a dramatic decrease in chemotherapy-induced gastrointestinal toxicities, significant potentiation of antitumor activity, and an increased therapeutic index of FUra administered on single dose, daily x 5 and weekly x 4 schedules. In contrast, IL-2 (400 microg/kg) significantly potentiated the toxicity of FUra administered as a single i.v. push, with minimal potentiation of the antitumor activity. Taken together, the results clearly demonstrated the ability of IL-15, but not IL-2, to provide significant improvement of the therapeutic index of FUra alone and in combination with LV. The clinical relevance of the results obtained in this model system needs to be confirmed.
- Published
- 1998
136. Novel cellular determinants for reversal of multidrug resistance in cells expressing P170-glycoprotein.
- Author
-
Yin MB, Guo B, Voigt W, Vanhoefer U, Gibbs JF, Skenderis BS, Frank C, Wrzosek C, and Rustum YM
- Subjects
- 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, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, DNA Damage, DNA, Neoplasm drug effects, Drug Resistance, Multiple physiology
- 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.
- Published
- 1998
- Full Text
- View/download PDF
137. Comparison of 5-fluoro-2'-deoxyuridine with 5-fluorouracil and their role in the treatment of colorectal cancer.
- Author
-
van Laar JA, Rustum YM, Ackland SP, van Groeningen CJ, and Peters GJ
- Subjects
- Antimetabolites, Antineoplastic metabolism, Cell Division, Clinical Trials as Topic, Colorectal Neoplasms metabolism, Floxuridine administration & dosage, Floxuridine metabolism, Fluorouracil administration & dosage, Fluorouracil metabolism, Humans, Tumor Cells, Cultured, Antimetabolites, Antineoplastic therapeutic use, Colorectal Neoplasms drug therapy
- Abstract
Despite more than 30 years of intensive studies on new drugs against advanced colorectal cancer, the fluoropyrimidines remain the drugs of choice for systemic treatment and for hepatic artery infusion (HAI). This overview describes new developments in advanced colorectal cancer chemotherapy, providing a rationale for more effective use of the fluoropyrimidines, with biochemical modulation, scheduling or by revealing biochemical mechanisms of action that correlate with antitumour activity. In human colorectal cancer cell lines and various animal tumour model systems 5-fluoro-2'-deoxyuridine (FdUrd) is more effective than 5-fluorouracil (5-FU). Comparably, FdUrd's modulation by leucovorin (LV) is more potent than 5-FU. In animal studies it is shown that intermittent high-bolus administration of FdUrd generates better antitumour activity, compared with equal toxic doses or any other schedule of 5-FU. These effects are related to prolonged-thymidylate synthase (TS) inhibition and the prevention of TS induction, rather than RNA incorporation. Preclinical studies with modulators such as N-phosphonacetyl-L-aspartate (PALA), WR-2721, mitomycin C and platinum derivatives provide a rationale for clinical use in the future. The first choice systemic chemotherapy of patients with advanced colorectal cancer remains 5-FU combined with LV. Some improvement in therapeutic efficacy has been achieved with locoregional HAI. In randomised studies HAI FdUrd improves the quality of life and survival as compared with optimal systemic therapy. Chronomodulation decreases toxicity, allowing dose intensification, while modulators such as LV or dexamethasone increase survival of patients treated with HAI FdUrd to 86% after 1 year. In conclusion, the clinical use of FdUrd has not been fully explored. Intermittent high-dose FdUrd, chronomodulation together with the use of modulators or drugs focused on prolonged TS inhibition, should be studied in large randomised studies.
- Published
- 1998
- Full Text
- View/download PDF
138. Rationale for treatment design: biochemical modulation of 5-fluorouracil by leucovorin.
- Author
-
Rustum YM, Cao S, and Zhang Z
- Subjects
- Animals, Clinical Trials as Topic, Drug Interactions, Fluorouracil administration & dosage, Humans, Leucovorin administration & dosage, Rats, Antineoplastic Combined Chemotherapy Protocols pharmacology, Fluorouracil pharmacology, Leucovorin pharmacology, Neoplasms drug therapy
- Abstract
Preclinical in vitro and in vivo results have demonstrated the conditions required for optimal modulation of 5-FU activity by LV. The ability to increase intracellular concentrations of higher chain length polyglutamates was a function of duration of longer exposure to LV rather than the dose. In rats bearing advanced colorectal tumors, the role of LV dosage was more clearly evident with the weekly 5-FU treatment schedule than with the daily schedule. Phase III clinical trials in patients with advanced colorectal cancer demonstrated that low-dose and high-dose LV (daily x 5) and weekly high-dose LV schedules yielded similar response rates with different toxicity profiles. A phase III trial demonstrated significant therapeutic advantages for a bimonthly schedule of high-dose LV over a monthly schedule of low-dose LV. Taken together, these results provide insight into LV biomodulation, but the optimal conditions for these regimens for individual patients remain undetermined. To date it has not been possible to identify the optimal conditions for modulation of 5-FU by LV in individual patients with advanced colorectal cancer, and response rates are comparable. A regimen that offers the opportunity to manage treatment-induced toxicity is recommended. With diarrhea being the primary dose-limiting toxicity with the weekly 5-FU and high-dose LV (manifested during the 2-3 weeks of treatment), management of toxicity can be achieved by delaying treatment, by dose reduction, and/or by treatment with octreotide47 without compromising efficacy. In contrast, with the daily x 5 schedule, multiple toxicities (mucositis [stomatitis], diarrhea, neutropenia, and hand and foot syndrome) are manifested regardless of the dose of LV administered. An additional advantage to the weekly schedule is that it provides the opportunity to use 5-FU/LV treatment in sequence or combination with other drugs, such as topoisomerase I inhibitors (CPT-11), antifolates (methotrexate, trimetrexate), and platins (oxaliplatin).
- Published
- 1998
139. Comparative antitumor efficacy of docetaxel and paclitaxel in nude mice bearing human tumor xenografts that overexpress the multidrug resistance protein (MRP)
- Author
-
Vanhoefer U, Cao S, Harstrick A, Seeber S, and Rustum YM
- Subjects
- Animals, Docetaxel, Fibrosarcoma drug therapy, Humans, Mice, Mice, Nude, Multidrug Resistance-Associated Proteins, Sarcoma, Experimental metabolism, Transplantation, Heterologous, Tumor Cells, Cultured, Up-Regulation, ATP-Binding Cassette Transporters biosynthesis, Antineoplastic Agents, Phytogenic pharmacology, Gene Expression Regulation, Neoplastic, Neoplasm Proteins biosynthesis, Paclitaxel analogs & derivatives, Paclitaxel pharmacology, Sarcoma, Experimental drug therapy, Taxoids
- Abstract
Background: Multidrug resistance has been associated with expression of the multidrug resistance protein (MRP). Recently, MRP-expression has been detected in human tumor samples of patients with breast cancer and non-small-cell lung cancer. Since taxoids are the most active drugs in the treatment of both tumor entities, the antitumor efficacies of paclitaxel and docetaxel were compared in nude mice bearing human tumor xenografts that express MRP., Materials and Methods: Athymic nude mice (nu/nu) bearing tumor xenografts of parental human sarcoma HT1080 or MRP-expressing HT1080/DR4 cells (as confirmed by Northern blot analysis) were treated with the maximum tolerated doses (MTD) of doxorubicin ([Dx] 10 mg/kg i.v. push), paclitaxel ([PC] 50 mg/kg three-hour i.v. infusion), or docetaxel ([DC] 40 mg/kg three-hour i.v. infusion). In vitro, the activity of doxorubicin, paclitaxel and docetaxel was evaluated by the sulphorhodamine B (SRB) assay using the pyridine analogue PAK-104P (5 microM), a potent inhibitor of MRP-function., Results: At their MTDs both taxoids showed significant activity against MRP-negative HT1080 xenografts with response rates of 80% (40% CR) for PC and 100% (60% CR) for DC. In contrast, DC was significantly more active than PC in nude mice bearing doxorubicin resistant MRP-expressing HT1080/DR4 tumor xenografts (overall response rates: 100% (60% CR) for DC; 10% (0% CR) for PC; 0% for Dx). Since treatment of mice with the MTD of PC or DC yielded similar overall toxicity (maximum weight loss for HT1080: PC 8.6 +/- 2.2%; DC 7.5 +/- 2.2% and for HT1080/DR4: PC 11.6 +/- 3.0%; DC 7.6 +/- 1.8%, respectively), these results demonstrate the increase in the therapeutic index for docetaxel against MRP-expressing tumors. In vitro, HT1080/DR4 cells were 270-fold, 6.4-fold and 2.8-fold more resistant than parental cells to doxorubicin, PC and DC, respectively. Pyridine analogue PAK-104P completely restored drug sensitivity to PC and DC, while no effect of PAK-104P on parental HT1080 cells was observed., Conclusions: Both taxoids, when given at their MTDs, showed significant efficacy against parental HT1080 tumor xenografts. However, docetaxel at its MTD was significantly more active against MRP-expressing tumor xenografts than paclitaxel. Furthermore, in vitro resistance of HT1080/DR4 cells was higher for PC (6.4-fold) than for DC (2.8-fold). Since PAK-104P completely restored sensitivity to both taxoids, the observed resistance appears to be related to MRP. These data suggest, that docetaxel is not as readily transported by MRP as paclitaxel leading to an increased therapeutic ratio in MRP-expressing tumors in vivo. Therefore, docetaxel may have therapeutic advantages in the clinical treatment of MRP-expressing tumors.
- Published
- 1997
- Full Text
- View/download PDF
140. Basic science research in postgraduate surgical training: difficulties encountered by clinical scientists.
- Author
-
Skenderis BS 2nd, Rustum YM, and Petrelli NJ
- Subjects
- Clinical Competence, Communication, Gene Expression Regulation, Neoplastic, Genes, p53 genetics, Humans, Internship and Residency, Laboratories, Physician-Patient Relations, Research Design, Students, Medical, Teaching methods, Tumor Suppressor Protein p53 genetics, Education, Medical, Graduate, General Surgery education, Research education, Science education
- Abstract
Background: Clinical training of physicians and surgeons involves teaching students and residents both the science and the art of medicine. The science of medicine involves clinical skills honed through reading textbooks and journals and experience in diagnosing and treating myriad diseases. The art of medicine involves the important communication skills necessary for a good doctor-patient relationship. A third aspect of training, basic science research, is frequently de-emphasized or omitted entirely. In general, training programs frequently do not allow protected time in a laboratory setting for the resident. The few weeks to months that some programs allow is insufficient to establish a thorough understanding of scientific method., Methods, Results: The article describes the introduction of one of the authors from a clinically oriented training background into the laboratory and outlines the pitfalls frequently encountered when a clinician embarks on basic science research training., Conclusion: The necessity for sufficient protected time in the laboratory away from clinical responsibilities is recognized.
- Published
- 1997
- Full Text
- View/download PDF
141. Evaluation of topoisomerase I catalytic activity as determinant of drug response in human cancer cell lines.
- Author
-
Voigt W, Vanhoefer U, Yin MB, Minderman H, Schmoll HJ, and Rustum YM
- Subjects
- Antineoplastic Agents, Phytogenic pharmacology, Camptothecin analogs & derivatives, Carbazoles pharmacology, Catalysis, Enzyme Inhibitors pharmacology, Glucosides pharmacology, Humans, Irinotecan, Topoisomerase I Inhibitors, Tumor Cells, Cultured, Camptothecin pharmacology, DNA Topoisomerases, Type I metabolism
- 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.
- Published
- 1997
142. p53 and WAF1 are induced and Rb protein is hypophosphorylated during cell growth inhibition by the thymidylate synthase inhibitor ZD1694 (Tomudex).
- Author
-
Yin MB, Voigt W, Panadero A, Vanhoefer U, Frank C, Pajovic S, Azizkhan J, and Rustum YM
- Subjects
- 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, 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
- 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.
- Published
- 1997
- Full Text
- View/download PDF
143. 5-Fluorouracil prodrug, ftorafur, modulated by uracil (UFT): preclinical and clinical prospective.
- Author
-
Cao S and Rustum YM
- Subjects
- Animals, Female, Fluorouracil administration & dosage, Rats, Rats, Inbred F344, Adenocarcinoma drug therapy, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Tegafur therapeutic use
- Published
- 1997
144. Mechanism-based improvement in the therapeutic selectivity of 5-FU prodrug alone and under conditions of metabolic modulation.
- Author
-
Rustum YM
- Subjects
- Administration, Oral, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Drug Administration Schedule, Drug Combinations, Drug Synergism, Leucovorin administration & dosage, Prodrugs administration & dosage, Rats, Tegafur metabolism, Fluorouracil administration & dosage, Tegafur administration & dosage, Uracil administration & dosage
- Abstract
To compare the antitumor activity of Tegafur (150 mg/kg/day) with continuous intravenous infusion of 5-fluorouracil (5-FU) (12.5 mg/kg/day) and with oral UFT (60 mg/kg/day) with and without low- or high-dose leucovorin (50 or 200 mg/kg/day), rats with advanced colon cancer were treated with Tegafur or UFT 3 times daily for 28 days and 5-FU by continuous intravenous infusion for 28 days. UFT alone had a complete remission (CR) rate of 38%, whereas Tegafur and 5-FU produced no CRs. When high-dose leucovorin was added, the CR rate for UFT increased to 75%; Tegafur plus high-dose leucovorin resulted in only a partial remission rate of 50%, with no CRs; low-dose leucovorin was not as effective as the high dose. Hence, UFT clearly offers significant therapeutic advantages over Tegafur and protracted infusion of 5-FU. High-dose leucovorin is essential for significant modulation of drug action in this tumor.
- Published
- 1997
- Full Text
- View/download PDF
145. Cellular determinants of resistance to indolocarbazole analogue 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 (NB-506), a novel potent topoisomerase I inhibitor, in multidrug-resistant human tumor cells.
- Author
-
Vanhoefer U, Voigt W, Hilger RA, Yin MB, Harstrick A, Seeber S, and Rustum YM
- Subjects
- 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, Carbazoles pharmacology, Drug Resistance, Multiple, Enzyme Inhibitors pharmacology, Fibrosarcoma drug therapy, Glucosides pharmacology, Ovarian Neoplasms drug therapy, Topoisomerase I Inhibitors
- 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.
- Published
- 1997
146. Combined action of paclitaxel and cisplatin against wildtype and resistant human ovarian carcinoma cells.
- Author
-
Levasseur LM, Greco WR, Rustum YM, and Slocum HK
- Subjects
- Cell Division drug effects, Cisplatin administration & dosage, Colonic Neoplasms pathology, Drug Resistance, Neoplasm, Female, Head and Neck Neoplasms pathology, Humans, Paclitaxel administration & dosage, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols pharmacology, Ovarian Neoplasms pathology
- Abstract
Purpose: The combination of paclitaxel (PTX) and cisplatin (DDP) shows good clinical efficacy against ovarian cancer. In order to examine the potential cellular basis for this, and provide leads as to how to optimize the combination, we examined the role of sequence of exposure to PTX and DDP on cell growth in vitro., Methods: Four human ovarian carcinoma cell lines, A121, A2780/WT, A2780/DX5B and A2780/CP3, two human head and neck carcinoma cell lines, A253 and FaDu, and the human ileocecal carcinoma cell line, HCT-8, were treated with PTX + DDP with seven schedules: (A) 96 h exposure to PTX + DDP; (B) 24 h PTX alone, then 72 h PTX + DDP; (C) 4 h DDP alone, then 92 h PTX + DDP; (D) 24 h PTX alone, 4 h DDP alone, then 68 h drug-free; (E) 4 h DDP alone, 24 h PTX alone, then 68 h drug-free; (F) 3 h PTX alone, 1 h DDP alone, then 92 h drug-free; and (G) 1 h DDP alone, 3 h PTX alone, then 92 h drug-free. Each of 66 two-drug experiments included five plates (440 randomly treated wells per experiment). Cell growth was measured by the sulforhodamine B assay. The nature and the intensity of the drug interactions were assessed by fitting a seven-parameter model to data with weighted nonlinear regression, enabling the estimation of an interaction parameter, alpha, with its standard error., Results: Overall there was very little departure from Loewe additivity: 43 experiments showed Loewe additivity, 10 showed Loewe antagonism, and 13 showed slight Loewe synergy. In vitro Loewe synergy was rare, was small when present, and reproducible only for the A121 and HCT-8 cells exposed to schedule D (24 h PTX prior to 4 h DDP). Isobolographic analysis showed complex combined-action surfaces with regions of local Loewe synergy and antagonism., Conclusion: It appears unlikely that the good clinical efficacy of the combination is primarily caused by a synergistic interaction at the cellular level.
- Published
- 1997
- Full Text
- View/download PDF
147. d,l-buthionine-(S,R)-sulfoximine potentiates in vivo the therapeutic efficacy of doxorubicin against multidrug resistance protein-expressing tumors.
- Author
-
Vanhoefer U, Cao S, Minderman H, Toth K, Skenderis BS 2nd, Slovak ML, and Rustum YM
- Subjects
- Animals, Buthionine Sulfoximine administration & dosage, Doxorubicin administration & dosage, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Drug Synergism, Female, Genes, MDR physiology, Glutathione blood, Humans, Immunohistochemistry, Mice, Mice, Nude, Neoplasm Transplantation, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Fibrosarcoma drug therapy
- Abstract
Intracellular glutathione (GSH) has been implicated as a regulatory determinant of multidrug resistance protein (MRP) function. The objective of the present study was to evaluate in vivo the ability of d,l-buthionine-(S,R)-sulfoximine (d,l-BSO), a potent inhibitor of GSH biosynthesis, to reverse MRP-mediated drug resistance to doxorubicin. Athymic nude mice (nu/nu) bearing advanced parental human fibrosarcoma HT1080 and MRP-expressing HT1080/DR4 tumors were treated with the maximum tolerated dose of doxorubicin (10 mg/kg, i. v. push). This therapy produced an overall response rate of 50% (20% complete response and 30% partial response) in mice bearing parental HT1080 xenografts, whereas no significant antitumor activity against HT1080/DR4 tumors was observed. Treatment of mice bearing HT1080 and HT1080/DR4 xenografts with a continuous i.v. infusion of nontoxic doses of d,l-BSO (300 and 600 mg/kg/day) produced a 60% reduction of GSH plasma levels and greater than 95% reduction in GSH tumor levels in both parental and multidrug-resistant tumors; however, this treatment possessed no in vivo antitumor activity by itself. Under these treatment conditions, a combination of d,l-BSO with the maximum tolerated dose of doxorubicin administered at 24 h during a 48-h i.v. infusion of d,l-BSO completely restored the response of MRP-expressing HT1080/DR4 tumors to doxorubicin (overall response rate, 63%; complete response rate, 38%) with no potentiation of host toxicity. The d,l-BSO-induced in vivo reversal of MRP-mediated drug resistance correlated in vitro with the restoration of intracellular doxorubicin retention in cultured HT1080/DR4 cells. Depletion of GSH by d,l-BSO in drug-sensitive HT1080 tumors that do not express MRP did not alter the in vivo response to doxorubicin. Using the same treatment schedule, dose, and administration of doxorubicin with and without d,l-BSO in nude mice bearing P-170 glycoprotein-expressing A2780/Dx5 tumors, no potentiation of the therapeutic index of doxorubicin was found, demonstrating the in vivo selectivity of d, l-BSO-induced GSH depletion on MRP-function. The data reported herein indicate that in vivo function of MRP as a mediator of doxorubicin resistance requires the presence of sufficient GSH pools. d,l-BSO may provide an example of an effective in vivo modulator of MRP-mediated drug resistance.
- Published
- 1996
148. A unique human ovarian carcinoma cell line expressing CD34 in association with selection for multidrug resistance.
- Author
-
Minderman H, Vanhoefer U, Toth K, Minderman MD, and Rustum YM
- Subjects
- DNA Topoisomerases, Type II analysis, Female, Flow Cytometry, Humans, Leukemia, Myeloid, Acute metabolism, Ovarian Neoplasms metabolism, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Antigens, CD34 metabolism, Drug Resistance, Multiple, Drug Resistance, Neoplasm, Neoplasm Proteins metabolism, Tumor Cells, Cultured metabolism
- Abstract
Background: The role of P-glycoprotein (Pgp) in multidrug resistance (MDR) is uncontested. Expression of Pgp on hematopoietic cells has been correlated with CD34 expression. For acute myeloid leukemia, the prognostic value of Pgp for clinical response is at best equivalent to that of CD34. The current study investigated whether expression of CD34 can be associated with selection for drug resistance., Methods: Several established MDR cell lines were screened by flow cytometry for expression of CD34. Human ovarian carcinoma cells (A2780), which simultaneously expressed CD34 and Pgp, were identified. Subsequent cloning resulted in a new cell line (A2780-Dx5c) that expressed CD34 in the absence of Pgp. Involvement of non-Pgp-mediated MDR mechanisms was assessed by immunohistochemistry (MRP and LRP), enzyme activity studies (glutathione pathway), cross-resistance patterns, and Northern blot (type II alpha topoisomerase)., Results: A2780-Dx5c was cross-resistant to doxorubicin, daunorubicin, idarubicin, and VP-16. However, unlike the Pgp-expressing cells, it was not cross-resistant to vincristine or amsacrine. The drug resistance was correlated with a decreased level of type II alpha topoisomerase in the A2780-Dx5c cell line compared with the parental cell line. No evidence was found of involvement of MRP, LRP, or the glutathione pathway with drug resistance in this cell line., Conclusions: A new cell line of nonhematopoietic and nonvascular endothelial origin that expresses CD34 in association with selection for MDR was cloned. A study of MDR mechanisms in this cell line revealed that reduced type II alpha topoisomerase levels were likely responsible for the MDR observed. A study of the causal relation between the selection of drug resistance and the expression of CD34 may provide insight into why CD34 correlates with poor clinical response in patients with acute myeloid leukemia.
- Published
- 1996
149. DNA damage and p53 induction do not cause ZD1694-induced cell cycle arrest in human colon carcinoma cells.
- Author
-
Matsui SI, Arredondo MA, Wrzosek C, and Rustum YM
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Cell Division drug effects, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Cyclin-Dependent Kinase Inhibitor p21, Cyclins biosynthesis, DNA Damage, DNA Replication drug effects, DNA, Neoplasm drug effects, G1 Phase drug effects, G2 Phase drug effects, Humans, S Phase drug effects, Tumor Cells, Cultured, Adenocarcinoma drug therapy, Antimetabolites, Antineoplastic pharmacology, Cell Cycle drug effects, Colonic Neoplasms drug therapy, Neoplasm Proteins biosynthesis, Quinazolines pharmacology, Thiophenes pharmacology, Thymidylate Synthase antagonists & inhibitors, Tumor Suppressor Protein p53 biosynthesis
- Abstract
Using four complementary approaches, ie., cell synchronization, bromodeoxyuridine labeling, and DNA and Western blot analyses, we investigated the underlying mechanism of cell cycle perturbation in response to ZD1694, a quinazoline-based antifolate thymidylate synthase inhibitor. With a single exposure at a concentration of 1 microM for 2 h, ZD1694 completely inhibits thymidylate synthase over 72 h and causes a sustained growth for at least 120 h, DNA damage, and p53 induction in human carcinoma cells. Although these cells displayed an S-phase block with the precise terminal arrest point depending on the timing of drug treatment in the cell cycle, their DNA-replicating machinery associated with polymerase alpha was preserved intact. When supplemented with exogenous dThd, these cells resumed an apparently normal S-phase progression for at least 4 h. Kinetic analyses based on synchronized cells indicate that S-phase arrest occurs first, preceding the induction of DNA double strand breaks and p53/p21. SW480 cells, in which p53mu failed to transduce p21, also exhibited the mode of S-phase arrest, essentially indistinguishable from that displayed by HCT-8 cells expressing the functional p53 (p53wt). That the DNA replication process is prerequisite for DNA double strand breaks was indicated by the following: (a) DNA damage occurred only when cells treated with ZD1694 progressed through S phase; and (b) the inhibition of DNA polymerase alpha by aphidicolin-blocked DNA damage. Based on the above, we conclude that S-phase arrest by ZD1694, with a subsequent damage of DNA double strands, is caused by the block of DNA synthesis in the middle of replication due to dTTP depletion and not by p53-mediated G1-G2 checkpoint mechanisms or p21-induced inactivation of the DNA-replicating machinery.
- Published
- 1996
150. MDR1 P-glycoprotein is expressed by endothelial cells of newly formed capillaries in human gliomas but is not expressed in the neovasculature of other primary tumors.
- Author
-
Tóth K, Vaughan MM, Peress NS, Slocum HK, and Rustum YM
- Subjects
- Adult, Brain blood supply, Brain metabolism, Brain Neoplasms pathology, Brain Neoplasms secondary, Capillaries metabolism, Drug Resistance, Multiple physiology, Endothelium, Vascular pathology, Glioma blood supply, Glioma pathology, Humans, Immunohistochemistry methods, Neoplasm Staging, Neovascularization, Pathologic pathology, ATP Binding Cassette Transporter, Subfamily B, Member 1 biosynthesis, Brain Neoplasms blood supply, Endothelium, Vascular metabolism, Glioma metabolism, Neovascularization, Pathologic metabolism
- Abstract
The expression of human MDR1 P-glycoprotein (Pgp) in the capillary endothelial cells of the central nervous system has been demonstrated. The brain capillary endothelial cells maintain the structure and function of the blood-brain barrier. Recently, the human MDR1 Pgp (and its mouse homologue MDR1a Pgp) has been shown to function as an important part of this barrier, pumping out xenobiotics from endothelial cells into the lumen of capillaries resulting in the protection of the brain parenchyma. To examine whether the endothelial cells of the newly formed capillaries during neoangiogenesis within malignant human brain tumors express MDR1 Pgp, 35 adult surgical brain tumor specimens (29 gliomas and 6 tumors metastatic to the brain) were obtained from previously untreated patients and studied by a new immunohistochemical sandwich method developed in our laboratory using the JSB-1 monoclonal antibody. JSB-1 is specific for the Pgp product of the human MDR1 (and not MDR3) gene. This sensitive method allows the detection of Pgp in capillary endothelial cells of normal brain in conventional paraffin sections after formalin fixation. The endothelial cells of the newly formed capillaries in 25 of 29 gliomas (86%) and 3 of 6 metastatic tumors, immunostained positive for MDR1 Pgp. The tumor cells in 7 of 35 cases were also positive for Pgp. In the 35 brain tumor cases investigated, the endothelial cells were Pgp positive in the tumor-brain border and in the brain further from the tumor. Capillary endothelial cells of neovasculature in 137 malignant tumors (non-brain) obtained from previously untreated patients showed no MDR1 Pgp expression. These results demonstrated that MDR1 Pgp is expressed not only in the capillaries of normal brain but also in the majority of the newly formed capillaries of brain tumors. Multidrug resistance of brain tumors may result not only from the expression of resistance markers in neoplastic cells but also from the MDR1 Pgp expression in endothelial cells of tumor capillaries. Pgp in this special localization can exclude chemotherapeutic agents from tumor cells that are located around the capillaries. The therapeutic benefit and selectivity of chemotherapeutic agents in combination with a Pgp-reversing agent should be evaluated.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.