Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Radiat Oncol Biol Phys ; 39(5): 1145-52, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9392557

ABSTRACT

PURPOSE: To evaluate the effects of biochemical modulation by N-(phosphonacetyl)-L-aspartate (PALA), 6-methylmercaptopurine riboside (MMPR), and 6-aminonicotinamide (6AN), (PALA + MMPR + 6AN is referred to as PMA) on tumor radiosensitivity, and evaluate the efficacy of the addition of 5-FU to the PMA + XRT regimen for enhancement of tumor response to radiation without exceeding normal tissue tolerance. METHODS AND MATERIALS: A first generation transplant of the CD8F1 spontaneous murine tumor was studied. 31P nuclear magnetic resonance spectroscopy was used to determine the interval between chemotherapy and radiation based on energy depletion. PMA was administered three times with fractionated XRT (15 Gy x 3 = 45 Gy) on days 1, 10, or 11, and 21. The addition of 5-fluorouracil (5-FU) at maximum tolerated doses was evaluated and intergroup comparisons were made for tumor growth delay, local control, and disproportionate normal tissue damage. RESULTS: The combination of 5-FU + XRT induced a tumor doubling time of 75.4 days (67.4-84.4) (p < 0.0001 compared to XRT), validating that in this tumor model, pretreatment with bolus i.p. 5-FU enhanced XRT. In comparison, mice treated with PMA + XRT had a tumor doubling time (TDT) > 123.2 days (109.4-138.7), (p < 0.0001 compared to 5-FU + XRT). The addition of 5-FU to PMA + XRT induced a doubling time of > 170.8 days (150.7-193.7) (p = 0.0002 compared to PMA + XRT). The doubling time for the PMA + XRT cohort and the PMA + 5-FU + XRT cohorts are underestimates since some of the tumor bearing mice continue to have a complete regression (CR). The CR rate (measured on day 250) for the PMA + 5-FU + XRT cohort was 31.7% compared to 0% for 5-FU + XRT and 10% for PMA + XRT (p < 0.05). Mortality and local effects induced by radiation in the PMA + XRT group were comparable to the toxicity for the PMA + 5-FU + XRT group indicating that the addition of 5-FU at 75 mg/kg to PMA + XRT was tolerated and induced both greater CR and tumor doubling times than XRT alone, 5-FU (150 mg/kg) + XRT, or PMA + XRT. CONCLUSIONS: PMA is superior to 5-FU as a radiosensitizer in the schedule studied. The combination of PMA + 5-FU further enhanced XRT without exceeding normal tissue tolerance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , 6-Aminonicotinamide/administration & dosage , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Cell Division , Combined Modality Therapy , Fluorouracil/administration & dosage , Mammary Neoplasms, Animal/pathology , Methylthioinosine/administration & dosage , Mice , Radiotherapy Dosage , Time Factors
2.
Invest New Drugs ; 15(2): 139-45, 1997.
Article in English | MEDLINE | ID: mdl-9220293

ABSTRACT

The results of several clinical trials support the hypothesis that biochemical modulation may enhance the antitumor activity of 5-Fluorouracil (5-FU). We have performed a phase I trial using a combination of three different biochemical modulators at the optimal dose established in previous clinical trials. The modulators include: phosphonacetyl-l-aspartate (PALA), which may increase 5-FU incorporation into RNA; leucovorin, which potentiates thymidylate synthase inhibition; and 6-methylmercaptopurine riboside (MMPR), which promotes the intracellular retention of fluorinated nucleotides. The treatment regimen consisted of PALA 250 mg/m2 day 1, followed 24 h later by MMPR 150 mg/m2 as an iv bolus, and the initiation of a 24-hour infusion of 5-FU along with leucovorin 50 mg/m2. This regimen was repeated weekly. Doses of 5-FU were escalated in cohorts of four or more patients from 2,000 to 2,600 mg/m2. Among 20 patients entered, the majority had colorectal cancer, and most had received prior 5-FU treatment. Toxicity was predominantly gastrointestinal, and diarrhea was dose-limiting at a 5-FU dose of 2600 mg/m2. There were three partial remissions observed, two of whom had colorectal cancer. Emerging data that casts doubt on the modulation value of PALA at this dose and schedule suggests that revision of this regimen be considered before Phase II trial.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Aspartic Acid/analogs & derivatives , Fluorouracil/therapeutic use , Leucovorin/pharmacology , Methylthioinosine/pharmacology , Phosphonoacetic Acid/analogs & derivatives , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/pharmacology , Colorectal Neoplasms/drug therapy , Drug Interactions , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Humans , Leucovorin/administration & dosage , Male , Methylthioinosine/administration & dosage , Middle Aged , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/pharmacology
3.
Cancer Invest ; 15(2): 111-20, 1997.
Article in English | MEDLINE | ID: mdl-9095206

ABSTRACT

The combination of N-(phosphonacetyl)-L-aspartate (PALA), 6-methylmercaptopurine riboside (MMPR), and 6-aminonicotinamide (6AN) has been shown to be an effective antineoplastic regimen and also to enhance the effects of other antineoplastic agents (1-4). To further enhance the effect of this combination, we investigated the effects of adding adriamycin, at its maximally tolerated dose, to this regimen. The response rate (complete regression+partial regression) for the four-drug regimen was higher than for the three-drug regimen, and the tumor growth delay was also significantly higher than for treatment with PALA, MMPR, 6AN, or after treatment with maximally tolerated doses of adriamycin alone (11 mg/kg). The addition of adriamycin to PALA, MMPR, 6AN did not result in enhancement of the effect of radiation, as measured by tumor growth delay studies and tumor control (complete and partial regression rate). The mechanism of action of the combination of PALA, MMPR, and 6AN is not known definitively, but a possible mechanism previously suggested is biochemical modulation of energy metabolism and inhibition of production of tumor ATP. Treatment with PALA, MMPR, 6AN, and adriamycin (at 2.5 hr post MMPR, 6AN) resulted in a nadir NTP/Pi value, as determined by 31P NMR spectroscopy, at approximately 10 hr post MMPR + 6AN (7.5 hr post adriamycin), which was not significantly different from the NTP/Pi value determined after treatment with the three-drug combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Magnetic Resonance Spectroscopy/methods , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/radiotherapy , Radiation-Sensitizing Agents/pharmacology , 6-Aminonicotinamide/administration & dosage , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Cell Division/drug effects , Disease Progression , Doxorubicin/administration & dosage , Drug Synergism , Evaluation Studies as Topic , Methylthioinosine/administration & dosage , Mice , Phosphorus Isotopes
4.
Anticancer Drugs ; 7(6): 655-9, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8913434

ABSTRACT

Paclitaxel alone is active against the CD8F1 murine spontaneous mammary cancer, and when administered following an ATP-depleting combination of N-(phosphonacetyl)-L-aspartate (PALA) + 6-methylmercaptopurine riboside (MMPR) + 6-aminonicotinamide (6-AN) (PMA) produced significantly enhanced partial tumor regressions over that produced by either paclitaxel alone at the maximal tolerated dose (MTD), or by the PMA drug combination alone, against advanced, first passage spontaneous murine breast tumors. The anticancer activity of paclitaxel is due to enhancement and stabilization of microtubule polymerization. Pertinently, microtubule disassembly (an ATP-dependent process) is known to sharply decrease in the presence of ATP depletion. Thus, the dramatic therapeutic enhancement observed with paclitaxel in combination with PMA is in agreement with biochemical expectations, since PMA has been shown to deplete ATP in CD8F1 tumor cells. The augmented therapeutic results were obtained with approximately one-third the MTD of paclitaxel as a single agent and suggest the potential clinical benefit of more effective treatment with lesser amounts of drug.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Paclitaxel/therapeutic use , 6-Aminonicotinamide/administration & dosage , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Female , Methylthioinosine/administration & dosage , Mice , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Remission Induction
5.
NMR Biomed ; 9(3): 114-20, 1996 May.
Article in English | MEDLINE | ID: mdl-8892397

ABSTRACT

The chemotherapeutic regimen of N-(phosphonacetyl)-L-aspartate (PALA) followed 17 h later by 6-methylmercaptopurine riboside (MMPR) and 6-aminonicotanamide (6AN) has been shown to be a potent sensitizer of anti-neoplastic therapy. We undertook this study to compare the therapeutic and metabolic effects of this triple drug combination vs one of its components, 6AN, in a murine mammary carcinoma. After treatment with PALA, MMPR and 6AN, a new peak was detected which was assigned to 6-phosphogluconate (6PG), which is a marker of inhibition of the pentose phosphate pathway at the 6-phosphogluconate dehydrogenase step. Treatment with PALA, MMPR and 6AN also induced a decrease in the ratios of nucleoside triphosphate/inorganic phosphate (NTP/Pi) and phosphocreatine/inorganic phosphate (PCr/Pi) similar to previous results with a different tumor model. These effects were most pronounced at 6 and 10 h. In addition, an increase in PME'/phosphocholine (PME' = downfield peak in the phosphomonoester region) was detected, which was expected because of the cytotoxic effect of this regimen. Treatment with 6AN alone also resulted in the detection of 6PG with a maximum intensity at 6 h post-6AN. Treatment with 6AN alone induced a smaller change in PME'/PC and failed to cause a decrease in PCr/Pi or NTP/Pi at 6 and 10 h. The enhanced response to the combination of PALA, MMPR and 6AN vs 6AN alone, both with regard to cytotoxicity and radiosensitization, may be due to energy depletion.


Subject(s)
6-Aminonicotinamide/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Pentose Phosphate Pathway/drug effects , Teratogens/pharmacology , 6-Aminonicotinamide/administration & dosage , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Cell Division/drug effects , Magnetic Resonance Spectroscopy/methods , Male , Methylthioinosine/administration & dosage , Mice , Mice, Inbred C3H , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Phosphorus
6.
Biochem Pharmacol ; 51(5): 621-7, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8615898

ABSTRACT

Treatment with a combination (PMA) of (N-phosphonacetyl)-L-aspartic acid (PALA), methylmercaptopurine riboside (MMPR), and 6-aminonicotinamide (6AN) induced partial regressions of CD8F1 murine mammary tumors and provided for tumor growth inhibition without regression of Colon 38 tumors. HPLC-nucleotide pool analysis of CD8 mammary tumors obtained at various times after treatment with PMA revealed that MMPR-5'-phosphate, which inhibits de novo purine nucleotide biosynthesis, was constant at levels of approximately 2.5 nmol/mg protein for 72 hr after treatment. In contrast, the MMPR-5'-phosphate levels of C38 tumors decreased from 24-hr levels at 1.5 nmol/mg protein with a half-time of about 24 hr. Treatment of CD8 tumor-bearing mice with iodotubercidin, a potent inhibitor of adenosine/MMPR kinase, at various times after PMA, reversed both the accumulation of high levels of MMPR-5'-phosphate and the number of partial tumor regressions. These data demonstrate that a cycle of MMPR rephosphorylation is active in the CD8 mammary tumor and suggest that this recycling of MMPR is important for the optimal effect of PMA treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Methylthioinosine/metabolism , 6-Aminonicotinamide/administration & dosage , Adenosine Kinase/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Mammary Neoplasms, Experimental/metabolism , Methylthioinosine/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives
7.
Biochem Pharmacol ; 50(11): 1943-8, 1995 Nov 27.
Article in English | MEDLINE | ID: mdl-8615876

ABSTRACT

DNA-damaging agents, e.g. Adriamycin (ADR), are reported to cause tumor regression by induction of apoptosis. A reduction in the intracellular content of ATP is part of the biochemical cascade of events that ultimately results in programmed death of the cell, or apoptosis. A chemotherapeutic three-drug combination (PMA) consisting of N-(phosphonacetyl)-L-aspartate (PALA) + 6-methylmercaptopurine riboside (MMPR) + 6-aminonicotinamide (6AN) significantly lowers levels of ATP in CD8F1 murine breast tumors in vivo and produces tumor regression by apoptosis. Addition of the DNA-damaging antitumor agent ADR to PMA was found to further significantly deplete ATP in CD8F1 murine breast tumors in vivo with a concomitant significant increase in the number of tumor regressions. The correlative biochemical and therapeutic results are consistent with, and support, the hypothesis that ATP depletion is a significant factor and, therefore, is a worthy therapeutic target in the production of apoptosis.


Subject(s)
Adenosine Triphosphate/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , 6-Aminonicotinamide/administration & dosage , Animals , Apoptosis , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Doxorubicin/administration & dosage , Mammary Neoplasms, Animal/metabolism , Methylthioinosine/administration & dosage , Mice , Mice, Inbred Strains , Neoplasm Transplantation , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Time Factors
8.
Invest New Drugs ; 12(4): 319-21, 1994.
Article in English | MEDLINE | ID: mdl-7775133

ABSTRACT

The biochemical modulators PALA, an inhibitor of aspartate transcarbamylase which depletes uridine nucleotide pools, and 6-methylmercaptopurine riboside (MMPR) which inhibits purine metabolism, selectively potentiate the antitumor activity of 5-fluorouracil (5-FU) in preclinical models. Based on a phase I trial of this combination, we performed a phase II trial in patients with advanced pancreatic cancer. PALA 250 mg/m2 was administered i.v. on day 1, followed 24h later by MMPR 150 mg/m2 as a bolus i.v. injection, and 5-FU 2300 mg/m2 by 24h infusion. Treatments were repeated weekly. Seventeen patients, all previously untreated with chemotherapy, were entered, of whom 14 are evaluable for response. Toxicity > or = grade 2 included nausea (6/17), vomiting (4/17), diarrhea (3/17), stomatitis (5/17), and neurotoxicity (2/17). Among 14 evaluable patients there were no partial responses, and two patients with stable disease. Pretreatment with PALA and MMPR is insufficient to enhance the activity of 5-FU in pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/secondary , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aspartate Carbamoyltransferase/antagonists & inhibitors , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Male , Methylthioinosine/administration & dosage , Middle Aged , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives
9.
Cancer Invest ; 12(3): 296-307, 1994.
Article in English | MEDLINE | ID: mdl-8187007

ABSTRACT

A quadruple drug combination--consisting of a triple-drug combination of N-(phosphonacetyl)-L-aspartate (PALA) + 6-methylmercaptopurine riboside (MMPR) + 6-amino-nicotinamide (6-AN), designed to primarily deplete cellular energy in tumor cells, + Adriamycin (Adria)--yielded significantly enhanced anticancer activity (i.e., tumor regressions) over that produced by either Adria alone at maximum tolerated dose (MTD) or by the triple-drug combination, against large, spontaneous, autochthonous murine breast tumors. The adenosine triphosphate (ATP)-depleting triple-drug combination administered prior to Adria resulted in a 100% tumor regression rate (12% complete regression; 88% partial regression) of spontaneous tumors. Histological examination of treated tumors demonstrated that the treatment-induced mechanism of cancer cell death was by apoptosis. The augmented therapeutic results (100% tumor regressions) were obtained with approximately one-half the MTD of Adria as a single agent and suggest the potential clinical benefit of longer, more effective, and safer treatment by low doses of Adria when combined with the triple-drug combination. Two likely mechanisms of action are discussed: (1) prevention of DNA repair; (2) complementary disruption of biochemical pathways by both the triple-drug combination and the biochemical cascade of apoptosis that is induced by a DNA-damaging anticancer agents such as Adria.


Subject(s)
Doxorubicin/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , 6-Aminonicotinamide/administration & dosage , Animals , Apoptosis , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Dose-Response Relationship, Drug , Drug Therapy, Combination , Energy Metabolism , Mammary Neoplasms, Experimental/metabolism , Methylthioinosine/administration & dosage , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives
10.
Cancer Res ; 52(15): 4074-81, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1379119

ABSTRACT

This report describes a highly active chemotherapeutic drug combination, consisting of N-(phosphonacetyl)-L-aspartate plus 6-methylmercaptopurine riboside plus 6-aminonicotinamide plus 5-fluorouracil, in CD8F1 mice bearing spontaneous, autochthonous, breast tumors or first-passage advanced transplants of these spontaneous tumors. The combination and sequence of administration of these drugs were selected on the basis of known potentiating biochemical interactions. High performance liquid chromatography and nuclear magnetic resonance spectroscopy measurements of biochemical changes resulting from treatment with N-(phosphonacetyl)-L-aspartate plus 6-methylmercaptopurine riboside plus 6-aminonicotinamide indicated a severe depletion of cellular energy levels in the treated tumors. 6-Aminonicotinamide produced a severe block of the pentose shunt, and 5-fluorouracil severely inhibited both thymidylate synthase and thymidine kinase in the treated tumors. This quadruple drug combination, administered on a 10-11-day schedule, produced an impressive partial tumor regression rate of 67% of large, spontaneous, autochthonous, murine breast tumors and a tumor regression rate of 74% of first-passage transplants of the spontaneous breast tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , 6-Aminonicotinamide/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Female , Fluorouracil/administration & dosage , Mammary Neoplasms, Experimental/metabolism , Methylthioinosine/administration & dosage , Mice , Mice, Inbred Strains , Neoplasm Transplantation , Orotic Acid/metabolism , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Phosphoribosyl Pyrophosphate/metabolism
11.
J Natl Cancer Inst ; 83(17): 1235-40, 1991 Sep 04.
Article in English | MEDLINE | ID: mdl-1714507

ABSTRACT

Preclinical and clinical studies demonstrate that the selective antitumor activity of fluorouracil (5-FU) is enhanced by agents which perturb certain intracellular nucleotide pools. We previously demonstrated that the combination of N-phosphonacetyl-L-aspartate (PALA), which depletes pyrimidine nucleotide pools, and 5-FU yielded a 43% response rate among 37 assessable patients with colorectal carcinoma. In preclinical tumor models, 6-methylmercaptopurine riboside (MMPR), an inhibitor of purine synthesis, elevates phosphoribosylpyrophosphate (PRPP) pools and promotes the anabolism of 5-FU to fluorinated nucleotides. In vivo, the addition of MMPR enhances the therapeutic efficacy of PALA-5-FU. In a phase I trial, we sought to determine the optimal dose and schedule of MMPR in combination with PALA (250 mg/m2 on day 1) and 5-FU (1300 mg/m2 by 24-hour infusion on day 2). MMPR (75-225 mg/m2) was given intravenously on day 1 to 27 patients with solid tumors (15 colorectal, seven breast, five other). Toxic effects were mild to moderate and included leukopenia, mucositis, nausea, or rash. Two of seven patients given MMPR at 225 mg/m2 had grade 3 diarrhea. PRPP was measured using a [14C]orotic acid 14CO2 release assay in tumor biopsy specimens obtained before and 12 hours and 24 hours after MMPR doses were given to 20 patients. The addition of MMPR elevated PRPP pools in human solid tumors. At 12 hours after treatment, two (50%) of four patients showed a twofold or greater elevation of PRPP at the MMPR dose level of 75 mg/m2; a similar elevation was observed in five (71%) of seven patients given 150 mg/m2 MMPR and in three (43%) of seven patients given 225 mg/m2 MMPR. At 24 hours after treatment, results for the respective dose levels of MMPR were two (33%) of six patients, one (20%) of five patients, and four (57%) of seven patients. Administration of the two highest MMPR dose levels appeared to result in a greater increase in tumor PRPP levels. However, toxicity was greater at the 225 mg/m2 dose level; therefore, the 150 mg/m2 dose level was favored. Tumor levels of PRPP decreased between 12 hours and 24 hours in nine (56%) of 16 patients. This time course indicates that MMPR should be administered at the beginning of the 24-hour infusion of 5-FU.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Methylthioinosine/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Biopsy , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation , Drug Interactions , Female , Fluorouracil/administration & dosage , Humans , Male , Middle Aged , Neoplasms/metabolism , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Phosphoribosyl Pyrophosphate/metabolism
12.
Med Oncol Tumor Pharmacother ; 5(2): 113-6, 1988.
Article in English | MEDLINE | ID: mdl-3412035

ABSTRACT

6-Methylmercaptopurine riboside (MMPR) and 5-fluorouracil (5-FU) were administered sequentially to 12 patients in a phase I clinical trial. Toxicities included mild nausea and vomiting, as well as reversible leukopenia and thrombocytopenia. Maximal accumulation of 6-methylmercaptopurine ribonucleoside 5'-monophosphate (MMPR-P), the active metabolite of MMPR, in patients' erythrocytes occurred between 2 and 6 h after the administration of MMPR and the degree of accumulation was dose-related. At 96 h after MMPR administration, MMPR-P was still detectable in patients' erythrocytes. Although no clinical responses were documented, a modified dosage schedule of these drugs should be pursued based on the pharmacokinetic data obtained.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Fluorouracil/administration & dosage , Inosine/analogs & derivatives , Methylthioinosine/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Evaluation , Drug Therapy, Combination , Female , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Humans , Male , Methylthioinosine/adverse effects , Methylthioinosine/pharmacokinetics , Middle Aged
14.
Cancer Chemother Pharmacol ; 13(2): 136-8, 1984.
Article in English | MEDLINE | ID: mdl-6205781

ABSTRACT

Fourty-four evaluable patients were treated with 6-methylmercaptopurine riboside (MMPR) at a dose of 20 mg/m2/day X 5 by continuous IV infusion (days 1-5 and 5-fluorouracil (5-FU) on an escalating dose schedule of 300-1519 mg/m2/day X 5 by continuous IV infusion (days 2-6). Dose-limiting oral mucositis occurred at a 5-FU dose of 1,381 mg/m2/day; other toxicities included nausea, vomiting, diarrhea, skin rash, and occasional myelosuppression. A partial and a complete response were observed in two previously untreated patients with metastatic colon carcinoma given the highest 5-FU doses (1,381 and 1,519 mg/m2/day). Bone marrow phosphoribosyl pyrophosphate (PRPP) levels monitored after 24 h of MMPR treatment indicated increases of 7.8- and 9.2-fold those found prior to therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Fluorouracil/administration & dosage , Inosine/analogs & derivatives , Methylthioinosine/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Evaluation , Female , Humans , Male , Middle Aged , Phosphoribosyl Pyrophosphate/analysis , RNA, Neoplasm/metabolism
15.
Cancer Lett ; 9(4): 305-11, 1980 Jun.
Article in English | MEDLINE | ID: mdl-7397683

ABSTRACT

Antileukemic activity of 5 kinds of sulfur-containing purine ribonucleosides were examined in the presence and absence of amphotericin B against L1210 in mice. Among these compounds, 6-methylthioninosine was potentiated by amphotericin B. 6-Methylthioinosine in combination with amphotericin B produced a 75% increase in the lifespans, which was greater than the increase in lifespans by 6-methylthioinosine (38%) or amphotericin B alone (2%). Antitumor effects of other sulfur-containing ribonucleosides, such as 6-thiocyanatoguanine, 6-thiocyanatopurine, 6-thiocyanatoinosine, and 6-methylthiopurine, were not augmented by amphotericin B.


Subject(s)
Amphotericin B/administration & dosage , Inosine/analogs & derivatives , Leukemia L1210/drug therapy , Methylthioinosine/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Drug Synergism , Male , Mice , Purine Nucleosides/administration & dosage , Thymidine/metabolism
16.
Cancer ; 45(5 Suppl): 1117-28, 1980 Mar 15.
Article in English | MEDLINE | ID: mdl-6986974

ABSTRACT

This review summarizes a body of information suggesting that proper metabolic modulation with certain metabolites can sensitize tumor cells to anti-metabolites, and others can de-sensitize (i.e. protect) normal cells from the toxicity of anti-metabolites. This new approach offers the possibility of increasing the selectivity of drug therapy, with the promise of a real advance in cancer chemotherapy. The metabolite thymidine (TdR), long used as a cell synchronizing agent, is known to exert this effect in vitro by metabolic modulation of a number of enzymes in the salvage pathway to DNA synthesis. Against this biochemical background, in vivo effects of TdR employed as an agent for cancer therapy are reviewed as follows: 1) TdR alone, and in combination with, 2) Methotrexate (MTX), or 3) 5-Fluorouracil (FU), or 4) Cytosine arabinoside (ara-C). TdR is shown in all instances either to protect against host toxicity (eg. MTX), or to potentiate the anti-tumor effect (eg. FU and ara-C). Findings are also presented that a sequential schedule of MTX prior to TdR prior to FU is important for the optimal therapeutic activity of these drugs. The biochemical basis for the MTX leads to FU augmentation is reportedly due to increased activation of FU by MTX (acting indirectely). On the basis of this biochemical insight, a completely different chemotherapeutic agent methyl-mercaptopurine raboside (MMPR) was substituted for MTX, resulting in a dramatic potentiation of anticancer activity. Metabolic modulation with still other metabolites (UR) and a hormone (testosterone) was demonstrated to protect from host toxicity due to certain anti-cancer agents without offsetting anti-tumor activity. The ability to prevent leukopenia by these means was particularly impressive. Clinical trials have been initiated with TdR alone, TdR + MTX, and TdR + FU; the available clinical data are summarized.


Subject(s)
Neoplasms/drug therapy , Thymidine/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Bone Marrow/drug effects , Clinical Trials as Topic , Cytarabine/administration & dosage , DNA, Neoplasm/biosynthesis , Drug Therapy, Combination , Fluorouracil/administration & dosage , Humans , Methotrexate/administration & dosage , Methylthioinosine/administration & dosage , Mice , Neoplasms/metabolism , Thymidine/administration & dosage , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...