Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 3(2): 309-15, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9815688

ABSTRACT

Uridine diphosphoglucose (UDPG) is a precursor of uridine that can be used as a rescuing agent from 5-fluorouracil (5FU) toxicity. Four doses of UDPG (2000 mg/kg i.p. or p.o. at 2, 6, 24, and 30 h after 5FU bolus) allowed the escalation of a weekly bolus of 5FU from 100 mg/kg (5FU100) to 150 mg/kg (5FU150) in healthy and tumor-bearing BALB/c, C57/BI, and CD8F1 (BALB/c x DBA/8) mice. 5FU150 without rescuing agents is not tolerated by the animals. When followed by UDPG, on the contrary, it is possible to increase the dose of 5FU even when it is modulated by leucovorin. Toxicity was the same for 5FU100 and 5FU150 + UDPG, and the nadir values (expressed as a percentage of pretreatment values) were 83 and 85% for weight, 45 and 45% for hematocrit, and 45 and 61% for leukocytes, respectively. Platelets were not affected by treatment. A protective effect was also shown for the gastrointestinal tract. The enzymes thymidine kinase, maltase, and sucrase were measured in the intestinal mucosa at different times after 5FU treatment with or without UDPG rescue. Even if the nadir values in enzyme activities were similar in mice receiving or not receiving UDPG, the pattern of recovery showed that cell repopulation was more rapid in the group treated with UDPG. 5FU150 + UDPG had enhanced antitumor activity against CD8F1 mammary carcinoma and against the resistant tumor Colon 26 (tumor doubling time 1.9 days for controls, 8.5 days for 5FU100, 13.7 days for 5FU150 + UDPG, and 15.9 days for 5FU150 + leucovorin + UDPG). We demonstrated that UDPG administered at 2, 24, and 30 h after 5FU100 does not reduce the antitumor activity of 5FU in two sensitive tumors (Colon 38 and Colon 26-10). In conclusion, UDPG is a promising rescuing agent for 5FU; it reduces the toxic side effects and increases the therapeutic index.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Fluorouracil/pharmacology , Protective Agents/pharmacology , Uridine Diphosphate Glucose/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/blood , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Digestive System/drug effects , Drug Interactions , Drug Screening Assays, Antitumor , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/blood , Leukocytes/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protective Agents/administration & dosage , Uridine Diphosphate Glucose/administration & dosage , Uridine Diphosphate Glucose/blood
3.
Cancer Chemother Pharmacol ; 40(5): 376-84, 1997.
Article in English | MEDLINE | ID: mdl-9272113

ABSTRACT

The drug combination N-(phosphonacetyl)-L-aspartic acid (PALA), methylmercaptopurine riboside (MMPR) and 6-aminonicotinamide (6AN), referred to as PMA, induces regressions of advanced CD8F1 murine mammary carcinomas in vivo. We demonstrated that CD8F1 tumor regressions were preceded by the appearance of apoptotic bodies, as observed by microscopic examination of morphology and TUNEL endlabeling, and fragmentation of DNA into nucleosomal "ladder" patterns. These indications of apoptosis were present as early as 6 h after simultaneous administration of MMPR and 6AN and further increased by over fivefold during the next 3 to 6 h, then remained at 7 to 12.8% (0.6 to 2.4% in saline-treated controls) of the cell population for at least 24 h after MMPR + 6AN administration. The 5'-phosphate derivative of MMRP, MMPR-5P, which inhibits de novo purine biosynthesis, was present at a "steady-state" level, and significant (40%) depletion of ATP had occurred by 3 h and both of these events preceded the onset of apoptosis. In addition, MMPR-5P was retained in CD8F1 tumors at a high level over a prolonged period (> 96 h) even as tumors were undergoing regression. The prolonged presence of MMPR-5P was important for optimal chemotherapeutic effect, since treatment with iodotubercidin (IodoT), an inhibitor of MMPR/adenosine kinase, 6 h after MMPR+6AN administration prevented the prolonged accumulation of MMPR-5P and reversed the regression of CD8F1 tumors. In addition, compared to the PMA-treated group, there was a significant restoration of ATP levels after treatment with IodoT. In individual PMA-treated CD8F1 tumors the degree of ATP depletion was found to correlate with the degree of tumor shrinkage at 24 h, after tumors had sufficient time to respond to treatment. These results define the time-course of drug-induced apoptosis in CD8F1 tumors, show that ATP depletion occurs prior to apoptosis and demonstrate that prolonged retention of MMPR-5P is associated with optimal chemotherapy. Collectively, these results suggest that the depletion of ATP by PMA treatment may be a component of the biochemical apoptotic cascade in the CD8F1 tumor.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , 6-Aminonicotinamide/administration & dosage , Animals , Antimetabolites, Antineoplastic/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Female , Mercaptopurine/administration & dosage , Mercaptopurine/analogs & derivatives , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Time Factors
4.
Cancer ; 78(9): 1988-95, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8909321

ABSTRACT

BACKGROUND: Dose intensification of 5-fluorouracil (5-FU) is complicated by increased toxicity. 5-FU is a fluorine-substituted pyrimidine analog of uracil. In preclinical studies, administration of oral uridine (Ur) has been shown to allow for dose intensification of 5-FU with enhancement of its antitumor activity. Therefore, a Phase I trial was designed aimed at dose intensification of 5-FU as a component of a modified 5-FU-doxorubicin-methotrexate (FAMTX) regimen using oral Ur rescue. METHODS: Methotrexate (MTX) was administered to all patients at a fixed dose of 1.5 g/m2. MTX was followed 24 hours later by escalating doses of 5-FU starting at 800 mg/m2 with leucovorin rescue. Cycles of 5-FU and MTX were repeated every 15 days. Every other cycle, patients received doxorubicin. "Adria cycles") at a dose of 30 mg/m2. Oral Ur was administered at a dose of 8 gm/m2 every 6 hours for 12 doses. In the first phase of the study, patients received Ur only if they developed Grade 3 or 4 hematologic toxicity. In the second phase, all patients received Ur 24 hours after 5-FU on all cycles. RESULTS: Without Ur rescue, the maximum tolerated dose (MTD) of 5-FU was 900 mg/m2 on the Adria cycles and 1.1 gm/m2 on the non-Adria cycles. With Ur, the MTD of 5-FU increased to 1.2 gm/m2 on the Adria cycles and to 1.6 gm/m2 on the non-Adria cycles. CONCLUSIONS: In this modified FAMTX regimen, oral Ur administration allowed for dose-intensification of 5-FU, with a 33% increase in the MTD of 5-FU on the Adria cycles and a 45% increase in the MTD of 5-FU dose on the non-Adria cycles.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antidotes/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Leucovorin/administration & dosage , Leukopenia/chemically induced , Leukopenia/prevention & control , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Middle Aged , Neutropenia/chemically induced , Neutropenia/prevention & control , Uridine/administration & dosage
5.
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
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.
Cancer Chemother Pharmacol ; 37(3): 229-34, 1996.
Article in English | MEDLINE | ID: mdl-8529282

ABSTRACT

Inhibition of pyrimidine and purine synthesis has been demonstrated to potentiate 5-fluorouracil (5-FU) activity in preclinical models. Low-dose phosphonacetyl-L-aspartate (PALA) potentiates the incorporation of 5-FU into RNA, without detectably increasing its toxicity. 6-Methylmercaptopurine riboside (MMPR) results in inhibition of purine biosynthesis with elevation of phosphoribosyl pyrophosphate (PRPP), which in turn is believed to increase the phosphorylation and intracellular retention of 5-FU. We conducted a phase I clinical trial to determine the maximum tolerated dose of 5-FU in combination with low-dose PALA and a biochemically-optimized dose of MMPR. The regimen consisted of PALA 250 mg/m2 given on day 1, followed 24 h later by MMPR 150 mg/m2, and escalating doses of 5-FU from 1625 to 2600 mg/m2 by 24 h continuous infusion. This regimen was repeated weekly. A group of 29 patients with a diagnosis of malignant solid tumor were entered; their median performance status was 1. The dose-limiting toxicity was mucositis, while other gastrointestinal toxicity was minimal. Two patients also experienced ischemic chest pain during the 5-FU infusion. The maximum tolerated dose of 5-FU in this combination was 2600 mg/m2. Several responses were observed including a complete remission in a previously treated breast cancer patient and two partial responses in breast and colon cancer. MMPR pharmacokinetics were obtained from urine analyses in 21 patients on this trial; there was no correlation between the pharmacokinetics of MMPR and the toxicity observed. This regimen was well tolerated and phase II trials are warranted using PALA 250 mg/m2, MMPR 150 mg/m2, and 5-FU 2300 mg/m2 by continuous infusion over 24 h.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Aspartic Acid/analogs & derivatives , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy , Phosphonoacetic Acid/analogs & derivatives , Thioinosine/analogs & derivatives , Thionucleotides/pharmacology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacokinetics , Aspartic Acid/pharmacology , Female , Fluorouracil/therapeutic use , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/metabolism , Phosphonoacetic Acid/pharmacology , RNA, Neoplasm , Thioinosine/pharmacokinetics , Thioinosine/pharmacology , Thionucleotides/pharmacokinetics
8.
Anticancer Drugs ; 7(1): 100-4, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8742105

ABSTRACT

A three-drug combination, PMA, consisting of (phosphonacetyl)-L-aspartic acid + 6-methylmercaptopurine riboside + 5-aminonicotinamide, preceding either 5-fluorouracil (5-FU) or adriamycin (Adr), produced tumor-regressing activity in a murine advanced breast tumor model not attainable with either 5-FU or Adr as single agents, or with any lesser combination of these drugs administered at maximally tolerated doses. Marked tumor-regressing activity was further increased significantly by using 5-FU and Adr together in conjunction with the modulatory biochemical conditioning (particularly ATP depletion) provided by pretreatment with PMA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Doxorubicin/administration & dosage , Fluorouracil/administration & dosage , Mammary Neoplasms, Animal/drug therapy , Mercaptopurine/administration & dosage , Mercaptopurine/analogs & derivatives , Mice , Niacinamide/administration & dosage , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives
9.
Oncol Rep ; 3(2): 317-22, 1996 Mar.
Article in English | MEDLINE | ID: mdl-21594366

ABSTRACT

An inhibitor of poly (ADP-ribose) polymerase, 1,5-dihydroxyisoquinoline (DHIQ), evaluated in vivo against a murine advanced breast cancer, significantly improved by 20% the PR rate of tumor-regressing chemotherapy. A detailed sequential biochemical cascade is proposed for chemotherapy-induced apoptosis, and the rationale for the utilization of the inhibitor is explained.

10.
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
11.
Int J Oncol ; 7(6): 1401-4, 1995 Dec.
Article in English | MEDLINE | ID: mdl-21552979

ABSTRACT

A positive correlation was found between increasing ATP depletion and enhanced tumor-regressing activity by combining PMA, a three drug combination (PALA, MMPR+6-AN), with FUra, or Adria, or FUra+Adria. For example, at 48 h post treatment, ATP was 68% of saline controls with a 10% tumor regression rate (PMA); ATP was 55% with a 60% regression rate (PMA+FUra); ATP was 54% with an 80% regression rate (PMA+Adria); and ATP was 30% with a;100% regression rate (PMA+FUra+Adria). The results give support to the suggestion that ATP depletion may be a significant factor in the production of chemotherapeutically-induced apoptosis.

12.
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
13.
Cancer Res ; 53(15): 3518-23, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-8339257

ABSTRACT

The combination of N-(phosphonacetyl)-L-aspartate, 6-methylmercaptopurine, and 6-aminonicotinamide has been shown to be an effective antineoplastic regimen and also to enhance the effects of other chemotherapeutic agents. The mechanism of action of this combination of drugs is not known definitively, but one possible mechanism is biochemical modulation of energy metabolism and inhibition of production of tumor ATP. Tumor-bearing mice were treated with N-(phosphonacetyl)-L-aspartate, followed 17 h later by 6-methylmercaptopurine and 6-aminonicotinamide. 31P nuclear magnetic resonance spectroscopic studies demonstrated a significant depletion of high energy phosphates at 10 h post-6-methylmercaptopurine and 6-aminonicotinamide. The addition of radiation at this time was shown to induce a significantly longer tumor growth delay and a greater number of regressions (including durable complete regressions) than either chemotherapy or radiation alone. The combination of chemotherapy and radiation was found to be supra-additive compared to the antineoplastic effects of either modality administered separately, without a measurable increase in host toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms, Experimental/therapy , 6-Aminonicotinamide/administration & dosage , Animals , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Combined Modality Therapy , Female , Mercaptopurine/administration & dosage , Mercaptopurine/analogs & derivatives , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives
14.
Cancer ; 71(5): 1875-81, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8448752

ABSTRACT

BACKGROUND: In a previous trial in which methotrexate and N-phosphonacetyl-L-aspartate (PALA) were used to modulate 5-fluorouracil (5-FU), four of six patients could not tolerate treatment at the 600 mg/m2 5-FU dose level because of mucositis, diarrhea, and a decrease in performance status. The current study examines the ability of uridine rescue to prevent such toxic effects in the same regimen and, thereby, allow additional dose escalation of 5-FU. METHODS: Twenty-nine patients with advanced malignant neoplasms received PALA and MTX, each at 250 mg/m2, followed 24 hours later by increasing bolus doses of 5-FU (600-750 mg/m2) with a leucovorin rescue (10 mg orally every 6 hours for eight doses) and uridine rescue (3 g/m2/hour, for a 72-hour infusion, 3 hours on, 3 hours off). Treatment was repeated weekly with either 2 weeks on, 2 weeks off, or 3 weeks on, 1 week off. RESULTS: Mucositis, which occurred in 4 of 12 patients treated at the 750 mg/m2 5-FU dose level, was the only significant chemotherapy-induced toxic effect. However, uridine-related central venous catheter complications (cellulitis in six patients and superior vena cava syndrome in one patient) precluded additional treatment on this protocol. CONCLUSIONS: In the current regimen, uridine allowed dose escalation of 5-FU to 750 mg/m2, which some patients tolerated on a 3-week on, 1-week off schedule. Because of the vascular toxic effects associated with intravenous uridine, the authors recommend additional studies with oral uridine to determine whether the increase in 5-FU dose that uridine allows is associated with improved response rates.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/adverse effects , Neoplasms/drug therapy , Uridine/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Diarrhea/chemically induced , Diarrhea/prevention & control , Drug Synergism , Female , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Male , Methotrexate/administration & dosage , Middle Aged , Mouth Mucosa , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Stomatitis/chemically induced , Stomatitis/prevention & control , Uridine/blood
15.
Cancer ; 70(7): 1988-92, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1381991

ABSTRACT

BACKGROUND: 5-Fluorouracil (5-FU) has modest activity as a single agent in a number of human adenocarcinomas. The technique of biochemical modulation has been used preclinically to increase the activity of 5-FU. METHODS: With doses based on a Phase I study, the authors performed a Phase II trial in patients with advanced metastatic adenocarcinoma of an unknown primary site using N-phosphonacetyl-l-aspartate (PALA), methotrexate (MTX), 5-FU, and leucovorin. In some patients, 5-phosphoriosyl-l-pyrophosphate (PRPP) and uridine triphosphate (UTP) metabolite pools were assayed before and after PALA and MTX to assess metabolite pool shifts. RESULTS: Twenty-one patients were treated in this Phase II trial. Toxicity was tolerable. Biopsy specimens of tumor tissue showed increases in PRPP and decreases in UTP levels in two of three and three of four patients, respectively. However, only one objective response was seen, which is not different from that expected with 5-FU alone. CONCLUSIONS: Expected PRPP and UTP metabolite pool shifts were seen when biochemical modulation was performed with these drugs according to this schedule. Because toxicity was mild, more intense dose schedules of 5-FU are planned for future studies.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms, Unknown Primary/drug therapy , Adenocarcinoma/metabolism , Adult , Antimetabolites, Antineoplastic/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Drug Evaluation , Female , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Male , Methotrexate/administration & dosage , Middle Aged , Neoplasms, Unknown Primary/metabolism , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Phosphoribosyl Pyrophosphate/metabolism , Uridine Triphosphate/metabolism
16.
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
17.
J Natl Cancer Inst ; 84(13): 1033-8, 1992 Jul 01.
Article in English | MEDLINE | ID: mdl-1376779

ABSTRACT

BACKGROUND: Increased response rates in studies of patients with colon cancer have indicated that the cytotoxic effects of fluorouracil (5-FU) are potentiated by leucovorin (LV) and by methotrexate (MTX). However, preliminary studies using a sequential combination of MTX, LV, and 5-FU showed no additional potentiation. PURPOSE: We hypothesized that the lack of additional cell kill with this combination could be due to competition of LV with MTX for cellular uptake and reduced folate polyglutamylation. We have tested this possibility by comparing the cytotoxicity of drug combinations containing MTX with that of drug combinations containing trimetrexate (TMTX), an antifolate that does not compete with LV for uptake or polyglutamylation. METHODS: Human lymphocytic leukemia CCRF-CEM cells were exposed to MTX or TMTX for 24 hours and to 5-FU during the last 4 hours of antifolate exposure. LV was administered 30 minutes before 5-FU. RESULTS: After 20 hours of exposure to TMTX or MTX, intracellular levels of phosphoribosyl pyrophosphate were elevated to a similar degree, and these levels did not decrease after a 30-minute exposure to LV. No additional cell kill was observed when LV was added to the MTX/5-FU combination, but cytotoxicity was enhanced when LV was added to the TMTX/5-FU combination. CONCLUSIONS: This study supports the hypothesis that the lack of additional cell kill when high-dose LV is added to the MTX/5-FU combination may be due to competition of MTX with LV for cellular uptake and/or competition of MTX or its polyglutamates with polyglutamylation of reduced folates. Inasmuch as TMTX does not compete with LV and reduced folates for uptake and polyglutamylation, the synergy obtained with the combination of TMTX plus 5-FU and high-dose LV further supports this hypothesis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Lymphoid/drug therapy , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Leucovorin/pharmacokinetics , Leukemia, Lymphoid/metabolism , Methotrexate/administration & dosage , Phosphoribosyl Pyrophosphate/metabolism , Trimetrexate/administration & dosage , Tumor Cells, Cultured
18.
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
19.
Pharm Res ; 6(10): 863-6, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2481854

ABSTRACT

The purpose of the present investigation was to determine whether a single bolus intravenous injection (2000 mg/kg) of uridine diphosphoglucose (UDPG) could affect levels of PRPP in a transplanted mammary adenocarcinoma and in liver of CD8FI mice. Six hours following a single intravenous injection of UDPG, 2000 mg/kg, tumor PRPP was lowered to 80 pmol/mg protein, a 53% decrease compared to saline control tumors. Liver was more sensitive than tumor to the 5-phosphoribosyl pyrophosphate (PRPP)-depleting effects of a single bolus intravenous injection of UDPG, since significantly lower levels of PRPP were found in liver, but not in tumor, at doses of 500-1000 mg/kg of UDPG. Maximal depression (30% of saline control) or PRPP occurred in liver 6 hr after intravenous UDPG at 1000-2000 mg/kg. Enhanced levels of UDPG in plasma (half-life less than 10 min) and tumor was detected at 30 min after intravenous UDPG at 2000 mg/kg. There was no detectable increase in endogenous levels of UDPG in liver at this time, probably as a result of rapid metabolism of UDPG by liver. At this same time, a twofold increase in uridine triphosphate (UTP) was measured in liver after intravenously administered UDPG. In contrast, the level of UTP was not increased significantly above control values in tumor. These data suggest the potential use of UDPG to elevate UTP pools in normal tissues in the delayed rescue of cancer chemotherapeutic drugs such as 5-fluorouracil which function as a uridine analogue in these tissues.


Subject(s)
Pentosephosphates/metabolism , Phosphoribosyl Pyrophosphate/metabolism , Uridine Diphosphate Glucose/pharmacology , Uridine Diphosphate Sugars/pharmacology , Uridine Triphosphate/metabolism , Animals , Chromatography, High Pressure Liquid , Liver/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasms, Experimental/metabolism , Orotic Acid
20.
Cancer Res ; 49(16): 4636-9, 1989 Aug 15.
Article in English | MEDLINE | ID: mdl-2472883

ABSTRACT

Based on an animal model to improve the antitumor activity of 5-fluorouracil (FUra), a Phase I study of N-(phosphonacetyl)-L-aspartate, methotrexate, FUra, and leucovorin was conducted on 44 patients. Methotrexate was given in an intermediate dose (250 mg/m2) to overcome potential drug resistance, and N-(phosphonacetyl)-L-aspartate was given at a low dose (250 mg/m2) in order to allow escalation of FUra to toxicity. These two drugs were given 24 h before FUra to enhance maximal incorporation of FUra into RNA. Two schedules of administration were used; one every other week and one weekly for 2 weeks. The every other week schedule was well tolerated, with minimal gastrointestinal and hematological toxicity. However, the weekly for 2 weeks schedule was more toxic with increased mucositis, diarrhea requiring therapy, and decreased performance status of 20% in 4 of 6 patients. There were no responders in the every other week schedule. There was one partial response and three patients with stable disease in four evaluable patients on the weekly for 2 weeks schedule. At 24 h post-N-(phosphonacetyl)-L-aspartate-methotrexate treatment, PRPP levels were doubled in bone marrow biopsies, and increased 2.5- to 25-fold in tumor biopsies. We have currently added uridine rescue to this combination with the hope of further escalating the dose of FUra.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Colorectal Neoplasms/drug therapy , Head and Neck Neoplasms/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/administration & dosage , Aspartic Acid/administration & dosage , Aspartic Acid/analogs & derivatives , Bone Marrow/analysis , Carcinoma, Squamous Cell/pathology , Colorectal Neoplasms/pathology , Drug Administration Schedule , Drug Evaluation , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/analysis , Fluorouracil/antagonists & inhibitors , Head and Neck Neoplasms/pathology , Humans , Leucovorin/therapeutic use , Male , Methotrexate/administration & dosage , Pentosephosphates/blood , Phosphonoacetic Acid/administration & dosage , Phosphonoacetic Acid/analogs & derivatives , Phosphoribosyl Pyrophosphate
SELECTION OF CITATIONS
SEARCH DETAIL
...