Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
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.
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.
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
6.
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
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.
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
9.
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
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 ; 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
12.
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
13.
J Natl Cancer Inst ; 80(7): 496-501, 1988 Jun 01.
Article in English | MEDLINE | ID: mdl-3259266

ABSTRACT

Almost all of the completed and ongoing phase III trials of the leucovorin/5-fluorouracil (LV/5-FU) combination have used either a single-agent 5-FU control arm in which the 5-FU was administered in a different schedule from the LV/5-FU arm or one in which the 5-FU was not at the maximally tolerated dose (MTD). Because both dose intensity and scheduling are known to affect drug activity, the LV/5-FU combination was evaluated in the preclinical CD8F1 murine model of advanced first-passage spontaneous breast tumors using the same dose (at MTD) and schedule for 5-FU alone and in the LV/5-FU combination arm. Overall, therapy with 5-FU at MTD was not improved by LV. Further, although the activity of 5-FU doses lower than the MTD could be increased by LV, the therapeutic result was comparable to that of single-agent 5-FU at MTD. In an evaluation with other modulators of 5-FU (e.g., uridine, PALA, methotrexate), therapy with various modulated 5-FU combinations at their MTD was not improved with LV. In conclusion, although LV can enhance the cytotoxicity of 5-FU in these in vivo preclinical studies, it does not confer enhanced selectivity to 5-FU, a conclusion at odds with many present clinical reports. Whether or not these murine findings have clinical relevance can be determined only by clinical trials designed with the MTD of 5-FU alone in the control arm, the MTD of 5-FU (or as close as tolerated) in the LV/5-FU arm, and identical schedules in both arms.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/administration & dosage , Leucovorin/administration & dosage , Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Female , Mice , Uridine/administration & dosage
14.
J Orthop Res ; 4(3): 273-80, 1986.
Article in English | MEDLINE | ID: mdl-3734935

ABSTRACT

Secreted and intracellular arylsulfatase B (ASB) activities were measured in normal and osteoarthritic (OA) human chondrocyte cultures in the absence and presence of monensin, ammonium chloride, and chloroquine. Of the three agents added, only monensin produced a significant stimulation of secreted enzyme activity. Osteoarthritic cells consistently exhibited a three-fold higher level of secreted specific ASB activity than did normal cells, with or without monensin. When compared with normal cells, OA cells also consistently exhibited a twofold heightened intracellular specific enzyme activity both in the absence or presence of monensin. With increasing dosage of monensin, secreted and intracellular ASB activity increased for both OA and normal cells. Total enzyme activity of secreted and intracellular ASB was found to be cell density dependent. No inhibition of secreted or intracellular ASB activity was observed for sparsely plated cultures. In contrast to sparse cultures, an inhibition of secreted ASB, with or without monensin, was observed in densely plated cultures. Intracellular total activity was not inhibited by high-density cultures. Secreted ASB activity was found to be time-dependent after passage. Enzyme activity was maximal at 6 h in both OA and normal cells and decreased by the end of 24 h both in serum-free medium and in serum-free medium with monensin. When compared with normal cells, OA cells expressed higher levels of ASB activity under all test conditions. This heightened activity therefore appears to be a property inherent in the OA chondrocyte.


Subject(s)
Cartilage, Articular/enzymology , Chondro-4-Sulfatase/metabolism , Furans/pharmacology , Monensin/pharmacology , Sulfatases/metabolism , Aged , Ammonium Chloride/pharmacology , Cartilage, Articular/cytology , Cell Count , Cells, Cultured , Chloroquine/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Lysosomes/drug effects , Lysosomes/enzymology , Male , Middle Aged , Osteoarthritis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...