Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Int J Oncol ; 29(3): 665-72, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16865283

ABSTRACT

Melatonin has been shown to have oncostatic effects on malignant melanoma in vitro and in vivo. We studied the growth suppressive effects of melatonin over a wide range of concentrations in four melanoma cell lines (SBCE2, WM-98, WM-164 and SKMEL-188) representative for different growth stages and phenotype. Melanoma cells were incubated with melatonin 10(-12)-10(-3) M, and proliferation and clonogenicity was assessed at 12 h and 14 days, respectively. We also determined the expression of cytosolic quinone oxidoreductases NQO1, NQO2 (known as MT3 receptor) and nuclear receptor RORalpha by RT-PCR. Melatonin at pharmacological concentrations (10(-3)-10(-7) M) suppressed proliferation in all melanoma cell lines. In SKMEL-188 cells cultured in serum-free media, melatonin at low concentrations (10(-12)-10(-10) M) also slightly attenuated the proliferation. The effects of pharmacological doses of melatonin were confirmed in the clonogenic assay. Expression of NQO1 was detected in all cell lines, whereas NQO2 and nuclear receptor RORalpha including its isoform RORalpha4 were present only in SBCE2, WM-164 and WM-98. Thus, melatonin differentially suppressed proliferation in melanoma cell lines of different behaviour. The intensity of the oncostatic response to melatonin could be related to the cell-line specific pattern of melatonin cellular receptors and cytosolic binding protein expression.


Subject(s)
Free Radical Scavengers/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Melatonin/pharmacology , NAD(P)H Dehydrogenase (Quinone)/genetics , Quinone Reductases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Trans-Activators/genetics , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Culture Media, Serum-Free , Cytosol/metabolism , Humans , Melanoma/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1 , Quinone Reductases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay
2.
J Pineal Res ; 40(1): 18-26, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16313494

ABSTRACT

Melatonin is a potent antioxidant and direct radical scavenger. As keratinocytes represent the major population in the skin and UV light causes damage to these cells, the possible protective effects of melatonin against UV-induced cell damage in HaCaT keratinocytes were investigated in vitro. Cells were preincubated with melatonin at graded concentrations from 10(-9) to 10(-3) m for 30 min prior to UV irradiation at doses of 25 and 50 mJ/cm2. Biological markers of cellular viability such as DNA synthesis and colony-forming efficiency as well as molecular markers of apoptosis were measured. DNA synthesis was determined by [3H]-thymidine incorporation into insoluble cellular fraction, clonogenicity through plating efficiency experiments and apoptosis by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. DNA synthesis experiments showed a strong protective effect by preincubation with melatonin at concentrations of 10(-4) m (P < 0.01) and 10(-3) m (P < 0.001). Additional postirradiation treatment with melatonin showed no increase in the pre-UV incubation protective effect. These results indicate that preincubation is a requirement for melatonin to exert its protective effects. The mechanism of melatonin's protective effect (10(-6) to 10(-3) m) includes inhibition of apoptosis as measured by TUNEL assay. Moreover, the biological significance of these effects is supported by clonogenic studies showing a significantly higher number of colonies in cultures treated with melatonin compared to controls. Thus, pretreatment with melatonin led to strong protection against UVB-induced damage in keratinocytes.


Subject(s)
Apoptosis/drug effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Melatonin/pharmacology , Cells, Cultured , Dose-Response Relationship, Radiation , Humans , In Situ Nick-End Labeling , Keratinocytes/metabolism , Thymidine/metabolism , Ultraviolet Rays
3.
J Chemother ; 17(4): 435-40, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16167524

ABSTRACT

Rhodamine-123, a lipophilic, cationic, rhodocyanine dye, has been reported to have carcinoma selective toxicity in vitro and in vivo. This phase I clinical trial established the safety and pharmacokinetics of Rhodamine-123 administered to men with hormone refractory prostate cancer. A single dose toxicity study of Rhodamine-123 determined the maximum tolerated dose. A multiple dose toxicity study assessed the safety of Rhodamine-123 at the maximum tolerated dose level. Transient and variable toxicities noted following Rhodamine-123 infusion resolved within 6 hours following infusion. Pharmacokinetic analyses of sera showed no accumulation of drug with repeated monthly administrations. Drug retention was confirmed in prostatic tissue following Rhodamine-123 administration. PSA doubling times lengthened variably suggesting drug efficacy but the data were not statistically significant. The maximum tolerated dose of Rhodamine-123 is 96 mg/m2. The drug can be safely administered at monthly intervals without detectable drug accumulation in serum. Rhodamine-123 is retained by prostatic tumor tissue.


Subject(s)
Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Rhodamine 123/pharmacokinetics , Rhodamine 123/therapeutic use , Aged , Dose-Response Relationship, Drug , Drug Administration Schedule , Follow-Up Studies , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Patient Selection , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Rhodamine 123/adverse effects , Risk Assessment , Survival Analysis , Treatment Outcome
4.
J Med Chem ; 44(7): 1028-34, 2001 Mar 29.
Article in English | MEDLINE | ID: mdl-11297449

ABSTRACT

N-Benzyladriamycin-14-valerate (AD 198) is a semisynthetic anthracycline with experimental antitumor activity superior to that of doxorubicin (DOX). AD 198, unlike DOX, only weakly binds DNA, is a poor inhibitor of topoisomerase II, and circumvents anthracycline-resistance mechanisms, suggesting a unique mechanism of action for this novel analogue. The phorbol ester receptors, protein kinase C (PKC) and beta2-chimaerin, were recently identified as selective targets for AD 198 in vitro. In vitro, AD 198 competes with [3H]PDBu for binding to a peptide containing the isolated C1b domain of PKC-delta (deltaC1b domain). In the present study molecular modeling is used to investigate the interaction of AD 198 with the deltaC1b domain. Three models are identified wherein AD 198 binds into the groove formed between amino acid residues 6-13 and 21-27 of the deltaC1b domain in a manner similar to that reported for phorbol-13-acetate and other ligands of the C1 domain. Two of the identified models are consistent with previous experimental data demonstrating the importance of the 14-valerate side chain of AD 198 in binding to the C1 domain as well as current data demonstrating that translocation of PKC-alpha to the membrane requires the 14-valerate substituent. In this regard, the carbonyl of the 14-valerate participates in hydrogen bonding to the deltaC1b while the acyl chain is positioned for stabilization of the membrane-bound protein-ligand complex in a manner analogous to the acyl chains of the phorbol esters. These studies provide a structural basis for the interaction of AD 198 with the deltaC1b domain and a starting point for the rational design of potential new drugs targeting PKC and other proteins with C1 domains.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Caenorhabditis elegans Proteins , Doxorubicin/chemistry , Isoenzymes/chemistry , Protein Kinase C/chemistry , Receptors, Drug/chemistry , Carrier Proteins , Doxorubicin/analogs & derivatives , Models, Molecular , Protein Binding , Protein Kinase C-delta
5.
Drug Resist Updat ; 4(3): 169-77, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11768330

ABSTRACT

The anthracycline antibiotics doxorubicin (Adriamycin; DOX) and daunorubicin (DNR) continue to be essential components of first-line chemotherapy in the treatment of a variety of solid and hematopoietic tumors. The overall efficacies of DOX and DNR are, however, impeded by serious dose-limiting toxicities, including cardiotoxicity, and the selection of multiple mechanisms of cellular drug resistance. These limitations have necessitated the development of newer anthracyclines whose structural and functional modifications circumvent these impediments. In this review, we will present recent strategies in anthracycline design and assess their potential therapeutic merits. Current anthracycline design has diverged to target either cytoplasmic or nuclear sites. Nuclear targets have been broadened to include not only topoisomerase II (topo II) inhibition through ternary complex stabilization and catalytic inhibition, but also topoisomerase I (topo I) inhibition and transcriptional inhibition. In contrast, cytoplasmic targeting focuses on anthracycline binding to protein kinase C (PKC) regulatory domain with consequent modulation of activity.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/drug effects , Cytoplasm/drug effects , Drug Delivery Systems/methods , Animals , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cytoplasm/enzymology , Cytoplasm/metabolism , Humans
6.
Laryngoscope ; 110(12): 2026-32, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11129014

ABSTRACT

OBJECTIVES/HYPOTHESIS: With some advanced squamous cell carcinomas (SCCs) of the head and neck, chemoradiation therapy may obviate the need for surgical intervention. However, both modalities are known to produce organ toxicities, and tumor insensitivity remains problematic. Thus there is a clear need for the development of new treatment strategies. Accordingly, preclinical studies to evaluate the use of valrubicin, a contact-safe, mechanistically novel antitumor agent, combined with low-dose radiation for the therapy of SCC have been conducted. METHODS: The comparative in vitro antitumor activities of valrubicin with or without irradiation versus cisplatin were evaluated using human-derived sensitive and cisplatin-resistant SCC cell lines. A hamster cheek pouch model of SCC was used to assess the efficacy of weekly intratumoral valrubicin injections with and without concurrent low-dose irradiation. RESULTS: Valrubicin cytotoxicity was found to be comparable in both sensitive and platinum-resistant cell lines and superior to cisplatin. The addition of minimally cytotoxic cell irradiation (300-450 cGy) resulted in prolonged G2/M cell cycle arrest and a supraadditive increase in apoptotic cell death. In hamsters, once a week x 3 intratumoral drug injections (3, 6, or 9 mg) were growth inhibitory; however, when valrubicin (6 mg) was combined with minimally cytotoxic irradiation (150, 250, or 350 cGy) significant tumor shrinkage was observed. CONCLUSIONS: Valrubicin produces supra-additive effects against SCC when combined with low-dose irradiation. This effect appears to correlate with the ability of valrubicin, a cytoplasmic-localizing drug, to inhibit protein kinase C. Therapeutic use of valrubicin against SCC could provide for reduced radiation doses with consequent improved efficacy and reduction in host toxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Doxorubicin/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Animals , Antineoplastic Agents/administration & dosage , Combined Modality Therapy , Cricetinae , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Injections, Intralesional , Models, Animal , Tumor Cells, Cultured
7.
Biochem Pharmacol ; 60(11): 1621-8, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11077044

ABSTRACT

N-Benzyladriamycin (AD 288) is a highly lipophilic, semi-synthetic congener of doxorubicin (DOX). Unlike DOX, which stimulates double-stranded DNA scission by stabilizing topoisomerase II/DNA cleavable complexes, AD 288 is a catalytic inhibitor of topoisomerase II, capable of preventing topoisomerase II activity on DNA. The concentration of AD 288 required to inhibit the topoisomerase II-catalyzed decatenation of linked networks of kinetoplast DNA was comparable to that for DOX. However, AD 288 did not stabilize cleavable complex formation or stimulate topoisomerase II-mediated DNA cleavage. In addition, AD 288 inhibited the formation of cleavable complexes by etoposide in a concentration-dependent manner. Human CCRF-CEM cells and murine J774. 2 cells exhibiting resistance against DOX, teniposide, or 3'-hydroxy-3'-deaminodoxorubicin through reduced topoisomerase II activity remained sensitive to AD 288. These studies suggest that AD 288 inhibits topoisomerase II activity by preventing the initial non-covalent binding of topoisomerase II to DNA. Since AD 288 is a potent DNA intercalator, catalytic inhibition is achieved by prohibiting access of the enzyme to DNA binding sites. These results also demonstrate that specific substitutions on the aminosugar of DOX can alter the mechanism of topoisomerase II inhibition.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Topoisomerase II Inhibitors , Catalysis , DNA/drug effects , DNA/metabolism , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Drug Resistance, Neoplasm , Etoposide/pharmacology , Humans , Tumor Cells, Cultured
8.
Pediatr Dent ; 22(6): 489-93, 2000.
Article in English | MEDLINE | ID: mdl-11132509

ABSTRACT

PURPOSE: This study was performed to determine the bioavailability and local tissue toxicological safety of flumazenil (Romazicon) when administered by oral submucosal (SM) as opposed to intravenous (i.v.) injection. METHODS: Six dogs each received SM flumazenil (0.2 mg), and their serum was collected at predetermined time intervals (0-2 h) and frozen (-70 degrees C). Seven days later, the dogs received an identical dose of i.v. flumazenil, and serum samples were again collected, as above. Comparative quantitation of flumazenil levels (i.v. vs. SM) was made using a sensitive HPLC assay (UV detection). Direct/local drug toxicity was visually scored by unbiased raters of color photographs (test and control mucosa) taken at 1, 2, and 7 days following SM flumazenil injection. An oral pathologist examined slides processed from control and treatment tissues (hematoxylin and eosin staining) taken (punch biopsy) 1 week following SM injection to compare with direct clinical scores. RESULTS: Serum flumazenil levels reached a plateau (8.5 +/- 1.5 ng/mL, mean +/- SD) within 4 min of SM drug injection and declined thereafter to -2 ng/mL by 2 h. Bioavailability of SM flumazenil was 101 +/- 14%, based upon measuring the area under the serum concentration-time curves over 1.5 h (AUC 0-1.5 h, SM vs. i.v. drug). Thus, serum drug levels following SM drug administration were broadly comparable to those obtained during the elimination phase of corresponding i.v. drug delivery. Regarding drug tissue toxicity, no evidence of direct drug toxicity was observed by unbiased raters of color photographs (test and control mucosa) taken at 1, 2, and 7 days following SM flumazenil injection. Following pathologic review, no difference was seen in the degree of inflammation between treatment and control tissue. CONCLUSION: At the quantity and concentration used, SM drug flumazenil administration appears to be both a safe and a viable alternative to bolus i.v. drug delivery and worthy of further investigation.


Subject(s)
Flumazenil/pharmacokinetics , GABA Modulators/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Biopsy , Chromatography, High Pressure Liquid , Coloring Agents , Confidence Intervals , Dogs , Flumazenil/administration & dosage , Flumazenil/blood , Flumazenil/toxicity , Follow-Up Studies , GABA Modulators/administration & dosage , GABA Modulators/blood , GABA Modulators/toxicity , Injections, Intravenous , Male , Models, Animal , Mouth Mucosa/drug effects , Mouth Mucosa/pathology , Observer Variation , Safety
9.
Cancer Chemother Pharmacol ; 43(5): 419-26, 1999.
Article in English | MEDLINE | ID: mdl-10100599

ABSTRACT

PURPOSE: N-Benzyladriamycin-14-valerate (AD 198) is a semisynthetic anthracycline analogue superior to doxorubicin (DOX) both in vitro and in experimental rodent tumor models, and with differing mechanistic properties from those of the parental antibiotic agent. In the present study, we examined the metabolic fate and hematotoxicity of AD 198 in rats, with a view to determining whether some of the therapeutic properties observed for this drug might be due to a DOX prodrug effect. METHODS: Samples of plasma, bile and urine were obtained at various times following intravenous (i.v.) [14C]-AD 198 administration to rats and were analyzed by reversed-phase HPLC with flow-fluorescence detection and complementary liquid scintillography. In other animals, red blood cell and white blood cell (WBC) counts were determined for blood obtained by retrobulbar sampling on selected days from groups of animals receiving either AD 198 or DOX at several dose levels, as well as from vehicle controls. RESULTS: Following a single iv dose of [14C]-AD 198 (5 mg/kg; equivalent to the optimal murine antitumor dose) in anesthetized rats, a triphasic plasma decay pattern for parental drug was evident with extremely rapid alpha and beta phases followed by a very long terminal elimination phase. Principal plasma products included N-benzyladriamycin (AD 288) and N-benzyladriamycinol (AD 298) together with very low levels of DOX and doxorubicinol (DOXOL). Analysis of bile from anesthetized and conscious animals receiving AD 198 revealed DOX to be the principal biliary fluorescent species together with low levels of AD 288, AD 298 and DOXOL; no parental drug was seen. By contrast, AD 288 was the principal urinary product, together with low levels of AD 298 and DOX; again, no parental drug was evident. Dose recovery (8 h) in the respective bile and urine of anesthetized rats was 12.4% and 13.2% based upon total fluorescence versus 1% and 15.3% of the administered radiolabel. In conscious animals, 13.4% of drug fluorescence was recovered in the bile (48 h), while in urine 16.6% and 77.1% of drug fluorescence and radiolabel, respectively, were eliminated over 72 h. The discrepancy between recovery of drug fluorescence and 14C was due to the production of nonfluorescent hippuric acid (benzoylglycine) and N-benzyl daunosamine as a consequence of hepatic and renal drug metabolism. In the separate hematotoxicity studies, AD 198 (24.6 mg/ kg i.v.; equivalent to the murine LD50 dose), produced a 45% reduction (nadir day 3-5) in WBC count, with recovery by day 10. By contrast, DOX (10 mg/kg i.v.; equivalent to the mouse highest nonlethal dose) produced an 80% decline in WBC with only partial recovery by day 17. CONCLUSIONS: By virtue of the low systemic DOX levels and low hematotoxicity observed in rats receiving AD 198, the in vivo therapeutic superiority of AD 198 cannot be attributed to substantial intracellular DOX generation. The conclusion that the therapeutic superiority of AD 198 compared to DOX results from the mechanistic differences between these two drugs is further supported by recent observations on their biochemical differences with regard to protein kinase C and topoisomerase II inhibition.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Animals , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/toxicity , Doxorubicin/analogs & derivatives , Doxorubicin/metabolism , Doxorubicin/pharmacology , Doxorubicin/toxicity , Female , Injections, Intravenous , Male , Rats , Rats, Sprague-Dawley
10.
Oncol Res ; 10(4): 209-17, 1998.
Article in English | MEDLINE | ID: mdl-9778692

ABSTRACT

N-(2-Chloroethyl)-N-nitrosoureidodaunorubicin (AD 312), a novel semisynthetic compound with combined anthracycline and nitrosourea alkylating functionalities, circumvents resistance conferred by either reduced DNA topoisomerase II (topo II) or increased P-glycoprotein expression with less myelosuppression and cardiotoxicity than adriamycin (doxorubicin; ADR). Cellular resistance to AD 312 could arise from a novel mechanism that confers resistance to both functions simultaneously, or one or more mechanisms common to anthracyclines and/or alkylating agents. The mechanism contributing to AD 312 resistance was investigated following selection of AD 312-resistant murine J774.2 macrophage-like cells and human NCI-H460 non-small-cell lung carcinoma cells. Murine J/312-400 (> 4.7-fold) and human H/312-40 cells (6.3-fold) were cross-resistant to topo II inhibitors (ADR, teniposide, etoposide) and nitrosoureas (carmustine, lomustine) but remained sensitive to vinblastine, colchicine, and camptothecin. There was approximately a twofold decrease in topo II decatenation activity and protein. Decreased net intracellular drug accumulation was not observed. There were no increases in glutathione content or glutathione-S-transferase activity. Increased O6-methylguanine-DNA methyltransferase (MGMT) activity (2.3-fold) was detected in J/312-400, and AD 312 resistance was partially reversed by O6-benzylguanine, a potent inhibitor of MGMT activity. The results suggest that AD 312 resistance arose through selective pressure by both cytotoxic functions in a serial manner.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type II/metabolism , Nitrosourea Compounds/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/drug effects , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Cells, Cultured , Drug Resistance, Neoplasm , Glutathione/metabolism , Glutathione Transferase/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Macrophages/drug effects , Macrophages/enzymology , Mice , Tumor Cells, Cultured
11.
Anticancer Drugs ; 9(1): 58-66, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9491793

ABSTRACT

N-benzyladriamycin-14-valerate (AD 198) is pharmacologically superior to Adriamycin (ADR) based upon comparable cytotoxicity, decreased cardiotoxicity and the ability of AD 198 to circumvent multidrug resistance conferred by either P-glycoprotein overexpression or reduced topoisomerase II activity. AD 198, however, suffers from systemic lability of the 14-O-valerate moiety to enzymatic and non-enzymatic cleavage to yield N-benzyladriamycin (AD 288), which is more similar to ADR in activity. The purpose of this study was to determine whether stability of the ester linkage could be achieved while preserving the favorable characteristics of AD 198 by using a series of N-benzylated ADR congeners containing 14-O-acyl substitutions of incrementally shorter carbon chain lengths. Results from this study indicate that the linear five-carbon valerate substitution is the minimum length necessary to circumvent P-glycoprotein and prevent inhibition of topoisomerase II activity. In addition, although AD 198 is not a pro-drug of AD 288, intracellular 14-O-acyl cleavage appears to contribute to the cytotoxicity of AD 198.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Animals , Antibiotics, Antineoplastic/metabolism , Cell Line/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Structure-Activity Relationship
12.
Cancer Chemother Pharmacol ; 37(5): 472-8, 1996.
Article in English | MEDLINE | ID: mdl-8599871

ABSTRACT

Lipophilic N-alkylanthracyclines such as AD 198 (N-benzyladriamycin-14-valerate) or AD 201 [N,N-di(n-propyl)adriamycin-14-valerate], which exert their cytotoxicity through mechanisms which are not yet fully defined, possess inherent abilities to circumvent multidrug resistance in vitro and in vivo, possibly though alterations in normal intracellular drug trafficking. As part of structure-activity studies with this class of agent, we have now examined the pharmacology of AD 202 [N,N-di(n-butyl)adriamycin-14-valerate], another analog possessing superior antitumor activity to doxorubicin in vivo and an ability to circumvent multidrug resistance in vitro. Following the administration of AD 202 (20 mg/kg, i.v.) to anesthetized rats, rapid drug distribution (T1/2 5 min) was followed by more gradual elimination (T1/2 3.6h). Plasma clearance of AD 202 (224 +/- 63.6 ml/min per kg) and steady state volume of distribution (25.7 +/- 11.1 l/kg) were indicative of extensive tissue sequestration and/or a large degree of extra-hepatic metabolism. The parent drug predominated in plasma until 20 min, thereafter N,N-di(n-butyl)adriamycin became the principal circulating anthracycline. The systemic exposure to this biotransformation product (area under the plasma concentration-time curve from time zero to 480 min AUC(0-480) 28 1672 ng.min/ml) was > tenfold higher than for the other detected plasma products (N-butyladriamycin-14-valerate, N-butyladriamycin, and three unidentified fluorescent signals; P1-3). Total urinary elimination over 8h was limited (1.9% of dose), occurring predominantly as N,N-di(n-butyl)adriamycin (1.2% of dose), N-butyladriamycin (0.4% of dose), and their corresponding 13-carbinol metabolites (<0.1% of dose each). Low levels of adriamycin (ADR), aglycones and two unidentified products were also seen. Parental AD 202 was found in urine only up to 1h. By contrast, hepatic elimination of parent drug was seen, albeit at low levels, through 8h. Excretion by this route (22% of dose) occurred principally as N-butyl-adriamycin (8% of dose), N-butyladraimycinol (2.1% of dose) with lower levels of N,N-di(n-butyl)adriamycin (1.6% of dose), N,N-di(n-butyl)adriamycin (0.8% of dose), and aglycones (4.3% of dose, combined). Other products included ADR (1.1% of dose) and two unidentified signals (3.4% of dose, combined). The relatively poor mass balance in these studies is attributed to prolonged intracellular retention (elimination T1/2 24.2h) of N,N-di(n-butyl)adriamycin. Thus, in common with other N-alkylanthracyclines, the pharmacology of AD 202 is complex but its therapeutic properties clearly are not derived from an ADR prodrug effect. Significant differences continue to be noted as to the metabolic fate of congeners of this class of anthracycline analogs.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/analogs & derivatives , Animals , Antibiotics, Antineoplastic/blood , Antibiotics, Antineoplastic/metabolism , Bile/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Doxorubicin/blood , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Rats , Rats, Sprague-Dawley
13.
Anticancer Drugs ; 5(6): 623-33, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7888699

ABSTRACT

The novel anthracycline N-benzyladriamycin-14-valerate (AD 198) circumvents P-glycoprotein (P-gp)- and altered topoisomerase II-mediated drug resistance. Nevertheless, AD 198-resistant (AD 198R) murine J774.2 cells overexpressed P-gp, were cross-resistant to other drugs through reduced accumulation and were rendered sensitive by continuous exposure to verapamil. Intracellular AD 198 was, however, similar in sensitive and resistant cells. Consequently, the ability of P-gp to confer AD 198 resistance was examined. It was observed that (i) AD 198 resistance in AD 198R cells grown without drug for 15 months declined by 60% with only a 10-15% loss of vinblastine cross-resistance and P-gp expression; (ii) a cloned AD 198R P388 mouse leukemic cell line did not express P-gp; and (iii) verapamil did not attenuate resistance against high-dose, short-term exposure to AD 198. Therefore, AD 198 resistance appeared to be P-gp-independent despite P-gp overexpression. Antioxidant enzyme and topoisomerase II activities remained unchanged between sensitive and resistance cells. These results suggest that AD 198 resistance was conferred by a novel mechanism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , DNA Topoisomerases, Type II/analysis , Doxorubicin/analogs & derivatives , Affinity Labels , Animals , Azides , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Dihydropyridines , Dose-Response Relationship, Drug , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance , Immunoblotting , Mice , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Verapamil/pharmacology
14.
Oncol Res ; 5(6-7): 229-34, 1993.
Article in English | MEDLINE | ID: mdl-7907237

ABSTRACT

N-Benzyladriamycin-14-valerate (AD 198)-resistant murine J774.2 macrophage-like cells (A300) exhibited a novel mechanism of resistance in which P-glycoprotein was overexpressed without decreased AD 198 accumulation. Cross-resistance to Adriamycin (ADR), N-benzyladriamycin, and Adriamycin-14-valerate was due, at least in part, to reduced accumulation, suggesting that circumvention of P-glycoprotein-mediated transport was associated with extreme lipophilicity conferred by both substitutions. Thus, unlike multidrug resistance mediated by either P-glycoprotein, the multidrug resistance-associated protein (MRP), or decreased topoisomerase II activity, cross-resistance in A300 cells was highly structure-specific. In order to further characterize the specificity of AD 198 resistance, the cytotoxicity, accumulation, and intracellular localization of a series of 3'-morpholinyl, 3'-deamino and halogenated ADR congeners that have been reported to circumvent MDR was determined in AD 198-resistant J774.2 and P388 AD 198-resistant cells. Cross-resistance correlating with increased AD 198 resistance was observed for 2'-bromo-4'-epi-hydroxy-daunomycin (13-fold), morpholinyl doxorubicin (24-fold), and 4'-iodo-4'-deoxydoxorubicin (2.8-fold), but was attributable to decreased accumulation. Cross-resistance to 3'-hydroxy-14-O-palmitoyl-doxorubicin (6-fold) was not due to reduced accumulation. No cross-resistance was observed for the highly cytotoxic metabolite of WP474, 3'-hydroxyldoxorubicin (hydroxyrubicin; WP159), nor for the much less cytotoxic 3'-O-benzylated congeners, including 3'-O-benzyl-doxorubicin-14-valerate. These findings indicate that AD 198 resistance confers cross-resistance to compounds that, like AD 198, localize in the cytoplasm but are metabolized to highly cytotoxic, nuclear-localizing compounds.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Drug Resistance , Epirubicin/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Carrier Proteins/physiology , Doxorubicin/pharmacology , Membrane Glycoproteins/physiology , Mice , Structure-Activity Relationship , Tumor Cells, Cultured
15.
Cancer Res ; 52(12): 3409-17, 1992 Jun 15.
Article in English | MEDLINE | ID: mdl-1350753

ABSTRACT

N-Benzyladriamycin-14-valerate (AD 198) is a highly lipophilic analogue of Adriamycin with novel cytotoxic mechanisms, greater in vivo antitumor activity, and the ability to circumvent multidrug resistance due to P-glycoprotein-mediated drug efflux or decreased topoisomerase II activity. To identify the mechanism(s) which may confer AD 198 resistance, J774.2 mouse macrophage-like cells were selected for growth in cytotoxic levels of AD 198 (AD 198R). AD 198R cells exhibited over-expression of the mdr1b (P-glycoprotein) gene, cross-resistance to Adriamycin and vinblastine, and potentiation of drug cytotoxicity by verapamil. However, net intracellular accumulation of AD 198 in AD 198R cells was unchanged compared to parental cells, while Adriamycin and vinblastine accumulations were reduced 40% and 95%, respectively. AD 198 was localized in the perinuclear region of the cytoplasm in both parental and AD 198R cells, with additional vesicular compartmentalization in AD 198R cells. Verapamil-induced reversal of AD 198 resistance coincided with some drug redistribution from cytoplasmic vesicles, but without redistribution of AD 198 into the nucleus. These results suggest that AD 198 resistance was not conferred through a P-glycoprotein-mediated reduction in intracellular drug accumulation but through other cytoplasmic mechanisms, including, but not limited to, drug compartmentalization.


Subject(s)
Doxorubicin/analogs & derivatives , Macrophages/metabolism , Membrane Glycoproteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Biotransformation , Cells, Cultured , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Drug Resistance/genetics , Flow Cytometry , Mice , Microscopy, Fluorescence , RNA, Messenger/metabolism , Verapamil/pharmacology , Vinblastine/metabolism
16.
Oncol Res ; 4(8-9): 341-7, 1992.
Article in English | MEDLINE | ID: mdl-1362503

ABSTRACT

N-Benzyladriamycin-14-valerate (AD 198) is a highly hydrophobic analogue of Adriamycin (ADR) which can circumvent multidrug resistance (MDR) in various cell lines. Unlike ADR, AD 198 avoids extrusion by P-glycoprotein (P-gp) in AD 198-resistant murine macrophage-like J774.2 cells and localizes in the cytoplasm. To determine the structural modification(s) responsible for these different characteristics, intracellular accumulation and distribution of ADR, AD 198, and the two half-substituted AD 198 congeners. N-benzyladriamycin (AD 288) and adriamycin-14-valerate (AD 48), were analyzed in AD 198-sensitive (J774.2) and -resistant (A300) cells. A300 cells exhibited cross-resistance to and reduced accumulation of ADR, AD 48, and AD 288. ADR and AD 288 rapidly localized in the nuclei of parental and A300 cells, while AD 48 and AD 198 localized in the cytoplasm. AD 48 redistributed into nuclei and cytoplasm of both cell lines, but AD 198 maintained a punctate cytoplasmic distribution in A300 cells. These results suggest that both the N-benzyl and C14-valerate substitutions of AD 198 are required for P-gp circumvention and stable cytoplasmic localization in A300 cells, probably as a result of differing intracellular drug trafficking.


Subject(s)
Doxorubicin/analogs & derivatives , Drug Resistance , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Biotransformation , Carrier Proteins/metabolism , Cell Division/drug effects , Cytoplasm/metabolism , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Membrane Glycoproteins/metabolism , Mice , Microscopy, Fluorescence , Neoplasm Proteins/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Cancer Chemother Pharmacol ; 28(1): 1-6, 1991.
Article in English | MEDLINE | ID: mdl-2040028

ABSTRACT

Based on previous clinical findings following systemic administration, as well as appropriate laboratory evidence, the novel lipophilic anthracycline analogue N-Trifluoroacetyladriamycin-14-valerate (AD 32) has been identified as an agent of potential value in the intravesical therapy of superficial bladder carcinoma. Toward this end, using a rat model, the present study was designed to evaluate the potential for toxicity of a therapeutic dose of AD 32 given intravesically. With regard to systemic toxicity, following a single intravesical dose of AD 32 (20 mg/kg), the total systemic drug exposure (O-6 h), expressed as the area under the plasma concentration-time curve, was 14.2 micrograms min ml-1, or less than 1% of the corresponding value obtained when the identical dose was injected intravenously (2,392 micrograms min ml-1). In separate studies, a single intravenous dose of AD 32 (20 mg/kg) given to normal animals produced only a 20% reduction in white blood cell counts as compared with a 60% reduction following the administration of a therapeutic dose of Adriamycin (5 mg/kg); no effect was seen for either drug on red blood cell production. Taken together, these results imply that systemic drug exposure following the intravesical instillation of a therapeutic dose of AD 32 would result in negligible (approximately 0.2%) hematotoxic potential. Furthermore, intravesical instillation of AD 32 (20 mg/kg) at a concentration (10 mg/ml) greater than that projected for use in humans resulted in no evidence of contact toxicity to the rat bladder urothelium. Thus, based on experimental and clinical consideration of safety and efficacy, AD 32 appears to be an excellent candidate for the intravesical treatment of superficial bladder cancer.


Subject(s)
Doxorubicin/analogs & derivatives , Leukocytes/drug effects , Administration, Intravesical , Animals , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/blood , Doxorubicin/pharmacology , Doxorubicin/toxicity , Epithelium/drug effects , Female , Injections, Intravenous , Leukocyte Count , Rats , Rats, Inbred Strains , Urinary Bladder/drug effects
18.
Biochem Pharmacol ; 40(7): 1441-8, 1990 Oct 01.
Article in English | MEDLINE | ID: mdl-2222502

ABSTRACT

Adriamycin (ADR; doxorubicin) and its highly lipophilic, less toxic analogue N-benzyl-adriamycin-14-valerate (AD 198) were found to inhibit rat heart and liver carnitine palmitoyltransferases of both mitochondrial outer and inner membranes. The outer membrane enzyme was more sensitive to inhibition by these drugs than the inner membrane enzyme, and AD 198 was a more potent inhibitor of these enzymes than ADR. Other analogues of ADR, N-trifluoroacetyladriamycin-14-valerate (AD 32) and N-trifluoroacetyladriamycin-14-O-hemiadipate (AD 143), which are documented as being noncardiotoxic, were also more potent inhibitors of the mitochondrial carnitine palmitoyltransferases than ADR. Overall, the cardiac mitochondrial carnitine palmitoyltransferases seemed to be slightly more sensitive to the inhibitory effects of ADR and its analogues than the liver enzyme. ADR was an uncompetitive inhibitor with respect to palmitoyl-CoA and a noncompetitive inhibitor with respect to carnitine for both mitochondrial outer and inner membrane enzymes. Our data suggest that mitochondria can take up ADR and concentrate it within the matrix, as is known to happen with other positively-charged compounds. More ADR was found associated with the mitochondrial inner membrane than with the outer membrane; this could be due to the greater protein content of the inner membrane rather than drug binding to cardiolipin. Although inhibition of cardiac inner membrane carnitine palmitoyltransferase has been implicated previously as part of the cardiotoxicity mechanism of ADR, the present findings with ADR and its noncardiotoxic analogues do not support this view.


Subject(s)
Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Doxorubicin/pharmacology , Mitochondria, Heart/enzymology , Mitochondria, Liver/enzymology , Animals , Doxorubicin/analogs & derivatives , Intracellular Membranes/drug effects , Male , Mitochondria, Heart/drug effects , Mitochondria, Liver/drug effects , Rats , Rats, Inbred Strains
19.
Cancer Chemother Pharmacol ; 27(3): 205-10, 1990.
Article in English | MEDLINE | ID: mdl-2265456

ABSTRACT

Little is known of the pharmacology of rhodamine 123 (RH-123), an agent reported to have carcinoma-selective experimental antitumor activity. Accordingly, using a high-performance liquid chromatographic assay system with fluorescence detection, we examined the plasma decay and the biliary and urinary elimination of parent drug and metabolites in female Sprague-Dawley rats receiving RH-123 at an intravenous dose (5 mg/kg) equivalent to the therapeutic dose used in murine tumor models. Following drug administration to unconscious animals, plasma levels of drug-associated fluorescence fell in a triphasic manner (t1/2 alpha, 15 min; t1/2 beta, 1 h; t1/2 gamma, 4.7 h). In plasma, unchanged drug predominated but lower levels of the deacylated metabolite rhodamine 110 (RH-110) and two unknowns were also detectable throughout the study. Drug fluorescence was recovered extensively in both urine and bile. In unconscious animals with ureteral cannulae, urinary excretion (11.4% of the dose in 6 h) occurred predominantly as unchanged RH-123 (97% of the total), with low levels of RH-110 (2.4%) and two unknowns (less than 0.6% combined) also being present. Similarly dosed conscious animals (without surgical intervention) housed in metabolic cages showed a comparable pattern of urinary excretion, with 11.9% of the drug dose being recovered in 6 h and 21.9%, by 48 h. Biliary drug elimination accounted for 8% of the delivered dose in 6 h in unconscious animals and for 11% by 36 h in conscious animals fitted with biliary cannulae. In contrast to urinary excretion, in which unchanged drug predominated, only 50% of the fluorescence recovered in bile was attributable to RH-123. The remainder was due to a number of products that were detectable throughout the study. Of these, one present at significant levels was identified as a glucuronide conjugate of RH-123, based on the liberation of parent drug when the purified metabolite was incubated with beta-glucuronidase or hydrolyzed with 1 N hydrochloric acid. Further studies with a radiolabeled form of RH-123 are necessary to establish the identity of the remaining unknowns disclosed in this work.


Subject(s)
Fluorescent Dyes/metabolism , Rhodamines/metabolism , Animals , Bile/metabolism , Chromatography, High Pressure Liquid , Female , Fluorescent Dyes/pharmacokinetics , Glucuronates/metabolism , Rats , Rats, Inbred Strains , Rhodamine 123 , Rhodamines/pharmacokinetics
20.
Br J Cancer ; 60(6): 819-26, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2605093

ABSTRACT

N-Benzyladriamycin-14-valerate (AD198) is a novel lipophilic anthracycline with greater in vivo antitumour activity than doxorubicin (DOX) in experimental model systems. Using sensitive and progressively DOX-resistant L1210 mouse leukaemia and B16-BL6 mouse melanoma lines, we have determined the cellular pharmacokinetics and cytotoxic response in vitro and in vivo of AD198. In the L1210 leukaemia model following 3 h drug exposure in vitro, the IC50 for AD198 was approximately 0.35 microgram ml-1 for the sensitive and 10-fold DOX resistant cells and 1.0 microgram ml-1 for the 40-fold DOX resistant cells. A similar pattern of cross-resistance to AD198 was also observed with the B16-BL6 melanoma, with and IC50 for AD198 with the sensitive and 10-fold DOX-resistant cells being similar, and about 2-fold higher with the 40-fold resistant cells. In the L1210 leukaemia model, cellular pharmacokinetics of AD198 revealed the following: (a) accumulation of AD198 was concentration but not time dependent, and cellular drug levels in the sensitive and resistant sublines were similar when treated with equimolar concentrations; (b) retention of AD 198 was 60% of the initial drug uptake and, in cells treated with the IC50 of AD198, cellular levels in the 40-fold DOX-resistant line were, as expected, 2-fold higher than in sensitive or 10-fold DOX-resistant cells; (c) in vitro biotransformation of AD 198 in the sensitive and resistant sublines was comparable. Studies in vivo with i.p. L1210 leukaemia (disseminating) and B16-BL6 melanoma (non-disseminating) tumour models evaluating therapeutic efficacy of DOX vs AD 198 in mice implanted with tumour i.p. on day 0 and treated i.p. on days 1-4 indicated: (a) DOX at 3 mg kg-1 administered once daily on days 1-4 resulted in a 55% ILS and 104% ILS with parent-sensitive B16-BL6 melanoma and L1210 leukaemia models respectively; however, similar doses of DOX in the resistant sublines were ineffective, with survival similar to the untreated control; (b) AD198 at 10-12.5 mg kg-1 day-1 for 4 days was extremely effective in the sensitive L1210 (189% ILS), and similar to DOX (61% ILS) in the sensitive B16-BL6; (c) AD198 (10-12.5 mg kg-1) was ineffective (survival similar to untreated control) in the 10-and 40-fold DOX-resistant L1210 leukaemia and 40-fold DOX resistant B16-BL6 melanoma, but produced a 76% ILS in the 10-fold DOX resistant B16-BL6 melanoma.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/analogs & derivatives , Doxorubicin/therapeutic use , Leukemia L1210/drug therapy , Melanoma, Experimental/drug therapy , Animals , Biotransformation , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Resistance , Drug Screening Assays, Antitumor , Female , In Vitro Techniques , Leukemia L1210/metabolism , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred Strains , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...