Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancer Chemother Pharmacol ; 69(4): 871-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22045498

ABSTRACT

PURPOSE: A phase I study was performed to determine the maximal tolerated dose (MTD), recommended dose (RD), safety and efficacy of vinflunine when combined with capecitabine in patients with metastatic breast cancer (MBC) previously treated with anthracyclines and taxanes, with pharmacokinetic blood sampling to test potential drug-drug interactions. PATIENTS AND METHODS: Sixteen patients with MBC who had received anthracyclines and taxanes in the neo/adjuvant setting, if progression occurred during or within 12 months of chemotherapy completion, and/or as first-line chemotherapy of MBC were enrolled. Vinflunine (VFL) was given on day 1 with capecitabine (CAPE) twice daily from days 1 to 14, every 3 weeks. Three dose levels (DL) were investigated (DL1: VFL 280 mg/m² and CAPE 1,650 mg/m²/day, DL2: VFL 320 mg/m² and CAPE 1,650 mg/m²/day and DL3: VFL 280 mg/m² and CAPE 2,000 mg/m²/day). RESULTS: The RD was established as vinflunine 280 mg/m² on day 1 plus capecitabine 1,650 mg/m²/day on days 1 to 14 given every 3 weeks. Dose-limiting toxicities were grade 4 neutropenia lasting at least 7 days for 2 patients, anorexia with fatigue for 1 patient and diarrhoea with fatigue, anorexia and febrile neutropenia for 1 patient. Neutropenia was the main toxicity of the combination, it was reported in 15 patients (93.8%) with grade 3 in 7 patients (43.8%) and 22.6% of cycles and grade 4 in 7 patients (43.8%) and 19.8% of cycles. Complications were rare with only one patient experiencing febrile neutropenia at DL exceeding the RD. The most frequent non-haematological toxicities were fatigue and gastrointestinal disorders; however, no grade 3 or 4 episode was observed at the RD. Hand-foot syndrome was reported in 5 patients (31.3%) and 22.6% of cycles, no episode of grade 3 was seen. Concerning pharmacokinetics, no modifications were detected for VFL, while slight accumulation between days 1 and 14 was observed for 5-FU formed from CAPE. The risk of clinical significant drug-drug interaction was considered weak. Objective partial responses were reported in 7 patients, yielding a response rate of 43.8% in the all-treated population according to the investigator assessment. CONCLUSIONS: The combination of vinflunine and capecitabine is safe and showed promising antitumour activity in MBC patients who have failed prior anthracyclines and taxanes. Further clinical development of this combination is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Adult , Aged , Anthracyclines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Capecitabine , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/analogs & derivatives , Fluorouracil/pharmacokinetics , Humans , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Taxoids/pharmacology , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinblastine/pharmacokinetics
2.
J Lipid Res ; 53(2): 282-91, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22058426

ABSTRACT

Synthesis inhibition is the basis for the treatment of type 1 Gaucher disease by the glucosylceramide synthase (GCS) inhibitor eliglustat tartrate. However, the extended use of eliglustat and related compounds for the treatment of glycosphingolipid storage diseases with CNS manifestations is limited by the lack of brain penetration of this drug. Property modeling around the D-threo-1-phenyl-2-decanoylamino-3-morpholino-propanol (PDMP) pharmacophore was employed in a search for compounds of comparable activity against the GCS but lacking P-glycoprotein (MDR1) recognition. Modifications of the carboxamide N-acyl group were made to lower total polar surface area and rotatable bond number. Compounds were screened for inhibition of GCS in crude enzyme and whole cell assays and for MDR1 substrate recognition. One analog, 2-(2,3-dihydro-1H-inden-2-yl)-N-((1R,2R)-1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-1-hydroxy-3-(pyrrolidin-1-yl)propan-2-yl)acetamide (CCG-203586), was identified that inhibited GCS at low nanomolar concentrations with little to no apparent recognition by MDR1. Intraperitoneal administration of this compound to mice for 3 days resulted in a significant dose dependent decrease in brain glucosylceramide content, an effect not seen in mice dosed in parallel with eliglustat tartrate.


Subject(s)
Brain/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glucosylceramides/metabolism , Glucosyltransferases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Brain/metabolism , Cells, Cultured , Dioxanes/chemical synthesis , Dioxanes/pharmacology , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical/methods , Indans/chemical synthesis , Indans/pharmacology , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Morpholines/chemistry , Vinblastine/pharmacokinetics
3.
Expert Opin Drug Saf ; 10(4): 645-53, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21524237

ABSTRACT

INTRODUCTION: Vinca alkaloid agents have been widely used in several different types of malignancies. However, cancer cells, ultimately, develop resistance to these agents. Therefore, the development of new agents with improved efficacy is warranted. Recently, a new synthetic vinca alkaloid, vinflunine, was developed through the addition of two fluor molecules by superacidic chemistry. AREAS COVERED: The authors describe the development of the new vinca alkaloid vinflunine from preclinical studies to the late-stage clinical trials, highlighting the most important clinical and safety data of vinflunine. In vitro and in vivo studies have shown a superior efficacy of vinflunine over other vinca alkaloids and with an improved safety profile. Early clinical trials have demonstrated a significant activity of vinflunine against different malignancies. Phase III trials showed that vinflunine increases survival in patients with advanced transitional cell carcinoma of the urothelium (TCCU) tract treated in the second-line and is as effective as docetaxel in second-line NSCLC. EXPERT OPINION: Vinflunine is currently approved in Europe for the treatment of second-line TCCU and is currently being developed in other malignancies. It has been shown to have predictable and manageable adverse effects, such as neutropenia, anemia, constipation and fatigue.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/chemically induced , Vinblastine/analogs & derivatives , Vinca Alkaloids/therapeutic use , Animals , Clinical Trials as Topic/methods , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Vinblastine/adverse effects , Vinblastine/pharmacokinetics , Vinblastine/therapeutic use , Vinca Alkaloids/adverse effects , Vinca Alkaloids/pharmacokinetics
4.
Nutr Cancer ; 61(4): 538-43, 2009.
Article in English | MEDLINE | ID: mdl-19838926

ABSTRACT

The effect of macelignan, a phytoestrogen, on P-gp function was investigated using multidrug resistant cancer cells overexpressing P-gp (NCI/ADR-RES) and the fluorescent P-gp substrates, daunorubicin and rhodamine 123. Macelignan (40 microM) increased the cellular accumulation of daunorubicin by approximately threefold in NCI/ADR-RES cells, whereas it did not alter the cellular accumulation of daunorubicin in MCF-7/sensitive cells. Similarly, the presence of macelignan also enhanced significantly (P < 0.05) the cellular accumulation of rhodamine 123 in a concentration-dependent manner in NCI/ADR-RES cells. Furthermore, cancer cells were more susceptible to the cytotoxicity of vinblastine, a P-gp substrate, in the presence of macelignan. Those results suggest that macelignan has inhibitory effects on P-gp mediated cellular efflux. However, P-gp activity did not affect the cellular accumulation of macelignan itself. Taken all together, macelignan was identified as a novel inhibitor of P-gp activity and may be a promising lead compound for the rational design of more efficacious drugs to reverse multidrug resistance in cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Lignans/pharmacology , Adenocarcinoma , Analysis of Variance , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biological Transport, Active/drug effects , Breast Neoplasms , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/pharmacokinetics , Dose-Response Relationship, Drug , Drug Design , Drug Resistance, Multiple , Drug Synergism , Female , Fluorescent Dyes/pharmacokinetics , Growth Inhibitors/pharmacokinetics , Growth Inhibitors/pharmacology , Humans , Lignans/pharmacokinetics , Phytoestrogens/pharmacokinetics , Phytoestrogens/pharmacology , Phytotherapy , Rhodamine 123/pharmacokinetics , Vinblastine/pharmacokinetics
5.
Clin Cancer Res ; 14(6): 1625-32, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18347163

ABSTRACT

Vinflunine (Javlor) is the first fluorinated microtubule inhibitor belonging to the Vinca alkaloids family. Vinflunine is obtained by semisynthesis using superacidic chemistry to selectively introduce two fluorine atoms at the 20' position of the catharanthine moiety. This compound has been selected for clinical development on the basis of encouraging preclinical activity that warrants study in patients with a wide spectrum of solid tumors. Clinically significant activity has been seen in phase II studies, mainly in the treatment of transitional cell carcinoma of the urothelial tract, non-small cell lung cancer, and carcinoma of the breast. Vinflunine is currently in phase III trial assessment in patients with (second line) transitional cell carcinoma of the urothelium and first-line advanced breast cancer. The efficacy of vinflunine in patients with advanced non-small cell lung cancer previously treated with a platinum-containing regimen was confirmed by a large phase III trial.


Subject(s)
Neoplasms/drug therapy , Tubulin Modulators/therapeutic use , Vinblastine/analogs & derivatives , Animals , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/pathology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Lung Neoplasms/drug therapy , Models, Biological , Neoplasms/metabolism , Treatment Outcome , Tubulin Modulators/adverse effects , Tubulin Modulators/pharmacokinetics , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Urothelium/pathology , Vinblastine/adverse effects , Vinblastine/pharmacokinetics , Vinblastine/therapeutic use
6.
Int J Pharm ; 348(1-2): 70-9, 2008 Feb 04.
Article in English | MEDLINE | ID: mdl-17719191

ABSTRACT

A lipid microsphere vehicle for vinorelbine (VRL) was designed to reduce the severe venous irritation caused by the aqueous intravenous formulation of VRL. Lipid microspheres (LMs) were prepared by high pressure homogenization. The physical stability was monitored by the appearance, particle size and zeta potential changes while the chemical stability was achieved by using effective antioxidants and monitored by long-term investigations. Safety tests were performed by testing rabbit ear vein irritation and a guinea pig hypersensitivity reaction. A pharmacokinetic study was performed by determining the drug levels in plasma up to 24h after intravenous administration of VRL-loaded LMs and conventional VRL aqueous injection separately. The VRL-loaded LMs had a particle size of 180.5+/-35.2nm with a 90% cumulative distribution less than 244.1nm, while the drug entrapment efficiency was 96.8%, and it remained stable for 12 months at 6+/-2 degrees C. The VRL-loaded LMs were less irritating and toxic than the conventional VRL aqueous injection. The pharmacokinetic profiles were similar and the values of AUC(0-t) were very close for the two formulations. A stable and easily mass-produced VRL-loaded LM preparation has been developed. It produces less venous irritation and is less toxic but has similar pharmacokinetics in vivo to the VRL aqueous injection currently commercially available.


Subject(s)
Lipids/chemistry , Microspheres , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Drug Hypersensitivity/etiology , Drug Stability , Ear/blood supply , Emulsions , Guinea Pigs , Hydrogen-Ion Concentration , Injections, Intravenous , Lecithins/chemistry , Male , Particle Size , Pharmaceutical Vehicles , Poloxamer/chemistry , Polysorbates/chemistry , Pressure , Rabbits , Rats , Rats, Wistar , Triglycerides/chemistry , Veins/drug effects , Veins/pathology , Vinblastine/chemistry , Vinblastine/pharmacokinetics , Vinorelbine
7.
Br J Cancer ; 97(8): 1071-6, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17912246

ABSTRACT

The purpose of this study was to investigate the utility of plasma pharmacokinetic and pharmacodynamic measures including plasma deoxynucleosides, homocysteine and methylmalonic acid concentrations in understanding the time course and extent of the inhibition of thymidylate synthase (TS) by pemetrexed in the context of a phase I/II combination study with vinorelbine. Eighteen patients received supplementation with folic acid and Vitamin B(12) 1 week before beginning treatment with pemetrexed and vinorelbine administered in a dose-escalating manner on a 21-day cycle. Heparinised blood samples were collected from consenting patients in the first cycle for pharmacokinetic analyses and in the first two cycles for determination of plasma thymidine, deoxyuridine, homocysteine and methylmalonic acid concentrations. These values were correlated with response and toxicity. Plasma deoxyuridine was used as a measure of TS inhibition, and concentrations of deoxyuridine were significantly elevated relative to baseline on days 1 (P<0.01), 2 (P<0.001) and 3 (P<0.05) after treatment at all pemetrexed dose levels (400-700 mg m(-2)). The magnitude of deoxyuridine elevation correlated with pemetrexed area under the plasma concentration-time curve (AUC) (r(2)=0.23, P<0.05). However, deoxyuridine concentrations returned to baseline between 8 and 15 days after treatment with pemetrexed, suggesting that inhibition of TS was not durable. Pemetrexed AUC correlated with the percentage decline (relative to baseline) in both platelets (r(2)=0.58, P<0.001) and leucocytes (r(2)=0.26, P<0.05) at day 8. Baseline homocysteine was also significantly correlated with these measures of haematological toxicity (r(2)=0.37, P<0.01 and r(2)=0.39, P<0.01, respectively). In addition, there was a significant reduction of plasma homocysteine on days 8 (P<0.005) and 15 (P<0.05) in cycle 1 compared to baseline values. The results suggest that the TS inhibitory effects of pemetrexed are short-lived and make the case for a more frequent schedule of administration such as every 2 weeks. The lack of protracted TS inhibition may be due to concomitant vitamin administration, and this may be the mechanism by which vitamins prevent life-threatening toxicity from pemetrexed. Baseline homocysteine concentration remains a predictive marker for haematological toxicity even following folate supplementation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Glutamates/administration & dosage , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Deoxyuridine/blood , Dietary Supplements , Dose-Response Relationship, Drug , Female , Folic Acid/therapeutic use , Glutamates/adverse effects , Glutamates/pharmacokinetics , Guanine/administration & dosage , Guanine/adverse effects , Guanine/pharmacokinetics , Homocysteine/blood , Humans , Male , Methylmalonic Acid/blood , Middle Aged , Pemetrexed , Thymidine/blood , Thymidylate Synthase/drug effects , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinblastine/pharmacokinetics , Vinorelbine , Vitamin B 12/therapeutic use
8.
Planta Med ; 72(7): 634-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16739070

ABSTRACT

Overexpression of the protein transporter P-glycoprotein (Pgp, MDR1) at the cell surface is a major cause of multidrug resistance (MDR) and poor response to treatment in cancer chemotherapy and therapy for leishmaniasis. The present study shows that conferone, a sesquiterpene coumarin ether isolated for the first time from Ferula schtschurowskiana, endemic in Uzbekistan, enhances the cell toxicity of vinblastine (VBL) in MDR1-transfected Madin-Darby canine kidney (MDCK-MDR1) cells. Conferone presents the advantage to mediate this effect at safe concentrations. At 10 microM, it efficiently competes with the photoactivatable cyclosporin A analogue (SDZ 212 - 122) for the binding to Pgp and accumulates [3H]-VBL to a higher extent than cyclosporin A or cnidiadin. [3H]-VBL accumulation is dose-dependent and correlates with the inhibition of Pgp photolabeling affinity, supporting the hypothesis that conferone sensitizes MDCK-MDR1 cells to VBL by competitively inhibiting drug efflux. In MDCK-MDR1 cells, [3H]-VBL accumulation appears to be almost completely dependent on inhibition of Pgp transport. However, the strict specificity of conferone to this efflux pump has to be demonstrated in cell lines expressing other protein transporters. Collectively, our findings identify conferone as a powerful modulator of Pgp transport and a promising molecule for the treatment of MDR malignancies and leishmaniasis. Complementary in vitro and in vivo studies are, however, needed to assess the value of conferone as a reversal drug in human therapy. Considering its high affinity for Pgp, conferone may have an additional usefulness as a tool for the design or the (hemi)synthesis of agents probing Pgp. To our knowledge, this is the first report identifying sesquiterpene coumarins from Ferula as possible drug candidates for the reversion of MDR encoded by the MDR1 gene or the synthesis of agents probing Pgp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adjuvants, Pharmaceutic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Coumarins/pharmacology , Ferula/chemistry , Vinblastine/pharmacology , Adjuvants, Pharmaceutic/isolation & purification , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Binding, Competitive , Biological Transport/drug effects , Cell Line , Coumarins/isolation & purification , Cyclosporine , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Genes, MDR , Humans , Photoaffinity Labels , Tritium , Vinblastine/pharmacokinetics
9.
Lung Cancer ; 49(3): 401-12, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15923057

ABSTRACT

PURPOSE: Pemetrexed and vinorelbine are active antineoplastic agents in non-small cell lung cancer (NSCLC). Phase I objectives include maximum tolerated dose (MTD) and recommended phase II dose determination, and pharmacokinetics of the pemetrexed-vinorelbine doublet in locally advanced or metastatic solid tumor patients (pts). Phase II objectives include tumor response evaluation, efficacy, and toxicity for first-line treatment of advanced NSCLC. EXPERIMENTAL DESIGN: Phase I pts received pemetrexed (day 1, 300-700 mg/m2) and vinorelbine (days 1 and 8, 15-30 mg/m2) every 21 days. Pharmacokinetics determined at cycle 1. Beginning with dose-level 3, folic acid and Vitamin B12 supplementation were given. RESULTS: Thirty-one phase I pts were enrolled. MTD was pemetrexed 700 mg/m2 and vinorelbine 30 mg/m2; and recommended phase II dose was pemetrexed 500 mg/m2 and vinorelbine 30 mg/m2. When administered in combination, pemetrexed and vinorelbine pharmacokinetics were consistent with single-agent administration. Thirty-seven (36 chemonaive) phase II NSCLC pts received pemetrexed-vinorelbine. Evaluable tumor response was 40%, with intent-to-treat 38%. One drug-related death occurred from febrile neutropenia with Staphylococcal infection. Grade 3/4 hematologic toxicities were neutropenia (65%) and febrile neutropenia (11%), while prevalent grade 3/4 non-hematologic toxicity was fatigue (27%). CONCLUSION: The pemetrexed-vinorelbine combination is well tolerated and shows activity as first-line treatment in advanced NSCLC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Glutamates/administration & dosage , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Vinblastine/analogs & derivatives , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Creatinine/metabolism , Dietary Supplements , Female , Glutamates/pharmacokinetics , Guanine/administration & dosage , Guanine/pharmacokinetics , Humans , Male , Maximum Tolerated Dose , Middle Aged , Pemetrexed , Time Factors , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/pharmacokinetics , Vinorelbine , Vitamin B 12/pharmacology
10.
Br J Pharmacol ; 143(7): 856-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15504753

ABSTRACT

We investigated the effects of grapefruit juice (GFJ) and orange juice (OJ) on drug transport by MDR1 P-glycoprotein (P-gp) and multidrug resistance protein 2 (MRP2), which are efflux transporters expressed in human small intestine. We examined the transcellular transport and uptake of [(3)H]vinblastine (VBL) and [(14)C]saquinavir in a human colon carcinoma cell line (Caco-2) and in porcine kidney epithelial cell lines transfected with human MDR1 cDNA and human MRP2 cDNA, LLC-GA5-COL150, and LLC-MRP2, respectively. In Caco-2 cells, the basal-to-apical transports of [(3)H]VBL and [(14)C]saquinavir were greater than those in the opposite direction. The ratio of basal-to-apical transport to apical-to-basal transport of [(3)H]VBL and [(14)C]saquinavir by Caco-2 cells was reduced in the presence of MK571 (MRPs inhibitor), verapamil (P-gp inhibitor), cyclosporin A (inhibitor of both), 50% ethyl acetate extracts of GFJ and OJ, or their components (6',7'-dihydroxybergamottin, bergamottin, tangeretin, hepatomethoxyflavone, and nobiletin). Studies of transport and uptake of [(3)H]VBL and [(14)C]saquinavir with MDR1 and MRP2 transfectants showed that VBL and saquinavir are transported by both P-gp and MRP2. GFJ and OJ components inhibited the transport by MRP2 as well as P-gp. However, their inhibitory potencies for P-gp or MRP2 were substrate-dependent. The present study has revealed that GFJ and OJ interact with not only P-gp but also MRP2, both of which are expressed at apical membranes and limit the apical-to-basal transport of VBL and saquinavir in Caco-2 cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Beverages , Citrus/chemistry , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Algorithms , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biological Transport, Active/drug effects , Caco-2 Cells , Calcium Channel Blockers/pharmacology , Cyclosporine/pharmacology , Food-Drug Interactions , HIV Protease Inhibitors/pharmacokinetics , Humans , Immunosuppressive Agents/pharmacology , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Plant Extracts/chemistry , Propionates/pharmacology , Quinolines/pharmacology , Saquinavir/pharmacokinetics , Transfection , Verapamil/pharmacology , Vinblastine/pharmacokinetics
11.
Pharm Res ; 19(6): 773-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12134946

ABSTRACT

PURPOSE: To investigate whether Madin-Darby canine kidney cells transfected with the human MRP2 gene (MDCK-MRP2) are a good model of the human intestinal mucosa. METHODS: MRP2 expression in Caco-2 cells was compared with the expression of this efflux transporter in MDCK-wild type (MDCK-WT) and MDCK-MRP2 cells using Western blotting methods. The polarized efflux activities of MRP2 in the MDCK-MRP2, MDCK-WT. MDCK cells transfected with the human MDR1 gene (MDCK-MDR1), and Caco-2 cells were compared using vinblastine as a substrate. Apparent Michaelis-Menten constants (K(M), Vmax) for the efflux of vinblastine in Caco-2 and MDCK-MRP2 cells were determined in the presence of GF120918 (2 microM), which inhibits P-glycoprotein but does not affect MRP2. Apparent inhibitory constants (K(I)) of known substrates/inhibitors of MRP2 were determined by measuring their effects on the efflux of vinblastine in these cell lines. RESULTS: MDCK-MRP2 cells expressed higher levels of MRP2 than MDCK-WT and Caco-2 cells as measured by Western blotting technique. Two isoforms of MRP2 expressed in Caco-2 and MDCK cells migrated at molecular weights of 150 kD and 190 kD. In MDCK-MRP2 cells, the 150 kD isoform appeared to be overexpressed. MDCK-MRP2 cell monolayers exhibited higher polarized efflux of vinblastine than Caco-2 and MDCK-WT cell monolayers. K(M) values for vinblastine in Caco-2 and MDCK-MRP2 cells were determined to be 71.8+/-11.6 and 137.3+/-33.6 microM. respectively, and Vmax values were determined to be (0.54+/-0.03 and 2.45+/-0.31 pmolcm(-2)s(-1), respectively. Known substrates/inhibitors of MRP2 showed differences in their ability to inhibit vinblastine efflux in Caco-2 cells as compared to MDCK-MRP2 cells CONCLUSIONS: These data suggest that MDCK-MRP2 cells overexpress only the 150 kD isoform of MRP2. The 190 kD isoform, which was also found in Caco-2 cells and MDCK-WT cells, was present in MDCK-MRP2 cells but not over expressed. The apparent kinetics constants and affinities of some MRP2 substrates were different in Caco-2 cells and MDCK-MRP2 cells. These differences in substrate activity could result from differences in the relative expression levels of the MRP2 isoforms present in Caco-2 cells and MDCK-MRP2 cells and/or differences in the partitioning of substrates in these two cell membrane bilayers.


Subject(s)
Cell Line/metabolism , Intestinal Mucosa/metabolism , Mitochondrial Proteins , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Transfection/methods , Animals , Caco-2 Cells/metabolism , Cell Line/physiology , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Multidrug Resistance-Associated Protein 2 , Ribosomal Proteins/genetics , Vinblastine/pharmacokinetics
12.
J Pharmacol Toxicol Methods ; 45(1): 71-7, 2001.
Article in English | MEDLINE | ID: mdl-11489667

ABSTRACT

Combinatorial synthesis and high-throughput pharmacology screening have greatly increased compound throughput in modern drug-discovery programs. For CNS drugs, it is also important to determine permeability to the blood--brain barrier. Yet, given the increased pace of discovery, it difficult to conduct this screen in a timely fashion. In this presentation, we describe several improvements to an existing CNS permeability screen, the bovine brain microvessel endothelial cell (BBMEC) model. By implementation of these incremental process improvements, we have achieved a robust, facile screen for determination of CNS permeability of multiple compounds.


Subject(s)
Blood-Brain Barrier , Brain/blood supply , Drug Evaluation, Preclinical/methods , Endothelium, Vascular/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biological Transport , Caffeine/pharmacokinetics , Cattle , Central Nervous System Agents/pharmacokinetics , Drug Combinations , Mannitol/pharmacokinetics , Microdialysis , Models, Biological , Permeability , Vinblastine/pharmacokinetics
13.
Br J Pharmacol ; 130(6): 1369-77, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10903978

ABSTRACT

1. The presence of inhibitors of drug efflux transporters, such as P-glycoprotein (P-gp), in grapefruit juice (GFJ) was confirmed based on the uptake of [(3)H]-vinblastine (VBL) by Caco-2 cells. 2. The uptake of [(3)H]-VBL by Caco-2 cells was significantly increased by the ethyl acetate extract of GFJ as well as by cyclosporin A. The extract was separated on a Cosmosil column and the eluate with 60% methanol increased [(3)H]-VBL uptake, while the activity to inhibit CYP3A4 was greatest in the 70 and 80% eluates. 3. These results show that the major inhibitor of efflux transport of VBL is different from that of CYP3A4. 4. Further separation of the 60% methanol eluate afforded dihydroxybergamottin (DHBG). Both ethyl acetate extract of GFJ and DHBG increased steady-state [(3)H]-VBL uptake by LLC-GA5-COL300 cells. Besides DHBG, other furanocoumarins contained in GFJ, such as bergamottin, FC726, bergaptol and bergapten, increased the steady-state uptake of [(3)H]-VBL by Caco-2 cells. 5. The order of inhibitory potency of these compounds was FC726>DHBG>bergamottin>bergapten>bergaptol . While, the IC(50) values for inhibition of CYP3A4 were 0.075, 0.45, 1.0, 1.0 and >20 microM, respectively. Bergaptol specifically inhibited VBL efflux. 6. DHBG was thus identified as a candidate for inhibitors of VBL transport, together with other furanocoumarins. Moreover, partly involvement of the P-gp inhibition was suggested. 7. Therefore, the inhibition of efflux transport of drugs as well as of drug metabolism by CYP3A4 could be an important cause of drug-GFJ interaction.


Subject(s)
Citrus/chemistry , Coumarins/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Vinblastine/pharmacokinetics , 3-O-Methylglucose/pharmacokinetics , 5-Methoxypsoralen , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acetates , Animals , Caco-2 Cells , Carbon Radioisotopes , Cell Line , Cell Line, Transformed , Coumarins/chemistry , Cyclosporine/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Furocoumarins/pharmacology , Humans , Hydroxylation/drug effects , Magnetic Resonance Spectroscopy , Methoxsalen/analogs & derivatives , Methoxsalen/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Mixed Function Oxygenases/metabolism , Phenylalanine/pharmacokinetics , Plant Extracts/chemistry , Plant Extracts/pharmacology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Testosterone/metabolism , Tritium
14.
Biol Pharm Bull ; 21(10): 1062-6, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9821810

ABSTRACT

We investigated the effect of components in grapefruit juice (GFJ) on the transport of vinblastine, a substrate of P-glycoprotein (P-gp), across Caco-2 cells. The apical to basolateral flux of [3H]vinblastine was increased in the presence of GFJ extracts. The steady-state uptake of [3H]vinblastine from the apical side was significantly increased in the presence of GFJ in a dose-dependent manner within the range of 2.5 to 50% (v/v) of GFJ. Although naringin and naringenin reduced apical efflux of [3H]vinblastine at the concentration present in GFJ and increased steady-state uptake from the apical side to 124 and 240%, respectively, the observed effect of naringin was not enough to account for the effect of GFJ and naringenin is not naturally present in GFJ. To investigate the effective components in GFJ, we examined the inhibitory effect of several organic solvent extracts of GFJ on the transport of [3H]vinblastine in Caco-2 cells. Organic solvent extracts of GFJ enhanced the apical to basolateral transcellular transport and inhibited the apical efflux. The permeability coefficient of apical to basolateral transport of [3H]vinblastine increased in the order of the ethyl acetate>diethyl ether>methylene chloride extracts of GFJ. Since the extracted amount of naringenin by ethyl acetate was less than that with the other organic solvents, the primary inhibitor in GFJ is suggested to be different from this flavonoid. The present study demonstrated the existence of inhibitory components in GFJ for the P-gp function in Caco-2 cells, which are distinct from known components such as naringin or naringenin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacokinetics , Beverages , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Citrus , Flavanones , Plant Extracts/pharmacology , Vinblastine/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antioxidants/pharmacology , Biological Transport/drug effects , Caco-2 Cells/enzymology , Cell Survival/drug effects , Cyclosporine/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Flavonoids/pharmacology , Food-Drug Interactions , Humans , Intestinal Absorption/drug effects , Mixed Function Oxygenases/metabolism , Tritium
15.
Clin Cancer Res ; 4(6): 1563-6, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626478

ABSTRACT

We analyzed the effect of high temperature (a 1-h incubation at 43 degrees C) on the accumulation and cytotoxicity of vinblastine and docetaxel in two model cell lines, K562 and MESSA, and their multidrug resistance (MDR) counterparts, K562/R7 and MESSA/Dx5. High temperature increased the amount of intracellular vinblastine and docetaxel significantly in MESSA cell and, to a much lesser extent, in K562 cells. MDR-positive cells retained a profound drug accumulation defect at 43 degrees C. Hyperthermia enhanced the cytotoxic effect of vinblastine (but not docetaxel) in both K562 and MESSA cells, but not in the MDR-positive variants. PSC833, a potent modulator of P-glycoprotein, induced high levels of drug accumulation in the two MDR-positive cell lines at both 37 degrees C and at 43 degrees C. PSC833 also significantly reduced the resistance levels of the two MDR-positive lines at both 37 degrees C and at 43 degrees C. The effect of hyperthermia on drug accumulation thus seems to depend on the cell line, whereas the effect on cytotoxicity depends on the type of compound. The MDR phenotype remains a therapeutic obstacle at 43 degrees C but is accessible to modulation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Drug Resistance, Multiple/physiology , Paclitaxel/analogs & derivatives , Taxoids , Vinblastine/pharmacokinetics , Vinblastine/toxicity , Antineoplastic Agents, Phytogenic/toxicity , Biological Transport , Cell Division/drug effects , Cell Survival/drug effects , Docetaxel , Humans , Hyperthermia, Induced , Leukemia, Erythroblastic, Acute , Paclitaxel/pharmacokinetics , Paclitaxel/toxicity , Sarcoma , Temperature , Tumor Cells, Cultured
16.
Arch Biochem Biophys ; 350(2): 340-7, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9473310

ABSTRACT

The intracellular concentration of many steroids and xenobiotics is influenced by the membrane protein P-glycoprotein (Pgp). It has been inferred that the intracellular retention of many drugs that upregulate Pgp or modulate Pgp function might also be affected by Pgp. However, the ability of Pgp to influence the translocation of these drugs needs to be established to understand Pgp's influence upon their pharmacological effect. We utilized two approaches to determine the interaction of several agents with Pgp: (a) an in vitro system, LLC-PK1 cell lines and derivative LLC cell lines stably expressing on the apical membrane either mouse mdr1a or human MDR1 Pgp grown as polarized epithelium in transwell culture to measure translocation of radiolabeled drugs; and (b) an in vivo system, mdr1a nullizygous and wild-type animals, to compare the contribution of Pgp to in vivo distribution of radiolabeled drugs. In combination these complementary approaches identified erythromycin as a drug whose intracellular retention is influenced by Pgp, while the intracellular accumulation and tissue distribution of retinoic acid and benzo(a)pyrene were unaffected by Pgp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP Binding Cassette Transporter, Subfamily B , ATP-Binding Cassette Transporters/physiology , Benzo(a)pyrene/pharmacokinetics , Erythromycin/pharmacokinetics , Tretinoin/pharmacokinetics , Animals , Biological Transport/physiology , Cell Line , Gene Expression/genetics , Humans , Mice , Mice, Knockout , Transfection/genetics , Vinblastine/pharmacokinetics
17.
Cancer Chemother Pharmacol ; 35(5): 391-6, 1995.
Article in English | MEDLINE | ID: mdl-7850920

ABSTRACT

Carboplatin is an alternative for cisplatin in the treatment of urothelial cancers. A pharmacologically guided phase I study of carboplatin in combination with methotrexate (30 mg/m2) and vinblastine (4 mg/m2) was conducted in ten patients by increment of the area under the plasma concentration versus time curve (AUC) for ultrafilterable carboplatin using the Calvert formula. The maximal tolerated AUC was 5 mg ml-1 min, with neutropenia being the dose-limiting toxicity. There was a significant linear correlation between the percentage of decrease in neutrophil count and the carboplatin AUC. Determination of the glomerular filtration rate by the isotopic method allowed us to adapt the dose of carboplatin given to patients suffering from urothelial cancer, who frequently have impaired renal function. The recommended AUC for phase II study is 4 mg ml-1 min.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Urologic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carboplatin/administration & dosage , Carboplatin/adverse effects , Carboplatin/pharmacokinetics , Cell Count/drug effects , Edetic Acid/metabolism , Female , Glomerular Filtration Rate/drug effects , Humans , Infusions, Intravenous , Kidney Neoplasms/drug therapy , Linear Models , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/pharmacokinetics , Middle Aged , Neutropenia/chemically induced , Neutrophils/drug effects , Thrombocytopenia/chemically induced , Ultrafiltration , Ureteral Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/pharmacokinetics
18.
Oncol Res ; 6(2): 59-70, 1994.
Article in English | MEDLINE | ID: mdl-7949466

ABSTRACT

Multidrug resistance (MDR) is the phenomenon in which cells become resistant to several classes of structurally and functionally diverse drugs after exposure to a single cytotoxic agent. One form of MDR is associated with the overexpression of a large plasma membrane phosphoglycoprotein, P-glycoprotein, which acts as an energy-requiring drug transport pump. Protein kinase C may participate in MDR through posttranslational modification of P-glycoprotein. The purpose of this study is to critically evaluate P-glycoprotein as a substrate for protein kinase C and to determine whether phosphorylation leads to changes in drug transport. Protein kinase C from rat brain phosphorylated immunoprecipitated P-glycoprotein in a manner dependent on the activation of the exogenous kinase. Phorbol 12-myristate 13-acetate (PMA) increased the phosphorylation of P-glycoprotein 6-fold and selectively decreased the accumulation of vinblastine in resistant MCF-7/AdrR cells. PMA selectively decreased the cellular association of vinblastine with MDR cells after brief periods of incubation, but only after critical concentrations of drug were achieved. The actions of PMA did not require new synthesis of P-glycoprotein. PMA had similar effects in MCF-7/BC-19, a cell line transfected with a cDNA for P-glycoprotein. Staurosporine inhibited the effects of PMA on the phosphorylation of P-glycoprotein and on the accumulation of vinblastine. These data demonstrated that immunoprecipitated P-glycoprotein can be a substrate for protein kinase C, and that phosphorylation of the transporter is associated with significant changes in drug transport.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Resistance, Multiple/physiology , Protein Kinase C/metabolism , Alkaloids/pharmacology , Animals , Cycloheximide/pharmacology , DNA, Complementary/genetics , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Humans , Phosphorylation , Rats , Staurosporine , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Cells, Cultured , Vinblastine/pharmacokinetics
19.
Onkologie ; 14(1): 7-12, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2057177

ABSTRACT

Vinorelbine (Navelbine) is a new, semisynthetic 5'Nor-vinca-alkaloid, modified on the catharantine ring, developed by Pierre Fabre Médicament. Vinorelbine is a potent as the other vinca alkaloids to inhibit mitotic microtubule polymerization. On the other hand, its activity is lower on axonal microtubule. Preclinical studies have shown its broad spectrum of activity in vitro and its antitumoral efficacy comparable or higher to that of other vinca alkaloids against murine tumors and in xenograft models. The main experimental toxicity of vinorelbine is a reversible leucopenia. No neurotoxicity was evidenced in rats, dogs and monkeys. After i.v. injection in patients, the plasma kinetic is described by a tricompartimental model with a high clearance, a very large volume of distribution and a long terminal half life, intermediate between vincristine and vinblastine. Tissue uptake of vinorelbine is very intense, probably related to its high liposolubility, leading to high tissue concentration compared to plasma. Phase I trial using weekly i.v. administration demonstrated a maximal tolerated dose (MTD) of 27.5 to 35.4 mg/m2 and the recommended dose was established at 30 mg/m2 weekly. In Phase II studies, Vinorelbine was shown to be effective in at least 4 types of cancer: Non-small cell lung cancer (remission rate: 33%), breast cancer (45%), advanced ovarian cancer (15% in heavily pretreated patients), Hodgkin's disease (90%). In all the trials, side effects are generally limited to a reversible and non-cumulative leucopenia. Neurotoxicity appears to be mild, similar to that observed with vinblastine and much less severe than with vincristine. No evidence of cardiac, pulmonary, renal, hepatic or other organ system toxicity has emerged.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents , Neoplasms/drug therapy , Tumor Cells, Cultured/drug effects , Vinblastine/analogs & derivatives , Animals , Drug Evaluation , Drug Evaluation, Preclinical , Humans , Neoplasms/blood , Neoplasms, Experimental/drug therapy , Vinblastine/adverse effects , Vinblastine/pharmacokinetics , Vinblastine/therapeutic use , Vinorelbine
20.
Nouv Rev Fr Hematol (1978) ; 31(2): 77-84, 1989.
Article in English | MEDLINE | ID: mdl-2671933

ABSTRACT

Vinorelbine (Navelbine) is a new semisynthetic vinca alkaloid which chemically differs from vinblastine by substitutions on the catharantine moiety of the molecule. It has shown promising experimental antitumor activity against experimental murine tumors as well as continuous cell lines of human neoplastic origin and human tumor xenografts in nude mice. Acute subacute and chronic toxicity extensively studied in rodents, dogs and primate has shown that hematotoxicity was almost the sole side-effect; neurotoxicity appears very limited. Almost exclusive affinity of NVB for mitotic tubulin and tubulin associated protein accounts for this pattern of toxicity. Phase I and II studies have been conducted in humans. Dose limiting side-effect appears to be neutropenia: the drug is slightly emetogenic, induces little alopecia, almost no neurotoxicity, and no other toxicity. Although preliminary, results of phase II studies already suggest significant activity of NVB in non small lung cancer (33% response rate in 78 evaluable patients), advanced breast cancer (53% response rate in 33 pts without significant chemotherapy for the target progression) and Hodgkin's disease (90% response rate after 4 weekly courses in 31 pts). Thus extensive pharmacological studies and ongoing clinical studies confirm that chemical modifications of the catharantine moiety of vinca alcaloid can lead to active agents with broader spectrum of activity and easily manageable side effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Dogs , Drug Evaluation , Drug Evaluation, Preclinical , Humans , Macaca mulatta , Mice , Mice, Nude , Microtubules/drug effects , Molecular Structure , Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Rats , Tumor Cells, Cultured/drug effects , Vinblastine/pharmacokinetics , Vinblastine/pharmacology , Vinblastine/therapeutic use , Vinblastine/toxicity , Vinorelbine
SELECTION OF CITATIONS
SEARCH DETAIL