Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Anticancer Res ; 20(2A): 849-52, 2000.
Article in English | MEDLINE | ID: mdl-10810365

ABSTRACT

We have previously shown that p27KiP1 plays a role in the tumor cell resistance of HT29 confluent monolayers to cytotoxic drugs in vitro. To determine whether p27KiP1 was a resistance factor to cytotoxic drugs in vivo we tested the effect of doxorubicin on p27KiP1-overexpressing HT29 tumors in nude mice. In this study we show that ectopic overexpression of p27KiP1 in HT29 human colon cancer cells decreases their tumorigenicity in vivo in nude mice. This decreased tumor growth was associated with increased p27KiP1 protein expression, studied by Western blotting in tumor extracts. Interestingly, the overexpressing-p27KiP1 tumors were significantly more resistant to intraveneous doxorubicin treatment than the control tumors. These results indicate that p27KiP1, which delays tumor growth could also increase tumor resistance to cytotoxic drugs in vivo.


Subject(s)
Cell Cycle Proteins , Cell Division/drug effects , Doxorubicin/toxicity , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Animals , Cyclin-Dependent Kinase Inhibitor p27 , Doxorubicin/therapeutic use , Enzyme Inhibitors/metabolism , HT29 Cells , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
2.
Ann Rheum Dis ; 59(2): 146-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666173

ABSTRACT

BACKGROUND: The multidrug resistance phenomenon results from the expression of P-glycoprotein (P-gp), a drug-efflux pump. Corticosteroids are substrates for P-gp, whose function can be inhibited by cyclosporin. This study evaluates the ability of cyclosporin to modulate dexamethasone uptake in multidrug resistant cells. METHODS: The K 562 cell line, which does not express P-gp and a P-gp expressing clone, K562/ADM, were used. Cells were incubated with H3-dexamethasone in the absence or presence of cyclosporin at various concentrations. Then, cells were washed, lysed, and radioactivity was measured. RESULTS: The uptake of dexamethasone alone was higher in sensitive than in resistant cells. Addition of cyclosporin induced a dose dependent increase of dexamethasone uptake in resistant cells, whereas the drug did not influence dexamethasone uptake in parental cells. CONCLUSION: Cyclosporin, at therapeutic concentrations induces a moderate, but significant increase in dexamethasone accumulation in multidrug resistant cells. Thus, cyclosporin might increase the intestinal absorption of corticosteroids or their accumulation in mononuclear cells, or both, thereby increasing their therapeutic efficacy.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Antirheumatic Agents/pharmacology , Cyclosporine/pharmacology , Dexamethasone/pharmacokinetics , Drug Resistance, Multiple , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Humans , K562 Cells/metabolism
3.
Leukemia ; 14(12): 2085-94, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11187897

ABSTRACT

Overexpression of P-glycoprotein (P-gp) in cancer cells reduces intracellular accumulation of various anticancer drugs including anthracyclines and vinca alkaloids. This multidrug resistance (MDR) phenotype can be reversed in vitro by a number of non-cytotoxic drugs. We have identified the quinine's isomer cinchonine as a potent MDR reversing agent, both in vitro and in animal models. Here, we report an open phase I dose escalation trial in patients with refractory or relapsed malignant lymphoid diseases. Cinchonine dihydrochloride was administered by continuous i.v. infusion for 48 h and escalated over five dose levels ranging from 15 to 35 mg/kg/d. Cinchonine infusion started 24 h before i.v. doxorubicin (25 mg/m2), vinblastine (6 mg/m2), cyclophosphamide (600 mg/m2) and methylprednisolone (1 mg/kg/d) (CHVP regimen) and lasted for 24 h after chemotherapy infusion. Thirty-four patients received 87 cycles of CHVP/cinchonine. The MTD of cinchonine administered by continuous i.v. infusion was 30 mg/kg/d. Prolonged cardiac repolarization was the main dose-limiting toxicity. No ventricular arrhythmia including 'torsade de pointes' was observed. An MDR reversing activity was identified in the serum from every patient and correlated with cinchonine serum level. When infused at 30 mg/kg/d, cinchonine demonstrated a limited influence on doxorubicin pharmacokinetic. We conclude that i.v. infusion of cinchonine might be started 12 h before MDR-related chemotherapy infusion and requires continuous cardiac monitoring but no reduction of cytotoxic drug doses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cinchona Alkaloids/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Lymphoproliferative Disorders/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cinchona Alkaloids/adverse effects , Cinchona Alkaloids/pharmacokinetics , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Electrocardiography , Female , Heart/drug effects , Humans , Leukopenia/chemically induced , Male , Middle Aged , Prednisone/administration & dosage , Recurrence , Teniposide/administration & dosage
4.
Bull Cancer ; 87 Spec No: 7-22, 2000 Dec.
Article in French | MEDLINE | ID: mdl-11250604

ABSTRACT

Aromatase is an enzymatic complex responsible for the conversion of androgens into estrogens; these hormones are important in development, reproduction, but also in the growth of estrogen-dependent cancer. This enzyme is present in 60-70% of the breast cancer. The aromatase inhibitors are important drugs in the breast cancer treatment of postmenopausal women. In order to study their in vivo activity, animal models have been developed, e.g. rat with tumour induced by 7,12-dimethylbenz[a]anthracene, PMSG-primed immature rat or athymic nude mice with aromatase transfected MCF-7 xenograft. In this review, we were interested in preclinical results obtained with both classes: steroidal and nonsteroidal inhibitors. The former group, as substrate analogs formestane or exemestane, are irreversible, selective and long-lasting inhibitors of aromatase. The nonsteroidal molecules, such as letrozole or anastrozole, are reversible inhibitors with high affinity. Finally, knowledge of the enzyme active site, with molecular modeling and site-directed mutagenesis, could be useful to develop new inhibitor families, more specific and potent in vivo.


Subject(s)
Androstenedione/analogs & derivatives , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors , Enzyme Inhibitors/therapeutic use , Aminoglutethimide/therapeutic use , Anastrozole , Androstadienes/therapeutic use , Androstenedione/therapeutic use , Animals , Aromatase/physiology , Drug Evaluation, Preclinical , Fadrozole/therapeutic use , Female , Humans , Letrozole , Models, Animal , Nitriles/therapeutic use , Rats , Structure-Activity Relationship , Triazoles/therapeutic use
5.
Anticancer Res ; 19(4C): 3575-81, 1999.
Article in English | MEDLINE | ID: mdl-10629655

ABSTRACT

To characterize the biological features of advanced breast cancer associated with poor chemotherapy response and worse prognosis, sequential tumor samples obtained from 75 patients receiving primary chemotherapy were analysed for MDR1 and TS gene expression before and after treatment. MDR1 gene expression was also analysed in 36 sequential normal samples. The levels of MDR1 and TS genes expression were determined by reverse transcription-PCR method, and examined in relation to p53 gene status, and the clinical outcome of the patients. After treatment, MDR1 expression levels were significantly enhanced in tumor (p = 0.0033) and normal (p = 0.0098) samples, whereas a significant decrease in TS expression was observed (p = 0.0054). There was no significant correlation between MDR1 or TS expressions and the presence of p53 mutations (detected in 24% of the cases), chemoresponsiveness, or survival. Only p53 mutations were associated with reduced disease-free survival (p = 0.0473). These results demonstrate that MDR1 and TS gene expressions were affected by drug exposure, but not by p53 gene status. Furthermore, the increase of MDR1 gene expression in normal and tumor tissues is in favor of an induced MDR1 expression rather than of a selection of resistant tumoral clones, which can be responsible for the absence of relationship of MDR1 expression with clinical outcome of advanced breast cancer patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Genes, p53 , Thymidylate Synthase/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Gene Expression/drug effects , Humans , Middle Aged , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
6.
Leukemia ; 11(7): 1086-94, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9204996

ABSTRACT

The wide discrepancies in the frequency of 'positive' samples for multidrug resistance (MDR) phenotype within the same type of tumor observed in the literature justified the need for the definition of consensus recommendations. To define standard techniques of MDR phenotype measurement, we ran a large multicentric evaluation of the different methods available. Thirty-six French centers participated in the study, and 742 samples of 2-10 x 10(6) viable cells were sent by overnight express mail between December 1993 and February 1996. The same batches of MRK16, 4E3 and UIC2 were used. Nineteen samples of leukemia (12 AML, 1 ALL, 6 lymphoproliferative syndromes) and six leukemic cell lines with different levels of MDR expression were tested. Five meetings reached agreement concerning the guidelines for each technique, except immunocytochemistry. The 19 fresh samples were tested by each center using one to four techniques among cytofluorometry, immunocytochemistry, functional tests and RT-PCR. Five samples were diagnosed as 'negative' according to local criteria, with few discordant results (0 to 16% of 'positive' results). For all the 14 remaining samples, large discrepancies were observed from center to center, and from one technique to another. No correlations could be found between techniques. Flow cytometric analysis of cells already exposed to MRK16 or control IgG2A, fixed in paraformaldehyde and sent to centers did not reduce the discrepancies between centers in two of the four samples with moderate expression, emphasizing the role of histogram interpretation. The use of alternative monoclonal antibodies (4E3 and UIC2) did not reduce the discrepancies observed. In a second step, the K562 parental cell line, a low resistant subline (K562/HHT100, x7 resistance index to DNR) and a high resistant subline (K562/HHT300, x125 resistance index to DNR) were sent blindly three times, with an increasing level of recommendations for flow cytometry. Dramatic improvements were observed in cytometric results when the result was expressed as the ratio of arithmetic mean of fluorescence of antibody (10 microg of MRK16)/arithmetic mean of fluorescence of control (10 microg IgG2A): the proportion of expected results increased from 61 to 100% for K562, and from 37 to 85% for K562/HHT100. For uptake and drug efflux measurements, the use of 1 h uptake of 0.1 microM of rhodamine, followed by 1 h efflux +/-10 microM of verapamil, permitted an increased reproducibility of the technique from 71 to 100% for K562 and K562/HHT100. Whatever the technique used, concordant results were obtained for K562/HHT300. The immunocytochemistry, using several antibodies (MRK16, JSB1 and C219) gave many non-interpretable results (44%), due to a frequent high background and discordant results between antibodies in the same centers, and discordant conclusions between centers. The group does not recommend this technique for circulating tumoral cells.


Subject(s)
Drug Resistance, Multiple , Leukemia/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Flow Cytometry , Humans , Immunophenotyping , Phenotype , Tumor Cells, Cultured
7.
Leukemia ; 11(7): 1095-106, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9204997

ABSTRACT

Since there is no consensus on the techniques for multidrug resistance (MDR) phenotype evaluation, many discrepancies concerning the importance and frequency of mdr1 gene expression in leukemias and solid tumors are observed in the literature. In order to establish an inter-laboratory consensus in France, a multicenter study was carried out to propose further guidelines for MDR phenotype evaluation. The techniques used by the 38 laboratories participating in the trial were: immunodetection (immunohisto and/or cytochemistry, flow cytometry), functional tests, reverse transcription-polymerase chain reaction (RT-PCR) or Northern blot. We present the results obtained by 19 laboratories concerning the measurement of mdr1 gene expression assessed by RT-PCR or Northern blot in: (1)19 samples of tumor cells obtained from leukemic patients; (2) six solid tumor samples obtained at surgery; (3) eight cell lines exhibiting variable levels of resistance, and; (4)10 preparations of RNA and of cDNA obtained from solid tumors. Standardization of the RT-PCR technique and preliminary results comparing RT-PCR with immunohistochemistry in solid tumors are also reported.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Leukemia/drug therapy , Neoplasms/drug therapy , Polymerase Chain Reaction/standards , DNA, Complementary/analysis , Humans , Immunohistochemistry , RNA/analysis
8.
Int J Cancer ; 63(2): 268-75, 1995 Oct 09.
Article in English | MEDLINE | ID: mdl-7591216

ABSTRACT

Azatoxin (NSC 640737), a synthetic molecule, was rationally designed as a topoisomerase-II inhibitor and was shown to be a potent cytotoxic agent that inhibits both tubulin and topoisomerase II. A structure-activity relationship study allowed to select 3 derivatives that inhibit either tubulin (methylazatoxin) only or topoisomerase II (fluoroanilinoazatoxin and nitroanilino-azatoxin) in MTT assays performed on K562 and K562/ADM cells; the latter, expressing P-glycoprotein, indicated cross-resistance of K562/ADM cells to all 4 compounds. DNA double-strand breaks induced by the 3 azatoxins that inhibit topoisomerase II in vitro were decreased in K562/ADM as compared with K562 cells. Nitroanilino-azatoxin was the only compound for which resistance and reduced DNA damage observed in K562/ADM cells was partially reversed by verapamil, suggesting that nitroanilinoazatoxin was a substrate for P-glycoprotein. These results were confirmed by testing the cytotoxic activity of azatoxins on P-glycoprotein-expressing rat colon-carcinoma DHDK12/TRb cells in the absence and the presence of verapamil. Cell-cycle and mitotic-index studies indicated that azatoxin- and methyl-azatoxin-induced M-phase arrest was less in K562/ADM than in K562 cells. The G2 block induced by fluoro- and nitroanilinoazatoxins was delayed in K562/ADM cells. Verapamil increased cell-cycle inhibition induced by nitroanilinoazatoxin in K562/ADM cells without modifying cell-cycle effects of fluoroanilinoazatoxin. These results (i) are consistent with the specific inhibition of topoisomerase II or tubulin by azatoxin derivatives in cells; (ii) indicate that the nitro group of nitroanilinoazatoxin allows recognition and efflux by the P-glycoprotein; and (iii) suggest that cross-resistance of K562/ADM cells to other azatoxin derivatives is not mediated by P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Drug Resistance, Multiple , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Topoisomerase II Inhibitors , Base Sequence , Cell Cycle/drug effects , Cell Survival/drug effects , Cytotoxins/pharmacology , DNA Damage/drug effects , DNA Primers/chemistry , Gene Expression/drug effects , Growth Inhibitors/pharmacology , Humans , Molecular Sequence Data , Mutagens/pharmacology , RNA, Messenger/genetics , Tumor Cells, Cultured , Verapamil/pharmacology
9.
Int J Cancer ; 61(6): 873-9, 1995 Jun 09.
Article in English | MEDLINE | ID: mdl-7790124

ABSTRACT

Colorectal adenocarcinomas are inherently resistant to anthracyclines and other topoisomerase-II inhibitors. Resistance to doxorubicin of colon cancer cells (Caco2) depends on 2 main mechanisms. The first is typical multi-drug resistance, characterized by the mdr1 gene and its product the P170 membrane glycoprotein. P170 effluxes anthracyclines out of cancer cells and is antagonized in vitro by verapamil. The second mechanism, which develops when cell-culture density increases, we have designated confluence-dependent resistance. Confluence-dependent resistance depends on the reduced topoisomerase II content of the G0/G1-phase cells which accumulate in the confluent population. We show here that short treatments of confluent Caco2 cells with slightly toxic concentrations of DNA-damaging agents (cisplatin, melphalan or mitomycin C) produced a transient accumulation of cells in S- and G2/M-phases of the cell cycle. Concomitantly with the increase in the S-phase population, the topoisomerase II cellular level and the sensitivity of cells to doxorubicin were greatly enhanced. Overcoming confluence-dependent resistance through S-phase accumulation and inhibition of multi-drug resistance by verapamil were fully additive, and a nearly complete reversal of confluent Caco2 cells' resistance to doxorubicin was obtained when both strategies were combined.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Drug Resistance, Multiple , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adenocarcinoma/pathology , Cell Cycle , Cisplatin/pharmacology , Colonic Neoplasms/pathology , DNA Topoisomerases, Type II/biosynthesis , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Humans , Melphalan/pharmacology , Mitomycins/pharmacology , Topoisomerase II Inhibitors , Tumor Cells, Cultured
10.
Leukemia ; 9(6): 1013-24, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7596166

ABSTRACT

Topoisomerase (topo) inhibitors induce enzyme-linked DNA breaks. Resulting DNA damage can lead to cell cycle arrest and/or cell death by apoptosis. The sensitivity of five human leukemic cell lines to topo I (camptothecin or CPT) and topo II (etoposide or VP-16) inhibitors varied widely (100-fold for CPT and 30-fold for VP-16). Three cell lines were more sensitive (BV173, HL60, U937) and two cell lines were resistant (K562, KCL22) to both drugs. None of these cell lines were selected for drug resistance and overexpressed mdr1 gene. Their sensitivity was not related to their doubling time nor to cell cycle repartition. The initial DNA damage (cleavable complexes) induced by topo I and II inhibitors was measured as DNA-protein crosslinks (DPC) using alkaline elution. Neither DPC level induced by 30-min treatment with CPT or VP-16 nor the levels of topo 1, topo II alpha and topo II beta mRNA were related to sensitivity. Electron microscopy and DNA fragmentation measured by filter elution and agarose gel electrophoresis demonstrated that apoptosis was induced by both drugs in the five cell lines. The kinetics of DNA fragmentation was related to cell sensitivity. At drug concentrations higher than IC50, DNA fragmentation increased very rapidly in the three sensitive, compared with the two resistant, cell lines. Continuous exposure to both drugs induced cell cycle arrest in either G2 or S phase that was related both to cell sensitivity and drug concentration. Comparison between cell lines indicated that the ability of cells to arrest cell cycle in G2 or S phase was related to their drug sensitivity and increased with cell resistance. In a given cell line, cell cycle progression was observed to be progressively inhibited by increasing drug concentrations. Treatment of synchronized cells demonstrated that highly cytotoxic drug concentration induced a complete inhibition of cell cycle progression. Altogether, these data suggest that the ability of leukemic cell lines to regulate cell cycle progression and to trigger apoptosis is more indicative of their sensitivity to topoisomerase poisons than cleavable complexes induced by these drugs.


Subject(s)
Apoptosis/physiology , Camptothecin/toxicity , Cell Cycle/physiology , Cell Survival/drug effects , Etoposide/toxicity , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Apoptosis/drug effects , Base Sequence , Cell Cycle/drug effects , Cell Line , DNA Damage , DNA Primers , DNA, Neoplasm/drug effects , DNA, Neoplasm/ultrastructure , Dose-Response Relationship, Drug , Humans , Leukemia , Microscopy, Electron , Molecular Sequence Data , Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Anticancer Drug Des ; 10(2): 103-18, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7710633

ABSTRACT

We have previously suggested that quinine and cinchonine could be good candidates for the clinical circumvention of multidrug resistance (MDR) in haematological malignancies because of their tolerance and their retained efficacy in serum. We have also shown that cinchonine was more efficient than quinine as an anti-MDR agent in vitro, ex vivo and in vivo after parenteral administration. Here, we report that cinchonine administered per os (po) is much more active than quinine po in circumventing MDR in rats bearing resistant colon tumours. The pharmacokinetics of cinchonine and quinine administered po in rat are shown to be very different. Cinchonine demonstrates a greater absolute bioavailability than quinine (44% versus 30%, respectively). Its serum concentration correlates with the anti-MDR activity measured ex vivo and in vivo. Cinchonine administered po does not significantly modify the pharmacokinetics of intravenous doxorubicin (DXR). However, cinchonine induces a significant increase of DXR uptake in organs which express the mdr1 gene (liver, kidney, lung). When associated with VAD (vincristine, adriamycin, dexamethasone) combined therapy in rats, cinchonine does not significantly increase the toxicity of the cytotoxic drugs. Based on these experimental data, a phase I clinical trial is currently in progress to test the tolerance of this potent MDR-reversing agent administered po.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Cinchona Alkaloids/pharmacology , Drug Resistance, Multiple , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Availability , Cinchona Alkaloids/administration & dosage , Cinchona Alkaloids/pharmacokinetics , Colonic Neoplasms/drug therapy , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Synergism , Female , Infusions, Intravenous , Quinones/pharmacokinetics , Quinones/pharmacology , Quinones/therapeutic use , Rats , Tissue Distribution , Tumor Cells, Cultured , Vincristine/administration & dosage
12.
Cancer Lett ; 85(2): 171-6, 1994 Oct 14.
Article in English | MEDLINE | ID: mdl-7954333

ABSTRACT

Confluence dependent resistance (CDR) is one of the principal mechanisms by which solid tumor cells resist anthracyclines. CDR is thought to be mediated by cell-cell contact which increases the fraction of non-proliferating resistant cells in a post confluence monolayer culture. As E-cadherin is a major Ca2+ dependent adhesion molecule, involved in cell-cell adhesion, differentiation and polarity of normal and cancerous epithelial cells, we decided to investigate its involvement in the CDR mechanism. In order to do this, we measured the intracellular accumulation and the cytotoxicity of doxorubicin (DXR) in four subclones, derived from the same parental murine mammary cell line (NMuMG), differing in their expression of E-cadherin. A significant reduction in DXR accumulation and cytotoxicity was observed in NM-f-ras-TD-CAMx, which expresses E-cadherin, suggesting that E-cadherin could play a role in the increase of drug resistance observed in confluent cancer cells.


Subject(s)
Cadherins/metabolism , Doxorubicin/toxicity , Mammary Glands, Animal/cytology , Animals , Biological Transport , Cell Adhesion , Cell Cycle , Cells, Cultured , Doxorubicin/metabolism , Drug Resistance , Epithelial Cells , Fibroblasts/cytology , Genes, ras , In Vitro Techniques , Mammary Glands, Animal/metabolism , Mice , Transfection
13.
Br J Cancer ; 70(4): 631-5, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7917908

ABSTRACT

Inherent resistance of colon cancer cells to cis-diamminedichloroplatinum(II) (CDDP) is partly attributed to reduced drug penetration through plasma membrane. Amphotericin B (AmB), a polyene antifungal antibiotic, has been shown to increase CDDP penetration and cytotoxicity on several non-digestive cancer cell lines. We demonstrated here that AmB dramatically increases the penetration of CDDP, and to a lesser extent that of carboplatin (Carbo-P) and oxaloplatin (L-OHP), in the primary resistant HT 29 human colon cancer cells when drug incubation is performed in serum-free medium. The cytotoxicity of CDDP but not that of Carbo-P and L-OHP was increased by AmB. However, AmB-induced potentiation of CDDP penetration and toxicity was almost completely abolished when cell incubation was performed in presence of human serum. We investigated whether the dilution of human serum by a high osmotic power gelatine solution (Lomol) could restore the positive effect of AmB on CDDP accumulation in HT 29 cells. Incubation of cells with CDDP and AmB in pure Lomol resulted in a 6-fold increase in platinum cellular content. However, addition of serum (25%) in Lomol solution reduced to only 2-fold the increase in platinum cellular content provoked by AmB. These disappointing results show that AmB is probably uninteresting as a modulator of CDDP resistance in clinical practice. The use of haemodilution to restore the positive AmB effect on platinum cellular accumulation cannot be warranted.


Subject(s)
Adenocarcinoma/drug therapy , Amphotericin B/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blood Proteins/pharmacology , Cisplatin/pharmacokinetics , Cisplatin/toxicity , Colonic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cell Membrane Permeability/drug effects , Culture Media, Serum-Free , Drug Resistance , Drug Synergism , Humans , Tumor Cells, Cultured/drug effects
15.
Anticancer Drug Des ; 9(2): 73-84, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8166930

ABSTRACT

Since liposomes are slowly resorbed from serous cavities, they may constitute a valuable tool for the treatment of peritoneal carcinomatosis. We prepared mitoxantrone (MXN)-liposomes with various lipid compositions and checked their antitumoral activity on a peritoneal carcinomatosis induced by a colon cancer cell injection (1 x 10(5) C51 cells) in BALB/c mice. MXN entrapment in liposomes was rapid and stable due to its high lipophilicity. MXN carried in phosphatidylcholine: cholesterol (2:1; G-liposomes) displayed a reduced toxicity in mice compared to the free drug. When tested at a non-toxic dose (2 mg/kg), MXN entrapped in G-liposomes proved to be as efficient as the free drug. At a higher MXN dose (3 mg/kg), both G-liposomes and phosphatidylcholine:cholesterol:dipalmitoylphosphatidylethanolamine (7:2:1) liposomes, loaded with MXN, significantly increased the life span of mice compared to the free drug and six other liposome formulations. Increase in the MXN therapeutic index, when used in the liposomal form, could then merit further clinical investigations in regard to patients with malignancies confined to serous cavities.


Subject(s)
Mitoxantrone/administration & dosage , Peritoneal Neoplasms/drug therapy , Animals , Carcinoma/drug therapy , Colonic Neoplasms , Drug Carriers , Electrochemistry , Female , Liposomes/chemistry , Male , Mice , Mice, Inbred BALB C , Mitoxantrone/pharmacokinetics , Mitoxantrone/toxicity , Neoplasm Transplantation , Tissue Distribution , Tumor Cells, Cultured
16.
Leukemia ; 8(1): 160-4, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8289482

ABSTRACT

We have previously suggested that quinine and cinchonine could be good candidates for clinical circumvention of multidrug resistance (MDR) in hematological malignancies because of their tolerance and their retained efficacy in serum. In the present study, we have used the well-characterized multidrug resistant human leukemic cell line K562/ADM to compare the effect in vitro of quinine and cinchonine on doxorubicin, mitoxantrone, and vincristine uptake and cytotoxicity. In serum-free medium, quinine induced a dose-dependent increase of doxorubicin uptake reaching about 200% at 40 microM, while it had a slight and no effect on mitoxantrone and vincristine uptake respectively. In the same conditions, cinchonine induced a rapid and significant increase in the accumulation of the three drugs, reaching a plateau phase between 5 and 10 microM. Quinine and cinchonine induced both potentiation of doxorubicin, vincristine and mitoxantrone cytotoxicity in K562/ADM cells. However, quinine reached a plateau phase at 10 microM, while cinchonine had a maximal effect at 5 microM and was significantly more potent at low concentrations. When diluted in plasma, cinchonine was less bound to proteins than quinine. The free fraction of alkaloids was 37-55% for cinchonine and 20-30% for quinine. Cinchonine-induced enhancement of vincristine cellular accumulation was little modified by plasma proteins. When incubated in whole blood, the fraction of cinchonine trapped in red blood cells was rapidly and completely exchangeable with plasma. We conclude that cinchonine is a stronger inhibitor of MDR than quinine.


Subject(s)
Cinchona Alkaloids/pharmacology , Leukemia, Myeloid/drug therapy , Quinine/pharmacology , Antineoplastic Agents/pharmacokinetics , Blood Proteins/metabolism , Cinchona Alkaloids/blood , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Interactions , Drug Resistance/genetics , Erythrocytes/metabolism , Humans , Leukemia, Myeloid/blood , Leukemia, Myeloid/metabolism , Mitoxantrone/pharmacokinetics , Mitoxantrone/pharmacology , Quinine/blood , Tumor Cells, Cultured/drug effects , Vincristine/pharmacokinetics , Vincristine/pharmacology
17.
Anticancer Res ; 13(2): 487-90, 1993.
Article in English | MEDLINE | ID: mdl-8517666

ABSTRACT

Sodium butyrate (NaBu) but not dimethylsulfoxide (DMSO) induced the synthesis of villin, a protein of the brush border microvilli cytoskeleton, in a rat colon cancer cell line. Neither NaBu nor DMSO altered mdr 1-mRNA expression or multidrug resistance (MDR)--associated cellular transport of doxorubicin. These results show that mdr 1 gene expression and activity are independent of other brush border proteins induced by differentiating agents at the apical pole of the epithelial cell.


Subject(s)
Butyrates/pharmacology , Carrier Proteins/biosynthesis , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Resistance/genetics , Gene Expression/genetics , Microfilament Proteins/biosynthesis , Animals , Biological Transport, Active/drug effects , Butyric Acid , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/metabolism , Colonic Neoplasms/pathology , Dimethyl Sulfoxide/pharmacology , Doxorubicin/pharmacokinetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Rats , Rats, Inbred Strains , Tumor Cells, Cultured/drug effects
18.
Cancer Res ; 52(10): 2797-801, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1581892

ABSTRACT

Circumvention of multidrug resistance is a new field of investigation in cancer chemotherapy, and safe and potent multidrug resistance inhibitors are needed for clinical use. We investigated several analogues of quinine for their ability to increase anthracycline uptake in resistant cancer cells. Cinchonine was the most potent inhibitor of anthracycline resistance in vitro, and its activity was little altered by serum proteins. Serum from rats treated with i.v. cinchonine produced greater uptake of doxorubicin in cancer cells (DHD/K12/PROb rat colon cells and K562/ADM human leukemic cells) than did serum from quinine-treated rats (ex vivo assay). Cinchonine was more effective than quinine in reducing tumor mass and increasing the survival of rats inoculated i.p. with DHD/K12/PROb cells and treated i.p. with deoxydoxorubicin. Moreover, the acute toxicity of cinchonine in rats and mice was lower than that of other quinine-related compounds. The lower toxicity and greater potentiation of in vivo anthracycline activity produced by cinchonine are favorable characteristics for its use as an anti-multidrug resistance agent in future clinical trials.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cinchona Alkaloids/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Cinchona Alkaloids/pharmacokinetics , Cinchona Alkaloids/toxicity , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Resistance, Microbial , Drug Synergism , Female , Male , Mice , Mice, Inbred BALB C , Quinine/pharmacokinetics , Quinine/pharmacology , Quinine/toxicity , Rats , Rats, Inbred Strains , Tumor Cells, Cultured/drug effects
19.
Int J Cancer ; 50(5): 677-82, 1992 Mar 12.
Article in English | MEDLINE | ID: mdl-1544702

ABSTRACT

In vitro sensitivity of HT29 human colon cancer cells to doxorubicin (DXR), vincristine (VCR), etoposide (VP16), cisplatin (CDDP), melphalan (L-PAM) and 5-fluorouracil (5FU) was markedly reduced when cell-culture density increased. For some drugs, confluence-dependent resistance (CDR) was partly due to decreased intracellular drug accumulation; the ratio of mean intracellular drug content of non confluent to confluent cells (NC/C) was 2.5 for DXR, 4.1 for VCR and 7.4 for VP16. Altered drug penetration with confluence could be related to decrease of plasma membrane fluidity as measured by the fluorescence polarization method. Reduction of drug intracellular accumulation was nil or weak for L-PAM (NC/C = 1.0), CDDP (NC/C = 1.2) and 5 FU (NC/C = 1.8). Even if drug concentration was adjusted in culture medium to produce similar intracellular drug content in confluent and non confluent cells, higher intrinsic resistance of confluent cells was still evidenced for DXR and VP16 but not for VCR, the only agent without direct interaction with DNA. DXR- and VP16-induced DNA breakage was also less important in confluent than in non-confluent cells. CDR appeared closely related to an increased proportion of non-cycling cells at confluence, as demonstrated by flow cytometry, expression of nuclear antigen recognized by Ki67 MAb and expression of topoisomerase II. CDR is probably a major factor in the poor sensitivity of colorectal adenocarcinomas to chemotherapy.


Subject(s)
Antineoplastic Agents/metabolism , Colonic Neoplasms/pathology , Drug Resistance , Cell Division , Colonic Neoplasms/metabolism , DNA Damage , Humans , Membrane Fluidity , Tumor Cells, Cultured
20.
Gastroenterol Clin Biol ; 16(3): 215-9, 1992.
Article in French | MEDLINE | ID: mdl-1582536

ABSTRACT

Animal models are useful in the evaluation of adjuvant or palliative treatment modalities of human colonic adenocarcinoma. In the present paper, the efficacy of 22 usual chemotherapeutic agents was evaluated in a model of peritoneal carcinomatosis of colonic origin in the BD IX rat. Mitomycin, cisplatine, carboplatine, cyclophosphamide, ifosfamide, and thiotepa were very effective agents on microscopic carcinomatosis (treatment given 3 days after an intraperitoneal inoculation of 1 x 10(6) DHD/K12/PROb cells). Intravenous administration was as effective as the intraperitoneal route, except for anthracyclines and 5-fluorouracil. Rats treated at early stages by thiotepa or cisplatin survived up to 4 months after cell injection and did not display tumor at autopsy. Administered late (15 days after cell injection), none of the drugs were able to cure the rats with carcinomatosis.


Subject(s)
Cisplatin/therapeutic use , Colonic Neoplasms/pathology , Mitomycin/therapeutic use , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Ifosfamide/administration & dosage , Ifosfamide/therapeutic use , Injections, Intraperitoneal , Injections, Intravenous , Male , Mitomycin/administration & dosage , Peritoneal Neoplasms/secondary , Rats , Thiotepa/administration & dosage , Thiotepa/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...