Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Curr Drug Targets ; 15(14): 1312-21, 2014.
Article in English | MEDLINE | ID: mdl-25382189

ABSTRACT

UNLABELLED: The active metabolite (JM118) of the oral platinum analog satraplatin (JM216) was investigated for potential synergism with erlotinib, an epidermal growth factor receptor (EGFR) inhibitor. JM118 sensitivity of 7 cancer cell lines (ovarian: 2008, A2780; colon: Lovo92, WiDr; lung: A549, SW1573; epidermoid: A431), was enhanced most pronounced when JM118 preceded erlotinib, which was associated with increased formation of DNA-platinum adducts. The combination increased G2/M phase accumulation and enhanced apoptosis. JM118 increased the phosphorylation of the cell cycle proteins CDK2 and CHK1 after 24 hr exposure. JM118/erlotinib enhanced Erk and Akt phosphorylation after 2 hr. JM118 significantly decreased the phosphorylation of PTEN, VEGFR, EPHA1, ERBB4, FGF-R, andSTAT3 by 20 (PTEN) to >90% (STAT3). CONCLUSION: Erlotinib enhanced the effects of JM118, even in cells with mutations in Ras. The mechanism of synergy involved a combination of effects on platinum-DNA adduct formation, cell cycle distribution and signaling.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Adducts/metabolism , Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , Apoptosis , Cell Cycle/drug effects , Cell Line, Tumor , Drug Synergism , Drug Therapy, Combination , Erlotinib Hydrochloride , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation/drug effects
2.
Springerplus ; 3: 732, 2014.
Article in English | MEDLINE | ID: mdl-25674464

ABSTRACT

UNLABELLED: We aimed to determine whether the multidrug-resistance-proteins MRP4 (ABCC4) and MRP5 (ABCC5) confer resistance to the antimetabolites cytarabine (Ara-C), gemcitabine (GEM), and the L-nucleoside analog troxacitabine. For this purpose we used HEK293 and the transfected HEK/MRP4 (59-fold increased MRP4) or HEK/MRP5i (991-fold increased MRP5) as model systems and tested the cells for drug sensitivity using a proliferation test. Drug accumulation was performed by using radioactive Ara-C, and for GEM and troxacitabine with HPLC with tandem-MS or UV detection. At 4-hr exposure HEK/MRP4 cells were 2-4-fold resistant to troxacitabine, ara-C and 9-(2-phosphonylmethoxyethyl)adenine (PMEA), and HEK/MRP5i to ara-C and PMEA, but none to GEM. The inhibitors probenecid and indomethacin reversed resistance. After 4-hr exposure ara-C-nucleotides were 2-3-fold lower in MRP4/5 cells, in which they decreased more rapidly after washing with drug-free medium (DFM). Trocacitabine accumulation was similar in the 3 cell lines, but after the DFM period troxacitabine decreased 2-4-fold faster in MRP4/5 cells. Troxacitabine-nucleotides were about 25% lower in MRP4/5 cells and decreased rapidly in MRP4, but not in MRP5 cells. Accumulation of GEM-nucleotides was higher in the MRP4/5 cells. IN CONCLUSION: MRP4 and MRP5 overexpression confer resistance to troxacitabine and ara-C, but not to GEM, which was associated with a rapid decline of the ara-C and troxacitabine-nucleotides in HEK/MRP4-5 cells.

3.
Invest New Drugs ; 30(5): 1908-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22002019

ABSTRACT

Cytarabine (ara-C) and gemcitabine (dFdC) are commonly used anticancer drugs, which depend on the equilibrative (ENT) and concentrative-nucleoside-transporters to enter the cell. To bypass transport-related drug resistance, lipophilic derivatives elacytarabine (CP-4055), ara-C-5'elaidic-acid-ester, and CP-4126, (CO 1.01) gemcitabine-5'elaidic-acid-ester, were investigated for the entry into the cell, distribution, metabolism and retention. The leukemic CEM-cell-line and its deoxycytidine-kinase deficient variant (CEM/dCK-) were exposed for 30 and 60 min to the radiolabeled drugs; followed by culture in drug-free medium in order to determine drug retention in the cell. The cellular fractions were analyzed with thin-layer-chromatography and HPLC. Elacytarabine and CP-4126 were converted to the parent compounds both inside and outside the cell (35-45%). The ENT-inhibitor dipyridamole did not affect their uptake or retention. Inside the cell Elacytarabine and CP-4126 predominantly localized in the membrane and cytosolic fraction, leading to a long retention after removal of the medium. In contrast, in cells exposed to the parent drugs ara-C and dFdC, intracellular drug concentration increased during exposure but decreased to undetectable levels after drug removal. In the dCK- cell line, no metabolism was observed. The concentrations of ara-CTP and dFdCTP reached a peak at the end of the incubation with the drugs, and decreased after drug removal; peak levels of dFdCTP were 35 times higher than ara-CTP and was retained better. In contrast, after exposure to elacytarabine or CP-4126, ara-CTP and dFdCTP levels continued to increase not only during exposure but also during 120 min after removal of the elacytarabine and CP-4126. Levels of ara-CTP and dFdCTP were higher than after exposure to the parent drugs. In conclusion, the lipophilic derivatives elacytarabine and CP-4126 showed a nucleoside-transporter independent uptake, with long retention of the active nucleotides. These lipophilic nucleoside analogues are new chemical entities suitable for novel clinical applications.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytarabine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Nucleotidases/metabolism , Antineoplastic Agents/metabolism , Cell Line , Cytarabine/metabolism , Cytarabine/pharmacokinetics , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Deoxycytidine Kinase/metabolism , Dipyridamole/metabolism , Drug Resistance, Neoplasm , Humans , Leukemia/metabolism , Nucleoside Transport Proteins/metabolism , Gemcitabine
4.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1168-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22132972

ABSTRACT

Many drugs that are currently used for the treatment of cancer have limitations, such as induction of resistance and/or poor biological half-life, which reduce their clinical efficacy. To overcome these limitations, several strategies have been explored. Chemical modification by the attachment of lipophilic moieties to (deoxy)nucleoside analogs should enhance the plasma half-life, change the biodistribution, and improve cellular uptake of the drug. Attachment of a lipophilic moiety to a phosphorylated (deoxy)nucleoside analog will improve the activity of the drugs by circumventing the rate-limiting activation step of (deoxy)nucleoside analogs. Encapsulating drugs in nanoparticles or liposomes protects the drug against enzymatic breakdown in the plasma and makes it possible to get lipophilic compounds to the tumor site. In this review, we discuss the considerable progress that has been made in increasing the efficacy of classic (deoxy)nucleoside and fluoropyrimidine compounds by chemical modifications and alternative delivery systems.


Subject(s)
Lipids/chemistry , Neoplasms/drug therapy , Nucleosides/therapeutic use , Prodrugs/therapeutic use , Pyrimidines/therapeutic use , Chemistry, Pharmaceutical , Humans , Nucleosides/chemistry , Nucleosides/pharmacology , Prodrugs/chemistry , Pyrimidines/chemistry
5.
Invest New Drugs ; 29(3): 456-66, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20066470

ABSTRACT

Gemcitabine is a deoxycytidine (dCyd) analog with activity in leukemia and solid tumors, which requires phosphorylation by deoxycytidine kinase (dCK). Decreased membrane transport is a mechanism of resistance to gemcitabine. In order to facilitate gemcitabine uptake and prolong retention in the cell, a lipophilic pro-drug was synthesized (CP-4126), with an elaidic fatty acid esterified at the 5'position. CP-4126 was tested in cell lines resistant to cytarabine, another dCyd analog or gemcitabine. Activity of gemcitabine and the derivative was comparable in the parent cell lines, while in dCK deficient cells all compounds were inactive. However, inhibition of nucleoside transport increased the IC(50) for gemcitabine up to 200-fold, but not for CP-4126, underlining the independence of a nucleoside transporter. For in vivo evaluation, nude mice bearing a human xenograft were treated intraperitoneally every third day for five doses at the maximal tolerated dose. In melanoma, sarcoma, lung, prostate, pancreatic and breast cancer xenografts, gemcitabine and CP-4126 were equally and highly effective; in four other xenografts moderately but equally active. In contrast to gemcitabine, CP-4126 could be administered orally, with a schedule and dose dependent toxicity and antitumor activity. In a colon cancer xenograft, antitumor activity of orally administered CP-4126 was equal to the intraperitoneally administered drug. In conclusion, CP-4126 is membrane transporter independent. Intraperitoneally administered CP-4126 was as effective as gemcitabine in several xenografts and CP-4126 is tolerated when orally administered. CP-4126 seems to be a promising new anticancer drug.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Fatty Acids/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Biological Availability , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytidine Deaminase , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Disease Models, Animal , Dogs , Humans , Mice , Nucleoside Deaminases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays , Gemcitabine
6.
Mol Cancer Ther ; 9(4): 1047-57, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20371715

ABSTRACT

Trifluorothymidine (TFT) is part of the novel oral formulation TAS-102, which is currently evaluated in phase II studies. Drug resistance is an important limitation of cancer therapy. The aim of the present study was to induce resistance to TFT in H630 colon cancer cells using two different schedules and to analyze the resistance mechanism. Cells were exposed either continuously or intermittently to TFT, resulting in H630-cTFT and H630-4TFT, respectively. Cells were analyzed for cross-resistance, cell cycle, protein expression, and activity of thymidine phosphorylase (TP), thymidine kinase (TK), thymidylate synthase (TS), equilibrative nucleoside transporter (hENT), gene expression (microarray), and genomic alterations. Both cell lines were cross-resistant to 2'-deoxy-5-fluorouridine (>170-fold). Exposure to IC(75)-TFT increased the S/G(2)-M phase of H630 cells, whereas in the resistant variants, no change was observed. The two main target enzymes TS and TP remained unchanged in both TFT-resistant variants. In H630-4TFT cells, TK protein expression and activity were decreased, resulting in less activated TFT and was most likely the mechanism of TFT resistance. In H630-cTFT cells, hENT mRNA expression was decreased 2- to 3-fold, resulting in a 5- to 10-fold decreased TFT-nucleotide accumulation. Surprisingly, microarray-mRNA analysis revealed a strong increase of secretory phospholipase-A2 (sPLA2; 47-fold), which was also found by reverse transcription-PCR (RT-PCR; 211-fold). sPLA2 inhibition reversed TFT resistance partially. H630-cTFT had many chromosomal aberrations, but the exact role of sPLA2 in TFT resistance remains unclear. Altogether, resistance induction to TFT can lead to different mechanisms of resistance, including decreased TK protein expression and enzyme activity, decreased hENT expression, as well as (phospho)lipid metabolism. Mol Cancer Ther; 9(4); 1047-57. (c)2010 AACR.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Equilibrative Nucleoside Transport Proteins/metabolism , Phospholipases A2, Secretory/metabolism , Thymidine Kinase/metabolism , Trifluridine/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Equilibrative Nucleoside Transport Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Oligonucleotide Array Sequence Analysis , Thymidine Kinase/genetics , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Trifluridine/chemistry , Trifluridine/metabolism , Up-Regulation/drug effects
7.
Mol Cancer Ther ; 6(10): 2807-15, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17938272

ABSTRACT

Basic fibroblast growth factor (bFGF) is a multifunctional protein and one of the most important growth factors in cutaneous melanoma development and progression. We hypothesized that high bFGF expression might be responsible for chemoresistance in advanced melanoma. M14 human melanoma cells expressing low levels of bFGF were successfully transfected with vectors encoding either the 18 kDa or all isoform proteins of bFGF. M14 cells and bFGF-overexpressing clones had a similar growth rate in vitro. Overexpression of 18 kDa or all isoform proteins of bFGF resulted in, respectively, 2.9- and 6.9-fold resistance against temozolomide. O6-alkylguanine-DNA-alkyltransferase (AGT) protein levels were highly elevated. Specific inhibition of AGT with O6-benzylguanine completely reversed the resistance in the 18 kDa clone, and partially in the clone overexpressing all isoforms. A methylation-specific PCR showed that at least in the 18 kDa overexpressing clone, increased AGT expression was the result of demethylation of the O6-methylguanine-DNA-methyltransferase promoter. In parental M14 cells, the demethylating agent 5-azacytidine generated AGT expression resulting in temozolomide resistance. Overexpression of all isoform proteins of bFGF, but not the 18 kDa isoform alone, resulted in 2.9-fold resistance against cisplatin, which could not be reversed by O6-benzylguanine. The expression levels of the mismatch repair proteins MSH2, MSH6, and MLH1 were not decreased, which likely excludes a defective mismatch repair system as a cause for cisplatin resistance. There were no changes in sensitivity to docetaxel and doxorubicin. In conclusion, bFGF overexpression can result in resistance against temozolomide mediated by demethylation of the O6-methylguanine-DNA-methyltransferase promoter.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Methylation , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Fibroblast Growth Factor 2/pharmacology , Melanoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Promoter Regions, Genetic/genetics , Antineoplastic Agents/pharmacology , Base Pair Mismatch , Blotting, Western , DNA Mismatch Repair , DNA Repair Enzymes/metabolism , Dacarbazine/pharmacology , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Luciferases/metabolism , Melanoma/enzymology , Melanoma/pathology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms , Temozolomide , Tumor Cells, Cultured/drug effects
8.
J Med Chem ; 50(9): 2249-53, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17419604

ABSTRACT

l-1,3-Dioxolane-cytidine, a potent anticancer agent against leukemia, has limited efficacy against solid tumors, perhaps due to its hydrophilicity. Herein, a library of prodrugs were synthesized to optimize in vitro antitumor activity against non-small cell lung cancer. N4-Substituted fatty acid amide prodrugs of 10-16 carbon chain length demonstrated significantly improved antitumor activity over l-1,3-dioxolane-cytidine. These in vitro results suggest that the in vivo therapeutic efficacy of l-1,3-dioxolane-cytidine against solid tumors may be improved with prodrug strategies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cytosine/analogs & derivatives , Dioxolanes/chemical synthesis , Lung Neoplasms/drug therapy , Prodrugs/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytosine/chemical synthesis , Cytosine/chemistry , Cytosine/pharmacology , Dioxolanes/chemistry , Dioxolanes/pharmacology , Drug Screening Assays, Antitumor , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Stereoisomerism , Structure-Activity Relationship
9.
Mol Pharmacol ; 66(4): 1054-60, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15269287

ABSTRACT

Platelet-derived endothelial cell growth factor/thymidine phosphorylase (PD-ECGF/TP) and interleukin-8 (IL-8) are angiogenic factors produced by tumor infiltrating macrophages. Here, we show that prolonged exposure of human monocytic/macrophage THP1 and U937 cells to sulfasalazine, an anti-inflammatory drug and inhibitor of nuclear factor-kappaB (NF-kappaB), resulted in down-regulation of PD-ECGF/TP and IL-8 (mRNA, protein and activity) along with elimination of their induction by tumor necrosis factor-alpha and interferon-gamma. Concomitantly, sulfasalazine-exposed cells were markedly resistant to 5'-deoxyfluorouridine, the last intermediate of capecitabine requiring activation by PD-ECGF/TP. This is the first report suggesting that disruption of NF-kappaB-dependent signaling pathways can provoke a marked and sustained down-regulation of macrophage-related angiogenic factors. However, this may also negatively affect capecitabine efficacy.


Subject(s)
Gene Expression/drug effects , Interleukin-8/metabolism , Macrophages/drug effects , Sulfasalazine/pharmacology , Thymidine Phosphorylase/metabolism , Angiogenesis Inducing Agents/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Humans , Interleukin-8/genetics , Macrophages/metabolism , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , NF-kappa B p50 Subunit , RNA, Messenger/metabolism , Receptors, Interferon/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Thymidine Phosphorylase/genetics , Transcription Factor RelA , Transcription Factors/metabolism , U937 Cells , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...