Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 20(3): 1247-51, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20022750

ABSTRACT

New hybrid molecules of isothiocyanate and progesterone and their metal complexes were synthesized exhibiting promising anti-proliferative and pro-apoptotic activity against breast and prostate cancer cells. These metal complex compounds exploited an existing cellular transport pathway for delivery of cytotoxic isothiocyanate moiety across cell membrane resulting in the inhibition of cell viability and inducing apoptosis. The highest apoptotic action was shown by the copper complex, which was mediated through the inhibition of Akt signaling similar to the one shown by isothiocyanate compounds. Our results underscore the possible role of metal redox cycling, and thus it is likely will open newer avenues for further optimization for the synthesis of novel active compounds through appropriate isothiocyanate pharmacophores.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Isothiocyanates/chemistry , Isothiocyanates/toxicity , Progesterone/chemistry , Progesterone/toxicity , Recombinant Fusion Proteins/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Humans , Isothiocyanates/administration & dosage , Progesterone/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry
2.
Cancer ; 110(12): 2775-84, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17948911

ABSTRACT

BACKGROUND: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have demonstrated clinical benefit in patients with nonsmall cell lung cancer (NSCLC), particularly those with tumors that have EGFR-TK domain mutations. Moreover, the EGFR and cyclooxygenase (COX)-2 pathways are known to enhance the procarcinogenic effects of each other in different tumor types. Therefore, it was hypothesized that tumor EGFR mutation status may influence the effectiveness of simultaneous EGFR and COX-2 inhibition in patients with NSCLC. METHODS: Three NSCLC cell lines with varying EGFR mutation status and sensitivities to EGFR-TKIs were selected: H3255 (L858R), H1650 (del E746-A750), and H1781 (wild-type EGFR). Cells were treated with erlotinib, gefitinib, or celecoxib alone, and the combination of both EGFR-TKI inhibitors with celecoxib. Cell survival and apoptosis was assessed and correlated with the expression of COX-2, EGFR, pEGFR, Akt, pAkt, expression, and derived prostaglandin E2 (PGE(2)). RESULTS: Celecoxib by itself was found to have no effects on cell growth or apoptosis in any of the cell lines. Erlotinib and gefitinib inhibited cell growth and induced apoptosis in both mutant cell lines and did so in H1781 cells at 10-fold higher concentrations. Celecoxib when added to erlotinib or gefitinib significantly enhanced the antiproliferative and proapoptotic effects in both mutant cell lines but had no additional effects in H1781 cells. Greater down-regulation of COX-2, EGFR, pEGFR, Akt, pAkt, and PGE(2) was found when H3255 cells were treated with the combination compared with any of the single agents alone. CONCLUSIONS: The results of the current study demonstrate that the effectiveness of the addition of celecoxib to an EGFR-TKI is significantly greater in NSCLC cells with EGFR mutations, which is likely due to more complete inhibition of both pathways.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Genes, erbB-1 , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Apoptosis/drug effects , Celecoxib , Cell Division/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Erlotinib Hydrochloride , Gefitinib , Humans , Prostaglandins E/metabolism , Pyrazoles/pharmacology , Quinazolines/pharmacology , Sulfonamides/pharmacology , Tumor Cells, Cultured
3.
J Inorg Biochem ; 101(10): 1517-24, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17689613

ABSTRACT

Copper conjugates of Schiff base derivatives of nimesulide (1), a well-known cyclooxygenase-2 (COX-2) inhibitor, were synthesized, structurally characterized and evaluated for their COX selectivity indices and cytotoxicities on pancreatic tumor, BxPC-3 (COX-2 positive) and MiaPaCa (COX-2 negative) cell lines. Copper conjugates exhibit distorted square planar geometries as revealed by the single crystal X-ray structure determination of Cu(L1)(2) and show significant growth inhibition in both cell lines (IC50 values 3-26 microM for COX-2 positive and 5-9 microM for COX-2 negative cell line) than the parent nimesulide (35 microM for COX-2 positive and >100 microM for COX-2 negative cell line). The mechanistic pathway for the biological activity involves inhibition of vascular endothelial growth factor (VEGF) and COX inhibition, as well as down regulation of antiapoptotic Bcl-2 and Bcl-(XL) proteins.


Subject(s)
Copper/chemistry , Cyclooxygenase 2/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Schiff Bases , Sulfonamides/chemistry , Vascular Endothelial Growth Factor A/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Electron Spin Resonance Spectroscopy , Humans , Models, Molecular , Spectrophotometry, Infrared
4.
Clin Cancer Res ; 13(9): 2811-21, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473215

ABSTRACT

PURPOSE: Pancreatic cancer is a highly aggressive disease that remains refractory to various chemotherapeutic agents. Because the proto-oncogene c-myc can modulate apoptosis in response to cytotoxic insults and is commonly overexpressed in pancreatic cancer, we investigated the value of c-myc as a potential modulator of cellular response to various chemotherapeutic agents. EXPERIMENTAL DESIGN: Stable overexpression or small interfering RNA (siRNA)-mediated knockdown of c-myc and restoration of cyclin D1 were done in the Ela-myc pancreatic tumor cell line. Cell viability after cisplatin treatment of c-myc-overexpressing, control, and siRNA-transfected cells was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and drug-induced apoptosis was measured by DNA fragmentation, sub-G(1), and poly(ADP-ribose) polymerase cleavage analyses. Protein expression profile after cisplatin treatment was determined by Western blotting and DNA binding activity of nuclear factor-kappaB was examined by electrophoretic mobility shift assay. RESULTS: Ectopic overexpression of c-myc in murine and human pancreatic cancer cell lines, Ela-myc and L3.6pl, respectively, resulted in increased sensitivity to cisplatin and other chemotherapeutic drugs. Increased sensitivity to cisplatin in c-myc-overexpressing cells was due, in part, to the marked increase in cisplatin-induced apoptosis. Conversely, down-regulation of c-myc expression in stable c-myc-overexpressing cells by c-myc siRNA resulted in decreased sensitivity to cisplatin-induced cell death. These results indicate an important role of c-myc in chemosensitivity of pancreatic cancer cells. The c-myc-induced cisplatin sensitivity correlated with inhibition of nuclear factor kappaB activity, which was partially restored by ectopic cyclin D1 overexpression. CONCLUSIONS: Our results suggest that the c-myc-dependent sensitization to chemotherapy-induced apoptosis involves suppression of cyclin D1 expression and nuclear factor kappaB activity.


Subject(s)
Cyclin D1/antagonists & inhibitors , Drug Resistance, Neoplasm , NF-kappa B/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Fragmentation , Drug Resistance, Neoplasm/genetics , Humans , Mice , NF-kappa B/metabolism , Proto-Oncogene Mas , RNA, Small Interfering/genetics
5.
Int J Cancer ; 120(4): 734-42, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17131340

ABSTRACT

Pancreatic cancer (PC) is characterized as one of the deadliest malignancies and its treatment is a great challenge to clinical oncologists. Expression of COX-2 is detectable in 75% of PCs among which 50% showed overexpression, suggesting the importance of COX-2 enzyme and its metabolic product prostaglandin E2 (PGE(2)) in PC. Here the authors report the synthesis and biological activity of a novel COX-2 inhibitor, FPA-306, and its effects on PC cells with different levels of COX-2 expression. Using MTT assay, the authors found a significant growth inhibition of BxPC-3 cells treated by FPA-306 with an IC(50) of 10 micromol/L, which was lower than that of ketoprofen (IC(50) = 35.4 micromol/L) and celecoxib (IC(50) > 100 micromol/L). There was no such effect found in MIAPaCa cell line, which does not express COX-2. The authors also found dose dependent reduction in cell survival and induction of apoptosis by FPA-306 treatment in BxPC-3 cells but not in MIAPaCa cells. These results were correlated with apoptosis data and secreted PGE(2) levels. The molecular modeling of FPA-306 in the COX-2 active site showed that FPA-306 is potentially able to inhibit the activity of enzyme by blocking the active site, thereby resulting in decreased PGE(2) production. The authors also found a significant reduction of COX-2 at the mRNA and protein levels together with downregulation of NF-kappaB DNA binding activity and its downstream genes, Bcl-2 and survivin. These results suggest that FPA-306 is an effective and potent agent in inhibiting the growth of PC cells.


Subject(s)
Apoptosis/drug effects , Copper/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Membrane Proteins/antagonists & inhibitors , Organometallic Compounds/pharmacology , Pancreatic Neoplasms/drug therapy , Binding Sites , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Dinoprostone/metabolism , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoenzyme Techniques , Membrane Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenylacetates/chemistry , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured/drug effects
6.
J Med Chem ; 49(24): 7242-6, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-17125278

ABSTRACT

We report the synthesis of novel 1:1 Schiff base copper complexes of quinoline-2-carboxaldehyde showing dose-dependent, antiproliferative, and proapoptotic activity in PC-3 and LNCaP prostate cancer cells. We found that quinoline thiosemicarbazone 2 (FPA-137) was the most potent and inhibited proteosome activity in intact human prostate cancer PC-3 and LNCaP cells (IC50 of 4 and 3.2 microM, respectively) compared to clioquinol and pyrrolidine dithiocarbamate (IC50 of 10 and 20 microM), supporting the novelty of 2.


Subject(s)
Aldehydes/chemistry , Copper , Organometallic Compounds/chemical synthesis , Proteasome Inhibitors , Quinolines/chemistry , Schiff Bases/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Male , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Prostatic Neoplasms , Semicarbazides/chemistry , Structure-Activity Relationship
7.
Cancer Res ; 66(20): 10064-72, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047070

ABSTRACT

Despite the initial efficacy of androgen deprivation therapy, most patients with advanced prostate cancer eventually progress to hormone-refractory prostate cancer, for which there is no curative therapy. Previous studies from our laboratory and others have shown the antiproliferative and proapoptotic effects of 3,3'-diindolylmethane (DIM) in prostate cancer cells. However, the molecular mechanism of action of DIM has not been investigated in androgen receptor (AR)-positive hormone-responsive and -nonresponsive prostate cancer cells. Therefore, we investigated the effects of B-DIM, a formulated DIM with greater bioavailability, on AR, Akt, and nuclear factor kappaB (NF-kappaB) signaling in hormone-sensitive LNCaP (AR+) and hormone-insensitive C4-2B (AR+) prostate cancer cells. We found that B-DIM significantly inhibited cell proliferation and induced apoptosis in both cell lines. By Akt gene transfection, reverse transcription-PCR, Western blot analysis, and electrophoretic mobility shift assay, we found a potential crosstalk between Akt, NF-kappaB, and AR. Importantly, B-DIM significantly inhibited Akt activation, NF-kappaB DNA binding activity, AR phosphorylation, and the expressions of AR and prostate-specific antigen, suggesting that B-DIM could interrupt the crosstalk. Confocal studies revealed that B-DIM inhibited AR nuclear translocation, leading to the down-regulation of AR target genes. Moreover, B-DIM significantly inhibited C4-2B cell growth in a severe combined immunodeficiency-human model of experimental prostate cancer bone metastasis. These results suggest that B-DIM-induced cell proliferation inhibition and apoptosis induction are partly mediated through the down-regulation of AR, Akt, and NF-kappaB signaling. These observations provide a rationale for devising novel therapeutic approaches for the treatment of hormone-sensitive, but more importantly, hormone-refractory prostate cancer by using B-DIM alone or in combination with other therapeutics.


Subject(s)
Androgen Receptor Antagonists , Indoles/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Down-Regulation/drug effects , Humans , Male , NF-kappa B/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostate-Specific Antigen/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/biosynthesis , Receptors, Androgen/metabolism , Signal Transduction/drug effects
8.
Int J Cancer ; 119(9): 2071-7, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16823852

ABSTRACT

Notch is an ancient cell signaling system that regulates cell fate specification, stem cell maintenance and initiation of differentiation in many tissues. It has been reported that Jagged-1, a Notch ligand, is significantly over expressed in metastatic prostate cancer compared to localized prostate cancer or benign prostatic tissues. Therefore, deregulation of Jagged-1 protein levels may play a role in prostate cancer cell growth and progression. Hence, the aim of our current study was to investigate the mechanistic role of Jagged-1 in prostate cancer cell growth and cell cycle progression. Our results show, for the first time, that down-regulation of Jagged-1 induces cell growth inhibition and S phase cell cycle arrest in prostate cancer cells, with reduced CDK2 kinase activity and increased p27 expression. These results suggest that Jagged-1 could be a potential therapeutic target for the treatment of prostate cancer.


Subject(s)
Calcium-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Prostatic Neoplasms/pathology , Cell Division , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Jagged-1 Protein , Male , Prostatic Neoplasms/enzymology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins
9.
J Med Chem ; 49(13): 3800-8, 2006 Jun 29.
Article in English | MEDLINE | ID: mdl-16789737

ABSTRACT

The synthesis and characterization of Schiff base derivatives of 3-formylchromone 3-6 (FPA-120 to FPA-123), the minimal biologically active structural motif of soy isoflavone, genistein, and their copper(II) complexes 7-10 (FPA-124 to FPA-127) are reported here. These copper complexes possess distorted square-planar geometries capable of stabilizing Cu2+/Cu+ redox forms. The molecular modeling study revealed that the key interaction of the metal complexes was with amino acids in the pleckstrin homology (PH) and the kinase domain of the PKB (Akt) protein. Copper complex 7 significantly forms stronger charge interactions in the kinase domain than genistein, leading to better stabilization in the active pocket. In vitro evaluation of copper complexes against hormone-independent and metastatic breast (BT20), prostate (PC-3), and K-ras mutant (COLO 357) and K-ras wild-type (BxPC-3) pancreatic cancer cells revealed that 7 was the most potent compound which exhibited PKB (Akt protein) inhibitory activities and caused NF-kappaB inactivation in a well-established orthotopic pancreatic tumor model using COLO 357 cells. An inverse relationship was observed between IC50 values of the anti-proliferative activities and the Cu2+/Cu+ redox couple for these compounds, which may provide a rapid screen for evaluating the efficacy of active metallodrugs affecting redox-sensitive transcription factors such as NF-kappaB and its upstream target, the PKB (Akt) pathway, in multiple cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chromones/chemical synthesis , Copper , Organometallic Compounds/chemical synthesis , Schiff Bases/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Chromones/chemistry , Drug Screening Assays, Antitumor , Female , Fluorescence Polarization , Genistein/analogs & derivatives , Genistein/chemical synthesis , Genistein/chemistry , Genistein/pharmacology , Humans , Mice , Mice, Nude , Models, Molecular , NF-kappa B/antagonists & inhibitors , Neoplasm Transplantation , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Oxidation-Reduction , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry , Schiff Bases/chemistry , Structure-Activity Relationship
10.
Cancer Res ; 65(15): 6934-42, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061678

ABSTRACT

Cancer chemotherapeutic strategies commonly require multiple agents. However, use of multiple agents contributes to added toxicity resulting in poor treatment outcome. Thus, combination chemotherapy must be optimized to increase tumor response and at the same time lower its toxicity. Chemotherapeutic agents are known to induce nuclear factor kappaB (NF-kappaB) activity in tumor cells, resulting in lower cell killing and drug resistance. In contrast, genistein has been shown to inhibit the activity of NF-kappaB and the growth of various cancer cells without causing systemic toxicity. We therefore investigated whether the inactivation of NF-kappaB by genistein before treatment of various cancer cells with chemotherapeutic agents could lead to better tumor cell killing as tested by in vitro studies using gene transfections and also by animal studies. PC-3 (prostate), MDA-MB-231 (breast), H460 (lung), and BxPC-3 (pancreas) cancer cells were pretreated with 15 to 30 micromol/L genistein for 24 hours and then exposed to low doses of chemotherapeutic agents for an additional 48 to 72 hours. We found that 15 to 30 micromol/L genistein combined with 100 to 500 nmol/L cisplatin, 0.5 to 2 nmol/L docetaxel, or 50 ng/mL doxorubicin resulted in significantly greater inhibition of cell growth and induction of apoptosis compared with either agent alone. Moreover, we found that the NF-kappaB activity was significantly increased within 2 hours of cisplatin and docetaxel treatment and that the NF-kappaB inducing activity of these agents was completely abrogated in cells pretreated with genistein. These results were also supported, for the first time, by animal experiments, p65 cDNA transfection and p65 small interfering RNA studies, which clearly showed that a specific target (NF-kappaB) was affected in vivo. Collectively, our results clearly suggest that genistein pretreatment inactivates NF-kappaB and may contribute to increased growth inhibition and apoptosis induced by cisplatin, docetaxel, and doxorubicin in prostate, breast, lung, and pancreatic cancer cells. Theses results warrant carefully designed clinical studies investigating the combination of soy isoflavones and commonly used chemotherapeutic agents for the treatment of human cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Genistein/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacology , Docetaxel , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Genistein/administration & dosage , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Taxoids/administration & dosage , Taxoids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...