Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 25(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38732265

ABSTRACT

Epigenetic alterations my play a role in the aggressive behavior of Non-Small Cell Lung Cancer (NSCLC). Treatment with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA, vorinostat) has been reported to interfere with the proliferative and invasive potential of NSCLC cells. In addition, the DNA methyltransferase inhibitor azacytidine (AZA, vidaza) can modulate the levels of the metastasis suppressor KiSS-1. Thus, since cisplatin is still clinically available for NSCLC therapy, the aim of this study was to evaluate drug combinations between cisplatin and SAHA as well as AZA using cisplatin-sensitive H460 and -resistant H460/Pt NSCLC cells in relation to KiSS-1 modulation. An analysis of drug interaction according to the Combination-Index values indicated a more marked synergistic effect when the exposure to SAHA or AZA preceded cisplatin treatment with respect to a simultaneous schedule. A modulation of proteins involved in apoptosis (p53, Bax) was found in both sensitive and resistant cells, and compared to the treatment with epigenetic agents alone, the combination of cisplatin and SAHA or AZA increased apoptosis induction. The epigenetic treatments, both as single agents and in combination, increased the release of KiSS-1. Finally, the exposure of cisplatin-sensitive and -resistant cells to the kisspeptin KP10 enhanced cisplatin induced cell death. The efficacy of the combination of SAHA and cisplatin was tested in vivo after subcutaneous inoculum of parental and resistant cells in immunodeficient mice. A significant tumor volume inhibition was found when mice bearing advanced tumors were treated with the combination of SAHA and cisplatin according to the best schedule identified in cellular studies. These results, together with the available literature, support that epigenetic drugs are amenable for the combination treatment of NSCLC, including patients bearing cisplatin-resistant tumors.


Subject(s)
Azacitidine , Cisplatin , Drug Resistance, Neoplasm , Epigenesis, Genetic , Kisspeptins , Lung Neoplasms , Vorinostat , Cisplatin/pharmacology , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Humans , Mice , Epigenesis, Genetic/drug effects , Kisspeptins/metabolism , Kisspeptins/pharmacology , Kisspeptins/genetics , Cell Line, Tumor , Vorinostat/pharmacology , Azacitidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Drug Synergism , Histone Deacetylase Inhibitors/pharmacology , Female
2.
Org Biomol Chem ; 21(32): 6572-6587, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37526931

ABSTRACT

N-Oxyamides of bioactive anionic glycoglycerolipids based on 2-O-ß-D-glucosylglycerol were efficiently prepared. However, the oxidation step of the primary hydroxyl group of the glucose moiety in the presence of the N-oxyamide function appeared to be a difficult task that was nevertheless conveniently achieved for the first time by employing a chemoenzymatic laccase/TEMPO procedure. The obtained N-oxyamides exhibited a higher inhibition of proliferation of ovarian carcinoma IGROV-1 cells in serum-free medium than in complete medium, similarly to the corresponding bioactive esters. Stability and serum binding studies indicated that the observed reduced activity of the compounds in complete medium could be mainly due to a binding effect of serum proteins rather than the hydrolytic degradation of glycoglycerolipid acyl chains. Furthermore, the results of the cellular studies under serum-free conditions suggested that the N-oxyamide group could increase the antiproliferative activity of a glycoglycerolipid independently of the presence of the anionic carboxylic group. Cellular studies in other cell lines besides IGROV-1 also support a certain degree of selectivity of this series of compounds for tumor cells with Akt hyperactivation.


Subject(s)
Ovarian Neoplasms , Proto-Oncogene Proteins c-akt , Female , Humans , Glycolipids/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Cell Line
3.
Toxicol Appl Pharmacol ; 310: 9-19, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27554045

ABSTRACT

The occurrence of drug resistance limits the efficacy of platinum compounds in the cure of ovarian carcinoma. Since microRNAs (miRNAs) may contribute to this phenomenon by regulating different aspects of tumor cell response, the aim of this study was to exploit the analysis of expression of miRNAs in platinum sensitive/resistant cells in an attempt to identify potential regulators of drug response. MiR-483-3p, which may participate in apoptosis and cell proliferation regulation, was found up-regulated in 4 platinum resistant variants, particularly in the IGROV-1/Pt1 subline, versus parental cells. Transfection of a synthetic precursor of miR-483-3p in IGROV-1 parental cells elicited a marked up-regulation of the miRNA levels. Growth-inhibition and colony-forming assays indicated that miR-483-3p over-expression reduced cell growth and conferred mild levels of cisplatin resistance in IGROV-1 cells, by interference with their proliferative potential. Predicted targets of miR-483-3p included PRKCA (encoding PKC-alpha), previously reported to be associated to platinum-resistance in ovarian carcinoma. We found that miR-483-3p directly targeted PRKCA in IGROV-1 cells. In keeping with this finding, cisplatin sensitivity of IGROV-1 cells decreased upon molecular/pharmacological inhibition of PKC-alpha. Overall, our results suggest that overexpression of miR-483-3p by ovarian carcinoma platinum-resistant cells may interfere with their proliferation, thus protecting them from DNA damage induced by platinum compounds and ultimately representing a drug-resistance mechanism. The impairment of cell growth may account for low levels of drug resistance that could be relevant in the clinical setting.


Subject(s)
MicroRNAs/genetics , Ovarian Neoplasms/drug therapy , Platinum Compounds/therapeutic use , Protein Kinase C-alpha/metabolism , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/genetics
4.
Bioorg Med Chem ; 22(3): 1089-103, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24398383

ABSTRACT

7-Azaindole-1-carboxamides were designed as a new class of PARP-1 inhibitors. The compounds displayed a variable pattern of target inhibition profile that, in part, paralleled the antiproliferative activity in cell lines characterized by homologous recombination defects. A selected compound (1l; ST7710AA1) showed significant in vitro target inhibition and capability to substantially bypass the multidrug resistance mediated by Pgp. In antitumor activity studies against the MX1 human breast carcinoma growth in nude mice, the compound exhibited an effect similar to that of Olaparib in terms of tumor volume inhibition when used at a lower dose than the reference compound. Treatment was well tolerated, as no deaths or significant weight losses were observed among the treated animals.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Multiple/drug effects , Enzyme Inhibitors/chemical synthesis , Female , HeLa Cells/drug effects , Homologous Recombination , Humans , Indoles/chemistry , Mice , Mice, Nude , Models, Molecular , Poly (ADP-Ribose) Polymerase-1 , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Bioconjug Chem ; 23(8): 1610-22, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22770429

ABSTRACT

The efficacy of taxane-based antitumor therapy is limited by several drawbacks which result in a poor therapeutic index. Thus, the development of approaches that favor selective delivery of taxane drugs (e.g., paclitaxel, PTX) to the disease area represents a truly challenging goal. On the basis of the strategic role of integrins in tumor cell survival and tumor progression, as well as on integrin expression in tumors, novel molecular conjugates were prepared where PTX is covalently attached to either cyclic AbaRGD (Azabicycloalkane-RGD) or AmproRGD (Aminoproline-RGD) integrin-recognizing matrices via structurally diverse connections. Receptor-binding assays indicated satisfactory-to-excellent α(V)ß(3) binding capabilities for most conjugates, while in vitro growth inhibition assays on a panel of human tumor cell lines revealed outstanding cell sensitivity values. Among the nine conjugate ensemble, derivative 21, bearing a robust triazole ring connected to ethylene glycol units by an amide function and showing excellent cell sensitivity properties, was selected for in vivo studies in an ovarian carcinoma model xenografted in immunodeficient mice. Remarkable antitumor activity was attained, superior to that of PTX itself, which was associated with a marked induction of aberrant mitoses, consistent with the mechanism of action of spindle poisons. Overall, the novel cRGD-PTX conjugates disclosed here represent promising candidates for further advancement in the domain of targeted antitumor therapy.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemical synthesis , Drug Design , Integrin alphaVbeta3/metabolism , Paclitaxel/chemistry , Peptides, Cyclic/chemical synthesis , Receptors, Vitronectin/metabolism , Amides/chemistry , Animals , Antineoplastic Agents/pharmacology , Azabicyclo Compounds/chemistry , Calibration , Cell Line, Tumor , Chemistry Techniques, Synthetic , Drug Carriers/chemistry , Drug Carriers/metabolism , Female , Humans , Inhibitory Concentration 50 , Mice , Paclitaxel/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Proline/analogs & derivatives , Proline/chemistry , Xenograft Model Antitumor Assays
6.
Transl Lung Cancer Res ; 11(7): 1315-1326, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35958339

ABSTRACT

Background: The secreted products of the metastasis suppressor gene KiSS1 may represent useful biomarkers in non-small cell lung cancer (NSCLC) but their levels in patients have remained poorly investigated. We previously found that forced expression of KiSS1 decreased the invasive capability of NSCLC drug-resistant cells and a pro-apoptotic role for KiSS1 has been proposed in head and neck cancer. Thus, we designed a translational investigation including a pilot study to analyze KiSS1 levels in liquid biopsies, and in vitro experiments to explore the biological relevance of KiSS1 modulation. Methods: KiSS1-derived peptide levels in liquid biopsies from 60 NSCLC patients were assayed by ELISA. Preclinical experiments were carried out using quantitative real time polymerase chain reaction (qRT-PCR), ELISA, annexin V-binding and caspase activation assays. Results: We compared KiSS1 release in 3 different matrices (serum, plasma and urine) and the highest levels were detectable in serum (range, 0-4.5 ng/mL). We observed increased levels of seric KiSS1 in NSCLC patients as compared to healthy donors. KiSS1 serum concentrations, after surgical procedure and/or adjuvant therapy. We observed differences among disease stages in urine samples. In preclinical models, KiSS1 mRNA levels were increased by short term exposure to azacytidine, enhanced KiSS1 release was induced by the combination of azacytidine and cisplatin and KiSS1-derived peptides enhanced cisplatin-induced apoptosis. KiSS1 increase was observed upon exposure neurons-enriched cultures to tumor cell conditioned medium. Conclusions: Our results showing a peculiar modulation of KiSS1 levels in liquid biopsies of NSCLC patients and a regulation of cisplatin-induced apoptosis by KiSS1-derived peptides support an involvement of KiSS1 in cell response to treatment and highlight its promising features as a potential biomarker in NSCLC.

7.
Front Cell Dev Biol ; 10: 1055067, 2022.
Article in English | MEDLINE | ID: mdl-36578788

ABSTRACT

The identification of therapeutic approaches to improve response to platinum-based therapies is an urgent need for ovarian carcinoma. Deubiquitinases are a large family of ubiquitin proteases implicated in a variety of cellular functions and may contribute to tumor aggressive features through regulation of processes such as proliferation and cell death. Among the subfamily of ubiquitin-specific peptidases, USP8 appears to be involved in modulation of cancer cell survival by still poorly understood mechanisms. Thus, we used ovarian carcinoma cells of different histotypes, including cisplatin-resistant variants with increased survival features to evaluate the efficacy of molecular targeting of USP8 as a strategy to overcome drug resistance/modulate cisplatin response. We performed biochemical analysis of USP8 activity in pairs of cisplatin-sensitive and -resistant cells and found increased USP8 activity in resistant cells. Silencing of USP8 resulted in decreased activation of receptor tyrosine kinases and increased sensitivity to cisplatin in IGROV-1/Pt1 resistant cells as shown by colony forming assay. Increased cisplatin sensitivity was associated with enhanced cisplatin-induced caspase 3/7 activation and apoptosis, a phenotype also observed in cisplatin sensitive cells. Increased apoptosis was linked to FLIPL decrease and cisplatin induction of caspase 3 in IGROV-1/Pt1 cells, cisplatin-induced claspin and survivin down-regulation in IGROV-1 cells, thereby showing a decrease of anti-apoptotic proteins. Immunohistochemical staining on 65 clinical specimens from advanced stage ovarian carcinoma indicated that 40% of tumors were USP8 positive suggesting that USP8 is an independent prognostic factor for adverse outcome when considering progression free survival as a clinical end-point. Taken together, our results support that USP8 may be of diagnostic value and may provide a therapeutic target to improve the efficacy of platinum-based therapy in ovarian carcinoma.

8.
Biochem Pharmacol ; 197: 114900, 2022 03.
Article in English | MEDLINE | ID: mdl-34995485

ABSTRACT

Deubiquitinases (DUBs) mediate the removal of ubiquitin from diverse proteins that participate in the regulation of cell survival, DNA damage repair, apoptosis and drug resistance. Previous studies have shown an association between activation of cell survival pathways and platinum-drug resistance in ovarian carcinoma cell lines. Among the strategies available to inhibit DUBs, curcumin derivatives appear promising, thus we hypothesized their use to enhance the efficacy of cisplatin in ovarian carcinoma preclinical models. The caffeic acid phenethyl ester (CAPE), inhibited ubiquitin-specific protease 8 (USP8), but not proteasomal DUBs in cell-free assays. When CAPE was combined with cisplatin in nine cell lines representative of various histotypes a synergistic effect was observed in TOV112D cells and in the cisplatin-resistant IGROV-1/Pt1 variant, both of endometrioid type and carrying mutant TP53. In the latter cells, persistent G1 accumulation upon combined treatment associated with p27kip1 protein levels was observed. The synergy was not dependent on apoptosis induction, and appeared to occur in cells with higher USP8 levels. In vivo antitumor activity studies supported the advantage of the combination of CAPE and cisplatin in the subcutaneous model of cisplatin-resistant IGROV-1/Pt1 ovarian carcinoma as well as CAPE activity on intraperitoneal disease. This study reveals the therapeutic potential of CAPE in cisplatin-resistant ovarian tumors as well as in tumors expressing USP8.


Subject(s)
Antineoplastic Agents/administration & dosage , Caffeic Acids/administration & dosage , Cisplatin/administration & dosage , Endopeptidases/biosynthesis , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/biosynthesis , Ovarian Neoplasms/enzymology , Phenylethyl Alcohol/analogs & derivatives , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/biosynthesis , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/drug therapy , Phenylethyl Alcohol/administration & dosage , Xenograft Model Antitumor Assays/methods
9.
J Proteome Res ; 10(3): 1191-207, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21142107

ABSTRACT

Novel agents characterized by the scaffold of the atypical retinoid ST1926, but containing different chemical functions (carboxylic or hydroxamic acid), exhibit potent proapoptotic activity. In the present paper, we show that the treatment of the IGROV-1 ovarian cancer cell line with compounds sharing structural features with ST1926 (ST1898, ST3595, ST3056) determines a strong inhibition of proliferation mainly due to apoptotic cell death. In an effort to understand the mechanism of action of these compounds, we performed a proteomics analysis of IGROV-1 total lysates and nuclear extracts. Using this approach, we found that deregulation of calcium homeostasis, oxidative stress, cytoskeleton reorganization, and deregulation of proteasome function may represent important pathways involved in response of IGROV-1 cells to the studied compounds. The most prominent effect was down-regulation of factors involved in protein degradation, an event more marked in cells treated with ST3595. In addition, we identified proteins specifically modulated by each treatment, including prohibitin and cochaperone P23 (ST1898), pre-mRNA splicing factor SF2p32 and clathrin light chain (ST3595), as well as Far upstream element (FUSE) binding protein 1 and DNA-binding protein B (ST3056). By identifying proteins modulated by novel proapoptotic agents, this study provides insights into critical aspects of their mechanism of action.


Subject(s)
Apoptosis/drug effects , Cell Line, Tumor/drug effects , Ovarian Neoplasms/pathology , Proteome/analysis , Proteomics/methods , Retinoids/chemistry , Retinoids/pharmacology , Cell Cycle/drug effects , Chromatography, High Pressure Liquid/methods , Electrophoresis, Gel, Two-Dimensional/methods , Female , Humans , Molecular Structure , Tandem Mass Spectrometry/methods
10.
BMC Genomics ; 12: 44, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21247416

ABSTRACT

BACKGROUND: The ubiquitin(Ub)-proteasome pathway is implicated in the regulation of a variety of cellular functions and plays a major role in stress response in eukaryotic cells, by targeting misfolded and damaged proteins for degradation. In addition, in the presence of DNA damage, the Ub-proteasome system regulates proteins involved in sensing, repairing, and/or tolerating the damage. Antitumor agents such as cisplatin can activate the pathway, but the role of specific pathway components in cell sensitivity/response to the drug is not known. Since platinum compounds represent clinically relevant antitumor agents and a major limitation to their use is the development of drug resistance, there is an urgent need for identifying targets for improving their efficacy. RESULTS: In the present study, we performed a genome-wide screening for sensitivity to cisplatin using non-essential haploid deletion mutants of the fission yeast Schizosaccharomyces pombe, belonging to a collection of haploid strains constructed through homologous recombination. Using this approach, we identified three Ub-proteasome mutants exhibiting hypersensitivity to cisplatin (ubp16, ubc13 and pmt3) and ten mutants (including ufd2, beta7 20S, rpt6/let1) resistant to the drug. In addition, the importance of lub1 gene emerged from the comparison between the present screening and gene expression profile data previously obtained in fission yeast. CONCLUSIONS: The factors identified in the present study allowed us to highlight most finely the close relationship between the Ub-proteasome system and DNA damage response mechanisms, thus establishing a comprehensive framework of regulators likely relevant also in higher eukaryotes. Our results provide the proof of principle of the involvement of specific genes modulated by cisplatin treatment in cell response to the drug, suggesting their potential role as targets for modulating cisplatin sensitivity. In this regard, the prospective identification of novel targets for modulation of cisplatin sensitivity in an eukaryotic model organism appears particularly intriguing towards the discovery of strategies to overcome cisplatin resistance in human tumors.


Subject(s)
Cisplatin/pharmacology , Proteasome Endopeptidase Complex/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/drug effects , Schizosaccharomyces/immunology , Ubiquitin/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
11.
Mol Pharm ; 7(1): 207-16, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19919086

ABSTRACT

Multinuclear platinum complexes are characterized by a peculiar DNA binding mode and higher cytotoxic potency than the mononuclear complexes, and efficacy against a wide range of preclinical tumor models. To reduce the high irreversible plasma protein binding and improve the chemical and metabolic drug stability, novel bis-platinum complexes were designed starting from the parent compound CT-3610. The novel second-generation bis-platinum complexes utilize alkylcarboxylate as leaving groups to improve their pharmacokinetic and pharmacodynamic profiles, thus overcoming the limitations of the previously developed multinuclear compounds. The selected compounds [CT-47518 and CT-47463, respectively (bis-capronate) platinum and (bis-butyrate) platinum], have similar in vitro degradation kinetics in human and murine plasma and, above all, an increased stability when compared to CT-3610, particularly in human plasma. In addition, both compounds exhibited a marked cytotoxic potency as compared with cisplatin and oxaliplatin. Interestingly, they were capable of overcoming resistance mediated by DNA mismatch repair defects in different cellular models. The complexes showed marked antitumor efficacy in Pt-refractory tumor xenografts, with remarkable activity in terms of tumor growth inhibition and tumor growth delay. The improved stability profile in human plasma compared to early bis- and triplatinum complexes together with the marked activity in cellular systems as well as in in vivo models, make CT-47518 and CT-47463 attractive candidates for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , DNA Adducts/metabolism , DNA Mismatch Repair , Drug Design , Drug Resistance, Neoplasm , Drug Stability , Female , Humans , Mice , Mice, Nude , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin , Xenograft Model Antitumor Assays
12.
Bioorg Med Chem ; 18(10): 3543-50, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20409723

ABSTRACT

The naturally occurring coumarins and resveratrol, attract great attention due to their wide range of biological properties, including anticancer, antileukemic, antibacterial and anti-inflammatory activities; moreover, their cancer chemopreventive property have been recently emphasized. A novel class of hybrid compounds, obtained by introducing a substituted trans-vinylbenzene moiety on a coumarin backbone, was synthesized and evaluated for the antitumor profile. A number of derivatives showed a good antiproliferative activity, in some cases higher to that of the reference compound resveratrol. The most promising compounds in this series were 14 and 17, endowed with excellent antiproliferative and proapoptotic activities. The present study suggests that the 7-methoxycoumarin nucleus, together with the 3,5-disubstitution pattern of the trans-vinylbenzene moiety, are likely promising structural features to obtain excellent antitumor compounds endowed with a apoptosis-inducing capability.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Stilbenes/pharmacology , Anticarcinogenic Agents , Antineoplastic Agents/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants , Cell Line, Tumor , Chimera , Coumarins/chemistry , Coumarins/pharmacology , Drug Design , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Oxides/pharmacology , Resveratrol , Stilbenes/chemistry , Structure-Activity Relationship
13.
Front Cell Dev Biol ; 8: 610, 2020.
Article in English | MEDLINE | ID: mdl-32754596

ABSTRACT

In spite of new knowledge on prostate cancer molecular landscape, this has been only partially translated to the therapeutic setting. The activation of Ras/Mitogen-activated protein kinase (MAPK) signaling plays an important role in progression of prostate cancer in which deregulation of histone deacetylases (HDAC) is frequent. Based on the notion that HDAC inhibitors may reactivate the expression of genes favoring cell response to drugs, the aim of this study was to investigate the interaction between the HDAC6-specific inhibitor ricolinostat (ACY1215) and the MEK-inhibitor selumetinib (AZD6244) to identify effective combinations in prostate cancer models. Using cell lines exhibiting differential activation of survival pathways (PC3, DU145, 22Rv1) and following different treatment schedules, a synergistic interaction was observed in all cell models, the drug combination being particularly effective in 22Rv1 cells. Marginal levels of apoptosis were observed in PC3 cells after combined treatment, whereas higher levels were achieved in DU145 and 22Rv1 cells. RNAi-mediated knockdown of HDAC6 in selumetinib-treated 22Rv1 cells resulted in increased apoptosis. Combined treatment suppressed the constitutively deregulated survival pathways in all cell lines. A decrease of androgen receptor (AR)-dependent gene (KLK2, DUSP1) mRNA levels was observed in 22Rv1 treated cells, associated with increased AR cytoplasmatic expression, suggesting AR signaling down-regulation, not involving Hsp90 acetylation. When a taxane was used in combination with AZD6244 and ACY1215 by a simultaneous schedule, a synergistic cytotoxic effect together with increased apoptosis was evidenced in all cell models. These results support a rational use of targeted agents to improve prostate cancer cell apoptotic response.

14.
Anticancer Res ; 39(7): 3803-3808, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262907

ABSTRACT

BACKGROUND: Platinum-based therapy represents the main pharmacological treatment for ovarian carcinoma. Since molecular targeting of receptor tyrosine kinases (RTK) affects factors that may modulate drug response, the aim of this study was to examine whether downstream targets of AXL RTK could be exploited to improve cell response to cisplatin. MATERIALS AND METHODS: Inhibitors of p38 (SB203580) and of signal transducer and activator of transcription 3 (stattic) were employed in combination with cisplatin in ovarian carcinoma cell lines. Apoptosis assay and western blot analysis were performed to evaluate cell response after treatment. RESULTS: SB203580 produced a synergistic effect in combination with cisplatin in cisplatin-resistant IGROV-1/Pt1 cells. In addition, a favorable drug interaction was observed in A2780 cells when pre-incubated with cisplatin prior to stattic. The analysis of cell response after combined treatment showed down-regulation of the pro-apoptotic protein BCL2-associated agonist of cell death (BAD). CONCLUSION: Our results support the notion that downstream targets of AXL in ovarian carcinoma cells can be exploited to increase cisplatin activity in ovarian carcinoma models.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Imidazoles/pharmacology , Ovarian Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Drug Synergism , Female , Humans , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Axl Receptor Tyrosine Kinase
15.
Cancer Lett ; 263(1): 140-4, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18261847

ABSTRACT

The present study was designed to explore the role of p73 in response to cisplatin. In contrast to cisplatin-resistant ovarian carcinoma cells, pharmacological drug concentrations, which caused induction of p53, induced a marginal increase of p73 in sensitive cells. The effect was more marked at high concentrations, with no evidence of p21(WAF1) induction. This behaviour, associated with substantial apoptosis, suggests cell inability to activate DNA damage checkpoint following extensive stress. Although p73 appears to be implicated mainly in response to high stress conditions, the available results support a cooperation between p53 and p73 in cisplatin-induced apoptosis in sensitive cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Female , Humans , Protein Binding , Tumor Protein p73
16.
Cancer Lett ; 266(2): 194-202, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18378394

ABSTRACT

The Schizosaccharomyces pombe rad17 is a checkpoint protein critical for maintenance of genomic stability. Since the loss of checkpoint control is a common feature of tumor cells, we investigated the biological function of the human homolog hRAD17. Expression of hRAD17 in a fission yeast rad17 deleted strain reduced growth of yeast colonies and caused slower progression through cell cycle. Immunoprecipitated hRad17 exhibited exonuclease activity. hRAD17 delayed growth of NIH3T3 fibroblasts transformed by the H-ras oncogene in nude mice. Our results support that hRAD17, similarly to other human genes involved in checkpoint mechanisms, plays a role in control of tumor growth.


Subject(s)
Cell Cycle Proteins/physiology , Neoplasms, Experimental/etiology , Animals , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Cell Growth Processes , Cell Line, Transformed , Exonucleases/isolation & purification , Exonucleases/metabolism , Female , G1 Phase , Gene Deletion , Humans , Immunoprecipitation , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasms, Experimental/pathology , Schizosaccharomyces/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/genetics
17.
Bioconjug Chem ; 19(11): 2270-9, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18839979

ABSTRACT

A series of camptothecin open-ring lactone tripartate conjugates were synthesized, in which polyamine side chains with different architecture (ethane-1,2-diamine, spermidine, homospermidine, spermine, and 4,8,13,17-tetraza-icosane-1,20-diamine) are linked to the 21-carboxylic function through an amidic bond, while the 17-CH(2)OH is acetylated. The rationale for the synthesis of these compounds was to explore the influence of the polyamine architecture on the activity of these CPT conjugates into cells, since the positively charged ammonium cations would favor interaction through electrostatic binding to the negatively charged DNA backbone. Topoisomerase I-mediated DNA cleavage assay was used to investigate the ability of these compounds to stimulate the DNA damage. The cleavage pattern was found to be similar to that of SN38 for all the new CPTs. The CPT tripartates were tested for growth inhibition ability against the human non-small-cell lung cancer carcinoma NCI-H460 cell line. Although these compounds were less potent than topotecan, SN38, and CPT after 1 h of treatment, the antiproliferative effects greatly increased after 72 h of exposure. The growth inhibition potency during long-term exposure is correlated with the number of charges of the 21-amide substituent. Both cleavage assay and in vitro effects support the interpretation that the compounds may have inhibitory activity also in the open-ring form. The architecture of the polyamine moiety is important for antiproliferative activity, and a balance between the hydrophilic and lipophilic properties of the polyamine is critical for CPT potency.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin/chemistry , Camptothecin/pharmacology , Lactones/chemistry , Polyamines/chemistry , Animals , Antineoplastic Agents/metabolism , Camptothecin/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Cleavage/drug effects , DNA Topoisomerases, Type I/metabolism , Humans , Time Factors
18.
Biochem Pharmacol ; 147: 93-103, 2018 01.
Article in English | MEDLINE | ID: mdl-29155058

ABSTRACT

The XPO1/CRM1 inhibitor selinexor (KPT-330), is currently being evaluated in multiple clinical trials as an anticancer agent. XPO1 participates in the nuclear export of FoxO-1, which we previously found to be decreased in platinum-resistant ovarian carcinoma. The aim of this study was to determine whether enriching FoxO-1 nuclear localization using selinexor would increase ovarian cancer cell sensitivity to cisplatin. Selinexor, as a single agent, displayed a striking antiproliferative effect in different ovarian carcinoma cell lines. A schedule-dependent synergistic effect of selinexor in combination with cisplatin was found in cisplatin-sensitive IGROV-1, the combination efficacy being more evident in sensitive than in the resistant cells. In IGROV-1 cells, the combination was more effective when selinexor followed cisplatin exposure. A modulation of proteins involved in apoptosis (p53, Bax) and in cell cycle progression (p21WAF1) was found by Western blotting. Selinexor-treated cells exhibited enriched FoxO-1 nuclear staining. Knock-down experiments with RNA interference indicated that FOXO1-silenced cells displayed a reduced sensitivity to selinexor. FOXO1 silencing also tended to reduce the efficacy of the drug combination at selected cisplatin concentrations. Selinexor significantly inhibited tumor growth, induced FoxO-1 nuclear localization and improved the efficacy of cisplatin in IGROV-1 xenografts. Taken together, our results support FoxO-1 as one of the key factors promoting sensitivity towards selinexor and the synergistic interaction between cisplatin and selinexor in ovarian carcinoma cells with selected molecular backgrounds, highlighting the need for treatment regimens tailored to the molecular tumor features.


Subject(s)
Cisplatin/administration & dosage , Forkhead Box Protein O1/metabolism , Hydrazines/administration & dosage , Karyopherins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Forkhead Box Protein O1/genetics , Humans , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Treatment Outcome , Xenograft Model Antitumor Assays/methods , Exportin 1 Protein
19.
Biochim Biophys Acta ; 1763(1): 93-100, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16473140

ABSTRACT

Development of resistance to platinum compounds may involve not only overexpression of defence mechanisms but also alterations in cellular response to the drug-induced genotoxic stress. To investigate the cellular bases of response to platinum compounds, we examined the profile of gene expression of ovarian carcinoma cells exhibiting sensitivity (A2780) or resistance (A2780/BBR3464) to platinum compounds. Using display PCR, we found that acquisition of resistance to the multinuclear platinum complex BBR3464 was associated with modulation of several transcripts, including up-regulation of the major substrate of protein kinase C (PKC), the myristoylated alanine-rich C kinase substrate (MARCKS). This feature was associated with PKCalpha down-regulation. To explore the role of PKCalpha in cellular sensitivity to platinum compounds, resistant cells were transfected with a PKCalpha-containing vector. PKCalpha-overexpressing resistant cells exhibited a decrease in sensitivity to cisplatin, whereas no significant change in sensitivity to BBR3464 was observed. A number of approaches designed to modulate the function or expression of PKCalpha support that the isoenzyme may play a role in determining resistance only to cisplatin but not to BBR3464, which is known to activate a different pathway of cell response. In conclusion, in spite of PKCalpha down-regulation in our model, its regulatory function was not apparently implicated in the development of resistance to platinum compounds and the present results do not support a general role of PKCalpha as a determinant of the resistance status.


Subject(s)
Drug Resistance, Neoplasm , Platinum Compounds/pharmacology , Protein Kinase C-alpha/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Drug Combinations , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Membrane Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Phenotype , Reproducibility of Results , Transfection
20.
Biochem Pharmacol ; 136: 40-50, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28404378

ABSTRACT

Ovarian carcinoma, the most common gynaecological cancer, is characterized by high lethality mainly due to late diagnosis and treatment failure. The efficacy of platinum drug-based therapy in the disease is limited by the occurrence of drug resistance, a phenomenon often associated with increased metastatic potential. Because the Tyr-kinase receptor Axl can be deregulated in ovarian carcinoma and plays a pro-metastatic/anti-apoptotic role, the aim of this study was to examine if Axl inhibition modulates drug resistance and aggressive features of ovarian carcinoma cells, using various pairs of cisplatin-sensitive and -resistant cell lines. We found that mRNA and protein levels of Axl were increased in the platinum-resistant IGROV-1/Pt1 and IGROV-1/OHP cell lines compared to the parental IGROV-1 cells. IGROV-1/Pt1 cells displayed increased migratory and invasive capabilities. When Axl was silenced, these cells exhibited reduced growth and invasive/migratory capabilities compared to control siRNA-transfected cells, associated with decreased p38 and STAT3 phosphorylation. In keeping with this evidence, pharmacological inhibition of p38 and STAT3 decreased IGROV-1/Pt1 invasive capability. Molecular inhibition of Axl did not sensitize IGROV-1/Pt1 cells to cisplatin, but enhanced ErbB3 activation in IGROV-1/Pt1 cells and suppressed the clonogenic capability of various ovarian carcinoma cell lines. The combination of cisplatin and AZD8931, a small molecule which inhibits ErbB3, produced a synergistic effect in IGROV-1/Pt1 cells. Thus, Axl targeting per se reduces invasive capability of drug-resistant cells, but sensitization to cisplatin requires the concomitant inhibition of additional survival pathways.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/genetics , Platinum Compounds/administration & dosage , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Female , Gene Knockdown Techniques/methods , Humans , Molecular Targeted Therapy/methods , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL