Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cancer Gene Ther ; 29(10): 1394-1404, 2022 10.
Article in English | MEDLINE | ID: mdl-35352023

ABSTRACT

Diffuse malignant peritoneal mesothelioma (DMPM) is a rare and rapidly lethal tumor, poorly responsive to conventional treatments. In this regards, the identification of molecular alterations underlying DMPM onset and progression might be exploited to develop novel therapeutic strategies. Here, we focused on miR-550a-3p, which we found downregulated in 45 DMPM clinical samples compared to normal tissues and whose expression levels were associated with patient outcome. Through a gain-of-function approach using miRNA mimics in 3 DMPM cell lines, we demonstrated the tumor-suppressive role of miR-550a-3p. Specifically, miRNA ectopic expression impaired cell proliferation and invasiveness, enhanced the apoptotic response, and reduced the growth of DMPM xenografts in mice. Antiproliferative and proapoptotic effects were also observed in prostate and ovarian cancer cell lines following miR-550a-3p ectopic expression. miR-550a-3p effects were mediated, at least in part, by the direct inhibition of HSP90AA1 and the consequent downregulation of its target proteins, the levels of which were rescued upon disruption of miRNA-HSP90AA1 mRNA pairing, partially abrogating miR-550a-3p-induced cellular effects. Our results show that miR-550a-3p reconstitution affects several tumor traits, thus suggesting this approach as a potential novel therapeutic strategy for DMPM.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , MicroRNAs , Peritoneal Neoplasms , Animals , Biomarkers , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/pharmacology , Humans , Lung Neoplasms/genetics , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Prognosis , RNA, Messenger
2.
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
3.
Cancers (Basel) ; 11(4)2019 Apr 13.
Article in English | MEDLINE | ID: mdl-31013908

ABSTRACT

Ovarian carcinoma, the most lethal gynecological cancer, is characterized by late diagnosis, with drug resistance limiting the efficacy of platinum-based therapy. Since some integrins are upregulated in cancer, including ovarian carcinoma, they represent a potential target for drug delivery. Receptor tyrosine kinases are also deregulated in cancer and their expression has been associated with drug resistance. Here, the antitumor effects of three conjugates possessing a selective binder of the extracellular portion of integrin αVß3 covalently linked to the tyrosine kinase inhibitor sunitinib were investigated in cisplatin-sensitive and -resistant ovarian carcinoma cells expressing both tyrosine kinase VEGFR2 and αVß3 at different levels. We found that one of the three compounds was active in inhibiting the growth of both drug-sensitive and -resistant cells in the micromolar range with a slightly increased potency in resistant cells as compared to sunitinib. The same compound markedly impaired cell migratory and invasive abilities and reduced paxillin phosphorylation. Antitumor activity studies in IGROV-1/Pt1 cells xenografted in nude mice revealed a striking activity of this conjugate versus sunitinib. Taken together, our results support the interest of integrin-targeted sunitinib conjugates for the treatment of drug-resistant tumors.

4.
Anticancer Res ; 38(10): 5783-5790, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30275200

ABSTRACT

BACKGROUND/AIM: The lipophilic, orally bioavailable camptothecin analogue gimatecan is characterized by improved efficacy over conventional camptothecins on human tumor xenografts. Gimatecan produced excellent outcomes orally with prolonged, low-dose, daily treatments. The aim of this study was to assess the antitumor efficacy of i.v. administered gimatecan. MATERIALS AND METHODS: The antitumor activity of gimatecan delivered i.v. q4dx4 was evaluated in nude mice bearing human tumors and compared to clinically available anticancer drugs. RESULTS: Intravenous administration of gimatecan showed superior efficacy with remarkable achievements at well tolerated doses. Moreover, prolonged treatments with i.v. administered gimatecan showed efficacy in models of cancer refractory to current therapeutic approaches, like an orthotopic brain tumor. CONCLUSION: Although the oral route is practical for gimatecan administration, the results support the interest in developing a suitable i.v. formulation in an attempt to further exploit the therapeutic potential of the compound.


Subject(s)
Astrocytoma/drug therapy , Bone Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Administration, Oral , Animals , Apoptosis/drug effects , Astrocytoma/pathology , Bone Neoplasms/pathology , Camptothecin/administration & dosage , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Molecules ; 23(2)2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29438315

ABSTRACT

BACKGROUND: Heat shock protein 90 (HSP90) is a well-known target for cancer therapy. In a previous work, some of us have reported a series of 3-aryl-naphtho[2,3-d]isoxazole-4,9-diones as inhibitors of HSP90. METHODS: In the present work, various compounds with new chromenopyridinone and thiochromenopyridinone scaffolds were synthesized as potential HSP90 inhibitors. Their binding affinity to HSP90 was studied in vitro. Selected compounds (5 and 8) were further studied in various tumor cell lines regarding their potential to cause cell growth inhibition, alter the cell cycle profile, inhibit proliferation, and induce apoptosis. Their effect on HSP90 client protein levels was also confirmed in two cell lines. Finally, the antitumor activity of compound 8 was studied in A431 squamous cell carcinoma xenografts in nude mice. RESULTS: Our results indicated that treatment with compounds 5 and 8 decreased the proliferation of tumor cell lines and compound 8 induced apoptosis. In addition, these two compounds were able to downregulate selected proteins known as "clients" of HSP90. Finally, treatment of xenografted mice with compound 5 resulted in a considerable dose-dependent inhibition of tumor growth. CONCLUSIONS: Our results show that two new compounds with a chromenopyridinone and thiochromenopyridinone scaffold are promising putative HSP90 inhibitors causing tumor cell growth inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Carcinoma, Squamous Cell/drug therapy , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Pyridones/pharmacology , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Apoptosis/genetics , Benzopyrans/chemical synthesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Female , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Inhibitory Concentration 50 , Isoxazoles/chemical synthesis , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyridones/chemical synthesis , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Survivin , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , raf Kinases/metabolism
6.
Cancer Lett ; 415: 187-197, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29225052

ABSTRACT

Synovial sarcoma (SS) is an aggressive tumor with propensity for lung metastases which significantly impact patients' prognosis. New therapeutic approaches are needed to improve treatment outcome. Targeting the heparanase/heparan sulfate proteoglycan system by heparin derivatives which act as heparanase inhibitors/heparan sulfate mimetics is emerging as a therapeutic approach that can sensitize the tumor response to chemotherapy. We investigated the therapeutic potential of a supersulfated low molecular weight heparin (ssLMWH) in preclinical models of SS. ssLMWH showed a potent anti-heparanase activity, dose-dependently inhibited SS colony growth and cell invasion, and downregulated the activation of receptor tyrosine kinases including IGF1R and IR. The combination of ssLMWH and the IGF1R/IR inhibitor BMS754807 synergistically inhibited proliferation of cells exhibiting IGF1R hyperactivation, also abrogating cell motility and promoting apoptosis in association with PI3K/AKT pathway inhibition. The drug combination strongly enhanced the antitumor effect against the CME-1 model, as compared to single agent treatment, abrogating orthotopic tumor growth and significantly repressing spontaneous lung metastatic dissemination in treated mice. These findings provide a strong preclinical rationale for developing drug regimens combining heparanase inhibitors/HS mimetics with IGF1R antagonists for treatment of metastatic SS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Pyrazoles/pharmacology , Receptors, Somatomedin/antagonists & inhibitors , Sarcoma, Synovial/drug therapy , Triazines/pharmacology , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Heparin, Low-Molecular-Weight/administration & dosage , Heparin, Low-Molecular-Weight/metabolism , Humans , Mice, SCID , Neoplasm Metastasis , Pyrazoles/administration & dosage , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Sarcoma, Synovial/metabolism , Sarcoma, Synovial/pathology , Sulfates , Triazines/administration & dosage
8.
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
9.
J Med Chem ; 60(7): 3082-3093, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28272894

ABSTRACT

Prostate cancer (PC) is the fifth leading cause of cancer death in men, and the androgen receptor (AR) represents the primary target for PC treatment, even though the disease frequently progresses toward androgen-independent forms. Most of the commercially available nonsteroidal antiandrogens show a common scaffold consisting of two aromatic rings connected by a linear or a cyclic spacer. By taking advantage of a facile, one-pot click chemistry reaction, we report herein the preparation of a small library of novel 1,4-substituted triazoles with AR antagonistic activity. Biological and theoretical evaluation demonstrated that the introduction of the triazole core in the scaffold of nonsteroidal antiandrogens allowed the development of small molecules with improved overall AR-antagonist activity. In fact, compound 14d displayed promising in vitro antitumor activity toward three different prostate cancer cell lines and was able to induce 60% tumor growth inhibition of the CW22Rv1 in vivo xenograft model. These results represent a step toward the development of novel and improved AR antagonists.


Subject(s)
Nonsteroidal Anti-Androgens/chemistry , Nonsteroidal Anti-Androgens/therapeutic use , Prostate/drug effects , Prostatic Neoplasms/drug therapy , Triazoles/chemistry , Triazoles/therapeutic use , Animals , Cell Line, Tumor , Drug Discovery , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Nude , Models, Molecular , Nonsteroidal Anti-Androgens/pharmacology , Prostate/metabolism , Prostate/pathology , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Triazoles/pharmacology
10.
Chemistry ; 23(24): 5842-5850, 2017 Apr 27.
Article in English | MEDLINE | ID: mdl-28300330

ABSTRACT

Synthetic tubulysins 24 a-m, containing non-hydrolysable N-substituents on tubuvaline (Tuv), were obtained in high purity and good overall yields using a multistep synthesis. A key step was the formation of differently N-substituted Ile-Tuv fragments 10 by using an aza-Michael reaction of azido-Ile derivatives 8 with the α,ß-unsaturated oxo-thiazole 5. A structure-activity relationship study using a panel of human tumour cell lines showed strong anti-proliferative activity for all compounds 24 a-m, with IC50 values in the sub-nanomolar range, which were distinctly lower than those of tubulysin A, vinorelbine and paclitaxel. Furthermore, 24 a-m were able to overcome cross-resistance to paclitaxel and vinorelbine in two tumour cell lines with acquired resistance to doxorubicin. Compounds 24 e and 24 g were selected as leads to evaluate their mechanism of action. In vitro assays showed that both 24 e and 24 g interfere with tubulin polymerization in a vinca alkaloid-like manner and prevent paclitaxel-induced assembly of tubulin polymers. Both compounds exerted antimitotic activity and induced apoptosis in cancer cells at very low concentrations. Compound 24 e also exhibited potent antitumor activity at well tolerated doses on in vivo models of diffuse malignant peritoneal mesothelioma, such as MESOII peritoneal mesothelioma xenografts, the growth of which was not significantly affected by vinorelbine. These results indicate that synthetic tubulysins 24 could be used as standalone chemotherapeutic agents in difficult-to-treat cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , Valine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HT29 Cells , Humans , Mice , Microscopy, Fluorescence , Neoplasms/drug therapy , Neoplasms/pathology , Paclitaxel/toxicity , Structure-Activity Relationship , Transplantation, Heterologous , Tubulin/chemistry , Tubulin Modulators/therapeutic use , Tubulin Modulators/toxicity , Valine/chemistry , Vinblastine/analogs & derivatives , Vinblastine/therapeutic use , Vinblastine/toxicity , Vinorelbine
11.
J Hematol Oncol ; 10(1): 19, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100259

ABSTRACT

BACKGROUND: The value of microRNAs (miRNAs) as novel targets for cancer therapy is now widely recognized. However, no information is currently available on the expression/functional role of miRNAs in diffuse malignant peritoneal mesothelioma (DMPM), a rapidly lethal disease, poorly responsive to conventional treatments, for which the development of new therapeutic strategies is urgently needed. Here, we evaluated the expression and biological effects of miR-34a-one of the most widely deregulated miRNAs in cancer and for which a lipid-formulated mimic is already clinically available-in a large cohort of DMPM clinical samples and a unique collection of in house-developed preclinical models, with the aim to assess the potential of a miR-34a-based approach for disease treatment. METHODS: miR-34a expression was determined by qRT-PCR in 45 DMPM and 7 normal peritoneum specimens as well as in 5 DMPM cell lines. Following transfection with miR-34a mimic, the effects on DMPM cell phenotype, in terms of proliferative potential, apoptotic rate, invasion ability, and cell cycle distribution, were assessed. In addition, three subcutaneous and orthotopic DMPM xenograft models were used to examine the effect of miR-34a on tumorigenicity. The expression of miRNA targets and the activation status of relevant pathways were investigated by western blot. RESULTS: miR-34a was found to be down-regulated in DMPM clinical specimens and cell lines compared to normal peritoneal samples. miR-34a reconstitution in DMPM cells significantly inhibited proliferation and tumorigenicity, induced an apoptotic response, and declined invasion ability, mainly through the down-regulation of c-MET and AXL and the interference with the activation of downstream signaling. Interestingly, a persistent activation of ERK1/2 and AKT in miR-34a-reconstituted cells was found to counteract the antiproliferative and proapoptotic effects of miRNA, yet not affecting its anti-invasive activity. CONCLUSIONS: Our preclinical data showing impressive inhibitory effects induced by miR-34a on DMPM cell proliferation, invasion, and growth in immunodeficient mice strongly suggest the potential clinical utility of a miR-34a-replacement therapy for the treatment of such a still incurable disease. On the other hand, we provide the first evidence of a potential cytoprotective/resistance mechanism that may arise towards miRNA-based therapies through the persistent activation of RTK downstream signaling.


Subject(s)
Lung Neoplasms/pathology , Mesothelioma/pathology , MicroRNAs/physiology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Proliferation , Down-Regulation , Heterografts , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mesothelioma/drug therapy , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , MicroRNAs/genetics , MicroRNAs/therapeutic use , Peritoneal Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Axl Receptor Tyrosine Kinase
12.
ChemMedChem ; 11(16): 1700-4, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27311681

ABSTRACT

A series of compounds containing the N-hydroxyoxindole scaffold were synthesized and evaluated for antitumor activity. The compounds showed potent antiproliferative activity against the wild-type p53 IGROV-1 ovarian carcinoma cell line and considerably lower efficacy against the mutant IGROV-1/Pt1 subline that lacks p53 function. The differential response of ovarian carcinoma cells depending on p53 status was also reflected in the varied susceptibility to apoptosis of the treated cell lines. These results support a role for the p53 transcription factor as a determinant of cytotoxicity. The therapeutic potential of the most promising compound of the series was evaluated in the treatment of an IGROV-1 xenograft growing as ascitic tumor in mice. Using intraperitoneal administration, daily treatment with the compound for four weeks produced a significant delay in the onset of ascites.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indoles/administration & dosage , Indoles/chemistry , Injections, Intraperitoneal , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Structure-Activity Relationship
13.
Tumori ; 102(2): 144-9, 2016.
Article in English | MEDLINE | ID: mdl-26917410

ABSTRACT

PURPOSE: Because the peptidyl-prolyl isomerase PIN1 interacts with multiple protein kinases and phosphoproteins into a network orchestrating the cellular response to various stimuli, there is an increasing interest in exploiting its potential as therapeutic target. In the present study, the effect of targeting PIN1 was investigated in 2 human cancer cell lines characterized by increased aggressive potential, high expression of erbB receptor family members, and defective p53. METHODS: PIN1 silencing was carried out in skin squamous cell carcinoma A431 cells displaying elevated EGFR/HER1 levels and in ovarian adenocarcinoma SKOV-3 cells displaying high levels of erbB2 (HER2). Nonoverlapping siRNA duplexes targeting different regions of PIN1 mRNA were transfected in tumor cells, which were analyzed using Western blotting for the expression of selected proteins. In vivo tumorigenicity studies were carried out in athymic nude mice. RESULTS: A431 and SKOV-3 cell systems were found to be a source of cells with increased aggressive potential, i.e., cancer stem cell-like cells, as defined by the capability to grow as spheres. A marked decrease of PIN1 levels and of sphere-forming capability was observed in PIN1-silenced cells. The expression of phospho-p38 decreased following PIN1 silencing in A431 and SKOV-3 cells, as well as phospho-EGFR levels in A431 - silenced cells. PIN1 inhibition prolonged latency and reduced tumor take and growth of SKOV-3 cells in nude mice. CONCLUSIONS: Our results support that PIN1 may be a valuable target to hit in cancer cells characterized by increased aggressive potential, overexpression of erbB receptor family members, and defective p53.


Subject(s)
ErbB Receptors/metabolism , Gene Silencing , Neoplasms/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/metabolism , Animals , Blotting, Western , Carcinogenicity Tests , Carcinoma, Squamous Cell/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , NIMA-Interacting Peptidylprolyl Isomerase , Ovarian Neoplasms/metabolism , Skin Neoplasms/metabolism , Up-Regulation
14.
Eur J Med Chem ; 112: 99-105, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-26890116

ABSTRACT

Modification of the cap group of biphenylacrylohydroxamic acid-based HDAC inhibitors led to the identification of a new derivative (3) characterized by an indolyl-substituted 4-phenylcinnamic skeleton. Molecular docking was used to predict the optimal conformation in the class I HDACs active site. Compound 3 showed HDAC inhibitory activity and antiproliferative activity against a panel of tumor cell lines, in the low µM range. The compound was further tested in vitro for acetylation of histone H4 and other non-histone proteins, and in vivo in a colon carcinoma model, showing significant proapoptotic and antitumor activities.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Drug Screening Assays, Antitumor , HCT116 Cells , Histone Deacetylases/metabolism , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/metabolism , Structure-Activity Relationship
15.
J Transl Med ; 14: 25, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26810896

ABSTRACT

BACKGROUND: Diffuse malignant peritoneal mesothelioma (DMPM) is a rare and locally aggressive disease. DMPM prognosis is dismal, mainly due to the lack of effective treatment options and the development of new therapeutic strategies is urgently needed. In this context, novel immunotherapy approaches can be explored in an attempt to improve DMPM patients' survival. METHODS: We tested the efficacy of CpG-oligodeoxynucleotides (CpG-ODN), synthetic DNA sequences recognized by Toll-like receptor 9 and able to induce innate/adaptive immune response, in two DMPM orthotopic xenografts (MesoII and STO), which properly recapitulate the dissemination pattern of the disease in the peritoneal cavity. Severe combined immunodeficiency mice carrying DMPM xenografts were treated at different stages of tumor development with i.p. delivered CpG-ODN1826 for 4 weeks. CpG-ODN1826-induced modulation in the composition of peritoneal immune infiltrate was assessed by flow cytometry. RESULTS: When administered to early-stage tumors (i.e., 4 days after i.p. DMPM cell injection in mice), the agent exhibited impressive efficacy against MesoII by completely inhibiting tumor take and ascites development (no evidence of tumor masses and ascites in 6/6 mice at necropsy), and also impaired STO tumor take and growth (4/6 tumor-free mice; i.p. tumor masses reduced by 94 % in the 2 remaining mice, P = 0.00005). Interestingly, when tested against late-stage STO tumors (i.e., 11 days after i.p. DMPM cell injection in mice), CpG-ODN1826 was still able to reduce the growth of i.p. tumor masses by 66 % (P = 0.0009). Peritoneal washings of tumor-bearing mice revealed a strong increase of macrophage infiltration together with a decrease in the presence of B-1 cells and a reduced IgM concentration after CpG-ODN1826 treatment. CONCLUSIONS: Our results indicate that locally administered CpG-ODN1826 is able to markedly affect the growth of both early- and late-stage DMPM orthotopic xenografts in the absence of severe side effects, and suggest a possible clinical role for the agent in the therapy of DMPM.


Subject(s)
Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromatography, Affinity , Female , Flow Cytometry , Humans , Immunity, Humoral/drug effects , Injections, Intraperitoneal , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mesothelioma/immunology , Mesothelioma/pathology , Mesothelioma, Malignant , Mice, SCID , Neoplasm Staging , Oligodeoxyribonucleotides/pharmacology , Treatment Outcome
16.
Oncotarget ; 6(15): 13119-32, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25948791

ABSTRACT

Survivin, which is highly expressed and promotes cell survival in diffuse malignant peritoneal mesothelioma (DMPM), exclusively relies on exportin 1 (XPO1/CRM1) to be shuttled into the cytoplasm and perform its anti-apoptotic function. Here, we explored the efficacy of Selective Inhibitors of Nuclear Export (SINE), KPT-251, KPT-276 and the orally available, clinical stage KPT-330 (selinexor), in DMPM preclinical models. Exposure to SINE induced dose-dependent inhibition of cell growth, cell cycle arrest at G1-phase and caspase-dependent apoptosis, which were consequent to a decrease of XPO1/CRM1 protein levels and the concomitant nuclear accumulation of its cargo proteins p53 and CDKN1a. Cell exposure to SINE led to a time-dependent reduction of cytoplasmic survivin levels. In addition, after an initial accumulation, the nuclear protein abundance progressively decreased, as a consequence of an enhanced ubiquitination and proteasome-dependent degradation. SINE and the survivin inhibitor YM155 synergistically cooperated in reducing DMPM cell proliferation. Most importantly, orally administered SINE caused a significant anti-tumor effect in subcutaneous and orthotopic DMPM xenografts without appreciable toxicity. Overall, we have demonstrated a marked efficacy of SINE in DMPM preclinical models that may relay on the interference with survivin intracellular distribution and function. Our study suggests SINE-mediated XPO1/CRM1 inhibition as a novel therapeutic option for DMPM.


Subject(s)
Antineoplastic Agents/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Karyopherins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Peritoneal Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Acrylamides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Hydrazines/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mesothelioma/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Oxadiazoles/pharmacology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Survivin , Thiazoles/pharmacology , Triazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Exportin 1 Protein
17.
Oncotarget ; 6(11): 8736-49, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25826089

ABSTRACT

Intrinsic and acquired tumor drug resistance limits the therapeutic efficacy of camptothecins (CPTs). Downregulation of the mitotic kinase PLK1 was found associated with apoptosis induced by SN38 (CPT11 active metabolite). We investigated the role of PLK1 in the cell response to CPTs in squamous cell carcinoma (SCC) and pediatric sarcoma cell lines and explored the therapeutic potential of the combination of CPT11 and the PLK1 inhibitor BI2536 in CPT-sensitive and -resistant tumor models. Gain- and loss-of-function experiments established a direct role for PLK1 in counteracting SN38 antiproliferative and pro-apoptotic effects. The ability to activate an efficient G2/M cell cycle checkpoint allowing PLK1 ubiquitination and degradation was found associated with SN38-induced apoptosis in SCC cells. However, the synergistic interaction between SN38 and BI2536 enhanced apoptosis in cell lines both sensitive and resistant to SN38-induced apoptotic cell death. A well-tolerated CPT11/BI2536 cotreatment resulted in improved antitumor effect against SCC xenografts in mice compared to single agent treatments. The increased apoptosis induction was reflected in a high rate of complete responses and cures in mice harboring SCC, including tumors with intrinsic or acquired resistance to CPTs. PLK1 inhibition represents a promising strategy to improve the antitumor efficacy of CPT11-based regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/physiology , Drug Resistance, Neoplasm/physiology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Prodrugs/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Pteridines/pharmacology , Rhabdomyosarcoma, Embryonal/pathology , Sarcoma, Ewing/pathology , Skin Neoplasms/pathology , Topoisomerase I Inhibitors/pharmacology , Uterine Cervical Neoplasms/pathology , Activation, Metabolic , Animals , Apoptosis/drug effects , Camptothecin/administration & dosage , Camptothecin/metabolism , Camptothecin/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Child , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Irinotecan , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Prodrugs/administration & dosage , Prodrugs/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proteolysis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pteridines/administration & dosage , Topoisomerase I Inhibitors/metabolism , Ubiquitination , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
18.
Int J Cancer ; 136(2): 299-309, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-24866585

ABSTRACT

Because available treatments have limited efficacy in triple-negative breast cancer (TNBC), the identification of new therapeutic strategies to improve patients' outcome is urgently needed. In our study, we investigated the effects of the administration of the small molecule selective survivin suppressant YM155, alone or in association with CD34+ cells transduced with a replication-deficient adenovirus encoding the human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene (CD34-TRAIL+ cells), in three TNBC cell models. YM155 exposure significantly impaired TNBC cell growth and selectively modulated survivin expression at both mRNA and protein level. In addition, co-culturing YM155-treated TNBC cells with CD34-TRAIL+ cells resulted in markedly increased cytotoxic effect and apoptotic response in comparison with single treatments. Such a chemosensitizing effect was observed only in TNBC cells inherently expressing DR5 and relied on the ability of YM155 to upregulate DR5 expression through a p38 MAPK- and CHOP-dependent mechanism. YM155/CD34-TRAIL+ combination also showed a significant inhibitory effect on the growth of DR5-expressing TNBC cells following xenotransplantation into NOD/SCID mice, in the absence of toxicity. Overall, our data (i) provide, for the first time, evidence that YM155 sensitizes TNBC cells to CD34-TRAIL+ cells-induced apoptosis by a mechanism involving the downregulation of survivin and the simultaneous p38 MAPK- and CHOP-mediated upregulation of DR5, and (ii) suggest the combination of YM155 with TRAIL-armed CD34+ progenitor cells as a promising therapeutic option for patients with TNBC expressing DR5.


Subject(s)
Imidazoles/pharmacology , Naphthoquinones/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor CHOP/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Membrane/metabolism , Cell Proliferation/drug effects , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Reactive Oxygen Species/metabolism , Survivin , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
J Med Chem ; 57(17): 7263-79, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25121586

ABSTRACT

The androgen receptor (AR) represents the primary target for prostate cancer (PC) treatment even when the disease progresses toward androgen-independent (AIPC) or castration-resistant (CRPC) forms. Because small chemical changes in the structure of nonsteroidal AR ligands determine the pharmacological responses of AR, we developed a novel stereoselective synthetic strategy that allows sterically hindered C2-substituted bicalutamide analogues to be obtained. Biological and theoretical evaluations demonstrate that C2-substitution with benzyl and phenyl moieties is a new, valuable option toward improving pan-antagonist behavior. Among the synthesized compounds, (R)-16m, when compared to casodex, (R)-bicalutamide, and enzalutamide, displayed very promising in vitro activity toward five different prostate cancer cell lines, all representative of CPRC and AIPC typical mutations. Despite being less active than (R)-bicalutamide, (R)-16m also displayed marked in vivo antitumor activity on VCaP xenografts and thus it may serve as starting point for developing novel AR pan-antagonists.


Subject(s)
Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms/prevention & control , Receptors, Androgen/chemistry , Active Transport, Cell Nucleus/drug effects , Androgen Receptor Antagonists/chemical synthesis , Animals , Apoptosis/drug effects , Blotting, Western , COS Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Models, Chemical , Models, Molecular , Molecular Structure , Mutation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Conformation/drug effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
20.
Oncotarget ; 5(12): 4516-28, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980831

ABSTRACT

Target-specific agents used in melanoma are not curative, and chemokines are being implicated in drug-resistance to target-specific agents. Thus, the use of conventional agents in rationale combinations may result in optimization of therapy. Because histone deacetylases participate in tumor development and progression, the combination of the pan-inhibitor SAHA and temozolomide might provide a therapeutic advantage. Here, we show synergism between the two drugs in mutant BRAF cell lines, in association with decreased phosphorylation of cell survival proteins (e.g., C-Jun-N-terminal-kinase, JNK). In the spontaneous ret transgenic mouse melanoma model, combination therapy produced a significant disease onset delay and down-regulation of Chemokine (C-C motif) ligand 2 (CCL2), JNK, and of Myeloid-derived suppressor cell recruitment. Co-incubation with a CCL2-blocking-antibody enhanced in vitro cell sensitivity to temozolomide. Conversely, recombinant CCL2 activated JNK in human tumor melanoma cells. In keeping with these results, the combination of a JNK-inhibitor with temozolomide was synergistic. By showing that down-regulation of CCL2-driven signals by SAHA and temozolomide via JNK contributes to reduce melanoma growth, we provide a rationale for the therapeutic advantage of the drug combination. This combination strategy may be effective because of interference both with tumor cell and tumor microenvironment.


Subject(s)
Chemokines/metabolism , Dacarbazine/analogs & derivatives , Histone Deacetylase Inhibitors/metabolism , Melanoma/genetics , Animals , Apoptosis , Cell Survival , Dacarbazine/metabolism , Disease Models, Animal , Drug Synergism , Humans , Mice , Mice, Transgenic , Temozolomide , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...