Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Eur J Pharm Sci ; 193: 106686, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38159687

ABSTRACT

As part of our efforts geared towards developing mechanism-based cancer sensitizing agents, we have previously synthesized and characterized novel deazaflavin analogs as potent tyrosyl DNA phosphodiesterase 2 (TDP2) inhibitors for combination treatments with topoisomerase II (TOP2) poisons. Interestingly, the sensitizing effect of a few analogs toward TOP2 poison etoposide (ETP) was associated with a significant increase in intracellular drug accumulation, which could be an alternative mechanism to boost the clinical efficacy of ETP in cancer chemotherapies. Hence, we evaluated more deazaflavin TDP2 inhibitors for their impact on drug retention in cancer cells. We found that all but one tested TDP2 inhibitors substantially increased the ETP retention in DT40 cells. Particularly, we identified an exceptionally potent analog, ZW-1226, which at 3 nM increased the intracellular ETP by 13-fold. Significantly, ZW-1226 also stimulated cellular accumulation of two other anticancer drugs, TOP2 poison teniposide and antifolate pemetrexed, and produced an effect more pronounced than those of ABC transporter inhibitors verapamil and elacridar in human leukemic CCRF-CEM cells toward ETP. Lastly, ZW-1226 potentiated the action of ETP in the sensitive human CCRF-CEM cells and a few resistant non-small-cell lung cancer (NSCLC) cells, including H460 and H838 cells. Collectively, the results of this study strongly suggest that deazaflavin analog ZW-1226 could be an effective cancer sensitizing agent which warrants further investigation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Poisons , Humans , DNA-Binding Proteins/genetics , Phosphoric Diester Hydrolases , Etoposide/pharmacology , DNA Topoisomerases, Type II/genetics
2.
Invest New Drugs ; 39(3): 636-643, 2021 06.
Article in English | MEDLINE | ID: mdl-33230623

ABSTRACT

In order to suppress 5' cap-mediated translation a highly available inhibitor of the interaction between the 5' mRNA cap and the eIF4E complex has been developed. 4Ei-10 is a member of the class of ProTide compounds and has elevated membrane permeability and is a strong active chemical antagonist for eIF4E. Once taken up by cells it is converted by anchimeric activation of the lipophilic 2-(methylthio) ethyl protecting group and after that Hint1 P-N bond cleavage to N7-(p-chlorophenoxyethyl) guanosine 5'-monophosphate (7-Cl-Ph-Ethyl-GMP). Using this powerful interaction, it has been demonstrated that 4Ei-10 inhibits non-small cell lung cancer (NSCLC) cell growth. In addition, treatment of NSCLC cells with 4Ei-10 results in suppression of translation and diminished expression of a cohort of cellular proteins important to maintaining the malignant phenotype and resisting apoptosis such as Bcl-2, survivin, and ornithine decarboxylase (ODC). Finally, as a result of targeting the translation of anti-apoptotic proteins, NSCLC cells are synergized to be more sensitive to the existing anti-neoplastic treatment gemcitabine currently used in NSCLC therapy.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Eukaryotic Initiation Factor-4E , Lung Neoplasms , Nucleotides , Prodrugs , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Interactions , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Lung Neoplasms/drug therapy , Prodrugs/pharmacology , Nucleotides/pharmacology , Nucleotides/therapeutic use , Gemcitabine
3.
Transl Oncol ; 13(7): 100782, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32422574

ABSTRACT

Oncolytic viruses have demonstrated efficacy in numerous tumor models including non-small cell lung cancer (NSCLC). One limitation of viral therapy for metastatic lung cancer is that systemic administration can be hindered by complement and antiviral immunity. Thus, we investigated whether ex vivo-infected blood outgrowth endothelial cells (BOECs) could be used to deliver VSV-IFNß in preclinical models of NSCLC. BOECs were obtained from human donors or C57/Bl6 mice. VSV was engineered to produce GFP or IFNß. Human and murine BOECs could be infected by VSV-GFP and VSV-IFNß. Infected BOECs resulted in killing of NSCLC cells in vitro and shielded VSV-IFNß from antibody neutralization. Mouse BOECs localized to lungs of mice bearing syngeneic LM2 lung tumors, and infected murine BOECs reduced tumor burden in this model. In an immune-deficient A549 xenograft model, mice treated with VSV-IFNß-infected human BOECs exhibited superior antitumor activity and survival of mice (n = 10, P < .05 compared to VSV-IFNß alone). We conclude that BOECs can be used as a carrier for delivery of oncolytic VSV-IFNß. This may be an effective strategy for clinical translation of oncolytic virotherapy for patients with metastatic NSCLC.

4.
Cancer Chemother Pharmacol ; 85(2): 425-432, 2020 02.
Article in English | MEDLINE | ID: mdl-31974652

ABSTRACT

Activated cap-dependent translation promotes cancer by stimulating translation of mRNAs encoding malignancy-promoting proteins. The nucleoside monophosphate Protide, 4Ei-10, undergoes intracellular uptake and conversion by Hint1 to form 7-Cl-Ph-Ethyl-GMP. 7-Cl-Ph-Ethyl-GMP is an analog of cap and inhibits protein translation by binding and sequestering eIF4E thus blocking eIF4E from binding to the mRNA cap. The effects of inhibiting translation initiation by disruption of the eIF4F complex with 4Ei-10 were examined in malignant mesothelioma (MM). In a cell-free assay system, formation of the eIF4F complex was disabled in response to exposure to 4Ei-10. Treatment of MM with 4Ei-10 resulted in decreased cell proliferation, increased sensitivity to pemetrexed and altered expression of malignancy-related proteins. In light of these findings, suppression of translation initiation by small molecule inhibitors like 4Ei-10 alone or in combination with pemetrexed represents an encouraging strategy meriting further evaluation in the treatment of MM.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mesothelioma/drug therapy , Mesothelioma/genetics , Carcinogenesis/drug effects , Cell Line, Tumor , Eukaryotic Initiation Factor-4F/genetics , Humans , Mesothelioma, Malignant , Pemetrexed/pharmacology , RNA, Messenger/genetics , Small Molecule Libraries/pharmacology
5.
Cancer Gene Ther ; 26(11-12): 411-418, 2019 11.
Article in English | MEDLINE | ID: mdl-30622322

ABSTRACT

The concept of using viruses to treat cancer has now shown proof of concept in several recent clinical trials, leading to the first FDA approval of virotherapy for melanoma last year. Vesicular stomatitis virus (VSV) is a promising oncolytic virus that has inhibitory effects on a number of cancer types including non-small cell lung cancer. One of the major mechanisms of resistance to VSV infection is the type I interferon (IFN) response, leading to the development of VSV expressing IFNß which will lead to resistance of viral replication in normal cells which have intact IFN signaling but allow replication in cancer cells with defective IFNß signaling. However, some cancer cells have intact or partially intact IFN signaling pathways leading to resistance to virotherapy. Here we utilized JAK/STAT inhibitor, ruxolitinib, in combination with VSV-IFNß to see if inhibition of JAK/STAT signaling will enhance VSV-IFNß therapy for lung cancer. We used five human and two murine NSCLC cell lines in vitro, and the combination treatment was assayed for cytotoxicity. We performed western blots on treated cells to see the effects of ruxolitinib on JAK/STAT signaling and PDL-1 expression in treated cells. Finally, the combination of VSV-IFNß and ruxolitinib was tested in an immune competent murine model of NSCLC. Ruxolitinib enhanced virotherapy in resistant and sensitive NSCLC cells. The addition of ruxolitinib inhibited STAT1 phosphorylation and to a lesser extent STAT3 phosphorylation. Ruxolitinib treatment prevented NSCLC cells from enhancing PDL-1 expression in response to virotherapy. Combination ruxolitinib and VSV-IFNß therapy resulted in a trend toward improved survival of mice without substantially effecting PDL-1 levels or levels of immune infiltration into the tumor. These results support further clinical evaluation of the combination of JAK/STAT inhibition with virotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Janus Kinase Inhibitors/pharmacology , Lung Neoplasms/metabolism , Oncolytic Virotherapy , Pyrazoles/pharmacology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Nitriles , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pyrimidines , Signal Transduction , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Cancer Gene Ther ; 26(5-6): 157-165, 2019 05.
Article in English | MEDLINE | ID: mdl-30420719

ABSTRACT

Hyperactivation of eIF4F-mediated translation occurs in many if not all cancers. As a consequence, cancer cells aberrantly enhance expression of malignancy-related proteins that are involved in cell cycle progression, angiogenesis, growth, and proliferation. With this in mind eIF4F is a promising molecular target for therapeutics that counteract pathological eIF4F activity. Here we used 4EGI-1, a small-molecule inhibitor of cap-mediated translation that disrupts formation of the eukaryotic initiation factor 4F (eIF4F) complex to treat non-small cell lung cancer (NSCLC). Treatment of cells with 4EGI-1 reduced cell proliferation, decreased cap-dependent complex formation, induced apoptosis, enhanced sensitivity to gemcitabine, and altered global cellular translation. Suppression of cap-dependent translation by 4EGI-1 resulted in diminished expression of oncogenic proteins c-Myc, Bcl-2, cyclin D1, and survivin, whereas ß-actin expression was left unchanged. In light of these results, small-molecule inhibitors like 4EGI-1 alone or with chemotherapy should be further evaluated in the treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Hydrazones/metabolism , Lung Neoplasms/genetics , Thiazoles/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Humans , Lung Neoplasms/pathology
7.
Invest New Drugs ; 36(2): 217-229, 2018 04.
Article in English | MEDLINE | ID: mdl-29116477

ABSTRACT

Deregulation of cap-dependent translation has been implicated in the malignant transformation of numerous human tissues. 4EGI-1, a novel small-molecule inhibitor of cap-dependent translation, disrupts formation of the eukaryotic initiation factor 4F (eIF4F) complex. The effects of 4EGI-1-mediated inhibition of translation initiation in malignant pleural mesothelioma (MPM) were examined. 4EGI-1 preferentially inhibited cell viability and induced apoptosis in MPM cells compared to normal mesothelial (LP9) cells. This effect was associated with hypophosphorylation of 4E-binding protein 1 (4E-BP1) and decreased protein levels of the cancer-related genes, c-myc and osteopontin. 4EGI-1 showed enhanced cytotoxicity in combination with pemetrexed or gemcitabine. Translatome-wide polysome microarray analysis revealed a large cohort of genes that were translationally regulated upon treatment with 4EGI-1. The 4EGI-1-regulated translatome was negatively correlated to a previously published translatome regulated by eIF4E overexpression in human mammary epithelial cells, which is in agreement with the notion that 4EGI-1 inhibits the eIF4F complex. These data indicate that inhibition of the eIF4F complex by 4EGI-1 or similar translation inhibitors could be a strategy for treating mesothelioma. Genome wide translational profiling identified a large cohort of promising target genes that should be further evaluated for their potential significance in the treatment of MPM.


Subject(s)
Genome, Human , Hydrazones/pharmacology , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Protein Biosynthesis/drug effects , RNA Caps/metabolism , Thiazoles/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Down-Regulation/drug effects , Eukaryotic Initiation Factor-4E/deficiency , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Humans , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Pemetrexed/pharmacology , Pemetrexed/therapeutic use , Phosphoproteins/metabolism , Phosphorylation/drug effects , Pleural Neoplasms/pathology , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Binding , Proteome/metabolism , Reproducibility of Results , Gemcitabine
8.
Oncotarget ; 8(38): 63096-63109, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28968974

ABSTRACT

Malignant mesothelioma has a poor prognosis for which there remains an urgent need for successful treatment approaches. Infection with the Edmonston vaccine strain (MV-Edm) derivative of measles virus results in lysis of cancer cells and has been tested in clinical trials for numerous tumor types including mesothelioma. Many factors play a role in MV-Edm tumor cell selectivity and cytopathic activity while also sparing non-cancerous cells. The MV-Edm receptor CD46 (cluster of differentiation 46) was demonstrated to be significantly higher in mesothelioma cells than in control cells. In contrast, mesothelioma cells are not reliant upon the alternative MV-Edm receptor nectin-4 for entry. MV-Edm treatment of mesothelioma reduced cell viability and also invoked apoptotic cell death. Forced expression of eIF4E or translation stimulation following IGF-I (insulin-like growth factor 1) exposure strengthened the potency of measles virus oncolytic activity. It was also shown that repression of cap-dependent translation by treatment with agents [4EASO, 4EGI-1] that suppress host cell translation or by forcing cells to produce an activated repressor protein diminishes the strength of oncolytic viral efficacy.

9.
Oncotarget ; 6(32): 33165-77, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26431376

ABSTRACT

Vesicular stomatitis virus (VSV) is a potent oncolytic virus for many tumors. VSV that produces interferon-ß (VSV-IFNß) is now in early clinical testing for solid tumors. Here, the preclinical activity of VSV and VSV-IFNß against non-small cell lung cancer (NSCLC) is reported. NSCLC cell lines were treated in vitro with VSV expressing green fluorescence protein (VSV-GFP) and VSV-IFNß. VSV-GFP and VSV-IFNß were active against NSCLC cells. JAK/STAT inhibition with ruxolitinib re-sensitized resistant H838 cells to VSV-IFNß mediated oncolysis. Intratumoral injections of VSV-GFP and VSV-IFNß reduced tumor growth and weight in H2009 nude mouse xenografts (p < 0.01). A similar trend was observed in A549 xenografts. Syngeneic LM2 lung tumors grown in flanks of A/J mice were injected with VSV-IFNß intratumorally. Treatment of LM2 tumors with VSV-IFNß resulted in tumor regression, prolonged survival (p < 0.0001), and cure of 30% of mice. Intratumoral injection of VSV-IFNß resulted in decreased tumor-infiltrating regulatory T cells (Treg) and increased CD8+ T cells. Tumor cell expression of PDL-1 was increased after VSV-IFNß treatment. VSV-IFNß has potent antitumor effects and promotes systemic antitumor immunity. These data support further clinical investigation of VSV-IFNß for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Interferon-beta/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Vesicular stomatitis Indiana virus/physiology , Animals , Carcinoma, Non-Small-Cell Lung/virology , Disease Models, Animal , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Lung Neoplasms/virology , Mice , Mice, Nude , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
10.
Anticancer Res ; 35(10): 5211-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26408679

ABSTRACT

BACKGROUND: Etoposide and other type-II human topoisomerase (TOPOII) poisons are widely used for the treatment of many different cancer types, including non-small cell lung cancer (NSCLC). However, there is a risk for the development of therapy-related secondary leukemia following treatment with these TOPOII poisons. Five to seven years is the typical latency period for the development of secondary leukemia. One of the strategies to overcome this issue is to develop agents that do not act as poisons but still effectively inhibit topoisomerase activity. This has led to the development of acridine-based agents, which are catalytic TOPOII inhibitors, that do not generate DNA strand breaks that can lead to secondary malignancies in in vitro tests. MATERIALS AND METHODS: In this study, we showd antiproliferative activity of a series of acridine-based catalytic inhibitors of TOPOII using four NSCLC cell lines (H460, A549, H2009 and H2030). Cells were treated with four acridine-based compounds for 72 h. RESULTS: The results indicate that these compounds inhibit NSCLC cell proliferation with half-maximal effective concentration (EC50) ranging from 8.15 to 42.09 µM. Combination therapy with cisplatin resulted in increased potency. Poly (ADP-ribose) polymerase cleavage and Guava Nexin assays confirm that the primary mode of cell death was by apoptosis. CONCLUSION: This current work is part of a series of studies for this panel of acridine-based compounds bearing TOPOII-inhibitory activity against different solid tumor types. The acridine-based agents were found to substantially reduce NSCLC cell viability and induce apoptosis. In addition, the acridine-based compounds sensitized cells to cisplatin as measured by cell viability. The results are consistent with prior work on mesothelioma, small-cell lung cancer and pancreatic cancer with this same panel of 9-aminoacridine derivatives. These findings support further development of this type of catalytic TOPOII inhibitor as a novel agent for NSCLC therapy.


Subject(s)
Aminacrine/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA Topoisomerases, Type II/metabolism , Lung Neoplasms/drug therapy , Topoisomerase II Inhibitors/pharmacology , Aminacrine/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Drug Synergism , Humans , Lung Neoplasms/enzymology , Molecular Targeted Therapy
11.
Genes Cancer ; 6(3-4): 144-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26000097

ABSTRACT

Malignant mesothelioma is a devastating disease with a poor prognosis for which there is a clear need for more successful therapeutic approaches. Triptolide, a diterpenoid triepoxide, is a highly effective agent against several cancer types in animal models. Owing to triptolide's poor solubility in water, a water-soluble analog, minnelide, was synthesized. Minnelide is a prodrug of triptolide and is activated by exposure to phosphatases that are found in all body tissues, including blood. Mesothelioma cells were treated in vitro with minnelide or its parent compound, triptolide. Minnelide and triptolide were both found to significantly reduce mesothelioma cell viability and induce apoptosis. The level of Hsp70, a protein that promotes cancer cell survival, was measured in mesothelioma cells before and after treatment with triptolide. Hsp70 levels were decreased in a dose-dependent manner. In addition, triptolide sensitized cells to gemcitabine and pemetrexed as measured by cell viability. Mice bearing mesothelioma flank tumors were treated with daily injections (28 d) of minnelide or saline solution and xenograft tumor growth recorded. Mice displayed significantly reduced tumor burden. These findings support the clinical evaluation of minnelide therapy for mesothelioma.

12.
J Thorac Oncol ; 9(8): 1101-10, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25157763

ABSTRACT

INTRODUCTION: Oncolytic virus therapy is a promising therapy for numerous tumor types. Edmonston-strain measles virus (MV) has been tested in clinical trials for ovarian cancer, glioma, and myeloma. Therefore, the antitumor activity of MV against non-small-cell lung cancer (NSCLC) was assessed. METHODS: Human NSCLC cells and immortalized lung epithelial cell lines, Beas2B, were infected with either MV-producing green fluorescent protein or MV-producing carcinoembryonic antigen. Cells were assessed for viability, induction of apoptosis by caspase and poly-ADP ribose polymerase cleavage, and for viral transgene production. The dependency of MV entry on CD46 and nectin-4 were determined using blocking antibodies. The role of host translational activity on viral replication was assessed by overexpression of eIF4E and translation inhibition. Antitumor activity was assessed by measuring treated NSCLC xenografts from flanks of nude mice. RESULTS: MV infection of NSCLC cells results in potent cell killing in most of the cell lines compared with immortalized Beas2B cells and induces apoptosis. MV infection was prevented by blocking of CD46, however independent of nectin-4 blockade. Tumor weights are diminished after intratumoral injections of MV-producing carcinoembryonic antigen in one of two cell lines and result in detectable viral transgene in serum of mice. CONCLUSIONS: These data indicate that MV is oncolytic for human NSCLC and this was independent of nectin-4 expression. Dysregulated protein translational machinery may play a role in determining tumor tropism in NSCLC. MV combined with gemcitabine could be explored further as chemovirotherapy for NSCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy , Tumor Burden , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoembryonic Antigen/drug effects , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Epithelial Cells/virology , Eukaryotic Initiation Factor-4E/metabolism , Green Fluorescent Proteins/genetics , Humans , Hydrazones/pharmacology , Lung , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Measles Vaccine , Measles virus/genetics , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/metabolism , Mice , Phosphoproteins/metabolism , Sirolimus/pharmacology , Thiazoles/pharmacology , Virus Replication , Gemcitabine
13.
Invest New Drugs ; 32(4): 598-603, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24711125

ABSTRACT

Deranged cap-mediated translation is implicated in the genesis, maintenance and progression of many human cancers including mesothelioma. In this study, disrupting the eIF4F complex by antagonizing the eIF4E-mRNA-cap interaction is assessed as a therapy for mesothelioma. Mesothelioma cells were treated with 4Ei-1, a membrane permeable prodrug that when converted to the active drug, 7-benzyl guanosine monophosphate (7Bn-GMP) displaces capped mRNAs from the eIF4F complex. Colony formation was measured in mesothelioma treated with 4Ei-1 alone or combined with pemetrexed. Proliferation was examined in cells treated with 4Ei-1. Binding to a synthetic cap-analogue was used to study the strength of eIF4F complex activation in lysates exposed to 4Ei-1. 4Ei-1 treatment resulted in a dose dependent decrease in colony formation and cell viability. Combination therapy of 4Ei-1 with pemetrexed further reduced colony number. Formation of eIF4F cap-complex decreased in response to 4Ei-1 exposure. 4Ei-1 is a novel prodrug that reduces proliferation, represses colony formation, diminishes association of eIF4F with the mRNA cap, and sensitizes mesothelioma cells to pemetrexed.


Subject(s)
Mesothelioma/drug therapy , Oncogene Proteins/antagonists & inhibitors , Prodrugs/pharmacology , Protein Biosynthesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Mesothelioma/genetics , Oncogene Proteins/genetics , Pemetrexed , Protein Biosynthesis/genetics , RNA, Messenger/genetics
14.
PLoS One ; 8(11): e81669, 2013.
Article in English | MEDLINE | ID: mdl-24260583

ABSTRACT

BACKGROUND: Aberrant cap-dependent translation is implicated in tumorigenesis in multiple tumor types including mesothelioma. In this study, disabling the eIF4F complex by targeting eIF4E with eIF4E-specific antisense oligonucleotide (4EASO) is assessed as a therapy for mesothelioma. METHODS: Mesothelioma cells were transfected with 4EASO, designed to target eIF4E mRNA, or mismatch-ASO control. Cell survival was measured in mesothelioma treated with 4EASO alone or combined with either gemcitabine or pemetrexed. Levels of eIF4E, ODC, Bcl-2 and ß-actin were assessed following treatment. Binding to a synthetic cap-analogue was used to study the strength of eIF4F complex activation following treatment. RESULTS: eIF4E level and the formation of eIF4F cap-complex decreased in response to 4EASO, but not mismatch control ASO, resulting in cleavage of PARP indicating apoptosis. 4EASO treatment resulted in dose dependent decrease in eIF4E levels, which corresponded to cytotoxicity of mesothelioma cells. 4EASO resulted in decreased levels of eIF4E in non-malignant LP9 cells, but this did not correspond to increased cytotoxicity. Proteins thought to be regulated by cap-dependent translation, Bcl-2 and ODC, were decreased upon treatment with 4EASO. Combination therapy of 4EASO with pemetrexed or gemcitabine further reduced cell number. CONCLUSION: 4EASO is a novel drug that causes apoptosis and selectively reduces eIF4E levels, eIF4F complex formation, and proliferation of mesothelioma cells. eIF4E knockdown results in decreased expression of anti-apoptotic and pro-growth proteins and enhances chemosensitivity.


Subject(s)
Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Mesothelioma/genetics , Oligonucleotides, Antisense/genetics , RNA, Messenger/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Cell Count , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/genetics , Eukaryotic Initiation Factor-4F/metabolism , Gene Expression , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Mesothelioma/metabolism , Mesothelioma/pathology , Molecular Targeted Therapy , Oligonucleotides, Antisense/metabolism , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Pemetrexed , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Gemcitabine
15.
J Thorac Oncol ; 8(9): 1142-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23883783

ABSTRACT

INTRODUCTION: For the majority of patients with non-small-cell lung cancer (NSCLC), response to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is suboptimal. In models of acquired resistance to EGFR-TKI, activation of Akt phosphorylation is frequently observed. Because Akt activation results in downstream initiation of cap-dependent protein translation, we hypothesized that a strategy of targeting cap-dependent translation in combination with erlotinib might enhance therapy. METHODS: NSCLC cells that are wild type for EGFR were assayed for sensitivity to erlotinib. Serum-starved NSCLC cells were assayed for EGFR signaling and downstream pathway activation by immunoblot after stimulation with epidermal growth factor. EGFR signaling and signaling mediators of cap-dependent translation were assayed by immunoblot under serum-replete conditions 24 hours after treatment with erlotinib. Finally, combination treatment with erlotinib and two different cap-dependent translation inhibitors were done to assess the effect on cell viability. RESULTS: EGFR signaling is coupled to activation of cap-dependent translation in EGFR wild-type cells. Erlotinib inhibits EGFR phosphorylation in EGFR-TKI resistant cells, however, results in activation of downstream signaling molecules including Akt and extracellular regulated kinase, ERK 1/2, resulting in maintenance of eukaryotic initiation factor 4F (eIF4F) activation. eIF4F cap-complex formation is maintained in erlotinib-resistant cells, but not in erlotinib-sensitive cells. Finally, using an antisense oligonucleotide against eukaryotic translation initiation factor 4E and a small-molecule inhibitor to disrupt eIF4F formation, we show that cap-dependent translation inhibition can enhance sensitivity to erlotinib. CONCLUSION: The results of these studies support further clinical development of translation inhibitors for treatment of NSCLC in combination with erlotinib.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , RNA Caps/drug effects , Signal Transduction/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Erlotinib Hydrochloride , Eukaryotic Initiation Factor-4F/metabolism , Humans , Immunoblotting , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Quinazolines/pharmacology , Tumor Cells, Cultured
16.
Invest New Drugs ; 30(4): 1443-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21789510

ABSTRACT

Human topoisomerase II (hTopoII) inhibitors are important chemotherapeutic agents in many different settings including treatment of malignant mesothelioma. Topoisomerase poisons, such as etoposide and doxorubicin, function by trapping the DNA-enzyme covalent complex producing DNA strand breaks which can ultimately lead to cancer cell death, as well as development of secondary malignancies. While these compounds have been used successfully in treating a wide variety of cancers, their use against mesothelioma has been limited. This study evaluates the anti-proliferative activity of series of acridine-based catalytic inhibitors of hTopoII using four mesothelioma cell lines (H513, H2372, H2461, and H2596). The results indicate these compounds inhibit malignant cell proliferation with EC(50) values ranging from 6.9 to 32 µM. Experiments are also performed that show that combination therapies may be used to increase potency. Based on the results of PARP cleavage and Guava Nexin assay, it is concluded that the primary mode of cell death is by apoptosis. The results are consistent with prior work involving pancreatic cancer and hTopoII catalytic inhibitors and suggest substituted acridines may hold promise in treating malignant mesothelioma.


Subject(s)
Acridines/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Mesothelioma/drug therapy , Mesothelioma/pathology , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Acridines/chemistry , Acridines/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Flow Cytometry , Humans , Topoisomerase II Inhibitors/chemistry
17.
Int J Oncol ; 40(2): 577-82, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22076560

ABSTRACT

Initiation of protein translation by the 5' mRNA cap is a tightly regulated step in cell growth and proliferation. Aberrant activation of cap-dependent translation is a hallmark of many cancers including non-small cell lung cancer. The canonical signaling mechanisms leading to translation initiation include activation of the Akt/mTOR pathway in response to the presence of nutrients and growth factors. We have previously observed that inhibition of c-jun N-terminal kinase (JNK) leads to inactivation of cap-dependent translation in mesothelioma cells. Since JNK is involved in the genesis of non-small cell lung cancer (NSCLC), we hypothesized that JNK could also be involved in activating cap-dependent translation in NSCLC cells and could represent an alternative pathway regulating translation. In a series of NSCLC cell lines, inhibition of JNK using SP600125 resulted in inhibition of 4E-BP1 phosphorylation and a decrease in formation of the cap-dependent translation complex, eIF4F. Furthermore, we show that JNK-mediated inhibition of translation is independent of mTOR. Our data provide evidence that JNK is involved in the regulation of translation and has potential as a therapeutic target in NSCLC.


Subject(s)
JNK Mitogen-Activated Protein Kinases/physiology , Protein Biosynthesis , RNA Caps/genetics , Adaptor Proteins, Signal Transducing/metabolism , Anthracenes/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Eukaryotic Initiation Factor-4G/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , RNA Caps/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
18.
Invest New Drugs ; 28(1): 20-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19132295

ABSTRACT

INTRODUCTION: Simocyclinone D-8 (SD8), a semi-synthetic compound derived from yeast, has been shown to decrease the proliferation of MCF-7 breast cancer cells. It has been shown to be a potent bacterial DNA gyrase inhibitor, a homologue of human topoisomerase II (hTopoII). We tested SD8 activity alone and in combination with cisplatin against malignant mesothelioma (MM) and non-small cell cancer (NSCLC) cell lines. METHODS: Inhibition of hTopoII supercoiling function by SD8 and a known hTopoII poison, etoposide, were done by in vitro assay using purified hTopoII and kinetoplast DNA as the substrate. The DNA products were analyzed by agarose gel electrophoresis after treatment with increasing concentrations of each drug. Mesothelioma cell lines (H2373, H2461 and H2596) and NSCLC cell lines (H2030, H460, and H2009) grown in RPMI with 10% calf serum were used. Non-malignant mesothelial cells, LP9, were grown in 1:1 ratio of MCDB:199E medium supplemented with 15% calf serum, 0.4 microg/mL hydrocortisone, and 15 ng/mL epidermal grown factor. Cell proliferation assays were performed in 96-well plates using the CCK-8 kit (Dojindo inc.). Cells were treated for 72 h with various SD8 concentrations and controls containing equal volume of the vehicle, DMSO. Treated cells were assayed for the induction of apoptosis with poly ADP-ribose polymerase-1 (PARP) cleavage assay. RESULTS: Biochemical assays revealed that the IC(50) for hTopoII inhibition was 100 microM for SD8 and 400 microM for etoposide. SD8 inhibited hTopoII function without inducing DNA cleavage events. SD8 inhibited the growth of NSCLC and Mesothelioma cells with IC(50) ranging from 75-125 microM. Furthermore, SD8 was not toxic to non-transformed primary mesothelial cell line, LP9 at the IC(50) doses. SD8 induced apoptosis in all cell lines tested. CONCLUSIONS: SD8 inhibits hTopoII in vitro without inducing DNA strands breaks and has significant activity against NSCLC and MM cell lines. While doses required for SD8 anticancer activity are unlikely to be achieved in vivo, chemical modifications to SD8 to increase its potency could lead to improved therapies for these diseases.


Subject(s)
Biocatalysis/drug effects , Enzyme Inhibitors/pharmacology , Topoisomerase II Inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/pharmacology , DNA Topoisomerases, Type II/metabolism , Drug Screening Assays, Antitumor , Glycosides/pharmacology , Humans
19.
J Thorac Oncol ; 2(9): 789-95, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17805054

ABSTRACT

INTRODUCTION: Mutations in Ras family genes are rare in malignant mesothelioma. The role of activation of the Ras signaling pathway in the pathogenesis of mesothelioma is not clear. METHODS: We studied the activation status of the Ras pathway and the status of other Ras-associated kinases in a panel of human mesothelioma cell lines. In addition, we tested the effect of inhibition of several kinase pathways on mesothelioma cell proliferation. The potential role of kinase signaling on the regulation of cap-dependent translation was also studied. RESULTS: In general, Ras-guanosine triphosphate (GTP) was higher in mesothelioma cell lines when compared with a nontransformed mesothelial cell line (LP9). Furthermore, known Ras effectors such as extracellular-regulated kinase 1/2, p38 mitogen-activated protein kinase, and c-Jun N-terminal kinase were found to be active in most of the mesothelioma cell lines tested. Exposure to specific inhibitors of extracellular-regulated kinase 1/2 (U0126) and c-Jun N-terminal kinase (SP600125) significantly decreased the proliferation of H2596 and H2373 cells compared with mock-treated cells. SP600125-mediated c-Jun N-terminal kinase inhibition, but not extracellular-regulated kinase 1/2 inhibition, resulted in a decrease in phosphorylation of 4E-BP1, consequently decreasing cap-dependent activation. CONCLUSIONS: These experiments provide a rationale for targeting Ras and associated signaling pathways in mesothelioma and also suggest cap-dependent translation as one mechanism by which Ras induces proliferation in this disease.


Subject(s)
DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Mesothelioma/genetics , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing/metabolism , Anthracenes/pharmacology , Apoptosis , Blotting, Western , Butadienes/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mesothelioma/enzymology , Mesothelioma/pathology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Nitriles/pharmacology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Polymerase Chain Reaction , Repressor Proteins , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
J Gastroenterol ; 42(3): 196-204, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17380277

ABSTRACT

BACKGROUND: Genetic instability is a hallmark of malignancy, and microsatellite instability is a widely appreciated mechanism of generating genetic changes. We have recently observed four markers clustered on chromosome 20 that showed the effects of microsatellite instability in the gastric adenocarcinoma cell line SNU-1. Each affected marker had alleles of three different sizes. The aim of this study was to investigate the origin for this high-density polymorphism on a single chromosome. METHODS: The high polymorphism located on chromosome 20 was confirmed using 37 additional markers. To further evaluate this finding, 15 clones of the cell line were generated and then assayed with the triallelic markers. RESULTS: All told, almost a third of the markers on chromosome 20 had triallelic patterns, but only 0.3% of the markers not on chromosome 20 showed this result. The number of clones showing allelic variation was an average of 50% greater for chromosome 20 markers than for markers elsewhere. A karyotype analysis showed that the progenitor cell line of SNU-1 was trisomic for chromosome 20, and the high polymorphism on that chromosome is almost certainly due to the trisomy. CONCLUSIONS: Not only are there more chromosome copies and therefore more gene copies subject to mutation in cells containing trisomy, but also more mutations may be passed on to the progeny. This elevated polymorphism increases the repertoire of genetic changes that could affect cellular growth, and may independently increase genomic instability.


Subject(s)
Adenocarcinoma/genetics , Chromosomes, Human, Pair 20/genetics , Polymorphism, Genetic , Stomach Neoplasms/genetics , Trisomy/genetics , Trisomy/pathology , Adenocarcinoma/pathology , Biomarkers, Tumor , Cell Line, Tumor/pathology , Humans , Karyotyping , Microsatellite Instability , Polymerase Chain Reaction , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...