Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38279277

ABSTRACT

Endometrial cancer is the most frequent malignant tumor of the female reproductive tract but lacks effective therapy. EphA2, a receptor tyrosine kinase, is overexpressed by various cancers including endometrial cancer and is associated with poor clinical outcomes. In preclinical models, EphA2-targeted drugs had modest efficacy. To discover potential synergistic partners for EphA2-targeted drugs, we performed a high-throughput drug screen and identified panobinostat, a histone deacetylase inhibitor, as a candidate. We hypothesized that combination therapy with an EphA2 inhibitor and panobinostat leads to synergistic cell death. Indeed, we found that the combination enhanced DNA damage, increased apoptosis, and decreased clonogenic survival in Ishikawa and Hec1A endometrial cancer cells and significantly reduced tumor burden in mouse models of endometrial carcinoma. Upon RNA sequencing, the combination was associated with downregulation of cell survival pathways, including senescence, cyclins, and cell cycle regulators. The Axl-PI3K-Akt-mTOR pathway was also decreased by combination therapy. Together, our results highlight EphA2 and histone deacetylase as promising therapeutic targets for endometrial cancer.


Subject(s)
Endometrial Neoplasms , Histone Deacetylase Inhibitors , Receptor, EphA2 , Animals , Female , Humans , Mice , Apoptosis , Cell Line, Tumor , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Histone Deacetylase Inhibitors/therapeutic use , Panobinostat/pharmacology , Panobinostat/therapeutic use , Phosphatidylinositol 3-Kinases , Molecular Targeted Therapy , Receptor, EphA2/antagonists & inhibitors
2.
Mol Cancer Ther ; 19(11): 2396-2406, 2020 11.
Article in English | MEDLINE | ID: mdl-32943548

ABSTRACT

Here, we examined the role of EP-100 [luteinizing hormone-releasing hormone (LHRH) ligand joined to a lytic peptide], improving the efficacy of immune checkpoint blockade. LHRH-R-positive murine ovarian cancer cells (ID8, IG10, IF5, and 2C12) were sensitive to EP-100 and were specifically killed at low micromolar levels through LHRH-R. EP-100 increased PD-L1 levels on murine ovarian cancer cells. In vivo syngeneic mouse models (ID8 and IG10) demonstrated that single-agent EP-100 reduced tumor volume, tumor weight, and ascites volume. The greatest reductions in tumor and ascites volume were observed with the combination of EP-100 with an anti-PD-L1 antibody. Immune profiling analysis showed that the population of CD8+ T cells, natural killer cells, dendritic cells, and macrophages were significantly increased in tumor and ascitic fluid samples treated with anti-PD-L1, EP-100, and the combination. However, monocytic myeloid suppressor cells, B cells, and regulatory T cells were decreased in tumors treated with anti-PD-L1, EP-100, or the combination. In vitro cytokine arrays revealed that EP-100 induced IL1α, IL33, CCL20, VEGF, and Low-density lipoprotein receptor (LDLR) secretion. Of these, we validated increasing IL33 levels following EP-100 treatment in vitro and in vivo; we determined the specific biological role of CD8+ T-cell activation with IL33 gene silencing using siRNA and Cas9-CRISPR approaches. In addition, we found that CD8+ T cells expressed very low level of LHRH-R and were not affected by EP-100. Taken together, EP-100 treatment had a substantial antitumor efficacy, particularly in combination with an anti-PD-L1 antibody. These results warrant further clinical development of this combination.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Peptide Fragments/pharmacology , Receptors, LHRH/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Molecular Targeted Therapy/methods , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 18(5): 969-979, 2019 05.
Article in English | MEDLINE | ID: mdl-30926640

ABSTRACT

EP-100 is a synthetic lytic peptide that specifically targets the gonadotropin-releasing hormone receptor on cancer cells. To extend the utility of EP-100, we aimed to identify effective combination therapies with EP-100 for ovarian cancer and explore potential mechanisms of this combination. A series of in vitro (MTT assay, immunoblot analysis, reverse-phase protein array, comet assay, and immunofluorescence staining) and in vivo experiments were carried out to determine the biological effects of EP-100 alone and in combination with standard-of-care drugs. EP-100 decreased the viability of ovarian cancer cells and reduced tumor growth in orthotopic mouse models. Of five standard drugs tested (cisplatin, paclitaxel, doxorubicin, topotecan, and olaparib), we found that the combination of EP-100 and olaparib was synergistic in ovarian cancer cell lines. Further experiments revealed that combined treatment of EP-100 and olaparib significantly increased the number of nuclear foci of phosphorylated histone H2AX. In addition, the extent of DNA damage was significantly increased after treatment with EP-100 and olaparib in comet assay. We performed reverse-phase protein array analyses and identified that the PI3K/AKT pathway was inhibited by EP-100, which we validated with in vitro experiments. In vivo experiment using the HeyA8 mouse model demonstrated that mice treated with EP-100 and olaparib had lower tumor weights (0.06 ± 0.13 g) than those treated with a vehicle (1.19 ± 1.09 g), EP-100 alone (0.62 ± 0.78 g), or olaparib alone (0.50 ± 0.63 g). Our findings indicate that combining EP-100 with olaparib is a promising therapeutic strategy for ovarian cancer.


Subject(s)
Ovarian Neoplasms/drug therapy , Peptides/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Receptors, LHRH/genetics , Animals , BRCA1 Protein/genetics , Cell Proliferation/drug effects , Cisplatin/pharmacology , DNA Damage/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Peptides/chemical synthesis , Phthalazines/pharmacology , Piperazines/pharmacology , Receptors, LHRH/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Neuro Oncol ; 18(4): 486-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995790

ABSTRACT

BACKGROUND: We recently demonstrated that brain endothelial cells and astrocytes protect cancer cells from chemotherapy through an endothelin-dependent signaling mechanism. Here, we evaluated the efficacy of macitentan, a dual endothelin receptor (ETAR and ETBR) antagonist, in the treatment of experimental breast and lung cancer brain metastases. METHODS: The effect of macitentan on astrocyte- and brain endothelial cell-mediated chemoprotective properties was measured in cytotoxic assays. We compared survival of mice bearing established MDA-MB-231 breast cancer or PC-14 non-small cell lung cancer (NSCLC) brain metastases that were treated with vehicle, macitentan, paclitaxel, or macitentan plus paclitaxel. Cell division, apoptosis, tumor vasculature, and expression of survival-related proteins were assessed by immunofluorescent microscopy. RESULTS: Cancer cells and tumor-associated endothelial cells expressed activated forms of AKT and MAPK in vehicle- and paclitaxel-treated groups in both metastasis models, but these proteins were downregulated in metastases of mice that received macitentan. The survival-related proteins Bcl2L1, Gsta5, and Twist1 that localized to cancer cells and tumor-associated endothelial cells in vehicle- and paclitaxel-treated tumors were suppressed by macitentan. Macitentan or paclitaxel alone had no effect on survival. However, when macitentan was combined with paclitaxel, we noted a significant reduction in cancer cell division and marked apoptosis of both cancer cells and tumor-associated endothelial cells. Moreover, macitentan plus paclitaxel therapy significantly increased overall survival by producing complete responses in 35 of 35 mice harboring brain metastases. CONCLUSIONS: Dual antagonism of ETAR and ETBR signaling sensitizes experimental brain metastases to paclitaxel and may represent a new therapeutic option for patients with brain metastases.


Subject(s)
Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Pyrimidines/pharmacology , Receptors, Endothelin/chemistry , Sulfonamides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Therapy, Combination , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , NIH 3T3 Cells , Receptors, Endothelin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 21(20): 4630-41, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26106074

ABSTRACT

PURPOSE: The objective of the study was to determine whether astrocytes and brain endothelial cells protect glioma cells from temozolomide through an endothelin-dependent signaling mechanism and to examine the therapeutic efficacy of the dual endothelin receptor antagonist, macitentan, in orthotopic models of human glioblastoma. EXPERIMENTAL DESIGN: We evaluated several endothelin receptor antagonists for their ability to inhibit astrocyte- and brain endothelial cell-induced protection of glioma cells from temozolomide in chemoprotection assays. We compared survival in nude mice bearing orthotopically implanted LN-229 glioblastomas or temozolomide-resistant (LN-229(Res) and D54(Res)) glioblastomas that were treated with macitentan, temozolomide, or both. Tumor burden was monitored weekly with bioluminescence imaging. The effect of therapy on cell division, apoptosis, tumor-associated vasculature, and pathways associated with cell survival was assessed by immunofluorescent microscopy. RESULTS: Only dual endothelin receptor antagonism abolished astrocyte- and brain endothelial cell-mediated protection of glioma cells from temozolomide. In five independent survival studies, including temozolomide-resistant glioblastomas, 46 of 48 (96%) mice treated with macitentan plus temozolomide had no evidence of disease (P < 0.0001), whereas all mice in other groups died. In another analysis, macitentan plus temozolomide therapy was stopped in 16 mice after other groups had died. Only 3 of 16 mice eventually developed recurrent disease, 2 of which responded to additional cycles of macitentan plus temozolomide. Macitentan downregulated proteins associated with cell division and survival in glioma cells and associated endothelial cells, which enhanced their sensitivity to temozolomide. CONCLUSIONS: Macitentan plus temozolomide are well tolerated, produce durable responses, and warrant clinical evaluation in glioblastoma patients.


Subject(s)
Dacarbazine/analogs & derivatives , Endothelin Receptor Antagonists/pharmacology , Glioblastoma/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Dacarbazine/pharmacology , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glioblastoma/pathology , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...