Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Gynecol Oncol ; 164(1): 170-180, 2022 01.
Article in English | MEDLINE | ID: mdl-34844776

ABSTRACT

BACKGROUND: Progress in immunotherapy use for gynecologic malignancies is hampered by poor tumor antigenicity and weak T cell infiltration of the tumor microenvironment (TME). Wnt/ß-catenin pathway modulation demonstrated patient benefit in clinical trials as well as enhanced immune cell recruitment in preclinical studies. The purpose of this study was to characterize the pathways by which Wnt/ß-catenin modulation facilitates a more immunotherapy-favorable TME. METHODS: Human tumor samples and in vivo patient-derived xenograft and syngeneic murine models were administered Wnt/ß-catenin modulating agents DKN-01 and CGX-1321 individually or in sequence. Analytical methods included immunohistochemistry, flow cytometry, multiplex cytokine/chemokine array, and RNA sequencing. RESULTS: DKK1 blockade via DKN-01 increased HLA/MHC expression in human and murine tissues, correlating with heightened expression of known MHC I regulators: NFkB, IL-1, LPS, and IFNy. PORCN inhibition via CGX-1321 increased production of T cell chemoattractant CXCL10, providing a mechanism for observed increases in intra-tumoral T cells. Diverse leukocyte recruitment was noted with elevations in B cells and macrophages, with increased tumor expression of population-specific chemokines. Sequential DKK1 blockade and PORCN inhibition decreased tumor burden as evidenced by reduced omental weights. CONCLUSIONS: Wnt/ß-catenin pathway modulation increases MHC I expression and promotes tumor leukocytic infiltration, facilitating a pro-immune TME associated with decreased tumor burden. This intervention overcomes common tumor immune-evasion mechanisms and may render ovarian tumors susceptible to immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Genital Neoplasms, Female/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Drug Synergism , Female , Genes, MHC Class I/genetics , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/therapy , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
2.
J Tissue Eng ; 12: 20417314211055015, 2021.
Article in English | MEDLINE | ID: mdl-34820113

ABSTRACT

Among gynecologic malignancies, ovarian cancer (OC) has the poorest survival rate, and its clinical management remains challenging due to the high rate of recurrence and chemoresistance. Improving survival for these patients is critical, although this requires the ability to translate preclinical studies to actual patient care: bench to bedside and back. Our objective was to develop a preclinical model that accurately represents tumor biology and its microenvironment. We utilized SKOV-3, OVCAR-8, and CS-99 cell lines to show that this model was suitable for in vitro assessment of cell proliferation. We tested OC cells independently and in co-culture with cancer associated fibroblasts (CAFs) or immune cells. Additionally, we used patient-derived ovarian carcinoma and carcinosarcoma samples to show that the system maintains the histologic morphology of the primary tissue after 7 days. Moreover, we tested the response to chemotherapy using both cell lines and patient-derived tumor specimens and confirmed that cell death was significantly higher in the treated group compared to the vehicle group. Finally, we immune profiled the 3-D model containing patient tissue after several days in the bioreactor system and revealed that the immune populations are still present. Our data suggest that this model is a suitable preclinical model to aid in research that will ultimately impact the treatment of patients with gynecologic cancer.

3.
Mol Cancer Ther ; 20(3): 602-611, 2021 03.
Article in English | MEDLINE | ID: mdl-33323456

ABSTRACT

The immunosuppressive effects of TGFß promotes tumor progression and diminishes response to therapy. In this study, we used ID8-p53-/- tumors as a murine model of high-grade serous ovarian cancer. An mAb targeting all three TGFß ligands was used to neutralize TGFß. Ascites and omentum were collected and changes in T-cell response were measured using flow. Treatment with anti-TGFß therapy every other day following injection of tumor cells resulted in decreased ascites volume (4.1 mL vs. 0.7 mL; P < 0.001) and improved the CD8:Treg ratio (0.37 vs. 2.5; P = 0.02) compared with untreated mice. A single dose of therapy prior to tumor challenge resulted in a similar reduction of ascites volume (2.7 vs. 0.67 mL; P = 0.002) and increased CD8:Tregs ratio (0.36 vs. 1.49; P = 0.007), while also significantly reducing omental weight (114.9 mg vs. 93.4 mg; P = 0.017). Beginning treatment before inoculation with tumor cells and continuing for 6 weeks, we observe similar changes and prolonged overall survival (median 70 days vs. 57.5 days). TGFß neutralization results in favorable changes to the T-cell response within the tumor microenvironment, leading to decreased tumor progression in ovarian cancer. The utilization of anti-TGFß therapy may be an option for management in patients with ovarian cancer to improve clinical outcomes and warrants further investigation.


Subject(s)
Ovarian Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Survival Analysis , Transfection
4.
Cancer Med ; 10(2): 709-717, 2021 01.
Article in English | MEDLINE | ID: mdl-33369199

ABSTRACT

OBJECTIVE: Patients with epithelial ovarian cancer (EOC) typically present with late-stage disease, posing a significant challenge to treatment. Although taxane and platinum-based chemotherapy plus surgical debulking are initially effective, EOC is marked by frequent recurrence with resistant disease. Immunotherapy represents an appealing treatment paradigm given the ability of immune cells to engage metastatic sites and impede recurrence; however, response rates to checkpoint blockade in ovarian cancer have been disappointing. Here, we tested whether class I HDAC inhibition can promote anti-tumor T cell responses in a spontaneous and nonspontaneous murine model of EOC. METHODS: We used the spontaneous Tg-MISIIR-Tag and nonspontaneous ID8 models of murine ovarian cancer to test this hypothesis. Whole tumor transcriptional changes were assessed using the nCounter PanCancer Mouse Immune Profiling Panel. Changes in select protein expression of regulatory and effector T cells were measured by flow cytometry. RESULTS: We found that treatment with the class I HDAC inhibitor entinostat upregulated pathways and genes associated with CD8 T cell cytotoxic function, while downregulating myeloid derived suppressor cell chemoattractants. Suppressive capacity of regulatory T cells within tumors and associated ascites was significantly reduced, reversing the CD8-Treg ratio. CONCLUSIONS: Our findings suggest class I HDAC inhibition can promote activation of intratumoral CD8 T cells, potentially by compromising suppressive networks within the EOC tumor microenvironment. In this manner, class I HDAC inhibition might render advanced-stage EOC susceptible to immunotherapeutic treatment modalities.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Ovarian Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Female , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Ther Adv Med Oncol ; 12: 1758835920913798, 2020.
Article in English | MEDLINE | ID: mdl-32313567

ABSTRACT

BACKGROUND: The Wnt/ß-catenin pathway is linked to tumorigenesis in a variety of tumors and promotes T cell exclusion and resistance to checkpoint inhibitors. We sought to determine whether a small molecule inhibitor of this pathway, WNT974, would impair tumor growth, affect gene expression patterns, and improve the immune response in human and murine ovarian cancer models. METHODS: Human ovarian cancer cells were treated with WNT974 in vitro. RNAseq libraries were constructed and differences in gene expression patterns between responders and nonresponders were compared to The Cancer Genome Atlas (TCGA). Mice with subcutaneous or intraperitoneal ID8 ovarian cancer tumors were treated with WNT974, paclitaxel, combination, or control. Tumor growth and survival were measured. Flow cytometry and ß-TCR repertoire analysis were used to determine the immune response. RESULTS: Gene expression profiling revealed distinct signatures in responders and nonresponders, which strongly correlated with T cell infiltration patterns in the TCGA analysis of ovarian cancer. WNT974 inhibited tumor growth, prevented ascites formation, and prolonged survival in mouse models. WNT974 increased the ratio of CD8+ T cells to T regulatory cells (Tregs) in tumors and enhanced the effector functions of infiltrating CD4+ and CD8+ T cells. Treatment also decreased the expression of inhibitory receptors on CD8+ T cells. Combining WNT974 with paclitaxel further reduced tumor growth, prolonged survival, and expanded the T cell repertoire. CONCLUSIONS: These findings suggest that inhibiting the Wnt/ß-catenin pathway may have a potent immunomodulatory effect in the treatment of ovarian cancer, particularly when combined with paclitaxel.

6.
Cancers (Basel) ; 12(3)2020 Mar 24.
Article in English | MEDLINE | ID: mdl-32213921

ABSTRACT

In ovarian cancer, upregulation of the Wnt/ß-catenin pathway leads to chemoresistance and correlates with T cell exclusion from the tumor microenvironment (TME). Our objectives were to validate these findings in an independent cohort of ovarian cancer subjects and determine whether inhibiting the Wnt pathway in a syngeneic ovarian cancer murine model could create a more T-cell-inflamed TME, which would lead to decreased tumor growth and improved survival. We preformed RNA sequencing in a cohort of human high grade serous ovarian carcinoma subjects. We used CGX1321, an inhibitor to the porcupine (PORCN) enzyme that is necessary for secretion of WNT ligand, in mice with established ID8 tumors, a murine ovarian cancer cell line. In order to investigate the effect of decreased Wnt/ß-catenin pathway activity in the dendritic cells (DCs), we injected ID8 cells in mice that lacked ß-catenin specifically in DCs. Furthermore, to understand how much the effects of blocking the Wnt/ß-catenin pathway are dependent on CD8+ T cells, we injected ID8 cells into mice with CD8+ T cell depletion. We confirmed a negative correlation between Wnt activity and T cell signature in our cohort. Decreasing WNT ligand production resulted in increases in T cell, macrophage and dendritic cell functions, decreased tumor burden and improved survival. Reduced tumor growth was found in mice that lacked ß-catenin specifically in DCs. When CD8+ T cells were depleted, CGX1321 treatment did not have the same magnitude of effect on tumor growth. Our investigation confirmed an increase in Wnt activity correlated with a decreased T-cell-inflamed environment; a relationship that was further supported in our pre-clinical model that suggests inhibiting the Wnt/ß-catenin pathway was associated with decreased tumor growth and improved survival via a partial dependence on CD8+ T cells.

7.
Cancer ; 124(24): 4657-4666, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30423192

ABSTRACT

BACKGROUND: Ovarian cancer is poorly immunogenic; however, increased major histocompatibility complex class II (MHCII) expression correlates with improved immune response and prolonged survival in patients with ovarian cancer. The authors previously demonstrated that the histone deacetylase inhibitor entinostat increases MHCII expression on ovarian cancer cells. In the current study, they evaluated whether entinostat treatment and resultant MHCII expression would enhance beneficial immune responses and impair tumor growth in mice with ovarian cancer. METHODS: C57BL/6 mice bearing intraperitoneal ID8 tumors were randomized to receive entinostat 20 mg/kg daily versus control. Changes in messenger RNA (mRNA) expression of 46 genes important for antitumor immunity were evaluated using NanoString analysis, and multicolor flow cytometry was used to measure changes in protein expression and tumor-infiltrating immune cells. RESULTS: Entinostat treatment decreased the growth of both subcutaneously and omental ID8 tumors and prolonged survival in immunocompetent C57BL/6 mice. NanoString analysis revealed significant changes in mRNA expression in 21 of 46 genes, including increased expression of the MHCI pathway, the MHCII transactivator (CIITA), interferon γ, and granzyme B. C57BL/6 mice that received entinostat had increased MHCII expression on omental tumor cells and a higher frequency of tumor-infiltrating, CD8-positive T cells by flow cytometry. In immunocompromised mice, treatment with entinostat had no effect on tumor size and did not increase MHCII expression. CONCLUSIONS: In the current murine ovarian cancer model, entinostat treatment enhances beneficial immune responses. Moreover, these antitumor effects of entinostat are dependent on an intact immune system. Future studies combining entinostat with checkpoint inhibitors or other immunomodulatory agents may achieve more durable antitumor responses in patients with ovarian cancer.


Subject(s)
Benzamides/administration & dosage , Histocompatibility Antigens Class II/genetics , Histone Deacetylase Inhibitors/administration & dosage , Ovarian Neoplasms/drug therapy , Pyridines/administration & dosage , Up-Regulation , Adaptive Immunity , Animals , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class II/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunocompromised Host , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Precision Medicine , Pyridines/pharmacology , Random Allocation , Trans-Activators/genetics , Xenograft Model Antitumor Assays
8.
Cancer Lett ; 436: 75-86, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30120964

ABSTRACT

Ovarian cancer is the fifth leading cause of cancer-related deaths among women in the United States. Although most patients respond to frontline therapy, virtually all patients relapse with chemoresistant disease. This study addresses the hypothesis that carboplatin or tamoxifen + FTY720, a sphingosine analogue, will minimize or circumvent drug-resistance in ovarian cancer cells and tumor models. In vitro data demonstrate that FTY720 sensitized two drug-resistant (A2780. cp20, HeyA8. MDR) and two high-grade serous ovarian cancer cell lines (COV362, CAOV3) to carboplatin, a standard of care for patients with ovarian cancer, and to the selective estrogen receptor modulator tamoxifen. FTY720 + tamoxifen was synergistic in vitro, and combinations of FTY720 + carboplatin or + tamoxifen were more effective than each single agent in a patient-derived xenograft model of ovarian carcinoma. FTY720 + tamoxifen arrested tumor growth. FTY720 + carboplatin induced tumor regressions, with tumor volumes reduced by ∼86% compared to initial tumor volumes. Anti-tumor efficacy was concomitant with increases in intracellular proapoptotic lipid ceramide. The data suggest that FTY720 + tamoxifen or carboplatin may be effective in treating ovarian tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Fingolimod Hydrochloride/pharmacology , Ovarian Neoplasms/drug therapy , Tamoxifen/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Carboplatin/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Fingolimod Hydrochloride/administration & dosage , Humans , Mice , Ovarian Neoplasms/pathology , Tamoxifen/administration & dosage
10.
Mol Cancer Res ; 16(5): 813-824, 2018 05.
Article in English | MEDLINE | ID: mdl-29523763

ABSTRACT

While high-grade serous ovarian carcinoma (HGSOC) is the most common histologic subtype of ovarian cancer, significant tumor heterogeneity exists. In addition, chemotherapy induces changes in gene expression and alters the mutational profile. To evaluate the notion that patients with HGSOC could be better classified for optimal treatment based on gene expression, we compared genetic variants [by DNA next-generation sequencing (NGS) using a 50 gene Ion Torrent panel] and gene expression (using the NanoString PanCancer 770 gene Panel) in the tumor from 20 patients with HGSOC before and after neoadjuvant chemotherapy (NACT). NGS was performed on plasma cell free DNA (cfDNA) on a select group of patients (n = 14) to assess the utility of using cfDNA to monitor these changes. A total of 86 genes had significant changes in RNA expression after NACT. Thirty-eight genetic variants (including SNPs) from 6 genes were identified in tumors pre-NACT, while 59 variants from 19 genes were detected in the cfDNA. The number of DNA variants were similar after NACT. Of the 59 variants in the plasma pre-NACT, only 6 persisted, whereas 33 of 38 specific variants in the tumor DNA remained unchanged. Pathway analysis showed the most significant alterations in the cell cycle and DNA damage pathways.Implications: Gene expression profiles at the time of interval debulking provide additional genetic information that could help impact treatment decisions after NACT; although, continued collection and analysis of matched tumor and cfDNA from multiple time points are needed to determine the role of cfDNA in the management of HGSOC. Mol Cancer Res; 16(5); 813-24. ©2018 AACR.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Neoadjuvant Therapy/methods , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Aged , Aged, 80 and over , Cohort Studies , Cystadenocarcinoma, Serous/pathology , Female , Humans , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/pathology
11.
Clin Cancer Res ; 23(21): 6529-6540, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28778862

ABSTRACT

Purpose: A hallmark of neoplasia is increased ribosome biogenesis, and targeting this process with RNA polymerase I (Pol I) inhibitors has shown some efficacy. We examined the contribution and potential targeting of ribosomal machinery in chemotherapy-resistant and -sensitive models of ovarian cancer.Experimental Design: Pol I machinery expression was examined, and subsequently targeted with the Pol I inhibitor CX-5461, in ovarian cancer cell lines, an immortalized surface epithelial line, and patient-derived xenograft (PDX) models with and without chemotherapy. Effects on viability, Pol I occupancy of rDNA, ribosomal content, and chemosensitivity were examined.Results: In PDX models, ribosomal machinery components were increased in chemotherapy-treated tumors compared with controls. Thirteen cell lines were sensitive to CX-5461, with IC50s 25 nmol/L-2 µmol/L. Interestingly, two chemoresistant lines were 10.5- and 5.5-fold more sensitive than parental lines. CX-5461 induced DNA damage checkpoint activation and G2-M arrest with increased γH2AX staining. Chemoresistant cells had 2- to 4-fold increased rDNA Pol I occupancy and increased rRNA synthesis, despite having slower proliferation rates, whereas ribosome abundance and translational efficiency were not impaired. In five PDX models treated with CX-5461, one showed a complete response, one a 55% reduction in tumor volume, and one maintained stable disease for 45 days.Conclusions: Pol I inhibition with CX-5461 shows high activity in ovarian cancer cell lines and PDX models, with an enhanced effect on chemoresistant cells. Effects occur independent of proliferation rates or dormancy. This represents a novel therapeutic approach that may have preferential activity in chemoresistant populations. Clin Cancer Res; 23(21); 6529-40. ©2017 AACR.


Subject(s)
Benzothiazoles/administration & dosage , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Naphthyridines/administration & dosage , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Benzothiazoles/adverse effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Naphthyridines/adverse effects , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA Polymerase I/antagonists & inhibitors , RNA Polymerase I/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Nucl Med Biol ; 47: 23-30, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28104527

ABSTRACT

INTRODUCTION: Novel therapies that effectively kill both differentiated cancer cells and cancer initiating cells (CICs), which are implicated in causing chemotherapy-resistance and disease recurrence, are needed to reduce the morbidity and mortality of ovarian cancer. These studies used monoclonal antibody (mAb) 376.96, which recognizes a B7-H3 epitope expressed on ovarian cancer cells and CICs, as a carrier molecule for targeted α-particle radioimmunotherapy (RIT) in preclinical models of human ovarian cancer. METHODS: mAb 376.96 was conjugated to the chelate 2-(4-isothiocyanotobenzyl)-1,4,7,10-tetraaza-1,4,7,10-tetra-(2-carbamoylmethyl)-cyclododecane (TCMC) and radiolabeled with 212Pb, a source of α-particles. In vitro Scatchard assays determined the specific binding of 212Pb-376.96 to adherent differentiated or non-adherent CIC-enriched ES-2 and A2780cp20 ovarian cancer cells. Adherent ovarian cancer cells and non-adherent CIC-enriched tumorspheres treated in vitro with 212Pb-376.96 or the irrelevant isotype-matched 212Pb-F3-C25 were assessed for clonogenic survival. Mice bearing i.p. ES-2 or A2780cp20 xenografts were injected i.p. with 0.17-0.70MBq 212Pb-376.96 or 212Pb-F3-C25 and were used for in vivo imaging, ex vivo biodistribution, and therapeutic survival studies. RESULTS: 212Pb-376.96 was obtained in high yield and purity (>98%); Kd values ranged from 10.6-26.6nM for ovarian cancer cells, with 104-105 binding sites/cell. 212Pb-376.96 inhibited the clonogenic survival of ovarian cancer cells up to 40 times more effectively than isotype-matched control 212Pb-F3-C25; combining 212Pb-376.96 with carboplatin significantly decreased clonogenic survival compared to either agent alone. In vivo imaging and biodistribution analysis 24h after i.p. injection of 212Pb-376.96 showed high peritoneal retention and tumor tissue accumulation (28.7% ID/g in ES-2 ascites, 73.1% ID/g in A2780cp20 tumors); normal tissues showed lower and comparable uptake for 212Pb-376.96 and 212Pb-F3-C25. Tumor-bearing mice treated with 212Pb-376.96 alone or combined with carboplatin survived 2-3 times longer than mice treated with 212Pb-F3-C25 or non-treated controls. CONCLUSION: These results support additional RIT studies with 212Pb-376.96 for future evaluation in patients with ovarian cancer.


Subject(s)
B7 Antigens/immunology , Epitopes/immunology , Lead Radioisotopes/therapeutic use , Ovarian Neoplasms/radiotherapy , Radioimmunotherapy/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cell Differentiation , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic , Female , Humans , Mice , Ovarian Neoplasms/pathology
13.
Oncotarget ; 7(52): 86803-86815, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27888804

ABSTRACT

Epithelial ovarian cancer (EOC) is the leading cause of gynecologic cancer mortality worldwide. Platinum-based therapy is the standard first line treatment and while most patients initially respond, resistance to chemotherapy usually arises. Major signaling pathways frequently upregulated in chemoresistant cells and important in the maintenance of cancer stem cells (CSCs) include Wnt/ß-catenin, mTOR, and STAT3. The major objective of our study was to investigate the treatment of ovarian cancer with targeted agents that inhibit these three pathways. Here we demonstrate that niclosamide, a salicylamide derivative, and two synthetically manufactured niclosamide analogs (analog 11 and 32) caused significant inhibition of proliferation of two chemoresistant ovarian cancer cell lines (A2780cp20 and SKOV3Trip2), tumorspheres isolated from the ascites of EOC patients, and cells from a chemoresistant patient-derived xenograft (PDX). This work shows that all three agents significantly decreased the expression of proteins in the Wnt/ß-catenin, mTOR and STAT3 pathways and preferentially targeted cells that expressed the ovarian CSC surface protein CD133. It also illustrates the potential of drug repurposing for chemoresistant EOC and can serve as a basis for pathway-oriented in vivo studies.


Subject(s)
Niclosamide/pharmacology , Ovarian Neoplasms/metabolism , Proteins/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Niclosamide/analogs & derivatives , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
14.
Oncotarget ; 5(18): 8750-64, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25209969

ABSTRACT

A cornerstone of preclinical cancer research has been the use of clonal cell lines. However, this resource has underperformed in its ability to effectively identify novel therapeutics and evaluate the heterogeneity in a patient's tumor. The patient-derived xenograft (PDX) model retains the heterogeneity of patient tumors, allowing a means to not only examine efficacy of a therapy, but also basic tenets of cancer biology in response to treatment. Herein we describe the development and characterization of an ovarian-PDX model in order to study the development of chemoresistance. We demonstrate that PDX tumors are not simply composed of tumor-initiating cells, but recapitulate the original tumor's heterogeneity, oncogene expression profiles, and clinical response to chemotherapy. Combined carboplatin/paclitaxel treatment of PDX tumors enriches the cancer stem cell populations, but persistent tumors are not entirely composed of these populations. RNA-Seq analysis of six pair of treated PDX tumors compared to untreated tumors demonstrates a consistently contrasting genetic profile after therapy, suggesting similar, but few, pathways are mediating chemoresistance. Pathways and genes identified by this methodology represent novel approaches to targeting the chemoresistant population in ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Mice, SCID , Middle Aged , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Patient Selection , Polymerase Chain Reaction , Precision Medicine , Predictive Value of Tests , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Oncotarget ; 5(16): 7065-80, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-25216523

ABSTRACT

The goal of this study was to determine whether combined targeted therapies, specifically those against the Notch, hedgehog and ubiquitin-proteasome pathways, could overcome ovarian cancer chemoresistance. Chemoresistant ovarian cancer cells were exposed to gamma-secretase inhibitors (GSI-I, Compound E) or the proteasome inhibitor bortezomib, alone and in combination with the hedgehog antagonist, LDE225. Bortezomib, alone and in combination with LDE225, was evaluated for effects on paclitaxel efficacy. Cell viability and cell cycle analysis were assessed by MTT assay and propidium iodide staining, respectively. Proteasome activity and gene expression were determined by luminescence assay and qPCR, respectively. Studies demonstrated that GSI-I, but not Compound E, inhibited proteasome activity, similar to bortezomib. Proteasome inhibition decreased hedgehog target genes (PTCH1, GLI1 and GLI2) and increased LDE225 sensitivity in vitro. Bortezomib, alone and in combination with LDE225, increased paclitaxel sensitivity through apoptosis and G2/M arrest. Expression of the multi-drug resistance gene ABCB1/MDR1 was decreased and acetylation of α-tubulin, a marker of microtubule stabilization, was increased following bortezomib treatment. HDAC6 inhibitor tubastatin-a demonstrated that microtubule effects are associated with hedgehog inhibition and sensitization to paclitaxel and LDE225. These results suggest that proteasome inhibition, through alteration of microtubule dynamics and hedgehog signaling, can reverse taxane-mediated chemoresistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biphenyl Compounds/pharmacology , Boronic Acids/pharmacology , Bridged-Ring Compounds/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Proteasome Inhibitors/pharmacology , Pyrazines/pharmacology , Pyridines/pharmacology , Taxoids/pharmacology , Biphenyl Compounds/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Female , Hedgehog Proteins/metabolism , Humans , Ovarian Neoplasms/metabolism , Pyrazines/administration & dosage , Pyridines/administration & dosage , Signal Transduction
16.
Gynecol Oncol ; 134(2): 331-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24844596

ABSTRACT

OBJECTIVE: BRCA-positive ovarian cancer patients derive benefit PARP inhibitors. Approximately 50% of ovarian cancer tumors have homologous recombination (HR) deficiencies and are therefore "BRCA-like," possibly rendering them sensitive to PARP inhibition. However, no predictive assay exists to identify these patients. We sought to determine if irradiation-induced Rad51 foci formation, a known marker of HR, correlated to PARP inhibitor response in an ovarian cancer model. METHODS: Ovarian cancer cell lines were exposed to PARP-inhibitor ABT-888 to determine effect on growth. Rad51 protein expression prior to irradiation was determined via Western blot. Cultured cells and patient-derived xenograft tumors (PDX) were irradiated and probed for Rad51 foci. In vivo PDX tumors were treated with ABT-888 and carboplatin; these results were correlated with the ex vivo ionizing radiation assay. RESULTS: Three of seven cell lines were sensitive to ABT-888. Sensitive lines had the lowest Rad51 foci formation rate after irradiation, indicating functional HR deficiency. Approximately 50% of the PDX samples had decreased Rad51 foci formation. Total Rad51 protein levels were consistently low, suggesting that DNA damage induction is required to characterize HR status. The ex vivo IR assay accurately predicted which PDX models were sensitive to PARP inhibition in vitro and in vivo. ABT-888 alone reduced orthotopic tumor growth by 51% in A2780ip2 cell line, predicted to respond by the ex vivo assay. Three PDX models' response also correlated with the assay. CONCLUSIONS: The ex vivo IR assay correlates with response to PARP inhibition. Analysis of total Rad51 protein is not a reliable substitute.


Subject(s)
Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/radiation effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carboplatin/pharmacology , Carboplatin/therapeutic use , Female , Humans , Tumor Cells, Cultured
17.
Clin Cancer Res ; 19(1): 170-82, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23147994

ABSTRACT

PURPOSE: Endoglin (CD105) is a membranous protein overexpressed in tumor-associated endothelial cells, chemoresistant populations of ovarian cancer cells, and potentially stem cells. Our objective was to evaluate the effects and mechanisms of targeting endoglin in ovarian cancer. EXPERIMENTAL DESIGN: Global and membranous endoglin expression was evaluated in multiple ovarian cancer lines. In vitro, the effects of siRNA-mediated endoglin knockdown with and without chemotherapy were evaluated by MTT assay, cell-cycle analysis, alkaline comet assay, γ-H2AX foci formation, and quantitative PCR. In an orthotopic mouse model, endoglin was targeted with chitosan-encapsulated siRNA with and without carboplatin. RESULTS: Endoglin expression was surprisingly predominantly cytoplasmic, with a small population of surface-positive cells. Endoglin inhibition decreased cell viability, increased apoptosis, induced double-stranded DNA damage, and increased cisplatin sensitivity. Targeting endoglin downregulates expression of numerous DNA repair genes, including BARD1, H2AFX, NBN, NTHL1, and SIRT1. BARD1 was also associated with platinum resistance, and was induced by platinum exposure. In vivo, antiendoglin treatment decreased tumor weight in both ES2 and HeyA8MDR models when compared with control (35%-41% reduction, P < 0.05). Endoglin inhibition with carboplatin was associated with even greater inhibitory effect when compared with control (58%-62% reduction, P < 0.001). CONCLUSIONS: Endoglin downregulation promotes apoptosis, induces significant DNA damage through modulation of numerous DNA repair genes, and improves platinum sensitivity both in vivo and in vitro. Antiendoglin therapy would allow dual treatment of both tumor angiogenesis and a subset of aggressive tumor cells expressing endoglin and is being actively pursued as therapy in ovarian cancer.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Platinum/pharmacology , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/genetics , Antineoplastic Agents/administration & dosage , Carcinoma, Ovarian Epithelial , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Repair , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Endoglin , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Platinum/administration & dosage , RNA Interference , Receptors, Cell Surface/genetics
18.
Mol Cancer Ther ; 11(7): 1587-97, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553355

ABSTRACT

The hedgehog pathway has been implicated in the formation and maintenance of a variety of malignancies, including ovarian cancer; however, it is unknown whether hedgehog signaling is involved in ovarian cancer chemoresistance. The goal of this study was to determine the effects of antagonizing the hedgehog receptor, Smoothened (Smo), on chemotherapy response in ovarian cancer. Expression of hedgehog pathway members was assessed in three pairs of parental and chemotherapy-resistant ovarian cancer cell lines (A2780ip2/A2780cp20, SKOV3ip1/SKOV3TRip2, HeyA8/HeyA8MDR) using quantitative PCR and Western blot analysis. Cell lines were exposed to increasing concentrations of two different Smo antagonists (cyclopamine, LDE225) alone and in combination with carboplatin or paclitaxel. Selective knockdown of Smo, Gli1, or Gli2 was achieved using siRNA constructs. Cell viability was assessed by MTT assay. A2780cp20 and SKOV3TRip2 orthotopic xenografts were treated with vehicle, LDE225, paclitaxel, or combination therapy. Chemoresistant cell lines showed higher expression (>2-fold, P < 0.05) of hedgehog signaling components compared with their respective parental lines. Smo antagonists sensitized chemotherapy-resistant cell lines to paclitaxel, but not to carboplatin. LDE225 treatment also increased sensitivity of ALDH-positive cells to paclitaxel. A2780cp20 and SKOV3TRip2 xenografts treated with combined LDE225 and paclitaxel had significantly less tumor burden than those treated with vehicle or either agent alone. Increased taxane sensitivity seems to be mediated by a decrease in P-glycoprotein (MDR1) expression. Selective knockdown of Smo, Gli1, or Gli2 all increased taxane sensitivity. Smo antagonists reverse taxane resistance in chemoresistant ovarian cancer models, suggesting combined anti-hedgehog and chemotherapies could provide a useful therapeutic strategy for ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Pyridines/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Smoothened Receptor , Xenograft Model Antitumor Assays
19.
Clin Cancer Res ; 18(3): 869-81, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22142828

ABSTRACT

PURPOSE: Within heterogeneous tumors, subpopulations often labeled cancer stem cells (CSC) have been identified that have enhanced tumorigenicity and chemoresistance in ex vivo models. However, whether these populations are more capable of surviving chemotherapy in de novo tumors is unknown. EXPERIMENTAL DESIGN: We examined 45 matched primary/recurrent tumor pairs of high-grade ovarian adenocarcinomas for expression of CSC markers ALDH1A1, CD44, and CD133 using immunohistochemistry. Tumors collected immediately after completion of primary therapy were then laser capture microdissected and subjected to a quantitative PCR array examining stem cell biology pathways (Hedgehog, Notch, TGF-ß, and Wnt). Select genes of interest were validated as important targets using siRNA-mediated downregulation. RESULTS: Primary samples were composed of low densities of ALDH1A1, CD44, and CD133. Tumors collected immediately after primary therapy were more densely composed of each marker, whereas samples collected at first recurrence, before initiating secondary therapy, were composed of similar percentages of each marker as their primary tumor. In tumors collected from recurrent platinum-resistant patients, only CD133 was significantly increased. Of stem cell pathway members examined, 14% were significantly overexpressed in recurrent compared with matched primary tumors. Knockdown of genes of interest, including endoglin/CD105 and the hedgehog mediators Gli1 and Gli2, led to decreased ovarian cancer cell viability, with Gli2 showing a novel contribution to cisplatin resistance. CONCLUSIONS: These data indicate that ovarian tumors are enriched with CSCs and stem cell pathway mediators, especially at the completion of primary therapy. This suggests that stem cell subpopulations contribute to tumor chemoresistance and ultimately recurrent disease.


Subject(s)
Adenocarcinoma/metabolism , Drug Resistance, Neoplasm/physiology , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction/physiology , AC133 Antigen , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aldehyde Dehydrogenase/analysis , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase 1 Family , Antigens, CD/analysis , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antineoplastic Agents/therapeutic use , Blotting, Western , Endoglin , Female , Gene Expression Profiling , Glycoproteins/analysis , Glycoproteins/biosynthesis , Humans , Hyaluronan Receptors/analysis , Hyaluronan Receptors/biosynthesis , Immunohistochemistry , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Laser Capture Microdissection , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peptides/analysis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Retinal Dehydrogenase , Reverse Transcriptase Polymerase Chain Reaction , Zinc Finger Protein Gli2
20.
Clin Cancer Res ; 17(17): 5674-85, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21753153

ABSTRACT

PURPOSE: Jagged1, a Notch ligand, is expressed on both tumor epithelial and endothelial cells and therefore may be amenable to dual targeting of the tumor stroma and malignant cell compartments of the tumor microenvironment. EXPERIMENTAL DESIGN: We describe in vitro effects of targeting of Jagged1 on ovarian cancer cells and in vivo effects of independent targeting of stromal and malignant cell Jagged1 using species-specific human or murine siRNA constructs incorporated into chitosan nanoparticles and delivered intravenously in an orthotopic mouse model. RESULTS: Jagged1 expression was prominent in SKOV3ip1 and IGROV-AF1, and significantly overexpressed in SKOV3TRip2, a taxane-resistant SKOV3 subclone. Jagged1 silencing with siRNA decreased cell viability and reversed taxane chemoresistance. In two different orthotopic ovarian cancer models, treatment with anti-human Jagged1 siRNA-CH reduced growth by 54.4% to 58.3% and with anti-murine Jagged1 siRNA-CH reduced growth by 41.7% to 48.8%. The combination of both species-specific constructs reduced tumor weight by 87.5% to 93.1% and sensitized SKOV3TRip2 tumors to docetaxel in vivo. Tumors showed reduced microvessel density with anti-murine Jagged1 constructs and decreased proliferation with anti-human Jagged1 siRNAs-CH. In addition, we show that Jagged1 downregulation does not sensitize cells to taxanes through a reduction in MDR1 expression, but at least in part by cross-talk with the GLI2 mediator of the Hedgehog pathway. CONCLUSIONS: Jagged1 plays dual roles in cancer progression through an angiogenic function in tumor endothelial cells and through proliferation and chemoresistance in tumor cells. Dual inhibition represents an attractive therapeutic strategy for ovarian and potentially other malignancies.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Receptors, Notch/metabolism , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bridged-Ring Compounds/pharmacology , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Docetaxel , Drug Resistance, Neoplasm , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA Interference , RNA, Small Interfering , Receptors, Notch/genetics , Serrate-Jagged Proteins , Stromal Cells , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
SELECTION OF CITATIONS
SEARCH DETAIL
...