19 results on '"YK-4-279"'
Search Results
2. The ETS Inhibitor YK-4-279 Suppresses Thyroid Cancer Progression Independent of TERT Promoter Mutations
- Author
-
Junyu Xue, Shiyong Li, Peijie Shi, Mengke Chen, Shuang Yu, Shubin Hong, Yanbing Li, Rengyun Liu, and Haipeng Xiao
- Subjects
YK-4-279 ,telomerase reverse transcriptase ,thyroid cancer ,apoptosis ,E-twenty-six transcription factor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Hotspot mutations in the core promoter region of the telomerase reverse transcriptase (TERT) gene have been well established to associate with aggressive clinical characteristics, radioiodine refractory, tumor recurrence, and mortality in thyroid cancer. Several E-twenty-six (ETS) transcription factors were reported to selectively bound to the mutant TERT promoter and activated TERT expression. In this study we aimed to investigate whether TERT promoter mutations confer sensitivity to ETS inhibitor YK-4-279 in thyroid cancer cells and whether this inhibitor could be served as a potential therapeutic agent for thyroid cancer. In vitro assays showed that YK-4-279 treatment sharply suppressed cell viability, colony formation, migration, and invasion, as well as induced cell cycle arrest and apoptosis in a panel of thyroid cancer cells. The cell viability after YK-4-279 treatment was similar between cell lines harboring mutant and wild-type TERT promoters. Furthermore, YK-4-279 treatment reduced both luciferase activity and mRNA expression of TERT independent of TERT promoter mutation status. Data from RNA-seq further revealed that YK-4-279 significantly affected biological processes including DNA replication and cell cycle. Reduced DNA helicase activity and decreased expression of several helicase genes were observed after YK-4-279 treatment. Moreover, YK-4-279 significantly inhibited tumor growth and induced apoptosis in a xenograft mice model. Thus, ETS inhibitor YK-4-279 suppressed TERT expression and conferred anti-tumor activity in a TERT promoter mutation-independent manner, and it could be a potential agent for the treatment of advanced thyroid cancers.
- Published
- 2021
- Full Text
- View/download PDF
3. The ETS Inhibitor YK-4-279 Suppresses Thyroid Cancer Progression Independent of TERT Promoter Mutations.
- Author
-
Xue, Junyu, Li, Shiyong, Shi, Peijie, Chen, Mengke, Yu, Shuang, Hong, Shubin, Li, Yanbing, Liu, Rengyun, and Xiao, Haipeng
- Subjects
THYROID cancer ,IODINE isotopes ,TELOMERASE reverse transcriptase ,CANCER invasiveness ,DNA helicases ,DNA replication ,DISEASE relapse - Abstract
Hotspot mutations in the core promoter region of the telomerase reverse transcriptase (TERT) gene have been well established to associate with aggressive clinical characteristics, radioiodine refractory, tumor recurrence, and mortality in thyroid cancer. Several E-twenty-six (ETS) transcription factors were reported to selectively bound to the mutant TERT promoter and activated TERT expression. In this study we aimed to investigate whether TERT promoter mutations confer sensitivity to ETS inhibitor YK-4-279 in thyroid cancer cells and whether this inhibitor could be served as a potential therapeutic agent for thyroid cancer. In vitro assays showed that YK-4-279 treatment sharply suppressed cell viability, colony formation, migration, and invasion, as well as induced cell cycle arrest and apoptosis in a panel of thyroid cancer cells. The cell viability after YK-4-279 treatment was similar between cell lines harboring mutant and wild-type TERT promoters. Furthermore, YK-4-279 treatment reduced both luciferase activity and mRNA expression of TERT independent of TERT promoter mutation status. Data from RNA-seq further revealed that YK-4-279 significantly affected biological processes including DNA replication and cell cycle. Reduced DNA helicase activity and decreased expression of several helicase genes were observed after YK-4-279 treatment. Moreover, YK-4-279 significantly inhibited tumor growth and induced apoptosis in a xenograft mice model. Thus, ETS inhibitor YK-4-279 suppressed TERT expression and conferred anti-tumor activity in a TERT promoter mutation-independent manner, and it could be a potential agent for the treatment of advanced thyroid cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
4. An inhibitor of endothelial ETS transcription factors promotes physiologic and therapeutic vessel regression.
- Author
-
Schafer, Christopher M., Gurley, Jami M., Kurylowicz, Katarzyna, Lin, Prisca K., Wen Chen, Elliott, Michael H., Davis, George E., Bhatti, Faizah, and Griffin, Courtney T.
- Subjects
- *
TRANSCRIPTION factors , *RETINAL blood vessels , *RETROLENTAL fibroplasia , *APOPTOSIS , *DIABETIC retinopathy - Abstract
During the progression of ocular diseases such as retinopathy of prematurity and diabetic retinopathy, overgrowth of retinal blood vessels results in the formation of pathological neovascular tufts that impair vision. Current therapeutic options for treating these diseases include antiangiogenic strategies that can lead to the undesirable inhibition of normal vascular development. Therefore, strategies that eliminate pathological neovascular tufts while sparing normal blood vessels are needed. In this study we exploited the hyaloid vascular network inmurine eyes,which naturally undergoes regression after birth, to gain mechanistic insights that could be therapeutically adapted for driving neovessel regression in ocular diseases. We found that endothelial cells of regressing hyaloid vessels underwent down-regulation of two structurally related E-26 transformation-specific (ETS) transcription factors, ETS-related gene (ERG) and Friend leukemia integration 1 (FLI1), prior to apoptosis. Moreover, the small molecule YK-4-279, which inhibits the transcriptional and biological activity of ETS factors, enhanced hyaloid regression in vivo and drove Human Umbilical Vein Endothelial Cells (HUVEC) tube regression and apoptosis in vitro. Importantly, exposure of HUVECs to sheer stress inhibited YK-4-279–induced apoptosis, indicating that low-flow vessels may be uniquely susceptible to YK-4-279–mediated regression.We tested this hypothesis by administering YK-4-279 to mice in an oxygen-induced retinopathy model that generates disorganized and poorly perfused neovascular tufts that mimic human ocular diseases. YK-4-279 treatment significantly reduced neovascular tufts while sparing healthy retinal vessels, thereby demonstrating the therapeutic potential of this inhibitor. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. The small molecule inhibitor YK-4-279 disrupts mitotic progression of neuroblastoma cells, overcomes drug resistance and synergizes with inhibitors of mitosis.
- Author
-
Kollareddy, Madhu, Sherrard, Alice, Park, Ji Hyun, Szemes, Marianna, Gallacher, Kelli, Melegh, Zsombor, Oltean, Sebastian, Michaelis, Martin, Jr.Cinatl, Jindrich, Kaidi, Abderrahmane, Malik, Karim, and Cinatl, Jindrich Jr
- Subjects
- *
NEUROBLASTOMA , *MITOSIS , *APOPTOSIS , *RAS oncogenes , *ANAPLASTIC lymphoma kinase , *DRUG resistance in cancer cells , *CANCER chemotherapy , *THERAPEUTICS , *PROTEIN metabolism , *ANTINEOPLASTIC agents , *AZEPINES , *CELL cycle , *CELL lines , *CELL physiology , *CELLULAR signal transduction , *CHROMOSOMES , *CYTOPLASM , *DRUG resistance , *DRUG synergism , *DOSE-effect relationship in pharmacology , *GENES , *GENETIC techniques , *HETEROCYCLIC compounds , *PACLITAXEL , *PROTEINS , *RESEARCH funding , *TIME , *TRANSFERASES , *VINCRISTINE , *INDOLE compounds , *PROTEIN kinase inhibitors , *PHARMACODYNAMICS - Abstract
Neuroblastoma is a biologically and clinically heterogeneous pediatric malignancy that includes a high-risk subset for which new therapeutic agents are urgently required. As well as MYCN amplification, activating point mutations of ALK and NRAS are associated with high-risk and relapsing neuroblastoma. As both ALK and RAS signal through the MEK/ERK pathway, we sought to evaluate two previously reported inhibitors of ETS-related transcription factors, which are transcriptional mediators of the Ras-MEK/ERK pathway in other cancers. Here we show that YK-4-279 suppressed growth and triggered apoptosis in nine neuroblastoma cell lines, while BRD32048, another ETV1 inhibitor, was ineffective. These results suggest that YK-4-279 acts independently of ETS-related transcription factors. Further analysis reveals that YK-4-279 induces mitotic arrest in prometaphase, resulting in subsequent cell death. Mechanistically, we show that YK-4-279 inhibits the formation of kinetochore microtubules, with treated cells showing a broad range of abnormalities including multipolar, fragmented and unseparated spindles, together leading to disrupted progression through mitosis. Notably, YK-4-279 does not affect microtubule acetylation, unlike the conventional mitotic poisons paclitaxel and vincristine. Consistent with this, we demonstrate that YK-4-279 overcomes vincristine-induced resistance in two neuroblastoma cell-line models. Furthermore, combinations of YK-4-279 with vincristine, paclitaxel or the Aurora kinase A inhibitor MLN8237/Alisertib show strong synergy, particularly at low doses. Thus, YK-4-279 could potentially be used as a single-agent or in combination therapies for the treatment of high-risk and relapsing neuroblastoma, as well as other cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
6. Synergistic Effect of YK-4-279 and Paclitaxel on A549 Cell Line
- Author
-
Sara Alkhatib, Emine Kandemiş, Gamze Gizir, and Gülay Bulut
- Subjects
lung cancer ,ERG ,EMT ,YK-4-279 ,paclitaxel ,synergistic effect ,General Works - Abstract
Lung cancer, among all cancer types around the world, is listed as the major cause of death with high mortality rate. ERG transcription factor has an important role in the Epithelial Mesenchymal Transition (EMT), which is one of the most important mechanisms in lung cancer progression. It is known that a small molecule inhibitor YK-4-279 is a potential inhibitor of ERG expression in prostate cancer. In this study, the possible synergistic effect of YK-4-279 and another anticancer drug, Paclitaxel is analysed in non-small cell lung cancer (NSCLC) cell line, A549.
- Published
- 2018
- Full Text
- View/download PDF
7. TK216 targets microtubules in Ewing sarcoma cells.
- Author
-
Povedano, Juan Manuel, Li, Vicky, Lake, Katherine E., Bai, Xin, Rallabandi, Rameshu, Kim, Jiwoong, Xie, Yang, De Brabander, Jef K., and McFadden, David G.
- Subjects
- *
EWING'S sarcoma , *MICROTUBULES , *TUBULINS , *CHROMOSOMAL proteins , *BINDING site assay , *CHROMOSOMAL translocation , *CHIMERIC proteins - Abstract
Ewing sarcoma (EWS) is a pediatric malignancy driven by the EWSR1-FLI1 fusion protein formed by the chromosomal translocation t(11; 22). The small molecule TK216 was developed as a first-in-class direct EWSR1-FLI1 inhibitor and is in phase II clinical trials in combination with vincristine for patients with EWS. However, TK216 exhibits anti-cancer activity against cancer cell lines and xenografts that do not express EWSR1-FLI1, and the mechanism underlying cytotoxicity remains unresolved. We apply a forward-genetics screening platform utilizing engineered hypermutation in EWS cell lines and identify recurrent mutations in TUBA1B , encoding ⍺-tubulin, that prove sufficient to drive resistance to TK216. Using reconstituted microtubule (MT) polymerization in vitro and cell-based chemical probe competition assays, we demonstrate that TK216 acts as an MT destabilizing agent. This work defines the mechanism of cytotoxicity of TK216, explains the synergy observed with vincristine, and calls for a reexamination of ongoing clinical trials with TK216. [Display omitted] • Recurrent TUBA1B mutations were identified in TK216-resistant Ewing sarcoma cells • TUBA1B mutations independently drove resistance to TK216 • TK216 suppressed tubulin polymerization in vitro in a stereo-selective manner • TK216 and vincristine act on microtubules through distinct binding mechanisms TK216 (ONCT-216) was developed to inhibit the fusion protein that drives Ewing sarcoma, EWSR1-FLI1. However, the mechanism underlying TK216-induced cytotoxicity remained elusive. Here, Povedano et al. demonstrate using forward genetics, biochemical reconstitution, and chemical probe competition that TK216 acts as a microtubule destabilizing agent. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. p53 Loss Mediates Hypersensitivity to ETS Transcription Factor Inhibition Based on PARylation-Mediated Cell Death Induction
- Author
-
Dinhof, Carina, Pirker, Christine, Kroiss, Philipp, Kirchhofer, Dominik, Gabler, Lisa, Gojo, Johannes, Lötsch-Gojo, Daniela, Stojanovic, Mirjana, Timelthaler, Gerald, Ferk, Franziska, Knasmüller, Siegfried, Reisecker, Johannes, Spiegl-Kreinecker, Sabine, Birner, Peter, Preusser, Matthias, and Berger, Walter
- Subjects
p53 ,YK-4-279 ,PARylation ,ETS1 ,parthanatos ,ETS factor inhibitor ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Article - Abstract
The small-molecule E26 transformation-specific (ETS) factor inhibitor YK-4-279 was developed for therapy of ETS/EWS fusion-driven Ewing&rsquo, s sarcoma. Here we aimed to identify molecular factors underlying YK-4-279 responsiveness in ETS fusion-negative cancers. Cell viability screenings that deletion of P53 induced hypersensitization against YK-4-279 especially in the BRAFV600E-mutated colon cancer model RKO. This effect was comparably minor in the BRAF wild-type HCT116 colon cancer model. Out of all ETS transcription factor family members, especially ETS1 overexpression at mRNA and protein level was induced by deletion of P53 specifically under BRAF-mutated conditions. Exposure to YK-4-279 reverted ETS1 upregulation induced by P53 knock-out in RKO cells. Despite upregulation of p53 by YK-4-279 itself in RKOp53 wild-type cells, YK-4-279-mediated hyperphosphorylation of histone histone H2A.x was distinctly more pronounced in the P53 knock-out background. YK-4-279-induced cell death in RKOp53-knock-out cells involved hyperPARylation of PARP1, translocation of the apoptosis-inducible factor AIF into nuclei, and induction of mitochondrial membrane depolarization, all hallmarks of parthanatos. Accordingly, pharmacological PARP as well as BRAFV600E inhibition showed antagonistic activity with YK-4-279 especially in the P53 knock-out background. Taken together, we identified ETS factor inhibition as a promising strategy for the treatment of notoriously therapy-resistant p53-null solid tumours with activating MAPK mutations.
- Published
- 2020
9. EWS-FLI1 and RNA helicase A interaction inhibitor YK-4-279 inhibits growth of neuroblastoma
- Author
-
Hao Wang, Cristian Coarfa, M. John Hicks, Yang Yu, Wei Huang, Saurabh Agarwal, Yanling Zhao, Sarah E. Woodfield, Roma H. Patel, Hong Zhang, Sanjeev A. Vasudevan, Yesenia Rojas, Jianhua Yang, Zhenghu Chen, Kimal Rajapakshe, Xin Xu, Angela Major, Jed G. Nuchtern, Wenjing Sun, Jason M. Shohet, Jingling Jin, and Yongfeng Wang
- Subjects
0301 basic medicine ,Gerontology ,medicine.medical_treatment ,chemotherapy ,doxorubicin ,neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Neuroblastoma ,medicine ,Cytotoxic T cell ,Doxorubicin ,YK-4-279 ,Chemotherapy ,Cell growth ,business.industry ,medicine.disease ,RNA Helicase A ,030104 developmental biology ,Oncology ,EWSR1 ,Cell culture ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer research ,business ,Research Paper ,medicine.drug - Abstract
// Wenjing Sun 1, 5 , Yesenia Rojas 1 , Hao Wang 2, 6 , Yang Yu 1 , Yongfeng Wang 3 , Zhenghu Chen 3 , Kimal Rajapakshe 4 , Xin Xu 3 , Wei Huang 3 , Saurabh Agarwal 3 , Roma H. Patel 1 , Sarah Woodfield 1 , Yanling Zhao 3 , Jingling Jin 1 , Hong Zhang 2 , Angela Major 7 , M. John Hicks 7 , Jason M. Shohet 3 , Sanjeev A. Vasudevan 1 , Cristian Coarfa 4 , Jianhua Yang 3 and Jed G. Nuchtern 1 1 Pediatric Surgery Division, Michael E. Debakey Department of Surgery, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA 2 Department of Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA 3 Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA 4 Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA 5 Laboratory of Medical Genetics, Harbin Medical University, Harbin, Heilongjiang 150081, China 6 Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China 7 Department of Pathology, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA Correspondence to: Jed G. Nuchtern, email: nuchtern@bcm.edu Keywords: neuroblastoma; YK-4-279; chemotherapy; doxorubicin; EWSR1 Abbreviations: NB, neuroblastoma; RHA, RNA helicase A; Dox, doxorubicin Received: July 12, 2017 Accepted: August 21, 2017 Published: October 19, 2017 ABSTRACT Treatment failure in high risk neuroblastoma (NB) is largely due to the development of chemotherapy resistance. We analyzed the gene expression changes associated with exposure to chemotherapy in six high risk NB tumors with the aid of the Connectivity Map bioinformatics platform. Ten therapeutic agents were predicted to have a high probability of reversing the transcriptome changes associated with neoadjuvant chemotherapy treatment. Among these agents, initial screening showed the EWS-FLI1 and RNA helicase A interaction inhibitor YK-4-279, had obvious cytotoxic effects on NB cell lines. Using a panel of NB cell lines, including MYCN nonamplified (SK-N-AS, SH-SY5Y, and CHLA-255), and MYCN amplified (NB-19, NGP, and IMR-32) cell lines, we found that YK-4-279 had cytotoxic effects on all lines tested. In addition, YK-4-279 also inhibited cell proliferation and anchorage-independent growth and induced cell apoptosis of these cells. YK-4-279 enhanced the cytotoxic effect of doxorubicin (Dox). Moreover, YK-4-279 was able to overcome the established chemoresistance of LA-N-6 NB cells. In an orthotopic xenograft NB mouse model, YK-4-279 inhibited NB tumor growth and induced apoptosis in tumor cells through PARP and Caspase 3 cleavage in vivo . While EWS-FLI1 fusion protein is not frequently found in NB, using the R2 public database of neuroblastoma outcome and gene expression, we found that high expression of EWSR1 was associated with poor patient outcome. Knockdown of EWSR1 inhibited the oncogenic potential of neuroblastoma cell lines. Taken together, our results indicate that YK-4-279 might be a promising agent for treatment of NB that merits further exploration.
- Published
- 2017
- Full Text
- View/download PDF
10. The Effects and Mechanism of YK-4-279 in Combination with Docetaxel on Prostate Cancer
- Author
-
Wu Xiaofeng, Zhiyun Du, Yan He, Wang Huaqian, Min Chen, Lin Yu, Kun Zhang, Xi Zheng, Susan Goodin, Huarong Huang, and Dongli Li
- Subjects
0301 basic medicine ,Oncology ,synergistic action ,Male ,medicine.medical_specialty ,Indoles ,Cell Survival ,Apoptosis ,Docetaxel ,urologic and male genital diseases ,Metastasis ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Cell Movement ,Internal medicine ,Cell Line, Tumor ,Survivin ,LNCaP ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Propidium iodide ,Neoplasm Metastasis ,Cell Proliferation ,YK-4-279 ,combination ,business.industry ,Cell growth ,General Medicine ,medicine.disease ,prostate cancer ,3. Good health ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Taxoids ,business ,medicine.drug ,Research Paper - Abstract
Background: Docetaxel is the first-line treatment for castration-resistant prostate cancer (CRPC). The limited survival benefit associated with the quick emergence of resistance and systemic toxicity diminishes its efficacy in high-dose monotherapy. YK-4-279 is a small molecule inhibitor of ETV1 that plays an important role in the progression of prostate cancer. The aim of this study was to evaluate the hypothesis that the combination of docetaxel and YK-4-279 will have a synergistic effect on inhibiting growth and accelerating apoptosis in human prostate cancer cells. Methods: Cell growth assessed using CCK-8 and trypan blue exclusion assays. Cell apoptosis was determined by morphological assessment in cells stained with propidium iodide. Standard scratch migration and Matrigel-coated transwell invasion assays were used to assess cell migration and invasion, respectively. Western blotting was used to investigate the levels of ETV1, AR, PSA, p-STAT3, survivin, Bcl-2, and p-Akt in prostate cancer cells. Results: The combination of low-dose docetaxel and YK-4-279 synergistically inhibited growth and induced apoptosis in human prostate cancer cells. The combination also more efficiently suppressed the migration and invasion of LNCaP and PC-3 cells. The combination of low-dose docetaxel and YK-4-279 caused a stronger decrease in the levels of ETV1, AR, PSA, p-STAT3, survivin, Bcl-2, and p-Akt in LNCaP cells and of p-Akt, Bcl-2, and p-STAT3 in PC-3 cells compared with either drug alone. Conclusions: These data suggest that the combination of docetaxel and YK-4-279 may be an effective approach for inhibiting the growth and metastasis of prostate cancer. This could permit a decrease in the docetaxel dose necessary for patients with CRPC and thereby lower its systemic toxicity.
- Published
- 2017
11. YK-4-279 effectively antagonizes EWS-FLI1 induced leukemia in a transgenic mouse model
- Author
-
Kristina M. Mueller, Heinrich Kovar, Metin Ozdemirli, Yasemin Saygideğer-Kont, Aykut Üren, Idil Temel, Tsion Z. Minas, Jeffrey A. Toretsky, Lukas Kenner, Tahereh Javaheri, Jenny Han, Haydar Çelik, Richard Moriggl, Michaela Schlederer, Sung-Hyeok Hong, and Hayriye V. Erkizan
- Subjects
Genetically modified mouse ,Pathology ,medicine.medical_specialty ,Chromatin Immunoprecipitation ,Indoles ,Oncogene Proteins, Fusion ,Blotting, Western ,Spleen ,Mice, Transgenic ,Real-Time Polymerase Chain Reaction ,Immunoenzyme Techniques ,Mice ,White blood cell ,medicine ,Animals ,Humans ,RNA, Messenger ,EWS-FLI1 ,ETS fusion proteins ,YK-4-279 ,business.industry ,erythoid leukemia ,Proto-Oncogene Protein c-fli-1 ,Reverse Transcriptase Polymerase Chain Reaction ,Myeloid leukemia ,Surface Plasmon Resonance ,medicine.disease ,Flow Cytometry ,Fusion protein ,Gene Expression Regulation, Neoplastic ,Leukemia ,Disease Models, Animal ,medicine.anatomical_structure ,Oncology ,Bone marrow ,Sarcoma ,Leukemia, Erythroblastic, Acute ,RNA-Binding Protein EWS ,business ,ewing sarcoma ,Research Paper - Abstract
Ewing sarcoma is an aggressive tumor of bone and soft tissue affecting predominantly children and young adults. Tumor-specific chromosomal translocations create EWS-FLI1 and similar aberrant ETS fusion proteins that drive sarcoma development in patients. ETS family fusion proteins and over-expressed ETS proteins are also found in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. Transgenic expression of EWS-FLI1 in mice promotes high penetrance erythroid leukemia with dense hepatic and splenic infiltrations. We identified a small molecule, YK-4-279, that directly binds to EWS-FLI1 and inhibits its oncogenic activity in Ewing sarcoma cell lines and xenograft mouse models. Herein, we tested in vivo therapeutic efficacy and potential side effects of YK-4-279 in the transgenic mouse model with EWS-FLI1 induced leukemia. A two-week course of treatment with YK-4-279 significantly reduced white blood cell count, nucleated erythroblasts in the peripheral blood, splenomegaly, and hepatomegaly of erythroleukemic mice. YK-4-279 inhibited EWS-FLI1 target gene expression in neoplastic cells. Treated animals showed significantly better overall survival compared to control mice that rapidly succumbed to leukemia. YK-4-279 treated mice did not show overt toxicity in liver, spleen, or bone marrow. In conclusion, this in vivo study highlights the efficacy of YK-4-279 to treat EWS-FLI1 expressing neoplasms and support its therapeutic potential for patients with Ewing sarcoma and other ETS-driven malignancies.
- Published
- 2015
12. The small molecule inhibitor YK-4-279 disrupts mitotic progression of neuroblastoma cells, overcomes drug resistance and synergizes with inhibitors of mitosis
- Author
-
Madhu, Kollareddy, Alice, Sherrard, Ji Hyun, Park, Marianna, Szemes, Kelli, Gallacher, Zsombor, Melegh, Sebastian, Oltean, Martin, Michaelis, Jindrich, Cinatl, Abderrahmane, Kaidi, and Karim, Malik
- Subjects
Prometaphase ,Indoles ,Time Factors ,Paclitaxel ,RNase A, ribonuclease A ,ERK, extracellular signal-regulated kinases ,Mitosis ,MTT, 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide ,Apoptosis ,Spindle Apparatus ,Antimitotic Agents ,Transfection ,Inhibitory Concentration 50 ,Neuroblastoma ,MEK, Mitogen-activated protein kinase kinase ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,TERT, Telomerase reverse transcriptase ,Humans ,Chemotherapy ,Kinetochores ,Protein Kinase Inhibitors ,ALK, Anaplastic Lymphoma kinase ,QVD, quinolyl-valyl-O-methylaspartyl-(-2,6-difluorophenoxy)- methyl ketone ,Aurora Kinase A ,GFP, green fluorescent protein ,YK-4-279 ,Drug resistance/synergy ,Dose-Response Relationship, Drug ,Cell Cycle ,SDS-PAGE, sodium-dodecyl sulphate-polyacrylamide gel electrophoresis ,Drug Synergism ,Azepines ,PBS, Phosphate-buffered saline ,Drug Resistance, Multiple ,Pyrimidines ,Drug Resistance, Neoplasm ,Vincristine ,EGF, Epidermal growth factor ,kDa, kilodaltons ,RNA Interference ,Original Article ,Tumor Suppressor Protein p53 ,pHH3, phospho-histone H3 (ser10) ,MAPK, mitogen-activated protein kinase ,Signal Transduction - Abstract
Neuroblastoma is a biologically and clinically heterogeneous pediatric malignancy that includes a high-risk subset for which new therapeutic agents are urgently required. As well as MYCN amplification, activating point mutations of ALK and NRAS are associated with high-risk and relapsing neuroblastoma. As both ALK and RAS signal through the MEK/ERK pathway, we sought to evaluate two previously reported inhibitors of ETS-related transcription factors, which are transcriptional mediators of the Ras-MEK/ERK pathway in other cancers. Here we show that YK-4-279 suppressed growth and triggered apoptosis in nine neuroblastoma cell lines, while BRD32048, another ETV1 inhibitor, was ineffective. These results suggest that YK-4-279 acts independently of ETS-related transcription factors. Further analysis reveals that YK-4-279 induces mitotic arrest in prometaphase, resulting in subsequent cell death. Mechanistically, we show that YK-4-279 inhibits the formation of kinetochore microtubules, with treated cells showing a broad range of abnormalities including multipolar, fragmented and unseparated spindles, together leading to disrupted progression through mitosis. Notably, YK-4-279 does not affect microtubule acetylation, unlike the conventional mitotic poisons paclitaxel and vincristine. Consistent with this, we demonstrate that YK-4-279 overcomes vincristine-induced resistance in two neuroblastoma cell-line models. Furthermore, combinations of YK-4-279 with vincristine, paclitaxel or the Aurora kinase A inhibitor MLN8237/Alisertib show strong synergy, particularly at low doses. Thus, YK-4-279 could potentially be used as a single-agent or in combination therapies for the treatment of high-risk and relapsing neuroblastoma, as well as other cancers., Highlights • The small molecule inhibitor YK-4-279 can trigger p53-independent apoptosis in neuroblastoma cell lines. • YK-4-279 induces mitotic arrest with multipolar, fragmented and unseparated spindles. • YK-4-279 acts differently from paclitaxel and vincristine. • YK-4-279 can overcome resistance to vincristine and synergize with other inhibitors of mitosis.
- Published
- 2017
13. Pharmacokinetic modeling optimizes inhibition of the ‘undruggable’ EWS-FLI1 transcription factor in Ewing Sarcoma
- Author
-
Julie S. Barber-Rotenberg, Sung-Hyeok Hong, Hayriye V. Erkizan, Peter J. Houghton, Jeffrey A. Toretsky, Aykut Üren, Bhaskar Kallakury, Sarah E. Youbi, Phillip Monroe, and S. Peter Hong
- Subjects
Male ,Indoles ,Proto-Oncogene Protein c-fli-1 ,Oncogene Proteins, Fusion ,Transcription, Genetic ,Oncogene Proteins ,Ewing Sarcoma ,pharmacokinetic modeling ,Antineoplastic Agents ,Apoptosis ,Sarcoma, Ewing ,rat xenograft ,Biology ,Rats, Sprague-Dawley ,Mice ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Transcription factor ,EWS-FLI1 ,030304 developmental biology ,YK-4-279 ,0303 health sciences ,medicine.disease ,Xenograft Model Antitumor Assays ,RNA Helicase A ,Fusion protein ,Virology ,Rats ,3. Good health ,Mice, Inbred C57BL ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Sarcoma ,RNA-Binding Protein EWS ,Research Paper - Abstract
// Sung-Hyeok Hong 1,* , Sarah. E. Youbi 1,* , S. Peter Hong 2 , Bhaskar Kallakury 3 , Phillip Monroe 2 , Hayriye V Erkizan 1 , Julie S. Barber-Rotenberg 1 , Peter Houghton 4 , Aykut Uren 1 , Jeffrey A. Toretsky 1 1 Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA 2 Battelle Memorial Institute, Health and Life Sciences, Columbus, OH, USA 3 Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA 4 Nationwide Children’s Research Institute, Center for Childhood Cancer, Columbus, USA * These authors contributed equally Correspondence: Jeffrey A. Toretsky, email: // Keywords : YK-4-279, EWS-FLI1, Ewing Sarcoma, rat xenograft, pharmacokinetic modeling Received : October 11, 2013 Accepted : November 9, 2013 Published : November 11, 2013 Abstract Transcription factors have long been deemed ‘undruggable’ targets for therapeutics. Enhanced recognition of protein biochemistry as well as the need to have more targeted approaches to treat cancer has rendered transcription factors approachable for therapeutic development. Since transcription factors lack enzymatic domains, the specific targeting of these proteins has unique challenges. One challenge is the hydrophobic microenvironment that affects small molecules gaining access to block protein interactions. The most attractive transcription factors to target are those formed from tumor specific chromosomal translocations that are validated oncogenic driver proteins. EWS-FLI1 is a fusion protein that results from the pathognomonic translocation of Ewing sarcoma (ES). Our past work created the small molecule YK-4-279 that blocks EWS-FLI1 from interacting with RNA Helicase A (RHA). To fulfill long-standing promise in the field by creating a clinically useful drug, steps are required to allow for in vivo administration. These investigations identify the need for continuous presence of the small molecule protein-protein inhibitor for a period of days. We describe the pharmacokinetics of YK-4-279 and its individual enantiomers. In vivo studies confirm prior in vitro experiments showing (S)-YK-4-279 as the EWS-FLI1 specific enantiomer demonstrating both induction of apoptosis and reduction of EWS-FLI1 regulated caveolin-1 protein. We have created the first rat xenograft model of ES, treated with (S)-YK-4-279 dosing based upon PK modeling leading to a sustained complete response in 2 of 6 ES tumors. Combining laboratory studies, pharmacokinetic measurements, and modeling has allowed us to create a paradigm that can be optimized for in vivo systems using both in vitro data and pharmacokinetic simulations. Thus, (S)-YK-4-279 as a small molecule drug is ready for continued development towards a first-in-human, first-in-class, clinical trial.
- Published
- 2013
- Full Text
- View/download PDF
14. Single Enantiomer of YK-4-279 Demonstrates Specificity in Targeting the Oncogene EWS-FLI1
- Author
-
Veselin S Dobrev, Lauren J. Scher, Yali Kong, Maksymilian Chruszcz, Christina L. Kobs, Hayriye V. Erkizan, Julie S. Barber-Rotenberg, Jeffrey A. Toretsky, Tara M. Snyder, Steven Summer, Aykut Üren, Philip J. Monroe, Steven J. Metallo, Milton L. Brown, Wladek Minor, Saravana P. Selvanathan, Perrer N. Tosso, S. Peter Hong, and Natalie L. South
- Subjects
Transcriptional Activation ,Proto-Oncogene Protein c-fli-1 ,Indoles ,Oncogene Proteins, Fusion ,Immunoprecipitation ,RHA ,Transplantation, Heterologous ,Sarcoma, Ewing ,Biology ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Cell Line, Tumor ,Animals ,Humans ,Transcription factor ,EWS-FLI1 ,030304 developmental biology ,Cell Proliferation ,YK-4-279 ,0303 health sciences ,Caspase 3 ,Stereoisomerism ,Molecular biology ,Small molecule ,RNA Helicase A ,Fusion protein ,Research Papers ,3. Good health ,Rats ,Oncology ,030220 oncology & carcinogenesis ,Enantiomer ,RNA-Binding Protein EWS ,Transcription Factors - Abstract
Julie S. Barber-Rotenberg 1 , Saravana P. Selvanathan 1 , Yali Kong 1 , Hayriye V. Erkizan 1 , Tara M. Snyder 2 , S. Peter Hong 3 , Christina L. Kobs 3 , Natalie L. South 3 , Steven Summer 3 , Philip J. Monroe 3 , Maksymilian Chruszcz 4 , Veselin Dobrev 5 , Perrer N. Tosso 1 , Lauren J. Scher 1 , Wladek Minor 4 , Milton L. Brown 1 , Steven J. Metallo 5 , Aykut Uren 1 , and Jeffrey A. Toretsky 1 1 Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC USA 2 AMRI, Pharmaceutical and Quality Services, Albany, NY USA 3 Battelle Memorial Institute, Health and Life Sciences, Columbus, OH USA 4 Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA USA 5 Department of Chemistry, Georgetown University, Washington, DC USA Received: February 17, 2012; Accepted: February 26, 2012; Published: February 29, 2012; Keywords: PRL-3 monoclonal antibody, PRL-3 mouse/human chimeric antibody, antibody therapy, intracellular oncoprotein Correspondence: Jeffrey A. Toretsky, // // Abstract Oncogenic fusion proteins, such as EWS-FLI1, are excellent therapeutic targets as they are only located within the tumor. However, there are currently no agents targeted toward transcription factors, which are often considered to be ‘undruggable.’ A considerable body of evidence is accruing that refutes this claim based upon the intrinsic disorder of transcription factors. Our previous studies show that RNA Helicase A (RHA) enhances the oncogenesis of EWS-FLI1, a putative intrinsically disordered protein. Interruption of this protein-protein complex by small molecule inhibitors validates this interaction as a unique therapeutic target. Single enantiomer activity from a chiral compound has been recognized as strong evidence for specificity in a small molecule-protein interaction. Our compound, YK-4-279, has a chiral center and can be separated into two enantiomers by chiral HPLC. We show that there is a significant difference in activity between the two enantiomers. (S)-YK-4-279 is able to disrupt binding between EWS-FLI1 and RHA in an immunoprecipitation assay and blocks the transcriptional activity of EWS-FLI1, while (R)-YK-4-279 cannot. Enantiospecific effects are also established in cytotoxicity assays and caspase assays, where up to a log-fold difference is seen between (S)-YK-4-279 and the racemic YK-4-279. Our findings indicate that only one enantiomer of our small molecule is able to specifically target a protein-protein interaction. This work is significant for its identification of a single enantiomer effect upon a protein interaction suggesting that small molecule targeting of intrinsically disordered proteins can be specific. Furthermore, proving YK-4-279 has only one functional enantiomer will be helpful in moving this compound towards clinical trials.
- Published
- 2012
15. Development of an Ewing sarcoma cell line with resistance to EWS‑FLI1 inhibitor YK‑4‑279.
- Author
-
Conn E, Hour S, Allegakoen D, Graham G, Petro J, Kouassi-Brou M, Hong SH, Selvanathan S, Çelik H, Toretsky J, and Üren A
- Subjects
- 12E7 Antigen genetics, 12E7 Antigen metabolism, Biomarkers, Tumor, Cell Line, Tumor, Dose-Response Relationship, Drug, Gene Expression, Humans, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm genetics, Indoles pharmacology, Oncogene Proteins, Fusion antagonists & inhibitors, Oncogene Proteins, Fusion genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Protein c-fli-1 antagonists & inhibitors, Proto-Oncogene Protein c-fli-1 genetics, RNA-Binding Protein EWS antagonists & inhibitors, RNA-Binding Protein EWS genetics, Sarcoma, Ewing genetics
- Abstract
Despite Ewing sarcoma (ES) being the second most common pediatric malignancy of bone and soft tissue, few novel therapeutic approaches have been introduced over the past few decades. ES contains a pathognomonic chromosomal translocation that leads to a fusion protein between EWSR1 and an ets family member, most often FLI1. EWS‑FLI1 is the most common type of fusion protein and is a well‑vetted therapeutic target. A small molecule inhibitor of EWS‑FLI1, YK‑4‑279 (YK) was developed with the intention to serve as a targeted therapy option for patients with ES. The present study investigated resistance mechanisms by developing an ES cell line specifically resistant to YK. The ES cell line A4573 was treated with YK to create resistant cells by long term continuous exposure. The results revealed that resistance in A4573 was robust and sustainable, with a >27‑fold increase in IC50 lasting up to 16 weeks in the absence of the compound. Resistant ES cells were still sensitive to standard of care drugs, including doxorubicin, vincristine and etoposide, which may be valuable in future combination treatments in the clinic. Resistant ES cells revealed an increased expression of CD99. RNA sequencing and qPCR validation of resistant ES cells confirmed an increased expression of ANO1, BRSK2 and IGSF21, and a reduced expression of COL24A1, PRSS23 and RAB38 genes. A functional association between these genes and mechanism of resistance remains to be investigated. The present study created a cell line to investigate YK resistance.
- Published
- 2020
- Full Text
- View/download PDF
16. Inhibition of ERG Activity in Patient-derived Prostate Cancer Xenografts by YK-4-279.
- Author
-
Winters B, Brown L, Coleman I, Nguyen H, Minas TZ, Kollath L, Vasioukhin V, Nelson P, Corey E, Üren A, and Morrissey C
- Subjects
- Animals, Apoptosis drug effects, Apoptosis genetics, Body Weight drug effects, Body Weight genetics, Cell Proliferation drug effects, Cell Proliferation genetics, Gene Expression drug effects, Gene Expression genetics, Heterografts metabolism, Humans, Male, Mice, Mice, SCID, Prostate-Specific Antigen metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Receptors, Mineralocorticoid genetics, Transcriptional Activation drug effects, Transcriptional Activation genetics, Transcriptional Regulator ERG metabolism, Tumor Burden drug effects, Tumor Burden genetics, Up-Regulation drug effects, Up-Regulation genetics, Heterografts drug effects, Indoles pharmacology, Prostatic Neoplasms drug therapy, Transcriptional Regulator ERG antagonists & inhibitors, Transcriptional Regulator ERG genetics
- Abstract
Background/aim: The aim of the current study was to determine the effects of the ERG small-molecule inhibitor YK-4-279 on ERG
+ prostate cancer patient-derived xenografts (PDX)., Materials and Methods: ERG activity was blocked using YK-4-279 in three subcutaneously-implanted ERG+ (LuCaP 23.1, 86.2 and 35) and one ERG- (LuCaP 96) PDX. Treated animals tumor volume (TV), body weight (BW) and serum prostate-specific antigen (PSA) were compared to vehicle-treated control animals. Gene expression, proliferation, apoptosis, microvessel density and ERG expression were also assessed., Results: Administration of YK-4-279 decreased TV (p=0.026), proliferation (p=0.0038) and PSA (p=0.022) in Severe Combined Immunodeficiency (SCID) mice bearing LuCaP 23.1 tumors. LuCaP 86.2, LuCaP 35 and LuCaP 96 showed no significant changes in TV, or PSA. Mineralocorticoid receptor (MR) and MR-direct target genes were up-regulated in treatment-resistant LuCaP 86.2 and LuCaP 35 PDX., Conclusion: YK-4-279 decreased ERG+ LuCaP 23.1 tumor growth, but not LuCaP 86.2 and LuCaP 35 ERG+ tumor growth., (Copyright© 2017, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
17. The Effects and Mechanism of YK-4-279 in Combination with Docetaxel on Prostate Cancer.
- Author
-
Yu L, Wu X, Chen M, Huang H, He Y, Wang H, Li D, Du Z, Zhang K, Goodin S, and Zheng X
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Docetaxel, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Neoplasm Metastasis, Neoplasm Proteins genetics, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Indoles administration & dosage, Prostatic Neoplasms, Castration-Resistant drug therapy, Taxoids administration & dosage
- Abstract
Background : Docetaxel is the first-line treatment for castration-resistant prostate cancer (CRPC). The limited survival benefit associated with the quick emergence of resistance and systemic toxicity diminishes its efficacy in high-dose monotherapy. YK-4-279 is a small molecule inhibitor of ETV1 that plays an important role in the progression of prostate cancer. The aim of this study was to evaluate the hypothesis that the combination of docetaxel and YK-4-279 will have a synergistic effect on inhibiting growth and accelerating apoptosis in human prostate cancer cells. Methods : Cell growth assessed using CCK-8 and trypan blue exclusion assays. Cell apoptosis was determined by morphological assessment in cells stained with propidium iodide. Standard scratch migration and Matrigel-coated transwell invasion assays were used to assess cell migration and invasion, respectively. Western blotting was used to investigate the levels of ETV1, AR, PSA, p-STAT3, survivin, Bcl-2, and p-Akt in prostate cancer cells. Results : The combination of low-dose docetaxel and YK-4-279 synergistically inhibited growth and induced apoptosis in human prostate cancer cells. The combination also more efficiently suppressed the migration and invasion of LNCaP and PC-3 cells. The combination of low-dose docetaxel and YK-4-279 caused a stronger decrease in the levels of ETV1, AR, PSA, p-STAT3, survivin, Bcl-2, and p-Akt in LNCaP cells and of p-Akt, Bcl-2, and p-STAT3 in PC-3 cells compared with either drug alone. Conclusions : These data suggest that the combination of docetaxel and YK-4-279 may be an effective approach for inhibiting the growth and metastasis of prostate cancer. This could permit a decrease in the docetaxel dose necessary for patients with CRPC and thereby lower its systemic toxicity., Competing Interests: Competing Interests: The authors have declared that no competing interest exists.
- Published
- 2017
- Full Text
- View/download PDF
18. YK-4-279 effectively antagonizes EWS-FLI1 induced leukemia in a transgenic mouse model.
- Author
-
Minas TZ, Han J, Javaheri T, Hong SH, Schlederer M, Saygideğer-Kont Y, Çelik H, Mueller KM, Temel I, Özdemirli M, Kovar H, Erkizan HV, Toretsky J, Kenner L, Moriggl R, and Üren A
- Subjects
- Animals, Blotting, Western, Chromatin Immunoprecipitation, Flow Cytometry, Humans, Immunoenzyme Techniques, Leukemia, Erythroblastic, Acute pathology, Mice, Mice, Transgenic, Oncogene Proteins, Fusion administration & dosage, Proto-Oncogene Protein c-fli-1 administration & dosage, RNA, Messenger genetics, RNA-Binding Protein EWS administration & dosage, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Surface Plasmon Resonance, Disease Models, Animal, Gene Expression Regulation, Neoplastic drug effects, Indoles pharmacology, Leukemia, Erythroblastic, Acute drug therapy, Leukemia, Erythroblastic, Acute etiology, Oncogene Proteins, Fusion antagonists & inhibitors, Oncogene Proteins, Fusion toxicity, Proto-Oncogene Protein c-fli-1 antagonists & inhibitors, Proto-Oncogene Protein c-fli-1 toxicity, RNA-Binding Protein EWS antagonists & inhibitors, RNA-Binding Protein EWS toxicity
- Abstract
Ewing sarcoma is an aggressive tumor of bone and soft tissue affecting predominantly children and young adults. Tumor-specific chromosomal translocations create EWS-FLI1 and similar aberrant ETS fusion proteins that drive sarcoma development in patients. ETS family fusion proteins and over-expressed ETS proteins are also found in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. Transgenic expression of EWS-FLI1 in mice promotes high penetrance erythroid leukemia with dense hepatic and splenic infiltrations. We identified a small molecule, YK-4-279, that directly binds to EWS-FLI1 and inhibits its oncogenic activity in Ewing sarcoma cell lines and xenograft mouse models. Herein, we tested in vivo therapeutic efficacy and potential side effects of YK-4-279 in the transgenic mouse model with EWS-FLI1 induced leukemia. A two-week course of treatment with YK-4-279 significantly reduced white blood cell count, nucleated erythroblasts in the peripheral blood, splenomegaly, and hepatomegaly of erythroleukemic mice. YK-4-279 inhibited EWS-FLI1 target gene expression in neoplastic cells. Treated animals showed significantly better overall survival compared to control mice that rapidly succumbed to leukemia. YK-4-279 treated mice did not show overt toxicity in liver, spleen, or bone marrow. In conclusion, this in vivo study highlights the efficacy of YK-4-279 to treat EWS-FLI1 expressing neoplasms and support its therapeutic potential for patients with Ewing sarcoma and other ETS-driven malignancies.
- Published
- 2015
- Full Text
- View/download PDF
19. Blocking the road, stopping the engine or killing the driver? Advances in targeting EWS/FLI-1 fusion in Ewing sarcoma as novel therapy.
- Author
-
Kovar H
- Subjects
- Animals, Antineoplastic Agents pharmacology, Bone Neoplasms genetics, Bone Neoplasms pathology, Chromatin Assembly and Disassembly physiology, DNA Repair physiology, Drug Design, Epigenesis, Genetic, Humans, Molecular Targeted Therapy, Sarcoma, Ewing genetics, Sarcoma, Ewing pathology, Bone Neoplasms drug therapy, Oncogene Proteins, Fusion metabolism, Proto-Oncogene Protein c-fli-1 metabolism, RNA-Binding Protein EWS metabolism, Sarcoma, Ewing drug therapy
- Abstract
Introduction: Ewing sarcoma (ES) represents the paradigm of an aberrant E-twenty-six (ETS) oncogene-driven cancer. It is characterized by specific rearrangements of one of five alternative ETS family member genes with EWSR1. There is experimental evidence that the resulting fusion proteins act as aberrant transcription factors driving ES pathogenesis. The transcriptional gene regulatory network driven by EWS-ETS proteins provides the oncogenic engine to the tumor. Therefore, EWS-ETS and their downstream machinery are considered ideal tumor-specific therapeutic targets., Areas Covered: This review critically discusses the literature on the development of EWS-ETS-directed ES targeting strategies considering current knowledge of EWS-ETS biology and cellular context. It focuses on determinants of EWS-FLI1 function with an emphasis on interactions with chromatin structure. We speculate about the relevance of poorly investigated aspects in ES research such as chromatin remodeling and DNA damage repair for the development of targeted therapies., Expert Opinion: This review questions the specificity of signature-based screening approaches to the identification of EWS-FLI1-targeted compounds. It challenges the view that targeting the downstream gene regulatory network carries potential for therapeutic breakthroughs because of resistance-inducing network rewiring. Instead, we propose to combine targeting of the fusion protein with epigenetic therapy as a future treatment strategy in ES.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.