Try a new search

Format these results:

Searched for:

person:chent15

in-biosketch:yes

Total Results:

65


Small-molecule targeting of brachyury transcription factor addiction in chordoma

Sharifnia, Tanaz; Wawer, Mathias J; Chen, Ting; Huang, Qing-Yuan; Weir, Barbara A; Sizemore, Ann; Lawlor, Matthew A; Goodale, Amy; Cowley, Glenn S; Vazquez, Francisca; Ott, Christopher J; Francis, Joshua M; Sassi, Slim; Cogswell, Patricia; Sheppard, Hadley E; Zhang, Tinghu; Gray, Nathanael S; Clarke, Paul A; Blagg, Julian; Workman, Paul; Sommer, Josh; Hornicek, Francis; Root, David E; Hahn, William C; Bradner, James E; Wong, Kwok K; Clemons, Paul A; Lin, Charles Y; Kotz, Joanne D; Schreiber, Stuart L
Chordoma is a primary bone cancer with no approved therapy1. The identification of therapeutic targets in this disease has been challenging due to the infrequent occurrence of clinically actionable somatic mutations in chordoma tumors2,3. Here we describe the discovery of therapeutically targetable chordoma dependencies via genome-scale CRISPR-Cas9 screening and focused small-molecule sensitivity profiling. These systematic approaches reveal that the developmental transcription factor T (brachyury; TBXT) is the top selectively essential gene in chordoma, and that transcriptional cyclin-dependent kinase (CDK) inhibitors targeting CDK7/12/13 and CDK9 potently suppress chordoma cell proliferation. In other cancer types, transcriptional CDK inhibitors have been observed to downregulate highly expressed, enhancer-associated oncogenic transcription factors4,5. In chordoma, we find that T is associated with a 1.5-Mb region containing 'super-enhancers' and is the most highly expressed super-enhancer-associated transcription factor. Notably, transcriptional CDK inhibition leads to preferential and concentration-dependent downregulation of cellular brachyury protein levels in all models tested. In vivo, CDK7/12/13-inhibitor treatment substantially reduces tumor growth. Together, these data demonstrate small-molecule targeting of brachyury transcription factor addiction in chordoma, identify a mechanism of T gene regulation that underlies this therapeutic strategy, and provide a blueprint for applying systematic genetic and chemical screening approaches to discover vulnerabilities in genomically quiet cancers.
PMID: 30664779
ISSN: 1546-170x
CID: 3610422

Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity

Vriens, Kim; Christen, Stefan; Parik, Sweta; Broekaert, Dorien; Yoshinaga, Kazuaki; Talebi, Ali; Dehairs, Jonas; Escalona-Noguero, Carmen; Schmieder, Roberta; Cornfield, Thomas; Charlton, Catriona; Romero-Pérez, Laura; Rossi, Matteo; Rinaldi, Gianmarco; Orth, Martin F; Boon, Ruben; Kerstens, Axelle; Kwan, Suet Ying; Faubert, Brandon; Méndez-Lucas, Andrés; Kopitz, Charlotte C; Chen, Ting; Fernandez-Garcia, Juan; Duarte, João A G; Schmitz, Arndt A; Steigemann, Patrick; Najimi, Mustapha; Hägebarth, Andrea; Van Ginderachter, Jo A; Sokal, Etienne; Gotoh, Naohiro; Wong, Kwok-Kin; Verfaillie, Catherine; Derua, Rita; Munck, Sebastian; Yuneva, Mariia; Beretta, Laura; DeBerardinis, Ralph J; Swinnen, Johannes V; Hodson, Leanne; Cassiman, David; Verslype, Chris; Christian, Sven; Grünewald, Sylvia; Grünewald, Thomas G P; Fendt, Sarah-Maria
Most tumours have an aberrantly activated lipid metabolism1,2 that enables them to synthesize, elongate and desaturate fatty acids to support proliferation. However, only particular subsets of cancer cells are sensitive to approaches that target fatty acid metabolism and, in particular, fatty acid desaturation3. This suggests that many cancer cells contain an unexplored plasticity in their fatty acid metabolism. Here we show that some cancer cells can exploit an alternative fatty acid desaturation pathway. We identify various cancer cell lines, mouse hepatocellular carcinomas, and primary human liver and lung carcinomas that desaturate palmitate to the unusual fatty acid sapienate to support membrane biosynthesis during proliferation. Accordingly, we found that sapienate biosynthesis enables cancer cells to bypass the known fatty acid desaturation pathway that is dependent on stearoyl-CoA desaturase. Thus, only by targeting both desaturation pathways is the in vitro and in vivo proliferation of cancer cells that synthesize sapienate impaired. Our discovery explains metabolic plasticity in fatty acid desaturation and constitutes an unexplored metabolic rewiring in cancers.
PMID: 30728499
ISSN: 1476-4687
CID: 3632262

Overcome LKB1 mutated cancer resistance to anti-PD1 treatment [Meeting Abstract]

Deng, Jiehui; Thennavan, Aatish; Pan, Yuanwang; Dolgalev, Igor; Chen, Ting; Silver, Heather; Harris, Matthew; Pyon, Val; Li, Fei; Lee, Chelsea; Tsirigos, Aristotelis; Rothenberg, Eli; Perou, Charles M.; Wong, Kwok-Kin
ISI:000488279402164
ISSN: 0008-5472
CID: 5381142

Targeting PKCδ as a Therapeutic Strategy against Heterogeneous Mechanisms of EGFR Inhibitor Resistance in EGFR-Mutant Lung Cancer

Lee, Pei-Chih; Fang, Yueh-Fu; Yamaguchi, Hirohito; Wang, Wei-Jan; Chen, Tse-Ching; Hong, Xuan; Ke, Baozhen; Xia, Weiya; Wei, Yongkun; Zha, Zhengyu; Wang, Yan; Kuo, Han-Pin; Wang, Chih-Wei; Tu, Chih-Yen; Chen, Chia-Hung; Huang, Wei-Chien; Chiang, Shu-Fen; Nie, Lei; Hou, Junwei; Chen, Chun-Te; Huo, Longfei; Yang, Wen-Hao; Deng, Rong; Nakai, Katsuya; Hsu, Yi-Hsin; Chang, Shih-Shin; Chiu, Tai-Jan; Tang, Jun; Zhang, Ran; Wang, Li; Fang, Bingliang; Chen, Ting; Wong, Kwok-Kin; Hsu, Jennifer L; Hung, Mien-Chie
Multiple mechanisms of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been identified in EGFR-mutant non-small cell lung cancer (NSCLC); however, recurrent resistance to EGFR TKIs due to the heterogeneous mechanisms underlying resistance within a single patient remains a major challenge in the clinic. Here, we report a role of nuclear protein kinase Cδ (PKCδ) as a common axis across multiple known TKI-resistance mechanisms. Specifically, we demonstrate that TKI-inactivated EGFR dimerizes with other membrane receptors implicated in TKI resistance to promote PKCδ nuclear translocation. Moreover, the level of nuclear PKCδ is associated with TKI response in patients. The combined inhibition of PKCδ and EGFR induces marked regression of resistant NSCLC tumors with EGFR mutations.
PMID: 30537515
ISSN: 1878-3686
CID: 3659292

RIP1 Kinase Drives Macrophage-Mediated Adaptive Immune Tolerance in Pancreatic Cancer

Wang, Wei; Marinis, Jill M; Beal, Allison M; Savadkar, Shivraj; Wu, Yue; Khan, Mohammed; Taunk, Pardeep S; Wu, Nan; Su, Wenyu; Wu, Jingjing; Ahsan, Aarif; Kurz, Emma; Chen, Ting; Yaboh, Inedouye; Li, Fei; Gutierrez, Johana; Diskin, Brian; Hundeyin, Mautin; Reilly, Michael; Lich, John D; Harris, Philip A; Mahajan, Mukesh K; Thorpe, James H; Nassau, Pamela; Mosley, Julie E; Leinwand, Joshua; Kochen Rossi, Juan A; Mishra, Ankita; Aykut, Berk; Glacken, Michael; Ochi, Atsuo; Verma, Narendra; Kim, Jacqueline I; Vasudevaraja, Varshini; Adeegbe, Dennis; Almonte, Christina; Bagdatlioglu, Ece; Cohen, Deirdre J; Wong, Kwok-Kin; Bertin, John; Miller, George
Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.
PMID: 30423296
ISSN: 1878-3686
CID: 3457042

Assessing Therapeutic Efficacy of MEK Inhibition in a KRAS G12C-Driven Mouse Model of Lung Cancer

Li, Shuai; Liu, Shengwu; Deng, Jiehui; Akbay, Esra A; Hai, Josephine; Ambrogio, Chiara; Zhang, Long; Zhou, Fangyu; Jenkins, Russell W; Adeegbe, Dennis O; Gao, Peng; Wang, Xiaoen; Paweletz, Cloud P; Herter-Sprie, Grit S; Chen, Ting; Gutierrez Quiceno, Laura; Zhang, Yanxi; Merlino, Ashley A; Quinn, Max M; Zeng, Yu; Yu, Xiaoting; Liu, Yuting; Fan, Lichao; Aguirre, Andrew J; Barbie, David A; Yi, Xianghua; Wong, Kwok-Kin
PURPOSE/OBJECTIVE:Despite the challenge to directly target mutant KRAS due to its high GTP affinity, some agents are under development against downstream signaling pathways, such as MEK inhibitors. However, it remains controversial whether MEK inhibitors can boost current chemotherapy in KRAS-mutant lung tumors in clinic. Considering the genomic heterogeneity among lung cancer patients, it is valuable to test potential therapeutics in KRAS-mutation driven mouse models. EXPERIMENTAL DESIGN/METHODS:We first compared the pERK1/2 level in lung cancer samples with different KRAS substitutions and generated a new genetically engineered mouse model whose tumor was driven by KRAS G12C, the most common KRAS mutation in lung cancer. Next, we evaluated the efficacy of selumetinib or its combination with chemotherapy, in KRAS G12C tumors compared to KRAS G12D tumors. Moreover, we generated KRAS G12C/p53 R270H model to explore the role of a dominant negative p53 mutation detected in patients in responsiveness to MEK inhibition. RESULTS:We determined higher pERK1/2 in KRAS G12C lung tumors compared to KRAS G12D. Using mouse models, we further identified that KRAS G12C tumors are significantly more sensitive to selumetinib compared with Kras G12D tumors. MEK inhibition significantly increased chemotherapeutic efficacy and progression-free survival of KRAS G12C mice. Interestingly, p53 co-mutation rendered KRAS G12C lung tumors less sensitive to combination treatment with selumetinib and chemotherapy. CONCLUSIONS:Our data demonstrate that unique KRAS mutations and concurrent mutations in tumor-suppressor genes are important factors for lung tumor responses to MEK inhibitor. Our preclinical study supports further clinical evaluation of combined MEK inhibition and chemotherapy for lung cancer patients harboring KRAS G12C and wildtype p53 status.
PMID: 29945997
ISSN: 1078-0432
CID: 3162862

Overcoming Resistance to Dual Innate Immune and MEK Inhibition Downstream of KRAS

Kitajima, Shunsuke; Asahina, Hajime; Chen, Ting; Guo, Sujuan; Quiceno, Laura Gutierrez; Cavanaugh, Jillian D; Merlino, Ashley A; Tange, Shoichiro; Terai, Hideki; Kim, Jong Wook; Wang, Xiaoen; Zhou, Shan; Xu, Man; Wang, Stephen; Zhu, Zehua; Thai, Tran C; Takahashi, Chiaki; Wang, Yujin; Neve, Richard; Stinson, Susanna; Tamayo, Pablo; Watanabe, Hideo; Kirschmeier, Paul T; Wong, Kwok-Kin; Barbie, David A
Despite extensive efforts, oncogenic KRAS remains resistant to targeted therapy. Combined downstream RAL-TBK1 and MEK inhibition induces only transient lung tumor shrinkage in KRAS-driven genetically engineered mouse models (GEMMs). Using the sensitive KRAS;LKB1 (KL) mutant background, we identify YAP1 upregulation and a therapy-induced secretome as mediators of acquired resistance. This program is reversible, associated with H3K27 promoter acetylation, and suppressed by BET inhibition, resensitizing resistant KL cells to TBK1/MEK inhibition. Constitutive YAP1 signaling promotes intrinsic resistance in KRAS;TP53 (KP) mutant lung cancer. Intermittent treatment with the BET inhibitor JQ1 thus overcomes resistance to combined pathway inhibition in KL and KP GEMMs. Using potent and selective TBK1 and BET inhibitors we further develop an effective therapeutic strategy with potential translatability to the clinic.
PMID: 30205046
ISSN: 1878-3686
CID: 3277732

Palbociclib resistance confers dependence on an FGFR-MAP kinase-mTOR-driven pathway in KRAS-mutant non-small cell lung cancer

Haines, Eric; Chen, Ting; Kommajosyula, Naveen; Chen, Zhao; Herter-Sprie, Grit S; Cornell, Liam; Wong, Kwok-Kin; Shapiro, Geoffrey I
CDK4 is emerging as a target in KRAS-mutant non-small cell lung cancer (NSCLC). We demonstrate that KRAS-mutant NSCLC cell lines are initially sensitive to the CDK4/6 inhibitor palbociclib, but readily acquire resistance associated with increased expression of CDK6, D-type cyclins and cyclin E. Resistant cells also demonstrated increased ERK1/2 activity and sensitivity to MEK and ERK inhibitors. Moreover, MEK inhibition reduced the expression and activity of cell cycle proteins mediating palbociclib resistance. In resistant cells, ERK activated mTOR, driven in part by upstream FGFR1 signaling resulting from the extracellular secretion of FGF ligands. A genetically-engineered mouse model of KRAS-mutant NSCLC initially sensitive to palbociclib similarly developed acquired resistance with increased expression of cell cycle mediators, ERK1/2 and FGFR1. In this model, resistance was delayed with combined palbociclib and MEK inhibitor treatment. These findings implicate an FGFR1-MAP kinase-mTOR pathway resulting in increased expression of D-cyclins and CDK6 that confers palbociclib resistance and indicate that CDK4/6 inhibition acts to promote MAP kinase dependence.
PMCID:6114982
PMID: 30167080
ISSN: 1949-2553
CID: 3257152

NK cells mediate synergistic antitumor effects of combined inhibition of HDAC6 and BET in a SCLC preclinical model

Liu, Yan; Li, Yuyang; Liu, Shengwu; Adeegbe, Dennis O; Christensen, Camilla L; Quinn, Max M; Dries, Ruben; Han, Shiwei; Buczkowski, Kevin; Wang, Xiaoen; Chen, Ting; Gao, Peng; Zhang, Hua; Li, Fei; Hammerman, Peter S; Bradner, James E; Quayle, Steven N; Wong, Kwok-Kin
Small cell lung cancer (SCLC) has the highest malignancy among all lung cancers, exhibiting aggressive growth and early metastasis to distant sites. For 30 years, treatment options for SCLC have been limited to chemotherapy, warranting the need for more effective treatments. Frequent inactivation of TP53 and RB1 as well as histone dysmodifications in SCLC suggest that transcriptional and epigenetic regulations play a major role in SCLC disease evolution. Here we performed a synthetic lethal screen using the BET inhibitor JQ1 and an shRNA library targeting 550 epigenetic genes in treatment-refractory SCLC xenograft models and identified HDAC6 as a synthetic lethal target in combination with JQ1. Combined treatment of human and mouse SCLC cell line-derived xenograft tumors with the HDAC6 inhibitor ricolinostat (ACY-1215) and JQ1 demonstrated significant inhibition of tumor growth; this effect was abolished upon depletion of NK cells, suggesting that these innate immune lymphoid cells play a role in SCLC tumor treatment response. Collectively, these findings suggest a potential new treatment for recurrent SCLC.
PMID: 29760044
ISSN: 1538-7445
CID: 3121342

Targeting HER2 Aberrations in Non-Small Cell Lung Cancer with Osimertinib

Liu, Shengwu; Li, Shuai; Hai, Josephine; Wang, Xiaoen; Chen, Ting; Quinn, Max M; Gao, Peng; Zhang, Yanxi; Ji, Hongbin; Cross, Darren; Wong, Kwok-Kin
PURPOSE/OBJECTIVE:HER2 (or ERBB2) aberrations, including both amplification and mutations, have been classified as oncogenic drivers that contribute to 2-6 percent of lung adenocarcinomas. HER2 amplification is also an important mechanism for acquired resistance to EGFR tyrosine kinase inhibitors (TKIs). However, due to limited preclinical studies and clinical trials, currently there is still no available standard of care for lung cancer patients with HER2 aberrations. To fulfill the clinical need for targeting HER2 in non-small cell lung cancer (NSCLC) patients, we performed a comprehensive pre-clinical study to evaluate the efficacy of a third-generation TKI, osimertinib (AZD9291).  Experimental Design:Three genetically modified mouse models (GEMMs) mimicking individual HER2 alterations in NSCLC were generated and osimertinib was tested for its efficacy against these HER2 aberrations in vivo. RESULTS:Osimertinib treatment showed robust efficacy in HER2wt overexpression and EGFR del19/HER2 models but not in HER2 exon 20 insertion tumors. Interestingly, we further identified that combined treatment with osimertinib and the BET inhibitor JQ1 significantly increased the response rate in HER2-mutant NSCLC while JQ1 single treatment did not show efficacy. CONCLUSIONS:Overall, our data indicated robust anti-tumor efficacy of osimertinib against multiple HER2 aberrations in lung cancer, either as a single agent or in combination with JQ1. Our study provides a strong rationale for future clinical trials using osimertinib either alone or in combination with epigenetic drugs to target aberrant HER2 in NSCLC patients.
PMID: 29298799
ISSN: 1078-0432
CID: 2899562