Searched for: in-biosketch:yes
person:chent15
Author Correction: LKB1 loss links serine metabolism to DNA methylation and tumorigenesis
Kottakis, Filippos; Nicolay, Brandon N; Roumane, Ahlima; Karnik, Rahul; Gu, Hongcang; Nagle, Julia M; Boukhali, Myriam; Hayward, Michele C; Li, Yvonne Y; Chen, Ting; Liesa, Marc; Hammerman, Peter S; Wong, Kwok Kin; Hayes, D Neil; Shirihai, Orian S; Dyson, Nicholas J; Haas, Wilhelm; Meissner, Alexander; Bardeesy, Nabeel
An Amendment to this paper has been published and can be accessed via a link at the top of the paper.
PMID: 31695192
ISSN: 1476-4687
CID: 5381322
Pan-Cancer Landscape and Analysis of ERBB2 Mutations Identifies Poziotinib as a Clinically Active Inhibitor and Enhancer of T-DM1 Activity
Robichaux, Jacqulyne P; Elamin, Yasir Y; Vijayan, R S K; Nilsson, Monique B; Hu, Lemei; He, Junqin; Zhang, Fahao; Pisegna, Marlese; Poteete, Alissa; Sun, Huiying; Li, Shuai; Chen, Ting; Han, Han; Negrao, Marcelo Vailati; Ahnert, Jordi Rodon; Diao, Lixia; Wang, Jing; Le, Xiuning; Meric-Bernstam, Funda; Routbort, Mark; Roeck, Brent; Yang, Zane; Raymond, Victoria M; Lanman, Richard B; Frampton, Garrett M; Miller, Vincent A; Schrock, Alexa B; Albacker, Lee A; Wong, Kwok-Kin; Cross, Jason B; Heymach, John V
We characterized the landscape and drug sensitivity of ERBB2 (HER2) mutations in cancers. In 11 datasets (n = 211,726), ERBB2 mutational hotspots varied across 25 tumor types. Common HER2 mutants yielded differential sensitivities to eleven EGFR/HER2 tyrosine kinase inhibitors (TKIs) in vitro, and molecular dynamics simulations revealed that mutants with a reduced drug-binding pocket volume were associated with decreased affinity for larger TKIs. Overall, poziotinib was the most potent HER2 mutant-selective TKI tested. Phase II clinical testing in ERBB2 exon 20-mutant non-small cell lung cancer resulted in a confirmed objective response rate of 42% in the first 12 evaluable patients. In pre-clinical models, poziotinib upregulated HER2 cell-surface expression and potentiated the activity of T-DM1, resulting in complete tumor regression with combination treatment.
PMID: 31588020
ISSN: 1878-3686
CID: 4130472
BORIS promotes chromatin regulatory interactions in treatment-resistant cancer cells
Debruyne, David N; Dries, Ruben; Sengupta, Satyaki; Seruggia, Davide; Gao, Yang; Sharma, Bandana; Huang, Hao; Moreau, Lisa; McLane, Michael; Day, Daniel S; Marco, Eugenio; Chen, Ting; Gray, Nathanael S; Wong, Kwok-Kin; Orkin, Stuart H; Yuan, Guo-Cheng; Young, Richard A; George, Rani E
The CCCTC-binding factor (CTCF), which anchors DNA loops that organize the genome into structural domains, has a central role in gene control by facilitating or constraining interactions between genes and their regulatory elements1,2. In cancer cells, the disruption of CTCF binding at specific loci by somatic mutation3,4 or DNA hypermethylation5 results in the loss of loop anchors and consequent activation of oncogenes. By contrast, the germ-cell-specific paralogue of CTCF, BORIS (brother of the regulator of imprinted sites, also known as CTCFL)6, is overexpressed in several cancers7-9, but its contributions to the malignant phenotype remain unclear. Here we show that aberrant upregulation of BORIS promotes chromatin interactions in ALK-mutated, MYCN-amplified neuroblastoma10 cells that develop resistance to ALK inhibition. These cells are reprogrammed to a distinct phenotypic state during the acquisition of resistance, a process defined by the initial loss of MYCN expression followed by subsequent overexpression of BORIS and a concomitant switch in cellular dependence from MYCN to BORIS. The resultant BORIS-regulated alterations in chromatin looping lead to the formation of super-enhancers that drive the ectopic expression of a subset of proneural transcription factors that ultimately define the resistance phenotype. These results identify a previously unrecognized role of BORIS-to promote regulatory chromatin interactions that support specific cancer phenotypes.
PMID: 31391581
ISSN: 1476-4687
CID: 4034372
Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor
To, Ciric; Jang, Jaebong; Chen, Ting; Park, Eunyoung; Mushajiang, Mierzhati; De Clercq, Dries J H; Xu, Man; Wang, Stephen; Cameron, Michael D; Heppner, David E; Shin, Bo Hee; Gero, Thomas W; Yang, Annan; Dahlberg, Suzanne E; Wong, Kwok-Kin; Eck, Michael J; Gray, Nathanael S; Jänne, Pasi A
Allosteric kinase inhibitors offer a potentially complementary therapeutic strategy to ATP-competitive kinase inhibitors due to their distinct sites of target binding. In this study, we identify and study a mutant-selective EGFR allosteric inhibitor, JBJ-04-125-02, which as a single agent can inhibit cell proliferation and EGFRL858R/T790M/C797S signaling in vitro and in vivo. However, increased EGFR dimer formation limits treatment efficacy and leads to drug resistance. Remarkably, osimertinib, an ATP-competitive covalent EGFR inhibitor, uniquely and significantly enhances the binding of JBJ-04-125-02 for mutant EGFR. The combination of osimertinib and JBJ-04-125-02 results in an increase in apoptosis, a more effective inhibition of cellular growth, and an increased efficacy in vitro and in vivo compared with either single agent alone. Collectively, our findings suggest that the combination of a covalent mutant-selective ATP-competitive inhibitor and an allosteric EGFR inhibitor may be an effective therapeutic approach for patients with EGFR-mutant lung cancer. SIGNIFICANCE: The clinical efficacy of EGFR tyrosine kinase inhibitors (TKI) in EGFR-mutant lung cancer is limited by acquired drug resistance, thus highlighting the need for alternative strategies to inhibit EGFR. Here, we identify a mutant EGFR allosteric inhibitor that is effective as a single agent and in combination with the EGFR TKI osimertinib.This article is highlighted in the In This Issue feature, p. 813.
PMCID:6664433
PMID: 31092401
ISSN: 2159-8290
CID: 4967652
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