Try a new search

Format these results:

Searched for:

person:chent15

in-biosketch:yes

Total Results:

65


Generation of genetically engineered mouse lung organoid models for squamous cell lung cancers allows for the study of combinatorial immunotherapy

Hai, Josephine; Zhang, Hua; Zhou, Jin; Wu, Zhong; Chen, Ting; Papadopoulos, Eleni; Dowling, Catríona M; Pyon, Val; Pan, Yuanwang; Liu, Jie B; Bronson, Roderick T; Silver, Heather; Lizotte, Patrick H; Deng, Jiehui; Campbell, Joshua D; Sholl, Lynette M; Ng, Christine; Tsao, Ming-Sound; Thakurdin, Cassandra; Bass, Adam J; Wong, Kwok-Kin
PURPOSE/OBJECTIVE:Lung squamous cell carcinoma (LSCC) is a deadly disease for which only a subset of patients responds to immune checkpoint blockade (ICB) therapy. Therefore, preclinical mouse models that recapitulate the complex genetic profile found in patients are urgently needed. EXPERIMENTAL DESIGN/METHODS:We used CRISPR genome editing to delete multiple tumor suppressors in lung organoids derived from Cre-dependent SOX2 knock-in mice. We investigated both the therapeutic efficacy and immunological effects accompanying combination PD-1 blockade and WEE1 inhibition in both mouse models and LSCC patient-derived cell lines. RESULTS:We show that multiplex gene editing of mouse lung organoids using the CRISPR-Cas9 system allows for efficient and rapid means to generate LSCCs that closely mimic the human disease at the genomic and phenotypic level. Using this genetically-defined mouse model and three-dimensional tumoroid culture system, we show that WEE1 inhibition induces DNA damage that primes the endogenous type I interferon and antigen presentation system in primary LSCC tumor cells. These events promote cytotoxic T cell-mediated clearance of tumor cells and reduce the accumulation of tumor-infiltrating neutrophils. Beneficial immunological features of WEE1 inhibition are further enhanced by the addition of anti-PD-1 therapy. CONCLUSIONS:We developed a mouse model system to investigate a novel combinatory approach that illuminates a clinical path hypothesis for combining ICB with DNA damage-inducing therapies in the treatment of LSCC.
PMID: 32209571
ISSN: 1078-0432
CID: 4358482

Use of ex vivo patient derived tumor organotypic spheroids to identify combination therapies for HER2mutant non small cell lung cancer

Ivanova, Elena; Kuraguchi, Mari; Xu, Man; Portell, Andrew; Taus, Luke J; Diala, Irmina; Lalani, Alshad S; Choi, Jihyun; Chambers, Emily S; Li, Shuai; Liu, Shengwu; Chen, Ting; Barbie, Thanh U; Oxnard, Geoffrey R; Haworth, Jacob; Wong, Kwok-Kin; Dahlberg, Suzanne E; Aref, Amir; Barbie, David A; Bahcall, Magda; Paweletz, Cloud P; Janne, Pasi A
PURPOSE/OBJECTIVE:system. EXPERIMENTAL DESIGN/METHODS:genetically engineered mouse model (GEMM). RESULTS:was more effective compared to single agent neratinib or trastuzumab and was associated with more robust inhibition of HER2 and downstream signaling. CONCLUSIONS:using PDX tumors. This approach may accelerate the identification and clinical development of therapies for targets with no or few existing models and/or therapies.
PMID: 32034078
ISSN: 1078-0432
CID: 4301622

PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer

Diskin, Brian; Adam, Salma; Cassini, Marcelo F; Sanchez, Gustavo; Liria, Miguel; Aykut, Berk; Buttar, Chandan; Li, Eric; Sundberg, Belen; Salas, Ruben D; Chen, Ruonan; Wang, Junjie; Kim, Mirhee; Farooq, Mohammad Saad; Nguy, Susanna; Fedele, Carmine; Tang, Kwan Ho; Chen, Ting; Wang, Wei; Hundeyin, Mautin; Rossi, Juan A Kochen; Kurz, Emma; Haq, Muhammad Israr Ul; Karlen, Jason; Kruger, Emma; Sekendiz, Zennur; Wu, Dongling; Shadaloey, Sorin A A; Baptiste, Gillian; Werba, Gregor; Selvaraj, Shanmugapriya; Loomis, Cynthia; Wong, Kwok-Kin; Leinwand, Joshua; Miller, George
Programmed cell death protein 1 (PD-1) ligation delimits immunogenic responses in T cells. However, the consequences of programmed cell death 1 ligand 1 (PD-L1) ligation in T cells are uncertain. We found that T cell expression of PD-L1 in cancer was regulated by tumor antigen and sterile inflammatory cues. PD-L1+ T cells exerted tumor-promoting tolerance via three distinct mechanisms: (1) binding of PD-L1 induced STAT3-dependent 'back-signaling' in CD4+ T cells, which prevented activation, reduced TH1-polarization and directed TH17-differentiation. PD-L1 signaling also induced an anergic T-bet-IFN-γ- phenotype in CD8+ T cells and was equally suppressive compared to PD-1 signaling; (2) PD-L1+ T cells restrained effector T cells via the canonical PD-L1-PD-1 axis and were sufficient to accelerate tumorigenesis, even in the absence of endogenous PD-L1; (3) PD-L1+ T cells engaged PD-1+ macrophages, inducing an alternative M2-like program, which had crippling effects on adaptive antitumor immunity. Collectively, we demonstrate that PD-L1+ T cells have diverse tolerogenic effects on tumor immunity.
PMID: 32152508
ISSN: 1529-2916
CID: 4349682

In vivo epigenetic CRISPR screen identifies Asf1a as an immunotherapeutic target in Kras-mutant lung adenocarcinoma

Li, Fei; Huang, Qingyuan; Luster, Troy A; Hu, Hai; Zhang, Hua; Ng, Wai-Lung; Khodadadi-Jamayran, Alireza; Wang, Wei; Chen, Ting; Deng, Jiehui; Ranieri, Michela; Fang, Zhaoyuan; Pyon, Val; Dowling, Catriona M; Bagdatlioglu, Ece; Almonte, Christina; Labbe, Kristen; Silver, Heather; Rabin, Alexandra R; Jani, Kandarp; Tsirigos, Aristotelis; Papagiannakopoulos, Thales; Hammerman, Peter S; Velcheti, Vamsidhar; Freeman, Gordon J; Qi, Jun; Miller, George; Wong, Kwok-Kin
Despite substantial progress in lung cancer immunotherapy, the overall response rate in KRAS-mutant lung adenocarcinoma (ADC) patients remains low. Combining standard immunotherapy with adjuvant approaches that enhance adaptive immune responses-such as epigenetic modulation of anti-tumor immunity-is therefore an attractive strategy. To identify epigenetic regulators of tumor immunity, we constructed an epigenetic-focused sgRNA library, and performed an in vivo CRISPR screen in a KrasG12D/P53-/- (KP) lung ADC model. Our data showed that loss of the histone chaperone Asf1a in tumor cells sensitizes tumors to anti-PD-1 treatment. Mechanistic studies revealed that tumor cell-intrinsic Asf1a deficiency induced immunogenic macrophage differentiation in the tumor microenvironment by upregulating GM-CSF expression and potentiated T cell activation in combination with anti-PD-1. Our results provide rationale for a novel combination therapy consisting of ASF1A inhibition and anti-PD-1 immunotherapy.
PMID: 31744829
ISSN: 2159-8290
CID: 4208912

CDK7 Inhibition Potentiates Genome Instability Triggering Anti-tumor Immunity in Small Cell Lung Cancer

Zhang, Hua; Christensen, Camilla L; Dries, Ruben; Oser, Matthew G; Deng, Jiehui; Diskin, Brian; Li, Fei; Pan, Yuanwang; Zhang, Xuzhu; Yin, Yandong; Papadopoulos, Eleni; Pyon, Val; Thakurdin, Cassandra; Kwiatkowski, Nicholas; Jani, Kandarp; Rabin, Alexandra R; Castro, Dayanne M; Chen, Ting; Silver, Heather; Huang, Qingyuan; Bulatovic, Mirna; Dowling, Catríona M; Sundberg, Belen; Leggett, Alan; Ranieri, Michela; Han, Han; Li, Shuai; Yang, Annan; Labbe, Kristen E; Almonte, Christina; Sviderskiy, Vladislav O; Quinn, Max; Donaghue, Jack; Wang, Eric S; Zhang, Tinghu; He, Zhixiang; Velcheti, Vamsidhar; Hammerman, Peter S; Freeman, Gordon J; Bonneau, Richard; Kaelin, William G; Sutherland, Kate D; Kersbergen, Ariena; Aguirre, Andrew J; Yuan, Guo-Cheng; Rothenberg, Eli; Miller, George; Gray, Nathanael S; Wong, Kwok-Kin
Cyclin-dependent kinase 7 (CDK7) is a central regulator of the cell cycle and gene transcription. However, little is known about its impact on genomic instability and cancer immunity. Using a selective CDK7 inhibitor, YKL-5-124, we demonstrated that CDK7 inhibition predominately disrupts cell-cycle progression and induces DNA replication stress and genome instability in small cell lung cancer (SCLC) while simultaneously triggering immune-response signaling. These tumor-intrinsic events provoke a robust immune surveillance program elicited by T cells, which is further enhanced by the addition of immune-checkpoint blockade. Combining YKL-5-124 with anti-PD-1 offers significant survival benefit in multiple highly aggressive murine models of SCLC, providing a rationale for new combination regimens consisting of CDK7 inhibitors and immunotherapies.
PMID: 31883968
ISSN: 1878-3686
CID: 4251032

Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway

Kurppa, Kari J; Liu, Yao; To, Ciric; Zhang, Tinghu; Fan, Mengyang; Vajdi, Amir; Knelson, Erik H; Xie, Yingtian; Lim, Klothilda; Cejas, Paloma; Portell, Andrew; Lizotte, Patrick H; Ficarro, Scott B; Li, Shuai; Chen, Ting; Haikala, Heidi M; Wang, Haiyun; Bahcall, Magda; Gao, Yang; Shalhout, Sophia; Boettcher, Steffen; Shin, Bo Hee; Thai, Tran; Wilkens, Margaret K; Tillgren, Michelle L; Mushajiang, Mierzhati; Xu, Man; Choi, Jihyun; Bertram, Arrien A; Ebert, Benjamin L; Beroukhim, Rameen; Bandopadhayay, Pratiti; Awad, Mark M; Gokhale, Prafulla C; Kirschmeier, Paul T; Marto, Jarrod A; Camargo, Fernando D; Haq, Rizwan; Paweletz, Cloud P; Wong, Kwok-Kin; Barbie, David A; Long, Henry W; Gray, Nathanael S; Jänne, Pasi A
Eradicating tumor dormancy that develops following epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) treatment of EGFR-mutant non-small cell lung cancer, is an attractive therapeutic strategy but the mechanisms governing this process are poorly understood. Blockade of ERK1/2 reactivation following EGFR TKI treatment by combined EGFR/MEK inhibition uncovers cells that survive by entering a senescence-like dormant state characterized by high YAP/TEAD activity. YAP/TEAD engage the epithelial-to-mesenchymal transition transcription factor SLUG to directly repress pro-apoptotic BMF, limiting drug-induced apoptosis. Pharmacological co-inhibition of YAP and TEAD, or genetic deletion of YAP1, all deplete dormant cells by enhancing EGFR/MEK inhibition-induced apoptosis. Enhancing the initial efficacy of targeted therapies could ultimately lead to prolonged treatment responses in cancer patients.
PMID: 31935369
ISSN: 1878-3686
CID: 4264372

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