Try a new search

Format these results:

Searched for:

in-biosketch:true

person:wongk11

Total Results:

344


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

Gain-of-Function RHOA Mutations Promote Focal Adhesion Kinase Activation and Dependency in Diffuse Gastric Cancer

Zhang, Haisheng; Schaefer, Antje; Wang, Yichen; Hodge, Richard G; Blake, Devon R; Diehl, J Nathaniel; Papageorge, Alex G; Stachler, Matthew D; Liao, Jennifer; Zhou, Jin; Wu, Zhong; Akarca, Fahire G; de Klerk, Leonie K; Derks, Sarah; Pierobon, Mariaelena; Hoadley, Katherine A; Wang, Timothy Cragin; Church, George; Wong, Kwok-Kin; Petricoin, Emanuel F; Cox, Adrienne D; Lowy, Douglas R; Der, Channing J; Bass, Adam J
Diffuse gastric cancer (DGC) is a lethal malignancy lacking effective systemic therapy. Among the most provocative recent results in DGC has been that of highly recurrent missense mutations in the GTPase RHOA. The function of these mutations has remained unresolved. We demonstrate that RHOAY42C, the most common RHOA mutation in DGC, is a gain-of-function oncogenic mutant and that expression of RHOAY42C with inactivation of canonical tumor suppressor Cdh1 induces metastatic DGC in a mouse model. Biochemically, RHOAY42C exhibits impaired GTP hydrolysis and enhances interaction with its effector ROCK. RHOAY42C mutation and Cdh1 loss induce actin/cytoskeletal rearrangements and activity of focal adhesion kinase (FAK), which activates YAP/TAZ, phosphoinositide 3-kinase (PI3K)/AKT and β-catenin. RHOAY42C murine models were sensitive to FAK inhibition and to combined YAP and PI3K pathway blockade. These results, coupled to sensitivity to FAK inhibition in patient-derived DGC cell lines, nominate FAK as a novel target for these cancers.
PMID: 31771969
ISSN: 2159-8290
CID: 4215912

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

The KDM5A/RBP2 histone demethylase represses NOTCH signaling to sustain neuroendocrine differentiation and promote small cell lung cancer tumorigenesis

Oser, Matthew G; Sabet, Amin H; Gao, Wenhua; Chakraborty, Abhishek A; Schinzel, Anna C; Jennings, Rebecca B; Fonseca, Raquel; Bonal, Dennis M; Booker, Matthew A; Flaifel, Abdallah; Novak, Jesse S; Christensen, Camilla L; Zhang, Hua; Herbert, Zachary T; Tolstorukov, Michael Y; Buss, Elizabeth J; Wong, Kwok-Kin; Bronson, Roderick T; Nguyen, Quang-De; Signoretti, Sabina; Kaelin, William G
More than 90% of small cell lung cancers (SCLCs) harbor loss-of-function mutations in the tumor suppressor gene RB1 The canonical function of the RB1 gene product, pRB, is to repress the E2F transcription factor family, but pRB also functions to regulate cellular differentiation in part through its binding to the histone demethylase KDM5A (also known as RBP2 or JARID1A). We show that KDM5A promotes SCLC proliferation and SCLC's neuroendocrine differentiation phenotype in part by sustaining expression of the neuroendocrine transcription factor ASCL1. Mechanistically, we found that KDM5A sustains ASCL1 levels and neuroendocrine differentiation by repressing NOTCH2 and NOTCH target genes. To test the role of KDM5A in SCLC tumorigenesis in vivo, we developed a CRISPR/Cas9-based mouse model of SCLC by delivering an adenovirus (or an adeno-associated virus [AAV]) that expresses Cre recombinase and sgRNAs targeting Rb1, Tp53, and Rbl2 into the lungs of Lox-Stop-Lox Cas9 mice. Coinclusion of a KDM5A sgRNA decreased SCLC tumorigenesis and metastasis, and the SCLCs that formed despite the absence of KDM5A had higher NOTCH activity compared to KDM5A+/+ SCLCs. This work establishes a role for KDM5A in SCLC tumorigenesis and suggests that KDM5 inhibitors should be explored as treatments for SCLC.
PMID: 31727771
ISSN: 1549-5477
CID: 4187012

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

CXCR7 reactivates ERK signaling to promote resistance to EGFR kinase inhibitors in NSCLC

Becker, Jeffrey H; Gao, Yandi; Soucheray, Margaret; Pulido, Ines; Kikuchi, Eiki; Rodríguez, María L; Gandhi, Rutu; Lafuente-Sanchis, Aranzazu; Aupí, Miguel; Alcácer Fernández-Coronado, Javier; Martín-Martorell, Paloma; Cremades, Antonio; Galbis-Caravajal, José M; Alcácer, Javier; Christensen, Camilla L; Simms, Patricia; Hess, Ashley; Asahina, Hajime; Kahle, Michael P; Al-Shahrour, Fatima; Borgia, Jeffrey A; Lahoz, Agustín; Insa, Amelia; Juan, Oscar; Janne, Pasi A; Wong, Kwok-Kin; Carretero, Julian; Shimamura, Takeshi
Although EGFR mutant-selective TKIs are clinically effective, acquired resistance can occur by reactivating ERK. We show using in vitro models of acquired EGFR TKI resistance with a mesenchymal phenotype that CXCR7, an atypical GPCR, activates the MAPK-ERK pathway via β-arrestin. Depletion of CXCR7 inhibited the MAPK pathway, significantly attenuated EGFR TKI resistance and resulted in mesenchymal to epithelial transition. CXCR7 overexpression was essential in reactivation of ERK1/2 for the generation of EGFR TKI resistant persister cells. Many NSCLC patients harboring an EGFR kinase domain mutation, who progressed on EGFR inhibitors, demonstrated increased CXCR7 expression. These data suggest that CXCR7 inhibition could considerably delay and prevent the emergence of acquired EGFR TKI resistance in EGFR mutant NSCLC.
PMID: 31273063
ISSN: 1538-7445
CID: 3968292

Innate αβ T cells Mediate Antitumor Immunity by Orchestrating Immunogenic Macrophage Programming

Hundeyin, Mautin; Kurz, Emma; Mishra, Ankita; Kochen Rossi, Juan Andres; Liudahl, Shannon M; Leis, Kenna R; Mehrotra, Harshita; Kim, Mirhee; Torres, Luisana E; Ogunsakin, Adesola; Link, Jason; Sears, Rosalie C; Sivagnanam, Shamilene; Goecks, Jeremy; Islam, Km Sadeq; Dolgalev, Igor; Savadkar, Shivraj; Wang, Wei; Aykut, Berk; Leinwand, Joshua; Diskin, Brian; Adam, Salma; Israr, Muhammad; Gelas, Maeliss; Lish, Justin; Chin, Kathryn; Farooq, Mohammad Saad; Wadowski, Benjamin; Wu, Jingjing; Shah, Suhagi; Adeegbe, Dennis O; Pushalkar, Smruti; Vasudevaraja, Varshini; Saxena, Deepak; Wong, Kwok-Kin; Coussens, Lisa M; Miller, George
Unconventional T lymphocyte populations are emerging as important regulators of tumor immunity. Despite this, the role of TCRαβ+CD4-CD8-NK1.1- innate αβ T-cells (iαβTs) in pancreatic ductal adenocarcinoma (PDA) has not been explored. We found that iαβTs represent ~10% of T-lymphocytes infiltrating PDA in mice and humans. Intra-tumoral iαβTs express a distinct TCR-repertoire and profoundly immunogenic phenotype compared to their peripheral counterparts and conventional lymphocytes. iαβTs comprised ~75% of the total intra-tumoral IL-17+ cells. Moreover, iαβT cell adoptive transfer is protective in both murine models of PDA and human organotypic systems. We show iαβT cells induce a CCR5-dependent immunogenic macrophage reprogramming, thereby enabling marked CD4+ and CD8+ T cell expansion/activation and tumor protection. Collectively, iαβTs govern fundamental intra-tumoral crosstalk between innate and adaptive immune populations and are attractive therapeutic targets.
PMID: 31266770
ISSN: 2159-8290
CID: 3968062

The combined effect of FGFR inhibition and PD-1 blockade promotes tumor-intrinsic induction of antitumor immunity

Palakurthi, Sangeetha; Kuraguchi, Mari; Zacharek, Sima J; Zudaire, Enrique; Huang, Wei; Bonal, Dennis M; Liu, Jeffrey; Dhaneshwar, Abha; Depeaux, Kristin; Gowaski, Martha R; Bailey, Dyane; Regan, Samuel N; Ivanova, Elena; Ferrante, Catherine; English, Jessie M; Khosla, Aditya; Beck, Andrew H; Rytlewski, Julie A; Sanders, Catherine; Laquerre, Sylvie; Bittinger, Mark A; Kirschmeier, Paul T; Packman, Kathryn; Janne, Pasi A; Moy, Christopher; Wong, Kwok-Kin; Verona, Raluca I; Lorenzi, Matthew V
The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti-PD-1 alone was ineffective, the erdafitinib and anti-PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C mutant genetically engineered mouse model (GEMM), which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased TCR clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti-PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti-PD-1 drives expansion of T-cell clones and immunological changes in the tumor microenvironment to support enhanced antitumor immunity and survival.
PMID: 31331945
ISSN: 2326-6074
CID: 3987902

Author Correction: A systems biology pipeline identifies regulatory networks for stem cell engineering

Kinney, Melissa A; Vo, Linda T; Frame, Jenna M; Barragan, Jessica; Conway, Ashlee J; Li, Shuai; Wong, Kwok-Kin; Collins, James J; Cahan, Patrick; North, Trista E; Lauffenburger, Douglas A; Daley, George Q
In the version of this article initially published, the second NIH grant "R24-DK49216" to author George Q. Daley contained an error. The grant number should have read U54DK110805. The error has been corrected in the HTML and PDF versions of the article.
PMID: 31312048
ISSN: 1546-1696
CID: 3977842