Try a new search

Format these results:

Searched for:

person:dengj06

in-biosketch:yes

Total Results:

48


The KRASG12C Inhibitor MRTX849 Reconditions the Tumor Immune Microenvironment and Sensitizes Tumors to Checkpoint Inhibitor Therapy

Briere, David M; Li, Shuai; Calinisan, Andrew; Sudhakar, Niranjan; Aranda, Ruth; Hargis, Lauren; Peng, David H; Deng, Jiehui; Engstrom, Lars D; Hallin, Jill; Gatto, Sole; Fernandez-Banet, Julio; Pavlicek, Adam; Wong, Kwok-Kin; Christensen, James G; Olson, Peter
KRASG12C inhibitors, including MRTX849, are promising treatment options for KRAS-mutant non-small cell lung cancer (NSCLC). PD-1 inhibitors are approved in NSCLC; however, strategies to enhance checkpoint inhibitor therapy (CIT) are needed. KRASG12C mutations are smoking-associated transversion mutations associated with high tumor mutation burden (TMB), PD-L1 positivity and an immunosuppressive tumor microenvironment. To evaluate the potential of MRTX849 to augment CIT, its impact on immune signaling and response to CIT was evaluated. In human tumor xenograft models, MRTX849 increased MHC class I protein expression and decreased RNA and/or plasma protein levels of immunosuppressive factors. In a KRASG12C-mutant CT26 syngeneic mouse model, MRTX849 decreased intratumoral myeloid-derived suppressor cells (MDSCs) and increased M1-polarized macrophages, dendritic cells, CD4+ and CD8+ T cells. Similar results were observed in lung KrasG12C-mutant syngeneic and a genetically engineered mouse (GEM) model. In the CT26 KrasG12C model, MRTX849 demonstrated marked tumor regression when tumors were established in immune-competent BALB/c mice; however, the effect was diminished when tumors were grown in T-cell deficient nu/nu mice. Tumors progressed following anti-PD-1 or MRTX849 single agent treatment in immune-competent mice; however, combination treatment demonstrated durable, complete responses (CRs). Tumors did not re-establish in the same mice that exhibited durable CRs when re-challenged with tumor cell inoculum, demonstrating these mice developed adaptive anti-tumor immunity. In a GEM model, treatment with MRTX849 plus anti-PD-1 led to increased progression-free survival compared to either single agent alone. These data demonstrate KRAS inhibition reverses an immunosuppressive tumor microenvironment and sensitizes tumors to CIT through multiple mechanisms.
PMID: 33722854
ISSN: 1538-8514
CID: 4837732

ULK1 inhibition overcomes compromised antigen presentation and restores antitumor immunity in LKB1 mutant lung cancer

Deng, Jiehui; Thennavan, Aatish; Dolgalev, Igor; Chen, Ting; Li, Jie; Marzio, Antonio; Poirier, John T; Peng, David; Bulatovic, Mirna; Mukhopadhyay, Subhadip; Silver, Heather; Papadopoulos, Eleni; Pyon, Val; Thakurdin, Cassandra; Han, Han; Li, Fei; Li, Shuai; Ding, Hailin; Hu, Hai; Pan, Yuanwang; Weerasekara, Vajira; Jiang, Baishan; Wang, Eric S; Ahearn, Ian; Philips, Mark; Papagiannakopoulos, Thales; Tsirigos, Aristotelis; Rothenberg, Eli; Gainor, Justin; Freeman, Gordon J; Rudin, Charles M; Gray, Nathanael S; Hammerman, Peter S; Pagano, Michele; Heymach, John V; Perou, Charles M; Bardeesy, Nabeel; Wong, Kwok-Kin
PMCID:8205437
PMID: 34142094
ISSN: 2662-1347
CID: 4917722

SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling

Fedele, Carmine; Li, Shuai; Teng, Kai Wen; Foster, Connor J R; Peng, David; Ran, Hao; Mita, Paolo; Geer, Mitchell J; Hattori, Takamitsu; Koide, Akiko; Wang, Yubao; Tang, Kwan Ho; Leinwand, Joshua; Wang, Wei; Diskin, Brian; Deng, Jiehui; Chen, Ting; Dolgalev, Igor; Ozerdem, Ugur; Miller, George; Koide, Shohei; Wong, Kwok-Kin; Neel, Benjamin G
KRAS is the most frequently mutated human oncogene, and KRAS inhibition has been a longtime goal. Recently, inhibitors were developed that bind KRASG12C-GDP and react with Cys-12 (G12C-Is). Using new affinity reagents to monitor KRASG12C activation and inhibitor engagement, we found that an SHP2 inhibitor (SHP2-I) increases KRAS-GDP occupancy, enhancing G12C-I efficacy. The SHP2-I abrogated RTK feedback signaling and adaptive resistance to G12C-Is in vitro, in xenografts, and in syngeneic KRASG12C-mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). SHP2-I/G12C-I combination evoked favorable but tumor site-specific changes in the immune microenvironment, decreasing myeloid suppressor cells, increasing CD8+ T cells, and sensitizing tumors to PD-1 blockade. Experiments using cells expressing inhibitor-resistant SHP2 showed that SHP2 inhibition in PDAC cells is required for PDAC regression and remodeling of the immune microenvironment but revealed direct inhibitory effects on tumor angiogenesis and vascularity. Our results demonstrate that SHP2-I/G12C-I combinations confer a substantial survival benefit in PDAC and NSCLC and identify additional potential combination strategies.
PMID: 33045063
ISSN: 1540-9538
CID: 4632492

Serial single-cell profiling analysis of metastatic TNBC during Nab-paclitaxel and pembrolizumab treatment

Deng, Jiehui; Thennavan, Aatish; Shah, Suhagi; Bagdatlioglu, Ece; Klar, Natalie; Heguy, Adriana; Marier, Christian; Meyn, Peter; Zhang, Yutong; Labbe, Kristen; Almonte, Christina; Krogsgaard, Michelle; Perou, Charles M; Wong, Kwok-Kin; Adams, Sylvia
PURPOSE/OBJECTIVE:Immunotherapy has recently been shown to improve outcomes for advanced PD-L1-positive triple-negative breast cancer (TNBC) in the Impassion130 trial, leading to FDA approval of the first immune checkpoint inhibitor in combination with taxane chemotherapy. To further develop predictive biomarkers and improve therapeutic efficacy of the combination, interrogation of the tumor immune microenvironment before therapy as well as during each component of treatment is crucial. Here we use single-cell RNA sequencing (scRNA-seq) on tumor biopsies to assess immune cell changes from two patients with advanced TNBC treated in a prospective trial at predefined serial time points, before treatment, on taxane chemotherapy and on chemo-immunotherapy. METHODS:Both patients (one responder and one progressor) received the trial therapy, in cycle 1 nab-paclitaxel given as single agent, in cycle 2 nab-paclitaxel in combination with pembrolizumab. Tumor core biopsies were obtained at baseline, 3 weeks (after cycle 1, chemotherapy alone) and 6 weeks (after cycle 2, chemo-immunotherapy). Single-cell RNA sequencing (scRNA-seq) of both cancer cells and infiltrating immune cells isolated were performed from fresh tumor core biopsy specimens by 10 × chromium sequencing. RESULTS:). In contrast, tumors from the patient with rapid disease progression showed a prevalent and persistent myeloid compartment. CONCLUSIONS:Our study provides a deep cellular analysis of on-treatment changes during chemo-immunotherapy for advanced TNBC, demonstrating not only feasibility of single-cell analyses on serial tumor biopsies but also the heterogeneity of TNBC and differences in on-treatment changes in responder versus progressor.
PMID: 32949350
ISSN: 1573-7217
CID: 4605282

Targeting HER2 Exon 20-Mutant Lung Adenocarcinoma with a Novel Tyrosine Kinase Inhibitor, Mobocertinib [Meeting Abstract]

Han, H.; Li, S.; Chen, T.; Fitzgerald, M.; Liu, S.; Peng, C.; Tang, K.; Cao, S.; Chouitar, J.; Wu, J.; Peng, D.; Deng, J.; Gao, Z.; Baker, T.; Li, F.; Zhang, H.; Pan, Y.; Ding, H.; Hu, H.; Pyon, V.; Thakurdin, C.; Papadopoulos, E.; Tang, S.; Gonzalvez, F.; Chen, H.; Rivera, V.; Brake, R.; Vincent, S.; Wong, K.
ISI:000709606500163
ISSN: 1556-0864
CID: 5184702

Epigenetic CRISPR screens identify Npm1 as a therapeutic vulnerability in non-small cell lung cancer

Li, Fei; Ng, Wai-Lung; Luster, Troy A; Hu, Hai; Sviderskiy, Vladislav O; Dowling, Catríona M; Hollinshead, Kate E R; Zouitine, Paula; Zhang, Hua; Huang, Qingyuan; Ranieri, Michela; Wang, Wei; Fang, Zhaoyuan; Chen, Ting; Deng, Jiehui; Zhao, Kai; So, Hon-Cheong; Khodadadi-Jamayran, Alireza; Xu, Mousheng; Karatza, Angeliki; Pyon, Val; Li, Shuai; Pan, Yuanwang; Labbe, Kristen; Almonte, Christina; Poirier, John T; Miller, George; Possemato, Richard; Qi, Jun; Wong, Kwok-Kin
Despite advancements in treatment options, the overall cure and survival rates for non-small cell lung cancers (NSCLC) remain low. While small-molecule inhibitors of epigenetic regulators have recently emerged as promising cancer therapeutics, their application in patients with NSCLC is limited. To exploit epigenetic regulators as novel therapeutic targets in NSCLC, we performed pooled epigenome-wide CRISPR knockout screens in vitro and in vivo and identified the histone chaperone nucleophosmin 1 (NPM1) as a potential therapeutic target. Genetic ablation of Npm1 significantly attenuated tumor progression in vitro and in vivo. Furthermore, KRAS-mutant cancer cells were more addicted to NPM1 expression. Genetic ablation of Npm1 rewired the balance of metabolism in cancer cells from predominant aerobic glycolysis to oxidative phosphorylation and reduced the population of tumor-propagating cells. Overall, our results support NPM1 as a therapeutic vulnerability in NSCLC.
PMID: 32646968
ISSN: 1538-7445
CID: 4518022

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

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

Pulsatile MEK Inhibition Improves Anti-tumor Immunity and T Cell Function in Murine Kras Mutant Lung Cancer

Choi, Hyejin; Deng, Jiehui; Li, Shuai; Silk, Tarik; Dong, Lauren; Brea, Elliott J; Houghton, Sean; Redmond, David; Zhong, Hong; Boiarsky, Jonathan; Akbay, Esra A; Smith, Paul D; Merghoub, Taha; Wong, Kwok-Kin; Wolchok, Jedd D
KRAS is one of the driver oncogenes in non-small-cell lung cancer (NSCLC) but remains refractory to current modalities of targeted pathway inhibition, which include inhibiting downstream kinase MEK to circumvent KRAS activation. Here, we show that pulsatile, rather than continuous, treatment with MEK inhibitors (MEKis) maintains T cell activation and enables their proliferation. Two MEKis, selumetinib and trametinib, induce T cell activation with increased CTLA-4 expression and, to a lesser extent, PD-1 expression on T cells in vivo after cyclical pulsatile MEKi treatment. In addition, the pulsatile dosing schedule alone shows superior anti-tumor effects and delays the emergence of drug resistance. Furthermore, pulsatile MEKi treatment combined with CTLA-4 blockade prolongs survival in mice bearing tumors with mutant Kras. Our results set the foundation and show the importance of a combinatorial therapeutic strategy using pulsatile targeted therapy together with immunotherapy to optimally enhance tumor delay and promote long-term anti-tumor immunity.
PMID: 30995478
ISSN: 2211-1247
CID: 3810572