Try a new search

Format these results:

Searched for:

person:dengj06

in-biosketch:yes

Total Results:

48


Pulsatile MEK inhibition improves antitumor immunity and T-cell function in Kras mutant lung cancer [Meeting Abstract]

Choi, H; Deng, J; Li, S; Silk, T; Brea, E J; Boiarsky, J; Akbay, E A; Smith, P D; Merghoub, T D; Wong, K -K; Wolchok, J D
KRAS is one of the most commonly identified driver oncogene in non-small cell lung cancer (NSCLC) and is commonly associated to NSCLC that is refractory to current available modalities of treatment. Targeted therapy to inhibit MEK has shown promising tumor growth control, but is followed by quick rebound of tumor growth. Recently, cancer immunotherapy has shown great promise by activating T-cells that are suppressed in the tumor microenvironment, especially through targeting co-inhibitory molecules and their counterparts. We sought to identify the most effective therapy for the treatment of KRAS mutant NSCLC patients by targeting cancer cells and activating immune infiltrates at the same time, by studying the impact of MEK inhibition on the immune microenvironment. We utilized Kras mutant lung cancer cell lines and Kras mutant transgenic lung cancer animals. We found that pulsatile treatment of MEK inhibitors activates T-cells and releases their proliferation suppression ex vivo and in vivo. Both selumetinib and trametinib showed highly increased CTLA-4 expression and mild increase of PD-1 in T-cells in pulsatile treatment, compared to continuous treatment in vivo. In addition, the pulsatile schedule alone showed superior antitumor effect and delayed drug resistance, in contrast with continuous dosing schedule. Based on the above observations, we combined pulsatile MEK inhibitor treatment and anti-CTLA-4 and found that it showed most prolonged survival of Kras tumor-bearing mice. All together, our findings will set the foundation for a combinatorial therapeutic strategy using pulsatile targeted therapy together with immunotherapy in patients, to optimally enhance tumor apoptosis and promote long-term immune response simultaneously
EMBASE:626516972
ISSN: 2326-6074
CID: 3729892

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

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

Autophagy sustains pancreatic cancer growth through both cell autonomous and non-autonomous mechanisms

Yang, Annan; Herter-Sprie, Grit; Zhang, Haikuo; Lin, Elaine Y; Biancur, Douglas; Wang, Xiaoxu; Deng, Jiehui; Hai, Josephine; Yang, Shenghong; Wong, Kwok-Kin; Kimmelman, Alec C
Autophagy has been shown to be elevated in pancreatic adenocarcinoma (PDAC) and its role in promoting established tumor growth has made it a promising therapeutic target. However, due to limitations of prior mouse models as well as the lack of potent and selective autophagy inhibitors, the ability to fully assess the mechanistic basis of how autophagy supports pancreatic cancer has been limited. To test the feasibility of treating PDAC using autophagy inhibition and further our understanding of the mechanisms of pro-tumor effects of autophagy, we developed a novel mouse model that allowed the acute and reversible inhibition of autophagy. We observed that autophagy inhibition causes significant tumor regression in an autochthonous mouse model of PDAC. A detailed analysis of these effects indicated that the tumor regression was likely multifactorial, involving both tumor cell intrinsic as well as host effects. Thus, our study supports autophagy inhibition in PDAC may have future utility in the treatment of pancreatic cancer and illustrates the importance of assessing complex biological processes in relevant autochthonous models.
PMCID:5835190
PMID: 29317452
ISSN: 2159-8290
CID: 2906432

Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids

Jenkins, Russell W; Aref, Amir R; Lizotte, Patrick H; Ivanova, Elena; Stinson, Susanna; Zhou, Chensheng W; Bowden, Michaela; Deng, Jiehui; Liu, Hongye; Miao, Diana; He, Meng Xiao; Walker, William; Zhang, Gao; Tian, Tian; Cheng, Chaoran; Wei, Zhi; Palakurthi, Sangeetha; Bittinger, Mark; Vitzthum, Hans; Kim, Jong Wook; Merlino, Ashley; Quinn, Max; Venkataramani, Chandrasekar; Kaplan, Joshua A; Portell, Andrew; Gokhale, Prafulla C; Phillips, Bart; Smart, Alicia; Rotem, Asaf; Jones, Robert E; Keogh, Lauren; Anguiano, Maria; Stapleton, Lance; Jia, Zhiheng; Barzily-Rokni, Michal; Cañadas, Israel; Thai, Tran C; Hammond, Marc R; Vlahos, Raven; Wang, Eric S; Zhang, Hua; Li, Shuai; Hanna, Glenn J; Huang, Wei; Hoang, Mai P; Piris, Adriano; Eliane, Jean-Pierre; Stemmer-Rachamimov, Anat O; Cameron, Lisa; Su, Mei-Ju; Shah, Parin; Izar, Benjamin; Thakuria, Manisha; LeBoeuf, Nicole R; Rabinowits, Guilherme; Gunda, Viswanath; Parangi, Sareh; Cleary, James M; Miller, Brian C; Kitajima, Shunsuke; Thummalapalli, Rohit; Miao, Benchun; Barbie, Thanh U; Sivathanu, Vivek; Wong, Joshua; Richards, William G; Bueno, Raphael; Yoon, Charles H; Miret, Juan; Herlyn, Meenhard; Garraway, Levi A; Van Allen, Eliezer M; Freeman, Gordon J; Kirschmeier, Paul T; Lorch, Jochen H; Ott, Patrick A; Hodi, F Stephen; Flaherty, Keith T; Kamm, Roger D; Boland, Genevieve M; Wong, Kwok-Kin; Dornan, David; Paweletz, Cloud Peter; Barbie, David A
Ex vivo systems that incorporate features of the tumor microenvironment (TME) and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations, and respond to ICB in short-term 3-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo. Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.
PMCID:5809290
PMID: 29101162
ISSN: 2159-8290
CID: 2916402

CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation

Deng, Jiehui; Wang, Eric S; Jenkins, Russell W; Li, Shuai; Dries, Ruben; Yates, Kathleen; Chhabra, Sandeep; Huang, Wei; Liu, Hongye; Aref, Amir R; Ivanova, Elena; Paweletz, Cloud P; Bowden, Michaela; Zhou, Chensheng W; Herter-Sprie, Grit S; Sorrentino, Jessica A; Bisi, John E; Lizotte, Patrick H; Merlino, Ashley A; Quinn, Max M; Bufe, Lauren E; Yang, Annan; Zhang, Yanxi; Zhang, Hua; Gao, Peng; Chen, Ting; Cavanaugh, Megan E; Rode, Amanda J; Haines, Eric; Roberts, Patrick J; Strum, Jay C; Richards, William G; Lorch, Jochen H; Parangi, Sareh; Gunda, Viswanath; Boland, Genevieve M; Bueno, Raphael; Palakurthi, Sangeetha; Freeman, Gordon J; Ritz, Jerome; Haining, W Nicholas; Sharpless, Norman E; Arthanari, Haribabu; Shapiro, Geoffrey I; Barbie, David A; Gray, Nathanael S; Wong, Kwok-Kin
Immune checkpoint blockade, exemplified by antibodies targeting the PD-1 receptor, can induce durable tumor regressions in some patients. To enhance the efficacy of existing immunotherapies, we screened for small molecules capable of increasing the activity of T cells suppressed by PD-1. Here, we show that short-term exposure to small-molecule inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) significantly enhances T-cell activation, contributing to antitumor effectsin vivo, due in part to the derepression of NFAT family proteins and their target genes, critical regulators of T-cell function. Although CDK4/6 inhibitors decrease T-cell proliferation, they increase tumor infiltration and activation of effector T cells. Moreover, CDK4/6 inhibition augments the response to PD-1 blockade in a novelex vivoorganotypic tumor spheroid culture system and in multiplein vivomurine syngeneic models, thereby providing a rationale for combining CDK4/6 inhibitors and immunotherapies.Significance:Our results define previously unrecognized immunomodulatory functions of CDK4/6 and suggest that combining CDK4/6 inhibitors with immune checkpoint blockade may increase treatment efficacy in patients. Furthermore, our study highlights the critical importance of identifying complementary strategies to improve the efficacy of immunotherapy for patients with cancer.Cancer Discov; 8(2); 216-33. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Jenkins et al., p. 196This article is highlighted in the In This Issue feature, p. 127.
PMCID:5809273
PMID: 29101163
ISSN: 2159-8290
CID: 2957522

Assessing efficacy of neratinib in HER2-driven lung cancer [Meeting Abstract]

Li, Shuai; Liu, Shengwu; Chen, Ting; Deng, Jiehui; Wu, Min; Kuraguchi, Mari; Cam Anh Tran; Kirschmeier, Paul T.; Avogadri-Connors, Francesca; Cutler, Richard E.; Lalani, Alshad S.; Wong, Kwok-Kin
ISI:000468819503394
ISSN: 0008-5472
CID: 5381122

Optimizing targeted therapy and immune checkpoint blockade therapy in Kras mutant lung cancer [Meeting Abstract]

Choi, Hyejin; Deng, Jiehui; Silk, Tarik; Powers, Ann; Boiarsky, Jonathan; Merghoub, Taha; Wong, Kwok-Kin; Wolchok, Jedd
ISI:000460207200047
ISSN: 2051-1426
CID: 4511802

Pre-clinical study using KRAS mutant mouse models for lung cancer immunotherapy together with MEK inhibition [Meeting Abstract]

Deng, Jiehui; Li, Shuai; Herter-Sprie, Grit; Smith, Paul A; Freeman, Gordon J; Engelman, Jeffrey A; Hammerman, Peter; Wong, Kwok-Kin
ISI:000389969804088
ISSN: 1538-7445
CID: 2589832

Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer

Herter-Sprie, Grit S; Koyama, Shohei; Korideck, Houari; Hai, Josephine; Deng, Jiehui; Li, Yvonne Y; Buczkowski, Kevin A; Grant, Aaron K; Ullas, Soumya; Rhee, Kevin; Cavanaugh, Jillian D; Neupane, Neermala Poudel; Christensen, Camilla L; Herter, Jan M; Makrigiorgos, G Mike; Hodi, F Stephen; Freeman, Gordon J; Dranoff, Glenn; Hammerman, Peter S; Kimmelman, Alec C; Wong, Kwok-Kin
Radiation therapy (RT), a critical modality in the treatment of lung cancer, induces direct tumor cell death and augments tumor-specific immunity. However, despite initial tumor control, most patients suffer from locoregional relapse and/or metastatic disease following RT. The use of immunotherapy in non-small-cell lung cancer (NSCLC) could potentially change this outcome by enhancing the effects of RT. Here, we report significant (up to 70% volume reduction of the target lesion) and durable (up to 12 weeks) tumor regressions in conditional Kras-driven genetically engineered mouse models (GEMMs) of NSCLC treated with radiotherapy and a programmed cell death 1 antibody (alphaPD-1). However, while alphaPD-1 therapy was beneficial when combined with RT in radiation-naive tumors, alphaPD-1 therapy had no antineoplastic efficacy in RT-relapsed tumors and further induced T cell inhibitory markers in this setting. Furthermore, there was differential efficacy of alphaPD-1 plus RT among Kras-driven GEMMs, with additional loss of the tumor suppressor serine/threonine kinase 11/liver kinase B1 (Stk11/Lkb1) resulting in no synergistic efficacy. Taken together, our data provide evidence for a close interaction among RT, T cells, and the PD-1/PD-L1 axis and underscore the rationale for clinical combinatorial therapy with immune modulators and radiotherapy.
PMCID:5033933
PMID: 27699275
ISSN: 2379-3708
CID: 2523722