Try a new search

Format these results:

Searched for:

in-biosketch:true

person:wongk11

Total Results:

344


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

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

Noncanonical agonist PPARγ ligands modulate the response to DNA damage and sensitize cancer cells to cytotoxic chemotherapy

Khandekar, Melin J; Banks, Alexander S; Laznik-Bogoslavski, Dina; White, James P; Choi, Jang Hyun; Kazak, Lawrence; Lo, James C; Cohen, Paul; Wong, Kwok-Kin; Kamenecka, Theodore M; Griffin, Patrick R; Spiegelman, Bruce M
The peroxisome-proliferator receptor-γ (PPARγ) is expressed in multiple cancer types. Recently, our group has shown that PPARγ is phosphorylated on serine 273 (S273), which selectively modulates the transcriptional program controlled by this protein. PPARγ ligands, including thiazolidinediones (TZDs), block S273 phosphorylation. This activity is chemically separable from the canonical activation of the receptor by agonist ligands and, importantly, these noncanonical agonist ligands do not cause some of the known side effects of TZDs. Here, we show that phosphorylation of S273 of PPARγ occurs in cancer cells on exposure to DNA damaging agents. Blocking this phosphorylation genetically or pharmacologically increases accumulation of DNA damage, resulting in apoptotic cell death. A genetic signature of PPARγ phosphorylation is associated with worse outcomes in response to chemotherapy in human patients. Noncanonical agonist ligands sensitize lung cancer xenografts and genetically induced lung tumors to carboplatin therapy. Moreover, inhibition of this phosphorylation results in deregulation of p53 signaling, and biochemical studies show that PPARγ physically interacts with p53 in a manner dependent on S273 phosphorylation. These data implicate a role for PPARγ in modifying the p53 response to cytotoxic therapy, which can be modulated for therapeutic gain using these compounds.
PMCID:5776997
PMID: 29295932
ISSN: 1091-6490
CID: 2966522

Suppression of adaptive responses to targeted cancer therapy by transcriptional repression

Rusan, Maria; Li, Kapsok; Li, Yvonne; Christensen, Camilla L; Abraham, Brian J; Kwiatkowski, Nicholas; Buczkowski, Kevin A; Bockorny, Bruno; Chen, Ting; Li, Shuai; Rhee, Kevin; Zhang, Haikuo; Chen, Wankun; Terai, Hideki; Tavares, Tiffany; Leggett, Alan L; Li, Tianxia; Wang, Yichen; Zhang, Tinghu; Kim, Tae-Jung; Hong, Sook-Hee; Poudel-Neupane, Neermala; Silkes, Michael; Mudianto, Tenny; Tan, Li; Shimamura, Takeshi; Meyerson, Matthew; Bass, Adam J; Watanabe, Hideo; Gray, Nathanael S; Young, Richard A; Wong, Kwok-Kin; Hammerman, Peter S
Acquired drug resistance is a major factor limiting the effectiveness of targeted cancer therapies. Targeting tumors with kinase inhibitors induces complex adaptive programs that promote the persistence of a fraction of the original cell population, facilitating the eventual outgrowth of inhibitor-resistant tumor clones. We show that the addition of a newly identified CDK7/12 inhibitor, THZ1, to targeted therapy enhances cell killing and impedes the emergence of drug-resistant cell populations in diverse cellular and in vivo cancer models. We propose that targeted therapy induces a state of transcriptional dependency in a subpopulation of cells poised to become drug tolerant, which THZ1 can exploit by blocking dynamic transcriptional responses, remodeling of enhancers and key signalling outputs required for tumor cell survival in the setting of targeted therapy. These findings suggest that the addition of THZ1 to targeted therapies is a promising broad-based strategy to hinder the emergence of drug-resistant cancer cell populations.
PMCID:5819998
PMID: 29054992
ISSN: 2159-8290
CID: 2742982

Combination of EGFR antibody with PD-1 pathway inhibitors improves anti-tumor efficacy and enhances intra-tumor immune response in preclinical mouse tumor models [Meeting Abstract]

Kandaswamy, Veena; Forest, Amelie; Deroose, Marianne; Schaer, David A.; Chen, Ting; Liu, Shengwu; Surguladze, David; Yao, Yung-mae; Doman, Thompson; Hall, Gerald; Wong, Kwok-Kin; Kalos, Michael; Novosiadly, Ruslan D.
ISI:000468819501427
ISSN: 0008-5472
CID: 5381132

Poziotinib overcomes de novo resistance of HER2 exon 20 mutations in NSCLC and other cancers: Preclinical studies and initial clinical testing [Meeting Abstract]

Robichaux, Jacqulyne P.; Elamin, Yasir Y.; Tan, Zhi; Negrao, Marelo Vailati; Routbort, Mark; Roeck, Brent; Li, Shuai; Liu, Shengwu; Chen, Ting; Ahnert, Jordi Rodon; Diao, Lixia; Nilsson, Monique B.; Zhang, Shuxing; Yang, Zane; Wang, Jing; Meric-Bernstam, Funda; Wong, Kwok-Kin; Heymach, John V.
ISI:000468819503390
ISSN: 0008-5472
CID: 5381152

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

Interplay between Notch1 and Notch3 promotes EMT and tumor initiation in squamous cell carcinoma

Natsuizaka, Mitsuteru; Whelan, Kelly A; Kagawa, Shingo; Tanaka, Koji; Giroux, Veronique; Chandramouleeswaran, Prasanna M; Long, Apple; Sahu, Varun; Darling, Douglas S; Que, Jianwen; Yang, Yizeng; Katz, Jonathan P; Wileyto, E Paul; Basu, Devraj; Kita, Yoshiaki; Natsugoe, Shoji; Naganuma, Seiji; Klein-Szanto, Andres J; Diehl, J Alan; Bass, Adam J; Wong, Kwok-Kin; Rustgi, Anil K; Nakagawa, Hiroshi
Notch1 transactivates Notch3 to drive terminal differentiation in stratified squamous epithelia. Notch1 and other Notch receptor paralogs cooperate to act as a tumor suppressor in squamous cell carcinomas (SCCs). However, Notch1 can be stochastically activated to promote carcinogenesis in murine models of SCC. Activated form of Notch1 promotes xenograft tumor growth when expressed ectopically. Here, we demonstrate that Notch1 activation and epithelial-mesenchymal transition (EMT) are coupled to promote SCC tumor initiation in concert with transforming growth factor (TGF)-beta present in the tumor microenvironment. We find that TGFbeta activates the transcription factor ZEB1 to repress Notch3, thereby limiting terminal differentiation. Concurrently, TGFbeta drives Notch1-mediated EMT to generate tumor initiating cells characterized by high CD44 expression. Moreover, Notch1 is activated in a small subset of SCC cells at the invasive tumor front and predicts for poor prognosis of esophageal SCC, shedding light upon the tumor promoting oncogenic aspect of Notch1 in SCC.
PMCID:5700926
PMID: 29170450
ISSN: 2041-1723
CID: 2792082

Synergy of WEE1 and mTOR Inhibition in Mutant KRAS-Driven Lung Cancers

Hai, Josephine; Liu, Shengwu; Bufe, Lauren; Do, Khanh; Chen, Ting; Wang, Xiaoen; Ng, Christine; Li, Shuai; Tsao, Ming-Sound; Shapiro, Geoffrey I; Wong, Kwok-Kin
Purpose:KRAS-activating mutations are the most common oncogenic driver in non-small cell lung cancer (NSCLC), but efforts to directly target mutant KRAS have proved a formidable challenge. Therefore, multitargeted therapy may offer a plausible strategy to effectively treat KRAS-driven NSCLCs. Here, we evaluate the efficacy and mechanistic rationale for combining mTOR and WEE1 inhibition as a potential therapy for lung cancers harboring KRAS mutations.Experimental Design: We investigated the synergistic effect of combining mTOR and WEE1 inhibitors on cell viability, apoptosis, and DNA damage repair response using a panel of human KRAS-mutant and wild type NSCLC cell lines and patient-derived xenograft cell lines. Murine autochthonous and human transplant models were used to test the therapeutic efficacy and pharmacodynamic effects of dual treatment.Results: We demonstrate that combined inhibition of mTOR and WEE1 induced potent synergistic cytotoxic effects selectively in KRAS-mutant NSCLC cell lines, delayed human tumor xenograft growth and caused tumor regression in a murine lung adenocarcinoma model. Mechanistically, we show that inhibition of mTOR potentiates WEE1 inhibition by abrogating compensatory activation of DNA repair, exacerbating DNA damage in KRAS-mutant NSCLC, and that this effect is due in part to reduction in cyclin D1.Conclusions: These findings demonstrate that compromised DNA repair underlies the observed potent synergy of WEE1 and mTOR inhibition and support clinical evaluation of this dual therapy for patients with KRAS-mutant lung cancers. Clin Cancer Res; 23(22); 6993-7005. (c)2017 AACR.
PMCID:5690829
PMID: 28821559
ISSN: 1078-0432
CID: 2784892

STK11/LKB1 loss of function genomic alterations predict primary resistance to PD-1/PD-L1 axis blockade in KRAS-mutant NSCLC [Meeting Abstract]

Skoulidis, F; Albacker, L; Hellmann, M; Awad, M; Gainor, J; Goldberg, M; Schrock, A; Gay, L; Elvin, J; Ross, J; Rizvi, H; Carter, B; Erasmus, J; Halpenny, D; Plodkowski, A; Long, N; Nishino-Habatu, M; Denning, W; Rodriguez-Canales, J; Villalobos, P; Parra, Cuentas E; Sholl, L; Sauter, J; Elamin, Y; Zhang, J; Leonardi, G; Wong, K; Stephens, P J; Papadimitrakopoulou, V; Wistuba, I; Wolchok, J; Shaw, A; Janne, P; Rudin, C; Miller, V; Heymach, J
Background: The genomic landscape of primary resistance to PD-1 blockade in lung adenocarcinoma (LUAD) is largely unknown. We previously reported that co-mutations in STK11/LKB1 (KL) or TP53 (KP) define subgroups of KRAS-mutant LUAD with distinct therapeutic vulnerabilities and immune profiles. Here, we present updated data on the clinical efficacy of PD-1/PD-L1 inhibitors in co-mutation defined KRAS mutant and wild-type LUAD patients and examine the relationship between genetic alterations in individual genes, tumor cell PD-L1 expression and tumor mutational burden (TMB) using cohorts form the SU2C/ACS Lung Cancer Dream Team and Foundation Medicine (FM). Method: The cohorts included 924 LUAD with NGS (FM cohort) and 188 patients with KRAS non-squamous NSCLC (SU2C cohort) who received at least one cycle of PD-1/PD-L1 inhibitor therapy and had available molecular profiling. Tumor cell PD-L1 expression was tested using E1L3N IHC (SU2C) and the VENTANA PD-L1 (SP142) assay (FM). TMB was defined as previously described and was classified as high (TMB-H), intermediate (TMB-I) or low (TMB-L). Result: 188 immunotherapy-treated (83.5% nivolumab, 11.7% pembrolizumab, 4.8% anti-PD1/PD-L1 plus anti-CTLA-4) pts with KRASmutant NSCLC were included in the efficacy analysis. The ORR differed significantly between the KL (8.8%), KP (35.9%) and K-only subgroups (27.3%) (P=0.0011, Fisher's exact test). KL LUAC exhibited significantly shorter PFS (mPFS 1.8m vs 2.7m, HR=0.53, 95% CI 0.34- 0.84, P<0.001, log-rank test) and OS (mOS 6.8m vs 15.6m, HR 0.53, 95% CI 0.34 to 0.84, P=0.0072, log rank test) compared to KRASmutant NSCLC with wild-type STK11. Loss-of function (LOF) genetic alterations in STK11 were the only significantly enriched event in PDL1 negative, TMB-I/H compared to PD-L1 high positive (TPS>=50%), TMB-I/H tumors in the overall FMI cohort (Bonferroni adjusted P=2.38x10-4, Fisher's exact test) and among KRAS-mutant tumors (adjusted P=0.05, Fisher's exact test). Notably, PD-1 blockade demonstrated activity among 10 PD-L1-negative KP tumors, with 3 PRs and 4SDs recorded. In syngeneic isogenic murine models PD-1 blockade significantly inhibited the growth of Kras mutant tumors with wild-type LKB1 (K), but not those with LKB1 loss (KL), providing evidence that LKB1 loss can play a causative role in promoting PD-1 inhibitor resistance. Conclusion: Loss of function genomic alterations in STK11 represents a dominant driver of de novo resistance to PD-1/PDL1 blockade in KRAS-mutant NSCLC. In addition to tumor PD-L1 status and tumor mutational burden precision immunotherapy approaches should take into consideration the STK11 status of individual tumors
EMBASE:620147130
ISSN: 1556-1380
CID: 2926662