Try a new search

Format these results:

Searched for:

in-biosketch:true

person:wongk11

Total Results:

344


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

Targeting PKCδ as a Therapeutic Strategy against Heterogeneous Mechanisms of EGFR Inhibitor Resistance in EGFR-Mutant Lung Cancer

Lee, Pei-Chih; Fang, Yueh-Fu; Yamaguchi, Hirohito; Wang, Wei-Jan; Chen, Tse-Ching; Hong, Xuan; Ke, Baozhen; Xia, Weiya; Wei, Yongkun; Zha, Zhengyu; Wang, Yan; Kuo, Han-Pin; Wang, Chih-Wei; Tu, Chih-Yen; Chen, Chia-Hung; Huang, Wei-Chien; Chiang, Shu-Fen; Nie, Lei; Hou, Junwei; Chen, Chun-Te; Huo, Longfei; Yang, Wen-Hao; Deng, Rong; Nakai, Katsuya; Hsu, Yi-Hsin; Chang, Shih-Shin; Chiu, Tai-Jan; Tang, Jun; Zhang, Ran; Wang, Li; Fang, Bingliang; Chen, Ting; Wong, Kwok-Kin; Hsu, Jennifer L; Hung, Mien-Chie
Multiple mechanisms of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been identified in EGFR-mutant non-small cell lung cancer (NSCLC); however, recurrent resistance to EGFR TKIs due to the heterogeneous mechanisms underlying resistance within a single patient remains a major challenge in the clinic. Here, we report a role of nuclear protein kinase Cδ (PKCδ) as a common axis across multiple known TKI-resistance mechanisms. Specifically, we demonstrate that TKI-inactivated EGFR dimerizes with other membrane receptors implicated in TKI resistance to promote PKCδ nuclear translocation. Moreover, the level of nuclear PKCδ is associated with TKI response in patients. The combined inhibition of PKCδ and EGFR induces marked regression of resistant NSCLC tumors with EGFR mutations.
PMID: 30537515
ISSN: 1878-3686
CID: 3659292

Intron retention is a source of neoepitopes in cancer

Smart, Alicia C; Margolis, Claire A; Pimentel, Harold; He, Meng Xiao; Miao, Diana; Adeegbe, Dennis; Fugmann, Tim; Wong, Kwok-Kin; Van Allen, Eliezer M
We present an in silico approach to identifying neoepitopes derived from intron retention events in tumor transcriptomes. Using mass spectrometry immunopeptidome analysis, we show that retained intron neoepitopes are processed and presented on MHC I on the surface of cancer cell lines. RNA-derived neoepitopes should be considered for prospective personalized cancer vaccine development.
PMID: 30114007
ISSN: 1546-1696
CID: 3241392

RIP1 Kinase Drives Macrophage-Mediated Adaptive Immune Tolerance in Pancreatic Cancer

Wang, Wei; Marinis, Jill M; Beal, Allison M; Savadkar, Shivraj; Wu, Yue; Khan, Mohammed; Taunk, Pardeep S; Wu, Nan; Su, Wenyu; Wu, Jingjing; Ahsan, Aarif; Kurz, Emma; Chen, Ting; Yaboh, Inedouye; Li, Fei; Gutierrez, Johana; Diskin, Brian; Hundeyin, Mautin; Reilly, Michael; Lich, John D; Harris, Philip A; Mahajan, Mukesh K; Thorpe, James H; Nassau, Pamela; Mosley, Julie E; Leinwand, Joshua; Kochen Rossi, Juan A; Mishra, Ankita; Aykut, Berk; Glacken, Michael; Ochi, Atsuo; Verma, Narendra; Kim, Jacqueline I; Vasudevaraja, Varshini; Adeegbe, Dennis; Almonte, Christina; Bagdatlioglu, Ece; Cohen, Deirdre J; Wong, Kwok-Kin; Bertin, John; Miller, George
Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.
PMID: 30423296
ISSN: 1878-3686
CID: 3457042

Clinical implications of monitoring nivolumab immunokinetics in non-small cell lung cancer patients

Osa, Akio; Uenami, Takeshi; Koyama, Shohei; Fujimoto, Kosuke; Okuzaki, Daisuke; Takimoto, Takayuki; Hirata, Haruhiko; Yano, Yukihiro; Yokota, Soichiro; Kinehara, Yuhei; Naito, Yujiro; Otsuka, Tomoyuki; Kanazu, Masaki; Kuroyama, Muneyoshi; Hamaguchi, Masanari; Koba, Taro; Futami, Yu; Ishijima, Mikako; Suga, Yasuhiko; Akazawa, Yuki; Machiyama, Hirotomo; Iwahori, Kota; Takamatsu, Hyota; Nagatomo, Izumi; Takeda, Yoshito; Kida, Hiroshi; Akbay, Esra A; Hammerman, Peter S; Wong, Kwok-Kin; Dranoff, Glenn; Mori, Masahide; Kijima, Takashi; Kumanogoh, Atsushi
BACKGROUND:The PD-1-blocking antibody nivolumab persists in patients several weeks after the last infusion. However, no study has systematically evaluated the maximum duration that the antibody persists on T cells or the association between this duration and residual therapeutic efficacy or potential adverse events. METHODS:To define the duration of binding and residual efficacy of nivolumab after discontinuation, we developed a simplified strategy for T cell monitoring and used it to analyze T cells from peripheral blood from 11 non-small cell lung cancer patients previously treated with nivolumab. To determine the suitability of our method for other applications, we compared transcriptome profiles between nivolumab-bound and nivolumab-unbound CD8 T cells. We also applied T cell monitoring in 2 nivolumab-treated patients who developed progressive lung tumors during long-term follow-up. RESULTS:Prolonged nivolumab binding was detected more than 20 weeks after the last infusion, regardless of the total number of nivolumab infusions (2-15 doses) or type of subsequent treatment, in 9 of the 11 cases in which long-term monitoring was possible. Ki-67 positivity, a proliferation marker, in T cells decreased in patients with progressive disease. Transcriptome profiling identified the signals regulating activation of nivolumab-bound T cells, which may contribute to nivolumab resistance. In 2 patients who restarted nivolumab, T cell proliferation markers exhibited the opposite trend and correlated with clinical response. CONCLUSIONS:Although only a few samples were analyzed, our strategy of monitoring both nivolumab binding and Ki-67 in T cells might help determine residual efficacy under various types of concurrent or subsequent treatment. TRIAL REGISTRATION/BACKGROUND:University Hospital Medical Information Network Clinical Trials Registry, UMIN000024623. FUNDING/BACKGROUND:This work was supported by Japan Society for the Promotion of Science KAKENHI (JP17K16045, JP18H05282, and JP15K09220), Japan Agency for Medical Research and Development (JP17cm0106310, JP18cm0106335 and JP18cm059042), and Core Research for Evolutional Science and Technology (JPMJCR16G2).
PMID: 30282824
ISSN: 2379-3708
CID: 3329272

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

BET bromodomain inhibition cooperates with PD-1 blockade to facilitate antitumor response in Kras-mutant non-small cell lung cancer

Adeegbe, Dennis O; Liu, Shengwu; Hattersley, Maureen; Bowden, Michaela; Zhou, Chensheng W; Li, Shuai; Vlahos, Raven; Grondine, Michael; Dolgalev, Igor; Ivanova, Elena; Quinn, Max M; Gao, Peng; Hammerman, Peter S; Bradner, James E; Diehl, J Alan; Rustgi, Anil K; Bass, Adam J; Tsirigos, Aristotelis; Freeman, Gordon J; Chen, Huawei; Wong, Kwok-Kin
KRAS mutation is present in approximately 30% of human lung adenocarcinomas. Although recent advances in targeted therapy have shown great promise, effective targeting of KRAS remains elusive, and concurrent alterations in tumor suppressors render KRAS-mutant tumors even more resistant to existing therapies. Contributing to the refractoriness of KRAS-mutant tumors are immunosuppressive mechanisms, such as increased presence of suppressive regulatory T cells (Tregs) in tumors and elevated expression of the inhibitory receptor PD-1 on tumor-infiltrating T cells. Treatment with BET bromodomain inhibitors is beneficial for hematologic malignancies, and they have Treg-disruptive effects in a non-small cell lung cancer (NSCLC) model. Targeting PD-1 inhibitory signals through PD-1 antibody blockade also has substantial therapeutic impact in lung cancer, although these outcomes are limited to a minority of patients. We hypothesized that the BET bromodomain inhibitor JQ1 would synergize with PD-1 blockade to promote a robust antitumor response in lung cancer. In the present study, using Kras+/LSL-G12D; Trp53L/L (KP) mouse models of NSCLC, we identified cooperative effects between JQ1 and PD-1 antibody. The numbers of tumor-infiltrating Tregs were reduced and activation of tumor-infiltrating T cells, which had a T-helper type 1 (Th1) cytokine profile, was enhanced, underlying their improved effector function. Furthermore, lung tumor-bearing mice treated with this combination showed robust and long-lasting antitumor responses compared to either agent alone, culminating in substantial improvement in the overall survival of treated mice. Thus, combining BET bromodomain inhibition with immune checkpoint blockade offers a promising therapeutic approach for solid malignancies such as lung adenocarcinoma.
PMID: 30087114
ISSN: 2326-6074
CID: 3226582

Author Correction: Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition [Correction]

Wong, Gabrielle S; Zhou, Jin; Bin Liu, Jie; Wu, Zhong; Xu, Xinsen; Li, Tianxia; Xu, David; Schumacher, Steven E; Puschhof, Jens; McFarland, James; Zou, Charles; Dulak, Austin; Henderson, Les; Xu, Peng; O'Day, Emily; Rendak, Rachel; Liao, Wei-Li; Cecchi, Fabiola; Hembrough, Todd; Schwartz, Sarit; Szeto, Christopher; Rustgi, Anil K; Wong, Kwok-Kin; Diehl, J Alan; Jensen, Karin; Graziano, Francesco; Ruzzo, Annamaria; Fereshetian, Shaunt; Mertins, Philipp; Carr, Steven A; Beroukhim, Rameen; Nakamura, Kenichi; Oki, Eiji; Watanabe, Masayuki; Baba, Hideo; Imamura, Yu; Catenacci, Daniel; Bass, Adam J
In the Supplementary Information originally published with this article, a lane was missing in the β-actin blot in Supplementary Fig. 2. The lane has been added. The error has been corrected in the Supplementary Information associated with this article.
PMID: 30093730
ISSN: 1546-170x
CID: 3226732

False positive plasma genotyping due to clonal hematopoiesis

Hu, Yuebi; Ulrich, Bryan; Supplee, Julianna; Kuang, Yanan; Lizotte, Patrick H; Feeney, Nora; Guibert, Nicolas; Awad, Mark M; Wong, Kwok-Kin; Janne, Pasi A; Paweletz, Cloud Peter; Oxnard, Geoffrey R
PURPOSE/OBJECTIVE:mutations, PBC and tumor NGS were available for comparison, and 5 were present in PBC but absent in tumor, consistent with CH. CONCLUSIONS:mutations detected in cfDNA are derived from CH not tumor. Clinicians ordering plasma genotyping must be prepared for the possibility that mutations detected in plasma, particularly in genes mutated in CH, may not represent true tumor genotype. Efforts to use plasma genotyping for cancer detection may need paired PBC genotyping so that CH-derived mutations are not misdiagnosed as occult malignancy.
PMID: 29567812
ISSN: 1078-0432
CID: 3001522

Overcoming Resistance to Dual Innate Immune and MEK Inhibition Downstream of KRAS

Kitajima, Shunsuke; Asahina, Hajime; Chen, Ting; Guo, Sujuan; Quiceno, Laura Gutierrez; Cavanaugh, Jillian D; Merlino, Ashley A; Tange, Shoichiro; Terai, Hideki; Kim, Jong Wook; Wang, Xiaoen; Zhou, Shan; Xu, Man; Wang, Stephen; Zhu, Zehua; Thai, Tran C; Takahashi, Chiaki; Wang, Yujin; Neve, Richard; Stinson, Susanna; Tamayo, Pablo; Watanabe, Hideo; Kirschmeier, Paul T; Wong, Kwok-Kin; Barbie, David A
Despite extensive efforts, oncogenic KRAS remains resistant to targeted therapy. Combined downstream RAL-TBK1 and MEK inhibition induces only transient lung tumor shrinkage in KRAS-driven genetically engineered mouse models (GEMMs). Using the sensitive KRAS;LKB1 (KL) mutant background, we identify YAP1 upregulation and a therapy-induced secretome as mediators of acquired resistance. This program is reversible, associated with H3K27 promoter acetylation, and suppressed by BET inhibition, resensitizing resistant KL cells to TBK1/MEK inhibition. Constitutive YAP1 signaling promotes intrinsic resistance in KRAS;TP53 (KP) mutant lung cancer. Intermittent treatment with the BET inhibitor JQ1 thus overcomes resistance to combined pathway inhibition in KL and KP GEMMs. Using potent and selective TBK1 and BET inhibitors we further develop an effective therapeutic strategy with potential translatability to the clinic.
PMID: 30205046
ISSN: 1878-3686
CID: 3277732