Searched for: in-biosketch:true
person:wongk11
IL-6 mediates cross-talk between activated fibroblasts and tumor cells in the tumor microenvironment
Karakasheva, Tatiana A; Lin, Eric W; Tang, Qiaosi; Qiao, Edmund; Waldron, Todd J; Soni, Monica; Klein-Szanto, Andres J; Sahu, Varun; Basu, Devraj; Ohashi, Shinya; Baba, Kiichiro; Giaccone, Zachary T; Walker, Sarah R; Frank, David A; Wileyto, E Paul; Long, Qi; Dunagin, Margaret; Raj, Arjun; Diehl, J Alan; Wong, Kwok-Kin; Bass, Adam J; Rustgi, Anil K
The tumor microenvironment (TME) plays a major role in the pathogenesis of multiple cancer types, including upper-gastrointestinal (GI) cancers that currently lack effective therapeutic options. Cancer-associated fibroblasts (CAF) are an essential component of the TME, contributing to tumorigenesis by secreting growth factors, modifying the extracellular matrix, supporting angiogenesis, and suppressing anti-tumor immune responses. Through an unbiased approach, we have established that IL-6 mediates crosstalk between tumor cells and CAF not only by supporting tumor cell growth, but also by promoting fibroblast activation. As a result, IL-6 receptor (IL-6Rα) and downstream effectors offer opportunities for targeted therapy in upper-GI cancers. IL-6 loss suppressed tumorigenesis in physiologically relevant 3D organotypic and 3D tumoroid models and murine models of esophageal cancer. Tocilizumab, an anti-IL-6Rα antibody, suppressed tumor growth in vivo in part via inhibition of STAT3 and MEK/ERK signaling. Analysis of a pan-cancer TCGA dataset revealed an inverse correlation between IL-6 and IL-6Rα overexpression and patient survival. Therefore, we expanded evaluation of tocilizumab to head-and-neck squamous cell carcinoma patient-derived xenografts and gastric adenocarcinoma xenografts, demonstrating suppression of tumor growth and altered STAT3 and ERK1/2 gene signatures. We used small molecule inhibitors of STAT3 and MEK1/2 signaling to suppress tumorigenesis in the 3D organotypic model of esophageal cancer. We demonstrate that IL-6 is a major contributor to the dynamic crosstalk between tumor cells and CAF in the TME. Our findings provide a translational rationale for inhibition of IL-6Rα and downstream signaling pathways as a novel targeted therapy in oral-upper-GI cancers.
PMCID:6125177
PMID: 29976575
ISSN: 1538-7445
CID: 3186182
Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors
Miao, Diana; Margolis, Claire A; Vokes, Natalie I; Liu, David; Taylor-Weiner, Amaro; Wankowicz, Stephanie M; Adeegbe, Dennis; Keliher, Daniel; Schilling, Bastian; Tracy, Adam; Manos, Michael; Chau, Nicole G; Hanna, Glenn J; Polak, Paz; Rodig, Scott J; Signoretti, Sabina; Sholl, Lynette M; Engelman, Jeffrey A; Getz, Gad; Jänne, Pasi A; Haddad, Robert I; Choueiri, Toni K; Barbie, David A; Haq, Rizwan; Awad, Mark M; Schadendorf, Dirk; Hodi, F Stephen; Bellmunt, Joaquim; Wong, Kwok-Kin; Hammerman, Peter; Van Allen, Eliezer M
Tumor mutational burden correlates with response to immune checkpoint blockade in multiple solid tumors, although in microsatellite-stable tumors this association is of uncertain clinical utility. Here we uniformly analyzed whole-exome sequencing (WES) of 249 tumors and matched normal tissue from patients with clinically annotated outcomes to immune checkpoint therapy, including radiographic response, across multiple cancer types to examine additional tumor genomic features that contribute to selective response. Our analyses identified genomic correlates of response beyond mutational burden, including somatic events in individual driver genes, certain global mutational signatures, and specific HLA-restricted neoantigens. However, these features were often interrelated, highlighting the complexity of identifying genetic driver events that generate an immunoresponsive tumor environment. This study lays a path forward in analyzing large clinical cohorts in an integrated and multifaceted manner to enhance the ability to discover clinically meaningful predictive features of response to immune checkpoint blockade.
PMCID:6119118
PMID: 30150660
ISSN: 1546-1718
CID: 3257072
mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis
Choe, Junho; Lin, Shuibin; Zhang, Wencai; Liu, Qi; Wang, Longfei; Ramirez-Moya, Julia; Du, Peng; Kim, Wantae; Tang, Shaojun; Sliz, Piotr; Santisteban, Pilar; George, Rani E; Richards, William G; Wong, Kwok-Kin; Locker, Nicolas; Slack, Frank J; Gregory, Richard I
N6-methyladenosine (m6A) modification of mRNA is emerging as an important regulator of gene expression that affects different developmental and biological processes, and altered m6A homeostasis is linked to cancer1-5. m6A modification is catalysed by METTL3 and enriched in the 3' untranslated region of a large subset of mRNAs at sites close to the stop codon5. METTL3 can promote translation but the mechanism and relevance of this process remain unknown1. Here we show that METTL3 enhances translation only when tethered to reporter mRNA at sites close to the stop codon, supporting a mechanism of mRNA looping for ribosome recycling and translational control. Electron microscopy reveals the topology of individual polyribosomes with single METTL3 foci in close proximity to 5' cap-binding proteins. We identify a direct physical and functional interaction between METTL3 and the eukaryotic translation initiation factor 3 subunit h (eIF3h). METTL3 promotes translation of a large subset of oncogenic mRNAs-including bromodomain-containing protein 4-that is also m6A-modified in human primary lung tumours. The METTL3-eIF3h interaction is required for enhanced translation, formation of densely packed polyribosomes and oncogenic transformation. METTL3 depletion inhibits tumorigenicity and sensitizes lung cancer cells to BRD4 inhibition. These findings uncover a mechanism of translation control that is based on mRNA looping and identify METTL3-eIF3h as a potential therapeutic target for patients with cancer.
PMID: 30232453
ISSN: 1476-4687
CID: 3301792
Palbociclib resistance confers dependence on an FGFR-MAP kinase-mTOR-driven pathway in KRAS-mutant non-small cell lung cancer
Haines, Eric; Chen, Ting; Kommajosyula, Naveen; Chen, Zhao; Herter-Sprie, Grit S; Cornell, Liam; Wong, Kwok-Kin; Shapiro, Geoffrey I
CDK4 is emerging as a target in KRAS-mutant non-small cell lung cancer (NSCLC). We demonstrate that KRAS-mutant NSCLC cell lines are initially sensitive to the CDK4/6 inhibitor palbociclib, but readily acquire resistance associated with increased expression of CDK6, D-type cyclins and cyclin E. Resistant cells also demonstrated increased ERK1/2 activity and sensitivity to MEK and ERK inhibitors. Moreover, MEK inhibition reduced the expression and activity of cell cycle proteins mediating palbociclib resistance. In resistant cells, ERK activated mTOR, driven in part by upstream FGFR1 signaling resulting from the extracellular secretion of FGF ligands. A genetically-engineered mouse model of KRAS-mutant NSCLC initially sensitive to palbociclib similarly developed acquired resistance with increased expression of cell cycle mediators, ERK1/2 and FGFR1. In this model, resistance was delayed with combined palbociclib and MEK inhibitor treatment. These findings implicate an FGFR1-MAP kinase-mTOR pathway resulting in increased expression of D-cyclins and CDK6 that confers palbociclib resistance and indicate that CDK4/6 inhibition acts to promote MAP kinase dependence.
PMCID:6114982
PMID: 30167080
ISSN: 1949-2553
CID: 3257152
EZH2-Mediated Primary Cilium Deconstruction Drives Metastatic Melanoma Formation
Zingg, Daniel; Debbache, Julien; Peña-Hernández, Rodrigo; Antunes, Ana T; Schaefer, Simon M; Cheng, Phil F; Zimmerli, Dario; Haeusel, Jessica; Calçada, Raquel R; Tuncer, Eylul; Zhang, Yudong; Bossart, Raphaël; Wong, Kwok-Kin; Basler, Konrad; Dummer, Reinhard; Santoro, Raffaella; Levesque, Mitchell P; Sommer, Lukas
Human melanomas frequently harbor amplifications of EZH2. However, the contribution of EZH2 to melanoma formation has remained elusive. Taking advantage of murine melanoma models, we show that EZH2 drives tumorigenesis from benign BrafV600E- or NrasQ61K-expressing melanocytes by silencing of genes relevant for the integrity of the primary cilium, a signaling organelle projecting from the surface of vertebrate cells. Consequently, gain of EZH2 promotes loss of primary cilia in benign melanocytic lesions. In contrast, blockade of EZH2 activity evokes ciliogenesis and cilia-dependent growth inhibition in malignant melanoma. Finally, we demonstrate that loss of cilia enhances pro-tumorigenic WNT/β-catenin signaling, and is itself sufficient to drive metastatic melanoma in benign cells. Thus, primary cilia deconstruction is a key process in EZH2-driven melanomagenesis.
PMID: 30008323
ISSN: 1878-3686
CID: 3193112
NK cells mediate synergistic antitumor effects of combined inhibition of HDAC6 and BET in a SCLC preclinical model
Liu, Yan; Li, Yuyang; Liu, Shengwu; Adeegbe, Dennis O; Christensen, Camilla L; Quinn, Max M; Dries, Ruben; Han, Shiwei; Buczkowski, Kevin; Wang, Xiaoen; Chen, Ting; Gao, Peng; Zhang, Hua; Li, Fei; Hammerman, Peter S; Bradner, James E; Quayle, Steven N; Wong, Kwok-Kin
Small cell lung cancer (SCLC) has the highest malignancy among all lung cancers, exhibiting aggressive growth and early metastasis to distant sites. For 30 years, treatment options for SCLC have been limited to chemotherapy, warranting the need for more effective treatments. Frequent inactivation of TP53 and RB1 as well as histone dysmodifications in SCLC suggest that transcriptional and epigenetic regulations play a major role in SCLC disease evolution. Here we performed a synthetic lethal screen using the BET inhibitor JQ1 and an shRNA library targeting 550 epigenetic genes in treatment-refractory SCLC xenograft models and identified HDAC6 as a synthetic lethal target in combination with JQ1. Combined treatment of human and mouse SCLC cell line-derived xenograft tumors with the HDAC6 inhibitor ricolinostat (ACY-1215) and JQ1 demonstrated significant inhibition of tumor growth; this effect was abolished upon depletion of NK cells, suggesting that these innate immune lymphoid cells play a role in SCLC tumor treatment response. Collectively, these findings suggest a potential new treatment for recurrent SCLC.
PMID: 29760044
ISSN: 1538-7445
CID: 3121342
Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition
Wong, Gabrielle S; Zhou, Jin; Liu, Jie Bin; 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
The role of KRAS, when activated through canonical mutations, has been well established in cancer 1 . Here we explore a secondary means of KRAS activation in cancer: focal high-level amplification of the KRAS gene in the absence of coding mutations. These amplifications occur most commonly in esophageal, gastric and ovarian adenocarcinomas2-4. KRAS-amplified gastric cancer models show marked overexpression of the KRAS protein and are insensitive to MAPK blockade owing to their capacity to adaptively respond by rapidly increasing KRAS-GTP levels. Here we demonstrate that inhibition of the guanine-exchange factors SOS1 and SOS2 or the protein tyrosine phosphatase SHP2 can attenuate this adaptive process and that targeting these factors, both genetically and pharmacologically, can enhance the sensitivity of KRAS-amplified models to MEK inhibition in both in vitro and in vivo settings. These data demonstrate the relevance of copy-number amplification as a mechanism of KRAS activation, and uncover the therapeutic potential for targeting of these tumors through combined SHP2 and MEK inhibition.
PMCID:6039276
PMID: 29808010
ISSN: 1546-170x
CID: 3136832
STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma
Skoulidis, Ferdinandos; Goldberg, Michael E; Greenawalt, Danielle M; Hellmann, Matthew D; Awad, Mark M; Gainor, Justin F; Schrock, Alexa B; Hartmaier, Ryan J; Trabucco, Sally E; Gay, Laurie; Ali, Siraj M; Elvin, Julia A; Singal, Gaurav; Ross, Jeffrey S; Fabrizio, David; Szabo, Peter M; Chang, Han; Sasson, Ariella; Srinivasan, Sujaya; Kirov, Stefan; Szustakowski, Joseph; Vitazka, Patrik; Edwards, Robin; Bufill, Jose A; Sharma, Neelesh; Ou, Sai-Hong I; Peled, Nir; Spigel, David R; Rizvi, Hira; Jimenez Aguilar, Elizabeth; Carter, Brett W; Erasmus, Jeremy; Halpenny, Darragh F; Plodkowski, Andrew J; Long, Niamh M; Nishino, Mizuki; Denning, Warren L; Galan-Cobo, Ana; Hamdi, Haifa; Hirz, Taghreed; Tong, Pan; Wang, Jing; Rodriguez-Canales, Jaime; Villalobos, Pamela A; Parra, Edwin R; Kalhor, Neda; Sholl, Lynette M; Sauter, Jennifer L; Jungbluth, Achim A; Mino-Kenudson, Mari; Azimi, Roxana; Elamin, Yasir Y; Zhang, Jianjun; Leonardi, Giulia C; Jiang, Fei; Wong, Kwok-Kin; Lee, J Jack; Papadimitrakopoulou, Vassiliki A; Wistuba, Ignacio I; Miller, Vincent A; Frampton, Garrett M; Wolchok, Jedd D; Shaw, Alice T; Jänne, Pasi A; Stephens, Philip J; Rudin, Charles M; Geese, William J; Albacker, Lee A; Heymach, John V
KRAS is the most common oncogenic driver in lung adenocarcinoma (LUAC). We previously reported that STK11/LKB1 (KL) or TP53 (KP) co-mutations define distinct subgroups of KRAS-mutant LUAC. Here, we examine the efficacy of PD-1 inhibitors in these subgroups. Objective response rates to PD-1 blockade differed significantly among KL (7.4%), KP (35.7%), and K-only (28.6%) subgroups (P<0.001) in the SU2C cohort (174 patients) with KRAS-mutant LUAC and in patients treated with nivolumab in the CheckMate-057 phase 3 trial (0% vs 57.1% vs 18.2%, P=0.047). In the SU2C cohort, KL LUAC exhibited shorter progression-free (P<0.001) and overall survival (P=0.0015) compared to KRASMUT;STK11/LKB1WT LUAC. Among 924 LUAC, STK11/LKB1 alterations were the only marker significantly associated with PD-L1 negativity in TMBIntermediate/High LUAC. The impact of STK11/LKB1 alterations on clinical outcomes with PD-1/PD-L1 inhibitors extended to PD-L1-positive NSCLC. In Kras-mutant murine LUAC models, Stk11/Lkb1 loss promoted PD-1/PD-L1 inhibitor resistance, suggesting a causal role. Our results identify STK11/LKB1 alterations as a major driver of primary resistance to PD-1 blockade in KRAS-mutant LUAC.
PMCID:6030433
PMID: 29773717
ISSN: 2159-8290
CID: 3121522
Targeting the human MUC1-C oncoprotein with an antibody-drug conjugate
Panchamoorthy, Govind; Jin, Caining; Raina, Deepak; Bharti, Ajit; Yamamoto, Masaaki; Adeebge, Dennis; Zhao, Qing; Bronson, Roderick; Jiang, Shirley; Li, Linjing; Suzuki, Yozo; Tagde, Ashujit; Ghoroghchian, P Peter; Wong, Kwok-Kin; Kharbanda, Surender; Kufe, Donald
Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly overexpressed on the surface of diverse human carcinomas and is an attractive target for the development of mAb-based therapeutics. However, attempts at targeting the shed MUC1 N-terminal subunit have been unsuccessful. We report here the generation of mAb 3D1 against the nonshed oncogenic MUC1 C-terminal (MUC1-C) subunit. We show that mAb 3D1 binds with low nM affinity to the MUC1-C extracellular domain at the restricted α3 helix. mAb 3D1 reactivity is selective for MUC1-C-expressing human cancer cell lines and primary cancer cells. Internalization of mAb 3D1 into cancer cells further supported the conjugation of mAb 3D1 to monomethyl auristatin E (MMAE). The mAb 3D1-MMAE antibody-drug conjugate (ADC) (a) kills MUC1-C-positive cells in vitro, (b) is nontoxic in MUC1-transgenic (MUC1.Tg) mice, and (c) is active against human HCC827 lung tumor xenografts. Humanized mAb (humAb) 3D1 conjugated to MMAE also exhibited antitumor activity in (a) MUC1.Tg mice harboring syngeneic MC-38/MUC1 tumors, (b) nude mice bearing human ZR-75-1 breast tumors, and (c) NCG mice engrafted with a patient-derived triple-negative breast cancer. These findings and the absence of associated toxicities support clinical development of humAb 3D1-MMAE ADCs as a therapeutic for the many cancers with MUC1-C overexpression.
PMCID:6124453
PMID: 29925694
ISSN: 2379-3708
CID: 3158242
Targeting HER2 Aberrations in Non-Small Cell Lung Cancer with Osimertinib
Liu, Shengwu; Li, Shuai; Hai, Josephine; Wang, Xiaoen; Chen, Ting; Quinn, Max M; Gao, Peng; Zhang, Yanxi; Ji, Hongbin; Cross, Darren; Wong, Kwok-Kin
PURPOSE/OBJECTIVE:HER2 (or ERBB2) aberrations, including both amplification and mutations, have been classified as oncogenic drivers that contribute to 2-6 percent of lung adenocarcinomas. HER2 amplification is also an important mechanism for acquired resistance to EGFR tyrosine kinase inhibitors (TKIs). However, due to limited preclinical studies and clinical trials, currently there is still no available standard of care for lung cancer patients with HER2 aberrations. To fulfill the clinical need for targeting HER2 in non-small cell lung cancer (NSCLC) patients, we performed a comprehensive pre-clinical study to evaluate the efficacy of a third-generation TKI, osimertinib (AZD9291). Experimental Design:Three genetically modified mouse models (GEMMs) mimicking individual HER2 alterations in NSCLC were generated and osimertinib was tested for its efficacy against these HER2 aberrations in vivo. RESULTS:Osimertinib treatment showed robust efficacy in HER2wt overexpression and EGFR del19/HER2 models but not in HER2 exon 20 insertion tumors. Interestingly, we further identified that combined treatment with osimertinib and the BET inhibitor JQ1 significantly increased the response rate in HER2-mutant NSCLC while JQ1 single treatment did not show efficacy. CONCLUSIONS:Overall, our data indicated robust anti-tumor efficacy of osimertinib against multiple HER2 aberrations in lung cancer, either as a single agent or in combination with JQ1. Our study provides a strong rationale for future clinical trials using osimertinib either alone or in combination with epigenetic drugs to target aberrant HER2 in NSCLC patients.
PMID: 29298799
ISSN: 1078-0432
CID: 2899562