Searched for: in-biosketch:yes
person:wongk11
Inhibitor-sensitive FGFR2 and FGFR3 mutations in lung squamous cell carcinoma [Case Report]
Liao, Rachel G; Jung, Joonil; Tchaicha, Jeremy; Wilkerson, Matthew D; Sivachenko, Andrey; Beauchamp, Ellen M; Liu, Qingsong; Pugh, Trevor J; Pedamallu, Chandra Sekhar; Hayes, D Neil; Gray, Nathanael S; Getz, Gad; Wong, Kwok-Kin; Haddad, Robert I; Meyerson, Matthew; Hammerman, Peter S
A comprehensive description of genomic alterations in lung squamous cell carcinoma (lung SCC) has recently been reported, enabling the identification of genomic events that contribute to the oncogenesis of this disease. In lung SCC, one of the most frequently altered receptor tyrosine kinase families is the fibroblast growth factor receptor (FGFR) family, with amplification or mutation observed in all four family members. Here, we describe the oncogenic nature of mutations observed in FGFR2 and FGFR3, each of which are observed in 3% of samples, for a mutation rate of 6% across both genes. Using cell culture and xenograft models, we show that several of these mutations drive cellular transformation. Transformation can be reversed by small-molecule FGFR inhibitors currently being developed for clinical use. We also show that mutations in the extracellular domains of FGFR2 lead to constitutive FGFR dimerization. In addition, we report a patient with an FGFR2-mutated oral SCC who responded to the multitargeted tyrosine kinase inhibitor pazopanib. These findings provide new insights into driving oncogenic events in a subset of lung squamous cancers, and recommend future clinical studies with FGFR inhibitors in patients with lung and head and neck SCC.
PMCID:3749739
PMID: 23786770
ISSN: 1538-7445
CID: 2269742
Metabolic and functional genomic studies identify deoxythymidylate kinase as a target in LKB1-mutant lung cancer
Liu, Yan; Marks, Kevin; Cowley, Glenn S; Carretero, Julian; Liu, Qingsong; Nieland, Thomas J F; Xu, Chunxiao; Cohoon, Travis J; Gao, Peng; Zhang, Yong; Chen, Zhao; Altabef, Abigail B; Tchaicha, Jeremy H; Wang, Xiaoxu; Choe, Sung; Driggers, Edward M; Zhang, Jianming; Bailey, Sean T; Sharpless, Norman E; Hayes, D Neil; Patel, Nirali M; Janne, Pasi A; Bardeesy, Nabeel; Engelman, Jeffrey A; Manning, Brendan D; Shaw, Reuben J; Asara, John M; Scully, Ralph; Kimmelman, Alec; Byers, Lauren A; Gibbons, Don L; Wistuba, Ignacio I; Heymach, John V; Kwiatkowski, David J; Kim, William Y; Kung, Andrew L; Gray, Nathanael S; Root, David E; Cantley, Lewis C; Wong, Kwok-Kin
The LKB1/STK11 tumor suppressor encodes a serine/threonine kinase, which coordinates cell growth, polarity, motility, and metabolism. In non-small cell lung carcinoma, LKB1 is somatically inactivated in 25% to 30% of cases, often concurrently with activating KRAS mutations. Here, we used an integrative approach to define novel therapeutic targets in KRAS-driven LKB1-mutant lung cancers. High-throughput RNA interference screens in lung cancer cell lines from genetically engineered mouse models driven by activated KRAS with or without coincident Lkb1 deletion led to the identification of Dtymk, encoding deoxythymidylate kinase (DTYMK), which catalyzes dTTP biosynthesis, as synthetically lethal with Lkb1 deficiency in mouse and human lung cancer lines. Global metabolite profiling showed that Lkb1-null cells had a striking decrease in multiple nucleotide metabolites as compared with the Lkb1-wild-type cells. Thus, LKB1-mutant lung cancers have deficits in nucleotide metabolism that confer hypersensitivity to DTYMK inhibition, suggesting that DTYMK is a potential therapeutic target in this aggressive subset of tumors.
PMCID:3753578
PMID: 23715154
ISSN: 2159-8290
CID: 1844122
Acquired substrate preference for GAB1 protein bestows transforming activity to ERBB2 kinase lung cancer mutants
Fan, Ying-Xin; Wong, Lily; Marino, Michael P; Ou, Wu; Shen, Yi; Wu, Wen Jin; Wong, Kwok-Kin; Reiser, Jakob; Johnson, Gibbes R
Activating mutations in the alphaC-beta4 loop of the ERBB2 kinase domain, such as ERBB2(YVMA) and ERBB2(G776VC), have been identified in human lung cancers and found to drive tumor formation. Here we observe that the docking protein GAB1 is hyper-phosphorylated in carcinomas from transgenic mice and in cell lines expressing these ERBB2 cancer mutants. Using dominant negative GAB1 mutants lacking canonical tyrosine residues for SHP2 and PI3K interactions or lentiviral shRNA that targets GAB1, we demonstrate that GAB1 phosphorylation is required for ERBB2 mutant-induced cell signaling, cell transformation, and tumorigenesis. An enzyme kinetic analysis comparing ERBB2(YVMA) to wild type using physiologically relevant peptide substrates reveals that ERBB2(YVMA) kinase adopts a striking preference for GAB1 phosphorylation sites as evidenced by approximately 150-fold increases in the specificity constants (kcat/Km) for several GAB1 peptides, and this change in substrate selectivity was predominantly attributed to the peptide binding affinities as reflected by the apparent Km values. Furthermore, we demonstrate that ERBB2(YVMA) phosphorylates GAB1 protein approximately 70-fold faster than wild type ERBB2 in vitro. Notably, the mutation does not significantly alter the Km for ATP or sensitivity to lapatinib, suggesting that, unlike EGFR lung cancer mutants, the ATP binding cleft of the kinase is not significantly changed. Taken together, our results indicate that the acquired substrate preference for GAB1 is critical for the ERBB2 mutant-induced oncogenesis.
PMCID:3675622
PMID: 23612964
ISSN: 1083-351x
CID: 2269772
A multicenter phase II study of ganetespib monotherapy in patients with genotypically defined advanced non-small cell lung cancer
Socinski, Mark A; Goldman, Jonathan; El-Hariry, Iman; Koczywas, Marianna; Vukovic, Vojo; Horn, Leora; Paschold, Eugene; Salgia, Ravi; West, Howard; Sequist, Lecia V; Bonomi, Philip; Brahmer, Julie; Chen, Lin-Chi; Sandler, Alan; Belani, Chandra P; Webb, Timothy; Harper, Harry; Huberman, Mark; Ramalingam, Suresh; Wong, Kwok-Kin; Teofilovici, Florentina; Guo, Wei; Shapiro, Geoffrey I
PURPOSE: Ganetespib is a novel inhibitor of the heat shock protein 90 (Hsp90), a chaperone protein critical to tumor growth and proliferation. In this phase II study, we evaluated the activity and tolerability of ganetespib in previously treated patients with non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Patients were enrolled into cohort A (mutant EGFR), B (mutant KRAS), or C (no EGFR or KRAS mutations). Patients were treated with 200 mg/m(2) ganetespib by intravenous infusion once weekly for 3 weeks followed by 1 week of rest, until disease progression. The primary endpoint was progression-free survival (PFS) at 16 weeks. Secondary endpoints included objective response (ORR), duration of treatment, tolerability, median PFS, overall survival (OS), and correlative studies. RESULTS: Ninety-nine patients with a median of 2 prior systemic therapies were enrolled; 98 were assigned to cohort A (n = 15), B (n = 17), or C (n = 66), with PFS rates at 16 weeks of 13.3%, 5.9%, and 19.7%, respectively. Four patients (4%) achieved partial response (PR); all had disease that harbored anaplastic lymphoma kinase (ALK) gene rearrangement, retrospectively detected by FISH (n = 1) or PCR-based assays (n = 3), in crizotinib-naive patients enrolled to cohort C. Eight patients (8.1%) experienced treatment-related serious adverse events (AE); 2 of these (cardiac arrest and renal failure) resulted in death. The most common AEs were diarrhea, fatigue, nausea, and anorexia. CONCLUSIONS: Ganetespib monotherapy showed a manageable side effect profile as well as clinical activity in heavily pretreated patients with advanced NSCLCs, particularly in patients with tumors harboring ALK gene rearrangement.
PMCID:3874465
PMID: 23553849
ISSN: 1078-0432
CID: 2269802
EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation
Beguelin, Wendy; Popovic, Relja; Teater, Matt; Jiang, Yanwen; Bunting, Karen L; Rosen, Monica; Shen, Hao; Yang, Shao Ning; Wang, Ling; Ezponda, Teresa; Martinez-Garcia, Eva; Zhang, Haikuo; Zheng, Yupeng; Verma, Sharad K; McCabe, Michael T; Ott, Heidi M; Van Aller, Glenn S; Kruger, Ryan G; Liu, Yan; McHugh, Charles F; Scott, David W; Chung, Young Rock; Kelleher, Neil; Shaknovich, Rita; Creasy, Caretha L; Gascoyne, Randy D; Wong, Kwok-Kin; Cerchietti, Leandro; Levine, Ross L; Abdel-Wahab, Omar; Licht, Jonathan D; Elemento, Olivier; Melnick, Ari M
The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting.
PMCID:3681809
PMID: 23680150
ISSN: 1878-3686
CID: 2269752
HIF1alpha and HIF2alpha independently activate SRC to promote melanoma metastases
Hanna, Sara C; Krishnan, Bhavani; Bailey, Sean T; Moschos, Stergios J; Kuan, Pei-Fen; Shimamura, Takeshi; Osborne, Lukas D; Siegel, Marni B; Duncan, Lyn M; O'Brien, E Tim 3rd; Superfine, Richard; Miller, C Ryan; Simon, M Celeste; Wong, Kwok-Kin; Kim, William Y
Malignant melanoma is characterized by a propensity for early lymphatic and hematogenous spread. The hypoxia-inducible factor (HIF) family of transcription factors is upregulated in melanoma by key oncogenic drivers. HIFs promote the activation of genes involved in cancer initiation, progression, and metastases. Hypoxia has been shown to enhance the invasiveness and metastatic potential of tumor cells by regulating the genes involved in the breakdown of the ECM as well as genes that control motility and adhesion of tumor cells. Using a Pten-deficient, Braf-mutant genetically engineered mouse model of melanoma, we demonstrated that inactivation of HIF1alpha or HIF2alpha abrogates metastasis without affecting primary tumor formation. HIF1alpha and HIF2alpha drive melanoma invasion and invadopodia formation through PDGFRalpha and focal adhesion kinase-mediated (FAK-mediated) activation of SRC and by coordinating ECM degradation via MT1-MMP and MMP2 expression. These results establish the importance of HIFs in melanoma progression and demonstrate that HIF1alpha and HIF2alpha activate independent transcriptional programs that promote metastasis by coordinately regulating cell invasion and ECM remodeling.
PMCID:3635738
PMID: 23563312
ISSN: 1558-8238
CID: 2269792
The pivotal role of IKKalpha in the development of spontaneous lung squamous cell carcinomas
Xiao, Zuoxiang; Jiang, Qun; Willette-Brown, Jami; Xi, Sichuan; Zhu, Feng; Burkett, Sandra; Back, Timothy; Song, Na-Young; Datla, Mahesh; Sun, Zhonghe; Goldszmid, Romina; Lin, Fanching; Cohoon, Travis; Pike, Kristen; Wu, Xiaolin; Schrump, David S; Wong, Kwok-Kin; Young, Howard A; Trinchieri, Giorgio; Wiltrout, Robert H; Hu, Yinling
Here, we report that kinase-dead IKKalpha knockin mice develop spontaneous lung squamous cell carcinomas (SCCs) associated with IKKalpha downregulation and marked pulmonary inflammation. IKKalpha reduction upregulated the expression of p63, Trim29, and keratin 5 (K5), which serve as diagnostic markers for human lung SCCs. IKKalpha(low)K5(+)p63(hi) cell expansion and SCC formation were accompanied by inflammation-associated deregulation of oncogenes, tumor suppressors, and stem cell regulators. Reintroducing transgenic K5.IKKalpha, depleting macrophages, and reconstituting irradiated mutant animals with wild-type bone marrow (BM) prevented SCC development, suggesting that BM-derived IKKalpha mutant macrophages promote the transition of IKKalpha(low)K5(+)p63(hi) cells to tumor cells. This mouse model resembles human lung SCCs, sheds light on the mechanisms underlying lung malignancy development, and identifies targets for therapy of lung SCCs.
PMCID:3649010
PMID: 23597566
ISSN: 1878-3686
CID: 2269782
Characterization of Torin2, an ATP-competitive inhibitor of mTOR, ATM, and ATR
Liu, Qingsong; Xu, Chunxiao; Kirubakaran, Sivapriya; Zhang, Xin; Hur, Wooyoung; Liu, Yan; Kwiatkowski, Nicholas P; Wang, Jinhua; Westover, Kenneth D; Gao, Peng; Ercan, Dalia; Niepel, Mario; Thoreen, Carson C; Kang, Seong A; Patricelli, Matthew P; Wang, Yuchuan; Tupper, Tanya; Altabef, Abigail; Kawamura, Hidemasa; Held, Kathryn D; Chou, Danny M; Elledge, Stephen J; Janne, Pasi A; Wong, Kwok-Kin; Sabatini, David M; Gray, Nathanael S
mTOR is a highly conserved serine/threonine protein kinase that serves as a central regulator of cell growth, survival, and autophagy. Deregulation of the PI3K/Akt/mTOR signaling pathway occurs commonly in cancer and numerous inhibitors targeting the ATP-binding site of these kinases are currently undergoing clinical evaluation. Here, we report the characterization of Torin2, a second-generation ATP-competitive inhibitor that is potent and selective for mTOR with a superior pharmacokinetic profile to previous inhibitors. Torin2 inhibited mTORC1-dependent T389 phosphorylation on S6K (RPS6KB1) with an EC(50) of 250 pmol/L with approximately 800-fold selectivity for cellular mTOR versus phosphoinositide 3-kinase (PI3K). Torin2 also exhibited potent biochemical and cellular activity against phosphatidylinositol-3 kinase-like kinase (PIKK) family kinases including ATM (EC(50), 28 nmol/L), ATR (EC(50), 35 nmol/L), and DNA-PK (EC(50), 118 nmol/L; PRKDC), the inhibition of which sensitized cells to Irradiation. Similar to the earlier generation compound Torin1 and in contrast to other reported mTOR inhibitors, Torin2 inhibited mTOR kinase and mTORC1 signaling activities in a sustained manner suggestive of a slow dissociation from the kinase. Cancer cell treatment with Torin2 for 24 hours resulted in a prolonged block in negative feedback and consequent T308 phosphorylation on Akt. These effects were associated with strong growth inhibition in vitro. Single-agent treatment with Torin2 in vivo did not yield significant efficacy against KRAS-driven lung tumors, but the combination of Torin2 with mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor AZD6244 yielded a significant growth inhibition. Taken together, our findings establish Torin2 as a strong candidate for clinical evaluation in a broad number of oncologic settings where mTOR signaling has a pathogenic role.
PMCID:3760004
PMID: 23436801
ISSN: 1538-7445
CID: 2269812
Combined use of ALK immunohistochemistry and FISH for optimal detection of ALK-rearranged lung adenocarcinomas [Case Report]
Sholl, Lynette M; Weremowicz, Stanislawa; Gray, Stacy W; Wong, Kwok-Kin; Chirieac, Lucian R; Lindeman, Neal I; Hornick, Jason L
INTRODUCTION: ALK gene rearrangements occur in approximately 5% of lung adenocarcinomas (ACAs), leading to anaplastic lymphoma kinase (ALK) overexpression and predicting response to targeted therapy. Fluorescence in situ hybridization (FISH) is the standard procedure for detection of ALK rearrangements in lung ACA but requires specialized equipment and expertise. Immunohistochemistry (IHC) for ALK protein overexpression is a promising screening modality, with reports of newer antibodies showing excellent sensitivity and specificity for ALK-rearranged lung ACA. METHODS: In this study, we analyzed ALK IHC (5A4 clone) in 186 cases from our clinical service and compared it with ALK FISH and EGFR and KRAS mutation status. RESULTS: Twelve cases had concordant ALK protein overexpression and ALK rearrangement by FISH. Three ALK-rearranged cases lacked ALK protein expression. Of these discrepant cases, one had a coexisting EGFR mutation and a subtle atypical ALK rearrangement manifested as a break in the 5' centromeric portion of the FISH probe. One case had a concurrent BRAF mutation. Follow-up testing on a metastasis revealed absence of the ALK rearrangement, with persistent BRAF mutation. In one ALK-rearranged protein negative case, very limited tissue remained for ALK IHC, raising the possibility of false negativity because of protein expression heterogeneity. Importantly, ALK protein expression was detected in one case initially thought not to have an ALK rearrangement. In this case, FISH was falsely negative because of interference by benign reactive nuclei. After correcting for these cases, ALK IHC was 93% sensitive and 100% specific as compared with FISH. CONCLUSIONS: ALK IHC improves the detection of ALK rearrangements when used together with FISH, and its use in lung ACA genetic testing algorithms should be considered.
PMCID:3573350
PMID: 23407557
ISSN: 1556-1380
CID: 2269822
KDM2B promotes pancreatic cancer via Polycomb-dependent and -independent transcriptional programs
Tzatsos, Alexandros; Paskaleva, Polina; Ferrari, Francesco; Deshpande, Vikram; Stoykova, Svetlana; Contino, Gianmarco; Wong, Kwok-Kin; Lan, Fei; Trojer, Patrick; Park, Peter J; Bardeesy, Nabeel
Epigenetic mechanisms mediate heritable control of cell identity in normal cells and cancer. We sought to identify epigenetic regulators driving the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), one of the most lethal human cancers. We found that KDM2B (also known as Ndy1, FBXL10, and JHDM1B), an H3K36 histone demethylase implicated in bypass of cellular senescence and somatic cell reprogramming, is markedly overexpressed in human PDAC, with levels increasing with disease grade and stage, and highest expression in metastases. KDM2B silencing abrogated tumorigenicity of PDAC cell lines exhibiting loss of epithelial differentiation, whereas KDM2B overexpression cooperated with KrasG12D to promote PDAC formation in mouse models. Gain- and loss-of-function experiments coupled to genome-wide gene expression and ChIP studies revealed that KDM2B drives tumorigenicity through 2 different transcriptional mechanisms. KDM2B repressed developmental genes through cobinding with Polycomb group (PcG) proteins at transcriptional start sites, whereas it activated a module of metabolic genes, including mediators of protein synthesis and mitochondrial function, cobound by the MYC oncogene and the histone demethylase KDM5A. These results defined epigenetic programs through which KDM2B subverts cellular differentiation and drives the pathogenesis of an aggressive subset of PDAC.
PMCID:3561797
PMID: 23321669
ISSN: 1558-8238
CID: 2269832