Try a new search

Format these results:

Searched for:

in-biosketch:true

person:wongk11

Total Results:

352


Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell lung cancer

Shimamura, Takeshi; Chen, Zhao; Soucheray, Margaret; Carretero, Julian; Kikuchi, Eiki; Tchaicha, Jeremy H; Gao, Yandi; Cheng, Katherine A; Cohoon, Travis J; Qi, Jun; Akbay, Esra; Kimmelman, Alec C; Kung, Andrew L; Bradner, James E; Wong, Kwok-Kin
PURPOSE: Amplification of MYC is one of the most common genetic alterations in lung cancer, contributing to a myriad of phenotypes associated with growth, invasion, and drug resistance. Murine genetics has established both the centrality of somatic alterations of Kras in lung cancer, as well as the dependency of mutant Kras tumors on MYC function. Unfortunately, drug-like small-molecule inhibitors of KRAS and MYC have yet to be realized. The recent discovery, in hematologic malignancies, that bromodomain and extra-terminal (BET) bromodomain inhibition impairs MYC expression and MYC transcriptional function established the rationale of targeting KRAS-driven non-small cell lung cancer (NSCLC) with BET inhibition. EXPERIMENTAL DESIGN: We performed functional assays to evaluate the effects of JQ1 in genetically defined NSCLC cell lines harboring KRAS and/or LKB1 mutations. Furthermore, we evaluated JQ1 in transgenic mouse lung cancer models expressing mutant kras or concurrent mutant kras and lkb1. Effects of bromodomain inhibition on transcriptional pathways were explored and validated by expression analysis. RESULTS: Although JQ1 is broadly active in NSCLC cells, activity of JQ1 in mutant KRAS NSCLC is abrogated by concurrent alteration or genetic knockdown of LKB1. In sensitive NSCLC models, JQ1 treatment results in the coordinate downregulation of the MYC-dependent transcriptional program. We found that JQ1 treatment produces significant tumor regression in mutant kras mice. As predicted, tumors from mutant kras and lkb1 mice did not respond to JQ1. CONCLUSION: Bromodomain inhibition comprises a promising therapeutic strategy for KRAS-mutant NSCLC with wild-type LKB1, via inhibition of MYC function. Clinical studies of BET bromodomain inhibitors in aggressive NSCLC will be actively pursued. Clin Cancer Res; 19(22); 6183-92. (c)2013 AACR.
PMCID:3838895
PMID: 24045185
ISSN: 1078-0432
CID: 1844022

Cetuximab response of lung cancer-derived EGF receptor mutants is associated with asymmetric dimerization

Cho, Jeonghee; Chen, Liang; Sangji, Naveen; Okabe, Takafumi; Yonesaka, Kimio; Francis, Joshua M; Flavin, Richard J; Johnson, William; Kwon, Jihyun; Yu, Soyoung; Greulich, Heidi; Johnson, Bruce E; Eck, Michael J; Janne, Pasi A; Wong, Kwok-Kin; Meyerson, Matthew
Kinase domain mutations of the EGF receptor (EGFR) are common oncogenic events in lung adenocarcinoma. Here, we explore the dependency upon asymmetric dimerization of the kinase domain for activation of lung cancer-derived EGFR mutants. We show that whereas wild-type EGFR and the L858R mutant require dimerization for activation and oncogenic transformation, the exon 19 deletion, exon 20 insertion, and L858R/T790M EGFR mutants do not require dimerization. In addition, treatment with the monoclonal antibody, cetuximab, shrinks mouse lung tumors induced by the dimerization-dependent L858R mutant, but exerts only a modest effect on tumors driven by dimerization-independent EGFR mutants. These data imply that different EGFR mutants show differential requirements for dimerization and that disruption of dimerization may be among the antitumor mechanisms of cetuximab.
PMCID:3903789
PMID: 24063894
ISSN: 1538-7445
CID: 2269712

Integrative radiogenomic profiling of squamous cell lung cancer

Abazeed, Mohamed E; Adams, Drew J; Hurov, Kristen E; Tamayo, Pablo; Creighton, Chad J; Sonkin, Dmitriy; Giacomelli, Andrew O; Du, Charles; Fries, Daniel F; Wong, Kwok-Kin; Mesirov, Jill P; Loeffler, Jay S; Schreiber, Stuart L; Hammerman, Peter S; Meyerson, Matthew
Radiotherapy is one of the mainstays of anticancer treatment, but the relationship between the radiosensitivity of cancer cells and their genomic characteristics is still not well defined. Here, we report the development of a high-throughput platform for measuring radiation survival in vitro and its validation in comparison with conventional clonogenic radiation survival analysis. We combined results from this high-throughput assay with genomic parameters in cell lines from squamous cell lung carcinoma, which is standardly treated by radiotherapy, to identify parameters that predict radiation sensitivity. We showed that activation of NFE2L2, a frequent event in lung squamous cancers, confers radiation resistance. An expression-based, in silico screen nominated inhibitors of phosphoinositide 3-kinase (PI3K) as NFE2L2 antagonists. We showed that the selective PI3K inhibitor, NVP-BKM120, both decreased NRF2 protein levels and sensitized NFE2L2 or KEAP1-mutant cells to radiation. We then combined results from this high-throughput assay with single-sample gene set enrichment analysis of gene expression data. The resulting analysis identified pathways implicated in cell survival, genotoxic stress, detoxification, and innate and adaptive immunity as key correlates of radiation sensitivity. The integrative and high-throughput methods shown here for large-scale profiling of radiation survival and genomic features of solid-tumor-derived cell lines should facilitate tumor radiogenomics and the discovery of genotype-selective radiation sensitizers and protective agents.
PMCID:3856255
PMID: 23980093
ISSN: 1538-7445
CID: 2269732

New cast for a new era: preclinical cancer drug development revisited

Herter-Sprie, Grit S; Kung, Andrew L; Wong, Kwok-Kin
Molecularly targeted agents promise to revolutionize therapeutics by reducing morbidity and mortality in patients with cancer. However, despite an urgent need for more effective anticancer compounds, current preclinical drug evaluations largely fail to satisfy the demand. New preclinical strategies, including the improvement of sophisticated mouse models and co-clinical study designs, are being used to augment the predictive value of animal-based translational cancer research. Here, we review the development of successful preclinical antineoplastic agents, their associated limitations, and alternative methods to predict clinical outcomes.
PMCID:3754257
PMID: 23999436
ISSN: 1558-8238
CID: 2269722

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