Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Emily Chan is active.

Publication


Featured researches published by Emily Chan.


Nature | 2012

Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors

Timothy R. Wilson; Jane Fridlyand; Yibing Yan; Elicia Penuel; Luciana Burton; Emily Chan; Jing Peng; Eva Lin; Yulei Wang; Jeffrey A. Sosman; Antoni Ribas; Jiang Li; John Moffat; Daniel P. Sutherlin; Hartmut Koeppen; Mark Merchant; Richard M. Neve; Jeffrey Settleman

Mutationally activated kinases define a clinically validated class of targets for cancer drug therapy. However, the efficacy of kinase inhibitors in patients whose tumours harbour such alleles is invariably limited by innate or acquired drug resistance. The identification of resistance mechanisms has revealed a recurrent theme—the engagement of survival signals redundant to those transduced by the targeted kinase. Cancer cells typically express multiple receptor tyrosine kinases (RTKs) that mediate signals that converge on common critical downstream cell-survival effectors—most notably, phosphatidylinositol-3-OH kinase (PI(3)K) and mitogen-activated protein kinase (MAPK). Consequently, an increase in RTK-ligand levels, through autocrine tumour-cell production, paracrine contribution from tumour stroma or systemic production, could confer resistance to inhibitors of an oncogenic kinase with a similar signalling output. Here, using a panel of kinase-‘addicted’ human cancer cell lines, we found that most cells can be rescued from drug sensitivity by simply exposing them to one or more RTK ligands. Among the findings with clinical implications was the observation that hepatocyte growth factor (HGF) confers resistance to the BRAF inhibitor PLX4032 (vemurafenib) in BRAF-mutant melanoma cells. These observations highlight the extensive redundancy of RTK-transduced signalling in cancer cells and the potentially broad role of widely expressed RTK ligands in innate and acquired resistance to drugs targeting oncogenic kinases.


Cancer Cell | 2013

Oncogenic ERBB3 Mutations in Human Cancers

Bijay S. Jaiswal; Noelyn M. Kljavin; Eric Stawiski; Emily Chan; Chaitali Parikh; Steffen Durinck; Subhra Chaudhuri; Kanan Pujara; Joseph Guillory; Kyle A. Edgar; Vasantharajan Janakiraman; Rolf-Peter Scholz; Krista K. Bowman; Maria N. Lorenzo; Hong Li; Jiansheng Wu; Wenlin Yuan; Brock A. Peters; Zhengyan Kan; Jeremy Stinson; Michelle Mak; Zora Modrusan; Charles Eigenbrot; Ron Firestein; Howard M. Stern; Krishnaraj Rajalingam; Gabriele Schaefer; Mark Merchant; Mark X. Sliwkowski; Frederic J. de Sauvage

The human epidermal growth factor receptor (HER) family of tyrosine kinases is deregulated in multiple cancers either through amplification, overexpression, or mutation. ERBB3/HER3, the only member with an impaired kinase domain, although amplified or overexpressed in some cancers, has not been reported to carry oncogenic mutations. Here, we report the identification of ERBB3 somatic mutations in ~11% of colon and gastric cancers. We found that the ERBB3 mutants transformed colonic and breast epithelial cells in a ligand-independent manner. However, the mutant ERBB3 oncogenic activity was dependent on kinase-active ERBB2. Furthermore, we found that anti-ERBB antibodies and small molecule inhibitors effectively blocked mutant ERBB3-mediated oncogenic signaling and disease progression in vivo.


Cancer Research | 2013

A Novel Tankyrase Small-Molecule Inhibitor Suppresses APC Mutation–Driven Colorectal Tumor Growth

Ted Lau; Emily Chan; Marinella Callow; Jo Waaler; Jason Boggs; Robert A. Blake; Steven Magnuson; Amy Sambrone; Melissa Schutten; Ron Firestein; Ondrej Machon; Vladimir Korinek; Edna F. Choo; Mark Merchant; Paul Polakis; Daniel Holsworth; Stefan Krauss; Mike Costa

Most colorectal cancers (CRC) are initiated by mutations of APC, leading to increased β-catenin-mediated signaling. However, continued requirement of Wnt/β-catenin signaling for tumor progression in the context of acquired KRAS and other mutations is less well-established. To attenuate Wnt/β-catenin signaling in tumors, we have developed potent and specific small-molecule tankyrase inhibitors, G007-LK and G244-LM, that reduce Wnt/β-catenin signaling by preventing poly(ADP-ribosyl)ation-dependent AXIN degradation, thereby promoting β-catenin destabilization. We show that novel tankyrase inhibitors completely block ligand-driven Wnt/β-catenin signaling in cell culture and display approximately 50% inhibition of APC mutation-driven signaling in most CRC cell lines. It was previously unknown whether the level of AXIN protein stabilization by tankyrase inhibition is sufficient to impact tumor growth in the absence of normal APC activity. Compound G007-LK displays favorable pharmacokinetic properties and inhibits in vivo tumor growth in a subset of APC-mutant CRC xenograft models. In the xenograft model most sensitive to tankyrase inhibitor, COLO-320DM, G007-LK inhibits cell-cycle progression, reduces colony formation, and induces differentiation, suggesting that β-catenin-dependent maintenance of an undifferentiated state may be blocked by tankyrase inhibition. The full potential of the antitumor activity of G007-LK may be limited by intestinal toxicity associated with inhibition of Wnt/β-catenin signaling and cell proliferation in intestinal crypts. These results establish proof-of-concept antitumor efficacy for tankyrase inhibitors in APC-mutant CRC models and uncover potential diagnostic and safety concerns to be overcome as tankyrase inhibitors are advanced into the clinic.


Cancer Discovery | 2013

Noncovalent Wild-type–Sparing Inhibitors of EGFR T790M

Ho-June Lee; Gabriele Schaefer; Timothy P. Heffron; Lily Shao; Xiaofen Ye; Steve Sideris; Shiva Malek; Emily Chan; Mark Merchant; Hank La; Savita Ubhayakar; Robert L. Yauch; Valentina Pirazzoli; Katerina Politi; Jeffrey Settleman

UNLABELLEDnApproximately half of EGFR-mutant non-small cell lung cancer (NSCLC) patients treated with small-molecule EGFR kinase inhibitors develop drug resistance associated with the EGF receptor (EGFR) T790M gatekeeper substitution, prompting efforts to develop covalent EGFR inhibitors, which can effectively suppress EGFR T790M in preclinical models. However, these inhibitors have yet to prove clinically efficacious, and their toxicity in skin, reflecting activity against wild-type EGFR, may limit dosing required to effectively suppress EGFR T790M in vivo. While profiling sensitivity to various kinase inhibitors across a large cancer cell line panel, we identified indolocarbazole compounds, including a clinically well-tolerated FLT3 inhibitor, as potent and reversible inhibitors of EGFR T790M that spare wild-type EGFR. These findings show the use of broad cancer cell profiling of kinase inhibitor efficacy to identify unanticipated novel applications, and they identify indolocarbazole compounds as potentially effective EGFR inhibitors in the context of T790M-mediated drug resistance in NSCLC.nnnSIGNIFICANCEnEGFR-mutant lung cancer patients who respond to currently used EGFR kinase inhibitors invariably develop drug resistance, which is associated with the EGFR T790M resistance mutation in about half these cases. We unexpectedly identified a class of reversible potent inhibitors of EGFR T790M that do not inhibit wild-type EGFR, revealing a promising therapeutic strategy to overcome T790M-associated drug-resistant lung cancers.


Cancer Research | 2012

FGFR3 Stimulates Stearoyl CoA Desaturase 1 Activity to Promote Bladder Tumor Growth

Xiangnan Du; Qian-Rena Wang; Emily Chan; Mark Merchant; Jinfeng Liu; Dorothy French; Avi Ashkenazi; Jing Qing

Fibroblast growth factor receptor 3 (FGFR3) belongs to a family of receptor tyrosine kinases that control cell proliferation, differentiation, and survival. Aberrant activation of FGFR3 via overexpression or mutation is a frequent feature of bladder cancer; however, its molecular and cellular consequences and functional relevance to carcinogenesis are not well understood. Through transcriptional profiling of bladder carcinoma cells subjected to short hairpin RNA knockdown of FGFR3, we identified a gene-signature linking FGFR3 signaling with de novo sterol and lipid biosynthesis and metabolism. We found that FGFR3 signaling promotes the cleavage and activation of the master transcriptional regulator of lipogenesis, sterol regulatory element-binding protein 1(SREBP1/SREBF1), in a PI3K-mTORC1-dependent fashion. In turn, SREBP1 regulates the expression of key lipogenic enzymes, including stearoyl CoA desaturase 1 (SCD1/SCD). SCD1 is the rate-limiting enzyme in the biosynthesis of monounsaturated fatty acids and is crucial for lipid homeostasis. In human bladder cancer cell lines expressing constitutively active FGFR3, knockdown of SCD1 by siRNA markedly attenuated cell-cycle progression, reduced proliferation, and induced apoptosis. Furthermore, inducible knockdown of SCD1 in a bladder cancer xenograft model substantially inhibited tumor progression. Pharmacologic inhibition of SCD1 blocked fatty acid desaturation and also exerted antitumor activity in vitro and in vivo. Together, these findings reveal a previously unrecognized role of FGFR3 in regulating lipid metabolism to maintain tumor growth and survival, and also identify SCD1 as a potential therapeutic target for FGFR3-driven bladder cancer.


Molecular Cancer Therapeutics | 2015

Modeling Targeted Inhibition of MEK and PI3 Kinase in Human Pancreatic Cancer

Melissa R. Junttila; Vidusha Devasthali; Jason H. Cheng; Joseph Castillo; Ciara Metcalfe; Anne C Clermont; Douglas Den Otter; Emily Chan; Hani Bou-Reslan; Tim C. Cao; William F. Forrest; Michelle Nannini; Dorothy French; Richard A. D. Carano; Mark Merchant; Klaus P. Hoeflich; Mallika Singh

Activating mutations in the KRAS oncogene occur in approximately 90% of pancreatic cancers, resulting in aberrant activation of the MAPK and the PI3K pathways, driving malignant progression. Significant efforts to develop targeted inhibitors of nodes within these pathways are underway and several are currently in clinical trials for patients with KRAS-mutant tumors, including patients with pancreatic cancer. To model MEK and PI3K inhibition in late-stage pancreatic cancer, we conducted preclinical trials with a mutant Kras-driven genetically engineered mouse model that faithfully recapitulates human pancreatic ductal adenocarcinoma development. Treatment of advanced disease with either a MEK (GDC-0973) or PI3K inhibitor (GDC-0941) alone showed modest tumor growth inhibition and did not significantly enhance overall survival. However, combination of the two agents resulted in a significant survival advantage as compared with control tumor-bearing mice. To model the clinical scenario, we also evaluated the combination of these targeted agents with gemcitabine, the current standard-of-care chemotherapy for pancreatic cancer. The addition of MEK or PI3K inhibition to gemcitabine, or the triple combination regimen, incrementally enhanced overall survival as compared with gemcitabine alone. These results are reminiscent of the survival advantage conferred in this model and in patients by the combination of gemcitabine and erlotinib, an approved therapeutic regimen for advanced nonresectable pancreatic cancer. Taken together, these data indicate that inhibition of MEK and PI3K alone or in combination with chemotherapy do not confer a dramatic improvement as compared with currently available therapies for patients with pancreatic cancer. Mol Cancer Ther; 14(1); 40–47. ©2014 AACR.


Cancer Research | 2017

Therapeutic Targeting of the CBP/p300 Bromodomain Blocks the Growth of Castration-Resistant Prostate Cancer

Lingyan Jin; Jesse Garcia; Emily Chan; Cecile de la Cruz; Ehud Segal; Mark Merchant; Samir Kharbanda; Ryan Raisner; Peter M. Haverty; Zora Modrusan; Justin Ly; Edna F. Choo; Susan Kaufman; Maureen Beresini; F. Anthony Romero; Steven Magnuson; Karen Gascoigne

Resistance invariably develops to antiandrogen therapies used to treat newly diagnosed prostate cancers, but effective treatments for castration-resistant disease remain elusive. Here, we report that the transcriptional coactivator CBP/p300 is required to maintain the growth of castration-resistant prostate cancer. To exploit this vulnerability, we developed a novel small-molecule inhibitor of the CBP/p300 bromodomain that blocks prostate cancer growth in vitro and in vivo Molecular dissection of the consequences of drug treatment revealed a critical role for CBP/p300 in histone acetylation required for the transcriptional activity of the androgen receptor and its target gene expression. Our findings offer a preclinical proof of concept for small-molecule therapies to target the CBP/p300 bromodomain as a strategy to treat castration-resistant prostate cancer. Cancer Res; 77(20); 5564-75. ©2017 AACR.


Molecular Cancer Therapeutics | 2017

Cotargeting of MEK and PDGFR/STAT3 Pathways to Treat Pancreatic Ductal Adenocarcinoma

Nisebita Sahu; Emily Chan; Felix Chu; Thinh Pham; Hartmut Koeppen; William F. Forrest; Mark Merchant; Jeffrey Settleman

Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal human diseases and remains largely refractory to available drug treatments. Insufficient targeting of the known oncogenic drivers and activation of compensatory feedback loops and inability to prevent metastatic spread contribute to poor prognosis for this disease. The KRAS-driven MEK pathway is mutationally activated in most pancreatic cancers and is an important target for therapeutics. Using a two-dimensional monolayer culture system as well as three-dimensional spheroid culture system, we conducted a screen of a large panel of anticancer agents and found that MAP2K (MEK) inhibitors were most effective in targeting PDAC spheroids in comparison with monolayer cultures. Combination treatment with an MEK inhibitor and the multikinase inhibitor ponatinib was effective in targeting pancreatic cancer cells both in monolayer and spheroids by effectively blocking signaling via the PDGFRα and MEK kinases, while also preventing the activation of STAT3- and S6-mediated compensatory feedback loops in cancer cells. Furthermore, using xenograft models, we demonstrate that cotreatment with a MEK inhibitor and ponatinib causes significant tumor regression. PDAC patient samples also provided evidence of increased STAT3 activation in PDAC tumors and MAPK1 (ERK) activation in liver metastases, implicating STAT3 and ERK as key drivers in primary tumors and metastases, respectively. These results reveal a combination drug treatment strategy that may be effective in pancreatic cancer. Mol Cancer Ther; 16(9); 1729–38. ©2017 AACR.


Cell Reports | 2018

Enhancer Activity Requires CBP/P300 Bromodomain-Dependent Histone H3K27 Acetylation

Ryan Raisner; Samir Kharbanda; Lingyan Jin; Edwin Jeng; Emily Chan; Mark Merchant; Peter M. Haverty; Russell Bainer; Tommy K. Cheung; David Arnott; E. Megan Flynn; F. Anthony Romero; Steven Magnuson; Karen Gascoigne

Acetylation of histone H3 at lysine 27 is a well-defined marker of enhancer activity. However, the functional impact of this modification at enhancers is poorly understood. Here, we use a chemical genetics approach to acutely block the function of the cAMP response element binding protein (CREB) bindingxa0protein (CBP)/P300 bromodomain in models of hematological malignancies and describe a consequent loss of H3K27Ac specifically from enhancers, despite the continued presence of CBP/P300 at chromatin. Using this approach to dissect the role of H3K27Ac at enhancers, we identify a critical role for this modification in the production of enhancer RNAs and transcription of enhancer-regulated gene networks.


Cancer Research | 2016

Abstract 4905: Effects of MAPK pathway inhibitors in the tumor immune microenvironment

Marcia Belvin; Erin Williams; Shiuh-Ming Luoh; Jeanne Cheung; Christine Orr; Emily Chan; Peter J.R. Ebert; Ira Mellman; Jeong Kim; Mark Merchant

Immunotherapeutic agents have shown great promise in the clinic in recent years and this has led to their approval as single agents or as immune doublet combinations in melanoma and lung cancer. In order to increase the extent of benefit from these agents and to extend immunotherapies to additional patients, combinations are being evaluated of immunotherapeutic agents with chemotherapy and targeted agents. Inhibitors of the mitogen-activated kinase protein kinase (MAPK) pathway, including BRAF and MEK inhibitors, have been approved in melanoma, and are being evaluated in additional indications. We evaluated the effects of MEK and ERK inhibitors on human T cells cultured in vitro as well as on the tumor microenvironment in preclinical syngeneic mouse models. In studies with human T cells in vitro, we find that MEK or ERK inhibition reduced proliferation of naive T cells, but had little effect on the proliferation of central memory cells, suggesting a differential requirement on MAPK signaling for proliferation of these T cell subsets. In vivo, MEK and ERK inhibition resulted in an increase in CD8+ infiltration in the tumor, as well as a decrease of markers of T cell exhaustion such as PD-1 and EOMES. The combination of MEK or ERK inhibitors plus anti-PD-L1 resulted in increased CD8+ effector function as measured by increased interferon gamma (IFNg) levels. In some cases, the MEK and ERK inhibitors showed differences in their pharmacodynamic effects as single agents; however, in combination with anti-PD-L1, the effects were similar. For example, single agent MEK inhibition but not single agent ERK inhibition increased the number of CD4+ helper and regulatory T cells (Tregs) in the tumor. However, both of these CD4+ subsets, as well as CD4+ IFNg levels, were increased with MEK or ERK inhibitors combined with anti-PD-L1. Similarly, single agent MEK inhibition decreased the number of infiltrating CD11b+Ly6G+ myeloid cells whereas single agent ERK inhibition increased the number of infiltrating CD11b+Ly6C+ myeloid cells. However, in combination with anti-PD-L1, these changes in myeloid populations were less apparent. Taken together, these data show that MAPK pathway suppression can modulate multiple immune cell subtypes within the tumor, with many of these changes expected to activate the immune infiltrate. Despite the increase in Tregs in response to ERK and MEK inhibition in combination with anti-PD-L1, these combinations were efficacious. This suggests that the other immune stimulatory effects of these treatments are sufficient to drive efficacy despite the presence of increased Tregs. Our data suggest that the combination of MAPK inhibition plus anti-PD-L1 inhibition, such as with atezolizumab, is a promising hypothesis to test in the clinic. Citation Format: Marcia P. Belvin, Erin Williams, Shiuh-Ming Luoh, Jeanne Cheung, Christine Orr, Emily Chan, Peter Ebert, Ira Mellman, Jeong Kim, Mark Merchant. Effects of MAPK pathway inhibitors in the tumor immune microenvironment. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4905.

Collaboration


Dive into the Emily Chan's collaboration.

Researchain Logo
Decentralizing Knowledge