Genetic Modifiers of EGFR Dependence in Non-Small Cell Lung Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Genetic Modifiers of EGFR Dependence in Non-Small Cell Lung Cancer Genetic modifiers of EGFR dependence in non-small cell lung cancer Tanaz Sharifniaa,b,c, Victor Rusua,d, Federica Piccionia, Mukta Bagula, Marcin Imielinskia,b,c, Andrew D. Cherniacka, Chandra Sekhar Pedamallua,c, Bang Wonga, Frederick H. Wilsona,c, Levi A. Garrawaya,c, David Altshulera,d,e, Todd R. Goluba, David E. Roota, Aravind Subramaniana, and Matthew Meyersona,b,c,1 aBroad Institute of Harvard and MIT, Cambridge, MA 02142; Departments of bPathology and dGenetics and Medicine, Harvard Medical School, Boston, MA 02115; cDepartment of Medical Oncology, Dana–Farber Cancer Institute, Boston, MA 02115; and eDepartment of Molecular Biology and Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114 Edited by Bert Vogelstein, Johns Hopkins University, Baltimore, MD, and approved November 18, 2014 (received for review June 29, 2014) Lung adenocarcinomas harboring activating mutations in the achieved via EGFR activation in MET-dependent cells (13), and epidermal growth factor receptor (EGFR) represent a common mo- analogous examples of reciprocal kinase switching have been lecular subset of non-small cell lung cancer (NSCLC) cases. EGFR reported in other kinase-driven cancer models (14, 15). These and mutations predict sensitivity to EGFR tyrosine kinase inhibitors other findings suggest that compensatory kinase switching may be (TKIs) and thus represent a dependency in NSCLCs harboring these a more general way in which oncogene-dependent cancers over- alterations, but the genetic basis of EGFR dependence is not fully come reliance on their primary driver kinase (14, 16), but the full- understood. Here, we applied an unbiased, ORF-based screen to range of kinases capable of mediating EGFR bypass has not been identify genetic modifiers of EGFR dependence in EGFR-mutant systematically studied. Recent advances in large-scale functional genetic libraries have NSCLC cells. This approach identified 18 kinase and kinase-related made it possible to query a wide range of genetic perturbations for genes whose overexpression can substitute for EGFR in EGFR- their ability to modulate specific cellular phenotypes in mammalian dependent PC9 cells, and these genes include seven of nine Src EGFR FGFR1 FGFR2 ITK NTRK1 NTRK2 MOS systems (17, 18). Using the model of -mutant, erlotinib- family kinase genes, , , , , , , sensitive NSCLC cells, we have performed a systematic ORF-based MST1R RAF1 , and . A subset of these genes can complement loss screen to identify kinase and kinase-related genes whose over- of EGFR activity across multiple EGFR-dependent models. Unbiased expression can complement loss of EGFR activity in an EGFR- gene-expression profiling of cells overexpressing EGFR bypass dependent context. Our findings indicate broad potential for EGFR genes, together with targeted validation studies, reveals EGFR- substitution in the setting of EGFR dependence, with compensatory independent activation of the MEK-ERK and phosphoinositide mechanisms commonly conferring EGFR-independent activation of 3-kinase (PI3K)-AKT pathways. Combined inhibition of PI3K-mTOR the PI3K-AKT and MEK-ERK signaling pathways. Importantly, this and MEK restores EGFR dependence in cells expressing each of approach has recovered known mechanisms of erlotinib resistance as the 18 EGFR bypass genes. Together, these data uncover a broad well as identified novel mediators of EGFR bypass in EGFR-mutant spectrum of kinases capable of overcoming dependence on EGFR NSCLC. These data support the idea that the EGFR-dependent and underscore their convergence on the PI3K-AKT and MEK-ERK state can be redundantly driven by diverse genetic inputs that signaling axes in sustaining EGFR-independent survival. commonly converge on shared downstream signaling nodes. Results epidermal growth factor receptor | non-small cell lung cancer | ORF An ORF-Based Screen Identifies 18 Genes Whose Expression Can Substitute for EGFR in an EGFR-Dependent NSCLC Cell Line. To he term “oncogene addiction” has been used to describe the identify genes that complement loss of EGFR activity in the Tphenomenon whereby tumor cells exhibit singular reliance on an oncogene or oncogenic pathway for their survival, despite the accumulation of multiple genetic lesions (1). In non-small Significance cell lung cancer (NSCLC), this principle is perhaps best exem- plified with the finding that epidermal growth factor receptor Non-small cell lung cancers (NSCLCs) harboring mutations in (EGFR) mutations predict response to EGFR tyrosine kinase the epidermal growth factor receptor (EGFR) gene are often inhibitors (TKIs) gefitinib and erlotinib, and thus represent a de- singularly reliant on EGFR activity for tumor cell survival, but pendency in the subset of tumors harboring these alterations (2–6). the genetic basis for this dependence is not fully understood. In However, though EGFR-mutant NSCLCs typically respond dra- this study, we have performed a screen to identify a spectrum matically to EGFR TKIs, clinical responses are not universal, even of kinase genes whose overexpression can overcome NSCLC within this genetically defined cohort, with the rate of objective cells’ reliance on EGFR. Using both unbiased and targeted response estimated to be ∼71% (5, 6). Furthermore, the over- approaches, we demonstrate that these genes commonly by- whelming majority of patients who initially respond to EGFR pass dependence on EGFR through reactivation of downstream inhibitors ultimately develop resistance to therapy (7). A deeper signaling pathways. understanding of the genetic underpinnings of EGFR addiction, and how EGFR-mutant cells can overcome reliance on EGFR, may Author contributions: T.S., A.S., and M.M. designed research; T.S. and M.B. performed research; V.R., B.W., F.H.W., L.A.G., D.A., T.R.G., D.E.R., and A.S. contributed new re- improve clinical outcomes. agents/analytic tools; T.S., V.R., F.P., M.B., M.I., A.D.C., and C.S.P. analyzed data; F.P. Here, we have applied an unbiased screening approach to iden- supervised screening; B.W. visualized data; F.H.W. and L.A.G. guided anaplastic lym- tify genetic modifiers of EGFR dependence in NSCLC. Mounting phoma kinase studies; and T.S. and M.M. wrote the paper. evidence supports the existence of several genetic modifiers of Conflict of interest statement: M.M. was a consultant and received research support from EGFR dependence in EGFR-mutant NSCLC, which can reduce Novartis, and receives research support from Bayer. M.M., L.A.G., and T.R.G. are equity the degree to which these tumors rely on EGFR and thereby holders in, and consultants for, Foundation Medicine. contribute to EGFR TKI resistance (8). Examples include ampli- This article is a PNAS Direct Submission. fication of the MET receptor tyrosine kinase (RTK) (9), activation 1To whom correspondence should be addressed. Email: matthew_meyerson@dfci. of the NF-κB pathway (8), amplification of the HER2 (ERBB2) harvard.edu. GENETICS RTK (10), amplification of the CRKL gene (11), and activation of This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. the AXL kinase (12). Notably, MET bypass can be reciprocally 1073/pnas.1412228112/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1412228112 PNAS | December 30, 2014 | vol. 111 | no. 52 | 18661–18666 Downloaded by guest on September 23, 2021 setting of EGFR dependence, we performed an ORF over- were assayed for erlotinib sensitivity across multiple drug doses expression screen in an EGFR-dependent NSCLC cell line in the using 72-h growth inhibition assays (Fig. 2A). For comparison, presence of erlotinib. A library of 589 ORFs encoding human the erlotinib-resistant EGFR-mutant positive controls EGFR- kinases and kinase-related proteins [Center for Cancer Systems Δ(E746-A750)-T790M (henceforth EGFR-ex19del-T790M) and Biology (CCSB)/Broad Institute Kinase ORF Collection] (17, EGFR-L858R-T790M were tested in parallel. Ectopic expression 18) was expressed via lentiviral transduction in the EGFR- of 18 of the 19 candidate bypass ORFs was capable of reducing mutant NSCLC cell line, PC9, which is sensitive to EGFR TKIs erlotinib sensitivity relative to cells transduced with an inert ORF A > with an IC50 of ∼30 nM (19). ORF-expressing PC9 cells were (Fig. 2 ). This reduced sensitivity corresponded to a twofold to treated with 300 nM erlotinib, 3 μM erlotinib, or DMSO for 72 h >400-fold shift in IC50 values (Table S1). One primary screening before being assayed for cell viability. Experimental ORFs were hit, YES1, failed to bypass EGFR dependence in our validation screened alongside positive control EGFR ORFs encoding the studies, leaving seven of nine Src family kinase genes confirmed to T790M “gatekeeper” mutation, which can promote resistance to confer the complementation phenotype (Fig. 2A and Table S1); EGFR TKIs (20), in cis with a canonical EGFR activating mu- and notably, this gene scored nearest to the cutoff we used to tation [EGFR-Δ(E746-A750)-T790M and EGFR-L858R-T790M] select ORFs for validation (Fig. 1). We noted that the known (21). Additional positive controls included ORFs encoding acti- EGFR bypass gene MET did not score in our primary screen, vating alleles of several MAPK family members. likely due to failure of the MET expression vector to express MET Cell viability under screening conditions was very low; the me- protein (Fig. S1B). Overall, we estimate that the vast majority of dian relative viability for all experimental ORFs was 12% at the screened ORFs express protein in PC9 cells (Fig. S1 C and D). 300-nM dose and 8% at the 3-μM dose (Fig. 1). Overexpression of The 18 genes capable of bypassing EGFR dependence in PC9 19 ORFs, of the 589 tested, led to a significant increase in viability cells were used to transduce four additional EGFR-mutant of erlotinib-treated PC9 cells, with viability of at least 39% in 300 NSCLC cell line models: the EGFR TKI-sensitive models nM erlotinib and at least 31% in 3 μM erlotinib (Fig. 1). The genes HCC827, HCC2935, and H3255 (22), and the partially sensitive whose ectopic expression led to greater viability in the presence of model HCC2279 (22, 23).
Recommended publications
  • TYKERB  Decreases in Left Ventricular Ejection Fraction (LVEF) Have Been Reported
    HIGHLIGHTS OF PRESCRIBING INFORMATION ---------------------------WARNINGS AND PRECAUTIONS------------------- These highlights do not include all the information needed to use TYKERB Decreases in left ventricular ejection fraction (LVEF) have been reported. safely and effectively. See full prescribing information for TYKERB. Confirm normal LVEF before starting TYKERB and continue evaluations TYKERB® (lapatinib) tablets, for oral use during treatment. (5.1) Initial U.S. Approval: 2007 TYKERB has been associated with hepatotoxicity. Monitor liver function tests before initiation of treatment, every 4 to 6 weeks during treatment, and as WARNING: HEPATOTOXICITY clinically indicated. Discontinue and do not restart TYKERB if patients See full prescribing information for complete boxed warning. experience severe changes in liver function tests. (5.2) Hepatotoxicity has been observed in clinical trials and postmarketing Dose reduction in patients with severe hepatic impairment should be experience. The hepatotoxicity may be severe and deaths have been considered. (2.2, 5.3, 8.7) reported. Causality of the deaths is uncertain. (5.2) Diarrhea, including severe diarrhea, has been reported during treatment. Manage with antidiarrheal agents, and replace fluids and electrolytes if --------------------------------INDICATIONS AND USAGE--------------------------- severe. (5.4) TYKERB is a kinase inhibitor indicated in combination with: (1) TYKERB has been associated with interstitial lung disease and pneumonitis. capecitabine for the treatment of patients with advanced or metastatic breast Discontinue TYKERB if patients experience severe pulmonary symptoms. cancer whose tumors overexpress human epidermal growth factor receptor 2 (5.5) (HER2) and who have received prior therapy, including an anthracycline, a TYKERB may prolong the QT interval in some patients. Consider taxane, and trastuzumab. electrocardiogram (ECG) and electrolyte monitoring.
    [Show full text]
  • Combining Paclitaxel and Lapatinib As Second-Line Treatment for Patients with Metastatic Transitional Cell Carcinoma: a Case Series
    ANTICANCER RESEARCH 32: 3949-3952 (2012) Combining Paclitaxel and Lapatinib as Second-line Treatment for Patients with Metastatic Transitional Cell Carcinoma: A Case Series STÉPHANE CULINE, ZINEB SELLAM, LINDA BOUAITA, ELIAS ASSAF, CATHERINE DELBALDO, MURIEL VERLINDE-CARVALHO and DAMIEN POUESSEL Department of Medical Oncology, Henri Mondor Hospital, Créteil, France Abstract. Background: Current first-line cisplatin-based trial comparing vinflunine with best supportive care (BSC) combination chemotherapy regimens provide interesting to BSC alone, an estimated difference in overall survival response rates but limited impact on survival for patients with (OS) of 2 months was reached in the intent-to-treat metastatic transitional cell carcinoma of the urothelium. Such population. However, a significant difference in OS was only results leave a significant patient population in need of salvage seen after removing patients who had major protocol therapy. Patients and Methods: As the epidermal growth factor violations (2). Therefore therapy for patients who fail first- receptors 1 and 2 (EGFR and HER2) are frequently line cisplatin-based chemotherapy remains a highly unmet overexpressed in urothelial carcinoma, we explored the medical need. feasibility of a combination of paclitaxel (80 mg/m2/week) and In a phase II study led by the French Genito-Urinary lapatinib (1,500 mg orally daily) for six patients who were Tumor group (GETUG), the activity of weekly paclitaxel as treated after failure of first-line platinum-based chemotherapy. second-line chemotherapy was assessed in 45 patients with Results: Only one out of six patients was able to receive the MTCCU. A low objective response rate (9%) along with a full doses during the first six weeks of treatment, while grade high rate of stabilization (38%) suggested limited impact as 2 or 3 diarrhea events required lapatinib dose reduction (one a single agent (3).
    [Show full text]
  • HER2-Positive Male Breast Cancer: an Update
    Breast Cancer: Targets and Therapy Dovepress open access to scientific and medical research Open Access Full Text Article REVIEW HER2-positive male breast cancer: an update Laura Ottini1 Abstract: Although rare, male breast cancer (MBC) remains a substantial cause for morbidity Carlo Capalbo2 and mortality in men. Based on age frequency distribution, age-specific incidence rate pattern, Piera Rizzolo1 and prognostic factor profiles, MBC is considered similar to postmenopausal breast cancer Valentina Silvestri1 (BC). Compared with female BC (FBC), MBC cases are more often hormonal receptor Giuseppe Bronte3 (estrogen receptor/progesterone receptor [ER/PR]) positive and human epidermal growth factor Sergio Rizzo3 receptor 2 (HER2) negative. Treatment of MBC patients follows the same indications as female postmenopausal with surgery, systemic therapy, and radiotherapy. To date, ER/PR and HER2 Antonio Russo3 status provides baseline predictive information used in selecting optimal adjuvant/neoadjuvant 1 Department of Experimental therapy and in the selection of therapy for recurrent or metastatic disease. HER2 represents Medicine, “Sapienza” University of ® Rome, Rome, Italy; 2Medical Oncology, a very interesting molecular target and a number of compounds (trastuzumab [Herceptin ; IDI-IRCCS, Rome, Italy; 3Department F. Hoffmann-La Roche, Basel, Switzerland] and lapatinib [Tykerb®, GlaxoSmithKline, London, of Surgical and Oncological Sciences, UK]) are currently under clinical evaluation. Particularly, trastuzumab, a monoclonal antibody Section of Medical Oncology, University of Palermo, Palermo, Italy which selectively binds the extracellular domain of HER2, has become an important therapeutic agent for women with HER2-positive (HER2+) BC. Currently, data regarding the use of trastuzumab in MBC patients is limited and only few case reports exist.
    [Show full text]
  • Lapatinib Has Been Associated with Hepatotoxicity
    HIGHLIGHTS OF PRESCRIBING INFORMATION ----------------------- WARNINGS AND PRECAUTIONS ---------------- These highlights do not include all the information needed to use • Decreases in left ventricular ejection fraction have been reported. TYKERB safely and effectively. See full prescribing information for Confirm normal LVEF before starting TYKERB and continue evaluations TYKERB. during treatment. (5.1) • Lapatinib has been associated with hepatotoxicity. Monitor liver function TYKERB (lapatinib) tablets tests before initiation of treatment, every 4 to 6 weeks during treatment, Initial U.S. Approval: 2007 and as clinically indicated. Discontinue and do not restart TYKERB if patients experience severe changes in liver function tests. (5.2) WARNING: HEPATOTOXICITY • Dose reduction in patients with severe hepatic impairment should be See full prescribing information for complete boxed warning. considered. (2.2, 5.3, 8.7) Hepatotoxicity has been observed in clinical trials and postmarketing • Diarrhea, including severe diarrhea, has been reported during treatment. experience. The hepatotoxicity may be severe and deaths have been Manage with anti-diarrheal agents, and replace fluids and electrolytes if reported. Causality of the deaths is uncertain. [See Warnings and severe. (5.4) Precautions (5.2).] • Lapatinib has been associated with interstitial lung disease and pneumonitis. Discontinue TYKERB if patients experience severe pulmonary symptoms. (5.5) ---------------------------RECENT MAJOR CHANGES -------------------- Boxed Warning. Month YEAR • Lapatinib prolongs the QT interval in some patients. Consider ECG and Hepatotoxicity. (5.2, 17.6) Month YEAR electrolyte monitoring. (5.6) Interstitial lung disease and pneumonitis. (5.5) August 2007 • Fetal harm can occur when administered to a pregnant woman. Women should be advised not to become pregnant when taking TYKERB.
    [Show full text]
  • Gene and Drug Matrix for Personalized Cancer Therapy
    CORRESPONDENCE LINK TO ORIGINAL ARTICLE ALK) for the treatment of non-small cell lung carcinoma with ALK translocations Gene and drug matrix for or for the treatment of neuroblastoma with ALK-activating mutations or personalized cancer therapy overexpression6,7. Although the numbers are currently Tim Harris small this matrix of ‘genes versus drugs’ is growing rapidly and will expand dramatically as our understanding of The recent Perspective by Richard Schilsky As a consequence of these gene profiling tumour mutations increases and as new (Nature Rev. Drug Discov. 9, 363–367; studies, physicians are being left with the inhibitors with different specificities emerge. 2010)1 suggests that the ‘future is now’ for increasingly complex question of which A more extensive analysis comparing many personalized medicine in the treatment of drugs to use to treat tumours that have one of the kinase inhibitors in late-stage cancer1. Examples of genetic analyses that or more of these cancer-associated molec- development against this same set of drive clinical decision-making in cancer ular defects. There are two aspects to this genes is available on request. include the use of imatinib in the treat- challenge: one is the ability to define the The therapeutic impact of personalized ment of chronic myeloid leukaemia with molecular defect or defects in the tumour, health care utilizing robust diagnostic assays BCR–ABL translocations; using gefitinib or and the other is access to available drugs and selected therapies will be considerable. erlotinib to treat lung cancer with epidermal on the market (or in clinical development) Information such as that provided by TABLE 1 growth factor receptor (EGFR) mutations; that are likely to be appropriate for the above will be needed to inform oncologists treating human epidermal growth factor treatment of that subclass of disease in and allow them to treat patients in a more receptor 2 (HER2/neu)-positive patients the context of other relevant (chemo) personalized way.
    [Show full text]
  • TARCEVA for Severe Renal These Highlights Do Not Include All the Information Needed to Use Toxicity
    HIGHLIGHTS OF PRESCRIBING INFORMATION patients at risk of dehydration. Withhold TARCEVA for severe renal These highlights do not include all the information needed to use toxicity. (5.2) TARCEVA® safely and effectively. See full prescribing information for • Hepatotoxicity: Occurs with or without hepatic impairment, including TARCEVA. hepatic failure and hepatorenal syndrome: Monitor periodic liver testing. Withhold or discontinue TARCEVA for severe or worsening liver tests. (5.3) TARCEVA (erlotinib) tablets, for oral use • Gastrointestinal perforations: Discontinue TARCEVA. (5.4) Initial U.S. Approval: 2004 • Bullous and exfoliative skin disorders: Discontinue TARCEVA. (5.5) • ---------------------------RECENT MAJOR CHANGES--------------------------­ Cerebrovascular accident (CVA): The risk of CVA is increased in patients with pancreatic cancer. (5.6) Indications and Usage, Non-Small Cell Lung Cancer (NSCLC) (1.1) 10/2016 • Microangiopathic hemolytic anemia (MAHA): The risk of MAHA is Dosage and Administration (2.1) 06/2016 increased in patients with pancreatic cancer. (5.7) Dosage and Administration, Dose Modifications (2.4) 05/2016 • Ocular disorders: Discontinue TARCEVA for corneal perforation, Warnings and Precautions, Cerebrovascular Accident (5.6) 10/2016 ulceration or persistent severe keratitis. (5.8) Warnings and Precautions, Embryo-fetal Toxicity (5.10) 10/2016 • Hemorrhage in patients taking warfarin: Regularly monitor INR in patients taking warfarin or other coumarin-derivative anticoagulants. (5.9) ---------------------------INDICATIONS
    [Show full text]
  • Fasting Potentiates the Anticancer Activity of Tyrosine Kinase Inhibitors by Strengthening MAPK Signaling Inhibition
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 14 Fasting potentiates the anticancer activity of tyrosine kinase inhibitors by strengthening MAPK signaling inhibition Irene Caffa1, Vito D’Agostino2, Patrizia Damonte1, Debora Soncini1, Michele Cea1, Fiammetta Monacelli1, Patrizio Odetti1,3, Alberto Ballestrero1,3, Alessandro Provenzani2, Valter D. Longo4,5 and Alessio Nencioni1,3 1 Department of Internal Medicine, University of Genoa, Genoa, Italy 2 Laboratory of Genomic Screening, Centre for Integrative Biology, CIBIO, University of Trento, Trento, Italy 3 IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy 4 Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 5 IFOM, FIRC Institute of Molecular Oncology, Milan, Italy Correspondence to: Valter D. Longo, email: [email protected] Correspondence to: Alessio Nencioni, email: [email protected] Keywords: tyrosine kinase inhibitors, fasting, MAPK pathway, E2F transcription factors, cell cycle regulation Received: March 04, 2015 Accepted: March 11, 2015 Published: March 18, 2015 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Tyrosine kinase inhibitors (TKIs) are now the mainstay of treatment in many types of cancer. However, their benefit is frequently short-lived, mandating the search for safe potentiation strategies. Cycles of fasting enhance the activity of chemo-radiotherapy in preclinical cancer models and dietary approaches based on fasting are currently explored in clinical trials. Whether combining fasting with TKIs is going to be potentially beneficial remains unknown.
    [Show full text]
  • Partial Response to Erlotinib in a Patient with Imatinib-Refractory
    Verma et al. Clin Sarcoma Res (2020) 10:28 https://doi.org/10.1186/s13569-020-00149-1 Clinical Sarcoma Research CASE REPORT Open Access Partial response to erlotinib in a patient with imatinib-refractory sacral chordoma Saurav Verma1, Surya Prakash Vadlamani1, Shamim Ahmed Shamim2, Adarsh Barwad3, Sameer Rastogi4* and S. T. Arun Raj2 Abstract Background: Chordoma is a rare, slow growing and locally aggressive mesenchymal neoplasm with uncommon distant metastases. It is a chemo-resistant disease with surgery and radiotherapy being the mainstay in treatment of localized disease. In advanced disease imatinib has a role. We report a case of metastatic sacral chordoma with symp- tomatic and radiological response to erlotinib post-progression on imatinib. Case presentation: A 48-year-old male with a sacral chordoma underwent partial sacrectomy followed by post- operative radiotherapy. Upon recurrence he received palliative radiotherapy to hemipelvis and was ofered therapy with imatinib. However, the disease was refractory to imatinib and he was started on treatment with erlotinib—show- ing a partial response on imaging at two months. He is currently doing well at 13 months since start of erlotinib. Conclusions: As seen in previously reported cases, erlotinib is a therapeutic option in advanced chordoma, even in imatinib refractory cases and thus warrants exploration of its therapeutic role in prospective clinical trials. Keywords: Chordoma, EGFR, Erlotinib Background adjuvant setting after a full or subtotal resection, and as Chordoma is a rare mesenchymal neoplasm which arises the primary treatment in unresectable disease. from the remnants of primitive notochord [1]. It accounts Chordoma responds poorly to cytotoxic chemotherapy.
    [Show full text]
  • LY2801653 Is an Orally Bioavailable Multi-Kinase Inhibitor with Potent
    Invest New Drugs (2013) 31:833–844 DOI 10.1007/s10637-012-9912-9 PRECLINICAL STUDIES LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and other oncoproteins, and displays anti-tumor activities in mouse xenograft models S. Betty Yan & Victoria L. Peek & Rose Ajamie & Sean G. Buchanan & Jeremy R. Graff & Steven A. Heidler & Yu-Hua Hui & Karen L. Huss & Bruce W. Konicek & Jason R. Manro & Chuan Shih & Julie A. Stewart & Trent R. Stewart & Stephanie L. Stout & Mark T. Uhlik & Suzane L. Um & Yong Wang & Wenjuan Wu & Lei Yan & Wei J. Yang & Boyu Zhong & Richard A. Walgren Received: 19 October 2012 /Accepted: 3 December 2012 /Published online: 29 December 2012 # The Author(s) 2012. This article is published with open access at Springerlink.com Summary The HGF/MET signaling pathway regulates a of a potent, orally bioavailable, small-molecule inhibitor wide variety of normal cellular functions that can be subverted LY2801653 targeting MET kinase. LY2801653 is a type-II to support neoplasia, including cell proliferation, survival, ATP competitive, slow-off inhibitor of MET tyrosine kinase apoptosis, scattering and motility, invasion, and angiogenesis. with a dissociation constant (Ki) of 2 nM, a pharmacodynamic −1 MET over-expression (with or without gene amplification), residence time (Koff) of 0.00132 min and t1/2 of 525 min. aberrant autocrine or paracrine ligand production, and mis- LY2801653 demonstrated in vitro effects on MET pathway- sense MET mutations are mechanisms that lead to activation dependent cell scattering and cell proliferation; in vivo anti- of the MET pathway in tumors and are associated with poor tumor effects in MET amplified (MKN45), MET autocrine prognostic outcome.
    [Show full text]
  • FLT3 Inhibitors in Acute Myeloid Leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2*
    Wu et al. Journal of Hematology & Oncology (2018) 11:133 https://doi.org/10.1186/s13045-018-0675-4 REVIEW Open Access FLT3 inhibitors in acute myeloid leukemia Mei Wu1, Chuntuan Li2 and Xiongpeng Zhu2* Abstract FLT3 mutations are one of the most common findings in acute myeloid leukemia (AML). FLT3 inhibitors have been in active clinical development. Midostaurin as the first-in-class FLT3 inhibitor has been approved for treatment of patients with FLT3-mutated AML. In this review, we summarized the preclinical and clinical studies on new FLT3 inhibitors, including sorafenib, lestaurtinib, sunitinib, tandutinib, quizartinib, midostaurin, gilteritinib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG 925, TTT-3002, and FF-10101. New generation FLT3 inhibitors and combination therapies may overcome resistance to first-generation agents. Keywords: FMS-like tyrosine kinase 3 inhibitors, Acute myeloid leukemia, Midostaurin, FLT3 Introduction RAS, MEK, and PI3K/AKT pathways [10], and ultim- Acute myeloid leukemia (AML) remains a highly resist- ately causes suppression of apoptosis and differentiation ant disease to conventional chemotherapy, with a me- of leukemic cells, including dysregulation of leukemic dian survival of only 4 months for relapsed and/or cell proliferation [11]. refractory disease [1]. Molecular profiling by PCR and Multiple FLT3 inhibitors are in clinical trials for treat- next-generation sequencing has revealed a variety of re- ing patients with FLT3/ITD-mutated AML. In this re- current gene mutations [2–4]. New agents are rapidly view, we summarized the preclinical and clinical studies emerging as targeted therapy for high-risk AML [5, 6]. on new FLT3 inhibitors, including sorafenib, lestaurtinib, In 1996, FMS-like tyrosine kinase 3/internal tandem du- sunitinib, tandutinib, quizartinib, midostaurin, gilteriti- plication (FLT3/ITD) was first recognized as a frequently nib, crenolanib, cabozantinib, Sel24-B489, G-749, AMG mutated gene in AML [7].
    [Show full text]
  • Genomic Aberrations Associated with Erlotinib Resistance in Non-Small Cell Lung Cancer Cells
    ANTICANCER RESEARCH 33: 5223-5234 (2013) Genomic Aberrations Associated with Erlotinib Resistance in Non-small Cell Lung Cancer Cells MASAKUNI SERIZAWA1, TOSHIAKI TAKAHASHI2, NOBUYUKI YAMAMOTO2,3 and YASUHIRO KOH1 1Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Sunto-gun, Shizuoka, Japan; 2Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Sunto-gun, Shizuoka, Japan; 3Third Department of Internal Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan Abstract. Background/Aim: Mechanisms of resistance to mutations develop resistance, usually within one year of epidermal growth factor receptor (EGFR)-tyrosine kinase treatment. Therefore, there is an urgent need to elucidate the inhibitors (TKIs) in non-small cell lung cancer (NSCLC) underlying mechanisms of resistance in such tumors to are not fully-understood. In this study we aimed to overcome this obstacle (11-14, 17, 24). Recent studies elucidate remaining unknown mechanisms using erlotinib- suggest that mechanisms of acquired resistance to EGFR- resistant NSCLC cells. Materials and Methods: We TKIs can be categorized into three groups: occurrence of performed array comparative genomic hybridization genetic alterations, activation of downstream pathways via (aCGH) to identify genomic aberrations associated with bypass signaling, and phenotypic transformation (15, 16, 21); EGFR-TKI resistance in erlotinib-resistant PC-9ER cells. therapeutic strategies to overcome these resistance Real-time polymerase chain reaction (PCR) and mechanisms are under development. However, although the immunoblot analyses were performed to confirm the results causes of acquired resistance to EGFR-TKIs have been of aCGH. Results: Among the five regions with copy investigated, in more than 30% of patients with acquired number gain detected in PC-9ER cells, we focused on resistance to EGFR-TKI treatment, the mechanisms remain 22q11.2-q12.1 including v-crk avian sarcoma virus CT10 unknown (15).
    [Show full text]
  • Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) Through Multikinase Inhibition Jessica D
    Published OnlineFirst February 9, 2018; DOI: 10.1158/1078-0432.CCR-17-1928 Cancer Therapy: Preclinical Clinical Cancer Research Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) through Multikinase Inhibition Jessica D. Lang1,William P.D. Hendricks1, Krystal A. Orlando2, Hongwei Yin1, Jeffrey Kiefer1, Pilar Ramos1, Ritin Sharma3, Patrick Pirrotte3, Elizabeth A. Raupach1,3, Chris Sereduk1, Nanyun Tang1, Winnie S. Liang1, Megan Washington1, Salvatore J. Facista1, Victoria L. Zismann1, Emily M. Cousins4, Michael B. Major4, Yemin Wang5, Anthony N. Karnezis5, Aleksandar Sekulic1,6, Ralf Hass7, Barbara C. Vanderhyden8, Praveen Nair9, Bernard E. Weissman2, David G. Huntsman5,10, and Jeffrey M. Trent1 Abstract Purpose: Small cell carcinoma of the ovary, hypercalcemic type three SWI/SNF wild-type ovarian cancer cell lines. We further (SCCOHT) is a rare, aggressive ovarian cancer in young women identified ponatinib as the most effective clinically approved that is universally driven by loss of the SWI/SNF ATPase subunits RTK inhibitor. Reexpression of SMARCA4 was shown to confer SMARCA4 and SMARCA2. A great need exists for effective targeted a 1.7-fold increase in resistance to ponatinib. Subsequent therapies for SCCOHT. proteomic assessment of ponatinib target modulation in Experimental Design: To identify underlying therapeutic vul- SCCOHT cell models confirmed inhibition of nine known nerabilities in SCCOHT, we conducted high-throughput siRNA ponatinib target kinases alongside 77 noncanonical ponatinib and drug screens. Complementary proteomics approaches pro- targets in SCCOHT. Finally, ponatinib delayed tumor dou- filed kinases inhibited by ponatinib. Ponatinib was tested for bling time 4-fold in SCCOHT-1 xenografts while reducing efficacy in two patient-derived xenograft (PDX) models and one final tumor volumes in SCCOHT PDX models by 58.6% and cell-line xenograft model of SCCOHT.
    [Show full text]