Gatekeeper mutations and intra-tumoural heterogeneity in FGFR2-translocated

Similar documents
New targets in endometrial and ovarian cancer

ArQule Jefferies Global Healthcare Conference June 2015

D Ross Camidge, MD, PhD

Osamu Tetsu, MD, PhD Associate Professor Department of Otolaryngology-Head and Neck Surgery School of Medicine, University of California, San

What do liquid biopsies offer us for breast cancer patients?

Corporate Medical Policy

Personalized Medicine: Lung Biopsy and Tumor

Is FGFR an Effective Target in Cholangiocarcinoma?

Lung Cancer Genetics: Common Mutations and How to Treat Them David J. Kwiatkowski, MD, PhD. Mount Carrigain 2/4/17

609G: Concepts of Cancer Genetics and Treatments (3 credits)

BLU-554, A Novel, Potent and Selective Inhibitor of FGFR4 for the Treatment of Liver Cancer

ALK Fusion Oncogenes in Lung Adenocarcinoma

LUNG CANCER. pathology & molecular biology. Izidor Kern University Clinic Golnik, Slovenia

Round Table: Tissue Biopsy versus Liquid Biopsy. César A. Rodríguez Hospital Universitario de Salamanca-IBSAL

Introduction to Cancer

RINDOPEPIMUT (CDX-110) IN GLIOBLASTOMA

Personalized oncology: the potential for tissue and cell-free DNA

WHAT IS NEW IN BILE DUCT CANCER IN THE LAST 12 MONTHS?

Is there a role for EGFR Tyrosine Kinase Inhibitors in recurrent glioblastoma?

Circulating Tumor DNA in GIST and its Implications on Treatment

RXDX-101 & RXDX-102. Justin Gainor, MD February 20 th, 2014

1.Basis of resistance 2.Mechanisms of resistance 3.How to overcome resistance. 13/10/2017 Sara Redaelli

Improving outcomes for NSCLC patients with brain metastases

The oncologist s point of view: the promise and challenges of increasing options for targeted therapies in NSCLC

Molecular Testing in Lung Cancer

The rise of the FGFR inhibitor in advanced biliary cancer: the next cover of time magazine?

Colorectal Cancer in 2017: From Biology to the Clinics. Rodrigo Dienstmann

ArQule CorporateUpdate

Selumetinib (AZD6244; ARRY ) Pediatric MATCH. George Kirk June 2016

Disclosures Genomic testing in lung cancer

Dr C K Kwan. Associate Consultant, Queen Elizabeth Hospital, HKSAR Honorary member, Targeted Therapy Team, ICR, UK

OTRAS TERAPIAS BIOLÓGICAS EN CPNM: Selección y Secuencia Óptima del Tratamiento

Circulating tumour DNA in breast cancer. Kathleen Burke, PhD Bioinformatics Postdoctoral Fellow Laboratory of Dr. Jorge Reis-Filho

Osimertinib (AZD9291) a science-driven, collaborative approach

The PI3K/AKT axis. Dr. Lucio Crinò Medical Oncology Division Azienda Ospedaliera-Perugia. Introduction

Novel treatments for SCC Andrés Felipe Cardona, MD MS PhD.

2015 EUROPEAN CANCER CONGRESS

Virtual Journal Club: Front-Line Therapy and Beyond Recent Perspectives on ALK-Positive Non-Small Cell Lung Cancer.

Disclosure. Summary. Circulating DNA and NGS technology 3/27/2017. Disclosure of Relevant Financial Relationships. JS Reis-Filho, MD, PhD, FRCPath

LUNG CANCER IN FOCUS. ALK Inhibitors in Non Small Cell Lung Cancer: How Many Are Needed and How Should They Be Sequenced?

IMMUNOTHERAPY FOR GASTROINTESTINAL CANCERS

Incorporating pharmacodynamic, response and patient selection biomarkers. Paul Elvin PhD Chief Translational Science Officer Aptus Clinical

Giorgio V. Scagliotti Università di Torino Dipartimento di Oncologia

Investor Presentation

Genetics of Cancer Lecture 32 Cancer II. Prof. Bevin Engelward, MIT Biological Engineering Department

Drug Discovery Platform: Cancer Cell Signaling. MET Inhibitor. Merestinib, LY Christensen JG, et al1; Eder JP, et al 2

33 rd Annual J.P. Morgan Healthcare Conference. January 2015

NCCN Non-Small Cell Lung Cancer V Meeting June 15, 2018

Diagnostic with alternative sample types (liquid biopsy)

MATCHMAKING IN ONCOLOGY CHALLENGES AND COMBINATION STRATEGY FOR NOVEL TARGETED AGENTS

Joachim Aerts Erasmus MC Rotterdam, Netherlands. Drawing the map: molecular characterization of NSCLC

Cancer and Tyrosine Kinase Inhibition

Cell-free tumor DNA for cancer monitoring

Treatment Landscape in R/R DLBCL Novel Targets and Strategies. Wyndham H. Wilson, M.D., Ph.D. Senior Investigator

FGFR1 amplification and FGFR gene fusion in resected squamous cell

Il Tumore del Fegato Prospettive Future nel Trattamento dei Tumori Gastrointestinali

A View to the Future: The Development of Targeted Therapy for Melanoma. Michael Davies, M.D., Ph.D.

MET Inhibitor. Merestinib, LY Drug Discovery Platform: Cancer Cell Signaling. Derived from Christensen JG, et al 1 ; Eder JP, et al.

2015 EUROPEAN CANCER CONGRESS

Resistance to anti-her2 therapies. Service d Oncologie Médicale

Supplementary Figure 1: Tissue of Origin analysis on 152 cell lines. (a) Heatmap representation of the 30 Tissue scores for the 152 cell lines.

Liquid Biopsy. Jesus Garcia-Foncillas MD PhD. Director

7/6/2015. Cancer Related Deaths: United States. Management of NSCLC TODAY. Emerging mutations as predictive biomarkers in lung cancer: Overview

SALSA MLPA probemix P315-B1 EGFR

NGS in tissue and liquid biopsy

EGFR: fundamenteel en klinisch

ARIAD Pharmaceuticals, Inc.

Activation of cellular proto-oncogenes to oncogenes. How was active Ras identified?

1. Q: What has changed from the draft recommendations posted for public comment in November/December 2011?

TARGETED THERAPY FOR LUNG CANCER. Patient and Caregiver Guide

CDx in oncology Prof. Christophe Le Tourneau, MD, PhD FEAM Geneva September 27, 2018

Genomic tests to personalize therapy of metastatic breast cancers. Fabrice ANDRE Gustave Roussy Villejuif, France

Spectrum Pharmaceuticals

Module 3: Pathway and Drug Development

National Horizon Scanning Centre. Vandetanib (Zactima) for locally advanced or metastatic medullary thyroid cancer. December 2007

Colorectal Cancer in the Coming Years: What Can We Expect?

EXAMPLE. - Potentially responsive to PI3K/mTOR and MEK combination therapy or mtor/mek and PKC combination therapy. ratio (%)

Ets-1 identifying polynucleotide sequence for targeted delivery of anti-cancer drugs

Tissue or Liquid Biopsy? ~For Diagnosis, Monitoring and Early detection of Resistance~

Triple Negative Breast Cancer. Eric P. Winer, MD Dana-Farber Cancer Institute Harvard Medical School Boston, MA October, 2008

Nucleic Acid Testing - Oncology. Molecular Diagnosis. Gain/Loss of Nucleic Acid. Objectives. MYCN and Neuroblastoma. Molecular Diagnosis

EGFR Antibody. Necitumumab, LY , IMC-11F8. Drug Discovery Platform: Cancer Cell Signaling

Molecular Diagnosis. Nucleic acid based testing in Oncology

2 nd line Therapy and Beyond NSCLC. Alan Sandler, M.D. Oregon Health & Science University

MET as a novel treatment target- the story of the sleeping beauty. Balazs Halmos M.D. Montefiore Medical Center/Albert Einstein College of Medicine

Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment with Targeted Tyrosine Kinase Inhibitors

Reviewers' comments: Reviewer #1 (Remarks to the Author):

Phase II Cancer Trials: When and How

Design of Clinical Trials with Molecularly Targeted Therapies. Gilberto Schwartsmann

LONDON CANCER NEW DRUGS GROUP RAPID REVIEW. Update on high dose imatinib for gastrointestinal stromal tumour (GIST) harbouring KIT exon 9 mutations

Andreas Wicki. University Hospital Basel Switzerland

CURRENT STANDARD OF CARE OF COLORECTAL CANCER: THE EVOLUTION OF ESMO CLINICAL PRACTICE GUIDELINES

Chapter 11. Cell Communication

Whole Genome and Transcriptome Analysis of Anaplastic Meningioma. Patrick Tarpey Cancer Genome Project Wellcome Trust Sanger Institute

La biopsia liquida. Aldo Scarpa. Anatomia Patologica e ARC-NET Centro di Ricerca Applicata sul Cancro

Complexity of FGFR signalling in metastatic urothelial cancer

Challenges in the clinical development of PI3K inhibitors

Progress Update June 2017 Lay Summary Funding: $6,000,000 Grant Funded: July 2015 Dream Team Members Dream Team Leader:

oncogenes-and- tumour-suppressor-genes)

Pfizer Presents Final Phase 2 Data on Investigational PARP Inhibitor Talazoparib in Patients with Germline BRCA-Positive Advanced Breast Cancer

Transcription:

Gatekeeper mutations and intra-tumoural heterogeneity in FGFR2-translocated cholangiocarcinoma. Elizabeth C. Smyth 1, Irina S. Babina 2, Nicholas C. Turner 2,3*. 1 GI Unit, The Royal Marsden Hospital, NHS Foundation Trust, London, SW3 6JJ, United Kingdom. Elizabeth.Smyth@rmh.nhs.uk 2 Molecular Oncology, Breast Cancer Now Research Centre, The Institute of Cancer Research, London, SW3 6JB, United Kingdom. Irina.Babina@icr.ac.uk 3 Breast Unit, The Royal Marsden Hospital, NHS Foundation Trust, London, SW3 6JJ, United Kingdom. *Corresponding Author: Nicholas C. Turner E: Nick.Turner@icr.ac.uk T: +44 207 153 5574 A: Molecular Oncology, Breast Cancer Now Research Centre, The Institute of Cancer Research, London, SW3 6JB, United Kingdom. Competing interests: Dr Nicholas Turner has received advisory board honoraria from AstraZeneca, Novartis and Servier. Dr Elizabeth Smyth and Dr Irina Babina declare no competing interests.

Summary FGFR2 genetic translocations are frequent in cholangiocarcinoma, yet despite initial sensitivity to FGFR inhibitors in clinic, patients quickly become resistant to targeted therapies. The work published by Goyal and colleagues demonstrates that acquisition of gatekeeper mutations in FGFR2 and intra-tumoral heterogeneity drive resistance in patients with FGFR2-translocated intrahepatic cholangiocarcinoma, which will have important implications for management of the disease in clinic. Editorial Oncogenic chromosomal rearrangements in the fibroblast growth factor receptors (FGFRs) present a potential therapeutic target, with translocations in FGFR2 found most frequently in cholangiocarcinoma (1,2) and translocations in FGFR3 prevalent in bladder and glioblastoma (2). Preclinical models of these fusion proteins have demonstrated sensitivity to FGFR inhibitors, and recent data from early phase clinical trials substantiated these findings in the clinic (3-5). However the extent of these translocations as true oncogenic drivers, and the potential mechanisms of resistance to therapy, have previously only been investigated preclinically. Goyal et al(6) demonstrate FGFR2 translocations in intrahepatic cholangiocarcinoma (ICC) are classic oncogene drivers, with response to FGFR inhibition in the clinic followed by development of common gatekeeper mutations in FGFR2 resulting in clinical resistance.

Cholangiocarcinoma has historically been dichotomised by anatomical location into intra- and extra-hepatic subtypes. Recent developments in genomic and transcriptomic technologies have revealed fundamental molecular differences between these anatomic sub-sites, reflecting distinct aetiologies (7). Intrahepatic cholangiocarcinoma, a disease with growing incidence for yet unknown reasons, harbours FGFR2 translocations in up to 16% of cases (1,2). Fusion proteins generated by genetic translocations generally fuse a variety of protein fragments to the cytoplasmic tail of FGFR2, thereby deleting the C-terminus. The fusion partners usually contain protein-binding domains, which most likely induce constitutive dimerization and ligand-independent activation of the FGFR kinase domain (2). Overexpression of FGFR fusion proteins results in increased sensitivity to FGFR inhibitors in vitro and in vivo models (2). Furthermore, early phase clinical trials have demonstrated clinical activity with a number of selective FGFR inhibitors, including JNJ-42756493(3), AZD4547(5), and NVP-BGJ398(4). In previously treated FGFR2- translocated cholangiocarcinoma patients, the selective pan-fgfr inhibitor NVP- BGJ398 revealed an objective response rate of 22% and median duration of treatment of approximately six months (4). As progression-free survival in most second line cholangiocarcinoma studies is in the region of three months, these results are promising. Despite these encouraging findings, FGFR2-translocated cholangiocarcinoma tumours appear to relatively swiftly develop acquired resistance to FGFR inhibitors. In this issue of Cancer Discovery, Goyal and colleagues report on a proposed mechanism of resistance to FGFR inhibition in three patients treated with NVP- BGJ398. Although the number of patients studied is small, these patients revealed

consistent acquisition of gatekeeper mutations in FGFR2 and polyclonal resistance reflective of diverse intra-tumoural genetic heterogeneity in cholangiocarcinoma. Using molecular barcoded sequencing on progression plasma DNA and tumour, they describe the acquisition of polyclonal FGFR2 mutations, with a gatekeeper p.v564f mutation found in all three patients. Mutations at this residue had been identified as potential gatekeeper mutation in prior preclinical studies (8,9), and this study confirms the importance of p.v564 residue in mediating resistance to targeted therapy. In total, the study identified three FGFR2 mutations that were common (p.n549h, p.v564f, and p.e565a, all in the kinase domain of the receptor) and further mutations unique to individual patients. Using computational modelling and functional in vitro studies, the authors show that the mutations identified have the potential to hinder binding of NVP-BJG398 either through stabilizing the kinase in an active or inactive conformation, producing an unfavourable binding conformation, or direct steric hindrance such as that created by the commonly present p.v564f gatekeeper mutation. Goyal et al. identified two other major factors contributing to resistance. Intra-tumoral genetic heterogeneity contributed substantially to acquired resistance in at least two of the three patients, although FGFR2 p.v564f was the only selected mutation detected in plasma, in the patient with the longest duration of response. Overt geographical intra-tumoural heterogeneity was further demonstrated from multiregion sequencing of a rapid autopsy, with lesion-specific acquisition of distinct resistance mutations. The study suggests that ICC has substantial intra-tumoral

genetic heterogeneity, which presents a major challenge to targeted therapy for these cancers. Amongst the lesion-specific mutations identified, acquired genetic loss of PTEN was selected in multiple lesions, confirming prior preclinical observation that silencing PTEN reduced sensitivity to FGFR inhibitors in FGFR2- amplified cancers (10). The study provides a rationale for developing third generation inhibitors that target FGFR2_V564F and other common mutations, to treat resistant tumours or cut-off one of the mechanisms to acquired resistance. Goyal et al. also profiled currently available first- and second-generation FGFR inhibitors to identify which may be more active against gatekeeper mutations. Here, the authors switched to using TEL- FGFR3 fusions in BaF3 cell lines for inhibitor screening. Although the kinase domains of FGFR2 and FGFR3 are highly similar, the use of FGFR3 as a model is a clear weakness in the manuscript. Nevertheless, the results are interesting. Polyclonal gatekeeper mutations develop in acquired resistant TEL-FGFR3 at low doses of NVP-BGJ398, but at higher doses only V555M mutation develops (equivalent to V564 in FGFR2). The authors proceeded to evaluate the differential sensitivity of BaF3 cells expressing FGFR3 resistance mutation to several other FGFR inhibitors, of which pan-fgfr inhibitor LY2874455 displayed the most activity against cells harbouring TEL-FGFR3_V555M mutant, and also in FGFR2_V564F. This finding may have direct clinical relevance in terms of treatment sequencing for cholangiocarcinoma patients in future.

The contribution of Goyal and colleagues to the field of FGFR2-translocated cholangiocarcinoma is important. Although the manuscript is limited by small sample size, shared gatekeeper mutations were characterized. Identification of similar gatekeeper mutations in EGFR and ALK-positive lung cancer has led to development of superior second and third generation EGFR and ALK tyrosine kinase inhibitors and improved survival for non-small cell lung cancer patients. It will be important to extend these observations into more patients, and into other pan-fgfr tyrosine kinase inhibitors. Identification of gatekeeper mutations confirms beyond doubt that cholangiocarcinomas are addicted to translocated FGFR2, emphasising the pressing clinical need to move inhibitors forward to pivotal studies. A more fundamental question that the current study raises is the utility of post-progression biopsies to evaluate mechanisms of resistance in the face of gross tumour heterogeneity. As the sensitivity of circulating DNA sequencing increases, this tool may become the primary method through which the genomic evolution of tumours is evaluated, as the limitations imposed by the spatial boundaries of biopsies may needlessly limit our capacity to fully comprehend the complexity of heterogeneous tumours.

References 1. Arai Y, Totoki Y, Hosoda F, Shirota T, Hama N, Nakamura H, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology 2014;59(4):1427-34 doi 10.1002/hep.26890. 2. Wu Y-M, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, et al. Identification of Targetable FGFR Gene Fusions in Diverse Cancers. Cancer Discovery 2013;3(6):636-47 doi 10.1158/2159-8290.cd-13-0050. 3. Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, et al. Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors. J Clin Oncol 2015;33(30):3401-8 doi 10.1200/jco.2014.60.7341. 4. Javle M, Shroff R, Zhu A, Sadeghi S, Choo S, Borad M, et al. A phase 2 study of BGJ398 in patients (pts) with advanced or metastatic FGFR-altered cholangiocarcinoma (CCA) who failed or are intolerant to platinum-based chemotherapy. J Clin Oncol 2016;34(suppl 4S; abstr 335). 5. Andre F, Ranson M, Dean E, Varga A, van der Noll R, Stockman PK, et al. Abstract LB-145: Results of a phase I study of AZD4547, an inhibitor of fibroblast growth factor receptor (FGFR), in patients with advanced solid tumors. Cancer Research 2013;73(8 Supplement):LB-145-LB- doi 10.1158/1538-7445.am2013-lb-145. 6. Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, et al. Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma. Cancer Discov 2016 doi 10.1158/2159-8290.CD-16-1000. 7. Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, et al. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol 2016;13(5):261-80 doi 10.1038/nrgastro.2016.51. 8. Blencke S, Zech B, Engkvist O, Greff Z, Orfi L, Horvath Z, et al. Characterization of a conserved structural determinant controlling protein kinase sensitivity to selective inhibitors. Chemistry & biology 2004;11(5):691-701 doi 10.1016/j.chembiol.2004.02.029. 9. Byron SA, Chen H, Wortmann A, Loch D, Gartside MG, Dehkhoda F, et al. The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia 2013;15(8):975-88. 10. Pearson A, Smyth E, Babina IS, Herrera-Abreu MT, Tarazona N, Peckitt C, et al. High-Level Clonal FGFR Amplification and Response to FGFR Inhibition in a Translational Clinical Trial. Cancer Discov 2016 doi 10.1158/2159-8290.cd- 15-1246.