11
Cancer Therapy: Preclinical Mechanisms of Primary Drug Resistance in FGFR1-Amplied Lung Cancer Florian Malchers 1 , Meryem Ercanoglu 2 , Daniel Schutte 1 , Roberta Castiglione 3 , Verena Tischler 1 , Sebastian Michels 4 , Ilona Dahmen 1 , Johannes Br agelmann 1,5 , Roopika Menon 6 , Johannes M. Heuckmann 6 , Julie George 1 , Sascha Ans en 4 , Martin L. Sos 1,5 , Alex Soltermann 7 , Martin Peifer 1,8 ,J urgen Wolf 4 , Reinhard Buttner 3 , and Roman K. Thomas 1,3,9 Abstract Purpose: The 8p12-p11 locus is frequently amplied in squa- mous cell lung cancer (SQLC); the receptor tyrosine kinase broblast growth factor receptor 1 (FGFR1) being one of the most prominent targets of this amplication. Thus, small mole- cules inhibiting FGFRs have been employed to treat FGFR1- amplied SQLC. However, only about 11% of such FGFR1- amplied tumors respond to single-agent FGFR inhibition and several tumors exhibited insufcient tumor shrinkage, compati- ble with the existence of drug-resistant tumor cells. Experimental Design: To investigate possible mechanisms of resistance to FGFR inhibition, we studied the lung cancer cell lines DMS114 and H1581. Both cell lines are highly sensitive to three different FGFR inhibitors, but exhibit sustained residual cellular viability under treatment, indicating a subpopulation of existing drug-resistant cells. We isolated these subpopulations by treating the cells with constant high doses of FGFR inhibitors. Results: The FGFR inhibitorresistant cells were cross-resis- tant and characterized by sustained MAPK pathway activation. In drug-resistant H1581 cells, we identied NRAS amplication and DUSP6 deletion, leading to MAPK pathway reactivation. Furthermore, we detected subclonal NRAS amplications in 3 of 20 (15%) primary human FGFR1-amplied SQLC speci- mens. In contrast, drug-resistant DMS114 cells exhibited tran- scriptional upregulation of MET that drove MAPK pathway reactivation. As a consequence, we demonstrate that rational combination therapies resensitize resistant cells to treatment with FGFR inhibitors. Conclusions: We provide evidence for the existence of diverse mechanisms of primary drug resistance in FGFR1- amplied lung cancer and provide a rational strategy to improve FGFR inhibitor therapies by combination treatment. Clin Cancer Res; 23(18); 552736. Ó2017 AACR. Introduction Genetically activated kinases have emerged as drug targets in lung adenocarcinoma with unprecedented therapeutic efcacy (1). Examples include mutant EGFR that can be effectively inhibited by EGFR inhibitors or rearranged ALK and ROS1 that are susceptible to ALK and ROS inhibition, respectively (24). In contrast, such genetically activated therapeutic targets had been lacking in squamous cell lung cancer (SQLC; ref. 5). We and others have discovered FGFR1 amplications in SQLC that were associated with sensitivity to FGFR inhibition in preclin- ical models (68). In a phase I clinical trial, 11% of patients with FGFR1-amplied SQLC treated with a highly selective and potent FGFR inhibitor, BGJ398, experienced a partial response (9). Additional clinical trials using the FGFR inhibitors, AZD4547 or JNJ-42756493 to treat solid tumors bearing FGFR1, 2, or 3 alterations yielded similar results. All FGFR inhibitors were associated with moderate toxicity including hyperphosphatemia, decreased appetite, con- stipation, fatigue, dry mouth, and nail toxicity (1, 10, 11). Of interest, FGFR2-amplied gastric cancers and FGFR2/3rear- ranged urothelial/bladder cancers appear to be the more sensitive to FGFR inhibition. While these observations support the notion that FGFR1 amplication associates with FGFR dependency in some cases, they question the overall generalizable conclusion that FGFR1 amplication always causes response to FGFR inhibi- tion. Of note, additional patients exhibited tumor shrinkage, but less than required for a partial response, thus suggesting insufcient tumor cell killing by FGFR inhibition alone (9, 11, 12). We therefore sought to identify mechanisms that underlie primary drug resistance in FGFR1-amplied lung cancer. 1 Department of Translational Genomics, Medical Faculty, University of Cologne, Cologne, Germany. 2 Center for Molecular Medicine, University of Cologne, Cologne, Germany. 3 Institute of Pathology, University of Cologne, Cologne, Germany. 4 Department I of Internal Medicine, Center of Integrated Oncology Cologne-Bonn, University Hospital Cologne, Cologne, Germany. 5 Molecular Pathology, Institute of Pathology, University of Cologne, Cologne, Germany. 6 NEO New Oncology GmbH, Cologne, Germany. 7 Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland. 8 Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany. 9 German Cancer Research Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK), Partner site Heidelberg, Germany. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Roman K. Thomas, Department of Translational Geno- mics, University of Cologne, Weyertal 115b, Cologne 50931, Germany. Phone: 0049 221 478 98771; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-17-0478 Ó2017 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 5527 on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

Mechanisms of Primary Drug Resistance in FGFR1-Amplified ...Cancer Therapy: Preclinical Mechanisms of Primary Drug Resistance in FGFR1-Amplified Lung Cancer Florian Malchers1, Meryem

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • Cancer Therapy: Preclinical

    Mechanisms of Primary Drug Resistance inFGFR1-Amplified Lung CancerFlorian Malchers1, Meryem Ercanoglu2, Daniel Sch€utte1, Roberta Castiglione3,Verena Tischler1, Sebastian Michels4, Ilona Dahmen1, Johannes Br€agelmann1,5,Roopika Menon6, Johannes M. Heuckmann6, Julie George1, Sascha Ans�en4,Martin L. Sos1,5, Alex Soltermann7, Martin Peifer1,8, J€urgen Wolf4,Reinhard B€uttner3, and Roman K. Thomas1,3,9

    Abstract

    Purpose: The 8p12-p11 locus is frequently amplified in squa-mous cell lung cancer (SQLC); the receptor tyrosine kinasefibroblast growth factor receptor 1 (FGFR1) being one of themost prominent targets of this amplification. Thus, small mole-cules inhibiting FGFRs have been employed to treat FGFR1-amplified SQLC. However, only about 11% of such FGFR1-amplified tumors respond to single-agent FGFR inhibition andseveral tumors exhibited insufficient tumor shrinkage, compati-ble with the existence of drug-resistant tumor cells.

    Experimental Design: To investigate possible mechanisms ofresistance to FGFR inhibition, we studied the lung cancer cell linesDMS114 and H1581. Both cell lines are highly sensitive to threedifferent FGFR inhibitors, but exhibit sustained residual cellularviability under treatment, indicating a subpopulation of existingdrug-resistant cells. We isolated these subpopulations by treatingthe cells with constant high doses of FGFR inhibitors.

    Results: The FGFR inhibitor–resistant cells were cross-resis-tant and characterized by sustained MAPK pathway activation.In drug-resistant H1581 cells, we identified NRAS amplificationand DUSP6 deletion, leading to MAPK pathway reactivation.Furthermore, we detected subclonal NRAS amplifications in 3of 20 (15%) primary human FGFR1-amplified SQLC speci-mens. In contrast, drug-resistant DMS114 cells exhibited tran-scriptional upregulation of MET that drove MAPK pathwayreactivation. As a consequence, we demonstrate that rationalcombination therapies resensitize resistant cells to treatmentwith FGFR inhibitors.

    Conclusions: We provide evidence for the existence ofdiverse mechanisms of primary drug resistance in FGFR1-amplified lung cancer and provide a rational strategy toimprove FGFR inhibitor therapies by combination treatment.Clin Cancer Res; 23(18); 5527–36. �2017 AACR.

    IntroductionGenetically activated kinases have emerged as drug targets in

    lung adenocarcinoma with unprecedented therapeutic efficacy(1). Examples include mutant EGFR that can be effectivelyinhibited by EGFR inhibitors or rearranged ALK and ROS1 thatare susceptible to ALK and ROS inhibition, respectively (2–4).

    In contrast, such genetically activated therapeutic targets hadbeen lacking in squamous cell lung cancer (SQLC; ref. 5). Weand others have discovered FGFR1 amplifications in SQLC thatwere associated with sensitivity to FGFR inhibition in preclin-ical models (6–8).

    In a phase I clinical trial, 11%of patients with FGFR1-amplifiedSQLC treated with a highly selective and potent FGFR inhibitor,BGJ398, experienced a partial response (9). Additional clinicaltrials using the FGFR inhibitors, AZD4547 or JNJ-42756493 totreat solid tumors bearing FGFR1, 2, or 3 alterations yieldedsimilar results. All FGFR inhibitors were associatedwithmoderatetoxicity including hyperphosphatemia, decreased appetite, con-stipation, fatigue, dry mouth, and nail toxicity (1, 10, 11). Ofinterest, FGFR2-amplified gastric cancers and FGFR2/3–rear-ranged urothelial/bladder cancers appear to be themore sensitiveto FGFR inhibition.

    While these observations support the notion that FGFR1amplification associates with FGFR dependency in somecases, they question the overall generalizable conclusion thatFGFR1 amplification always causes response to FGFR inhibi-tion. Of note, additional patients exhibited tumor shrinkage,but less than required for a partial response, thus suggestinginsufficient tumor cell killing by FGFR inhibition alone(9, 11, 12). We therefore sought to identify mechanisms thatunderlie primary drug resistance in FGFR1-amplified lungcancer.

    1Department of Translational Genomics, Medical Faculty, University of Cologne,Cologne, Germany. 2Center for Molecular Medicine, University of Cologne,Cologne, Germany. 3Institute of Pathology, University of Cologne, Cologne,Germany. 4Department I of Internal Medicine, Center of Integrated OncologyCologne-Bonn, University Hospital Cologne, Cologne, Germany. 5MolecularPathology, Institute of Pathology, University of Cologne, Cologne, Germany.6NEONewOncologyGmbH, Cologne, Germany. 7Institute of Surgical Pathology,University Hospital Zurich, Zurich, Switzerland. 8Center for Molecular MedicineCologne (CMMC), University of Cologne, Cologne, Germany. 9German CancerResearch Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK),Partner site Heidelberg, Germany.

    Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

    Corresponding Author: Roman K. Thomas, Department of Translational Geno-mics, University of Cologne, Weyertal 115b, Cologne 50931, Germany. Phone:0049 221 478 98771; E-mail: [email protected]

    doi: 10.1158/1078-0432.CCR-17-0478

    �2017 American Association for Cancer Research.

    ClinicalCancerResearch

    www.aacrjournals.org 5527

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://crossmark.crossref.org/dialog/?doi=10.1158/1078-0432.CCR-17-0478&domain=pdf&date_stamp=2017-8-26http://clincancerres.aacrjournals.org/

  • Materials and MethodsCell culture and reagents

    H1581 (from p.99 to p.108) and DMS114 (from p.75 to p.83)cell lines were purchased from ATCC and cultured using RPMI,supplemented with 10% FCS. Cell line authentication was doneby short tandem repeat analysis (Supplementary Fig. S1). Adher-ent cells were routinely passaged by washing with PBS buffer andsubsequent incubation in Trypsin/EDTA. Trypsin was inactivatedby addition of culture medium and cells were plated or 1/6diluted. Suspension cell lines were passaged by suitable dilutionof the cell suspension. All cells were cultured at 37�Cand5%CO2.BGJ398, AZD4547, JNJ-42756493, trametinib, crizotinib, andEMD1214063 were obtained from Selleck Chemicals. They werediluted in DMSO, aliquoted, and stored as 10 mmol/L stocksat �80�C.

    Generation of FGFR inhibitor–resistant cellsH1581 and DMS114 cells were seeded at about 80% conflu-

    ence in T75 cell culture flasks (RPMI 10% FCS). FGFR inhibitorswere added at a constant concentration of 1 mmol/L. After48–72 hours, medium was removed and fresh medium withoutinhibitor was added until cells recovered. Procedure was repeateduntil cells were not affected by the 1 mmol/L FGFR inhibitortreatment.

    Limited dilution assaySupernatant fromH1581 cell culture was collected, centrifuged

    at 300 � g for 5 minutes and mixed with normal cell culturemedium to a total volume of 500 mL (RPMI, 10% FBS). H1581cells were washed with PBS, trypsinized, and 1,500 cells wereadded to the prepared medium. Suspension (250 mL) was seededperwell in ten 96-well plates (0.75 cells perwell). After twoweeks,supernatant was removed and each well was washed with 100-mLPBS. PBS was removed and 55-mL trypsin was added. After cellswere trypsinized, 55-mL RPMI (10% FBS) was added and 50-mLsuspension per well was used to inoculate two new wells. After-wards, 40-mL medium was added to each well and plates wereincubated overnight. Next day, plates were treated with 10-mLmedium containing 5,000 nmol/L BGJ398 (500 nmol/L finalconcentration). Corresponding plates were treated with 10-mLmedium containing DMSO followed by 100-mL CellTiter Glo(Promega) treatment and viability measurement. After 72-hourincubation, BGJ398-treated cells were also treated with 100-mLCellTiter Glo and viability was measured, followed by 72-hourBGJ/0-hour DMSO viability ratio calculation.

    Viability assays and compound activity predictionCell lines were plated as triplicates into sterile 96-well plates at

    1,500 cells/well density as described previously (13). After 24hours of incubation, compounds were added at increasing

    dosages, ranging from 10 mmol/L to 0.002 mmol/L together witha separateDMSOcontrol. Topredict synergy,we added the secondinhibitor in constant doses of 20, 50, 100, or 500 nmol/L. After96-hour incubation at 37�C, relative cell viability was determinedby comparing the ATP content of each well, assessed by CellTiterGloAssay (Promega), to the content of theDMSOcontrol. Finally,half-maximal growth-inhibitory concentrations (GI50) were cal-culated by the package "ic50" (R programming language; ref. 14).Combination index (CI) was calculated using CI ¼ cI1/IC50 I1 þcI2/IC50 I2 (15).

    DNA/RNA extraction and next-generation sequencingDNA was isolated using the Gentra Puregene DNA extraction

    kit (Qiagen) following themanufacturers' protocol. IsolatedDNAwas hydrated in TE buffer and stored at�80�C. RNA was isolatedusing the Qiagen RNAeasy Mini Kit according to manufacturers'protocol and stored at �80�C. One microgram of RNA wastranscribed into cDNA using Superscript III reverse transcriptase(Invitrogen, #18064) for qPCR use. Whole-exome sequencingwas performed with the SureSelectXT All Exon kit (Agilent)following manufacturers' protocol. Exon-enriched libraries weresubjected to paired-end sequencing with a read-length of 2 �100 bp. Libraries were sequenced to aminimum coverage of 60�.RNA-sequencing (RNA-seq) was performed using cDNA librariesprepared from poly(A)-selected RNA (Illumina TruSeq protocolfor mRNA). The libraries were then sequenced with a 2 � 100 bppaired-end protocol to a minimum mean coverage of 30�. Datawere processed and analyzed as described previously (16, 17). Alldata have been deposited at the European Genome-phenomeArchive under the accession code EGAS00001002491.

    Quantitative real-time PCRQuantitative real-time PCR was performed using a 7300 Real-

    Time PCR System (Applied Biosystems) and Power SYBR GreenPCR Master Mix (Applied Biosystems) following the manufac-turers' protocol. DCt values were determined using the 7300System Software (Applied Biosystems) using GADPH as referencecontrol. Gene expression was calculated by DDCt method.

    ImmunoprecipitationOne day prior treatment, cells were seeded to 70% confluence

    on 6-cm dishes. The next day, cells were treated for 24 hours withdistinct inhibitor concentrations or DMSO. Cells were washedwith coldPBS and lysed in lysis buffer (Cell Signaling Technology)supplemented with protease (Roche) and phosphatase inhibitor(Calbiochem) cocktails. After 20 minutes of incubation on ice,lysates were centrifuged at 18,000 � g for 25 minutes. Proteinconcentration in supernatants was measured using BCA ProteinAssay (Thermo Fisher Scientific). Equivalent amounts of protein(30–60 mg) were denatured at 95�C and separated on 4%–12%SDS-PAGE followed by blotting on nitrocellulose membranes(Amersham Hybond-C Extra). The following antibodies wereused for immunoblotting: Anti-RAS (ab108602, Abcam); GST-HRP (Sc-459, SantaCruz Biotechnology); phospho-AKT (#9271),total AKT (#9272), phospho-ERK (#9106), total ERK (#9102),and phosphor-FRS2 (#3221) all from Cell Signaling Technology.

    Active RAS pull-down assayRas pull-down activation assay was conducted according to the

    protocol of Cytoskeleton (cat #BK008). Two-hundred micro-grams of cell lysate proteins were incubated with 5 mL of Raf-RBD

    Translational Relevance

    In clinical trials, FGFR1-amplified lung cancer patientsexperienced limited benefit from FGFR inhibition. Here, weshow two different mechanisms that cause subclonal emer-gence of resistance, thus providing a rationale for combinationdrug treatment.

    Malchers et al.

    Clin Cancer Res; 23(18) September 15, 2017 Clinical Cancer Research5528

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • beads for 1 hour at 4�C on a rotator. Afterwards, the beads werepelleted by centrifugation at 5000 � g at 4�C for 1 minute.Supernatant was carefully removed and beads were washed with500mLofwashbuffer (25mmol/L Tris pH7.5, 30mmol/LMgCl2,40 mmol/L NaCl) followed by centrifugation at 5,000 � g at 4�Cfor 3minutes. Supernatant was removed and 4 mL of 5� Laemmlisample buffer was added to the pellet. After a 10-minute boilingstep at 95�C, samples were ready for immunoprecipitation.

    Crystal violet stainingA total of 3� 105 cells per well were seeded in 6-well plates and

    incubated overnight. Cells were exposed to DMSO and indicatedinhibitor for 96 hours at 37�C. After treatment, cells were washedtwice with 1-mL PBS per well and fixed with 500-mL 4 % para-formaldehyde (PFA) for 15 minutes at room temperature. Aftertwo washing steps with PBS, cells were stained with crystalviolet (Sigma). Stock crystal violet solution was prepared with0.5 g in 100-mL distilled water. Stock solution was diluted 1:5 in10% ethanol. After, removing the crystal violet and conductingtwo washing steps with PBS, plates were ready for scanning(HP Scanjet G4050).

    Human phospho-receptor tyrosine kinase arrayThe human phosphor-receptor tyrosine kinase (pRTK) Array

    Kit from R&D Systems was used. Cells were seeded into 10-cmdishes with 1 � 106 cells per dish. After 24-hour incubation at37�C, cells were washed twice with 3-mL cold PBS. Lysis buffer(150–200 mL) was added, scraped, and incubated on ice for 30minutes. Samples were centrifuged at 14,000 rpm for 5 minutesand supernatantswere transferred into anew cold 1.5-mL reactiontube. pRTK arrays were blocked for 1 hour at room temperature.Two-hundred micrograms protein per sample was diluted andincubated on a platform shaker together with the array overnightat 4�C following the manufacturers' protocol. After incubation,the array was washed three times in 20-mL washing buffer.Diluted anti-phospho-tyrosine-HRP detection antibody wasadded to the array and incubated for 2 hours at room temperatureon a rocking platform shaker. After three additional washingsteps, the Chemi Reagent Mix was pipetted onto the array andplaced between transparency film and a light-sensitive film (GEHealthcare) to detect the signals.

    NRAS FISHH1581 and H1581_BGJr cells were trypsinized, washed, fixed

    overnight in 4% formalin and embedded in paraffin (FFPE). Two-micron thick cuts of the FFPE blocks and additional tumormicroarrays with 163 different SQLC tumors plus available cor-responding tumor-normal were transferred on SuperFrost plusslides. According to the hybridization protocol, NRAS-FISH wasperformed using the Agilent Life Sciences SureFISH 1p13.2 NRASRD probe (G100205R). Probe signals of 60 different cells wereevaluated by two different experimenters and copy number aswell as signal ratio was calculated. A sample was referred to besubclonalNRAS amplified if 5%of the cells had a copy number of6 or more observed by both experimenters.

    ResultsA subclonal population of FGFR inhibitor–resistant cells

    We first determined half-maximal inhibitory concentrations(GI50 values) of the FGFR1-amplified, FGFR inhibitor–sensitive

    cell lines H1581 and DMS114 against three selective and pot-ent FGFR inhibitors, AZD4542, BGJ398, and JNJ-42756493(5, 18–20). Confirming earlier observations, both cell lineswere highly sensitive to FGFR inhibition with GI50 values in therange of 2 to 20 nmol/L and 50 to 300 nmol/L for the H1581and DMS114 cell lines, respectively (Fig. 1A). However, in bothcell lines, we predicted the presence of a subpopulation of cellswith primary resistance to FGFR inhibition because of theapparent inability of all FGFR inhibitors to fully reduce cellularviability. (Fig. 1A). After 96 hours of treatment with 0.1 to3 mmol/L of the FGFR inhibitors, the cells exhibited only subtlechanges in viability causing a plateau in the dose–responsecurves (Fig. 1A and B). We therefore treated H1581 andDMS114 cells with cycles of 1 mmol/L of each FGFR inhibitorto isolate these resistant cell populations (Fig. 1C; ref. 21). After8–12 weeks of culture, the cells were entirely resistant to allFGFR inhibitors tested (Fig. 1D and E). Short tandem repeatanalysis confirmed that the resistant cells originate from theparental cell line (Supplementary Fig. S1). As expected, FGFRinhibitor treatment led to decreased ERK phosphorylation inparental cells (2–4, 6–8). Of interest, all resistant cells exhibitedsustained ERK phosphorylation under 1 mmol/L of FGFRinhibitor treatment (Fig. 1E). Furthermore, resistant DMS114cells showed slightly increased AKT phosphorylation, whereasH1581 cells exhibited no detectable levels of phosphorylatedAKT (Fig. 1E). Mutations in the open reading frame of FGFR1that might have caused resistance were not found. All FGFRinhibitor–resistant cells displayed sustained ERK activation inthe presence of drug thereby indicating a possible relevance ofMAPK pathway activation in mediating resistance to FGFRinhibition.

    NRAS amplification induces resistance to FGFR inhibition inH1581 cells

    As a next step, we performed whole-exome and whole-tran-scriptome sequencing of the parental and the BGJ398-resistantcells. We analyzed the sequencing data as previously described,matching the sequencing results of the BGJ398-resistant cells tothe data of the parental cells (5, 9, 16, 22). In the case of resistantH1581 cells (H1581_BGJr), we detected a focal (6 kbp) ampli-fication with a copy number of 20 on chromosome 1p12including NRAS and a chromosomal arm level loss on 12pincluding DUSP6 (Fig. 2A; Supplementary Fig. S2). The ampli-fication on chromosome 1q12 led to 19-fold transcriptionalupregulation of NRAS (Fig. 2B; Supplementary Fig. S2A). Fur-thermore, the arm level loss on chromosome 12p resulted insignificant transcriptional downregulation of DUSP6 (Supple-mentary Fig. S2B), a negative regulator of the MAPK signalingpathway (6–8, 23, 24). We therefore hypothesized that NRASamplification caused resistance to FGFR inhibition throughMAPpathway activation, possibly in concert with loss of DUSP6. Insupport of this hypothesis, we observed that H1581_BGJrcells displayed strong enrichment of activated GTP-bound RAS(Fig. 2C). We next treated H1581 and H1581_BGJr cells with theFGFR inhibitor, BGJ398, or the MEK inhibitor, trametinib, orwith a combination of both (Fig. 2D). As expected, H1581 cellswere highly sensitive to inhibition with 50 nmol/L BGJ398while the H1581_BGJr cells remained unaffected by such treat-ment (Fig. 2D). The viability of H1581 and H1581_BGJr cellswas largely unaffected by treatment with 50 nmol/L of trame-tinib alone. In contrast, the combination of both inhibitors led

    FGFR Inhibitor Resistance

    www.aacrjournals.org Clin Cancer Res; 23(18) September 15, 2017 5529

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • to a significant reduction of cell growth and viability in bothcell lines, as assessed in viability as well as in clonogenic assays(Fig. 2D and E). However, while H1581 cells exhibited only amoderate benefit of the combination treatment (P¼ 0.004), theH1581_BGJr cells were highly sensitive (P ¼ 2 � 10�13).

    We stably transduced H1581 cells with wild-type NRAS to testthe hypothesis thatNRAS overexpression causedMAPK activationand drug resistance. Empty vector and KRAS G12V constructsserved as controls (Fig. 2F; refs. 9, 25). As predicted, RAS-trans-duced H1581 cells demonstrated a severalfold increased GI50value under treatment with FGFR inhibitors, compared with theempty vector control (Fig. 2G). Furthermore, combined treatmentof H1581 cells ectopically expressing NRAS or KRAS G12V withBGJ398 and20nmol/L of trametinib led to downregulationof theMAPK signaling pathway and cytotoxicity (Fig. 2F–H).

    To obtain an estimate of the abundance of the drug-resistantcell population in the original parental cell line, we performed alimited dilution assay and identified three different populationsof cells (Fig. 2i). As expected, the majority of H1581 colonypopulations (82%) were sensitive to the inhibitor. However,16% of the colonies exhibited a moderate increase of viabilityunder 500 nmol/L FGFR inhibitor treatment and might be "per-sistent" cells (i.e., cells that survive drug treatment, but do notproliferate in the presence of drug). Of interest, nearly 2% of thecolonies kept proliferating in the presence of drug (25 of 1621

    colonies). These colonies reached an average of 4-fold viabilityincrease during treatment. Thus, FGFR1-amplified H1581 cellsharbor subpopulations of cells with primary resistance to FGFRinhibition.

    In summary, treatment of H1581 cells with FGFR inhibitorstriggers the outgrowth of a preexisting drug-resistant subpopula-tion of cells. The resistant subpopulation demonstrated high-levelamplification of NRAS, RAS, and MAPK activation as well asresistance to FGFR inhibition. Of note, resistant cells can beeffectively killed by combined FGFR and MEK inhibition.

    MET overexpression and activation induce FGFR inhibitorresistance in DMS114 cells

    We analyzed whole-exome sequencing data of the originalDMS114 and the isolated resistant subpopulation (DMS114_BGJr),but could not identify a genomic alteration that might explainresistance of DMS114_BGJr cells (Supplementary Fig. S3). How-ever, transcriptome sequencing revealed an at least 11-fold tran-scriptional upregulation of MET (Fig. 3A), which was one of themost upregulated transcripts in DMS114_BGJr cells (Supplemen-tary Fig. S4). We also found a strong increase of MET phosphor-ylation in DMS114_BGJr cells suggesting that MET activationmight cause resistance to FGFR inhibition, similar to other settingsof drug resistance (e.g., resistance to EGFR activation; refs. 1, 10,11, 26; Fig. 3B). In support of this notion, combined treatment of

    Figure 1.

    Generation of FGFR inhibitor–resistant cells. A, FGFR dependency evaluation in FGFR1-amplified cell lines H1581 (red) and DMS114 (green) using the FGFRinhibitors AZD4547, BGJ398, and JNJ-42756493 by measuring cellular ATP content after 96 hours. Viability data were individually pooled for thethree different FGFR inhibitors. B, Residual cell viability between 0.1 to 3 mmol/L after 96 hours treatment of AZD4547, BGJ398, and JNJ-42756493 wasquantified and merged for the cell lines H1581 (red) and DMS114 (green). C, Schematic overview to generate FGFR-resistant cells. Cells were treatedwith constant dose of 1 mmol/L AZD4547, BGJ398, or JNJ-42756493. After 48–72 hours the inhibitor was removed and fresh medium without inhibitor wasadded until cells recovered. The procedure was repeated until the resistant cells showed similar growth kinetics under treatment as untreated cells. D, FGFRdependency was evaluated in the parental cell lines H1581 (red), DMS114 (green), and resistant cell lines DMS114_BGJr (black), DMS114_JNJr (brown),H1581_BGJr (grey), H1581_AZDr (purple), and H1581_JNJr (orange) using the FGFR inhibitors BGJ398 by measuring cellular ATP content after 96 hours.E, Continuous ERK phosphorylation in FGFR inhibitor resistant cells under 24-hour treatment with 1 mmol/L BGJ398 assessed by immunoblotting.

    Malchers et al.

    Clin Cancer Res; 23(18) September 15, 2017 Clinical Cancer Research5530

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • Figure 2.

    NRAS amplification as a resistance factor in H1581 cells. A, H1581 and H1581_BGJr whole exome sequencing data were matched and analyzed for copy numberchanges. Copy number gain (red) and copy number loss (blue) are illustrated below the indicated human chromosomes 1 to 22 (top). The focal amplifiedregion on chromosome 1 and the broad deleted region on chromosome 12 is zoomed in (bottom). Dashed lines indicate the genomic position of NRASand DUSP6. B, H1581 and H1581_BGJr expression values generated from RNA sequencing data are plotted as FPKM values. NRAS FPKM values are highlighted(red). C, Active RAS pull-down assay from BGJ398 treated and untreated H1581, H1581_BGJr, and NIH3T3 KRAS G12C cells (positive control) followed byimmunoprecipitation. Blots were stained with total KRAS antibody and GST-HRP antibody as loading control. D, Crystal violet clonogenic assay of H1581 andH1581_BGJr cells. Cells were plated on 6-well plates and treated 96 hours with DMSO, 50 nmol/L of BGJ398, trametinib, or in combination. E, Quantification ofclonogenic assay (crystal violet, black bars) and ATP based viability assay (gray bars) of H1581 and H1581_BGJr cells after 96-hour treatment with DMSO,50 nmol/L of BGJ398, trametinib, or in combination. P values were calculated using the two-tailed Student t test and significant results (P < 0.05) are indicatedwith � , highly significant results (P < 0.0005) are indicated with �� . F, FGFR dependency evaluation in parental (H1581, blue), empty vector (H1581 e.V., red),NRAS (H1581 NRASwt, green), and KRAS G12V (H1581 KRASG12V, purple) retroviral transduced H1581 cells using the FGFR inhibitors BGJ398 (left) alone or incombination with 20 nmol/L trametinib (right) by measuring cellular ATP content after 96 hours. G, Quantification of GI50 values in H1581 e.V. (red),H1581 NRASwt (green), and H1581 KRASG12V (purple) cells. Cells were treated with BGJ398 alone or in combination with 20 nmol/L trametinib. Viability wasassessed after 96 hours by ATP-based viability assay. H, Immunoprecipitation of H1581, H1581 e.V., H1581 NRASwt, and H1581 KRASG12V cells. Cells were treatedfor 24 hours with DMSO, 50 nmol/L of BGJ398, trametinib, or in combination. I, Limited dilution assay of H1581 cells. H1581 cells were seeded in 96-wellplates at a concentration of 0.75 cells per well and treated with 500 nmol/L of BGJ398 for 72 hours. The ratio between 72-hour treated and 0-hour untreatedH1581 single-cell colonies was calculated. Highly significant results (P < 0.0005) are indicated by �� and were calculated by two-tailed Student t test.

    FGFR Inhibitor Resistance

    www.aacrjournals.org Clin Cancer Res; 23(18) September 15, 2017 5531

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • Figure 3.

    MET activation as a resistance factor in DMS114 cells. A, DMS114 and DMS114_BGJr expression values generated from RNA sequencing data are plotted asFPKM values. MET FPKM values are highlighted (red). B, Lysates of the DMS114 (top) and DMS114 BGJ398–resistant cells (bottom) were analyzed byhuman phospho-receptor tyrosine kinase array. The arrow indicates the phosphorylated MET (pMET) signal. C, FGFR dependency evaluation in DMS114BGJ398 resistant (top) and PC9 cells (control, bottom) using the FGFR inhibitors BGJ398 or the MET inhibitor crizotinib alone or in combination. Thecombination was screened using increasing concentrations of BGJ398 (blue) or crizotinib (red) combined with constant concentrations of crizotinib(50 nmol/L yellow, 100 nmol/L black) or BGJ398 (50 nmol/L green, 100 nmol/L purple). D, Crystal violet clonogenic assay of DMS114_BGJr and PC9 cells.Cells were plated on 6-well plates and treated for 96 hours with DMSO, 500 nmol/L of BGJ398, crizotinib, or in combination. E, Quantification of clonogenicassay (crystal violet, black bars) and ATP-based viability assay (gray bars) of DMS114_BGJr and PC9 cells after 96-hour treatment with DMSO, 500 nmol/L ofBGJ398, crizotinib, or in combination. P values were calculated using the two-tailed Student t test and highly significant results (P < 0.0005) are indicatedwith �� . F, Immunoprecipitation of DMS114_BGJr and PC9 cells. Cells were treated for 24 hours with DMSO, 500 nmol/L of BGJ398, crizotinib, or incombination. G, Calculated combination index (CI) values are plotted as a boxplot. The background (light purple to purple) indicates antagonistic/additivity(CI > 0.9) effects or slight (CI ¼ 0.7–0.9), moderate (CI ¼ 0.5–0.7), and strong synergy (CI < 0.5). In three independent experiments, the cell linesHCC827GR, HCC827, DMS114_BGJr, DMS114, and PC9 cells (annotated top) were screened using the FGFR inhibitors BGJ398, the MET inhibitors crizotinib orEMD1214063 alone or in combination. The combinations were screened using increasing concentrations of the inhibitors BGJ398, crizotinib or EMD1214063combined with constant concentrations (50 and 100 nmol/L) of crizotinib/EMD1214063 or BGJ398 (annotated bottom). GI50 values for each singleexperiment were calculated followed by calculation of the combination index (CI ¼ cI1/IC50 I1 þ cI2/IC50 I2). P values were calculated using the two-tailedStudent t test and highly significant results (P < 0.0005) are indicated with �� and not significant results are indicated with n.s. H, CT before (top left,baseline) and after 12 weeks (top right, partial response), 44 weeks (bottom left, partial response), and 76 weeks (bottom right, progressive disease)of BGJ398 therapy. Arrows highlight target lesion for evaluation of tumor response. I, Pathologic examination of a rebiopsy taken after 76 weeks of BGJ398therapy. IHC HE stain (top) and phosphorylated MET stain (middle) and MET dual-color FISH with indicated copy number (bottom).

    Malchers et al.

    Clin Cancer Res; 23(18) September 15, 2017 Clinical Cancer Research5532

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • DMS114_BGJr cellswithBGJ398 and theMET inhibitor, crizotinib,led to depletion of pERK and effective cytotoxicity (Fig. 3C–F).We next calculated synergistic interactions using the FGFR inhib-itor BGJ398 in combination with the MET inhibitors, crizotinib orEMD1214063, a potent and selective c-Met inhibitor (9, 11,12, 27). To this end, we measured the effects of several differentFGFR/MET inhibitor combinations in comparison with the singledrug and quantified the combination index (CI) and the isobolo-gram derived from Loewe additivity models (13, 15, 28). As apositive control, we used EGFR-mutant HCC827 cells that aresensitive to EGFR inhibition and a derivative cell line, HCC827GR,which is resistant to erlotinib because of a MET amplification(14, 29). We treated the cells with the EGFR inhibitor, erlotinib, orthe MET inhibitor, EMD1214063, alone or with a combination ofboth. For the HCC827 and HCC827GR cell lines, we calculatedaverage CI ¼ 0.78 and CI ¼ 0.09 (P ¼ 2.195e�06), respectively,confirming strong synergistic effects of the EGFR/MET inhibitorcombination inHCC827GR cells (Fig. 3G). In the case of DMS114cells, we calculated an average CI¼ 1.24 indicating no benefit for aFGFR/MET inhibitor combination. However, we detected robustsynergistic FGFR/MET inhibitor interactions inDMS114_BGJr cells(Fig. 3G). For the BGJ398/crizotinib combination, we calculated aCI ¼ 0.45 and for the BGJ398/EMD1214063 combination a CI ¼0.22 (P ¼ 2.3e�04; Fig. 3G). Thus, combined FGFR and METinhibitionwas highly effective in DMS114_BGJr cells, supporting arole of MET in causing resistance to FGFR inhibition. As expected,EGFR-mutant PC9 cells were unaffected by the combination treat-ment (CI > 1).

    Thus, the drug-resistant subpopulation of DMS114 cells over-express and activate MET, thus leading to FGFR inhibitor resis-tance. Furthermore, the resistant subpopulation can be effectivelytreated by combined FGFR and MET inhibition.

    MET activation in a patient relapsing after initial response toFGFR inhibition

    MET amplification is a known resistance mechanism in EGFR-mutant lung cancer (15, 29, 30) and our preclinical finding oftranscriptional MET upregulation in DMS114_BGJr cells raisedthe possibility that MET may mediate resistance to FGFR inhibi-tion as well. In a phase I clinical trial using the FGFR inhibitor,BGJ398, a patient was diagnosed with a T4N2M0 squamous celllung cancer tumor (9, 16, 17). The tumor exhibited an amplifi-cation of FGFR1with a copynumber ratio of 5.8, as determined byFISH. The patient experienced a confirmed partial responseaccording to RECIST 1.1 criteria upon daily treatment withBGJ398 (100 mg) with a reduction of 51% in the target lesions(Fig. 3H). Disease progression occurred after 17 months oftreatment (Fig. 3H; Supplementary Fig. S5A). We performedcomprehensive tumor genome characterization using NEO com-prehensive genomic sequencing technology on a tumor biopsytaken before initiation of anti-FGFR therapy (5, 8, 18–20, 23).Wedetected a TP53 (p.R248L) mutation and a CDKN2A (p.G23fs)frame-shift mutation. Furthermore, we confirmed the FGFR1amplification found by FISH (copy number: 5; SupplementaryFig. S5b), but did not detect MET amplification (SupplementaryFig. S5C). After the tumor progressed under therapy with BGJ398,rebiopsy was performed. We detected a decreased FGFR1 copynumber ratio of 3.1 and a low-level amplification ofMET by FISH(copy number, 3.7). Furthermore, we observed MET activation inapproximately 50% of tumor cells as determined by IHC stainingof pMET (Fig. 3I). These observations suggest that MET activation

    may drive tumor progression in FGFR inhibitor sensitive squa-mous cell lung cancer tumor patients, similar to EGFR-mutantlung cancer. However, we cannot exclude the possibility that METwas activated before the FGFR inhibitor therapy was initiatedbecause of the lack of sufficient tumor specimen.

    Subclonal NRAS amplification in FGFR1-amplified primarysquamous cell lung tumors

    We next sought to test whether the results observed in vitroare also of potential relevance clinically. We hypothesized thatNRAS amplification might be a subclonal event in H1581 cells,inducing FGFR inhibitor resistance. We therefore labeled theNRAS locus in the H1581 and H1581_BGJr cells by FISH(Fig. 4A). Confirming our sequencing results, NRAS was highlyamplified in the H1581_BGJr cells. We detected an NRAS copynumber range of 4 to 29 signals with an average copy numberof 14 (Fig. 4A). As hypothesized, we detected clear subclonalNRAS amplifications in the parental H1581 cells as well. Weobserved 2 to 8 NRAS signals in the H1581 population with anaverage of 3.4 signals per cell (Fig. 4A). However, the ratio ofNRAS and centromere signals was 1 suggesting, compatiblewith a triploid genome.

    We next tested whether subclonalNRAS amplification can alsooccur in primary FGFR1-amplified squamous cell lung cancer toexplore whetherNRAS amplificationmight in general be a poten-tial resistance mechanism in this tumor entity. To this end, weperformed NRAS FISH on 163 primary tumor specimens (all ofsquamous histology; refs. 21, 31). Twenty of these tumors wereknown to be FGFR1 amplified (CN > 4). We scored subclonalNRAS amplification if at least 5% of the cells exhibited six ormore NRAS signals. We detected subclonal NRAS amplificationsin 3 (15%) FGFR1-amplified squamous cell lung cancer tumors(Fig. 4B). Of note, 30% of the FGFR1-amplified primary tumorsexhibited NRAS polysomy, suggesting that ploidy could be adriver of NRAS amplification in primary squamous cell lungcancer tumors with FGFR1 amplification.

    In summary, subclonal NRAS-amplified cells exist in theH1581 cell line and in primary FGFR1-amplified squamous celllung cancers. Thus,NRAS amplification might underlie resistanceto FGFR inhibition in FGFR1-amplified lung cancer.

    DiscussionEarly clinical trials testing FGFR inhibitors suggest that a subset

    of FGFR1-amplified lung tumors depend on FGFR signaling fortheir survival (9, 11). However, even in the amplified tumorsresponse rates were low and several tumors exhibited shrinkage toa lesser extent than required for a response. Thus, molecularresistance mechanisms may exist before treatment that limits theoverall efficacy of FGFR inhibitors in these patients.

    Here we show that subpopulations of FGFR1-amplified lungcancer cells exhibit primary resistance to FGFR inhibition. Wedemonstrate that distinct molecular mechanisms can underliesuch primary resistance to FGFR inhibition that may cause insuf-ficient tumor shrinkage in patients. Without harboringmutationsin the FGFR1 gene, the cell lines examined in this study exhibitedcross-resistance to all FGFR inhibitors tested. These resultssuggest that increasing FGFR inhibitor potency will not overcomeFGFR inhibitor resistance.

    Drug-resistant H1581 cells harbor amplified NRAS associat-ed with transcriptional upregulation of NRAS and increased

    FGFR Inhibitor Resistance

    www.aacrjournals.org Clin Cancer Res; 23(18) September 15, 2017 5533

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • GTP-bound RAS, thus causing MAPK activation. As a conse-quence, ectopic NRAS overexpression in H1581 cells inducedresistance to FGFR inhibition. Accordingly, combined FGFRand MEK inhibition was still able to effectively kill drug-resistant H1581 cells. In contrast, resistant DMS114 cells exhib-ited sustained MAPK signaling through MET activation. In thesecells, MET was transcriptionally upregulated and activated. Inline with this observation, combined FGFR and MET inhibition

    was highly effective in these cells. Supporting a role for thismechanism in patients, we found amplification of MET in apatient with acquired resistance to the FGFR inhibitor, BGJ398.Furthermore, we observed subclonal copy number gains ofNRAS in FGFR1-amplified primary human lung cancers. Theseresults suggest that NRAS amplification might be a generalmechanism of resistance to FGFR inhibition. Of note, bothmechanisms described here involved sustained MAPK

    Figure 4.

    Subclonal NRAS amplification in a subset of FGFR1-amplified SQLC tumors. A, Dual-color FISH (NRAS, green; control, red) of H1581 BGJ398–resistantcells (left) and parental H1581 cells (right). Pictures were taken with a 600-fold magnification and zoomed in for cells of interest. B, FISH analysis (NRAS, green;control, red) of three subclonal NRAS amplified (top) and three NRAS nonamplified samples (controls, bottom). Pictures were taken with a 600-foldmagnification and zoomed in for cells of interest.

    Malchers et al.

    Clin Cancer Res; 23(18) September 15, 2017 Clinical Cancer Research5534

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • activation under treatment, thus reinforcing the notion that thispathway is critically involved in signaling downstream ofactivated FGFR1. Furthermore, adding MAPK pathway inhibi-tors to FGFR inhibitors may be therapeutically beneficial.

    Our findings shed light on possible mechanisms of primaryresistance in FGFR1-amplified lung cancer. Similar mechanismsmay also induce acquired resistance in such tumors and it will beimportant to analyze tumor specimens after clinical relapse to testthis hypothesis.

    In summary, comprehensive molecular characterization oftumors and molecularly informed choice of combination drugregimens may help overcoming resistance to FGFR inhibition inFGFR1-amplified lung cancer, thereby improving therapeuticefficacy in this detrimental tumor type.

    Disclosure of Potential Conflicts of InterestF. Malchers is a consultant/advisory board member for NEO New Oncology

    GmbH. S. Michels is a consultant/advisory board member for Boeringer-Ingelheim, Novartis, and Roche. M.L. Sos reports receiving commercial researchgrants from Novartis. J. Wolf is a consultant/advisory board member forAstraZeneca, Bristol-Myers Squibb, Boehringer-Ingelheim, Clovis, Lilly, MSD,Novartis, Pfizer, and Roche. R.K. Thomas has ownership interests (includingpatents) at AstraZeneca, Bayer, Novartis, and Roche, is a consultant/advisoryboard member for AstraZeneca, Bayer, Boehringer-Ingelheim, Clovis, Daiichi-Sankyo, Johnson & Johnson, Lilly, Merck, MSD, New Oncology, Puma, Roche,and Sanofi-Aventis. No potential conflicts of interest were disclosed by the otherauthors.

    Authors' ContributionsConception and design: F. Malchers, J.M. Heuckmann, M.L. Sos, J. Wolf,R.K. ThomasDevelopment of methodology: F. Malchers, A. Soltermann, R. B€uttnerAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): F. Malchers, M. Ercanoglu, D. Sch€utte, R. Castiglione,V. Tischler, S. Michels, I. Dahmen, R. Menon, J.M. Heuckmann, A. Soltermann,J. Wolf, R. B€uttner

    Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): F. Malchers, M. Ercanoglu, D. Sch€utte, R. Castiglione,V. Tischler, J. Br€agelmann, R. Menon, J.M. Heuckmann, J. George, M.L. Sos, M.Peifer, J. Wolf, R. B€uttnerWriting, review, and/or revision of themanuscript: F.Malchers, R. Castiglione,S.Michels, J. Br€agelmann, J. George,M.L. Sos, A. Soltermann, J.Wolf, R. B€uttner,R.K. ThomasAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): F. Malchers, M.S. Ercanoglu, J. George, S. Ans�enStudy supervision: F. Malchers, J. Wolf, R.K. Thomas

    AcknowledgmentsWe thank the computing center of the University of Cologne (RRZK) for

    providing the CPU time on the DFG-funded supercomputer 'CHEOPS', as wellas for the support. We thank Tim Perera and Eli Jovcheva for stimulatingdiscussions and for having contributed to the initiation of this study.

    Grant SupportThis work was supported by the Federal German Ministry of Science and

    Education (BMBF) through the e:Med program (grant no. 01ZX1303A and01ZX1603A, to R.K. Thomas, J. Wolf, R. B€uttner and M. Peifer and grant no.01ZX1406, to M.L. Sos andM. Peifer), the EFRE initiative (grant no. LS-1-1-030to R.K. Thomas, M.L. Sos, R. B€uttner, and J. Wolf), by the German Cancer Aid(Deutsche Krebshilfe, grant ID: 109679 to R.K. Thomas, M. Peifer, andR. B€uttner), by the state North Rhine Westphalia (NRW) and by the EuropeanUnion (European Regional Development Fund: Investing In Your Future)through the PerMed NRW initiative (grant 005-1111-0025 to R.K. Thomas,J.Wolf, andR. B€uttner) andby theGermanConsortium for Translational CancerResearch (DKTK) Joint Funding program (to R.K. Thomas). V. Tischler is therecipient of a joint ERS/EMBO Long-Term Research fellowship no. LTRF 2014-2951 and a Swiss Cancer League postdoctoral research fellowship. J. Georgereceived funding as part of the IASLC Young Investigator award.

    The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisementin accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    Received February 21, 2017; revised April 29, 2017; accepted June 13, 2017;published OnlineFirst June 19, 2017.

    References1. The Cancer Genome Atlas Network. Comprehensive molecular profiling

    of lung adenocarcinoma. Nature 2014;511:543–50.2. Mok TS, Wu Y-L, Thongprasert S, Yang C-H, Chu D-T, Saijo N, et al.

    Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N EnglJ Med 2009;361:947–57.

    3. Solomon BJ, Mok T, KimD-W,Wu Y-L, Nakagawa K, Mekhail T, et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl JMed 2014;371:2167–77.

    4. Shaw AT, Ou S-HI, Bang Y-J, Camidge DR, Solomon BJ, Salgia R, et al.Crizotinib in ROS1-rearranged non–small-cell lung cancer. N Engl J Med2014;371:1963–71.

    5. Hammerman PS, Voet D, Cibulskis K, McKenna A, Lander ES, Gabriel S,et al. Comprehensive genomic characterization of squamous cell lungcancers. Nature 2012;489:519–25.

    6. Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, et al.Frequent and focal FGFR1 amplification associates with therapeuticallytractable FGFR1 dependency in squamous cell lung cancer. Sci TranslatMed 2010;2:62ra93–3.

    7. Dutt A, Ramos AH, Hammerman PS, Mermel C, Cho J, Sharifnia T, et al.Inhibitor-Sensitive FGFR1 amplification in human non-small cell lungcancer. PLoS ONE 2011;6:e20351.

    8. Malchers F, Dietlein F, Schottle J, Lu X, Nogova L, Albus K, et al. Cell-autonomous and non-cell-autonomousmechanisms of transformation byamplified FGFR1 in lung cancer. Cancer Discov 2014;4:246–57.

    9. Nogova L, Sequist LV, Perez Garcia JM, Andre F, Delord J-P, Hidalgo M,et al. Evaluation of BGJ398, a fibroblast growth factor receptor 1-3kinase inhibitor, in patients with advanced solid tumors harboringgenetic alterations in fibroblast growth factor receptors: results of a

    global phase I, dose-escalation and dose-expansion study. J Clin Oncol2017;35:157–65.

    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 inhibi-tion in a translational clinical trial. Cancer Discov 2016;6:838–51.

    11. 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-fibroblastgrowth factor receptor inhibitor, in patients with advanced solid tumors.J Clin Oncol 2015;33:3401–8.

    12. Pearson A, Smyth E, Babina IS, Herrera-Abreu MT, Tarazona N, Peckitt C,et al. High-level clonal FGFR amplification and response to FGFR inhibi-tion in a translational clinical trial. Cancer Discov 2016;6:838–51.

    13. SosML, Michel K, Zander T, Weiss J, Frommolt P, Peifer M, et al. Predictingdrug susceptibility of non–small cell lung cancers based on genetic lesions.J Clin Invest 2009;119:1727–40.

    14. SosML, Michel K, Zander T, Weiss J, Frommolt P, Peifer M, et al. Predictingdrug susceptibility of non–small cell lung cancers based on genetic lesions.J Clin Invest 2009;119:1727–40.

    15. Zhao L, Au JL-S,WientjesMG.Comparisonofmethods for evaluating drug-drug interaction. Front Biosci 2010;2:241–9.

    16. Peifer M, Fern�andez-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al.Integrative genome analyses identify key somatic driver mutations ofsmall-cell lung cancer. Nat Genet 2012;44:1104–10.

    17. Fern�andez-Cuesta L, Peifer M, Lu X, Sun R, cacute LO, Seidel D, et al.Frequent mutations in chromatin-remodelling genes in pulmonary carci-noids. Nat Comm 2014;5:1–7.

    18. Gavine PR, Mooney L, Kilgour E, Thomas AP, Al-Kadhimi K, Beck S, et al.AZD4547: an orally bioavailable, potent, and selective inhibitor of the

    FGFR Inhibitor Resistance

    www.aacrjournals.org Clin Cancer Res; 23(18) September 15, 2017 5535

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • fibroblast growth factor receptor tyrosine kinase family. Cancer Res 2012;72:2045–56.

    19. Squires M, Ward G, Saxty G, Berdini V, Cleasby A, King P, et al. Potent,selective inhibitors of fibroblast growth factor receptor define fibroblastgrowth factor dependence in preclinical cancer models. Mol Cancer Ther2011;10:1542–52.

    20. Guagnano V, Furet P, Spanka C, Bordas V, Le Douget M, Stamm C, et al.Discovery of 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea (NVP-BGJ398), a potent and selective inhibitor of the fibroblast growthfactor receptor family of receptor tyrosine kinase. J Med Chem 2011;54:7066–83.

    21. Liao RR. Characterizing mechanisms of acquired resistance to targetedtherapy in FGFR1-amplified lung squamous cell carcinoma. Cancer Res2013;73:1538–7445.

    22. George J, Lim JS, Jang SJ, Cun Y, Ozreti�c L, Kong G, et al. Comprehensivegenomic profiles of small cell lung cancer. Nature 2015;524:47–53.

    23. Caunt CJ, Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs).FEBS J 2012;280:489–504.

    24. Ercan D, Xu C, Yanagita M, Monast CS, Pratilas CA, Montero J, et al.Reactivation of ERK signaling causes resistance to EGFR kinase inhibitors.Cancer Discov 2012;2:934–47.

    25. Manchado E, Weissmueller S, Morris JP, Chen C-C, Wullenkord R, Lujam-bio A, et al. A combinatorial strategy for treating KRAS-mutant lung cancer.Nature 2016;534:647–51.

    26. Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solidtumours: learning from lung cancer. Nature 2014;11:473–81.

    27. Medova M, Pochon B, Streit B, Blank-Liss W, Francica P, Stroka D,et al. The novel ATP-competitive inhibitor of the MET hepatocytegrowth factor receptor EMD1214063 displays inhibitory activityagainst selected MET-mutated variants. Mol Cancer Ther 2013;12:2415–24.

    28. Foucquier J, Guedj M. Analysis of drug combinations: current methodo-logical landscape. Pharmacol Res Perspect 2015;3:e00149.

    29. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al.MET amplification leads to gefitinib resistance in lung cancer by activatingERBB3 signaling. Science 2007;316:1039–43.

    30. Bean J, Brennan C, Shih JY, Riely G. MET amplification occurs withor without T790M mutations in EGFR mutant lung tumorswith acquired resistance to gefitinib or erlotinib. PNAS 2007;104:20932–37.

    31. The Clinical Lung Cancer Genome Project (CLCGP) and NetworkGenomic Medicine (NGM). A genomics-based classification of humanlung tumors. Sci Translat Med 2013;5:209ra153–3.

    Clin Cancer Res; 23(18) September 15, 2017 Clinical Cancer Research5536

    Malchers et al.

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/

  • 2017;23:5527-5536. Published OnlineFirst June 19, 2017.Clin Cancer Res Florian Malchers, Meryem Ercanoglu, Daniel Schütte, et al. Cancer

    -Amplified LungFGFR1Mechanisms of Primary Drug Resistance in

    Updated version

    10.1158/1078-0432.CCR-17-0478doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2017/06/17/1078-0432.CCR-17-0478.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://clincancerres.aacrjournals.org/content/23/18/5527.full#ref-list-1

    This article cites 31 articles, 10 of which you can access for free at:

    Citing articles

    http://clincancerres.aacrjournals.org/content/23/18/5527.full#related-urls

    This article has been cited by 2 HighWire-hosted articles. Access the articles at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected]

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/23/18/5527To request permission to re-use all or part of this article, use this link

    on June 2, 2021. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst June 19, 2017; DOI: 10.1158/1078-0432.CCR-17-0478

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-17-0478http://clincancerres.aacrjournals.org/content/suppl/2017/06/17/1078-0432.CCR-17-0478.DC1http://clincancerres.aacrjournals.org/content/23/18/5527.full#ref-list-1http://clincancerres.aacrjournals.org/content/23/18/5527.full#related-urlshttp://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/23/18/5527http://clincancerres.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice