h3b-6527 is a potent and selective inhibitor of ... · therapeutics, targets, and chemical biology...

16
Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4 in FGF19-Driven Hepatocellular Carcinoma Jaya Julie Joshi 1 , Heather Coffey 1 , Erik Corcoran 1 , Jennifer Tsai 1 , Chia-Ling Huang 1 , Kana Ichikawa 1 , Sudeep Prajapati 1 , Ming-Hong Hao 1 , Suzanna Bailey 1 , Jeremy Wu 1 , Victoria Rimkunas 1 , Craig Karr 1 ,Vanitha Subramanian 1 , Pavan Kumar 1 , Crystal MacKenzie 1 , Raelene Hurley 1 , Takashi Satoh 1 , Kun Yu 1 , Eunice Park 1 , Nathalie Rioux 1 , Amy Kim 1 , Weidong G. Lai 2 , Lihua Yu 1 , Ping Zhu 1 , Silvia Buonamici 1 , Nicholas Larsen 1 , Peter Fekkes 1 , John Wang 1 , Markus Warmuth 1 , Dominic J. Reynolds 1 , Peter G. Smith 1 , and Anand Selvaraj 1 Abstract Activation of the broblast growth factor receptor FGFR4 by FGF19 drives hepatocellular carcinoma (HCC), a disease with few, if any, effective treatment options. While a number of pan-FGFR inhibitors are being clinically evaluated, their application to FGF19-driven HCC may be limited by dose- limiting toxicities mediated by FGFR13 receptors. To evade the potential limitations of pan-FGFR inhibitors, we generat- ed H3B-6527, a highly selective covalent FGFR4 inhibitor, through structure-guided drug design. Studies in a panel of 40 HCC cell lines and 30 HCC PDX models showed that FGF19 expression is a predictive biomarker for H3B-6527 response. Moreover, coadministration of the CDK4/6 inhib- itor palbociclib in combination with H3B-6527 could effec- tively trigger tumor regression in a xenograft model of HCC. Overall, our results offer preclinical proof of concept for H3B-6527 as a candidate therapeutic agent for HCC cases that exhibit increased expression of FGF19. Cancer Res; 77(24); 69997013. Ó2017 AACR. Introduction Hepatocellular carcinoma (HCC), which accounts for up to 90% of all primary liver cancers, is a major health issue with an increasing rate of incidence and over 780,000 new cases diag- nosed globally each year (1). HCC is a heterogeneous disease with generally poor prognosis and sorafenib, a multikinase inhibitor, is the only approved systemic therapy for advanced HCC patients in the United States (2). Several multikinase inhibitors have been clinically tested in HCC but none have improved activity over sorafenib, and with limited additional treatment options, there is a need for more effective HCC therapies (3). Recent genomic studies of HCC have identied several oncogenic pathways including mutations in the TERT promoter, CTNNB1, TP53, and ARID1A (4). These studies have also identied focal amplication of 11q13.3 chromosomal locus that contains genes including broblast growth factor 19 (FGF19) and cyclin D1 (CCND1), suggesting FGF19 as a poten- tial driver oncogene in HCC (5). FGF19 is a gut-secreted endocrine hormone that acts in the liver through the receptor tyrosine kinase FGFR4 to regulate bile acid synthesis (6). Downstream signaling, upon activation of FGFR4 by FGF19, leads to the inhibition of hepatic enzyme cholesterol 7a-hydroxylase (CYP7A1) expression, resulting in reduced bile acid synthesis (6). This activation of FGFR4 by FGF19 requires the cofactor KLB, which is normally bound to FGFR4 (7). Consistent with the notion that FGF19 is a driver oncogene in HCC, trans- genic mice overexpressing FGF19 form liver tumors and genetic ablation of FGFR4 prevents tumor formation (8). In the onco- genic setting, where FGF19 can act in an autocrine/paracrine fashion, MAPK pathway activity and several proliferative genes including EGR1 and c-Fos are known to be regulated by FGF19FGFR4 signaling (9, 10). These data suggest targeting FGFR4 would have therapeutic benet in HCC with altered FGF19 signaling. While a number of pan-FGFR inhibitors are being clinically evaluated in a variety of indications, they display poor potency against FGFR4 potentially limiting their application to FGF19-driven HCC (11). Furthermore, pan-FGFR inhibitors with FGFR4 potency would likely be limited by their on-target FGFR13mediated dose-limiting toxicities (hyperphosphatemia and soft-tissue mineralization), suggesting a need to improve FGFR4 selectivity (11). Emerging studies show that selective inhibition of FGFR4 by antibody and small-molecule approaches are being pursued given the strong link between FGF19, FGFR4, and onco- genesis in HCC (9, 12). Here, we describe the identication of H3B-6527, a highly selective and covalent small-molecule inhib- itor of FGFR4 and provide preclinical evidence that HCC with increased FGF19 expression is sensitive to H3B-6527 monother- apy and CDK4/6 inhibitor combination therapy. H3B-6527 is currently undergoing evaluation in a phase I clinical trial in HCC (ClinicalTrials.gov Identier: NCT02834780). 1 H3 Biomedicine, Cambridge, Massachusetts. 2 Eisai, Andover, Massachusetts. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). J.J. Joshi, H. Coffey, and E. Corcoran contributed equally to this article. Corresponding Authors: Anand Selvaraj, H3 Biomedicine, 300 Technology Square, Cambridge, MA 02139. Phone: 617-252-5065; Fax: 617-252-5098; E-mail: [email protected]; and Peter G. Smith, [email protected] doi: 10.1158/0008-5472.CAN-17-1865 Ó2017 American Association for Cancer Research. Cancer Research www.aacrjournals.org 6999 on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 16-Jun-2020

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

Therapeutics, Targets, and Chemical Biology

H3B-6527 Is a Potent and Selective Inhibitor ofFGFR4 in FGF19-DrivenHepatocellular CarcinomaJaya Julie Joshi1, Heather Coffey1, Erik Corcoran1, Jennifer Tsai1, Chia-Ling Huang1,Kana Ichikawa1, Sudeep Prajapati1, Ming-Hong Hao1, Suzanna Bailey1, Jeremy Wu1,Victoria Rimkunas1, Craig Karr1,Vanitha Subramanian1, Pavan Kumar1, Crystal MacKenzie1,Raelene Hurley1, Takashi Satoh1, Kun Yu1, Eunice Park1, Nathalie Rioux1, Amy Kim1,Weidong G. Lai2, Lihua Yu1, Ping Zhu1, Silvia Buonamici1, Nicholas Larsen1, Peter Fekkes1,JohnWang1, MarkusWarmuth1, Dominic J. Reynolds1, Peter G. Smith1, andAnand Selvaraj1

Abstract

Activation of the fibroblast growth factor receptor FGFR4 byFGF19 drives hepatocellular carcinoma (HCC), a disease withfew, if any, effective treatment options. While a number ofpan-FGFR inhibitors are being clinically evaluated, theirapplication to FGF19-driven HCC may be limited by dose-limiting toxicities mediated by FGFR1–3 receptors. To evadethe potential limitations of pan-FGFR inhibitors, we generat-ed H3B-6527, a highly selective covalent FGFR4 inhibitor,through structure-guided drug design. Studies in a panel of

40 HCC cell lines and 30 HCC PDX models showed thatFGF19 expression is a predictive biomarker for H3B-6527response. Moreover, coadministration of the CDK4/6 inhib-itor palbociclib in combination with H3B-6527 could effec-tively trigger tumor regression in a xenograft model of HCC.Overall, our results offer preclinical proof of concept forH3B-6527 as a candidate therapeutic agent for HCC casesthat exhibit increased expression of FGF19. Cancer Res; 77(24);6999–7013. �2017 AACR.

IntroductionHepatocellular carcinoma (HCC), which accounts for up to

90% of all primary liver cancers, is a major health issue with anincreasing rate of incidence and over 780,000 new cases diag-nosed globally each year (1). HCC is a heterogeneous diseasewith generally poor prognosis and sorafenib, a multikinaseinhibitor, is the only approved systemic therapy for advancedHCC patients in the United States (2). Several multikinaseinhibitors have been clinically tested in HCC but none haveimproved activity over sorafenib, and with limited additionaltreatment options, there is a need for more effective HCCtherapies (3). Recent genomic studies of HCC have identifiedseveral oncogenic pathways including mutations in the TERTpromoter, CTNNB1, TP53, and ARID1A (4). These studies havealso identified focal amplification of 11q13.3 chromosomallocus that contains genes including fibroblast growth factor 19(FGF19) and cyclin D1 (CCND1), suggesting FGF19 as a poten-tial driver oncogene in HCC (5).

FGF19 is a gut-secreted endocrine hormone that acts in the liverthrough the receptor tyrosine kinase FGFR4 to regulate bile acidsynthesis (6). Downstream signaling, upon activation of FGFR4by FGF19, leads to the inhibition of hepatic enzyme cholesterol7a-hydroxylase (CYP7A1) expression, resulting in reduced bileacid synthesis (6). This activation of FGFR4 by FGF19 requires thecofactor KLB, which is normally bound to FGFR4 (7). Consistentwith the notion that FGF19 is a driver oncogene in HCC, trans-genic mice overexpressing FGF19 form liver tumors and geneticablation of FGFR4 prevents tumor formation (8). In the onco-genic setting, where FGF19 can act in an autocrine/paracrinefashion, MAPK pathway activity and several proliferative genesincluding EGR1 and c-Fos are known to be regulated by FGF19–FGFR4 signaling (9, 10). These data suggest targeting FGFR4would have therapeutic benefit in HCC with altered FGF19signaling. While a number of pan-FGFR inhibitors are beingclinically evaluated in a variety of indications, they display poorpotency against FGFR4 potentially limiting their application toFGF19-driven HCC (11). Furthermore, pan-FGFR inhibitors withFGFR4 potency would likely be limited by their on-target FGFR1–3–mediated dose-limiting toxicities (hyperphosphatemia andsoft-tissue mineralization), suggesting a need to improve FGFR4selectivity (11). Emerging studies show that selective inhibition ofFGFR4 by antibody and small-molecule approaches are beingpursued given the strong link between FGF19, FGFR4, and onco-genesis in HCC (9, 12). Here, we describe the identification ofH3B-6527, a highly selective and covalent small-molecule inhib-itor of FGFR4 and provide preclinical evidence that HCC withincreased FGF19 expression is sensitive to H3B-6527 monother-apy and CDK4/6 inhibitor combination therapy. H3B-6527 iscurrently undergoing evaluation in a phase I clinical trial in HCC(ClinicalTrials.gov Identifier: NCT02834780).

1H3 Biomedicine, Cambridge, Massachusetts. 2Eisai, Andover, Massachusetts.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

J.J. Joshi, H. Coffey, and E. Corcoran contributed equally to this article.

Corresponding Authors: Anand Selvaraj, H3 Biomedicine, 300 TechnologySquare, Cambridge,MA02139. Phone: 617-252-5065; Fax: 617-252-5098; E-mail:[email protected]; and Peter G. Smith,[email protected]

doi: 10.1158/0008-5472.CAN-17-1865

�2017 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 6999

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

Materials and MethodsH3B-6527

The synthetic procedure for the preparation of H3B-6527 canbe found in patent application WO 2015057938 (compound108, pages 42–45). Detailed structure–activity relationship stud-ies will be described elsewhere.

CCLE scatter plotThe log2-transformed mRNA expression of FGF19, FGFR4, and

KLB of 24 CCLE HCC cell lines were purchased from Omicsoft(http://www.omicsoft.com/).

RNA-seq, cluster, and pathway analysisThe RNA-seq data was generated by the Illumina HiSeq 4000

RNA Sequencing platform. The fastq files were aligned to bothhuman (GRCh37) and the mouse genomes (GRCm38) usingSTAR (13) and the aligned BAM files were processed by the in-house xenotool to remove mouse reads and generate the highquality human BAM files. The transcripts abundance was thenestimated using Kallisto (14). The RNA-seq data was normalizedand log2 transformed in R v3.2.3 statistical environment (http://www.r-project.org) using edgeR v3.16.5 Bioconductor library(15) with trimmed mean of M values (TMM) normalization. Theexpression profiles of 24-hour time points were used in the geneexpression analysis. We excluded nonprotein–coding genes andfiltered out low expressed genes [genes with log2 (tpm) < 2 in allsamples] as well as low variation genes (genes with variation < 0.1across all samples). A total of 5,843 genes were left for the geneexpression analysis. For each treatment group, the genes wereranked by fold changes of the treatment comparing with vehicleand the Gene Set Enrichment Analysis preranked was performedusing the ranked gene list to obtain the enriched hallmark genesets (H collection in v5.2 MSigDB) in each group (16, 17).Hallmarks with nominal P < 0.01 in at least one treatment groupwere reported.

Kinase assay and competition binding assayFor kinase assay, the His-TEV-FGFR4 kinase (447–753) was

cloned into pFastBac1 (Thermo Fisher Scientific) and expressed inSF9 insect cells. Insect cells were infected with baculovirus at amultiplicity of infection (MOI) of approximately 10 and har-vested after 72 hours. Protein was purified by Ni-NTA chroma-tography followed by size-exclusion chromatography with asephacryl S-300 column equilibrated in 20 mmol/L Tris pH 8,20 mmol/L NaCl, and 2 mmol/L TCEP. Peak fractions werepooled and flash frozen in liquid N2. Active FGFR1 kinase waspurchased (Thermo Fisher Scientific). IC50 measurements weremade using a FRET-based assay using Lance ULight poly-GT as asubstrate (PerkinElmer). All measurements were performed atroom temperature and run in the presence of 20 pmol/L FGFR1 or300 pmol/L FGFR4 kinase in a reaction buffer containing20 mmol/L HEPES pH 7.5, 10 mmol/L MgCl2, 1 mmol/L EGTA,0.2% Brij35, 0.2 mg/mL BSA, 1� Roche phosphatase inhibitors,and 2 mmol/L DTT. FGFR1 and FGFR4 were preincubated for1 hour with inhibitor and the kinase reaction was initiatedby addition of poly-GT substrate (100 nmol/L) and ATP(50 mmol/L). Reactions were quenched after 1 hour in Lancedetection buffer supplemented with 10 mmol/L EDTA. Substrateconversion was monitored using the LANCE Eu-W1024-antiphosphotyrosine antibody (2 nmol/L) with an Envision plate

reader. Curves were fit using GraphPad Prism software. Forcompetition binding assay, the DiscoverX kinome scan servicewas utilized (https://www.discoverx.com/services/drug-discovery-development-services/kinase-profiling/kinomescan).

Mass spectrometryFor mass spectrometry experiments, 3 mmol/L FGFR4 was

incubated overnight at 4�C with 10 mmol/L H3B-6527 in 50mmol/L HEPES pH 7.5 and 100 mmol/L NaCl. The protein wasdesalted and concentratedmanually using anOPTI-TRAP column(Optimize Technologies) and eluted in 50% acetonitrile in 0.1%HCOOH. Samples were analyzed by nanocapillary liquid chro-matography mass spectrometry on an Easy-nLC 1000 HPLCsystem coupled to a QExactive mass spectrometer (Thermo Sci-entific), using an easy spray microflow emitter (Thermo Scientif-ic). The intact protein is eluted at a flow rate of 300 nL per minutewith a step gradient starting at 5% and finishing at 50% of buffer(100% acetonitrile in 0.1% formic acid) over 25 minutes. Datawas analyzed using BioPharma Finder 1.0 (Thermo Scientific)using amass tolerance of 0.05 kDa and a cut-off threshold of 15%.

CrystallographyTheHis-TEV-FGFR1-Y563C kinase (458–765)was coexpressed

with l-phosphatase in E. coli using a bicistronic construct clonedinto pET-28a (EMD Millipore). In this construct, the P-loopmutation C488A and C-lobe mutation C584S were included toimprove protein behavior and yields (18). Soluble protein wasobtained by overnight induction at 20�C using 0.1mmol/L IPTG.Cells were harvested and protein was purified using Ni-NTAchromatography followed by overnight TEV cleavage of theHis-tag and a polishing subtractive Ni-NTA step to remove theTEV. The flow through was concentrated and injected on asephacryl S-300 column equilibrated in 20 mmol/L Tris pH 8,20 mmol/L NaCl, and 2 mmol/L TCEP. Peak fractions werepooled and concentrated to approximately 14 mg/mL and flashfrozen in liquid N2. Crystals were obtained at 4�C from 2 þ 2 mLhanging drops equilibrated over a reservoir containing 14%–19%PEG10K, 300mmol/L (NH4)2SO4, 100mmol/LMESpH6.5, and5% ethylene glycol (19). They grew to full size in 1–3 weeks andwere then soaked for 2 weeks in a 10-mL drop containing 9.5 mL ofthe reservoir solution and 0.5 mL of 30 mmol/L H3B-6527formulated in 90% DMSO. The final soaking solution was there-fore 1.5 mmol/L compound with 4.5% DMSO. Crystals werefrozen in the soaking solution supplemented with 20% ethyleneglycol and data were collected at the Advanced Photon Source,LS-CAT 21-ID-G (Supplementary Table S1). The structure wassolved bymolecular replacement usingMOLREP (20) and refinedusing Refmac (21) with ligand coordinates generated using JLi-gand (22). Figure 1D was generated using Chimera (23). Coor-dinates have been deposited in the Protein Data Bank withaccession code 5VND.

Cell culture and compound treatmentHep3B cells (purchased from ATCC), weremaintained in Eagle

minimum essential medium (ATCC, 30-2003) with 10% FBS.JHH-7 cells (JHH-7 was purchased from Japan Health SciencesFoundation) were maintained in William E Medium (ThermoFisher Scientific, 12551-032) with 10% FBS. Both cell lines wereincubated in ahumidified incubator at 37�Cwith 5%CO2. For thedose response, time course, and wash out experiments, cells wereharvestedduring the logarithmic growthphase, seeded into 6-well

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7000

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

plates (2-mL media/well), and incubated until they reached 80%confluence. Compounds were dissolved in DMSO to yield stockconcentrations of 10 mmol/L. DMSO was used as the diluent forall the compounddilutions. Thefinal percentage ofDMSO in eachwell was 0.1% to 0.2%.Wash out was conducted by aspirating thetreatmentmedia from the cells, washing the cells with freshmediathree times, and thenmaintaining the cells in freshmedia. The 40HCC cell line panel and 625 cell line panel was conducted usingthe services of Crown Bioscience (http://www.crownbio.com/oncology/in-vitro-services) and Chempartner (http://www.chempartner.com/biology/cell-biology/cancer-cell-line-panel-screen),respectively. The authenticity of cell lines was confirmed by STR(short tandem repeat) analysis provided by both companies.

CYP7A1 measurementsRNA was extracted and purified from 50 to 100 mg tumor

fragments using Tri reagent (Ambion) and Omni Bead Ruptor.cDNA synthesis and qRT-PCR was performed using SuperScriptEnzymeMix (Thermo Fisher Scientific) and TaqManGene Expres-sion kit. The assay was performed on the ViiA7TM Real-Time PCRSystem (Thermo Fisher Scientific). qRT-PCR data were analyzedusing the DDCt method, where each sample was first normalizedby subtracting the 18S endogenous control Ct value from theCYP7A1 Ct value, then subtracting that value from the averagevehicle control Ct value for that time point. Fold increase wasdetermined by raising 2 to the DDCt value. Results were graphedusing GraphPad Prism. Data are expressed as mean � SEM.

Western blottingTo extract protein from the cells forWestern blotting, the 6-well

plateswere placed on ice, themediawere aspirated from thewells,and the cells were washed two times with cold 1� PBS. Cells weredirectly harvested with sample buffer (2� NuPAGE LDS samplebuffer, 2�NuPAGE reducing agent, 2�HALT protease inhibitor,2�HALT phosphatase inhibitor, and 2� EDTA), probe sonicatedat 10% amplitude for 10 seconds, and then heated at 70�C for 10minutes. Samples were loaded and resolved on 4%–15% Crite-rion TGX precast gels (Bio-Rad). Proteins were transferred ontonitrocellulose membrane using the IBlot 2 Dry Blotting system(Life Technologies). Blots were blocked for 1 hour in LI-CORblocking buffer and probed overnight on a shaker at 4�C withpERK1/2 (Thr202/Tyr204; D13.14.4E) XP Rabbit mAb (CellSignaling Technology, 4370) and the antibody was diluted1:1,000. After washing three times with 1� PBS-T, they wereprobed with IRDye 800CW Donkey anti-Rabbit IgG (HþL) sec-ondary (LI-COR), diluted 1:10,000, for 1 hour at room temper-ature with shaking. Blots were washed again three times with 1�PBS and then imaged with the Odyssey Infrared Imaging System.The blots were reprobed with p44/42 MAPK (ERK1/2; 3A7)Mouse mAb (Cell Signaling Technology, 9107) similar to above,followed by IRDye 680LT Goat anti-Mouse IgG (HþL) secondary(LI-COR Biosciences), and imaged again.

Viability assay and caspase assayFor both assays, cells were harvested during the logarithmic

growth phase and seeded into 96-well plates. Compounds weredissolved in DMSO to yield stock concentrations of 10 mmol/L.DMSO was used as the diluent for all the compound dilutions.Thefinal percentage ofDMSO in eachwell was 0.1%. The viabilityassay was performed 72 hours after compound treatment byadding 50 mL of CellTiter-Glo Viability Assay reagent (Promega)

and caspase assay was performed 24 hours posttreatment byadding 100 mL of Caspase-Glo 3/7 Reagent (Promega) to eachwell. Plates were shaken for 2minutes on amicroplate shaker andluminescencemeasured on the Envision plate reader. For viabilityassay, GI50 (the concentration of test article at which cell growth isinhibited by 50% compared with vehicle-treated cells) was cal-culated according to the publications from NCI (24). Data forcaspase assay are expressed relative to DMSO-treated cells.

IHCFive-micron sections of formalin-fixed paraffin-embedded

xenograft tissue samples were stained for pERK1/2 (Thr202/Tyr204; Cell Signaling Technology, catalog no. 4370) expressionusing a BOND-MAXTM Autostainer (Leica Microsystems) and aLeica ST5020 Multistainer (Leica Microsystems). Briefly, slideswere baked at 60�C for 30 minutes, deparaffinized with BondDewax solution, and rehydrated. Heat-mediated epitope retrievalwas performed in Leica Bond ER1 retrieval at 100�C for 20minutes. Endogenous peroxidase activity was blocked with 3%H2O2 for 15 minutes. Slides were incubated with primary anti-body (pERK1/2 final concentration 1.25 mg/mL) for 2 hours.Immunostaining was detected with Polymer HRP reagent anddeveloped with the DAB Refine chromogen. Slides were counter-stained with hematoxylin, dehydrated, andmounted withMicro-mount medium. The whole slides were digitized using the Scan-Scope XTTM (Aperio Technologies/Leica Biosystems) and imageanalysis was performed using the Aperio ImageScopeTM softwareversion 11.1.2.760 (Aperio Technologies/Leica Biosystems). ForFGF19 IHC, 5-mm slides were stained on the Leica Bond Rx withFGF19 D1N3R mAb (Cell Signaling Technology, catalog no.83348) similarly to the pERK1/2 IHC protocol described above.FGF19-positive tissues are defined as those staining with � 1%positive tumor cells.

FGF19 ELISAFGF19 levels weremeasured in the supernatants (150mL) using

a commercial human FGF19 ELISA kit as directed by the manu-facturer (BioVendor). In the event, the signal was beyond thelinear range of the assay, the supernatantwas diluted, and thefinalconcentration was corrected for the dilution.

Preparation of H3B-6527, palbociclib, and sorafenib for in vivoexperiments

H3B-6527 was formulated in 0.5% methylcellulose (Sigma,CAS: 9004-67-5) and 0.2% Tween80 (Sigma, catalog no. P4780).Palbociclib was formulated in 25 mmol/L sodium bicarbonateand 15 mmol/L lactic acid solution in distilled water, with 2%Cremaphor. Sorafenib was formulated in equal parts Cremaphorand ethanol as the stock solution. Before use, the stockwas dilutedin sterile water at 1:3.

Subcutaneous xenograft generation, dosing, and measurementof antitumor activity

The BALB/c nu/nu female mice approximately 8-week-old,weighing 18–20 g were obtained from Jackson Laboratory. Forthe human hepatocellular cancer cell line Hep3B xenografts,Hep3B cells were cultured in RPMI1640 medium containing10% FBS at 37�C in a 5% CO2 atmosphere and kept in expo-nential growth phase. The human hepatocellular cancer line JHH-7 was cultured in William E medium with 10% FBS (GIBCO10099) at 37�C in a 5%CO2 atmosphere and kept in exponential

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7001

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

Figure 1.

Biochemical and structural characterization of H3B-6527. A, Kinome profiling of H3B-6527 at 10 mmol/L with 395 kinases. Red circles, areas of activity as apercentage of control. B, Eleven-point H3B-6527 dose–response curves for kinases identified from the kinome profiling. C, Intact mass spectrometryshows that the untreated protein (top) is 100% modified (bottom) in the presence of H3B-6527. The chemical structure is shown in the inset. D, Crystalstructure of H3B-6527 bound in the ATP pocket (PDB 5VND). H-bonds are shown as dashed lines. The refined electron density map (2Fo-Fc) contoured at1.0 s is continuous between the ligand and the hinge cysteine (C563). The residues (P483-V493) of the p-loop were cropped for clarity.

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7002

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

growth phase. For harvesting, the cells were washed with PBS,incubated with 0.25% trypsin-EDTA, and suspended in a 1:1mixture of RPMI1640 medium containing 10% FBS andMatrigel(Corning) at a final concentration of 5 � 107 cells/mL for Hep3Band 2.5� 107 cells/mL for JHH-7. To generate xenografts, 0.1-mLof the inoculum was injected subcutaneously into the right flankregion ofmice, giving afinal concentration of 5� 106 cells/mousefor Hep3B and 2.5� 106 cells/mouse for JHH-7. When the meantumor volume (TV) reached approximately 170 mm3, mice weresorted into treatment groups with 8 animals per group. H3B-6527, palbociclib, or the combination was administered orallyeither as once-daily or twice-daily schedule as indicated in thefigures. The 30 patient-derived xenograft (PDX)models' one-doseefficacy study and the 4PDXmodels' dose–response efficacy studywere conducted using 3 animals and 8 animals per group, respec-tively. For these PDX studies, 6- to 8-week-old femaleNu/Numiceweighing 16–19 g were purchased from Beijing Vital River Lab-oratory Animal Technology Co. Ltd and housed under specificpathogen-free (SPF) conditions. All PDX efficacy studies wereconducted using the contract research organization servicesoffered by Shanghai ChemPartner (25). The authenticity of thePDX models was verified using the pathology analysis providedby Chempartner.

Body weights were measured daily and tumor measurementswere performed twice weekly. Mice with >20% body weight losscomparedwith their day 1bodyweight or bearing tumorswith thelongest diameter >2,000 mm were immediately euthanized toprevent any pain or suffering for the animal as according toInstitutional Animal Care and Use Committee guidelines definedby the H3 Biomedicine Animal Care and Use Program and studyprotocol. The tumor volume (TV) in mm3 was calculated accord-ing to the following formula: TV¼ length� width2 � 0.5 length:largest diameter of tumor (mm)width: diameter perpendicular tolength (mm). The tumor growth inhibition % (TGI) was calcu-lated according to the following formula: Tumor growth inhibi-tion%(TGI)¼ [(Average control TVdayX� Treatment TVdayX)/(Average control TV day X)] � 100, where day X is any day of thetreatment.

The antitumor effects of the treatment, partial (PR) andcomplete regression (CR), stable (SD), and progressive (PD)disease were defined as follows, PD: 3 consecutive measure-ments >120% of starting volume or 3 consecutive increasingmeasurements from best response; SD: 3 consecutive measure-ments >50% and <120% of starting volume; PR: 3 consecutivemeasurements <50% of starting volume; CR: 3 consecutivemeasurements <30 mm3.

Orthotopic xenograft generation, luciferin injection, andimaging

Hep3B cells stably expressing the Luciferase gene (Hep3B-Luc)were inoculated at the left lobe of the liver of female BALB/c nudemice. Hep3B-Luc cells (1.25 � 106) in 25-mL PBS supplementedwith 50% Matrigel were used for inoculation. Bioluminescentimagingwas performedon thefirst day of the study and continuedon a weekly basis. Treatment with H3B-6527 at various dosesgiven orally twice a daywas started 12days after inoculationwhenthe mean signal intensity was 9.68 � 106 photons/second. Lucif-erin injections were prepared by dissolving 15mg/mL of luciferin(D-Luciferin, Firefly, potassium salt) in DPBS and sterile filteredthrough a 0.2-mm filter. Each mouse received 150 mg/kgof luciferin by intraperitoneal injection. After 10 minutes,

anesthetized mice were placed in the imaging machine (XenogenIVIS Lumina XR) and imaged ventrally to detect luminescencesignal. Exposure time was 60 seconds. Images were quantified asphotons/ second using the Living Image software.

ResultsH3B-6527 is a potent, selective covalent FGFR4 inhibitor

The ATP-binding domains of the FGFR family are known tobe highly homologous (26). We sought to gain selectivity forthe FGFR4 paralog by targeting Cys552, present in the hingeregion of the ATP-binding pocket, unique within the FGFRfamily and found in only 6 other kinases. We evaluatedwhether an electrophilic small molecule that covalentlybound Cys552 would be selective for FGFR4 versus otherfamily members and across the kinome. Using a structure-based design approach focusing on kinase hinge-bindingmotifs, we discovered templates that present an appropriatevector for attachment of an electrophile to engage Cys552(27, 28). After iterative rounds of design and synthesis, wechose to focus on an N-aryl-N0-pyrimidin-4-yl urea scaffold,where the introduction of an acrylamide to the ortho-positionof the N-aryl substituent provided a compound capable offorming a covalent bond with Cys552 in FGFR4, capturing theresidue via hetero-Michael addition of the free thiol. Subse-quent optimization of potency and physicochemical proper-ties led to the incorporation of dichlorodimethoxyphenyl andN-ethyl piperazine substituents, ultimately leading to thediscovery of H3B-6527.

H3B-6527 was tested at 10 mmol/L against 395 distinctkinases in a competition binding assay that revealed FGFR4as the top hit (Fig. 1A). Six other kinases, FGFR1-3, TAOK2,JNK2, and CSF1R were also identified with varying bindingcapacity to H3B-6527 (Fig. 1A). A follow up functional bio-chemical assay demonstrated robust inhibition of the targetkinase FGFR4 with an IC50 value of <1.2 nmol/L and at least250-fold selectivity over FGFR1-3 (IC50 values of 320, 1,290and 1,060 nmol/L respectively; Fig. 1B). TAOK2, JNK2, andCSF1R were also less sensitive to H3B-6527 treatment with IC50

values of 690, >10,000, and >10,000 nmol/L, respectively(Fig. 1B). Protein mass spectrometry can provide a directreadout of potential covalent modification of a protein andthe stoichiometry of the interaction. When incubated withH3B-6527, FGFR4 underwent 100% monoalkylation, confirm-ing the stoichiometry and covalency of the binding mode(Fig. 1C). The complex was further characterized using a FGFR1surrogate crystallization system in which the key hinge residueTyr563 was mutated to cysteine. This simple alteration inFGFR1 effectively mirrors the ATP-binding pocket in FGFR4and was used to interrogate the binding modes of FGFR4inhibitors (to be described elsewhere). The complex withH3B-6527 was determined at 2.1 Å resolution (Fig. 1D; Sup-plementary Table S1). Consistent with mass spectrometry data,the continuous electron density between the thiolate of Cys563and the acrylamide of H3B-6527 demonstrates the formationof a covalent bond (Fig. 1D). The structure shows how exploi-tation of this unusual hinge Cys interaction enables H3B-6527to achieve >250-fold selectivity over FGFR1. This selectivityarises not only from the unique engagement of the cysteine butalso from the steric clash that would occur between the acryl-amide and the Tyr residue present at the corresponding

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7003

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

position in FGFR1–3. The compound binds the ATP-bindingpocket like other type 1 kinase inhibitors, including the bindingmode observed for the covalent FGFR4 inhibitor BLU9931 (9).The N-aryl-N0-pyrimidin-4-yl core forms two hydrogen bondswith the hinge (Fig. 1D), while the dichlorodimethoxyphenylmoiety anchors the compound in the back pocket next to thegatekeeper Val561, Lys514, and Asp641. The solubilizing ethyl-piperazinyl group is located at the solvent accessible region asanticipated. Thus, these data provide a structural rationale forthe potency and selectivity of H3B-6527.

H3B-6527 inhibits FGFR4 signaling, proliferation, and leads toapoptosis in a HCC cell line

As a first step to understand the effect of H3B-6527 on HCCcell lines, we probed the FGFR4 signaling pathway in a HCCcell line, Hep3B. This cell line overexpresses FGF19 as a result ofFGF19 gene amplification and is a suitable model to studyFGF19–FGFR4 signaling (29). Signaling through FGFR4 leadsto phosphorylation and activation of extracellular-signal-regulated kinases (ERK1/2), and also leads to downregulationof CYP7A1 mRNA levels. Thus, both pERK1/2 (Thr202/Tyr204)and CYP7A1 can serve as pharmacodynamic markers for FGFR4pathway activity (9). First, we tested several concentrations ofH3B-6527 in a 24-hour treatment and found a robust increase(approximately 7-fold in comparison with DMSO vehicle) inCYP7A1 transcripts, as measured by qRT-PCR, at 1 nmol/L (Fig.2A). Higher concentrations of 10, 100 and 1,000 nmol/L led togreater than a 100-fold increase in CYP7A1 transcripts levels,suggesting strong inhibition of the FGFR4 pathway by H3B-6527 (Fig. 2A). Next, we tested the acute effects of 1-hourtreatment of H3B-6527 and measured pERK1/2 levels by West-ern blotting. The levels of pERK1/2 decreased in a dose-dependent manner with maximal inhibition occurring at100 nmol/L (Fig. 2B). To evaluate the kinetics of FGFR4inhibition, Hep3B cells were treated with H3B-6527 at 100and 300 nmol/L for 0.5, 1, 2, 4, 8, and 24 hours and pERK1/2levels were measured. A rapid decrease in pERK1/2 levels wasobserved with maximal inhibition at 2 hours at both concen-trations and a modest recovery of levels was observed at 8 and24 hours possibly due to pathway rewiring (Fig. 2C). Toevaluate the covalent binding properties of H3B-6527 to FGFR4in cells, Hep3B cells were treated with 1 mmol/L H3B-6527 for 1hour and then washed several times with fresh media to removethe compound. The levels of pERK1/2 were measured up to 24hours post-wash and H3B-6527 showed continued inhibitionup to 8 hours consistent with covalent interaction and irre-versible inhibition of FGFR4 (Fig. 2D). This was in contrast tothe treatment of a noncovalent pan-FGFR inhibitor, PD173074,where pERK1/2 levels recovered to vehicle (H3B-6527 untreat-ed) levels as early as 1 hour post-wash. To test the antiproli-ferative properties of H3B-6527, Hep3B cells were treated for72 hours with increasing concentrations and their growth wasmeasured by an ATP-based cell viability assay. H3B-6527treatment resulted in a GI50 value (concentration of inhibitorthat led to a 50% reduction in viability) of 25 nmol/L (Fig. 2E).Furthermore, H3B-6527 treatment of Hep3B cells led to robustactivation of caspase-3/7, an apoptotic marker, in a concentra-tion-dependent manner, indicating FGFR4 inhibition by H3B-6527 leads to cell death in HCC cell lines (Fig. 2F). In summary,these data show that H3B-6527 inhibits proliferation and leadsto apoptosis in a HCC cell line by inhibiting FGFR4 signaling.

Pharmacokinetics/pharmacodynamics and antitumor activityofH3B-6527 in theHep3BhumanHCCxenograftmousemodel

As H3B-6527 has shown FGFR4 pathway modulation inHep3B cells in vitro, we evaluated the pharmacokinetic andpharmacodynamic properties of H3B-6527 in Hep3B xenograftsgrown in female nude immunocompromised mice. Three doselevels of H3B-6527 (30, 100, and 300 mg/kg) and a controlvehicle group were tested in this study. The study was designedsuch thatfive repeat oral doses ofH3B-6527were givenon a twice-daily (a dose every 12 hours) schedule and, on day 3, plasma andtumor samples were collected at designated time points andanalyzed for H3B-6527 parent compound concentration. H3B-6527 showed dose-proportional plasma exposures and greaterthan dose-proportional tumor exposures within the dose rangeevaluated (Fig. 3A). A dose-dependent increase in tumor CYP7A1mRNA levels was observed and the maximum levels for the 30,100, and 300 mg/kg dose groups were 46-fold (at 4 hours), 180-fold (at 4 hours), and 509-fold (at 24 hours), respectively, relativeto the vehicle levels (Fig. 3A). For the 30 and100mg/kgH3B-6527dose groups, the tumor CYP7A1 levels peaked at 4 hours and thendecreased with time. For the 300 mg/kg dose group, tumorCYP7A1 levels remained elevated across each time point (Fig.3A). Thus, the increase of CYP7A1 levels induced by H3B-6527was dose-dependent, with higher doses leading to a more sus-tained response. Inhibition of pERK1/2 (Thr202/Tyr204) byH3B-6527was also dose dependent, with higher doses leading tomoresustained inhibition (Fig. 3B). For the 30 and 100 mg/kg dosegroups, the peak inhibition of pERK1/2 was observed at 4 hoursand for the 300mg/kg the peak inhibitionwas observed already at1 hour after administration of H3B-6527 (Fig. 3B). Followingpeak inhibition, the pERK1/2 levels recovered rapidly in the30 mg/kg group while a slower recovery was observed in the100 mg/kg group probably due to H3B-6527 clearance (Fig. 3A).No pERK1/2 recovery was seen in the 300 mg/kg group and thelevels remained inhibited across each time point likely due to thepresence of high levels of H3B-6527 even at later time points(Fig. 3A). In summary, the H3B-6527 pharmacodynamicresponse as measured by CYP7A1 mRNA and pERK1/2 proteinlevels was dose dependent, with higher doses leading to sustainedresponses (Fig. 3A and B).

To test the antitumor properties of H3B-6527, nude micebearing Hep3B subcutaneous xenografts were treated orally twicedaily for 15 days. Treatment with H3B-6527 inhibited Hep3Bxenograft growth, with the 300 and 100 mg/kg twice-daily dosessignificantly inhibiting growth and inducing CR or PR in 8 of 8and6of 8ofmice, respectively (TGIof 115and109%,P<0.05, forthe 300 and 100 mg/kg treatment groups, respectively; Fig. 3C).Treatment with 30 mg/kg twice-daily H3B-6527 did not signif-icantly inhibit xenograft growth (TGI of 37%, P > 0.05; Fig. 3C).The vehicle-treated group lost approximately 10% body weight(BW) at day 15 although this BW loss is likely a result of HEP3Btumor-induced cachexia (Fig. 3D; ref. 9). The BW loss induced byHep3B xenograftswas prevented at 300 and100mg/kgH3B-6527where tumor regression was observed on treatment (Fig. 3D). Alldose levels of H3B-6527 were well tolerated as no drug-inducedBW loss was observed on treatment (Fig. 3D). Next, we tested theantitumor activity of H3B-6527 in an orthotopic liver tumorxenograft model, which is thought to provide a more clinicallyrelevant setting than subcutaneous models (30). Nude micebearing Hep3B orthotopic xenografts in liver were treated orallyon a twice-daily schedule for 28 days and tumor growth was

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7004

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 7: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

measured by the luciferase reporter that has been stably expressedin Hep3B cells. Treatment with H3B-6527 significantly inhibitedtumor growth at the 300 and 100 mg/kg twice-daily dose (Sup-plementary Fig. S1A). Treatment with 30 and 10 mg/kg did notinhibit tumor growth in this study. All dose levels were well

tolerated as no drug-induced BW loss was observed (Supplemen-tary Fig. S1B). In summary, these data show that oral treatment ofH3B-6527, twice daily, inhibited xenograft growth in a dose-dependentmanner in nudemice,with the 300or 100mg/kg twicedaily, significantly inhibiting tumor growth in both Hep3B

Figure 2.

H3B-6527 effects in the HCC cell line Hep3B in vitro. A, Relative expression of CYP7A1 24 hours post H3B-6527 treatment at indicated doses. B, pERK1/2(Thr202/Tyr204) levels 1 hour post H3B-6527 treatment at indicated doses. C, pERK1/2 (Thr202/Tyr204) levels following 100 and 300 nmol/L H3B-6527 atindicated time points. D, pERK1/2 (Thr202/Tyr204) levels at indicated time points following washout of H3B-6527 and PD173074. Initial treatment of bothcompounds was for 1 hour, denoted by "0 hour." ERK1/2 levels served as loading control for B–D. E, Representative H3B-6527 dose–response curve for ATP-basedcell viability assay. F, Representative H3B-6527 dose–response curve of caspase-3/7–based apoptosis assay.

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7005

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 8: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

subcutaneous andorthotopic xenograftmodel and causing tumorregressions in the subcutaneous xenograft model.

FGF19 expression is a predictor of H3B-6527 sensitivityHaving established the sensitivity of the Hep3B HCCmodel to

H3B-6527, we sought to comprehensively study the sensitivity ofH3B-6527 by utilizing multiple HCC models. First, 40 HCC celllines were treated with increasing concentrations of H3B-6527and growth inhibition was measured by cell viability assay. Anycell line with a GI50 value of <2,000 nmol/L was considered asensitive cell line to H3B-6527 and four cell lines met thiscriterion: Hep3B, SNU-878, Huh-7, and JHH-7 (SupplementaryTable S2). CCLE RNA-seq data were available for 24 of the 40 celllines including the four sensitive lines (31, 32). Examination ofthese data revealed that the four H3B-6527–sensitive cell linesexpress high levels of FGF19 mRNA (Supplementary Fig. S2;

ref. 9). We evaluated the FGF19 protein levels of these 24 celllines and found the same four cell lines express the highest levelsof FGF19 in culture media (Fig. 4A). We measured the FGF19protein levels in the remaining 16HCC cell lines that did not haveRNA-seq data and found they failed to produce any detectablelevels of FGF19 protein (Fig. 4A). Among these 40HCC lines, onecell line, Huh-1, produced detectable butmuch lower FGF19 thanthe above mentioned 4 cell lines and was not sensitive to H3B-6527 (Fig. 4A; Supplementary Table S2). Consistent with thenotion that FGFR4 is themajor receptor for FGF19, these 40HCCcell line panel data indicate FGF19 overexpression can lead toFGFR4 dependence and H3B-6527 sensitivity in vitro.

We next sought to independently confirm these observationsusing in vivomodels. To this end, we utilized 30HCCPDXmodelsand tested the antitumor activity of H3B-6527 by oral treatmentusing 500 mg/kg twice-daily dose. Any model where H3B-6527

Figure 3.

H3B-6527 effects in a HCC cell line Hep3Bsubcutaneous xenografts in female nudemice.A,Changes in tumor CYP7A1 levels atindicated time points along with tumorand plasma H3B-6527 concentrationsafter oral dosing of H3B-6527 at indicateddoses and schedule. [� P < 0.05 vs.corresponding time point vehicle control(one-way ANOVA, followed by Dunnettpost hoc test)]. B, Representative stainingfor phospho-ERK1/2 (Thr202/Tyr204) intumor xenograft tissues after oral dosing ofH3B-6527 at indicated doses and timepoints. Tumor volume (C) and bodyweightmeasurements (D) at indicated days for15 days following oral dosing of H3B-6527at indicated doses and schedule. BID,twice-daily dose; PO, oral dosing. Datarepresent the mean � SEM (n ¼ 8).� , P < 0.05 versus vehicle control on thelast day of treatment (multiple unpairedt tests with significance determined usingthe Holm–Sidak method).

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7006

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 9: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

treatment resulted in tumor stasis or tumor regression is consid-ered a responder and 10 of 30 models fit this criterion with aresponse rate of approximately 33% (Supplementary Figs. S3 andS4). As our 40 HCC cell line panel viability data suggested thatFGF19overexpression is a strongpredictor ofH3B-6527 response,

we analyzed the HCC PDX models for FGF19 expression usingFGF19 IHC. The results of this IHC experiment showed that 8 of10 responders are positive for FGF19 staining and establishesFGF19 overexpression as a predictive biomarker for H3B-6527response (Fig. 4B). There were 2 responding models that did notshow any FGF19 staining by IHC (Fig. 4B, see Discussion). Mostof the nonresponders, 16 of 20, showed no FGF19 staining andonly 4 of 20 showed positive FGF19 staining (Fig. 4B, seeDiscussion). Using the same IHC assay, we then analyzed 231primary HCC samples to evaluate the frequency of FGF19 pos-itivity in HCC and 44 samples (�20%) scored positive based onsimilar scoring methods used for the PDX models (Fig. 4C). Tostrengthen the single-dose PDX models study above, we thenperformed a dose–response study by choosing 4 PDX models,3 responding, and one nonresponding model, using four doselevels of H3B-6527. This study showed that the respondermodelsindeed were sensitive to lower doses of H3B-6527 (Fig. 5; Sup-plementary Fig. S4). In this dose–response study, we includedsorafenib, the standard of care inHCC, as a comparator at itsMTDin mice and found that H3B-6527 exerts superior efficacy com-pared with sorafenib (Fig. 5A–D; Supplementary Fig. S5A–S5D;ref. 9). Taken together, the 40 HCC cell line panel and 30 HCCPDX models demonstrate that FGF19 overexpression is a predic-tive biomarker for H3B-6527 response and evaluation of 231primary HCC samples suggest approximately 20% of HCCpatients may potentially benefit from H3B-6527 treatment.

Having established FGF19 as a strong predictive biomarker forH3B-6527 response in HCC, we searched for additional diseasemodels where the FGF19/FGFR4 axis plays a critical role inviability. To this end, we applied H3B-6527 in a dose–responsestudy to a 625 cell line panel representing 40 cancer types andmeasured their viability. As before, any cell line with a GI50 valueof <2,000 nmol/L was considered sensitive to H3B-6527 and 12cell lines met this criterion. Validating our earlier observation, 4HCC cell lines, Hep3B, JHH-7, SNU-878, and Huh-7 showedsensitivity to H3B-6527 (Supplementary Table S3). We identifiedan additional 8 H3B-6527–sensitive cell lines; SJRH30, RH41,MDA-MB-453, KG1, NALM1, LP1, HEMC SS, and MFE280 (Sup-plementary Table S3). SJRH30 and RH41 are rhabdomyosarcoma(RMS) cell lines that express high levels FGFR4 due to directtranscriptional activation by the PAX3-FOXO1 oncogenic fusionand were previously shown to be dependent on FGFR4 (33).MDA-MB-453, a breast cancer cell line expresses an activated formof FGFR4, FGFR4-Y367C, which confers dependence on FGFR4(34). The FGFR4 dependence in the KG1 leukemia line, NALM1lymphoma line, LP1 myeloma line, HEMC bone cancer cell line,and MFE280 endometrial cancer line is a novel and intriguingfinding (see Discussion). H3B-6527 did not hit any other line inthe 625 cell line panel demonstrating both the selectivity of H3B-6527 and the selective dependence on FGFR4 across cancer types.

Palbociclib enhances H3B-6527 antitumor activityThe data described so far indicate that H3B-6527 as a single

agent is efficacious in FGF19-overexpressing HCC models. TheFGFR4 pathway is thought to restrict proliferation of cells byarresting the G1–S-phase transition through modulation of thecyclin D–CDK4/6 axis, suggesting combined inhibition of FGFR4and CDK4/6 may provide increased antitumor activity (35). Wechose the FGF19-overexpressing JHH-7 cell line xenografts infemale nude immunocompromised mice to conduct a combina-tion efficacy study. Hep3B xenografts were not utilized as this cell

Figure 4.

FGF19 expression and its correlation to the H3B-6527 sensitivity. A, FortyHCC cell lines tested in a dose response and GI50 measured. Separately,FGF19 levels were measured by ELISA and correlated to GI50. Red diamonds,H3B-6527–sensitive lines. B, Representative IHC images of FGF19 protein inPDX models. R, responder (shaded light red); NR, nonresponder to H3B-6527.Association of FGF19 expression and H3B-6527 response by a c2 statisticaltest (P ¼ 0.0016), with Yate correction (P ¼ 0.0057). C, Representative IHCimages of FGF19 protein of primary HCC samples tested with the sameFGF19 IHC assay as above.

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7007

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 10: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

line has minimal CDK4/6 activity, likely due to chronic loss of Rbexpression and relies on other CDKs for G1–S progression ren-dering them unsuitable for CDK4/6 inhibitor studies (36). As afirst step toward conducting the combination study, we appliedH3B-6527 single agent at a high dose of 500 mg/kg twice daily toJHH-7 xenografts and found that oral H3B-6527 treatment led totumor stasis (Fig. 6A). Next, we reduced the H3B-6527 total dailydose by half and applied 500 mg/kg once daily in combinationwith palbociclib, an approvedCDK4/6 inhibitor, at theMTDdoseof 100 mg/kg once daily, both by oral administration. Thiscombination resulted in enhanced tumor growth inhibition ascompared with the single-agent arms. Furthermore, the combi-nation led to tumor regressions, whereas the H3B-6527 admin-istered as a single agent twice daily led only to tumor stasis,indicating the combination could increase the best possible

response achievable with FGFR4 inhibition (Fig. 6A and B). Thecombination treatment is well tolerated in mice as no drug-induced body weight loss was observed during the course of thestudy (Supplementary Fig. S6A and S6B). Examination of the TheCancer Genome Atlas (TCGA) cohort revealed the majority ofpatients with FGF19-overexpressing HCC expressed appreciablelevels of Rb, CDK4, andCDK6, suggesting potential benefit of thiscombination for the subset (Supplementary Fig. S7).

To probe the mechanism of this enhanced combinationeffect, we conducted RNA-seq analysis of JHH-7 cell linexenograft tumors following treatment of mice with one doseof either single agents or the combination. Unsupervised clus-tering of differentially expressed genes showed a distinct pat-tern in the combination group when compared with the single-agent groups (Fig. 6C). We then performed pathway

Figure 5.

H3B-6527 antitumor effect in four HCC PDX models grown in female nude mice. Tumor volume measurements of HCC PDX6 (A), HCC PDX9 (B), HCC PDX15(C), and HCC PDX26 (D) at indicated days following oral dosing of H3B-6527 or sorafenib at indicated doses and schedule. QD, once daily; BID, twice dailydose; PO, oral dosing. Data represent the mean � SEM (n ¼ 8). � , P < 0.05 versus vehicle control on the last day of treatment (multiple unpaired t tests withsignificance determined using the Holm–Sidak method).

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7008

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 11: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

Figure 6.

Antitumor effects and RNA-seq analysis following H3B-6527 and palbociclib as single agents or in combination in JHH-7 HCC cell line xenografts grown infemale nudemice.A andB, Tumor volumemeasurements of JHH-7 xenografts at indicated doses and schedule. QD, once daily; BID, twice daily dose; PO, oral dosing.Data represent themean�SEM (n¼8). � ,P<0.05 versus vehicle control on the last dayof treatment (multiple unpaired t testswith significancedeterminedusing theHolm–Sidak method). C, Hierarchical clustering analysis of expressed genes. The samples are ordered by treatment. D, Functional annotation using Gene SetEnrichment Analysis. � , functions with nominal P < 0.01.

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7009

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 12: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

enrichment analysis of differentially expressed genes, whichshowed bile acid metabolism as the top upregulated pathwayin H3B-6527 single-agent group, a well-known effect specific tothe FGFR4 signaling pathway (Fig. 6D) (37). E2F targets scoredas the top downregulated pathway; an effect that is commonamong many receptor tyrosine kinases including FGFR4 (38).Palbociclib single agent also showed significant downregula-tion of E2F targets as would be expected for a CDK4/6 inhibitor(39). Consistent with the convergence of FGFR4 and CDK4/6functions on Rb–E2F axis, the combination groups showedstronger effects on E2F targets as compared with the single-agent groups (Fig. 6D). Moreover, the combination groupsshowed stronger downregulation of mTORC1 signaling andMYC targets in comparison with the single-agent groups andcould explain the increased antitumor activity observed inefficacy experiments (Fig. 6A and B). Taken together, theseobservations suggest that palbociclib combines well withH3B-6527 to enhance H3B-6527 efficacy in FGF19-overexpres-sing HCC models.

DiscussionPatient selection and highly selective drugs are critical factors

for the successful clinical development of targeted anticanceragents (3). The remarkable success of biomarker-defined patientenrichment strategies in the treatment of solid tumors withtargeted agents has not yet penetrated HCC (3, 40). Sorafeniband regorafenib are the only approved first- and second-linesystemic therapies, respectively, in HCC and it is important tonote that genomic predictors of their activity are not known, likelydue to theirmultikinase inhibitormode of action (40, 41).On thebasis of their lack of potency against FGFR4, it can be hypothe-sized that sorafenib and regorafenib will be unsuitable for thetreatment of FGF19-driven HCC (42). Recently, the anti-PD-1antibody nivolumab has shown durable objective responses witha response rate of approximately 20% and has the potential tobecome an approved therapy in HCC (43). One criterion that hasclearly emerged as a predictor for response to immunotherapyacross indications is the mutational load of tumors (44). Weexamined the mutational load of FGF19-overexpressing patientsamples and found no enrichment of high mutation load, sug-gesting immunotherapy may be inadequate as a single agent forthis HCC subset (45).

In this study, we have shown that inhibiting FGFR4 using aselective small molecule may provide therapeutic benefit to asubset of HCC patients with FGF19 overexpression. Althoughpan-FGFR inhibitors are undergoing clinical evaluation for avariety of indications, they are likely unsuitable for FGF19-drivenHCC due to their low potency against FGFR4 (11). We exploitedthe unique hinge cysteine in the FGFR4 kinase domain to design acovalent inhibitor, H3B-6527, that specifically inhibits FGFR4and largely spares other FGFRs. Covalent inhibitors directedagainst a cysteine have recently been developed for other kinasessuch as Bruton tyrosine kinase (BTK) and EGFR and their successin the clinic has renewed the enthusiasm for covalent inhibitors incancer therapy (46). Currently, there are three other FGFR4-selective covalent compounds undergoing preclinical and clinicalevaluation where the safety and efficacy as a monotherapy will beexplored (47–50).

The covalent binding of H3B-6527 to FGFR4 was confirmeddirectly by mass spectrometry and crystallography. Cellular

washout studies supported the covalent mechanism of actionand the in vivo xenograft studies showed that H3B-6527 is orallybioavailable and pharmacokinetic/pharmacodynamic correlateswith antitumor activity at well-tolerated doses in mice. It isthought that persistent FGFR4 inhibition may lead to excessiveproduction of bile acids resulting in chronic diarrhea, thus lim-iting the application of FGFR4 inhibitors in the clinic (51).Wedidnot observe any H3B-6527 treatment related diarrhea or bodyweight loss in mice during the efficacy studies. Factors that couldhave contributed to this beneficial therapeutic index in miceinclude shorter half-life of H3B-6527 and differences in thehalf-life of FGFR4 receptor between tumor and liver. An impor-tant element that is thought to complicate HCC therapies is theunderlying cirrhosis in the majority of patients. Total bile acidlevels are known to be reduced in cirrhosis due to the inhibition ofCyp7A1 in the liver by inflammation (52). Currently, it is notknownhow FGFR4 inhibition and the potential alterations in bileacidmetabolism could impact cirrhosis and affect the outcome ofthis mode of treatment. Efforts to model this preclinically can bechallenging due to the vast differences in bile acid pools andregulation between mice and humans (53).

The selectivity of H3B-6527 to FGFR4 is further illustrated bythe 625 cell line panel viability assay where only 12 cell linesshowed sensitivity. Among these 12 cells lines, 7 cell lines,belonging to HCC, RMS, and breast cancer lineages have knownFGF19/FGFR4 dependence. Transcriptomic and genomic altera-tions that lead to FGFR4 dependence (FGF19 overexpression inHCC cell lines and PAX3-FOXO1 fusion in RMS cell lines) are alsopresent in primary samples supporting the notion that H3B-6527would potentially be useful in these indications. In contrast, theFGFR4-Y367C–activating mutation that confers FGFR4 depen-dence in the MDA-MB-453 breast cancer cell line is not present inTCGAprimary breast cancer samples (34, 54). For the remaining 5lines representing leukemia, lymphoma, myeloma, bone, andendometrial lineages, dependence on other FGFRs has beendescribed but the existence of FGFR4 dependence was not known(29, 55). We examined the mutational and expression status ofFGF19–FGFR4 pathway components using the publicly availablegenomic datasets, but did not uncover any meaningful altera-tions. Future work involving unbiased next-generation sequenc-ing and data mining may uncover the basis for this H3B-6527sensitivity. Uncovering the basis for this intriguing FGFR4 depen-dence will potentially broaden the application of H3B-6527 tocancers involving these five lineages.

Initial analysis of 40HCC cancer cell lines suggested that FGF19overexpression is a good predictor for H3B-6527 response. Pre-vious studies have indicated that FGFR4 and KLB expression arealso determining factors for FGFR4 inhibitor response (9, 29);however, our analysis using the publicly available RNA-seq dataclearly showed that FGF19 overexpression, as a single biomarker,can predict H3B-6527 response inHCC cell lines. It is noteworthythat FGFR4 and KLB genes are highly expressed in the liver andaccordingly the majority of HCC primary samples also displayhigh level expression of FGFR4 and KLB. Although human can-cer–derived cell lines are themostwidely usedmodels to study thebiology of cancer, the clinical relevance of these models has beencontinuously questioned. Thus, we employed PDX models thatare thought to faithfully capture the genotype–response correla-tion (56). Our study with 30 PDX models again strongly impli-cated FGF19 overexpression as a single predictive biomarker forH3B-6527 response. This discovery will help shape and simplify

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7010

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 13: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

the patient selection strategies for FGFR4 inhibitor therapy. Themechanism that drives FGF19overexpression in a limited numberof HCC cases (5%) is thought to be the genomic amplification ofthe 11Q13 locus that contains the FGF19 gene (9, 35, 57).However, other yet-to-be-identified mechanisms must exist aswe and others have identified approximately 20% to 30%ofHCCoverexpress FGF19 (Fig. 4C; ref. 9). Of note, the sensitivity ofFGF19 detection assay being appliedwill determine the frequencyof FGF19 overexpression in HCC patients. Although the majorityof FGF19-overexpressing PDX models (8 of 12 models)responded to H3B-6527 in our studies, there were some (4 of12 models) FGF19-overexpressing nonresponders. Genomicanalysis did not reveal any alterations in FGFR4, KLB, CCND1,and Rb that could explain this lack of response necessitatingfurther exploration work. While the majority of FGF19-negativePDX models (16 of 18) showed no response to H3B-6527, twomodels (2 of 18) showed clear response in the absence of anydetectable FGF19 staining. A search for activating mutations inFGFR4 or alternative ligands that could activate FGFR4 failed toidentify any aberrations and the basis of sensitivity of these 2models to H3B-6527 remains unknown. Unbiased next-genera-tion sequencing with follow up validation may be needed todiscover the underlying biology conferring sensitivity to H3B-6527 in the absence of FGF19 overexpression.

Our work suggests that H3B-6527 can inhibit both cell prolif-eration and induce apoptosis in HCC cell line models. AlthoughH3B-6527 can induce tumor regression in many PDX models,potentially due to apoptosis, somemodels showed tumor stasis asthe best response. This observation led us to search for a combi-nation partner to improve the response andwe tested the CDK4/6inhibitor, palbociclib, as a potential partner due to its recentsuccess in the breast cancer setting and on the rationale that themajority of growth factor pathways exert their effect through thecyclin D–CDK4/6 axis (58). These experiments led to the discov-ery that palbociclib can enhance H3B-6527 efficacy and promotetumor regression in JHH-7 model where H3B-6527 as a singleagent can only lead to tumor stasis. Gene expression analysissuggested that the combination leads to stronger effects on E2Ftargets, mTORC1, and MYC pathways that could explain theincreased efficacy seen in the combination as compared with thesingle agents. Palbociclib has been tested in aHCC clinical trial asa single agent but showed only modest activity as a single agent(59). It is noteworthy that palbociclib became a breakthroughtherapy in the breast cancer setting as a combination partner toletrazole and in HCC a similar combination approach as

described by our studies may be required for significant improve-ment in clinical efficacy. Our studies using primary HCC samplessuggest that a substantial fraction of HCC patients havealtered FGF19 expression that could potentially benefit fromH3B-6527 monotherapy. Furthermore, the increased activityobserved with H3B-6527 in combination with CDK4/6 inhibi-tion provides a rationale for exploration of this combination inearly clinical trials.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: E. Corcoran, S. Prajapati, M.-H. Hao, L. Yu, P. Zhu,N. Larsen, J. Wang, M. Warmuth, D.J. Reynolds, P.G. Smith, A. SelvarajDevelopment of methodology: H. Coffey, E. Corcoran, J. Tsai, S. Prajapati,M.-H. Hao, S. Bailey, C. Karr, V. Subramanian, E. Park, N. Larsen, P. Fekkes,D.J. Reynolds, P.G. Smith, A. SelvarajAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J.J. Joshi, H. Coffey, E. Corcoran, J. Tsai, K. Ichikawa,M.-H. Hao, S. Bailey, V. Rimkunas, C. Karr, V. Subramanian,W.G. Lai, N. LarsenAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J.J. Joshi, H. Coffey, J. Tsai, C.-L. Huang, K. Ichikawa,M.-H. Hao, S. Bailey, J. Wu, V. Rimkunas, C. Karr, V. Subramanian, P. Kumar,C. Mackenzie, K. Yu, E. Park, N. Rioux, W.G. Lai, L. Yu, S. Buonamici, N. Larsen,D.J. Reynolds, P.G. Smith, A. SelvarajWriting, review, and/or revision of the manuscript: J.J. Joshi, H. Coffey,C.-L. Huang, J. Wu, V. Rimkunas, P. Kumar, N. Rioux, A. Kim, W.G. Lai,S. Buonamici, N. Larsen, M. Warmuth, D.J. Reynolds, P.G. Smith, A. SelvarajAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): H. Coffey, S. Bailey, W.G. Lai, P. Zhu, J. Wang,D.J. ReynoldsStudy supervision: C. Karr, W.G. Lai, L. Yu, P. Fekkes, J. Wang, D.J. Reynolds,P.G. Smith, A. SelvarajOther (synthesis of compounds): T. SatohOther (oversaw chemistry): D.J. Reynolds

AcknowledgmentsWe would like to thank Namita Kumar and Galina Kuznetsov for the IHC

analysis and Andrew Cook for critical reading of the manuscript.

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received June 22, 2017; revised August 23, 2017; accepted October 10, 2017;published online December 15, 2017.

References1. Torrecilla S, Llovet JM. New molecular therapies for hepatocellular carci-

noma. Clin Res Hepatol Gastroenterol 2015;39Suppl 1:S80–5.2. Cheng AL, Shen YC, Zhu AX. Targeting fibroblast growth factor

receptor signaling in hepatocellular carcinoma. Oncology 2011;81:372–80.

3. Llovet JM, Villanueva A, Lachenmayer A, Finn RS. Advances in targetedtherapies for hepatocellular carcinoma in the genomic era. Nat Rev ClinOncol 2015;12:408–24.

4. Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic landscape andbiomarkers of hepatocellular carcinoma. Gastroenterology 2015;149:1226–39.

5. Pinyol R, Nault JC, Quetglas IM, Zucman-Rossi J, Llovet JM. Molecularprofiling of liver tumors: classification and clinical translation for decisionmaking. Semin Liver Dis 2014;34:363–75.

6. Jones S. Mini-review: endocrine actions of fibroblast growth factor 19. MolPharm 2008;5:42–8.

7. Lin BC, Wang M, Blackmore C, Desnoyers LR. Liver-specific activities ofFGF19 require Klotho beta. J Biol Chem 2007;282:27277–84.

8. French DM, Lin BC, Wang M, Adams C, Shek T, Hotzel K, et al. TargetingFGFR4 inhibits hepatocellular carcinoma in preclinical mouse models.PLoS One 2012;7:e36713.

9. Hagel M, Miduturu C, Sheets M, Rubin N, Weng W, Stransky N, et al. Firstselective small molecule inhibitor of FGFR4 for the treatment of hepato-cellular carcinomas with an activated FGFR4 signaling pathway. CancerDiscov 2015;5:424–37.

10. Uriarte I, Latasa MU, Carotti S, Fernandez-Barrena MG, Garcia-Irigoyen O,Elizalde M, et al. Ileal FGF15 contributes to fibrosis-associated hepatocel-lular carcinoma development. Int J Cancer 2015;136:2469–75.

H3B-6527 for FGF19-Driven HCC

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7011

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 14: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

11. Packer LM, Pollock PM. Paralog-specific kinase inhibition of FGFR4:adding to the arsenal of anti-FGFR agents. Cancer Discov 2015;5:355–7.

12. Desnoyers LR, Pai R, Ferrando RE, Hotzel K, Le T, Ross J, et al. TargetingFGF19 inhibits tumor growth in colon cancer xenograft and FGF19transgenic hepatocellular carcinoma models. Oncogene 2008;27:85–97.

13. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR:ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.

14. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilisticRNA-seq quantification. Nat Biotechnol 2016;34:525–7.

15. RobinsonMD,McCarthyDJ, SmythGK. edgeR: a Bioconductor package fordifferential expression analysis of digital gene expression data. Bioinfor-matics 2010;26:139–40.

16. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J,et al. PGC-1alpha-responsive genes involved in oxidative phosphorylationare coordinately downregulated in human diabetes. Nat Genet 2003;34:267–73.

17. SubramanianA, TamayoP,Mootha VK,Mukherjee S, Ebert BL,GilletteMA,et al. Gene set enrichment analysis: a knowledge-based approach forinterpreting genome-wide expression profiles. Proc Natl Acad Sci U S A2005;102:15545–50.

18. Mohammadi M, Schlessinger J, Hubbard SR. Structure of the FGF receptortyrosine kinase domain reveals a novel autoinhibitory mechanism. Cell1996;86:577–87.

19. Eathiraj S, Palma R, Volckova E, Hirschi M, France DS, Ashwell MA, et al.Discovery of a novelmodeof protein kinase inhibition characterizedby themechanism of inhibition of human mesenchymal-epithelial transitionfactor (c-Met) protein autophosphorylation by ARQ 197. J Biol Chem2011;286:20666–76.

20. Vagin A, Teplyakov A. Molecular replacement with MOLREP. Acta Crystal-logr D Biol Crystallogr 2010;66:22–5.

21. Murshudov GN, Vagin AA, Dodson EJ. Refinement of macromolecularstructures by the maximum-likelihood method. Acta Crystallogr D BiolCrystallogr 1997;53:240–55.

22. Lebedev AA, Young P, Isupov MN, Moroz OV, Vagin AA, Murshudov GN.JLigand: a graphical tool for the CCP4 template-restraint library. ActaCrystallogr D Biol Crystallogr 2012;68:431–40.

23. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, MengEC, et al. UCSF Chimera–a visualization system for exploratory researchand analysis. J Comput Chem 2004;25:1605–12.

24. Shoemaker RH. TheNCI60 human tumour cell line anticancer drug screen.Nat Rev Cancer 2006;6:813–23.

25. Xin H, Wang K, Hu G, Xie F, Ouyang K, Tang X, et al. Establishment andcharacterization of 7 novel hepatocellular carcinoma cell lines frompatient-derived tumor xenografts. PLoS One 2014;9:e85308.

26. Tan L, Wang J, Tanizaki J, Huang Z, Aref AR, Rusan M, et al. Develop-ment of covalent inhibitors that can overcome resistance to first-gen-eration FGFR kinase inhibitors. Proc Natl Acad Sci U S A 2014;111:E4869–77.

27. van Linden OP, Kooistra AJ, Leurs R, de Esch IJ, de Graaf C. KLIFS: aknowledge-based structural database to navigate kinase-ligand interactionspace. J Med Chem 2014;57:249–77.

28. Urich R, Wishart G, Kiczun M, Richters A, Tidten-Luksch N, Rauh D, et al.De novo design of protein kinase inhibitors by in silico identification ofhinge region-binding fragments. ACS Chem Biol 2013;8:1044–52.

29. Guagnano V, Kauffmann A, Wohrle S, StammC, Ito M, Barys L, et al. FGFRgenetic alterations predict for sensitivity to NVP-BGJ398, a selective pan-FGFR inhibitor. Cancer Discov 2012;2:1118–33.

30. Richmond A, Su Y. Mouse xenograft models vs. GEM models for humancancer therapeutics. Dis Model Mech 2008;1:78–82.

31. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S,et al. The Cancer Cell Line Encyclopedia enables predictive modelling ofanticancer drug sensitivity. Nature 2012;483:603–7.

32. Wilks C, Cline MS, Weiler E, Diehkans M, Craft B, Martin C, et al. TheCancerGenomicsHub (CGHub): overcoming cancer through the power oftorrential data. Database 2014;2014:article ID bau093.

33. Li SQ, Cheuk AT, Shern JF, Song YK, Hurd L, Liao H, et al. Targeting wild-type and mutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534). PLoS One 2013;8:e76551.

34. Roidl A, Foo P, Wong W, Mann C, Bechtold S, Berger HJ, et al. The FGFR4Y367Cmutant is a dominant oncogene inMDA-MB453 breast cancer cells.Oncogene 2010;29:1543–52.

35. Sawey ET, ChanrionM, Cai C,WuG, Zhang J, Zender L, et al. Identificationof a therapeutic strategy targeting amplified FGF19 in liver cancer byOncogenomic screening. Cancer Cell 2011;19:347–58.

36. Rivadeneira DB, Mayhew CN, Thangavel C, Sotillo E, Reed CA, Grana X,et al. Proliferative suppression by CDK4/6 inhibition: complex function ofthe retinoblastoma pathway in liver tissue and hepatoma cells. Gastroen-terology 2010;138:1920–30.

37. Yu C, Wang F, Kan M, Jin C, Jones RB, Weinstein M, et al. Elevatedcholesterol metabolism and bile acid synthesis in mice lacking membranetyrosine kinase receptor FGFR4. J Biol Chem 2000;275:15482–9.

38. Katz M, Amit I, Yarden Y. Regulation of MAPKs by growth factors andreceptor tyrosine kinases. Biochim Biophys Acta 2007;1773:1161–76.

39. O'Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6inhibitors. Nat Rev Clin Oncol 2016;13:417–30.

40. Llovet JM, Hernandez-Gea V. Hepatocellular carcinoma: reasons for phaseIII failure and novel perspectives on trial design. Clin Cancer Res2014;20:2072–9.

41. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenibfor patients with hepatocellular carcinoma who progressed on sorafenibtreatment (RESORCE): a randomised, double-blind, placebo-controlled,phase 3 trial. Lancet 2017;389:56–66.

42. Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, et al.Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol2011;29:1046–51.

43. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al.Nivolumab in patients with advanced hepatocellular carcinoma (Check-Mate 040): an open-label, non-comparative, phase 1/2 dose escalation andexpansion trial. Lancet 2017;389:2492–502.

44. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy.Science 2015;348:69–74.

45. Cancer Genome Atlas Research Network. Comprehensive and integrativegenomic characterization of hepatocellular carcinoma. Cell 2017;169:1327–41.

46. Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs.Nat Rev Drug Discov 2011;10:307–17.

47. Bruce Ruggeri MS, Yang Y-O, Juvekar A, Lu L, Condon S, DiMatteo D, et al.The novel FGFR4-selective inhibitor INCB062079 is effcacious in modelsof hepatocellular carcinoma harboring FGF19 amplifcation [abstract]. In:Proceedings of the AACRAnnualMeeting; 2017 Apr 1–5;Washington, DC.Philadelphia (PA): AACR; 2017. Abstr nr 1234.

48. Diana Graus Porta AW, Fairhurst RA, Wartmann M, Stamm C, Reimann F,Buhles A, et al. NVP-FGF401, a frst-in-class highly selective and potentFGFR4inhibitor for the treatment of HCC [abstract]. In: Proceedings of theAACRAnnualMeeting; 2017Apr 1–5;Washington,DC. Philadelphia (PA);2017. Abstr nr 2098.

49. Kim R, Sharma S, Meyer T, Sarker D, Macarulla T, Sung M, et al. First-in-human study of BLU-554, a potent, highly-selective FGFR4 inhib-itor designed for hepatocellular carcinoma (HCC) with FGFR4pathway activation [abstract]. In: Proceedings of the EORTC-NCI-AACR Molecular Targets and Cancer Therapeutics Symposium;2016 Nov 29–Dec 02; Munich, Germany. Philadelphia (PA): AACR;2017. Abstr nr 105A.

50. Fairhurst RA, Knoepfel T, Leblanc C, Buschmann N, Gaul C, Blank J, et al.Approaches to selective fibroblast growth factor receptor 4 inhibitionthrough targeting the ATPpocket middle-hinge region. Med Chem Com-mun 2017;8:1604–13.

51. Mellor HR. Targeted inhibition of the FGF19-FGFR4 pathway in hepato-cellular carcinoma; translational safety considerations. Liver Int 2014;34:e1–9.

52. Ridlon JM, Alves JM, Hylemon PB, Bajaj JS. Cirrhosis, bile acids and gutmicrobiota: unraveling a complex relationship. Gut Microbes 2013;4:382–7.

53. Chiang JY. Bile acids: regulation of synthesis. J Lipid Res 2009;50:1955–66.54. Cancer Genome Atlas Network. Comprehensive molecular portraits of

human breast tumours. Nature 2012;490:61–70.55. Nakanishi Y, Akiyama N, Tsukaguchi T, Fujii T, Sakata K, Sase H, et al. The

fibroblast growth factor receptor genetic status as a potential predictor of

Joshi et al.

Cancer Res; 77(24) December 15, 2017 Cancer Research7012

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 15: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

the sensitivity to CH5183284/Debio 1347, a novel selective FGFR inhib-itor. Mol Cancer Ther 2014;13:2547–58.

56. Cho SY, KangW,Han JY,Min S, Kang J, Lee A, et al. An integrative approachto precision cancer medicine using patient-derived xenografts. Mol Cells2016;39:77–86.

57. SchulzeK, ImbeaudS, Letouze E, Alexandrov LB,Calderaro J, Rebouissou S,et al. Exome sequencing of hepatocellular carcinomas identifies new

mutational signatures and potential therapeutic targets. Nat Genet2015;47:505–11.

58. Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, et al. Palbocicliband letrozole in advanced breast cancer. N Engl J Med 2016;375:1925–36.

59. Littman SJ, Brus C, Burkart A. A phase II study of palbociclib (PD-0332991)in adult patients with advanced hepatocellular carcinoma. J Clin Oncol2015:33; 277.

www.aacrjournals.org Cancer Res; 77(24) December 15, 2017 7013

H3B-6527 for FGF19-Driven HCC

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 16: H3B-6527 Is a Potent and Selective Inhibitor of ... · Therapeutics, Targets, and Chemical Biology H3B-6527 Is a Potent and Selective Inhibitor of FGFR4inFGF19-DrivenHepatocellularCarcinoma

2017;77:6999-7013. Cancer Res   Jaya Julie Joshi, Heather Coffey, Erik Corcoran, et al.   FGF19-Driven Hepatocellular CarcinomaH3B-6527 Is a Potent and Selective Inhibitor of FGFR4 in

  Updated version

  http://cancerres.aacrjournals.org/content/77/24/6999

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2018/01/31/77.24.6999.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/77/24/6999.full#ref-list-1

This article cites 54 articles, 13 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/77/24/6999.full#related-urls

This article has been cited by 3 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://cancerres.aacrjournals.org/content/77/24/6999To request permission to re-use all or part of this article, use this link

on June 23, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from