13
Small Molecule Therapeutics PI3K Inhibitors Synergize with FGFR Inhibitors to Enhance Antitumor Responses in FGFR2 mutant Endometrial Cancers Leisl M. Packer 1 , Xinyan Geng 1 , Vanessa F. Bonazzi 1 , Robert J. Ju 1 , Clare E. Mahon 1 , Margaret C. Cummings 2 , Sally-Anne Stephenson 3 , and Pamela M. Pollock 1 Abstract Improved therapeutic approaches are needed for the treat- ment of recurrent and metastatic endometrial cancer. Endome- trial cancers display hyperactivation of the MAPK and PI3K pathways, the result of somatic aberrations in genes such as FGFR2, KRAS, PTEN, PIK3CA, and PIK3R1. The FGFR2 and PI3K pathways, have emerged as potential therapeutic targets in endometrial cancer. Activation of the PI3K pathway is seen in more than 90% of FGFR2 mutant endometrial cancers. This study aimed to examine the efcacy of the pan-FGFR inhibitor BGJ398 with pan-PI3K inhibitors (GDC-0941, BKM120) and the p110a-selective inhibitor BYL719. We assessed synergy in three FGFR2 mutant endometrial cancer cell lines (AN3CA, JHUEM2, and MFE296), and the combination of BGJ398 and GDC-0941 or BYL719 showed strong synergy. A signicant increase in cell death and decrease in long-term survival was seen when PI3K inhibitors were combined with BGJ398. Importantly, these effects were seen at low concentrations correlating to only partial inhibition of AKT. The combination of BGJ398 and GDC-0941 showed tumor regressions in vivo, whereas each drug alone only showed moderate tumor growth inhibition. BYL719 alone resulted in increased tumor growth of AN3CA xenografts but in combination with BGJ398 resulted in tumor regression in both AN3CA- and JHUEM2-derived xeno- grafts. These data provide evidence that subtherapeutic doses of PI3K inhibitors enhance the efcacy of anti-FGFR therapies, and a combination therapy may represent a superior therapeu- tic treatment in patients with FGFR2 mutant endometrial cancer. Mol Cancer Ther; 16(4); 63748. Ó2017 AACR. Introduction Endometrial cancer is the most common gynecologic malig- nancy in developed countries, and its incidence is increasing in postmenopausal women (1). In 2016, the American Cancer Society estimated that about 10,500 U.S. women will die of endometrial cancer (2). Treatment options for patients with recurrent or persistent endometrial cancer are limited to radiation and chemotherapy, which offer limited clinical benet. As a result, the average survival of patients with metastatic or recurrent endometrial cancer is only 7 to 12 months (3). Thus, there is a need for more effective therapies with reduced side effects, as well as predictive biomarkers to identify patients most likely to respond to these treatment options. Our group and others have identied activating somatic muta- tions in FGFR2 in about 10% of patients presenting with primary endometrioid endometrial cancer (49). With regard to the endometrial cancer subtypes identied by The Cancer Genome Atlas (TCGA), FGFR2 mutations occur at a similar frequency in the microsatellite instability (MSI) hypermutated subtype as well as the copy numberlow subtype, which has also been described as those tumors with no specic molecular aberration (NSMP) (7, 10). More recently, mutational analysis in a large multi-institute cohort has revealed that FGFR2 mutations are more common in the tumors of patients who present with late-stage disease (17%) as well as those who progress (progressed, recurred, or died from disease; 26%) (11). In multivariate analysis where age, grade, and stage were also taken into account, the presence of an FGFR2 mutation was associated with decreased progression-free survival and decreased endometrial cancerspecic survival (11). Preclinical studies by our group and others have shown that FGFR2 mutant endometrial cancer cells are highly sensitive to a range of FGFR inhibitors including PD173074 (5, 12) ponatinib (13, 14), BGJ398, dovitinib (15), and AZD4547 (16). The major- ity (93%) of FGFR2 mutant endometrial cancers also harbor muta- tions in the PI3K pathway (PIK3CA, PIK3CB, PIK3R1, PIK3R2, PTEN, AKT1) (7). Western blot analyses of FGFR2 mutant endome- trial cancer cell lines show that FGFR inhibitors fail to completely block PI3K pathway activation (12, 15, 16). Although these in vitro studies had shown classic oncogene addiction in FGFR2 mutant endometrial cancer cell lines, in vivo studies with 30 mg/kg 1 Endometrial Cancer Laboratory, Queensland University of Technology (QUT), Translational Research Institute, Queensland, Australia. 2 School of Medicine, University of Queensland Centre for Clinical Research, Queensland, Australia. 3 Eph Receptor Biology Group, Queensland University of Technology (QUT), Translational Research Institute, Queensland, Australia. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). L.M. Packer and X. Geng contributed equally to this study. Corresponding Author: Pamela M. Pollock, Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology (QUT) at the Translational Research Institute, 37 Kent Road Woolloongabba QLD 4102, Australia. Phone: 617-34437237; Fax: 617-3443-7779; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-16-0415 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 637 on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

PI3K Inhibitors Synergize with FGFR Inhibitors to Enhance ...Small Molecule Therapeutics PI3K Inhibitors Synergize with FGFR Inhibitors to Enhance Antitumor Responses in FGFR2mutant

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Small Molecule Therapeutics

    PI3K Inhibitors Synergize with FGFR Inhibitors toEnhance Antitumor Responses in FGFR2mutant

    Endometrial CancersLeisl M. Packer1, Xinyan Geng1, Vanessa F. Bonazzi1, Robert J. Ju1, Clare E. Mahon1,Margaret C. Cummings2, Sally-Anne Stephenson3, and Pamela M. Pollock1

    Abstract

    Improved therapeutic approaches are needed for the treat-ment of recurrent and metastatic endometrial cancer. Endome-trial cancers display hyperactivation of the MAPK and PI3Kpathways, the result of somatic aberrations in genes such asFGFR2, KRAS, PTEN, PIK3CA, and PIK3R1. The FGFR2 andPI3K pathways, have emerged as potential therapeutic targets inendometrial cancer. Activation of the PI3K pathway is seen inmore than 90% of FGFR2mutant endometrial cancers. This studyaimed to examine the efficacy of the pan-FGFR inhibitorBGJ398 with pan-PI3K inhibitors (GDC-0941, BKM120) andthe p110a-selective inhibitor BYL719. We assessed synergy inthree FGFR2mutant endometrial cancer cell lines (AN3CA,JHUEM2, and MFE296), and the combination of BGJ398 andGDC-0941 or BYL719 showed strong synergy. A significant

    increase in cell death and decrease in long-term survival wasseen when PI3K inhibitors were combined with BGJ398.Importantly, these effects were seen at low concentrationscorrelating to only partial inhibition of AKT. The combinationof BGJ398 and GDC-0941 showed tumor regressions in vivo,whereas each drug alone only showed moderate tumor growthinhibition. BYL719 alone resulted in increased tumor growth ofAN3CA xenografts but in combination with BGJ398 resulted intumor regression in both AN3CA- and JHUEM2-derived xeno-grafts. These data provide evidence that subtherapeutic doses ofPI3K inhibitors enhance the efficacy of anti-FGFR therapies,and a combination therapy may represent a superior therapeu-tic treatment in patients with FGFR2mutant endometrial cancer.Mol Cancer Ther; 16(4); 637–48. �2017 AACR.

    IntroductionEndometrial cancer is the most common gynecologic malig-

    nancy in developed countries, and its incidence is increasing inpostmenopausal women (1). In 2016, the American CancerSociety estimated that about 10,500 U.S. women will die ofendometrial cancer (2). Treatment options for patients withrecurrent or persistent endometrial cancer are limited to radiationand chemotherapy,whichoffer limited clinical benefit. As a result,the average survival of patients with metastatic or recurrentendometrial cancer is only 7 to 12 months (3). Thus, there is aneed for more effective therapies with reduced side effects, as well

    as predictive biomarkers to identify patients most likely torespond to these treatment options.

    Our group and others have identified activating somatic muta-tions in FGFR2 in about 10% of patients presenting with primaryendometrioid endometrial cancer (4–9). With regard to theendometrial cancer subtypes identified by The Cancer GenomeAtlas (TCGA), FGFR2mutations occur at a similar frequency in themicrosatellite instability (MSI) hypermutated subtype as well asthe copy number–low subtype, which has also been described asthose tumors with no specific molecular aberration (NSMP)(7, 10).More recently,mutational analysis in a largemulti-institutecohort has revealed that FGFR2 mutations are more common inthe tumors of patientswhopresentwith late-stagedisease (17%)aswell as those who progress (progressed, recurred, or died fromdisease; 26%) (11). In multivariate analysis where age, grade, andstage were also taken into account, the presence of an FGFR2mutation was associated with decreased progression-free survivaland decreased endometrial cancer–specific survival (11).

    Preclinical studies by our group and others have shown thatFGFR2mutant endometrial cancer cells are highly sensitive to arange of FGFR inhibitors including PD173074 (5, 12) ponatinib(13, 14), BGJ398, dovitinib (15), and AZD4547 (16). The major-ity (93%) of FGFR2mutant endometrial cancers also harbor muta-tions in the PI3K pathway (PIK3CA, PIK3CB, PIK3R1, PIK3R2,PTEN, AKT1) (7). Western blot analyses of FGFR2mutant endome-trial cancer cell lines show that FGFR inhibitors fail to completelyblock PI3Kpathway activation (12, 15, 16). Although these in vitrostudies had shown classic oncogene addiction in FGFR2mutant

    endometrial cancer cell lines, in vivo studies with 30 mg/kg

    1Endometrial Cancer Laboratory, Queensland University of Technology (QUT),Translational Research Institute, Queensland, Australia. 2School of Medicine,University of Queensland Centre for Clinical Research, Queensland, Australia.3Eph Receptor Biology Group, Queensland University of Technology (QUT),Translational Research Institute, Queensland, Australia.

    Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

    L.M. Packer and X. Geng contributed equally to this study.

    Corresponding Author: Pamela M. Pollock, Institute of Health and BiomedicalInnovation, School of Biomedical Science, Queensland University of Technology(QUT) at the Translational Research Institute, 37 Kent Road WoolloongabbaQLD 4102, Australia. Phone: 617-34437237; Fax: 617-3443-7779; E-mail:[email protected]

    doi: 10.1158/1535-7163.MCT-16-0415

    �2017 American Association for Cancer Research.

    MolecularCancerTherapeutics

    www.aacrjournals.org 637

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://crossmarksupport.crossref.org/http://crossmarksupport.crossref.org/http://crossmarksupport.crossref.org/http://mct.aacrjournals.org/

  • BGJ398 showed that FGFR inhibition alone in the FGFR2mutant

    AN3CA cell line led to a delay in tumor growth but not tumorregression (15). More recently, a similar study evaluating 30 mg/kg AZD4547 in AN3CA xenografts in vivo did show tumor regres-sion (16), consistent with earlier studies performed in our labo-ratory using twice daily dosing of PD173074 (data not shown).

    The PI3K pathway regulates proteins involved in cell cycle,survival, andmetabolism. It is thought to be the most commonlyactivated signaling pathway in human cancer, and endometrioidendometrial cancers have the highest frequency (80%–90%) ofsomatic mutations affecting this pathway (7, 8). In most tumortypes, loss of PTEN and activation of PIK3CA are mutuallyexclusive events; however, endometrial cancer is unusual in thatmany tumors carry aberrations in multiple members of thissignaling pathway (7).

    There are several different classes of PI3K pathway inhibitorsdesigned to target this pathway at one or more nodes and theseinclude pan-PI3K, isoform-specific PI3K, mTOR, AKT, dual PI3K/mTOR, and dualmTORC1/mTORC2 inhibitors (reviewed in 17).Unfortunately, many inhibitors targeting this pathway haveshown disappointing results in phase II/III clinical trials, and thishas been attributed to a small therapeutic window accompaniedby on-target toxicity from inhibiting this pathway in normaltissues, as well as a lack of predictive biomarkers to better identifythe patients who will respond (18, 19).

    In this study, we chose to evaluate BGJ398 (infigratinib), anorally bioavailable selective pan-FGFR inhibitor currently beingevaluated in phase II trials as a single agent in several FGFR-dependentmalignancies (NCT02160041,NCT02150967) aswellas the pan-PI3K inhibitor (BKM120) and the p110a-selectiveinhibitor BYL719 (alpelisib), all developed by Novartis. Of directrelevance to this project, there is currently a phase Ib expansiontrial evaluating the efficacy of BGJ398 þ BYL719 in breast andlung cancers (NCT01928459). As BKM120 has been shown topossess off-target effects at concentrations above 1 mmol/L (20),we also assessed BGJ398 in combinationwith the class I pan-PI3Kinhibitor GDC-0941 (pictilisib). This research shows that partialabrogation of signaling through the PI3K pathway enhances theefficacy of BGJ398 in FGFR2mutant endometrial cancer modelsin vitro and in vivo.

    Materials and MethodsCell lines, culture conditions, and inhibitors

    AN3CA, MFE296, and JHUEM2 were obtained from ATCC(2005), ECACC (2007), and RikenCell Bank (2012), respectively.AN3CA, JHUEM2, and MFE296 were authenticated by shorttandem repeat (STR) profiling at the sequencing facility of TheQIMRBerghoferMedical Research Institute in 2013 and 2016 andpassaged less than 20 times since authentication. AN3CA andMFE296 cells were grown in MEM-a and JHUEM2 cells in 1:1DMEM:HamF12, supplemented with 10% FBS, 1% penicillin/streptomycin, and 0.1mmol/L nonessential amino acids. Accord-ing to the Cancer Cell Line Encyclopedia, the cell lines harborthe following mutations: AN3CA expresses FGFR2 N550Kand K310R, PIK3R1 p.557_561REIDK>Q, and PTEN p.R130fs;JHUEM2 expresses FGFR2 C383R, PIK3CA p.V344G, p.E978K,PIK3R1 p.N707del, and PTEN p.N212_splice; and MFE296 cellsharbor FGFR2 N550K, PIK3CA p.I20M, p.P539R, and PTEN p.R130Q, and p.T321fs�23. Kinase inhibitors (BGJ398, GDC-0941,BKM120, and BYL719) were purchased from Selleck Chemicals

    for in vitro experiments and from Synkinase for in vivo studies.Structures of compounds are shown in Supplementary Fig. S1B.

    Cell viability assayCell viability was assessed by sulforhodamine B (SRB) staining.

    Briefly, 3,000 cells were seeded in a 96-well plate. The followingday, cells were treatedwith half-log dilutions of drug (1 nmol/L to10 mmol/L). After 96 hours, cells were fixed in methanol, stainedwith SRB, solubilizedwith 10mmol/L Tris and absorbance read at492 nm. Values were normalized to DMSO control. IC50 valuesare the mean of 3 independent experiments and were calculatedusing nonlinear regression analysis with variable slope in Graph-Pad Prism v6.0.

    Chou–Talalay drug combination studySynergy between BGJ398 and the PI3K inhibitors was assessed

    using themethodology proposed byChou and Talalay (21). Drugconcentrations were in a series of 2-fold dilutions above andbelow the IC50 of each drug. One day after seeding, cells weretreated with BGJ398, GDC-0941, BYL719, BKM120 alone or incombination for 96 hours. All experiments were repeated 3independent times. The combination index and fraction affectedwas calculated by CalcuSyn, v2.0 (Biosoft).

    Colony-forming assayCells (600–1,000) were seeded in 6-well plates and the fol-

    lowing day treated with DMSO or inhibitors for 72 hours. Cellswere washed 3 times in PBS and grown in full-growthmedium for10 to 16 days, fixed withmethanol, and stained with crystal violet(0.1% in 25% methanol). Colonies were counted, and the meanof 2 (JHUEM2) or 3 (AN3CA,MFE296) independent experiments(each performed in triplicate) was plotted as a percentage of theDMSO control.

    Assessment of apoptosisCells (4 � 105) were seeded in 6-well plates overnight. On the

    second day, cells were treated with the indicated drugs or DMSOfor 72 hours. Floating and attached cells were collected andanalyzed for Annexin V and propidium iodide staining accordingto the manufacturer's instructions (FITC Annexin V ApoptosisDetection Kit II, BD Biosciences) using BD LSR II and FlowJo,v10.7.

    siRNA-mediated depletion of p110a and p110bAN3CA and JHUEM2 cells (3.5 � 105) were reversed-trans-

    fected with 10 nmol Dharmacon ON-TARGETplus siRNA poolstargeting p110a/PIK3CA and p110b/PIK3CB or a nontargetingcontrol (D-001810-10) using Lipofectamine RNAiMAX in serum-freemedia in a 6-well plate. Full-growthmedia was added after 24hours. Forty-eight hours posttransfection, cells were treated withDMSO, 0.3 mmol/L GDC-0941, or 0.6 mmol/L BYL719 for 6 hoursand then lysed and subjected to Western blot analysis. Quanti-fication of band intensities from duplicate (JHUEM2) and trip-licate (AN3CA) experiments (normalized to tubulin) was per-formed using ImageJ.

    Immunoprecipitation, Western blot analysis, and antibodiesProteins were harvested using RIPA buffer [50mmol/L Tris, pH

    7.4, 150 mmol/L NaCl, 1 mmol/L EDTA, 1% IGEPAL, 0.1% SDS,0.5% sodium deoxycholate, 1 mmol/L sodium orthovanadate,1mmol/LNaF, 1mmol/L phenylmethylsulfonylfluoride (PMSF),

    Packer et al.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics638

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • 10 mg/mL aprotinin, and leupeptin]. For immunoprecipitation, 1mg of protein lysate was precleared with Protein A SureBeadsbeads (Bio-Rad) for 30 minutes before incubating the lysate withanti-FGFR2 (C-17) antibody (Santa Cruz) and Protein A Sure-Beads beads overnight at 4�C. Western blotting was performedusing standard protocols. The following antibodies were used:p110a (#4249), p110b (#3011), PARP (#9542), pFRS2a(Tyr436) (#3861), pAKT (Ser473) (#4060), AKT (#2920),pERK(Thr202/Tyr204) (#4695), ERK (#9107), pS6 (Ser240/244) (#2215), S6 (#2317), p-Tyr-100 (#9411) from Cell Signal-ing Technology; pERK (Thr202/Tyr204) (mouse), tubulin(T9026) from Sigma; FGFR2 Bek-C17 (sc-122), ERK2 (sc-154),FRS2 (sc-8318), and GAPDH (sc-32233) antibodies from SantaCruz Biotechnology, Inc. Secondary antibodies IRDye 680LTDonkey anti-Rabbit IgG (C31024-04) and IRDye 800CWDonkeyanti-mouse IgG (#C30904-02) were from LI-COR Corporate.

    In vivo murine xenograft modelSix-week-old female NOD/SCID mice (15–18 g) were pur-

    chased from the Animal Resources Centre (Canningvale, WA,Australia) and hosted in the pathogen-free Biological ResourceFacility (BRF) of the Translational Research Institute (Brisbane,Australia). Mice were maintained and handled under asepticconditions and were allowed access to food and water ad libitum.In vivo animal studies were performed according to institution-approved protocols (TRI/160/14/AUC) and guidelines for main-tenance of animals and endpoint of tumor studies were followed.Xenografts of AN3CA and JHUEM2 endometrial cancer cell lineswere established by subcutaneously injecting 1 � 106 viable cellsin growth factor–reducedMatrigel (#354230, BDBiosciences) 1:1with PBS into the flank of the mice. Perpendicular tumor dia-meters were measured by a single observer using Vernier scalecalipers, and tumor volumeswere calculated using the formula [(x� y2)/2]. JHUEM2 and AN3CA xenografts were allowed to growfor 10 and 14 days, respectively (to allow formation of tumorswith mean xenograft volume � 150 mm3). Mice were thenstratified into treatment groups with one tumor per mouse onthe basis of their weight and tumor volume. Mice (8/group) weretreated for 3 weeks via oral gavage, 5 days on/1 day off, of (i)vehicle control [100 mmol/L acetic acid/sodium acetate buffer,pH4.6/PEG300 (1:1)]; (ii) BGJ398, 20mg/kg; (iii)GDC-0941, 75mg/kg; (iv) BYL719, 12.5 mg/kg; (v) BGJ398 þ GDC-0941; and(vi) BGJ398þBYL719. Bodyweight was recorded for each animalevery other day to monitor potential toxicities. Additional ani-mals (4/group)were treated for 4days, with theirfinal treatment 6hours prior to tumor collection. Part of the tumorwas snap-frozenand then lysed in RIPA lysis buffer (2.5 mL/mg) for Western blotanalysis and the other part fixed in 4% paraformaldehyde.

    Immunohistochemical staining of mouse xenograftsTumors were fixed in 4% paraformaldehyde solution over-

    night, paraffin-embedded, and cut into 5-mm-thick sections. Sec-tionswere deparaffinized, rehydrated, followedby antigen retriev-al with CC1 buffer at 100�C for 64 minutes using the VentanaDiscovery Ultra. Slides were blocked with Discovery Inhibitor for8 minutes, incubated with Anti-Rabbit Cleaved Caspase-3 anti-body (#9661; Cell Signaling Technology) for 1 hour at 37�C,followed by secondary anti-Rabbit HQ and anti-HQ HRP. Thesignal was detected with DAB substrate (Discovery ChomoMapkit) followed by a hematoxylin counterstain. All images weretakenwithOlympus IX73 inverted Fluorescencemicroscopefitted

    with XM10 monochrome camera. Histopathologic scoring ofcleaved caspase-3 was performed on 5 fields (�400 magnifica-tion) for each of the 4 tumors treated with the different drug/savoiding areas of marked necrosis. Identification of positive cellswas performed blinded and independently on a multiheadermicroscope by M.C. Cummings, V.F. Bonazzi, and P.M. Pollockand averaged for each sample and condition. Data for caspasepositivity for each drug treatment are presented as a ratio overvehicle control.

    Statistical analysisThe in vitro data were analyzed using one-way ANOVA with

    Tukey multiple comparison to test all treatment combinations.Differences in xenograft volumebetween groupswere assessed forsignificance using a repeated 2-way ANOVA. P-values, calculatedwith Prism (GraphPad), are coded by asterisks:

  • Combination treatment of AN3CA, JHUEM2, and MFE296 cellswith BGJ398 and either GDC-0941 or BYL719 resulted inenhanced inhibition of cell proliferation compared with BGJ398alone. The combination of BGJ398 with GDC-0941 or BYL719was synergistic (combination index values < 0.7) at all concen-trations in JHUEM2 and MFE296 and in all but the lower 2concentrations in AN3CA (Fig. 1C, F, I). The BGJ398 and BKM120combination had a more subtle effect, with synergy seen only atthe highest concentrations (Supplementary Fig. S1D–S1F). Theseresults suggest that dual treatment with BGJ398 and either GDC-0941 or BYL719 is more synergistic at inhibiting proliferation ofFGFR2mutant endometrial cancer cells than BGJ398 combinedwith BKM120.

    Co-targeting FGFR2 and PI3K signaling reduces long-term cellsurvival

    Clonogenic assayswere performed to further examine the effectof the combination treatments on long-term cell survival and todetermine whether synergy could be seen at clinically relevantdoses (Fig. 2). Plasma concentrations of BGJ398 in phase I trialpatients were found to have aCmin�Cmax range of approximately100 to 450 nmol/L (22), and as such, 100 and 300 nmol/Lconcentrations were assessed, as these represented a low- andmid-range concentration, respectively. The reportedCmax ofGDC-0941, BYL719, and BKM120 were 2.07 mmol/L (23), 2.3 mmol/L(24), and 1.8 mmol/L (25), respectively; therefore 0.3, 0.6, and1 mmol/L were initially assessed. When combined with BGJ398,

    Figure 1.

    Synergistic inhibition of cell viability by combined treatment with BGJ398 and a PI3K inhibitor. Growth inhibition induced by the FGFR inhibitor BGJ398 andthe PI3K inhibitors alone or in combination. AN3CA (A–C), JHUEM2 (D–F), and MFE296 (G–I) cells were treated with the indicated doses of BGJ398, GDC-0941, andBYL719 alone or in combination for 96 hours, and an SRB assay was subsequently performed. Data are presented as a percentage of the control, in whichcells were treated with 0.1% (v/v) DMSO. Points represent themean of 3 independent experiments (each performed in triplicate). Error bars represent SEM, and lineswere fitted using nonlinear regression analysis. Interaction of BGJ398 and GDC-0941 (red circles) or BYL719 (black squares) in AN3CA (C), JHUEM2 (F), andMFE296(I). Median effect analysis (CalcuSyn software) was used to evaluate the interaction between the inhibitor combinations. Horizontal dotted lines indicate theboundaries for each interaction classification.

    Table 1. Half maximal inhibitory concentration (IC50) of inhibitors in FGFR2-mutant endometrial cancer cell lines

    InhibitorsRange of concentrationsused in IC50 calculation

    IC50 in AN3CA,nmol/L

    IC50 in JHUEM2,nmol/L

    IC50 in MFE296,nmol/L

    BGJ398 0.1 nmol/L–1 mmol/L 30 20 80GDC-0941 1 nmol/L–10 mmol/L 140 355 630BYL719 1 nmol/L–10 mmol/L 1,730 530 4,060BKM120 1 nmol/L–10 mmol/L 320 260 695

    Packer et al.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics640

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • even low concentrations of the PI3K inhibitors caused a substan-tial reduction in colony formation (Supplementary Fig. S2A andS2B), despite the small reduction in AKT phosphorylation seenwith the lower drug concentrations (Supplementary Fig. S1). Forsubsequent analysis, we used 0.3 mmol/L GDC-0941, 0.6 mmol/LBYL719, and0.6mmol/L BKM120. These concentrationswerewellbelow the plasma Cmax values (often close to the Cmin) such thatevidence of synergism might open new avenues for using thesedrugs at subtherapeutic doses.

    Treatment with 0.3 mmol/L BGJ398 significantly reduced col-ony formation by about 70% (P 0.0001), but not by any of the PI3Kinhibitors alone. The combination ofGDC-0941or BKM120withBGJ398 further reduced the colony formation by 15% (nonsig-nificant) and 10% (nonsignificant), respectively, but no addi-tional benefit was seen when BYL719was combinedwith BGJ398(Fig. 2E and F). It should be noted that for this assay, cells weretreated with single agents or combinations for 72 hours afterwhich cells were washed to remove residual drugs before plating,

    Figure 2.

    Dual targeting of the FGFR and PI3K pathways leads to synergistic inhibition of long-term survival and enhanced cell death. Clonogenic survival assays in AN3CA(A and B), JHUEM2 (C and D), MFE296 (E and F) treated with the indicated doses (mmol/L) of BGJ398 (BGJ), GDC-0941 (GDC), and BYL719 (BYL) alone or incombination for 72 hours. Cells were then cultured for 16 days without inhibitors and stained with crystal violet. Pictures are representative of 3 independentexperiments. Colonies were counted and expressed as a percentage of the DMSO control. The mean of 3 independent experiments (each performed intriplicate) for AN3CA (B), JHUEM2 (D), and MFE296 (F) is shown along with SD. Percentage of apoptotic cells in AN3CA (G), JHUEM2 (H), and MFE296 (I) treatedwith DMSO, 0.3 mmol/L BGJ398 (BGJ), 0.3 mmol/L GDC-0941 (GDC), and 0.6 mmol/L BYL719 (BYL) alone or in combination for 72 hours. Apoptosis wasdetected by staining cells with Annexin V and propidium iodide. The mean percentage of apoptotic cells from 2 (JHUEM2) or 3 (AN3CA, MFE296) independentexperiments (each performed in triplicate) is shown along with SD. Statistical significance between the indicated groups according to a one-way ANOVAis shown. ���� , P < 0.0001; ��� , P < 0.001; �� , P < 0.01; �, P < 0.05.

    FGFR and PI3K Inhibitors in FGFR2mutant Endometrial Cancer

    www.aacrjournals.org Mol Cancer Ther; 16(4) April 2017 641

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • Packer et al.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics642

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • so this assay underrepresents the cell death that would be seenfollowing continuous drug exposure.

    BGJ398 synergizes with PI3K inhibition to induce cell deathTo test the hypothesis that BGJ398 combined with a PI3K

    inhibitor enhances apoptosis, Annexin V positivity was assessedfollowing 72-hour drug treatment. Treatment with BGJ398alone induced approximately 30% to 40% cell death in AN3CA(P < 0.05), JHUEM2 (P < 0.0001), and MFE296 (P < 0.05),compared with 10% in the vehicle control (Fig. 2G–I). Thesingle-agent PI3K inhibitors had little effect on cell death in anycell line at the low concentrations chosen. The combination ofBGJ398 with any of the PI3K inhibitors induced significantlymore cell death than BGJ398 alone. These data demonstrate thatcombining the FGFR inhibitor BGJ398with apan-PI3Kor p110a/PIK3CA-selective inhibitor not only reduces cell proliferation andlong-term survival but also enhances the effect of BGJ398 ininducing cell death in BGJ398- sensitive endometrial cancer cells.

    The combination of BGJ398 and a PI3K inhibitor causedenhanced inhibition of AKT and downstream target S6

    To understand the molecular basis of the synergistic cell deathinduced by the combination of BGJ398 and PI3K inhibitors, wemeasured the response of key downstream targets to the individ-ual inhibitors and combinatorial treatments after 1, 8, and 24hours (Fig. 3A and B; Supplementary Fig. S3). Phosphorylation ofERK, a downstream marker of FGFR activity, is totally abrogatedby 0.3 mmol/L BGJ398 at all time points in all 3 cell lines. BGJ398slightly blocks AKT phosphorylation in JHUEM2 and MFE296and shows little effect on AKT phosphorylation in AN3CA cells.

    In JHUEM2, all 3 PI3K inhibitors inhibit AKT activity to asimilar extent, leading to dephosphorylation of S6 at 8 and 24hours. In AN3CA and MFE296 cells, GDC-0941 is the mosteffective inhibitor of AKT and S6 with strongest inhibition seenat the earlier 2 time points. BYL719 has little effect on AKT or S6activity in AN3CA cells. This may be explained by the lack of aPIK3CA mutation in AN3CA cells, the presence of which likelysensitizes JHUEM2 cells to this isoform-selective inhibitor. Theinhibition of the PI3K pathway by single agents is short-lived,with phospho-S6 levels returning to almost normal by 24hours inAN3CA and JHUEM2. The dephosphorylation of S6 in responseto dual targeting of the FGFR and PI3K pathways was greatest at 8hours and still evident at 24 hours in all 3 cell lines.

    Preferential signaling of JHUEM2 cells through p110a/PIK3CATo determine whether AN3CA and JHUEM2 cells show pref-

    erential signaling through p110a/PIK3CA or p110b/PIK3CB,siRNA knockdown of each gene was performed (Fig. 3C). Aspredicted, knockdown of p110a in JHUEM2 (PIK3CAmutant)resulted in almost complete inhibition of AKT and S6, suggest-ing that p110a regulates PI3K pathway activity in these cells. In

    contrast, while p110a knockdown in AN3CA (PIK3CAWT) cellsinhibited AKT (though to a lesser extent than JHUEM2), thisdid not translate into an equivalent inhibition of the down-stream effector S6, suggesting that AN3CA does not solely relyon p110a/PIK3CA for PI3K pathway activation.

    Knockdown of p110b resulted in an unexpected increase in theexpression of p110a in both cell line models (Fig. 3D), whichresulted in an increase in the activation of AKT and S6 (Fig. 3C),unlike knockdown of p110a, which did not alter p110b levels(Fig. 3D). Knockdown of both p110a and p110b abrogated thisactivation of AKT signaling. The pan-PI3K inhibitor GDC-0941almost completely inhibits AKT/S6 signaling in AN3CA, more sothan the combined siRNAs (which elicit only partial knockdown),suggesting that AN3CA relies on both p110a and p110b for PI3Kpathway activation.

    Combined BGJ398 and PI3K inhibition inducedmarked tumorregression in FGFR2mutant xenograft models in vivo

    We then studied the antitumor activity of the BGJ398 þ GDC-0941 and BGJ398 þ BYL719 combinations in AN3CA- andJHUEM2-derived murine xenografts. BGJ398, GDC-0941, andBYL719 alone and in combination were well-tolerated with nosignificant weight loss observed throughout the course of treat-ment (Supplementary Fig. S4A and S4B). While BGJ398 has beenused at concentrations ranging from5 to 45mg/kg in vivo (26), weutilized a concentration of 20mg/kg to detect increased efficacy inour combination studies. As expected, 20 mg/kg BGJ398 resultedin significantly delayed tumor growth in both cell line modelscompared with the control group (P < 0.001, Fig. 4A–C). GDC-0941 (75 mg/kg) and BYL719 (12.5 mg/kg) administered assingle agents had surprising opposite effects. GDC-0941 inhibitedtumor growth to a similar extent as BGJ398, consistent withincreased pathway inhibition by 75 mg/kg GDC-0941 in vivocompared with the lower concentrations utilized for the in vitrostudies (Fig. 4D). In contrast, BYL719monotherapy unexpectedlyenhanced the growth of AN3CA-derived tumors (P < 0.0001).

    Combinatory treatment of BGJ398 þ GDC-0941 and BGJ398þ BYL719 resulted in a marked inhibition of tumor growth inboth AN3CA (P < 0.001, P < 0.05, respectively) and JHUEM2xenograft models, compared with the BGJ398-treated groups.Indeed, these combinations caused complete or partial tumorregression, with no palpable tumor present in 5 of 8 and 4 of 8mice with AN3CA and 2 of 8 and 1 of 8 mice with JHUEM2xenografts treated with BGJ398 þ GDC-0941 and BGJ398 þBYL719, respectively (Supplementary Fig. S4B).

    Biochemically, BGJ398-treated xenografts show partial inhibi-tion of AKT and almost complete inhibition of ERK at 4 days (Fig.4D). AN3CA xenografts treated with GDC-0941 for 4 days showmarked reduction in pAKT, pS6, and p4EBP-1 levels, confirminginhibition of the PI3K pathway. BYL719 treatment also resulted ina reduction in pAKT and pS6 levels, albeit not to the extent seen

    Figure 3.Inhibition of FGFR and PI3K pathways by BGJ398, GDC-0941, BYL719. and BKM120. AN3CA (A) and JHUEM2 (B) cells were treated for the indicated times withDMSO, 0.3 mmol/L BGJ398 (BGJ), 0.3 mmol/L GDC-0941 (GDC), 0.6 mmol/L BYL719 (BYL), and 0.6 mmol/L BKM120 (BKM) alone or in combination. Cell lysateswere immunoblotted with antibodies for phospho-AKT (Ser473), total AKT, phospho-ERK (Thr202/Tyr204), ERK2, phospho-S6 (Ser240/244), total S6,total PARP, and cleaved PARP. Tubulin was detected as the loading control. Western blot analysis of AN3CA and JHUEM2 (C) cells transfected with siRNApools targeting p110a and p110b and a nontargeting (NT) control for 48 hours and treated with 0.3 mmol/L GDC-0941 (GDC) or 0.6 mmol/L BYL719 (BYL) for 6 hours.The mean band intensity of pAKT and pS6 (normalized to tubulin) are shown, along with SD. The mean level of p110a with p110b knockdown (D) and p110bfollowing p110a knockdown (normalized to tubulin) from 3 independent experiments along with SD is also shown.

    FGFR and PI3K Inhibitors in FGFR2mutant Endometrial Cancer

    www.aacrjournals.org Mol Cancer Ther; 16(4) April 2017 643

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • with GDC-0941. The combination of BGJ398 þ GDC-0941caused a stronger reduction in pAKT, pS6, and p4EBP1 thanGDC-0941 alone. These results are consistent with the cell deathand clonogenic data, which confirm that a synergistic effect occurswhen blocking FGFR and PI3K pathways.

    Histologic analysis of the AN3CA tumors, which had beentreated for 4 days, revealed very frequent mitoses in both thevehicle control and in the BYL719 tumors (data not shown).Onlyoccasional apoptotic bodies were observed in the GDC-0941–treated tumors, whereas a high number of apoptotic bodies wereseen in the tumors treated with BGJ398 þ GDC-0941 (data notshown). Both combination treatments showed broad, confluentareas of necrosis.

    Assessment of cell death markers following dual targeting ofFGFR and PI3K pathways

    The single-agent and combination treatments were assessedfor their effect on the apoptotic marker PARP and cleavedcaspase-3 (Figs. 3A and B and 4E). In AN3CA cells, cleavageof PARP resulting in 25- and 89-kDa fragments, is morepronounced following treatment with BGJ398 þ GDC-0941at 8 and 24 hours (Fig. 3A). This is consistent with a significantincrease in caspase-3 cleavage by the BGJ398 þ GDC-0941combination in AN3CA-derived xenografts and the presence ofnumerous apoptotic bodies compared with BGJ398 and GDC-0941 single treatments (Fig. 4E). This high level of caspasecleavage is not observed with the BGJ398 þ BYL719 combina-tion in the AN3CA xenografts, despite large areas of tumornecrosis evident histologically, suggesting different mechan-isms of cell death may be occurring.

    Similar Annexin positivity was seen with all 3 combinations(Fig. 2G), suggesting that BYL719 and BKM120 might elicit adifferent cell death mechanism. The inhibition of AKT caused byBYL719 alone or in combination with BGJ398 is much less thanthat caused by GDC-0941. It is possible that only minor abro-gation of AKT signaling is required to kill AN3CA cells when FGFRis also inhibited. In contrast to the results observed in AN3CAcells, a strong induction of PARP cleavage in response to BGJ398was seen in JHUEM2 cells, which was enhanced equally in all 3PI3K inhibitor combinations treatments. This PARP cleavagecoincides with similar levels of Annexin V positivity seen at 72hours in all 3 combinations (Fig. 2H).

    DiscussionSmall-molecule inhibitors that target oncogenic drivers of

    tumorigenesis are becoming standard therapies in many cancertypes. Given the high frequency of PI3K aberrations in endome-trial cancer, several phase II trials evaluating PI3K inhibitors assingle agents have been undertaken, with overall disappointingresults (27). Anumber of FGFR inhibitors have shown remarkableclinical responses in a subset of other FGFR-dependent malig-nancies (28, 29), but to date, only multi-kinase inhibitors such asdovitinib have been tested in endometrial cancer (30). No com-plete responses were documented in the latter; however; nohyperphosphatemia was reported, bringing into question wheth-er sufficient inhibition of the FGFR receptors was obtained.Although FGFR inhibitors have been shown to induce cell deathin FGFR2mutant endometrial cancer cell lines with concomitantPI3K pathway activation (12), it is reasonable to assume thatthe co-occurrence of activating PI3K pathway mutations may

    limit the extent and durability of tumor responses to single-agentFGFR inhibitors.

    Here we show that multiple inhibitors targeting the PI3Kpathway enhance the efficacy of the FGFR inhibitor BGJ398 inFGFR2mutant endometrial cancers. Notably, we show that lowdoses of PI3K inhibitors, correlating with only partial inhibitionof AKT phosphorylation, synergize with FGFR inhibition toachieve cell death and tumor shrinkage in vivo. Our data suggestisoform-specific inhibition of p110a by BYL719 has differenteffects in AN3CA and JHUEM2 cells. In JHUEM2 cells, carrying anactivating PIK3CA mutation, BYL719 resulted in partial AKTinhibition, a small decrease in pS6 phosphorylation (Fig. 3B),and a reduction in tumor growth in vivo (Fig. 4B). In AN3CA cells,BYL719 had less of an effect on pAKT (Figs. 3A and 4D), with littleto no reduction of phosphorylated pS6, suggesting that inhibitionof the PI3K/AKT/mTORC1 pathway by this p110a-specific inhib-itor is incomplete. Unexpectedly, BYL719 single-agent treatmentincreased growth of AN3CA-derived xenografts (Fig. 3A), which isblocked by the addition of BGJ398. This suggests that whateverprosurvival pathway is being activated by BYL719 in AN3CAtumors, it is blocked by pan-FGFR inhibition. Activation ofparallel pathways has previously been implicated in resistanceto PI3K inhibitors (reviewed in 31), leading to the belief thatcombination therapies are required to overcome such feedbackloops. Further studies are required to understand the molecularbasis of BYL719-induced tumor growth in AN3CA xenografts.

    The differential reliance of AN3CA and JHUEM2 on p110a/PIK3CA may explain their differential response to BYL719.BYL719 inhibits AKT/S6 signaling to a greater extent in JHUEM2cells, resulting in PARP cleavage when combined with BGJ398 inJHUEM2 cells (Fig. 3B), which are more reliant on p110a thanAN3CA (Fig. 3C). In contrast, PI3K signaling is only partiallyinhibited by BYL719 in AN3CA cells, which fails to cause PARPcleavage (Fig. 3A) or caspase-dependent cell death in combina-tion with BGJ398 (Fig. 4E). These results suggest that completePI3K inhibition is required to induce caspase-dependent celldeath. The fact that BGJ398 þ BYL719 induces tumor regressionto the same extent as BGJ398 þ GDC-0941 suggests that onlypartial inhibition of the PI3K pathway is needed to lower theapoptotic threshold of FGFR inhibitors. Furthermore, the resultssuggest that cell death induced by BYL719 is caspase-independentand may also be PI3K-independent.

    Our results show that in the context of endometrial cancer S6 isregulated by both the PI3K and FGFR2 pathways, with thecombination treatments reducing levels of phosphorylated S6more than the individual treatments (Fig. 3). The sustainedinhibition of S6 by combination treatment is likely the result ofinhibiting both the PI3K and FGFR2 pathways. Together withprevious studies targeting the PI3K pathway alone or in combi-nation with MEK inhibition, these results indicate that levels ofphosphorylated S6 may be an effective biomarker of response totargeting these key survival pathways (32–34).

    Our data in endometrial cancer are supported by similarcombination studies in endometrial cancer and other cancers.Specifically in endometrial cancer, Gozgit and colleagues reportedsynergy between the multi-kinase inhibitor ponatinib and themTOR inhibitor ridaforolimus (35). In liver cancer, the additionof the mTOR inhibitor RAD001 to dovitinib also showed anincrease in growth inhibition of Hep3B xenografts (36), and theaddition of the mTOR inhibitor rapamycin to BGJ398 resultedin increased tumor growth inhibition in a subcutaneous and a

    Packer et al.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics644

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • Figure 4.

    PI3K inhibition improves antitumor efficacy when given in combination with BGJ398. AN3CA (A) and JHUEM2 (B) xenografts were established in nude miceand stratified into 6 groups (8/group) treated for the indicated number of days with vehicle, 20 mg/kg BGJ398, 75 mg/kg GDC-0941, 12.5 mg/kg BYL719,BGJ398 þ GDC-0941, and BGJ398 þ BYL719. Mean tumor volumes are shown along with SE. Representative tumors including the smallest and largest from eachgroup are shown.C, Tumor growth of AN3CA and JHUEM2 xenografts assessed at 21 days of treatmentwith inhibitors described inA. Protein lysates fromAN3CA (D)xenografts taken from mice treated with the above doses of BGJ398, GDC-0941, or BYL719 for 4 days were lysed and subjected to Western blot analysis forphospho-AKT (Ser473), total AKT, phospho-ERK (Thr202/Tyr204), total ERK, phospho-S6 (Ser240/244), total S6, phospho-4EBP1 (Thr37/46), and total4EBP1. Tubulinwas detected as the loading control.E, Immunohistochemical staining of cleaved caspase-3 in AN3CAxenografts treated for 4 dayswith the indicateddrugs, along with the mean of caspase-3–positive cells counted in 5 fields (�400 magnification) in 4 tumors (presented as a ratio over vehicle control).

    FGFR and PI3K Inhibitors in FGFR2mutant Endometrial Cancer

    www.aacrjournals.org Mol Cancer Ther; 16(4) April 2017 645

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • syngeneic orthotopic model (37). In FGFR1-amplified lungcancer, the combination of BGJ398 and GDC-0941 led toenhanced growth suppression in vivo (38). Furthermore, a recentstudy in FGFR1-amplified lung and bladder cancer cells foundthat the FGFR inhibitor AZD4547 caused synergistic inductionof cell death in vitro when combined with an mTOR inhibitor(either AZD8055 or KU0063794) or a pan-PI3K inhibitor(GDC-0941) (29).

    Early trials using pan-PI3K inhibitors were associated with on-target toxicity. Thus, considerable effort has gone into developingPI3K isoform-selective inhibitors and the identification of pre-dictive biomarkers for these isoform-selective PI3K inhibitors.Initial studies utilizing both genetically engineeredmousemodelsand panels of cell lines from multiple cancer types showed thatPTEN-deficient breast and prostate cancer cells preferentiallysignal through p110b (39–41). Moreover, clinical resistance toBYL719 in a metastatic breast cancer lesion carrying an activatingPIK3CA mutation was attributed to loss of PTEN in this specificlesion (42). More recently, there have been reports that PI3Kisoform usage in the context of PTEN loss is dependent on thegenetic context by which the PI3K pathway is activated. Tumorswhere the PI3K pathway was activated by either activated KRAS(43) or the polyoma middle T antigen (44) showed a reliance onp110a even in the presence of concurrent PTEN loss, in contrast toearlier studies where the reliance on p110b occurred only inmodels with PTEN loss.

    Our attempt to determine whether preferential signalingthrough p110a or p110bwas taking place in AN3CA or JHUEM2cells was somewhat hindered by the unexpected activation of AKTfollowing p110b depletion.While unexpected, our results were inline with those of Utermark and colleagues (45), showing thatincreased AKT activation in a conditional knockout of p110b inmurine mammary epithelium in transgenic models of breastcancer driven by the polyoma middle T antigen or Her2/Neuresulted in enhanced tumor growth. The authors presented dataindicating that there was competition between p110a and p110bfor binding to the oncogenic receptor and removal of p110ballows increased binding of the more active p110a, leading toincreased pathway activation (45). In the endometrial cancer linesexamined, the increased AKT activation could be due to both anincrease in p110a expression (Fig. 3C and D) as well as higheractivation of AKT elicited by p110a. Whether the ablation ofp110b resulted in an increase in p110a expression was notassessed in themurinemodels, but thedata presentedhere suggesta level of compensatory crosstalk that has not been previouslyreported.

    As endometrial tumors are unique in that they often carryaberrations in multiple members of the PI3K pathway, iden-tifying biomarkers of response to isoform-specific PI3K inhi-bitors in endometrial cancer has proven difficult. The study byWeigelt and colleagues investigating a number of drugs target-ing different nodes of the PI3K pathway in a large panel ofendometrial cancer cell lines showed that PTEN-null endome-trial cancers require inhibition of both p110a and p110b toreduce cell viability and suggest that the preferential use ofspecific catalytic subunits of PI3K may also depend on tissuecontext (17). This is in line with clinical data using mTORinhibitors in endometrial cancer where no association betweenspecific mutations and clinical responses was observed (46).Our in vitro data are consistent with that of Weigelt andcolleagues (17), with single-agent pan-PI3K inhibitors GDC-

    0941 and BKM120 showing greater activity than BYL719 in avariety of assays. However, perhaps surprisingly, when com-bined with FGFR inhibition, p110a-specific inhibition byBYL719 induced similar tumor growth inhibition to pan-PI3Kinhibition with GDC-0941.

    A variety of studies have demonstrated that crosstalk betweenthe MAPK and PI3K signaling pathways are associated withresistance to targeted therapies (47–49). Thus, we hypothesizethat dual inhibition of both the FGFR/MAPK axis and PI3Ksignaling pathways will not only induce more tumor cell deathbut also result in a decrease in intrinsic and acquired resistance. Ina similar manner, we would also hypothesize that pan-PI3Kinhibition may well prevent the emergence of resistance viaaltered p110 isoform usage.

    Although many companies are developing inhibitors againstthese pathways, only a few companies have inhibitors againstboth FGFR and the PI3K in clinical development. ArQule, Inc.has a specific FGFR inhibitor (ARQ087) and a pan-AKT inhib-itor (ARQ092/ARQ751) and Astra Zeneca also has a pan-FGFRinhibitor (AZD4547) and a pan-AKT inhibitor (AKT5363). Atthis time, Novartis has the only combination in clinical trials(BYL719 þ BGJ398) with enrolment focused on those patientswith an activating PIK3CA mutation. Our data, and that ofWeigelt and colleagues (17), would suggest that pan-PI3Kinhibition is more efficacious than isoform-specific PIK3CAinhibition in endometrial cancer. As with all combinationtherapies, there remains a need to determine whether thetoxicities seen with inhibition of either the PI3K or FGFRpathways are additive or synergistic when blocked together.Our in vitro data suggest that subtherapeutic inhibition of thePI3K pathway may be effective in combination, allowing lowerdoses and ideally less toxicity.

    Disclosure of Potential Conflicts of InterestP.M. Pollock has ownership interest in a patent on the detection of FGFR2

    mutations in endometrial cancer. No potential conflicts of interest weredisclosed by the other authors.

    Authors' ContributionsConception and design: L.M. Packer, X. Geng, V.F. Bonazzi, P.M. PollockDevelopmentofmethodology: L.M. Packer, X.Geng, V.F. Bonazzi, P.M. PollockAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): L.M. Packer, X. Geng, V.F. Bonazzi, R. Ju, C. Mahon,M.C. CummingsAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): L.M. Packer, X. Geng, V.F. Bonazzi, C. Mahon,S.-A. Stephenson, P.M. PollockWriting, review, and/or revision of the manuscript: L.M. Packer, X. Geng,V.F. Bonazzi, M.C. Cummings, S.-A. Stephenson, P.M. PollockAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): L.M. Packer, X. Geng, V.F. Bonazzi, P.M. PollockStudy supervision: L.M. Packer, V.F. Bonazzi, S.-A. Stephenson, P.M. Pollock

    Grant SupportL.M. Packer has been supported byNHMRCCJMartin Fellowship (443038).

    P.M. Pollock has been supported by anNHMRCCDF2 Fellowship (#1032851).The study is supported by a Cancer Australia Grant (#1087165).

    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 27, 2016; revised December 1, 2016; accepted January 4, 2017;published OnlineFirst January 23, 2017.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics646

    Packer et al.

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • References1. Saso S, Chatterjee J, Georgiou E, Ditri AM, Smith JR, Ghaem-Maghami S.

    Endometrial cancer. BMJ 2011;343:d3954.2. American Cancer Society. Cancer facts & figures 2016. Atlanta, GA: Amer-

    ican Cancer Society;2016.3. Obel JC, FribergG, FlemingGF. Chemotherapy in endometrial cancer. Clin

    Adv Hematol Oncol 2006;4:459–68.4. Pollock PM, Gartside MG, Dejeza LC, Powell MA, Mallon MA, Davies H,

    et al. Frequent activating FGFR2 mutations in endometrial carcinomasparallel germline mutations associated with craniosynostosis and skeletaldysplasia syndromes. Oncogene 2007;26:7158–62.

    5. Dutt A, Salvesen HB, Chen TH, Ramos AH, Onofrio RC, Hatton C, et al.Drug-sensitive FGFR2mutations in endometrial carcinoma. ProcNatl AcadSci U S A 2008;105:8713–7.

    6. Byron SA, Gartside M, Powell MA, Wellens CL, Gao F, Mutch DG, et al.FGFR2 point mutations in 466 endometrioid endometrial tumors: rela-tionship with MSI, KRAS, PIK3CA, CTNNB1 mutations and clinicopatho-logical features. PLoS ONE 2012;7:e30801.

    7. Cancer Genome Atlas ResearchNetwork, Kandoth C, Schultz N, CherniackAD, Akbani R, Liu Y, et al. Integrated genomic characterization of endo-metrial carcinoma. Nature 2013;497:67–73.

    8. Cheung L, Hennessy B, Li J, Yu S, Myers A, Djordjevic B. High frequency ofPIK3R1 and PIK3R2 mutations in endometrial cancer elucidates a novelmechanism for regulation of PTEN protein stability. Cancer Discov 2011;1:170–85.

    9. Krakstad C, Birkeland E, Seidel D, Kusonmano K, Petersen K, Mjos S, et al.High-throughput mutation profiling of primary and metastatic endome-trial cancers identifies KRAS, FGFR2 and PIK3CA to be frequently mutated.PLoS ONE 2012;7:e52795.

    10. Stelloo E, Nout RA, Osse EM, Jurgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC,et al. Improved risk assessment by integratingmolecular and clinicopatho-logical factors in early-stage endometrial cancer-combined analysis of thePORTEC cohorts. Clinical Cancer Res 2016;22:4215–24.

    11. Jeske Y, Ali S, Byron S, Gao F, Mannel R, Ghebre R, et al. FGFR2 mutationsare associated with poor outcomes in endometrioid endometrial cancer:An NRG oncology/gynecologic oncology group study. Submitted toGynaecological Oncology 2017. In Press.

    12. Byron SA, Gartside MG, Wellens CL, Mallon MA, Keenan JB, Powell MA,et al. Inhibition of activated fibroblast growth factor receptor 2 in endo-metrial cancer cells induces cell death despite PTEN abrogation. Cancer Res2008;68:6902–7.

    13. Gozgit JM, Squillace RM, Wongchenko MJ, Miller D, Wardwell S, Mohem-mad Q, et al. Combined targeting of FGFR2 and mTOR by ponatinib andridaforolimus results in synergistic antitumor activity in FGFR2 mutantendometrial cancer models. Cancer Chemother Pharmacol 2013;71:1315–23.

    14. Kim DH, Kwak Y, Kim ND, Sim T. Antitumor effects and molecularmechanisms of ponatinib on endometrial cancer cells harboring activatingFGFR2 mutations. Cancer Biol Ther 2016;17:65–78.

    15. Konecny GE, Kolarova T, O'Brien NA, Winterhoff B, Yang G, Qi J, et al.Activity of the fibroblast growth factor receptor inhibitors dovitinib(TKI258) andNVP-BGJ398 inhuman endometrial cancer cells.Mol CancerTher 2013;12:632–42.

    16. Kwak Y, Cho H, Hur W, Sim T. Antitumor Effects and Mechanisms ofAZD4547 on FGFR2-deregulated endometrial cancer cells. Mol CancerTher 2015;14:2292–302.

    17. Weigelt B, Warne PH, Lambros MB, Reis-Filho JS, Downward J. PI3Kpathway dependencies in endometrioid endometrial cancer cell lines. ClinCancer Res 2013;19:3533–44.

    18. Rodon J, Dienstmann R, Serra V, Tabernero J. Development of PI3Kinhibitors: lessons learned from early clinical trials. Nat Rev Clin Oncol2013;10:143–53.

    19. Yap TA, Bjerke L, Clarke PA,Workman P. Drugging PI3K in cancer: refiningtargets and therapeutic strategies. Curr Opin Pharmacol 2015;23:98–107.

    20. Brachmann SM, Kleylein-Sohn J, Gaulis S, Kauffmann A, Blommers MJ,Kazic-Legueux M, et al. Characterization of the mechanism of action of thepan class I PI3K inhibitor NVP-BKM120 across a broad range of concen-trations. Mol Cancer Ther 2012;11:1747–57.

    21. Chou T, Talalay P. Quantitative analysis of dose-effect relationships: thecombined effects of multiple drugs or enzyme inhibitors. Adv EnzymeRegul 1984;22:27–55.

    22. Sequist L, Cassier P, Varga A, Tabernero J, Schellens J, Delord J-P. Phase Istudy of BGJ398, a selective pan-FGFR inhibitor in genetically preselectedadvanced solid tumors [abstract]. In: Proceedings of the AnnualMeeting ofthe American Association for Cancer Research; 2014 Apr 5–9; San Diego,CA. Philadelphia, PA: AACR; 2014. Abstract nrCT326.

    23. Sarker D, Ang JE, Baird R, Kristeleit R, Shah K, Moreno V, et al. First-in-human phase I study of pictilisib (GDC-0941), a potent pan-class Iphosphatidylinositol-3-kinase (PI3K) inhibitor, in patients with advancedsolid tumors. Clin Cancer Res 2015;21:77–86.

    24. De Buck SS, Jakab A Fau - BoehmM, BoehmMFau - Bootle D, Bootle D Fau- Juric D, Juric D Fau - Quadt C, Quadt C Fau - Goggin TK, et al. Populationpharmacokinetics and pharmacodynamics of BYL719, a phosphoinositide3-kinase antagonist, in adult patients with advanced solidmalignancies. BrJ Clin Pharmacol 2014;78:543–55.

    25. Rodon J, Brana I, Siu LL, De JongeMJ, Homji N,Mills D, et al. Phase I dose-escalation and -expansion studyof buparlisib (BKM120), anoral pan-ClassI PI3K inhibitor, in patients with advanced solid tumors. Invest NewDrugs2014;32:670–81.

    26. 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.

    27. Bregar AJ, Growdon WB. Emerging strategies for targeting PI3K in gyne-cologic cancer. Gynecol Oncol 2016;140:333–44.

    28. Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dienstmann R, et al.Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, andpharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol2014;25:2244–51.

    29. 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.

    30. Konecny GE, Finkler N, Garcia AA, Lorusso D, Lee PS, Rocconi RP, et al.Second-linedovitinib (TKI258) inpatientswith FGFR2-mutated or FGFR2-non-mutated advanced or metastatic endometrial cancer: a non-rando-mised, open-label, two-group, two-stage, phase 2 study. Lancet Oncol2015;16:686–94.

    31. Klempner SJ, Myers AP, Cantley LC. What a tangled web we weave:emerging resistance mechanisms to inhibition of the phosphoinositide3-kinase pathway. Cancer Discov 2013;3:1345–54.

    32. O'BrienC,Wallin JJ, SampathD,GuhaThakurtaD, SavageH, Punnoose EA,et al. Predictive biomarkers of sensitivity to the phosphatidylinositol 3'kinase inhibitor GDC-0941 in breast cancer preclinical models. ClinCancer Res 2010;16:3670–83.

    33. Zhong H, Sanchez C, Spitzer D, Plambeck-Suess S, Gibbs J, Hawkins WG,et al. Synergistic effects of concurrent blockade of PI3K andMEK pathwaysin pancreatic cancer preclinical models. PLoS ONE 2013;8:e77243.

    34. Haagensen EJ, Kyle S, Beale GS, Maxwell RJ, Newell DR. The synergisticinteraction ofMEK and PI3K inhibitors ismodulated bymTOR inhibition.Br J Cancer 2012;106:1386–94.

    35. Gozgit JM, Wong MJ, Moran L, Wardwell S, Mohemmad QK, NarasimhanNI, et al. Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor withactivity inmultiple FGFR-amplified ormutated cancermodels.Mol CancerTher 2012;11:690–9.

    36. Chan SL, Wong CH, Lau CP, Zhou Q, Hui CW, Lui VW, et al. Preclinicalevaluation of combined TKI-258 and RAD001 in hepatocellular carcino-ma. Cancer Chemother Pharmacol 2013;71:1417–25.

    37. Scheller T, Hellerbrand C, Moser C, Schmidt K, Kroemer A, Brunner SM,et al.mTOR inhibition improvesfibroblast growth factor receptor targetingin hepatocellular carcinoma. Br J Cancer 2015;112:841–50.

    38. Kotani H, Ebi H, Kitai H, Nanjo S, Kita K, Huynh TG, et al. Co-activereceptor tyrosine kinases mitigate the effect of FGFR inhibitors in FGFR1-amplified lung cancers with low FGFR1 protein expression. Oncogene2015;35:3587–97.

    39. Jia S, Liu Z, Zhang S, Liu P, Zhang L, Lee SH, et al. Essential roles of PI(3)K-p110beta in cell growth,metabolism and tumorigenesis. Nature 2008;454:776–9.

    40. Wee S, Wiederschain D, Maira SM, Loo A, Miller C, deBeaumont R, et al.PTEN-deficient cancers depend on PIK3CB. Proc Natl Acad Sci U S A2008;105:13057–62.

    41. Edgar KA, Wallin Jj Fau - Berry M, Berry M Fau - Lee LB, Lee Lb Fau - PriorWW, Prior Ww Fau - Sampath D, Sampath D Fau - Friedman LS, et al.

    FGFR and PI3K Inhibitors in FGFR2mutant Endometrial Cancer

    www.aacrjournals.org Mol Cancer Ther; 16(4) April 2017 647

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • Isoform-specific phosphoinositide 3-kinase inhibitors exert distinct effectsin solid tumors. Cancer Res 2010;70:1164–72.

    42. Juric D, Castel P, GriffithM,GriffithOL,WonHH, Ellis H, et al. Convergentloss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature2015;518:240–4.

    43. Schmit F, Utermark T, Zhang S, Wang Q, Von T, Roberts TM, et al. PI3Kisoformdependence of PTEN-deficient tumors can be altered by the geneticcontext. Proc Natl Acad Sci U S A 2014;111:6395–400.

    44. Utermark T, Schmit F, Lee SH, Gao X, Schaffhausen BS, Roberts TM. Thephosphatidylinositol 3-kinase (PI3K) isoform dependence of tumor for-mation is determined by the genetic mode of PI3K pathway activationrather than by tissue type. J Virol 2014;88:10673–9.

    45. Utermark T,RaoT,ChengH,WangQ, Lee SH,WangZC, et al. Thep110alphaand p110beta isoforms of PI3K play divergent roles in mammary glanddevelopment and tumorigenesis. Genes Dev 2012;26:1573–86.

    46. Myers AP.New strategies in endometrial cancer: targeting the PI3K/mTOR pathway–the devil is in the details. Clin Cancer Res 2013;19:5264–74.

    47. Packer LM, Rana S, Hayward R, O'Hare T, Eide CA, Rebocho A, et al.Nilotinib and MEK inhibitors induce synthetic lethality through paradox-ical activation of RAF in drug-resistant chronic myeloid leukemia. CancerCell 2011;20:715–27.

    48. Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, Alimonti A, et al.Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest 2008;118:3065–74.

    49. Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H,et al. Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J Clin Invest2008;118:3051–64.

    Mol Cancer Ther; 16(4) April 2017 Molecular Cancer Therapeutics648

    Packer et al.

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/

  • 2017;16:637-648. Published OnlineFirst January 23, 2017.Mol Cancer Ther Leisl M. Packer, Xinyan Geng, Vanessa F. Bonazzi, et al.

    Endometrial CancersmutantAntitumor Responses in FGFR2PI3K Inhibitors Synergize with FGFR Inhibitors to Enhance

    Updated version

    10.1158/1535-7163.MCT-16-0415doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2017/01/21/1535-7163.MCT-16-0415.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://mct.aacrjournals.org/content/16/4/637.full#ref-list-1

    This article cites 46 articles, 21 of which you can access for free at:

    Citing articles

    http://mct.aacrjournals.org/content/16/4/637.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://mct.aacrjournals.org/content/16/4/637To request permission to re-use all or part of this article, use this link

    on June 5, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst January 23, 2017; DOI: 10.1158/1535-7163.MCT-16-0415

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-16-0415http://mct.aacrjournals.org/content/suppl/2017/01/21/1535-7163.MCT-16-0415.DC1http://mct.aacrjournals.org/content/16/4/637.full#ref-list-1http://mct.aacrjournals.org/content/16/4/637.full#related-urlshttp://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/16/4/637http://mct.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