12
Small Molecule Therapeutics Effective Targeting of Estrogen ReceptorNegative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101 Sahra Borges 1 , Edith A. Perez 1,2 , E. Aubrey Thompson 1 , Derek C. Radisky 1 , Xochiquetzal J. Geiger 3 , and Peter Storz 1 Abstract Invasive ductal carcinomas (IDC) of the breast are associated with altered expression of hormone receptors (HR), ampli- cation or overexpression of HER2, or a triple-negative pheno- type. The most aggressive cases of IDC are characterized by a high proliferation rate, a great propensity to metastasize, and their ability to resist to standard chemotherapy, hormone therapy, or HER2-targeted therapy. Using progression tissue microarrays, we here demonstrate that the serine/threonine kinase protein kinase D3 (PKD3) is highly upregulated in estrogen receptor (ER)negative (ER ) tumors. We identify direct binding of the ER to the PRKD3 gene promoter as a mechanism of inhibition of PKD3 expression. Loss of ER results in upregulation of PKD3, leading to all hallmarks of aggressive IDC, including increased cell proliferation, migra- tion, and invasion. This identies ER breast cancers as ideal for treatment with the PKD inhibitor CRT0066101. We show that similar to a knockdown of PKD3, treatment with this inhibitor targets all tumorigenic processes in vitro and decreases growth of primary tumors and metastasis in vivo. Our data strongly support the development of PKD inhibitors for clinical use for ER breast cancers, including the triple- negative phenotype. Mol Cancer Ther; 14(6); 111. Ó2015 AACR. Introduction Invasive ductal carcinomas (IDC) of the breast are among the most aggressive types of cancer affecting women. IDCs with the worst outcome are associated either with loss of expression of hormone receptors, estrogen receptor (ER) and progesterone receptor (PR), overexpression, or amplication of the human epidermal growth factor receptor-2 (HER2/ErbB2), or a triple- negative phenotype [lack of expression of HER2 and both hormone receptors (HR]. The most aggressive cases of IDC are characterized by a high proliferation rate, a great propensity to metastasize, and their ability to resist to standard chemother- apy, hormone therapy, or HER2-directed agents such as tras- tuzumab (1, 2). Particularly, triple-negative breast cancers (TNBC), which represent approximately 20% of all cases, are difcult to treat, because molecular targets are lacking (3, 4). Cytotoxic radio- and chemotherapy currently are the only options for patients with TNBC (5). However, the rate of recurrence is high after such treatment (6). The high resistance to therapy has been partially attributed to tumors that show mesenchymal and stem cell features, for which no specic targeted therapies are available (79). Thus, identication of tissue biomarkers that may lead to novel therapeutic strategies is of utmost importance. The protein kinase D (PKD) family members PKD1, PKD2, and PKD3 have been implicated in the progression of breast cancer. PKD1 contributes to breast cancer cell proliferation (10), but inhibits the invasive phenotype (11, 12). This is mediated by inhibition of epithelial-to-mesenchymal transition (EMT) through phosphorylation of Snail (1316), downregulation of the expression of several matrix metalloproteinases (MMP), and negative-regulation of F-actin reorganization at the leading edge at multiple levels (12, 13, 17, 18). Consequently, PRKD1 (encod- ing PKD1) is silenced by hypermethylation in the most aggressive breast cancers, including the TNBC subtype (11, 19). In contrast to PKD1, the two other isoforms PKD2 and PKD3, in breast cancer cell lines seem to drive all aspects of oncogenic transformation, including cell proliferation, migration, invasion, and chemore- sistance (2022). Similar opposing functions in breast cancer have been described for other kinases such as members of the Akt/ PKB kinase family (23, 24). However, how subtypes of the same kinase family, which recognize the same substrate phosphoryla- tion motif, can have opposite cellular functions remains unclear. On the basis of the recent studies for PKD enzymes, it seems that a number of different parameters, such as their relative level of expression or activity, their cellular localization, and/or their ability to form complexes, can differentially inuence cellular phenotypes (25). Using progression tissue microarrays (TMA), here we dem- onstrate that a switch toward the isoform PKD3 is associated with the aggressiveness of breast cancer. Although PKD1 is downregulated and PKD2 is expressed homogeneously at low 1 Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida. 2 Department of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida. 3 Laboratory Medicine and Pathology, Mayo Clinic, Jackson- ville, Florida. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Peter Storz, Mayo Clinic, Grifn Room 306, 4500 San Pablo Road, Jacksonville, FL 32224. Phone: 904-953-6909; Fax: 904-953-0277; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-14-0945 Ó2015 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org OF1 Research. on January 13, 2020. © 2015 American Association for Cancer mct.aacrjournals.org Downloaded from Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

Small Molecule Therapeutics

Effective Targeting of Estrogen Receptor–Negative Breast Cancers with the ProteinKinase D Inhibitor CRT0066101Sahra Borges1, Edith A. Perez1,2, E. Aubrey Thompson1, Derek C. Radisky1,Xochiquetzal J. Geiger3, and Peter Storz1

Abstract

Invasive ductal carcinomas (IDC) of the breast are associatedwith altered expression of hormone receptors (HR), amplifi-cation or overexpression of HER2, or a triple-negative pheno-type. The most aggressive cases of IDC are characterized by ahigh proliferation rate, a great propensity to metastasize, andtheir ability to resist to standard chemotherapy, hormonetherapy, or HER2-targeted therapy. Using progression tissuemicroarrays, we here demonstrate that the serine/threoninekinase protein kinase D3 (PKD3) is highly upregulated inestrogen receptor (ER)–negative (ER�) tumors. We identifydirect binding of the ER to the PRKD3 gene promoter as a

mechanism of inhibition of PKD3 expression. Loss of ERresults in upregulation of PKD3, leading to all hallmarks ofaggressive IDC, including increased cell proliferation, migra-tion, and invasion. This identifies ER� breast cancers as idealfor treatment with the PKD inhibitor CRT0066101. We showthat similar to a knockdown of PKD3, treatment with thisinhibitor targets all tumorigenic processes in vitro anddecreases growth of primary tumors and metastasis in vivo.Our data strongly support the development of PKD inhibitorsfor clinical use for ER� breast cancers, including the triple-negative phenotype. Mol Cancer Ther; 14(6); 1–11. �2015 AACR.

IntroductionInvasive ductal carcinomas (IDC) of the breast are among the

most aggressive types of cancer affecting women. IDCs with theworst outcome are associated either with loss of expression ofhormone receptors, estrogen receptor (ER) and progesteronereceptor (PR), overexpression, or amplification of the humanepidermal growth factor receptor-2 (HER2/ErbB2), or a triple-negative phenotype [lack of expression of HER2 and bothhormone receptors (HR]. The most aggressive cases of IDC arecharacterized by a high proliferation rate, a great propensity tometastasize, and their ability to resist to standard chemother-apy, hormone therapy, or HER2-directed agents such as tras-tuzumab (1, 2). Particularly, triple-negative breast cancers(TNBC), which represent approximately 20% of all cases, aredifficult to treat, because molecular targets are lacking (3, 4).Cytotoxic radio- and chemotherapy currently are the onlyoptions for patients with TNBC (5). However, the rate ofrecurrence is high after such treatment (6). The high resistanceto therapy has been partially attributed to tumors that show

mesenchymal and stem cell features, for which no specifictargeted therapies are available (7–9). Thus, identification oftissue biomarkers that may lead to novel therapeutic strategiesis of utmost importance.

The protein kinaseD (PKD) familymembers PKD1, PKD2, andPKD3 have been implicated in the progression of breast cancer.PKD1 contributes to breast cancer cell proliferation (10), butinhibits the invasive phenotype (11, 12). This is mediated byinhibition of epithelial-to-mesenchymal transition (EMT)through phosphorylation of Snail (13–16), downregulation ofthe expression of several matrix metalloproteinases (MMP), andnegative-regulation of F-actin reorganization at the leading edgeat multiple levels (12, 13, 17, 18). Consequently, PRKD1 (encod-ing PKD1) is silenced by hypermethylation in themost aggressivebreast cancers, including the TNBC subtype (11, 19). In contrast toPKD1, the two other isoforms PKD2 and PKD3, in breast cancercell lines seem to drive all aspects of oncogenic transformation,including cell proliferation, migration, invasion, and chemore-sistance (20–22). Similar opposing functions in breast cancerhave been described for other kinases such asmembers of the Akt/PKB kinase family (23, 24). However, how subtypes of the samekinase family, which recognize the same substrate phosphoryla-tion motif, can have opposite cellular functions remains unclear.On the basis of the recent studies for PKD enzymes, it seems that anumber of different parameters, such as their relative level ofexpression or activity, their cellular localization, and/or theirability to form complexes, can differentially influence cellularphenotypes (25).

Using progression tissue microarrays (TMA), here we dem-onstrate that a switch toward the isoform PKD3 is associatedwith the aggressiveness of breast cancer. Although PKD1 isdownregulated and PKD2 is expressed homogeneously at low

1Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.2Department of Hematology/Oncology, Mayo Clinic, Jacksonville,Florida. 3Laboratory Medicine and Pathology, Mayo Clinic, Jackson-ville, Florida.

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

Corresponding Author: Peter Storz, Mayo Clinic, Griffin Room 306, 4500 SanPablo Road, Jacksonville, FL 32224. Phone: 904-953-6909; Fax: 904-953-0277;E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-14-0945

�2015 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org OF1

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 2: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

levels in different breast cancer subtypes as well as in normaltissue, PKD3 is highly upregulated in ER-negative (ER�) tumors.We identify estrogen-dependent signaling as the mechanismof inhibition of PKD3 expression in ER-expressing ductal cancercells. Loss of ER results in upregulation of PKD3, leading toincreased cell proliferation, migration, and invasion. These dataidentify ER� breast cancers as ideal cancers for treatment withthe PKD inhibitor CRT0066101, because they express little orno PKD1 and high levels of PKD3. We show that, similar to aknockdown of PKD3, treatment with this inhibitor targetsmost tumorigenic processes in vitro, and also decreases growthof primary tumors and prevents metastasis in vivo. Thus, becauseit can be given orally, it may be developed for treatmentof PKD1-negative breast cancers, including the triple-negativephenotype.

Materials and MethodsCell lines, antibodies, and reagents

The MDA-MB-231-luc2 cell line was obtained from PerkinEl-mer in May 2009. All other cells lines were obtained from theAmerican Type Culture Collection in August 2008. All cell lineswere not further authenticated. MCF-7, MDA-MB-231, andT47D cells were maintained in Dulbecco's Modified EagleMedium (DMEM) with 10% fetal bovine serum (FBS). MDA-MB-468 and HCC1954 were maintained in RPMI-1640 with10% FBS. BT20 cells were maintained in Eagle's minimal essen-tial medium with 10% FBS, 2 mmol/L L-glutamine, 1.5 g/Lsodium bicarbonate, 0.1 mmol/L nonessential amino acids(NEAA), and 1 mmol/L sodium pyruvate. MCF-10A cells weremaintained in DMEM/Ham's F-10 medium (50:50 v/v) with 5%horse serum, 20 ng/mL EGF, 0.5 mg/mL hydrocortisone, 100ng/mL cholera toxin, 10 mg/mL insulin, and 1% penicillin/streptomycin. NEAAs were obtained from Mediatech, EGF fromPeproTech, and insulin and hydrocortisone from Sigma-Aldrich.Anti-b-actin antibody was obtained from Sigma-Aldrich; anti-Ki67 from Dako; anti-cleaved PARP, anti-cleaved caspase-3, andanti-MMP9 from Cell Signaling Technology; anti-COX-2 fromCayman Chemical; anti-smooth muscle actin (SMA), anti-GFP,and anti-cyclin D1 from Abcam; anti-Snail from Abgent; anti-vimentin (for Western blotting) from Santa Cruz Biotechnology;and anti-N-cadherin and anti-vimentin [for immunohistochem-istry (IHC)] from Epitomics. The rabbit polyclonal antibodyspecific for PKD2 was from Upstate Biotechnology and therabbit polyclonal antibody specific for PKD3 used for immu-noblotting was from Bethyl Laboratories. The mouse monoclo-nal antibody specific for PKD3 (H00023683-M01) used forIHC was from Abnova. The mouse monoclonal antibody spe-cific for PKD1 was raised by Creative Biolabs/Creative Dynamicsand is further described in ref. (11). Secondary horseradishperoxidase (HRP)–linked antibodies were obtained from RocheApplied Science. Luciferin was obtained from Gold Biotechnol-ogy. Fulvestrant and b-estradiol (E2) were purchased fromSigma-Aldrich. The PKD-specific inhibitor CRT0066101 wasobtained from Cancer Research Technology.

Lentiviral shRNA expression and shRNA constructsSpecific lentiviral expression constructs for short hairpin RNA

(shRNA) targeting human PKD3 were purchased from Sigma-Aldrich (MISSION shRNA Plasmid DNA). Constructs used wereNM_005813.x-3393s1c1 (labeled as PKD3-shRNA#1) andNM_005813.x-2494s1c1 (labeled as PKD3-shRNA#2). Lentivirus

was produced in HEK293FT cells using the ViraPower LentiviralExpression System (Life Technologies). MDA-MB-231 cellswere infected with PKD3-shRNA lentivirus to generate stable celllines. After infection, cell pools were selected using puromycin(1 mg/mL) for 15 days.

Plasmids and transfectionsTo generate a PKD3 promoter-luciferase reporter, the human

PRKD3 promoter region (�1000 to þ3) was cloned in pGL3plasmid from Promega via BglII and XhoI restriction sites, using50-TTTTTTGTCCCTTCTGTTTTTGAT-30 and 50-GACGGAAAGAAA-TTAGAAAATTTT-30 as primers. The pRL-CMV-Renilla luciferaseplasmid was from Promega. The ERa (pEGFP-C1-ERa; #28230)expression plasmid was from Addgene. The pSuper-PKD2-shRNAplasmid was obtained by cloning the oligonucleotides 50-GAT-CCCCGTTCCCTGAGTGTGGCTTCTTCAAGAGAGAAGCCACAC-TCAGGGAACTTTTTGGAAA-30 and 50-AGCTTTTCCAAAAAGTT-CCCTGAGTGTGGCTTCTCTCTTGAAGAAGCCACACTCAGGGA-ACGGG-30 into pSuper. GenJet from SignaGen was used fortransfection of breast cancer cells.

Cell lysates and Western blot analysisCells were washed twice with ice-cold phosphate-buffered

saline (PBS; 140 mmol/L NaCl, 2.7 mmol/L KCl, 8 mmol/LNa2HPO4, 1.5 mmol/L KH2PO4, pH 7.2) and lysed with BufferA (50 mmol/L Tris–HCl, pH 7.4, 1% Triton X-100, 150 mmol/LNaCl, 5 mmol/L EDTA, pH 7.4) plus Protease Inhibitor Cocktail(Sigma-Aldrich). Lysates were used for Western blot analysis asdescribed previously (12).

ImmunofluorescenceCells were seeded in 8-well ibiTreat m-Slides (ibidi) and treated

as indicated. Before fixation with 4% paraformaldehyde (20min-utes, 4�C), cells were washed twice with PBS. Fixed cells werewashed three times in PBS, permeabilizedwith 0.1%Triton X-100in PBS (2 minutes, room temperature), and then blocked withblocking solution [3% bovine serum albumin (BSA) and 0.05%Tween 20 in PBS] for 30minutes at room temperature. F-actinwasstained with Alexa Fluor 633–Phalloidin (Life Technologies),nuclei with 40,6-diamidino-2-phenylindole dihydrochloride(DAPI; Sigma-Aldrich) in blocking solution. After extensivewashes with PBS, cells were mounted in ibidi mounting medium(ibidi). Samples were examined using an IX81 DSU SpinningDisc Confocal from Olympus with a 40� objective.

Proliferation, migration, and invasion assaysTranswell migration and invasion assays were performed as

described previously (12). Briefly, Transwell chambers were leftuncoated (migration assay) or coated with Matrigel (2 mg/well;BD Biosciences), dried overnight, and rehydrated for 1 hour with40 mL of tissue culture media. Cells were harvested, washed oncewith media containing 1% BSA, and resuspended in mediacontaining 0.1% BSA. Then, 100,000 cells were seeded per Trans-well insert. NIH-3T3 conditioned medium served as a chemoat-tractant in the lower chamber. Remaining cells were used tocontrol the expression of genes of interest by Western blot anal-ysis. After 16 hours, cells on top of the Transwell insert wereremoved and cells that hadmigrated/invaded to the lower surfaceof the filters were fixed in 4% paraformaldehyde, stained withDAPI, and counted. For impedance-based real-time chemotacticassays, cells were seeded onto an E-Plate (for proliferation assays)

Borges et al.

Mol Cancer Ther; 14(6) June 2015 Molecular Cancer TherapeuticsOF2

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 3: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

or a CIM-Plate 16 Transwell (for migration/invasion assays) fromRoche Applied Science. After attachment, cell migration or inva-sion (coating of top well with 2 mg of Matrigel) toward NIH-3T3conditioned media was continuously monitored in real time forthe indicated times using the xCELLigence RTCA DP Instrumentfrom Roche Applied Science.

Patient samples, TMAs, and IHCTissue samples were initially collected with the approval of the

Mayo Clinic Institutional Review Board (IRB) under protocolMC0033. Written informed consent for the use of these tissuesin research was obtained from all participants. Generation of theTMA was performed under protocol 09-001642. Therefore, allunique patient identifiers and confidential data were removedand tissue samples were de-identified. The Mayo Clinic IRBassessed the protocol 09-001642 as minimal risk and waived theneed for further consent. All data were analyzed anonymously.TMAs were deparaffinized (1 hour at 60�C), dewaxed in xylene(five times for 4 minutes), and gradually rehydrated with ethanol(100%, 95%, and 75%, twice with each concentration for 3minutes). The rehydrated TMA sections were rinsed in water andsubjected to hematoxylin and eosin (H&E) staining or to antigenretrieval in citrate buffer (pH 6.0) as previously described (26).Slides were treated with 3% hydrogen peroxide (5 minutes) toreduce endogenous peroxidase activity and washed with PBScontaining 0.5% Tween 20 (PBS–Tween 20). Proteins of interestwere detected using indicated specific antibodies diluted in PBS–Tween 20 and visualized using the EnVisionþDual Link LabelledPolymer Kit following the manufacturer's instructions (Dako).Images were captured using the Aperio ScanScope slide scanner(Aperio).

Orthotopic tumor models and treatmentAnimal experiments were performed under protocols A43213

and A17313 approved by the Mayo Clinic Institutional AnimalCare and Use Committee (IACUC). Female nonobese diabeticsevere combined immunodeficiency (NOD scid) mice wereanesthetized, and 500,000 cells washed three times in PBS andmixed with 30 mL of complete Matrigel (BD Biosciences) wereinjected into the fourth mammary gland on the right side ofeach animal. As indicated, cell lines used were MDA-MB-231.Luc (MDA-MB-231-luc2 from PerkinElmer; additionally expres-sing luciferase), MDA-MB-231.Luc stably expressing controlshRNA (scr-shRNA), or two different shRNAs specifically target-ing PKD3 (PKD3-shRNA#1 and PKD3-shRNA#2). For studieswith CRT0066101, mice were treated orally with 80 mg/kgCRT0066101 diluted in a 5% dextrose saline solution (Sig-ma-Aldrich) or 5% dextrose saline solution alone (control)every other day starting 14 days after cell injection. Body weightand tumor volume (caliper measurement) were determinedonce per week. The presence of metastases was detected usingthe IVIS Spectrum Imaging System (PerkinElmer). At the end-point, primary tumors and sites of metastasis were removed andanalyzed as indicated.

Luciferase reporter assayCells were transfected with PRKD3 promoter-luciferase re-

porter (2 mg), Renilla luciferase reporter (0.1 mg), and pEGFP-ERa expression construct (2 mg) in 6-well plates, as indicated.Twenty-four hours after transfection, cells were washed twicewith ice-cold PBS, scraped in 250-mL Passive Lysis Buffer

(Promega), and centrifuged (13,000 rpm, 10 minutes, 4�C).Assays for luciferase activity were performed according to thePromega Luciferase assay protocol and measured using a Ver-itas luminometer (Symantec). Luciferase activity of the PRKD3promoter-luciferase reporter was normalized to Renilla luci-ferase activity. Expression of proteins was controlled by West-ern blot analysis.

Chromatin immunoprecipitationChromatin immunoprecipitation (ChIP) assays were per-

formed using the Imprint Chromatin Immunoprecipitation Kitfrom Sigma-Aldrich according to the manufacturer's protocol.Five micrograms of primary antibody (anti-GFP, Abcam) orrabbit IgG control was used for ChIPs. Immunoprecipitates wereanalyzed by PCR using the primer sets 50-TGACAATGCCTGT-CAGCTTC-30 and 50-AAACGCGAATGTGACCCTAC-30 to amplifya 243 bp fragment (ERa site 1) or 50-TGATAGACACGCTCGC-GACT-30 and 50-TGCCGGGAGCTGTAGTTCCT-30 to amplify a199-bp fragment (ERa site 2) of the human PRKD3 promoter.

BioinformaticsEvaluation of expression of PRKD3 in annotated breast cancer

datasets was performed using the Nextbio server (www.nextbio.com; ref. 27). KM Plotter data were obtained using the currentrelease of Kaplan–Meier Plotter [www.kmplot.com; (28); 2012version, n ¼ 2,978], interrogating using Affymetrix ID:"211084_x_at," survival set at distant metastasis-free survival,auto select best cutoff set at checked, follow-up threshold set atall, and array quality control set at exclude biased arrays.

Statistical analysisGraphPad Prism version 4.0c software (GraphPad Software)

was used for all statistical analyses. Statistical significance (P <0.05) was determined using a two-tailed Student t test andstandard deviations.

ResultsPKD3 is highly expressed in ER� breast cancers and correlateswith aggressiveness

Loss of gene expression of PRKD1 (encoding for PKD1) is amarker for aggressive breast cancer (11, 17). Using isoform-specific antibodies, we determined the expression pattern ofPKD1 and the two oncogenic versions of this kinase family,PKD2 and PKD3, in normal breast (n ¼ 60 samples) and TNBC(n ¼ 40 samples). Whereas PKD1 is the main isoform expressedin normal breast, TNBC show an isoform switch toward expres-sion of PKD3 (Fig. 1A). PKD2 generally was weakly expressed,but is also slightly downregulated in TNBC. In order to evaluatewhether increased PKD3 expression is indeed linked to thetriple-negative phenotype, we evaluated annotated clinicalbreast cancer datasets from 13 different studies. In all studies,triple-negative biopsy samples showed significantly increasedexpression of PRKD3 as compared with triple-positive (ERþ/PRþ/HER2þ) biopsies (Supplementary Table S1). Using TheGene Set Analysis Cell Lines module of GOBO (29, 30), wethen analyzed a panel of 51 breast cancer cell lines and alsofound a reverse correlation of PRKD3 gene expression betweenbasal and luminal breast cancer types (Fig. 1B, left). A similarreverse correlation was found between TNBC and HR-positive

Targeting of ER� Breast Cancer with CRT0066101

www.aacrjournals.org Mol Cancer Ther; 14(6) June 2015 OF3

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 4: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

cell lines (Fig. 1B, right), indicating that loss of HR expressionand increased PRKD3 expression may be linked. To test this, weanalyzed ER-positive (ERþ; n ¼ 44) and ER� (n ¼ 41) patienttumors for PKD3 expression and confirmed that increased PKD3expression is linked to loss of ER expression (Fig. 1C andSupplementary Fig. S1). The negative correlation between ERand PKD3 was bolstered by meta-analysis of 24 publishedstudies in which we found that PRKD3 shows significantlyhigher expression in ER� versus ERþ breast cancers (Supple-mentary Table S2). We next assessed the association of PRKD3expression with prognosis in breast cancer patients, and foundthat elevated PRKD3 expression levels in ER� tumors (n ¼1,353) were associated with significantly decreased distantmetastasis-free survival (Fig. 1D).

ERa regulates PKD3 expression levels in breast cancer cellsTo determine whether ERa could directly affect PRKD3

promoter activity, we performed luciferase reporter assays usinga PRKD3 promoter-luciferase gene reporter. Reintroductionof ERa into MDA-MB-231 (ER�/PR�/HER2�) cells led to asignificant decrease (approximately 40%) of PRKD3 gene pro-moter activity (Fig. 2A). These data were confirmed with BT20,another TNBC cell line. BT20 cells, when transfected with ERa,showed an approximately 75% decrease of PRKD3 expressionand this was further decreased, when media were supplement-ed with E2 (Fig. 2B). We analyzed the PRKD3 promotersequence and identified two potential ER-binding sites, 780bases (ERa site 1) or 318 bases (ERa site 2), upstream of thetranscription start site (Fig. 2C). To test whether ERa directly

HER2–, ER+ HER2+, ER+ HER2+, ER–

Invasive ductal carcinoma

0

1

2

3

ER+ ER–

PK

D3

inte

nsi

ty

(AU

)

*

0

–1

–2

1

2

-

n = 25 n = 15

PR

KD

3 L

og

2 ex

pre

ssio

n

PR

KD

3 L

og

2 ex

pre

ssio

n

0

–1

–2

1

2

n = 12 n = 14 n = 25

0

1

2

3

Normal TNBC

*PK

D1

inte

nsi

ty(A

U)

Normal TNBC

PK

D2

inte

nsi

ty(A

U)

*

1.5

0.5

0

1

0

1

2

3

4

Normal TNBC

PK

D3

inte

nsi

ty(A

U)

*

PK

D1

PK

D2

PK

D3

NormalA B

C

D

TNBC

ER-

Dis

tan

t m

etas

tasi

s-

free

su

rviv

al

Time (years)

Low PRKD3 High PRKD3

0

0.0

0.2

0.4

0.6

0.8

1.0

5 10 15 20

HR = 1.68 (1.14–2.47) log-rank P = 0.0079

Figure 1.PKD3 is highly expressed in ER� breast cancers and correlates with aggressiveness. A, TMA slides containing human TNBC (n ¼ 40) and normal humanbreast tissue samples (n ¼ 60) were analyzed for PKD1, PKD2, and PKD3 expression using isoform-specific antibodies. Scale bar, 100 mm. Statisticalanalysis of PKD1, PKD2, and PKD3 intensity was performed using Aperio positive pixel count algorithm in the Imagescope software (Aperio). B, analysisof PRKD3 gene expression in breast cancer cells lines using the Gene Set Analysis Cell Lines module of GOBO. Cell lines were grouped into Basal A(n ¼ 12), Basal B (n ¼ 14), or Luminal (n ¼ 25) subtypes (left), or TNBC (n ¼ 25) and HR-positive (n ¼ 15) subtypes (right). C, TMA slidescontaining histologically confirmed IDC of indicated subtypes were analyzed for PKD3 expression using an isoform-specific antibody. Scale bar, 100 mm.Statistical analysis of PKD3 intensity in ERþ or ER� groups (n ¼ 44 for ERþ; n ¼ 41 for ER�) was performed using Aperio positive pixel countalgorithm in the Imagescope software (Aperio). D, the Kaplan–Meier plot depicting the impact of levels of PRKD3 gene expression on the metastasis-freesurvival of patients (n ¼ 1,353) with ER� breast cancer. In A and C, P values were acquired with the Student t test using Prism v5 software. � , P < 0.05,statistical significance. In A and C, representative pictures from each group are depicted.

Borges et al.

Mol Cancer Ther; 14(6) June 2015 Molecular Cancer TherapeuticsOF4

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 5: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

binds to the PKD3 promoter at these sites, we performed ChIPand confirmed the direct binding of ERa to both of the twopredicted ER-binding sites (Fig. 2D). Next, we determined

whether presence of ERa translates to a decrease in PKD3protein expression. Therefore, we reintroduced ERa in triple-negative MDA-MB-231 and BT20 cells, as well as HCC1954

0

50

100

150PR

KD

3 pr

omot

er a

ctiv

ity(%

of c

ontro

l)

ERα

ERα

ERα

ERαERα

ERα

ERα

ERαERα

46

WB: anti-GFP80

WB: anti-β-actin

kDa

*

Luciferase Assay0

25

50

75

100

125

PRK

D3

prom

oter

act

ivity

(% o

f con

trol)

46

80

– – + + β-Estradiol

**

ERα

β-Actinβ-Actin

WB: anti-GFP

WB: anti-β-actin

kDa

Luciferase Assay

#

TGGTGGTAGGGTCACATTCGCGTT TAAAGG

–785 –756

CGGGGCAGAGACAACCCCGACCTCCCGCC

–324 –296 ERα site 2

PRKD3 promoter

ERα site 1 ERα-bound PRKD3 promoter

PCR: PRKD3promoter (site 1)

Input control

PCR: PRKD3promoter (site 2)

ERα-bound PRKD3 promoter

PCR: PRKD3promoter (site 1)

PCR: PRKD3promoter (site 2)

Input control

PKD3

β-Actin

WB: anti-PKD3

MDA-MB-231 HCC1954BT20

kDa

80

80

46

ERα

WB: anti-GFP

WB: anti-β-actin

46

WB: anti-β-actin

β-Actin

PKD3WB: anti-PKD3

T47D

kDa

80

BT20MDA-MB-231

A B

C D

E F

Figure 2.ERa directly regulates PKD3 expression in breast cancer cells. A, MDA-MB-231 cells were transfected with PRKD3 promoter-luciferase, Renilla luciferasereporters and vector control or GFP-tagged ERa, as indicated. Forty-eight hours after transfection, reporter gene luciferase assays were performed.� , P < 0.05, statistical significance. Cell lysates were also analyzed by Western blot analysis for expression of ERa (anti-GFP), or b-actin (anti-b-actin)as a loading control. B, BT20 cells were cultivated in phenol-red–free media and transfected with PRKD3 promoter-luciferase and Renilla luciferasereporters and vector control or ERa, as indicated. Forty-eight hours after transfection, BT20 cells were stimulated with 10 nmol/L E2 or vehicle (control)for 6 hours and reporter gene luciferase assays were performed. �, P < 0.05, statistical significance as compared with unstimulated vector control; #, statisticalsignificance as compared with unstimulated ERa-transfected cells. Cell lysates were also analyzed by Western blot analysis for expression of ERa (anti-GFP),or b-actin (anti-b-actin) as a loading control. C, schematic representation of potential ERa-binding sites (in grey italic) in the PRKD3 promoter region �1000to �0. D, chromatin IP. MDA-MB-231 cells were transfected with vector control or GFP-tagged ERa. After cross-linking, the ERa–DNA complexes wereimmunoprecipitated using an anti-GFP antibody. Precipitates were analyzed by PCR for the ERa-bound PRKD3 promoter. A PCR for the PRKD3 promoterusing the input DNA served as a control (input control). E, ER� cell lines MDA-MB-231, BT20, and HCC1954 were transfected with vector control or GFP-taggedERa. Forty-eight hours after transfection, cell lysates were analyzed by Western blot analysis for the expression of PKD3 and ERa (anti-GFP).Staining for b-actin (anti-b-actin) served as a loading control. F, the ERþ cell line T47D was treated with 100 nmol/L fulvestrant or DMSO (control) for24 hours and cell lysates were analyzed by Western blot analysis for the expression of PKD3 or b-actin (anti-b-actin).

Targeting of ER� Breast Cancer with CRT0066101

www.aacrjournals.org Mol Cancer Ther; 14(6) June 2015 OF5

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 6: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

(ER�/PR�/HER2þ) cells. As expected, the presence of ERa ledto a significant decrease in PKD3 expression, and this wasindependent of the HER2 amplification status (Fig. 2E). Even-tually, we treated the ERþ cell line T47D with fulvestrant, acompound that leads to nuclear export and degradation ofER (31, 32). As expected, treatment of T47D cells with fulves-trant increased PKD3 expression further indicating a role forER as a regulator of PKD3 expression (Fig. 2F).

The knockdown of PKD3 decreases cancer cell proliferation,migration, and invasion in vitro and in vivo

To test the impact of PKD3 on breast cancer cell behavior, weused the invasive breast cancer cells MDA-MB-231 (ER�, PR�,

HER2�), which express high levels of PKD3, low levels of PKD2,and no PKD1 (Supplementary Fig. S2; Supplementary Table S3;ref. 11). A knockdown of basal PKD3 expression using twodifferent PKD3-specific shRNA sequences significantly decreasedMDA-MB-231 cell numbers over a time period of 60 hours(Fig. 3A and B and Supplementary Fig. S3). Similar results wereobserved in another ER� cell line, HCC1954, which show asimilar PKD expression pattern (Supplementary Fig. S2; datanot shown). Next, we tested the role of PKD3 on the invasivephenotype. The knockdown of PKD3 in MDA-MB-231 cells ledto a dramatic decrease in directed cell migration (Fig. 3C andSupplementary Fig. S4), and similar effects were observed forcell invasion through extracellular matrix (Fig. 3D). This confirms

WB: anti-PKD3

WB: anti-β-actin

kDa

80

46 β-Actin

PKD3

0

20

40

60

80

100

120

Cel

l inv

asio

n (%

of c

ontro

l)

*

*Transwell Invasion Assay

Scr-shRNA

PKD3-shRNA#1

PKD3-shRNA#2

0

5,000

10,000

15,000

20,000

25,000

Cel

l are

a (s

quar

e pi

xel)

*

*

Scr-shR

NA

PK

D3-shR

NA#1

PK

D3-shR

NA#2

LungsLymph nodes

Time (h)2 4 6 8 10

Cel

l mig

ratio

norm

aliz

ed c

ell i

ndex

1.0

1.5

2.0

2.5

3.0

3.5

4.0scr-shRNAPKD3-shRNA#1PKD3-shRNA#2

* *

Time (h)

A B

C

F G

D E

0 10 20 30 40 50 601

2

3

4

5

scr-shRNAPKD3-shRNA#1PKD3-shRNA#2

* *

**

n = 5 n = 5 n = 5

**

n = 5 n = 5 n = 5

Cel

lsΔ

Nor

mal

ized

cel

l ind

exM

etas

tase

s (m

m2 )

Met

asta

sis

Ave

rage

siz

e (μ

m2 )

50,000

40,000

30,000

20,000

10,000

15

10

5

0

0

Figure 3.Knockdownof PKD3 decreases cancer cellproliferation, migration, and invasion invitro and in vivo. A–E, MDA-MB-231 cellswere infected with lentivirus harboringcontrol shRNA (scr-shRNA) or twodifferent shRNAs specifically targetingPKD3 (PKD3-shRNA#1 and PKD3-shRNA#2). A, 48 hours after initialinfection, a fraction of the cells was lysedand PKD3 knockdown was verified byWestern blot analysis. B, cellswere seededin E-plates and numbers of attachedcells were continuously monitored in realtime for 60 hours using an xCELLigenceRTCA DP instrument. Error bars (gray)represent three experiments. C, cellswere seeded in CIM-plates and (afterattachment) directed cell migration wascontinuously monitored in real time for10 hours using an xCELLigence RTCA DPinstrument. Error bars (gray) representthreeexperiments. D, cellswere seededonMatrigel-coated Transwell filters andTranswell invasion assayswere performedas described in Materials and Methods. E,cells were fixed and stained with DAPI(blue) and phalloidin (red). Bars, 10 mm.Representative pictures from each groupare depicted. The cell area (graph) wasmeasured and analyzed using the ImageJsoftware. In B–E, the results presented arethe mean�SEM. P values were calculatedusing a two-tailed Student t test.� , P < 0.05, statistical significance. F andG,MDA-MB-231.Luc cells stably expressingcontrol shRNA (scr-shRNA) or twodifferent shRNAs specifically targetingPKD3 (PKD3-shRNA#1 and PKD3-shRNA#2) were injected into the mfp offemale NOD scid mice. In F, the presenceof metastasis in lymph nodes and lungswas detected by immunostaining with anantibody specific for human vimentin(detects human cancer cells).Representative pictures from each groupare depicted. Bars, 200 mm for lymphnodes; 1 mm for lungs. In G, the number ofpulmonary metastases per mm2, or theaverage size of pulmonary metastases inmm2 was quantified from five fields eachlung. � , P < 0.05, statistical significance.

Borges et al.

Mol Cancer Ther; 14(6) June 2015 Molecular Cancer TherapeuticsOF6

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 7: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

that PKD3 is the major isoform driving motility (and prolifera-tion) in these cells. Interestingly, along with a decreased motility,we noticed a dramatic increase in cell spreading and altered F-actin organization when PKD3 was knocked down (Fig. 3E). Totest whether the knockdown of PKD3 can affect breast tumormetastasis in vivo, we orthotopically implantedMDA-MB-231.Luccells either stably expressing scrambled shRNA control or twodifferent specific shRNA sequences for PKD3 into the mammaryfat pad (mfp) of female NOD scid mice. To exclude that PKD3effects on cell proliferation affect results onmetastasis, endpointsof the experiment were set for each individual mouse at a primarytumors size of 700 � 100 mm3. At the endpoint, tumors andtissues of potential sites of metastasis were extracted. As a control,primary tumors were analyzed by IHC with a monoclonal anti-body specific for PKD3 (Supplementary Fig. S5A). As predicted byour in vitro experiments, primary tumor growth was significantlyslowerwhenPKD3 expressionwas decreased (Supplementary Fig.S5A). Analysis of expression of Ki67 and cleaved caspase-3 stain-ing indicated that thiswas due to adecrease in cell proliferation, aswell as an increase in cell death (Supplementary Fig. S5B andS5C). Interestingly, PKD3-shRNA tumors showed reduced localinvasion when compared with control (scr-shRNA) tumors (Sup-plementary Fig. S5D). Next, we determined metastasis to distantorgans by immunohistochemical staining for human vimentin asa marker for human cancer cells. We found that PKD3 down-regulation dramatically decreased cancer cells infiltration tolymph nodes and lungs (Fig. 3F). Furthermore, mice implantedwith PKD3-shRNA cells had significantly fewer and smallermetastases to their lungs compared with controls (scr-shRNA; Fig. 3G). Taken together, our data indicate that PKD3plays an important role in breast tumor growth, progression, andmetastasis.

The PKD inhibitor CRT0066101 decreases cancer cellproliferation, migration, and invasion in vitro

Next, we tested the impact of PKD3 inhibition on cell pro-liferation and the invasive phenotype. Because PKD3 is upregu-lated and as such can be targeted in ER� breast cancer indepen-dently of the HER2 status, we decided to test two different celllines, MDA-MB-231 as a model for TNBC and HCC1954 as amodel for ER�, HER2þ breast cancer. We used the pan-PKDinhibitor CRT0066101, which has been shown to have antican-cer activity in pancreatic, prostate, and colorectal cancer cells (33,34). CRT0066101 induced a significant decrease in cell prolif-eration in both cell lines (Fig. 4A and D and Supplementary Fig.S6). In a similar fashion to PKD3 depletion, inhibition of PKDactivity with CRT0066101 also blocked directed cell migration(Fig. 4B and E and Supplementary Fig. S7) and invasion (Fig. 4Cand F). Similar as observed with the PKD3 knockdown in Fig. 3E,along with a decreased motility, we noticed increased spreadingand altered F-actin organization of cells that were treated withCRT0066101 (Fig. 4G and H). Overall, data obtained with PKD3knockdown and CRT0066101 were similar, although both celllines also express PKD2 that may have similar functions as PKD3in regard to regulation of cell migration and invasion (Supple-mentary Fig. S8).

CRT0066101 decreases primary tumor size, local invasiveness,and metastasis in vivo

Next, we tested whether CRT0066101 can be used as an effi-cient strategy for the treatment of tumor growth andmetastasis of

ER� cancers in vivo. Therefore, we orthotopically implantedMDA-MB-231 cells into the mfp of female NOD scid mice. After estab-lishment of primary tumors (day 14 after cell implantation),mice were treated with 80 mg/kg CRT0066101 (oral administra-tion, everyotherday)or vehicle control. At the endpoint (10weeksafter cell implantation), primary tumors and tissues of potentialsites ofmetastasis were extracted. As in a previous studymodelingpancreatic cancer (33), no toxicity was detected at this dosage ofCRT0066101 and no significant changes in body weight ordamage in tissue was observed (not shown). Treatment of micewith CRT0066101 did result in a significant decrease of primarytumor size and weight (Fig. 5A and Supplementary Fig. S9),associated with an approximately 50% decrease in tumor cellproliferation (Fig. 5B), and an increase of apoptosis (Fig. 5C). Ofnote, the effects of CRT0066101 on tumor cell viability andproliferation that were observed in vivo were in line with effectsobserved in vitro (Fig. 4). Additional analysis of tumor edges aswell as the connection of tumor cells to the normal adjacentmouse mammary tissue showed a reduced local invasion intumors treated with the PKD inhibitor (Fig. 5D). This decreasein invasiveness correlated with decreased expression of COX-2(Fig. 5E), which previously was associated with local invasion ofbreast cancer cells, as well as metastasis to the lungs (35). Indeed,IVIS imaging of animals indicated that CRT0066101 may affectmetastasis to distant organs (Fig. 6A). Immunohistochemicalanalysis for human cancer cells (IHC for anti-human vimentin)at the endpoint indicated a dramatic decrease of infiltration oftumor cells into lymph nodes in CRT0066101-treated mice (Fig.6B). Similarly, lungmetastaseswere fewer innumbers and smallerin size (Fig. 6B–D). Metastases in the lungs of the treated miceshowed a significant lower expression of Ki67, indicating that thedecrease in average size ofmetastasesmay be due to CRT0066101effects on the ability of tumor cells to proliferate in their newenvironment (Fig. 6E).

DiscussionSilencing of PKD1 and increased expression of the oncogenic

versions of this kinase family, PKD2 and PKD3, has beendescribed to contribute to progression of several epithelial can-cers, including breast cancer (11, 13, 16, 17, 20–22), gastric cancer(36), pancreatic cancer (37, 38), colorectal cancer (34), andprostate cancer (14, 39–42). We here show that the switch fromPKD1 to PKD3 expression defines the transition to an aggressivebreast tumor phenotype. PKD1 previously had been shown tomaintain the epithelial phenotype bypreventing EMT (13, 14, 16)and to negatively regulate cell migration (12), invasion (17), andmetastatic progression of breast cancer (11, 19). Consequently, ininvasive breast cancers, PKD1 expression is downregulated bypromoter hypermethylation (11, 36). The signaling mechanismsby which other PKD isoforms are (up)regulated at the transcrip-tional levels have not been identified so far.

Analysis of cell lines, TMAs from patient samples (Fig. 1), andannotated clinical breast cancer datasets showed significantlyhigher PRKD3 gene expression in ER� or TNBC biopsies (Sup-plementary Table S1). A detailed analysis suggested that increasedPKD3 expression is mainly due to loss of ER expression and notdependent on the HER2 status of tumors (Fig. 1C and Supple-mentary Table S2). This led to the questions whether the ER couldbe a direct negative regulator of PRKD3 expression; or whetherobserved reverse expression between both molecules is

Targeting of ER� Breast Cancer with CRT0066101

www.aacrjournals.org Mol Cancer Ther; 14(6) June 2015 OF7

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 8: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

correlative. By reexpressing ER in ER� cell lines (Fig. 2E) orinhibiting ER expression in ERþ T47D cells (Fig. 2F), we clearlydemonstrate that PKD3 repression depends on ER activity. As amechanism of regulation, we demonstrate that ER decreasesPKD3 expression through direct binding to the PRKD3 promoterat two different ER-binding sites (Fig. 2C and D). Thus, our datademonstrate a direct negative regulation of a gene by this receptor.Although ER is mostly known for its positive effect on genetranscription, some studies have demonstrated that it can alsoact as a repressor of gene expression (43, 44). For example, ER canrepress a cytochrome P450-encoding gene (CYP1A1) by targetingDnmt3B DNA methyltransferase (44).

PKD3 has been implicated in all aspects of tumor formationand progression, such as mediating proliferation, survival, andinvasiveness, in different cancers (20, 22, 39, 45). However,relatively little is known about the molecular mechanisms bywhich PKD3may drive carcinogenesis. PKD3 previously has been

shown to mediate activation of Akt, leading to prolonged acti-vation of extracellular signal–regulated kinase (ERK) 1/2 (39).Furthermore, S6 kinase 1 (S6K1), a member of the mammaliantarget of rapamycin complex 1 signaling cascade (mTORC1), wasidentified as a downstream target of PKD3 tomediate its effect oncell proliferation in TNBC cell lines (22). Another target involvedmaybe theG-protein–coupled receptor kinase-interacting protein1 (GIT1), a key mediator of PKD3-induced cell spreading andproliferation (46).

Besides describing a previously unknown regulation of PKD3expression that can be linked to aggressiveness of breast cancers,we also tested the use of PKD inhibitors in breast cancers thatmainly express PKD3. Our data and previous work implicate thatideal targets to test the efficacy of PKD inhibitors would be ER� ortriple-negative, invasive breast cancers, in which upregulation ofPKD3 is accompanied by the loss of PKD1 expression.CRT0066101 is a selective and potent PKD inhibitor that targets

*

0

20

40

60

80

100

120

Cel

l inv

asio

n (%

of c

ontro

l)

Transwellinvasion assay

MDA-MB-231

0

20

40

60

80

100

120

Cel

l mig

ratio

n (%

of c

ontro

l)

Transwell migration assay

*

HCC1954

0

20

40

60

80

100

120

Cel

l inv

asio

n(%

of c

ontro

l)Transwell invasion assay

*

HCC1954

4 6 8 10 12C

ell m

igra

tion

Δ no

rmal

ized

cel

l ind

ex

1.0

1.1

1.2

1.3

1.4

1.5

*

MDA-MB-231

20 40 60 80 1000

2

4

6

8

10

12

14

*

HCC1954

Time (h)0 10 20 30 40 50 60

Cel

ls

Δ no

rmal

ized

cel

l ind

ex

2

4

6

8 DMSOCRT0066101

*

MDA-MB-231A B C

D E

G H

F

0

5,000

10,000

15,000

20,000

25,000

Cel

l are

a (s

quar

e pi

xel)

DMSO (control)

CRT0066101

*

HCC1954

0

5,000

10,000

15,000

20,000

25,000

30,000

Cel

l are

a (s

quar

e pi

xel)

DMSO (control)

CRT0066101

*

MDA-MB-231

Cel

ls

Δ no

rmal

ized

cel

l ind

ex

Time (h)

Time (h)

DMSOCRT0066101

DMSOCRT0066101 Figure 4.

The PKD inhibitor CRT0066101decreases the cell proliferation and theinvasive phenotype of ER� breast cancercells. A and D, indicated cell lines wereseeded in E-plates and (afterattachment) treated with 2.5 mmol/LCRT0066101 or DMSO (control).Numbers of attached cells werecontinuously monitored in real time forindicated times using an xCELLigenceRTCA DP instrument. Error bars (gray)represent three experiments. B, MDA-MB-231 cells were seeded in CIM-platesand treatedwith 2.5mmol/LCRT0066101orDMSO(control). After attachment, cellmigrationwas continuouslymonitored inreal time for indicated time using anxCELLigence RTCA DP instrument. Errorbars (gray) represent three experiments.C and F, indicated cell lines were seededon Matrigel-coated Transwell filters andtreated with 2.5 mmol/L CRT0066101 orDMSO (control). Transwell invasionassays were performed as described inMaterials and Methods. E, HCC1954 wereseeded on Transwell filters and treatedwith 2.5 mmol/L CRT0066101 or DMSO(control). Transwell migration assayswere performed as described inMaterials and Methods. G and H,indicated cell lines were treated with2.5 mmol/L CRT0066101 or DMSO(control) for 16 hours. Cells were fixedand stained with DAPI (blue) andphalloidin (red). Bar, 10 mm.Representative pictures fromeachgroupare depicted. The cell areawasmeasuredand analyzed using the ImageJ software.In A–H, the results presented are themean � SEM. P values were calculatedusing a two-tailed Student t test.� , P < 0.05, statistical significance.

Borges et al.

Mol Cancer Ther; 14(6) June 2015 Molecular Cancer TherapeuticsOF8

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 9: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

all three isoforms in a low nanomolar range (i.e., IC50 for PKD3is 2 nmol/L). In a previous work, it has been shown to be activein vivo in orthotopic animal models for pancreatic cancer andcolorectal cancer (33, 34). Because it can be orally administer-ed and has no side effects in mice, when used at doses thatinhibit PKD (33, 34), it is an inhibitor that could be developedfor clinical use.

Our data not only show that CRT0066101 can block all aspectsof the tumor phenotype in PKD1-negative/PKD3-positive breastcancer cells in vitro (Fig. 4), but also demonstrate in vivo relevanceby showing that CRT0066101 significantly inhibits primarytumor growth, local invasion, and metastasis to distant organsin vivo (Figs. 5 and 6). It is also important to note that the sameevents were obtained with specific knockdown of PKD3 showingthat this kinase is the main target in ER� cancer cells (Fig. 3).Although our in vitro data using shRNA or CRT0066101 clearlydemonstrate effects on cell proliferation,migration, and invasion,it is possible that additional tumorigenic functions are affected by

knockdown or inhibition of PKD3 in vivo. For example, PKDsignaling previously has been implicated in angiogenesis(38, 47, 48) and it is very possible that the decrease in size ofthe primary tumors that we observed in response to CRT0066101treatment is partly due to blocking of angiogenesis.

It should be noted that for breast cancers that undergo a switchfrom PKD1 to PKD3, two strategies are possible, either to reex-press PKD1, or to inhibit PKD3 (discussed in ref. 49). We recentlyhave tested the first strategy, and shown that reverting the epige-netic silencing of PKD1with theDNAmethyltransferase inhibitordecitabine can dramatically reduce the invasive and metastaticpotential of triple-negative orthotopic tumors in vivo (11, 19).What is still ill-defined is how PKD1, once it is reexpressed ininvasive breast cancers, can exert a protective effect and antagonizePKD3 functions. Because the ESR1 gene promoters also can besilenced by DNA methylation (50), the simplest explanation forthis may be that decitabine treatment also upregulates ER, whichthen decreases PKD3 expression.

Vehicle

A

B

D E

C

CRT0066101

*

0

40

80

120

Ki6

7+ c

ells

(rel

ativ

e pe

rcen

tage

)

Ki67

Vehi

cle

CR

T006

6101

Vehi

cle

CR

T006

6101

Cleaved caspase-3

0

100

200

300

400

500

600

700

800

900

*

Cle

aved

cas

pase

-3+

cells

(rel

ativ

e pe

rcen

tage

)

n = 6 n = 6

*

Tum

or v

olum

e (m

m3 )

*

Tum

or w

eigh

t(g

)

Vehicle CRT0066101Ve

hicl

eC

RT0

0661

01COX-2

0

60

120

180

*CO

X-2+

cel

ls

(rel

ativ

e pe

rcen

tage

)

1,500

1,000

500

0

2.5

2.0

1.5

1.0

0.5

0.0n = 6 n = 6

Figure 5.CRT0066101 inhibits tumor growth andlocal tumor cell invasion in vivo. A, MDA-MB-231.Luc cells were injected into themfp of female NOD scid mice. Afterestablishment of primary tumors, atweek 2, mice were treated orally with 80mg/kg CRT0066101 or vehicle everyother day for an additional period of 8weeks. Tumor growth was continuouslymonitored using the IVIS Spectrumimaging system. Left, a representativepicture of mice from each group (n ¼ 6per group) at week 7. At the endpoint(week 10), tumor volume and weightwere measured. � , P < 0.05, statisticalsignificance. B and C, primary tumors ofdifferent treatment groups were stainedby IHC for the expression of Ki67 (B) andcleaved caspase-3 (C). Bars, 50 mm. D,samples of primary tumor were stainedwith H&E. Bar, 200 mm. Areas wheretumors connect with mouse mammarygland tissue were enhanced. E, primarytumors were stained by IHC for theexpression of COX2. Bars, 50 mm. In B,C, and E, statistical analysis wasperformed using Aperio positive pixelcount algorithm in the Imagescopesoftware. P values were acquired withthe Student t test using Prism v5software. �, P < 0.05, statisticalsignificance. In B–E, representativepictures from each group are depicted.

Targeting of ER� Breast Cancer with CRT0066101

www.aacrjournals.org Mol Cancer Ther; 14(6) June 2015 OF9

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 10: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

In conclusion, our study provides a rationale that supports theuse of PKD inhibitors such as CRT0066101 for treatment ofpatients diagnosed with ER� or TNBC. Key for treatment withPKD inhibitors is a downregulation of PKD1 and upregulationof the oncogenic version PKD3 (or PKD2). This requires devel-oping reliable techniques that could be used in clinical settingsto determine PKD1, 2, and 3 expression status before treatmentdecisions are made.

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

Authors' ContributionsConception and design: S. Borges, E.A. Perez, E.A. Thompson, P. StorzAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): E.A. Perez, E.A. Thompson, X.J. GeigerAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. Borges, E.A. Perez, E.A. Thompson, D.C. Radisky,P. StorzWriting, review, and/or revision of the manuscript: S. Borges, E.A. Perez,D.C. Radisky, X.J. Geiger, P. Storz

Administrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): E.A. PerezStudy supervision: P. Storz

AcknowledgmentsThe authors thank members of the Storz laboratory for helpful discussions

and the Luther and Susie Harrison Foundation for their support.

Grant SupportThis work was supported by the NIH (GM086435 and CA140182) to

P. Storz, the Bankhead-Coley Program of the Florida Department of Health(1BG11) to P. Storz, the Donna Foundation (26.2 with Donna) to E.A. Perezand E.A. Thompson, and the Mayo Clinic Breast Cancer SPORE (CA116201)to P. Storz and D.C. Radisky.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received October 31, 2014; revised April 2, 2015; accepted April 2, 2015;published OnlineFirst April 7, 2015.

References1. Carey LA. Directed therapy of subtypes of triple-negative breast cancer.

Oncologist 2010;15(Suppl 5):49–56.2. Perou CM. Molecular stratification of triple-negative breast cancers.

Oncologist 2010;15(Suppl 5):39–48.3. Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis

of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative,and HER2-negative invasive breast cancer, the so-called triple-negativephenotype: a population-based study from the California cancer Registry.Cancer 2007;109:1721–8.

4. Rakha EA, El-Sayed ME, Green AR, Lee AH, Robertson JF, Ellis IO.Prognostic markers in triple-negative breast cancer. Cancer 2007;109:25–32.

5. Schneider BP, Winer EP, Foulkes WD, Garber J, Perou CM, Richardson A,et al. Triple-negative breast cancer: risk factors to potential targets. ClinCancer Res 2008;14:8010–8.

6. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al.Triple-negative breast cancer: clinical features and patterns of recurrence.Clin Cancer Res 2007;13:4429–34.

VehicleA B C

D E

CRT0066101

Mouse #1 Mouse #2

Mouse #1 Mouse #2

0

25

50

75

100

*

Ki6

7+ c

ells

(rel

ativ

e pe

rcen

tage

)

LungLymph node

Vehi

cle

CR

T006

6101

Staining: anti-human Vimentin

H&E Ki67

Vehi

cle

CR

T006

6101

n = 6 n = 6

12

8

4

0

*

Lung

met

asta

ses

(num

bers

per

mm

2 )

Lung

met

asta

ses

(ave

rage

siz

e in

μm

2 )

n = 6 n = 6

*

20,000

15,000

10,000

5,000

0

Figure 6.CRT0066101 inhibits metastasis todistant organs. A, metastasis wascontinuously monitored using the IVISSpectrum imaging system in vivo.Representative pictures of mice fromeach treatment group are depicted. B,the presence of metastasis in lymphnodes and lungswas analyzed by IHC forhuman vimentin. Bar, 500 mm for lymphnodes; 1 mm for lungs. C and D, thenumber of pulmonary metastases permm2 (C), or the average size ofpulmonary metastases in mm2 (D) wasquantified from five fields each lung.� , P < 0.05, statistical significance. E,lungs were stained with H&E andexpression of Ki67 was detected by IHC.The percentage of Ki67-positive cellswas determined using Aperio positivepixel count algorithm in the Imagescopesoftware. Bar, 50 mm. For B, D, and E,the values are mean � SEM. P valueswere acquired with the Student t testusing Prism v5 software. � , P < 0.05,statistical significance. In B and E,representative pictures from eachgroup are depicted.

Borges et al.

Mol Cancer Ther; 14(6) June 2015 Molecular Cancer TherapeuticsOF10

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 11: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

7. Irshad S, Ellis P, Tutt A. Molecular heterogeneity of triple-negative breastcancer and its clinical implications. Curr Opin Oncol 2011;23:566–77.

8. Karihtala P, Auvinen P, Kauppila S, Haapasaari KM, Jukkola-Vuorinen A,Soini Y. Vimentin, zeb1 and Sip1 are up-regulated in triple-negative andbasal-like breast cancers: associationwith an aggressive tumour phenotype.Breast Cancer Res Treat 2013;138:81–90.

9. Sarrio D, Rodriguez-Pinilla SM, Hardisson D, Cano A, Moreno-Bueno G,Palacios J. Epithelial–mesenchymal transition in breast cancer relates to thebasal-like phenotype. Cancer Res 2008;68:989–97.

10. Karam M, Legay C, Auclair C, Ricort JM. Protein kinase D1 stimulatesproliferation and enhances tumorigenesis of MCF-7 human breast cancercells through a MEK/ERK-dependent signaling pathway. Exp Cell Res2012;318:558–69.

11. Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z, Anastasiadis PZ, et al.Pharmacologic reversion of epigenetic silencing of the PRKD1 promoterblocks breast tumor cell invasion and metastasis. Breast Cancer Res2013;15:R66.

12. Eiseler T, Doppler H, Yan IK, Kitatani K, Mizuno K, Storz P. Protein kinaseD1 regulates cofilin-mediated F-actin reorganization and cell motilitythrough slingshot. Nat Cell Biol 2009;11:545–56.

13. Bastea LI, Doppler H, Balogun B, Storz P. Protein kinase D1 maintains theepithelial phenotype by inducing a DNA-bound, inactive SNAI1 transcrip-tional repressor complex. PLoS ONE 2012;7:e30459.

14. Du C, Zhang C, Hassan S, Biswas MH, Balaji KC. Protein kinase D1suppresses epithelial-to-mesenchymal transition through phosphoryla-tion of snail. Cancer Res 2010;70:7810–9.

15. Eiseler T, Kohler C, Nimmagadda SC, Jamali A, Funk N, Joodi G, et al.Protein kinase D1 mediates anchorage-dependent and -independentgrowth of tumor cells via the zinc finger transcription factor Snail1. J BiolChem 2012;287:32367–80.

16. Zheng H, Shen M, Zha YL, Li W, Wei Y, Blanco MA, et al. PKD1 phos-phorylation-dependent degradation of SNAIL by SCF-FBXO11 regulatesepithelial-mesenchymal transition and metastasis. Cancer Cell 2014;26:358–73.

17. Eiseler T, Doppler H, Yan IK, Goodison S, Storz P. Protein kinase D1regulates matrix metalloproteinase expression and inhibits breast cancercell invasion. Breast Cancer Res 2009;11:R13.

18. Peterburs P, Heering J, Link G, Pfizenmaier K, Olayioye MA, Hausser A.Protein kinase D regulates cell migration by direct phosphorylation of thecofilin phosphatase slingshot 1 like. Cancer Res 2009;69:5634–8.

19. Borges S, Doppler HR, Storz P. A combination treatment with DNAmethyltransferase inhibitors and suramin decreases invasiveness of breastcancer cells. Breast Cancer Res Treat 2014;144:79–91.

20. Hao Q, McKenzie R, Gan H, Tang H. Protein kinases D2 and D3 are novelgrowth regulators in HCC1806 triple-negative breast cancer cells. Antican-cer Res 2013;33:393–9.

21. Chen J, Lu L, Feng Y, Wang H, Dai L, Li Y, et al. PKD2mediates multi-drugresistance in breast cancer cells through modulation of P-glycoproteinexpression. Cancer Lett 2011;300:48–56.

22. Huck B, Duss S, Hausser A, Olayioye MA. Elevated protein kinase D3(PKD3) expression supports proliferation of triple-negative breast cancercells and contributes to mTORC1-S6K1 pathway activation. J Biol Chem2014;289:3138–47.

23. Chin YR, Toker A. Akt isoform-specific signaling in breast cancer: uncover-ing an anti-migratory role for palladin. Cell Adh Migr 2011;5:211–4.

24. Yoeli-Lerner M, Yiu GK, Rabinovitz I, Erhardt P, Jauliac S, Toker A. Aktblocks breast cancer cell motility and invasion through the transcriptionfactor NFAT. Mol Cell 2005;20:539–50.

25. Doppler H, Bastea LI, Borges S, Spratley SJ, Pearce SE, Storz P. Proteinkinase d isoforms differentially modulate cofilin-driven directed cellmigration. PLoS ONE 2014;9:e98090.

26. Liou GY, Doppler H, Necela B, Krishna M, Crawford HC, Raimondo M,et al. Macrophage-secreted cytokines drive pancreatic acinar-to-ductalmetaplasia through NF-kappaB and MMPs. J Cell Biol 2013;202:563–77.

27. Kupershmidt I, Su QJ, Grewal A, Sundaresh S, Halperin I, Flynn J, et al.Ontology-based meta-analysis of global collections of high-throughputpublic data. PLoS ONE 2010;5:e13066.

28. Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q, et al. Anonline survival analysis tool to rapidly assess the effect of 22,277 genes on

breast cancer prognosis using microarray data of 1,809 patients. BreastCancer Res Treat 2010;123:725–31.

29. Neve RM,Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al. A collection ofbreast cancer cell lines for the study of functionally distinct cancer subtypes.Cancer Cell 2006;10:515–27.

30. RingnerM, FredlundE,Hakkinen J, BorgA, Staaf J.GOBO: gene expression-based outcome for breast cancer online. PLoS ONE 2011;6:e17911.

31. Fawell SE, White R, Hoare S, SydenhamM, Page M, Parker MG. Inhibitionof estrogen receptor-DNA binding by the "pure" antiestrogen ICI 164,384appears to bemediated by impaired receptor dimerization. Proc Natl AcadSci U S A 1990;87:6883–7.

32. Kumar V, Chambon P. The estrogen receptor binds tightly to its re-sponsive element as a ligand-induced homodimer. Cell 1988;55:145–56.

33. Harikumar KB, Kunnumakkara AB, Ochi N, Tong Z, Deorukhkar A, SungB, et al. A novel small-molecule inhibitor of protein kinase D blockspancreatic cancer growth in vitro and in vivo. Mol Cancer Ther 2010;9:1136–46.

34. Wei N, Chu E, Wipf P, Schmitz JC. Protein kinase d as a potentialchemotherapeutic target for colorectal cancer. Mol Cancer Ther 2014;13:1130–41.

35. Minn AJ, Gupta GP, Siegel PM, Bos PD, Shu W, Giri DD, et al. Genes thatmediate breast cancer metastasis to lung. Nature 2005;436:518–24.

36. Kim M, Jang HR, Kim JH, Noh SM, Song KS, Cho JS, et al. Epigeneticinactivation of protein kinase D1 in gastric cancer and its role in gastriccancer cell migration and invasion. Carcinogenesis 2008;29:629–37.

37. Wille C, Kohler C, Armacki M, Jamali A, Gossele U, Pfizenmaier K, et al.Protein kinase D2 induces invasion of pancreatic cancer cells by regulatingmatrix metalloproteinases. Mol Biol Cell 2014;25:324–36.

38. Azoitei N, Pusapati GV, Kleger A, Moller P, Kufer R, Genze F, et al. Proteinkinase D2 is a crucial regulator of tumour cell-endothelial cell communi-cation in gastrointestinal tumours. Gut 2010;59:1316–30.

39. Chen J, Deng F, Singh SV, Wang QJ. Protein kinase D3 (PKD3) contributesto prostate cancer cell growth and survival through a PKCepsilon/PKD3pathway downstream of Akt and ERK 1/2. Cancer Res 2008;68:3844–53.

40. Zou Z, Zeng F, Xu W, Wang C, Ke Z, Wang QJ, et al. PKD2 and PKD3promote prostate cancer cell invasion by modulating NF-kappaB- andHDAC1-mediated expression and activation of uPA. J Cell Sci 2012;125:4800–11.

41. Jaggi M, Rao PS, Smith DJ, Hemstreet GP, Balaji KC. Protein kinase Cmu isdown-regulated in androgen-independent prostate cancer. Biochem Bio-phys Res Commun 2003;307:254–60.

42. Jaggi M, Rao PS, Smith DJ, Wheelock MJ, Johnson KR, Hemstreet GP, et al.E-cadherin phosphorylation by protein kinase D1/protein kinase C{mu}is associated with altered cellular aggregation and motility in prostatecancer. Cancer Res 2005;65:483–92.

43. Gevry N, Hardy S, Jacques PE, Laflamme L, Svotelis A, Robert F, et al.Histone H2A.Z is essential for estrogen receptor signaling. Genes Dev2009;23:1522–33.

44. Marques M, Laflamme L, Gaudreau L. Estrogen receptor alpha can selec-tively repress dioxin receptor-mediated gene expression by targeting DNAmethylation. Nucleic Acids Res 2013;41:8094–106.

45. Lavalle CR, Bravo-Altamirano K, Giridhar KV, Chen J, Sharlow E, Lazo JS,et al. Novel protein kinase D inhibitors cause potent arrest in prostatecancer cell growth and motility. BMC Chem Biol 2010;10:5.

46. Huck B, Kemkemer R, Franz-WachtelM,Macek B,Hausser A,OlayioyeMA.GIT1 phosphorylation on serine 46 by PKD3 regulates paxillin traffickingand cellular protrusive activity. J Biol Chem 2012;287:34604–13.

47. Ochi N, Tanasanvimon S, Matsuo Y, Tong Z, Sung B, Aggarwal BB, et al.Protein kinase D1 promotes anchorage-independent growth, invasion,and angiogenesis by human pancreatic cancer cells. J Cell Physiol2011;226:1074–81.

48. Rozengurt E. Protein kinase D signaling: multiple biological functions inhealth and disease. Physiology. (Bethesda) 2011;26:23–33.

49. Borges S, Storz P. Protein kinase D isoforms: new targets for therapy ininvasive breast cancers? Expert Rev Anticancer Ther 2013;13:895–8.

50. Ottaviano YL, Issa JP, Parl FF, Smith HS, Baylin SB, Davidson NE. Meth-ylation of the estrogen receptor gene CpG island marks loss of estrogenreceptor expression in human breast cancer cells. Cancer Res 1994;54:2552–5.

www.aacrjournals.org Mol Cancer Ther; 14(6) June 2015 OF11

Targeting of ER� Breast Cancer with CRT0066101

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945

Page 12: Effective Targeting of Estrogen Receptor Negative Breast ... · Small Molecule Therapeutics Effective Targeting of Estrogen Receptor– Negative Breast Cancers with the Protein Kinase

Published OnlineFirst April 7, 2015.Mol Cancer Ther   Sahra Borges, Edith A. Perez, E. Aubrey Thompson, et al.   Cancers with the Protein Kinase D Inhibitor CRT0066101

Negative Breast−Effective Targeting of Estrogen Receptor

  Updated version

  10.1158/1535-7163.MCT-14-0945doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2015/04/07/1535-7163.MCT-14-0945.DC1

Access the most recent supplemental material at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://mct.aacrjournals.org/content/early/2015/05/18/1535-7163.MCT-14-0945To request permission to re-use all or part of this article, use this link

Research. on January 13, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst April 7, 2015; DOI: 10.1158/1535-7163.MCT-14-0945