il15ra drives antagonistic mechanisms of cancer ......tumor and stem cell biology il15ra drives...

14
Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched Triple-Negative Breast Cancers Pierfrancesco Marra 1 , Sumi Mathew 1 , Anita Grigoriadis 1 , Yin Wu 2 , Fernanda Kyle-Cezar 2 , Johnathan Watkins 1 , Mamunur Rashid 3 , Emanuele De Rinaldis 4 , Sonya Hessey 1 , Patrycja Gazinska 1 , Adrian Hayday 2 , and Andrew Tutt 1 Abstract Despite its aggressive nature, triple-negative breast cancer (TNBC) often exhibits leucocyte inltrations that correlate with favorable prognosis. In this study, we offer an explanation for this apparent conundrum by dening TNBC cell subsets that overexpress the IL15 immune receptor IL15RA. This receptor usually forms a heterotrimer with the IL2 receptors IL2RB and IL2RG, which regulates the proliferation and differentiation of cytotoxic T cells and NK cells. However, unlike IL15RA, the IL2RB and IL2RG receptors are not upregulated in basal-like TNBC breast cancer cells that express IL15RA. Mechanistic investigations indicated that IL15RA signaling activated JAK1, STAT1, STAT2, AKT, PRAS40, and ERK1/2 in the absence of IL2RB and IL2RG, whereas neither STAT5 nor JAK2 were activated. RNAi-mediated attenuation of IL15RA established its role in cell growth, apoptosis, and migration, whereas expression of the IL15 cytokine in IL15RA-expressing cells stimulated an autocrine signaling cascade that promoted cell proliferation and migration and blocked apoptosis. Notably, coexpression of IL15RA and IL15 was also sufcient to activate peripheral blood mononuclear cells upon coculture in a paracrine signaling manner. Overall, our ndings offer a mechanistic explanation for the paradoxical association of some high-grade breast tumors with better survival outcomes, due to engagement of the immune stroma. Cancer Res; 74(17); 490821. Ó2014 AACR. Introduction Tumor inltration by a variety of immune cells, includ- ing cytotoxic T cells, regulatory T cells, natural killer (NK) cells, neutrophils, and macrophages, is a common feature of many cancers (1). Although tumor inltration by cyto- toxic lymphocytes is generally correlated with a favorable outcome, substantial evidence exists that other types of leukocytes can instead promote tumorigenesis by supplying cytokines (such as IL6, TNF, and IL1b) that stimulate tumor proliferation, tissue invasion, and/or angiogenesis (2). Tumor cells interact with the immune system and have evolved strategies to neutralize immune attack through stim- ulating immune checkpoint receptors CTLA4 and PD-1, down- regulating MHC class I molecules, interfering with the per- forin/granzyme cytotoxicity functions of T cells or TGFb- mediated inactivation of T-cell responses (3). Immune-cell inltration in breast cancer is, in general, associated with a favorable prognosis (4). However, the mechanisms that underlie this observation and their vari- ation among breast cancers, are poorly understood. Breast cancer is recognized as being a heterogeneous disease in both its malignant and stromal cell compartments, with immune-cell inltration being particularly associated with some subgroups. On the basis of gene-expression proles, breast cancer can be divided into subgroups commonly referred to as intrinsic subtypes that include an HER2-positive, a basal and two luminal subtypes (5). Recent evidence (6) has revealed an even greater complexity within breast cancer but has consistently identied a transcriptome belonging to a "basal" group that overlaps substantially, although not entirely, with that of immunohistochemically dened estrogen (ER), progesterone (PR), and HER2 receptornegative breast cancers, referred to as triple-negative breast cancer (TNBC). In ER-positive breast cancers, expression of proliferative biomarkers is largely associated with a poor prognosis (7). This is not the case in 1 Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital, King's College London School of Medicine, London, United Kingdom. 2 Peter Gorer Department of Immunobiology, King's College of London, London, United Kingdom. 3 Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom. 4 NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College of London, London, United Kingdom. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: A. Tutt, King's College of London, Guy's Hospital, London SE1 9RT, UK. Phone: 44-0-207-188-9881; Fax: 44-0-207-188- 3666; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-14-0637 Ó2014 American Association for Cancer Research. Cancer Research Cancer Res; 74(17) September 1, 2014 4908 Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association for https://bloodcancerdiscov.aacrjournals.org Downloaded from

Upload: others

Post on 16-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

Tumor and Stem Cell Biology

IL15RA Drives Antagonistic Mechanisms of CancerDevelopment and Immune Control in Lymphocyte-EnrichedTriple-Negative Breast Cancers

Pierfrancesco Marra1, Sumi Mathew1, Anita Grigoriadis1, Yin Wu2, Fernanda Kyle-Cezar2,Johnathan Watkins1, Mamunur Rashid3, Emanuele De Rinaldis4, Sonya Hessey1,Patrycja Gazinska1, Adrian Hayday2, and Andrew Tutt1

AbstractDespite its aggressive nature, triple-negative breast cancer (TNBC) often exhibits leucocyte infiltrations that

correlate with favorable prognosis. In this study, we offer an explanation for this apparent conundrum bydefining TNBC cell subsets that overexpress the IL15 immune receptor IL15RA. This receptor usually forms aheterotrimer with the IL2 receptors IL2RB and IL2RG, which regulates the proliferation and differentiationof cytotoxic T cells and NK cells. However, unlike IL15RA, the IL2RB and IL2RG receptors are not upregulatedin basal-like TNBC breast cancer cells that express IL15RA. Mechanistic investigations indicated that IL15RAsignaling activated JAK1, STAT1, STAT2, AKT, PRAS40, and ERK1/2 in the absence of IL2RB and IL2RG,whereas neither STAT5 nor JAK2 were activated. RNAi-mediated attenuation of IL15RA established its role incell growth, apoptosis, and migration, whereas expression of the IL15 cytokine in IL15RA-expressing cellsstimulated an autocrine signaling cascade that promoted cell proliferation and migration and blockedapoptosis. Notably, coexpression of IL15RA and IL15 was also sufficient to activate peripheral bloodmononuclear cells upon coculture in a paracrine signaling manner. Overall, our findings offer a mechanisticexplanation for the paradoxical association of some high-grade breast tumors with better survival outcomes,due to engagement of the immune stroma. Cancer Res; 74(17); 4908–21. �2014 AACR.

IntroductionTumor infiltration by a variety of immune cells, includ-

ing cytotoxic T cells, regulatory T cells, natural killer (NK)cells, neutrophils, and macrophages, is a common featureof many cancers (1). Although tumor infiltration by cyto-toxic lymphocytes is generally correlated with a favorableoutcome, substantial evidence exists that other typesof leukocytes can instead promote tumorigenesis bysupplying cytokines (such as IL6, TNF, and IL1b) thatstimulate tumor proliferation, tissue invasion, and/orangiogenesis (2).

Tumor cells interact with the immune system and haveevolved strategies to neutralize immune attack through stim-ulating immune checkpoint receptors CTLA4 and PD-1, down-regulating MHC class I molecules, interfering with the per-forin/granzyme cytotoxicity functions of T cells or TGFb-mediated inactivation of T-cell responses (3).

Immune-cell infiltration in breast cancer is, in general,associated with a favorable prognosis (4). However, themechanisms that underlie this observation and their vari-ation among breast cancers, are poorly understood. Breastcancer is recognized as being a heterogeneous disease inboth its malignant and stromal cell compartments, withimmune-cell infiltration being particularly associated withsome subgroups.

On the basis of gene-expression profiles, breast cancer canbe divided into subgroups commonly referred to as intrinsicsubtypes that include an HER2-positive, a basal and twoluminal subtypes (5). Recent evidence (6) has revealed an evengreater complexity within breast cancer but has consistentlyidentified a transcriptome belonging to a "basal" group thatoverlaps substantially, although not entirely, with that ofimmunohistochemically defined estrogen (ER), progesterone(PR), and HER2 receptor–negative breast cancers, referred toas triple-negative breast cancer (TNBC). In ER-positive breastcancers, expression of proliferative biomarkers is largelyassociated with a poor prognosis (7). This is not the case in

1Breakthrough Breast Cancer Research Unit, Department of ResearchOncology, Guy's Hospital, King's College London School of Medicine,London, United Kingdom. 2Peter Gorer Department of Immunobiology,King's College of London, London, United Kingdom. 3Wellcome TrustSanger Institute, Wellcome Trust Genome Campus, Cambridge, UnitedKingdom. 4NIHR Biomedical Research Centre, Guy's and St Thomas'NHS Foundation Trust and King's College of London, London, UnitedKingdom.

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

Corresponding Author:A. Tutt, King's College of London, Guy's Hospital,London SE1 9RT, UK. Phone: 44-0-207-188-9881; Fax: 44-0-207-188-3666; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-14-0637

�2014 American Association for Cancer Research.

CancerResearch

Cancer Res; 74(17) September 1, 20144908

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 2: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

ER-negative breast cancers, which are more uniformly highlyproliferative and in which immune response signatures havebeen shown to have a higher prognostic significance (8). Evenamong basal tumors there is still significant histologic, immu-nohistologic, and prognostic heterogeneity (9). Indeed, somemorphologically recognized groups show excellent long-termprognosis despite a basal expression profile and aggressivehistopathologic features. One such example is medullarybreast carcinoma, which shares many features with theTNBC/basal subtype, including similarity in the gene-expres-sion profile, highly proliferative phenotype, and TP53 muta-tions (10). Medullary carcinoma also shows numerous lym-phoid cells in the stroma (11) and expression of an immuneprofile, including TH1-associated cytokine receptors such asIL15RA, their associated cytokines, including IL15, and genesinvolved in cytotoxicity (12).IL15RA usually exists in a heterotrimeric receptor with

IL2RB and IL2RG, which also contribute to the trimeric IL2receptor (13). IL15RA is able to bind IL15 with high affinity inthe absence of the beta and gamma subunits. This allows thereceptor to function in cis, i.e., the three subunits being on thesame cell, or in trans, with the IL15RA subunit bound to IL15 onone cell and the beta and gamma subunits on an adjacent cell.In the latter case, the IL15/IL15RA complex is transpresentedto the beta and gamma subunit with consequent activation ofintracellular signaling (14).IL15-dependent signaling has been implicated in the mod-

ulation of adaptive immune responses (15), the activationand maintenance of distinct lymphocyte populations (16) andthe inhibition of apoptosis in multiple systems (17). Further-more, IL15-dependent signaling has been shown to play a rolein hematologic malignancies, in which there is evidence thatan IL15/IL15RA autocrine growth-stimulation loop plays arole in the progression of human T-cell lymphotrophic virus1 (HTLV-1)–associated adult T-cell leukemia/lymphoma (14).There are several reports of IL15 and/or IL15 receptor

expression in solid tumor–derived cell lines or tissues. Afunctional heterotrimeric IL15 receptor has been described inrenal cancer (18) and IL15RA expression, has been found inmelanoma models (19).In the present study, we find IL15RA to be frequently

amplified and describe elevated expression of both IL15RAand its ligand IL15, in the absence of IL2RB and ILRG, inbasal and immune-modulatory forms (20) of TNBC and inbreast cancer cell lines (BCCL). We investigate the conse-quences of this coordinated expression, describing a tumor-promoting IL15 and IL15RA-dependent autocrine signalingmechanism that drives cell growth, colony formation andcell migration, and protects from apoptosis. We also showthat IL15/IL15RA expression on the plasma membrane ofBCCLs drives signaling in peripheral blood mononuclearcells (PBMC) making BCCLs visible to the immune systemand potentially leading to an antitumor immune response.In doing so, we provide a potential mechanistic explanationfor the paradox in which some basal breast cancers show ahigh mitotic index, aggressive morphologic appearances andyet engage an effective immune-cell response with an asso-ciated impact on prognosis.

Materials and MethodsCell lines

BCCLs were obtained from the ATCC, HMEC from LifeTechnologies, and Natural Killer YT cells from Dr. S. John(KCL, London, UK). Growth conditions were as recommendedby the suppliers. Cells were authenticated by short tandemrepeat analysis and matched to the German Collection ofMicroorganisms and Cell Cultures (DSMZ)—database.

DNA copy-number and gene-expression analysisAbsolute DNA copy number was obtained for 172 cancers in

our TNBC-enriched Guy's Hospital breast-cancer cohort (21),using Affymetrix SNP6.0 genome profiles and human genomebuild hg18 mapping. The Tumor Aberration Prediction Suitealgorithm (22) was applied to minimize influence of normaltissue. IL15RA and IL15 levels were extracted from the Guy'sHospital (21), METABRIC (23), TCGA (The Cancer GenomeAtlas; ref. 6) 579 TNBC cohorts (TNBC579_GSE31519; //;ref. 24), as well as two BCCL datasets (25, 26). PAM50 classi-fication was as described for the Guy's (27), METABRIC (23),and TCGA cohorts (6). TNBC subtypes were established usingthe online TNBC-type program (28). IL15RA and IL15 expres-sion was compared using Pearson correlation. All analyseswere performed with R software (www.r-project.org/).

Quantitative PCR analysesMethodology for quantitative PCR (qPCR) is described in

Supplementary Methods.

Western blottingWestern blots were processed as described in Supplemen-

tary Methods.

Immunofluorescence and confocal microscopyCells were processed as described in Supplementary Meth-

ods. Confocal images were acquired by an A1R Si Confocalsystem (Nikon Corp.) equipped with a �100 objective. Opticalsections were taken at 1 Airy U at 1-mm intervals, and thenreconstructed in a Z-stack. Images assembled in Adobe Photo-shop CS5.1.

ImmunohistochemistryFormalin-fixed paraffin-embedded (FFPE) sections 3-mm-

thick were processed as described in Supplementary Methodsthen stainedwith theMAB147 IL15RAantibody (R&DSystems)at 10 ng/mL for 2 hours. Images from digitalized scans of theglass slide specimens were obtained at magnification �20(0.45 mm/pixel resolution) using a Hamamatsu Nanozoomer2.0 HT.

XenotransplantsUM13,MC1, and UM2 patient-derived xenotransplants were

established from primary breast tumors at the University ofMichigan (Ann Arbor, MI) as described by Cariati and collea-gues (29). Once established in the mouse mammary fat pad,xenotransplants were characterized by immunohistochemis-try (IHC): UM13 was ER-positive and -negative for PR, HER2,

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4909

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 3: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4910

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 4: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

EGFR, and cytokeratin 5/6.MC1 andUM2were ER-, PR-, HER2-negative but EGFR- and CK5/6–positive.

Small interfering RNA–mediated silencingHuman Silencer Select small interfering RNAs (siRNA),

including a nontargeting negative control siRNA, were pur-chased from Ambion (Life Technologies) as detailed in Sup-plementary Methods and transfected at 50 nmol/L by RNAi-Max (Life Technologies). Knockdown efficacy was assessed72 hours posttransfection.

Small hairpin RNA–mediated gene silencingMISSION small hairpin RNA (shRNA) Library–derived

clones (Sigma-Aldrich) were used as detailed in SupplementaryMethods. Lentiviral particles were produced and titrated asdescribed in Supplementary Methods.

Cell population growth assaysCells were seeded at 8� 103 per well in a 96-well plate. Effect

on cell growth was assessed using Hoechst 33342 nucleic acidstain (Life Technologies) at the times indicated after seeding,typically 24 hours (T0), 48 hours (T1), 72 hours (T2), and96 hours (T3), using the FLUOstar Omega plate reader (BMGLABTECH). Hoechst 33342 readings were standardized againstT0 to give a relative growth value.

Caspase activation analysesCaspase-3/7 activity was measured at 24 hours after seeding

or as indicated, using the caspase-Glo-3/7 assay (Promega).

Colony formation assaysA total of 1 � 103 or 2 � 103 cells per well were plated in

6-well plates and cultured in their ideal media for 15 daysbefore staining with 0.5% crystal violet in 1% formaldehydefor 20 minutes. Cells were then washed with PBS and imaged.

Stimulation of cells with recombinant IL15 and analysisof changes in protein expression and phosphorylationCells were grown to 50% confluence then serum

starved in normal growth medium for 3 hours before

addition of rIL15 (Peprotech) at the indicated concentra-tions for 15 minutes at 37�C. The proteins werethen harvested by scraping the cells in Laemmli bufferwith 10% b-mercaptoethanol, and separated by SDS–PAGE.

Phospho-kinase arrayThe Human Phospho-Kinase Array (R&D Systems) was

used according to the manufacturer's instructions. Briefly, aprotein lysate was obtained from approximately 5 � 105

control or IL15-stimulated cells in the specific lysis bufferedprovided. Lysates were then incubated with the filtersprovided and phosphorylation levels of the individual sitesin the array revealed by chemoluminescence on a radio-graphic film, and quantified by densitometry using ImageJsoftware.

Data and statistical analysisData and statistical analyses have been performed using

Microsoft Excel and GraphPad Prism software.

Cell migration assaysCell migration was assessed using Transwell inserts (96-well

microplates, 8-mm pore; Corning-Costar) coated with bovinecollagen type I (Invitrogen) and seeded with 25 � 103 cellsresuspended in 90 mL RPMI 0.1% FBS. The microplate insertswere placed on the receiver plate containing medium supple-mented with 2% FBS and incubated for 4 hours or overnight at37�C. Migrated cells were detached from the inserts by trypsinand counted using a plate reader after permeabilization with0.01% Triton X-100 and DNA labeling by SYTOX Green(Invitrogen).

Cell adhesion assaysA total of 10 � 103 cells were seeded on a thin layer of

collagen type I (Invitrogen) and incubated at 37�C for 60minutes. Plates were then washed three times with PBS andthe remaining attached cells were then permeabilized with0.01% Triton X-100, DNA labeled by SYTOX Green (Invitrogen)and counted using a plate reader.

Figure 1. Copy number, mRNA, and protein expression of IL15RA in breast cancer and cell models. A, chromosome 10 frequency plot of amplificationand deletion across 111 TNBCs and 61 non-TNBCs profiled with Affymetrix SNP6.0 microarray. The percentage of tumors in which each segment isamplified (defined as more than 4 copies) is plotted in green for TNBCs and yellow for non-TNBCs, whereas deletions are shown in red and purplefor TNBCs or non-TNBCs, respectively. The location of IL15RA is indicated by a black line. B, IL15RA gene expression across breast carcinomasof the Guy's TNBC–enriched BC, METABRIC and TCGA cohorts is shown. The classification of the three cohorts into molecularly defined breastcancer subtypes was based on their previously published PAM50 centroid correlation [Basal in red, HER2 in pink, Luminal A (LumA) in dark blue,Luminal B (LumB) in light blue, and normal-like (NL) in green]. Mean-centered expression values (log2 scale) are shown on the y-axis. C, IL15RA geneexpression across Guy's TNBCs and the 579 TNBCs (GSE31519) is shown using Vanderbilt TNBC subtype classification. [TNBC subtypes arerepresented pink for basal-like 1 (BL1), blue basal-like 2 (BL2), orange immunomodulatory (IM), grey mesenchymal (M), magenta mesenchymalstem-like (MSL), green luminal androgen receptor (LAR)]. D, relative expression of IL15RA, IL2RB, and IL2RG across 51 basal-like and luminal-likeBCCLs, each dot represents a cell line. E, FACS analysis of IL15RA, IL2RB, and IL2RG surface expression on HCC38, HCC1143, and MDA-MB-231(basal-like, high IL15RA, red) or BT549, HCC1428, and SKBR3 (luminal-like, low IL15RA, green), and HMEC (nonmalignant, IL15RA-low) with YTNK cells, CD56þ PBMC and lymphocytes (IL15RA, IL2RB, and IL2RG) as positive controls. F, total protein lysates from nonmalignant HMEC, luminal(green) or basal (red) BCCLs, and YT cells (positive control), were probed with anti-IL15RA, IL2RB, and IL2RG antibodies and GAPDH as aloading control. G, 3D reconstruction of serial confocal z-axis sections from a nonpermeabilized HCC1143 cell stained for IL15RA (red) and DNA(Hoechst 33342, blue). A top view and two orthogonal views along the yellow bars are shown. White arrows indicate IL15RA staining thatpredominantly decorates the contour of the cells. H, IHC images of HCC1143 cells either untreated (left) or knocked down for IL15RA (right) bytransfection of IL15R siRNA#0 as described in Materials and Methods, and stained with the MAB147 monoclonal antibody against IL15RA(R&D Systems). I, IHC images of the MC1 and UM2 (ER-, PR-, and HER2-negative, CK5/6 and EGFRþ) and UM13 (ERþ, PR� and HER2�, and K5/6 andEGFR�) PDX models and normal breast tissue, stained with the MAB147 antibody against IL15RA.

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4911

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 5: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4912

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 6: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

Flow-cytometry analysisCultured cells were detached with Accutase (PAA Labora-

tories) and washed twice with PBS before staining. PBMCwere isolated by Ficoll gradient centrifugation of whole bloodfromhealthy volunteers andwashed twice with PBS. Cells werefirst stained with LIVE/DEAD Aqua (Molecular Probes) for 20minutes at room temperature, then washed twice with FACSbuffer (PBS, 2% FCS, 2 mmol/L EDTA) before staining in 1:50anti-IL15Ra PE (BioLegend clone JM7A4), anti-IL2Rb APC(BioLegend clone TU27), and anti-IL2Rg Brilliant Violet 421(BD Biosciences clone AG184) or paired isotype controls inFACS buffer for 20 minutes at 4�C. Cells were washed twicewith FACS buffer before fixing in CellFIX (BD Biosciences) andacquisition on a BD FACS Canto II.

ELISAA total of 2 � 106 cells were seeded in 1 mL of culture

medium in 6-well plates and incubated overnight. Superna-tants were harvested and centrifuged to remove debris. Cellswere detached with Accutase (PAA Laboratories) and countedbefore lysis in 300mL of lysis buffer (25mmol/L Tris-HCl pH7.4,150mmol/LNaCl, 1%Triton X-100, and 5%glycerol). Cellswereincubated on ice with intermittent vortexing every 10 minutesfor 60 minutes. Lysates and supernatants were assayed forIL15 by sandwich ELISA (R&D Systems Human IL15 DuoSet)as per themanufacturer's instructions. IL15 concentration wasnormalized to 106 cells.

PBMC and breast cancer cell coculturePBMC were taken from healthy adult donors after the

informed consent approved by the Institutional Review Board.Mononuclear cells were obtained by Ficoll gradient centrifu-gation (GE Healthcare). A total of 5 � 104 breast cancer cellswere plated in a 96-well plate and incubated overnight. Thefollowing day, the same number of PBMC was added. After 30minutes, PBMC were harvested and stained with pSTAT5antibody (BD Biosciences) and analyzed by flow cytometry.When indicated, a blocking anti-IL15 monoclonal mouse anti-body (MAB2471; R&D Systems) was added at 10 mg/mL toBCCLs 30 minutes before coculture with PBMC.

ResultsAmplification and overexpression of IL15RA in subsets ofTNBCs and BCCLsSeveral studies (23, 6) have shown that the most distal

region on chromosome 10p is frequently amplified in TNBCs.

Investigating the copy number of chromosome 10 in ourGuy's TNBC–enriched BC cohort (27), using AffymetrixSNP6.0-derived genomic profiles, we confirmed the presenceof an amplicon encompassing approximately 10 Mbp of themost distal region of the short arm of chromosome 10(10:82,824-8,738,800; hg18). Within this amplicon, a numberof genes had an absolute copy number of greater than 4 in atleast 30% of TNBC cases (Fig. 1A; Supplementary Table S1).Analysis of these genes revealed some with features sugges-tive of a possible role as cancer drivers. Among these, IL15RAattracted our interest as it encodes the alpha subunit of theIL15 receptor known to be involved in the signaling cascadecontrolling proliferation and activation of immune cells.IL15RA showed an absolute copy number of greater than 4in 43% (48/111) of TNBC in contrast with 18% (11/61) of non-TNBC (Fisher exact test; P < 0.001). To investigate whetherthe abundance of IL15RA expression is enhanced speci-fically in TNBCs, its expression was extracted from thetranscriptomes of the Guy's TNBC–enriched BC collection,the invasive ductal breast carcinomas in METABRIC (23), andthe TCGA breast cancer cohorts (Fig. 1B; ref. 6). Using thePAM50 breast cancer subtype classification published foreach cohort, increased IL15RA expression was consistentlyobserved in basal and in normal-like breast cancer subtypes.Given the heterogeneity within TNBCs, we investigatedIL15RA expression across previously defined TNBC subtypes(20) in the Guy's cohort and a publicly available dataset of579 TNBCs (GSE31519; Fig. 1C; ref. 24). Higher IL15RAexpression was observed in the immunomodulatory subtype,characterized by expression of genes involved in severalimmune-cell signaling pathways that also significantly over-lap with gene signatures for medullary breast cancers (20).

To identify cellular models, we analyzed expression ofcomponents of the IL15 receptor in 51 BCCLs (26) findingsignificantly higher expression of IL15RA, but not IL2RB orIL2RG, in "basal" versus "luminal" BCCLs (t test, P < 0.001; Fig.1D). To confirm localization and presence of IL15RA andabsence of IL2RB and IL2RG on basal-like BCCLs, FACSanalyses were performed using antibodies against cell sur-face–exposed epitopes on nonpermeabilized cells. Plasmamembrane IL15 receptor subunit levels were consistent withexpression data. Only IL15RA was detected on the plasmamembrane of the basal-like BCCLs HCC1143, HCC38, andMDA-MB-231, with all components being absent on the lumi-nal-like BCCLs BT549, HCC1428, and SKBR3, despite all com-ponents being detected on positive control YT, NK, or PBMC

Figure 2. IL15RA silencing impairs cell growth and colony formation and increases apoptosis in BCCLs expressing high levels of IL15RA. Analysis of cellpopulation growth of MDA-MB-231 (high IL15RA expressing; A) and SKBR3 (low IL15RA expressing; B) cells transduced with control shRNA or shRNAstargeting IL15RA. C,MDA-MB-231 colony formation assay onplastic. Cells were either untreated (control) or transducedwith lentivirus driving expression of anonspecific shRNA (ns), or shRNAs targeting IL15RA (IL15R sh1, sh2, and sh3), then incubated for 15 days before being fixed/stained and imaged. D,quantification of shRNA-mediated IL15RA knockdown in MDA-MB-231 cells by qPCR (top) and immunoblotting (bottom). Analysis of cell populationgrowth of MDA-MB-231 (E), HCC1143 (F), SKBR3 (G), and BT549 (H) cells transfected with control siRNA (nontargeting) or siRNAs targeting IL15RA (IL15RsiRNA#0, IL15 siRNA#2). MDA-MB-231 and HCC1143 cells express high levels of IL15RA and BT549 and SKBR3, low levels. I, quantification of siRNA-mediated IL15RA knockdown in MDA-MB-231 cells by qPCR (top) and immunoblotting (bottom). J, caspase-3/7 activation was assessed in eitheruntreated MDA-MB-231 and HCC1143 cells or cells transduced with a control shRNA (nontargeting) or shRNAs targeting IL15RA, 24 hours after seedingthe cells for the cell population growth assay, equivalent to 7 days posttransduction. K, the effect of IL15 on caspase-3/7 activation was assessed inHCC1143 cells. Cells were serum starved in the presence of increasing amounts of IL15 as indicated and incubated for 24 hours before assessing caspaseactivity. Data representative of at least two independent experiments, values are average � SD; sample size, 3; �, P < 0.05; ��, P < 0.01; ���, P < 0.001.

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4913

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 7: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

(Fig. 1E). Immunoblotting of whole-cell lysates from BCCLsand nonmalignant HMECS confirmed that IL15RA could bedetected and was enriched in basal-like BCCLs, whereas nei-ther IL2RB nor IL15RA could be detected except in the YT cells(Fig. 1F).

Plasma membrane localization of IL15RA was furtherconfirmed by both confocal microscopy (Fig. 1G) and IHCin sections of FFPE-embedded HCC1143 cells, in whichplasma membrane staining was shown to be specific by itssubstantial diminution by siRNA-mediated IL15RA silencing(Fig. 1H). To examine IL15RA localization in relation to theplasma membrane in patient-derived xenotransplant (PDX)tissue in which we had demonstrated presence or absence ofIL15RA expression on immunoblot (data not shown), weexamined sections of these PDXs stained using the sameIL15RA antibodies. This again showed a plasma membraneexpression pattern in basal-like PDXs, which was almostabsent in a non–basal-like ER-positive PDX and normalbreast tissue (Fig. 1I).

Silencing of IL15RA results in a growth defect in BCCLsHaving confirmed surface expression of an apparently

isolated IL15RA receptor component in basal-like BCCLs,we then examined its function. We silenced IL15RA by RNAiin cell lines expressing either high or low levels of IL15RA.The IL15RA shRNAs induced significant impairment ofgrowth in the high IL15RA–expressing MDA-MB-231 butnot in the low-expressing SKBR3 cell line (Fig. 2A and B).We also analyzed the effect of IL15RA knockdown onadherent cell colony formation. After a 15-day incubation,MDA-MB-231 cells knocked down for IL15RA showed asignificantly reduced number of colonies compared withcontrol cells (Fig. 2C and D). To test a broader range ofcells and an orthogonal RNAi methodology, we transfectedsiRNAs targeting IL15RA in MDA-MB-231 and HCC1143cells and confirmed impaired cell population growth(Fig. 2E and F). No effect was observed in the lowIL15RA–expressing SKBR3 and BT549 cell lines (Fig. 2G,H, and I).

IL15 and IL15RA have antiapoptotic effects in BCCLsIL15 signaling can protect against lethal apoptosis (17).

We asked whether the reduction in cell population growthand colony formation, after IL15RA silencing might be inpart a consequence of apoptosis. We observed substantialactivation of caspase-3/7 upon IL15RA knockdown in MDA-MB-231 and HCC1143 cells (Fig. 2J). Addition of recombi-nant IL15 to HCC1143 cells cultured in 1% rather than 10%FBS resulted in dose-dependent inhibition of serum-depri-vation induced caspase-3/7 activation (Fig. 2K), suggestingthat IL15, via IL15RA, inhibits apoptosis.

IL2RB and IL2RG silencing does not affect BCCLsThe absence of detectable beta and gamma subunits of

the heterotrimeric IL15 receptor in BCCLs suggests thatthe alpha subunit alone might be sufficient to drive thedescribed effects on cell population growth. To furtherexclude the possibility that the IL2RB or IL2RG subunits

contribute to the influence of IL15RA on BCCL growth, wesilenced IL2RB and IL2RG. This failed to show any signif-icant effect in the growth rate of HCC1143 cells but induceda significant defect in IL2-stimulated proliferation of con-trol YT cells, which express the full heterotrimeric receptor(Fig. 3).

An autocrine IL15/IL15RA signal drives cell populationgrowth in BCCLs

Previous work has shown that IL15 and IL15RA bind to eachother along the secretory pathway, remaining in a complexupon arrival at the plasmamembrane (30). Indeed an autocrineIL15/IL15RA–mediated signaling mechanism occurs in T-celllymphoma (14). We, therefore, investigated whether TNBCsand BCCLs express IL15, how its expression is correlated withIL15RA expression and to what extent this is required forIL15RA-mediated growth.

Within the 579 TNBCs cohort (GSE31519; ref. 24), wefound a positive correlation between the mRNA levels ofIL15RA and IL15 (Fig. 4A; correlation, 0.51; P ¼ 2.2e�16).Similarly, in some cell lines, the expression of these twogenes was correlated (Fig. 4B; correlation, 0.36; P ¼ 0.007;ref. 26) and on average showed a higher expression of theligand and receptor in cells of basal-like rather than luminal-like subtypes (Fig. 4B and C).

Because expression of IL15 mRNA does not always cor-relate with the protein (14), we assessed IL15 protein levelsin BCCLs by ELISA and found higher levels in basal-like celllines that expressed higher levels of IL15RA (Fig. 4D). Thisled us to hypothesize that cytokine and receptor expressedby the same cells might signal using an autocrine mecha-nism promoting proliferation of BCCLs. To test this, weknocked down IL15 expression using shRNAs and observed asignificant impairment of growth in MDA-MB-231 andHCC1143 (high IL15RA and IL15) but not in SKBR3 cells(low IL15RA and IL15; Fig. 4E–G). We examined the effect ofknockdown of endogenous IL15 on levels of spontaneousapoptosis in HCC1143 cells and showed significant caspaseactivation (Fig. 4H).

IL15- and IL15RA-dependent intracellular signaling inBCCLs

Having observed a reduction in growth and an increasein apoptosis upon IL15RA knockdown in basal BCCLs,we sought to identify the mechanisms downstream ofIL15RA controlling these functions. In T cells, the IL15heterotrimeric receptor signals through JAK1, JAK3, andSTAT3 or STAT5, thereby regulating cell proliferationand inhibiting apoptosis (31). We analyzed the activityof a range of intracellular signaling kinases in HCC1143cells using a validated phospho-antibody array. The stim-ulation of HCC1143 cells with recombinant IL15 signi-ficantly increased phosphorylation of ERK1/2, STAT1,STAT2, AKT, and PRAS40 (Fig. 5A and B). ERK1/2 andSTAT1 phosphorylation were independently validatedin lysates from IL15-stimulated HCC1143 cells using adifferent set of phospho-protein antibodies and Westernblots (Fig. 5C).

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4914

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 8: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

It has previously been shown that the IL15 receptor signalsthrough JAK1 and JAK3 inT lymphocytes (31), although inmastcells, IL15 activates JAK2 and STAT6 through a poorly defined60 to 65 kD IL15 receptor subunit (IL15RX; ref. 32).We assessedchanges in JAK1 and JAK2 phosphorylation upon IL15 stim-ulation. JAK1 but not JAK2 phosphorylation was induced byIL15 stimulation in HCC1143, MDA-MB-231 and HCC1954, butnot in SKBR3, suggesting a dependency upon IL15RA (Fig. 5D).We also observed no change in STAT3, STAT5, and STAT6

phosphorylation, which are known to be downstream of theheterotrimeric receptor. This findingwas validated byWesternblotting HCC1143 cell lysates using independent antibodies(Fig. 5E).

To confirm IL15RA-dependent activation of JAK1 andERK1/2, we knocked down the expression of IL15RA usingsiRNAs in HCC1954 cells, then stimulated with recombinantIL15 and observed substantial reduction in the levels ofIL15-stimulated phospho-JAK1 and ERK1/2 (Fig. 5F).

Figure 3. IL2RB and IL2RGknockdown does not affect growthof BCCLs. Cell population growthassay of HCC1143 cells (A and B)and IL2-stimulated growth of YTNK cells (C and D), followingsiRNA-mediated knockdown ofIL2RB (A and C) and IL2RG (B andD). YT NK cells were grown in theabsence or presence of 20 ng/mLIL2 and the raw data obtained fromthe cells grown without IL2 weresubtracted from those ofstimulated cells. The difference isreferred to as IL2-dependentgrowth and shown in the graphs (CandD). IL2 stimulation of HCC1143did not produce any increase inthe cell population growth rate(data not shown). The efficacy ofsiRNAs targeting, IL2RB (E), andIL2RG (F) was assessed in YTcells by qPCR. Data representativeof at least two independentexperiments; values are average �SD; sample size, 3; �, P < 0.05;��, P < 0.01; ���, P < 0.001.

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4915

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 9: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

IL15RA regulates cell adhesion and motility viaJAK1/STAT1 independent of its effect on ERK1/2

IL15 signaling has been implicated in T-cell adhesion andmotility (14). We hypothesized that interfering with IL15/IL15RA function might impair cell adhesion and/or motilityin BCCLs. We tested MDA-MB-231 and HCC1954 cells in a

standard Transwell 2D-motility assay, performed on a thinlayer of collagen type I. IL15RA knockdown induced a 35%to 50% reduction in the number of cells migrating (Fig. 6A).We then investigated whether the migration defect observedwas due, at least in part, to a defect in adhesion andconfirmed a defect in adhesion of IL15RA-silenced MDA-

Figure 4. IL15 is expressed byTNBCs and basal BCCLs and itssilencing specifically impairsgrowth in high IL15RA–expressingcell lines. Correlation of gene-expression levels between IL15RAand IL15 across A, 579 TNBC(GSE31519) and B, 51 BCCLs. ThePearson r values and P values forsignificance are shown. C, relativegene expression of IL15 in 51BCCLdichotomized into the basal-like (triangles) and luminal-like(dots) phenotypes, whereby eachsymbol represents a cell line. D,ELISA analysis of IL15 levels in asubset of basal or luminal BCCLs.HaCat cells were included as apositive control. IL15 levels wereassessed in whole cells (lysate). Eto G, cell population growth assaysof MDA-MB-231 (E), HCC1143 (F),(high levels of both IL15 andIL15RA), and SKBR3 (G) cells (lowlevels of both IL15 and IL15RA)following IL15 knockdown by twodifferent shRNA species (IL15 sh26and IL15 sh27). H, caspase-3/7activation was assessed inHCC1143 cells transduced with acontrol shRNA (nontargeting) orshRNAs targeting IL15, 24 hoursafter seeding the cells for the cellpopulation growth assay,equivalent to 7 daysposttransduction. I, quantificationof shRNA-mediated IL15knockdown in MDA-MB-231 cellsby qPCR. Data representative ofat least two independentexperiments; values areaverage � SD; sample size,3; �, P < 0.05; ��, P < 0.01;���, P < 0.001.

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4916

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 10: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

MB-231 cells to the collagen layer (Fig. 6B). Nevertheless, thesmall degree by which adhesion was reduced suggests thatthis does not fully account for the reduction in migrationand that other processes in cell migration are affected byIL15 signaling in these cells.As the JAK1/STAT1 pathway plays a role in cell motility

(33), we investigated the possible influence of the previ-ously observed IL15-mediated JAK1/STAT1 activation onBCCL motility by silencing JAK1 expression. We observed asignificant reduction in HCC1954 cell migration (Fig. 6C)but no concomitant reduction in cell population growth(Fig. 6D and E). In parallel, we observed abrogation ofIL15-dependent STAT1 phosphorylation, whereas no sub-

stantial change occurred in the phosphorylation of ERK1/2(Fig. 6F).

These findings suggest a split signaling cascade downstreamof IL15RA in BCCLs, in which IL15-dependent activation ofJAK1 leads to phosphorylation of STAT1 and subsequentregulation of cell motility. At the same time ERK1/2 activationoccurs by a JAK1/STAT1–independentmechanism and ismostlikely responsible for promoting cell proliferation.

The presence of an IL15RA/IL15 complex on the surfaceof BCCLs stimulates a signaling response in immune cells

Given the established role of IL15 and IL15RA as promotersof T-cell recruitment and activation (14), we asked whether the

Figure 5. IL15 signals throughIL15RA to promotephosphorylation of ERK1/2, JAK1,STAT1, STAT2, AKT, and PRAS40in BCCLs. A, HCC1143 cells (highIL15RA), were stimulated with 100ng/mL of IL15 for 15 minutes, thenlysed and protein phosphorylationassessed using a human phospho-protein array. Phospho-proteinpatterns were consistent in twoindependent experiments; for a fulldescription of the phospho-antibodies included in the arrayplease refer to SupplementaryFig. S1. B, IL15 induced increasedERK1/2, AKT, STAT2, PRAS40,and STAT1 phosphorylation asassessed by densitometry in onerepresentative experiment; errorbars, � SD of the densitometryreadings from the experimentalreplicates. C, phospho-proteinarray results were validated byWestern blotting using a differentset of phospho-antibodies and awider set of BCCLs: either highIL15RA, expressing such asHCC1954 and HCC1143, or lowIL15RA, expressing such asSKBR3. D,Western blot analysis ofJAK1 and JAK2 phosphorylation inresponse to IL15 stimulation inHCC1954, HCC1143, and MDA-MB-231 (high IL15RA) or SKBR3(low IL15RA) cell lines. E, Westernblot analysis of STAT3, STAT5, andSTAT6 phosphorylation inresponse to IL15 stimulation inHCC1143 cells. F, Western blotanalysis of JAK1 and ERK1/2phosphorylation in response toIL15 stimulation following siRNA-mediated IL15RA knockdown(IL15R siRNA #0, #1, and #2) inHCC1954 cells; nontargetingsiRNAs used as negative control.

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4917

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 11: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

levels of IL15 being expressed by basal-like BCCLs, if secretedor exposed by IL15RA on the cell surface were sufficient toactivate a response in PBMC. BCCLs expressing differentlevels of IL15RA/IL15 were cocultured with PBMC and thephosphorylation of STAT5 was measured by flow cytometry inthe gated PBMC population. We observed increased STAT5phosphorylation following exposure of PBMC to HCC1143and MDA-MB-231 but not to IL15/IL15RA low-expressingSKBR3 and T47D. The concomitant addition of a blocking

anti-IL15 antibody or the silencing of IL15 or IL15RA throughshRNAs, resulted in a significant reduction of STAT5 phos-phorylation in PBMC. This indicates a specific role for IL15/IL15RA produced by the cancer cell leading to STAT5 phos-phorylation in the PBMC (Fig. 7). Furthermore, conditionedmedium from MDA-MB-231 cells did not induce STAT5phosphorylation in PMBC, suggesting transpresentation, asopposed to secreted IL15, is required for PBMCactivation (datanot shown).

Figure 6. IL15RA knockdownimpairs motility of basal BCCLs bya JAK1/STAT1–dependent, ERK1/2–independent mechanism. A,MDA-MB-231 and HCC1954 cellmigration was investigated using astandard Transwell assay in cellstransfected with a control siRNA(nontargeting) and siRNAstargeting IL15RA (IL15R siRNA#0,IL15R siRNA#2). B, MDA-MB-231cells were seeded on a thin layer ofcollagen type I and adhesionassessed following transfectionwith a control siRNA (nontargeting)and siRNAs targeting IL15RA(IL15R siRNA#0, IL15R siRNA#2).C, Western blot analysis showingefficacy of JAK1 knockdown 72hours posttransfection. D, MDA-MB-231 and HCC1954 cellmigration was investigated using astandard Transwell assay in cellstransfected with a control siRNA(nontargeting) and siRNAstargeting JAK1 (JAK1 siRNA#47,JAK1 siRNA48). E, HCC1954 cellpopulation growthwas assessed incells transfected with a controlsiRNA (nontargeting) and siRNAstargeting JAK1 (JAK1 siRNA#47,JAK1 siRNA48). F, the impact ofJAK1 knockdown on IL15-inducedphosphorylation of STAT1 andERK1/2 was assessed in HCC1954cells by Western blot analysis. G,quantitation by densitometry of theIL15-induced phosphorylation ofSTAT1 as showed in F. H,quantitation by densitometry of theIL15-induced phosphorylation ofERK1/2 as showed in F. Data arerepresentative of at least twoindependent experiments; valuesare average � SD; sample size, 3;�, P < 0.05; ��, P < 0.01;���, P < 0.001.

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4918

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 12: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

DiscussionAlthough expression of IL15RA occurs in several cell types,

expression of a functional heterotrimeric IL15 receptor com-prising IL2RB and IL2RG ismostly restricted to immune cells: Tlymphocytes, B lymphoblasts, and NK cells (34). This is con-sistent with our finding of undetectable levels of the beta andgamma subunits by flow-cytometry orWestern blot analysis inBCCLs. Our demonstration of the high frequency of amplifi-cation of IL15RA and of expression IL15RA protein on thesurface of subgroups of TNBCs and BCCLs suggests that thiscomponent of the receptor may drive malignant phenotypes,despite isolation from other components of the classicalreceptor, in a significant proportion of TNBCs. As expected,we did not find a strong correlation between IL15RA copynumber and its expression in tumors (data not shown) becauseof the confounding effect of infiltration of leucocytes thatusually express this receptor. IL15RA knockdown experimentsusing multiple RNAi methodologies confirmed a role forIL15RA in the regulation of cell growth, migration, and apo-ptosis in basal BCCLs that express it at higher levels.A positive correlation between IL15 and IL15RA transcript,

in both tumors and BCCLs, prompted us to investigate andconfirm by ELISA, that IL15 protein was expressed in basalBCCLs that also express IL15RA. We then confirmed thatIL15, known to be capable of binding to IL15RA intracellularlyfor cotransport to the plasma membrane (30), was also re-quired for growth, suggesting an IL15/IL15RA autocrine–driv-en malignant phenotype in BCCLs.

To our knowledge, this is the first report of such a role forthe IL15/IL15RA complex in the absence of a full heterotri-meric receptor, although an IL15/IL15RA-driven malignantphenotype has been suggested in myeloma and T-cell lym-phoma (which also express IL2RB and IL2RG; refs. 35, 36) andmelanoma (in which the expression of IL2RB and IL2RG wasneither verified nor disproved; ref. 19).

We then aimed to characterize IL15-dependent signalingin TNBCs. In lymphocytes, the signaling cascade originatingfrom the IL15 heterotrimeric receptor involves activation ofJAK1/3 and STAT3/5 (31). Further data suggested IL15-dependent activation of the PI3K/AKT pathway (37) andthe NFkB pathway in neutrophils (38) or myeloid cells (39).IL15RA also interacts with and signals through SYK inneutrophils (40). Evidence obtained in epithelial cells sug-gested a signaling function for IL15/IL15RA in the absence ofIL2RB (41), whereas in melanoma, IL15RA can bind andsignal through TRAF (19).

Upon stimulation of BCCLs with recombinant IL15, weobserved activation of JAK1, STAT1, and STAT2, as well asERK1/2 and AKT and PRAS40. Intriguingly, we did not observeactivation of other known effectors such as STAT3, STAT5, orSTAT6. We speculate that this is a consequence of the lack ofIL2RB and IL2RG that have been reported to specificallymediate the activation of many of these effectors (31).

Our observation of ERK1/2 activation is consistent withan IL15RA-dependent stimulation of proliferation in TNBCs.The lack of any evidence of activation of MEK might beexplained by our observation of IL15-dependent activationof AKT/PRAS40, which may lead to MEK-independentERK1/2 activation, as has been suggested in melanoma(42). Interestingly AKT phosphorylates PRAS40 specificallyat T246 (43), the same residue that we found phosphorylatedin response to IL15 stimulation. AKT activation can inhibitapoptosis (44), and we speculate that this mechanism maybe responsible for the inhibition of apoptosis observed inBCCLs upon IL15 stimulation.

JAK1/STAT1 signaling is known to play a role in cell motility(45, 33). This is consistent with our observation of impairedmigration of IL15RA/IL15–expressing cells upon JAK1 knock-down in the absence of any significant reduction in growthrate. We also found JAK1 silencing inhibits STAT1 activationbut not ERK activation consistent with lack of effect of JAK1silencing on cell growth.

It is increasingly apparent that the interface betweenimmune cells and tumor cells can induce powerful effectson prognosis. Several reports have indicated positive prog-nostic correlations with evidence of effective engagement ofboth humoral and cytotoxic T-cell–adaptive immuneresponses (8), whereas others have shown that expressionof inflammatory cytokines is associated with a poor prog-nosis (1).

There is growing evidence, suggesting inflammatory cyto-kines can regulate the growth of breast cancer cells. Acooperative role for IL6 and IL8 in promoting TNBC growthhas recently been suggested, and although the expressionof IL15RA and IL15 in TNBCs and TNBC-derived cell lineswas also noted, it was not further investigated (46). This,

Figure 7. IL15RA/IL15–expressingBCCLs elicit STAT5phosphorylation inPBMC upon cocultivation. Flow-cytometry analysis of STAT5phosphorylation in PBMC cocultured 1:1 with BCCLs or stimulated withrecombinant IL15 (rIL15; 100 pg/mL) for 30 minutes. IL15-blockingantibody (aIL15) effects were assessed by adding 30 minutes beforecoculture. The effect of IL15RA knockdown was also assessedfollowing transduction and selection of MDA-MB-231 cells with shRNAstargeting IL15RA (IL15R sh3) or IL15 (IL15 sh26 and IL15 sh27) as well asa control shRNA (nontargeting). Data are representative of at least twoindependent experiments; values are average � SD; sample size, 3;�, P < 0.05; ��, P < 0.01; ���, P < 0.001.

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4919

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 13: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

together with evidence of IL6 stimulation of IL15 expressionobserved in HaCaT cells (47), suggests cooperation amongproinflammatory cytokines to promote cancer cell growthdespite known roles in stimulation of the immune response.

We show that the endogenous expression of IL15RA andIL15 by basal BCCLs stimulates a pSTAT5 signaling responsein freshly isolated PBMC in coculture. This phenomenon isabolished by an inhibitory IL15 antibody as well as by silencingof either IL15RA or IL15. Taken together, this evidence pointsto PBMC activation by IL15 expressed by basal BCCLs co-expressing IL15RA. This most likely occurs via transpresenta-tion of IL15 by IL15RA, as knockdown of IL15RA inhibitedpSTAT5 signaling in PBMC. IL15 is known to be a potentinducer of the activatory receptor NK group 2D (NKG2D)receptor on cytotoxic lymphocytes and is an important surv-ival factor for NK cells (48, 49). Indeed NKG2D-mediatedantitumor immunity is enhanced by IL15 (50).

Our data suggest a dual effect of IL15RA amplification andits consequent high-level expression. IL15RA coexpressed withIL15 in the absence of IL2RB and IL2RG, on the one hand drivescancer proliferation, protection from apoptosis, and enhancescell migration, whereas on the other hand is capable ofactivating signaling in immune cells that, when not edited,suppressed or compromised by other intrinsic tumor or stro-mal factors, may lead to a relatively better prognosis.

A number of clinical trials (NCT01727076 andNCT01885897) are currently investigating the effects of IL15administration with an aim to stimulate anticancer immuneresponses. Our data add further insight into the complexitiesof IL15 signaling in some breast tumors, not only in immunecells in the tumor stroma, but also within malignant cellsin which IL15 and IL15RA may also drive hallmarks ofmalignancy. This may explain why some basal breast car-cinomas with aggressive high-grade malignant features arealso intensely infiltrated by immune cells and why thesehigh-grade cancers have a paradoxically good prognosiscompared with other basal breast cancers (11). If an IL15transpresentation-induced activation of immune cells isblocked or edited by immune checkpoints at the immuneand cancer cell interface but cancer cell–intrinsic IL15/IL15RA drivers of the malignant phenotype remain intact,we speculate there may be adverse effects on prognosis. Ourdata indicating IL15-dependent cancer cell signaling suggestthat IL15 therapies should be investigated cautiously in

IL15RA-expressing cancers as there may be the potentialto drive tumor growth

A more detailed understanding of the signaling pathwayinvolved in IL15/IL15RA signaling in cancer cells, and theanticancer immunologic mechanisms that can be enhancedby IL15, would aid identification of cancer cell–intrinsic targetsdownstream of IL15/IL15RA that might be inhibited to blockprotumorigenic effects while retaining the antitumor immu-nostimulatory effects of an IL15-based therapy.

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

DisclaimerThe views expressed are those of the author(s) and not necessarily those of

the NHS, the NIHR or the Department of Health.

Authors' ContributionsConception and design: P. Marra, A. Grigoriadis, A. Hayday, A. TuttDevelopment of methodology: P. Marra, A. Grigoriadis, F. Kyle-Cezar,M. Rashid, A. Hayday, A. TuttAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): P. Marra, S. Mathew, A. Grigoriadis, Y. Wu, S. Hessey,P. GazinskaAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): P. Marra, S. Mathew, A. Grigoriadis,Y. Wu, F. Kyle-Cezar, J. Watkins, M. Rashid, E. De Rinaldis, P. Gazinska,A. Hayday, A. TuttWriting, review, and/or revision of themanuscript: P.Marra, A. Grigoriadis,Y. Wu, F. Kyle-Cezar, A. Hayday, A. TuttAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): S. Mathew, P. GazinskaStudy supervision: P. Marra, A. Tutt

AcknowledgmentsThe authors thank Dr. Susan John (Peter Gorer Department of Immunobiol-

ogy King's College of London) for providing us with her expertise and reagentsand Sorapun Vorapon for help with proof editing.

Grant SupportThis research was supported by the Breakthrough Breast Cancer Research

Unit funding at King's College London and by the National Institute for HealthResearch Biomedical Research Centre based at Guy's and St Thomas' NHSFoundation Trust and King's College London. Patient tissue samples wereprovided by Guy's and St Thomas' Breast Tissue and Data Bank, which issupported by the Department of Health via the National Institute for HealthResearch comprehensive Biomedical Research Centre award.

The costs of publication of this article were defrayed in part 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 March 4, 2014; revised May 16, 2014; accepted June 5, 2014;published OnlineFirst June 30, 2014.

References1. Galon J, Angell HK, Bedognetti D, Marincola FM. The continuum of

cancer immunosurveillance: prognostic, predictive, and mechanisticsignatures. Immunity 2013;39:11–26.

2. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflamma-tion. Nature 2008;454:436–44.

3. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integratingimmunity's roles in cancer suppression and promotion. Science2011;331:1565–70.

4. Denkert C, Loibl S, Noske A, Roller M, Muller BM, Komor M, et al.Tumor-associated lymphocytes as an independent predictor ofresponse to neoadjuvant chemotherapy in breast cancer. J Clin Oncol2010;28:105–13.

5. PerouCM,Sorlie T, EisenMB, vandeRijnM, JeffreySS, ReesCA, et al.Molecular portraits of human breast tumours. Nature 2000;406:747–52.

6. Comprehensive molecular portraits of human breast tumours. Nature2012;490:61–70.

7. Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer:classification, prognostication, and prediction. Lancet 378:1812–23.

8. Irshad S, Grigoriadis A, Lawler K, Ng T, Tutt A. Profiling the immunestromal interface in breast cancer and its potential for clinical impact.Breast Care 2012;7:273–80.

9. Rakha EA, Ellis IO, Reis-Filho JS. Immunohistochemical heterogeneityof breast carcinomas negative for estrogen receptors, progesterone

Marra et al.

Cancer Res; 74(17) September 1, 2014 Cancer Research4920

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 14: IL15RA Drives Antagonistic Mechanisms of Cancer ......Tumor and Stem Cell Biology IL15RA Drives Antagonistic Mechanisms of Cancer Development and Immune Control in Lymphocyte-Enriched

receptors and Her2/neu (basal-like breast carcinomas). Mod Pathol2008;21:1060–1.

10. Sabatier R, Finetti P, Cervera N, Lambaudie E, Esterni B, Mamessier E,et al. A gene expression signature identifies two prognostic subgroupsof basal breast cancer. Breast Cancer Res Treat 2011;126:407–20.

11. Rakha EA, Aleskandarany M, El-Sayed ME, Blamey RW, Elston CW,Ellis IO, et al. The prognostic significance of inflammation and med-ullary histological type in invasive carcinoma of the breast. Eur JCancer 2009;45:1780–7.

12. Bertucci F, Finetti P, Cervera N, Charafe-Jauffret E, Mamessier E,Adelaide J, et al. Gene expression profiling shows medullary breastcancer is a subgroup of basal breast cancers. Cancer Res 2006;66:4636–44.

13. Waldmann T. The contrasting roles of IL2 and IL15 in the life and deathof lymphocytes: implications for the immunotherapy of rheumatolog-ical diseases. Arthritis Res 2002;4:S161–7.

14. Budagian V, Bulanova E, Paus R, Bulfone-Paus S. IL15/IL15 receptorbiology: a guided tour through an expanding universe. CytokineGrowth Factor Rev 2006;17:259–80.

15. Lodolce J, Burkett P, Koka R, Boone D, Chien M, Chan F, et al.Interleukin-15 and the regulation of lymphoid homeostasis. Mol Immu-nol 2002;39:537–44.

16. Becknell B, Caligiuri MA. Interleukin-2, interleukin-15, and their roles inhuman natural killer cells. Adv Immunol 2005;86:209–39.

17. Bulfone-Paus S, Ungureanu D, Pohl T, Lindner G, Paus R, Ruckert R,et al. Interleukin-15 protects from lethal apoptosis in vivo. Nat Med1997;3:1124–8.

18. Tejman-Yarden N, Zlotnik M, Lewis E, Etzion O, Chaimovitz C, Douv-devani A. Renal cells express a functional interleukin-15 receptor.Nephrol, Dial, Transplant 2005;20:516–23.

19. Pereno R, Giron-Michel J, Gaggero A, Cazes E, Meazza R, Monetti M,et al. IL15/IL15Ralpha intracellular trafficking in humanmelanoma cellsand signal transduction through the IL15Ralpha. Oncogene 2000;19:5153–62.

20. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, ShyrY, et al. Identification of human triple-negative breast cancer subtypesand preclinical models for selection of targeted therapies. J ClinInvestigation 2011;121:2750–67.

21. de Rinaldis E, Gazinska P, Mera A, Modrusan Z, Fedorowicz GM,Burford B, et al. Integrated genomic analysis of triple-negative breastcancers reveals novel microRNAs associated with clinical and molec-ular phenotypes and sheds light on the pathways they control. BMCGenomics 2013;14:643.

22. Rasmussen M, Sundstrom M, Goransson Kultima H, Botling J, MickeP, Birgisson H, et al. Allele-specific copy number analysis of tumorsamples with aneuploidy and tumor heterogeneity. GenomeBiol 2011;12:R108.

23. Curtis C, Shah SP, Chin S-F, Turashvili G, Rueda OM, Dunning MJ,et al. The genomic and transcriptomic architecture of 2,000 breasttumours reveals novel subgroups. Nature 2012;486:346–52.

24. Karn T, Pusztai L, Holtrich U, Iwamoto T, Shiang CY, Schmidt M, et al.Homogeneous datasets of triple negative breast cancers enable theidentification of novel prognostic and predictive signatures. PloS ONE2011;6:e28403.

25. Grigoriadis A, Mackay A, Noel E, Wu PJ, Natrajan R, Frankum J, et al.Molecular characterisation of cell linemodels for triple-negative breastcancers. BMC Genomics 2012;13:619.

26. Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al. Acollectionof breast cancer cell lines for the studyof functionally distinctcancer subtypes. Cancer cell 2006;10:515–27.

27. GazinskaP,Grigoriadis A, Brown JP,Millis RR,Mera A, Gillett CE, et al.Comparison of basal-like triple-negative breast cancer defined bymorphology, immunohistochemistry and transcriptional profiles. ModPathol 2013;26:955–66.

28. Chen X, Fau - Li J, Li J, Fau - Gray WH, Gray WH, Fau - Lehmann BD,et al. TNBCtype: a subtyping tool for triple-negative breast cancer.

29. Cariati M, Marlow R, Dontu G. Xenotransplantation of breast cancers.Methods in Mol Biology 2011;731:471–82.

30. Duitman EH, Orinska Z, Bulanova E, Paus R, Bulfone-Paus S. How acytokine is chaperoned through the secretory pathway by complexing

with its own receptor: lessons from interleukin-15 (IL15)/IL15 receptoralpha. Mol Cell Biol 2008;28:4851–61.

31. Johnston JA, BaconCM, FinbloomDS,ReesRC, KaplanD, ShibuyaK,et al. Tyrosine phosphorylation and activation of STAT5, STAT3, andJanus kinases by interleukins 2 and 15. Proc Natl Acad Sci U S A1995;92:8705–9.

32. MasudaA,Matsuguchi T, Yamaki K, Hayakawa T, KuboM, LaRochelleWJ, et al. Interleukin-15 induces rapid tyrosine phosphorylation ofSTAT6 and the expression of interleukin-4 in mouse mast cells. J BiolChem 2000;275:29331–7.

33. Malilas W, Koh SS, Kim S, Srisuttee R, Cho IR, Moon J, et al. Cancerupregulated gene 2, a novel oncogene, enhances migration and drugresistance of colon cancer cells via STAT1 activation. Int J Oncol2013;43:1111–6.

34. Su AI, Wiltshire T, Batalov S, Lapp H, Ching KA, Block D, et al. A geneatlas of the mouse and human protein-encoding transcriptomes. ProcNatl Acad Sci U S A 2004;101:6062–7.

35. Tinhofer I, Marschitz I, Henn T, Egle A, Greil R. Expression of functionalinterleukin-15 receptor and autocrine production of interleukin-15 asmechanisms of tumor propagation in multiple myeloma. Blood 2000;95:610–8.

36. Dobbeling U, Dummer R, Laine E, Potoczna N, Qin JZ, Burg G.Interleukin-15 is an autocrine/paracrine viability factor for cutaneousT-cell lymphoma cells. Blood 1998;92:252–8.

37. Lai YG, HouMS, Lo A, Huang ST, Huang YW, Yang-Yen HF, et al. IL15modulates the balance between Bcl-2 and Bim via a Jak3/1-PI3K-Akt-ERK pathway to promote CD8alphaalphaþ intestinal intraepitheliallymphocyte survival. Eur J Immunol 2013;43:2305–16.

38. McDonald PP, Russo MP, Ferrini S, Cassatella MA. Interleukin-15(IL15) induces NF-kappaB activation and IL8 production in humanneutrophils. Blood 1998;92:4828–35.

39. ChenowethMJ,MianMF, Barra NG, Alain T, SonenbergN, Bramson J,et al. IL15 can signal via IL15Ralpha, JNK, and NF-kappaB to driveRANTES production by myeloid cells. J Immunol 2012;188:4149–57.

40. Ratthe C, Girard D. Interleukin-15 enhances human neutrophil phago-cytosis by a Syk-dependent mechanism: importance of the IL15Ral-pha chain. J leukocyte Biol 2004;76:162–8.

41. Stevens AC, Matthews J, Andres P, Baffis V, Zheng XX, Chae DW,et al. Interleukin-15 signals T84 colonic epithelial cells in theabsence of the interleukin-2 receptor beta-chain. The Am J Physiol1997;272:G1201–8.

42. Aksamitiene E, Kholodenko BN, Kolch W, Hoek JB, Kiyatkin A. PI3K/Akt-sensitive MEK-independent compensatory circuit of ERK activa-tion in ER-positive PI3K-mutant T47D breast cancer cells. Cell Signal2010;22:1369–78.

43. HaarEV, LeeS-i, Bandhakavi S,GriffinTJ,KimD-H. Insulin signalling tomTOR mediated by the Akt/PKB substrate PRAS40. Nat Cell Biol2007;9:316–23.

44. Morgensztern D, McLeod HL. PI3K/Akt/mTOR pathway as a target forcancer therapy. Anticancer Drugs 2005;16:797–803.

45. Andl CD, Mizushima T, Oyama K, Bowser M, Nakagawa H, Rustgi AK.EGFR-induced cell migration is mediated predominantly by the JAK-STAT pathway in primary esophageal keratinocytes. Am J PhysiolGastrointest Liver Physiol 2004;287:G1227–37.

46. Hartman ZC, Poage GM, den Hollander P, Tsimelzon A, Hill J, Panu-pinthuN, et al. Growth of triple-negative breast cancer cells relies uponcoordinate autocrine expression of the proinflammatory cytokines IL6and IL8. Cancer Res 2013;73:3470–80.

47. Tao QS, Huang HL, Chai Y, Luo X, Zhang XL, Jia B, et al. Interleukin-6upregulates the expression of interleukin-15 is associatedwithMAPKsand PI3-K signaling pathways in the human keratinocyte cell line,HaCaT. Mol Biol Reports 2012;39:4201–5.

48. Roberts AI, Lee L, Schwarz E, Groh V, Spies T, Ebert EC, et al. NKG2Dreceptors induced by IL15 costimulate CD28-negative effector CTL inthe tissue microenvironment. J Immunol 2001;167:5527–30.

49. Ma A, Koka R, Burkett P. Diverse functions of IL2, IL15, and IL7 inlymphoid homeostasis. Ann Rev Immunol 2006;24:657–79.

50. Sutherland CL, Rabinovich B, Chalupny NJ, Brawand P, Miller R,Cosman D. ULBPs, human ligands of the NKG2D receptor, stimulatetumor immunity with enhancement by IL15. Blood 2006;108:1313–9.

www.aacrjournals.org Cancer Res; 74(17) September 1, 2014 4921

IL15RA Drives Malignancy and Immune Cell Activation in TNBC

Cancer Research. by guest on September 2, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from