+ All Categories
Home > Documents > M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor...

M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor...

Date post: 01-Aug-2020
Category:
Upload: others
View: 36 times
Download: 0 times
Share this document with a friend
9
Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment Eva Van Overmeire 1,2 , Beno ^ t Stijlemans 1,2 , Felix Heymann 3 , Jiri Keirsse 1,2 ,Yannick Morias 1,2 , Yvon Elkrim 1,2 , Lea Brys 1,2 , Chlo e Abels 1,2 , Qods Lahmar 1,2 , Can Ergen 3 , Lars Vereecke 4,5 , Frank Tacke 3 , Patrick De Baetselier 1,2 , Jo A. Van Ginderachter 1,2 , and Damya Laoui 1,2 Abstract Tumors contain a heterogeneous myeloid fraction comprised of discrete MHC-II hi and MHC-II lo tumor-associated macrophage (TAM) subpopulations that originate from Ly6C hi monocytes. However, the mechanisms regulating the abundance and pheno- type of distinct TAM subsets remain unknown. Here, we inves- tigated the role of macrophage colony-stimulating factor (M-CSF) in TAM differentiation and polarization in different mouse tumor models. We demonstrate that treatment of tumor-bearing mice with a blocking anti-M-CSFR monoclonal antibody resulted in a reduction of mature TAMs due to impaired recruitment, extrav- asation, proliferation, and maturation of their Ly6C hi monocytic precursors. M-CSFR signaling blockade shifted the MHC-II lo / MHC-II hi TAM balance in favor of the latter as observed by the preferential differentiation of Ly6C hi monocytes into MHC-II hi TAMs. In addition, the genetic and functional signatures of MHC-II lo TAMs were downregulated upon M-CSFR blockade, indicating that M-CSFR signaling shapes the MHC-II lo TAM phenotype. Conversely, granulocyte macrophage (GM)-CSFR had no effect on the mononuclear tumor inltrate or relative abundance of TAM subsets. However, GM-CSFR signaling played an important role in ne-tuning the MHC-II hi pheno- type. Overall, our data uncover the multifaceted and opposing roles of M-CSFR and GM-CSFR signaling in governing the phenotype of macrophage subsets in tumors, and provide new insight into the mechanism of action underlying M-CSFR blockade. Cancer Res; 76(1); 3542. Ó2015 AACR. Introduction Tumors are often highly inltrated with inammatory cells such as tumor-associated macrophages (TAM). TAMs are now considered to promote tumor growth in multiple ways, including induction of angiogenesis, remodeling of the extracellular matrix, stimulation of cancer cell proliferation and metastasis, and inhi- bition of adaptive immunity (1), the clinical signicance of which is evidenced by the correlation between high TAM density and worse outcome in most cancer types (2). Ly6C hi monocytes differentiate into distinct TAM subsets with specialized functions and a different intratumoral localization: normoxic M1-like MHC-II hi TAM and hypoxic M2-like MHC-II lo TAM (3, 4). As hypoxia is not the main driver of TAM inltration and differentiation (4), we considered myelopoietic growth factor receptor signaling as governor of TAM heterogeneity. In this respect, M-CSFdriven macrophage activation leads to the expres- sion of a substantial part of the M2 transcriptome, while granu- locyte macrophage colony-stimulating factor (GM-CSF) induces a M1 phenotype (5, 6). As such, blocking M-CSFR (CSF1R, CD115) signaling in myometrial macrophages stimulated the occurrence of an M1-like MHC-II hi population at the expense of M2-like MHC-II lo macrophages in the pregnant mouse uterus (7). M-CSFR signaling was recently also shown to be critical for TAM differentiation and shaping the M1/M2 TAM phenotype (811). However, which aspects of the monocyte-to-TAM differen- tiation pathway are inuenced by M-CSFR and whether GM-CSF also skews the TAM phenotype remain largely unexplored (12). Using mouse tumors of different histologic origin, we show here, that Ly6C hi monocytes depend on M-CSFR signaling for their recruitment and extravasation to tumors and differentiation to mature TAM. M-CSFR signaling governs the phenotype of M2- like MHC-II lo TAM, and its blockade resulted in a preferential differentiation of monocytes to M1-like MHC-II hi TAM. Although GM-CSF receptor (GM-CSFR, CSF-2R, or CD116) signaling is not determining monocyte recruitment and differentiation, it ne tunes the molecular prole of MHC-II hi TAM, hence uncovering 1 Laboratory of Myeloid Cell Immunology,VIB, Brussels, Belgium. 2 Lab- oratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium. 3 Department of Medicine III, RWTH Uni- versity-Hospital Aachen, Aachen, Germany. 4 Unit of Molecular Signal Transduction in Inammation, Inammation Research Center, VIB, Ghent, Belgium. 5 Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). J.A. Van Ginderachter and D. Laoui share senior authorship. Corresponding Author: Jo A. Van Ginderachter, Lab of Cellular and Molecular Immunology, VIB-Vrije Universiteit Brussel, Building E8, Pleinlaan 2, Brussels B-1050, Belgium. Phone: 322-629-1979; Fax: 322-629-1981; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-15-0869 Ó2015 American Association for Cancer Research. Cancer Research www.aacrjournals.org 35 on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869
Transcript
Page 1: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

Priority Report

M-CSF and GM-CSF Receptor SignalingDifferentially Regulate Monocyte Maturation andMacrophage Polarization in the TumorMicroenvironmentEvaVanOvermeire1,2, Beno�t Stijlemans1,2, FelixHeymann3, Jiri Keirsse1,2,YannickMorias1,2,Yvon Elkrim1,2, Lea Brys1,2, Chlo�e Abels1,2, Qods Lahmar1,2, Can Ergen3, Lars Vereecke4,5,Frank Tacke3, Patrick De Baetselier1,2, Jo A. Van Ginderachter1,2, and Damya Laoui1,2

Abstract

Tumors contain a heterogeneous myeloid fraction comprisedof discrete MHC-IIhi andMHC-IIlo tumor-associated macrophage(TAM) subpopulations that originate from Ly6Chi monocytes.However, the mechanisms regulating the abundance and pheno-type of distinct TAM subsets remain unknown. Here, we inves-tigated the role ofmacrophage colony-stimulating factor (M-CSF)in TAMdifferentiation and polarization in differentmouse tumormodels. We demonstrate that treatment of tumor-bearing micewith a blocking anti-M-CSFR monoclonal antibody resulted in areduction of mature TAMs due to impaired recruitment, extrav-asation, proliferation, and maturation of their Ly6Chi monocyticprecursors. M-CSFR signaling blockade shifted the MHC-IIlo/MHC-IIhi TAM balance in favor of the latter as observed by the

preferential differentiation of Ly6Chi monocytes into MHC-IIhi

TAMs. In addition, the genetic and functional signatures ofMHC-IIlo TAMs were downregulated upon M-CSFR blockade,indicating that M-CSFR signaling shapes the MHC-IIlo TAMphenotype. Conversely, granulocyte macrophage (GM)-CSFRhad no effect on the mononuclear tumor infiltrate or relativeabundance of TAM subsets. However, GM-CSFR signalingplayed an important role in fine-tuning the MHC-IIhi pheno-type. Overall, our data uncover the multifaceted and opposingroles of M-CSFR and GM-CSFR signaling in governing thephenotype of macrophage subsets in tumors, and provide newinsight into the mechanism of action underlying M-CSFRblockade. Cancer Res; 76(1); 35–42. �2015 AACR.

IntroductionTumors are often highly infiltrated with inflammatory cells

such as tumor-associated macrophages (TAM). TAMs are nowconsidered to promote tumor growth inmultiple ways, includinginduction of angiogenesis, remodeling of the extracellular matrix,stimulation of cancer cell proliferation and metastasis, and inhi-bition of adaptive immunity (1), the clinical significance of whichis evidenced by the correlation between high TAM density andworse outcome in most cancer types (2).

Ly6Chi monocytes differentiate into distinct TAM subsets withspecialized functions and a different intratumoral localization:normoxic M1-like MHC-IIhi TAM and hypoxic M2-like MHC-IIlo

TAM (3, 4). As hypoxia is not the main driver of TAM infiltrationanddifferentiation (4), we consideredmyelopoietic growth factorreceptor signaling as governor of TAM heterogeneity. In thisrespect, M-CSF–drivenmacrophage activation leads to the expres-sion of a substantial part of the M2 transcriptome, while granu-locytemacrophage colony-stimulating factor (GM-CSF) induces aM1 phenotype (5, 6). As such, blockingM-CSFR (CSF1R, CD115)signaling in myometrial macrophages stimulated the occurrenceof an M1-like MHC-IIhi population at the expense of M2-likeMHC-IIlomacrophages in the pregnantmouse uterus (7).M-CSFRsignaling was recently also shown to be critical for TAMdifferentiation and shaping the M1/M2 TAM phenotype (8–11). However, which aspects of the monocyte-to-TAM differen-tiation pathway are influenced by M-CSFR and whether GM-CSFalso skews the TAM phenotype remain largely unexplored (12).

Using mouse tumors of different histologic origin, we showhere, that Ly6Chi monocytes depend on M-CSFR signaling fortheir recruitment and extravasation to tumors and differentiationto mature TAM. M-CSFR signaling governs the phenotype of M2-like MHC-IIlo TAM, and its blockade resulted in a preferentialdifferentiation ofmonocytes toM1-likeMHC-IIhi TAM. AlthoughGM-CSF receptor (GM-CSFR, CSF-2R, or CD116) signaling is notdetermining monocyte recruitment and differentiation, it finetunes the molecular profile of MHC-IIhi TAM, hence uncovering

1LaboratoryofMyeloid Cell Immunology,VIB, Brussels, Belgium. 2Lab-oratory of Cellular and Molecular Immunology, Vrije UniversiteitBrussel, Brussels, Belgium. 3Department of Medicine III, RWTH Uni-versity-Hospital Aachen, Aachen, Germany. 4Unit of Molecular SignalTransduction in Inflammation, Inflammation Research Center, VIB,Ghent, Belgium. 5Department of Biomedical Molecular Biology,GhentUniversity, Ghent, Belgium.

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

J.A. Van Ginderachter and D. Laoui share senior authorship.

Corresponding Author: Jo A. Van Ginderachter, Lab of Cellular and MolecularImmunology, VIB-Vrije Universiteit Brussel, Building E8, Pleinlaan 2, BrusselsB-1050, Belgium. Phone: 322-629-1979; Fax: 322-629-1981; E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-15-0869

�2015 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 35

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 2: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

the differential role played byM-CSFR and GM-CSFR signaling inmacrophage differentiation and polarization in tumors.

Materials and MethodsDetailed Materials and Methods can be found in the Supple-

mentary Information

Mice, cell lines, and tumor modelsFemaleC57BL/6mice andubiquitin-GFPmice (C57BL/6)were

purchased from Janvier and Jackson, respectively. Cx3cr1gfp/þ

(C57BL/6), GM-CSFR�/� (Csf2rb�/�) (C57BL/6), Il4ra�/�

(C57BL/6), and MMTV-PyMT (FVB) mice were respectively pro-vided by Frank Tacke (Aachen University, Germany), MelanieGreter (University of Zurich, Germany), Frank Brombacher (Uni-versity of Cape Town, South Africa), and Massiliano Mazzone(KULeuven, Belgium). Procedures followed the guidelines ofthe Belgian Council for Laboratory Animal Science. C57BL/6 orBalb/c mice were injected subcutaneously with 3 � 106 3LL-RLewis lung carcinoma or TS/A breast carcinoma cells, respectively.

Tumor preparation, flow cytometry, and cell sortingTumor single-cell suspensions were prepared as described (3).

Antibodies for cell surface staining are listed in SupplementaryTable S1. To prevent aspecific binding, TAMs were preincubatedwith rat anti-mouse CD16/CD32 (2.4G2, BD Biosciences). Deltamedian fluorescence intensity (DMFI) was calculated as: (MFIstaining)� (MFI isotype staining). FACSdatawere acquired usinga BD FACSCanto II (BD Biosciences) and analyzed using FlowJo(Tree Star, Inc.). To purify monocytes or TAM, cells were sortedusing a BD FACSAria II (BD Biosciences) from 4 to 8 pooledtumors or pooled blood from 6 to 8 mice.

Anti–M-CSFR antibody administrationAnti–M-CSFR antibody (300 mg per intravenous injection;

clone AFS98) or isotype control antibody [anti-human CD44Hermes-1 (rat IgG2a)] producing hybridomas were originallyprovided by Miriam Merad and antibodies were prepared in-house. Treatment schedules ofmice are listed in the legendof eachfigure. Briefly, mice were either treated every two days startingfrom tumor inoculation or received one shot of antibody beforesacrifice depending on the experiment.

Measurement of IL34, M-CSF, GM-CSF, and CCL2 productionConcentrations of cytokines and chemokines were measured

by ELISA following the manufacturer's protocol (R&D Systems;see Supplementary Information).

Adoptive bone marrow and monocyte transfersBlood and bone marrow Ly6ChiGFPþ monocytes were sorted

from ubiquitin-GFP mice. A total of 106 cells were intravenouslyinjected in 3LL-R tumor-bearing mice. The fate of the GFPþ

progeny was determined 48 hours later. For bone marrow adop-tive transfers, 107 bone marrow cells from ubiquitin-GFP micewere injected 4 hours before checking GFPþLy6Chiþ monocytepresence in blood and tumor.

Extravasation of monocytesMice were intravenously injected 1minute before sacrifice with

1 mg rat anti-CD45/PeCy7 (30-F11; eBioscience) or rat IgG2b

isotype control/PECy7 (eB149/10H5; eBioscience) as wasdescribed before (7, 13).

Intravital imagingIntravital imaging was performed following a previously

reported protocol (14) with minor modifications (Supplemen-tary Information).

In vitro phagocytosis assayFor in vitro latex uptake, freshly isolated tumor single-cell

suspentions were cultured for 40 minutes at 4�C (control) or37�C, in the presence of yellow–green latex microspheres (Poly-sciences; 0.5mm)diluted 1:5,000. Latex uptake by tumor CD11bþ

Ly6G� cells was assessed via flow cytometry.

Bromodeoxyuridine labelingTumor-bearing mice were given an intraperitoneal injection of

1 mg bromodeoxyuridine (BrdUrd) 4 hours before sacrifice.Tumors were collected and BrdUrd intracellular staining wasperformed following the manufacturer's instructions (BD Bio-sciences; Supplementary Information).

RNA extraction, cDNA preparation, and quantitative real-timePCR

These experiments were performed as described before (3).RNA was extracted using TRIzol (Invitrogen) and was reversetranscribed with oligo(dT) and SuperScript II RT (Invitrogen).Quantitative real-time PCR was performed in an iCycler, with iQSYBR Green Supermix (Bio-Rad). Primer sequences are listed inSupplementary Table S2. PCR cycles consisted of 10 94�C, 4500

55�C, 10 72�C. Gene expression was normalized using ribosomalprotein S12 (Mrps12) as a housekeeping gene.

Statistical analysisSignificance was determined by the Student t test or ANOVA

followed by a post test using GraphPad Prism 6.0 software. A Pvalue < 0.05 was considered statistically significant. All graphsshow mean � SEM.

ResultsM-CSFR signaling blockade impairs the extravasation of tumor-infiltrating Ly6Chi monocytes

Treatment with blocking anti-M-CSFR monoclonal antibody(mAb) AFS98, but not an isotype control mAb, caused a signif-icant reduction of the CD11bþLy6G�SiglecF� infiltrate in subcu-taneous 3LL-R lung carcinoma tumors (Fig. 1A, gating strategy inSupplementary Fig. S1A), subcutaneous TS/A mammary carcino-ma tumors, and transgenic MMTV-PyMT mammary carcinomas(Supplementary Fig. S1B).

To assess whether anti-M-CSFR treatment impaired the extrav-asation of monocytes into 3LL-R tumors, we injected anti-CD45/PECy7 intravenously to label all intravascular hematopoietic cells(7, 13) and one minute later (a time span that is too short forlabeled cells to extravasate) quantified labeled versus unlabeledLy6Chi monocytes in excised tumors. Significantly, more Ly6Chi

monocytes remained in the tumor vasculature upon M-CSFRblockade (Supplementary Fig. S1C). These findings were corrob-orated via intravitalmicroscopy of anti-M-CSFR–treated tumors inCX3CR1gfp/þ mice (Fig. 1B). Reduced monocyte recruitment andextravasation could be secondary to an anti-M-CSFR–mediated

Van Overmeire et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research36

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 3: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

decrease in local CCL2 production, as was demonstrated before inother tissues (7). However, when GFPþ bone marrow cells weretransferred intravenously to tumor-bearing mice pretreated withanti-M-CSFR, the recruitment of GFPþLy6Chi monocytes intotumors was not significantly reduced compared with controlmAb–pretreated mice, arguing against major chemoattractantalterations caused by anti-M-CSFR (Fig. 1C). Accordingly, CCL2protein levels in the tumor were not influenced by M-CSFRblockade (Supplementary Fig. S1D).

To assess whether M-CSFR signaling blockade has monocyte-intrinsic effects, GFPþ bone marrow cells were pretreated withanti-M-CSFR in vitro before transfer into tumor-bearing mice.Significantly less anti-M-CSFR–bound GFPþLy6Chi monocytescould be traced back in the tumor (Fig. 1C). Importantly, theirnumbers did not differ in peripheral blood, excluding monocyteelimination in the circulation as underlying mechanism for theirreduced tumor penetrance (Supplementary Fig. S1E). Notably,the expression of adhesion molecules known to be involved in

monocyte extravasation (15) were not affected by anti-M-CSFR(Supplementary Fig. S1F). Hence, M-CSFR blockade lowersmonocyte recruitment and extravasation to tumors in a mono-cyte-intrinsic fashion.

M-CSFR signaling blockade impairs the proliferation ofperipheral and tumor-infiltrating Ly6Chi monocytes

M-CSFR signaling may drive monocyte proliferation and assuch contribute to tumor-infiltrating mononuclear cell numbers.BrdUrd pulse labeling in combination with an intravenous injec-tion of CD45-PeCy7 1 minute before sacrifice revealed that theproliferative capacity of Ly6Chimonocytes in isotype-treatedmicedropped while migrating from peripheral blood (11.7%� 2.5%)to the tumor vasculature (4.8%� 0.9%) and finally to the tumormass (0.27% � 0.05%; Fig. 1D). In all sites, the proliferation ofLy6Chi monocytes depended on M-CSFR signaling, as it signifi-cantly dropped after M-CSFR blockade (Fig. 1D). Of note, cost-aining with AnnexinV/7AAD revealed that M-CSFR antibody

Figure 1.M-CSFR regulates the behavior of tumor-infiltrating monocytes. A, the absolute number of CD11bþLy6G�SiglecF� cells was determined in 3LL-R tumor single-cellsuspensions after treatment of tumor-bearing mice with either isotype antibody or a-M-CSFR antibody every two days from day 0 postinjection until sacrifice(day 14 postinjection). Results are representative of three independent experimentswith n�4. B, representative intravital images of isotype-treated anda-M-CSFR–treated 3LL-R tumors. 3LL-R tumor-bearing CX3CR1-GFPþ/� reporter mice received one shot of isotype antibody or a-M-CSFR antibody just before imagingthe tumor (green, CX3CR1þ cells; red, labeled vessels). The percentage of CX3CR1þ cells in the parenchyma or in the tumor blood vessels was analyzed.Two fields were analyzed per animal, with two animals per group. C, influence of M-CSFR blockade on monocyte recruitment to the tumor. One million GFPþ bonemarrow cells were adoptively transferred to 11-day-old 3LL-R tumor-bearing mice. GFPþ bone marrow cells were either treated with a-M-CSFR antibodyfor 30 minutes and washed before transfer (a-M-CSFR–treated cells) or mice were injected with a-M-CSFR antibody 12 hours before GFPþ bone marrowtransfer (a-M-CSFR–treated recipients). Four hours after GFPþ bonemarrow transfer, micewere sacrificed and tumorswere collected. Graphs show the percentageofGFPþLy6Chimonocyteswithin the total amount ofGFPþ cells thatwere foundback in the tumor. Results are representative of three independent experimentswithn � 4. D, effect of M-CSFR blockade on monocyte proliferation. 11-day-old 3LL-R tumor-bearing mice were given sequentially: (i) a-M-CSFR antibody orisotype antibody intravenously 12 hours before sacrifice, (ii) one BrdUrd shot (or left untreated as control) 4 hours before sacrifice, and (iii) CD45-PeCy7 orisotype PeCy7 antibody 1 minute before sacrifice. BrdUrd incorporation was measured using intracellular flow cytometry. The graph shows the percentage ofBrdUrdþ cells within the Ly6Chi peripheral blood monocytes or Ly6Chi tumor-associated monocytes. The latter was divided in a CD45-PECy7–labeled(i.e., tumor blood vessel monocytes) and a CD45-PECy7–unlabeled population (i.e., tumor mass monocytes). Results are representative of three independentexperiments with n � 4. All experiments, graphs show mean � SEM. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001.

M-CSF and GM-CSF Receptor in Determining the TAM Phenotype

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 37

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 4: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

treatment had no effect on the viability of Ly6Chi monocytes norTAM subsets (Supplementary Fig. S2). Thus, M-CSFR signalingblockade impairs the recruitment and proliferation of Ly6Chi

monocytes, resulting in a reduced mononuclear tumor infiltrate.Notably, the amount of Ly6Chi monocytes was unaltered in theperipheral blood and spleen and was even increased in the bonemarrow afterM-CSFR blockade (Supplementary Fig. S3), possiblyresulting from the combined effect of a lower proliferation rateand reduced recruitment to the tumor.

M-CSFR signaling blockade impedes the differentiation ofLy6Chi monocytes into MHC-IIlo TAM

We next assessed whether M-CSFR signaling affected the dif-ferentiation pathway of the Ly6Chi monocytes that were still ableto infiltrate the tumor. Upon anti-M-CSFR treatment, proportion-ally more monocytes and immature TAM (gates 1 þ 2 þ 3) andless mature TAM (gates 4 þ 5, with especially a strong effect onMHC-IIlo TAM in gate 4) were found within the CD11bþ

Ly6G�SiglecF� tumor infiltrate (Fig. 2A and Supplementary Fig.S4A and S4B), suggesting a monocyte-to-macrophage differenti-ation block. This conclusion is corroborated by the diminishedgene expression level of themacrophage differentiation regulatorsPu1.1 andMafb in tumor-associated Ly6Chi monocytes from anti-M-CSFR–treated mice (Fig. 2B). In this context, it is important tonote that especially M-CSF, but much less the alternative M-CSFR

ligand IL34, is secreted in the tumor microenvironment (Supple-mentary Fig. S5A) and that blocking the M-CSFR tends to reducethe presence of this cytokine in tumors (Supplementary Fig. S5B).

Interestingly, mainly the generation of MHC-IIlo TAM washampered upon M-CSFR blockade, as the MHC-IIlo/MHC-IIhi

TAM balance shifted from an excess of MHC-IIlo TAM towardsequal amounts of both TAM subsets (Fig. 2A and SupplementaryFig. S4). Notably, also the presence of tumor-associated dendriticcells (gate 6) was not affected. Accordingly, the differentiationpathway of adoptively transferred GFPþLy6Chimonocytes shiftedfrom yielding mainly MHC-IIlo TAM in isotype-treated mice topredominantly MHC-IIhi TAM upon anti-M-CSFR treatment (Fig.2C). Importantly, the altered MHC-IIlo/MHC-IIhi TAM balancecaused by M-CSFR blockade was also seen in TS/A (Supplemen-tary Fig. S6).

M-CSFR and GM-CSFR signaling play opposing roles indetermining the phenotype of MHC-IIlo and MHC-IIhi TAM

Finally, we wondered whether M-CSFR signaling also influ-ences the phenotype of the mature TAM subsets. Both MHC-IIlo

andMHC-IIhi TAMexpressed theM-CSFR, but the expression levelin MHC-IIlo TAM was significantly higher (Fig. 2D), possiblyexplaining the greater sensitivity of this subset to M-CSFR block-ade. When analyzing a panel of genes we previously identified asbeing associated with the distinct TAM subsets, it became clear

Figure 2.M-CSFR regulates thematuration andpreferential differentiation of Ly6Chimonocytes toMHC-IIlo TAM. A, single-cell suspensions of 13-day-old 3LL-R tumors treatedwith isotype or a-M-CSFR antibody were stained for myeloid populations. Ly6C versus MHC-II plots of the gated CD11bþLy6G�SiglecF� population areshown and the indicated populations are 1, Ly6Chi monocytes; 2, MHC-IIlo immature macrophage (M�); 3, MHC-IIhi immature M�; 4, MHC-IIlo TAM;5, MHC-IIhi TAM; and 6, tumor-associated dendritic cells (TADC). Graphs show the percentage of all these subsets within the CD11bþLy6G�SiglecF�

gate. B, expression of themacrophagematuration markers Pu.1 andMafb in sorted Ly6Chi monocytes from 13-day-old 3LL-R tumors grown in isotype or a-M-CSFR–treated animals was assessed using qRT-PCR. The expression of each gene was normalized based on the S12 housekeeping gene. C, GFPþLy6Chi monocyteadoptive transfer. Ly6Chi monocytes were sorted from blood and bone marrow of ubiquitin-GFP reporter mice and incubated with either a-M-CSFRantibody or isotype antibody for 30 minutes. GFPþLy6Chi monocytes were intravenously injected in 9-day-old 3LL-R tumor bearing mice. The fate of the GFPþ

progeny was detected in tumors after 48 hours. The Ly6C versus MHC-II plot was gated on GFPþCD11bþLy6G� cells. Graphs show the percentage ofMHC-IIlo and MHC-IIhi TAM within the GFPþCD11bþLy6G� cells. D, expression of M-CSFR and GM-CSFR by tumor-associated myeloid subpopulations.Single-cell suspensions of 13-day-old 3LL-R tumors were stained for M-CSFR and GM-CSFR. Values are represented as DMFI ¼ MFI marker � MFI isotypecontrol. Graphs show mean � SEM. All experiments, n � 3. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001.

Van Overmeire et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research38

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 5: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

that most (but not all) MHC-IIlo TAM-associated genes (3, 4),such asMrc1, Arg1, Cd163, andMmp9were downregulated in theMHC-IIlo TAM upon M-CSFR inhibition, while several MHC-IIhi

TAM-associated genes, such as Mgl2, Cdh1, Cldn1, Retnla, Ptgs2,and Ccl17 were upregulated in these cells (Fig. 3A). Hence, M-CSFR blockade enhanced the MHC-IIhi TAM gene signature inMHC-IIlo TAM. Accordingly, the protein expression level of mar-kers highly expressed on MHC-IIlo TAM (MMR, SR-A and IL4Ra)significantly dropped after M-CSFR blockade (Fig. 3B). The dropin IL4Ra expression may imply that part of the anti-M-CSFR–mediated effects on gene expression are due to diminished IL4Rasignaling. Although some genes were indeed regulated by IL4Rain the tumor microenvironment, the effects of M-CSFR blockadeare largely maintained in IL4Ra-deficient mice and are thus IL4/IL13-independent (Supplementary Fig. S7).Notably, the inherentphagocytic capacity of the MHC-IIlo TAM, which is one of itshallmark functions (3), was strongly diminished by M-CSFR

blockade as evidenced by lower uptake of latex beads ex vivo (Fig.3C). In addition, the capacity of both MHC-IIlo and MHC-IIhi

TAM to stimulate na€�ve CD4þ and CD8þ T cells was increasedfollowing M-CSFR blockade, further evidencing a functional shift(Supplementary Fig. S8).

M-CSF and GM-CSF were reported to have opposing effects onthe macrophage activation state (5, 6). Both TAM subsetsexpressed high levels of theGM-CSFR, with the highest expressionseen on the MHC-IIhi population (Fig. 2D). To assess whetherGM-CSF regulates the TAM phenotype, 3LL-R tumors were inoc-ulated in GM-CSFR–deficient mice. Notably, the abundance ofthe mononuclear tumor infiltrate was comparable between GM-CSFRKOandWT tumors, the Ly6Chimonocytes expressed similarlevels ofmaturationmarkers (Fig. 3D) and the relative abundanceof TAM subsets was unaltered (Fig. 3D). These data illustrate thatGM-CSF does neither regulate monocyte extravasation nor theirintratumoral differentiation. However, several genes that are

Figure 3.M-CSFR predominantly affects the MHC-IIlo TAM phenotype. A, expression of typical MHC-IIlo TAM- and MHC-IIhi TAM-associated genes in TAM populations sortedfrom 13-day-old 3LL-R tumors, treated every two days with either isotype antibody or a-M-CSFR antibody, was assessed using qRT-PCR. The expressionof each gene was normalized based on the S12 housekeeping gene. B, MMR, IL4Ra, SR-A, and CD11c expression was assessed by flow cytometry on MHC-IIlo

and MHC-IIhi TAM from 13-day-old 3LL-R tumors treated with either isotype antibody or a-M-CSFR antibody every two days. Values are represented asDMFI¼ MFI marker �MFI control. C, tumor single-cell suspensions of 13-day-old 3LL-R tumor-bearing mice, treated with isotype antibody or a-M-CSFR antibody,were cultured in vitro at 37�C in the absence (control) or presence of fluorescent latex beads for 40 minutes. The percentage of latexþ cells withinthe Ly6Chi monocyte, MHC-IIlo TAM, andMHC-IIhi TAM gates is given. D, single-cell suspensions of 13-day-old 3LL-R tumors grown inWT or GM-CSFR–deficient micewere stained for myeloid subsets. Graphs show the absolute number of CD11bþLy6G�SiglecF� and the percentage of Ly6Chi monocytes, MHC-IIlo, andMHC-IIhi TAM within the CD11bþLy6G�SiglecF� population. Expression of maturation markers in sorted Ly6Chi monocytes from 13-day-old 3LL-R tumors grownin WT or GM-CSFR–deficient mice was assessed using qRT-PCR. The expression of each gene was normalized on the basis of the S12 housekeeping gene.All experiments, graphs show mean � SEM, n � 3. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001.

M-CSF and GM-CSF Receptor in Determining the TAM Phenotype

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 39

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 6: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

highly expressed by MHC-IIhi TAM (3, 4), such as Mgl2, Cdh1,Cldn1, Retnla, Ptgs2, Ccl17, and IL1bwere strongly downregulatedin GM-CSFR KO MHC-IIhi TAM (Fig. 4A). Conversely, genes thatare typically associated with the MHC-IIlo TAM were eitherunchanged or upregulated in the GM-CSFR KO MHC-IIhi popu-lation (except Arg1). A similar general trend was observed in GM-CSFR KOMHC-IIlo TAM (Fig. 4A). At the protein expression level,the MHC-IIhi TAM marker CD11c dropped in GM-CSFR KOMHC-IIhi TAM, while surface markers associated with a MHC-IIlo

TAM signature (IL4Ra, MMR, and SRA) increased or remainedunaltered (Fig. 4B). In addition, the phagocytic capacity of bothTAM subsets was enhanced in the absence of GM-CSFR, whilehaving no effect on the Ly6Chi monocytes (Fig. 4C).

Interestingly, treatment of GM-CSFR–deficient mice withblocking anti-M-CSFR mAb caused a significant reduction of the

CD11bþLy6G�SiglecF� tumor infiltrate (Supplementary Fig.S9A), hampered the generationofMHC-IIlo TAM(SupplementaryFig. S9B), and enhanced the MHC-IIhi TAM gene and proteinsignature in MHC-IIlo TAM (Supplementary Fig. S9C and S9D),similar to blocking M-CSFR signaling in WT mice (Figs. 1A, 2A,and 3A and B). Hence, the induction of a more M1-like TAMphenotype upon M-CSFR blockade is GM-CSF–independent.

Together, these data validate the distinct role of M-CSFR andGM-CSFR signaling in macrophage polarization in tumors in vivo(Fig. 4D).

DiscussionStrong reductions in the number of TAM have been reported in

various tumor models upon blocking M-CSFR signaling using

Figure 4.GM-CSFR predominantly affects the MHC-IIhi TAM phenotype. A, expression of typical MHC-IIlo TAM- and MHC-IIhi TAM-associated genes in TAM populations sortedfrom 13-day-old 3LL-R tumors grown in WT or GM-CSFR–deficient mice was assessed using qRT-PCR. The expression of each gene was normalized on thebasis of the S12 housekeeping gene. B, MMR, IL4Ra, SR-A, and CD11c expression was assessed by flow cytometry on MHC-IIlo and MHC-IIhi TAM from 13-day-old3LL-R tumors grown in WT and GM-CSFR–deficient mice. Values are represented as DMFI ¼ MFI marker � MFI control. C, tumor single-cell suspensions of13-day-old 3LL-R tumor-bearing mice, grown in WT or GM-CSFR–deficient mice, were cultured in vitro at 37�C in the absence (control) or presence offluorescent latex beads for 40 minutes. The percentage of latexþ cells within the Ly6Chi monocyte, MHC-IIlo TAM, and MHC-IIhi TAM gates is given. All experiments,graphs show mean � SEM, n � 3. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001. D, overview figure. Ly6Chi monocytes proliferate and extravasateinto the tumor in a M-CSFR–dependent manner. These cells then differentiate into either MHC-IIlo or MHC-IIhi TAM, whereby the former critically depend onM-CSFR signaling. MHC-IIlo and MHC-IIhi TAM differentially express M-CSFR and GM-CSFR, whose signaling affects the molecular and functional phenotypeof these cells. The table represents the effect of M-CSFR blockade or GM-CSFR deficiency on gene expression of the TAM subsets in a color code. Genestypically associated with MHC-IIlo or MHC-IIhi TAMs are listed. Red/orange indicates that expression of the gene increases in treated/KO subsets; lightgreen/green shows a drop in treated/KO subsets compared with their WT counterparts.

Van Overmeire et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research40

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 7: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

several M-CSFR blockers (12). However, the underlying mechan-isms remain largely unexplored. In this study, we show that M-CSFR, but not GM-CSFR, signaling, is responsible for the recruit-ment, extravasation, proliferation, and maturation of tumor-infiltrating Ly6Chi monocytes. The exact mechanism by whichM-CSFR signaling regulates monocyte extravasation remainsunknown, but appears not to be CCL2 driven like in some othertissues (7). However, we cannot exclude the possibility that theprocess of extravasation is driven by other M-CSFR–regulatedchemokines in these tumors. Another M-CSFR–regulated mole-cule, involved in monocyte and macrophage migration andactivation is the GTPase Rac2 (16), which could be an interestingcandidate for further investigation.

We also show that M-CSFR, but not GM-CSFR, inhibitionabrogated the differentiation of Ly6Chi monocytes to M2-likeMHC-IIlo TAM.M-CSFRblockadewaspreviously shown to inducea shift in phenotype from MHC-IIlo to MHC-IIhi TAM in asyngeneic mouse model of BRAFV600E-driven melanoma (9),to drive the repolarization of protumoral M2 to M1 TAM in amouse glioblastoma model (8) and to cause the preferentialdepletion of M2-like MMRhi TAM (10). Notably, M2-likeMMRhi TAM may be MHC-IIhi in some models and are alsodepletedbyM-CSFRblockade (11, 17).Overall, these data suggestthat M-CSFR blockade preferentially depletes mature M2-likeTAM. Administration of M-CSFR blocking mAbs to patients withdiffuse-type giant cell tumors even led to clinical responses,validating M-CSF and its receptor as a therapeutic target (18).

We now show for the first time that M-CSFR and GM-CSFRsignaling are in fact opposing driving forces in the tumor micro-environment, regulating the MHC-IIlo and MHC-IIhi TAM phe-notype, respectively. While M-CSFR predominantly regulated thedifferentiation andM2-like properties of MHC-IIlo TAM in a GM-CSFR–independent fashion, GM-CSFR fine-tuned the M1-likeMHC-IIhi phenotype. A differential expression of the respectivereceptorsmaymediate part of this effect.However, as distinct TAMsubsets are differentially locatedwithin the tumor, with theMHC-IIlo TAM associated with more hypoxic regions (3, 4), a locor-egional production of M-CSF and GM-CSF in different compart-ments of the tumor microenvironment may also be involved.Interestingly, many of the genes that were shown to be regulated

by GM-CSFR in MHC-IIhi TAM in the current study were reportedbefore as typical GM-CSF–induced genes in bone marrow–derived macrophages in vitro (Cldn1, Cdh1, Ccl17, IL1b; ref. 19).Hence, even in the complex tumormicroenvironment harboring amultitude of potential macrophage-regulating cues, M-CSFR andGM-CSFR signaling are important for skewing macrophagepolarization.

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

Authors' ContributionsConception and design: E. Van Overmeire, P. De Baetselier, J.A. Van Ginder-achter, D. LaouiDevelopment of methodology: E. Van Overmeire, F. Heymann, C. Ergen,J.A. Van GinderachterAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): E. Van Overmeire, B. Stijlemans, F. Heymann,J. Keirsse, Y. Morias, C. Abels, Q. Lahmar, L. Vereecke, F. Tacke, D. LaouiAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): E. Van Overmeire, F. Heymann, Y. Morias, C. Ergen,J.A. Van Ginderachter, D. LaouiWriting, review, and/or revision of the manuscript: E. Van Overmeire,F. Heymann, P. De Baetselier, J.A. Van Ginderachter, D. LaouiAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): B. Stijlemans, Y. Elkrim, L. Brys, C. Ergen,F. TackeStudy supervision: P.De Baetselier, J.A. Van Ginderachter, D. Laoui

AcknowledgmentsThe authors thank Ella Omasta, Marie-Th�er�ese Detobel, Maria Slazak, and

Nadia Abou for technical and administrative assistance.

Grant SupportThis work was supported by a doctoral grant from FWO-Vlaanderen (E. Van

Overmeire, C. Abels and J.A. Van Ginderachter), a scholarship from "StichtingEmmanuel van der Schueren" (E. Van Overmeire and D. Laoui), a grant from"Stichting tegen Kanker" (P. De Bateselier and J.A. Van Ginderachter), and agrant from Vlaamse Liga tegen Kanker (J.A. Van Ginderachter).

Received March 30, 2015; revised September 16, 2015; accepted October 10,2015; published OnlineFirst November 16, 2015.

References1. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression

and metastasis. Cell 2010;141:39–51.2. Zhang QW, Liu L, Gong CY, Shi HS, Zeng YH, Wang XZ, et al. Prognostic

significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature. PLoS ONE 2012;7:e50946.

3. Movahedi K, Laoui D, Gysemans C, BaetenM, Stange G, Van den Bossche J,et al. Different tumor microenvironments contain functionally distinctsubsets of macrophages derived from Ly6C(high) monocytes. Cancer Res2010;70:5728–39.

4. Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, et al.Tumor hypoxia does not drive differentiation of tumor-associated macro-phages but rather fine-tunes the M2-like macrophage population. CancerRes 2014;74:24–30.

5. Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte-macro-phage colony-stimulating factor (CSF) and macrophage CSF-dependentmacrophage phenotypes display differences in cytokine profiles and tran-scription factor activities: implications for CSF blockade in inflammation. JImmunol 2007;178:5245–52.

6. Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M, Vaisberg E,et al. Human IL-23-producing type 1 macrophages promote but IL-10-

producing type 2 macrophages subvert immunity to (myco)bacteria. ProcNatl Acad Sci U S A 2004;101:4560–5.

7. Tagliani E, Shi C, Nancy P, Tay CS, Pamer EG, Erlebacher A. Coordinateregulation of tissuemacrophage and dendritic cell population dynamics byCSF-1. J Exp Med 2011;208:1901–16.

8. Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, QuailDF, et al. CSF-1R inhibition alters macrophage polarization and blocksglioma progression. Nat Med 2013;19:1264–72.

9. Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, et al. Inhibition of CSF-1receptor improves the antitumor efficacy of adoptive cell transfer immu-notherapy. Cancer Res 2014;74:153–61.

10. Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, et al. CSF1/CSF1Rblockade reprograms tumor-infiltratingmacrophages and improvesresponse to T-cell checkpoint immunotherapy inpancreatic cancermodels.Cancer Res 2014;74:5057–69.

11. Strachan DC, Ruffell B, Oei Y, Bissell MJ, Coussens LM, Pryer N, et al.CSF1R inhibition delays cervical and mammary tumor growth inmurine models by attenuating the turnover of tumor-associated macro-phages and enhancing infiltration by CD8 T cells. Oncoimmunology2013;2:e26968.

M-CSF and GM-CSF Receptor in Determining the TAM Phenotype

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 41

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 8: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

12. Laoui D, Van Overmeire E, De Baetselier P, Van Ginderachter JA, Raes G.Functional relationship between tumor-associatedmacrophages andmac-rophage colony-stimulating factor as contributors to cancer progression.Front Immunol 2014;5:489.

13. Van Gassen N, Van Overmeire E, Leuckx G, Heremans Y, De Groef S, Cai Y,et al. Macrophage dynamics are regulated by local macrophage prolifer-ation and monocyte recruitment in injured pancreas. Eur J Immunol2015;45:1482–93.

14. Heymann F,NiemietzPM, Peusquens J, ErgenC, KohlheppM,Mossanen JC,et al. Long term intravitalmultiphotonmicroscopy imaging of immune cellsin healthy and diseased liver using CXCR6.Gfp reportermice. J Vis Exp 2015;(97). doi: 10.3791/52607.

15. Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immu-nity 2014;41:694–707.

16. Joshi S, Singh AR, Zulcic M, Bao L, Messer K, Ideker T, et al. Rac2 controlstumor growth, metastasis and M1-M2 macrophage differentiation in vivo.PLoS ONE 2014;9:e95893.

17. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al.Targeting tumor-infiltrating macrophages decreases tumor-initiating cells,relieves immunosuppression, and improves chemotherapeutic responses.Cancer Res 2013;73:1128–41.

18. Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V,et al. Targeting tumor-associated macrophages with anti-CSF-1Rantibody reveals a strategy for cancer therapy. Cancer Cell 2014;25:846–59.

19. LaceyDC, Achuthan A, FleetwoodAJ, DinhH, Roiniotis J, Scholz GM, et al.Defining GM-CSF- and macrophage-CSF-dependent macrophageresponses by in vitro models. J Immunol 2012;188:5752–65.

Cancer Res; 76(1) January 1, 2016 Cancer Research42

Van Overmeire et al.

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869

Page 9: M-CSF and GM-CSF Receptor Signaling Differentially ... · Priority Report M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization

2016;76:35-42. Published OnlineFirst November 16, 2015.Cancer Res   Eva Van Overmeire, Benoît Stijlemans, Felix Heymann, et al.   MicroenvironmentMonocyte Maturation and Macrophage Polarization in the Tumor M-CSF and GM-CSF Receptor Signaling Differentially Regulate

  Updated version

  10.1158/0008-5472.CAN-15-0869doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2015/11/13/0008-5472.CAN-15-0869.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/76/1/35.full#ref-list-1

This article cites 18 articles, 9 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/76/1/35.full#related-urls

This article has been cited by 17 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/76/1/35To request permission to re-use all or part of this article, use this link

on October 12, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 16, 2015; DOI: 10.1158/0008-5472.CAN-15-0869


Recommended