+ All Categories
Home > Documents > Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai...

Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai...

Date post: 20-Jul-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
12
Small Molecule Therapeutics Colony-Stimulating Factor 1 Receptor Blockade Inhibits Tumor Growth by Altering the Polarization of Tumor-Associated Macrophages in Hepatocellular Carcinoma Jian-Yang Ao 1,2,3 , Xiao-Dong Zhu 1,2 , Zong-Tao Chai 1,2 , Hao Cai 1,2 , Yuan-Yuan Zhang 1,2 , Ke-Zhi Zhang 4 , Ling-Qun Kong 5 , Ning Zhang 1,2 , Bo-Gen Ye 1,2 , De-Ning Ma 1,2 , and Hui-Chuan Sun 1,2 Abstract Colony-stimulating factor-1 (CSF-1) and its receptor, CSF- 1R, regulate the differentiation and function of macrophages and play an important role in macrophage inltration in the context of hepatocellular carcinoma. The therapeutic effects of CSF-1R blockade in hepatocellular carcinoma remain unclear. In this study, we found that CSF-1R blockade by PLX3397, a competitive inhibitor with high specicity for CSF-1R tyrosine kinase, signicantly delayed tumor growth in mouse models. PLX3397 inhibited the proliferation of macrophages in vitro, but intratumoral macrophage inltration was not decreased by PLX3397 in vivo. Gene expression proling of tumor- associated macrophages (TAM) showed that TAMs from the PLX3397-treated tumors were polarized toward an M1-like phenotype compared with those from vehicle-treated tumors. In addition, PLX3397 treatment increased CD8 þ T-cell inl- tration, whereas CD4 þ T-cell inltration was decreased. Fur- ther study revealed that tumor cellderived CSF-2 protected TAMs from being depleted by PLX3397. In conclusion, CSF-1R blockade delayed tumor growth by shifting the polarization rather than the depletion of TAMs. CSF-1R blockade warrants further investigation in the treatment of hepatocellular carci- noma. Mol Cancer Ther; 16(8); 154454. Ó2017 AACR. Introduction Hepatocellular carcinoma is one of the leading causes of cancer-related death worldwide (1). Many cancers, including hepatocellular carcinoma, arise from sites of infection and chronic inammation (2). Inammation has been recognized as having a role in tumor initiation and tumor progression (3, 4). A tumor microenvironment largely shaped by inam- matory cells is favorable for tumor growth and progression (5). Tumor-associated macrophages (TAM) are a major component of inammatory cellular inltrates in tumors and play a pivotal role in tumor progression in inammation-related cancer (610). Macrophage activation in response to different agents has long been recognized as M1 and M2 macrophages. Differential cytokine production is a key feature of polarized macrophages (11). The M1 phenotype macrophages typically produce IL12b and IL23, whereas M2 macrophages produce IL10, chemokine (C-C motif) ligand (CCL)17, and CCL22 (6, 12). Our previous studies found that colony-stimulating factor 1 (CSF-1) expression and TAM density (CD68 or CSF-1 receptor, CSF-1R) in the adjacent liver tissue are associated with patient survival after resection of hepatocellular carcino- ma (1315), suggesting that CSF-1/CSF1R may play an impor- tant role in tumor progression and macrophage polarization in hepatocellular carcinoma (16). CSF-1 is a cytokine that controls the differentiation and function of macrophages via its receptor, CSF-1R. Ligand bind- ing activates the receptor kinase through a process of oligo- merization and transphosphorylation. Cellular changes occur in macrophages under different circumstances, which contri- butes to the heterogeneity and variability of TAMs (17). This plasticity is a hallmark of myeloid cells, particularly those of the monocytemacrophage lineage (18). Activated forms of macro- phages, known as M1 and M2, are linked to lineage-determin- ing growth factors in T helper 1 (Th1) and T helper 2 (Th2) cells (19). CSF-1 and granulocytemacrophage CSF (GMCSF, or CSF-2) are often used to obtain mature macrophages, and they are also involved in the polarization of anti-inammatory/ protumorigenic and pro-inammatory/antitumorigenic macro- phages, respectively (11, 20, 21). We previously found that depletion of TAMs enhanced the antitumor effect of sorafenib in liver cancer models (22, 23), and a clinical trial has been initiated in our hospital to test the safety of a 1 Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China. 2 Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China. 3 Department of Hepatobiliary Surgery, the First Afliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China. 4 Department of Hepatobiliary Surgery, Taizhou People's Hospital, Taizhou, Jiangsu, China. 5 Department of Hepatobiliary Surgery, Binzhou Medical College Afliated Hospital, Binzhou, Shandong, China. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of Surgery, Zhongshan Hos- pital, Fudan University, Shanghai 200032, China. Phone: 86-021-54237965; Fax: 86-21-64037181; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-16-0866 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 16(8) August 2017 1544 on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866
Transcript
Page 1: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

Small Molecule Therapeutics

Colony-Stimulating Factor 1 Receptor BlockadeInhibitsTumorGrowthbyAltering thePolarizationof Tumor-Associated Macrophages inHepatocellular CarcinomaJian-Yang Ao1,2,3, Xiao-Dong Zhu1,2, Zong-Tao Chai1,2, Hao Cai1,2, Yuan-Yuan Zhang1,2,Ke-Zhi Zhang4, Ling-Qun Kong5, Ning Zhang1,2, Bo-Gen Ye1,2, De-Ning Ma1,2, andHui-Chuan Sun1,2

Abstract

Colony-stimulating factor-1 (CSF-1) and its receptor, CSF-1R, regulate the differentiation and function of macrophagesand play an important role in macrophage infiltration in thecontext of hepatocellular carcinoma. The therapeutic effects ofCSF-1R blockade in hepatocellular carcinoma remain unclear.In this study, we found that CSF-1R blockade by PLX3397, acompetitive inhibitor with high specificity for CSF-1R tyrosinekinase, significantly delayed tumor growth in mouse models.PLX3397 inhibited the proliferation of macrophages in vitro,but intratumoral macrophage infiltration was not decreasedby PLX3397 in vivo. Gene expression profiling of tumor-

associated macrophages (TAM) showed that TAMs from thePLX3397-treated tumors were polarized toward an M1-likephenotype compared with those from vehicle-treated tumors.In addition, PLX3397 treatment increased CD8þ T-cell infil-tration, whereas CD4þ T-cell infiltration was decreased. Fur-ther study revealed that tumor cell–derived CSF-2 protectedTAMs from being depleted by PLX3397. In conclusion, CSF-1Rblockade delayed tumor growth by shifting the polarizationrather than the depletion of TAMs. CSF-1R blockade warrantsfurther investigation in the treatment of hepatocellular carci-noma. Mol Cancer Ther; 16(8); 1544–54. �2017 AACR.

IntroductionHepatocellular carcinoma is one of the leading causes

of cancer-related death worldwide (1). Many cancers, includinghepatocellular carcinoma, arise from sites of infection andchronic inflammation (2). Inflammation has been recognizedas having a role in tumor initiation and tumor progression(3, 4). A tumor microenvironment largely shaped by inflam-matory cells is favorable for tumor growth and progression (5).Tumor-associated macrophages (TAM) are a major componentof inflammatory cellular infiltrates in tumors and play apivotal role in tumor progression in inflammation-relatedcancer (6–10). Macrophage activation in response to different

agents has long been recognized as M1 and M2 macrophages.Differential cytokine production is a key feature of polarizedmacrophages (11). The M1 phenotype macrophages typicallyproduce IL12b and IL23, whereas M2 macrophages produceIL10, chemokine (C-C motif) ligand (CCL)17, and CCL22(6, 12). Our previous studies found that colony-stimulatingfactor 1 (CSF-1) expression and TAM density (CD68 or CSF-1receptor, CSF-1R) in the adjacent liver tissue are associatedwith patient survival after resection of hepatocellular carcino-ma (13–15), suggesting that CSF-1/CSF1R may play an impor-tant role in tumor progression and macrophage polarization inhepatocellular carcinoma (16).

CSF-1 is a cytokine that controls the differentiation andfunction of macrophages via its receptor, CSF-1R. Ligand bind-ing activates the receptor kinase through a process of oligo-merization and transphosphorylation. Cellular changes occurin macrophages under different circumstances, which contri-butes to the heterogeneity and variability of TAMs (17). Thisplasticity is a hallmark of myeloid cells, particularly those of themonocyte–macrophage lineage (18). Activated forms of macro-phages, known as M1 and M2, are linked to lineage-determin-ing growth factors in T helper 1 (Th1) and T helper 2 (Th2) cells(19). CSF-1 and granulocyte–macrophage CSF (GMCSF, orCSF-2) are often used to obtain mature macrophages, and theyare also involved in the polarization of anti-inflammatory/protumorigenic and pro-inflammatory/antitumorigenic macro-phages, respectively (11, 20, 21).

We previously found that depletion of TAMs enhanced theantitumor effect of sorafenib in liver cancermodels (22, 23), and aclinical trial has been initiated in our hospital to test the safety of a

1Department of Liver Surgery and Transplantation, Liver Cancer Institute andZhongshan Hospital, Fudan University, Shanghai, China. 2Key Laboratory ofCarcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.3Department of Hepatobiliary Surgery, the First Affiliated Hospital of WenzhouMedical University, Wenzhou, Zhejiang, China. 4Department of HepatobiliarySurgery, Taizhou People's Hospital, Taizhou, Jiangsu, China. 5Department ofHepatobiliary Surgery, Binzhou Medical College Affiliated Hospital, Binzhou,Shandong, China.

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

J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article.

Corresponding Author: Hui-Chuan Sun, Professor of Surgery, Zhongshan Hos-pital, Fudan University, Shanghai 200032, China. Phone: 86-021-54237965; Fax:86-21-64037181; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-16-0866

�2017 American Association for Cancer Research.

MolecularCancerTherapeutics

Mol Cancer Ther; 16(8) August 20171544

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 2: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

combination treatment using sorafenib with zoledronic acid(ClinicalTrials.gov identifier: NCT01259193). Another approachis the deactivation of TAMs by a CSF-1R inhibitor, which wasstudied in diffuse-type giant cell tumor (24) and glioblastomamultiforme (25). PLX3397, a competitive inhibitor with highspecificity for CSF-1R tyrosine kinase, has been tested in severalstudies, with promising therapeutic results (26–33).

In the present study, we tested the effect of PLX3397 inhepatocellular carcinomamousemodels and found that PLX3397treatment delayed tumor growth. This delay was mediated byaltering TAM polarization rather than by causing TAM depletion.

Materials and MethodsCell lines

Four human hepatocellular carcinoma cell lines (HepG2,MHCC97-H, MHCC97-L, and HCCLM3), one nontransformedhuman hepatocyte cell line (L-02), and onemouse hepatocellularcarcinoma cell line (Hepa1-6) were used in this study. MHCC97-H, MHCC97-L, and HCCLM3 were established at our institute(34, 35).HepG2 andHepa1-6were obtained fromATCC.Humanhepatic stellate cell line LX2 was obtained from ShanghaiAdvanced Research Institute, Chinese Academy of Sciences. Thesecell lines were maintained in DMEM (Gibco BRL) supplementedwith 10% (v/v) FBS (Gibco BRL).

Human monocyte THP-1 cells obtained from ATCC werecultured in RPMI1640 (Invitrogen) containing 10% FBS andsupplemented with 10 mmol/L Hepes (Gibco BRL). THP-1was differentiated into macrophages by 24-hour incubationwith 160 nmol/L phorbol 12-myristate 13-acetate (PMA; Sig-ma) followed by 24-h incubation in RPMI medium. Macro-phages were further polarized to M1 macrophages by incu-bation with 10 pg/mL of lipopolysaccharide (LPS; Sigma)and 20 ng/mL of IFNg (R&D Systems) and are referred to asM(LPSþIFNg) cells. M2 macrophages were obtained by incu-bation with 20 ng/mL of IL4 (R&D Systems) and are referredto as M(IL4) cells (36).

A human umbilical vein endothelial cell (HUVEC) line waspurchased from Allcells and was grown in Allcells completedmedium supplemented with 10% FBS. All cell lines obtainedfrom the cell bank were tested for authentication using shorttandem repeat fingerprinting and were passaged for fewer than 6months.

Isolation of TAMs and T cells from tumor tissuesIsolated TAMs were obtained from orthotopically implanted

hepa1-6 tumors in C57BL/6 mice. Specimens were minced withscissors and digested by incubation for 1 hour at 37�C in high-glucose DMEM (Life Technologies) containing 0.1% collagenaseIV (Sigma). After being washed inmedium plus 10% FBS, the cellsuspensionwas forced through a graded series ofmeshes to obtainsingle-cell suspensions and centrifuged at 400 g for 30 to 40minutes at 18 to 20�C on layers of Lympholyte-M (CedarlaneLabs), after which the upper layer was removed, thus leavingthe lymphocyte layer undisturbed at the interface. TAMs weresortedusing FACS (BectonDickinson)withCD45þ, CD11bþ, andF4/80þ (ref. 37; Becton Dickinson) according to the protocollisted at http://www.immgen.org/Protocols/ImmGen%20Cell%20prep%20and%20sorting%20SOP.pdf. TAMs growing inDMEM supplemented with 10% FBS were tested to determinetheir biological characteristics.

CD8þ T and CD4þ T cells were purified using the CD8þ orCD4þ T Cell Isolation Kit from Miltenyi Biotec, respectively. Thepurified T-cell suspension was labeled with CFSE (Invitrogen). Atotal of 1� 105 CFSE labeled CD8þ or CD4þ T cells were added toCD3/CD28 coated plates (1.0 mg/mL each, eBioscience) to induceT-cell proliferation. Analysis of cells was performed on a BDLSRFortessa cell analyzer after labeling with fluorescent antibo-dies and 7AAD (BD Bioscience) to exclude dead cells.

ELISAIFNg concentrations in the supernatants were measured by

ELISA Kit (R&D Systems) according to themanufacturer's instruc-tion. We collected the total cell protein to assess the different cellnumbers of the different groups. An equal volume of lysis bufferwas added before the total cellular protein extracted, and thenbicinchoninic acid (BCA) assay was used to measure the proteinconcentration. Thereafter, the IFNg concentrationwasnormalizedto the total cellular protein.

Flow cytometry analysisLymphocytes isolated from tumors were washed in PBS

containing 0.5% (w/v) BSA. After cells were counted, single-cell suspensions in FACS buffer (1% IgG free BSA in PBS;Jackson Immunoresearch) were incubated with 1 mL of FcBlock (Becton Dickinson) for every 1 million cells for at least15 minutes at 4�C. Surface staining was performed in the darkfor 30 minutes at 4�C in staining buffer. Cells were thenincubated for 30 minutes at 4�C with the appropriate antibodyor with a control in PBS containing 0.5% (w/v) BSA. Cells wereanalyzed on a fluorescence activated cell sorter (BD LSRFor-tessa; Becton Dickinson). Surface markers used for theseexperiments included F4/80 clone REA126 FITC (1:100, Mil-tenyi Biotec), CD11b clone M1/70 PE-Cy7 (1:125, BD Phar-mingen), Ly6G clone 1A8 Alexa Fluor 700 (1:125, BD Phar-mingen), and MHC Class II clone M5/114.15.2 APC (1:100,Miltenyi Biotec). Directly conjugated mouse immunoglobulinG1k was used for isotype controls. Cells were then washedtwice with staining buffer followed by fixation in 1% parafor-maldehyde (VWR). Cells were analyzed on a BD LSRFortessacell analyzer and gating was performed with FlowJo analysissoftware (TreeStar). Biexponential transformation was adjust-ed manually when necessary.

Obtaining CSF1- andCSF2-stimulatedmacrophages frombonemarrow–derived macrophages

Femur and tibia bones were harvested from 8-week-old wild-type (WT) C57BL/6mice. Bonemarrowwas flushed out into coldPBS (Life Technologies) plus 2% heat-inactivated FBS, passedthrough a needle five times to dissociate the cells, and then passedthrough a 70-mm cell strainer (Becton Dickinson) to remove cellclumps, bone, hair, and other cells/tissues. After addition of threevolumes of NH4Cl solution (0.8% NH4Cl solution; BeyotimeInstitute of Biotechnology, Jiangsu, China), the mixture wasincubated on ice for 10 minutes to remove red blood cells; thecells were then spun down and resuspended in cold PBS with 2%FBS (38). The harvested cells were cultured in DMEM containing10% FBS and supplemented with 10 ng/mL recombinant mouseCSF1 (R&D Systems) or 10 ng/mL recombinant mouse CSF2(R&D Systems) for 7 days to obtain CSF1- or CSF2-induced bonemarrow–derived macrophages (BMDMs): M(CSF1) or M(CSF2)cells, respectively.

CSF1R Blockade and Macrophage Polarization

www.aacrjournals.org Mol Cancer Ther; 16(8) August 2017 1545

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 3: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

Immunohistochemical assayFrozen sections (5 mm) of tumor samples were used to deter-

mine the cells with dual expression of CD68 (1:250, AbD Serotec;1:200, Abcam) and Fizz1 (1:100; Abcam), cells with dual expres-sion of CD68 and MHC-II (1:100; Abcam), or cells with dualexpression of CD68 and CSF-1R (1:100; Genetex) by immuno-fluorescent staining and were then imaged by confocal laserscanning microscopy.

Macrophages growing on glass coverslips were fixed in 4%paraformaldehyde for 15 minutes, rinsed three times with PBSfor 5 minutes each time, and incubated in a protein-blockingsolution for 30 minutes at room temperature. After incubationwith the primary antibody against CD68 and F4/80 (1:100;Abcam), CD68 and CSF-1R (1:100; Novus Biologicals), CD68and CSF2Ra (1:100; Abcam), or CD68 and CSF2Rb (1:100;Abcam) overnight at 4�C followed by incubation with thesecondary antibody (Alexa Fluor 488 donkey anti-rat, AlexaFluor 546 donkey anti-rabbit, 1:300; Jackson Immunoresearch)at 37�C for 2 hours, the cells were counterstained with DAPI(Beyotime). Cells on slides not incubated with primary anti-bodies served as negative controls.

IHC assay was performed on 6-mm sections of paraffin-embed-ded Hepa1-6 tumor tissues using antibodies against CD8 (1:100;Abcam) and CD4 (1:100; Abcam). For observing the staining foreach antibody, a uniform setting was applied for all slides. CD68-orCSF-1R-positive areas in thephotographsweremeasuredby theLeicaQwin Plus, and themacrophage density in each photographwas calculated as CD68-positive area/total area. The positivestaining area valuesweredetermined asdescribedpreviously (13).

Quantitative real-time PCR analysisTotal RNA from L-02, hepatocellular carcinoma cell lines

(MHCC97-H, MHCC97-L, HCCLM3, and HepG2) and Hepa1-6, mouse M(CSF1), M(CSF2), M(LPSþIFNg), and M(IL4) cellswas extracted and reverse-transcribed onto single-stranded cDNAusing the iScript cDNA Synthesis Kit (Bio-Rad Laboratories). ForqPCR, primers were designed by Sangon Biotech, and theirefficiency was tested on a genomic DNA dilution series. qPCRwas performed with the Applied Biosystems 8100 HT SequenceDetection System (Applied Biosystems). Expression of the glyc-eraldehyde-3 phosphate dehydrogenase gene was used to nor-malize the expression of each gene. The primer sequences used todetermine the expression of the target genes are listed in Supple-mentary Tables S1 and S2.

Western blot analysisCells were lysed with RIPA Lysis Buffer (Santa Cruz Biotech-

nology) containing protease inhibitors (Beyotime Institute ofBiotechnology). The protein concentration was determined usinga bicinchoninic acid assay (Beyotime Institute of Biotechnology)and equalized before loading. Aliquots of 25 to 50 mg of proteinwere separated by SDS-PAGE and transferred onto polyvinylidenedifluoridemembranes (Millipore). Membranes were blocked andblotted with the relevant antibodies. Horseradish peroxidase–conjugated secondary antibodies were detected with an enhancedchemiluminescence reagent (Beyotime). Antibodies against phos-phorylated CSF-1R (Cell Signaling Technology, CST), CSF-1R(CST), and CSF2rb (Abcam) were used to determine their expres-sion. GAPDH was used as a loading control. All antibody dilu-tions were 1:1,000, except for the GAPDH antibody, which wasused at a dilution of 1:5,000.

Cell proliferation assayAliquots of the cell suspension were inoculated into a 96-well

plate (3� 103 to 5� 103 cells in 100 mL/well) and then incubatedin a humidified incubator (37�C, 5% CO2). Ten microliters ofCCK-8 solution (Dojindo Laboratories, Kumamoto, Japan) wasadded to each well, and the plate was the incubated for 2 to 4hours (37�C, 5%CO2). Absorbance at 450 nmwasmeasured by amicroplate reader.

Animal studiesMale C57BL/6 mice (6 weeks old) and male BALB/c mice (6

weeks old) were obtained from the Shanghai Institute of MateriaMedica, Chinese Academy of Science, and housed under specificpathogen-free conditions. The experimental protocol wasapproved by the Shanghai Medical Experimental Animal CareCommission.

Hepa1-6 cells (6 � 106 cells) in 200 mL of normal saline wereimplanted by subcutaneous injection to obtain subcutaneoustumors. TwentyC57BL/6micewere treated byorthotopic implan-tation of 1 mm3 tumor into the liver from a subcutaneouslygrowing one. Three days after implantation, the mice were ran-domized into two PLX3397-treated groups (50 mg/kg/day, oraladministration) or two vehicle-treated groups. Each group includ-ed fivemice. Themouse body weights were measured every week.One PLX3397-treated group and one vehicle-treated group werecontinuously observed for survival analysis. In the other twogroups, tumors were resected after 5 weeks to obtain TAMs; theremaining tumor tissues were stored in 4% paraformaldehydesolution for further study. Tumor volume was calculated accord-ing to the following formula: tumor volume¼ (largest diameter�perpendicular height2)/2.

HepG2 cells (5 � 105 cells) or HCCLM3 (5 � 105 cells) in200 mL of normal saline were implanted by subcutaneous injec-tion to obtain subcutaneous tumors. Nude mice were treated byorthotopic implantation of 1 mm3 tumor into the liver from asubcutaneously growing one. Twelve Balb/c nude mice withorthotopic HepG2 or 10 Balb/c nude mice with orthotopicHCCLM3 tumors were randomized into PLX3397-treated andvehicle-treated groups. Threedays after implantation, PLX339750mg/kg/day or vehicle solution was orally administered by gavage.Body weights were measured every week. Tumors were removedafter 5 weeks and stored in a 4% paraformaldehyde solution.

Molecular characteristics of macrophagesTHP-1 derived macrophages, including M(LPSþINFg) and M

(IL4) cells, were processed in Trizol and then analyzed using theAffymetrix U133 Array platform. M(CSF1) cells, M(CSF2) cells,and TAMs from mouse tumors were processed in Trizol within 4hours after sorting and then analyzedusingwhole-mouse genomeAffymetrix Mouse Gene 1.0 ST Arrays. Data files are available atthe GEO database https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc¼GSE95407. The M1 macrophage-related genes for MHCII (HLA-DRA), TNF, IL1B, IL12B, IL23A, CD80, CD86, IL6, andCXCL10 and theM2macrophage-related genes forCCL17,MRC1,IL10, and IL4Rwere used to characterizemacrophage polarization(19). Hierarchical clustering analysis using the aforementionedpanel of genes was performed using MeV v4.6.0 software (http://www.tm4.org/; TM4). The matrix was presented graphically bycoloring each gene expression on the basis of a measured colorrange: lower limit "�6" was blue, upper limit "6" was red, andmidpoint value "0" was white. A hierarchical clustering algorithm

Ao et al.

Mol Cancer Ther; 16(8) August 2017 Molecular Cancer Therapeutics1546

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 4: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

usually required two main steps, which were repeated to find thestrains that were the most similar: the M1/M2-associated generesult values from the same strain were assigned to its own cluster,and the two clusters that were closest to each other were mergeduntil only one large cluster resulted.

Reagents and antibodiesOther reagents and antibodies used in this study are shown in

Supplementary Table S3.

Statistical analysisStatistical analysis was performed with SPSS for Windows

(version 20.0; SPSS Inc.). Quantitative variables were analyzedby the independent samples t test. Overall survival and time torecurrence were assessed using the Kaplan–Meier method andcompared with the log-rank test. Statistical significance wasdefined by P < 0.05. Western blots were analyzed with ImageJ(NIH, Bethesda, MD). Cell migration was analyzed by Image-ProPlus as described previously (39). Pearson correlationwas used asa distance metric and the complete linkage method was used inhierarchical clustering.

ResultsBone marrow–derived monocytes were polarized toward theM2-like or M1-like phenotype by CSF1 or CSF2 stimulation

M(CSF1) cells (CSF1-induced BMDMs) had a spindle appear-ance with a colony growth pattern, whereas M(CSF2) cells (CSF2-

induced BMDMs) had a more rounded appearance with a patternof individual growth (Supplementary Fig. S1A and S1B). FACSassay showed that more than 95% of M(CSF1) and M(CSF2)cells were positive for F4/80 (Supplementary Fig. S1). Immuno-cytofluorescence assay further confirmed that M(CSF1) and M(CSF2) cells were positive for the markers specific to macrophag-es, including CD68, F4/80, and CSF-1R (Fig. 1A and B). CSF2Raand CSF-2Rb expression was found on M(CSF1) and M(CSF2)cells by immunocytochemistry assay (Fig. 1C and D). However,BMDMs without CSF-1 stimulation were not positive for CD68,F4/80, and CSF-1R and were suspended in the culture medium.

To establishmolecular signatures shared by human andmousemacrophages, we conducted a whole-genome expression profil-ing assay (40, 41), which identified differentially expressed genesin M1-polarized macrophages (THP-1–derived macrophagesstimulated by LPSþIFNg) and M2-polarized macrophages (stim-ulated by IL4). Of these, expression of M1-associated genes suchas HLA-DRA, TNF, IL1B, IL12B, IL23A, CD80, CD86, IL6, andCXCL10 was higher, whereas expression of M2-associated genessuch as CCL17, MRC1, IL10, and IL4RAwas lower in comparisonwith M(LPSþIFNg) and M(IL4) cells, which is consistent withprevious reports (11, 19, 42).

Whole mouse genome expression profiling identified 1,631genes that were upregulated and 2,539 downregulated genes inM(CSF2) cells comparedwithM(CSF1) cells.Of these, expression ofM1-polarized macrophage-related genes such as MHC-II, Tnf,Il1b, Il12b, Il23a, Cd80, Cd86, Il6, and Cxcl10 was increased and

Figure 1.

Expression of a series of macrophage markers were found in CSF1- and CSF2-induced BMDMs. Confocal laser scanning microscopy images of CSF1-inducedBMDMs [M(CSF1) cells] and CSF2-induced BMDMs [M(CSF2) cells]. F4/80 (green), CSF-1R (green), and CD68 (red) were coexpressed in M(CSF1) cells (A) and M(CSF2) cells (B). CSF2Ra (green), CSF2Rb (green), and CD68 (red) were also coexpressed in M(CSF1) cells (C) and M(CSF2) cells (D). Scale bar ¼ 100 mm.

CSF1R Blockade and Macrophage Polarization

www.aacrjournals.org Mol Cancer Ther; 16(8) August 2017 1547

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 5: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

expression of M2-polarized macrophage-related genes such asCcl17, Mrc1, Il10, and Il4ra was decreased in the M(CSF2) cells,compared with their counterparts, suggesting that M(CSF2) cellswere more likely to be M1-polarized macrophages, whereas M(CSF1) cells were more likely to be M2-polarized macrophages.The expression of the typical markers for M1- or M2-polarizedmacrophages was validated by qPCR to confirm the gene expres-sion levels (Supplementary Fig. S2A and S2B).

PLX3397 suppressed tumor growth without depletion of TAMinfiltration in vivo

In vitro, a population doubling assay showed that the IC50 ofPLX3397 (ref. 33; Fig. 2A), a highly selective CSF-1R inhibitor,for M(CSF1) cells was 22 nmol/L (Fig. 2B), which is consistentwith a previous report (43). A similar inhibitory effect of exog-enous CSF-1R antibody onM(CSF1) cells was observed (Fig. 2C).Marked suppression of CSF-1R phosphorylation (p-CSF-1R) inM(CSF1) cells could be achieved with PLX3397 (Fig. 2D). In

contrast, the same doses of PLX3397 showed no antiproliferationeffect on Hepa1-6, MHCC97-H, HCCLM3, HepG2, HUVEC,T cells, fibroblasts cells, and M(CSF2) cells (SupplementaryFig. S3A–S3G). In accordance with these results, very low CSF-1R expression was detected in hepatocyte (L-02), hepatoma(MHCC97-H, MHCC97-L, HCCLM3, Hepa1-6, and HepG2), orendothelial cell lines (HUVEC) as comparedwithM(CSF1) andM(CSF2) cells (Supplementary Fig. S3H).

In an orthotopic C57BL/6 model with Hepa1-6 tumor cells,PLX3397 treatment suppressed tumor growth by 70% withoutaffecting mouse body weight compared to the vehicle-treatedmice (Fig. 3A–C). PLX3397 treatment also prolonged survival ofthe tumor-bearing mice (median survival time, 8.0 weeks versus11.3 weeks; P ¼ 0.026; Fig. 3D).

We next examined the key mechanism of the antitumor effectsof PLX3397. The number of CD68-positive or CSF-1R-positivemacrophages in the tumors from the PLX3397-treated and vehi-cle-treatedmicewere not statistically different (Fig. 4A andB). The

Figure 2.

CSF-1R blockade led toCSF1-inducedBMDM[M(CSF1) cells] death by phosphorylated CSF-1R inhibition.A,Chemical structure of PLX3397. PLX3397 is an oral, potentreceptor tyrosine kinase inhibitor of CSF-1R. Molecular weight: 417.81. Chemical formula: C20H15ClF3N5. B, A population doubling assay showed that IC50

concentration of PLX3397 inM(CSF1) cells was 22 nmol/L.C,Culturemediumwith different conditionswere performed to determineM(CSF1) survival. A neutralizingantibody against CSF-1R (10 mg/mL, GeneTex, AFS98) led to M(CSF1) cell death in DMEM containing 10% FBS and supplemented with 10 ng/mL recombinant mouseCSF1 in a similar manner with 22 nmol/L, 220 nmol/L PLX3397 and the culturemediumwithout CSF1, compare withM(CSF1) culture in DMEM containing 10% FBS andsupplemented with 10 ng/mL CSF1, as determined by CCK8 assays (n ¼ 3 replicates, � , P < 0.01). D, Western blot analysis of M(CSF1) cells, which werecultured in medium without CSF-1 for 12 hours before stimulation then followed CSF-1 addition for the time points indicated (1.5, 3, and 5 minutes). A progressiveincrease of phosphorylated CSF-1R expression resulted, whichwas effectively inhibited by 22 nmol/L PLX3397; CSF-1R expressionwas not changed. In lane 1,markedby #, M(CSF1) cells were continuously cultured with CSF-1. GAPDH was used as a loading control.

Ao et al.

Mol Cancer Ther; 16(8) August 2017 Molecular Cancer Therapeutics1548

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 6: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

number of cells with CD31 (P ¼ 0.589) or a-Smooth MuscleActin (SMA) (P ¼ 0.913) expression did not differ betweenthe two groups (Supplementary Fig. S4A–S4F), suggesting thatPLX3397 may have no effect on the infiltration of macrophagesor tumor angiogenesis in vivo.

Tumor-derived CSF-2 promoted TAM polarization toward anM1-like phenotype under treatment with PLX3397

As PLX3397 treatment didNOTdeplete TAM infiltration in vivo,whether the phenotype of TAMs changed or cytokines protectTAM from depletion in the microenvironment was further inves-tigated. To study whether the phenotype of TAMs was affected byPLX3397, we conducted gene expression profiling on the isolatedTAMs. We found 4,342 upregulated genes and 4,570 downregu-lated genes in the PLX3397-treated TAMs compared with thevehicle-treated TAM. The expression profile in TAMs isolated fromthe PLX3397-treated tumors was similar to that of M1-polarizedmacrophages, whereas the gene expression profile of TAMs fromthe vehicle-treated tumors was similar to that of M2-polorizedmacrophages (Supplementary Fig. S5A, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc¼GSE95407). The expression of

the typical markers for M1- or M2-polarized macrophages werevalidated by qPCR (Supplementary Fig. S5B).

An immunocytofluorescence assay was conducted to exam-ine the expression of Mrc1, MHC-II, and CD68 in the PLX3397-or vehicle-treated tumors. The results showed increased expres-sion of MHC-II-positive macrophages and decreased expres-sion of Mrc1-positive macrophages (Supplementary Fig. S5Cand S5D). Taken together, these results indicated that PLX3397promoted a shift in polarization of macrophages from M2to M1.

Because TAMs from the PLX3397-treated tumors showed anM1-like phenotype, we hypothesized that this transition waspromoted by the cytokines secreted in the tumor microenviron-ment. The isolated macrophages from the PLX3397- and vehicle-treated tumors were not different in proliferation (Fig. 5A). Theremaining nonmacrophage cells from the tumor tissue werecultured for 48 hours, and the conditioned medium (CM) wascollected. The CMof nonmacrophage cells from PLX3397-treatedmice, but not from the vehicle-treated mice, protected the M(CSF1) cells from the antiproliferative effects of PLX3397 in vitro(Fig. 5B). These results indicated that some cytokines from the

Figure 3.

CSF-1R inhibition restricted tumor growth and prolonged mouse survival in an allograft C57BL/6 mouse model with orthotopic implanted hepa1-6 cells. A, After5-week treatment with PLX3397 or vehicle, the mice were killed and the liver tissue was obtained. B, The mean tumor volume in the PLX3397-treated group wassignificantly lower than in the vehicle-treated group (P ¼ 0.045). C, Mice treated with PLX3397 had a similar body weight compared with those in thecontrol group at 5weeks (P¼0.712).D, The survival timeswere compared between themice continuously treatedwith PLX3397 and those that received the vehicle.The cumulative survival plots showed that PLX3397 significantly prolonged the survival timeof the tumor-bearingmice (P¼0.026). Themice of treatedPLX3397 andvehicle group were die through cancer cachexia and no lung, peritoneal cavity metastasis was found.

CSF1R Blockade and Macrophage Polarization

www.aacrjournals.org Mol Cancer Ther; 16(8) August 2017 1549

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 7: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

intratumoral microenvironment protected TAMs from the anti-proliferative effects of PLX3397.

An antibody array (AAM-ANG-1; Raybiotech) was conductedto identify the differentially expressed cytokines in the CM ofnonmacrophage cells from PLX3397-treated and vehicle-treatedtumors. The results showed that the expression of CSF2, granu-locyte CSF 3 (CSF3), IL3 (multi-CSF), and IFNg were higher in theCM of nonmacrophages from the PLX3397-treated mice thanfrom the vehicle-treated mice (Fig. 5C and D). Furthermore, apopulation doubling assay showed that CSF2, IL3, and IFNgprotected M(CSF1) cells from the inhibitory effects of PLX3397.Among these cytokines, CSF2 was the most potent (Fig. 5E–H).These results suggested that CSF2 signals could be an importantsurvival factor for macrophages under a CSF-1R blockade. Inaccordance with these results, PLX3397 did not inhibit M(CSF2)cells in a population doubling assay at concentrations of 22nmol/L and 50,000 nmol/L (Supplementary Fig. S6A); however, IC50 ofPLX3397 for M(CSF1) cells in medium containing 10 ng/mLCSF1 was 22 nmol/L, whereas IC50 of PLX3397 for M(CSF1) cellsin medium containing 10 ng/mL CSF2 was 19,495 nmol/L(Supplementary Fig. S6B and S6C), suggesting that CSF2 pro-tected macrophages from PLX3397 treatment.

PLX3397 treatment changed the intratumoralmicroenvironment

We also studied the effects of PLX3397 on immune cells inthe mouse model by immunostaining the antigen presentingmacrophages (F4/80þMHC IIþ), myeloid-derived suppressor

cells (MDSCs, CD11bþGr1þ) cells, and the CD8þ and CD4þ Tcells. We found that the proportion of antigen presentingmacrophages was elevated (P ¼ 0.041), whereas myeloid-derived suppressor cells were decreased (P ¼ 0.029) in thetumor tissues (Supplementary Fig. S7A and S7B). Furthermore,PLX3397 treatment increased the number of CD8þ cell (P ¼0.023) and decreased the number of CD4þ cells in tumorscompared to the vehicle treatment (P ¼ 0.002; SupplementaryFig. S7C and S7D). Furthermore, as mentioned above, wefound that PLX3397 did not affect the proliferation of fibro-blasts and T cells directly in vitro (Supplementary Fig. S3H andS3J–S3M).

PLX3397 treatment inhibited tumor growth in two xenograftmodels of hepatocellular carcinoma

In xenograft models derived from two human hepatoma celllines, HepG2 and HCCLM3, PLX3397 treatment suppressedtumor growth by 33% and 84%, respectively (P ¼ 0.026 and0.047, respectively; Supplementary Fig. S8A–S8D). In addition,there was no difference in CD68þ pan-macrophages, but therewas an increased expression ofM1-associatedmarkerMHC-II anda decreased expression of M2-associated marker MRC1 in thePLX3397-treated tumors compared to the vehicle-treated tumors(Supplementary Fig. S9).

DiscussionThis study showed that PLX3397 treatment induced the

transition of M2-polarized macrophages to M1-polarized

Figure 4.

PLX3397 suppressed tumor growthwithout depletion of TAM infiltrationbut promoted TAM polarization towardan M1-like phenotype. In the allograftmouse models, the densities of CD68-and CSF-1R-positive cells in the tumorstreated with PLX3397 or vehicle werenot significantly different (P ¼ 0.915and 0.842, respectively) (A and B).

Ao et al.

Mol Cancer Ther; 16(8) August 2017 Molecular Cancer Therapeutics1550

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 8: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

macrophages in tumors, which was mediated by inhibition ofCSF-1R on TAMs, delayed tumor growth, and prolonged sur-vival of tumor-bearing mice.

Our findings support the idea that M(CSF2) cells are similar toM1-macrophages, whereas M(CSF1) cells are similar to M2-macrophages, which is consistent with a previous report (25).Other authors have indicated that significant differences in tran-scriptome level remained in M(CSF1) cells and M2 macrophages(19, 21, 44); however, the transcriptome data from the presentstudy showed that the gene expression signature determined by apanel of genes in M(CSF1) cells was similar to that of M2macrophages, and that M(CSF2) cells was similar to M1 macro-phages. Using the same signature, we showed that TAMs from thePLX3397-treated tumors were similar to the M(CSF2) cells andTAMs from theuntreated tumorwere similar to theM(CSF1) cells.We also showed that the M2-polarized macrophages were trans-formed to M1-polarized macrophages. These results are in agree-ment with previous data (40, 41).

Several studies have suggested that CSF1 is the major chemoat-tractant in cancers, attracting TAMs to the neoplastic microenvi-ronment and differentiating them to protumorigenic types(13, 45, 46). In this study, we demonstrated that blockade ofCSF-1R signaling in TAMs affects tumor progression in multiplehepatoma models by polarization of TAMs toward the M1 phe-notype in animal hepatomamodels. TAMswere not depleted, andthey survived CSF-1R inhibitor treatment, which was consistentwith findings showing that M(CSF1) cells could survive in CSF2-containing medium with CSF-1R inhibition. The cell shape andsensitivity to the CSF-1R inhibitor also changed in line withadding CSF2 to the medium. One interesting finding was thatexpression of CSF2, IL3, and IFNg in the tumor microenviron-

ment was increased when PLX3397 treatment was applied totumors. In this study, we did not attempt to identify the source ofCSF2 in the tumor microenvironment. Other authors havereported similar findings (25, 47) supporting other survivalfactors that were increased when the tumor was treated withCSF-1R inhibitors. The authors suggested that tumor cells mightbe a source of CSF2 that was affected by CSF-1R inhibitortreatment. Notably, Swierczak and colleagues have reported thatinhibition of CSF-1R/CSF-1 signaling by AFS-98 (a CSF-1R anti-body) increased the granulocyte CSF (G-CSF; CSF3) level in theserum of mice with breast tumors (47) and was associated withincreased metastasis of mammary tumors in the animal model. Itwas also reported that increased CSF2 promoted tumor growththrough paracrine action on stromal cells in skin carcinoma (48)and promoted epithelial–mesenchymal transition andmetastasisin breast cancer (49). Furthermore, Ries and colleagues reportedRG7155 (aCSF-1R antibody) depletesmacrophages in vitro and invivo (50). These discrepancies may indicate that the CSF1/CSF-1Rsystem may play different roles in different types of cancers. Arecent study demonstrated a collaborative interaction betweenmacrophage and stromal cells (fibroblasts) in forming a favorableenvironment for neuroblastoma development (51). Although wehave showed that fibroblasts were not directly affected byPLX3397 in the present study, it would be interesting to exploreif the effect of PLX3397 on tumor growth is partially mediated byfibroblasts or other mesenchymal stromal cells (52), which maybe affected by polarized macrophages.

PLX3397 treatment results in more antigen presenting macro-phages and CD8þ cells and fewer MDSCs and CD4þ cells intumors. These findings suggested antitumor immunity in themicroenvironment has been ameliorated by CSF-1R inhibition,

Figure 5.

Tumor-derived CSF-2 promoted TAM polarization toward an M1-like phenotype under the treatment of PLX3397. A, A population doubling assay found that TAMsfrom PLX3397 and vehicle treatments had no difference in proliferation. B, The remaining non-macrophage cells from the tumor tissue were cultured for 48 hours,and the CM was collected. A population doubling assay found that PLX3397-treated non-TAM CM could support M(CSF1) cell survival with 22 nmol/LPLX3397, whereas vehicle non-TAMCM could not support M(CSF1) cell survival with 22 nmol/L PLX3397.C andD,Antibody array identified that CSF2, CSF3, IL3, andIFNg were higher in PLX3397-treated non-TAMCM than the vehicle group.E–H,Apopulation doubling assay found that 10 ng/mLCSF2, 10 ng/mL IFNg , and 10 ng/mLIL3 could support M(CSF1) cell survival with 22 nmol/L PLX3397. In these protective factors for M(CSF1) cells treated with CSF-1R inhibitor, CSF2 was themost potent among them.

CSF1R Blockade and Macrophage Polarization

www.aacrjournals.org Mol Cancer Ther; 16(8) August 2017 1551

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 9: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

which is consistent with depletion of TAMs come out with moreactivated CTLs (53).

In summary, we found that treatment with CSF-1R inhibitorPLX3397 delayed tumor growth in both xenograft and allograftmodels, and this delay was probably mediated by the transitionfrom M2 macrophages to M1 macrophages in a TAM populationinduced by blockade of the CSF1/CSF-1R signal pathway (Fig. 6).It would be interesting to investigate the effects of the CSF-1Rinhibitor on hepatocellular carcinoma patients because systemictreatment for hepatocellular carcinoma is not yet sufficient.

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

Authors' ContributionsConception and design: J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, L.-Q. Kong, H.-C. SunDevelopment of methodology: J.-Y. Ao, Z.-T. Chai, H.-C. SunAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J.-Y. Ao, X.-D. Zhu, H. Cai, N. Zhang, B.-G. Ye,D.-N. MaAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J.-Y. Ao, X.-D. Zhu, H. Cai, H.-C. Sun

Writing, review, and/or revision of the manuscript: J.-Y. Ao, X.-D. Zhu,Z.-T. Chai, H.-C. SunAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): J.-Y. Ao, Y.-Y. Zhang, K.-Z. Zhang, H.-C. SunStudy supervision: H.-C. Sun

AcknowledgmentsWe thank Gideon E. Bollag, PhD, and Parmveer Singh from Plexxikon Inc.,

for supplying the PLX3397 compound and scientific expertise.

Grant SupportThis studywas jointly supported by grants from theNational Natural Science

Foundation of China (No. 81372655, to X.D. Zhu and No. 81472224 and81672326 to H.C. Sun) and the Leading Investigator Program of Shanghaimunicipal government (17XD1401100 to H.C. Sun), the National Key BasicResearch Program (973 project) (2015CB554005 to H.C. Sun) from theMinistry of Science and Technology of China.

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

ReceivedDecember 14, 2016; revisedMarch 23, 2017; acceptedMay19, 2017;published OnlineFirst June 1, 2017.

References1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer

statistics, 2012. CA Cancer J Clin 2015;65:87–108.2. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860–7.3. Wan S, Kuo N, Kryczek I, Zou W, Welling TH. Myeloid cells in hepatocel-

lular carcinoma. Hepatology 2015;62:1304–12.4. Wang L, Fu H, Nanayakkara G, Li Y, Shao Y, Johnson C, et al. Novel

extracellular and nuclear caspase-1 and inflammasomes propagate inflam-

mation and regulate gene expression: a comprehensive database miningstudy. J Hematol Oncol 2016;9:122.

5. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflam-mation in the initiation and promotion of malignant disease. Cancer Cell2005;7:211–7.

6. Capece D, Fischietti M, Verzella D, Gaggiano A, Cicciarelli G, Tessitore A,et al. The inflammatory microenvironment in hepatocellular carcinoma: a

Figure 6.

Adiagrammatic illustration summarized the findings of this study. Both CSF1 andCSF2 can support the cell differentiation of BMDMs. However, BMDMs stimulated bythe CSF1/CSF-1R pathway (M(CSF1) cells) showed an M2-like phenotype (alternative activation), whereas BMDMs stimulated by CSF2/CSF2Ra and CSF2Rbpathway [M(CSF2) cells] showed an M1-like phenotype (classical activation). When the CSF1/CSF-1R signaling pathway was blocked, the CSF2/CSF-2R signalingpathway alternatively dominated the differentiation of M(CSF1) cells, and these cells showed an M1-like phenotype.

Ao et al.

Mol Cancer Ther; 16(8) August 2017 Molecular Cancer Therapeutics1552

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 10: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

pivotal role for tumor-associated macrophages. BioMed Res Int 2013;2013:187204.

7. Mantovani A, Germano G, Marchesi F, Locatelli M, Biswas SK. Cancer-promoting tumor-associatedmacrophages: new vistas and open questions.Eur J Immunol 2011;41:2522–5.

8. Allavena P, Mantovani A. Immunology in the clinic review series; focuson cancer: tumour-associated macrophages: undisputed stars of theinflammatory tumour microenvironment. Clin Exp Immunol 2012;167:195–205.

9. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progressionand metastasis. Cell 2010;141:39–51.

10. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms totherapy. Immunity 2014;41:49–61.

11. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polar-ization: tumor-associated macrophages as a paradigm for polarized M2mononuclear phagocytes. Trends Immunol 2002;23:549–55.

12. Gordon S. Alternative activation of macrophages. Nat Rev Immunol2003;3:23–35.

13. Zhu XD, Zhang JB, Zhuang PY, Zhu HG, Zhang W, Xiong YQ,et al. High expression of macrophage colony-stimulating factor inperitumoral liver tissue is associated with poor survival aftercurative resection of hepatocellular carcinoma. J Clin Oncol 2008;26:2707–16.

14. Jia JB,WangWQ, SunHC, Zhu XD, Liu L, Zhuang PY, et al. High expressionof macrophage colony-stimulating factor-1 receptor in peritumoral livertissue is associated with poor outcome in hepatocellular carcinoma aftercurative resection. Oncologist 2010;15:732–43.

15. Kong LQ, Zhu XD, Xu HX, Zhang JB, Lu L, Wang WQ, et al. The clinicalsignificance of the CD163þ and CD68þ macrophages in patients withhepatocellular carcinoma. PLoS One 2013;8:e59771.

16. Chai ZT, Zhu XD, Ao JY, Wang WQ, Gao DM, Kong J, et al. microRNA-26asuppresses recruitment of macrophages by down-regulating macrophagecolony-stimulating factor expression through the PI3K/Akt pathway inhepatocellular carcinoma. J Hematol Oncol 2015;8:56.

17. 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.

18. Galdiero MR, Bonavita E, Barajon I, Garlanda C, Mantovani A, Jaillon S.Tumor associated macrophages and neutrophils in cancer. Immunobiol-ogy 2013;218:1402–10.

19. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al.Macrophage activation and polarization: nomenclature and experimentalguidelines. Immunity 2014;41:14–20.

20. 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.

21. Joshi S, Singh AR, Zulcic M, Bao L, Messer K, Ideker T, et al. Rac2 controlstumor growth, metastasis and M1-M2macrophage differentiation in vivo.PLoS One 2014;9:e95893.

22. Zhang W, Zhu XD, Sun HC, Xiong YQ, Zhuang PY, Xu HX, et al. Depletionof tumor-associated macrophages enhances the effect of sorafenib inmetastatic liver cancermodels by antimetastatic and antiangiogenic effects.Clin Cancer Res 2010;16:3420–30.

23. Zhu XD, Sun HC, Xu HX, Kong LQ, Chai ZT, Lu L, et al. Antiangiogenictherapy promoted metastasis of hepatocellular carcinoma by suppressinghost-derived interleukin-12b in mouse models. Angiogenesis 2013;16:809–20.

24. Cassier PA, Gelderblom H, Stacchiotti S, Thomas D, Maki RG, Kroep JR,et al. Efficacy of imatinib mesylate for the treatment of locally advancedand/or metastatic tenosynovial giant cell tumor/pigmented villonodularsynovitis. Cancer 2012;118:1649–55.

25. 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.

26. Coniglio SJ, Eugenin E, Dobrenis K, Stanley ER, West BL, Symons MH,et al. Microglial stimulation of glioblastoma invasion involves epider-mal growth factor receptor (EGFR) and colony stimulating factor 1receptor (CSF-1R) signaling. Mol Med (Cambridge, Mass) 2012;18:519–27.

27. De I, Nikodemova M, Steffen MD, Sokn E, Maklakova VI, Watters JJ, et al.CSF1 overexpression has pleiotropic effects on microglia in vivo. Glia2014;62:1955–67.

28. He Y, Rhodes SD, Chen S, Wu X, Yuan J, Yang X, et al. c-Fms signalingmediates neurofibromatosis Type-1 osteoclast gain-in-functions. PLoSOne 2012;7:e46900.

29. 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.

30. Patwardhan PP, Surriga O, Beckman MJ, de Stanchina E, DematteoRP, Tap WD, et al. Sustained inhibition of receptor tyrosine kinasesand macrophage depletion by PLX3397 and rapamycin as a potentialnew approach for the treatment of MPNSTs. Clin Cancer Res2014;20:3146–58.

31. Prada CE, Jousma E, Rizvi TA, Wu J, Dunn RS, Mayes DA, et al. Neuro-fibroma-associated macrophages play roles in tumor growth and responseto pharmacological inhibition. Acta Neuropathol 2013;125:159–68.

32. SluijterM, van der Sluis TC, van der Velden PA, VersluisM,West BL, van derBurg SH, et al. Inhibition of CSF-1R supports T-cell mediated melanomatherapy. PLoS One 2014;9:e104230.

33. TapWD,Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C,Healey JH, et al.Structure-guided blockade of CSF1R kinase in tenosynovial giant-celltumor. N Engl J Med 2015;373:428–37.

34. Li Y, Tang Y, Ye L, Liu B, Liu K, Chen J, et al. Establishment of ahepatocellular carcinoma cell line with unique metastatic character-istics through in vivo selection and screening for metastasis-relatedgenes through cDNA microarray. J Cancer Res Clin Oncol 2003;129:43–51.

35. Li Y, Tian B, Yang J, Zhao L, Wu X, Ye SL, et al. Stepwise metastatichuman hepatocellular carcinoma cell model system with multiplemetastatic potentials established through consecutive in vivo selectionand studies on metastatic characteristics. J Cancer Res Clin Oncol2004;130:460–8.

36. Genin M, Clement F, Fattaccioli A, Raes M, Michiels C. M1 and M2macrophages derived from THP-1 cells differentially modulate theresponse of cancer cells to etoposide. BMC Cancer 2015;15:577.

37. Bain CC, Bravo-Blas A, Scott CL, Gomez Perdiguero E, Geissmann F, HenriS, et al. Constant replenishment from circulatingmonocytes maintains themacrophage pool in the intestine of adult mice. Nat Immunol 2014;15:929–37.

38. Ying W, Cheruku PS, Bazer FW, Safe SH, Zhou B. Investigation of macro-phage polarization using bone marrow derived macrophages. J Vis Exp2013;76:e50323.

39. Chai ZT, Kong J, Zhu XD, Zhang YY, Lu L, Zhou JM, et al. MicroRNA-26ainhibits angiogenesis by down-regulating VEGFA through the PIK3C2al-pha/Akt/HIF-1alpha pathway in hepatocellular carcinoma. PLoS One2013;8:e77957.

40. Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profilingof the human monocyte-to-macrophage differentiation and polarization:new molecules and patterns of gene expression. J Immunol 2006;177:7303–11.

41. Martinez FO, Helming L, Milde R, Varin A, Melgert BN, Draijer C, et al.Genetic programs expressed in resting and IL-4 alternatively activatedmouse and human macrophages: similarities and differences. Blood2013;121:e57–69.

42. Vogel DY, Vereyken EJ, Glim JE, Heijnen PD, Moeton M, van derValk P, et al. Macrophages in inflammatory multiple sclerosislesions have an intermediate activation status. J Neuroinflammation2013;10:35.

43. DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, MaddenSF, et al. Leukocyte complexity predicts breast cancer survival andfunctionally regulates response to chemotherapy. Cancer Discov2011;1:54–67.

44. Fleetwood AJ, Dinh H, Cook AD, Hertzog PJ, Hamilton JA. GM-CSF-and M-CSF-dependent macrophage phenotypes display differentialdependence on type I interferon signaling. J Leukoc Biol 2009;86:411–21.

45. Goswami S, Sahai E, Wyckoff JB, Cammer M, Cox D, Pixley FJ, et al.Macrophages promote the invasion of breast carcinoma cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop. Cancer Res2005;65:5278–83.

www.aacrjournals.org Mol Cancer Ther; 16(8) August 2017 1553

CSF1R Blockade and Macrophage Polarization

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 11: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

46. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, et al. A paracrineloop between tumor cells and macrophages is required for tumor cellmigration in mammary tumors. Cancer Res 2004;64:7022–9.

47. Swierczak A, Cook AD, Lenzo JC, Restall CM, Doherty JP, Anderson RL,et al. The promotion of breast cancer metastasis caused by inhibition ofCSF-1R/CSF-1 signaling is blocked by targeting the G-CSF receptor. CancerImmunol Res 2014;2:765–76.

48. Mueller MM, Peter W, Mappes M, Huelsen A, Steinbauer H, Boukamp P,et al. Tumor progression of skin carcinoma cells in vivo promoted by clonalselection, mutagenesis, and autocrine growth regulation by granulocytecolony-stimulating factor and granulocyte-macrophage colony-stimulat-ing factor. Am J Pathol 2001;159:1567–79.

49. Su S, Liu Q, Chen J, Chen J, Chen F, He C, et al. A positive feedback loopbetween mesenchymal-like cancer cells and macrophages is essential tobreast cancer metastasis. Cancer Cell 2014;25:605–20.

50. Ries CH, CannarileMA,Hoves S, Benz J,Wartha K, Runza V, et al. Targetingtumor-associated macrophages with anti-CSF-1R antibody reveals a strat-egy for cancer therapy. Cancer Cell 2014;25:846–59.

51. Hashimoto O, Yoshida M, Koma Y, Yanai T, Hasegawa D, Kosaka Y,et al. Collaboration of cancer-associated fibroblasts and tumour-asso-ciated macrophages for neuroblastoma development. J Pathol 2016;240:211–23.

52. Lou G, Song X, Yang F, Wu S, Wang J, Chen Z, et al. Exosomesderived from miR-122-modified adipose tissue-derived MSCs increasechemosensitivity of hepatocellular carcinoma. J Hematol Oncol 2015;8:122.

53. 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.

Mol Cancer Ther; 16(8) August 2017 Molecular Cancer Therapeutics1554

Ao et al.

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866

Page 12: Colony-Stimulating Factor 1 Receptor Blockade ...J.-Y. Ao, X.-D. Zhu, Z.-T. Chai, and H. Cai contributed equally to this article. Corresponding Author: Hui-Chuan Sun, Professor of

2017;16:1544-1554. Published OnlineFirst June 1, 2017.Mol Cancer Ther   Jian-Yang Ao, Xiao-Dong Zhu, Zong-Tao Chai, et al.   Macrophages in Hepatocellular CarcinomaGrowth by Altering the Polarization of Tumor-Associated Colony-Stimulating Factor 1 Receptor Blockade Inhibits Tumor

  Updated version

  10.1158/1535-7163.MCT-16-0866doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2017/06/01/1535-7163.MCT-16-0866.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/16/8/1544.full#ref-list-1

This article cites 53 articles, 13 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/16/8/1544.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mct.aacrjournals.org/content/16/8/1544To request permission to re-use all or part of this article, use this link

on September 13, 2020. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst June 1, 2017; DOI: 10.1158/1535-7163.MCT-16-0866


Recommended