CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105...

13
Microenvironment and Immunology CSF1R Signaling Blockade Stanches Tumor-Inltrating Myeloid Cells and Improves the Efcacy of Radiotherapy in Prostate Cancer Jingying Xu 1,5 , Jemima Escamilla 1,5 , Stephen Mok 1 , John David 1 , Saul Priceman 6 , Brian West 7 , Gideon Bollag 7 , William McBride 2 , and Lily Wu 1,3,4,5 Abstract Radiotherapy is used to treat many types of cancer, but many treated patients relapse with local tumor recurrence. Tumor-inltrating myeloid cells (TIM), including CD11b (ITGAM) þ F4/80 (EMR1) þ tumor- associated macrophages (TAM), and CD11b þ Gr-1 (LY6G) þ myeloid-derived suppressor cells (MDSC), respond to cancer-related stresses and play critical roles in promoting tumor angiogenesis, tissue remodeling, and immunosuppression. In this report, we used a prostate cancer model to investigate the effects of irradiation on TAMs and MDSCs in tumor-bearing animals. Unexpectedly, when primary tumor sites were irradiated, we observed a systemic increase of MDSCs in spleen, lung, lymph nodes, and peripheral blood. Cytokine analysis showed that the macrophage colony-stimulating factor CSF1 increased by two-fold in irradiated tumors. Enhanced macrophage migration induced by conditioned media from irradiated tumor cells was completely blocked by a selective inhibitor of CSF1R. These ndings were conrmed in patients with prostate cancer, where serum levels of CSF1 increased after radiotherapy. Mechanistic investigations revealed the recruitment of the DNA damage-induced kinase ABL1 into cell nuclei where it bound the CSF1 gene promoter and enhanced CSF1 gene transcription. When added to radiotherapy, a selective inhibitor of CSF1R suppressed tumor growth more effectively than irradiation alone. Our results highlight the importance of CSF1/CSF1R signaling in the recruitment of TIMs that can limit the efcacy of radiotherapy. Furthermore, they suggest that CSF1 inhibitors should be evaluated in clinical trials in combination with radiotherapy as a strategy to improve outcomes. Cancer Res; 73(9); 278294. Ó2013 AACR. Introduction Radiotherapy is one of the primary treatments for prostate cancer. Approximately 50% of patients are treated with radio- therapy either alone or in combination with other therapies (1). Data from Cancer of the Prostate Strategic Urologic Research Endeavor identied that 63% of patients experienced biochem- ical prostate-specic antigen (PSA) recurrence after radiother- apy (2). In fact, D'Amico and colleagues determined that a high rate of PSA velocity pretreatment is signicantly associated with a shorter time to not only PSA recurrence but also prostate cancer-specic mortality after radiotherapy (3), and most of the recurrence is local (4). Several studies have addressed the importance of hypoxia and the SDF-1/CXCR4 axis in promoting tumor regrowth after radiotherapy in brain tumor and breast cancer (5, 6). However, a better understand- ing of the mechanisms of tumor regrowth is needed to achieve increased local control by radiotherapy in prostate cancer and improve the cure rate of this disease. Solid tumors contain a signicant population of tumor- inltrating myeloid cells (TIM; ref. 7). TIMs are now recogniz- ed as important mediators of not only tumor progression and metastasis (8), but also therapeutic resistance (9, 10), through promoting angiogenesis and suppressing antitumor immune responses (11, 12). The protumorigenic role of "alternatively" activated macrophages has been well established (12). Recent- ly, another specic subtype of TIMs, namely myeloid-derived suppressor cells (MDSC), is receiving great attention in cancer research. MDSCs comprise a heterogeneous population of immature myeloid cells that originate in the bone marrow and are recruited to the tumor by a diverse array of cytokine and chemokine signals. Similar to tumor-associated macro- phages (TAM; ref. 8), MDSCs have been shown to generate an environment favorable for tumors by heightening immuno- suppression, angiogenesis, and invasion (1315). Various cell surface markers are used to identify TIM subsets: TAMs can be Authors' Afliations: Departments of 1 Molecular and Medical Pharma- cology, 2 Radiation Oncology, 3 Urology, and 4 Pediatrics; 5 Institute of Molecular Medicine, David Geffen School of Medicine, University of Cali- fornia Los Angeles, Los Angeles, California; 6 Department of Cancer Immu- notherapeutics and Tumor Immunology, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, California; and 7 Plexxikon Inc., Berkeley, California Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Lily Wu, Departments of Molecular & Medical Pharmacology, Urology, 33-118 CHS, UCLA School of Medicine, Los Angeles, CA 90095-1735. Phone: 310-794-4390; Fax: 310-825-6267; E-mail: [email protected]. doi: 10.1158/0008-5472.CAN-12-3981 Ó2013 American Association for Cancer Research. Cancer Research Cancer Res; 73(9) May 1, 2013 2782

Transcript of CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105...

Page 1: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

Microenvironment and Immunology

CSF1R Signaling Blockade Stanches Tumor-InfiltratingMyeloid Cells and Improves the Efficacy of Radiotherapy inProstate Cancer

Jingying Xu1,5, Jemima Escamilla1,5, Stephen Mok1, John David1, Saul Priceman6, Brian West7,Gideon Bollag7, William McBride2, and Lily Wu1,3,4,5

AbstractRadiotherapy is used to treat many types of cancer, but many treated patients relapse with local tumor

recurrence. Tumor-infiltrating myeloid cells (TIM), including CD11b (ITGAM)þF4/80 (EMR1)þ tumor-associated macrophages (TAM), and CD11bþGr-1 (LY6G)þ myeloid-derived suppressor cells (MDSC),respond to cancer-related stresses and play critical roles in promoting tumor angiogenesis, tissue remodeling,and immunosuppression. In this report, we used a prostate cancer model to investigate the effects ofirradiation on TAMs and MDSCs in tumor-bearing animals. Unexpectedly, when primary tumor sites wereirradiated, we observed a systemic increase of MDSCs in spleen, lung, lymph nodes, and peripheral blood.Cytokine analysis showed that the macrophage colony-stimulating factor CSF1 increased by two-fold inirradiated tumors. Enhanced macrophage migration induced by conditioned media from irradiated tumorcells was completely blocked by a selective inhibitor of CSF1R. These findings were confirmed in patients withprostate cancer, where serum levels of CSF1 increased after radiotherapy. Mechanistic investigations revealedthe recruitment of the DNA damage-induced kinase ABL1 into cell nuclei where it bound the CSF1 genepromoter and enhanced CSF1 gene transcription. When added to radiotherapy, a selective inhibitor of CSF1Rsuppressed tumor growth more effectively than irradiation alone. Our results highlight the importance ofCSF1/CSF1R signaling in the recruitment of TIMs that can limit the efficacy of radiotherapy. Furthermore,they suggest that CSF1 inhibitors should be evaluated in clinical trials in combination with radiotherapy as astrategy to improve outcomes. Cancer Res; 73(9); 2782–94. �2013 AACR.

IntroductionRadiotherapy is one of the primary treatments for prostate

cancer. Approximately 50% of patients are treated with radio-therapy either alone or in combinationwith other therapies (1).Data from Cancer of the Prostate Strategic Urologic ResearchEndeavor identified that 63% of patients experienced biochem-ical prostate-specific antigen (PSA) recurrence after radiother-apy (2). In fact, D'Amico and colleagues determined that a highrate of PSA velocity pretreatment is significantly associatedwith a shorter time to not only PSA recurrence but also

prostate cancer-specific mortality after radiotherapy (3), andmost of the recurrence is local (4). Several studies haveaddressed the importance of hypoxia and the SDF-1/CXCR4axis in promoting tumor regrowth after radiotherapy in braintumor and breast cancer (5, 6). However, a better understand-ing of the mechanisms of tumor regrowth is needed to achieveincreased local control by radiotherapy in prostate cancer andimprove the cure rate of this disease.

Solid tumors contain a significant population of tumor-infiltrating myeloid cells (TIM; ref. 7). TIMs are now recogniz-ed as important mediators of not only tumor progression andmetastasis (8), but also therapeutic resistance (9, 10), throughpromoting angiogenesis and suppressing antitumor immuneresponses (11, 12). The protumorigenic role of "alternatively"activated macrophages has been well established (12). Recent-ly, another specific subtype of TIMs, namely myeloid-derivedsuppressor cells (MDSC), is receiving great attention in cancerresearch. MDSCs comprise a heterogeneous population ofimmature myeloid cells that originate in the bone marrowand are recruited to the tumor by a diverse array of cytokineand chemokine signals. Similar to tumor-associated macro-phages (TAM; ref. 8), MDSCs have been shown to generate anenvironment favorable for tumors by heightening immuno-suppression, angiogenesis, and invasion (13–15). Various cellsurface markers are used to identify TIM subsets: TAMs can be

Authors' Affiliations: Departments of 1Molecular and Medical Pharma-cology, 2Radiation Oncology, 3Urology, and 4Pediatrics; 5Institute ofMolecular Medicine, David Geffen School of Medicine, University of Cali-fornia Los Angeles, Los Angeles, California; 6Department of Cancer Immu-notherapeutics and Tumor Immunology, Beckman Research Institute andCity of Hope Comprehensive Cancer Center, Duarte, California; and7Plexxikon Inc., Berkeley, California

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

Corresponding Author: Lily Wu, Departments of Molecular & MedicalPharmacology, Urology, 33-118 CHS, UCLA School of Medicine, LosAngeles, CA 90095-1735. Phone: 310-794-4390; Fax: 310-825-6267;E-mail: [email protected].

doi: 10.1158/0008-5472.CAN-12-3981

�2013 American Association for Cancer Research.

CancerResearch

Cancer Res; 73(9) May 1, 20132782

Page 2: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

identified by CD11b and F4/80, andMDSCs by CD11b and Gr-1coexpression in murine models (11, 16). Macrophage colony-stimulating factor (M-CSF or CSF1) is a potent growth factorthat promotes the differentiation, proliferation, and migrationof monocytes/macrophages via signaling through its receptortyrosine kinase CSF1R (cFMS; refs. 17, 18). We recently showedthat TAMs and MDSCs form a spectrum of bone marrow-derived myeloid cells dependent on CSF1/CSF1R signaling forrecruitment into the tumor and that they play critical roles intumor growth (15). In addition, DeNardo and colleagueshighlighted the importance of CSF1/CSF1R signaling in therecruitment of TAMs in breast cancer and further showed thatCSF1R blockade can inhibit TAMs in chemotherapy andimprove treatment outcome (19).ABL1 (c-Abl) is an ubiquitously expressed nonreceptor

tyrosine kinase that has been implicated in many cellularprocesses including cell migration, differentiation, apoptosis,and gene regulation (20–22). ABL1 has also been implicated inthe proliferation and metastasis of melanoma and breastcancer cells (23–25). In the present study, we show that theinfiltration of TIMs is significantly enhanced by local irradia-tion of prostate cancer. In addition, CSF1 mRNA and CSF1secretion is increased following radiotherapy through anABL1-dependent mechanism. We further showed that block-ade of CSF1/CSF1R signaling effectively reduces TIMs infiltra-tion to tumors, thereby achieving more effective tumor growthsuppression after irradiation. The rational combination ther-apy reported here may provide a more effective and durabletreatment strategy for patients with prostate cancer.

Materials and MethodsCell cultureMurine macrophage RAW264.7 cells (American Type Cul-

ture Collection), rasþmyc-transformed RM-1, and RM-9 pros-tate tumor cells (kind gifts from Dr. Timothy C. Thompson,Baylor College ofMedicine, Houston, TX), human glioblastomacell lines U87 and U251 [a kind gift from Dr. Paul Mischel,University of California Los Angeles (UCLA), Los Angeles, CA],human breast cancer cell lineMDA-MB-283, andmousemalig-nant peripheral nerve sheath tumor cells (MMPNST, a kind giftfrom Dr. Hong Wu, UCLA) were cultured in Dulbecco's Mod-ified Eagle's Medium (DMEM) containing 10% FBS, 100 U/mLpenicillin, and 15mmol/L HEPES at 37�Cwith 5% CO2. Humanprostate cancer cell lines CWR and LNCaP, and the humancarcinoma cell line A549 were cultured in RPMI-1640 mediumcontaining 10% FBS, 100 U/mL penicillin, and 15 mmol/LHEPES at 37�C with 5% CO2. Cell lines were periodicallyauthenticated by morphologic inspection and tested negativefor mycoplasma contamination by PCR tests.

Chromatin precipitationTreated Myc-CaP cells were cross-linked with 1% formal-

dehyde at room temperature for 15 minutes. The cells werethen washed with PBS and processed according to the man-ufacturer's instruction using Pierce Agarose ChIP Kit (ThermoScientific). c-Abl antibody K-12 (Santa Cruz Biotechnology)and RNA polymerase II (Santa Cruz Biotechnology) were used

for immunoprecipitation. The following primers were used fordetecting CSF1 promoter sequences: Forward: 50ATGTGTCAGTGCCTGTGAGTGTGT30, Reverse: 50GCCAGGGTGATTTCC-CATAAACCA 30; CSF1 control sequences: Forward, 50TGCAA-GAAGCACCCATGAAATGGC30, Reverse: 50ATGCCAAAGCCT-GCAGTTAAACCC30.

Human serum assessmentSera, collected before and after radiotherapy, from human

prostate cancer patients were obtained from by the Depart-ment of Radiation Oncology, UCLA Medical Center Hospital,with informed consent according to US federal law and areexempt from consideration by the UCLA Administrative Panelon Human Subjects in Medical Research. Analysis of CSF1 wasdone by Eve Technologies using the human CSF1 multiplex kit(Bio-Rad).

In vivo tumor modelsC57BL6 male mice (4 to 8 weeks old) were purchased

from Jackson Laboratory (Bar Harbor). RM-1 (2.5 � 105

cells), RM-9 (2.5 � 105 cells), or Myc-CaP (2 � 106 cells)were implanted subcutaneously in the thigh, and treatmentwas initiated when tumors reached 4 mm in diameter. Allanimal experiments were approved by the UCLA Institu-tional Animal Care and Use Committee and conformed toall local and national animal care guidelines and regula-tions. Tumor size was measured by digital calipers daily orevery 2 days, depending on the model. Mice were sacrificedand tissues were analyzed at the ethical tumor size limit of1.5 cm in diameter. For GW2580 treatment, mice weretreated with control diluent (0.5% hydroxypropyl methylcel-lulose; Sigma-Aldrich; 0.1% Tween20 in distilled H2O) orGW2580 (160 mg/kg) by oral gavage beginning on the sameday irradiation treatment started. PLX3397 was provided infood chow together with daily food consumption.

In vitro migration assayRAW264.7 (1.5� 105 cells) were seeded in cell culture inserts

(8 mm pore size; BD Falcon) in DMEM containing 0.1% FBSwith or without 1,000 nmol/L GW2580. Inserts were placed in24-well plates with tumor-conditioned media collected 48hours after irradiation treatment (3 Gy). After 6 hours, migrat-ed cells were immediately fixed in 3% formaldehyde andstained with 4,60-diamidino-2-phenylindole (DAPI). Nine fieldsper well at �4 magnification were quantified using ImageJVersion 1.34s (NIH).

ImmunohistochemistryTissues were harvested and fixed in 3% paraformaldehyde

overnight. Sections (5 mm) were stained with the followingantibodies: anti-F4/80 (1:500; Serotec), anti-Gr-1 (1:100;eBioscience), or anti-CD31 (1:300; BD Biosciences) antibodies.Histology was conducted, processed, and quantified as previ-ously described (15). The samples were analyzed using anOlympus BX41 fluorescentmicroscope fitted with a Q-ImagingQICAM FAST 1394 camera. Images were captured at�4, �10,or � 20 magnification using QCapture Pro Version 5.1 (MediaCybernetics), and quantified using ImageJ Version 1.34s (NIH).

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2783

Page 3: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

Immunofluorescence microscopyCells seeded on cover slips were fixed with 3% formaldehyde

and incubated with c-Abl antibody (K-12, 1:100; Santa CruzBiotechnology) followed by AlexaFluor 488 rabbit anti-mouse(Invitrogen), and then AlexaFluor 568 phalloidin and DAPI(Invitrogen). Mounting medium (Pro-Long Gold AntifadeReagent; Invitrogen) was applied and cover slips were sealedwith clear nail polish. Fluorescent images were acquired atroom temperature on a confocal microscope, LSM710 (CarlZeiss).

Flow cytometric analysisTo prepare single-cell suspensions for flow cytometry, har-

vested tissues (tumors or lungs) were dissected into approx-imately 1 to 3 mm3 fragments and digested with 80 U/mLcollagenase (Invitrogen) in DMEM containing 10% FBS for 1.5hours at 37�C while shaking. Spleens and lymph nodes weregently dissociated between 2 glass slides for single-cell isola-tion. Peripheral blood was isolated directly into BD VacutainerK2 EDTA tubes (BD Biosciences). After red blood cell lysis(Sigma-Aldrich), single-cell suspensionswerefiltered and incu-bated for 30 minutes on ice with the following: APC, PerCP-Cy5.5, PE, and APC-e780-conjugated antibodies (CD11b, Gr-1,CSF1R, and F4/80) were purchased from eBioscience (1:200).Ly6C (1:200) was purchased from BD Biosciences. DAPI waspurchased from Invitrogen. Cells were washed twice beforeanalysis on the BD LSR-II flow cytometer (Beckman Coulter).Data were analyzed with FlowJo software (TreeStar).

Local irradiationIrradiation was carried out using a Gulmay X-ray machine

(300 kV, 10 mA) with a dose rate of 1.84 Gy/min. When tumorsreached 4 to 5 mm in diameter, mice were anesthetized andirradiated with a daily dose of 3 Gy for 5 days to the tumor areawith the rest of body shielded.

Real-time reverse transcriptase-PCR analysisTotal cellular RNA was extracted from cells using Tri

Reagent (Sigma Aldrich). RNA was isolated according to theTRIzol procedure. RNA was quantified and assessed for purityby UV spectrophotometry and gel electrophoresis. RNA (1 mg)was reverse-transcribed using the iScript cDNA synthesis kit(Bio-Rad) according to the manufacturer's instructions. Foreach sample, 1 mL cDNA (�20 ng) was amplified using SyberGreen 2�MasterMix (Bioline) and 10 mmol/L primers (Primersequences are listed in Supplementary Table S1). The reactionwas run on the My IQ single color iCycler real time PCRmachine (Bio-Rad). Samples were amplified using the follow-ing cycling conditions: 40 cycles of 95�C/15 s, 60�C/30 s, and72�C/30 s. Gene expression was determined by theDCt methodand normalized to b-actin expression.

SDS-PAGEFor experiments using concentrated media, cells were plat-

ed in 150 mm tissue culture dish in 0% FBS DMEM overnight.Twenty milliliters of media were collected 48 hours afterirradiation and subjected to media concentration using aprotein concentrator (Pierce Biotechnology) at a speed of

4,500 � g for 30 minutes. Total volume was normalizedbetween samples and 40 mL was loaded onto 4% to 12%continuous gradient Tris–glycine gel (Invitrogen). For ABL1cleavage experiments, cells were plated in 6-well platesovernight and collected after irradiation at the time indicat-ed. Cells were lysed in radioimmunoprecipitation assay buf-fer (Upstate) containing proteinase inhibitor cocktail (Sigma),sonicated briefly, and centrifuged for 10 minutes at 12,000� g.Twenty mmicrograms of cell lysate was resolved on a 4%to 12% continuous gradient Tris–glycine gel (Invitrogen). Thegels were then transferred to a polyvinylidene difluoride(PVDF) membrane (Millipore) and incubated with primaryantibodies: anti-ABL1 (K-12, rabbit polyclonal; Santa CruzBiotechnology), dilution 1:1,000; anti-CSF1 (H-300, rabbit poly-clonal, dilution 1:20,000; Santa Cruz Biotechnology), dilution1:1,000; anti-GAPDH (A-3, mouse monoclonal; Santa CruzBiotechnology).

Statistical analysisData are presented as mean� SEM. Statistical comparisons

between groups were carried out using the Student t test.

ResultsLocal irradiation enhances myeloid cell infiltration totumors

Abundant evidence points to the infiltrating myeloid cellsexerting significant influences on tumor cell aggression andthe immunologic environment. Therefore, we studied therecruitment of irradiation-induced TIMs in 2 immunocompe-tent murine prostate cancer models, namely RM-1 and Myc-CaP syngeneic in the C57/BL6 and FVB strain, respectively.Thesemodels provide distinct host genetic background, tumorgrowth rate, degree of myeloid cells infiltration, and responseto irradiation to broaden the perspectives on this issue.Wefirstexamined the recruitment of TIMs to tumors after irradiationin the RM-1, a Ras-, and Myc-transformed murine prostaticcancer model (26), with moderate level of TIMs infiltration. Asshown in Fig. 1, irradiation effectively delayed the tumorgrowth by approximately 7 days (Fig. 1A). Control tumors andirradiated tumors were collected when they reached similarsizes (day 13 for control tumors and day 19 for irradiatedtumors) and processed to assess their content of TIMs. Irra-diation significantly induced the infiltration of F4/80þCD11bþ

TAMs (Fig. 1B) and Gr-1þCD11bþ MDSCs (Fig. 1C) to thetumors. Immunohistochemical staining further confirmedthis increase of TIMs in the tumors (Fig. 1D). Recent reportsfurther distinguished the myeloid subsets within MDSCs asconsisting of MO-MDSC (CD11bhily6Clo) and PMN-MDSC(CD11bloly6Chi) with different functional characteristics (27).We found that both MO-MDSCs and PMN-MDSCs were sig-nificantly induced by irradiation, with MO-MDSCs showing alarger increase (Fig. 1E). Likewise, the irradiation-inducedTIMs recruitment was also observed in the RM-9 tumor, aC57BL6 compatible model derived in the same manner asRM-1, and Myc-CaP tumor, a myc oncogene-driven modelimplantable in FVB host (Supplementary Figs. 1 and 2). Takentogether, these data show that local irradiation enhances

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2784

Page 4: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

recruitment of both TAMs and MDSCs to tumors in severalmurine prostate cancer models.

Local irradiation enhances systemic myeloid cellexpansionTo better characterize the potential systemic impact of local

tumor irradiation, we examined the infiltration of MDSCs andTAMs in peripheral tissues at different time points afterirradiation, providing insights on the dynamics and kineticsof the myeloid cell recruitment process. The level of CD11bþ

F4/80þ macrophages were low in lungs, spleens, lymph nodes,and blood; therefore, we focused on CD11bþGr-1þ MDSCs inthe systemic sites and analyzed the content of CD11bþF4/80þ

macrophages only in the tumor (Fig. 2B). As shown in Fig. 2,before irradiation, the baseline levels of MDSCs in tumors,blood samples, and spleens were 1.5 � 0.9%, 30.1 � 1.3%, and3.4 � 1.1%, respectively, and the levels in the lungs and lymphnodes were negligible. In untreated mice, MDSC levels stay thesame or only mildly increase over time (Fig. 2). In irradiatedmice, within 2 days after irradiation, MDSCs in the peripheralblood doubled to 69.0� 6.8% (Fig. 2C), whereas MDSCs stayedrelatively stable in spleens, lymph nodes, and lungs (Fig. 2D–2F). In the irradiated tumor, there was a sustained low level oftumoral MDSCs throughout and beyond the duration of irra-diation although they increased nearly 4-fold (1.5 � 0.9% to6.5 � 1.9%) in nonirradiated tumors. On day 12, 2 days after

Figure 1. Local irradiation enhances myeloid infiltration into tumors. Subcutaneous RM-1 tumors were collected, processed to single-cell suspension, andassayed by FACS and immunohistochemistry for CD11bþGr-1þ MDSCs and CD11bþF4/80þ TAMs. A, growth curve of RM-1 tumors with or withoutirradiation. B, FACS plots and quantification of TAMs in tumor. C, FACS plots and quantification for MDSCs in tumor. D, representative F4/80 stainingof RM-1 tumors from control and irradiation-treated mice. E, effect of irradiation on the 2 subsets of MDSCs: MO-MDSC and PMN-MDSC; n ¼ 4 foreach group.

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2785

Page 5: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

cessation of tumor irradiation,MDSC levels reached their nadirin the tumors, but had begun to rise in the other organs. By day15, a dramatic increase in the MDSC population was observedin the tumor, spleen, and lymph node, with levels reaching apeak of approximately 15% in all 3 sites (Fig. 2A, D, and F)before falling on day 17. In the blood and lungs, the trend ofMDSC elevation continued from day 15 onward, reaching aremarkable level of 80.0� 5.3% and 30.7� 14.6%, respectively,on day 17. Collectively, these data suggest that local tumorirradiation induces the expansion of MDSCs and their subse-quent influx into different organs in a time-dependentmanner.

Irradiation induces macrophage migration andexpression of protumorigenic genes

We next used an in vitro culture system to aid in dissectingthe complex cross-signaling between different cellular com-ponents in the tumor microenvironment after irradiation. Wefirst examined the effects of conditionedmedia from irradiatedtumor cells on themigration ability ofmacrophages. RAW264.7murine macrophages were analyzed in a Transwell migrationassay with conditionedmedia from irradiated or nonirradiatedmurine prostate cancer cells (RM-1 and Myc-CaP) as themigration stimulus. Conditioned media from irradiated tumorcells induced a nearly 2-fold greater number of RAW264.7 cellsto migrate across the Transwell filter compared with nonir-radiated controls (Fig. 3A; data not shown for Myc-CaP).

A large volume of work points to the plasticity of tissuemacrophages that are educated by tumor environmental cuesto promote tumor growth (8). Therefore, we interrogatedwhether irradiation could skew bone marrow-derived macro-phages (BMDM) toward the gene expression profiles of pro-

tumorigenic macrophages. Direct irradiation with 3 Gy (Fig.3B) and indirect effects transmitted through coculturingBMDMs with irradiated tumor-conditioned media (Fig. 3C)both polarized BMDMs toward a protumorigenic phenotype,as we observed increased expression of Arg1, Fizz, IL-1b, IL-10,MMP-9, VEGF-A, CD206, and CSF1, and decreased expressionof inflammatory genes such as iNOS and IL-12. As a reflection oftheir immunosuppressive roles in tumors, protumorigenicmacrophages also typically exhibit lower MHCII expression(28). By fluorescence-activated cell sorting (FACS) analysis, all(100%) of the CD11bþF4/80þ macrophages from irradiatedtumors displayed low MHCII expression, whereas only 50%of the macrophages in nonirradiated tumors were MHCIIlow

(Fig. 3D). These data suggest that both direct and indirecteffects of irradiation can have profound effects on macro-phages by skewing them toward a protumorigenic subtypewith enhanced migration ability and gene expression profilethat favor tumor growth.

CSF1 expression is increased by irradiationNext, we examined whether tumor cell irradiation can alter

the expression of cytokines known to participate in myeloidcell recruitment (6, 29). The expression of CSF1, CCL2, CCL5,and SDF-1 was examined in Myc-CaP cells 24 hours after 3 Gyof irradiation. Among these cytokines examined, CSF1 showedthe highest expression and the most significant increase inirradiated over untreated tumor cells (Fig. 4A). We furtherevaluated secreted levels of CSF1 protein, which was elevatedin conditioned media from irradiated tumor cells (Fig. 4B).IL-34 is a recently discovered second CSF1R ligand that func-tions similarly to CSF1 (30). However the expression of IL-34

Figure 2. A–F, local irradiation enhances systemic myeloid cell expansion. Control mice were sacrificed on days 6, 8, and 12. Irradiated mice weresacrificed on days 6, 8, 12, 15, and 17. Tumors, blood, spleens, lungs, and lymph nodes were collected for FACS analysis for MDSC and TAMpopulation; n ¼ 4 for each time point.

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2786

Page 6: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

was 100-fold lower than CSF1 in our 2 prostate cancer systems(RM-1 and Myc-CaP model, data not shown) and, thus, we didnot pursue IL-34 further. The ability of irradiation to augmentCSF1 expression appeared to be a general phenomenon. Intotal, we tested 9murine and human cancer cell lines and 8 of 9showed an increase in CSF1 after irradiation (Fig. 4A and E;Supplementary Fig. S3). Consistent with results from cellculture experiments, irradiated tumors (the same cohortas Fig 1A) showed a significant increase in CSF1 gene expres-sion compared with untreated tumors (Fig. 4C). Importantly,an increase in CSF1 was also observed in the serum of patientswith prostate cancer after radiotherapy (Fig. 4D), supportingthe clinical relevance of CSF1 increase seen in our murinemodels.To explore the issue of cross-talk between tumor cells and

macrophages on the CSF1 axis, we cocultured RAW264.7macrophages or BMDMs with RM-1 tumor cells and observedan increase in the magnitude of CSF1 expression more thanthat in either cell grown alone, especially in the irradiationsetting (Fig. 4E and F). Furthermore, the addition of a highlyselective CSF1R kinase inhibitor GW2850 (31) resulted in acomplete negation of increasedmacrophagemigration towardirradiated tumor-conditioned media (Fig. 4G). Collectively,these data show that irradiation increases CSF1 expression

in tumors, which is amplified by tumor–macrophage interac-tions. The heightened CSF1 production induced by irradiationin turn drives macrophage migration and recruitment intoirradiated tumors.

Irradiation enhances CSF1production through anABL1-dependent mechanism

Given our finding shown above that irradiation booststumoral CSF1 expression, we sought to interrogate a signaltransduction pathway implicated in irradiation-induced tran-scriptional regulation of CSF1. ABL1, a nonreceptor tyrosinekinase, is known for mediating apoptosis and cycle arrest afterirradiation (32). It has been reported that ABL1 can also berecruited to the promoter region of CSF1 and regulates CSF1gene expression in concert with AP-1 (33). Thus, we examinedin detail the kinetics of irradiation-induced CSF1 expressionand ABL1 activation in our system. First, detailed analysis ofirradiation-induced gene expression of CSF1 in Myc-CaP cellsshowed the increase in CSF1 RNA initiated at 4 hours afterirradiation (Fig. 5A). Next, we profiled the activation of ABL1protein in response to irradiation. We observed that two ABL1cleavage products, 75 kDa and 60 kDa product, emerged at 2hours after irradiation (Fig. 5B). Further examination of thesubcellular localization of ABL1 by confocal microscopy

Figure 3. Irradiation increases cell migration and induces protumorigenic genes in macrophages. A, RAW264.7 macrophages were seeded in 8 mmTranswell inserts, and tumor-conditioned media (collected 48 hours after 3Gy irradiation) were placed in the bottom. Cells were allowed to migrate towardthebottom for 6hours. Then, cellswere fixedandstainedwithDAPI. Representative imagesofmigrated cells are shownand theywerequantifiedusing ImageJsoftware (n ¼ 3). B, effect of irradiation on bone marrow-derived macrophages. Bone marrows were collected and induced to macrophages by CSF1(10 ng/mL) for 6 days. Cells were counted, seeded, and subjected to 3Gy irradiation. Cells were collected 24 hours later, and RT-PCR was carried outto detect RNA for the protumorigenic and inflammatory genes noted. C, bone marrow-derived macrophages as prepared above were cultured in 50%tumor-conditioned media þ 50% complete DMEM for 24 hours. Cells were collected and assayed by RT-PCR for the genetic markers noted. D, tumorscollected as shown in Fig. 1A were analyzed by FACS for MHCII expression on CD11bþF480þ macrophages. An increase in MHCII low-expressingmacrophage population was observed. (�, significant changes with P < 0.05).

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2787

Page 7: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

revealed that before the radiation insult, ABL1 was predom-inantly located in the cytoplasm with very little ABL1 immu-nocytochemical signal registered in the nucleus (Fig. 5C, left).As early as 1 hour after irradiation, a noticeable portion, but notall, of the ABL1 protein has translocated to the nucleus (Fig. 5C,middle), and this is maintained at 4 hours (Fig. 5C, right). Tofurther substantiate the functional impact of ABL1 on the CSF1gene expression, we analyzed the binding of ABL1 to the CSF1promoter by a chromatin immunoprecipitation assay (ChIP).An increase in ABL1 binding to the CSF1 promoter wasobserved as early as 1 hour after irradiation and peaked at 2hours, before slowly declining (Fig. 5D). When looking into thekinetics of RNA polymerase II binding to the CSF1 promoter,it first showed a significant decrease at 1 hour and an increasestarting 2 hours post-irradiation (Fig. 5E). This bell-shapedkinetics of RNA polymerase activity could be attributed to theradiation-induced DNA damage causing an initial inhibitionon transcription (34). The timing of ABL1 nuclear transloca-tion, it's binding to the CSF1 promoter, and the binding of

RNA polymerase II to the CSF1 promoter preceded thechanges in CSF1 mRNA level. These findings support theinvolvement of ABL1 in the regulation of CSF1 expression inresponse to irradiation.

Next, we used anABL1-targeted siRNA and a small-moleculeinhibitor to further confirm its regulatory role on CSF1 in oursystem. The irradiation-induced CSF1 expression in MycCaPcells was significantly inhibited by the addition of the ABL1kinase inhibitor, STI-571 (5 mmol/L; Fig 5F). A similar resultwas also observed in the human prostate cancer cell lineCWR22Rv1 (Supplementary Fig. S4). Using a migration assay,we found that STI-571–treated conditioned media displayeddecreased ability to promote macrophage migration (Fig. 5G).Furthermore, the addition of GW2580 to macrophages (topchamber) did not further retard macrophage migrationthroughout STI-571 treatment of tumor cells (Fig. 5G), suggest-ing STI-571 andGW2580may be targeting the same pathway toregulate migration. Like most of the pharmacologic proteinkinase inhibitors, STI-571 is not completely specific. STI-571

Figure 4. CSF1 expression is increased by irradiation. A, MycCaP cells were plated overnight and irradiated with 3 Gy. Cells were collected 24 hours after,and assayed by RT-PCR for factors known to recruit myeloid cells: CSF1, CCL2, SDF1, and CCL5. CSF1 is shown to have the highest expressionandmost significant increase. B, conditionedmedia fromMyc-CaP cells 48 hours after irradiation were collected, concentrated by centrifugation, normalized,and analyzed by SDS-PAGE. Secreted CSF1 was detected at 50 kDa. Relative expression level quantified by ImageJ was shown below the blot. C, tumorscollected as shown in Fig. 1A were assayed by RT-PCR for CSF1 mRNA expression. D, serum samples from patients with prostate cancer pre- andpostradiotherapy were analyzed by ELISA for CSF1. E, RM-1 cells and macrophage cell line RAW264.7 were cocultured overnight and irradiated with3 Gy. Cells were collected after 24 hours and analyzed by RT-PCR for CSF1 mRNA. F, RM-1 cells and BMDM were cocultured for 4 hours and irradiatedwith 3 Gy. Cells were collected after 24 hours and analyzed by RT-PCR for CSF1 mRNA. G, RAW264.7 macrophage migration assay was conductedusing conditioned media in the lower chamber and then collected as in B. GW2580 (1 mmol/L) was added to the top chamber. Quantification of 9 fields issummarized in F. Representative images of migrated cells were shown in G.

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2788

Page 8: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

also inhibits c-Kit, platelet-derived growth factor receptor, andCSF1R (35). Thus, we used siRNA-mediated knockdown ofABL1 to examine its effect on CSF1. Because of the lowefficiency of transfecting murine prostate cancer cells, weused the human CWR22Rv1 cell line for this experiment. TheABL1 siRNA was able to decrease ABL1 mRNA expressionto 30% of the normal level in the CWR22Rv1 cells (Fig. 5H,right). As expected, ABL1 siRNA effectively blocked the CSF1mRNA induction by irradiation (Fig. 5H, left). These datasuggest that upon irradiation, ABL1 is activated, translocatesto the nucleus, binds to the promoter region of CSF1, andpromotes its expression.

Blockade of CSF1/CSF1R signaling retards tumorregrowth after local irradiationCSF1 is a potent cytokine well known to promote myeloid

cell proliferation, differentiation, and migration. In a recent

study, we showed that blockade of CSF1/CSF1R signaling caneffectively inhibit TIM function and recruitment to tumors(15). Thus, in this study, we investigated whether blockingCSF1R can also reduce recruitment of TIMs and diminish theirprotumorigenic influences in the radiotherapy setting. Thecombined irradiation and CSF1R blockade treatment was firsttested with the selective CSF1R inhibitor, GW2580. Significantreductions in TIM populations in RM-1 tumors were observedwith GW2580 treatment (160 mg/kg/d), which augmentedthe efficacy of irradiation by achieving more effective suppres-sion of tumor growth than irradiation alone (SupplementaryFig. S5A–E; data not shown). To further substantiate thisrational combination strategy, we used a recently describedsmall-molecule CSF1R kinase inhibitor PLX3397. This inhibitorwas shown to be a highly potent inhibitor of CSF1R (cFMS)with IC50 of 20 nmol/L and it is under active clinical investi-gation for several types of cancers (19). Here, RM-1 prostate

Figure 5. Irradiation induces CSF1 production through an ABL1-dependent mechanism. A, CSF1 mRNA expression in Myc-CaP cells 0, 1, 2, 4, 6, 8, and24 hours after irradiation (3 Gy). B, ABL1 is cleaved by irradiation in vitro. A total of 4� 105 Myc-CaP cells were plated in 6-well plate overnight and irradiatedwith 3 Gy the next day. Cells were collected at 1, 2, 4, and 8 hours after irradiation. Cells lysates were normalized and assayed by SDS-PAGE. Westernblotting was done and probed with ABL1 (K-12) antibody for both full-length and cleaved ABL1. Two cleavage products, measuring 60 and 75 kDa,weredetected.C, confocal images ofMyc-CaP cells at 1 and 4hours after irradiation. Green, ABL1; red, actin;white, nucleus (DAPI). D andE,Myc-CaPcells 0,1, 2, and 4 hours after irradiation were fixed in 3% PFA and processed for ChIP assay using c-Abl antibody (D) and RNA polymerase II antibody (E) asdescribed in Materials and Methods. Rabbit IgG was used as negative control. F, Myc-CaP cells were irradiated with 3 Gy and STI-571 (5 mmol/L) wasadded right after. Cells were collected 24 hours later and analyzed by RT-PCR for CSF1 mRNA expression. G, migration assay using conditioned mediacollected as in F. GW2580 was added to the top chamber to examine the additive effects between STI-571 and GW2580. H, CSF1-mRNA in human prostatecancer cells after ABL1 directed RNAi. CWR22Rv1 cells were transfected with ABL1 siRNA and negative control (NC, nonspecific siRNA). Cells wereirradiated 30 hours after siRNA treatment and collected 24 hours after irradiation. Expression level of ABL1 was reduced to 30% of control after specificsiRNA treatment (right). CSF1 mRNA was analyzed by RT-PCR with and without irradiation.

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2789

Page 9: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

tumor-bearing mice were treated with control (chow), localirradiation (3Gy � 5 days), PLX3397 (drug chow), or thecombination. As shown in Fig. 6A, PLX3397 alone has littleeffect on tumor growth compared with the control group.Irradiation reduced tumor size by 43% at day 10, 1 day aftercessation of irradiation (P < 0.001). The irradiated tumor sizeswere stabilized for a short duration, and subsequently resumedan aggressive tumor growth rate, whereas the combinedirradiation-and-PLX3397-treated group maintained a muchslower growth rate (Fig. 6A). Both flow cytometry and histo-logic analyses of tumors revealed a significant reduction ofCD11bþGr-1þ MDSCs and CD11bþF4/80þ macrophages intumors as well as in spleens of both PLX3397-treated groups,with more pronounced effects observed in the combinationtreatment (Fig. 6B–E, and G). Interestingly, both subsets ofMDSCs, monocytic and polymorphonuclear, were reduced byPLX3397 (Fig. 6F), a result that differed from our previousfindings with GW2580 treatment in 3LL tumors where onlythe monocytic subtype of MDSCs was inhibited (15). At themolecular level, CSF1R blockade significantly reduced radio-therapy-inducedCSF1,MMP9, andArg1 (Fig. 6H–J). The latter 2genes are known to be involved in cancer progression andmetastasis by promoting tissue remodeling, angiogenesis, andimmunosuppression (15). Similar reductions in the expressionof CSF1 and Arg1 were also observed in irradiated tumorstreated with GW2580, along with a significant reduction in themacrophage chemotactic factor, CCL2 (Supplementary Fig.S5D and S5E). In summary, we observed that prostate tumor-directed irradiation can potently induce the influx of TIMs,which in turn can thwart treatment efficacy. The addition ofpotent CSF1R inhibitors such as PLX3397 and GW2580 canprevent the influx of TIMs and halt their protumorigenicfunctions, thus leading to more effective and durable tumorgrowth control.

DiscussionIn the present study, we show that the recruitment of

TIMs to prostate tumors is highly induced by local irradi-ation in several immunocompetent mouse models. We findelevated expression of CSF1, an important cytokine formacrophage survival, migration, and differentiation, intumor cells after irradiation, which was also observed inthe serum of prostate cancer patients after radiotherapy.Increased CSF1 expression was mediated, at least partially,by the ABL1 tyrosine kinase. On irradiation, ABL1 wasactivated and translocated to the nucleus, where it boundto the promoter region of CSF1 to upregulate its expression.We further showed that blockade of CSF1R with selectivesmall-molecule kinase inhibitors, such as GW2580 andPLX3397, greatly inhibit TIMs infiltration and significantlydelay tumor regrowth after irradiation. These results suggestthat disrupting the protumorigenic contributions of hostinnate immune cells, namely MDSCs and macrophages,through blockade of the CSF1/CSF1R axis can be a prom-ising approach for developing rational and more effectivecombination cancer therapies. A schema of irradiation-induced expression of CSF1 and recruitment of TIMs and

the impact of CSF1R inhibition on TIMs' modulation oftumor regrowth is shown in Fig. 7.

The use of immunocompetent murine prostate tumor mod-els in this study allowed us to directly assess the contributionsof host immune cells, in particular, the distinct myeloid sub-populations, to tumor progression after therapy. Our resultsshowed that themajor impact of CSF1R blockade is directed atthe tumor microenvironment, namely TIMs. Interestingly,CSF1R has been shown to be expressed and can contributeto the oncogenesis of several types of cancer, including pros-tate cancer (36, 37). Therefore, the blockade of the CSF1/CSF1Raxis could potentially have a direct, suppressive impact ontumor cells, albeit unlikely as the RM-1 and Myc-CaP tumorsused here express negligible levels of CSF1R on the basis ofsensitive RT-PCR analyses (data not shown). We also believeIL-34, a newly identified ligand for CSF1R having similarfunctions in stimulating macrophage proliferation and migra-tion (30, 38), is unlikely to play a significant role as itsexpression level is 100-fold lower than CSF1 in our models(data not shown). Recent findings from Dr. Hong Wu andcolleagues using the PTEN-knockout transgenic prostatic car-cinoma model revealed that intratumoral MDSCs expansioncontributes to tumor progression and that CSF1R blockadewas an effective means to suppress the infiltration and func-tion of MDSCs in this spontaneous murine prostate cancermodel (data not shown).

It is promising that our initial exploratory study on 10consecutive patients with prostate cancer, who recently under-went radiotherapy, also yielded data supportive of the CSF1axis being involved. Although our data suggests that serumCSF1 could potentially be a biomarker of TIMs recruitment,there are several considerations that caution against thispremature conclusion. First, a wide range of serum CSF1 levelwas detected in patients (Fig. 4D). This issue could likely beattributed to the different infection or inflammation status ofthe patients. Second, the timing of patient specimen procure-ment after radiotherapy was not uniform between patientsin the small cohort tested. We did observe an increase in aMDSC population (CD11bþCD15þ) in the peripheral blood ofa few patients whose serum CSF1 increased from pre- topostradiotherapy (data not shown). Because of the heteroge-neity issues, data from a much larger cohort of patients willbe needed to fully validate the concept put forth here. Weare actively pursuing these studies with more standardizedtime points of peripheral blood collection in patients withprostate cancer undergoing radiotherapy.

Our findings are consistent with CSF1 being an importantstimulus for the influx of TIMs to tumors especially in responseto irradiation. However, our study does not exclude otherpathways that may also be involved in this complex inflam-matory cascade. For instance, several recent articles highlight-ed the role of the SDF-1/CXCR4 axis in local irradiation-induced influx of TIMs to tumors (5, 6). The authors showedthat irradiation induced hypoxia through destruction of endo-thelial cells and the microvasculature, and the resultantincreased expression of HIF-1a, in turn, induced the expres-sion of CXCR4 and SDF-1, which then mediated the recruit-ment of TIMs to tumors. Likewise, CCL2 has also been

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2790

Page 10: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

implicated in the recruitment of bonemarrow-derivedmyeloidcells into tumors and this axis can also modulate prostatecancer growth and metastasis to bone (39, 40). However, weobserved negligible SDF-1 and CCL2 expression with or with-

out irradiation in our tumor cells (Fig. 4A). The migration/recruitment of TIMs is a complex process that is likely regu-lated by several pathways, especially in the context of differenttumor types, host genetic background, and stimulus induced

Figure 6. CSF1/CSF1R blockade inhibits tumor growth after irradiation. A, growth curve of subcutaneous RM-1 tumors treated with radiotherapy(3 Gy � 5 days), PLX3397 (in food chow), or combination as indicated. Tumors were measured daily by caliper. B and C, FACS analysis of CD11bþF4/80þ

macrophages and CD11bþGr-1þ MDSCs in tumor, collected at end points (tumor, n ¼ 6 � for each cohort). D and E, FACS analysis of CD11bþF4/80þ

macrophages andCD11bþGr-1þMDSCs in spleen, at the same termination timeas above. F, FACSanalysis forMDSCsubsets,MO-MDSC, andPMN-MDSCwith single or combination treatment. G, representative immunohistochemsitry staining for F4/80 on tumor sections with single or combination treatment.H–J, RT-PCR analysis of mRNA extracted from tumors for CSF1, MMP9, and Arg1.

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2791

Page 11: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

by different therapeutic settings or progression status. Ofinterest, we consistently observed a decrease in CCL2 levelwith GW2580 or PLX3397 treatment (data not shown; Supple-mentary Fig. 5F). This finding is also in accordance with arecent study that showed the removal of CSF1 significantlydecreases CCL2 (41). An intriguing possibility could be thatCSF1 is an upstream regulator of other cytokines like CCL2.Clearly, the influence of the CSF1/CSF1R axis and its cross-talkwith other cytokine/chemokine pathways in the recruitment ofTIMs deserves further investigation.

A unique aspect of this study is the finding that the ABL1pathway mediates the heightened CSF1 transcription inducedby irradiation. Several factors have been implicated in theregulation of CSF1 gene expression, including PDGF (42), ABL1(33), IFN-g (43), and AP-1, CTF/NF-1, SP1, SP3 (44), and nuclearactin (45) in different cell types and contexts. Among these,ABL1 is known to respond to irradiation or DNA damage viathe DNA-PK, ATM, and p53 pathway (46–48). ABL1 contains acatalytic domain as well as a NES and 3 NLS motifs. Severalgroups reported that ABL1 localizes to both nucleus andcytoplasm and can shuttle between these 2 compartments(49). In our study, we observed that a portion of cytoplasmicABL1 is activated and translocated to the nucleus as early as 1hour after irradiation (Fig. 5B). TheABL1DNA-binding domainis critical for its biologic function (50), yet no classical DNA-binding motifs have been identified so far. The few ABL1transcriptional targets identified include p21 and CSF1(33, 51). Here, we showed that ABL1 binds to the promoterregion of CSF1 and activates CSF1 gene transcription. A

previous study suggested that ABL1 forms a complex withAP-1, a transcription factor composed of c-jun and c-fos, in theregulation of CSF1 (33). On the basis of this finding, a potentialfeedback loop in CSF1R-dependent cells is that the blockade ofCSF1R signaling inhibits c-fos, which further downregulatesCSF1 expression. This mechanism might explain why weobserved a decrease in CSF1 expression in tumors afterPLX3397 treatment (Fig. 6H).

In summary, the data presented in this study show thatirradiation induces CSF1 through an ABL1-dependent mech-anism in prostate cancer. The heightened CSF1 serves acritical role in the systemic recruitment of protumorigenicmyeloid cells to irradiated tumors. Therefore, the blockadeof TIMs in combination with local irradiation of prostatetumors displays an augmented and more durable responsethan irradiation alone in preclinical models. We believe thatcotargeting the CSF1/CSF1R pathway with local irradiationof prostate tumors will be a promising strategy for clinicaltranslation.

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

Authors' ContributionsConception and design: J. Xu, J. Escamilla, J. David, S.J. Priceman, L. WuDevelopment of methodology: J. Xu, J. DavidAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J. Xu, J. DavidAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J. Xu, J. Escamilla, B.L. West

Radiation

Tumor Stromal TIMs Monocytes CSF1 ABL1

Bone marrow

CSF1R inhibitor

A

B

Bone marrow

ABL1

ABL1

CSF1 Figure 7. Model of CSF1 expressioninduced by irradiation, promotingTIMs recruitment and tumorregrowth. A, tumor irradiationactivates ABL1,which translocatesto the nucleus, binding to thepromoter region of CSF1, andupregulates its gene expression.Additional TIMs are recruited totumor sites due to the increase inCSF1 and they can thus promotetumor growth. B, when tumor-bearing mice were treated with asmall-molecule CSF1R kinaseinhibitor, CSF1/CSF1R signalingwas inhibited, resulting indecreased infiltration of TIMs,reducing their tumor growth–promoting influences.

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2792

Page 12: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

Writing, review, and/or revision of the manuscript: J. Xu, J. Escamilla, S.J.Priceman, B.L. West, G. Bollag, W.H. McBride, L. WuAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): J. Xu, S. Mok, W.H. McBrideStudy supervision: J. Xu

AcknowledgmentsThe authors thank Drs. Steven Bensinger, John Colicelli, and Arnie Berk for

helpful discussions. The authors also thank UCLA JCCC and UCLA CTSI(1UL1RR033176), respectively, for providing postdoctoral fellowship supportto J. Xu and translational infrastructural services.

Grant SupportThis work was supported by DOD CDMRP W81XWH-12-1-0206 (L. Wu),

NCI SPORE P50 CA092131, and Margaret E. Early Medical Trust awardL. Wu).

The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisementin accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received October 22, 2012; revised January 4, 2013; accepted January 21, 2013;published OnlineFirst February 15, 2013.

References1. Delaney G, Jacob S, Featherstone C, Barton M. The role of radio-

therapy in cancer treatment: estimating optimal utilization from areview of evidence-based clinical guidelines. Cancer 2005;104:1129–37.

2. Agarwal PK, Sadetsky N, Konety BR, Resnick MI, Carroll PR.Treatment failure after primary and salvage therapy for prostatecancer: likelihood, patterns of care, and outcomes. Cancer 2008;112:307–14.

3. D'Amico AV, Renshaw AA, Sussman B, Chen MH. Pretreatment PSAvelocity and risk of death fromprostate cancer following external beamradiation therapy. JAMA 2005;294:440–7.

4. Bianco FJ Jr, Scardino PT, Stephenson AJ, Diblasio CJ, Fearn PA,Eastham JA. Long-term oncologic results of salvage radical prosta-tectomy for locally recurrent prostate cancer after radiotherapy. Int JRadiat Oncol Biol Phys 2005;62:448–53.

5. Kozin SV, Kamoun WS, Huang Y, Dawson MR, Jain RK, Duda DG.Recruitment of myeloid but not endothelial precursor cells facil-itates tumor regrowth after local irradiation. Cancer Res 2010;70:5679–85.

6. Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh GR, Brown JM.Inhibition of vasculogenesis, but not angiogenesis, prevents the recur-rence of glioblastoma after irradiation in mice. J Clin Invest2010;120:694–705.

7. Hanahan D, Coussens LM. Accessories to the crime: functions of cellsrecruited to the tumor microenvironment. Cancer Cell 2012;21:309–22.

8. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progres-sion and metastasis. Cell 2010;141:39–51.

9. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, et al.Tumor refractoriness to anti-VEGF treatment is mediated byCD11bþGr1þ myeloid cells. Nat Biotechnol 2007;25:911–20.

10. MuthanaM,RodriguesS,ChenYY,WelfordA,HughesR, TazzymanS,et al. Macrophage delivery of an oncolytic virus abolishes tumorregrowth and metastasis after chemotherapy or irradiation. CancerRes 2013;73:490–5.

11. MurdochC,MuthanaM,Coffelt SB, LewisCE. The role ofmyeloid cellsin the promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618–31.

12. Lewis CE, Pollard JW. Distinct role of macrophages in different tumormicroenvironments. Cancer Res 2006;66:605–12.

13. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells:linking inflammation and cancer. J Immunol 2009;182:4499–506.

14. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as reg-ulators of the immune system. Nat Rev Immunol 2009;9:162–74.

15. Priceman SJ, Sung JL, Shaposhnik Z, Burton JB, Torres-Collado AX,MoughonDL, et al. Targeting distinct tumor-infiltratingmyeloid cells byinhibiting CSF-1 receptor: combating tumor evasion of antiangiogenictherapy. Blood 2010;115:1461–71.

16. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y,et al. Expansion of myeloid immune suppressor GrþCD11bþ cells intumor-bearing host directly promotes tumor angiogenesis. CancerCell 2004;6:409–21.

17. Hamilton JA. Colony-stimulating factors in inflammation and autoim-munity. Nat Rev Immunol 2008;8:533–44.

18. Chitu V, Stanley ER. Colony-stimulating factor-1 in immunity andinflammation. Curr Opin Immunol 2006;18:39–48.

19. DeNardo D, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF,et al. Leukocyte complexity predicts breast cancer survival and func-tionally regulates response to chemotherapy. Cancer Discov 2011;1:52–65.

20. Wang JY. Nucleo-cytoplasmic communication in apoptotic responseto genotoxic and inflammatory stress. Cell Res 2005;15:43–8.

21. Ren X, Xu J, Cooper JP, Kang MH, Erdreich-Epstein A. c-Abl is anupstream regulator of acid sphingomyelinase in apoptosis induced byinhibition of integrins alphavbeta3 and alphavbeta5. PLoS ONE2012;7:e42291.

22. Xu J, Millard M, Ren X, Cox OT, Erdreich-Epstein A. c-Abl mediatesendothelial apoptosis induced by inhibition of integrins alphavbeta3and alphavbeta5 and by disruption of actin. Blood 2010;115:2709–18.

23. Ogawa Y, Kawamura T, Furuhashi M, Tsukamoto K, Shimada S.Improving chemotherapeutic drug penetration in melanoma by ima-tinib mesylate. J Dermatol Sci 2008;51:190–9.

24. Ganguly SS, Fiore LS, Sims JT, Friend JW, Srinivasan D, Thacker MA,et al. c-Abl and Arg are activated in human primary melanomas,promote melanoma cell invasion via distinct pathways, and drivemetastatic progression. Oncogene 2012;31:1804–16.

25. Srinivasan D, Sims JT, Plattner R. Aggressive breast cancer cells aredependent on activated Abl kinases for proliferation, anchorage-inde-pendent growth and survival. Oncogene 2008;27:1095–105.

26. Baley PA, Yoshida K, Qian W, Sehgal I, Thompson TC. Progression toandrogen insensitivity in a novel in vitro mouse model for prostatecancer. J Steroid Biochem Mol Biol 1995;52:403–13.

27. Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 2008;181:5791–802.

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

29. Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating therecruitment of macrophages into hypoxic areas of tumors and otherischemic tissues. Blood 2004;104:2224–34.

30. Lin H, Lee E, Hestir K, Leo C, Huang M, Bosch E, et al. Discovery of acytokine and its receptor by functional screening of the extracellularproteome. Science 2008;320:807–11.

31. Conway JG, McDonald B, Parham J, Keith B, Rusnak DW, Shaw E,et al. Inhibition of colony-stimulating-factor-1 signaling in vivowith theorally bioavailable cFMS kinase inhibitor GW2580. Proc Natl Acad SciU S A 2005;102:16078–83.

32. Yuan ZM, Huang Y, Ishiko T, Kharbanda S, Weichselbaum R, Kufe D.Regulation of DNA damage-induced apoptosis by the c-Abl tyrosinekinase. Proc Natl Acad Sci U S A 1997;94:1437–40.

33. Chen C, Shang X, Cui L, Xu T, Luo J, Ba X, et al. L-selectin ligation-induced CSF-1 gene transcription is regulated by AP-1 in a c-Ablkinase-dependent manner. Hum Immunol 2008;69:501–9.

34. RockxDA,MasonR, vanHoffenA,BartonMC,Citterio E,BregmanDB,et al. UV-induced inhibition of transcription involves repression oftranscription initiation and phosphorylation of RNApolymerase II. ProcNatl Acad Sci U S A 2000;97:10503–8.

35. Dewar AL, Cambareri AC, Zannettino AC, Miller BL, Doherty KV,Hughes TP, et al. Macrophage colony-stimulating factor receptorc-fms is a novel target of imatinib. Blood 2005;105:3127–32.

36. Ide H, Seligson DB, Memarzadeh S, Xin L, Horvath S, Dubey P, et al.Expression of colony-stimulating factor 1 receptor during prostate

Targeting CSF1R to Block TIMs Improves Efficacy of Radiation

www.aacrjournals.org Cancer Res; 73(9) May 1, 2013 2793

Page 13: CSF1R Signaling Blockade Stanches Tumor-Infiltrating ... · RAW264.7(1.5 105 cells)wereseededincellcultureinserts (8 mm pore size; BD Falcon) in DMEM containing 0.1% FBS with or

development and prostate cancer progression. Proc Natl Acad SciU S A 2002;99:14404–9.

37. Wrobel CN, Debnath J, Lin E, Beausoleil S, Roussel MF, Brugge JS.Autocrine CSF-1R activation promotes Src-dependent disruption ofmammary epithelial architecture. J Cell Biol 2004;165:263–73.

38. Wei S, Nandi S, Chitu V, Yeung YG, Yu W, Huang M, et al. Functionaloverlap but differential expression of CSF-1 and IL-34 in their CSF-1receptor-mediated regulation of myeloid cells. J Leukoc Biol 2010;88:495–505.

39. Sawanobori Y, Ueha S, Kurachi M, Shimaoka T, Talmadge JE, Abe J,et al. Chemokine-mediated rapid turnover of myeloid-derived sup-pressor cells in tumor-bearing mice. Blood 2008;111:5457–66.

40. Li X, Loberg R, Liao J, Ying C, Snyder LA, Pienta KJ, et al. A destructivecascademediated byCCL2 facilitates prostate cancer growth in bone.Cancer Res 2009;69:1685–92.

41. El Chartouni C, Benner C, Eigner M, Lichtinger M, Rehli M. Transcrip-tional effects of colony-stimulating factor-1 in mouse macrophages.Immunobiology 2010;215:466–74.

42. Wittrant Y, Bhandari BS, Abboud H, Benson N, Woodruff K, MacDou-gall M, et al. PDGF up-regulates CSF-1 gene transcription in amelo-blast-like cells. J Dent Res 2008;87:33–8.

43. Tsuchimoto D, Tojo A, Asano S. A mechanism of transcriptionalregulation of the CSF-1 gene by interferon-gamma. Immunol Invest2004;33:397–405.

44. Harrington M, Konicek BW, Xia XL, Song A. Transcriptional regu-lation of the mouse CSF-1 gene. Mol Reprod Dev 1997;46:39–44;discussion -5.

45. Song Z, Wang M, Wang X, Pan X, Liu W, Hao S, et al. Nuclear actin isinvolved in the regulation of CSF1 gene transcription in a chromatinrequired, BRG1 independentmanner. J Cell Biochem2007;102:403–11.

46. Hamer G, Gademan IS, Kal HB, de Rooij DG. Role for c-Abl and p73in the radiation response of male germ cells. Oncogene 2001;20:4298–304.

47. Kharbanda S, Bharti A, Pei D, Wang J, Pandey P, Ren R, et al. Thestress response to ionizing radiation involoves c-Abl-dependentphosphorylation of SHPTP1. Proc Natl Acad Sci U S A 1996;93:6898–901.

48. Baskaran R, Wood LD, Whitaker LL, Canman CE, Morgan SE, Xu Y,et al. Ataxia telangiectasia mutant protein activates c-Abl tyrosinekinase in response to ionizing radiation. Nature 1997;387:516–9.

49. Lewis JM, Baskaran R, Taagepera S, Schwartz MA, Wang JY. Integrinregulation of c-Abl tyrosine kinase activity and cytoplasmic-nucleartransport. Proc Natl Acad Sci U S A 1996;93:15174–9.

50. Kipreos ET, Wang JY. Cell cycle-regulated binding of c-Abl tyrosinekinase to DNA. Science 1992;256:382–5.

51. Jing Y,WangM, TangW,Qi T, GuC, Hao S, et al. c-Abl tyrosine kinaseactivates p21 transcription via interaction with p53. J Biochem 2007;141:621–6.

Xu et al.

Cancer Res; 73(9) May 1, 2013 Cancer Research2794