combination of cd40 agonism and csf-1r blockade ...research article combination of cd40 agonism and...

14
Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor Immunity Karla R. Wiehagen 1 , Natasha M. Girgis 1,2 , Douglas H. Yamada 1 , Andressa A. Smith 1 , Szeman Ruby Chan 1 , Iqbal S. Grewal 1 , Michael Quigley 1,3 , and Raluca I. Verona 1 Abstract Efcacious antitumor immune responses must overcome multiple suppressive mechanisms in the tumor micro- environment to control cancer progression. In this study, we demonstrate that dual targeting of suppressive myeloid populations by inhibiting CSF-1/CSF-1R signaling and activa- tion of antigen-presenting cells with agonist anti-CD40 treatment confers superior antitumor efcacy and increased survival compared with monotherapy treatment in preclinical tumor models. Concurrent CSF-1R blockade and CD40 agonism lead to profound changes in the composition of immune inltrates, causing an overall decrease in immuno- suppressive cells and a shift toward a more inammatory milieu. Anti-CD40/antiCSF-1Rtreated tumors contain decreased tumor-associated macrophages and Foxp3 þ regula- tory T cells. This combination approach increases maturation and differentiation of proinammatory macrophages and den- dritic cells and also drives potent priming of effector T cells in draining lymph nodes. As a result, tumor-inltrating effector T cells exhibit improved responses to tumor antigen rechal- lenge. These studies show that combining therapeutic approaches may simultaneously remove inhibitory immune populations and sustain endogenous antitumor immune responses to successfully impair cancer progression. Cancer Immunol Res; 5(12); 110921. Ó2017 AACR. Introduction To escape detection by the immune system, tumors foster an immunosuppressive environment and produce signals that sub- due antitumor immune responses (1). Tumor-associated macro- phages (TAMs) are a heterogeneous population of myeloid cells that differentiate in the tumor microenvironment and become sensitized to tumor-derived suppressive signals (2). TAMs, in turn, act to prevent local immune responses by inefciently presenting antigen and generating suppressive signals such as IL10, indolea- mine-pyrrole 2,3-dioxygenase (IDO), and TGFb that inhibit lym- phocyte function and metabolism (3). Therefore, for adaptive immune responses to persist, tumor-inltrating lymphocytes (TILs) must overcome a suppressive cytokine milieu and mechan- isms of tolerance propagated by TAMs to successfully attack cancerous cells. CSF-1 (M-CSF) is a cytokine that supports differentiation, proliferation, and function of monocyte and macrophage popu- lations (4). In tumors, CSF-1 promotes immune suppression by expanding and promoting differentiation of myeloid-derived suppressor cells (MDSC) and alternatively activated MHC II lo TAMs that express its cognate receptor, CSF-1R (5, 6). High CSF- 1R expression on inltrating immune cells in the TME correlates with immune dysfunction and increased immunosuppression (7, 8), as well as poor prognosis in breast, ovarian, pancreatic cancers, and lymphoma (911). Although inhibiting the CSF-1/ CSF-1R axis alone constrains tumor growth in preclinical models by partially decreasing TAMs and encouraging T-cell inltration (1216), it has often proven insufcient to clear tumors. Pairing CSF-1R blockade with chemotherapeutic agents or checkpoint inhibitors has enhanced efcacy of the monotherapy activity of CSF-1R (17). CD40 is a highly conserved costimulatory receptor expressed on antigen-presenting cells (APCs) including dendritic cells (DC), macrophages, monocytes, B cells, as well as a number of non- hematopoietic cell types and some tumor cells (18, 19). The ligand for CD40 (CD40L) is expressed on activated T cells and B cells and, under inammatory conditions, is upregulated on innate immune cells as well as endothelial and epithelial cells (2024). CD40 ligation drives "licensing" (maturation and acti- vation) of APCs, enabling effective antigen presentation and stimulation of CD4 þ Th1 and CD8 þ T cells. This is a key step in mounting antitumor responses, as supported by emerging preclinical and clinical data with agonist anti-CD40 in multiple tumor types (18, 20, 25). However, a number of studies have shown that potency of CD40 agonists can be enhanced by combinations with other immune modulatory therapies such as TLR agonists, cytokines including type I interferons and IL2, adoptive cell transfer, and chemotherapy (18, 2630). A study 1 Janssen Research and Development, Spring House, Pennsylvania. 2 Constella- tion Pharmaceuticals, Cambridge, Massachusetts. 3 Bristol-Myers Squibb, Prin- ceton, New Jersey. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). Corresponding Author: Raluca I. Verona, Janssen R&D, 1400 McKean Road, Spring House, PA 19477. Phone: 215 628-5211; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-17-0258 Ó2017 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 1109 on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Upload: others

Post on 17-Aug-2020

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

Research Article

Combination of CD40 Agonism and CSF-1RBlockade Reconditions Tumor-AssociatedMacrophages and Drives Potent AntitumorImmunityKarla R.Wiehagen1, Natasha M. Girgis1,2, Douglas H. Yamada1, Andressa A. Smith1,Szeman Ruby Chan1, Iqbal S. Grewal1, Michael Quigley1,3, and Raluca I. Verona1

Abstract

Efficacious antitumor immune responses must overcomemultiple suppressive mechanisms in the tumor micro-environment to control cancer progression. In this study,we demonstrate that dual targeting of suppressive myeloidpopulations by inhibiting CSF-1/CSF-1R signaling and activa-tion of antigen-presenting cells with agonist anti-CD40treatment confers superior antitumor efficacy and increasedsurvival compared with monotherapy treatment in preclinicaltumor models. Concurrent CSF-1R blockade and CD40agonism lead to profound changes in the compositionof immune infiltrates, causing an overall decrease in immuno-suppressive cells and a shift toward a more inflammatory

milieu. Anti-CD40/anti–CSF-1R–treated tumors containdecreased tumor-associated macrophages and Foxp3þ regula-tory T cells. This combination approach increases maturationand differentiation of proinflammatory macrophages and den-dritic cells and also drives potent priming of effector T cellsin draining lymph nodes. As a result, tumor-infiltrating effectorT cells exhibit improved responses to tumor antigen rechal-lenge. These studies show that combining therapeuticapproaches may simultaneously remove inhibitory immunepopulations and sustain endogenous antitumor immuneresponses to successfully impair cancer progression. CancerImmunol Res; 5(12); 1109–21. �2017 AACR.

IntroductionTo escape detection by the immune system, tumors foster an

immunosuppressive environment and produce signals that sub-due antitumor immune responses (1). Tumor-associated macro-phages (TAMs) are a heterogeneous population of myeloid cellsthat differentiate in the tumor microenvironment and becomesensitized to tumor-derived suppressive signals (2). TAMs, in turn,act to prevent local immune responses by inefficiently presentingantigen and generating suppressive signals such as IL10, indolea-mine-pyrrole 2,3-dioxygenase (IDO), and TGFb that inhibit lym-phocyte function and metabolism (3). Therefore, for adaptiveimmune responses to persist, tumor-infiltrating lymphocytes(TILs)must overcome a suppressive cytokinemilieu andmechan-isms of tolerance propagated by TAMs to successfully attackcancerous cells.

CSF-1 (M-CSF) is a cytokine that supports differentiation,proliferation, and function of monocyte and macrophage popu-lations (4). In tumors, CSF-1 promotes immune suppression by

expanding and promoting differentiation of myeloid-derivedsuppressor cells (MDSC) and alternatively activated MHC IIlo

TAMs that express its cognate receptor, CSF-1R (5, 6). High CSF-1R expression on infiltrating immune cells in the TME correlateswith immune dysfunction and increased immunosuppression(7, 8), as well as poor prognosis in breast, ovarian, pancreaticcancers, and lymphoma (9–11). Although inhibiting the CSF-1/CSF-1R axis alone constrains tumor growth in preclinical modelsby partially decreasing TAMs and encouraging T-cell infiltration(12–16), it has often proven insufficient to clear tumors. PairingCSF-1R blockade with chemotherapeutic agents or checkpointinhibitors has enhanced efficacy of the monotherapy activity ofCSF-1R (17).

CD40 is a highly conserved costimulatory receptor expressedon antigen-presenting cells (APCs) including dendritic cells (DC),macrophages, monocytes, B cells, as well as a number of non-hematopoietic cell types and some tumor cells (18, 19). Theligand for CD40 (CD40L) is expressed on activated T cells andB cells and, under inflammatory conditions, is upregulated oninnate immune cells as well as endothelial and epithelial cells(20–24). CD40 ligation drives "licensing" (maturation and acti-vation) of APCs, enabling effective antigen presentation andstimulation of CD4þ Th1 and CD8þ T cells. This is a key stepin mounting antitumor responses, as supported by emergingpreclinical and clinical data with agonist anti-CD40 in multipletumor types (18, 20, 25). However, a number of studies haveshown that potency of CD40 agonists can be enhanced bycombinations with other immune modulatory therapies such asTLR agonists, cytokines including type I interferons and IL2,adoptive cell transfer, and chemotherapy (18, 26–30). A study

1Janssen Research and Development, Spring House, Pennsylvania. 2Constella-tion Pharmaceuticals, Cambridge, Massachusetts. 3Bristol-Myers Squibb, Prin-ceton, New Jersey.

Note: Supplementary data for this article are available at Cancer ImmunologyResearch Online (http://cancerimmunolres.aacrjournals.org/).

Corresponding Author: Raluca I. Verona, Janssen R&D, 1400 McKean Road,Spring House, PA 19477. Phone: 215 628-5211; E-mail: [email protected]

doi: 10.1158/2326-6066.CIR-17-0258

�2017 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org 1109

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 2: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

also demonstrated that CD40 agonism following CSF-1R block-ade leads to enhanced antitumor efficacy, suggesting these twotherapies may cooperate when coadministered (30).

In this study, we sought to investigate whether dual targeting ofsuppressive myeloid populations via CSF-1/CSF-1R inhibitionand APC activation with agonist anti-CD40 treatment confersenhanced antitumor efficacy compared with monotherapy treat-ments. Tumor profiling after anti-CD40/anti–CSF-1R treatmentrevealed significant changes in the composition of CD11bþ

myeloid populations, decreased frequencies of regulatory Foxp3þ

T cells (Treg), and increased infiltrationof effector TILs. Analysis ofthe cytokinemilieu in combination-treated tumors demonstratedan inflammatory TME, which corresponded with improved con-trol of tumor burden. The combination of starving TAMs whileactivating APCs resulted in a cascade of proinflammatoryresponses and significant gains in effector T-cell responses andtumor clearance. Thus, the combination of anti-CD40 and anti–CSF-1R simultaneously removes inhibitory immune populationsand drives endogenous antitumor immune responses to improvetumor clearance and significantly lengthen overall survival.

Materials and MethodsTumor cell lines

CT26 andMC-38 cell lineswere obtained fromATCC.Cell lineswere authenticated and certified by ATCC (February 2015) andregularly tested by Analytical Biological Services, Inc. MAP/RAPPCR results (February 2016) were negative, and <1 EU/mL bylimulus amebocyte lysate (LAL; ATCC,ABS). CT26andMC-38 celllineswere cultured in vitrounder conditions specifiedby the ATCCand passaged up to 3 times before implant.

Syngeneic tumor studiesIn all in vivo studies, age-matched female mice from Charles

River Laboratories were implanted subcutaneously with 5 � 105

cultured cells of the indicated tumor line suspended in PBS.Animal welfare (grooming, activity) was monitored daily, andanimal weights and caliper measurements to calculate tumorvolume (L � L � W/2) were collected at least twice per weekuntil end of the study or until tumor burden limit approached2,000 mm3. All studies were conducted in accordance withinternal Institutional AnimalCare andUseCommittees of JanssenR&D and in compliance with federal guidelines.

Female Balb/c mice, 6 to 9 weeks old, bearing subcutaneousCT26 tumors 50 to 100 mm3 in volume were randomized intogroups of 10 to 12mice and treatedwith 200 mg anti-IgG2a (clone2A3; BioXcell), 100 mg anti-CD40 (clone FGK4.5; BioXcell), or200 mg anti-CSF1R (clone AFS98; BioXcell) per dose. The com-bination group received 100mg FGK4.5 and200mgAFS98 given atthe same time but in separate injections on opposite sides of theabdomen (treatment was not premixed before injection). Doseswere spaced four days apart for a total of three doses. CD8þ T-celldepletion studies were carried out with randomized groups ofmice bearing CT26 tumors, which were treated with 200 mg anti-CD8 (clone 2.43; BioXcell) or PBS intraperitoneally 24 hoursbefore immunotherapy was initiated and concurrently withimmunotherapy. Dosing began day 11 and continued everyfour days as described above. For MC-38 tumor studies, femaleC57BL/6 mice, 6 to 9 weeks old, harboring MC-38 tumors 75 to125 mm3 in volume were randomized and treated identically tothe CT26 studies.

Tumor and tissue processingTumors were excised and dissociated into single-cell suspen-

sions using theMiltenyi TumorDissociationKit formurine tissues(catalog no. 130-096-730) per the manufacturer's instructions.Sampleswerefiltered andmechanically processed through70-mmnylon filters. The following draining lymph nodes (dLN) werepooled per mouse: ipsilateral brachial, axillary, and inguinallymph nodes. Spleens or pooled dLNs were processed through70-mm nylon filters with syringe plungers. Samples were stainedfor flow cytometry the same day, and remaining suspensions werestored on ice at 4�C overnight for subsequent analyses.

Flow cytometry and antibodiesFor flow cytometry profiling, 2� 106 tumor samples or 1� 106

lymphoid tissue samples were plated in tissue culture nontreatedpolystyrene round bottom96-well plates (Corning). Viability wasassessed by Live/Dead Fixable Aqua Dead Cell Stain (Thermo-Fisher). Antibody cocktails were assembled with the followingantibodies:

Myeloid stains. FITC-CD206 (clone C068C2), PE-IL4Ra (cloneI015F8), PE-CD115 (clone AFS98), PE-NOS2 (clone 5C1B52),PE-CCR7 (clone 4B12), PE-IL10R (clone 1B1.3a), PE-IL10 (cloneJES3-12G8), PE-IL6 (clone MP5-20F3), PE-CD103 (clone 2E7),PerCPCy5.5-F4/80 (clone BM8), PerCPCy5.5-CD80 (clone 16-10A1), PerCPCy5.5-TNFa (clone MP6-XT22), PECy7-CD103(clone 2E7), APC-IDO (clone 2E2/IDO1), APC-TGF/LAP (cloneTW7-16B4), APC-CD86 (clone GL-1), APC-F4/80 (clone BM8),AF700-Ly6C (clone HK1.4), APCCy7-I-A:I-E (MHC II; clone M5/114.15.2), BV421-Ly6G (clone 1A8), BV421-TGFb/LAP (cloneTW7-16B4), BV605-CD24 (clone 30-F1); BV650- or BV711-CD11b (clone M1/70), BV786-CD11c (clone N418), BUV395-CD45 (clone 30-F11), and dump stain BV711 for CD90.2 (clone53-2.1) or TCRb (cloneH57-597), Siglec-F (clone E50-2440), andgdTCR (clone V65).

Lymphoid stains. FITC-Granzyme B (clone GB11), FITC-CD8(clone 53-6.7), PE-IFNg (clone XMG1.2), PE–T-bet (clone4B10), PerCPCy5.5-CD4 (clone GK1.5), PECy7-Foxp3 (cloneFJK-16s), PECy7-Eomes (clone Dan11mag), APC-IFNg (cloneXMG1.2), APC–TIM-3 (clone RMT3-23 or B8.2C12), AF700-CD90.2 (clone 53-2.1), AF700-CD4 (clone GK1.5), AF700-CD44(clone IM7), APCCy7-CD90.2 (clone 53-2.1), APCCy7-CD4(clone GK1.5), Pacific Blue-Foxp3 (clone FJK-16s), BV605–PD-1 (clone 29F.1A12), BV650-CD4, BV711-dump, BV786-CD8,BV786-CD62L, BUV395-CD45 (clone 30-F11), and BV711 forCD11b, CD11c, CD49b (clone DX5), gdTCR, and Siglec-F. AH1-tetramer: PE-conjugated H-2Ld MuLV-gp70 tetramer (SPSY-VYHQF; MBL International Corporation).

All antibodies were sourced from BD Biosciences, Biolegend,R&Dsystems, or eBioscience. Intracellular stainingwas performedafter cellular fixation and permeabilization using either the BDCell Fix (catalog no. 554714) or eBioscience Transcription Factor/Foxp3 staining buffer set (catalog no. 00-5523-00) according tothe manufacturer's instructions. All flow cytometry data werecollected on BD Fortessa instruments and analyzed by FlowJosoftware v9.9.4 (TreeStar).

Ex vivo functional assaysT-cell stimulation. Tumor and lymphoid tissues were processedinto single-cell suspensions, and 2 � 106 tumor samples or

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1110

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 3: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

1 � 106 lymphoid tissue samples were plated in tissue culturenontreated polystyrene round bottom 96-well plates (Corn-ing). Stimulation cocktail containing BD GolgiPlug (catalogno. 555029) and BD GolgiStop (catalog no. 554724) wasprepared per manufacturer's instruction (final concentration1 mL/mL for each), with or without AH1 peptide (final con-centration of 5 mg/mL). Cell stimulation cocktail (500�;catalog no. 00-4970-93; eBiosciences) served as the positivecontrol. All conditions were incubated at 37�C for four hours.

TAM intracellular staining. Tumor suspensions were plated 2 �106perwellwithGolgiStop andGolgiPlug as stated above for fourhours at 37�C. Cells were surfaced-stained, fixed, and permeabi-lized. Sampleswere then stained intracellularly for IL6, TNFa, andnitric oxide synthase 2 (NOS2, iNOS). TAMs (viable, Dump�

CD11bþ Ly6G� Ly6Clo F4/80þ) were assessed for IL6, TNFa, andNOS2.

TME cytokine measurementThe day after treatment ended, tumors were weighed and

homogenized with a MP FastPrep-24 instrument at 4.5 m/s for45 seconds. Samples were filtered, centrifuged to collect thesoluble fraction, and stored at �80�C. Analytes were measuredusing the Meso Scale Diagnostics Biomarker Group 1 Mouse 27-plex kit (catalog no. K15069L-2; Meso Scale Diagnostics). Resultswere analyzed by the MSD Discovery Workbench software. Cali-brators 5, 7, and 8 were used as per manufacturer's instructionsand served as controls.

Statistics and graphing with GraphPad Prism v6On all graphs, mean and standard error of the mean (SEM)

are shown unless otherwise indicated. Differences betweentreatment groups measured at endpoint were determined usinganalysis of variance (ANOVA) followed by post hoc all-pairwisecomparison tests with a Tukey adjustment for multiple com-parisons. Survival analysis was performed using log-rank test.Statistical analyses were run using GraphPad Prism version 6(GraphPad Software). Statistical significance was accepted at P< 0.05. All comparisons stated in results are versus isotype,unless otherwise specified.

ResultsCombined anti-CD40 and CSF-1R blockade improves tumorcontrol and survival

Tumor-infiltrating myeloid cells, including TAMs, suppressantitumor immunity, and these populations rely on CSF-1/CSF-1R signaling for survival, development, and differentiation.To determine which myeloid subsets depend on the CSF-1/CSF-1R axis, CSF-1R expression was assessed in distinct populationsof myeloid cells in established, untreated CT26 tumors. We useda gating strategy previously described by Movahedi and collea-gues, which uses Ly6C and MHC class II (MHC II) expression todiscriminate populations in the Ly6Chi and Ly6Clo gates (31, 32).Specifically, in this analysis, MHC II expression distinguishesLy6Chi monocytes (Ly6Chi MHC IIlo) from Ly6Cint TAMs (Ly6Cint

MHC IIhi; Supplementary Fig. S1A). Ly6Clo F4/80þ TAMs can beseparated into proinflammatory MHC IIhi TAMs (Ly6Clo F4/80þ

MHC IIhi) and alternatively activated, suppressive MHC IIlo

macrophages (Ly6Clo F4/80þ MHC IIlo; Supplementary Fig.S1A). Comparedwith isotype control staining,DCs, Ly6CintMHCIIhi TAMs, Ly6Clo MHC IIhi, and MHC IIlo TAMs demonstrated

increased surface expression of CSF-1R, whereas Ly6Gþ neutro-philsmaintained lower expression (Supplementary Fig. S1B). Thehighest frequencies of CSF-1Rhi cells were detected in Ly6Glo

Ly6CintMHC IIhi and Ly6Clo TAMs,with no significant differencesin CSF-1R expression between MHC IIhi or MHC IIlo TAMs(Supplementary Fig. S1B). Together, these expression profilescorrelate with data in the literature and suggest mature TAMswould be sensitive to CSF-1R blockade.

We hypothesized that blocking the CSF-1/CSF-1R pathway inCT26 tumors would result in significant changes in TAMs, andthat couplingCSF-1R inhibitionwithCD40 agonismwould resultin enhanced antitumor activity. Anti–CSF-1Rmonotherapy failedto significantly control CT26 tumor burden compared with iso-type control, whereas anti-CD40 agonist treatment delayed tumorgrowth (Fig. 1). Treatment with the combination resulted in themost significant control of tumor growth and extended survival(Fig. 1A–C and E). Similar results were seen in MC-38 tumors.Combination anti-CD40/anti–CSF-1R treatment elicited superiorcontrol of tumor growth compared with isotype ormonotherapy-treated groups. As in CT26 studies, anti–CSF-1R monotherapytreatment failed to limit MC-38 tumor burden, whereas anti-CD40 monotherapy significantly controlled progression but notto the extent of combination treatment (Fig. 1F and G). Althoughthese therapies targeted myeloid cells that expressed CD40 andCSF-1R, depletion of CD8þ T cells in the CT26 model abrogatedthe antitumor efficacy of anti-CD40 monotherapy and combina-tion treatment, as well as eliminated survival benefits observedwith combination treatment (Fig. 1C–E), suggesting adaptiveimmune responses are critical for these responses.

Combination immunotherapy results in reduced TAMs andalters myeloid populations

To understand mechanisms underlying the improved efficacyand survivalmediated by anti-CD40 and anti–CSF-1R therapy,weprofiled the immune composition of tumors harvested at twotime points during treatment: 24 hours after the first dose and 24hours after thefinal dose. At both timepoints, groups that receivedanti-CD40 or combination anti-CD40/anti–CSF-1R exhibitedsimilar frequencies of CD45þ hematopoietic cells compared withisotype controls. However, tumors treated with anti–CSF-1Rmonotherapy contained approximately 10% fewer CD45þ cells(Supplementary Fig. S2A). No changes were observed in thefrequencies of CD11bþ cells, targeted by anti-CD40 and anti–CSF-1R, at either time point across treatment groups (Supple-mentary Fig. S2B).

We used Ly6C and Ly6G expression to subdivide CD11bþ

populations into monocytes, macrophages, and neutrophils andidentified broad changes in the immune infiltrates across groups.Combination-treated tumors exhibited the greatest reduction inLy6Gþ cells, which includes neutrophils and polymorphonuclearMDSCs, and Ly6Clo F4/80þ TAMs (Fig. 2A and B). In contrast,Ly6Chi cells increased significantly over isotype in all treatmentgroups. The greatest increase was seen in combination-treatedtumors, which was also significantly greater than both mono-therapies (Fig. 2B).

According to this model of subset analysis, the Ly6Cþ fractionconsists of bothmonocytes and Ly6Cint tumor–associatedmacro-phages, but the accumulation of Ly6Chi cells in combination-treated tumors was not due to proportionate increases of both celltypes (Fig. 2C–E). Analysis of MHC II expression revealed anonsignificant increase in frequencies of Ly6Chi MHC IIlo

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1111

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 4: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

monocytes in monotherapy- and combination-treated tumors(Fig. 2D) and that increased Ly6Cint MHC IIhi TAMs were pri-marily responsible for the overall increase in Ly6Cþ cells (Fig. 2Dand E). Both monotherapies elicited increases in Ly6Cint macro-phages within tumors as well, with anti-CD40 treatment eliciting2-fold more Ly6Cint macrophages than isotype treatment (Fig.2E). The most significant changes were found in combination-treated tumors, where 4-foldmore Ly6Cint macrophages accumu-lated over the course of treatment compared with isotype controlsand were significantly higher than both monotherapies (Fig. 2E).

Approximately 70%ofCD11bþLy6G�myeloid-lineage cells inisotype-treated tumors consisted of Ly6Clo TAMs, which weredetected in both MHC IIhi and MHC IIlo populations (Fig. 2C, Fand G). All treatment approaches decreased frequencies of MHCIIlo TAMs compared with the isotype group, with the fewest MHCIIlo TAMs detected after combination treatment (Fig. 2C and F).Relative to isotype controls, we observed a 33%, 50%, and 70%reduction in MHC IIlo suppressive TAMs in anti–CSF-1R, anti-CD40, and combination-treated groups, respectively (Fig. 2F).Frequencies of MHC IIhi TAMs in monotherapy groups were notsignificantly different from isotype-treated groups, whereas nearlyhalf of MHC IIhi TAMs were reduced with combination treatment(Fig. 2G). Together, these data indicate that anti–CSF-1R andCD40 agonism differentially affect TAM and monocyte popula-tions and that anti-CD40/anti–CSF-1R combination treatmentsignificantly skews the composition of tumor-infiltratingCD11bþ

myeloid cells.

CD40 agonism or combination immunotherapy increasesproinflammatory TAMs

Low frequencies of Ly6Clo TAM subsets led us to investigatethe polarization status of the total Ly6Clo F4/80þ TAM popu-lation. We first assessed expression of cell-surface markersassociated with inflammatory MHC IIhi TAMs and alternativelyactivated MHC IIlo macrophages on total Ly6Clo F4/80þ TAMsafter treatment concluded. Typically, alternatively activatedTAMs have high expression of IL4Ra, MMR (CD206), MerTK,and IL10R, consistent with MHC IIlo TAM sensitivity to sup-pressive signals. TAMs from tumors treated with anti-CD40 orcombination anti-CD40/anti–CSF-1R exhibited decreasedexpression of IL4Ra, IL10R, and CD206 compared with anti-CSF-1R and isotype groups (Fig. 3A–D). Expression of inhib-itory ligands, PD-L1 and PD-L2, did not change significantlyacross treatment groups in the majority of the experimentsperformed (Fig. 3E and F). In contrast, TAMs in anti-CD40 andcombination-treated tumors expressed markers consistent withactivation (CD83) and maturation (CD80 and CD86) of proin-flammatory MHC IIhi macrophages (Fig. 3G–I), with highestexpression observed in anti-CD40 and combination-treatedgroups, consistent with CD40 ligation-induced activation(ref. 33; Fig. 3G–I). Anti–CSF-1R monotherapy elicitedincreased expression of CD80 and CD83 markers comparedwith isotype controls, but did not affect activation markers inalternatively activated MHC IIlo TAMs. These data suggest thatcombination treatment aids polarization of remaining TAM

Figure 1.

Combination immunotherapy witha CD40 agonist and CSF-1R blockade controls tumor burden.A, Tumor volumes over time of Balb/c mice implantedwith CT26 atday 0. Dates of treatment indicated by arrows. �, P < 0.05; �� , P < 0.005; ��� , P < 0.0003; ���� , P < 0.0001. B, Individual tumor volume traces per mouse.n ¼ 12 per group. Data are representative of four independent experiments. C and D, Tumor volumes of Balb/c mice implanted with CT26 tumors with andwithout CD8þ T cell depletion. C, Isotype-control antibody (intact CD8þ T cells) and D, anti-CD8 antibody. Data are representative of two independentexperiments. E, Kaplan–Meier analysis of survival. Pairwise comparison of tumor growth with anti-CD40 or combination treatment P ¼ 0.011. F, Tumor volumes ofC57BL/6 mice implanted with MC-38 at day 0. Dates of treatment indicated by arrows. � , P < 0.05; �� , P < 0.005; ��� , P < 0.0003; ���� , P < 0.0001. G, Individualtumor volume traces per mouse. n ¼ 10 per group. F and G, Data are representative of three independent experiments. A–G, Isotype (white), anti–CSF-1R (blue),anti-CD40 (green), and combination (red). Significance between treatment groups measured at endpoint was determined using ANOVA followed bypost hoc all-pairwise comparison tests with a Tukey adjustment for multiple comparisons.

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1112

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 5: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

populations from the MHC IIlo suppressive TAM phenotypetoward MHC IIhi proinflammatory TAMs.

Based on expression of markers affiliated with proinflamma-tory TAMs, we hypothesized that macrophages from anti-CD40and combination anti–CD40/anti–CSF-1R would be primed togenerate proinflammatory signals. Anti-CD40 and anti-CD40/anti–CSF-1R treatment induced increased frequencies of TNFaþ

andNOS2þTAMs comparedwith isotype- or anti–CSF-1R-treatedgroups (Fig. 3J–L). Latency-associated protein (LAP) was evalu-

ated as a surrogate for the immunosuppressive cytokine TGFb.Anti-CD40 and combination treatment decreased the fraction ofTAMs poised to produce TGFb in tumors, whereas anti–CSF-1Rmonotherapy was not appreciably different from isotype treat-ment (Fig. 3M).

IL10/IL10R signaling is associated with suppression of antitu-mor immune responses (34, 35). Reduced frequencies of IL10-producing TAMs were observed in all treated groups comparedwith isotype controls (Fig. 3N), with only the combination-

Figure 2.

CD40 and combination immunotherapyskews the composition of monocyte andmacrophage populations. A,Representative plots of monocytes,macrophages, and neutrophils. Events aregated on live, singlet, size- and lineage-excluded CD45þ CD11bþ tumor-infiltrating cells. B, Frequency of Ly6Gþ,Ly6Chi Ly6G�, or Ly6Clo Ly6G� cells.Compiled from four independentexperiments. n � 15 for each group.� , P < 0.05; �� , P < 0.01; ��� , P < 0.005;���� , P < 0.0005. C, Representative plotsof monocytes, Ly6Cint macrophages, andTAMs. Populations depicted correlatewith the schematic (far right). D–G,Compiled frequencies of indicatedpopulations across groups. n � 5.� , P < 0.05; �� , P� 0.003; ��� , P� 0.0006;���� , P < 0.0001. (A–G) isotype (white),anti–CSF-1R (blue), anti-CD40 (green),combination (red). Significance betweentreatment groups measured at endpointwas determined using ANOVA followedby post hoc all-pairwise comparison testswith a Tukey adjustment for multiplecomparisons. Data are representativeof three experiments.

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1113

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 6: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

treated group reaching significance. Therefore, combination treat-ment reduced levels of LAP/TGFb, IL10, and IL10R in suppressiveTAM populations, increased frequencies of activated MHC IIhi

macrophages, and skewed the TME toward a proinflammatoryphenotype.

Combination therapy induces accumulation of activatedCD103þ DCs in dLNs

Efficient antigen presentation by DCs is critical to primingeffective antitumor T-cell responses, andCD40 agonism is knownto drive DC activation and maturation (36, 37). In anti-CD40 or

anti–CD40/anti-CSF-1R-treated tumors harvested after the end oftreatment, CD83, CD80, and CD86 expression was increased onintratumoral DCs compared with isotype-treated DCs, whereasanti–CSF-1R monotherapy did not significantly change the acti-vation status of these APCs (Fig. 4E). Lower frequencies of DCs(CD24hi MHC IIhi CD11cþ) were observed in tumors from anti-CD40 and combination-treated groups compared with isotypecontrols 24 hours after start of treatment (Fig. 4A). We hypoth-esized that DCs could be trafficking to lymphoid tissues afteractivation via CD40 agonism and, therefore, examined dLNs 24hours after the initial dose. Consistent with our hypothesis,

Figure 3.

Combination therapy promotes aproinflammatory phenotype inremaining TAMs. A–I, Geometric meanfluorescent intensity (gMFI) of totalTAMs (CD11bþ Ly6G� Ly6Clo F4/80þ)from tumors harvested after treatmentregimen. gMFI of (A) IL4Ra, (B) IL10R,(C) CD206, (D) MerTK, (E) PD-L1,(F) PD-L2, (G) CD83, (H) CD80, and(I) CD86 on TAMs. � , P < 0.05;�� , P � 0.003; ��� , P � 0.0006;���� ,P<0.0001. J, Intracellular stainingfor cytokine or effector moleculesTNFa and NOS2. Relative frequenciesof TAMs stained for (K) NOS2,(L) TNFa, (M) LAP (pro-TGFb), and(N) IL10. A–N, Isotype (white),anti–CSF-1R (blue), anti-CD40 (green),combination (red). Significancebetween treatment groups measuredat endpoint were determined usingANOVA followed by post hocall-pairwise comparison tests with aTukey adjustment for multiplecomparisons. Data are representativeof three experiments.

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1114

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 7: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

Figure 4.

In dLNs, CD103þ DCs express increased markers ofactivation and accumulate in dLN after one dose.Twenty-four hours after initial dosing, mice weresacrificed and tumors and dLNs were harvested. Aand B, Frequency of DCs (CD24hi MHC IIhi CD11cþ) oftotal CD45þ infiltrates in (A) tumors or (B) dLNs. Cand D, Frequency of CD103þ DCs of total DCs in (C)tumor or (D) dLNs. E, Expression of activationmarkers on intratumoral DCs at the conclusion oftreatment. F–H, Expression of activation markers onDCs in the dLNs from animals treated with isotype(gray filled histogram), anti–CSF-1R (blue line), anti-CD40 (green line), or combination (red line).F, CCR7 expression and compiled frequencies ofCCR7þ DC in dLNs. G and H, Expression andcompiled gMFI of (G) CD80 or (H) CD86 on DC indLNs. (A–H) � , P < 0.05; ��, P < 0.01; ��� , P < 0.0005;���� , P < 0.0001. (A–H) isotype (white), anti–CSF-1R(blue), anti-CD40 (green), combination (red).Significance between treatment groupsmeasured atendpoint was determined using ANOVA followed bypost hoc all-pairwise comparison tests with a Tukeyadjustment for multiple comparisons. Data arerepresentative of four experiments.

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1115

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 8: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

significant DC accumulation was observed in the dLNs of animalstreatedwith anti-CD40or combination therapy but not isotype oranti–CSF-1R monotherapy (Fig. 4B).

CD103 expression marks a subset of nonlymphoid classicalDCs that have been implicated in cross presentation of tumorantigens andT-cell priming in lymphoid tissues (38, 39). CD103þ

DCs were reduced in tumors one day after anti-CD40 or combi-nation treatment began butmade up a significant fraction of totalDCs in matched dLNs at the same time point (Fig. 4C and D). Ahigher proportion of these DCs in the dLNs expressed markers ofmigration and maturation: CCR7, CD83, CD80, and CD86 (Fig.4F–H). Together, these findings demonstrate anti-CD40 agonismwith CSF-1R blockade drives accelerated maturation and sus-tained activation of DCs and macrophages in tumors andincreased frequencies of mature DCs in draining lymph nodes.

Enhanced CD8þ T-cell effector function in the dLNs ofcombination-treated mice

Although the individual treatments in this combinationapproach targeted myeloid cells, control of tumor growth byanti-CD40/anti-CSF-1R therapy wasmitigated when CD8þ T cellswere depleted in vivo. Therefore, adaptive immune responses arecritical for tumor rejection mediated by anti-CD40/anti-CSF-1Rtherapy. Given the increased accumulation and activation of DCsin the dLNs of anti-CD40 and anti-CD40/anti–CSF-1R-treatedmice, we next investigated whether T cells in dLNs and tumorsmounted improved antitumor effector responses. After treatmentconcluded, increased frequencies of CD8þ T cells were detected inthe dLNs of anti-CD40 and anti-CD40/anti–CSF-1R-treated ani-mals but not after anti–CSF-1R treatment alone (Fig. 5A).

Because CD103þ DCs are capable of trafficking tumor antigento dLNs (38) and increased CD103þDCs were found in the dLNsof combination-treated animals, we sought to evaluate tumorantigen–specific CD8þ T-cell responses in dLNs. CT26 tumor cellsexpress AH1 peptide, which is found in an envelope glycoprotein(gp70) of murine leukemia virus. Tetramer staining revealed anincreased frequency of AH1-specific CD8þ T cells in the dLNs ofanti-CD40/anti–CSF-1R-treated mice after treatment concluded(Fig. 5B), indicating that combination treatment expanded thetumor AH1-specific T cells.

Responses against AH1 serve as a surrogate measurement oftumor antigen–specific CD8þ T-cell activity. To assess T-cellfunction, CD8þ T cells from dLNs were rechallenged with AH1peptide ex vivo. Only CD44hi effector CD8þ T cells from combi-nation-treated mice, but not from either monotherapy, exhibitedimproved T-cell function on rechallenge (Fig. 5C–F). Significantlyhigher frequencies of IFNgþ CD44hi or TNFaþ CD44hi CD8þ Tcells were found in the dLNs of combination-treated animalscompared with monotherapy or isotype groups (Fig. 5C–F).

Along with increased effector function, CD8þ T cells alsodemonstrated increased expression of activation markers andupregulation of checkpoint molecules PD-1, TIM-3, LAG-3, andTIGIT (Supplementary Fig. S3A and S3B). Patterns of increasedPD-1 and TIM-3 coexpression on CD8þ T cells were similar inanti-CD40 and anti-CD40/anti-CSF-1R-treated groups, whereas<1% of CD8þ T cells expressed either PD-1 or TIM-3 in the dLNsof anti–CSF-1R- or isotype-treated groups. CD8þ T cells fromanti-CD40 or combination-treated animals expressed multiplecheckpoint markers, whereas <3% of CD8þ T cells expressedmore than one of these receptors after anti–CSF-1R or isotypetreatment (Supplementary Fig. S3B). More CD8þ T cells from

the dLNs of combination-treated animals exhibited an activat-ed cell-surface phenotype and increased expression of the T-boxtranscription factor (T-bet) and Eomesodermin (Eomes), twotranscription factors critical for effector function (Supplemen-tary Fig. S3C–S3F). Anti-CD40 treatment was sufficient to driveincreased expression of these transcription factors relative toisotype and anti–CSF-1R-treated groups but not to the extentobserved in combination-treated groups. Taken together, anti-CD40/anti–CSF-1R treatment triggers enhanced APC primingresulting in expanded antigen-specific effector CD8þ T cells indLNs.

Combined immunotherapy increasedCD8þ TILs and enhancedCD8þ effector responses

We next investigated how improved antigen responses indLNs of treated animals correlated to effector lymphocyteresponses in tumors. First, we assessed frequencies of total Tcells of CD45þ infiltrating immune cells in tumors. Aftertreatment concluded, anti–CD40/anti-CSF-1R tumors con-tained more T cells on average than isotype-treated tumors,whereas anti–CSF-1R and anti-CD40 monotherapies drovesmaller but reproducible increases in T-cell frequencies (Fig.5G). Within the Thy1þ fraction, CD8þ TIL frequencies weresignificantly increased across all treatment groups comparedwith isotype-treated tumors, with the most significant increasedetected in the anti-CD40/anti–CSF-1R group that exhibitedalmost 40% more CD8þ TILs compared with isotype controls(Fig. 5H). Increased Thy1þ and CD8þ T cells resulted inincreased CD8þ T cells within the total CD45þ immune infil-trate in tumors treated with anti-CD40/anti–CSF-1R therapy(Supplementary Fig. S2C). Moreover, the absolute number ofCD8þ T cells was increased per gram of tumor after combina-tion or anti-CD40 treatment (Supplementary Fig. S2F). Thesedata demonstrate that more CD8þ T cells infiltrate tumors afterCD40 agonism or CSF-1R blockade, but the combination ismore effective than either monotherapy. However, unlike thefindings in dLNs, the proportion of AH1 tetramerþ CD8þ cellswas not significantly altered in tumors across treatment groups(Fig. 5I).

To determine whether intratumoral T-cell effector responsesimproved with combination immunotherapy, we assessed thephenotype and function of CD8þ TILs in CT26 tumors by mea-suring expression of activation markers in total CD8þ TILs andAH1 tetramerþ CD8þ T cells. Similar to CD8þ T cells in dLNs,activation markers and checkpoint molecules, PD-1, TIM-3,TIGIT, and LAG-3, were increased on CD8þ TILs after anti-CD40or combination immunotherapy, relative to isotype-treatedtumors (Supplementary Fig. S3G and S3H). Combination treat-ment led to increased frequencies ofCD8þTILs coexpressing PD-1and TIM-3, and nearly half of CD8þ TILs coexpressed multiplecheckpoint molecules, indicating enhanced effector activation(Supplementary Fig. S3H).

Restimulation with AH1 peptide ex vivo revealed that anti-CD40/anti–CSF-1R combination immunotherapy elicited signif-icantly higher frequencies of antigen-specificCD8þTILs primed toproduce IFNg , TNFa, or granzyme B in response to tumor antigen(Fig. 5J). A higher proportion of CD8þ TILs from combination-treated tumors were also able to mount polyfunctional responsesin response to AH1 peptide (Fig. 5J). Together, these data suggestthat anti-CD40/anti–CSF-1R immunotherapy enhances both thequality and quantity of CD8þ TILs.

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1116

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 9: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

Tumor-infiltrating regulatory T cells are decreased in tumorswith combination treatment

Anti-CD40/anti–CSF-1R treatment resulted in increased fre-quencies and improved function of CD8þ TILs, as well asdecreased frequencies of Foxp3� conventional CD4þ TILs.Although frequencies of CD4þ T cells were decreased in theimmune infiltrate of anti-CD40 or combination-treated tumors,the number of helper T cells per gram of tumor was increased,reflecting overall increased immune infiltration induced by anti-CD40 or combination treatment (Supplementary Fig. S2F). Addi-tionally, the remaining helper CD4þ Foxp3� T cells in both dLNs

and tumors treated with anti-CD40/anti–CSF-1R exhibited sig-nificantly increased activation and effector function. After treat-ment concluded, more CD4þ T cells in combination-treatedanimals produced IFNg , TNFa, and IL2 in unstimulated ex vivocultures (Fig. 6A and B). Anti–CSF-1R monotherapy had a role indriving improved CD4þ helper responses compared with isotypecontrols (Fig. 6B).

Previous reports described decreased immunosuppressiveFoxp3þ Tregs in tumor models after either anti–CSF-1R or anti-CD40 treatment (40, 41). We assessed whether similarchanges were observed in this subset of CD4þ T cells after

Figure 5.

Combination immunotherapy improvedtumor antigen–specific responses in dLNsand tumors. After treatment concluded,tumors were harvested and dLNs werepooled per animal. A, Frequency of CD8þ

T cells in dLNs compiled from threeindependent experiments. n� 12 for eachgroup. B, Frequency of AH1 tetramerþ

CD8þ T cells in dLNs. C, Intracellularstaining for IFNg and TNFa afterrechallenge ex vivo with AH1 peptide.Numbers within gates represent relativefrequencies of total CD44hi CD8þ T cellsfrom dLNs. D–F, Frequencies of CD44hi

CD8þ T cells in dLNs that produced (D)IFNg , (E) TNFa, or (F) IFNg and TNFa inresponse to AH1 peptide rechallenge.A–F: � ,P<0.05; �� ,P<0.01; ��� ,P <0.005;���� , P < 0.0005. Significance wasdetermined using ANOVA followed bypost hoc all-pairwise comparison testswith a Tukey adjustment for multiplecomparisons. Representative of fourindependent experiments. G, Compiledfrequency of Thy1þ cells of total CD45þ

immune infiltrates in tumors. n � 13 foreach group. H, Compiled frequencies ofCD8þ T cells in tumors of Thy1þ events.n � 13 for each group. I, Compiledfrequency of AH1-tetramer antigen-specific CD8þ T cells of total Thy1þ

dump� events in tumors. n � 22 pergroup. J, Compiled frequencies of IFNgþ,TNFaþ, or IFNgþ TNFaþ, or GranzymeBþ CD8þ TILs after rechallenge with AH1peptide. G–J: � , P < 0.05; �� , P � 0.01;��� , P � 0.001; ���� , P < 0.0001. A–J,Isotype (white), anti–CSF-1R (blue),anti-CD40 (green), combination (red).Significance was determined usingANOVA followed by post hoc all-pairwisecomparison tests with a Tukeyadjustment for multiple comparisons.G–I, Data are compiled from threestudies. J, Data are representativeof four experiments.

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1117

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 10: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

anti-CD40/anti–CSF-1R treatment. Consistent with publisheddata, tumors treatedwith either anti–CSF-1Ror anti-CD40mono-therapy consistently exhibited decreased frequencies of CD4þ

Foxp3þ Tregs of total lymphocytes or total CD45þ immune cellsafter treatment concluded (Fig. 6C and E; Supplementary Fig.S2E), and the most dramatic decrease was seen after anti-CD40/

anti–CSF-1R combination treatment, which had a higher ratio ofhelper CD4+ T cells to T regs in combination-treated animals.A 4-fold higher ratio of CD8þ TILs to Tregs was seen incombination-treated tumors compared with isotype controls(Fig. 6F). Although frequencies of AH1 tetramerþ CD8þ TILs didnot significantly change, combination treatment resulted in a

Figure 6.

Combination treatment increasesactivation of tumor-infiltrating CD4þ

helper T cells and significantly reducesFoxp3þ Tregs. After treatmentconcluded, tumors were analyzed forchanges in CD4þ TILs. A, Representativeplots of intracellular staining for IFNg andTNFa by unstimulated CD4þ Foxp3�

TILs. Numbers within gates representrelative frequencies of total CD4þ

Foxp3� TILs. B, Frequencies of CD4þ

Foxp3� helper TILs that produced (left)IFNg , (center) TNFa, or (right) IL2 atbaseline. C, Representative plots ofFoxp3� T cells and Foxp3þ Tregs inCD4þ TILs. Numbers indicate relativefrequency of gated events of totalCD4þ TILs. A–C, Data are representativeof four independent experiments.D–G, Results compiled from threeindependent experiments n � 15.Frequency of (D) CD4þ Foxp3� T-helperTILs or (E) CD4þ Foxp3þ TILs ofintratumoral T cells.F,Ratio ofCD8þTILsto Tregs in tumors. G, Ratio of AH1tetramerþ CD8þ TILs to Tregs in tumors.� , P < 0.05; �� , P � 0.01; ��� , P � 0.001;���� , P < 0.0001. A–G, Significancebetween treatment groups measured atendpoint was determined using ANOVAfollowed by post hoc all-pairwisecomparison tests with a Tukeyadjustment for multiple comparisons.

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1118

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 11: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

significantly higher ratio of AH1-specific CD8þ TILs to Tregs (Fig.6G). Together, these data suggest that helper CD4þ TILs andeffector CD8þ TILs may be able to sustain improved antitumorfunctions due to the decreased Treg frequency in the TME.

The cytokine milieu is more inflammatory with combinationtreatment

Changes we observed in both myeloid and lymphoid popula-tions demonstrate that anti-CD40/anti-CSF-1R immunotherapyresulted in significant alterations to the immune infiltrate in CT26tumors. To determine the sum effect of the observed changes onthe TME cytokine milieu, we measured cytokine analytes inhomogenized tumors and normalized analyte concentration pergram of tumor harvested. Proinflammatory cytokines TNFa,IFNg , IL6, IL23, and IL12 were increased in tumors that receivedanti-CD40 or combination immunotherapy (Supplementary Fig.S4A). In anti-CD40 or combination-treated tumors, increasedGM-CSF was also observed, which supports MHC IIhi TAMpolarization. In contrast, levels of immunosuppressive cytokineIL10 were decreased in all treatment groups compared withisotype-treated tumors, including anti-CSF-1R treatment alone(Supplementary Fig. S4B). Mean levels of IL4 were also reduced�30% in anti-CD40 and combination-treated tumors comparedwith isotype controls, but the amount of IL4 was relativelyminimal in all groups (Supplementary Fig. S4B). These datasuggest that the total effect of combination treatment skewed theTME toward increased inflammatory cytokine production andreduced suppressive signals. Together, the phenotype and func-tional analysis of TAMs, monocytes, DCs, and tumor-infiltratingT cells demonstrates that anti-CD40/anti–CSF-1R immunother-apy drives a cumulative polarization of the TME toward anantitumor, proinflammatory environment, resulting in improvedimmune responses, overall enhanced antitumor efficacy, andextended survival.

DiscussionIn this report, we investigated the efficacy of combining

CD40 agonism and CSF-1R blockade to modulate endogenousantitumor immunity and control tumor growth. We demon-strated enhanced inhibition of tumor growth and extendedsurvival with combination treatment compared with mono-therapies. The improved antitumor efficacy with anti-CD40/anti–CSF-1R therapy was characterized by significant changesto the immune infiltrate that disrupted suppressive immunemechanisms and promoted inflammatory innate and adaptiveantitumor responses.

The most dramatic changes in the immune infiltrate after anti-CD40/anti–CSF-1R treatment were observed in macrophage andmonocyte populations. Consistent with the highest CSF-1Rexpression on Ly6Clo TAMs, combining anti–CSF-1R inhibitionand CD40 agonism resulted in significantly reduced frequenciesof MHC IIhi and MHC IIlo TAMs in tumors. We observed aconcomitant increase in MHC IIhi Ly6Cint macrophages, whichsuggested that combination therapy reduced the suppressivetumor-educated TAMs, while leaving newly differentiated, proin-flammatory macrophages to repopulate the TME. The remainingmacrophages in tumors had higher expression of costimulatorymolecules CD80 and CD86, inflammatory cytokines, and lowerlevels of MHC IIlo markers, including IL10R. IL10 signaling,through its receptor IL10R, not only drives IL10 production by

Foxp3þ Tregs, but also antagonizes IL1- and TNFa-induced path-ways and suppresses transcription of proinflammatory genes in awide range of immune cells (34). Since increased expression ofIL-10R is associated with poor control of tumor progression, thedecreased IL-10R levels observed across cell types suggest thatcombination therapy supports resistance to IL-10-dependentsuppression. Macrophages and myeloid subsets infiltrating theTME also demonstrated upregulated costimulatory moleculesthat potentially propagated effector T-cell responses within thetumor. Together, these data demonstrated broad changes in thebalance of suppressive and inflammatory myeloid subsets func-tioning in tumors and illustrated how targeting CD40 pathways,together with CSF-1R inhibition, generated potent downstreameffects beyond TAM depletion.

Although anti-CD40/anti–CSF-1R therapy does not directlytarget T cells, it has a profound effect on boosting T-cell responses.To investigate how combination treatment affected antigen pre-senting cells responsible for initiating T-cell responses, weexplored changes in DCs across treatment groups. We foundCD40 agonism alone or in combination with anti–CSF-1Rreduced frequencies of DCs in tumors but increased frequenciesin dLNs. DCs exhibited an activated, mature APC phenotype withincreased levels of MHC II, CD80, and CD86 in both tumor anddLNs. Studies from other groups have described CD103þ DCsimmune surveillance and migration through nonlymphoid tis-sues. Upon receiving activating signals,migratoryDCs traffic fromsurrounding tissues to dLNs to present antigens that primelymphocytes. Accumulation of CD103þ DCs was most increasedwith combination therapy, which may indicate CD40 agonismimproves activation of CD103þ DCs residing in tissues aroundtumor sites, enabling them to pick up and process tumor antigensfor T-cell priming. Consistent with this hypothesis, AH1-specificCD8þ T-cell responses were increased in magnitude. Our obser-vation that T cells in combination-treated mice exhibited greaterlevels of activation and function than those in the anti-CD40monotherapy group indicates that skewing of the macrophagepopulation by CSF-1R contributes to the overall antitumorresponse. A prominent effect of combination treatment includedlower quantities of IL10. IL10 signaling in DCs potently inhibitsmaturation and antagonizes IL12 production, resulting in pooractivation of T cells and increased promotion of Treg differenti-ation (34, 42). Alternatively,wehypothesized that blockingCSF-1signals may alter the composition of DCs by reducing frequenciesof suppressive tolerogenic DCs that rely on this cytokine fordevelopment and survival. Together, these direct and indirecteffects of anti-CD40/anti–CSF-1R treatment may increase fre-quencies of mature immunogenic DCs and improve T-cell prim-ing. Therefore, the quantity and quality of T-cell responses foundin combination-treated animals could result from improvedAPCsthat couple more efficient antigen presentation with increasedexpression of costimulatory molecules. Our data highlight howgenerating antitumor T-cell responses not only within the tumor,but also in local lymphoid tissues, may support long-lived adap-tive immunity.

We found combination therapy boosted the function not onlyof CD8þ cytotoxic T cells but also of conventional CD4þ T cells.These activated helper CD4þ T cells exhibited an inflammatoryTh1 differentiation profile, based on production of prototypicalTh1 cytokines IFNg and TNFa. The shift in inflammatory cyto-kines seen in combination-treated tumors is likely driven not onlyby myeloid and CD8þ effector activity but also by increased Th1

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1119

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 12: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

effector function. CD40 ligand signaling has a critical role inpreservingCD4þT cell help (37, 43). IncreasedCD4þhelper T-cellfunction could be induced by enhanced antigen presentation andMHC–peptide complex stability via CD83 upregulation, costi-mulation with increased expression of CD80 and CD86, and IL12production (20, 21, 37).Decreased frequencies of Tregs could alsocontribute to the increased magnitude of CD4þ T-cell effectorfunction in anti-CD40/anti–CSF-1R-treated tumors. Future inves-tigation will focus on whether combination therapy directlyreduces Treg numbers by stimulating Th1 differentiation orchanging availability of cytokines critical for maintaining Tregs'Foxp3 expression and transcriptional programs (44). As suppres-sive TAMswere eradicated andmore inflammatory cells infiltratedcombination-treated tumors, we found lower levels of IL10 andIL4 and higher levels of IL12, IFNg , and TNFa in tumors, whichmay lend to the instability of Foxp3þ Treg numbers and supportdifferentiation of Th1 CD4þ T cells.

Combination therapy induced significant activation of lym-phocytes, concomitant with upregulation of checkpoint recep-tors PD-1, TIM-3, TIGIT, and LAG-3. Although coinhibitoryreceptors normally contribute to contraction of immuneresponses following pathogen clearance, checkpoint moleculesin the context of cancer and chronic infection are associatedwith T-cell exhaustion and immune dysfunction. A criticalquestion emerging from these mechanistic data is how toreconcile improved antitumor efficacy with TILs that pheno-typically resemble exhausted lymphocytes vulnerable to intra-tumoral suppressive mechanisms. T-cell exhaustion is notpurely a product of expression of inhibitory receptors, buta complex, progressive syndrome that develops over time inenvironments with multiple obstacles, including reducedCD4þ T cell help, chronic exposure to antigen, and inhibitorysignaling via checkpoint ligands and suppressive cytokines (45,46). Although coexpression of checkpoint receptors was ele-vated on both CD4þ and CD8þ T cells following combinationtherapy, we found that expression of checkpoint ligands, PD-L1or PD-L2, was not significantly different in the anti-CD40/anti-CSF-1R group from isotype controls. It has also been shownthat increased CD4þ T cell help preserves CD8þ T-cell effectorfunctions during chronic infection or in cancer (47). Therefore,exhaustion may be avoided despite upregulation of inhibitoryreceptors associated with immune dysfunction in cancer. None-theless, given the expression of numerous checkpoint receptorson T cells, antitumor efficacy of the combination of CD40agonism and CSF-1R blockade may be further improved byincluding checkpoint blockade. Checkpoint blockade has com-plemented TAM-directed therapies in various tumor models(48–50), and future experiments will investigate whetherincluding anti-PD-1 checkpoint blockade in combination withanti-CD40/anti–CSF-1R combination therapy will provideadditional benefits.

The present studies demonstrated the advantage of combin-ing CD40 agonism with CSF-1R blockade to produce the most

effective tumor control and extend survival. Using a rigorousflow cytometry approach that enables in-depth characterizationof distinct immune subsets, we have extensively profiled thephenotypic and functional changes in tumor-associated mye-loid and lymphocyte populations in response to treatment.Previous reports have described the effects of CD40 agonist oranti–CSF1R monotherapy alone in murine tumor models byusing the general phenotypic markers, CD11b and Gr-1, tocharacterize changes in the myeloid infiltrate. Our dataimproved the resolution on the heterogeneous myeloid popu-lations that infiltrate tumors and detailed the manner in whichanti-CD40/anti–CSF-1R immunotherapy altered endogenousimmunity. Tumors treated with both anti-CD40 and anti–CSF-1R maintained features of both monotherapies, but thecombination regimen increased the magnitude of changes inmyeloid and lymphoid responses. Our data highlight howsuccessful antitumor efficacy might require both the removalof suppressive populations and the delivery of activating sig-nals for immune cells in the TME. The cumulative effect ofCD40 agonism and CSF-1R inhibition conditions immuneinfiltrates to counteract immunosuppression in the tumorenvironment and successfully attack tumor cells.

Disclosure of Potential Conflicts of InterestM. Quigley has ownership interest in Bristol-Myers Squibb. No potential

conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design:K.R.Wiehagen,N.M. Girgis, D.H. Yamada, I.S. Grewal,M. Quigley, R.I. VeronaDevelopment of methodology: K.R. Wiehagen, N.M. Girgis, D.H. YamadaAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): K.R. Wiehagen, N.M. GirgisAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): K.R. Wiehagen, N.M. Girgis, D.H. Yamada,A.A. Smith, S.R. Chan, I.S. Grewal, M. Quigley, R.I. VeronaWriting, review, and/or revision of the manuscript: K.R. Wiehagen,D.H. Yamada, A.A. Smith, S.R. Chan, I.S. Grewal, M. Quigley, R.I. VeronaAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): K.R. Wiehagen, A.A. SmithStudy supervision: K.R. Wiehagen, I.S. Grewal, M. Quigley, R.I. Verona

AcknowledgmentsThe authors acknowledge the contributions of Cassandra L. Lowenstein,

Nikki A. DeAngelis, and Catherine A. Ferrante in support of ex vivo experi-ments. Amy Wong, Leopoldo L. Luistro, Darlene J. Pizutti, and Diana Chinsupported in vivo studies. We thank Enrique Zudaire and Jonathan Cenna forexpert advice and helpful discussions, and Enrique Zudaire for criticalreading of this article.

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

Received May 22, 2017; revised September 1, 2017; accepted October 24,2017; published OnlineFirst November 2, 2017.

References1. Palucka AK, Coussens LM. The basis of oncoimmunology. Cell 2016;

164:1233–47.2. 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.

3. Hoves S, Krause SW, SchutzC,HalbritterD, Scholmerich J,HerfarthH, et al.Monocyte-derived human macrophages mediate anergy in allogeneic Tcells and induce regulatory T cells. J Immunol 2006;177:2691–8.

4. Stanley ER, Chitu V. CSF-1 receptor signaling in myeloid cells. Cold SpringHarbPerspect Biol 2014;6. pii: a021857. doi: 10.1101/cshperspect.a021857.

Wiehagen et al.

Cancer Immunol Res; 5(12) December 2017 Cancer Immunology Research1120

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 13: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

5. 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.

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

7. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development,homeostasis and disease. Nature 2013;496:445–55.

8. Gordon S,Martinez FO.Alternative activationofmacrophages:mechanismand functions. Immunity 2010;32:593–604.

9. Kurahara H, Shinchi H, Mataki Y, Maemura K, Noma H, Kubo F, et al.Significance of M2-polarized tumor-associated macrophage in pancreaticcancer. J Surg Res 2011;167:e211–9.

10. Beck AH, Espinosa I, Edris B, Li R, Montgomery K, Zhu S, et al. Themacrophage colony-stimulating factor 1 response signature in breastcarcinoma. Clin Cancer Res 2009;15:778–87.

11. Espinosa I, Beck AH, Lee CH, Zhu S, Montgomery KD, Marinelli RJ, et al.Coordinate expression of colony-stimulating factor-1 and colony-stimu-lating factor-1-related proteins is associated with poor prognosis in gyne-cological and nongynecological leiomyosarcoma. Am J Pathol2009;174:2347–56.

12. 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.

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

14. 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.

15. 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.

16. StrachanDC, Ruffell B,Oei Y, BissellMJ, Coussens LM, PryerN, et al. CSF1Rinhibition delays cervical and mammary tumor growth in murine modelsby attenuating the turnover of tumor-associatedmacrophages and enhanc-ing infiltration by CD8þ T cells. Oncoimmunology 2013;2:e26968.

17. Ries CH, Hoves S, Cannarile MA, Ruttinger D. CSF-1/CSF-1R targetingagents in clinical development for cancer therapy. Curr Opin Pharmacol2015;23:45–51.

18. Beatty GL, Li Y, Long KB. Cancer immunotherapy: activating innate andadaptive immunity through CD40 agonists. Expert Rev Anticancer Ther2017;17:175–86.

19. Posner MR, Cavacini LA, UptonMP, Tillman KC, Gornstein ER, Norris CMJr. Surface membrane-expressed CD40 is present on tumor cells fromsquamous cell cancer of the head and neck in vitro and in vivo andregulates cell growth in tumor cell lines. Clin Cancer Res 1999;5:2261–70.

20. Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. T-cellhelp for cytotoxic T lymphocytes ismediated byCD40-CD40L interactions.Nature 1998;393:480–3.

21. Toes RE, Schoenberger SP, van der Voort EI, Offringa R, Melief CJ. CD40-CD40Ligand interactions and their role in cytotoxic T lymphocyte primingand anti-tumor immunity. Semin Immunol 1998;10:443–8.

22. Grammer AC, Bergman MC, Miura Y, Fujita K, Davis LS, Lipsky PE. TheCD40 ligand expressed by human B cells costimulates B cell responses.J Immunol 1995;154:4996–5010.

23. Carbone E, Ruggiero G, Terrazzano G, Palomba C, Manzo C, Fontana S,et al. A newmechanism of NK cell cytotoxicity activation: the CD40-CD40ligand interaction. J Exp Med 1997;185:2053–60.

24. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G, et al. CD40 ligand on activated platelets triggers an inflam-matory reaction of endothelial cells. Nature 1998;391:591–4.

25. Bennett SR, Carbone FR, Karamalis F, Flavell RA,Miller JF, HeathWR. Helpfor cytotoxic-T-cell responses is mediated by CD40 signalling. Nature1998;393:478–80.

26. Nowak AK, Robinson BW, Lake RA. Synergy between chemotherapy andimmunotherapy in the treatment of established murine solid tumors.Cancer Res 2003;63:4490–6.

27. Ahonen CL, Doxsee CL, McGurran SM, Riter TR, Wade WF, Barth RJ, et al.CombinedTLR andCD40 triggering induces potentCD8þT cell expansionwith variable dependence on type I IFN. J Exp Med 2004;199:775–84.

28. MurphyWJ, Welniak L, Back T, Hixon J, Subleski J, Seki N, et al. Synergisticanti-tumor responses after administration of agonistic antibodies to CD40and IL-2: coordination of dendritic and CD8þ cell responses. J Immunol2003;170:2727–33.

29. Liu C, Lewis CM, Lou Y, Xu C, Peng W, Yang Y, et al. Agonistic antibody toCD40 boosts the antitumor activity of adoptively transferred T cells in vivo.J Immunother 2012;35:276–82.

30. Byrne KT, Leisenring NH, Bajor DL, Vonderheide RH. CSF-1R-dependentlethal hepatotoxicitywhen agonisticCD40 antibody is givenbefore but notafter chemotherapy. J Immunol 2016;197:179–87.

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

32. Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J,et al. Reprogramming tumor-associated macrophages by antibody tar-geting inhibits cancer progression and metastasis. Cell Rep 2016;15:2000–11.

33. Rakhmilevich AL, Alderson KL, Sondel PM. T-cell-independent antitumoreffects of CD40 ligation. Int Rev Immunol 2012;31:267–78.

34. Saraiva M,O'Garra A. The regulation of IL-10 production by immune cells.Nat Rev Immunol 2010;10:170–81.

35. Sato T, Terai M, Tamura Y, Alexeev V, Mastrangelo MJ, Selvan SR. Inter-leukin 10 in the tumor microenvironment: a target for anticancer immu-notherapy. Immunol Res 2011;51:170–82.

36. Quezada SA, Jarvinen LZ, Lind EF, Noelle RJ. CD40/CD154 interactions atthe interface of tolerance and immunity. Annu Rev Immunol 2004;22:307–28.

37. Grewal IS, Flavell RA. The role of CD40 ligand in costimulation and T-cellactivation. Immunol Rev 1996;153:85–106.

38. Roberts EW, Broz ML, Binnewies M, Headley MB, Nelson AE, Wolf DM,et al. Critical role for CD103(þ)/CD141(þ) dendritic cells bearing CCR7for tumor antigen trafficking and priming of T cell immunity inmelanoma.Cancer Cell 2016;30:324–36.

39. Hansen M, Andersen MH. The role of dendritic cells in cancer. SeminImmunopathol 2017;39:307–16.

40. Byrne KT, Vonderheide RH. CD40 stimulation obviates innate sensors anddrives T cell immunity in cancer. Cell Rep 2016;15:2719–32.

41. 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.

42. Li H, Shi B. Tolerogenic dendritic cells and their applications in transplan-tation. Cell Mol Immunol 2015;12:24–30.

43. Whitmire JK, Flavell RA, Grewal IS, Larsen CP, Pearson TC, Ahmed R.CD40-CD40 ligand costimulation is required for generating antiviral CD4T cell responses but is dispensable for CD8 T cell responses. J Immunol1999;163:3194–201.

44. Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al.Molecular antagonism and plasticity of regulatory and inflammatory T cellprograms. Immunity 2008;29:44–56.

45. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection andcancer. Trends Immunol 2015;36:265–76.

46. Zarour HM. Reversing T-cell dysfunction and exhaustion in cancer. ClinCancer Res 2016;22:1856–64.

47. Church SE, Jensen SM, Antony PA, Restifo NP, Fox BA. Tumor-specificCD4þ T cells maintain effector and memory tumor-specific CD8þ T cells.Eur J Immunol 2014;44:69–79.

48. Eissler N,Mao Y, BrodinD, Reutersward P, Andersson SvahnH, Johnsen JI,et al. Regulation ofmyeloid cells by activated T cells determines the efficacyof PD-1 blockade. Oncoimmunology 2016;5:e1232222.

49. Luheshi NM, Coates-Ulrichsen J, Harper J, Mullins S, Sulikowski MG,Martin P, et al. Transformation of the tumour microenvironment by aCD40 agonist antibody correlates with improved responses to PD-L1blockade in a mouse orthotopic pancreatic tumour model. Oncotarget2016;7:18508–20.

50. Zippelius A, Schreiner J, Herzig P, Muller P. Induced PD-L1 expressionmediates acquired resistance to agonistic anti-CD40 treatment. CancerImmunol Res 2015;3:236–44.

www.aacrjournals.org Cancer Immunol Res; 5(12) December 2017 1121

CD40 Agonist with CSF-1R Blockade Combination Immunotherapy

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258

Page 14: Combination of CD40 Agonism and CSF-1R Blockade ...Research Article Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor

2017;5:1109-1121. Published OnlineFirst November 2, 2017.Cancer Immunol Res   Karla R. Wiehagen, Natasha M. Girgis, Douglas H. Yamada, et al.   ImmunityTumor-Associated Macrophages and Drives Potent Antitumor Combination of CD40 Agonism and CSF-1R Blockade Reconditions

  Updated version

  10.1158/2326-6066.CIR-17-0258doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerimmunolres.aacrjournals.org/content/suppl/2017/11/01/2326-6066.CIR-17-0258.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerimmunolres.aacrjournals.org/content/5/12/1109.full#ref-list-1

This article cites 49 articles, 16 of which you can access for free at:

  Citing articles

  http://cancerimmunolres.aacrjournals.org/content/5/12/1109.full#related-urls

This article has been cited by 10 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

  Permissions

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

.http://cancerimmunolres.aacrjournals.org/content/5/12/1109To request permission to re-use all or part of this article, use this link

on November 24, 2020. © 2017 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 2, 2017; DOI: 10.1158/2326-6066.CIR-17-0258