15
Tumor Biology and Immunology AntiPD-L1 Treatment Results in Functional Remodeling of the Macrophage Compartment Huizhong Xiong 1 , Stephanie Mittman 1 , Ryan Rodriguez 1 , Marina Moskalenko 1 , Patricia Pacheco-Sanchez 1 , Yagai Yang 1 , Dorothee Nickles 2 , and Rafael Cubas 1 Abstract © 2019 American Association for Cancer Research T cells T cells aPDL1 MHCI and MHCII ARGI iNOS CD40 CD86 proinflammatory cytokines Phagocytosis pathways Proinflammatory Suppressive Macrophage Without treatment, tumor macrophages maintain a suppressive phenotype. Following anti-PD-L1 treatment, increased IFN signaling remodels the macrophage compartment towards a more proinflammatory phenotype, which can enhance T-cell responses. Remodeling of the macrophage compartment is driven by IFN following anti-PD-L1 treatment. Macrophage Cancer cells ARGI IFNγ IFNγ Checkpoint inhibitors like anti-PD1/PD-L1 have demonstrated signicant therapeutic efca- cy in a subset of patients partly through reinvig- oration of CD8 T cells. However, their impact on myeloid cells remains largely unknown. Here, we report that antiPD-L1 treatment favorably impacts the phenotype and function of tumor macrophages by polarizing the macrophage com- partment toward a more proinammatory phe- notype. This phenotype was characterized by a decrease in Arginase-I (ARG1) expression and an increase in iNOS, MHCII, and CD40 expression. Whole-transcriptome proling further conrmed extensive polarization of both tumor monocytes and macrophages from a suppressive to a proin- ammatory, immunostimulatory phenotype. This polarization was driven mainly through IFNg and was associated with enhanced T-cell activity. Transfer of monocytes into antiPD-L1treated tumor-bearing mice led to macrophage differentiation into a more proinammatory phenotype, with an increase in CD8 T cells expres- sing granzyme-B and an increase in the CD8/Treg ratio compared with control-treated mice. Although in responsive tumor models, antiPD-L1 treatment remodeled the macrophage compartment with benecial effects on T cells, both macrophage reprogramming and depletion were needed to maximize antiPD-L1 responses in a tumor immune contexture with high macrophage burden. Our results demonstrate that antiPD-L1 treatment can favorably remodel the macrophage compartment in responsive tumor models toward a more proinammatory phenotype, mainly through increased IFNg levels. They also suggest that directly targeting these cells with reprogramming and depleting agents may further augment the breadth and depth of response to antiPD-L1 treatment in less responsive or more macrophage-dense tumor microenvironments. Signicance: This work demonstrates that increased IFNg signaling following antiPD-L1 treatment can remodel the macrophage compartment to enhance T-cell responses. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1493/F1.large.jpg. 1 Department of Translational Oncology, Genentech, South San Francisco, Cali- fornia. 2 Bioinformatics and Computational Biology, Genentech, South San Francisco, California. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Rafael Cubas, Genentech, 1 DNA Way MS-50, South San Francisco, CA 94080. Phone: 650-225-1539; Fax: 650-225-1411; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-18-3208 Ó2019 American Association for Cancer Research. Cancer Research www.aacrjournals.org 1493 on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

Tumor Biology and Immunology

Anti–PD-L1 Treatment Results in FunctionalRemodeling of the Macrophage CompartmentHuizhong Xiong1, Stephanie Mittman1, Ryan Rodriguez1, Marina Moskalenko1,Patricia Pacheco-Sanchez1, Yagai Yang1, Dorothee Nickles2, and Rafael Cubas1

Abstract

© 2019 American Association for Cancer Research

T cells

T cells

aPDL1

MHCI and MHCII

ARGI iNOS CD40 CD86

proinflammatory cytokines

Phagocytosispathways

Proinflammatory

SuppressiveMacrophage

Without treatment, tumor macrophages maintain a suppressive phenotype.

Following anti-PD-L1 treatment, increased IFN signaling remodels the macrophage compartment towards amore proinflammatory phenotype, which can enhance T-cell responses.

Remodeling of the macrophage compartment is driven by IFN following anti-PD-L1 treatment.

Macrophage

Cancer cells

ARGIIFNγ

IFNγ

Checkpoint inhibitors like anti-PD1/PD-L1have demonstrated significant therapeutic effica-cy in a subset of patients partly through reinvig-oration of CD8 T cells. However, their impact onmyeloid cells remains largely unknown. Here, wereport that anti–PD-L1 treatment favorablyimpacts the phenotype and function of tumormacrophages by polarizing themacrophage com-partment toward a more proinflammatory phe-notype. This phenotype was characterized by adecrease in Arginase-I (ARG1) expression and anincrease in iNOS, MHCII, and CD40 expression.Whole-transcriptome profiling further confirmedextensive polarization of both tumor monocytesand macrophages from a suppressive to a proin-flammatory, immunostimulatory phenotype.This polarization was driven mainly throughIFNg and was associated with enhanced T-cellactivity. Transfer of monocytes into anti–PD-L1–treated tumor-bearing mice led to macrophagedifferentiation into a more proinflammatoryphenotype,with an increase inCD8Tcells expres-sing granzyme-B and an increase in the CD8/Tregratio compared with control-treated mice.Although in responsive tumor models, anti–PD-L1 treatment remodeled the macrophagecompartment with beneficial effects on T cells,both macrophage reprogramming and depletion were needed to maximize anti–PD-L1 responses in a tumor immunecontexture with high macrophage burden. Our results demonstrate that anti–PD-L1 treatment can favorably remodel themacrophage compartment in responsive tumormodels toward amore proinflammatory phenotype, mainly through increasedIFNg levels. They also suggest that directly targeting these cells with reprogramming and depleting agents may further augmentthe breadth and depth of response to anti–PD-L1 treatment in less responsive or more macrophage-dense tumormicroenvironments.

Significance: This work demonstrates that increased IFNg signaling following anti–PD-L1 treatment can remodel themacrophage compartment to enhance T-cell responses.

Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1493/F1.large.jpg.

1Department of Translational Oncology, Genentech, South San Francisco, Cali-fornia. 2Bioinformatics and Computational Biology, Genentech, South SanFrancisco, California.

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

CorrespondingAuthor:Rafael Cubas, Genentech, 1 DNAWayMS-50, South SanFrancisco, CA 94080. Phone: 650-225-1539; Fax: 650-225-1411; E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-18-3208

�2019 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 1493

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 2: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

IntroductionTumor-associated macrophages (TAM) are abundantly found

in tumors and contribute critically to the immune set point (1–3).The activation state of macrophages is generally characterized asM1 (classic) and M2 (alternative; refs. 4, 5). M1 macrophages aremannose receptor low (CD206low), express high levels of MHCII(MHCIIhigh), and are characterized by high expression of induc-ible nitric oxide synthase (iNOS), as well as costimulatory mole-cules like CD40, CD86, and various proinflammatory cyto-kines (6). M2 macrophages, on the other hand, are CD206high

MHCIIlow, and release anti-inflammatory and immunosuppres-sive mediators such as Arginase-I (ARG1), IL10, and TGFb (7).

In the cancer setting, TAMs display remarkable heterogeneityand plasticity (1, 2, 4). The definition ofM1 andM2, derived fromin vitro polarization assays, lies at two extremes and is oftenconsidered inaccurate for describing the complexity of TAMfunctional states in vivo. Thus, in our work, we refer to TAMs aseitherM1-like,M1/M2 intermediate, orM2-like to reflect differentdegrees of mixed phenotypes within this spectrum (2, 8). M1-likemacrophages have beenpositively associatedwith overall survival(OS) in a variety of human cancers, whereasM2-likemacrophagesare poor prognostic indicators (9). Recent studies have shed lighton the complexity ofmacrophage function. Fcg receptors and IL1bhave been reported to be involved in phagocytosis-induced PDL1and IDO expression (10), and iNOS has been shown to play bothanti- and protumoral roles (11–13) despite continuing to serve asa commonM1-associatedmarker (14–16). These results highlightthenecessity to examine the expressionofmultiplemarkers and tointegrate whole transcriptome analysis for a more accurate eval-uation of macrophage functional state.

Given the importance of macrophages as major immune mod-ulators in the tumor microenvironment, macrophage-targetingstrategies are under intense investigation with two mainapproaches: depletion and repolarization (17, 18). Variousdepleting agents that block monocyte recruitment and/or TAMdifferentiation are currently being tested (2). Repolarization ofmacrophages aims at changing the phenotype of TAMs from animmune-suppressive to an immune-stimulatory state. Properlyeducated TAMs can have notable beneficial characteristics, expres-sing high levels of MHC, costimulatory molecules, secretingproinflammatory cytokines, and actively killing antibody-opso-nized cancer cells through phagocytosis (19, 20). RepolarizedTAMs could therefore become efficient antigen-presenting cellsactivating CD8 and CD4 T cells in situ (21). TAM repolarizationstrategies include agonistic anti-CD40 (aCD40), blockade ofCSF1 signaling, selective inhibition of PI3Kg , deletion of Dicer,as well as targeting surface markers expressed by suppressivemacrophages (2, 8, 16, 21–28). Repolarization and depletionhave recently been combined to achieve an optimal stimulatoryphenotype on macrophages; however, its implication on tumorsensitization to checkpoint inhibitors remains unclear (27).

Checkpoint inhibitors like anti-PD1/PD-L1 (aPD1/PDL1) andanti-CTLA4 (aCTLA4) have proven effective and durable in asubset of patients in a variety of cancers (29–31). Extensive workhas mainly focused on exploring T-cell functionality and theirreinvigoration mechanisms following treatment (3, 32–36).Despite numerous efforts at understanding the mechanism ofaction of checkpoint inhibitors and macrophage polarization,little is known about the connection between the two. A recentstudy demonstrated profound remodeling of monocyte and

macrophage subsets after aPD1/aCTLA4 treatment, with anincrease in iNOS and a decrease in CD206 expression, which waspartially dependent on IFNg (37). These results suggest that TAMscan actively respond to microenvironment changes mediated bycheckpoint blockade and thus should be considered in the designof combinatorial strategies.

Here, we demonstrate that PDL1 blockade in responsive tumormodels led to an overall remodeling of the macrophage com-partment toward a more proinflammatory phenotype, an effectthat was observed in both monocytes and recently differentiatedmacrophages. We further demonstrate that macrophage polari-zation following aPDL1 treatment can beneficially impact T-cellresponses and that differential manipulation of macrophagesbased on the tumor immune contexture can be used to signifi-cantly augment the activity of aPDL1.

Materials and MethodsAnimal study oversight

All animal studies were reviewed and approved byGenentech'sInstitutional Animal Care and Use Committee. Mice whosetumors exceeded acceptable size limits (2,000 mm3) or becameulcerated were euthanized and removed from the study.

MiceEight– to 10-week-old female C57BL/6 or Balb/c mice were

obtained from Charles River Laboratories. The mice were housedat Genentech in standard rodent microisolator cages and wereacclimated to study conditions for at least 3 days before tumor cellimplantation. Animals were 8- to 10-week-old with an averageweight of 19.68 g. Only animals that appeared to be healthy andfree of obvious abnormalities were used for the study.

Cell linesThe murine colon adenocarcinoma MC38 cell line was

obtained from former GNE colleague Rink Offringa and murinemammary carcinoma EMT6 and JC cell lines were obtainedfrom ATCC, tested for Mycoplasma using MycoAlert MycoplasmaDetection Kit (Lonza), kept at low passage and maintained incomplete RPMI1640 medium (HyClone) supplemented with10% heat-inactivated premium grade FBS (VWR) and 2.05mmol/L L-glutamine.

Syngeneic tumor studiesTumor cells were harvested in log-phase growth and resus-

pended in HBSS-containing Matrigel (BD Biosciences) at a 1:1ratio. For MC38, cells were implanted subcutaneously in the rightunilateral thoracic area of C57BL/6 mice at 0.1 � 106 MC38 cellsin 100 mL. For EMT6 and JC studies, Balb/c mice were inoculatedin the left mammary fat pad #5 with 0.1 � 106 cells in 100 mL.Tumors were monitored until they became established andreached a mean tumor volume of approximately 190 mm3. Micewith tumors in the range of 130 to 250 mm3 were then random-ized into treatment groups. Treatment was initiated the next daywith either isotype (gp120) or aPDL1 (clone 6E11) antibodiesgiven at 10 mg/kg i.v. for the first dose followed by 5 mg/kg i.p.,thereafter twice a week for 3 weeks (for efficacy studies). Whenindicated, anti-mouse IFNg rat IgG1 (BioXCell, clone: XMG1.2)was administered at 250 mg in 100 mL PBS intraperitoneally, twicea week. In the corresponding studies aCD40 (FGK45) was dosedintraperitoneally once at 4mg/kg and aCSF1R (2G2)was dosed at

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1494

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 3: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

30 mg/kg i.v. once. For tumor volume and body weight measure-ments, see Supplementary Methods.

Adoptive monocyte transferBone marrows from CD45.1 mice (The Jackson Laboratory,

stock no. 002014) were harvested and monocytes were purifiedusing Miltenyi Monocyte Isolation Kit (BM; catalog no. 130-100-629). A total of 2 � 106 monocytes were injected intravenouslyinto the tail vein of each MC38-tumor–bearing CD45.2 recipientmice. Isotype or aPDL1 antibodies were administered 5 days priorto cell transfer. Two days later, tumors were collected and digestedfor staining and FACS analysis.Monocytes andmacrophagesweredistinguished by Ly6C and F4/80 staining.

Flow cytometryTo generate single-cell suspensions, tumors were collected, cut

into 2–4mmpieces, anddigested for 30minutes using themurineTumorDissociationKit fromMiltenyi (Miltenyi Biotec) followingthe manufacturer's instructions (catalog no. 130-096-730).Tumor homogenates were filtered through a 70-mm nylon filter(Corning) and washed twice with RPMI1640media. After the lastwash, cells were resuspended in staining buffer (PBSþ 0.5% FCSþ 5 mmol/L EDTA). Cells were surface-stained for 25 minutes at4�C. For intracellular staining, cells were fixed and permeabilizedusing the eBioscience Foxp3 Fix/Perm buffer kit. For detection ofIFNg and TNFa, cells were stimulated with Cell StimulationCocktail for 5 hours. After 1 hour, GolgiStop was added to thecells. FACS analyses were performed using a Symphony flowcytometer (BD Biosciences). Data were analyzed using FlowJosoftware (Tree Star Inc.).

AntibodiesFor animal studies murine IgG1 aPDL1 (clone 6E11), murine

IgG1 anti-gp120 isotype control, rat IgG2a aCD40 (clone FGK45),and murine IgG1 anti-CSF1R (clone 2G2) antibodies were used.Antibodies were stored in 20 mmol/L histidine acetate, 240mmol/L sucrose, and 0.02% polysorbate 20, pH 5.5 and dilutedinPBSprior touse. Antibodies used forflowcytometry are listed inthe Supplementary Methods.

Sublethal irradiation and mixed bone marrow chimerasRecipient CD45.1 C57BL/6 mice (The Jackson Laboratory,

stock no. 002014) were sublethally irradiated with 450 rads byusing a Cesium 137 source. One day after irradiation, donorCD45.2 or IFNgR-deficient mice (The Jackson Laboratory, stockno. 003288) were euthanized and bone marrow was collectedfrom femurs and tibias. Irradiated recipientmice were injected viatail veinwith donor 2� 106 bonemarrow cells in 100 mL in sterilePBS/mouse. Tumor inoculation and tissue processing were per-formed as described previously.

Preparation of dissociated tumor supernatantMC38 tumors were digested as described previously. Tumor

homogenates were filtered through a 70-mm nylon filter (Corn-ing) and centrifuged twice at 2,000 rpm for 5 minutes at 4�C. Toremove treatment antibodies, tumor supernatants were incubatedwith Toyopearl (Tosoh Bioscience) for 5 minutes at room tem-perature (1:10 dilution) and centrifuged. Supernatants were thenfiltered through a 0.22-mm Vacuum Filter Unit (Corning, catalogno. 431153) on ice, aliquoted into tubes, and stored at �80�Cbefore use.

OT-II purification and CFSE labelingSpleens and lymph nodes from OT-II mice (The Jackson

Laboratory, stock no. 004194) were harvested. Na€�ve OT-II cellswere purified with Naive CD4þ T Cell Isolation Kit (MiltenyiBiotec, catalog. no. 130-104-453). Cells were counted and labeledwith CFSE using CellTrace CFSE Cell Proliferation Kit (ThermoFisher Scientific, C34554) as described in the SupplementaryMethods.

Bone marrow–derived macrophage polarization and OT-IIcoculture assay

Bone marrow–derived macrophage (BMDM) differentiation isdescribed in the Supplementary Methods. On day 7 of differen-tiation, BMDMs were replenished with DMEM þ 10% FBS þPenicillin/Streptomycin/GlutaMAX without M-CSF. Dissociatedtumor supernatant media was added for macrophage polariza-tion. When indicated, recombinant mouse IFNg (PeproTech,10ng/mL) or anti-mouse IFNg rat IgG1 (BioXCell, clone: XMG1.2,20 mg/mL) was added to the medium. On day 9, BMDMs weregently harvested, washed with PBS, plated in 96-well plates at 0.2� 106 cells/well, and loaded with OVA323-339 (AnaSpec) at 10 mg/mL for 3 hours at 37�C. After 3 extensive washes, CFSE-labeledna€�ve OT-II cells were added to the BMDM culture at 0.2 � 106

cells/well for 4 days before harvesting for FACS analysis.

Statistical analysisAll data were presented as means� SD. Comparisons between

tumor models, time points, or treatments were generated usingnonparametric, Mann–Whitney tests, whereas cell subset com-parisons within a given groupwere performed using paired t tests.For correlations, two-tailed nonparametric Spearman correlationanalysis was used. Prism 6.0 (GraphPad) was used to process allthe statistical analyses. Uncorrected P values were presented tovalidate RNA sequencing data.

RNA sequencing for transcriptome analysisMC38 tumors of similar size between isotype and aPDL1-

treated groups were collected, digested, and cell lysates stained(see Methods above). Macrophages and monocytes were sortedusing a BDFACSAria. RNAwas extracted and quantifiedwith RNAPlus Mini Kit (Qiagen) according to the manufacturer's protocol.Concentration of RNA samples was determined using NanoDrop8000 (Thermo Fisher Scientific) and integrity of RNA was deter-mined by Fragment Analyzer (Advanced Analytical Technolo-gies). tRNA (0.1 mg) was used as an input material for librarypreparation using TruSeq Stranded Total RNA Library Prep Kit(Illumina). Library establishment is described in the Supplemen-tary Methods.

Differential gene expression and gene set enrichment analysisAfter normalization using R/Bioconductor package DESeq2

(38), differential gene expression between cell types isolated fromcontrol-treated versus aPDL1-treated tumorswas performedusingR/Bioconductor package limma (39). Gene set enrichment anal-ysis (GSEA) was performed for MSigDB collections c2, c5, andc7 (40) using camera (41) as implemented in the multiGSEA Rpackage (http://github.com/lianos/multiGSEA). Enriched genesets were filtered for those passing an FDR cutoff of 0.01 and agene set size ofn<150. For visualization, only the 10most up- and10 most downregulated gene ontology (GO) sets are shown.These were manually grouped into broad functional categories

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1495

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 4: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

based on the genes that contributed most to the enrichment. Acomplete list of enriched gene sets is provided as SupplementaryTable S1. Calculation of gene set scores and differential expressionanalysis for select genes and gene sets is described in the Supple-mentary Methods. (GEO accession number: GSE125383).

ResultsTumor macrophages have a suppressive phenotype thatcorrelates with tumor burden

To investigate the effects of aPDL1 treatment on TAMs, we firstfocused on the baseline characteristics of the myeloid compart-ment in the MC38 tumor model given its partial responsivenessto treatment and its significant macrophage infiltration.Ly6C�Ly6G�F4/80þ TAMs and Ly6Cþ inflammatory monocyteswere present at high densities in tumors (52% and 22%, respec-tively) as the major tumor immune infiltrate (SupplementaryFig. S1A). TAMs in MC38 tumors expressed high levels of ARG1and low but detectable levels of iNOS, whereas monocytes highlyexpressed iNOS and low levels of ARG1 (SupplementaryFig. S1B). Ly6Cint cells expressed moderate levels of both ARG1and iNOS, suggesting a transitional state (Supplementary Fig.S1B; refs. 2, 17). The majority of TAMs expressed high levels ofboth MHCII and CD206 and were identified as an M1/M2intermediate subset. MHCIIþCD206� and MHCII�CD206þ

TAMs were described as M1-like and M2-like, respectively(Fig. 1A, left). Further analysis showed that the M2-like andM1/M2 intermediate subsets had high ARG1 expression, whereasM1-like and M1/M2 intermediate subsets had high iNOS levels(Fig. 1A, right). This indicated that the M1/M2 intermediatesubset displayed both immunosuppressive and proinflammatoryphenotypes. Noteworthy, MHCII and iNOS levels on TAMs, aswell as the M1-like frequency, were negatively associated withtumor burden (Fig. 1B). In contrast, CD206, ARG1 expression,and M2-like frequency were positively correlated with tumor size(Fig. 1C). Dynamically, the composition of TAMs shifted to amore suppressive M2-like phenotype as tumors progressed andtumor burden increased (Fig. 1D and E) consistent with previousreports (1, 4, 6).

aPDL1 treatment drives TAM polarization toward a moreproinflammatory phenotype along with increased CD8 T-cellabundance and activity

Following aPDL1 treatment, MC38 tumor growth was signif-icantly delayedwith10%complete responses (CR) in treatedmice(Fig. 2A). We then examined the expression of various functionalmarkers on TAMs 7 days after treatment initiation and at a timepoint both were treated and control tumors were still of compa-rable size (Supplementary Fig. S1C). aPDL1 treatment reducedARG1 levels (Fig. 2B Supplementary Fig. S1C) mainly on ARG1-high–expressing M1/M2 intermediate and M2-like subsets(Fig. 2B). Conversely, iNOS, MHCII, and CD40 levels on TAMs,as well as iNOS levels on monocytes, increased following treat-ment (Fig. 2C and D; Supplementary Fig. S1C). CD8 T-cellinfiltration, granzyme-B (GZMB) production, and the CD8/Tregratio were also significantly elevated by treatment, consistent withprevious reports of aPDL1-induced reinvigoration of CD8 T cells(Fig. 2E; refs. 31, 36). Interestingly, ARG1 levels on TAMs werenegatively associated with the density, activation, and prolifera-tion of CD8 T cells, whereas iNOS expression on TAMs waspositively associated with the abundance and function of CD8

and CD4 T cells (Fig. 2F–G; Supplementary Fig. S1D and S1E),indicating a connection between T-cell activation and TAM polar-ization in tumors.

We next evaluated whether macrophage remodeling followingaPDL1 treatment was contributed in part by newly differentiatedmacrophages from monocyte precursors. Monocytes were adop-tively transferred intomice with establishedMC38 tumors treatedwith aPDL1 or isotype control antibodies. A similar level ofmonocyte recruitment and TAM differentiation was observed inthe tumors after 2 days (Supplementary Fig. S1F). However,recently differentiated TAMs in aPDL1-treatedmice were enrichedin M1-like macrophages when compared with differentiatedTAMs in isotype-treated mice (Fig. 2H) with lower levels of ARG1and higher levels of iNOS and CD40 expression (Fig. 2I). Inaddition, recently recruited monocytes also displayed a moreproinflammatory phenotype with elevated iNOS and CD40expression in aPDL1-treated recipient tumors (Fig. 2J). Impor-tantly, this induction of newly differentiated M1-like macro-phages in aPDL1-treated mice coincided with a higher level ofCTL infiltration, GZMB production, and a significant increase inthe CD8/Treg ratio (Supplementary Fig. S1G), suggesting that thephenotypic changes imparted on newly differentiated TAMs pro-vided a beneficial effect on the T-cell compartment. Together,these results corroborate that aPDL1 treatment induces a rapidand substantial change in the tumor microenvironment, whichleads to a proinflammatory phenotype on monocytes and differ-entiating TAMs. Thus, the phenotypic changes observed in TAMsfollowing aPDL1 treatment are in part contributed by newlydifferentiated monocyte-derived macrophages that are imprintedwith a more proinflammatory phenotype.

aPDL1 treatment leads to genome-wide TAM polarizationtoward a more proinflammatory phenotype

To expand our understanding and complement the phenotypicanalysis of TAMs using cell surface markers, we performed whole-transcriptome profiling of macrophages sorted from aPDL1and control-treated tumors. GSEA revealed profound genome-wide repolarization, evidenced by an elevated expression of theantigen presentation machinery including various gene setsmade up of MHC molecules and phagocytosis/opsonization–related Fcg receptors (Fig. 3A; complete lists of enriched gene setsin Supplementary Table S1). Furthermore, predefined genesets for IFNg downstream, proinflammatory signaling and che-mokine expression, TLR/NFkB, and the autophagy pathway werealso upregulated in macrophages following aPDL1 treatment(Fig. 3B; Supplementary Fig. S2A and S2B). Interestingly, GSEAuncovered a concomitant downregulation of numerous collagen-related gene sets, potentially reflecting tissue remodeling, as wellas development-related gene sets in macrophages (Fig. 3A).aPDL1 treatment had a similar effect on intratumoral monocytes(Supplementary Fig. S2C and S2D), indicating a substantial andsignificant influence of PDL1 blockade on bothmacrophages andtheir precursors in agreement with a recent publication (37).

aPDL1–mediated remodeling of themacrophage compartmentis IFNg dependent

To identify the factor(s) linking aPDL1-induced T-cell reinvig-oration and TAM polarization, we performed cytokine profileanalysis of MC38 tumor lysates. We observed amarked upregula-tion of IFNg following aPDL1 treatment (Fig. 4A), which wasmainly produced by intratumoral CD8 T cells (Fig. 4B). IFNg was

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1496

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 5: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

P = 0.0009

R: 0.4

0 10 20 30 40 500.0

0.2

0.4

0.6

0.8

% ARG1+ Macrophages

Tum

or

we

igh

t (g

)

0 20 40 600.0

0.2

0.4

0.6

0.8

% M2-Like

Tum

or

we

igh

t (g

)P < 0.0001

R: 0.66

CD206 MFI on Macrophages

Tum

or

we

igh

t (g

)

P < 0.0001

R: 0.42

0 2,000 4,000 6,000 8,000

0.0

0.2

0.4

0.6

0.8

0 20 40 60 800.0

0.2

0.4

0.6

0.8

% M1-Like

Tum

or

we

igh

t (g

) P < 0.0001

R: −0.37

% iNOS+ Macrophages

Tum

or

we

igh

t (g

) P < 0.0001

R: −0.36

0 2 4 6 80.0

0.2

0.4

0.6

0.8

MHCII MFI on Macrophages

Tum

or

we

igh

t (g

) P < 0.0001

R: −0.48

0 20,000 40,000 60,000 80,000 100,0000.0

0.2

0.4

0.6

0.8

CD206

MH

CII

0

1,000

2,000

3,000

4,000

5,000

MFI

of A

RG

1

****

*****

M1-

Like

M1/

M2

Inte

rm.

M2-

Like

0

1,000

2,000

3,000

4,000

MFI

of i

NO

S

ns *

*

M1-

Like

M1/

M2

Inte

rm.

M2-

Like

A

B

C

D

E

CD206

MH

CII

Gated on F4/80+ CD11b+ macrophages

220 mm3 760 mm3 1,200 mm3,

0

20

40

60

80

% M

1-L

ike

**

**

220 mm3 760 mm3 1,200 mm3

0

50

100

% M

1/M

2 In

term

ed

iate *

**

220 mm3 760 mm3 1,200 mm3 220 mm3 760 mm3 1,200 mm3

0

20

40

60

80

% M

2-L

ike

P = 0.06*

F4/80+ Macrophages

Like

-1

M

M1/M2 interm.

M2-Like

Figure 1.

Tumor macrophages have a suppressive phenotype that correlates with tumor burden. MC38 tumors were collected 1 day after they reached an average size ofapproximately 180 mm3 and single-cell suspensions were prepared for flow cytometry analysis. Single-cell suspensions were stained and intratumoralmacrophages were defined as CD45þDump�CD11bþCD11c�F4/80þ cells. A, Left, representative FACS plot showing M1-like, M1/M2 intermediate, and M2-likesubsets based on MHCII and CD206 staining. Right, mean fluorescent intensity (MFI) of ARG1 and iNOS onmacrophage subsets. B, Correlation between M1markers (MHCII and iNOS) and M1-like frequency and tumor weight. C, Correlation between M2markers (CD206 and ARG1) and M2-like frequency and tumorweight. D, Representative FACS plots of M1/M2 subsets based on CD206/MHCII staining onmacrophages from tumors of different sizes. E, Comparison of M1-like(left), M1/M2-intermediate (middle), or M2-like frequencies (right) from D. N¼ 20 (220mm3 and 760mm3) and 40 (120 mm3), respectively. � , P < 0.05;�� , P < 0.01; ����, P < 0.0001. ns, nonsignificant.

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1497

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 6: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

0 20 40 608

163264

128256512

1,0242,0484,096

Days

CR: 10%0 20 40 60

8163264

128256512

1,0242,0484,096

Days

mm( e

mulov romuT

3 )

mm( e

mulov romuT

3 )

aPDL1Isotype

CR: 0%

Isotyp

e

aPDL1

0

10

20

30

40

% A

RG

1+

Isotyp

e

aPDL1

Isotyp

e

aPDL1

Isotyp

e

aPDL1

0

20

40

60

**

M1-Like M1/M2 Interm. M2-Like

Isotyp

e

aPDL1

0

10,000

20,000

30,000

40,000

50,000

MH

CII

MFI

*

Isotyp

e

aPDL1

0

5,000

10,000

15,000

CD

40 M

FI *

0

10

20

30

40

Isotyp

e

aPDL1

Mono.

***

Isotyp

e

aPDL1

Isotyp

e

aPDL1

Isotyp

e

aPDL1

0

2

4

6

8

****

M1-Like M1/M2 Interm. M2-Like

0

2

4

6

8

Isotyp

e

aPDL1

% iN

OS+

***

Macro.

E

0

10

20

30

% C

D8

in C

D45

**

Isotyp

e

aPDL1

0

500

1,000

1,500

2,000

2,500

CD

8 #/m

g tu

mor

*

Isotyp

e

aPDL1

0

20

40

60

80

100

% G

ZMB

+ in

CD

8

**

Isotyp

e

aPDL1

0

1

2

3

4

5

CD

8/Tr

eg r

atio

Isotyp

e

aPDL1

***

0 200 400 600 8000

10

20

30

CD8 T Cell # /mg

% A

RG

1+

Mac

roph

ages P = 0.02

R: −0.38

0 200 400 600 8000

1

2

3

4

CD8 T Cell # /mg

% iN

OS+

M

acro

phag

es

P = 0.0003R: 0.65

0 2,000 4,000 6,000 8,0000

10

20

30

40

GZMB MFI on CD8 T Cells

%

AR

G1+

M

acro

phag

es P = 0.0003R: −0.53 P < 0.0001

R: −0.57

0 20 40 60 800

10

20

30

40

% Ki67+ CD8 T Cells

%

AR

G1+

M

acro

phag

es

0 20 40 60 800

10

20

30

40

% ICOS+ CD8 T Cells

% A

RG

1+

Mac

roph

ages P = 0.001

R: −0.33

% ICOS+ CD8 T Cells

%

iNO

S+

Mac

roph

ages

P = 0.0002R: 0.58

0 20 40 60 800

1

2

3

4

GZMB MFI on CD8 T Cells

%

iNO

S+

Mac

roph

ages

P < 0.0001R: 0.58

0 2,000 4,000 6,000 8,0000

1

2

3

4

% Ki67+ CD8 T Cells

%

iNO

S+

Mac

roph

ages

P = 0.001R: 0.47

0 20 40 60 800

1

2

3

4

D

F

A

B C

G

H IM1-LikeM1/M2 IntermediateM2-Like

Isotype donor aPDL1 Donor

***

0

20

40

60

% A

RG1+

Isotyp

e

aPDL1

0

5

10

15

% iN

OS+

Isotyp

e

aPDL1

0

5

10

15

20

% iN

OS+

J

Isotyp

e

aPDL1

* **

0

1,000

2,000

3,000

4,000

CD

40 M

FI

Isotyp

e

aPDL1

*

Monocyte-derived macrophages

0

500

1,000

1,500

2,000

CD

40 M

FI

Isotyp

e

aPDL1

Donor monocytes

*

Figure 2.

aPDL1 treatment drives TAM polarization toward a more proinflammatory phenotype along with increased CD8 T-cell numbers. Mice were inoculated withMC38 tumor cells and once tumors reached a volume of approximately 180 mm3 (day 0), mice with similarly sized tumors were grouped into treatment arms.The next day (day 1), mice were dosed with aPDL1 or isotype control antibody at 10 mg/kg i.v. for the first dose and 5 mg/kg i.p., thereafter twicea week for 3 weeks. Tumors were also collected 7 days after treatment initiation, digested, and single-cell suspensions used for FACS analysis.A, Tumor volume (mm3) of control (black) or aPDL1 (red)-treated mice is shown on a log2 scale; N ¼ 10. (Continued on the following page.)

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1498

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 7: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

critical for aPDL1-mediated efficacy, as IFNg blockade abolishedaPDL1-mediated antitumor responses and the abundance of IFNgwas negatively correlated with tumor weight and the frequency ofM2-like cells (Supplementary Fig. S3A–S3C). We therefore exam-ined TAM polarization during aPDL1 treatment with blockade ofIFNg , a known driver of M1 differentiation (4, 8, 42), andobserved abrogation of M1-like skewing with an increased fre-quency of suppressive M2-like macrophages (Fig. 4C). Moreover,

IFNg blockade abated the propolarization changes in ARG1,iNOS, and MHCII expression following aPDL1 treatment andreduced the overall levels of CD40 expression (Fig. 4D).

We next investigated the capacity of the macrophage compart-ment to respond to changes in environmental IFNg by transferringIFNgR�/� bone marrow into sublethally irradiated wild-typeanimals. Following reconstitution, TAMs consisted of a mixedpopulation of either IFNgR positive (IFNgRþ/þ) or negative

(Continued.) B, Frequency of arginase-expressing cells (ARG1þ) in total macrophages and macrophage subsets following treatment. C, Frequency of iNOS-expressing cells in total macrophages, macrophage subsets, and monocytes. D, Expression levels (mean fluorescent intensity, MFI) of MHCII and CD40 onmacrophages following treatment. E, Frequency and absolute number of intratumoral CD8 T cells, frequency of GZMB-producing cells, and the CD8/Tregratio. F, Correlation between the frequency of ARG1þ macrophages and CD8 T-cell abundance or GZMB, Ki67, and ICOS levels. G, Correlation between thefrequency of iNOSþ macrophages and CD8 T-cell abundance or GZMB, Ki67, and ICOS levels. N ¼ 26. H, CD45.1 monocytes (donor) were adoptivelytransferred into CD45.2 MC38 tumor–bearing animals (recipient) treated with isotype or aPDL1 antibodies. Two days after transfer, tumors were harvestedand stained for FACS analysis. H, Pie charts depicting the proportion of M1/M2 subsets in recently differentiated donor-derived TAMs. I, Frequency ofARG1þ and iNOSþ cells as well as CD40 MFI on donor-derived TAMs. J, Frequency of iNOSþ cells and CD40 MFI on donor-derived monocytes. N ¼ 5;� , P < 0.05; �� , P < 0.01; ��� , P < 0.001.

A Gene set Top−scoring genes

complex_of_collagen_trimersextracellular_matrix_structural_constituentneuron_fate_specificationmetanephric_nephron_developmenttongue_developmentcollagen_trimerspinal_cord_motor_neuron_cell_fate_specificationplatelet_derived_growth_factor_bindingproximal_distal_pattern_formationnephron_tubule_formationimmunoglobulin_bindingrespiratory_burstigg_bindingregulation_of_b_cell_receptor_signaling_pathwaymhc_class_ii_protein_complexantimicrobial_humoral_responsebeta_2_microglobulin_bindingdetection_of_other_organismpositive_regulation_of_natural_killer_cell_mediated_immunitymhc_protein_complex

Col11a2, Col3a1, Col6a1, Dcn, Col11a1, Col5a2, ...Col11a2, Col3a1, Col12a1, Eln, Lamc1, Col11a1, ...Gli2, Pax6, Hoxc10, Hoxd10, Gli3, Lbx1, ...Cd34, Lamb2, Smo, Kif26b, Pdgfra, Irx2, ...Wnt10a, Six4, Lef1, Gli3, Cyp26b1, Bnc2, ...Col11a2, Col3a1, Col6a1, Col12a1, C1qtnf6, Col6a2, ...Gli2, Pax6, Hoxc10, Hoxd10, Gli3, Nkx6−1, ...Col3a1, Col6a1, Col5a1, Col4a1, Pdgfra, Pdgfrb, ...Lrp4, Gli2, Aldh1a2, Hoxd9, Irx3, Hoxa9, ...Irx3, Irx2, Sox9, Osr1, Irx1, Six1, ...Fcgr1, Fcgr4, Fcgr2b, Fcgrt, Fcgr3, Fcer1g, ...Slc11a1, Cybb, Ncf1, Ncf2, Pik3cd, Hck, ...Fcgr1, Fcgr4, Fcgr2b, Fcgrt, Fcgr3, Fcer1g, ...Plcl2, Cmtm3, Cd300a, Ptpn6, Lyn, Nfam1, ...H2−DMa, H2−DMb1, Cd74, H2−Ab1, H2−DMb2, H2−Eb1, ...H2−T23, H2−K1, H2−D1, App, H2−M3, H2−Q4, ...H2−T23, H2−K1, H2−D1, Cd1d1, H2−M3, H2−Q4, ...H2−T23, Nlrc4, Nod1, H2−K1, Tlr4, Tlr6, ...H2−T23, H2−K1, H2−D1, Cd226, H2−M3, H2−Q4, ...H2−T23, H2−K1, H2−DMa, H2−DMb1, Cd74, H2−Ab1, ...

4 2 2 4

−log10 adj. P

by

aP

DL1

tre

atm

en

tUp

reg

ula

ted

Do

wn

reg

ula

ted

MHC complexFCγ receptorCollagenDevelopment

Isotype aPDL1

−5

−4

−3

−2

−1

0

IFN Pathways

Ge

ne

sig

na

ture

sco

re

P = 0.00794

Isotype aPDL1

01

23

4

MHC Class II family

Ge

ne

sig

na

ture

sco

re

P = 0.00794

Isotype aPDL1

−5

−4

−3

−2

−1

ProinflammatoryG

en

e s

ign

atu

re s

core

P = 0.01587

Isotype aPDL1

−2.

5−

2.0

−1.

5−

1.0

−0.

50.

0

Chemotaxis

Ge

ne

sig

na

ture

sco

re

P = 0.00794

B

Figure 3.

aPDL1 treatment leads to genome-wide TAM polarization toward a more proinflammatory phenotype. TAMswere sorted from untreated or aPDL1-treated MC38tumors 7 days after treatment. RNAwas subsequently purified for high-throughput sequencing. A, Select GO gene sets enriched in genes differentially expressedin macrophages isolated from control versus aPDL1-treated tumors. Shown are adjusted�log10 P values for enrichment of the ten most significantly up- andsignificantly downregulated GO gene sets in aPDL1-treated macrophages. Gene sets that are inferred to reflect key underlying biological processes are colored.Only the top six genes per set (ranked by single-gene P value) are shown. B, Expression of predefined gene sets (see Supplementary Table S1) representingpathways of interest in isotype versus aPDL1-treated macrophages. Expression of all genes within a set was summarized into one score. Differential expression ofgene set scores was assessed using two-sidedWilcoxon rank-sum tests. N¼ 5/group.

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1499

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 8: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

Isotype aPDL10

20

40

60

80

% IF

Nγ+

in C

D8

T C

ells

*

IFNγ

CD8

CD4

NK

CD45-

0

20

40

60

0

2,000

4,000

6,000

8,000

10,000

+γNFI f o

%IF

M γNFI

CD8 NK CD45-CD4

CD8 NK CD45-CD4

0

2,000

4,000

6,000IF

of T

umor

g/)lm/gp(

Isotype aPDL1

****

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

20

40

60

80

% M

1-Li

ke

********

* ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

20

40

60

80

100

% M

1/M

2 In

term

edia

te * ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

20

40

60

80

100

% M

2-Li

ke

********

*ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

10

20

30

40

% A

RG1

+ in

Mac

roph

ages

* **ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

2

4

6

% iN

OS+

in M

acro

phag

es ** *****

ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

10

20

30

% iN

OS+

in M

onoc

ytes **** ****

****

ns

* **

ns

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

0

20000

40000

60000

80000

MH

CII

MFI

on

Mac

roph

ages

CD

40 M

FI o

n M

acro

phag

es

0

20

40

60

% M

1-Li

ke

**

IFNγ R +/+ -/-

% M

1/M

2 In

term

edia

te **

0

20

40

60

80

+/+ -/-0

20

40

60

80

100

% M

2-Li

ke

***

+/+ -/- IFNγR +/+ -/-

0

20

40

60

***

% A

RG

1+ in

Mac

roph

ages

IFNγR +/+ -/- +/+ -/-ISO aPDL1

***

*

MH

CII

MFI

on

Mac

roph

ages

0

10,000

20,000

30,000

40,000

50,000

+/+ -/- +/+ -/-ISO aPDL1

****

**

% iN

OS+

in M

onoc

ytes

0

10

20

30

+/+ -/- +/+ -/-ISO aPDL1

***

**

% iN

OS+

in M

acro

phag

es

0

1

2

3

4

5

+/+ -/- +/+ -/-ISO aPDL1

****

**

0

5,000

10,000

15,000

CD

40 M

FI o

n M

acro

phag

es

+/+ -/- +/+ -/-ISO aPDL1

F

E

D

C

A B

Isotyp

e

aPDL1

aIFNg

aPDL1+

aIFNg

6,000

8,000

10,000

12,000 ** ***

M1-Like

M1/M2 Intermediate

M2-Like

Figure 4.

aPDL1 remodeling of the macrophage compartment is IFNg-mediated and dependent on IFNgR expression on TAMs. A, IFNg levels as measured by Luminex fromMC38 tumor supernatant from isotype or aPDL1-treated tumors. B, Frequency of IFNgþ cells among intratumoral CD8 T cells and IFNg-expressing cell subsetsand mean fluorescent intensity (MFI) following treatment. C and D,MC38 tumors were treated with isotype or aPDL1 antibodies in the presence or absence ofneutralizing anti-IFNg . C, Frequency of M1/M2 subsets following treatment. D, Frequency of ARG1þ and iNOSþmacrophages (or monocytes), as well as MHCIIexpression onmacrophages. E,WT and IFNgR-deficient mixed bone marrow chimeras were inoculated with MC38 and once tumors became established, micewere randomized and treated with aPDL1 or isotype control antibody. Frequency of M1/M2 subsets amongWT (þ/þ) and IFNgR-deficient (�/�) macrophages.F, Frequency of ARG1þ and iNOSþmacrophages as well as MHCII and CD40 levels. �, P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001; ns, nonsignificant.

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1500

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 9: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

(IFNgR�/�) cells.Weobserved apronounced shift to a suppressiveM2-like phenotype in IFNgR�/� TAMs at baseline (Fig. 4E). ARG1levels were also increased while iNOS, MHCII, and CD40 expres-sion was significantly decreased in IFNgR�/� macrophages com-pared with their IFNgRþ/þ counterparts, which persisted evenafter aPDL1 treatment (Fig. 4F). These results corroborate themajor role of intratumoral IFNg levels in mediating the pheno-typic skewing of the macrophage compartment in tumors. Col-lectively, these findings demonstrate a direct cross-talk betweenIFNg , mainly produced by CD8 T cells following aPDL1 treat-ment, and remodeling of the macrophage compartment.

Dissociated tumor supernatant from aPDL1-treated tumorsdrives BMDM polarization that enhances T-cell proliferationand activation

To further interrogate the relationship between the tumormicroenvironment following aPDL1 treatment and the polari-zation of TAMs, we developed a BMDM polarization assay usingdissociated tumor supernatant from ex vivo–digested MC38tumors. Dissociated tumor supernatant from untreated tumorsled to robust M2-like polarization, with its effect graduallyweakening with serial dilution (Supplementary Fig. S4A). Exog-enous IFNg robustly polarized BMDMs in the presence ofdissociated tumor supernatant, reducing ARG1 levels andincreasing iNOS, MHCII, and CD40 expression (SupplementaryFig. S4B and S4C). Importantly, dissociated tumor supernatantfrom aPDL1-treated tumors, moderately but significantly polar-ized macrophages from M2-like to an intermediate state andelevated their expression of MHCII and CD40, as well as MHCI,CD86, and FcgRI (Fig. 5A), consistent with the gene expressionprofiling in sorted macrophages from aPDL1-treated tumorsin vivo (Fig. 3; Supplementary Fig. S2B). In addition, dissociatedaPDL1-treated tumor supernatant decreased ARG1 levels whileincreasing iNOS expression (Supplementary Fig. S4D), albeit thefrequency of iNOS-producing BMDMs was low. Importantly,combining dissociated tumor supernatant from aPDL1-treatedtumors with IFNg blockade reverted most of the phenotypicchanges observed, suggesting that IFNg is a major driver ofBMDM polarization in dissociated tumor supernatants fromaPDL1-treated tumors.

We next examined the functional output of polarizedBMDMs by coculture of OVA323-339–loaded BMDMs withCFSE-labeled OT-II cells. BMDMs polarized with dissociatedtumor supernatant from aPDL1-treated tumors, or tumor super-natant supplemented with exogenous IFNg , induced robust OT-II proliferation as determined by CFSE dilution (Fig. 5B; Sup-plementary Fig. S4E and S4F). In addition, these OT-II cellssignificantly upregulated their expression of the activationmarkers CD69 and CD25 and showed increased coexpressionof CD44 and ICOS when compared with OT-II cells coculturedwith control polarized BMDMs (Fig. 5B; SupplementaryFig. S4F). Furthermore, blockade of IFNg during the course ofBMDM polarization with dissociated tumor supernatant fromaPDL1-treated tumors, partially abrogated the subsequent pro-liferation and activation of OT-II cells (Fig. 5B). These resultsillustrate that increased IFNg levels following aPDL1 treatmentcan impact not only the phenotype of TAMs, but more impor-tantly, enhance their ability to promote T-cell activation andproliferation. Tumor macrophages therefore represent an indi-rect downstream target of aPDL1 treatment in responsivetumors, which can form a positive feedback loop to T cells.

Remodeling of the macrophage compartment is observed inaPDL1-responsive tumor models

To expand our understanding of aPDL1-mediatedmacrophageremodeling, we studied two additional tumor models: EMT6 thatalso responds to aPDL1, and JC that is refractory to treatment.These two tumor models have substantial TAM infiltration butexhibit different overall immune profiles and TAM phenotypes(Supplementary Fig. S5A and S5B). Following aPDL1 treatment,there was a shift from M2-like to M1-like macrophages in EMT6,but not in JC tumors (Supplementary Fig. S5C). Furthermore,there was a significant decrease in the absolute numbers of ARG1-producing macrophages and an increase in the frequency ofiNOS-producing TAMs and monocytes, as well as their MHCIIand CD40 expression in EMT6 similar to MC38, but this was notobserved in JC tumors (Supplementary Fig. S6A–S6D). Theseobservations indicate a possible and consistent remodeling ofthe macrophage compartment in aPDL1-responsive tumor mod-els that is absent in a nonresponsive tumor context. Thiswas likelydue, in part, to a lack of IFNg induction following aPDL1 treat-ment in JC tumors (Supplementary Fig. S6E and S6F) and a moresuppressive TAM phenotype given the lower IFNg levels at base-line in JC when compared with MC38 and EMT6 tumors.

TAMphenotype and density influences the level ofmacrophageremodeling

Given the direct polarizing effect of agonistic aCD40 onTAMs (8, 22, 27), we investigated the capacity of aCD40 to furtherpush the remodeling mediated by aPDL1 in different tumorimmune contextures. To that end, we used macrophage-richMC38 and EMT6 tumors (Supplementary Fig. S1A and S5A),with MC38 having a higher baseline abundance of macrophageswith increased ARG1 expression and decreased iNOS levels whencompared with macrophages in EMT6 tumors (Fig. 6A). Bothmodels respond to aPDL1 treatment, but antitumor activity(Fig. 6B) and the remodeling effect of aPDL1 on TAMs were morepronounced in EMT6 than MC38 tumors (Fig. 2A–C; Supple-mentary Fig. S6A–S6C). This led us to speculate that furtherpolarizing macrophages with aCD40 would combine more effec-tively with aPDL1 in the EMT6 tumor model given its lowermacrophage burden and less suppressive phenotype. Indeed, assoon as 48 hours after a single dose of aCD40, a significantremodeling of the macrophage compartment was observed inEMT6, characterized by potentiated M1-like polarization and amarked increase in MHCII and MHCI expression with decreasedCD206 and ARG1 (Fig. 6C). Some combinatorial effects withaPDL1were alreadynoticeable (i.e., CD206 andARG1), but iNOSlevels were not substantially changed at this time point. On theother hand, aCD40 led to milder M1-like polarization on macro-phages in MC38, mainly associated with a reduction in CD206(Fig. 6D). There were no combinatorial effects with aPDL1 basedon the markers examined. These results suggest that reinforcingthe polarization of macrophages by combining aPDL1 withaCD40 can further unleash antitumor activity, but the combina-torial effect is likely dependent on various parameters includingthe abundance and baseline phenotype of TAMs.

Combining aCD40 with aCSF1R has recently been shown toaugment responses when compared with either treatmentalone (27) and a single dose of aCSF1R was sufficient to signif-icantly reduce both macrophage and monocyte numbers intumors, without affecting CD8 T-cell abundance (Fig. 7A). Wethus speculated that in the MC38 tumor model, in which

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1501

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 10: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

combination of aPDL1 with aCD40 was insufficient to signifi-cantly remodel the macrophage compartment, reducing the sup-pressive macrophage burden by polarizing and depleting TAMswould maximize the antitumor activity mediated by aPDL1. LikeaCD40, aCSF1R only mildly synergized with aPDL1 treatment,implying the limited benefit of eithermacrophage polarization ordepletion in MC38 (Fig. 7B and C). Combination of aCD40 withaCSF1R, however, elicited a robust antitumor response, resultingin 70% CRs and corroborating the immune-suppressive charac-teristic of TAMs in this tumor model. Remarkably, the combina-tion of aCD40 and aCSF1R with aPDL1 demonstrated the stron-gest combinatorial activity, resulting in 90% CRs and clearlyimproving the efficacy of aPDL1 or any other two-agent combina-tions. These results suggest that in tumor immune contextures thatare characterized by a high density of suppressive TAMs, remodel-

ing of the macrophage compartment by aPDL1 or even a moredirect polarizing agent like aCD40 might not be enough toenhance responses. Here, both macrophage depletion and polar-ization will likely be needed to efficiently reduce macrophageburden and further unleash aPDL1-mediated T-cell responses.

DiscussionTAMs are shaped by the tumor microenvironment and they, in

turn, actively shape theirmicroenvironment. Tumormacrophagescan have diverse cross-talk with other immune cell subsets,and their phenotype and function can vary among tumortypes and stages of disease. In this study, we show that aPDL1treatment can remodel the macrophage compartment, resultingin a more proinflammatory phenotype in both monocytes and

**** ****

MH

CI M

FI

epyt

osI

1LD

Pa

− −

1LD

Pa

+

6,000

8,000

10,000

12,000

14,000

** ***

MC38 Supernatant

anti-IFNγ

0

10

20

30

40

50

** * * *

% C

D4

4+

ICO

S+

CFS

E M

F I

MC38 Supernatant

Iso

typ

e

aP

DL1

anti-IFNγ − −

1LD

Pa

+

Iso

typ

e

aP

DL1

− -

aP

DL1

+

0

200,0000

400,0000

600,0000

800,0000

0

20

40

60

80

100

* *

% C

D6

9+

Iso

typ

e

aP

DL1

− −

aP

DL1

+

60

65

70

75

80

85* *

% C

D2

5+

Iso

typ

e

aP

DL1

− −

aP

DL1

+

55

60

65

70

75

80

85

CFSE

Isotype

aPDL1

aPDL1+aIFNγ

0

2,000

4,000

6,000

8,000

2,500

3,000

3,500

4,000

50

60

70

80**** ******** **** **** ****

% M

1/M

2-I

nte

rme

dia

te

% M

2-Li

ke

MH

CII

MFI

CD

40

MFI

* ***

MC38 SupernatantIs

oty

pe

aP

DL1

anti-IFNγ − −

1LD

Pa

+

epyt

osI

1LD

Pa

− −

1LD

Pa

+

Iso

typ

e

aP

DL1

− −

aP

DL1

+

Iso

typ

e

aP

DL1

− −

aP

DL1

+

A

B

6,000

7,000

8,000

9,000

10,000

CD

86

MFI

** ***

epyt

osI

1LD

Pa

− −1L

DP

a+

5,000

6,000

7,000

8,000

9,000 * *

Iso

typ

e

aP

DL1

− −

aP

DL1

+

FcγR

I MF

I

Figure 5.

Dissociated tumor supernatant from aPDL1-treated tumors drives BMDM polarization that enhances T-cell proliferation and activation. MC38 tumors fromisotype or aPDL1-treated mice were digested and supernatant collected for subsequent assays. BMDMswere generated and polarized with tumor supernatant inthe presence or absence of a neutralizing aIFNg antibody. A, Frequency of M1/M2 subsets and MHCII, CD40, MHCI, CD86, and FcgRI (CD64) expression levels onBMDMs polarized with supernatant from isotype or aPDL1-treated tumors. B, BMDMs were loaded with OVA323-339 and cocultured with CFSE-labeled na€�ve OT-IIcells. OT-II cell proliferation and activation were determined by CFSE dilution, CD25, and CD69 staining, as well as CD44 and ICOS coexpression, respectively.Lines connect data acquired from the same bonemarrow donor. Paired t tests were performed. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001. MFI, meanfluorescent intensity.

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1502

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 11: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

BIsotype anti-PD-L1

C

Isotype anti-PD-L1

anti-CD40 Combo

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

MC38EMT6

CR: 10%

CR: 0% CR: 10%

CR: 70%

A

Day Day

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

CR: 10%

CR: 10%

CR: 10%

CR: 0%

anti-CD40 Combo

Vo

lum

e (m

m3

)

Vo

lum

e (m

m3

)

Vo

l um

e (m

m3

)

Vo

lum

e (m

m3

)

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

0 10 20 30 40 508

16

32

64

128

256

512

1,024

2,048

4,096

DayDay

Isoty

pe

aCD40

aPDL1

Combo

0

20,000

40,000

60,000

80,000

MH

CII

MFI

on

Mac

roph

ages ***

***

EMT6 D MC38

Isoty

pe

aCD40

aPDL1

Combo

0

5

10

15

M1-

Like

%

**

*

Isoty

pe

aCD40

aPDL1

Combo

0

20

40

60

80

100

M1/

M2

Inte

rmed

iate

%

Isoty

pe

aCD40

aPDL1

Combo

0

20

40

60

80

100

M2-

Like

%

Isoty

pe

aCD40

aPDL1

Combo

0

10,000

20,000

30,000

40,000

50,000

MH

CII

MFI

on

Mac

roph

ages

Isoty

pe

aCD40

aPDL1

Combo

0

20,000

40,000

60,000

MH

CI M

FI o

n M

acro

phag

es

Isoty

pe

aCD40

aPDL1

Combo

0

1,000

2,000

3,000

4,000

5,000

CD

206

MFI

on

Mac

roph

ages

****

***

0

2,000

4,000

6,000

8,000

Isoty

pe

aCD40

aPDL1

Combo

AR

G1

MFI

on

Mac

roph

ages

****

0

2,000

4,000

6,000

8,000

10,000

Isoty

pe

aCD40

aPDL1

Combo

iNO

S M

FI o

n M

acro

ph

ages

**

0

2,000

4,000

6,000

8,000

10,000

Isoty

pe

aCD40

aPDL1

Combo

iNO

S M

FI o

n M

on

ocy

tes

*

Isoty

pe

aCD40

aPDL1

Combo

2,000

3,000

4,000

5,000

6,000

CD

206

MFI

on

Mac

roph

ages

**

***

*

0

5,000

10,000

15,000

20,000

Isoty

pe

aCD40

aPDL1

Combo

MH

CI M

FI o

n M

acro

phag

es ****

Isoty

pe

aCD40

aPDL1

Combo

0

2

4

6

8

M1-

Like

% ***

**

Isoty

pe

aCD40

aPDL1

Combo

0

10

20

30

M1/

M2

Inte

rmed

iate

% ***

Isoty

pe

aCD40

aPDL1

Combo

40

60

80

100

M2-

Like

%

***

0

5,000

10,000

15,000

20,000

Isoty

pe

aCD40

aPDL1

Combo

iNO

S M

FI o

n M

on

ocy

tes

Isoty

pe

aCD40

aPDL1

Combo

1,500

2,000

2,500

3,000

3,500

4,000

AR

G1

MFI

on

Mac

roph

ages

***

****

****

0

2,000

4,000

6,000

8,000

10,000

Isoty

pe

aCD40

aPDL1

Combo

iNO

S M

FI o

n M

acro

ph

ages

MC38 EMT6

Mac

rop

hag

e #

/mg

**

0

2,000

4,000

6,000

8,000

10,000

iNO

S+ M

acro

ph

age

#/m

g

MC38 EMT6

***

MC38 EMT6

0

500

1,000

1,500

AR

G1

+ M

acro

ph

age

#/m

g

0

50

100

150

Figure 6.

Directed TAM polarization with aCD40 combines effectively with aPDL1 in the EMT6 tumor model. Mice were inoculated with either MC38 or EMT6 tumor cellsand once tumors reached a volume of approximately 180 mm3 (day 0), mice with similarly sized tumors were randomized into treatment groups. The next day(day 1), mice were dosed with either isotype control (anti-gp120), aPDL1, or a single dose of aCD40. A, Absolute number of total, ARG1þ, and iNOSþ

macrophages per mg of tissue in MC38 and EMT6 tumors at baseline. B, Tumor volume (mm3) of control (black), aPDL1 (blue), aCD40 (red), or combo (green)-treated EMT6 and MC38 tumor–bearing mice is shown on a log2 scale; N¼ 10/group. C and D, Pharmacodynamic analysis of tumor macrophages 48 hoursafter treatment initiation evaluating the M1/M2-like subset frequency and the expression of MHCI, MHCII, CD206, ARG1, and iNOS. � , P < 0.05; �� , P < 0.01;��� , P < 0.001; ���� , P < 0.0001. MFI, mean fluorescent intensity.

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1503

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 12: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

macrophages. This was evidenced not only by changes in canon-ical M1/M2 marker expression like ARG1, iNOS, MHCII, andCD40, but also by increased expression of genes associated withantigen uptake, processing and presentation machinery, proin-flammatory cytokines and T-cell–recruiting chemokines. We fur-ther demonstrate that TAM polarization was largely mediated byincreased IFNg signaling and required IFNgR expression onTAMs.We also find that while remodeling of the macrophage compart-ment was observed in responsive MC38 and EMT6 tumors, thiswas not evident in nonresponsive JC, which had lower IFNginduction following treatment. Our results are in agreement with

recent findings by Gubin and colleagues who observed a similarpolarization and remodeling of the macrophage compartmentlargely mediated by IFNg following aPD1 and aCTLA4 treatmentand that initiated at themonocytic precursor stage (37). Together,our results confirm that the phenotype of the intratumoral mac-rophage compartment is influenced by the cytokine milieu thatcan be altered by treatment and provides new insights into themechanism of action of checkpoint inhibitors.

Importantly, we demonstrate that aPDL1-mediated macro-phage polarization had a beneficial effect on T-cell responsesincreasing their proliferation and cytolytic potential.

anti-CD40B

CR: 90%

anti−PD-L1 anti−CSF1R

anti−PD-L1 + anti−CD40 + anti−CSF1R

anti−CD40 + anti-CSF1Ranti−PD-L1 + anti-CD40anti−PD-L1 + anti-CSF1R

C

Isotype

CR: 70%CR: 30% CR: 50%

CR: 20%0 20 40 60

8163264

128256512

1,0242,0484,096

0 20 40 608

163264

128256512

1,0242,0484,096

A

0 20 40 608

163264

128256512

1,0242,0484,096

Days

Days Days Days

Days

Days Days Days

Volu

me

(mm

3 )Vo

lum

e (m

m3 )

Volu

me

(mm

3 )

0 20 40 608

163264

128256512

1,0242,0484,096

0 20 40 608

163264

128256512

1,0242,0484,096

0 20 40 608

163264

128256512

1,0242,0484,096

0 20 40 608

163264

128256512

1,0242,0484,096

0 20 40 608

163264

128256512

1,0242,0484,096

0 20 400

20

40

60

80

100

Time

Perc

ent s

urvi

val

IsoaPDL1aCD40aCSF1RaPDL1 + aCD40aPDL1 + aCSF1RaCD40 + aCSF1RaPDL1 + aCD40 + aCSF1R

3/10

5/106/10

7/10

9/10

Isoty

pe

aCSF1R

Isoty

pe

aCSF1R

0

2,000

4,000

6,000

8,000

Day 3 Day 6

Mac

roph

age

# /m

g tu

mor

*

**

Isoty

pe

aCSF1R

Isoty

pe

aCSF1R

0

1,000

2,000

3,000

Day 3 Day 6

**

*

Mon

ocyt

e #

/mg

tum

or

Isoty

pe

aCSF1R

Isoty

pe

aCSF1R

0

50

100

150

200

Neu

trop

hil #

/mg

tum

or NS*

Day 3 Day 6

Isoty

pe

aCSF1R

Isoty

pe

aCSF1R

0

1,000

2,000

3,000

4,000

5,000

Day 3 Day 6

CD

8 T

cel

l # /m

g tu

mor

NS

NS

Figure 7.

TAM polarization and depletion combine effectively with aPDL1 treatment in the MC38 tumor model. Mice were inoculated with MC38 and once tumors reached avolume of approximately 180 mm3 (day 0), mice with similarly sized tumors were randomized into treatment groups. The next day (day 1), mice were dosed witheither isotype control, aPDL1, aCD40 (4mg/kg single dose), or aCSF1R (30 mg/kg single dose) alone or in combination. A, Abundance of intratumoral immuneinfiltrates 3 and 6 days after a single dose of aCSF1R. B, Tumor volume (mm3) of control (black), aPDL1 (blue), aCD40 (red), aCSF1R (orange), combination of two(green), or three agents (purple)-treated MC38 tumor–bearing mice is shown on a log2 scale. C, Survival curves from treatment groups in B. N¼ 10 mice pergroup. � , P < 0.05; �� , P < 0.01; NS, nonsignificant.

Xiong et al.

Cancer Res; 79(7) April 1, 2019 Cancer Research1504

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 13: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

Functionally, BMDMs polarized with dissociated tumor superna-tant from aPDL1-treated tumors were significantly more effectiveat driving theproliferation andactivationofOVA-specific T cells invitro than control-treated BMDMs, suggesting a beneficial, func-tional consequence of macrophage polarization induced byaPDL1-mediated microenvironment changes. This was furtherevidenced by transfer of monocytes into aPDL1-treated tumorsthat led to the differentiation of TAMs toward a more proin-flammatory phenotype as well as an increase in CTL infiltrationand GZMB expression. Polarization of macrophages followingaPDL1 treatment can therefore improve T-cell responses. Ourresults highlight the beneficial impact of aPDL1 in shaping themacrophage compartment in responsive tumor models, which inturn can provide a positive feedback loop to T cells, thus support-ing the notion for directly targeting these innate immune cells tofurther augment T-cell activity.

Agonistic aCD40 has been shown to rapidly activate macro-phages and enhance their tumoricidal capacity alone or in com-bination with other TAM-targeting approaches (2, 22, 27, 28). Inour studies, combining aPDL1 with a single dose of aCD40 led toan appreciable enhancement in antitumor activity demonstratinga further reduction in suppressive mechanisms that control anti-tumor responses when direct macrophage polarization was com-binedwith aPDL1.However, although a single dose of aCD40wassufficient to significantly augment responses in EMT6 tumors, thecombinatorial effect with aPDL1 was milder in MC38. It isimportant to point out that the observed combinatorial activitywith aCD40 could also be due to activation of dendritic cells,B cells, or other nonimmune cells–expressingCD40, but at least inthe models tested, macrophages represent the major immuneinfiltrate and are likely playing a major role in the response toaCD40 (Supplementary Figs. S1A and S5A).

Given that a high density of TAMs could represent a largersuppressive barrier to overcome for antitumor activity, reducingthe TAM compartment has recently been suggested (28) and islikely needed to enhance aPDL1 responses in tumor immunecontextures highly infiltrated by macrophages. Indeed, weobserved that combining aPDL1 with aCD40 and aCSF1R max-imized antitumor activity in the macrophage-rich MC38 tumormodel, whereas combining aPDL1 with either aCD40 or aCSF1Rshowed only a mild combinatorial response. Importantly, thecombination of aCD40 with aCSF1R in the absence of aPDL1significantly augmented responses, demonstrating the contribu-tion of TAMs to inhibiting antitumor responses.

Together, our results demonstrate that aPDL1 can impart aphenotypic change on the macrophage compartment mainlythrough increased IFNg levels and that this recently appreciatedmechanism of action can be further augmented by direct macro-phage targeting approaches. However, in the clinical setting,tumors characterized by a high level of suppressive TAMs couldderive more benefit from aPDL1 treatment by combining mac-rophage polarization and depleting strategies. This approachcould help proestablish a lower threshold for antitumor activity,potentially expanding the proportion and depth of respondingindividuals to checkpoint blockade.

Disclosure of Potential Conflicts of InterestH. Xiong, S. Mittman, R. Rodriguez, M. Moskalenko, P. Pacheco-Sanchez,

Y. Yang, D. Nickles, and R. Cubas have ownership interest (including stocksand patents) in Roche.

Authors' ContributionsConception and design: H. Xiong, R. CubasDevelopment of methodology: H. Xiong, S. Mittman, R. Rodriguez, R. CubasAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): H. Xiong, S. Mittman, R. Rodriguez, M. Moskalenko,P. Pacheco-Sanchez, Y. Yang, R. CubasAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): H. Xiong, S. Mittman, M. Moskalenko, P. Pacheco-Sanchez, Y. Yang, D. Nickles, R. CubasWriting, review, and/or revision of themanuscript:H. Xiong, M.Moskalenko,P. Pacheco-Sanchez, Y. Yang, D. Nickles, R. CubasAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): S. Mittman, R. Rodriguez, M. Moskalenko,P. Pacheco-Sanchez, Y. Yang, D. Nickles, R. CubasStudy supervision: R. Cubas

AcknowledgmentsThe authors would like to thank the core dosers and the cell line core group at

Genentech for their contribution to this project as well as John Silva and SharonYee for their help with the anti-CSF1R study. They would also like to thank theflow cytometry core group for their help with cell sorting.

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 October 11, 2018; revised December 12, 2018; accepted January 17,2019; published first January 24, 2019.

References1. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to

therapy. Immunity 2014;41:49–61.2. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associ-

ated macrophages as treatment targets in oncology. Nat Rev Clin Oncol2017;14:399–416.

3. Chen D, Mellman I. Elements of cancer immunity and the cancer-immuneset point. Nature 2017;541:321–30.

4. Biswas SK, Mantovani A. Macrophage plasticity and interaction withlymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11:889–96.

5. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. Thechemokine system in diverse forms of macrophage activation and polar-ization. Trends Immunol 2004;25:677–86.

6. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophageactivation: time for reassessment. F1000Prime Rep 2014;6:13.

7. Lawrence T, Natoli G. Transcriptional regulation of macrophage polariza-tion: enabling diversity with identity. Nat Rev Immunol 2011;11:750–61.

8. Baer C, Squadrito M, Laoui D, Thompson D, Hansen S, Kiialainen A, et al.Suppression of microRNA activity amplifies IFN-g-induced macrophageactivation and promotes anti-tumour immunity. Nat Cell Biol 2016;18:790–802.

9. Thorsson V, Gibbs D, Brown S, Wolf D, Bortone D, Yang T-H, et al. Theimmune landscape of cancer. Immunity 2018;48:812—30.

10. Su S, Zhao J, Xing Y, Zhang X, Liu J, Ouyang Q, et al. Immune checkpointinhibition overcomes ADCP-induced immunosuppression by macro-phages. Cell 2018;175:442–457.

11. Bronte V, Zanovello P. Regulation of immune responses by L-argininemetabolism. Nat Rev Immunol 2005;5:nri1668.

12. LechnerM, Lirk P, Rieder J. Inducible nitric oxide synthase (iNOS) in tumorbiology: the two sides of the same coin. Semin Cancer Biol 2005;15:277–89.

13. Burke AJ, Sullivan FJ, Giles FJ, Glynn SA. The yin and yang of nitric oxide incancer progression. Carcinogenesis 2013;34:503–12.

Anti–PD-L1 Treatment Can Remodel the Macrophage Compartment

www.aacrjournals.org Cancer Res; 79(7) April 1, 2019 1505

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 14: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

14. Georgoudaki A-M, Prokopec KE, Boura VF, Hellqvist E, Sohn S, €Ostling J,et al. Reprogramming tumor-associated macrophages by antibody target-ing inhibits cancer progression and metastasis. Cell Reports 2016;15:2000–11.

15. Gao J, Ward J, Pettaway C, Shi L, Subudhi S, Vence L, et al. VISTA is aninhibitory immune checkpoint that is increased after ipilimumab therapyin patients with prostate cancer. Nat Med 2017;23:551–55.

16. De Henau O, Rausch M, Winkler D, Campesato LF, Liu C, Cymerman DH,et al. Overcoming resistance to checkpoint blockade therapy by targetingPI3Kg in myeloid cells. Nature 2016;539:443–7.

17. Qian B-ZZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2recruits inflammatory monocytes to facilitate breast-tumour metastasis.Nature 2011;475:222–5.

18. Franklin RA, LiaoW, Sarkar A, KimMV, BivonaMR, Liu K, et al. The cellularandmolecular origin of tumor-associatedmacrophages. Science 2014;344:921–5.

19. McCracken MN, Cha AC, Weissman IL. Molecular pathways: activating Tcells after cancer cell phagocytosis from blockade of CD47 "Don't Eat Me"signals. Clin Cancer Res 2015;21:3597–601.

20. Tseng D, Volkmer J-PP, Willingham SB, Contreras-Trujillo H, Fathman JW,Fernhoff NB, et al. Anti-CD47 antibody-mediated phagocytosis of cancerby macrophages primes an effective antitumor T-cell response. Proc NatlAcad Sci U S A 2013;110:11103–8.

21. 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.

22. Beatty G, Chiorean E, Fishman M, Saboury B, Teitelbaum U, Sun W, et al.CD40 agonists alter tumor stroma and show efficacy against pancreaticcarcinoma in mice and humans. Science 2011;331:1612–6.

23. 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.

24. 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.

25. KanedaM, Messer K, Ralainirina N, Li H, Leem C, Gorjestani S, et al. PI3Kgis amolecular switch that controls immune suppression. Nature 2016;539:437–42.

26. Holmgaard RB, Brachfeld A, Gasmi B, Jones DR, Mattar M, Doman T, et al.Timing of CSF-1/CSF-1R signaling blockade is critical to improvingresponses to CTLA-4 based immunotherapy. Oncoimmunology 2016;5:e1151595.

27. Hoves S, Ooi C-H, Wolter C, Sade H, Bissinger S, Schmittnaegel M, et al.Rapid activation of tumor-associated macrophages boosts preexistingtumor immunity. J Exp Med 2018;215:859–76.

28. Perry C,Mu~noz-Rojas A,MeethK, KellmanL, Amezquita R, Thakral D, et al.Myeloid-targeted immunotherapies act in synergy to induce inflammationand antitumor immunity. J Exp Med 2018;215:877.

29. Sharma P, Allison J. The future of immune checkpoint therapy. Science2015;348:56–61.

30. Brahmer J, Tykodi S, Chow L, HwuW-J, Topalian S, Hwu P, et al. Safety andactivity of anti–PD-L1 antibody in patients with advanced cancer. N Engl JMed 2012;366:2455–65.

31. Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. MPDL3280A(anti-PD-L1) treatment leads to clinical activity in metastatic bladdercancer. Nature 2014;515:558–62.

32. Pauken K, Sammons M, Odorizzi P, Manne S, Godec J, Khan O, et al.Epigenetic stability of exhausted T cells limits durability of reinvigorationby PD-1 blockade. Science 2016;354:1160–65.

33. Huang A, Postow M, Orlowski R, Mick R, Bengsch B, Manne S, et al. T-cellinvigoration to tumour burden ratio associated with anti-PD-1 response.Nature 2017;545:60–65.

34. Sen D, Kaminski J, Barnitz R, Kurachi M, Gerdemann U, Yates K, et al.The epigenetic landscape of T cell exhaustion. Science 2016;354:1165–69.

35. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al.Mismatch-repair deficiency predicts response of solid tumors to PD-1blockade. Science 2017;357:409–13.

36. Herbst RS, Soria J-CC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al.Predictive correlates of response to the anti-PD-L1 antibody MPDL3280Ain cancer patients. Nature 2014;515:563–7.

37. Gubin MM, Esaulova E, Ward JP, Malkova ON, Runci D, Wong P, et al.High-dimensional analysis delineates myeloid and lymphoid compart-ment remodeling during successful immune-checkpoint cancer therapy.Cell 2018;175:1014–30.

38. Love M, Huber W, Anders S. Moderated estimation of fold changeand dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.

39. Ritchie M, Phipson B, Wu D, Hu Y, Law C, Shi W, et al. limma powersdifferential expression analyses for RNA-sequencing and microarray stud-ies. Nucleic Acids Res 2015;43:e47.

40. SubramanianA, TamayoP,Mootha VK,Mukherjee S, Ebert BL,GilletteMA,et al. Gene set enrichment analysis: a knowledge-based approach forinterpreting genome-wide expression profiles. Proc Natl Acad Sci USA2005;102:15545–50.

41. Wu D, Smyth G. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res 2012;40:e133.

42. DulucD, CorvaisierM, Blanchard S, Catala L,Descamps P,Gamelin E, et al.Interferon-gamma reverses the immunosuppressive and protumoral prop-erties and prevents the generation of human tumor-associated macro-phages. Int J Cancer 2009;125:367–73.

Cancer Res; 79(7) April 1, 2019 Cancer Research1506

Xiong et al.

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208

Page 15: Anti PD-L1 Treatment Results in Functional Remodeling of ...€¦ · ratio compared with control-treated mice. Although in responsive tumor models, anti– PD-L1 treatment remodeled

2019;79:1493-1506. Published OnlineFirst January 24, 2019.Cancer Res   Huizhong Xiong, Stephanie Mittman, Ryan Rodriguez, et al.   Macrophage Compartment

PD-L1 Treatment Results in Functional Remodeling of the−Anti

  Updated version

  10.1158/0008-5472.CAN-18-3208doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2019/01/24/0008-5472.CAN-18-3208.DC1

Access the most recent supplemental material at:

   

  Overview

Visual 

http://cancerres.aacrjournals.org/content/79/7/1493/F1.large.jpgA diagrammatic summary of the major findings and biological implications:

   

  Cited articles

  http://cancerres.aacrjournals.org/content/79/7/1493.full#ref-list-1

This article cites 41 articles, 11 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/79/7/1493.full#related-urls

This article has been cited by 8 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/79/7/1493To request permission to re-use all or part of this article, use this link

on September 13, 2020. © 2019 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2019; DOI: 10.1158/0008-5472.CAN-18-3208