Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1 -derived CXCL9 and CXCL10 are critically required for anti-tumor immune responses 2 following immune checkpoint blockade

3 Imran G. House1,2,+, Peter Savas2,3,+, Junyun Lai1,2, Amanda X.Y. Chen1,2, Amanda J. Oliver1,2, Zhi L. 4 Teo2,3, Kirsten L. Todd1,2 Melissa A. Henderson1,2, Lauren Giuffrida1,2, Emma V. Petley1,2, Kevin Sek1,2, 5 Sherly Mardiana1,2, Tuba N. Gide4, Camelia Quek4, Richard A. Scolyer4,5, Georgina V. Long4,6,7 James S. 6 Wilmott4, Sherene Loi2,3, Phillip K. Darcy1, 2, 8, 9* and Paul A. Beavis1,2*

7 1Cancer Immunology Program, Peter MacCallum Centre, Melbourne, 3000, Victoria, Australia. 8 2Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia. 9 3Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, 10 Australia. 4The University of Sydney, Institute Australia, Sydney, NSW, Australia. 5Royal 11 Prince Alfred Hospital, Sydney, NSW, Australia.6 Royal North Shore Hospital, Sydney, NSW, Australia. 12 7Mater Hospital, North Sydney, NSW, Australia. 8Department of Pathology, University of Melbourne, 13 Parkville. 9Department of Immunology, Monash University, Clayton. +Contributed equally, *Contributed 14 equally as senior authors.

15 Key words: cancer, anti-PD-1, anti-CTLA-4, CXCL9, CXCL10, CXCR3

16 Address correspondence and reprint requests to Paul A. Beavis ([email protected]) or Phillip K. 17 Darcy ([email protected]), Cancer Immunology Program, Peter MacCallum Cancer Centre, 18 Victoria, Australia. Telephone (613) 85595051

19 This work was funded by a Program Grant from the National Health and Medical Research Council 20 (NHMRC; Grant number 1132373) and a National Breast Cancer Foundation Grant (IIRS-19-016 19-22). 21 P. A. Beavis is supported by National Breast Cancer Foundation Fellowship (ID# ECF-17-005). P. K. 22 Darcy is supported by an NHMRC Senior Research Fellowship (APP1136680).

23 Conflict of interest: The authors declare no conflict of interest.

24

25

1

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

26 Statement of Translational Relevance

27 Treatment with immune checkpoint inhibitors has been highly successful in a subset of tumor types,

28 particularly those with increased levels of immune infiltrate. However, the mechanism and the immune

29 cell subsets critical for successful clinical responses are not fully understood. This study demonstrates that

30 the CXCL9/10 are indispensable for robust responses to immune checkpoint inhibitors (anti-

31 PD-1 and anti-CTLA-4) and, in particular, that CXCL9/10-secreting are critical for their

32 therapeutic efficacy. Moreover, we identified a novel transcriptional signature in macrophages that was

33 associated with positive patient responses to immune checkpoint inhibitors that was lacking in non-

34 responding patients. This is of significant interest to the field, challenging the paradigm that macrophages

35 are pro-tumoral and detrimental to anti-tumor immune responses through immunosuppressive

36 mechanisms, including expression of PD-L1. These findings indicate that the intratumoral macrophage

37 phenotype is a key feature in determining responses to immune checkpoint inhibitors that can be exploited

38 either diagnostically or therapeutically.

2

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

39 Abstract

40 Purpose: Response rates to immune checkpoint blockade (ICB; anti-PD-1/anti-CTLA-4) correlate with

41 the extent of tumor immune infiltrate but the mechanisms underlying the recruitment of T cells following

42 therapy are poorly characterized. A greater understanding of these processes may see the development of

43 therapeutic interventions that enhance recruitment and, consequently, improved patient outcomes.

44 We therefore investigated the chemokines essential for immune cell recruitment and subsequent

45 therapeutic efficacy of these immunotherapies.

46 Experimental design: The chemokines upregulated by dual PD-1/CTLA-4 blockade were assessed using

47 nanosting-based analysis with results confirmed at the level by flow cytometry and cytometric

48 bead array. Blocking/neutralizing confirmed the requirement for key chemokines/ and

49 immune effector cells. Results were confirmed in patients treated with immune checkpoint inhibitors using

50 single-cell RNAseq and paired survival analyses.

51 Results: The CXCR3 ligands, CXCL9 and CXCL10, were significantly upregulated following dual PD-

52 1/CTLA-4 blockade and both CD8+ T cell infiltration and therapeutic efficacy were CXCR3 dependent.

53 In both murine models and patients undergoing immunotherapy, macrophages were the predominant

54 source of CXCL9 and their depletion abrogated CD8+ T cell infiltration and the therapeutic efficacy of

55 dual immune checkpoint blockade. Single cell RNA-seq analysis of patient TILs revealed that

56 CXCL9/10/11 was predominantly expressed by macrophages following ICB and we identified a distinct

57 macrophage signature that was associated with positive responses to immune checkpoint blockade.

58 Conclusions: These data underline the fundamental importance of macrophage-derived CXCR3 ligands

59 for the therapeutic efficacy of ICB and highlight the potential of manipulating this axis to enhance patient

60 responses.

3

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

61 Introduction

62 Immunotherapy, and in particular immune checkpoint therapy, has emerged as a powerful treatment

63 strategy in several cancer types (1). Immune checkpoint inhibitors such as anti-PD-1, anti-PD-L1 and anti-

64 CTLA-4 used alone or in combination provide durable responses in a subset of patients. However, it is not

65 fully understood which parameters influence the likelihood of patient response (1, 2). Factors that have

66 been reported to predict responses to immune checkpoint inhibitors include the mutational load of the

67 tumor (3), the expression of target ligands such as PD-L1 and the presence of high number of TILs (4). It

68 is now clear that the number of CD8+ T cells at the tumor site is a strong predictor of response to anti-PD-

69 1 therapy in melanoma (4). However, a large proportion of patients exhibit an immune-exclusion or

70 immune-desert phenotype and a number of strategies are currently being developed to enhance immune

71 infiltrate in this patient subtype (5). The efficacy of immune checkpoint blockade depends upon the

72 recruitment of tumor-infiltrating and yet the mechanism by which this occurs is largely

73 unknown. Since the trafficking of immune cells, including T cells, is modulated by : chemokine

74 interactions, this prompted us to evaluate the chemokine networks responsible for the recruitment

75 of T cells following immune checkpoint blockade. Chemokines interact with their respective chemokine

76 receptors and this binding leads to the activation of intracellular signaling pathways which result in the

77 migration of the target cells towards the source of the chemokine. T cells express a range of chemokine

78 receptors including CCR2, CCR4, CCR5 and CXCR3 which respond to a range of chemokines. In the

79 context of cancer, the ligands of CCR5 (CCL5) and CXCR3 (CXCL9, CXCL10 and CXCL11) have been

80 shown to associate with levels of TIL infiltrate in human (6-8) and the production of CXCL9 and

81 CXCL10 has been associated with improved responses to chemotherapy (9-11) and adoptive cellular

82 therapy (12-14). CXCL11 binds to CXCR3 with the highest affinity, followed by CXCL10 and CXCL9

4

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

83 (15, 16) and notably CXCL11 binds to the CXCR3 receptor at a distinct binding site of the receptor (17),

84 potentially leading to diverse signaling outcomes (18).

85 In the current study, an unbiased analysis of the maximally upregulated chemokines following dual PD-

86 1/CTLA-4 blockade revealed that CXCL9 and CXCL10 were significantly upregulated in the tumor

87 microenvironment following therapy. Neutralization of CXCR3, the receptor for CXCL9 and CXCL10,

88 abrogated the therapeutic efficacy of dual PD-1/CTLA-4 blockade and was associated with reduced

89 infiltration and activation phenotype of CD8+ T cells in the tumor and draining lymph nodes. This

90 induction of CXCL9 was dependent on the production of IFNγ and TNFα since blockade of these cytokines

91 reduced the production of CXCL9 following therapeutic intervention. CRISPR-Cas9 mediated knockout

92 of CXCL9/10 in tumor cells revealed that production of these chemokines by the tumor cells was not

93 required for the efficacy of immune checkpoint blockade, inferring an important role for host-derived

94 chemokines. Interrogation of the source of CXCL9 revealed that macrophages were a major source of

95 CXCL9 chemokine and that depletion of these cells significantly impaired the efficacy of dual PD-

96 1/CTLA-4 blockade. Furthermore, investigation of the relevance of this axis in patients treated with either

97 atezolizumab (anti-PD-L1) or nivolumab (anti-PD-1) respectively revealed that the production of CXCL9

98 and CXCL10 was highly associated with improved patient prognosis and correlated with an

99 signature, underlining the importance of IFNγ in mediating this effect. Notably, single cell analysis of

100 patient tumors confirmed that macrophages were a major source of CXCL9 both prior to and after immune

101 checkpoint blockade. These studies reveal the critical importance of CXCL9 induction post

102 immunotherapy, allowing for the effective trafficking and activation of T cells. This suggests that

103 enhancing CXCL9 production by macrophages could be an effective therapeutic strategy to enhance the

104 efficacy of immunotherapy, particularly in patients with low levels of immune cells.

105

5

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

106 Materials and Methods

107 Cell lines and mice

108 The C57BL/6 mouse breast carcinoma cell line AT-3, was obtained from Dr. Trina Stewart (Griffith

109 University, Queensland) and transduced to express chicken ovalbumin as previously described (19).

110 B16F10-ova tumor cells were obtained from Dr. Jason Waithman (Telethon Kid’s Institute), MC38 tumor

111 cells were obtained from Dr. Nicole Haynes (Peter MacCallum Cancer Centre) and E0771 tumor cells

112 were obtained from Prof. Robin Anderson (Olivia Newton-John Cancer Research Institute).Tumor lines

113 were verified to be mycoplasma negative by PCR analysis and were actively passaged for less than 6

114 months. Tumor cells were grown in DMEM supplemented with 10% FCS, glutamax and

115 penicillin/streptomycin. For in vivo experiments, the indicated number of cells were resuspended in PBS

116 and injected sub-cutaneously (100 μl) or into the fourth mammary fat pad (E0771; 20µl). C57BL/6 WT

117 mice were obtained from the Walter and Eliza Hall Institute of Medical Research (Melbourne) and BATF3-

118 /- mice were bred in house. All animal experiments were approved by the Animal Experimental Ethics

119 Committee (Peter MaCallum Cancer Centre; Protocol E582).

120 Immunohistochemistry

121 Tumors were harvested and fixed overnight in 10 % neutral buffered formalin (NBF). Fixed samples were

122 embedded in paraffin and 4 μm sections were cut onto glass slides. Before staining, samples were dewaxed

123 in xylene and antigens retrieved at 125 °C for 3 minutes in 10 mM citrate buffer (Sigma) pH6. For staining,

124 Perkin Elmer OPAL reagents were used as per kit instructions. Antibodies and fluorophores were used in

125 the following order: CD3 (abcam SP7, 1:200) 690; foxP3 (ebioscience FJK-16s, 10 μg/ml) 540; PD-1

126 (abcam EPR20665, 0.8 μg/ml) 620; CD8 (ebioscience 4SM15, 5 μg/ml) 570; CK8 (abcam EP1628Y, 0.2

6

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

127 μg/ml) 520; DAPI. Multiplexing IHC images were visualized using the Perkin Elmer Vectra 3 microscope

128 and analysed using InForm v2.4.0 (Perkin Elmer) and HALO v2.3 (Indica labs).

129 Antibodies and treatment of tumor bearing mice

130 Antibodies to PD-1 (RMP1-14), CTLA-4 (9H10), isotype control (2A3), the depleting anti-F4/80

131 (CI:A3-1) and the neutralizing antibodies for CXCR3 (CXCR3-173), IFN (H22) and TNFα (TN3-19.12)

132 were purchased from BioXcell (West Lebanon, USA). Mice were treated once every 4 days with either

133 isotype control (2A3), anti-PD-1 (RMPI-14) and/or anti-CTLA-4 (9H10) for up to 4 doses.

134 Analysis of tumor-infiltrating immune subsets

135 Seven days post treatment, tumors were excised and digested post-mortem using a cocktail of 1 mg/ml

136 collagenase type IV (Sigma-Aldrich) and 0.02 mg/ml DNAase (Sigma-Aldrich). After digestion at 37ºC

137 for 30 minutes, cells were passed through a 70 μm filter twice. Inguinal lymph nodes were also harvested

138 and cells filtered through a 70 μm filter. Cells were then analyzed by flow cytometry as previously

139 described (19) and Fixable Yellow (ThermoFisher) used as a viability dye.

140 Intracellular staining

141 For detection of IFNγ and TNFα production by T cells ex vivo, TILs cells were cultured for 3 hours with

142 10 ng/ml PMA 1 µg/ml Ionomycin in the presence of Golgi Plug (BD Pharmingen) and Golgi Stop (BD

143 Pharmingen). After 3 hours cells were analysed by flow cytometry. For the detection of CXCL9, cells

144 were cultured for 3 hours in Golgi Plug/Stop in the absence of PMA/Ionomycin.

145

146

7

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

147 Cytometric Bead Array

148 Analysis of cytokine and chemokine concentrations was performed using either the Biolegend Legendplex

149 kit (#74041) or paired cytometric bead array reagents (BD Biosciences) as per manufacturer’s instructions.

150 expression analysis

151 RNA prepared from tumor cells was quantified and quality assessed using Tapestation analysis. Gene

152 count were determined by Peter MacCallum Cancer Centre Advanced Genomica Core by nCounter XT

153 assay (Mouse Pan Cancer Immune Profiling; Nanostring technologies 115000142) and analyzed by

154 nSolver 4.0 software as per manufacturer’s instructions.

155 Default quality control and normalisation steps showed all samples were adequate for further analysis.

156 Differential expression between treatment groups was performed with the R package limma [version

157 3.36.5](20), using TMM (21)normalisation and transformation of counts with ‘voom’ (22). To create the

158 mean-difference plot, the average expression of each gene was plotted against log2 of the fold change

159 between groups. with an adjusted p value less than 0.05 calculated by the differential expression

160 methodology of limma were highlighted.

161 Four clinical cohorts were utilised for analysis of the prognostic impact of chemokine expression. Gide et

162 al. analysed a cohort of metastatic melanoma patients treated with immune checkpoint blockade and

163 classified as responders or non-responders (23). The cohort consisted of 41 biopsies at baseline and 9

164 biopsies on-treatment from patients treated with anti-PD1 monotherapy, and 32 biopsies at baseline and 9

165 biopsies on-treatment from patients treated with combination anti-PD1 and anti-CTLA4. Bulk RNASeq

166 and survival data were available for all patients, and 56 patients had immunohistochemical quantification

167 of intra-tumoral CD8 T cells in baseline samples. IMvigor210 was a phase 2 trial investigating PD-L1

168 blockade with atezolizumab 1200mg 3-weekly in metastatic urothelial carcinoma patients that had

8

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

169 progressed on prior platinum-based chemotherapy. Overall survival data and whole transcriptome

170 RNASeq was available for 348 cases (24). The METABRIC cohort consists of primary breast cancers that

171 underwent microarray profiling, and have long term survival follow up available. Triple

172 negative cases were selected according to the absence of estrogen receptor staining and absence of HER2

173 amplification determined by SNP array. Gene expression data are available as log transformed normalized

174 expression values. The TCGA Melanoma cohort consists of primary that underwent RNASeq

175 with the data provided as transcripts per million (TPM). All survival analyses were done with the R

176 package ‘survival’. In each cohort, cases were divided into two groups: cases where expression of the gene

177 in question was above the median, and cases where gene expression was equal to or below the median.

178 The overall survival function of each group was estimated with the Kaplan-Meier method and compared

179 using the log-rank test. Survival curve plots were generated with the R package ‘survminer’

180 Bulk RNASeq

181 Differential expression results between tumors from responders and non-responders was obtained from

182 the R package ‘IMvigor210CoreBiologies' associated with the original publication (24) and available at

183 http://research-pub.gene.com/IMvigor210CoreBiologies/. No modifications to the published differential

184 expression analysis were made. In a second analysis of gene sets associated with CXCL9 and CXCL10

185 expression, the IMvigor cohort was re-analysed from raw count data. The count matrix was filtered to

186 remove un-expressed genes followed by principle component analysis to identify outlier samples which

187 were excluded. Counts were transformed to counts per million and samples were ranked by CXCL9 or

188 CXCL10 expression and split into two groups above and below the median expression. Competitive gene

189 set testing between high and low expression groups was performed with limma’s ‘camera’ function (25),

190 using Reactome gene sets from the Molecular Signatures Database version 5.6 C2 collection and GO term

191 associated gene sets from the C5 collection (26). Enrichment barcode plots were generated with a modified

9

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

192 version of limma’s ‘barcodeplot’ function. Log10 transformed gene counts per million were used for

193 correlations and survival analyses with the cohort split by median gene expression as described. The Gide

194 et al. melanoma dataset described in the previous section was analysed as per the original study, with log10

195 transformed counts per million used for correlations and survival analyses (23). Data from another Data

196 from another cohort of metastatic melanoma treated with checkpoint blockade associated with Riaz et al

197 (27) were obtained from https://github.com/riazn/bms038_analysis/. Differential expression between

198 baseline and on-treatment biopsies was performed with limma, using the duplicateCorrelation function for

199 repeat biopsy samples. Adjusted p values were calculated according to the method of Benjamini and

200 Hochberg. A score was calculated per sample from the mean of log10 transformed gene

201 expression values for CD8A, CD8B, GZMA, GZMB, PRF1 (28).

202 Single Cell mRNASeq Datasets

203 Four independent cohorts of single cell mRNASeq on immune infiltrates from human tumors were

204 analysed: metastatic melanoma studies by Sade-Feldman et al. (29) and Jerby-Arnon et al. (30), head and

205 neck carcinoma study by Puram et al. (31); carcinoma study by Zilionis et al. (32). See the associated

206 publications for further details and data availability. The Sade-Feldman dataset also contains clinical

207 information and outcome after treatment with checkpoint blockade. The R package Seurat (version 3.0)

208 was used to analyse these datasets (33). Raw data consisted of gene expression values in transcripts per

209 million (TPM). Log2(TPM/10 + 1) was used for downstream analysis (30). For each dataset, variable

210 genes were selected prior to dimensionality reduction with PCA. Significant principle components were

211 selected with Seurat’s JackStraw permutation testing. These components were supplied to the UMAP

212 algorithm (https://arxiv.org/abs/1802.03426) to generate a two-dimensional projection for visualisation

213 purposes. The head and neck data from Puram et al. displayed batch effects related to different sample

214 preparation methods. This effect was corrected using the data integration method of Seurat. No batch

10

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

215 effects were seen related to known experimental factors in the other studies. For each dataset, the cluster

216 assignments determined in the original publications were maintained. All single cell plots were generated

217 using log transformed TPM/10 values. Differential expression between groups of cells was performed

218 using MAST (34), and adjusted p values calculated with the Bonferroni correction. In the Sade-Feldman

219 et al. dataset, cases were classified in various ways regarding response, by response of the lesion which

220 was biopsied and the overall clinical response. The latter was used in classifying a case as a responder or

221 non-responder. Two cases were classified as treatment resistance, as they develop progressive disease after

222 an initial brief response. These cases were excluded to provide more distinct biology for comparison of

223 responders and non-responders. A gene signature was created from differential expression results by

224 selecting genes with an adjusted p value < 0.05 and log2 fold change > 2. The per cell signature score was

225 calculated by the sum of the expression of upregulated signature genes minus the expression of

226 downregulated signature genes, using scaled data in Seurat.

227 Single cell mRNASeq from the CT26 melanoma syngeneic model was also analysed in a similar fashion

228 to the above (35). As this data is unique molecular identifier based, the sctransform normalization and

229 variance stabilization method was used.

230 Data Availability

231 nanoString data generated in this study is included as a supplementary file.

232 METABRIC processed gene expression data is available at www.cbioportal.org, and clinical data is

233 available as supplementary table 1 in Rueda et al. (36).

234 TCGA melanoma gene expression data is available at www.cbioportal.org under the TCGA pan-cancer

235 atlas 2018 dataset, and curated survival data was obtained from Table S1 in Liu et al. (37).

11

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

236 IMvigor210 data can be found in the R package at http://research-

237 pub.gene.com/IMvigor210CoreBiologies/

238 Riaz et al. metastatic melanoma data is available at

239 https://github.com/riazn/bms038_analysis/tree/master/data.

240 Gide et al. metastatic melanoma data is available in the European Nucleotide Archive under accession

241 PRJEB23709.

242 Single cell mRNASeq data are available from the Gene Expression Omnibus; Sade-Feldman et al.

243 melanoma single cell data under accession GSE120575, Jerby-Arnon et al. melanoma single cell data

244 under accession GSE115978; Puram et al. head and neck data under GSE103322; Zilionis et al. lung

245 cancer data GSE127465; Kumar et al. CT26 syngeneic model data GSE121861

246 qReal Time PCR Analysis

247 RNA was isolated using an RNeasy mini kit (Qiagen) following which cDNA was generated using M-

248 MLV Reverse Transcriptase (Promega) as per the manufacturers’ instructions. qRT-PCRs were performed

249 using SensiFAST SYBR (Bioline) and performed on an StepOnePlus (Applied Biosystems). Primers for

250 as follows: CXCL9 Fwd 5’- ATCTTCCTGGAGCAGTGTGG-3’, Rev 5’-

251 AGTCCGGATCTAGGCAGGTT-3’: CXCL10 Fwd 5’-GTGAGAATGAGGGCCATAGG-3’, Rev 5’-

252 GGCTAAACGCTTTCATTAAATTC-3’: CXCR3 Fwd: 5’-GCCAAGCCATGTACCTTGAG-3’, Rev 5’-

253 TCAGGCTGAAATCCTGTGG-3’: GAPDH Fwd 5’- AACTTTGGCATTGTGGAAGG-3’, Rev 5’-

254 GGATGCAGGGATGATGTTCT-3’: RPL27 Fwd 5’- AAGCCGTCATCGTGAAGAACA-3’. Rev 5’-

255 CTTGATCTTGGATCGCTTGGC -3’.

256 Western Immunoblotting

12

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

257 Protein lysates were generated in 1% NP40 lysis buffer and boiled and resolved in Laemmli buffer in

258 reducing conditions using 10% acrylamide SDS-PAGE. Protein was then transferred to PVDF membrane

259 (Millipore) and probed with antibodies for CXCL9 (R & D Biosystems) and CXCL10 (R & D Biosystems)

260 and secondary antibodies conjugated with HRP. X-ray film was used to detect chemiluminescence.

261 CRISPR knockout cell line generation

262 To generate knockout cell lines, 37 pmoles sgRNA (Synthego) and 270 pmoles recombinant Cas9 (IDT)

263 were incubated together for 10 minutes at room temperature. sgRNA/Cas9 RNPs were then electroporated

264 into AT-3ova cells (3 x 105) using a Lonza SG Cell line kit and 4D-Nucleofector. Gene knockout was

265 confirmed 48 hours later by Western immunoblot analysis. Two sgRNAs were used per gene. Sequences

266 used were as follows: CXCL9 g1:auuuguaguggaucgugccu and g2: aaccugccuagauccggacu: CXCL10

267 g1:ugacgggccagugagaauga and g2: ugagcagagaugucugaauc.

268 Statistical analysis

269 Statistical differences were analyzed by one-way ANOVA, two-way ANOVA or unpaired t-test, where

270 appropriate, as stated in text. p<0.05 was considered significant.

271

272

13

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

273 Results

274 Intratumoral CXCL9 and CXCL10 expression is increased following immune checkpoint blockade

275 To investigate the chemokines required for effective therapeutic responses following immune checkpoint

276 blockade we utilized the AT-3ova model of triple negative breast cancer, which we have previously shown

277 to be responsive to dual PD-1/CTLA-4 blockade (19). As expected, when mice bearing AT-3ova tumors

278 were treated with anti-PD-1 and anti-CTLA-4 at day 14 post tumor inoculation, robust anti-tumor immune

279 responses were observed (Figure 1A, B), whilst single agent treatment with either anti-PD-1 or anti-

280 CTLA-4 resulted in limited anti-tumor activity (Supplementary Figure 1A). Since treatment of tumors

281 with anti-PD-1/anti-CTLA-4 is known to be associated with an increase in tumor-infiltrating lymphocytes

282 (19, 38), we investigated the chemokines that were upregulated in this context by performing gene

283 expression analysis on tumor samples obtained from mice treated with 2A3 isotype control or anti-PD-1

284 and anti-CTLA-4. This analysis revealed significant upregulation of numerous cytokines and chemokines

285 associated with T cell infiltration with CXCL9 (p = 0.001) and CXCL10 (p = 0.0001) being two of the

286 most significantly upregulated and abundant chemokines following dual immune checkpoint blockade

287 (Figure 1C). Similarly, CXCR3, the receptor for CXCL9/CXCL10, was also significantly upregulated in

288 tumors treated with anti-PD-1/anti-CTLA-4 (Supplementary Figure 1B). Analysis of tumor supernatants

289 confirmed that several chemokines including CCL5, CCL22, CCL17, CXCL9 and CXCL10 were

290 significantly upregulated at the protein level following dual blockade of PD-1 and CTLA-4 (Figure 1D)

291 whereas blockade of either PD-1 or CTLA-4 alone resulted in modest increases in chemokine production

292 (Supplementary Figure 1C).

293 We next analyzed the relevance of these chemokines in the context of melanoma patients treated with

294 either anti-PD-1 alone (Nivolumab or Pembrolizumab) or dual anti-PD-1/anti-CTLA-4 ()

14

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

295 therapy as previously described (23). Strikingly, the expression of CXCL9, CXCL10 and CXCL11 was

296 significantly higher in tumors isolated from responders at baseline (Figure 1E) and on therapy (Figure

297 1F). Furthermore high expression of CXCL9 and CXCL10, in particular, was associated with improved

298 patient survival and increased numbers of intratumoral CD8+ T cells in these patients treated with immune

299 checkpoint blockade (Figure 1G-H). Analysis of a separate cohort of melanoma patients treated

300 exclusively with Nivolumab single agent therapy revealed that CXCL9, CXCL10 and CCL5 were also

301 amongst the most highly upregulated genes in this cohort (Supplementary Figure 2A) (27). Moreover,

302 analysis of gene expression data from the IMvigor 210 trial indicated that the expression of CXCL9 (p=

303 0.0023), CXCL10 (p = 0.0074) and CXCL11 (P=0.029) were significant prognostic indicators in

304 metastatic urothelial carcinoma patients treated with atezolizumab, were expressed to a greater extent in

305 responders than non-responders and correlated with the extent of T cell infiltrate (Supplementary Figure

306 2B-D). In contrast, the expression of other chemokines CCL5, CCL2, CCL17 and CCL22 did not correlate

307 with survival (Supplementary Figure 2B), despite also correlating with immune cell infiltrate

308 (Supplementary Figure 2D). In contrast, using data from both the METABRIC triple negative breast

309 cancer (Supplementary Figure 3A; n= 279) and the TCGA melanoma cohorts (Supplementary Figure

310 3B; n = 369), we identified that, in patients treated with standard of care therapies, CXCL9, CXCL10,

311 CXCL11, CCL2, CCL17, CCL22 and CCL5 expression were all significantly associated with improved

312 patient survival (Supplementary Figure 3A, 3B) and was associated with a significant increase in the

313 cytotoxic T cell gene score within patient’s tumors, suggesting that the increased survival of patients with

314 high levels of these chemokines was likely an immune-dependent phenomenon (Supplementary Figure

315 3C). Taken together, this data indicates that responses to immune checkpoint blockade are associated with

316 the expression of the CXCR3 ligands CXCL9 and CXCL10, particularly patients treated with the

15

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

317 combination of anti-PD-1 and anti-CTLA-4. This led us to investigate the requirement for these

318 chemokines in the therapeutic effect evoked by immune checkpoint blockade.

319 CXCR3 neutralization abrogates the efficacy of immune checkpoint blockade and prevents the

320 infiltration of CD8+ T cells following therapy

321 To investigate the role of CXCL9 and CXCL10 in the therapeutic effect mediated by dual PD-1/CTLA-4

322 blockade, we first utilized a blocking for the CXCR3 receptor (Figure 2A).

323 Strikingly the blockade of CXCR3 significantly reduced the efficacy of anti-PD-1/anti-CTLA-4 treatment

324 leading to enhanced tumor growth (Figure 2B), reduced survival of mice (Figure 2C) and increased size

325 and weight of tumors when tumors were excised at day 7 post treatment (Figure 2D, E). To determine

326 whether this effect was broadly applicable to other cancer models we also investigated this in the context

327 of mice bearing MC38, E0771 or B16F10-ova tumors. In all cases, the anti-tumor effect mediated by dual

328 agent anti-PD-1 and anti-CTLA-4 therapy was reduced by co-administration of an anti-CXCR3 antibody

329 (Figure 2F-H). Given that CXCR3 ligands are also important in the context of patients treated with

330 atezolizumab as a single agent (Supplementary Figure 2), we further assessed the importance of CXCR3

331 in a model that was sensitive to PD-1 inhibition alone. To this end, we treated less established AT-3ova

332 tumors (Day 7 post inoculation) using anti-PD-1 alone, which also resulted in significant therapeutic

333 effects that were CXCR3 dependent (Supplementary Figure 2E). Taken together, these data demonstrate

334 the critical importance for the CXCR3/CXCR3 ligand axis for the therapeutic efficacy of anti-PD1/CTLA-

335 4 therapy, and next sought to investigate the underlying mechanism.

336 We have previously shown that dual PD-1/CTLA-4 blockade significantly enhances both CD8+ and CD4+

337 T cell effector functions in this model and that the therapeutic efficacy of this combination therapy is

338 dependent on both CD8+ and CD4+- cells (19). We therefore investigated the effect of CXCR3

16

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

339 blockade on the number and activation phenotype of these subsets in the context of dual PD-1/CTLA-4

340 blockade. In agreement with previous observations, including our own (19, 38-40), dual PD-1/CTLA-4

341 blockade led to a significant increase in the proportion and number of CD8+ T cells (Figure 3A;

342 Supplementary Figure 4A). Dual blockade therapy also significantly increased the frequency of

343 CD3+CD4+foxp3- T cells (Figure 3B) whilst the numbers of other populations including NK

344 Cells (Figure 3C) and CD4+foxp3+ T cells were unaffected (Figure 3D). Strikingly, blockade of CXCR3

345 significantly reduced the number of CD8+ T cells and CD4+foxp3- cells following dual PD-1/CTLA-4

346 blockade, as determined by both flow cytometry and immunofluorescent analysis (Figure 3A, B, E). By

347 contrast, the frequency of other immune cell populations including CD4+foxp3+ and NK1.1+ NK cells were

348 not significantly reduced by CXCR3 blockade (Figure 3C-D). Given that the expression of CXCR3

349 ligands has previously been linked to T cell priming (41) as well as playing a role in trafficking (13), we

350 investigated whether blockade of these receptors may also potentially impact the function of T cells within

351 the tumor microenvironment in addition to their recruitment to the tumor. As expected, we observed that

352 the combination of PD-1 and CTLA-4 significantly enhanced the production of both IFNγ and TNFα

353 intratumorally in both the CD8+ and CD4+foxp3- subsets (Figure 3F), consistent with our previous

354 observations (19). Notably CXCR3 blockade significantly reduced the production of IFNγ and TNFα of

355 CD8+ and CD4+foxp3- T cells, but not NK cells, in the setting of dual PD-1/CTLA-4 blockade (Figure 3F,

356 Supplementary Figure 4B), suggesting that intratumoral expression of CXCR3 ligands enhanced their

357 activation. However, CXCR3 blockade did not affect the expression of other markers associated with T

358 cell activation on either CD8+ or CD4+foxp3- cells including Ki67 (proliferation), PD-1/TIM-3 or tbet

359 (Figure 3G, Supplementary Figure 4C). We next sought to identify whether the importance of CXCR3

360 for the therapeutic effects of dual PD-1/CTLA-4 blockade stemmed from enhanced CXCR3-dependent

361 recruitment of T cells to the tumor site and/or local effects of CXCL9/10 on T cell function. To address

17

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

362 this we repeated our therapeutic studies in the context of S1PR blockade, using FTY720. Treatment of

363 mice with FTY720 significantly reduced the therapeutic efficacy of dual PD-1 and CTLA-4 blockade, to

364 a similar extent as anti-CXCR3 (Figure 3H). Moreover, blockade of CXCR3 did not affect the capacity

365 of OT-I T cells to produce cytokines when cocultured with tumor cells in vitro (Supplementary Figure

366 4D). Taken together, this data suggests that CXCR3-mediated enhancement of T cell cytokine production

367 in vivo is associated with enhanced migration of T cells to the tumor site which creates a more favorable

368 environment for T cell activation.

369 CXCR3 blockade affects the priming of CD8+ T cells within the tumor draining lymph node

370 Given that we have previously shown that the therapeutic effects of anti-PD-1/anti-CTLA-4 are dependent

371 on both CD4+foxp3- and CD8+ T cells and are associated with activation of these cells in the tumor draining

372 lymph nodes (DLNs) (19), we further investigated the phenotype of these cells in the DLNs in the context

373 of CXCR3 blockade. To investigate a potential role for CXCL9/CXCL10: CXCR3 interactions we first

374 evaluated the expression of these genes in cells isolated from DLNs of tumor bearing mice following PD-

375 1/CTLA-4 blockade. This analysis revealed that CXCL9 and CXCR3 were significantly upregulated

376 within tumor DLNs (Figure 4A-B). We therefore investigated the effect of CXCR3 blockade on T cell

377 activation within tumor DLNs. In line with our previous observations, dual blockade of PD-1 and CTLA-

378 4 led to the activation of both CD8+ and CD4+foxp3- T cells within DLNs in terms of IFNγ production,

379 proliferation and expression of tbet and PD-1 (Figure 4C-F). Strikingly, and similarly to our observations

380 within the tumor microenvironment, blockade of CXCR3 significantly reduced the production of IFNγ by

381 CD8+ T cells isolated from tumor DLNs (Figure 4C), whilst other parameters such as the expression of

382 Ki67, tbet and PD-1 were unaffected (Figure 4D-F). Interestingly, whilst CXCR3 blockade did not

383 modulate the expression of IFNγ (Figure 4C) or tbet (Figure 4E) by CD4+foxp3- cells in the DLNs, it did

384 increase their expression of Ki67 (Figure 4D) and PD-1 (Figure 4F). Taken together, this data indicates

18

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

385 that dual PD-1/CTLA-4 blockade not only induces CXCR3-dependent migration of CD8+ T cells into the

386 tumor site but also that CXCR3 ligands modulate the activation of both CD8+ and CD4+foxp3- cells both

387 at the tumor site and within draining lymph nodes.

388 Intratumoral CXCL9 is predominantly secreted from macrophages following immune checkpoint

389 blockade

390 We next assessed the mechanism and cell types that were responsible for the generation of CXCR3 ligands

391 following immune checkpoint blockade. One possibility was that the production of CXCL9 and CXCL10

392 was mediated by tumor cells, since AT-3ova cells produced CXCL9 and CXCL10 following stimulation

393 with IFNγ and TNFα in vitro (Supplementary Figure 5A-B). To assess the importance of tumor-derived

394 CXCL9/CXCL10 for the therapeutic effect of dual PD-1/CTLA-4 blockade, we generated AT-3ova cells

395 that did not express CXCL9 or CXCL10 using CRISPR-Cas9 mediated deletion of these genes. The

396 resulting cells produced negligible amounts of CXCL9/10 in vitro following dual TNFα and IFNγ

397 stimulation (Supplementary Figure 5C). However, loss of CXCL9 and CXCL10 production by AT-3ova

398 tumor cells did not significantly affect their responsiveness to dual PD-1/CTLA-4 blockade (Figure 5A)

399 nor the level of CXCL9 within the tumor microenvironment following therapy (Figure 5B). This data

400 suggested that host cells were the major source of CXCL9 in the context of combination therapy which

401 was critical for the therapeutic effect. Accordingly, we assessed the cell types responsible for the

402 production of CXCL9 and CXCL10 in tumors of mice undergoing immune checkpoint therapy. Following

403 dual PD-1/CTLA-4 blockade, on a per cell basis CXCL9 was predominantly expressed in a

404 /macrophage (Mo/Mac) population, defined as CD11b+Ly6CintCD11c+F4/80+ (Figure 5C-D;

405 Supplementary Figure 6A). Moreover, dual anti-PD-1/anti-CTLA-4 treatment evoked a strong

406 infiltration and/or differentiation of macrophages as shown by an increased proportion of these cells

407 (Supplementary Figure 6B-C) and a significant upregulation of a macrophage gene signature in treated

19

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

408 tumors (Figure 5E). Moreover, these cells expressed high levels of CD64 and MHCII, phenotypic markers

409 associated with /macrophages thus confirming their identity as macrophages (Supplementary

410 Figure 6D). Therefore, although CXCL9 production was increased in CD103+ cDC1s

411 (CD11c+CD103+CD11b-) and tumor/stromal cells (CD45-) following dual PD-1/CTLA-4 blockade

412 (Figure 5C-D), given their increased frequency relative to CD103+ DCs (Supplementary Figure 6C, E),

413 Mo/Macs were the predominant source of CXCL9. Indeed, Mo/Macs accounted for 38.1% of CXCL9

414 positive cells following dual PD-1/CTLA-4 blockade with conventional CD11b+CD11c+MHCII+ DCs

415 (27.5%) and CD45.2- cells (stroma/tumor; 14.7%) making up the other major CXCL9+ populations

416 (Figure 5F; Supplementary Figure 6F). These experiments also revealed that B cells were not a major

417 source of CXCL9 (Supplementary Figure 6G). Macrophages were also a major source of CXCL9 in

418 MC38 tumors (Figure 5G) and analysis of an independent, previously generated single cell RNA-seq

419 dataset of tumor-infiltrating immune cells in the CT26 tumor model (35) revealed that CXCL9 and

420 CXCL10 were predominantly expressed by immune cells that also co-expressed the macrophage markers

421 F4/80 and CD64 (Figure 5H), confirming this observation was applicable to multiple tumor models. To

422 analyse CXCL10 in the context of dual PD-1/CTLA-4 blockade in the absence of an antibody suitable for

423 flow cytometry, we also FACS sorted the relevant immune populations and analysed CXCL9/10 at the

424 transcriptional level. This analysis confirmed that Mo/Mac cells produced significantly more CXCL9 in

425 the context of dual checkpoint blockade and that these cells also produced large amounts of CXCL10

426 mRNA following therapy (Supplementary Figure 6A).

427 Production of CXCL9 and CXCL10 is IFN dependent

428 Given the known key role of IFNγ and TNFα in modulating CXCL9/10 expression in alternative contexts

429 (13, 42), we next assessed their importance in driving chemokine expression in vivo in the context of

430 immune checkpoint blockade. We found that IFNγ and TNFα levels were significantly enhanced in the

20

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

431 context of dual checkpoint blockade (in comparison to single agent PD-1 or CTLA-4 blockade) and that

432 both CXCL9 and CXCL10 expression was highly correlated with IFNγ and TNFα production (Figure

433 6A). We next sought to confirm these observations in the IMvigor210 trial cohort where CXCL9 and

434 CXCL10 expression was associated with improved responses to Atezolizumab (anti-PDL1)

435 (Supplementary Figure 2B). Both CXCL9 and CXCL10 high tumors were significantly enriched for both

436 IFNγ and TNFα gene signatures (Figure 6B). To formally test the requirement of IFNγ and TNFα for

437 CXCL9 production following dual checkpoint blockade, we analyzed the production of CXCL9 by

438 Mo/Macs and CD11b+CD103+DCs in the context of either IFNγ or TNFα neutralization. These results

439 revealed that IFNγ was critical for CXCL9 production whilst no significant difference was observed

440 following TNFα neutralization (Figure 6C). To confirm this directly, we stimulated tumor infiltrating

441 leukocytes ex vivo with IFN and/or TNFα which revealed that macrophages, DCs and CD45- cells

442 upregulated CXCL9 ex vivo in response to IFNγ stimulation alone (Figure 6D). Consistent with our

443 previous data, macrophages isolated from tumors in mice treated with anti-PD-1 and anti-CTLA-4 secreted

444 high amounts of CXCL9 which could not be further enhanced by IFN, suggesting that this IFN pathway

445 was already activated in vivo. Notably, these effects were also observed in BATF3-/- mice, confirming that

446 these effects were independent of CD103+ DCs (Figure 6E). However, we did observe that the overall

447 levels of CXCL9 production by macrophages were lower in BATF3-/- mice, suggesting that CD103+ DCs

448 indirectly contributed to the production of CXCL9 by these cells. Taken together, these data demonstrated

449 that Mo/Mac cells are the major source of tumor derived CXCL9 and CXCL10 after dual PD-1/CTLA-4

450 blockade and that this expression is driven by IFNγ that is induced following combination therapy.

451

452 Macrophages are essential for dual PD-1/CTLA4 therapeutic efficacy

21

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

453 To investigate the requirement for macrophages for CXCL9/10 production following dual PD-1/CTLA-4

454 blockade, we depleted these cells using an anti-F4/80 antibody (Supplementary Figure 6H) which

455 resulted in a complete abrogation of the therapeutic efficacy of dual PD-1/CTLA-4 blockade (Figure 7A).

456 Depletion of macrophages also attenuated the increased production of CXCL9 (Figure 7B) and infiltration

457 of CD8+ and CD4+foxp3- cells within tumors following dual PD-1/CTLA-4 blockade (Figure 7C).

458 Similarly, IFNγ neutralization also attenuated increased infiltration of CD8+ and CD4+foxp3- cells,

459 highlighting the link between IFNγ, macrophages and CXCL9 production. To investigate the clinical

460 significance of macrophage derived CXCR3 ligands (CXCL9/CXCL10/CXCL11) we next sought to

461 determine the source of these chemokines in human tumors and their prognostic significance.

462 Analysis of single cell RNA-seq datasets in melanoma (29), lung carcinoma (32) and head and neck cancer

463 cohorts (31) revealed that macrophages were the predominant cell type responsible for the production of

464 CXCL9, CXCL10 and CXCL11 (Figure 7D-E; Supplementary Figure 7A-B). Of note, within the lung

465 carcinomas studied by Zilonis et al. (32), multiple and macrophage (tMac1-9) sub-

466 populations were defined but CXCL9-11 were more highly expressed by CD68 expressing macrophages

467 than dendritic cells, including the CLEC9A+ cDC1 population (Figure 7E). This was consistent with all

468 human datasets analyzed where CXCL9-11 production was also detected in other immune cell populations

469 such as dendritic cells, but consistently to a lesser extent than macrophages (Figure 7D, Supplementary

470 Figure 7A-B). Utilizing the melanoma dataset of Sade-Feldman et al., derived from single cell RNA-seq

471 analysis before and after immune checkpoint blockade, we were able to investigate the expression of these

472 chemokines in the context of immunotherapy (29). Differential gene expression analysis of macrophages

473 in responders and non-responders revealed that CXCR3 ligands, and in particular CXCL10 and CXCL11,

474 were more abundantly expressed in macrophages from responding patients (Figure 7F-G). Further

475 analysis of the macrophage transcriptome identified a unique macrophage gene signature that included

22

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

476 CXCL10 and CXCL11 which was more abundant in macrophages derived from patients that responded to

477 immune checkpoint blockade (Figure 7H, Supplementary Figure 7C). Taken together, the data obtained

478 from these patient samples strongly supports our preclinical observation that macrophages are a major

479 source of CXCR3 ligands and are associated with responses to immune checkpoint blockade.

480

23

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

481 Discussion

482 Understanding the processes which govern responsiveness to immune checkpoint therapy as well as the

483 transition from non-T cell infiltrated to inflamed tumors are two major frontiers in the field of cancer

484 immunotherapy. A number of factors can contribute to these parameters, including the local tumor

485 microenvironment, stromal stiffness, the production of factors which promote an immune exclusion

486 phenotype (24, 43, 44) and an ineffective or inappropriate production of chemokines which either promote

487 the recruitment of immunosuppressive subtypes (45, 46) or a failure to mount effective recruitment of anti-

488 tumor T cells. In this study, we investigated chemokine production within tumors following effective

489 therapy with immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4). This analysis showed that

490 CXCL9 production was significantly induced in an IFNγ dependent manner following therapy both at the

491 transcriptional and protein level and that the production of CXCR3 ligands was critical for therapeutic

492 efficacy. The requirement for the CXCL9:CXCR3 axis for the therapeutic efficacy of these

493 immunotherapeutic treatments is consistent with previous data indicating that CXCL9/CXCL10: CXCR3

494 interactions govern immune cell infiltration into tumors prior to therapy (47) and following chemotherapy

495 (10, 48, 49), IL-2 treatment (50, 51), adoptive cellular therapy (12, 52, 53), bispecific antibodies (54) and

496 that CXCR3-/- mice fail to elicit effective anti-tumor immune responses post-vaccination and anti-PD-1

497 (55). These studies have shown that CXCL9 and CXCL10 can modulate anti-tumor immune responses

498 evoked following several therapeutic approaches. Although our murine studies did not address the role of

499 CXCL11 (as C57BL/6 mice do not express CXCL11), the data obtained from cancer patients suggests that

500 this chemokine is worthy of further attention in future studies. Notably, our data indicates that CXCL11

501 expression correlates with improved responses to atezolizumab and was the most significantly upregulated

502 gene in macrophages isolated from patients responding to immune checkpoint blockade. Therefore, further

503 studies investigating the role of CXCL11 in human cancers, and in particular its function relative to

24

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

504 CXCL9 and CXCL10 would be of high relevance. Although our study focuses on the role of CXCR3

505 ligands in the response to immune checkpoint blockade, it does not preclude a role for other important

506 chemokines such as CCL5, the expression of which was shown to correlate with enhanced anti-tumor

507 immune responses and improved patient survival, particularly in the context of high CXCL9 expression

508 (56). However, whilst CCL5 can potentially enhance anti-tumor through the recruitment of

509 CCR5+ T cells and NK cells, pro-tumoral roles of CCL5 have also been reported (57, 58). Therefore

510 strategies to enhance CXCL9/10/11 production may be more favourable to those designed to enhance

511 CCL5 expression.

512 Our data indicating the importance of CXCL9/10/11: CXCR3 interactions for the efficacy of immune

513 checkpoint blockade in vivo was supported by transcriptome analysis of patients treated with either

514 atezolizumab (anti-PD-L1) or nivolumab (anti-PD-1), or a combination of ipilimumab and a PD-1

515 blocking antibody. In all cases, CXCL9/10/11 production was highly correlated with improved patient

516 survival and their production was associated with an IFNγ gene signature. Given that single cell RNA-seq

517 analysis revealed that CXCR3 is highly expressed on CD8+ and CD4+ tumor-infiltrating lymphocytes in

518 triple negative breast cancer (59), it is highly likely that local production of CXCL9/10 can modulate T

519 cell recruitment and activation in human cancers. Interestingly, a similar observation has been reported in

520 a study of 45 melanoma patients treated with ipilimumab where the increased expression of IFNγ

521 dependent genes including CXCL9-11 was observed in patients undergoing a clinical response (60). Our

522 analysis supports that these chemokines are also increased following anti-PD-1, that they are crucial for

523 therapeutic activity and correlate with patient prognosis. Therefore strategies to enhance the expression

524 and/or activity of CXCL9/10 may be expected to enhance the therapeutic efficacy of immune checkpoint

525 blockade. Indeed, the use of a Dipeptidylpeptidase 4 (DPP4, CD26) inhibitor to preserve CXCL10

526 bioactivity was shown to enhance the efficacy of adoptive cellular therapy and immune checkpoint

25

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

527 blockade (61, 62). Similarly, the localized application of a COX-2 inhibitor was shown to enhance

528 therapeutic responses to anti-PD-1, associated with an increase in expression of CXCL9 and CXCL10 (63)

529 and epigenetic modulators (EZH2i and DNMT1i) were shown to enhance anti-tumor immune responses

530 associated with increased expression of CXCL9 and CXCL10 (64). However, given the broad non-specific

531 nature of these inhibitors, it is clear that more specific therapeutics to modulate the CXCL9/CXCL10

532 chemokines would be of significant interest.

533 Having established that CXCR3 was crucial to the anti-tumor efficacy of dual PD-1/CTLA-4 blockade we

534 next investigated the underlying mechanisms. We observed that CXCR3 blockade reduced CD8+ T cell

535 infiltrate into tumors following dual PD-1/CTLA-4 blockade whilst the numbers of other immune cell

536 populations such as CD4+ Tregs and NK cells were not affected. This was somewhat surprising given that

537 these cells express CXCR3 and expression of CXCL10 was shown to promote NK cell infiltration into the

538 NK sensitive RMA-S cell line (65). However, this data may either reflect a higher dependency of CD8+ T

539 cells on CXCR3 for their migration into the tumor site following immune checkpoint blockade than other

540 immune cell subtypes and/or our use of a T cell sensitive model where NK cells are unlikely to play a

541 major role in the control of tumor growth. This hypothesis is consistent with the fact that CXCR3

542 expression was increased following T cell activation by dual PD-1/CTLA-4 blockade. We did not

543 investigate the role of CXCL9 and CXCL10 in recruiting other immune cell types, including myeloid cells.

544 Intriguingly, since monocytes themselves express CXCR3 it is possible that CXCL9-10 production by

545 monocytes/macrophages leads to the recruitment of more monocytes in a positive feedback loop.

546 Previous studies have shown that CXCL9 and CXCL10 can be secreted by multiple cell types including

547 monocytes, endothelial cells, fibroblasts, inflammatory macrophages dendritic cells (particularly the cDC1

548 subtype) and tumor cells themselves (9, 12, 13, 66-68). In the context of cancer, previous data have

549 suggested that tumor cells (68), CD103+ DCs (9, 12) (69) or CD11b+ cells (67) are major sources of

26

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

550 CXCL9/10 within the tumor microenvironment. Notably, whilst we did observe that dual PD-1/CTLA-4

551 blockade enhanced CXCL9/10 production by CD103+ DCs and the bulk CD11b+ population, our data

552 indicates that macrophages are the predominant source of CXCL9 and CXCL10 in the context of immune

553 checkpoint blockade both in mice and in patients. This was supported by single-cell RNA-seq in patient

554 datasets and alternative murine tumor models suggesting this is a pathway conserved in multiple tumor

555 types.

556 Our analysis of single cell RNA-seq data indicated that macrophages secreted higher levels of CXCL9-11

557 than dendritic cells in multiple cancer cell types. This was also observed in the context of immunotherapy

558 where the presence of CXCL10 or CXCL11 positive macrophages was a marker for benefit from

559 immunotherapy. Although CXCL9 was not prognostic within this patient cohort, we cannot exclude the

560 possibility that the low CXCL9 counts arose through technical limitation rather than a reduced expression

561 of CXCL9 relative to the other CXCR3 ligands CXCL10 and CXCL11. Our data suggest that the

562 production of CXCL9 and CXCL10 by the tumor cells themselves are dispensable for the therapeutic

563 effect of anti-PD-1 and anti-CTLA-4 and that knockout of these genes from tumor cells had minimal

564 impact on the overall concentration of these chemokines, particularly CXCL9, in the context of dual

565 combination therapy. However we cannot exclude a role for tumor-derived CXCL9/CXCL10 in tumors

566 with low immune infiltrate where their overall contribution to CXCL9/CXCL10 levels maybe more

567 pronounced. Interestingly our data also suggests a greater increase in CXCL10 mRNA, relative to CXCL9

568 mRNA, in CD45- cells following combination therapy. Although this population includes tumor cells and

569 other host cells such as stroma, it would be interesting to confirm this at the protein level in future

570 experiments, particularly since we observed a significant decrease in CXCL10 protein levels within the

571 tumor microenvironment following CRISPR-Cas9 mediated deletion of CXCL10 from tumor cells.

27

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

572 Moreover, our data does not discount the importance of CXCL9/10 from other cell types including CD103+

573 DCs which has previously been reported in the context of adoptive cellular therapy and immune checkpoint

574 blockade (12, 69). The production of CXCL9/10 by these cells can enhance T cell priming by facilitating

575 DC: T cell interactions and result in enhanced anti-tumor immunity. However, our data indicates a clear

576 role for CXCR3 dependent migration of T cells into tumors and that the predominant cellular source of

577 CXCL9/10 is macrophages. We believe one explanation for this apparent discrepancy is that whilst

578 macrophages are critical for CXCR3-dependent recruitment of T cells through their production of

579 CXCL9/10 in the tumor, CXCL9/10 production by other myeloid subsets, including CD103+ DCs is likely

580 to be important for T cell priming both within the tumor and in the draining lymph node. Therefore, whilst

581 our studies do not preclude an important role for CXCL9/10 production by DCs in promoting T cell

582 activation, they clearly indicate that macrophages are responsible for a significant proportion of these

583 chemokines. Notably, the requirement for immune cell trafficking into tumors following immune

584 checkpoint blockade is consistent with the emergence of new antigen specific immune cells at the tumor

585 site following therapy (70).

586 Although it was recently described that anti-TIM-3 therapy enhances CXCL9/10 production by CD103+

587 DCs through direct modulation of TIM-3 expressed on CD103+ DCs (9), such a direct mechanism is

588 unlikely to contribute to the efficacy of PD-1 and CTLA-4 blockade, as PD-1 and CTLA-4 are not

589 expressed on either DCs or the tumoral macrophages in our study. Instead our data suggest that T cell

590 mediated increases in IFNγ and TNFα production are critical for this effect. Indeed, we observed that the

591 production of CXCL9 was dependent on IFNγ and TNFα, which is consistent with the role of STAT1 and

592 NFκB in enhancing the transcriptional activity of CXCL9 (71, 72) and with a previous report that indicated

593 that anti-PD-1 enhanced CXCL10 production from intratumoral CD11b+ cells within tumors in an IFNγ

594 dependent manner in the context of adoptive cellular therapy (67). Transcriptome analysis of patient’s

28

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

595 tumors post immune checkpoint blockade indicated a robust correlation between the expression of IFNγ-

596 related genes and CXCL9/CXCL10, suggesting that this mechanism is also important in patients. Given

597 that mutations in both the JAK-STAT (73-75) pathway and in TNF signaling (76) have previously been

598 reported as resistance mechanisms for immune checkpoint inhibitors, our data suggests that a failure to

599 secrete CXCL9/CXCL10 by these tumors may represent an additional mechanism of immune escape.

600 Interestingly, the role of the CXCR3 receptor in the response to dual PD-1/CTLA-4 blockade was not

601 limited to CD8+ T cell trafficking to the tumor site since CXCR3 blockade also modulated T cell

602 functionality in the DLNs. CXCR3 mRNA expression was increased on both CD4+ and CD8+ subsets

603 following dual PD-1/CTLA-4 blockade and, although we did not observe increased expression of CXCR3

604 protein on CD8+ T cells following dual PD-1/CTLA-4 blockade, this is potentially explained by CXCR3

605 internalization which has previously been reported (77). Blockade of CXCR3 reduced the production of

606 IFNγ by CD8+ T cells in DLNs and affected CD4+foxp3- T cell differentiation as shown by enhanced

607 expression of PD-1 and the proliferation marker Ki67. The mechanism underlying this is yet to be fully

608 determined but a role for CXCR3 ligands in promoting DC: T cell interactions within interfollicular

609 regions of lymph nodes following vaccination has previously been described (41, 78). Thus, it is tempting

610 to speculate that CXCL9/CXCL10: CXCR3 interactions may play a similar role in tumor DLNs by

611 promoting the aggregation of tumor specific T cells and antigen presenting DCs. Interestingly in these

612 studies showing a role for CXCR3 ligands in lymph node priming, the production of CXCL10 was

613 predominantly by the CD11b+ lymph node resident dendritic cells whilst CXCL9 was predominantly

614 expressed by stromal cells. Whilst we did not observe significant expression of CXCL10 by CD11b+ cells

615 in our analysis we cannot exclude that the expression of CXCL10 was restricted to an alternative immune

616 subset and/or expressed at a different timepoint than analyzed in our experiments. Nevertheless, our data

617 suggest a role for CXCL9/CXCR3 interactions within DLNs in the context of immunotherapy and this

29

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

618 could potentially play a role in the spatial organisation and consequent activation of both CD8+ and CD4+

619 T cells within the DLNs, as has been reported in the context of vaccination (78, 79) (41). Such interactions

620 have not been studied within the tumor microenvironment but this would be of significant interest given

621 our data suggest that the expression of CXCL9 by host immune cells is critical for the therapeutic effect

622 of immune checkpoint blockade.

623 In summary, our data indicate that the production of CXCR3 ligands is critical for the therapeutic effect

624 of dual anti-PD-1/anti-CTLA-4 therapy and identifies macrophages as the predominant source of CXCL9

625 both in preclinical models and in patients treated with immunotherapy. These cells were predicative of

626 response to immune checkpoint blockade and thus novel strategies to enhance CXCL9/10 production,

627 particularly by macrophages, have the potential to enhance the efficacy of immunotherapy in patients.

628 Author Contributions

629 PAB, IGH and PKD designed the experiments and wrote the manuscript. IGH, PS, JL, PAB and PKD

630 analyzed the data. IGH, PS, JL, AC, AO, ZLT, KLT, MAH, LG, EVP, KS, SM and PAB performed the

631 experiments. SL provided intellectual input and read the manuscript.

632 Acknowledgments

633 The authors would like to acknowledge the assistance of the Animal Facility technicians at the Peter

634 MacCallum Cancer Centre.

635 References

636 1. A. Ribas, J. D. Wolchok, Cancer immunotherapy using checkpoint blockade. Science 359, 1350-1355 (2018). 637 2. P. Sharma, J. P. Allison, Immune checkpoint targeting in cancer therapy: toward combination strategies 638 with curative potential. Cell 161, 205-214 (2015). 639 3. N. A. Rizvi et al., Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in 640 non-small cell lung cancer. Science 348, 124-128 (2015).

30

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

641 4. P. C. Tumeh et al., PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 642 568-571 (2014). 643 5. T. F. Gajewski et al., Cancer Immunotherapy Targets Based on Understanding the T Cell-Inflamed Versus 644 Non-T Cell-Inflamed Tumor Microenvironment. Adv Exp Med Biol 1036, 19-31 (2017). 645 6. C. Denkert et al., Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or 646 without carboplatin in human epidermal receptor 2-positive and triple-negative primary 647 breast cancers. J Clin Oncol 33, 983-991 (2015). 648 7. Q. Ding et al., CXCL9: evidence and contradictions for its role in tumor progression. Cancer Med 5, 3246- 649 3259 (2016). 650 8. T. J. Zumwalt, M. Arnold, A. Goel, C. R. Boland, Active secretion of CXCL10 and CCL5 from colorectal cancer 651 microenvironments associates with GranzymeB+ CD8+ T-cell infiltration. Oncotarget 6, 2981-2991 (2015). 652 9. A. de Mingo Pulido et al., TIM-3 Regulates CD103(+) Dendritic Cell Function and Response to 653 Chemotherapy in Breast Cancer. Cancer Cell 33, 60-74 e66 (2018). 654 10. M. Hong et al., Chemotherapy induces intratumoral expression of chemokines in cutaneous melanoma, 655 favoring T-cell infiltration and tumor control. Cancer Res 71, 6997-7009 (2011). 656 11. K. Specht et al., Expression profiling identifies genes that predict recurrence of breast cancer after adjuvant 657 CMF-based chemotherapy. Breast Cancer Res Treat 118, 45-56 (2009). 658 12. S. Spranger, D. Dai, B. Horton, T. F. Gajewski, Tumor-Residing Batf3 Dendritic Cells Are Required for 659 Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 31, 711-723 e714 (2017). 660 13. R. Tokunaga et al., CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation - A target for novel cancer 661 therapy. Cancer Treat Rev 63, 40-47 (2018). 662 14. D. Bedognetti et al., CXCR3/CCR5 pathways in metastatic melanoma patients treated with adoptive 663 therapy and -2. Br J Cancer 109, 2412-2423 (2013). 664 15. Y. Weng et al., Binding and functional properties of recombinant and endogenous CXCR3 chemokine 665 receptors. J Biol Chem 273, 18288-18291 (1998). 666 16. K. E. Cole et al., Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine 667 with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med 187, 668 2009-2021 (1998). 669 17. R. A. Colvin, G. S. Campanella, J. Sun, A. D. Luster, Intracellular domains of CXCR3 that mediate CXCL9, 670 CXCL10, and CXCL11 function. J Biol Chem 279, 30219-30227 (2004). 671 18. M. Metzemaekers, V. Vanheule, R. Janssens, S. Struyf, P. Proost, Overview of the Mechanisms that May 672 Contribute to the Non-Redundant Activities of Interferon-Inducible CXC 3 Ligands. 673 Front Immunol 8, 1970 (2017). 674 19. P. A. Beavis et al., Dual PD-1 and CTLA-4 Checkpoint Blockade Promotes Antitumor Immune Responses 675 through CD4(+)Foxp3(-) Cell-Mediated Modulation of CD103(+) Dendritic Cells. Cancer Immunol Res 6, 676 1069-1081 (2018). 677 20. M. E. Ritchie et al., limma powers differential expression analyses for RNA-sequencing and microarray 678 studies. Nucleic Acids Res 43, e47 (2015). 679 21. M. D. Robinson, A. Oshlack, A scaling normalization method for differential expression analysis of RNA-seq 680 data. Genome Biol 11, R25 (2010). 681 22. C. W. Law, Y. Chen, W. Shi, G. K. Smyth, voom: Precision weights unlock linear model analysis tools for 682 RNA-seq read counts. Genome Biol 15, R29 (2014). 683 23. T. N. Gide et al., Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti- 684 PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 35, 238-255 e236 (2019). 685 24. S. Mariathasan et al., TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion 686 of T cells. Nature 554, 544-548 (2018). 687 25. D. Wu, G. K. Smyth, Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic 688 Acids Res 40, e133 (2012).

31

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

689 26. A. Subramanian et al., Gene set enrichment analysis: a knowledge-based approach for interpreting 690 genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545-15550 (2005). 691 27. N. Riaz et al., Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 171, 692 934-949 e916 (2017). 693 28. P. Jiang et al., Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat 694 Med 24, 1550-1558 (2018). 695 29. M. Sade-Feldman et al., Defining T Cell States Associated with Response to Checkpoint Immunotherapy in 696 Melanoma. Cell 175, 998-1013 e1020 (2018). 697 30. L. Jerby-Arnon et al., A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint 698 Blockade. Cell 175, 984-997 e924 (2018). 699 31. S. V. Puram et al., Single-Cell Transcriptomic Analysis of Primary and Metastatic Tumor Ecosystems in Head 700 and Neck Cancer. Cell 171, 1611-1624 e1624 (2017). 701 32. R. Zilionis et al., Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid 702 Populations across Individuals and Species. Immunity 50, 1317-1334 e1310 (2019). 703 33. A. Butler, P. Hoffman, P. Smibert, E. Papalexi, R. Satija, Integrating single-cell transcriptomic data across 704 different conditions, technologies, and species. Nat Biotechnol 36, 411-420 (2018). 705 34. G. Finak et al., MAST: a flexible statistical framework for assessing transcriptional changes and 706 characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol 16, 278 (2015). 707 35. M. P. Kumar et al., Analysis of Single-Cell RNA-Seq Identifies Cell-Cell Communication Associated with 708 Tumor Characteristics. Cell Rep 25, 1458-1468 e1454 (2018). 709 36. O. M. Rueda et al., Dynamics of breast-cancer relapse reveal late-recurring ER-positive genomic subgroups. 710 Nature 567, 399-404 (2019). 711 37. J. Liu et al., An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome 712 Analytics. Cell 173, 400-416 e411 (2018). 713 38. M. A. Curran, W. Montalvo, H. Yagita, J. P. Allison, PD-1 and CTLA-4 combination blockade expands 714 infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad 715 Sci U S A 107, 4275-4280 (2010). 716 39. C. Boutros et al., Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev 717 Clin Oncol, (2016). 718 40. C. Twyman-Saint Victor et al., Radiation and dual checkpoint blockade activate non-redundant immune 719 mechanisms in cancer. Nature 520, 373-377 (2015). 720 41. J. R. Groom et al., CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T 721 helper 1 cell differentiation. Immunity 37, 1091-1103 (2012). 722 42. P. Guirnalda, L. Wood, R. Goenka, J. Crespo, Y. Paterson, -induced intratumoral 723 expression of CXCL9 alters the local distribution of T cells following immunotherapy with Listeria 724 monocytogenes. Oncoimmunology 2, e25752 (2013). 725 43. M. Binnewies et al., Understanding the tumor immune microenvironment (TIME) for effective therapy. 726 Nat Med 24, 541-550 (2018). 727 44. L. L. van der Woude, M. A. J. Gorris, A. Halilovic, C. G. Figdor, I. J. M. de Vries, Migrating into the Tumor: a 728 Roadmap for T Cells. Trends Cancer 3, 797-808 (2017). 729 45. T. M. Nywening et al., Targeting both tumour-associated CXCR2(+) and CCR2(+) macrophages 730 disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal 731 adenocarcinoma. Gut 67, 1112-1123 (2018). 732 46. J. Li et al., Tumor Cell-Intrinsic Factors Underlie Heterogeneity of Immune Cell Infiltration and Response to 733 Immunotherapy. Immunity 49, 178-193 e177 (2018). 734 47. A. V. Gorbachev et al., CXC chemokine ligand 9/ induced by IFN-gamma production by tumor 735 cells is critical for T cell-mediated suppression of cutaneous tumors. J Immunol 178, 2278-2286 (2007).

32

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

736 48. R. Zhang et al., Combination of MIG (CXCL9) chemokine gene therapy with low-dose cisplatin improves 737 therapeutic efficacy against murine carcinoma. Gene Ther 13, 1263-1271 (2006). 738 49. L. Hannesdottir et al., Lapatinib and doxorubicin enhance the Stat1-dependent antitumor immune 739 response. Eur J Immunol 43, 2718-2729 (2013). 740 50. J. Pan et al., CXCR3/CXCR3 ligand biological axis impairs RENCA tumor growth by a mechanism of 741 immunoangiostasis. J Immunol 176, 1456-1464 (2006). 742 51. K. L. Reckamp et al., Expression of CXCR3 on mononuclear cells and CXCR3 ligands in patients with 743 metastatic renal cell carcinoma in response to systemic IL-2 therapy. J Immunother 30, 417-424 (2007). 744 52. X. Wang et al., Human IP10-scFv and DC-induced CTL synergistically inhibit the growth of glioma in a 745 xenograft model. Tumour Biol 35, 7781-7791 (2014). 746 53. M. E. Mikucki et al., Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking 747 across tumour vascular checkpoints. Nat Commun 6, 7458 (2015). 748 54. J. Li et al., IFNgamma-induced Chemokines Are Required for CXCR3-mediated T-Cell Recruitment and 749 Antitumor Efficacy of Anti-HER2/CD3 Bispecific Antibody. Clin Cancer Res 24, 6447-6458 (2018). 750 55. Z. S. Chheda, R. K. Sharma, V. R. Jala, A. D. Luster, B. Haribabu, Chemoattractant Receptors BLT1 and CXCR3 751 Regulate Antitumor Immunity by Facilitating CD8+ T into Tumors. J Immunol 197, 2016- 752 2026 (2016). 753 56. D. Dangaj et al., Cooperation between Constitutive and Inducible Chemokines Enables T Cell Engraftment 754 and Immune Attack in Solid Tumors. Cancer Cell 35, 885-900 e810 (2019). 755 57. S. Zhang et al., CCL5-deficiency enhances intratumoral infiltration of CD8(+) T cells in colorectal cancer. 756 Cell Death Dis 9, 766 (2018). 757 58. Y. Ban et al., Targeting Autocrine CCL5-CCR5 Axis Reprograms Immunosuppressive Myeloid Cells and 758 Reinvigorates Antitumor Immunity. Cancer Res 77, 2857-2868 (2017). 759 59. P. Savas et al., Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset 760 associated with improved prognosis. Nat Med 24, 986-993 (2018). 761 60. R. R. Ji et al., An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer 762 Immunol Immunother 61, 1019-1031 (2012). 763 61. R. Barreira da Silva et al., Dipeptidylpeptidase 4 inhibition enhances lymphocyte trafficking, improving 764 both naturally occurring tumor immunity and immunotherapy. Nat Immunol 16, 850-858 (2015). 765 62. J. Decalf et al., Inhibition of DPP4 activity in humans establishes its in vivo role in CXCL10 post-translational 766 modification: prospective placebo-controlled clinical studies. EMBO Mol Med 8, 679-683 (2016). 767 63. Y. Li et al., Hydrogel dual delivered celecoxib and anti-PD-1 synergistically improve antitumor immunity. 768 Oncoimmunology 5, e1074374 (2016). 769 64. D. Peng et al., Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. 770 Nature 527, 249-253 (2015). 771 65. M. Wendel, I. E. Galani, E. Suri-Payer, A. Cerwenka, accumulation in tumors is dependent 772 on IFN-gamma and CXCR3 ligands. Cancer Res 68, 8437-8445 (2008). 773 66. I. C. Hirako et al., Splenic differentiation and emergence of CCR5(+)CXCL9(+)CXCL10(+) monocyte-derived 774 dendritic cells in the brain during cerebral malaria. Nat Commun 7, 13277 (2016). 775 67. W. Peng et al., PD-1 blockade enhances T-cell migration to tumors by elevating IFN-gamma inducible 776 chemokines. Cancer Res 72, 5209-5218 (2012). 777 68. H. Bronger et al., Modulation of CXCR3 ligand secretion by E2 and inhibitors 778 in human breast cancer. Breast Cancer Res 14, R30 (2012). 779 69. M. T. Chow et al., Intratumoral Activity of the CXCR3 Chemokine System Is Required for the Efficacy of 780 Anti-PD-1 Therapy. Immunity, (2019). 781 70. K. E. Yost et al., Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 25, 1251- 782 1259 (2019).

33

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

783 71. Y. Ohmori, R. D. Schreiber, T. A. Hamilton, Synergy between interferon-gamma and - 784 alpha in transcriptional activation is mediated by cooperation between signal transducer and activator of 785 transcription 1 and nuclear factor kappaB. J Biol Chem 272, 14899-14907 (1997). 786 72. D. L. Clarke et al., TNFalpha and IFNgamma synergistically enhance transcriptional activation of CXCL10 in 787 human airway smooth muscle cells via STAT-1, NF-kappaB, and the transcriptional coactivator CREB- 788 binding protein. J Biol Chem 285, 29101-29110 (2010). 789 73. J. M. Zaretsky et al., Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N 790 Engl J Med 375, 819-829 (2016). 791 74. D. S. Shin et al., Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov 7, 188- 792 201 (2017). 793 75. R. T. Manguso et al., In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 794 547, 413-418 (2017). 795 76. C. J. Kearney et al., Tumor immune evasion arises through loss of TNF sensitivity. Sci Immunol 3, (2018). 796 77. A. Meiser et al., The chemokine receptor CXCR3 is degraded following internalization and is replenished 797 at the cell surface by de novo synthesis of receptor. J Immunol 180, 6713-6724 (2008). 798 78. M. C. Woodruff et al., Trans-nodal migration of resident dendritic cells into medullary interfollicular 799 regions initiates immunity to influenza vaccine. J Exp Med 211, 1611-1621 (2014). 800 79. J. Lian, A. D. Luster, Chemokine-guided cell positioning in the lymph node orchestrates the generation of 801 adaptive immune responses. Curr Opin Cell Biol 36, 1-6 (2015).

802

803

34

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

804 Figure legends

805 Figure 1: CXCL9 and CXCL10 are upregulated upon dual PD-1 and CTLA4 blockade and correlate with

806 survival in patient cohorts treated with immune checkpoint blockade

807 A) C57BL/6 mice were injected s.c. with 5 x 105 AT-3ova cells and allowed to establish for 14 days, following

808 which they were treated on days 14, 18, 22 and 26 with either dual treatment with anti-PD-1 (200 μg/mouse) and

809 anti-CTLA4 (150 μg/mouse) (P+C) or isotype control (ISO) (200 μg/mouse). Data shown as the mean ± SEM of 6-

810 8 mice per group of a representative experiment (n>3). B) Mouse survival is shown as combined data of two

811 independent experiments. **** P<0.0001 (determined by Two-way ANOVA and Mantel-Cox test). C-D Seven

812 days post treatment, tumors were harvested (n=3 per group) and C) gene expression was analysed by NanoString

813 and presented as an MA plot D) Tumors were cultured in PBS for 4 hours at 37ºC (2 µl per mg of tissue) and

814 chemokine expression determined by cytometric bead array. * p<0.05 **P<0.005, Multiple t-test. N = 18-19 mice

815 per group. E-H) Analysis of patient datasets as described in (23). E) Differential gene expression analysis comparing

816 responders (n=6) to non-responders (n=3) in baseline and F) on-treatment biopsies from melanoma patients

817 receiving combination nivolumab/pembrolizumab and ipilimumab. Benjamini-Hochberg adjusted p values shown.

818 G) Kaplan-Meier curves showing overall survival of 72 melanoma patients treated with anti-PD-1 or anti-PD-1 and

819 anti-CTLA-4 therapy stratified by high and low expression of indicated chemokines. Survival curves were compared

820 with the log-rank test. H) Correlation of expression of indicated chemokine and numbers of intratumoral CD8+ T

821 cells in 56 biopsies of metastatic melanoma taken prior to checkpoint blockade, with each case annotated by

822 response to therapy. Correlation tested with Pearson’s product moment correlation coefficient.

823 Figure 2: CXCR3 blockade abrogates therapeutic efficacy of dual PD-1 and CTLA4 blockade

824 A) C57BL/6 mice were injected s.c. with 5 x 105 AT-3ova cells and allowed to establish for 14 days before dual

825 treatment with anti-PD-1 (200 μg/mouse) and anti-CTLA4 (150 μg/mouse) ± anti-CXCR3 (200 μg/mouse). CXCR3

826 blockade was given days 13, 14, 18, 22 and 26. B) Tumor growth and C) survival was assessed. Data shown as the

827 mean ± of 6-8 mice/group of a representative experiment of n=3. Survival is shown as combined data of two

35

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

828 independent experiments. **P<0.005, * p<0.05 (determined by two-way ANOVA and Mantel-Cox test). Data points

829 for ISO and P+C treated are included in Figure 1B. D-E) Representative D) sizes and E) weights of tumors harvested

830 seven days post treatment. A representative experiment of n>3 is shown. F-H) C57BL/6 mice were injected with 3

831 x 105 MC38 cells (s.c.) and allowed to establish for 12 days (n = 6 mice per group) G) 1.5 x 105 E0771 cells (intra

832 mammary fat pad) and allowed to establish for 3 days (n = 5 – 8 per group of representative experiment n=2) or H)

833 2 x 105 B16F10-OVAdim cells (s.c.) and allowed to establish for 4 days (n = 16-17 mice per group) before dual

834 treatment with anti-PD-1/anti-CTLA4 ± anti-CXCR3, as per B, except for MC38 which received only two doses of

835 combined therapy.

836 Figure 3: CXCR3 blockade inhibits dual PD-1 and CTLA4 blockade-induced CD8+ T cell migration and

837 activation

838 C57BL/6 mice were injected s.c. with 5 x 105 AT-3ova cells and allowed to establish for 14 days before dual

839 treatment with anti-PD-1 and anti-CTLA4 ± anti-CXCR3 as per Figure 2. On day 21 (seven days post treatment)

840 TIL infiltrate was analysed as a percentage of CD45+ cells; A) CD8+ T cells B) CD4+foxp3- C) NK1.1+ cells D)

841 CD4+foxp3+ cells. E) T cell infiltrate quantified by immunofluorescent staining. Shown are representative images

842 and quantification of three tumors per group. F) IFNγ and TNFα expression in CD8+ and CD4+foxp3- T cells was

843 assessed. Shown is concatenated data from 6 mice (left) and quantification from two independent experiments

844 (right). G) Ki67, PD-1 and TIM3 expression was determined in CD8+ and CD4+foxp3- T cells cells. A-D, F, G)

845 Data represents combined data of two independent experiments with n=11-15 per group. H) Where indicated, mice

846 were dosed with either the S1PR1 inhibitor FTY720 (25 mg/kg) or DMSO control on days 13 and 14 post tumor

847 inoculation, and every two days subsequently. Data in right panel shows numbers of CD8+ and CD4+ T cells

848 collected from blood three days post initial FTY720 administration. Data is represented as the mean ± SEM of 6-7

849 mice per group (**** P<0.0001 *** P<0.001 ** P<0.01 1way, n.s. not significant, one-way ANOVA or two-way

850 ANOVA).

851

36

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

852 Figure 4: CXCR3 and its ligands modulate T cell priming within the tumor draining lymph nodes

853 Draining lymph nodes (DLNs) were harvested from AT-3ova tumor bearing C57BL/6 mice following treatment

854 with dual anti-PD-1 and anti-CTLA4 ± anti-CXCR3 as per Figure 2. A) Indicated immune cell populations were

855 isolated using FACS and CXCL9, CXCL10 and CXCR3 expression was assessed by qRT-PCR. Expression is

856 shown relative to the Rpl27 housekeeping gene and is representative of triplicate qRT-PCR reactions pooled from

857 n=4 mice per group. B-F) CD8+ and CD4+foxp3- T cells were assessed by flow cytometry for expression of B)

858 CXCR3 C) IFNγ D) Ki67 E) Tbet and F) PD-1. (* P<0.01, ** P<0.01, n.s. not significant, one-way ANOVA C-F

859 or unpaired t test B). Data is representative of n=12 per group from two pooled experiments.

860 Figure 5: Macrophages are the major source of intratumoral CXCL9 and CXCL10 following dual

861 combination therapy

862 C57BL/6 mice were injected s.c. with either A) 5 x 105 control AT-3ova (Mock KO) or CXCL9/CXCL10 double

863 KO AT-3ova cells and allowed to establish for 14 days. Mice were then treated with either dual anti-PD-1 (200

864 μg/mouse) and anti-CTLA4 (150 μg/mouse) or isotype control (200 μg/mouse). Treatment was repeated on days

865 18, 22 and 26. On day 21, tumors were collected from mice and CXCL9 and CXCL10 production assessed by

866 chemokine bead array as per Figure 1. A-B. Data shown is the mean ± SEM of 5-6 mice per group, of a

867 representative experiment (n=2). * P<0.05, n.s. not significant assessed by one way ANOVA. C-D, F) Intracellular

868 CXCL9 staining was performed ex vivo in the presence of golgi plug/golgi stop for 4 hours at 37ºC. n = 3 mice per

869 group, 2 representative experiments. C) CXCL9 production per cell type before and after therapy D) CXCL9

870 staining in indicated populations shown as representative histograms. E) Tissue enrichment scores for the top 200

871 genes upregulated in tumors treated with anti-PD-1 and anti-CTLA-4 relative to isotype control treated mice. F) Pie

872 charts indicating the source of CXCL9 by cell type. Each pie represents one mouse from a representative experiment.

873 G) C57BL/76 mice were injected with MC38 sub-cutaneously and 19 days post inoculation expression of CXCL9

874 by tumor infiltrating myeloid cells was determined. (H) Expression of Cxcl9, Cxcl10, Adgrel (F4/80), Fcgr1 (CD64)

875 and Itgae (CD103) in CT26 tumor-infiltrating immune cells as determined by single cell RNA-seq (35). UMAP

37

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

876 embedding of single cells in macrophage cluster (larger population) and fibroblast cluster (smaller population

877 bottom left) identified as per the original study are shown, with colour intensity representing normalized gene

878 expression level.

879 Figure 6 IFN is sufficient to induce CXCL9 expression from intratumoral macrophages

880 A. Correlation of IFN or TNFα and CXCL9/CXCL10 production as determined by cytometric bead array of tumor

881 samples obtained as per Figure 1. Data is represented as n= 6 mice per group from 2 independent experiments. R2

882 values determined by linear regression analysis. B) IFN and TNFα gene signature enrichment scores for CXCL9

883 and CXCL10 high tumors in urothelial cancer patients treated with Atezolizumab (ImVigor210 trial) C) AT-3ova

884 tumor bearing mice were treated as per Figure 5A with the additional treatment of anti-IFN or anti-TNFα (250

885 µg/mouse) on days 14 and 18. Expression of CXCL9 in MoMacs and CD103+ dendritic cells determined in n = 6

886 mice per group. D-E AT-3ova tumor bearing C57BL/6 wild type or BATF3-/- mice were treated as per Figure 5A.

887 Seven days post treatment tumor-infiltrating lymphocytes were dissociated and stimulated for 3 hours with 1 ng/ml

888 IFNγ and/or TNFα in the presence of Golgi Plug/Stop. Expression of CXCL9 in Mo/Macs or CD103+ DCs was

889 determined by flow cytometry. * P<0.05, ** P<0.01, ***P<0.001, not significant, one way ANOVA.

890 Figure 7: Generation of CXCR3 ligands by macrophages is critical for the efficacy of immune checkpoint

891 blockade

892 A-C) C57BL/6 mice were injected s.c. with 5 x 105 AT-3ova cells and allowed to establish for 14 days before dual

893 treatment with anti-PD-1, anti-CTLA4 and/or anti-F4/80 (100 µg/mouse) as per Figure 2. A) Tumor size (Left) and

894 survival (Right) of n = 6 mice per group from a representative experiment of n =2. B) CXCL9 production by tumors

895 ex vivo following therapy, n= 3-7 per group. C) FACS plots from concatenated samples (top) and individual data

896 points (n =8-14 per group; bottom) are shown. D-E) Expression of CXCL9, CXCL10, CXCL11, CD68 and CLEC9a

897 by cell subset in all patients at baseline in D) a cohort of metastatic melanoma patients and E) a cohort of lung

898 cancer patients that underwent single cell RNASeq (Melanoma; 5928 cells from 19 patients, Lung Carcinoma;

899 Violin plots are shown with single cell expression values overlayed). F) Single cell differential gene expression

38

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

900 analysis in macrophages in baseline melanoma samples from responders vs non responders. G) box plot of CXCL9,

901 CXCL10 and CXCL11 expression level in individual macrophages from baseline melanoma cases in relation to

902 immune checkpoint blockade response. Significance determined by unpaired t test. H) Proportion of macrophages

903 displaying chemokine signature in responders and non-responders.

904

905

39

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Adj. pval Figure 1 <0.05 >0.05 A B C 4 Ccl24 Cxcl3 Ccl17 Cxcl11 Ccl9

) ISO Ccl11 Cxcl1 2 Ccl1 2 Ccl5 Cxcl10 100 100 P+C Cklf Ccl6 Cxcl2 P+C Increased P+C Ccl19 Cxcl9

**** Ccl3 Ccl22 Ccl8

ISO **** logFC 0 50 Ccl7 50 Ccl12 Cx3cl1 −2

P+C Xcl1

Percent Survival 0 20 40 60 80 Decreased 4 8 12 16 Tumor size (mm 0 Days post treatment AveExpr 0 5 10 15 20 25 Absolute Expression P+C Days post treatment D E Responders vs. Non-Responders Pooled data- Heat Map Individual data points Differential gene expression Baseline

12 CCL5 15000 CCL20 * ISO 1500 P+C Adj. pval CCL11 ** 9 <0.05 CCL17 ** 5000 >0.05 CXCL1 CCL2 1000 4000 CXCL9 ** 6

pg/ml 3000 CXCL10 CXCL9 ** CCL3 CXCL10 2000 −log10(FDR) CCL4 3 CXCL11 500 ** CXCL13 1000 CCL5 CXCL5 CCL22 0 ** −2 0 2 ISO P+C pg/ml log2FC CCL3 CCL2 CCL4 CCL5 CCL11

CCL20 CCL17 Decreased Increased CCL22 CXCL1 CXCL9 CXCL5 CXCL10 CXCL13 in responders in responders F G Responders vs. Non-Responders 1.00 CXCL9 high 1.00 CXCL10 high 1.00 CXCL11 high | || Differential gene expression | CXCL9 low CXCL10 low | CXCL11 low | | ||| | | |||||| ||| | ||| |||| | ||| | ||| On therapy 0.75 | | |||| ||| | |||| 0.75 | 0.75 || | | | |||| ||| | |||| | | | || || | || ||||||| | | || || |||| 25 || || 0.50 ||| 0.50 || |||||| | 0.50 | Adj. pval ||| | | || | || <0.05 | 20 >0.05 0.25 p = 0.00048 0.25 p = 0.00099 0.25 p = 0.097 Survival probability Survival probability Survival probability | |

15 0.00 0.00 0.00 0 12 24 36 48 0 12 24 36 48 0 12 24 36 48 OS Time (months) OS Time (months) OS Time (months)

CXCL10 10 CXCR3 high 1.00 || −log10(FDR) | CCL5 CXCR3 low | |||||||| CXCL11 |||| | 0.75 | ||| || ||| CXCL9 5 || | ||||| 0.50 0 || −10 −5 0 5 | | || | 0.25 p = 0.0042 −log2FC Survival probability 0.00 Decreased Increased 0 12 24 36 48 in responders in responders OS Time (months) CXCL9 CXCL10 CXCL11 H 5 4 4 2 3 response 3 0 2 2 NR 1 1 −2 0 0 R −4 −1 −1 Normalised expression Normalised expression 10 100 1000 10 100 1000 Normalised expression 10 100 1000 Intratumoural CD8 Intratumoural CD8 Intratumoural CD8 (cells/mm2) Log scale (cells/mm2) Log scale (cells/mm2) Log scale Downloaded from clincancerres.aacrjournals.orgp = 2.08E-07 on September 27, 2021.p ©= 5.64E-062019 American Associationp = 2.17E-05 for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Figure 2Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A Treatment timeline AT3 ovadim injection Isotype control (200 µg/mouse) or (5 x 105 cells/mouse) αPD-1 (200 µg/mouse) + αCTLA-4 (150 µg/mouse)

Day -14 -1 0 4 8 12

αCXCR3 (200 µg/mouse)

B AT-3ova C AT-3ova ISO

ISO + anti-CXCR3 100 P+C *** 100 ISO )

2 ISO + anti-CXCR3 P+C + anti-CXCR3 P+C 50 P+C + anti-CXCR3 ** 50

Percent Survival 0 20 40 60 80 Tumor size (mm 0 Days post treatment 0 5 10 15 20 25

Days post treatment

D E ** ISO ** 100 P+C

ISO + 50 anti-CXCR3 P+C +anti-CXCR3

Tumor weight (mg) 0 10 mm ISO P+C ISO + P+C + anti-CXCR3 anti-CXCR3

F G H MC38 E0771 B16F10-OVA

ISO ISO

**** ISO + anti-CXCR3 ISO + anti-CXCR3 **** P+C P+C ) P+C + anti-CXCR3 ) P+C + anti-CXCR3 2 100 2 80 200 100 ISO ISO + anti-CXCR3 60 P+C 40 100 50 P+C + anti-CXCR3 * 20 0 Tumor size (mm Tumor size (mm 0 0 Percent Survival 0 2 4 6 8 10 12 0 2 4 6 8 10 12 10 20 30 40 50 Days post treatment Days post treatment Days post treatment

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Figure 3Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A CD8+ T cells B CD4+foxp3- C NK Cells D CD4+foxp3+ cells n.s. n.s. 80 **** *** 30 ** 15 20 60 15 20 10 40 10

%CD8+ 10 5 20 %NK1.1+ 5 %CD4+foxp3-

0 0 0 %CD4+foxp3+ 0 ISO ISO ISO ISO P+C P+C P+C ISO P+C P+C P+C P+C P+C ISO+ ISO+ ISO+ ISO+ anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 ISO P+C anti-CXCR3 anti-CXCR3 E P+C CD8+ CD4+ CD4+foxp3+ ) ) ) 2 2 2 25 25 1500 *** ** 20 20 n.s. 1000 15 15 ISO+anti-CXCR3 P+C + anti-CXCR3 10 10 500 5 5 0 0 0 Cell density (Cells/mm Cell density (Cells/mm Cell density (Cells/mm ISO ISO ISO P+C P+C P+C P+C P+C P+C ISO+ ISO+ ISO+

100μm anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 CD3 Pooled data F ISO P+C ISO+ anti-CXCR3 P+C anti-CXCR3 100 ** ** 80 CD8+ 60 +/TNF α +

γ 40 20 %IFN 0 n.s.

TNF α ** 100 ** 80 60

CD4+foxp3- +/TNF α +

γ 40 20 %IFN 0 ISO P+C P+C IFNγ ISO+ anti-CXCR3 anti-CXCR3 G Proliferation Immune Checkpoints H CD8+ CD4+foxp3- CD8+ CD4+foxp3- ISO + DMSO **** n.s. ISO + FTY720 500 n.s. ** ISO + FTY720+anti-CXCR3 400 ** 100 n.s. 80 n.s. 30 15 P+C + DMSO 300 ** ** P+C + FTY720 200 80 60 10 P+C+ FTY720+anti-CXCR3 100 20 P+C + DMSO+anti-CXCR3

60 CD8+/ υ l blood

) 0

40 2

%Ki67+ 40 **** 10 5 100 800

20 %PD-1+TIM3+ 20 * 600 0 0 0 0 50 ** 400 ISO ISO ISO ISO P+C P+C P+C

P+C 200 P+C P+C P+C CD4+/ υ l blood P+C ISO+ ISO+ ISO+

ISO+ 0 0 Tumor size (mm anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 0 5 10 15 Days post treatment DMSO Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer FTY720 Research. Figure 4 Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 A Author manuscripts have been peer reviewed and accepted for publicationB but have not yet been edited. CXCL9 CXCL10 CXCR3 CD8+ CD4+foxp3- CD4+foxp3+ n.s. 40 ** 60 * 20 ISO 20 ISO 20 ISO 10 P+C P+C P+C 30 15 15 15 40 10 10 10 20 5 %CXCR3+ %CXCR3+ 5 5 5 %CXCR3+ 20 10 0 0 0 0 0 0 Relative Expression to Rpl27 to Expression Relative Relative Expression to Rpl27 Relative Expression to Rpl27 CD4+ CD4+ CD8+ CD8+ CD4+ CD8+ ISO ISO ISO P+C P+C P+C CD11c+ CD103+ MoMac CD11b+ Mo/Mac CD11b+ Mo/Mac CD11c+ CD103+ CD11b+ CD11c+ CD103+

C CD8+ CD4+foxp3- D CD8+ CD4+foxp3-

25 * * 10 * n.s. 15 * n.s. 15 * * 20 8 + 10 + 10 15 6 %Ki67 %Ki67 %IFN γ + %IFN γ + 10 4 5 5 5 2 0 0 0 0 ISO ISO P+C P+C ISO ISO P+C P+C P+C P+C ISO+ ISO+ P+C P+C ISO+ ISO+ anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3

E CD8+ CD4+foxp3- F CD8+ CD4+foxp3- * n.s. 5 * n.s. 5 * n.s. * * 10 15 4 4 3 3 10 %PD-1+ %PD-1+ %Tbet+ 5 %Tbet+ 2 2 5 1 1 0 0 0 0 ISO ISO ISO P+C P+C ISO P+C

P+C P+C P+C P+C ISO+ ISO+ P+C ISO+ ISO+ anti-CXCR3 anti-CXCR3 anti-CXCR3

anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3 anti-CXCR3

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Figure 5

Author Manuscript PublishedB OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868C A Author manuscripts have been peerMock reviewed KO CXCL9/CXCL10and accepted for KOpublication but have not yet been edited. Mock KO ISO 150 Mock KO P+C 1000 250 CXCL9 (Protein) )

2 CXCL9/10KO ISO 80 ISO ** CXCL9/10KO P+C 800 200 P+C *** 100 60 ** 600 150 *** 40 400 100 50 CXCL9 pg/ml CXCL10 pg/ml Tumor size (mm Tumor 20 200 50 Percent CXCL9+ ** *

0 0 0 0 0 10 20 30 ISO ISO P+C Days post treatment P+C CD45- CD45+ CD11c+ CD11c+ CD103+ CD11b+ Mo/Mac D E CD45+ CD11c+CD103+ Mo/Mac GSEA analysis

ISO Macrophage anti-PD-1 Treg Cells anti-CTLA-4 NK Cells P+C T lymphocyte Neutophil Dendritic cell B lymphocyte 0 20 40 60 10 CXCL9 Log- (q-value) F G MC38 CXCL9 Protein Expression

CD45-

CD8+ T cell Lin CD64 ISO Lin+CD8- Mo/Mac CD103+ DCs CXCL9 CD11b CD11c+Ly6c-MHCII+CD103-

P+C CD11b+Ly6CintCD11c- Other MHCII

H Adgre1 CD11c Cxcl9 Cxcl10 (F4/80) -3 -3 -3 -4 -4 -4 -5 3 -5 3 -5 2 2 2 -6 1 -6 1 -6 1

UMAP_2 -7 0 UMAP_2 -7 0 UMAP_2 -7 0 -8 -8 -8 -4 -2 0 2 4 -4 -2 0 2 4 -4 -2 0 2 4 UMAP_1 UMAP_1 UMAP_1 Fcgr1 Itgae (CD64) -3 -3 (CD103) -4 -4 3 -5 2 -5 1.0 -6 1 -6 0.5

UMAP_2 -7 0 UMAP_2 -7 0.0 -8 -8 -4 -2 0 2 4 -4 -2 0 2 4 UMAP_1 UMAP_1

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Figure 6 Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 A Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. ISO PD1 CTLA4 P+C 400 200 400 2 200 r2 -0.65 r2 -0.82 r -0.83 r2 -0.85 300 150 300 150

200 100 200 100 IFNγ pg/mL TNFα pg/mL IFNγ pg/mL 100 TNFα pg/mL 50 100 50

0 0 0 0 1 2 3 4 5 1 2 3 4 5 0.5 1.51.0 0.5 1.51.0 CXCL9 ng/mL CXCL9 ng/mL CXCL10 ng/mL CXCL10 ng/mL

B CXCL9: IFNγ enrichment CXCL10: IFNγ enrichment C -26 -6 Mo/Mac CD103+ DC FDR 1x10 FDR 3x10 n.s. 80 *** 80 n.s. * *** 60 60 * 40 40

CXCL9: TNFα enrichment CXCL10: TNFα enrichment 20 20 Percent CXCL9+ -8 Percent CXCL9+ FDR 1x10-31 FDR 2x10 0 0 ISO ISO Control Control anti-IFN γ anti-IFN γ anti-TNF α anti-TNF α P+C P+C

D Mo/Mac CD103+ DC CD45- Mo/Mac E 2A3 2A3 2A3 P+C Mo/Mac 60 80

40 40 40 60 40 40 20 20 20 20 20 Percent CXCL9+ Percent CXCL9+ Percent CXCL9+ Percent CXCL9+ Percent CXCL9+ 0 0 0 0 0 IFN γ IFN γ IFN γ IFN γ IFN γ IFN γ TNF α TNF α TNF α TNF α Control Control Control Control Control Control IFN γ/ TNF α IFN γ/ TNF α IFN γ/ TNF α IFN γ/ TNF α WT BATF3-/-

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Figure 7 A Author Manuscript Published OnlineFirst on October 21,B 2019; DOI: 10.1158/1078-0432.CCR-19-1868 ISOAuthor manuscripts have been peerISO reviewed and accepted for publicationn.s. but have not yet been edited. ISO+ αF4/80 ISO+ αF4/80 500 * **

) P+C 2 P+C 400 P+C + P+C + αF4/80 αF4/80 **** 300 100 100 200 50 100

50 CXCL9 pg/ml 0 0 0 Tumor size (mm ISO P+C

10 201550 25 Percent Survival 10 20 30 40 Days post treatment Days post treatment α F4/80 P+C+ α IFN γ ISO+ P+C+ α F4/80 C D Melanoma ISO P+C P+C + αF4/80 CXCL9 CXCL10 CXCL11 Tregs CD8+ CD8+ Plasma cells CD8+ Monocytes/Macrophages 23.2% 40.7% 30.2% Memory T−cells Lymphocytes exhausted/cell−cycle Lymphocytes CD4 Exhausted/HS CD8+ T−cells Exhausted CD8+ T−cells Dendritic cells Cytotoxicity (Lymphocytes) B−cells CD8 1 3 5 1 3 10 1 3 10 Log Expression Level CD8+ CD4+foxp3- CD4+foxp3+ E 80 *** ** 15 ** * 20 Lung carcinoma CXCL9 CXCL10 60 15 30 10 30 10 40 10 10 5 20 5 3 3 1 1 0 0 0 tDC1 tDC1 tDC2 tpDC tDC3 tDC2 tpDC tDC3 % of CD45+ cells Log Expression Level Log Expression Level tMac5 tMac4 tMac1 tMac2 tMac3 tMac5 tMac4 tMac1 tMac2 tMac3 tMac6 tMac7 tMac8 tMac9 tMac6 tMac7 tMac8 tMac9 tMono1 tMono2 tMono3 tMono1 tMono2 tMono3 ISO ISO ISO tMonoDC tMonoDC P+C P+C P+C CXCL11

5 P+C+ α IFN γ P+C+ α IFN γ P+C+ α IFN γ 3 P+C+ α F4/80 P+C+ α F4/80 P+C+ α F4/80

1 tDC1 tDC2 tpDC tDC3 Log Expression Level tMac5 tMac4 tMac1 tMac2 tMac3 tMac6 tMac7 tMac8 tMac9 tMono1 tMono2 tMono3 tMonoDC F Macrophage gene DE CD68 Clec9a Responders vs 10 5 3 Non-responders 3 Adj. pval < 0.05 1 1 > 0.05 Log Expression Level tDC1 tDC1 tDC2 tDC2 tpDC tpDC tDC3 tDC3 Log Expression Level tMac5 tMac5 tMac4 tMac4 tMac1 tMac2 tMac3 20 tMac1 tMac2 tMac3 tMac6 tMac7 tMac8 tMac9 tMac6 tMac7 tMac8 tMac9 tMono1 tMono2 tMono3 tMono1 tMono2 tMono3 tMonoDC tMonoDC

val) CXCL11 CCL3L1 Non- Signature G H Responders 10 CCL4L2 CXCL10 responders Expression CXCL3 CCL8 Mono/ Mono/ Mono/ CXCL1 CCL3L3 CCL4L1 100

−log10(p CXCL16 CCL4 CCL2 Macrophages Macrophages Macrophages Low CCL5 CXCL9 4e -07 5e -06 0.7 High 0 CCL20 CXCL2 CCL7 75 −5.0 −2.5 0.0 2.5 avg logFC 10 10 10 50

25 Decreased Increased 5 5 5 in responders in responders CXCL9 CXCL11 CXCL10

Percent of macrophages 0 0 P8 P4

0 P3 0 P2 P6 P31 P24 P29 NR R NR R NR R P12 P15 P27 P20 P26

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 21, 2019; DOI: 10.1158/1078-0432.CCR-19-1868 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Macrophage derived CXCL9 and CXCL10 are required for anti-tumor immune responses following immune checkpoint blockade

Imran G House, Peter Savas, Junyun Lai, et al.

Clin Cancer Res Published OnlineFirst October 21, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-1868

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2019/10/19/1078-0432.CCR-19-1868.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2019/10/19/1078-0432.CCR-19-1868. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2019 American Association for Cancer Research.