<<

Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Research Article Cancer Immunology Research A Transcriptionally Distinct CXCL13þCD103þCD8þ T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Cancer Hagma H. Workel1, Joyce M. Lubbers1, Roland Arnold2, Thalina M. Prins1, Pieter van der Vlies3, Kim de Lange3, Tjalling Bosse4, Inge C. van Gool4, Florine A. Eggink1, Maartje C.A. Wouters5, Fenne L. Komdeur1, Elisabeth C. van der Slikke1, Carien L. Creutzberg6, Arjan Kol1, Annechien Plat1, Mark Glaire7, David N. Church7,8, Hans W. Nijman1, and Marco de Bruyn1

Abstract

þ The CXCL13 mediates recruitment of B cells to CD8 T cells from peripheral blood activated in the presence tumors and is essential for the formation of tertiary lymphoid of TGFb upregulated CD103 and secreted CXCL13. Converse- structures (TLSs). TLSs are thought to support antitumor ly, inhibition of TGFb signaling abrogated CXCL13 þ þ þ immunity and are associated with improved prognosis. How- production. CXCL13 CD103 CD8 TILs correlated with ever, it remains unknown whether TLSs are formed in response B-cell recruitment, TLSs, and neoantigen burden in six cohorts to the general inflammatory character of the tumor microen- of human tumors. Altogether, our findings indicated that vironment, or rather, are induced by (neo)-specific TGFb plays a noncanonical role in coordinating immune adaptive immunity. We here report on the finding that the responses against human tumors and suggest a potential role þ þ þ þ þ TGFb-dependent CD103 CD8 tumor-infiltrating T-cell (TIL) for CXCL13 CD103 CD8 TILs in mediating B-cell recruit- subpopulation expressed and produced CXCL13. Accordingly, ment and TLS formation in human tumors.

Introduction Unfortunately, only a subset of patients currently benefit from treatment. ICIs are more likely to be effective in patients with a inhibitors (ICIs) targeting programmed þ preexisting anticancer immune response, most notably a CD8 death 1 (PD-) or its receptor, programmed death 1 (PD- cytotoxic T-cell response against tumor neoantigens (4). 1), have elicited unprecedented long-term disease remissions in Responsive tumors harbor significantly more predicted neoan- advanced and previously treatment-refractory cancers (1–3). tigens (5, 6) and display evidence of a coordinated immune response comprising T cells, dendritic cells (DCs), and B cells (7). In diseases that parallel tumor development, such as chronic 1 Department of Obstetrics and Gynecology, University Medical Center Gronin- inflammatory conditions, this coordinated infiltration by differ- gen, University of Groningen, Groningen, the Netherlands. 2Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom. ent immune cell subsets is frequently associated with tertiary – 3Department of Genetics, University Medical Center Groningen, University of lymphoid structures (TLSs) an ectopic form of lymphoid tissue. Groningen, Groningen, the Netherlands. 4Department of Pathology, Leiden TLSs exhibit features of regular lymph nodes, including high University Medical Center, Leiden University, Leiden, the Netherlands. 5Trev endothelial venules, a T-cell zone with mature DCs, and a ger- and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, minal center with follicular DCs and B cells (8). Several studies 6 Canada. Department of Radiation Oncology, Leiden University Medical Center, have reported the presence of TLSs in tumors, which was generally Leiden University, Leiden, the Netherlands. 7Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics and Oxford Cancer found to be associated with greater immune control of cancer 8 growth and improved prognosis (9, 10). For several malignancies, Centre, University of Oxford, Oxford, United Kingdom. NIHR Oxford Biomedical þ Research Centre, Oxford University Hospitals NHS Foundation Trust and John the combination of TLS presence and high CD8 T-cell infiltration Radcliffe Hospital, Oxford, United Kingdom. was found to associate with superior prognosis, whereas high þ Note: Supplementary data for this article are available at Cancer Immunology CD8 T-cell infiltration alone associated with poor or moderate Research Online (http://cancerimmunolres.aacrjournals.org/). prognosis (11, 12). These observations highlight the importance H.H. Workel and J.M. Lubbers are the co-first authors of this article. of a coordinated immune response, including TLS formation, in anticancer immunity. H.W. Nijman and M. de Bruyn are the co-senior authors of this article. To date, the molecular determinants of tumor TLS formation Corresponding Author: Marco de Bruyn, University of Groningen, University remain incompletely understood. Current data suggest that TLS Medical Center Groningen, Hanzeplein 1, Groningen 9713GZ, the Netherlands. formation results from a complex interplay between DCs, T cells, Phone: 3105-0361-3174; Fax: 3105-0361-1806; E-mail [email protected] B cells, and supporting stromal cells. Interplay among these cells doi: 10.1158/2326-6066.CIR-18-0517 relies on reciprocal and chemokine signaling, including 2019 American Association for Cancer Research. chemokine [C-X-C motif] ligand 13 (CXCL13), receptor activator

784 Cancer Immunol Res; 7(5) May 2019

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

of nuclear factor k B (ligand)(RANK/RANKL), ab Analysis of TCGA mRNA sequencing data (LTab), and chemokine (C-C motif) ligand 21 (CCL21; ref. 13). RSEM-normalized mRNA-seq data and clinical data from uter- A central role for CXCL13 in this process is suggested by the ine corpus endometrial carcinoma (UCEC), ovarian cancer, breast inability of B cells to home and accumulate into lymphoid cancer, and adenocarcinoma were downloaded from fire- aggregates (14) and generate functional lymphoid tissue (15, browse.org on March 13, 2017 (UCEC) and July 14, 2017 (ovarian 16) in CXCL13-knockout mice and the observation that cancer, breast cancer, lung adenocarcinoma). RSEM mRNA CXCL13 alone is sufficient to generate lymphoid tissues (17– sequencing expression data were log2þ1 transformed, and 19). Nevertheless, a key outstanding question remains whether with zero reads in all samples were removed. POLE-EDM, MSI, and tumor-associated TLSs are formed in response to the general MSS cases were identified in the endometrial cancer data. The inflammatory character of the , or mononucleotide and dinucleotide marker panel analysis status rather, are induced by (neo)antigen-specific adaptive immuni- was provided by The Cancer Genome Atlas (TCGA), and mutations ty. Two studies suggest that a subset of CXCL13-producing in the exonuclease domain of POLE were determined previous- þ CD8 tumor-infiltrating (TIL) may link (neo) ly (26). Heatmaps were constructed in R (version 3.3.1) with antigen recognition to TLS formation (7, 20). packages gplots and ggplots. The set of TLSs was reported þ þ Here, we report the finding that TGFb receptor signaling previously (10, 27). Gene sets for the CD8 CD103 and þ þ – licensed CD8 T cells to produce and secrete CXCL13 upon CD8 CD103 signatures were derived from the sequencing concurrent T-cell receptor (TCR) stimulation. Induction of data (Supplementary Table S1). Spearman correlations between þ þ CXCL13 was paralleled by upregulation of CD103, a marker for the TLS signature, the CD8 CD103 signature, and the þ – tissue-resident TILs. Accordingly, bulk and single-cell RNA CD8 CD103 signature were visualized in correlation plots using sequencing identified exclusive expression of CXCL13 in human the Corrplot package (Version 0.77) in R. CXCL13highCD103high þ – þ CD103 , but not CD103 , CD8 TILs. In line with these data, the versus CXCL13lowCD103low groups in MSS UCEC were based on þ presence of CD103 CTLs correlated to B-cell recruitment and median expression of CXCL13 (4.35698) and ITGAE (8.086985). TLSs in tumors with a high mutational load. This discovery sheds Survival curves were constructed with R packages survival (version on how B cells could be recruited to tumors by CTLs, 2.41-3) and survminer (version 0.4.3). All analyses were performed identifying a noncanonical role for TGFb in the orchestration of in R (version 3.4.0), with exception of the construction of the a coordinated immune response against human (neo)antigen- heatmap in Fig. 3C, which was made in R version 3.3.1. rich tumors. Our findings also identify CD103 and B cells as potential biomarkers for ICI in epithelial malignancies. IHC To assess the presence of TLSs in uterine cancer, we stained for fi Materials and Methods the B-cell marker CD20 on whole tissue sections. Formalin- xed, paraffin-embedded (FFPE) slides were deparaffinized and rehy- Patients drated in graded ethanol. Antigen retrieval was initiated with a Tumor tissue from four patients with stage IIIC high-grade preheated 10 mmol/L citrate buffer (pH ¼ 6) and endogenous serous ovarian cancer was collected during primary cytoreductive peroxidase activity was blocked by submerging sections in a surgery, prior to chemotherapy, and from one patient with stage 0.45% hydrogen peroxide solution. Slides were blocked in PBS IV high-grade serous ovarian cancer during interval debulking containing 1% human serum and 1% BSA. Slides were incubated upon three cycles of chemotherapy. Written informed consent overnight with anti-CD20 (0.63 mg/L; clone L26, catalog number was obtained from all patients. Selection of patients with uterine M0755, Dako) at 4C. Subsequently, slides were incubated with fl cancer was described previously (21). Brie y, uterine cancer a ready-to-use peroxidase-labeled polymer for 30 minutes tissue was obtained from patients involved in the PORTEC- (Envisionþ/HRP anti-mouse, 2 drops, catalog number K4001, ¼ 0 1 (22) and PORTEC-2 (23) studies (n 57), the uterine cancer Dako). Signal was visualized with 3,3 diaminobenzidin (DAB) ¼ series (n 67) from Leiden University Medical Center (LUMC, solution, and slides were counterstained with hematoxylin. ¼ Leiden, the Netherlands), and uterine cancer series (n 26) from Appropriate washing steps with PBS were performed in-between the University Medical Center Groningen (UMCG, Groningen, incubation steps. Sections were embedded in Eukitt mounting the Netherlands) in accordance with local medical ethical guide- medium (Sigma Aldrich), and slides were scanned on a Hama- fi lines (24). Tumor material was xed in formalin and embedded matsu digital slide scanner (Hamamatsu Photonics). The number fi þ in paraf n. Tumor material from 119 patients was available for of CD20 (dense) follicles in each slide was quantified in analysis. Mutations in the exonuclease domain of polymerase NDPview2 software by two independent observers who were epsilon (POLE-EDM) and microsatellite instability status were blinded to clinicopathologic data. known from previous studies (24). Of the tumors available for IHC for CD8 was performed previously in this cohort (24). To this study, 42 tumors were POLE wild-type and microsatellite assess the survival effect of CD103 infiltration in mismatch stable (MSS), 38 were POLE wild-type with microsatellite insta- repair–proficient (pMMR) cancers, we used a staining for CD103 bility (MSI), and 39 were POLE-EDM. POLE-EDM statuses did on tissue microarray slides of uterine cancer (28). FFPE slides were not cooccur with microsatellite instability. All cases were of prepared as described above and incubated with rabbit-anti endometrioid histology (EEC), and the number of low-grade human CD103 [1 mg/L; anti-E7-, clone ERPR4166(2), and high-grade tumors was spread equally over the three molec- catalog number Ab129202, Abcam), followed by a ready-to-use ular groups. Selection of patients with ovarian cancer and CD103 peroxidase-labeled polymer (Envisionþ/HRP anti-rabbit, cat.no. staining was reported previously (25). Ethical approval for K4003, Dako) and Biotin Tyramide working solution (TSA , tumor molecular analysis was granted at LUMC, UMCG, Perkin Elmer), streptavidin-HRP (TSA kit, Perkin Elmer), and 3,30- and by Oxfordshire Research Ethics Committee B (approval no. diaminobenzidin/hematoxylin. Positively stained cells were 05\Q1605\66). quantified per core and adjusted for core surface. Patients with

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 785

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

at least two cores with a minimum of 20% tumor were Table S4], enabling identification of individual cells after pooled included for analysis. All slides were counted manually by two RNA sequencing. individuals who were blinded for clinicopathologic data. In addition to single-cell wells, small bulk populations of 20 þ cells were sorted per microplate well. Per patient, 40 single CD8 þ – Multicolor immunofluorescence T cells (20 wells CD103 , 20 wells CD103 ) and 20 small bulk 20- þ – FFPE slide preparation and antigen retrieval were performed cell populations (10 wells CD103 , 10 wells CD103 ) were as described above. Next, slides were incubated overnight at sorted. After sorting, the plate was vortexed and spun down 4C with primary antibody and subsequently incubated with briefly, and incubated at 72C for 3 minutes. After this step, the the appropriate secondary antibody for 45 minutes at room plate was kept cool. The transcriptomes were amplified by a temperature (Supplementary Table S2). Specificsignalwas modified SMART-Seq2 protocol using SmartScribe reverse tran- amplified using the TSA Cyanine 5 (Cy5) detection kit (Perkin scriptase (Westburg-Clontech, CL639537), based on a previously Elmer, NEL705A001KT) or the TSA Cyanine 3 (Cy3) and published protocol (29). In brief, custom primers were designed Fluorescein detection kit (Perkin Elmer, 753001KT), according that included a PCR-primer recognition site for pre-PCR, a Unique to the manufacturer's protocols. To allow multiple amplifica- Molecular Identifier (UMI), a cell barcode (see Supplementary tions on the same slide, primary HRP labels were destroyed Table S4), and a poly T-stretch. Each cell (or pool) was tagged with between incubations by washing with 0.01 mol/L hydrochloric an oligo dT primer, including the UMI and cell barcode. A acid for 10 minutes. Appropriate washing steps with PBS template switching pre-PCR was used to generate cDNA. Pools containing 0.05% Tween20 (Sigma-Aldrich) were performed are made of single cells or pools with unique cell barcodes during the procedure. For embedding, Prolong Diamond anti- and 500 pg of the pools was tagmentated and barcoded using fade mounting medium with or without DAPI was used a N7xx index and custom P5 hybrid primer: (AATGATACG- (Invitrogen/Thermo Fisher Scientific, P36962 and P36961). GCGACCACCGAGATCTACACGCCTGTCCGCGGAAGCAGTG- Finally, slides were scanned at room temperature using the GTATCAACGCAGAGTAC) according the Illumina Nextera XT TissueFAXS acquisition software and microscope (TissueGnostics) DNA Sample Preparation Kit Protocol (Illumina, catalog no. with the following specifications: Zeiss EC "Plan-Neofluar" FC-131-1096). The pools were purified using AMPure beads, in 40/1.30 Oil, DIC objective, CMOS-color camera PL-B623 a ratio of 0.6:1, to remove primer dimers. Presence and size Pixelink (3.1 Megapixels), EXFO Excite 120 PC fluorescence distribution of the obtained PCR product were checked on a illumination and Chroma ET Dapi (49000), Chroma ET CY3 PerkinElmer LabChip GX high-sensitivity DNA chip. A super pool (49004), Chroma ET Cy5 (49006), and Chroma FITC (49011) was created by equimolar pooling (1 nmol/L) of the Nextera filter sets. Overlay images were produced using Adobe Photoshop products, and the samples were sequenced on Illumina Next- software. Seq500 2500 using 50 bp paired-end reads, one read for the mRNA transcript, and the other for the cell-barcode. The obtained mRNA sequencing RNA-sequencing data were demultiplexed into individual FASTQ Ovarian tumors from two patients were cut into pieces of files. The obtained single-end reads were aligned to human <1mm3 and placed in a T75 culture flask (Nunc EasYFlask Cell reference genome 37 [GRCh37.p13 (GCA_000001405.14), top- Culture Flasks, catalog no. 156499, Thermo Fisher Scientific) with level built] using STAR (version 2.5.2). digestion medium, consisting of RPMI (Gibco), 10% FBS (Gibco), RNA-SeQC (version 1.1.8) was then used to assess the quality collagenase type IV (1 mg/mL; Gibco), and recombinant human of single cells. Data were visualized and clear low-quality outliers DNase (12.6 mg/mL; Pulmozyme, Roche) for overnight digestion were identified on the basis of number of transcripts, uniquely at room temperature. After digestion, the suspension was strained mapped reads, mapping rate, expression profiling efficiency and through a 70-mm filter and washed with PBS. Cells were centri- exonic rate, and these were removed from further analysis. All cells fuged over a Ficoll-Paque gradient (GE Healthcare Bio-Sciences that did not meet one of the following criteria were removed: AB) and lymphocytes were isolated from between the two layers. <10,000 transcripts detected, <500,000 uniquely mapped reads, After a wash with PBS the cells were pelleted. Total cell pellet was <1,000 genes detected, a mapping rate of <0.5, an expression suspended in 1 mL FBS with 10% dimethylsulfoxide (Merck) and profiling efficiency of <0.4, or an exonic rate of <0.5 (Supple- stored in liquid nitrogen until further use. mentary Table S5). mRNA expression values for small bulk Prior to sequencing, tumor digests were thawed on ice, washed populations of 20 cells are shown in transcripts per kilobase with AIM-V medium (Gibco) with 5% pooled human serum million (TMP) and for single cells as fragments per kilobase (PHS, One Lambda) and centrifuged at 1,000 g. The total cell million (FKPM) and log2þ1 transformed. Differential expression pellets were resuspended in AIM-V with 5% PHS, and cells were in the 20-cell populations was analyzed with DESeq2 (version þ incubated with CD3-BV421, CD4-PerCP-Cy5.5, CD8a-APCe- 1.16.1) to obtain insight into the differences between CD103 – þ Fluor780, CD8b-PEcy7, TCRab-APC, CD103-FITC, and CD56- and CD103 CD8 T cells. For this analysis, expression values PE antibodies at 4C for 45 minutes (Supplementary Table S3). for each sample have been obtained using RSEM (version1.3.0, þ – þ þ – After gating for CD3 CD4 CD8ab TCRab CD56 cells, with Bowtie 2, version 2.2.5, nonstranded and with the single – þ CD103 and CD103 single cells were sorted on a Beckman cell prior activated to account for drop-out genes) and have Coulter Astrios cytometer directly into 4 mL lysis buffer (2 mLof been computed for the Gencode 19 transcriptome annotation 0.2% Triton X-100, Sigma-Aldrich, catalog no. T9284) with 5% for GRCh37 (reference index built with –polyA activated). recombinant RNase inhibitor (Westburg-Clontech, catalog no. Genes with a Benjamini–Hochberg FDR <0.01 and log2 fold 2313A), 1 mLof10mmol/L barcoded oligo dT primer, and 1 mL4 change >1 were selected for further analysis. Differentially 10 mmol/L dNTP mix in 96-well PCR plates. Each well contained expressed genes were visualized in a Volcano plot (DESeq2, a unique indexed Oligo dT primer [custom designed by P. van der version 1.16.1). The accession number for the sequencing data Vlies (UMGC, Groningen, the Netherlands); Supplementary reported in this study is GSE127888.

786 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

ELISA using the Human Chemokine Antibody Array- TILs from three high-grade serous ovarian cancer digests were Membrane, ab169812, Abcam; Supplementary Table S6). In brief, stained and sorted as described for mRNA sequencing. The number –coated membranes were incubated with of sorted T cells for the three patients were 163 103,216 103,and supernatant overnight at 4C. Per condition, 450 mL of supernatant þ 154 103 for CD4 cells; 82 103,38 103,and83 103 for was diluted with 1,050 mL buffer, provided by the manufacturer, þ – CD8 CD103 ;and207 103,120 103,and146 103 for and 1,500 mL was added (R&D Systems), or 1,000 mL was added þ þ CD8 CD103 Tcells.SortedTcellsremainedunstimulatedor undiluted (Abcam). Captured were visualized using were activated either with a stimulation cocktail containing phorbol chemiluminescent detection reagents, provided by the manufac- myristate acetate (PMA, 40.5 mmol/L) and ionomycin (670 mmol/L, turers. Appropriate washing steps using wash buffers provided by 500 dilution, Invitrogen, 00-4970-93) or with Dynabeads the manufacturers were performed in-between incubation steps. (2 mL/1 105 cells, T-activator CD3/CD28 beads, 11131D, Gibco). Membranes were imaged on the Bio-Rad ChemiDoc MP Imaging þ Peripheral blood CD8 T cells were isolated from blood of four System, and densitometric analysis of chemokine spots was per- healthy volunteers (Sanquin, written informed consent was formed using the Protein Array Analyzer plugin for ImageJ (30). obtained) by a Ficoll-Paque gradient followed by magnetic- þ activated cell sorting with a CD8 T-cell–negative selection kit Statistical analyses þ þ (purity >90%, MagniSort Human CD8 Enrichment Kit, Differentially expressed genes in CD103 CD8 versus – þ catalog no. 8804-6812-74; Thermo Fisher Scientific). Peripheral CD103 CD8 T cells sorted from human ovarian tumors were þ blood CD8 T cells were incubated in 100 mL AIM-V medium with determined by DESeq2 for 20-cell populations. Genes with a 5 or without Dynabeads (2 mL/1 10 cells) for activation, recom- Benjamini–Hochberg FDR <0.01 and log2 fold change >1 were binant TGFb1 (rTGFb1, 100 ng/mL, PeproTech), TGFb1 receptor selected for further analysis. Differences in FPKM values of single inhibitor (10 mmol/L, SB431542, Sigma Aldrich/Merck), or a cells were assessed by a Mann–Whitney U test. Differences in þ combination of these. Similar experiments were performed with number of CD20 follicles on FFPE slides of molecular subgroups the addition of IL2 (100 IU/mL, Novartis Pharmaceuticals). For of endometrial cancer were determined by a nonparametric þ the dose–response curve, peripheral blood CD8 T cells from Kruskal–Wallis test, followed by Dunn post hoc analysis. We three healthy donors were incubated with or without Dynabeads analyzed the TCGA mRNA sequencing data and compared differ- (2 mL/1 105 cells) for activation and with recombinant TGFb1at ences in between molecular subgroups of endo- doses ranging from 0 to 100 ng/mL (rTGFb1, PeproTech). All cells metrial cancer with a nonparametric Kruskal–Wallis test and a post were cultured in AIM-V medium with 5% pooled human serum hoc Dunn test. Differences in survival were determined by a log- (catalog no. A25761, One Lambda) in 96-well plates containing test. CXCL13 production was analyzed using a Kruskal– 1 105 cells per condition. After 7 days, plates were centrifuged Wallis comparison with a post hoc Dunn test, or, for the dose– and supernatant was collected for ELISA. response curve, with a two-way ANOVA followed by a post hoc CXCL13 sandwich ELISA experiments were performed accord- Bonferroni test. The chemokine arrays were analyzed using a ing to manufacturer's protocol (human CXCL13/BLC/BCA-1 Kruskal–Wallis test with a post hoc Dunn test. The survival effect DuoSet ELISA DY801, R&D Systems, Abingdon, United King- of CD103 in pMMR uterine cancer was assessed by Kaplan–Meier dom). In brief, Nunc MaxiSorp flat-bottom plates (Invitrogen) analysis and log-rank test by comparing "above median CD103 were coated with a capture antibody, followed by incubation with expression" and "equal to or below median CD103 expression" cell supernatant. Per condition, 70 mL of supernatant was diluted (cutoff 16.14) patient groups. Uni- and multivariate analyses were with 40 mL 1% BSA in PBS, after which 100 mL was added to the performed by disease-specific Cox regression survival analyses well. Binding of CXCL13 was detected using secondary antibody, [Enter for univariate and Backward and Forward (both LR and streptavidin-HRP, and TMB 1-Component Microwell Peroxidase conditional) methods for multivariate analyses]. All statistical Substrate (SureBlue, KPL/SeraCare). Substrate conversion was analyses were performed using R version 3.4.0 or GraphPad Prism stopped after 20 minutes with 0.01 mol/L hydrogen chloride. (GraphPad Software Inc.). A P value of <0.05 was used as a cutoff Plates were washed with PBS plus 0.05% Tween20 in-between for significance. incubations. Optical density values were obtained using a micro- plate reader set to 450 nm (Bio-Rad iMark Microplate reader). Finally, the derived CXCL13 concentrations (pg/mL) were mul- Results þ tiplied by 1.57 to correct for diluting. AIM-V medium only was Epithelial CD8 T cells associate with an activated and used as a negative control. exhausted transcriptional signature We and others have previously shown that intraepithelial þ – þ Chemokine arrays CD103 , but not stromal CD103 , CD8 TILs are promising þ CD8 T cells were isolated from blood of three healthy targets for ICI therapy (25, 29, 31, 32). To understand the donors as described above in the ELISA section. Per condition, underlying transcriptional changes in these two cell populations, 5 105 cells were cultured in AIM-V medium with 5% PHS in a we performed mRNA sequencing on single- and 20-cell pools of þ 24-well plate. Cells were either incubated for 7 days in medium CD8 T cells isolated from human tumors. We chose ovarian alone, with rTGFb1 (100 ng/mL, PeproTech), with Dynabeads cancer as a model tumor because of its large tumor bulk, avail- (2 mL/1 105 cells, T-activator CD3/CD28 beads, 11131D, ability of pretreatment tissue, and a high number of distinct þ – þ þ Gibco), or with both rTGFb1 and Dynabeads. Samples were CD103 and CD103 infiltrating CD8 cells. CD8 TILs were þ þ þ centrifuged, and supernatants were collected to analyze produc- defined on the basis of a CD3 /TCRab /CD8ab /CD56 /CD4 tion of chemokines on chemokine arrays, according to manufac- phenotype (Fig. 1A). Post hoc t-distributed stochastic neighbor þ turer's instructions (31 chemokines using the Proteome Profiler embedding (t-SNE) confirmed the presence of unique CD103 – Human Chemokine Array Kit, ARY017, R&D Systems and 38 and CD103 CTL populations in these tumors (Supplementary

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 787

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

− − A CD3+ TCRαβ+ CD56 CD4 CD8α+ CD8β+ CD56-PE CD3 BV421 TCR αβ APC CD8β PE-Cy7 CD4 PerCP-Cy5.5

CD3 BV421 CD3 BV421 CD8α APC-eFluor780 CD3 BV421 CD103 FITC − + BCCD103 vs. CD103 RNAseq 40 7 CXCL13 PDCD1 LAYN VTCN1 CTLA4 LAYN 6 8 10 **** ITGAE TNFRSF18 ns 30 KIR2DL4 P ) P ) 5 TIGIT 8 HAVCR2 6 GZMB GAGE12H 4 6 20 LAG3 CCL3 4 (TPM+1) (TPM+1) (adjusted (adjusted 3 2 2 4 10 10 GMPR2 GNGT2 Log 2 2 Log 10

− Log 2 − Log GAGE12I 1 0 0 − + − + 0 0 3 03 -40 -20 0 20 40 -20 -10 0 10 20 10 Log (fold change) Log (fold change) CD CD103 CD103 CD1 2 − 2 − CD103+ vs. CD103 CD103+ vs. CD103

D CXCL13 E F Patient 1 Patient 2 Patient 3 ** 12 **** 100

80 8 60 (%) + (TPM+1)

2 + + 40

PD-1 APC PD-1 PD-1 4 PD-1 Log 20

0 0 − − + CD103 FITC +

CD103 CD103 CD103 CD103 H I G PDCD1 CXCL13 CD103+ TILs CD4+ TILs 8 20 25 1,200 1,200 * ** * 20 6 15 * 15 800 800 4 10

(RPKM+1) 10 CXCL13 2 (RPKM+1) (RPKM+1) 2 2

log 5 400 400 2 5 CXCL13 (pg/mL) CXCL13 (pg/mL) Log Log 0 P < 0.05 0 0 0 0 − − + + 02468

PDCD1 PMA PMA CD103 CD103 CD103 CD103 log2 (RPKM+1) Medium Medium

Anti-CD3/CD28 Anti-CD3/CD28

Figure 1. CD103þ CD8þ TILs display a distinct phenotype and gene signature characterized by exhaustion genes and CXCL13 expression. A, Gating strategy of CD103þ and CD103– CD8þ T cells according to TCRab, CD3, CD56, CD4, CD8a, CD8b, and CD103 expression. One exemplary ovarian tumor is depicted. B, Volcano plot of up- or downregulated genes in CD103þ and CD103– TILs as determined by RNA sequencing, annotated by a set of exhaustion and chemokine-related genes.

Significance was determined as Benjamini–Hochberg FDR <0.01 and log2 fold-change >1. Expression of exhaustion-related genes (C) PDCD1 and LAYN and (D) þ – chemokine CXCL13 in the CD103 and CD103 subsets (n ¼ 20). The lines indicate median Log2 (TPMþ1) values IQR. E, Cell surface expression of CD103 and PD-1 in gated CD8þ TILs from exemplary ovarian tumor digests (n ¼ 3) determined by flow cytometry. The red line represents threshold for PD-1 positivity based on isotype control. F, Cell surface expression of PD-1 on CD103þ and CD103– gated CD8þ TIL subsets determined by flow cytometry (n ¼ 8). Graphs indicate – þ median and IQR. G, Expression of PDCD1 and CXCL13 in single CD103 (n ¼ 12) and CD103 TILs (n ¼ 12). Lines indicate median Log2 (FPKMþ1) IQR. Significance in F and G was determined as Mann–Whitney U test (, P < 0.05; , P < 0.01). H, Correlation of PDCD1 and CXCL13 expression in CD103þ TILs (n ¼ 24). I, Secretion of CXCL13 by CD103þ TILs and control CD4þ TILs after stimulation overnight at 37C using anti-CD3/CD28 T-cell activation beads (2 mL Dynabeads per 1 105 cells) or PMA/ionomycin (500 dilution according to manufacturer's instructions), determined by ELISA Mean plus SEM of TILs from ovarian tumors (n ¼ 3) are depicted. Significance was determined by Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.05; ns, not significant).

788 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

þ Fig. S1A–S1C). The transcriptome of CD103 CTLs was charac- receptor 1 (TGFbR1) signaling (25, 38). TGFb is a reported terized by an activation and exhaustion signature (Supplementary inducer of exhaustion-related genes such as PD-1 in T cells (39), Table S7) with significant upregulation of GZMB (granzyme B), which is also in-line with the transcriptional and phenotypical þ þ HAVCR2 (T-cell immunoglobulin and mucin domain 3, TIM3), profile that we obtained for CD103 CD8 TILs. Therefore, we þ LAG3 (-activation gene 3), TNFRSF18 (glucocorticoid- hypothesized that TGFb might stimulate CD8 T cells to secrete þ induced TNFR-related protein, GITR), KIR2DL4 (killer cell immu- CXCL13. To investigate this, we activated peripheral blood CD8 noglobulin-like receptor 2DL4), TIGIT (T-cell immunoreceptor T cells from healthy donors with anti-CD3/anti-CD28–conjugated with Ig and ITIM domain), and CTLA4 (CTL attenuator 4) in the beads in the presence or absence of recombinant TGFb1 (rTGFb1) þ þ 20-cell pools (Fig. 1B). CD103 CTLs expressed GNGT2 (G protein and measured secretion of CXCL13. Resting CD8 T cells did subunit gamma transducin 2), encoding a G protein gamma not produce or secrete CXCL13 (Fig. 2A), nor did they express family member expressed in lymph nodes and that is CD103 at the cell surface. Activation using anti-CD3/anti-CD28– involved in GTPase activity (Fig. 1B). The expression of these conjugated beads induced minimal secretion of CXCL13 (Fig. 2A) markers is in-line with our earlier work demonstrating that the and a minor upregulation of CD103 (Fig. 2B). Secretion of þ intraepithelial CD103 T cells likely represent CTLs that have CXCL13 and expression of CD103 was inhibited by coincubation undergone activation and/or exhaustion (25, 29). In contrast, with a TGFbR1 inhibitor, suggesting autocrine TGFb signaling was – CD103 CTLs displayed a more quiescent phenotype with a high required for this induction of CXCL13 (Fig. 2A and B). Accord- þ differential expression of the V-set domain–containing T-cell acti- ingly, activation of CD8 T cells in the presence of rTGFb1 induced vation inhibitor 1 (VTCN1), a known suppressor of T-cell function significant CXCL13 secretion (Fig. 2A) and expression of CD103 (Fig. 1B). These cells differentially expressed GAGE12H, GAGE12I, on the CD8 T-cell surface (Fig. 2B). Again, induction of CXCL13 and GMPR2 (guanosine monophosphate reductase 2), involved in and CD103 were inhibited by coincubation with a TGFbR1 þ cell energy metabolism (Fig. 1B). Finally, CD103 , but not inhibitor (Fig. 2A and B). Induction of CXCL13 by TGFb was – CD103 , cells were characterized by expression of genes previously dose-dependent and induced CXCL13 secretion at 0.1 ng/mL þ associated (33) with exhausted CD8 T cells (Fig. 1C). rTGFb1 with a peak at 10 ng/mL rTGFb1 (Fig. 2C). Because IL2 þ inhibits the secretion of CXCL13 in follicular helper CD4 T þ CD103 CTLs differentially express the B-cell recruiting cells (40), we also examined whether IL2 impacted CXCL13 þ þ chemokine CXCL13 secretion by CD8 T cells. In contrast to CD4 T cells, induction þ In addition to the activated and exhausted gene signature, of CXCL13 in CD8 T cells was not inhibited by IL2 (Fig. 2D), þ CD103 CTLs were also characterized by significantly upregulated suggesting underlying differences in CXCL13 gene regulation and/ þ þ expression of the TLS-inducing CXCL13 (Fig. 1B and D; P < or IL2 signaling between CD8 and CD4 cells. On the basis of our þ 0.0001). Although traditionally considered a CD4 follicular findings, we concluded that TGFb was sufficient for CXCL13 þ helper T-cell (TFH) gene, CXCL13 is also expressed in subsets of induction in activated CD8 T cells. þ CD8 TILs from hepatocellular carcinoma, , breast, Next, we determined whether TGFb also modulates secretion and non–small cell (34–37). In the latter, CXCL13 was of other chemokines. Analysis of chemokine mRNA expression þ þ – identified in a transcriptionally unique PD-1–high (PD-1T) CD8 in CD103 versus CD103 TILs revealed a significant upregula- T-cell population that predicted response to ICI therapy (34). We, tion of CCL3, CCL5, and CXCL13 and a trend toward over- þ therefore, determined whether ovarian tumor-infiltrating expression of CCL3L1, CCL4L2, CCL20, and CXCL9 in CD103 þ þ CD103 and PD-1T CD8 populations overlapped phenotypi- TILs (Fig. 2E). Subsequent analysis of 47 chemokines secreted þ þ cally. PDCD1 was differentially expressed in CD103 over from peripheral blood CD8 T cells revealed activation-depen- – CD103 cells on the mRNA level, although this did not dent production of CCL4, CCL5, CXCL9, and CXCL10 (Fig. 2F reach statistical significance in the 20-cell pools (Fig. 1B and and G; Supplementary Fig. S2A–S2C). As before, significantly C). Nevertheless, at the protein level, PD-1 was expressed almost higher CXCL13 was secreted upon activation of T cells in the þ þ exclusively on the cell surface of CD103 CD8 T cells (Fig. 1E and presence of rTGFb1 (Fig. 2H and I). In contrast, rTGFb1didnot F), although considerable heterogeneity existed between patients affect the induction of other chemokines (Fig. 2I). Taken with regards to PD-1 expression (Fig. 1E). Accordingly, analysis of together, our data indicated that TGFb was a specificinducer þ – þ þ the CD103 and CD103 CD8 single-cell mRNA sequencing of CXCL13 in CD8 Tcellsandidentified a hallmark chemo- þ þ data revealed that PDCD1 was heterogeneously expressed in kine pattern for CD103 CD8 TILs. þ – CD103 cells but absent in CD103 cells (Fig. 1G). In contrast, þ þ CXCL13 was homogenously expressed in almost all CXCL13 CD103 CTLs associated with a high mutation load, þ þ CD103 CD8 T cells (Fig. 1G). In line with the above, CXCL13 B-cell infiltration, and TLSs and PDCD1 transcripts were significantly, although poorly, corre- CXCL13 is a key driver of B-cell recruitment and TLS formation þ lated (Fig. 1H). Finally, we assessed whether CD103 TILs secreted in cancer and autoimmune diseases (16, 41–44). As such, we þ þ þ CXCL13 protein, using CD4 TILs obtained by flow cytometry– speculated the CXCL13 CD103 TIL population would be þ þ þ based sorting as controls. CD103 CD8 TILs and CD4 TILs involved in recruitment of B cells and TLS formation across readily secreted CXCL13 upon ex vivo activation with anti-CD3/ human tumors. We retrospectively analyzed a cohort of 125 þ þ anti-CD28-conjugated beads or PMA/ionomycin (Fig. 1I). (high-grade serous) ovarian tumors for CD103 cells and CD20 þ þ B cells using a tissue microarray. CD103 and CD20 cells were þ þ TGFb primes cytotoxic CD8 T cells to secrete CXCL13 in vitro detected in most patients, with some CD20 cells forming aggre- þ We next sought to define the molecular mechanism underlying gates in close proximity to CD103 cells in the tumor epithelium þ þ production of CXCL13 by CD103 CD8 TILs. Previously, we and (Fig. 3A). Indeed, division of patients by high (>26.3), interme- þ þ others have demonstrated that induction of CD103 on CD8 T diate (8.6-26.3), or low (<8.6) CD103 cell infiltration revealed a cells is dependent on concurrent T-cell receptor (TCR) and TGFb significant association with the number of B cells (Fig. 3B). Next,

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 789

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

A Sorted peripheral blood CD8+ T cells B Sorted peripheral blood CD8+ T cells 750 * * Anti-CD3/CD28 Anti-CD3/CD28 Anti-CD3/CD28 * Anti-CD3/CD28 +SB431542 +TGFβ +TGFβ+SB431542

500

250 CXCL13 (pg/mL) CD3 PE

0 Anti-CD3/CD28 - - - + + + + TGFβ - - + - - + + CD103 FITC SB431542 -+-- + - +

+ C Sorted peripheral D Sorted peripheral blood CD8+ T cells + IL2 E RNA Expression tumor-infiltrating CD8 T cells + 1,200 blood CD8 T cells 1,200 ** 8 CD103+ Resting ****

**** - 6 Anti-CD3/CD28 4 **** * 800 800 * **** 2 0 vs. CD103 + -2 (fold change)

400 400 2 * -4 CXCL13 (pg/mL) CXCL13 (pg/mL) Log CD103 -6 CD103- 0 0 -8 Anti-CD3/CD28 - + - + + 1 3 7 6 0 2 8 2 0 6 7 1 0 -4 -3 -2 -1 0 1 2 L2 L L4 L5 1 2 L 13 1 1 L C CL3 C 4L2 L L2 L2 L C L C TGFβ - - - + + C C L3L C L4L1L CC CCLC X CL CL C CL3 C C C CC CC CC X X X3 Log10 ng/mL (TGFβ) C C C C C CXCL9 SB431542 --+- + CXCL1CXCC C C FG + 6 **** Protein secretion by activated peripheral blood CD8 T cells Anti-CD3/ Anti-CD3/ Resting CD28 Resting CD28 4 ******** CCL3 CXCL9 * CCL4 CXCL10 2 CCL5 CXCL13

(fold change) 0 CCL17 2 Anti-CD3/ CCL20 Resting

Log CD28 -2 POS

anti-CD3/CD28 vs. resting CCL22

2 5 8 3 6 7 4 6 7 8 ) 1 5 6 7 8 9 a b 3 6 1 CCL24 NEG L L4 L7 1 1 20 22 2 2 2 -3 L L L 2 1 L L1 C CL C L L1 L L L23 L2 L25 1 C CL C CL L10 L11 1 C C CCL1CC CCL3C C C CCL C C C C C CL CL CL X X C C L12 L CL X CCL11C CCL15CC CCLCCL18CCL19C CC C C C C C C CL CXCLCXCCX C CX C X C X X3 X C CX XC X C CXCL1C C C C

RO ( H G + I 6 **** Protein secretion by activated peripheral blood CD8 T cells + TGFβ 3 * **** **** CXCL13 4 * **** **** 2 **** ** Resting 2 1 TGFβ (fold change) 0 (fold change)

2 Anti-CD3/ 2 0 CD28 vs. anti-CD3/CD28 Log Log

-2 anti-CD3/CD28+TGFβ +TGFβ -1 ) anti-CD3/CD28+TGFβ vs. resting 2 3 5 6 7 9 2 4 5 6 8 5 6 7 8 9 0 1 a b 6 1 L L L7 L8 1 20 23 2 2 2 27 3 L L L L 1 1 2 L1 L 4 5 0 2 0 3 CL1C CL C C C L13 L1 L18 L L2 L L L 1- C C C CL L L 12 1 L1 C C L3 2 2 L9 1 1 CL11 C C C C C CL CL CL2L C L C 3 C CL CL L17 L C L L C C C CCL4C C C XCL1 C CL X C C C CL C C C CC CCL15CC CCL1C CCLC C C C C CC C C C CX CXCCX CX CX X XCL13X X CC C C CX XC X CXCC X C C C C C (CXC C CX

GRO

Figure 2. TGFb-dependent induction of CXCL13 in CD8þ T cells and CD103þ TIL chemokine profile. A, Secretion of CXCL13 by sorted peripheral blood CD8þ T cells after stimulation for 7 days using anti-CD3/CD28 T cell activation beads (2 mL per 1 105 cells), recombinant TGFb1 (100 ng/mL), and/or TGFb1 receptor inhibitor SB431542 (10 mmol/L), determined by ELISA. Depicted are mean plus SEM of CD8þ T cells from 4 donors. Significance was determined using Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.05). B, Cell surface expression of CD103 on sorted CD8þ T cells after stimulation using anti-CD3/CD28 T-cell activation beads, recombinant TGFb1, and/or TGFb1 receptor inhibitor SB431542 as described for A. One exemplary donor is depicted. C, Secretion of CXCL13 as determined by ELISA of CD8þ T cells after stimulation using anti-CD3/CD28 T-cell activation beads as described for A and/or varying concentrations of recombinant TGFb1 as depicted on the x-axis. Mean plus SD from 3 donors is depicted. Significance was determined by a two-way ANOVA followed by a post hoc Bonferroni test (, P < 0.05; , P < 0.0001). D, Secretion of CXCL13 by sorted peripheral blood CD8þ T cells after stimulation using anti-CD3/CD28 T-cell activation beads, recombinant TGFb1, and/or TGFb1 receptor inhibitor SB431542 (as described for A) in the presence of IL2 (100 IU/mL), determined by ELISA. Depicted are mean plus SEM of CD8þ T cells from 4 donors. Significance was determined using Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.05). E, Up- or downregulated chemokines between CD103þ and CD103– TILs determined by differential expression of RNA sequencing of 20-cell pools (n ¼ 20). Dashed red line indicates 2-fold change; dashed blue line indicates 0.5-fold change. Error bars indicate fold change SE. Significance was determined using Benjamini–Hochberg FDR. F, Secretion of chemokines by CD8þ T cells after 7 days stimulation using anti-CD3/CD28 T-cell activation beads (2 mL per 1 105 þ cells) determined by a chemokine array kit. n ¼ 3. Shown are log2 fold changes of the mean compared with resting unstimulated CD8 T cells. Dashed red line indicates 2-fold change; dashed blue line indicates 0.5-fold change. Error bars indicate SEM. G, Exemplary chemokine array data from anti-CD3/CD28- activated CD8þ T cells of one donor depicting chemokines with 2-fold and 0.5-fold change compared with resting CD8þ T cells from F. H and I, Secretion of þ chemokines by CD8 T cells after stimulation using anti-CD3/CD28 T-cell activation beads and recombinant TGFb1(n ¼ 4). Shown are log2 fold changes of the mean compared with resting unstimulated CD8þ T cells (H) or to anti-CD3/CD28–activated CD8þ T cells (I). Dashed red line indicates 2-fold change; dashed blue line indicates 0.5-fold change. Error bars indicate SEM. Significance in F–I was determine using Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.05; , P < 0.01; , P < 0.0001).

790 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

A Ovarian tissue microarray B Ovarian tumors *** CD103 CD20 CD20 CD103 300 * 2 200

100 B Cells per mm

0 t w n lo i gh 3 hi 10 03 103 D 1 C D CD C C POLE-EDM/MSI D UC TCGA POLE-EDM UC Tumor sections UC Tumor sections MSS MSI 100 50 *** 33/36 ** CD20 CCL2 CCL3 CCL4 CCL5 80 40 CCL8 28/38 CCL17 CCL18 CCL19 CCL20 CCL21 60 CCL22 30 CCR2 CCR4 20/42 CCR5 CCR7 CD3E CD4 40 20 CD5 CD8A CD19 CD28 CD38 CD40 20 10 CD40LG # of B-Cell structures CD62L B-Cell structures (%) CD68 CD80 CD86 CD200 0 0 CSF2 CTLA4 CXCL9 MSS MSI POLE- CXCL10 MSS MSI POLE- CXCL11 CXCL13 EDM CXCR3 EDM FasLG FBLN7 GNLY HLADRA ICAM3 F ICOS IFNG IGSF6 CD11c PNAd CD20 PNAd CD20 CD11c IL2 IL1R1 IL1R2 IL2RA IL10 IL12B IL15 IL16 IL18 IRF4 ITGAL ITGAD ITGA4 LTA PDCD1 PRF1 SDC1 SGPP2 SH2D1A STAT5A section 1 TBX21 TIGIT TNF CD38 CD79A CD138 CD79A DNA CD38 CD138 TRAF6 VCAM1 Relative expression -4 2 40-2

E MSS MSI POLE-EDM

CD3 CD8 CD103 CD8 DNA CD3 CD103

Figure 3. B cells, tertiary lymphoid structures, and correlation to CD103þCD8þ T cells in human tumors. A and B, IHC analysis of CD103þ and CD20þ cells in ovarian cancer. Exemplary image of a CD20þ aggregate in close proximity to CD103þ cells in the tumor epithelium (A) and B-cell infiltration in CD103-low, CD103-intermediate, and CD103-high tumors stratified by tertile (n ¼ 125; B). Significance was determined using Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.05; , P < 0.001). C, Gene expression of B cell, tertiary lymphoid structure, , CD8þ T cell, and CD4þ follicular helper T-cell–associated genes in TCGA data of uterine cancer (UCEC, n ¼ 546). Tumors were ranked according to molecular subtype and then by CD20 expression. D and E, Prevalence and number of B-cell aggregates (D) in MSS, MSI, and POLE-EDM uterine cancers. E, One exemplary image of each subtype is depicted. Significance was determine using Kruskal–Wallis comparison with a post hoc Dunn test (, P < 0.01; , P < 0.001). F, Immunofluorescent analysis of CD20 aggregates in uterine cancer. CD20 aggregate expression of characteristic TLS markers CD11c, PNAD, CD20, CD38, CD79A, CD138, CD3, and CD8, one exemplary image is depicted.

þ þ we assessed whether CD103 CD8 T cells might also link neoan- MSI tumors compared with MSS tumors (24). In-line with the tigen-specific T-cell responses to B-cell–driven immune responses. above, MSS tumors mostly lacked B-cell– and TLS-related genes, To address this, we determined whether tumors with a high whereas MSI and POLE-EDM tumors were enriched for these þ neoantigen load and concomitantly high CD103 TIL infiltrate genes (Fig. 3C). To confirm these findings, we further analyzed were enriched for B-cell– and TLS-associated genes in mRNA an independent cohort of MSS, MSI, and POLE-EDM tumors from þ sequencing data from The Cancer Genome Atlas (TCGA). Specif- patients with uterine cancer for the presence of CD20 B cells by ically, we analyzed uterine cancer samples stratified by neoantigen IHC. B cells were predominantly observed in large aggregations in load. In brief, four distinct molecular subtypes can be distin- the tumor and surrounding stroma (Fig. 3D and E). In-line with guished in uterine cancer: microsatellite stable (MSS), microsat- the TCGA data, only 48% (20/42) of MSS tumors were found ellite unstable (MSI), polymerase epsilon exonuclease domain to have B-cell aggregates, whereas 74% (28/38) of MSI and 92% mutated (POLE-EDM) tumors, and -mutant tumors. We have (33/36) of POLE-EDM tumors contained B-cell aggregates previously demonstrated an increased number of mutations, (Fig. 3D). Quantification per tumor revealed a significant increase þ predicted neoantigens, and (CD103 ) T cells in POLE-EDM and in the number of B-cell aggregates when comparing MSS to

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 791

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

þ þ POLE-EDM and MSI to POLE-EDM tumors (Fig. 3D, P < 0.001 and CXCL13 CD103 CD8 cells correlate to improved survival P < 0.01, respectively). To confirm that the observed B-cell irrespective of neoantigen burden þ þ aggregates were phenotypically similar to TLSs, we performed To assess whether CXCL13 CD103 CTLs were also correlated multicolor immunofluorescence. Indeed, B-cell aggregates showed with improved survival, we analyzed clinical outcomes in two the typical characteristics of lymphoid tissues, as determined by cohorts of patients with uterine cancer. We focused on tumors the presence of (HEV), germinal B-cell with a low neoantigen burden because a high neoantigen load is centers, and DCs surrounded by a rim of T cells (Fig. 3F). associated with better survival. First, we analyzed TCGA endo- On the basis of the above findings, we speculated that metrial cancers of the MSS subtype and correlated high CXCL13 þ þ CXCL13 CD103 CTL genes would associate with TLS genes and CD103 (ITGAE) gene expression to survival. CXCL13high across human epithelial tumors. To assess this, we analyzed TCGA ITGAEhigh MSS tumors had a significantly improved survival as mRNA expression data of ovarian, uterine, lung, and breast cancer compared with CXCL13lowITGAElow MSS tumors from TCGA þ þ using the differentially expressed genes from CXCL13 CD103 (Supplementary Fig. S3A). To confirm this survival benefit for – – þ þ and CXCL13 CD103 CTL cells identified by our mRNA sequenc- CXCL13 CD103 CTLs in neoantigen low tumors, we analyzed þ þ ing. TLS genes correlated with CXCL13 CD103 , but not FFPE tumor tissue of an independent cohort of pMMR uterine – – CXCL13 CD103 , CTL genes across all four tumor types (Fig. 4). cancers by IHC (Supplementary Table S8). We were unable to þ þ þ Taken together, our data suggested that CXCL13 CD103 CTLs detect CXCL13 protein expression in CD103 CTLs, consistent þ promote migration of B cells to tumors and the formation of TLSs with our previous finding that almost all CD103 cells express across tumor types. CXCL13 on the mRNA level (Fig. 1B, D, and G), but express and

Ovarian cancer Uterine cancer G1 T1 T1 A Y T T5A RVELD2 O7A T86 T8 T86 A3 VCR2 TF G3 A AD2B A A Y TA3 AVCR2 T A RT7 R AYN A AKMIP1 T CCL2 CD80 CD86 CCL22 CCL2 CD86 GNL GNLY CCL22 CCL3 CCL4 CCL5 CCL8 CCL3 CCL4 CCL5 CCL8 CD3E CD40LG CD40 CD80 CXCL10 CCR2 CD200 CD28 CD38 CD3E CD40LG CD40 CD68 CD8A CTLA4 CXCL10 CXCL11 CXCL13 CXCR3 FBLN7 ICAM3 ICOS IFNG IGSF6 IL15 IL16 IL18 MADCAM1 MS4A1 CCL17 CCL18 CCR4 CCL17 CCL18 CD8A CTLA4 CXCL11 CXCL13 ICAM3 ICOS IFNG IGSF6 IL12B IL18 CCR4 CCR5 CD4 CD5 IL12B CCL19 CCL20 CCL21 CCR2 CCR5 CCR7 CD19 CD200 CD28 CD4 CD5 CD68 CCL19 CCL21 CXCL9 CXCR3 IL10 IL15 TNF CCR7 CD19 CXCL9 IL10 IL1R1 IL1R2 IL2RA IRF4 ITGAL LTA PDCD1 PRF1 SDC1 SGPP2 SH2D1A STAT5A TIGIT TNF CD38 CCL20 FBLN7 IL16 IL1R1 IL1R2 IL2RA IRF4 ITGA4 ITGAD ITGAL L MADCAM1 MS4A1 PDCD1 PRF1 SDC1 SGPP2 SH2D1A S TIGIT ITGA4 ITGAD TBX21 VCAM1 APOBEC3G BATF CCDC82 CD79B CPNE7 FGD3 GALNT2 GNGT2 GZMB GZMH HA TBX21 TRAF6 VCAM1 APOBEC3G AT B CPNE7 CPT1B CTLA4 CXCL13 DNMT1 FGD3 GALNT2 GNGT2 H L TRAF6 CPT1B CTLA4 CXCL13 DNMT1 CCDC82 CD79B GZMB GZMH HLA−DQA1 JAKMIP1 LT PRMT7 CCDC104 LAYN ATAD2B HIP1 HLA−DQA1 HOPX ITGAE J HIP1 HOPX ITGAE M SPG7 CDK5RAP2 DLA LIG1 TIGIT TNFRSF18 TRAF5 ABCB7 ANXA3 CDH1 EPHA4 EXT2 FAM21A FLNB PRMT7 SPG7 TIGIT TNFRSF18 TRAF5 ABCB7 ALDH7A1 ANXA3 CCDC104 CDH1 CDK5RAP2 CDKN2D COBLL1 CYB5R2 CDKN2D COBLL1 CYB5R2 DLAT MARVELD2 EXT2 FAM21A FLNB MA KIR2DL4 KIR2DL4 KR LAG3 ALDH7A1 ARHGAP29 BAIAP2 LIG1 LTA MYO7A ARHGAP29 BAIAP2 EPHA4 QPRT K L GMPR2 GTSF1 USP53 DNAJB4 ELF3 GMPR2 JUP KRT7 KR LAMB1 DSG2 M MXI1 OCLN SETD5 SETDB1 SHF SLC25A4 SORBS3 SVIL TDG TPD52 TRIM2 ULK4 USP53 VTCN1 ZNF33B IL7R JUP K NKRF NTHL1 OCLN POLR3H QPRT SLC25A4 SORBS3 SUV420H1 SVIL TPD52 ZBTB10 DSG2 GLO1 LPGA DNAJB4 ELF3 MEG3 NTHL1 POLR3H SUV420H1 TPRG1L ZBTB10 KRT8 LAMB1 LPG MEG3 MT PRKAG1 SETD5 TDG TPRG1L ULK4 ZNF33B GTSF1 IL7R NKRF PRKA SCNN1A SEPHS2 GLO1 MXI1 SCNN1A SEPHS2 SETDB1 SHF TRIM2 VTCN1

CCL17 CCL17 CCL18 CCL18 CCL19 CCL19 CCL20 CCL20 CCL21 CCL21 CCL22 CCL22 CCL2 TLS CCL2 TLS CCL3 CCL3 CCL4 CCL4 CCL5 CCL5 CCL8 CCL8 CCR2 CCR2 CCR4 + + CCR4 + + CCR5 CCR5 CCR7 CD103 CD8 CCR7 CD103 CD8 CD19 CD19 CD200 CD200 CD28 CD28 CD38 CD38 CD3E CD3E CD40LG - + CD40LG - + CD40 CD40 CD4 CD103 CD8 CD4 CD103 CD8 CD5 CD5 CD68 CD68 CD80 CD80 CD86 CD86 CD8A CD8A CTLA4 CTLA4 CXCL10 CXCL10 CXCL11 CXCL11 CXCL13 CXCL13 CXCL9 CXCL9 CXCR3 CXCR3 FBLN7 FBLN7 GNLY GNLY ICAM3 ICAM3 ICOS ICOS IFNG IFNG IGSF6 IGSF6 IL10 IL10 IL12B IL12B IL15 IL15 IL16 IL16 IL18 IL18 IL1R1 IL1R1 IL1R2 IL1R2 IL2RA IL2RA IRF4 IRF4 ITGA4 ITGA4 ITGAD ITGAD ITGAL 1 ITGAL 1 LTA LTA MADCAM1 MADCAM1 MS4A1 MS4A1 PDCD1 PDCD1 PRF1 PRF1 SDC1 SDC1 SGPP2 SGPP2 SH2D1A SH2D1A STAT5A STAT5A TBX21 TBX21 TIGIT TIGIT TNF TNF TRAF6 TRAF6 VCAM1 VCAM1 APOBEC3G APOBEC3G ATAD2B ATAD2B BATF BATF CCDC82 CCDC82 CD79B CD79B CPNE7 CPNE7 CPT1B CPT1B CTLA4 CTLA4 CXCL13 CXCL13 DNMT1 DNMT1 FGD3 FGD3 GALNT2 GALNT2 GNGT2 GNGT2 GZMB GZMB GZMH GZMH HAVCR2 HAVCR2 HIP1 HIP1 HLA−DQA1 HLA−DQA1 HOPX HOPX

ITGAE Correlation ITGAE Correlation JAKMIP1 JAKMIP1 KIR2DL4 KIR2DL4 KRT86 KRT86 LAG3 LAG3 LAYN LAYN LIG1 LIG1 LTA LTA MYO7A MYO7A PRMT7 PRMT7 SPG7 SPG7 TIGIT TIGIT TNFRSF18 TNFRSF18 TRAF5 TRAF5 ABCB7 ABCB7 ALDH7A1 ALDH7A1 ANXA3 0 ANXA3 0 ARHGAP29 ARHGAP29 BAIAP2 BAIAP2 CCDC104 CCDC104 CDH1 CDH1 CDK5RAP2 CDK5RAP2 CDKN2D CDKN2D COBLL1 COBLL1 CYB5R2 CYB5R2 DLAT DLAT DNAJB4 DNAJB4 DSG2 DSG2 ELF3 ELF3 EPHA4 EPHA4 EXT2 EXT2 FAM21A FAM21A FLNB FLNB GLO1 GLO1 GMPR2 GMPR2 GTSF1 GTSF1 IL7R IL7R JUP JUP KRT7 KRT7 KRT8 KRT8 LAMB1 LAMB1 LPGAT1 LPGAT1 MARVELD2 MARVELD2 MEG3 MEG3 MTA3 MTA3 MXI1 MXI1 NKRF NKRF NTHL1 NTHL1 OCLN OCLN POLR3H POLR3H PRKAG1 PRKAG1 QPRT QPRT SCNN1A SCNN1A SEPHS2 SEPHS2 SETD5 SETD5 SETDB1 SETDB1 SHF SHF SLC25A4 SLC25A4 SORBS3 SORBS3 SUV420H1 SUV420H1 SVIL SVIL TDG TDG TPD52 TPD52 TPRG1L TPRG1L TRIM2 TRIM2 ULK4 ULK4 USP53 -1 USP53 -1 VTCN1 VTCN1 ZBTB10 ZBTB10 ZNF33B ZNF33B

Lung cancer Breast cancer G1 A Y T T5A LY RT O7A T86 T7 A3 VCR2 AT TF G3 AT5A A AD2B A A YO7A A T A RT7 AKMIP1 TA CCL2 GN CXCR3 FBLN7 GNL MADCAM1 MS4A1 CCL22 CCL3 CCL4 CCL5 CCL8 CD80 CD86 CCL22 CCL2 CCL3 CCL4 CCL5 CCL8 CD68 CD80 CD86 ICAM3 ICOS IFNG IGSF6 IL10 IL12B IL15 IL16 IL18 IL2RA IRF4 ITGA4 SDC1 SGPP2 SH2D1A ICAM3 ICOS IFNG IGSF6 IL15 IL16 IL18 IL2RA IRF4 CCL17 CCL18 CCL19 CCL20 CCL21 CCR2 CCR4 CCR5 CCR7 CD19 CD200 CD28 CD38 CD40LG CD40 CD4 CD5 CD68 CD8A CTLA4 CXCL10 CXCL11 CXCL13 CXCL9 CXCR3 CCL17 CCL18 CCL19 CCL21 CCR2 CCR4 CCR5 CD200 CD28 CD38 CD3E CD40LG CD40 CD4 CD5 CD8A CTLA4 CXCL10 CXCL11 CXCL13 SDC1 SGPP2 SH2D1A ST IL1R1 IL1R2 ITGAD ITGAL LTA MADCAM1 MS4A1 PDCD1 PRF1 S IL10 IL12B IL1R1 IL1R2 ITGA4 ITGAD ITGAL LT CD3E FBLN7 TBX21 TIGIT TNF CPNE7 CPT1B CTLA4 CXCL13 CCL20 CCR7 CD19 CXCL9 PDCD1 PRF1 TIGIT TNF TRAF6 VCAM1 APOBEC3G AT B DNMT1 FGD3 GALNT2 GNGT2 VCAM1 APOBEC3G BATF CCDC82 CD79B CPNE7 CPT1B CTLA4 CXCL13 DNMT1 FGD3 GALNT2 GZMB GZMH H TBX21 CCDC82 CD79B GZMB GZMH HAVCR2 HIP1 HLA−DQA1 HOPX ITGAE J TRAF6 ATAD2B GNGT2 HIP1 HLA−DQA1 HOPX ITGAE JAKMIP1 LAYN PRMT7 TRAF5 LAYN TRAF5 ALDH7A1 DLA KIR2DL4 KR LAG3 LIG1 L M SPG7 TIGIT TNFRSF18 ABCB7 KIR2DL4 KRT86 LA LIG1 LT MY ZBTB10 ALDH7A1 ANXA3 ARHGAP29 BAIAP2 CCDC104 CDH1 CDK5RAP2 CDKN2D COBLL1 CYB5R2 DL EPHA4 EXT2 FAM21A FLNB JUP MARVELD2 PRMT7 SPG7 TIGIT TNFRSF18 ABCB7 ANXA3 ARHGAP29 BAIAP2 CCDC104 CDH1 CDK5RAP2 CDKN2D COBLL1 CYB5R2 ELF3 EPHA4 EXT2 FAM21A FLNB LAMB1 MARVELD2 TDG ZNF33B QP SETD5 SLC25A4 SORBS3 SUV420H1 SVIL TDG TPD52 TPRG1L TRIM2 ZNF33B DNAJB4 DSG2 ELF3 GLO1 GMPR2 GTSF1 IL7R KR KRT8 LAMB1 LPGAT1 MEG3 NKRF NTHL1 OCLN POLR3H GLO1 GMPR2 KRT8 LPGAT1 MEG3 MTA3 MXI1 OCLN QPRT SETD5 SETDB1 SHF SLC25A4 SORBS3 TPD52 TPRG1L TRIM2 ULK4 USP53 VTCN1 ZBTB10 SCNN1A SEPHS2 SETDB1 SHF ULK4 USP53 VTCN1 MT MXI1 PRK DNAJB4 DSG2 GTSF1 IL7R JUP K NKRF NTHL1 POLR3H PRKAG1 SCNN1A SEPHS2 SUV420H1 SVIL

CCL17 CCL17 CCL18 CCL18 CCL19 CCL19 CCL20 CCL20 CCL21 CCL21 CCL22 CCL22 CCL2 TLS CCL2 TLS CCL3 CCL3 CCL4 CCL4 CCL5 CCL5 CCL8 CCL8 CCR2 CCR2 CCR4 + + CCR4 + + CCR5 CCR5 CCR7 CD103 CD8 CCR7 CD103 CD8 CD19 CD19 CD200 CD200 CD28 CD28 CD38 CD38 CD3E CD3E CD40LG - + CD40LG - + CD40 CD40 CD4 CD103 CD8 CD4 CD103 CD8 CD5 CD5 CD68 CD68 CD80 CD80 CD86 CD86 CD8A CD8A CTLA4 CTLA4 CXCL10 CXCL10 CXCL11 CXCL11 CXCL13 CXCL13 CXCL9 CXCL9 CXCR3 CXCR3 FBLN7 FBLN7 GNLY GNLY ICAM3 ICAM3 ICOS ICOS IFNG IFNG IGSF6 IGSF6 IL10 IL10 IL12B IL12B IL15 IL15 IL16 IL16 IL18 IL18 IL1R1 IL1R1 IL1R2 IL1R2 IL2RA IL2RA IRF4 IRF4 ITGA4 ITGA4 ITGAD ITGAD ITGAL 1 ITGAL 1 LTA LTA MADCAM1 MADCAM1 MS4A1 MS4A1 PDCD1 PDCD1 PRF1 PRF1 SDC1 SDC1 SGPP2 SGPP2 SH2D1A SH2D1A STAT5A STAT5A TBX21 TBX21 TIGIT TIGIT TNF TNF TRAF6 TRAF6 VCAM1 VCAM1 APOBEC3G APOBEC3G ATAD2B ATAD2B BATF BATF CCDC82 CCDC82 CD79B CD79B CPNE7 CPNE7 CPT1B CPT1B CTLA4 CTLA4 CXCL13 CXCL13 DNMT1 DNMT1 FGD3 FGD3 GALNT2 GALNT2 GNGT2 GNGT2 GZMB GZMB GZMH GZMH HAVCR2 HAVCR2 HIP1 HIP1 HLA−DQA1 HLA−DQA1 HOPX HOPX

ITGAE Correlation ITGAE Correlation JAKMIP1 JAKMIP1 KIR2DL4 KIR2DL4 KRT86 KRT86 LAG3 LAG3 LAYN LAYN LIG1 LIG1 LTA LTA MYO7A MYO7A PRMT7 PRMT7 SPG7 SPG7 TIGIT TIGIT TNFRSF18 TNFRSF18 TRAF5 TRAF5 ABCB7 ABCB7 ALDH7A1 ALDH7A1 ANXA3 0 ANXA3 0 ARHGAP29 ARHGAP29 BAIAP2 BAIAP2 CCDC104 CCDC104 CDH1 CDH1 CDK5RAP2 CDK5RAP2 CDKN2D CDKN2D COBLL1 COBLL1 CYB5R2 CYB5R2 DLAT DLAT DNAJB4 DNAJB4 DSG2 DSG2 ELF3 ELF3 EPHA4 EPHA4 EXT2 EXT2 FAM21A FAM21A FLNB FLNB GLO1 GLO1 GMPR2 GMPR2 GTSF1 GTSF1 IL7R IL7R JUP JUP KRT7 KRT7 KRT8 KRT8 LAMB1 LAMB1 LPGAT1 LPGAT1 MARVELD2 MARVELD2 MEG3 MEG3 MTA3 MTA3 MXI1 MXI1 NKRF NKRF NTHL1 NTHL1 OCLN OCLN POLR3H POLR3H PRKAG1 PRKAG1 QPRT QPRT SCNN1A SCNN1A SEPHS2 SEPHS2 SETD5 SETD5 SETDB1 SETDB1 SHF SHF SLC25A4 SLC25A4 SORBS3 SORBS3 SUV420H1 SUV420H1 SVIL SVIL TDG TDG TPD52 TPD52 TPRG1L TPRG1L TRIM2 TRIM2 ULK4 ULK4 USP53 -1 USP53 -1 VTCN1 VTCN1 ZBTB10 ZBTB10 ZNF33B ZNF33B

Figure 4. CXCL13þCD103þ TIL genes correlate with TLS-associated genes across cancer types. Spearman correlation of genes associated with TLS, CD103þCD8þ cells, and – þ CD103 CD8 cells in ovarian (n ¼ 307), uterine (n ¼ 546), lung (n ¼ 517), and breast (n ¼ 1,100) cancer. Correlation plots depict relative correlation of log2þ1 transformed mRNA sequencing data from TCGA.

792 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

secrete the protein only after reactivation (Fig. 1I). Therefore, we suppression (40, 47–49), was essential for the induction of þ þ analyzed the effect of CXCL13 CD103 CTL infiltration on CXCL13. Under homeostatic conditions, TGFb1 is abundantly clinical outcome in this cohort by proxy, using CD103 staining. present in epithelial tissue and controls the epithelial localization We observed a significant survival benefit for patients with mis- of resident memory immune subsets, such as the intraepithelial match repair–proficient tumors infiltrated by a high number of lymphocytes in the colon (50). In epithelial cancers, we suggest þ þ CD103 cells over tumors infiltrated by a low number of CD103 that TGFb1 has a similar role in promoting not only recruitment, þ cells (Supplementary Fig. S3B). This effect was independent of signaling, and retention of CD8 T cells via CD103 expres- other clinical variables, as demonstrated by multivariate analysis sion (51), but also stimulating immunity via attraction of C-X- þ þ (Table 1). As such, our data demonstrated that (CXCL13 ) C chemokine receptor type 5 (CXCR5) immune cells through þ CD103 CTLs are associated with improved clinical outcome, CXCL13 signaling. independent of neoantigen burden. CXCL13 is the key molecular determinant of TLS forma- tion (17–19), ectopic lymphoid structures that are thought to enable efficient local priming of T cells by DCs (9). Hereby, the Discussion time-consuming migration of DCs and T cells to and from lymph In this study, we reported on the finding that TGFb stimu- nodes may be circumvented, augmenting local antitumor immu- þ lates activated CD8 T cells to produce CXCL13, a known nity. In-line with this, characteristic components of TLSs, such as inducer of TLSs (17–19). This production of CXCL13 was HEVs and B cells, are found to be generally associated with an paralleled by the induction of CD103 on the cell surface of improved prognosis (10), and plasma B cells in the TLSs are þ þ þ CD8 cells in vitro. CD103 CD8 T cells isolated directly from thought to enhance antitumor responses by production and human tumors expressed CXCL13 mRNAandsecretedCXCL13 subsequent accumulation of antitumor antibodies, potentially protein upon ex vivo reactivation. The presence of B-cell and leading to antibody-dependent and opsoniza- þ þ þ TLS genes was increased in mutated, CXCL13 CD103 CD8 tion (12). Thus, TLSs may orchestrate a joint T- and B-cell response T-cell–enriched human tumors from TCGA, and the absolute to improve antitumor immunity. number of B cells associated with a high load of predicted Because TLSs were more abundant in tumors with a high þ neoantigens in an independent cohort of uterine cancers and a mutational load, we postulated that activated CD103 CTLs were cohort of high-grade serous ovarian cancers. Our findings shed involved in the migration of B cells to tumors via production of þ light on the link between CD8 T-cell activation and the CXCL13. This is supported by our observations that mutated, þ migration of B cells into the tumor. Our data also identified CD8 T-cell–rich tumors showed higher expression of CXCL13 CD103 and B cells as potential biomarkers of interest for and ITGAE (CD103) and that they presented with significantly cancer immunotherapy. higher numbers of B cells. In accordance, a higher degree of TCR þ CXCL13 is generally associated with DCs and TFH (40, 45, 46). clonality within CD8 T cells correlates with a higher number of Nevertheless, single-cell sequencing of exhausted, tumor-infiltrat- TLSs in non–small cell lung cancer (52). These TLSs may represent ing T cells of cancer, breast cancer, lung cancer, and mela- an ongoing immune response that is insufficient to halt tumor noma does support expression of CXCL13 in TILs (34–36). We progression at an early time point. It would, therefore, be of great found that TGFb1, a cytokine mostly associated with immune interest to study the induction and formation of TLSs in

Table 1. Cox regression survival analysis Disease-specific survival, n ¼ 189 Univariate Multivariate HR (95% CI) P HR (95% CI) P Age of diagnosis (cont) 1.022 (0.989–1.056) 0.197 Grade of differentiation Good/moderate ref ref ref ref Poor/undifferentiated 4.934 (2.307–10.550) <0.0001 2.694 (1.192–6.091) 0.017 FIGO stage I ref ref ref ref II 1.425 (0.261–7.780) 0.683 1.016 (0.184–5.617) 0.985 III 8.117 (2.646–24.906) <0.0001 4.403 (1.349–14.371) 0.014 IV 22.707 (7.218–71.432) <0.0001 15.697 (4.762–51.735) <0.0001 Myometrial invasion No Ref ref Yes 3.425 (1.568–7.480) 0.002 LVSI No ref ref Yes 5.617 (2.650–11.907) <0.0001 Lymph node metastasis No ref ref Yes 4.620 (1.911–11.171) 0.001 No sampling 0.775 (0.299–2.011) 0.601 CD103 Low ref ref ref ref High 0.249 (0.107–0.581) 0.001 0.273 (0.114–0.652) 0.003 NOTE: CD103 - low is lower than or equal to median expression; high is higher than median expression. All values in boldface are significant at a level of P < 0.05. Abbreviations: FIGO, International Federation of Gynecology and Obstetrics; ref, reference.

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 793

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

þ developing cancer lesions and to determine whether CD8 T-cell CXCL13 chemokine by activated T cells and subsequently to the infiltration precedes TLS formation. formation of lymphoid structures. TLSs may, therefore, reflect an þ þ In-line with previous work (24, 29, 53), CD103 CTLs from ongoing CD8 T-cell response in cancer. As such, TLSs may be human tumors were also characterized by an activation- and used as a biomarker to predict response to ICI, and these structures exhaustion-related gene expression signature, with differential may be used as a general biomarker for response to immuno- expression of granzymes and well-known immune checkpoint therapy because TLS were found to identify patients with pan- þ molecules, such as CTLA4. CD103 CTLs expressed several addi- creatic cancer who responded to therapeutic vaccination (58). tional immune checkpoint genes currently under clinical inves- Taken together, we demonstrated that TGFb1 induces coex- þ tigation, such as TIM3, LAG3, and TIGIT. This observed phenotype pression of CXCL13 and CD103 in CD8 T cells, potentially þ þ is concordant with a reported subset of PD-1–high (PD-1T) CD8 linking CD8 T-cell activation to B-cell migration and TLS for- T cells in lung cancer (34). The PD-1T subset was transcriptionally mation. Our findings provide a perspective on how (neo) distinct and characterized by expression of CXCL13. This subset could promote the formation of TLSs in human tumors. Accord- þ also expressed higher ITGAE, although cell surface expression of ingly, CD103 cells and B cells should be considered as a potential CD103 was not examined. Nevertheless, our analysis of the gene predictive or response biomarker for ICI therapy. þ expression profile reported for PD-1T CD8 cells (34) revealed overlapping overexpression of approximately 200 genes with Disclosure of Potential Conflicts of Interest þ þ CD103 CD8 cells, suggesting these populations might repre- H.W. Nijman reports receiving commercial research funding from Aduro þ þ sent the same T-cell subset. Importantly, CXCL13 PD-1T CD8 Biotech Europe and BionTech, has ownership interest in ViciniVax, and is a cells are associated with response to ICI (34). consultant/advisory board member for ViciniVax. M. de Bruyn reports þ T þ receiving commercial research funding from Aduro Biotech Europe and The association between CXCL13 PD-1 CD8 cells and fl þ European Industrial Doctorate (I-Direct). No potential con icts of interest response to ICI is in-line with the observation that CD103 CTLs were disclosed by the other authors. significantly expand upon treatment with nivolumab or pembro- lizumab (anti–PD-1) in tumor specimens of patients with Disclaimer advanced-stage metastatic melanoma (54). Accordingly, an article The views expressed are those of the authors and not necessarily those of the by Riaz and colleagues demonstrates that tumors from patients NHS, the NIHR, the Department of Health, or the Wellcome Trust. who responded to nivolumab treatment differentially expressed genes such as CXCL13, CTLA4, TIM3, LAG3, PDCD1, GZMB, and Authors' Contributions TNF receptor superfamily member 9 (TNFRSF9), all genes over- Conception and design: H.H. Workel, J.M. Lubbers, T. Bosse, F.L. Komdeur, þ – expressed in CD103 versus CD103 CTLs (7). Pretreatment, but H.W. Nijman, M. de Bruyn not on-treatment, CXCL13 was differentially expressed in respon- Development of methodology: P. van der Vlies, T. Bosse, M.C.A. Wouters, M. de Bruyn ders versus nonresponders in this study (7). This may be explained Acquisition of data (provided animals, acquired and managed patients, by the low basal CXCL13 secretion we observed in the exhausted, provided facilities, etc.): H.H. Workel, J.M. Lubbers, P. van der Vlies, þ CD103 CTLs freshly isolated from untreated human tumors. In I.C. Van Gool, F.A. Eggink, F.A. Eggink, M.C.A. Wouters, F.L. Komdeur, their exhausted state, CTLs might accumulate mRNA encoding C.L. Creutzberg, A. Kol, A. Plat, M. Glaire several key effector molecules that is translated only upon reac- Analysis and interpretation of data (e.g., statistical analysis, biostatistics, tivation (e.g., by ICI). Consistent with this, Riaz and colleagues computational analysis): H.H. Workel, J.M. Lubbers, R. Arnold, T.M Prins, – E.C. van der Slikke, M. Glaire, D.N. Church observed an increase in the number of B-cell related genes on- Writing, review, and/or revision of the manuscript: H.H. Workel, J.M. Lubbers, treatment in responding patients (7), perhaps hinting at B-cell R. Arnold, P. van der Vlies, T. Bosse, M.C.A. Wouters, A. Kol, D.N. Church, recruitment and the formation of TLSs in these patients upon ICI- H.W. Nijman, M. de Bruyn mediated release of CXCL13. This hypothesis is supported Administrative, technical, or material support (i.e., reporting or organizing by the observed increases in serum CXCL13 and concomitant data, constructing databases): P. van der Vlies, K. de Lange, I.C. Van Gool þ depletion of CXCR5 B cells from the circulation in patients Study supervision: H.W. Nijman, M. de Bruyn Other (chair of International TransPROTEC collaboration): C.L. Creutzberg treated with anti–CTLA-4 and/or anti–PD-1 (55). Our data, therefore, suggest that ICIs are of particular interest for patients Acknowledgments þ þ fi with a high CXCL13 CD103 CTL in ltration pretreatment The authors would like to thank Henk Moes, Geert Mesander, Johan Teunis, across malignancies. Joan Vos, and Niels Kouprie for their technical assistance. This work was Several combination immunotherapy regimes that promote supported by Dutch Cancer Society/Alpe d'Huzes grant UMCG 20146719 þ CD8 T-cell infiltration and TLS formation may also function via (to M. de Bruyn), Dutch Cancer Society Young Investigator Grant 10418 (to þ CD8 T-cell–dependent production of CXCL13. For instance, T. Bosse), Jan Kornelis de Cock Stichting grants (to F.L. Komdeur, F.A. Eggink, and H.H. Workel), Nijbakker-Morra Stichting and Studiefonds Ketel1 grants (to combined therapy with antiangiogenic and immunotherapeutic H.H. Workel), the Oxford NIHR Comprehensive Biomedical Research Centre, agents in mice stimulated the transformation of tumor blood core funding to the Wellcome Trust Centre for Human Genetics from the vessels into intratumoral HEVs, which subsequently enhanced the Wellcome Trust (090532/Z/09/Z), a Wellcome Trust Clinical Training Fellow- þ infiltration and activation of CD8 T cells and the destruction of ship (to M. Glaire), and a Health Foundation/Academy of Medical Sciences tumor cells (56, 57). These T cells formed structures around HEVs Clinician Scientist Fellowship award (to D.N. Church). that closely resembled TLSs (56, 57). One of these studies found þ that induction of TLSs was dependent on both CD8 T cells and The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked (56). However, the exact intratumoral mechanism advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate of action remains unclarified. Because macrophages produce this fact. TGFb in a chronically inflamed environment (40), we hypothe- size that the macrophages in these studies may have generated a Received July 30, 2018; revised December 3, 2018; accepted March 6, 2019; TGFb-enriched environment, thus, leading to the production of published first March 14, 2019.

794 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

TGFb Induces CXCL13 in CD103þ Tumor-Infiltrating T Cells

References 1. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. candidates for checkpoint inhibition. Oncoimmunology 2017;6: PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lym- e1264565. phoma. N Engl J Med 2015;372:311–9. 22. Nout RA, Bosse T, Creutzberg CL, Jurgenliemk-Schulz€ IM, Jobsen JJ, 2. Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, et al. Safety and Lutgens LCHW, et al. Improved risk assessment of endometrial cancer by tumor responses with lambrolizumab (Anti–PD-1) in melanoma. N Engl J combined analysis of MSI, PI3K-AKT, Wnt/b- and P53 pathway Med 2013;369:134–44. activation. Gynecol Oncol 2012;126:466–73. 3. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. 23. Creutzberg CL, van Putten WL, Koper PC, Lybeert ML, Jobsen JJ, Warlam- Nivolumab versus docetaxel in advanced nonsquamous non–small-cell Rodenhuis CC, et al. Surgery and postoperative radiotherapy versus surgery lung cancer. N Engl J Med 2015;373:1627–39. alone for patients with stage-1 endometrial carcinoma: multicentre ran- 4. Pardoll DM. The blockade of immune checkpoints in cancer immuno- domised trial. PORTEC Study Group. Post Operative Radiation Therapy in therapy. Nat Rev Cancer 2012;12:252–64. Endometrial Carcinoma. Lancet 2000;355:1404–11. 5. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, et al. 24. Van Gool IC, Eggink FA, Freeman-Mills L, Stelloo E, Marchi E, De Bruyn M, Genetic basis for clinical response to CTLA-4 blockade in melanoma. et al. POLE proofreading mutations elicit an antitumor immune response N Engl J Med 2014;371:2189–99. in endometrial cancer. Clin Cancer Res 2015;21:3347–55. 6. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. 25. Komdeur FL, Wouters MCA, Workel HH, Tijans AM, Leffers N, Helfrich W, Mutational landscape determines sensitivity to PD-1 blockade in non– et al. CD103 þ intraepithelial T cells in high-grade serous ovarian cancer are small cell lung cancer 2016;348:124–9. phenotypically diverse TCRab þ CD8ab þ T cells that can be targeted for 7. Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor cancer immunotherapy. 2016;7:75130–44. and microenvironment evolution during immunotherapy with nivolu- 26. Rayner E, van Gool IC, Palles C, Kearsey SE, Bosse T, Tomlinson I, et al. A mab. Cell 2017;171:934–949.e15. panoply of errors: polymerase proofreading domain mutations in cancer. 8. Dieu-Nosjean MC, Goc J, Giraldo NA, Sautes-Fridman C, Fridman WH. Nat Rev Cancer 2016;16:71–81. Tertiary lymphoid structures in cancer and beyond. Trends Immunol 2014; 27. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, 35:571–80. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the 9. Dieu-Nosjean MC, Giraldo NA, Kaplon H, Germain C, Fridman immune landscape in human cancer. Immunity 2013;39:782–95. WH, Sautes-Fridman C. Tertiary lymphoid structures, drivers of the 28. Workel HH, Komdeur FL, Wouters MCA, Plat A, Klip HG, Eggink FA, et al. anti-tumor responses in human cancers. Immunol Rev 2016;271: CD103 defines intraepithelial CD8þ PD1þ tumour-infiltrating lympho- 260–75. cytes of prognostic significance in endometrial adenocarcinoma. Eur J 10. Sautes-Fridman C, Lawand M, Giraldo NA, Kaplon H, Germain C, Fridman Cancer 2016;60:1–11. WH, et al. Tertiary lymphoid structures in cancers: Prognostic value, 29. Picelli S, Faridani OR, Bjorklund€ ÅK, Winberg G, Sagasser S, Sandberg R. regulation, and manipulation for therapeutic intervention. Front Immunol Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc 2014;9: 2016;7:1–11. 171–81. 11. Giraldo NA, Becht E, Pages F, Skliris G, Verkarre V, Vano Y, et al. Orches- 30. Carpentier G, Henault E. Protein Array Analyzer for ImageJ. Proceedings of tration and prognostic significance of immune checkpoints in the micro- the ImageJ User and Developer Conference, Centre de Recherche Public environment of primary and metastatic renal cell cancer. Clin Cancer Res Henri Tudor, ed., (ISBN 2-919941-11-9), pp. 238–240, 2010. 2015;21:3031–40. 31. Komdeur FL, Prins TM, van de Wall S, Plat A, Wisman GBA, Hollema H, 12. Kroeger DR, Milne K, Nelson BH. Tumor-infiltrating plasma cells are et al. CD103þ tumor-infiltrating lymphocytes are tumor-reactive intrae- associated with tertiary lymphoid structures, cytolytic T-cell responses, pithelial CD8þ T cells associated with prognostic benefit and therapy and superior prognosis in ovarian cancer. Clin Cancer Res 2016;22: response in cervical cancer. Oncoimmunology 2017;6:1–14. 3005–15. 32. Djenidi F, Adam J, Goubar A, Durgeau A, Meurice G, de Montpreville V, þ þ 13. Pimenta EM, Barnes BJ. Role of tertiary lymphoid structures (TLS) in anti- et al. CD8 CD103 tumor–infiltrating lymphocytes are tumor-specific tumor immunity: potential tumor-induced /chemokines that tissue-resident memory T cells and a prognostic factor for survival in lung regulate TLS formation in epithelial-derived cancers. Cancers 2014;6: cancer patients. J Immunol 2015;194:3475–86. 969–97. 33. Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, et al. Global 14. Ansel KM, Harris RBS, Cyster JG. CXCL13 is required for B1 cell homing, characterization of T cells in non-small-cell lung cancer by single-cell natural antibody production, and body cavity immunity. Immunity 2002; sequencing. Nat Med 2018;24:978–85. 16:67–76. 34. Thommen DD, Koelzer V, Herzig P, Roller A, Trefny M, Kiialainen A, et al. A þ þ 15. Van De Pavert SA, Olivier BJ, Goverse G, Vondenhoff MF, Greuter M, Beke transcriptionally and functionally distinct PD-1 CD8 T cell pool with P, et al. Chemokine is essential for initiation and is predictive potential in non-small cell lung cancer treated with PD-1 induced by retinoic acid and neuronal stimulation. Nat Immunol 2009;10: blockade. Nat Med 2018;24:994–1004. 1193–9. 35. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of 16. Ansel KM, Ngo VN, Hyman PL, Luther SA, Forster€ R, Sedgwlck JD, et al. A infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell chemokine-driven positive feedback loop organizes lymphoid follicles. 2017;169:1342–56. Nature 2000;406:309–14. 36. Tirosh I, Izar B, Prakadan SM, Ii MHW, Treacy D, Trombetta JJ, et al. 17. Luther SA, Ansel KM, Cyster JG. Overlapping roles of CXCL13, 7 Dissecting the multicellular exosystem of metastatic melanoma by single- receptor a, and CCR7 ligands in lymph node development. J Exp Med cell RNA-seq. Science 2016;352:189–96. 2003;197:1191–8. 37. Gu-Trantien C, Migliori E, Buisseret L, De Wind A, Brohee S, Garaud S, et al. 18. Luther SA, Lopez T, Bai W, Hanahan D, Cyster JG. BLC expression in CXCL13-producing T FH cells link immune suppression and adaptive pancreatic islets causes recruitment and lymphotoxin-dependent memory in human breast cancer. JCI Insight 2017;2:1–17. lymphoid neogenesis. Immunity 2000;12:471–81. 38. Mokrani M, Klibi J, Bluteau D, Bismuth G, Mami-Chouaib F. Smad and 19. Gr€abner R, Lotzer€ K, Dopping€ S, Hildner M, Radke D, Beer M, et al. NFAT pathways cooperate to induce CD103 expression in human CD8 T Lymphotoxin b receptor signaling promotes tertiary lymphoid organo- lymphocytes. J Immunol 2014;192:2471–9. genesis in the aorta adventitia of aged ApoE / mice. J Exp Med 2009;206: 39. Park B V., Freeman ZT, Ghasemzadeh A, Chattergoon MA, Rutebemberwa 233–48. A, Steigner J, et al. TGFb1-mediated SMAD3 enhances PD-1 expression on 20. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S. A antigen-specific T cells in cancer. Cancer Discov 2016;6:1366–81. transcriptionally and functionally distinct PD-1 þ CD8 þ T cell pool with 40. Kobayashi S, Watanabe T, Suzuki R, Furu M, Ito H, Ito J, et al. TGF-b induces predictive potential in non-small cell lung cancer treated with PD-1 the differentiation of human CXCL13-producing CD4þ T cells. Eur J blockade. Nat Med 2018;24:994–1004. Immunol 2016;46:360–71. 21. Eggink FA, Van Gool IC, Leary A, Pollock PM, Crosbie EJ, Mileshkin L, et al. 41. Amft N, Curnow SJ, Scheel-Toellner D, Devadas A, Oates J, Crocker J, et al. Immunological profiling of molecularly classified high-risk endometrial Ectopic expression of the B cell-attracting chemokine BCA-1 (CXCL13) on cancers identifies POLE-mutant and microsatellite unstable carcinomas as endothelial cells and within lymphoid follicles contributes to the

www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 795

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

Workel et al.

establishment of germinal center-like structures in Sjogren's€ syndrome. 51. Boutet M, Gauthier L, Leclerc M, Gros G, De Montpreville V, Theret N, et al. Arthritis Rheum 2001;44:2633–41. TGFb signaling intersects with CD103 integrin signaling to promote 42. Steinmetz OM, Panzer U, Kneissler U, Harendza S, Lipp M, Helmchen U, T-Lymphocyte accumulation and antitumor activity in the lung tumor et al. BCA-1/CXCL13 expression is associated with CXCR5-positive B-cell microenvironment. Cancer Res 2016;76:1757–69. cluster formation in acute renal transplant rejection. Kidney Int 2005;67: 52. Zhu W, Germain C, Liu Z, Sebastian Y, Devi P, Knockaert S, et al. A high 1616–21. density of tertiary lymphoid structure B cells in lung tumors is associated þ 43. Henry RA, Kendall PL. CXCL13 blockade disrupts B lymphocyte organi- with increased CD4 T cell receptor repertoire clonality. Oncoimmunol- zation in tertiary lymphoid structures without altering B cell receptor bias ogy 2015;4:e1051922. or preventing diabetes in nonobese diabetic mice. J Immunol 2010;185: 53. Ganesan A-P, Clarke J, Wood O, Garrido-Martin EM, Chee SJ, Mellows T, 1460–5. et al. Tissue-resident memory features are linked to the magnitude of 44. Marraco SF, Neubert N, Verdeil G, Speiser D, Vergassola M, Altan-Bonnet G, cytotoxic T cell responses in human lung cancer. Nat Immunol 2017;18: et al. CXCL13-producing TFH cells link immune suppression and adaptive 940–50. memory in human breast cancer. Front Immunol 2017;6:1–17. 54. Edwards J, Wilmott JS, Madore J, NurGideT,QuekC,TaskerA,etal. 45. Rasheed A-U, Rahn H-P, Sallusto F, Lipp M, Muller€ G. Follicular B helper T CD103þ tumor-resident CD8þ T cells are associated with improved cell activity is confined to CXCR5hiICOShi CD4 T cells and is independent survival in immunotherapy naive melanoma patients and expand of CD57 expression. Eur J Immunol 2006;36:1892–903. significantly during anti-PD-1 treatment. Clin Cancer Res 2018;24: 46. Vissers JLM, Hartgers FC, Lindhout E, Figdor CG, Adema GJ. BLC (CXCL13) 3036–45. is expressed by different subsets in vitro and in vivo. Eur J 55. Das R, Bar N, Ferreira M, Newman AM, Zhang L, Bailur JK, et al. Early B cell Immunol 2001;31:1544–9. changes predict autoimmunity following combination immune check- 47. Wrzesinski SH, Wan YY, Flavell RA. Transforming -b and the point blockade. J Clin Invest 2018;128:2–7. immune response: Implications for anticancer therapy. Clin Cancer Res 56. Johansson-Percival A, He B, Li ZJ, Kjellen A, Russell K, Li J, et al. De novo 2007;13:5262–70. induction of intratumoral lymphoid structures and vessel normalization 48. Blobe G, Schiemann WP, Lodish HF. Role of transforming growth factor enhances immunotherapy in resistant tumors. Nat Immunol 2017;18: beta in human disease. N Engl J Med 2000;342:1350–8. 1207–17. 49. Johnston CJC, Smyth DJ, Dresser DW, Maizels RM. TGF-b in tolerance, 57. Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, et al. development and regulation of immunity. Cell Immunol 2015;299: Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor 14–22. immunity through HEV formation. Sci Transl Med 2017;9:eaak9679. 50. Konkel JE, Maruyama T, Carpenter AC, Xiong Y, Zamarron BF, Hall BE, et al. 58. Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, et al. Immuno- Control of the development of CD8aaþ intestinal intraepithelial lym- therapy converts nonimmunogenic pancreatic tumors into immunogenic phocytes by TGF-b. Nat Immunol 2011;12:312–20. foci of immune regulation. Cancer Immunol Res 2014;2:616–31.

796 Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst March 14, 2019; DOI: 10.1158/2326-6066.CIR-18-0517

A Transcriptionally Distinct CXCL13+CD103+CD8+ T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Human Cancer

Hagma H. Workel, Joyce M. Lubbers, Roland Arnold, et al.

Cancer Immunol Res 2019;7:784-796. Published OnlineFirst March 14, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/2326-6066.CIR-18-0517

Supplementary Access the most recent supplemental material at: Material http://cancerimmunolres.aacrjournals.org/content/suppl/2019/03/13/2326-6066.CIR-18-0517.DC1

Cited articles This article cites 55 articles, 15 of which you can access for free at: http://cancerimmunolres.aacrjournals.org/content/7/5/784.full#ref-list-1

Citing articles This article has been cited by 8 HighWire-hosted articles. Access the articles at: http://cancerimmunolres.aacrjournals.org/content/7/5/784.full#related-urls

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 Department Subscriptions at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerimmunolres.aacrjournals.org/content/7/5/784. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerimmunolres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research.