Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

TUMOR CELL-DERIVED IL-1 PROMOTES DESMOPLASIA AND

IMMUNE SUPPRESSION IN PANCREATIC

Shipra Das1, Beny Shapiro1, Emily A. Vucic1, Sandra Vogt1 and Dafna Bar-Sagi1,*

1 Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of

Medicine, New York, NY 10016, USA

*Correspondence: e-mail: [email protected]

Telephone: 212-263-2095

Address: 530 First Avenue, HCC 15, Executive Offices

New York, NY 10016

Running Title: IL-1 promotes immune suppression in .

Competing Interests Statement: The authors declare no potential conflicts of interest.

1

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

Pancreatic ductal adenocarcinoma (PDA) is an aggressive malignancy typified by a highly stromal and weakly immunogenic that promotes tumor evolution and contributes to therapeutic resistance. Here, we demonstrate that PDA tumor cell-derived pro-inflammatory , interleukin- (IL- ) is essential for the establishment of the pro-tumorigenic PDA microenvironment.

Tumor cell-derived IL- promoted the activation and secretory phenotype of quiescent pancreatic stellate cells (PSC) and established an immunosuppressive milieu mediated by M2 macrophages,

MDSCs, CD1dhiCD5+ regulatory B cells and Th17 cells. Loss of tumor cell-derived IL-1 signaling in tumor stroma enabled intra-tumoral infiltration and activation of CD8+ cytotoxic T cells, attenuated growth of pancreatic neoplasia and conferred survival advantage to PDA bearing mice. Accordingly, antibody-mediated neutralization of IL- significantly enhanced the anti-tumor activity of -PD-1, and was accompanied by increased tumor infiltration of CD8+ T cells. Tumor cell expression of IL- in vivo was driven by microbial-dependent activation of toll-like receptor 4 (TLR4) signaling and subsequent engagement of the NLRP3 inflammasome. Collectively, these findings identify a hitherto unappreciated role for tumor cell-derived IL- in orchestrating an immune modulatory program that supports pancreatic tumorigenesis.

Significance: Findings identify a new modality for immune evasion in PDA that depends on IL- production by tumor cells through TLR4-NLRP3 inflammasome activation. Targeting this axis might provide an effective PDA therapeutic strategy.

2

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

INTRODUCTION

Pancreatic ductal adenocarcinoma (PDA) is a highly lethal malignancy with a mortality rate approaching the rate of incidence (1). In addition to lack of efficient early diagnosis methods, disease survival is compromised by resistance to conventional chemotherapy and immunotherapeutic strategies that are proving effective in the treatment of other (2,3). It is becoming increasingly recognized that this recalcitrance is largely attributable to an elaborate network of tumor-stromal interactions that are orchestrated by paracrine factors released by the tumor epithelium, activated and immune cells

(4,5). Identification and functional characterization of such factors and the processes they control is therefore an essential prerequisite for rational development of strategies that can circumvent therapeutic barriers and improve immune responsiveness of PDA tumors.

The cytokine interleukin 1 (IL-1 is an inflammatory mediator that is frequently upregulated in a variety of cancers and its production is associated with poor prognosis (6,7). Upregulation of either IL1B expression or post-translational processing in head and neck squamous carcinoma, , lung cancer and results in increased tumor infiltration of immunosuppressive macrophages and myeloid-derived suppressor cells (MDSC), thereby promoting immune evasion and tumor development

(8-10). Other pro-tumorigenic effects of IL-1 have been attributed to the induction of neoangiogenesis

(11) and the regulation of expression in stromal cells of soluble mediators that enhance tumor cell survival and (7). These effects are mediated by IL-1 -dependent signaling cascades that under conditions of IL-1 over-abundance result in the sustained activation of NF- B and mitogen activated protein kinase (MAPK) pathways (7).

Several lines of evidence suggest a role for IL1- in pancreatic cancer development and progression.

Increased pancreatic levels of IL-1 are observed in association with pancreatitis, a well-established PDA risk factor (12). High intra-tumoral and serum IL-1 levels in pancreatic cancer patients correlate with poor overall survival and increased chemoresistance (13-15). In mouse models of PDA, adipocyte-

3

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

secreted IL-1 is found to promote obesity-induced pancreatic carcinogenesis and drug resistance through recruitment of tumor-associated neutrophils (16). In addition, regulatory pathways that control IL-1 production in PDA-associated myeloid cells have been reported to support tumor progression by promoting immune tolerance (17,18). Overall, several lines of evidence suggest a heterotypic distribution of IL-1 expression in PDA with implications in disease pathogenesis. Thus, in this study we sought to elucidate the mechanisms underlying the regulation and function of IL-1 in PDA, with an eye on assessing its potential as a therapeutic target.

Here, we identify the tumor cell compartment as a prominent source of IL-1 production in human and mouse PDA through activation of the TLR4-NLRP3 inflammasome signaling pathway. Targeted depletion of IL-1 in established mouse models demonstrates acute dependency of pancreatic cancer evolution on tumor cell-derived IL-1 through pro-tumorigenic modulation of the stroma and immune microenvironment. Overall, our study identifies IL-1 as an attractive target that may improve PDA response to therapeutic strategies, including immunotherapy.

4

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

MATERIAL AND METHODS

Animals All mouse protocols were reviewed and approved by the Institutional Animal Care and Use

Committee of the New York University (NYU) Grossman School of . The LSL-KrasG12D/+, LSL-

Trp53R172H/+, and p48Cre/+ mice strains have been described previously (19,20). Eight- to 10-week-old wild type (WT) C57BL/6 (stock 027) mice were purchased from The Charles River Laboratories. Eight- to 10- week-old IL1r1-/- (stock # 003245) mice were purchased from The Jackson Laboratories. All mice were on a C57BL/6 genetic background. Female mice were used for orthotopic injections of KRasG12D-PDEC and KPC cells (21,22). Briefly, mice were anesthetized using a ketamine (100 mg/kg)/Xylazine (10 mg/kg) cocktail administered via intraperitoneal injection. After making a small incision on the left abdominal wall, either 106 KrasG12D-PDEC or 5 X 104 KPC cells in ice-cold PBS mixed at 1:1 dilution with Matrigel (#354234, Corning) in a volume of 50 L were injected into the tail of the pancreas using a

28-gauge needle. For PSC co-implantation experiments, 106 KrasG12D-PDEC cells and 105 PSCs were mixed in 50 l of ice-cold PBS: Matrigel (1:1) and injected into the tail of the pancreas using a 28-gauge needle. The incision was closed using 5-0 Vicryl RAPIDE sutures (Ethicon) for the body-wall and 4-0

PROLENE sutures (Ethicon) for the skin. All animals were given buprenorphine (0.1 mg/kg) for pain relief directly after surgery and once a day for three days post-surgery. Mice were euthanized by carbon dioxide induced narcosis 2 weeks post implantation of KRasG12D-PDEC. For KPC cells, mice were euthanized 2 weeks and 4 weeks post implantation for flow cytometry analysis and tumor volume assessment, respectively. KPC tumors were measured using digital caliper (VWR) at the endpoint and tumor volume was calculated using the formula 2/6.

Anti-PD-1, anti-IL-1 treatment and CD8+ depletion. Mice were orthotopically injected into the pancreas with 5 X 104 KPC cells. On day 7 post-injection, mice were intraperitoneally administered either

10 mg/kg anti PD-1 (Novartis), 10 mg/kg anti-IL-1 Novartis or IgG control (Novartis) antibody diluted in 2 Thereafter, anti-PD-1 antibody was administered on days 9, 11 and 16 and anti-IL-1 was administered every 2 days. For CD8+ T- -CD8 (BioXCell,

5

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

clone 53-6.7) or an IgG isotype control antibody (BioXCell, clone 2A3) diluted in 2 were administered intraperitoneally daily starting 3 days prior to tumor cell injection and every 5 days after tumor cell injection. Efficiency of CD8+ T-cell depletion was assessed by flow cytometry.

Murine Bacterial Depletion Mouse gut microbiota depletion was performed as described previously

(23). Briefly, 6-week old WT mice were administered 0.1 mg/ml Amphotericin-B (Sigma) by oral gavage every 12 hours for three days. Subsequently, water flasks were supplemented with 1g/L ampicillin (Fisher

Bioreagents) and antibiotic cocktail containing 5 mg/ml vancomycin (Cayman Chemical Company), 10 mg/ml neomycin (Sigma) and 10 mg/ml metronidazole (Sigma) was administered by oral gavage once a day for 2 weeks pre-surgery and then for the duration of the experiment. Fresh antibiotic cocktail was mixed every day and ampicillin and water was renewed every 7th day. To assay for microbial depletion, fecal pellets were collected from mice at Day 0 and Day 14 of antibiotic treatment. DNA was isolated from fecal pellets with QIAamp DNA Stool Mini Kit (Qiagen) as per manufacturer instructions. Bacterial

16S DNA gene quantification was assessed by quantitative PCR as described previously (24).

Cell Lines Isolation, culture and adenoviral infection of PDEC were carried out as previously described

(21). The KPC cell line (line 4662) was a kind gift from Dr. R.H. Vonderheide. The immortalized PSC cell line was a kind gift from Dr. A.C. Kimmelman. Isolation and culture of primary PSCs was carried out as previously described (25). Cell lines were not authenticated and were tested for Mycoplasma contamination every 4 months. Scramble control and shRNAs against IL1B, NLRP3 and TLR4 were cloned into the lentiviral pLKO.1 hygro vector obtained from Addgene (#24150). shRNA sequences used were as follows scramble: GCGACATCCTCATCTCGTTAGTA; IL-1 -sh1:

GTGGTCAGGACATAATTGACTTC, IL-1 -sh2: GCAGCACATCAACAAGAGCTTCA; NLRP3-sh1:

AGCCTGAGCTGACTATAGTCTTC, NLRP3-sh2: CTTGAAGATGTGGACCTCAAGAA; TLR4-sh1:

GCCAATCCTAAGAATGCTATA. Lentiviral particles were generated by transfecting HEK-293T cells with the pLKO.1 vector, the packaging construct (psPAX2), and the envelope plasmid (pMD2G).

6

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Supernatants containing viral particles were collected over a period of 48 hours and stored at 4C.

Following final collection, supernatants were filtered through a 0.45-µm syringe filter and concentrated using 100 MWCO Amicon Ultra centrifugal filters (Millipore). A multiplicity of infection of 15 was used for lentiviral infection of KRasG12D-PDEC or KPC cells in the presence and infected cells were selected using 150 g/ml hygromycin (Sigma).

Human Data Generation

130 human PDAC tumor (n=75) and Adj Norm (n=55) mRNA expression profiles generated on the same array (Affymetrix GeneChip Human Genome U133 Plus 2.0) were downloaded from

GEO (https://www.ncbi.nlm.nih.gov/geo/) (GSE15471, GSE16515). Adj Norm samples clustering with PDAC tumors, PDAC tumor profiles clustering with subsets of Adj Norm samples and duplicates were discarded (as previously described (26), for a remainder of n= 74 tissues (n= 50 PDAC tumor and n= 24 Adj Norm). Raw data were processed and normalized in one batch using a GC-content background correction Robust Multi-array Average (RMA) algorithm (GC-RMA), performed in R: A language and environment for statistical computing.

Unpaired t test were generated in GraphPad Prism (GraphPad Software, CA, USA, www.graphpad.com).

Human Pancreas Specimens For the purposes of analyzing IL1B, NLRP3 and TLR4 expression pattern, we examined 8-10 patient PDAC lesions and corresponding adjacent normal tissue samples. Samples consisted of 5- -fixed, paraffin embedded (FFPE) blocks provided by the Center for Biospecimen Research and Development at NYU Langone Health. This study was conducted in accordance with the Declaration of Helsinki; all samples were anonymized prior to being transferred to the investigator's laboratory and therefore met exempt human subject research criteria.

7

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Histology and Mouse pancreata were fixed in 10% buffered formalin (Fisher

Scientific) overnight and embedded in paraffin as described earlier (21). Trichrome staining was performed at NYU Grossman School of Medicine Histopathology Core Facility. For

peroxide/methanol for 15 minutes, and antigen retrieval was performed in 10 mM sodium citrate/0.05%

Tween-20 (pH 6.0) for 15 minutes in a microwave oven. Blocking was done in 10% serum/1% BSA/0.5%

Tween-20 for 1 hour at room temperature, followed by incubation with the primary antibodies diluted in

2% BSA overnight at 4°C. Primary antibodies are detailed in Supplementary Data. After incubating with secondary biotinylated antibodies and ABC solution (both from Vector Laboratories), sections were developed with DAB peroxidase substrate kit (Vector Laboratories) according to manufacturer instructions. After counterstaining with Harris hematoxylin (Sigma), slides were subjected to an alcohol dehydration series and mounted with Permount (Fisher). Slides were examined on a Nikon Eclipse 80i microscope and images were analysed to measure stained area using ImageJ software.

Immunofluorescence FFPE sections were deparaffinized and rehydrated, permeabilized with TBS/0.1%

Tween-20 and washed in PBS. Citrate buffer antigen retrieval (10 mM sodium citrate/0.05% Tween-20, pH 6.0) was performed in a microwave for 15 minutes. Blocking was performed in 10% serum/1%

BSA/0.5% Tween-20/PBS for 1 hour at room temperature. Primary antibodies were diluted in 2%

BSA/0.5% Tween-20/PBS and incubated on sections overnight at 4°C. Primary antibodies are detailed in

Supplementary Data. Secondary antibodies (Alexa Fluor labeled; 1:1000, Invitrogen) were diluted in 2%

BSA/PBS, and incubated on sections for 1 hour at room temperature. Sections were washed with PBS and stained with DAPI. Slides were examined and imaged on a Nikon Eclipse Ti2 microscope.

Flow Cytometry Single-cell suspensions were prepared from pancreas as previously described (27). For isolation of tumor-infiltrating lymphocytes, tumor tissue was minced into 1 to 2 mm pieces and digested with collagenase IV (1.25 mg/mL, Worthington) and 0.1% trypsin inhibitor from soybean (Sigma), in complete RPMI for 25 minutes at 37°C. For isolation and FACS analysis of epithelial and

8

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

compartments, minced tumor tissue was digested with Pronase (0.2 mg/mL, Roche), Collagenase P (0.5 mg/ml, Roche) and DNase I (0.5 mg/mL, Roche). Cells were suspended in 1%FBS/PBS, passed through a

(eBiosciences). Single cell suspensions were blocked with anti-CD16/CD32 antibody (Fc Block, BD Biosciences) for 5 minutes on ice, and labeled with monoclonal antibodies against mouse antigens as detailed in Supplementary Data. All samples were acquired on LSR II (BD Bioscience) at NYU Flow Cytometry Core Facility and analyzed by FlowJo version 10.2 (TreeStar, Inc.). Cell sorting using a BD FACS ARIA II sorter was performed to isolate Ep-

CAM+ cells, CD140a+ fibroblasts and CD45+ cells, and >95% purity of sorted cells was achieved.

Quantitative RT-PCR For RNA isolation from tumors, pancreata processed to single cell suspension were stained for flow cytometry. CD45-CD34-CD140a+Ep-CAM- fibroblasts were FACS sorted using a

100- instructions (RNeasy Mini Kit, QIAGEN). To check knockdown in KRasG12D-PDEC and KPC cells, 105 cells were lysed in 350 instructions (RNeasy Mini Kit, QIAGEN). -transcribed using the

Quantitect Reverse Transcription Kit (Qiagen). Subsequently, specific transcripts were amplified by

SYBR Green PCR Master Mix (USB) using a Stratagene Mx 3005P thermocycler. Where fold expression is specified, comparative CT method was used to quantify gene expression. Expression was normalized to

GAPDH. Primers used for QPCR are detailed in Supplementary Data.

Supernatant collection and cytokine analysis For cytokine analysis of mouse pancreata, the tissues were harvested, before supernatant collection. Mouse IL-1 protein levels were determined by Mouse IL-1 Quantikine

ELISA Kit (R&D Systems) .

9

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Statistical analysis At least 7 to 15 mice were used in each group, and the experiments were repeated a minimum 2 times to validate reproducibility. Group means were compared with Student t tests.

Significance in variations between two groups was determined by an unpaired Student t test (two- tailed). Statistical analyses were performed using GraphPad Prism software (version 7.0d), and data are presented as mean ± SD. P < 0.05 was considered statistically significant.

10

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

RESULTS

Tumor cell-derived IL-1 promotes pancreatic tumorigenesis.

To assess IL-1 production in PDA, we first examined microarray data from 50 PDA patient tumors and

24 adjacent normal tissue samples. Our analysis revealed significant upregulation of IL1- expression in

PDA tumors relative to normal adjacent pancreatic tissue (Fig. 1A). We next assessed the distribution pattern of IL1- by immunohistochemical staining of tumor sections from patient PDA samples.

Consistent with previous reports documenting the expression of IL-1 by innate immune cells and fibroblasts (15,16,18), robust IL-1 staining was detected in the tumor stroma (Supplementary Fig. S1A).

However, unexpectedly, we also observed significant IL-1 staining in the ductal epithelium (Fig. 1B).

Similarly, robust expression of IL-1 in the CK8+ epithelial tumor cell compartment was revealed by immunofluorescence staining of pancreata from the slowly progressive KrasG12D; p48Cre (KC) mouse model of pancreatic neoplasia (19) and the KrasG12D;p53R172H;p48Cre (KPC) invasive PDA mouse model

(20) (Fig. 1C and D; Supplementary Fig. S1B). The relative levels of IL-1 production by stromal

(CD140a+ fibroblasts and CD45+ immune cells) and tumor cells (EpCAM+ epithelium) from KC mice was further analyzed by flow cytometry (Fig. 1E; Supplementary Fig. S1C). In agreement with the immunohistochemical data, the epithelial compartment displayed the highest levels of IL-1 production

(Fig. 1E).

Next we sought to investigate the functional significance of tumor-cell derived IL-1 . Utilizing a RNAi strategy, two independent short hairpin (sh) sequences targeting IL1B were introduced into pancreatic ductal epithelial cells derived from either KrasG12D mice (KRasG12D-PDEC) or

KrasG12D;Trp53R172H;p48Cre mice (KPC) (21,22). Knockdown efficiency was ascertained by qPCR and immunofluorescence staining (Supplementary Fig. S1D-S1F). Orthotopic injection of IL-1 -sh KRasG12D-

PDEC into pancreata of syngeneic mice led to grafts that displayed a significant reduction of CK8+ pancreatic intraepithelial neoplasia (PanIN)-like lesions, relative to scramble control (Fig. 1F;

11

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Supplementary Fig. S1G). A role for tumor cell-derived IL-1 was also evident in the context of more advanced lesions, with IL-1 -depleted KPC cells forming significantly smaller tumors upon orthotopic implantation (Fig. 1G and H). Furthermore, IL-1 knockdown increased the survival of mice bearing orthotopic KPC tumors (Fig. 1I). Overall, these results establish a pro-oncogenic role for tumor cell- derived IL-1 in pancreatic cancer.

Tumor cell-derived IL-1 induces a tolerogenic immune state in the PanIN microenvironment.

Given the well-established role of IL1- as an inflammatory mediator, we tested whether tumor cell- derived IL-1 promotes pancreatic oncogenesis through its interactions with the tumor microenvironment

(TME) by implanting KRasG12D-PDEC or KPC cells into the pancreas of IL-1 receptor (IL1r1) null mice

(28). Absence of IL-1 signaling in the pancreatic stroma phenocopied the depletion of tumor cell- derived IL-1 , with reduced growth of orthotopic KRasG12D-PDEC grafts and KPC tumors in IL1r1 null mice relative to wild type control (Supplementary Fig. S1H-S1K). In addition, the overall survival of

KPC cell-implanted animals was extended in IL1r1 null mice, relative to wild type control

(Supplementary Fig. S1L). Notably, surface expression of IL1r1 is nearly undetectable in KrasG12D-PDEC cells (Supplementary Fig. S1M). These observations suggest a paracrine role for tumor cell-derived IL-1 and therefore prompted us to investigate the fibro-inflammatory effects of tumor cell-derived IL-1 on the

TME.

Flow cytometric analysis of pancreatic grafts formed by IL-1 -sh KRasG12D-PDEC revealed a pronounced alteration of the TME immune landscape, relative to the scramble control. Specifically, depletion of tumor cell-derived IL-1 significantly decreased stromal accumulation of CD11b+F4/80+ tumor- associated macrophages (TAMs, Fig. 2A), CD11b+Gr1+ myeloid-derived suppressor cells (MDSC, Fig.

+ + hi + 2B), CD11b Ly6G tumor-associated neutrophils (TANs, Fig. 2C), CD1d CD5 regulatory B cells (Bregs,

+ + Fig. 2D) and CD4 ROR t TH17 cells (Fig. 2E). Additionally, knockdown of tumor cell-derived IL-1 also decreased the CD206+ M2 polarized state of stromal TAMs (Fig. 2F). No significant changes were

12

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

+ + + + observed in stromal recruitment of CD4 FoxP3 regulatory T cells, CD4 helper T (TH) cells, and CD19

B cells, upon IL-1 knockdown (Supplementary Fig. S2A-S2C).

The immune cell profile resulting from the suppression of IL-1 production is consistent with a role for tumor cell-derived IL-1 in constraining anti-tumor immunity in PDA. For instance, TANs have been reported to downregulate T cell infiltration in the PDA stroma (29). In addition, M2 TAMs, MDSCs and

hi + CD1d CD5 Breg are immunosuppressive cell populations that have been shown to inhibit the tumor lytic

+ activity of CD8 Tc cells (21,27,30). Indeed, loss of stromal immunosuppressive sub-populations in IL-

1 depleted KRasG12D-PDEC grafts was accompanied by a significant increase in tumor infiltration (Fig.

+ 2G) and activation of CD8 Tc cells, as measured by IFN and granzyme B expression (Fig. 2H and I).

Similar immune changes were also observed in tumors formed by orthotopic transplantation of IL-1 -sh

KPC cells (Supplementary Fig. S2D-S2L), indicating a role for IL-1 in shaping the immune microenvironment in advanced disease as well.

Together these observations implicate tumor cell-derived IL-1 in promoting the establishment of an immunosuppressive microenvironment. Notably, we observed a reduction in IL-1 -expressing CD45+ immune cells present in IL-1 -sh KRasG12D-PDEC pancreatic grafts relative to the scramble control

(Supplementary Fig. S2M) suggesting a feedforward mechanism wherein the tumor-derived IL-1 could dictate the abundance of stromal-derived IL-1 .

Tumor cell-derived IL-1 promotes immunosuppression in part by regulating activation and secretory phenotype of pancreatic stellate cells.

In addition to the immune changes described above, we also observed a significant decrease in stromal , as detected by deposition, in IL-1 -sh KRasG12D-PDEC pancreatic grafts

(Supplementary Fig. S3A). Since the most prominent source of collagen deposition in the are cancer associated fibroblasts (CAFs) which are generated by the activation of pancreatic

13

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

stellate cells (PSCs), we assessed the state of PSC activation in IL-1 -sh pancreatic grafts by assaying for the activation marker -SMA (31). While prominent -SMA staining was observed in the microenvironment of scramble control KRasG12D-PDEC grafts and KPC tumors, the IL-1 -sh KRasG12D-

PDEC grafts and IL-1 -sh KPC tumors were largely devoid of -SMA staining (Fig. 3A and B). In contrast, no change in the abundance of Vimentin-positive fibroblasts was detected in the IL-1 -sh

KRasG12D-PDEC grafts (Fig. 3A), indicating that tumor cell-derived IL-1 is required for PSC activation and not viability. The potential relevance of this activation mechanism to our observations is supported by the findings that these cells display surface expression of IL-1 receptor as determined by FACS analysis (Supplementary Fig. S3B).

To distinguish between direct and indirect effect of tumor-derived IL-1 on PSC activation, we isolated primary PSCs from wildtype or IL1r1 null mice and co-implanted each with KRasG12D-PDECs in IL1r1 null mice. Under these conditions, all components of the host stroma are IL1r1 null and therefore by definition unresponsive to tumor cell-derived IL-1 . We found that co-implanted wild-type PSCs successfully underwent activation, as detected by -SMA staining (Supplemental Fig. S3C). These results implicate tumor cell-derived IL-1 as the primary driver of PSC activation. Consistent with this interpretation, the IL1r1 null PSCs derived from pancreata of IL1r1 null mice failed to undergo activation when co-implanted with tumor cells (Supplemental Fig. S3C). The functional significance of the tumor- derived IL-1 /PSCs axis for pancreatic tumor growth is indicated by the observation that the growth defect of KrasG12D-PDEC pancreatic grafts in IL1r1 null mice could be rescued by co-implanted wild type

PSCs but not IL1R1 null PSCs (Supplemental Fig. S3D).

The pro-tumorigenic effects of CAFs is well documented and is mediated by multiple paracrine mechanisms (31,32). In addition to secreting extracellular matrix proteins and growth factors, the transition of PSCs from a quiescent to an activated state has been shown to be accompanied by the induction of an inflammatory program with upregulation of and chemokines such as IL-6,

14

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

CCL2, CCL5 and CCL8 (32). To investigate the effect of tumor cell-derived IL-1 depletion on the inflammatory secretome of CAFs, we sorted CAFs from scramble control and IL-1 -sh KRasG12D-PDEC pancreatic grafts using the fibroblast marker CD140 (Supplementary Fig. S3E) and analyzed them for expression of previously characterized cytokines (33) . Loss of tumor cell-derived IL-1 significantly downregulated the expression of several inflammatory cytokines, relative to scramble control (Fig. 3C).

Moreover, flow cytometric analysis of CD140a CAFs derived from IL-1 -sh KRasG12D-PDEC grafts showed a significant decrease in Ly6C expression, a surface glycoprotein that marks cytokine-producing

CAFs (34) (Supplementary Fig. S3F). Our results thus indicate that tumor cell-derived IL-1 promotes the activation and shapes the secretory phenotype of CAFs.

Many of the cytokines produced by CAFs in response to IL-1 are known modulators of immune cell function (35). We therefore postulated that tumor cell-derived IL-1 -mediated PSC activation may in turn contribute to the establishment of an immune suppressive TME. To test this hypothesis, we co-implanted immortalized CAFs isolated from KPC tumors (36) with IL-1 -sh KRasG12D-PDEC in wild type mice pancreata. Successful co-implantation was verified by restoration of stromal -SMA staining (Fig. 3D),

+ + increase in Ly6C CD140a cell population (Supplementary Fig. S3F) and increased collagen deposition

(Supplementary Fig. S3G). Significantly, CAF co-implantation with IL-1 -sh KRasG12D-PDEC specifically rescued the decrease in macrophage recruitment and M2 TAM polarization induced by loss of tumor cell-derived IL-1 (Fig. 3E and F; Supplementary Fig S3H-S3K) as well as restored the inactive state of CD8+ Tc cells and the decrease in CD8+ Tc cell infiltration (Fig. 3G and H). This phenotype is consistent with the observed IL-1 -dependent production by CAFs of CCL2 and CCL5 (Fig. 3C), which promote macrophage infiltration and M2 polarization (37,38), and CXCL12 (Fig. 3C), which is known to

+ + impede tumor infiltration of CD8 Tc cells (39). To determine the functional significance of CD8 T cell exclusion in the pro-tumorigenic role of tumor cell-derived IL-1 , we depleted CD8+ T cells in mice prior to orthotopic implantation of scramble or IL-1 -sh KRasG12D-PDECs (Supplementary Fig. S4A). CD8+ T

15

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cell depletion completely rescued tumor growth defect of IL-1 -sh KRasG12D-PDEC pancreatic grafts

(Fig. 4A and B; Supplementary Fig. S4B) indicating that the oncogenic role of tumor cell-derived IL-1 is mediated through immune suppression of CD8+ T cell infiltration and activity.

IL-1 neutralization sensitizes pancreatic tumors to anti-PD-1 checkpoint therapy.

The poor response of pancreatic tumors to immune checkpoint blockade has been primarily attributed to its immunosuppressive microenvironment and poor CD8+ T cell infiltration (3). Since depletion of tumor- derived IL-1 significantly increases CD8+ T cell infiltration and activity, we reasoned that IL-1 neutralization may sensitize PDA tumors to PD-1 checkpoint blockade. To this end, orthotopic KPC tumor bearing mice were treated with neutralizing antibodies against IL-1 and PD-1 (Fig. 4C). Indeed, addition of -IL-1 treatment significantly enhanced the anti-tumor activity of -PD-1 (Fig. 4D;

Supplementary Fig. S4C). As predicted, combined treatment of -IL-1 and -PD-1 resulted in increased tumor infiltration of CD8+ T cells, relative to vehicle control or -PD-1 alone (Fig. 4E).

IL-1 production in pancreatic tumor cells is mediated by the NLRP3 inflammasome.

Having established its importance in pancreatic tumorigenesis, we next wanted to dissect the molecular pathway regulating IL-1 production in tumor cells. In innate immune cells, IL-1 mRNA is translated to produce an inactive precursor pro-IL-1 form, which is further processed to yield the mature secreted form of the cytokine by a multimeric protein complex called the inflammasome (6). The most well characterized inflammasomes are comprised of a Nod-like receptor protein family pyrin-domain containing (NLRP) protein that serves as an activation sensor, which associates with apoptosis-associated speck-like proteins containing a CARD complex (ASC) protein (40). In complex with NLRP, ASC recruits pro-caspase-1 which auto-catalyses its cleavage to active caspase-1. Active caspase-1 in turn cleaves pro-IL-1 to produce the functional IL-1 protein. Of the various NLRP proteins that can form inflammasomes, the NLRP3 inflammasome appears most relevant to our study since its activation was found to be necessary for induction of pancreatitis (41), a major risk factor for PDA development.

16

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Additionally, NLRP3 inflammasome activity is associated with malignancies such as colon cancer and melanoma (42). We therefore analyzed the activation status of the NLRP3 inflammasome axis in the pancreatic tumor epithelium. A robust presence of NLRP3 was detected in the tumor epithelial compartment of KC mice pancreata (Fig. 5A). NLRP3 expression in these tumor cells strongly correlated with the expression of cleaved caspase-1 (Fig. 5B), a product of active inflammasomes. Moreover,

NLRP3 was found to co-localize with phospho-ASC (Y223) in speck-like aggregates (43) in tumor cells of both KC and KPC mouse pancreata (Fig. 5C; Supplementary Fig. S5A) as well as in human PDA samples (Fig. 5D), further validating the presence of active NLRP3 inflammasomes in these cells.

To determine whether the NLRP3 inflammasome is the primary source of pro-IL-1 processing in tumor cells, we knocked down NLRP3 expression in KRasG12D-PDEC and KPC cells using two independent short hairpins (Supplementary Fig. S5B and S5C). Depletion of NLRP3 in the transformed ductal epithelia significantly reduced cleaved caspase-1 expression and IL-1 production in IL-1 -sh KRasG12D-

PDEC pancreatic grafts, relative to scramble control (Fig. 5E-G; Supplementary Fig. S5D). This was accompanied by a decrease in growth of IL-1 -sh KRasG12D-PDEC pancreatic grafts (Fig. 5H;

Supplementary Fig. S5E) as well as decreased tumor growth and increased overall survival of orthotopic

IL-1 -sh KPC tumor-bearing mice (Fig. 5I-K). Together, these results implicate the NLRP3 inflammasome in the production of tumor cell-derived IL-1 and define a tumor-supportive role for

NLRP3 in pancreatic cancer.

Tumor-derived IL-1 expression is regulated by TLR4 and the pancreatic microbiome.

NLRP3 inflammasome assembly and the post-translational processing of IL-1 that ensues is predominantly regulated by toll-like receptors (TLRs) through induction of IL1B and NLRP3 expression in response to pathogens or cellular damage (44). Members of the TLR family are expressed in various cancers and have been shown to promote tumor growth (45). In pancreatic cancer, while most TLRs have been shown to be expressed largely in stromal cells, elevated TLR4 expression has been reported in the

17

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

tumor cell compartment of patient PDA samples and shown to be correlated with reduced survival

(46,47). Consistent with these findings, analysis of a panel of patient PDA samples revealed significant upregulation of tumor-associated TLR4 expression, relative to adjacent normal tissue (Fig. 6A), and immunohistochemical analysis demonstrated robust TLR4 expression in tumor cells in human PDA as well as KC and KPC mouse pancreata (Fig. 6B and C; Supplementary Fig. S6A). To determine whether

IL-1 production in tumor cells is TLR4-driven, we employed RNAi to stably knockdown TLR4 expression in KRASG12D-PDEC (Supplementary Fig. S6B). Transformed ductal epithelium of TLR4-sh

KRasG12D-PDEC pancreatic grafts had significantly reduced IL-1 production, relative to scramble control

(Fig. 6D and E). Moreover, TLR4 knockdown in KRasG12D-PDEC and KPC cells decreased growth of orthotopic KRasG12D-PDEC pancreatic grafts (Fig. 6F; Supplementary Fig. S6C) and KPC tumors, respectively (Fig. 6G and H; Supplementary Fig. S6D) and increased overall survival of TLR4-sh KPC orthotopic tumor bearing mice, relative to scramble control (Fig. 6I). We conclude TLR4 thus serves as a critical regulator of tumor cell-derived IL-1 production and pancreatic tumorigenesis.

Having identified TLR4 as the receptor that controls IL-1 production in pancreatic tumor cells, we next searched for possible cues in the pancreatic microenvironment that can induce TLR4 signaling. Recent reports on the existence of a complex pancreatic microbiome (23) prompted us to hypothesize that microbial-derived ligands which are known to activate TLR4 signaling (17) could be responsible for inducing IL-1 production in pancreatic tumor cells. To test this hypothesis, we treated wild-type mice with an antibiotic cocktail for 3 weeks to ablate their microbiome prior to implantation of KRasG12D-

PDEC (Supplementary Fig. S6E). KRasG12D-PDEC grafts formed in antibiotic-treated mice indeed displayed a significant reduction in tumor cell IL-1 expression without affecting TLR4 level, relative to vehicle-treated control mice (Fig. 6J and K; Supplementary Fig. 6F). Overall, our results indicate a role for the pancreatic microbiome in initiating a signaling cascade, likely through TLR4, to activate IL-1 production in pancreatic tumor cells.

18

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

DISCUSSION

In the present study, we demonstrate a role for tumor cell-derived IL-1 in promoting pancreatic oncogenesis by paracrine induction of heterotypic stromal interactions. Specifically, we show that tumor- derived IL-1 is critical for shaping the tolerogenic immune landscape of PDA by promoting stromal accumulation of immunosuppressive cell populations. These include M2 polarized macrophages, tumor-

hi + associated neutrophils, IL-17 producing TH17 cells, MDSCs and CD1d CD5 regulatory B cells. In addition, we report that tumor-derived IL-1 regulates PDA-associated desmoplasia by promoting activation of quiescent pancreatic stellate cells.

IL-1 is a member of the IL-1 family of pro-inflammatory cytokines, which also includes the co-founding member, IL-1 (7) Both IL-1 and IL-1 are critical immune regulators that signal through a common cell surface receptor (IL1R1-IL1RAcP) to activate two main pathways: IKK NF-

MAPK/JNK/ERK (7). Despite considerable functional homology, the two cytokines differ appreciably in several aspects. While IL-1 is predominantly a cytosolic or membrane-bound protein constitutively expressed in epithelial, endothelial and immune cells, IL-1 is a secretory protein chiefly produced by immune cells only in response to inflammatory cues (7). In PDA, previous studies have predominantly categorized IL-1 signaling into tumor cell production of IL-1 (48) and stromal production of IL-1 (16-

18). In fact, IL-1 protein is reportedly undetectable in PDA cell lines and organoids in vitro (33,49).

Consistent with these reports, we too did not detect IL-1 production by KRasG12D-PDEC and KPC cells cultured ex vivo. We did however observe a robust in vivo production of the IL-1 protein in the tumor cell compartment of human and murine PDA. This suggests the existence within the pancreatic tumor microenvironment of regulatory cues that can induce the activation of the toll-like receptor signaling pathway in tumor cells to drive IL1B expression and post-translational processing. This conclusion is

19

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

supported by our finding that the TLR4/NLRP3 inflammasome signaling axis is active in pancreatic tumor cells and is required for the production of IL-1 by these cells.

The TLR4 receptor is a specific sensor of exogenous microbial ligands such as lipopolysaccharides (LPS) as well as endogenous ligands termed damage-associated molecular patterns (DAMPs), derived from host tissue or cells (46). Significantly, the pancreatic microenvironment has been shown to be rich in such endogenous TLR4 ligands including HMGB-1 and S100A9 which can activate TLR4 signaling in tumor cells (17). Furthermore, the recently described PDA-associated microbiome has been shown to be rich in microbial ligands capable of activating the toll-like receptor pathway (23). Our finding that bacterial dysbiosis leads to inhibition of tumor cell-derived IL-1 production indicates that the pancreatic microbiome plays a significant role in inducing IL-1 production in transformed cells, likely through the toll-like receptor pathway. Moreover, it raises the possibility that the pro-oncogenic role of the microbiome in pancreatic cancer (23) could be in part mediated by the activation of TLR4-mediated IL-

1 production in the tumor cell compartment.

We have established that the post-translational mechanisms that drive IL-1 processing and maturation in the tumor cells require the NLRP3 inflammasome. The precise nature of the TLR4-induced signals that promote the assembly and activation of the NLRP3 inflammasome in pancreatic tumor cells remain to be determined. In monocytes, TLR4 ligation can induce ATP release which in turn triggers NLRP3 inflammasome assembly via the ATP-gated ionotropic receptor P2X7 (P2X7R) (40). Interestingly,

P2X7R is highly expressed in pancreatic cancer cells (50) suggesting that a co-option by the tumor cells of this immune cell-specific signaling axis might be responsible for the NLRP3 inflammasome activation.

As indicated by our loss of function and rescue experiments, the secretion by pancreatic tumor cells of IL-

1 instigates sweeping changes in the fibro-inflammatory pancreatic milieu in part by modulating PSC function. PSCs have been implicated in the regulation of a plethora of pro-tumorigenic processes including tumor cell growth and metabolic adaptation and metastasis (31,32). Recently, Öhlund et al.

20

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

have described the presence of two distinct intra-tumoral CAFs subpopulations in PDA: myofibroblastic cancer-associated fibroblasts (myCAF) with elevated -SMA expression and inflammatory cancer- associated fibroblasts CAF (iCAF) expressing an array of cytokines and chemokines (51). Our data indicate a role for IL-1 in regulating the secretory phenotype of inflammatory CAFs. Specifically, we demonstrate the dependence of stromal CASFs on tumor-derived IL-1 for the production of cytokines and chemokines with documented roles in subverting anti-tumor immunity. These include the chemokines

CCL2 and CCL5 which regulate chemotaxis of monocytes, and, in the context of pancreatic cancer, have been found to regulate macrophage infiltration and M2 polarization (37,38), as well as the chemokine

CXCL12 which is known to inhibit intra-tumoral accumulation of CD8+ T cells (39). In accordance with this IL-1 -dependent secretory profile, we found that upregulation of M2-TAMs and restriction of CD8+

Tc cell tumor infiltration is dependent on stromal PSCs. Overall, our study delineates epistatic interactions between tumor cell-derived IL-1 and PSCs that are critical for the establishment of immune tolerance in pancreatic cancer.

+ The low immunogenicity of pancreatic cancer due to poor tumor-infiltration of CD8 Tc cells is considered a major factor responsible for the failure of checkpoint immunotherapy in PDA (3). As

+ demonstrated by our studies, neutralizing IL-1 promotes intra-tumoral CD8 Tc cell infiltration and function and sensitizes PDA to checkpoint immunotherapy. Hence therapeutic strategies that target IL-1 may increase the efficacy of immune checkpoint inhibitors in pancreatic cancer. It is noteworthy that in a recent analysis of pancreatic cystic (PCN) in patients, intracystic bacterial load as well as increased IL-1 protein levels were detected in cystic precursors to pancreatic cancer called intraductal papillary mucinous neoplasms (IPMNs), relative to non-IPMN PCNs (52). This study in combination with our current findings suggest that IL-1 production might be an early event in pancreatic tumorigenesis, the targeting of which could be used to impede disease progression.

21

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Acknowledgement

The authors thank L.J. Taylor for help with article preparation, Mark Choi for technical assistance and members of the Bar-Sagi lab for valuable discussions and comments. The KPC cell line (line 4662) was a kind gift from Dr. R.H. Vonderheide. The immortalized CAF cell line isolated from KPC tumors was a kind gift from Dr. A.C. Kimmelman. We thank Novartis for providing the mouse anti-PD1 and anti-IL- antibodies and isotype controls for this study. The TROMA-I antibody against cytokeratin-8 developed by Brulet P et al., Institut Pasteur was obtained from the Developmental Studies Hybridoma Bank, created by the NICHD of the NIH and maintained at The University of Iowa, Department of Biology.

boratory and the Experimental

Pathology Research Laboratory, which are supported in part by grant P30CA016087 from the National

Institutes of Health/National Cancer Institute, for providing cell sorting/flow cytometry technologies and histochemistry support respectively as well as the Center for Biospecimen Research and

Development for providing patient pancreatic tumor tissue sections. This work was supported by

NIH/NCI grant CA210263 (D. Bar-Sagi) and by a Stand Up To Cancer-Lustgarten Foundation

Pancreatic Cancer Convergence Dream Team Name Translational Research Grant (SU2C-AACR-

DT14-14). Stand Up to Cancer is a division of the Entertainment Industry Foundation administered by the American Association for Cancer Research, the Scientific Partner of SU2C. B. Shapiro was supported by NIH grant T32GM115313. E.A. Vucic was supported by a Canadian Institutes of Health

Research Fellowship (146792).

22

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

REFERENCES

1. Simoes PK, Olson SH, Saldia A, Kurtz RC. Epidemiology of pancreatic adenocarcinoma. Chin Clin Oncol 2017;6:24. 2. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691-703. 3. Johnson BA, 3rd, Yarchoan M, Lee V, Laheru DA, Jaffee EM. Strategies for Increasing Pancreatic Tumor Immunogenicity. Clin Cancer Res 2017;23:1656-69. 4. Cullis J, Das S, Bar-Sagi D. Kras and Tumor Immunity: Friend or Foe? Cold Spring Harb Perspect Med 2018;8 5. Padoan A, Plebani M, Basso D. Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int J Mol Sci 2019;20 6. Bent R, Moll L, Grabbe S, Bros M. Interleukin-1 Beta-A Friend or Foe in Malignancies? Int J Mol Sci 2018;19 7. Apte RN, Dotan S, Elkabets M, White MR, Reich E, Carmi Y, et al. The involvement of IL-1 in tumorigenesis, tumor invasiveness, metastasis and tumor-host interactions. Cancer metastasis reviews 2006;25:387-408. 8. Chen L, Huang CF, Li YC, Deng WW, Mao L, Wu L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck . Cell Mol Life Sci 2018;75:2045-58. 9. Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. Inflammation induces myeloid- derived suppressor cells that facilitate tumor progression. Journal of immunology 2006;176:284- 90. 10. Jiang H, Gebhardt C, Umansky L, Beckhove P, Schulze TJ, Utikal J, et al. Elevated chronic inflammatory factors and myeloid-derived suppressor cells indicate poor prognosis in advanced melanoma patients. Int J Cancer 2015;136:2352-60. 11. Saijo Y, Tanaka M, Miki M, Usui K, Suzuki T, Maemondo M, et al. Proinflammatory cytokine IL-1 beta promotes tumor growth of Lewis lung carcinoma by induction of angiogenic factors: in vivo analysis of tumor-stromal interaction. Journal of immunology 2002;169:469-75. 12. Mayer J, Rau B, Gansauge F, Beger HG. Inflammatory mediators in human acute pancreatitis: clinical and pathophysiological implications. Gut 2000;47:546-52. 13. Maker AV, Katabi N, Qin LX, Klimstra DS, Schattner M, Brennan MF, et al. Cyst fluid interleukin- 1beta (IL1beta) levels predict the risk of carcinoma in intraductal papillary mucinous neoplasms of the pancreas. Clin Cancer Res 2011;17:1502-8. 14. Mitsunaga S, Ikeda M, Shimizu S, Ohno I, Furuse J, Inagaki M, et al. Serum levels of IL-6 and IL- 1beta can predict the efficacy of gemcitabine in patients with advanced pancreatic cancer. British journal of cancer 2013;108:2063-9.

23

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

15. Zhang D, Li L, Jiang H, Li Q, Wang-Gillam A, Yu J, et al. Tumor-Stroma IL1beta-IRAK4 Feedforward Circuitry Drives Tumor Fibrosis, Chemoresistance, and Poor Prognosis in Pancreatic Cancer. Cancer research 2018;78:1700-12. 16. Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M, et al. Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. Cancer Discov 2016;6:852-69. 17. Ochi A, Nguyen AH, Bedrosian AS, Mushlin HM, Zarbakhsh S, Barilla R, et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. The Journal of experimental medicine 2012;209:1671-87. 18. Daley D, Mani VR, Mohan N, Akkad N, Pandian G, Savadkar S, et al. NLRP3 signaling drives macrophage-induced adaptive immune suppression in pancreatic carcinoma. The Journal of experimental medicine 2017;214:1711-24. 19. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer cell 2003;4:437- 50. 20. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer cell 2005;7:469-83. 21. Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer cell 2012;21:836-47. 22. Tseng WW, Winer D, Kenkel JA, Choi O, Shain AH, Pollack JR, et al. Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clin Cancer Res 2010;16:3684-95. 23. Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov 2018;8:403-16. 24. Reikvam DH, Erofeev A, Sandvik A, Grcic V, Jahnsen FL, Gaustad P, et al. Depletion of murine intestinal microbiota: effects on gut mucosa and epithelial gene expression. PloS one 2011;6:e17996. 25. Apte M. Isolation of quiescent pancreatic stellate cells from rat and human pancreas Pancreapedia: Exocrine Pancreas Knowledge Base 2011;10.3998/panc.2011.10 26. Gadaleta E, Cutts RJ, Kelly GP, Crnogorac-Jurcevic T, Kocher HM, Lemoine NR, et al. A global insight into a cancer transcriptional space using pancreatic data: importance, findings and flaws. Nucleic Acids Res 2011;39:7900-7. 27. Pylayeva-Gupta Y, Das S, Handler JS, Hajdu CH, Coffre M, Koralov SB, et al. IL35-Producing B Cells Promote the Development of Pancreatic Neoplasia. Cancer Discov 2016;6:247-55. 28. Glaccum MB, Stocking KL, Charrier K, Smith JL, Willis CR, Maliszewski C, et al. Phenotypic and functional characterization of mice that lack the type I receptor for IL-1. Journal of immunology 1997;159:3364-71. 29. Chao T, Furth EE, Vonderheide RH. CXCR2-Dependent Accumulation of Tumor-Associated Neutrophils Regulates T-cell Immunity in Pancreatic Ductal Adenocarcinoma. Cancer Immunol Res 2016;4:968-82. 30. Arnold JN, Magiera L, Kraman M, Fearon DT. Tumoral immune suppression by macrophages expressing fibroblast activation protein-alpha and heme oxygenase-1. Cancer Immunol Res 2014;2:121-6. 31. Mekapogu AR, Pothula SP, Pirola RC, Wilson JS, Apte MV. Multifunctional role of pancreatic stellate cells in pancreatic cancer. J Annals of Pancreatic Cancer 2019;2:10.

24

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

32. Fu Y, Liu S, Zeng S, Shen H. The critical roles of activated stellate cells-mediated , metabolism and onco-immunology in pancreatic ductal adenocarcinoma. Mol Cancer 2018;17:62. 33. Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, Tiriac H, et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2014;159:80-93. 34. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, et al. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFbeta to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov 2019;9:282-301. 35. Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nature reviews Immunology 2017;17:559-72. 36. Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman MH, Zhang L, et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016;536:479- 83. 37. Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 2013;19:3404-15. 38. Aldinucci D, Colombatti A. The inflammatory chemokine CCL5 and cancer progression. Mediators Inflamm 2014;2014:292376. 39. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP- expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2013;110:20212-7. 40. He Y, Hara H, Nunez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci 2016;41:1012-21. 41. Zhang GX, Wang MX, Nie W, Liu DW, Zhang Y, Liu HB. P2X7R Blockade Prevents NLRP3 Inflammasome Activation and Pancreatic Fibrosis in a Mouse Model of Chronic Pancreatitis. Pancreas 2017;46:1327-35. 42. Zhiyu W, Wang N, Wang Q, Peng C, Zhang J, Liu P, et al. The inflammasome: an emerging therapeutic oncotarget for cancer prevention. Oncotarget 2016;7:50766-80. 43. Hara H, Tsuchiya K, Kawamura I, Fang R, Hernandez-Cuellar E, Shen Y, et al. Phosphorylation of the adaptor ASC acts as a molecular switch that controls the formation of speck-like aggregates and inflammasome activity. Nature immunology 2013;14:1247-55. 44. Netea MG, van de Veerdonk FL, Kullberg BJ, Van der Meer JW, Joosten LA. The role of NLRs and TLRs in the activation of the inflammasome. Expert Opin Biol Ther 2008;8:1867-72. 45. Dajon M, Iribarren K, Cremer I. Toll-like receptor stimulation in cancer: A pro- and anti-tumor double-edged sword. Immunobiology 2017;222:89-100. 46. Vaz J, Andersson R. Intervention on toll-like receptors in pancreatic cancer. World journal of gastroenterology 2014;20:5808-17. 47. Zhang JJ, Wu HS, Wang L, Tian Y, Zhang JH, Wu HL. Expression and significance of TLR4 and HIF- 1alpha in pancreatic ductal adenocarcinoma. World journal of gastroenterology 2010;16:2881-8. 48. Ling J, Kang Y, Zhao R, Xia Q, Lee DF, Chang Z, et al. KrasG12D-induced IKK2/beta/NF-kappaB activation by IL-1alpha and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. Cancer cell 2012;21:105-20. 49. Tjomsland V, Spangeus A, Valila J, Sandstrom P, Borch K, Druid H, et al. Interleukin 1alpha sustains the expression of inflammatory factors in human pancreatic cancer microenvironment by targeting cancer-associated fibroblasts. Neoplasia 2011;13:664-75. 50. Giannuzzo A, Pedersen SF, Novak I. The P2X7 receptor regulates cell survival, migration and invasion of pancreatic ductal adenocarcinoma cells. Mol Cancer 2015;14:203.

25

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

51. Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and in pancreatic cancer. The Journal of experimental medicine 2017;214:579-96. 52. Gaiser RA, Halimi A, Alkharaan H, Lu L, Davanian H, Healy K, et al. Enrichment of oral microbiota in early cystic precursors to invasive pancreatic cancer. Gut 2019;68:2186-94.

26

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

FIGURE LEGENDS

Figure 1. Tumor cell-derived IL-1 is required for pancreatic oncogenesis. A. mRNA transcript levels of IL-1 in PDA compared to normal adjacent tissue from publicly available human transcriptomic data (74 patients: n=50 PDA and n=24 adjacent normal). Each data point indicates an individual tissue sample. Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). B.

Representative immunohistochemical detection of IL-1 expression in sections from human PDA and adjacent normal tissues (n=10 patient samples). C. Representative immunofluorescence detection of IL-

1 expression on a section from a 4 month-old KC mouse pancreas (N=12; CK8 red; IL-1 green; and

DAPI blue). D. Representative immunofluorescence detection of IL-1 expression on a section from a 2- month-old KPC mouse pancreas (N=8 at 2-4 mo; CK8, red; IL-1 green; DAPI, blue). E. Quantification of flow cytometric analysis of distribution of IL-1 expression in epithelial (EpCAM+), fibroblast

(CD140a+) and immune (CD45+) cells sorted from pancreata of 1-2 month-old KC mice (N=7). Error bars indicate SD; P-values determined by Student t test (two-tailed, unpaired). F. Graph indicates quantification of percentage of CK8+ signal per lesion from immunohistochemical staining with CK8 antibody on sections of orthotopic pancreatic grafts 2 weeks after implantation of KRasG12D-PDEC expressing scrambled shRNA (scr-sh) control or IL-1 shRNAs (IL-1 -sh1, IL-1 -sh2) in wild type

(WT) mice. (N=8-9). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 3 independent experiments. G. Representative tumors 4 weeks after orthotopic implantation of KPC cells expressing scrambled shRNA (scr-sh) or IL-1 shRNA (IL-1 -sh) in pancreata of WT mice. H. Graph represents quantification of G, indicating tumor volume (N=15).

Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 3 independent experiments. I. Kaplan Meier curve for survival analysis of mice bearing pancreatic tumors derived from orthotopically implanted scr-sh or IL-1 -sh KPC cells (N=8). P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments.

p < 0.001; p < 0.0001.

27

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2. Tumor cell-derived IL-1 promotes immune suppression in the pancreatic tumor microenvironment. A-C. Representative flow cytometry plots and quantification (graph, bottom) from orthotopic pancreatic grafts of scr-sh or IL-1 -sh KRasG12D-PDEC in WT mice 2 weeks post-implantation analyzing A, CD11b+F4/80+ tumor-associated macrophages, B, CD11b+Gr1+ myeloid-derived suppressor cells (MDSC), and C, CD11b+Ly6G+ tumor-associated neutrophil populations. For A-C, graphs indicate immune sub-populations as a percentage of CD45+ cells (N=5-8). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments.

D-F. Representative flow cytometry plots and quantification (graph, bottom) from orthotopic pancreatic grafts of scr-sh or IL-1 -sh KRasG12D-PDEC in WT mice 2 weeks post-implantation analyzing D, CD5+

+ hi + regulatory B (Breg) cells measured as a percentage of total CD19 CD1d B cells (N=6), E, ROR t TH17

+ + cells measured as a percentage of total CD4 TH cells (N=8-9), F, CD206 M2 polarized macrophages measured as a percentage of total stromal macrophages (N=5-6). Data representative of 2 (D, E) or 3 (F) independent experiments. For D-F, error bars indicate SD; P-values determined by the Student t test (two- tailed, unpaired). G. Immunohistochemical (IHC) detection of CD8+ Tc cells on sections of scr-sh and IL-

1 -sh KRasG12D-PDEC grafts in WT mice, 2 weeks post-orthotopic implantation. Representative images are shown. Graph depicts quantification of IHC, indicating the average percentage of CD8+ cells per field of view (FOV) of the implant (4-6 10X FOV per animal, N=5). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments.

H-I. Representative flow cytometry plots and quantification (graph, right) from orthotopic pancreatic grafts of scr-sh or IL-1 -sh KRasG12D-PDEC in WT mice 2 weeks post-implantation analyzing activated cytotoxic CD8+ T cells (Tc) as measured by H, IFN and I, Granzyme B (GzmB) expression.

Quantification of IFN + and GzmB+ cells is represented as a percentage of total CD8+ Tc cells (N=5-7).

Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. p < 0.05; p < 0.01; p < 0.001; p < 0.0001.

28

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 3. Tumor cell-derived IL-1 regulates pancreatic stellate cell activation and inflammatory functions. A. Immunohistochemical detection of -SMA and vimentin expression on sections of scr-sh and IL-1 -sh KRasG12D-PDEC grafts in WT mice, 2 weeks post-orthotopic implantation (N=7).

Representative images are shown. B. Immunofluorescence detection of -SMA expression on sections of scr-sh and IL-1 -sh KPC tumors in WT mice, 4 weeks post-orthotopic implantation (N=10; CK8, red; -

SMA, green; DAPI blue). Representative images are shown. C. Levels of cytokine mRNA in CD140a+

CAFs sorted from scr-sh or IL-1 -sh KRasG12D-PDEC grafts in WT mice, 2 weeks post-orthotopic implantation (N=9) analyzed by quantitative PCR. Results show mean ± SD of three biological replicates, each with 3 technical replicates. P-values determined by Student t test (two-tailed, unpaired). D.

Immunofluorescence detection of -SMA expression on sections of orthotopic pancreatic grafts of scr-sh,

IL-1 -sh KRasG12D-PDEC and IL-1 -sh KRasG12D-PDEC co-implanted with CAFs in WT mice, 2 weeks post-implantation (N=7; CK8, red; -SMA, green; DAPI, blue). Representative images are shown. E-G.

Representative flow cytometry plots (left) and quantification (graph, right) from orthotopic pancreatic grafts 2 weeks after implantation of scr-sh, IL-1 -sh KRasG12D-PDEC and IL-1 -sh KRasG12D-PDEC co- implanted with CAFs of E, CD11b+F4/80+ macrophages measured as a percentage of total CD45+ cells

(N=7), F, CD206+ M2 polarized macrophages measured as a percentage of total stromal macrophages

(N=7), G, activated cytotoxic CD8+ T cells measured by IFN + cells as a percentage of total CD8+ Tc cells (N=7). For E-G, error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. H. Immunohistochemical (IHC) detection of

CD8+ T cells on sections of pancreatic grafts of scr-sh, IL-1 -sh KRasG12D-PDEC and IL-1 -sh KRasG12D-

PDEC co-implanted CAFs, 2 weeks post-orthotopic implantation (N=7). Representative images are shown. Graph depicts quantification of IHC, indicating the average percentage of CD8+ cells per FOV of the implant (4-6 10X FOV per animal, N=7). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. p < 0.05; p < 0.01; p <

0.001; p < 0.0001; NS, not significant.

29

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 4. IL-1 neutralization sensitizes PDA tumors to PD-1 checkpoint blockade. A. Schematic of anti-CD8 antibody treatment regimen. Anti-CD8 or control IgG antibody (red arrow) was administered every day for three days prior to orthotopic implantation of KrasG12D-PDECs, and then every five days hence. B. Graph depicts quantification of immunohistochemical analysis of CK8 staining on sections of orthotopic pancreatic grafts 2 weeks post-implantation of scr-sh or IL-1 -sh KRasG12D-PDEC and indicates percentage of CK8+ signal per lesion (N=8). Error bars indicate SD; P-values determined by the

Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. C. Schematic of anti-IL-1 and anti-PD-1 antibody treatment regimen. Treatment was initiated one week post orthotopic implantation of KPC cells. Green and red arrows indicate anti-PD-1 and anti-IL-1 antibody administration, respectively. D. Graph represents quantification of analysis in C, indicating tumor weight

(N=8). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. E. Representative flow cytometry plots (left) of KPC tumors treated with vehicle control, anti-PD-1 antibody alone, anti-IL-1 antibody alone or both anti-PD-1 and anti-IL-1 antibody, indicating tumor infiltrating CD8+ T cells. Graphs depict quantitation of FACS analysis, represented as either percentage of CD45+ immune cells (top right, N=8) or absolute number of

CD8+ T cells relative to tumor weight (bottom right, N=7). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. p < 0.05;

p < 0.01; p < 0.001; p < 0.0001.

Figure 5. pro-IL-1 processing in pancreatic tumor cells is regulated by the NLRP3 inflammasome.

A-B. Immunohistochemical detection on sections of 2-4-month-old KC mouse pancreata of A, NLRP3 expression (N=8) and B, cleaved caspase-1 expression (N=8). Insets show respective isotype controls.

Representative images are shown. C. Immunofluorescence detection of NLRP3 and phospho-ASC

(Y144) co-localization in a 2-month-old KPC mouse pancreas (N=8; NLRP3 red, p-Asc green, DAPI blue). Representative image is shown. D. Immunofluorescence detection of NLRP3 and phospho-Asc

(Y144) co-localization on a section of human PDA tissue (N=8; NLRP3 red, p-Asc green, DAPI blue).

30

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Representative image is shown. 36 ±3.42 % ductal epithelium co-stained positively for NLRP3 and phospho-Asc, as measured using Image J (3-5 FOV/sample, N=8). E. Immunohistochemical detection of

NLRP3 and cleaved caspase-1 expression on sections of orthotopic pancreatic grafts 2 weeks post- implantation of KRasG12D-PDEC expressing scramble shRNA (scr-sh) control or NLRP3 shRNA

(NLRP3-sh) in WT mice (N=6). Representative images are shown. F. ELISA analysis for assessing IL-1 protein production in scr-sh or NLRP3-sh KRasG12D-PDEC in orthotopic pancreatic grafts 2 weeks post- implantation in WT mice (N=4). P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. G. Graph depicts quantification of immunofluorescence detection of IL-1 on orthotopic pancreatic graft sections, 2 weeks post-implantation of scr-sh or

NLRP3-sh KRasG12D-PDEC in WT mice. Represented as percentage of IL-1 positive epithelium per

FOV of the implant (4-6 FOV per animal, N=8). Error bars indicate SD; P-values determined by the

Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. H. Graph depicts quantification of immunohistochemical analysis of CK8 staining on sections of orthotopic pancreatic grafts 2 weeks post-implantation of scr-sh or NLRP3-sh KRasG12D-PDEC and indicates percentage of

CK8+ signal per lesion (N=8). Error bars indicate SD; P-values determined by the Student t test (two- tailed, unpaired). Data representative of 2 independent experiments. I. Representative tumors 4 weeks after orthotopic implantation of KPC cells expressing scrambled shRNA (scr-sh) or NLRP3 shRNA

(NLRP3-sh) in pancreata of WT mice. J. Graph represents quantification of I, indicating tumor volume

(N=7). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. K. Kaplan Meier curve for survival analysis of mice bearing orthotopically implanted pancreatic tumors derived from scr-sh or NLRP3-sh KPC cells (N=7). P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments.

p < 0.001; p < 0.0001.

Figure 6. The pancreatic microbiome drives IL-1 expression in tumor cells through TLR4 signaling. A. mRNA transcript level of TLR4 in PDA compared to normal adjacent tissue from publicly

31

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

available human transcriptomic data (74 patients: n=50 PDA and n=24 adjacent normal). Each data point indicates an individual tissue sample. Error bars indicate SD; P-values determined by the Student t test

(two-tailed, unpaired). B. Immunohistochemical detection of TLR4 expression on a section from human

PDA tissue (n=8 patient samples). Representative image is shown. 67 ± 8.94 % ductal epithelium co- stained positively for TLR4, as measured using Image J (3-5 FOV/sample, N=8). C.

Immunohistochemical detection of TLR4 expression on a section from a 4-month-old KC mouse pancreas

(N=8). Representative image is shown. D. Immunofluorescence detection of IL-1 expression on sections of orthotopic pancreatic grafts 2 weeks post-implantation of KRasG12D-PDEC expressing scramble shRNA

(scr-sh) control or TLR4 shRNA (TLR4-sh) in WT mice (N=7; CK8, red; IL-1 green; DAPI, blue).

Representative images are shown. E. Graph depicts quantification of data in D, represented as a percentage of IL-1 positive epithelium per FOV of the implant (4-6 FOV per animal, N=7). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. F. Graph depicts quantification of immunohistochemical analysis of CK8 staining on sections of orthotopic pancreatic grafts 2 weeks post-implantation of scr-sh or TLR4-sh

KRasG12D-PDEC, and indicates percentage of CK8+ signal per lesion (N=7). Error bars indicate SD; P- values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. G. Representative tumors 4 weeks after orthotopic implantation of KPC cells expressing scr- sh or TLR4-sh in pancreata of WT mice. H. Graph represents quantification of G indicating tumor volume (N=8). Error bars indicate SD; P-values determined by the Student t test (two-tailed, unpaired).

Data representative of 2 independent experiments. I. Kaplan Meier curve for survival analysis of mice bearing pancreatic tumors derived from orthotopically implanted scr-sh or TLR4-sh KPC cells (N=7). P- values determined by the Student t test (two-tailed, unpaired). Data representative of 2 independent experiments. J. Immunofluorescence analysis of IL-1 expression on sections of orthotopic pancreatic grafts 2 weeks post-implantation of KRasG12D-PDEC in WT mice treated with vehicle control or antibiotic cocktail (N=7; CK8 red, IL-1 green, DAPI blue). Representative images are shown. K. Graph depicts

32

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

quantification of data in J, represented as a percentage of IL-1 -positive epithelium per FOV of the implant (4-6 FOV per animal, N=7). Error bars indicate SD; P-values determined by the Student t test

(two-tailed, unpaired). p < 0.001; p < 0.0001.

33

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Figure 1

A B Adj Nml PDA 15 100 ****

**** 80 10 60 staining 40 5

IL-1 20

0 (% ductal epithelium/FOV) 0 Adj Nml PDA Adj Nml PDA C D E KC KPC 80 DAPI DAPI 60

40 expression IL-1 IL-1

20 of IL-1 % cellular distribution CK8 CK8 0 Epithelial Fibroblast Immune F G

50 **** *** IL-1 -sh 40 scr-sh 30 area/lesion + 20

10 % CK8 0 scr-sh IL-1 - IL-1 - sh1 sh2 H I 2000

) scr-sh IL-1 -sh 3 *** 100 1500

1000 **p=0.002 50 500 Percent survival Tumor volumeTumor (mm 0 0 scr-sh IL-1 -sh 0 20 40 60 80 Days

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2

A scr-sh B scr-sh C scr-sh D scr-sh E scr-sh F scr-sh 12.50 12.50 12.1 55.90 11.1 59.70

IL-1 -sh1 IL-1 -sh1 IL-1 -sh1 IL-1 -sh1 IL-1 -sh1 IL-1 -sh1 4.92 7.517.51 5.00 29.70 4.09 31.50

IL-1 -sh2 IL-1 -sh2 IL-1 -sh2 IL-1 -sh2 IL-1 -sh2 IL-1 -sh2 4.26 5.57 5.36 26.60 4.96 26.60 F4/80 CD1d FSC-H Gr1 FSC-H Ly6G Ly6G CD11b CD11b CD11b CD5 ROR t CD206

* ) + ) * )

* + * * 30 30 30 hi 25 * 60 ** 80 * + + * + + + 20 * 20 20 reg 20 60 CD11b

CD1d 40 * B +

+ 15 +

* (%CD4 40 + 10 10 10 10 t 20 % CD45 % CD45 % CD45 20 CD5 5 0 0 0 0 0 0 ROR (%CD19 hi + (% F4/80 macrophage MDSC neutrophil CD1d CD5 TH17 CD206 M2 TAMs Breg scr-sh IL-1 -sh1 IL-1 -sh2 G H CD8; 20X scr-sh IL-1 -sh1 IL-1 -sh2 ) + 25 ** 2.32 12.50 12.40 20 * 15

(%CD8 10 + scr-sh 5 FSC-H

IFN 0 IFN + **** IFN TC 400 **** 300 I scr-sh IL-1 -sh1 IL-1 -sh2 ) + -sh1 60 **** 200 4.05 31.3 24.9 *** cells/FOV c

T 40

IL-1 100 + (%CD8

0 + + 20 CD8 CD8 TC

FSC-H 0 GzmB + -sh2 GzmB GzmB TC IL-1

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Figure3 H D A

Downloaded from CD8; 20X IL-1 -sh+CAF IL-1 -sh scr-sh IL-1 -sh2 IL-1 -sh1 scr-sh CK8 Author manuscriptshavebeenpeerreviewedandacceptedforpublicationbutnotyetedited. Author ManuscriptPublishedOnlineFirstonJanuary8,2020;DOI:10.1158/0008-5472.CAN-19-2080 -SMA scr-sh -SMA DAPI cancerres.aacrjournals.org G E F FSC-H FSC-H F4/80 IFN CD206 CD206 CD11b scr-sh scr-sh c-hIL-1 scr-sh

IL-1 vimentin 85.90 3.96 18.50 -sh on September 26, 2021. © 2020American Association for Cancer Research. IL-1 IL-1 C B -sh -sh s IL-1 -sh 23.30 51.50

IL-1 mRNA fold change 4.53 0.0 0.5 1.0 1.5 CK8 -SMA DAPI CCL2 CCL5 CCL8 CCL12 IL-6 IL-6 IL-1 CCL8CCL12 CCL2 CCL5 -sh+CAF *** IL-1 IL-1 KPC -sh+CAF -sh+CAF -sh+CAF -scr-sh * 83.80 5.82 14.80 scr-sh * CD8+ T cells/FOV

100 200 c 300 400 500 + CD206+ + + 0 IFN F4/80 CD11b + + + + * (%CD8 ) (% F4/80 CD11b 100 ) (%CD45 cells) 10 20 30 10 20 30 20 40 60 80 0

0 0 s

KPC

c

r -

*

s

s s s

s s s

IL-1

h

c c c

****

h h h

-IL-1

r r r

- - - ** ** ** IL-1 IL-1 IL-1 IL-1 ** -sh NS -sh s IL-1 -sh s IL-1 -sh s IL-1 -sh s IL-1 -sh **** ****

* * CXCL12 ** +CAF +CAF +CAF +CAF -sh -sh -sh -sh Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Figure 4 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A - -CD8 or C - -PD-1 - -IL-1 Isotype KRasG12D-PDEC KPC implantation Analysis implantation Analysis

B D ** 40 **** 500 ** **** * 400 30 300 20 area/lesion

+ 200 10 100 tumor weight (mg) % CK8 0 0 scr-sh IL-1 -sh IL-1 -sh Veh -PD-1 -IL-1 -PD-1 + -CD8 + -IL-1 E 20 *** Vehicle -PD-1 *** 15

c * T 5.59 7.11 cells) + ** + 10

CD8 5 (%CD45

FSC-H 0 CD8 Veh -PD-1 -IL-1 -PD-1 + -IL-1 -IL1 -PD-1 + -IL-1 * 150 * 9.01 15 100 * /tumor weight

C * 50 T + FSC-H CD8 0 #CD8 Veh -PD-1 -IL-1 -PD-1 + -IL-1

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5

A C E KC NLRP3 p-ASC DAPI scr-sh NLRP3-sh KPC

Isotype B D KC NLRP3 p-ASC DAPI HumanPDA

Isotype F G H **** 1.5 80 **** 40 **** **** ****

protein ** 60 30 1.0

40 20 area/lesion + epithelium/FOV

0.5 + 20 10 % CK8 %IL-1 Foldchange IL-1 0.0 0 0 scr-shNLRP3 NLRP3 scr-shNLRP3 NLRP3 scr-sh NLRP3 NLRP3 sh1 sh2 sh1 sh2 sh1 sh2 I J K 2000 scr-sh NLRP3-sh )

3 100 *** **p=0.001 NLRP3-sh 1500

scr-sh 1000 50

500 Percent survival Tumor volumeTumor (mm 0 0 scr-sh NLRP3-sh 0 20 40 60 80 Days

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 FigureAuthor 6 manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A 8 **** B PDA C KC

6

4

2

0 Nml PDA D E F scr-sh TLR4-sh

80 60 **** ****

DAPI 60 40 40 area/lesion IL-1 + epithelium/FOV

+ 20 20 % CK8 CK8 %IL-1 0 0 scr-sh TLR4-sh scr-sh TLR4-sh G H I 2000 scr-sh TLR4-sh )

3 100 *** *p=0.016 TLR4-sh 1500

scr-sh 1000 50

500 Percent survival Tumor volume(mm Tumor 0 0 scr-sh TLR4-sh 0 20 40 60 80 Days J K Vehicle Antibiotic 80 ****

60 DAPI

40 epithelium/FOV IL-1 + 20 %IL-1 CK8 0 Vehicle Antibiotic

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 8, 2020; DOI: 10.1158/0008-5472.CAN-19-2080 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Tumor Cell-Derived IL-1β Promotes Desmoplasia and Immune Suppression in Pancreatic Cancer

Shipra Das, Beny Shapiro, Emily A Vucic, et al.

Cancer Res Published OnlineFirst January 8, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-2080

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2020/01/07/0008-5472.CAN-19-2080.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://cancerres.aacrjournals.org/content/early/2020/01/07/0008-5472.CAN-19-2080. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research.