Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Cancer Research (Research Article)

IL33 is a key driver of treatment resistance of cancer

Chie Kudo-Saito1, Takahiro Miyamoto1,2, Hiroshi Imazeki1,2, Hirokazu Shoji2,

Kazunori Aoki1, Narikazu Boku2

1 Department of Immune Medicine, National Cancer Center Research Institute,

Tokyo, Japan

2 Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital,

Tokyo, Japan

Running title: IL33 in evolutional transformation of cancer

Corresponding author: Chie Kudo-Saito, Department of Immune Medicine,

National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo

104-0045, Japan. Phone: +81-3-3542-2511; E-mail: [email protected].

Conflicts of interest: The authors have no competing financial interests.

1

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

Recurrence and treatment resistance are major causes of cancer-associated death.

There has been a growing interest in better understanding epithelial-mesenchymal transition (EMT), stemness of cancer cells, and exhaustion and dysfunction of the immune system for which numerous genomic, proteomic, microenvironmental, and immunological mechanisms have been demonstrated. However, practical treatments for such patients have not yet been established. Here we identified -33 (IL33) as a key driver of polyploidy followed by rapid proliferation after treatment. IL33 induction transformed tumor cells into polyploid giant cells, showing abnormal cell cycle without cell division accompanied by Snail deregulation and p53 inactivation; small progeny cells were generated in response to treatment stress.

Simultaneously, soluble IL33 was released from tumor cells, leading to expansion of ST2-expressing cells including IL17RB+GATA3+ cells, which promoted tumor progression and metastasis directly and indirectly via induction of immune exhaustion and dysfunction. Blocking IL33 with a specific mAb in murine IL33+ metastatic tumor models abrogated negative consequences and successfully elicited anti-tumor efficacy induced by other combined treatments. Ex vivo assays

2

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

using tumor tissues and PBMCs of cancer patients validated the clinical relevancy of these findings. Together, these data suggest that targeting the IL33-ST2 axis is a promising strategy for diagnosis and treatment of patients likely to be resistant to treatments in the clinical setting.

Significance

Findings indicate the functional role of IL33 in cancer polyploidy contributes to intrinsic and extrinsic mechanisms underlying treatment failure.

Introduction

Recurrence and treatment resistance are the major causes of cancer-associated death. The topics that most resonated has been epithelial-mesenchymal transition

(EMT) that enables tumor escape by conferring mesenchymal and stem properties such as high motility and dormancy (1), and numerous genomic, proteomic, microenvironmental and immunological mechanisms have been demonstrated (2,3).

However, little is known about the precise mechanisms underlying the metastatic colonization of cancer stem cells (CSCs) after terminating EMT within the niche,

3

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and thus practical treatment strategies targeting CSCs have been not established yet. Although its reversal MET has been believed as the sequential step in the mechanism (4), relapsed and metastatic tumors appear more aggressive than primary tumors, implying a possible different mechanism.

We have been investigating the interplay between cancer EMT and host immunity, and previously demonstrated that tumor metastasis (lung and bone marrow) is a possible risk factor of resistance to immune checkpoint inhibitors (ICIs) using mouse metastasis models (5). Particularly in the bone metastasis models, which were implanted with murine melanoma B16-F10 cells transduced with an

EMT-governing transcriptional factor snail (designated F10-snail+), tumor growth and metastasis were adversely promoted by ICI treatment just like hyperprogression reported in the clinical settings (6). In this study, we harvested and biologically and immunologically analyzed B16-F10 cells obtained from the metastatic bone marrow of the implanted mice, and attempted to define the molecular mechanisms underlying the adverse effect on tumors.

4

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Materials and Methods

Mice

Five-week-old female C57BL/6 mice were purchased from Charles River

Laboratories (Japan), and were maintained under pathogen-free conditions. The mice were used according to the protocols approved by the Animal Care and Use

Committee at the National Cancer Center Research Institute.

Cell lines

Human breast cancer cell lines (MCF7 and MDA-MB-231) were purchased from

ATCC, and were authenticated by short tandem repeat profiling. Murine melanoma

B16-B10 cells were purchased from the Cell Resource Center for Biomedical

Research at Tohoku University in Japan. We used B16-F10 cells transfected with plasmid vector pcDNA3.1(+) encoding neomycin-resistant with or without murine snail (F10-snail+ or F10-mock) that we established before (7). All tumor cells were tested for Mycoplasma negativity using a Hoechst-staining detection (MP

5

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Biomedicals) and were expanded and frozen in liquid nitrogen to avoid changes occurred by a long-term culture until use in experiments.

Establishment of B16-F10 subclones

F10-mock cells were harvested from subcutaneous tumors (F10-primary) and femur bone marrow (F10-BM) of mice 25 days after implantation and were cultured in 10%

FBS/DMEM with Geneticin (Merck) to select tumor cells. The details were described in the Supplementary Methods.

Establishment of IL33 transfectants

B16-F10 cells and MCF7 cells were transfected with plasmid vector pCMV6-ENTRY (Origene Technologies) encoding murine or human il33 by electroporation (0.4 kV, 25 µFD), and were cultured in 10% FBS/DMEM with

Geneticin. The details were described in the Supplementary Methods.

Functional analysis of tumor cells

6

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

We assessed cellular functions: cell proliferation (2 days) by cell count or WST-1 assay (Takara), cell adhesion (1 hour) using fibronectin-coated multiwell plates

(Corning), and cell invasion (8 hours) using a transwell chamber with a

Matrigel-coated membrane (Corning) as described before (7). To determine cell cycle, after fixation with ethanol, cells were treated with prodium iodide (PI; 50

µg/ml) and RNase A (200 µg/ml) for 30 min, and were analyzed by flow cytometry.

Cells were stimulated with IL13 (1 ng/ml; Peprotech) for 2 days, and were tested for adhesion and invasion. To determine chemo-sensitivity, tumor cells were treated with Paclitaxel (Wako), 5-Fluorouracil (5-Fu; Wako) or Gemcitabine (Wako) for 3 days (0 - 100 µg/ml, two-fold serial dilution), and the data were indicated as the percentage of untreated control (100%). Cell death was analyzed by flow cytometry after staining with PI and Annexin V. For tracking cell division, PKH67-labelled cells were used. In the in vivo experiments, tumor cells were s.c. (3x105 cells) and i.v.

(3x105 cells) implanted into mice, and tumor volume (0.5 x Length x Width2, mm3) was measured. For a convenience of observation and quantification, we assessed lung metastasis by counting the number of tumor metastatic nodules in lung, albeit tumor metastases in many tissue organs of the mice.

7

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

siRNA transfection

For il33 knockdown, we used two siRNAs targeting distinct il33 sequences or one scrambled sequence as a negative control (Invitrogen; Supplementary Methods).

The siRNAs were complexed with jetPEI (PolyPlus) according to the manufacturer’s instruction before transfection. The transfection efficacy was validated by RT-PCR 1

- 2 days after transfection.

Flow cytometric analysis

After Fc blocking, cells were stained with the immunofluorescence-conjugated antibodies (Supplementary Methods). For intracellular staining, cells were treated with Cytofix/Cytoperm solution (BD) before the staining. In mouse study, data were acquired using the FACSCalibur cytometer (Becton-Dickinson), and were analyzed by Cellquest software (BD Biosciences). In clinical study, data were acquired using a BD LSR Fortessa X-20 cytometer (Becton-Dickinson), and were analyzed by

FlowJo software (BD Biosciences). Before defining the specific molecular expressions, debris was firstly excluded by FSC/SSC followed by gating CD45+

8

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

leukocytes, and immunofluorescence intensity was compared to the isotype control

(Supplementary Fig. S1).

Functional analysis of IL17RB+ cells

IL17RB+ cells were sorted from SPCs of mice 7 days after 5-Fu treatment using a

BD IMag system (BD Biosciences) with anti-mouse IL17RB mAb (R&D) and the secondary anti-rat Ig magnetic beads (BD Biosciences). The purity (> 90%) of the

IL17RB+ cells was validated by flow cytometry. The IL17RB+ cells (1x106/ml) were cultured in 10%FBS/RPMI1640 at 37 oC for 3 days, and the supernatant fluid was harvested and filtrated (ø0.22 µm) before stock at 4 oC. IL13 in the supernatant was measured using an ELISA kit (R&D). The IL17RB+ supernatant or IL13 (1 ng/ml;

Peprotech) were added to a CTL induction system with splenic CD3+ T cells, gp70 peptide (MBL), APCs (inactivated bulk SPCs) in the presence or absence of anti-mouse IL13 mAb (1 µg/ml; R&D). Six days later, the sorted CD8+ T cells were tested for IFNγ production (24 hrs) and cytotoxic activity (ET ratio = 25:1, 4 hrs) as described before (7). The IL17RB+ cells (3x105) were co-injected with B16-F10 cells

(3x105) in mice, and tumor volume was measured. In a setting, anti-mouse IL13

9

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

mAb (20 µg) was intratumorally (i.t.) injected in the mice on days 4 and 8 after co-injection.

In vivo therapy

To evaluate the anti-tumor efficacy on both subcutaneous tumor growth and metastasis mimicking metastatic cancer patients, tumor cells were both s.c. (3x105 cells) and i.v. (3x105 cells) implanted in mice. The mice were intraperitoneally (i.p.) treated with PBS or 5-Fu (20 mg/kg; Wako) on days 4 - 8 after tumor implantation, or were i.t. treated with anti-mouse PD1 mAb (BioLegend), anti-mouse IL33 mAb

(R&D), or the isotype control (R&D) on day 5 (n = 5 - 10 per experiment). Tumor volume was measured twice a week. Metastatic nodules in lung were counted, and subcutaneous tumors and spleens were harvested for assays on day 14 - 18.

Clinical analysis

For immunohistochemical analysis, we purchased paraffin-embedded tissue sections (normal mammary tissues, and primary and metastatic tumor tissues) of

Stage II - III breast cancer patients from SuperBioChips, and stained with

10

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

immunofluorescence-conjugated anti-human IL33 mAb (R&D), anti-human IL17RB mAb (R&D), or the isotype IgG (BioLegend) as described before (5). The immunofluorescence intensity was automatically measured as pixel counts at two fields per section using a LSM700 Laser Scanning Microscope (Carl Zeiss), and the average was plotted in graphs. For flow cytometric analysis, EDTA-added peripheral blood was collected from healthy donors (n = 4) and patients with Stage

IV metastatic colorectal cancer (n = 9; age 57 - 79) after receiving written informed consent (November 2018 and June 2019), according to the protocol approved by the Institutional Review Board at the National Cancer Center. PBMCs were isolated by Ficoll (Nacalai), and were stained with the specific mAbs (Supplementary

Methods). All activities were conducted in accordance with the ethical principles of the Declaration of Helsinki.

Statistical analysis

Data indicate mean ± SD unless otherwise specified. Significant differences (P value < 0.05) were statistically evaluated using GraphPad Prism 7 software (MDF).

Data between two groups were analyzed by the unpaired two-tailed Student’s t test.

11

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Data among multiple groups were analyzed by one-way ANOVA, followed by the

Bonferroni post-hoc test for pairwise comparison of groups based on the normal distributions. Non-parametric groups were analyzed by the Mann-Whitney test.

Mouse survival was analyzed by the Kaplan-Meier method and ranked according to the Mantel-Cox log- test. Correlation between two factors was evaluated by the nonparametric Spearman’s rank test.

Results

IL33 is involved in chemoresistance of tumor cells

To compare tumor properties between primary sites and metastatic sites, B16-F10 cells were both subcutaneously (s.c.) and intravenously (i.v.) implanted into mice, and tumor cells were harvested from the subcutaneous tumors (F10-primary) and the metastatic bone marrow (F10-BM) on day 25 after implantation. The F10-BM cells slightly expressed an epithelial marker E-cadherin, but highly expressed a mesenchymal marker Snail in the cytoplasm, but not in the nucleus, implying de-regulation state (Fig. 1A). The F10-BM cells showed significant lower

12

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

proliferative activity and higher resistance to treatment with 5-Fluorouracil (5-Fu) with a slight increase at low doses as compared to F10-primary cells and F10-snail+ cells derived from bone marrow (P < 0.05; Fig. 1B). In the in vivo setting, the

F10-BM tumor also significantly slowly grew than F10-primary tumor (P = 0.002), but the growth was adversely promoted by 5-Fu treatment (days 4 - 8; P = 0.002 versus PBS treatment), although F10-snail+ tumors were just unresponsive to the treatment (Fig. 1C). These suggest that the F10-BM cells acquire a distinct property from that of the F10-primary after metastasis. The F10-BM tumors showed significant increase in ST2/ST2L+ cells including c-kit+FceRIa+ mast cells (P = 0.002 versus PBS treatment), CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs; P

= 0.005), and IL17RB+GATA3+ type 2 innate lymphoid cells (ILC2s; P = 0.005) after the treatment (Fig. 1D). This observation raised a possibility of its ligand IL33 release from the progressing F10-BM tumors. Indeed, higher IL33 production was observed in the F10-BM cells compared to other clones, and IL33 knockdown with the specific siRNAs significantly reduced the chemoresistance (P < 0.05 versus control siRNA; Fig. 1E). Similar results were observed using highly bone-metastatic breast cancer IL33+ MDA-MB-231 cells after siRNA-il33 transfection (P < 0.02

13

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

versus control siRNA; Supplementary Fig. S2). We chose breast cancer cell lines because bone metastasis is most frequently seen in breast cancer in clinical settings (8,9). These results suggest IL33 is involved in the treatment-induced tumor progressive mechanisms.

IL33 drives tumor progression after treatment

IL33 is a member of the IL1 family expressed in some types of cells such as endothelial cells and fibroblasts, and is associated with diseases including allergy, infection and cancer (10,11). However, the role of IL33 in cancer is still controversial, albeit IL33 upregulation in many types of cancers (12). To address this issue, we established IL33 transfectants using B16-F10 cells (F10-TR; Fig. 2) and human breast cancer MCF7 cells (MCF7-TR; Supplementary Fig. S3). IL33 overexpression generated large cells with low proliferative property (P < 0.001 versus F10-mock) and high 5-Fu-resistant property in vitro (Fig. 2A) and in vivo (Fig. 2B). The 5-Fu treatment remarkably promoted F10-TR tumor growth and metastasis, and significantly shortened mouse survival (P = 0.0001 versus F10-mock; Fig. 2B).

These suggest IL33 plays a key role in the treatment-induced tumor progression,

14

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

albeit likely a better prognostic factor under tranquility. In the F10-TR-implanted mice, ST2+ cells, particularly ILC2s, were significantly expanded not only in the tumor tissues (P = 0.003 versus PBS treatment) but also in the spleen (P < 0.0001) after the treatment (Fig. 2C and Supplementary Fig. S4A). ST2+ increase was also observed within F10-mock tumor tissues regardless of treatment. This might be potentially mediated by chemokines such as CXCL12 for CXCR4 that is commonly expressed in mast cells, MDSCs and ILC2s (13). CTLs were impaired in the

F10-TR-implanted mice, and the CTL activities were further reduced by the treatment (IFNγ production, P = 0.002 versus PBS treatment; cytotoxicity, P =

0.001; Fig. 2D). These suggest IL33 induction confers a progressive property to tumor cells via not only biological self-transformation but also immunological damage possibly mediated by ST2+ cells.

IL33 regulates cancer polyploidy

We next examined whether IL33 could be associated with polyploidy because nuclear IL33 is known but its functional roles remain unclear (14), and IL33 overexpression transformed tumor cells into a giant size (Fig. 2A) that is a

15

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

representative feature of polyploidy, by which giant cells are generated through mis-regulation of canonical G1–S–G2–M cell cycle without cell division followed by unrestrained propagation (15). F10-TR cells contained significantly more ≧ 4N

DNA contents (P < 0.001 versus F10-mock; Fig. 3A), and generated smaller progeny cells by stimulation with a low dose of 5-Fu albeit cell death in F10-mock cells (Fig. 3B). Aurka (16) and Aurkb (17) regulate polyploidization followed by inactivation of p53 (18). Indeed, Aurka/b increased and phosphorylated p53-Ser15

(p53-p) decreased in the F10-TR cells (P < 0.002 versus F10-mock), and IL33 knockdown reduced Aurka/b expression but increased p53-p expression in the

F10-TR cells (Fig. 3C). These suggest IL33 expression, potentially in the nucleus, plays a key role in polyploidization followed by rapid proliferation in response to treatment stress.

IL33-inducible IL17RB+ cells promote tumor progression

ST2+ cells remarkably increased in the F10-TR-implanted mice particularly after treatment. We further pursued the observation because the ST2+ cells would likely play important roles in the IL33+ tumor progression mechanisms. Since ST2 is

16

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expressed not only in ILC2s (10,11), mast cells and MDSCs (19) but also in NK (20) and Tregs (21), we conducted antibody-mediated ablation experiments to determine the key effector cells using immunodeficient nu/nu mice treated with anti-asialo GM1, indicating absence of functional T cells and NK/NKT cells.

5-Fu-induced F10-TR tumor progression was partly retarded in the absence of

FceRIa+ cells and CD11b+ cells, but not NK/NKT cells and T cells (Supplementary

Fig. S4B). In co-injection experiments, however, greater impact on tumor progression was provided by IL17RB+ cells derived from F10-TR-implanted mice

(designated TR-IL17RB cells) compared to FceRIa+ cells and CD11b+ cells (P <

0.001; Supplementary Fig. S4C). These suggest ST2+ cells, especially ILC2s, could play important roles in the IL33+ tumor progression. The TR-IL17RB cells highly produced IL13, suggesting these cells could be ILC2s (Fig. 4A). Stimulation with a low dose of IL13 that did not affect F10-mock cells remarkably enhanced F10-TR invasion (Fig. 4B). Anti-IL13 mAb injection into the TR-IL17RB-coinjected tumors significantly suppressed the growth (P = 0.010; Fig. 4C). These suggest IL13 is involved in the ILC2-induced tumor progressive mechanisms.

17

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

In the in vitro CTL induction, addition of the TR-IL17RB supernatant generated exhausted and dysfunctional CTLs expressing PD1, Tim3 and Tigit (Fig. 4D).

However, anti-IL13 mAb addition to the culture generated potent CTLs, suggesting

ILC2-induced IL13 could impair CTLs (P < 0.004; Fig. 4D). Since IL13R is not generally expressed on T cells except some cases (22), the CTL dysfunction observed might be indirectly induced by other IL13R+ cells such as M2 macrophages and MDSCs contained in the APCs used. Anti-IL13 mAb injection significantly suppressed 5-Fu-induced F10-TR tumor progression and metastasis

(P < 0.02 versus 5-Fu only; Fig. 4E). These suggest ILC2s contribute to IL33+ tumor progression via immune exhaustion and dysfunction partly mediated by IL13.

Blocking IL33 elicits anti-tumor immunity in mice with IL33+ tumors

Unexpected hyperprogression is a serious problem in cancer therapy with ICIs (6).

In the mouse models implanted with F10-BM tumors that is similar the F10-TR tumors (Supplementary Fig. S5), anti-PD1 treatment significantly promoted tumor growth, and shortened mouse survival (P < 0.0001 versus control), albeit significantly effective in the F10-primary models (P = 0.0017; Fig. 5A). In the

18

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

anti-PD1-treated mice, cell infiltration was hardly seen in the F10-BM tumors (Fig.

5B and C), and splenic CTLs were still dysfunctional (Fig. 5D and E). In contrast, anti-IL33 treatment did not cause tumor progression, and the mouse survival was slightly but significantly prolonged (P = 0.004 versus control; Fig. 5A). In the anti-IL33-treated mice, tumor specific CD8+ T cells increased within tumors, and splenic CTL activities were significantly elevated (Fig. 5). Combination of anti-PD1 treatment synergistically enhanced the anti-IL33 efficacy on tumor growth (P =

0.0003 versus anti-IL33 monotherapy) and mouse survival (P = 0.0036). The combinatory regimen was also significantly effective in the F10-primary models (P =

0.047 versus anti-PD1 monotherapy). These results provide a potential of blocking

IL33 for successfully eliciting anti-tumor immunity in combination with other therapeutics in the treatment of IL33+ tumors.

Clinical relevancy of the IL33-ST2/IL17RB axis

We next validated the findings using clinical samples. We firstly conducted immunohistochemical analysis using tumor tissues obtained from patients with breast cancer. IL33 expression was upregulated in primary and metastatic tumor

19

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

tissues compared to noncancerous portions (P < 0.05; Fig. 6A), and the IL33 intensity was correlated with IL17RB+ cell infiltration particularly in the Stage III patients with tumor metastasis (P = 0.0005) rather than the Stage II patients (P =

0.0886). IL17RB+ cells were hardly seen in the tumors if IL33 was expressed only in the nuclei. These suggest a potential causal relationship between IL33 positivity in tumors and increase of IL17RB+ cells within the tumor microenvironment.

We further analyzed PBMCs for ST2+ cells and IL17RB+ cells by flow cytometry, since the cells could potentially play critical roles in systemic immunity as well as local immunity. We used PBMCs obtained from patients with metastatic colorectal cancer (mCRC) because of only one available for us. It has been demonstrated that

IL33 upregulation is associated with CRC pathogenesis (23), and hyperprogression is seen in CRC patients after anti-PD1/PDL1 treatment albeit a few in number (24).

ST2+ cells (P = 0.008) and IL17RB+ cells (P = 0.021) were significantly increased in mCRC patients compared to healthy donors, and the increase of these cells, particularly ST2+ cells (P = 0.005), was correlated with decrease of CD3+CD8+ T cells (Fig. 6B). These suggest a potential causal relationship between ST2/IL17RB+

20

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expansion and CD8+ T-cell reduction in systemic immunity. Taken together, targeting IL33 and the consequent ST2/IL17RB+ cells may be a promising strategy of diagnosis and treatment of patients who are likely resistant to treatments in the clinical settings.

Discussion

We identified IL33 as a determinant of treatment resistance of cancer. IL33 induction in tumor cells transforms into polyploid giant cells showing abnormal cell cycle without cell division accompanied by Snail deregulation and p53 inactivation, and its small progeny cells are generated in response to treatment stress.

Simultaneously, soluble IL33 is released from the tumor cells, and expands ST2+ cells including IL17RB+GATA3+ ILC2s that promote tumor progression and metastasis directly and indirectly through induction of immune exhaustion and dysfunction partly mediated by IL13. In the mouse IL33+ metastatic tumor models, however, blocking IL33 with the specific mAb abrogates the negative consequences, and successfully elicits anti-tumor efficacy induced by other treatment combined. In clinical samples, IL33 positivity in tumors is significantly correlated with increase of

21

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

the IL17RB+ cells within the tumor milieu, and increase of ST2+ cells and IL17RB+ cells is correlated with reduction of CD8+ T cells in PBMCs. This suggests clinical relevancy of the basic findings. Thus, this study reveals the functional role of IL33 in cancer polyploidy that could be intrinsic tumor biological and extrinsic immunological mechanisms underlying treatment failure.

Although many studies demonstrated the roles of IL33 in cancer, it is still controversial (12). For example, treatments with mAbs specific for IL33 (25), ST2

(26) and IL1RAP (27) significantly suppressed tumor progression through reduction of tumor-associated macrophages and Tregs in mouse tumor models. On the other hand, IL33 induction in melanoma suppressed tumor progression (28). We found that IL33 expression in tumor cells retards proliferation and growth, whereas aberrant mitotic progression is caused by treatment stress. In contrast, stable silencing of IL33 in tumor cells restricts cell proliferation resulting in poor engraftment and growth of the tumors in mice probably because of damaging cells not only internally via reduction of Aurka/b that is required for mitotic control, but also externally via loss of ST2+ cell support. Therefore, the tumor suppressive

22

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

activity reported might be explained by the low proliferative property due to polyploidization. The authors did not conduct therapeutic experiments, in which tumor progression might be seen as shown in our study. IL33 overexpression simultaneously induced ST2 and IL1RAP expressions (Fig. 2A, Supplementary Fig.

S3), and IL1RAP knockdown enhanced chemo-sensitivity of IL33+ tumor cells

(Supplementary Fig. S2). The receptor coexpression may intensify the treatment resistance of IL33+ tumor cells in an autocrine manner.

Cancer polyploidy has been demonstrated as cancer stemness because of producing unrestrained propagation that undermines genomic stability (15,29).

Polyploid giant cancer cells are pathologically observed in clinical tumor tissues, and the incidence is associated with chemoresistance (30) and poor prognosis of cancer patients (31). However, there are no practical managements of the polyploidization and the rapid proliferation. Our findings that IL33 plays a critical role in cancer polyploidy provided a possible biomarker and a potentially druggable target in cancer therapy. Combination with polyploidy-associated Aurka/b inhibitors

(32) may be potentially enhance the anti-IL33 therapeutic efficacy.

23

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

This study provides new insights into the IL33 roles in recurrence and treatment resistance of cancer. Targeting IL33-driven polyploidy could, at least in part, overcome treatment failure in clinical settings.

Acknowledgements

C. Kudo-Saito was financially supported by Grants-in-Aid for Scientific Research

KAKENHI (21590445 and 26430122), and Japan Agency for Medical Research and

Development AMED-P-CREATE (106209).

24

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1. Patel P, Chen EI. Cancer stem cells, tumor dormancy, and metastasis. Front

Endocrinol (Lausanne) 2012;3:125

2. Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial-mesenchymal

transition: new insights in signaling, development, and disease. J Cell Biol

2006;172:973-81

3. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal

transitions in development and disease. Cell 2009;139:871-90

4. Chaffer CL, Thompson EW, Williams ED. Mesenchymal to epithelial

transition in development and disease. Cells Tissues Organs 2007;185:7-19

5. Kudo-Saito C, Ishida A, Shouya Y, Teramoto K, Igarashi T, Kon R, et al.

Blocking the FSTL1-DIP2A Axis Improves Anti-tumor Immunity. Cell Rep

2018;24:1790-801

6. Champiat S, Ferrara R, Massard C, Besse B, Marabelle A, Soria JC, et al.

Hyperprogressive disease: recognizing a novel pattern to improve patient

management. Nat Rev Clin Oncol 2018;15:748-62

25

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

7. Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Cancer metastasis is

accelerated through immunosuppression during Snail-induced EMT of

cancer cells. Cancer Cell 2009;15:195-206

8. Riihimaki M, Hemminki A, Sundquist J, Hemminki K. Patterns of metastasis

in colon and rectal cancer. Sci Rep 2016;6:29765

9. Fornetti J, Welm AL, Stewart SA. Understanding the Bone in Cancer

Metastasis. J Bone Miner Res 2018;33:2099-113

10. Liew FY, Girard JP, Turnquist HR. Interleukin-33 in health and disease. Nat

Rev Immunol 2016;16:676-89

11. Griesenauer B, Paczesny S. The ST2/IL-33 Axis in Immune Cells during

Inflammatory Diseases. Front Immunol 2017;8:475

12. Wasmer MH, Krebs P. The Role of IL-33-Dependent in the

Tumor Microenvironment. Front Immunol 2016;7:682

13. Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors:

positioning cells for host defense and immunity. Annu Rev Immunol

2014;32:659-702

26

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14. Sundlisaeter E, Edelmann RJ, Hol J, Sponheim J, Kuchler AM, Weiss M, et

al. The alarmin IL-33 is a notch target in quiescent endothelial cells. Am J

Pathol 2012;181:1099-111

15. Zhang S, Mercado-Uribe I, Xing Z, Sun B, Kuang J, Liu J. Generation of

cancer stem-like cells through the formation of polyploid giant cancer cells.

Oncogene 2014;33:116-28

16. Wu CC, Yu CT, Chang GC, Lai JM, Hsu SL. Aurora-A promotes gefitinib

resistance via a NF-kappaB signaling pathway in p53 knockdown lung

cancer cells. Biochem Biophys Res Commun 2011;405:168-72

17. Wu L, Ma CA, Zhao Y, Jain A. Aurora B interacts with NIR-p53, leading to

p53 phosphorylation in its DNA-binding domain and subsequent functional

suppression. J Biol Chem 2011;286:2236-44

18. Aylon Y, Oren M. p53: guardian of ploidy. Mol Oncol 2011;5:315-23

19. Jovanovic IP, Pejnovic NN, Radosavljevic GD, Pantic JM, Milovanovic MZ,

Arsenijevic NN, et al. Interleukin-33/ST2 axis promotes breast cancer growth

and metastases by facilitating intratumoral accumulation of

27

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

immunosuppressive and innate lymphoid cells. Int J Cancer

2014;134:1669-82

20. Milovanovic M, Volarevic V, Radosavljevic G, Jovanovic I, Pejnovic N,

Arsenijevic N, et al. IL-33/ST2 axis in inflammation and immunopathology.

Immunol Res 2012;52:89-99

21. Schiering C, Krausgruber T, Chomka A, Frohlich A, Adelmann K, Wohlfert

EA, et al. The alarmin IL-33 promotes regulatory T-cell function in the

intestine. Nature 2014;513:564-8

22. Yang L, Xu LZ, Liu ZQ, Yang G, Geng XR, Mo LH, et al. Interleukin-13

interferes with activation-induced t-cell apoptosis by repressing p53

expression. Cell Mol Immunol 2016;13:669-77

23. Landskron G, De la Fuente Lopez M, Dubois-Camacho K, Diaz-Jimenez D,

Orellana-Serradell O, Romero D, et al. /ST2 Axis Components

Are Associated to Desmoplasia, a Metastasis-Related Factor in Colorectal

Cancer. Front Immunol 2019;10:1394

28

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24. Ji Z, Peng Z, Gong J, Zhang X, Li J, Lu M, et al. Hyperprogression after

immunotherapy in patients with malignant tumors of digestive system. BMC

Cancer 2019;19:705

25. Wang K, Shan S, Yang Z, Gu X, Wang Y, Wang C, et al. IL-33 blockade

suppresses tumor growth of human lung cancer through direct and indirect

pathways in a preclinical model. Oncotarget 2017;8:68571-82

26. Akimoto M, Maruyama R, Takamaru H, Ochiya T, Takenaga K. Soluble IL-33

receptor sST2 inhibits colorectal cancer malignant growth by modifying the

tumour microenvironment. Nat Commun 2016;7:13589

27. Agerstam H, Karlsson C, Hansen N, Sanden C, Askmyr M, von Palffy S, et

al. Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in

xenograft models of acute myeloid leukemia. Proc Natl Acad Sci U S A

2015;112:10786-91

28. Long A, Dominguez D, Qin L, Chen S, Fan J, Zhang M, et al. Type 2 Innate

Lymphoid Cells Impede IL-33-Mediated Tumor Suppression. J Immunol

2018;201:3456-64

29

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

29. Tommasi S, Besaratinia A, Wilczynski SP, Pfeifer GP. Loss of Rassf1a

enhances p53-mediated tumor predisposition and accelerates progression to

aneuploidy. Oncogene 2011;30:690-700

30. Xuan B, Ghosh D, Cheney EM, Clifton EM, Dawson MR. Dysregulation in

Actin Cytoskeletal Organization Drives Increased Stiffness and Migratory

Persistence in Polyploidal Giant Cancer Cells. Sci Rep 2018;8:11935

31. Fei F, Zhang D, Yang Z, Wang S, Wang X, Wu Z, et al. The number of

polyploid giant cancer cells and epithelial-mesenchymal transition-related

are associated with invasion and metastasis in human breast

cancer. J Exp Clin Cancer Res 2015;34:158

32. Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel

therapy targets in cancers. Oncotarget 2017;8:23937-54

30

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1.

IL33 expression is associated with chemoresistance of tumors. A, Morphology and

EMT-related molecular expressions of tumor cells. B16-F10 cells were isolated from subcutaneous tumors (F10-primary) or bone marrow (F10-BM) of mice 25 days after both subcutaneous (s.c.) and intravenous (i.v.) implantation. As another control, snail-transduced B16-F10 cells (F10-snail+) were isolated from bone marrow. Scale bar = 50 µm. B, Chemoresistance of the F10-BM cells. Tumor cells were cultured with or without 5-Fu for 3 days. Data are depicted as the percentage of untreated proliferation (n = 3). C, Enhancement of in vivo F10-BM tumor growth by chemotherapy. Mice were intraperitoneally (i.p.) injected with PBS (open bars) or

5-Fu (20 mg/kg; closed bars) on days 4 - 8 after tumor implantation (n = 5). Tumor volume on day 14. D, Increase of ST2L+ cells in the F10-BM tumors after chemotherapy. Tumor-infiltrating cells (TILs) were analyzed for ST2L+ cells by immunostaining (scale = 50 µm) and flow cytometry (n = 5) on day 14. E,

Enhancement of chemosensitivity by IL33 knockdown. F10-BM cells were

31

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

transfected with il33-specific siRNAs (#1 and #2), and were cultured with or without

5-Fu. The knockdown efficacy was validated by RT-PCR (photos) and ELISA (IL33 production; n = 3). Open circles, F10-primary. Closed squares, F10-BM + control siRNA. Open triangles, F10-BM + siRNA-il33. *P < 0.01, **P < 0.05. Graphs show mean ± SD. Representative data of five independent experiments.

Figure 2.

IL33 drives tumor progression after treatment. A, IL33 overexpression confers chemoresistance. B16-F10 cells were transduced with murine il33 (F10-TR1 and

F10-TR2), and were tested for IL33 receptor expressions (scale = 50 µm), IL33 production (3 days; n = 3), and cell proliferation with or without 5-Fu (3 days; n = 3).

B, Chemotherapy worsens the F10-TR tumor progression. Mice were treated with

PBS or 5-Fu on days 4 - 8 after tumor implantation (n = 5). Lung metastasis on day

14 (n = 5). Mouse survival (n = 10). C, Possibly ST2L+ cells are expanded in the

F10-TR-implanted mice after 5-Fu treatment. TILs and spleen cells (SPCs) were analyzed by flow cytometry on day 14 (n = 5). D, Chemotherapy impairs CTLs in the

F10-TR-implanted mice (n = 3). In the cytotoxic assay, F10-mock cells were used

32

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

as a target (ET ratio = 25:1). *P < 0.01, **P < 0.05. Graphs show mean ± SD.

Representative data of three independent experiments.

Figure 3.

IL33 expression induces polyploidy in tumor cells. A, Abnormal cell cycle in the

F10-TR cells. PI-stained DNA contents were quantified by flow cytometry (n = 3).

Photo shows IL33 expression in a large DAPI+ nucleus of F10-TR cells (scale = 20

µm). B, Chemo-treatment generates small progeny cells of the F10-TR cells.

PKH67-labeled tumor cells were treated with 5-Fu (5 µg/ml) for 24 hrs, and analyzed for cell division and Annexin V+ apoptosis by flow cytometry. C,

Expression of polyploidy-associated molecules, Aurka, Aurkb and p53. Total p53 and phosphorylated p53-ser15 (p53-p) expressions were analyzed by flow cytometry (n = 3) and immunostaining (photos). *P < 0.01. Bar graphs show mean ±

SD. Data in each panel are representative of three independent experiments.

Figure 4.

33

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL33-inducible IL17RB+ cells promote tumor progression. A, IL13 production from

IL17RB+ cells (n = 3). IL17RB+ cells were sorted from SPCs of F10-mock-implanted mice (Mock-17RB) and F10-TR-implanted mice (TR-17RB) 7 days after 5-Fu treatment. B, High sensitivity of F10-TR cells to IL13. Tumor cells were tested for adhesive and invasive properties after treatment with IL13 (1 ng/ml; n = 3). Photos show morphological changes (scale = 50 µm). C, IL17RB+ cells promote tumor progression. Tumor cells were co-implanted with IL17RB+ cells (1:1) in mice, and anti-IL13 mAb or control IgG (20 µg) was intratumorally (i.t.) injected on days 4 and

8 after co-implantation (tumor volume on day 14; n = 5). D, IL17RB+ cells impair

CTLs partly via IL13 released. TR-17RB supernatant or IL13 was added to the CTL induction system in the presence of anti-IL13 mAb or control IgG (1 µg/ml). The cells were analyzed by flow cytometry, and the sorted CD8+ T cells were tested for cellular functions (n = 3). E, Blocking IL13 suppresses 5-Fu-induced F10-TR tumor progression. Anti-IL13 blocking mAb or control IgG (20 µg) was i.t. injected in the mice on days 4 and 8 (5-Fu treatment = days 4 - 8). Data on day 14 (n = 5). *P <

0.01, **P < 0.05. Bar graphs show mean ± SD. Representative data of three independent experiments.

34

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 5.

Anti-tumor immunity induced by blocking IL33 in mice. A, Anti-IL33 treatment enhances anti-PD1-induced efficacy. Mice were i.t. injected with anti-PD1 mAb and/or anti-IL33 mAb (50 µg) on day 5 after tumor implantation (tumor growth, n = 5; mouse survival, n = 10). B, The number of TILs and SPCs of the mice on day 18 (n

= 5). Anti-IL33 treatment increases tumor-specific CD8+ T cells within tumors (C), following reduction of immune exhausted CD8+ T cells (D) and generation of potent

CTLs (E, n = 3) in spleen of the F10-TR-implanted mice. *P < 0.01, **P < 0.05.

Graphs show mean ± SD. Representative data of two independent experiments.

Figure 6.

Clinical relevancy of the IL33-ST2/IL17RB axis. A, Significant correlation between

IL33 positivity and IL17RB+ cell accumulation within tumor microenvironment.

Primary tumor tissues (Stage II, n = 23; Stage III, n = 14), metastatic lymph nodes

(n = 9) and the neighboring noncancerous mammary tissues (Stage III, n = 8) obtained from breast cancer patients were immunohistochemically analyzed for

35

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL33 and IL17RB expressions. Immunofluorescence intensity is depicted as pixel counts (IL33 < 76, IL17RB < 443 in normal tissues). Representative photos are shown (scale = 100 µm). *P < 0.01, **P < 0.05 versus normal tissues. B, Significant reversal correlation between ST2+/IL17RB+ cells and CD8+ T cells in peripheral blood. PBMCs obtained from healthy donors (n = 4, open circles) and patients with

Stage IV metastatic colorectal cancer (mCRC, n = 9; closed circles) were analyzed by flow cytometry. Bar graphs show mean ± SD. P values in the scatter plot graphs were analyzed by the nonparametric Spearman’s rank test.

36

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 10, 2020; DOI: 10.1158/0008-5472.CAN-19-2235 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL33 is a key driver of treatment resistance of cancer

Chie Kudo-Saito, Takahiro Miyamoto, Hiroshi Imazeki, et al.

Cancer Res Published OnlineFirst March 10, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-2235

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2020/03/10/0008-5472.CAN-19-2235.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/2020/03/10/0008-5472.CAN-19-2235. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research.