Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The IL-18 antagonist IL-18 Binding is produced in the human ovarian cancer microenvironment

Grazia Carbotti1§, Gaia Barisione1, Anna Maria Orengo1, Antonella Brizzolara1, Irma Airoldi2, Marina Bagnoli3, Patrizia Pinciroli3, Delia Mezzanzanica3, Maria Grazia Centurioni4, Marina Fabbi1*, Silvano Ferrini1*

1Department of Integrated Oncological Therapies, IRCCS AOU San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy; 2Laboratory of Oncology, IRCCS Istituto G. Gaslini, Genoa Italy; 3Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; 4Department of Surgery, IRCCS AOU San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy.

Running title: IL-18BP in ovarian cancer

Key words: IL-18 Binding Protein, ovarian cancer, immune escape, IL-27, EBI3

Financial support: AIRC (Associazione Italiana per la Ricerca sul Cancro, IG5509, IG13018 and IG13518), Ministry of Health Project ‘Tumori Femminili’ and Compagnia di San Paolo.

§Grazia Carbotti is enrolled in the Doctorate School of Genetics, University of Genoa, Italy.

*Equal contributors and correspondence: Silvano Ferrini and Marina Fabbi, UOC Terapia Immunologica, IRCCS AOU San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Largo R. Benzi 10, 16132 Genova, ITALY Phone: 0039-010-5737-372; Fax:-374; Email: [email protected], [email protected]

Conflict of interest: no conflict of interest to disclose.

Word count of text: 4667

Total number of figures and tables: 6

Relevance Statement: IL-18 is an immune-enhancing , which is being studied in clinical trials of immunotherapy. IL-18BP is a natural antagonist of IL-18 and limits IL-18 biological activity. Here we show that IL-18BP is produced by both tumor cells and tumor- associated leukocytes. Expression of IL-18BP in ovarian cancer cells in vitro is induced by such as IFN-γ and IL-27, which may play a role in the tumor environment. The high local levels of IL-18BP at the tumor site may limit the induction of an efficient immune response by either endogenous or therapeutic IL-18.

1

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

PURPOSE: (IL)-18 is an immune-enhancing cytokine, which induces Interferon

(IFN)-γ production, Th1-responses and anti-tumor effects. In turn, IFN-γ stimulates IL-18

Binding Protein (BP) production, which blocks IL-18 activity. In view of the potential use of

IL-18 in epithelial ovarian cancer (EOC) immunotherapy, here we studied IL-18BP

expression and its regulation by cytokines in EOC cells in vitro and in vivo.

EXPERIMENTAL DESIGN: Expression and production of IL-18BP in EOC cell lines,

primary ovarian carcinomas and the corresponding normal tissues, patients’ serum and

ascites were investigated by immunochemistry, ELISA, screening of expression

profiles and RT-PCR.

RESULTS: Analysis of gene expression profiles revealed that IL18BP mRNA is increased

in EOC tumors compared to normal ovary cells. Release of IL-18BP was detectable in EOC

sera and at greater extent in the ascites, indicating production at the tumor site. Indeed,

immunochemical analyses on cells isolated from the ascites and on tumor sections indicated

that IL-18BP is expressed in both tumor cells and tumor-associated leukocytes, which

displayed a CD3-CD20-NKp46-CD13+CD14low phenotype. EOC cell lines do not

constitutively express IL-18BP. However, its release is inducible both by IFN-γ stimulation

in vitro and by xenotransplantation of EOC cells in immune-deficient mice, suggesting a

role for the microenvironment. In vitro experiments and immunochemistry indicated that

also IL-27 is involved in IL-18BP up-regulation in EOC cell lines and primary cells through

STAT1 activation. Altogether these data indicate that IL-18BP, which is produced in EOC

in response to micro-environmental factors, may inhibit endogenous or exogenous IL-18

activity.

2

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

Epithelial ovarian cancer (EOC) represents 80% of all ovarian malignancies, is frequently

diagnosed at advanced stages and has a poor 5-year survival rate (1). Several evidences

indicate that T lymphocytes capable of recognizing EOC cells are present at the tumor site

(2, 3) and that -mediated immunity may impact on clinical course of EOC (3).

However, similarly to other tumors, EOC has the ability to escape from immune system

control through several mechanisms (4, 5).

Several cytokines and chemokines were found to be elevated in serum and in the ascites of

EOC patients and have been implicated in tumor development, angiogenesis (6, 7),

progression (8), drug resistance (9) or immune suppression (10). Some of these cytokines

have been considered as serological biomarkers of EOC (11). Among them, IL-18 was

proposed as potential biomarker by gene expression profiling (12) and, indeed, IL-18 levels

were elevated in serum and ascites of EOC patients (12, 13).

IL-18 is a pro-inflammatory and immune-enhancing cytokine, which induces IFN-γ

production by T and NK cells, mediates Th1 polarization and is involved in the defense from

pathogens (14-16). IL-18 is synthesized as an inactive precursor (pro-IL-18), which is

converted to a mature form (mat-IL-18) by caspase-1 (17). IL-18 is released from cells as

both pro- and mat-IL-18 forms, but only mat-IL-18 can bind to IL-18R. Moreover, in pre-

clinical tumor models mat-IL-18 exhibited anti-tumor properties through the induction of an

immune response, whereas pro-IL-18 had no activity (18, 19).

EOC cell lines release pro-IL-18 but not mat-IL-18, due to defective caspase-1 expression or

activation, while normal ovarian epithelial cells secrete mat-IL-18 (20). Indeed, IL-18

present at high levels in EOC ascites is predominantly the inactive pro-IL-18 (13), although

we cannot exclude that low levels of mat-IL-18, eventually present, may contribute to the

immune response.

3

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL-18 Binding Protein (IL-18BP) is an inhibitor of IL-18 activity, as it binds the mature

form of this cytokine and blocks its interaction with IL-18R (21) and its isoform “a” is the

mostly expressed form (22). IL-18BP is produced by monocytes and macrophages (23) and

by prostatic (24) and colorectal tumor cells (25) in response to IFN-γ stimulation. IFN-γ is

the physiological inducer of IL-18BP, which in turn inhibits IL-18 biological activity in a

negative feed-back loop. IL-18BP accumulates in the serum in chronic renal failure, where it

may contribute to the defective immune response (26).

To further address the biological role of endogenous IL-18 in human EOC we performed an

integrated analysis of IL-18 and IL-18BP in EOC patients. The study of IL-18BP seemed

also relevant, as IL-18-based immunotherapy is being evaluated in EOC patients (27, 28)

(NCT00659178) and high levels of IL-18BP may interfere with this treatment. We found

that indeed IL-18BP levels are elevated in the serum and particularly in the ascites of EOC

patients and that IL-18BP is expressed by EOC cells and by reactive leukocytes. Our data

also suggest that IL-18BP production may be the outcome of a cross-talk between tumor

cells and the microenvironment, involving IFN-γ and IL-27, a member of the IL-12 cytokine

family (29, 30). Altogether these data support an immune-regulatory role for IL-18BP in

EOC, as it may limit the activity of endogenous or therapeutic IL-18.

4

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Materials and methods

Cells and Cell treatments

The human EOC cell lines SKOV3 (ATTC), A2780 (ICLC), A2774 (IST Genoa) and

OVCAR5 (INT Milan) were grown in RPMI 1640, with L-glutamine, 10% FCS and

antibiotics (Lonza). NK-92 cells (ATCC) were grown in medium containing 600 IU IL-2

(Novartis). A vial of each cell line master stock was recently genotyped using the Cell IDTM

System (Promega) and the GeneMapper® software, version 4.0.

Cells (50x103/well) were seeded in 24-well plates in culture medium. The day after, culture

medium was replaced with medium with or without human recombinant IFN-γ (PeproTech),

human recombinant IL-27 (R&D System) or human recombinant IL-35 (Enzo Life

Sciences). For IL-27R blocking experiments an anti-gp130 antibody was added (mAb228,

R&D System). Treatment was carried on for 48 hours. Conditioned media were then

collected, centrifuged, and used for IL-18BP detection.

RT-PCR Analysis of IL18BP mRNA Expression

Cells were detached by trypsin, washed and total RNA was isolated by the NucleoSpin RNA

II kit (Macherey-Nagel) and reverse-transcribed using the SuperScript II Reverse

Transcriptase (Invitrogen). Quantitative RT-PCR analysis was performed using the

following primers: POLR2A upper primer GACAATGCAGAGAAGCTGG, lower primer

GCAGGAAGACATCATCATCC; GAPDH upper primer GAAGGTGAAGGTCGGAGT,

lower primer CATGGGTGGAATCATATTGGAA; IL18BP upper primer

GTGTCCAGCATTGGAAGTGACC, lower primer GGAGGTGCTCAATGAAGGAACC.

Amplification was carried out by the Mastercycler® ep realplex4 instrument (Eppendorf

International) using the iQTM SYBR® Green Supermix system (Bio-Rad Laboratories).

Relative quantification of mRNAs was calculated by the ΔΔCt method. For semi-

5

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

quantitative RT-PCR, 2 μl of cDNA were separately amplified with 0.25 U of Taq DNA

Polymerase (Roche) in the presence of 1 μmol/L of the following primers: IL18BP upper

primer ACCATGAGACACAACTGGACACCAG, lower primer

TTAACCCTGCTGCTGTGGACTGCTG; housekeeping gene ACTB upper primer

GGCATCGTGATGGACTCCG, lower primer GCTGGAAGGTGGACAGCGA in a

Eppendorf Mastercycler ep Gradient S. PCR products were analyzed on 1% agarose gel

stained with ethidium bromide.

Patients

Clinical samples were obtained upon written informed consent and previous approval by the

Institutional Review Board from patients and tumor-free age-matched (Median=60 years

Range=43-75) women. All 55 patients were with evidence of disease and untreated or off-

treatment since at least 2 months (Table 1). Ascitic fluids were collected during surgical

procedures. Tumors histopathology, grade and stage were assigned according to the

International Federation of Gynecology and Obstetrics (FIGO) criteria.

EOC Xenotransplant model

Female homozygous non-obese-diabetic (NOD)/SCID mice (Jackson Laboratory) were bred

in-house. Nude mice were from Janvier. The experiments were performed according to the

National Regulation on Animal Research Resources and approved by the institutional

Review Board. Six-weeks old NOD/SCID animals were injected i.p. with 5x106 SKOV3 or

A2774 cells. When ascites developed, animals were euthanized and blood and ascites

collected. Nu/Nu mice were surgically implanted in the left ovary with 2x106 SKOV3 cells.

Tumour masses were excised and fixed in 10% buffered formalin.

6

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

ELISA for IL-18, IL-18BP, IFN-γ

Samples were tested with commercially available ELISA kits for human IL-18 (MBL), IL-

18BPa (DuoSet R&D Systems) and IFN-γ (Quantikine, R&D Systems). Assays were

performed in duplicates, and background values were subtracted.

Immunochemistry

Immunochemistry (IC) detection of IL-18BP was performed on sections of formaldehyde-

fixed paraffin-embedded cell pellets or tumors explanted from mice or on commercially

available tissue microarrays of EOC patients (Super Bio Chips). Antigen-retrieval was

performed with high pH citrate buffer in microwave oven. The sections were immunostained

using rabbit anti-IL-18BP (clone EP1088Y, Epitomics), anti-EBI3 (Novus Biologicals

Europe) or anti-IL27A (LifeSpan BioSciences) overnight at 4°C. The antibody complex was

revealed with the EnVision+ System-Peroxidase (Dako) and 3-amino-9-ethylcarbazole

(AEC, Calbiochem). The sections were counterstained with modified Mayer’s ematoxylin

and mounted in PermaFluor (Thermo Scientific). The sections were observed with a Nikon

Eclipse 80i light microscope equipped with a color camera imaging head, using a 40x

objective.

Public EOC datasets of gene expression

We explored IL18BP gene expression in our dataset (Iorio, GSE19532) and in public EOC

datasets processed through the Affymetrix HG U133 Plus 2.0 arrays (Tothill, GSE9891 and

Anglesio GSE12172). Only type II tumors (31) were considered for all the three datasets

when exploring pattern of correlation among different relevant . We considered only

the platforms including probes 222868_s_at because this is the only probe that comprises the

IL-18BPa isoform. The Tothill dataset consists of 18 low malignant potential (LMP), 10

Type I and 210 Type II EOC; the Anglesio dataset consists of 30 LMP and 58 Type II EOC

7

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cases; Iorio dataset included 17 EOC high grade tumors, 2 preparations of normal ovarian

surface epithelial cells (OSE) and 6 EOC cell lines. Raw data were downloaded from GEO

and normalized through the RMA algorithm (by using the Expression Console software,

Affymetrix) except for Iorio dataset for which the processed matrix was downloaded from

GEO.

Statistical analysis

The normal distribution of the data was verified before applying parametric tests by

transforming data to logarithms. The one-way ANOVA analysis of variance and appropriate

multiple comparison tests were used to compare expression levels between patients and

control subjects. The paired Student’s t test was used when appropriate. Parametric methods

were used to examine the correlation among gene or protein expression levels (Pearson’s

correlation coefficient). An alpha level of 0.05 was used for all statistical tests. GraphPad

Prism 5.0d software and R statistical language (URL: http://www.R-project.org; version

12.2) were used.

Western blot analysis

For the analysis of phosphorylated , 107 EOC cells were incubated 10 min at 37°C

with or without 20 ng/ml of rIL-27 in 0.5 ml of medium. Cells, were lysed in 100l of buffer

containing 1 mM Na Orthovanadate. Lysates were resolved under reducing conditions by

10% SDS-PAGE and analyzed by western blotting using rabbit anti-phospho-STAT1

(pY701) anti-serum (Cell Signaling Technology) or anti- actin or tubulin mAb (Sigma) and

chemiluminescence detection.

8

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Results

Gene expression of IL18BP in EOC tumors

IL18BP mRNA was first analysed in our gene expression dataset (Iorio) including primary

EOC tumors, tumor cell lines and normal ovarian surface epithelial (OSE) cells. A

significantly higher expression intensity of IL18BP was observed in tumor samples than in

EOC cell lines (P<0.0001) and in OSE cells (P=0.01) (Fig. 1A). In addition, IL18BP

expression was significantly higher in type II (high grade) tumors (31) relative to low

malignant potential tumors, in two independent datasets (Tothill and Anglesio, P=0.0043

and P<0.0001, respectively), suggesting a possible relationship of high IL18BP expression

with malignancy (Fig. 1B).

IL-18BP levels are elevated in EOC sera and ascites

We then tested IL-18BP serum levels in 48 EOC patients (Table 1), all with evidence of

disease and untreated, in 7 patients at relapse, and in 13 age-matched healthy women by

ELISA for IL-18BPa, the major isoform of IL-18BP (22). IL-18BP serum levels were higher

in both untreated (onset: M±SD=11.06±5.7; Median=9.45 ng/ml, P=0.03) and in relapsing

EOC patients (M±SD=11.76±3.2; Median=11.21 ng/ml, P=0.01) than in healthy women

(M±SD=7.4±3.5; Median=6.86 ng/ml) (Fig. 1C). By stratifying patients in stage I/II and

stage III/IV, no significant difference between the two groups was observed (P=0.43),

suggesting that IL-18BP is altered at early stages of EOC and that IL-18BP serum levels are

independent from tumor burden (Fig. 1C). In addition, when patients were stratified in type I

and in type II, according to a recent classification (31), no difference was observed between

the two groups (data not shown). Consistently, no differences were observed by stratifying

patients in accordance to tumor grading (data not shown). Performance of serum IL-18BP as

9

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

classifier, evaluated by Receiver Operating Characteristic analysis, yielded an area under the

curve of 0.734, suggestive of a poor performance (Fig. S1).

To gain information on the possible tumor origin of the high serum IL-18BP levels, we

tested 18 EOC serum and ascites pairs, collected at the same time. By paired analysis, IL-

18BP levels were significantly higher in the ascites than in serum of the same patients

(M±SD=31.9±14.7, Median 32.8 vs M±SD=11.7±5.9, Median 10.5 ng/ml P<0.0001) (Fig.

1D), thus suggesting that IL-18BP derives from the tumor site, where it could limit the IL-

18-driven immune response.

Analyses of IL-18BP correlation with IL-18 and IFN-γ

It is known that IL-18BP inhibits IL-18 as the result of a negative feed-back loop mediated

by IL-18-induced IFN-γ (21, 23). In agreement with our previous data (13), IL-18 ELISA

levels were elevated in EOC serum and ascites (Fig. S2A and B).

We therefore first evaluated whether there was any relationship between IL18 and IL18BP

mRNA in two public access datasets of ovarian cancers and found a moderate, yet

significant correlation (Anglesio dataset P=0.0056; Tothill dataset P<0.0001), (Fig. 2A). In

addition, a correlation was also found for IFNG and IL18BP and for IFNG and IL18 in the

Tothill dataset (IFNG vs IL18 P<0.0001; IL18BP vs IFNG P<0.0001), although only a

minority of patients showed elevated levels of IFNG expression (Fig. 2B).

However, when we investigated possible correlations at the protein level, we found no

significant correlation between IL-18 and IL-18BP in the sera (r= 0.12, P=ns by Pearson’s

correlation) and in the ascites (r=0.3, P=ns) of EOC patients (Fig. S2A and B). Moreover,

IFN-γ levels were low or undetectable in ascites and sera from EOC patients, in agreement

with previous reports, and showed no correlation with IL-18 and IL-18BP (Fig. S2C).

Although microarray data suggest that the IL-18/IFN-γ axis may be involved in the

10

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

regulation of IL-18BP expression in the tumor tissue of a few patients, no evidence of this

regulation was found in sera and ascites.

Both EOC cells and reactive cells from the microenvironment express IL-18BP

To address the possible cellular origin of the elevated IL-18BP in EOC we performed

immunochemical analyses on cells isolated from the ascites and on tissue arrays. Among

cells present in EOC ascites, tumor cell nests showed positivity for IL-18BP, although a

stronger expression was found in tumor-associated leukocytes (Fig. 3A). Staining appeared

specific as no reactivity was found on a contiguous section stained with secondary

antiserum. Immunohistochemical analyses of tissue microarrays revealed that most EOC

tumors expressed IL-18BP, irrespective of the tumor histotype, although at variable intensity

in different tumors. Both neoplastic cells and infiltrating leukocytes showed expression of

IL-18BP (Fig. 3B).

The IL-18BP-expressing leukocytes showed monocyte- or granulocyte-like morphological

features and had a non-lymphoid (CD3-CD20-NKP46-) but myeloid (CD13+CD14-/low)

surface phenotype (Fig. S3). These features may be consistent with “myeloid-derived

suppressor cells” (32). Altogether these data indicate that different cell populations express

IL-18BP in the EOC microenvironment. By contrast normal ovary and Fallopian tube

showed no expression of IL-18BP by immunohistochemistry (Fig. 3C).

IFN-γ and IL-27 up-regulate IL-18BP in human EOC cell lines

Different from EOC cells present in tumor specimen, four EOC cell lines showed no

constitutive expression of IL-18BP mRNA or protein (Fig. 4). However, culture in the

presence of IFN-γ increased IL-18BP protein secretion (Fig. 4A) and IL18BP mRNA (Fig.

4B) expression in EOC cell lines. In addition, although human IL-18BP was undetectable in

11

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

sera, it was found in the ascites of nude mice bearing orthotopic xenotransplants of the

human A2774 and SKOV3 cell lines (Fig. 4C), further suggesting that EOC cells can

contribute to IL-18BP production in vivo. Indeed, A2774 and SKOV3 cells grown in

immune deficient mice showed IL-18BP expression by immunohistochemistry, while IL-

18BP was virtually undetectable in the same EOC cell lines in vitro (Fig. S4). These

findings suggest that factor(s) present in the microenvironment are responsible for

production of IL-18BP in vivo. Although IFN-γ mediates IL-18BP expression in EOC cell

lines in vitro, it should not be involved in vivo, as it was virtually undetectable in human

ascites from patients and, moreover, mouse IFN-γ is inactive on human cells. These

considerations prompted us to examine whether other cytokines, known to be elevated in

EOC patients, such as IL-6, TNF-α, VEGF-A, EGF, IL-18 and IL-8 could mediate IL-18BP

expression in EOC cell lines, but no one proved active (data not shown).

A recent report indicated that the heterodimeric cytokine IL-27, consisting of EBI3 and IL-

27 chains, could mediate IL-18BP production by human keratinocytes in an IFN-γ-

independent manner (33). We then tested whether IL-27 or the related cytokine IL-35 could

mediate IL-18BP expression in EOC cells. Indeed, IL-27 induced IL-18BP secretion (Fig.

5A; S5A-B) in four EOC cell lines in a dose-dependent fashion, while IL-35 showed no

activity (not shown). IL-27 also increased IL-18BP mRNA expression (Fig. S5C)

Importantly, the IL-18BP-containing supernatant of IL-27-stimulated A2780 cells

significantly inhibited IL-18 bioactivity in a concentration-dependent manner, as detected

through IFN-γ release by the human NK cell line NK-92. Controls such as IL-27-containing

medium or the supernatant from unstimulated EOC cells produced no inhibition (Fig 5B).

As IL-18BP expression is activated through the STAT1 pathway, we also analyzed STAT1

activation by IL-27 in EOC cells. Indeed, IL-27 activates STAT1 signaling in EOC cell lines

(Fig. 5C; S5D), as reported for other IL-27-sensitive cell types (33, 34). In addition, western

12

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

blot analysis showed that STAT1 was constitutively tyrosine-phosphorylated in cells

isolated from ascites ex-vivo and was further activated by in vitro treatment with IL-27 (Fig.

5C). Confocal microscopy confirmed constitutive STAT1 phosphorylation, which increased

with IL-27 stimulation, in both EPCAM+ EOC and EPCAM- reactive cells (Fig. S5E).

Consistently, cells isolated from ascites showed spontaneous IL-18BP secretion in culture,

which could be further enhanced by in vitro IL-27 stimulation (Fig. 5A).

As IL-27 activity is mediated through a heterodimeric receptor consisting of gp-130 and

WSX-1 molecules we asked whether antibodies neutralizing gp-130 could inhibit the effect

of IL-27. Anti-gp130 mAb significantly inhibited IL-27-mediated IL-18BP production in

two different EOC cell lines, further supporting the involvement of the IL-27/IL-27R

pathway in IL-18BP regulation (Fig. 5A).

IL-27A and EBI3 are expressed in EOC tissues

Further analyses of two microarray datasets of EOC indicated a correlation between the

expression of EBI3 and IL18BP mRNA in EOC primary tumors (Fig. S6A). In addition,

although IL18BP mRNA expression showed no significant correlation with outcome (not

shown), high levels of EBI3 expression correlated with a shorter relapse-free survival (Fig

S6B). The correlation between IL18BP and EBI3 gene expression suggested a possible

paracrine loop of IL-18BP induction in vivo. This hypothesis was also suggested by the use

of an anti-IL-27A (p28) specific antibody in IC, which revealed IL-27 expression

predominantly in a fraction of tumor-associated leukocytes isolated from the ascites and in

tissue microarrays, while tumor cell nests appeared negative (Fig. 5C, D). Also EBI3

protein showed a similar distribution both in ascites (Fig. S7A) and within tumor tissues

(Fig. S7B). Altogether our data are consistent with a paracrine activation of IL-18BP

expression in the tumor microenvironment.

13

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Discussion

In this study we show that IL-18BP levels are elevated in the serum of EOC patients and are

even higher in the ascites, reaching 4-fold higher levels than those found in normal serum.

This finding suggested a local production of IL-18BP in the microenvironment of EOC.

Indeed, immunochemical analyses showed that IL-18BP is expressed by neoplastic cells of

different EOC histotypes and by tumor-associated leukocytes with myeloid features.

Therefore the high concentration of IL-18BP in the tumor environment of EOC may limit

the effect of endogenous or exogenously administered IL-18. On the other hand, IL-18BP

also binds the anti-inflammatory cytokine IL-37 (35), which may suppress the host immune

response (36) and this may result in a beneficial effect for the host. However, to our

knowledge, no evidence for IL-37 expression in ovarian cancer has been provided so far.

Elevated IL-18BP levels were recently described also in serum of patients with prostatic

(24) and pancreatic cancer (37). In the latter, the concomitantly elevated levels of free-IL-18

in the serum suggested the existence of a biological paradox, in view of the immune-

enhancing properties of IL-18. Indeed, increased levels of both IL-18 and its natural

inhibitor IL-18BP, were also found in SLE patients, where biologically active free IL-18

was still higher than in controls and was a marker of disease activity and a potential

contributor to autoimmunity (38).

It was previously demonstrated that high levels of IL-18 are present in serum and ascites of

EOC patients (12) and that pro-IL-18 is largely predominant (13). In fact, although IL-18

ELISA preferentially recognized mat-IL-18 in sera, this assay also detected pro-IL-18,

although with a reduced sensitivity. Therefore the presence of “free IL-18” in EOC, could be

explained by the predominance of pro-IL-18, which is unable to bind IL-18BP and is

detected by IL-18 ELISA. It is likely that a similar situation may occur in other tumors

where alterations of IL-18 processing have been reported (39).

14

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

As the presence of mat-IL-18 in the ascites of EOC could not be formally excluded and mat-

IL-18 is an inducer of IL-18BP, via IFN-γ production (25, 40), we explored possible

correlations between immune-reactive IL-18 and IL-18BP or IFN-γ levels. No significant

correlation was found in the ascites and serum of EOC patients, and IFN-γ levels were very

low to undetectable in the ascites. However, a correlation between IL18 and IL18BP mRNA

levels was found in two independent datasets of EOC gene expression profiles. Moreover

IFNG mRNA showed a correlation with IL18BP in one dataset, although IFNG mRNA was

elevated only in a minority of cases. These data suggest that the IL-18/IFN-γ loop may be

active in the tumor tissue microenvironment in some patients and that other factor(s) may

participate in the induction of IL-18BP expression.

Several evidences suggested that IL-18BP production is a result of the interaction between

EOC cells and the microenvironment. Human EOC cell lines do not produce IL-18BP in

culture but once grafted in immune-deficient mice they display IL-18BP expression,

suggesting a role for factor(s), which function across the species. This is not the case for

IFN-γ, in view of its species-specificity. As other cytokines, which are elevated in the ascites

of EOC failed to induce IL-18BP expression in vitro, we focused on IL-27, which was

recently shown to stimulate IL-18BP expression in human keratinocytes (33).

IL-27 is a member of the IL-12 family that may have pro- or anti-inflammatory properties in

different systems (29, 30). IL-27 is a heterodimeric cytokine, composed of p28 and EBV-

induced gene 3 (EBI3), which up-regulates IL-12R expression and is relevant for Th1

polarization (41, 42). However, the precise contribution of IL-27 to immune response,

inflammation and cancer is still poorly understood. On one hand, IL-27 has pro-

inflammatory effects through the induction of CXCL10 in macrophages in inflammatory

skin disorders (43). However, IL-27 may limit the pro-inflammatory and immune-enhancing

activities of IL-18 in the skin through IL-18BP induction (33) and may dampen

15

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

autoimmunity, as Il27-/- mice were more susceptible to experimental autoimmune

encephalomyelitis (44).

Here we show that IL-27 induces the expression of IL-18BP mRNA and protein in human

EOC cell lines in culture and activates STAT1 signaling in these cells, while IL-35, another

EBI3 containing cytokine (30), was inactive. Such activity was specifically induced through

the IL-27R complex as indicated by the significant inhibition of IL-18BP induction upon

treatment with a neutralizing antibody against the gp130 chain. A potential role of IL-27 in

vivo was suggested by the expression of IL-27A and EBI3 found by immunochemistry in

tumor-associated leukocytes in both ascites and tumor tissues and by the correlation between

EBI3 and IL18BP mRNA expression in two different EOC datasets. Interestingly high EBI3

expression correlated with a shorter PFS in type II tumors. The finding that IL18BP gene

expression had no significant correlation with relapse-free survival of patients with type II

tumors, may reflect the multiplicity of components driving clinical outcome. A correlation

with IL27A mRNA expression could not be found (data not shown), but this may relate to

technical limitations, as only one probeset was present in the arrays. The possible role of IL-

27 in vivo was reinforced by the detection of constitutive STAT1 activation in both

neoplastic and reactive cells isolated from the ascites, in the absence of measurable IFN-γ

levels. Moreover, these ascites cells showed spontaneous secretion of IL-18BP in culture,

which could be further enhanced by the addition of exogenous IL-27. Consistently also

STAT1 phosphorylation was increased by IL-27.

Our study may open new perspectives on understanding the role of IL-27 in cancer, since its

involvement in the anti-tumor immune response is still poorly understood. In some

hematological neoplasia including multiple myeloma (45) and acute leukemias (46, 47) IL-

27 displays direct and indirect anti-tumor effects. We thus hypothesize that in EOC IL-27

may be part of an immune-regulatory network, which limits the induction of Th1 responses

16

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and IFN-γ production in the microenvironment by inhibiting IL-18 activity. In support of

this concept, studies in murine models highlighted a predominant role of IL-27 as an

immune-regulatory and anti-inflammatory agent that generates and maintains Treg cell

functions (48) and induces IL-10 production by T lymphocytes (49).

Although the role of IL-18 in tumor cell biology has been debated (50), pre-clinical studies

indicated that IL-18 displays anti-tumor activity through its ability to trigger IFN-γ

production and to favour the induction of a Th1 response (18, 19). Therefore recombinant

IL-18 is undergoing testing in clinical trials of cancer immunotherapy (27, 28) and in

particular a clinical phase I study of IL-18 and doxil in advanced stage EOC

(NCT00659178) has recently concluded patient recruitment. It might be possible that the

high local levels of IL-18BP present in EOC may limit the biological effects of low levels of

endogenous IL-18 or of therapeutically administered IL-18, particularly at the tumor site.

17

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1: Expression of IL18BP mRNA and secretion of IL-18BP protein in EOC.

A: Normalized IL18BP gene expression levels in EOC tumors, cell lines and OSE.

Expression levels were significantly higher in tumor samples (one-way ANOVA and

Bonferroni’s post-test). B: IL18BP mRNA expression was significantly higher in high-grade

tumors (Type II) relative to low malignant potential tumors (LMP), in two independent

datasets by two-sided unpaired Student’s t test (Anglesio dataset) and one-way ANOVA and

Tukey’s post-test (Tothill dataset). Boxes and whiskers represent median and quartiles with

minimum and maximum. C: IL-18BP ELISA levels were significantly higher in sera from

EOC patients at diagnosis (n. 48) or at relapse (n. 7) than in age matched female controls (n.

13) by Kruskal-Wallis test. IL-18BP levels were similar in stage I/II and stage III/IV patients

D: IL-18BP levels were higher in the ascites than in sera simultaneously collected from the

same patient (by two-sided paired Student’s t test). Mean and SD are indicated in C and D.

Figure 2: Analysis of the correlation between IL18, IL18BP and IFNG mRNA levels in high

grade (Type II) tumors. A: A significant correlation was found between IL18 and IL18BP in

both datasets. B: A correlation between IL18 and IFNG, and between IL18BP and IFNG

was also found only in the Tothill dataset. Pearson’s correlation coefficients are shown (r).

Lines represent the best fit linear regression analysis with the 95% CI.

Figure 3: Immunochemical analysis of IL-18BP expression in EOC and in normal tissue

counterparts. A: IL-18BP staining of cells isolated from ascites. In the upper panel both

tumor cell nests and particularly tumor-associated leukocytes show staining for IL-18BP.

Negative control of a contiguous section stained only with secondary antibody is shown.

Bar=100μm. Arrows indicate tumor cell nests. Arrowheads indicate tumor-associated

18

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

leukocytes. B: EOC Tissue array staining. Different EOC histotypes show expression of IL-

18BP in tumor cells and infiltrating leukocytes. C: Normal ovary and Fallopian tube show

virtually no positivity for IL-18BP staining. Negative controls of contiguous sections are

shown for each panel.

Figure 4: IL-18BP expression by EOC cell lines. A: IL-18BP secretion is detected by

ELISA in culture supernatants following EOC cell treatment with IFN-γ (100 and 1000

U/ml for 24 h. * P<0.05, ** P<0.01 by two-sided unpaired Student’s t-test). B: IFN-γ

induced IL18BP mRNA expression in EOC cell lines as detected by RT-PCR analysis. C:

Human IL-18BP ascites levels were measured in NOD-SCID mice bearing orthotopic

xenotransplants of the SKOV3 (n. 5) and A2774 (n. 2) EOC cell lines.

Figure 5: Involvement of IL-27 in IL-18BP expression in EOC cells. A: IL-18BP secretion

is detected by ELISA in culture supernatants following A2780 and A2774 EOC cell

stimulation with IL-27 (20 ng/ml for 48h P<1E-05 vs untreated control). Anti-gp130 mAb

(from 0.5 to 500 ng/ml) significantly inhibits IL-18BP secretion mediated by IL-27

stimulation (* P<0.05, ** P<0.001, *** P<1E-05). Black bars show constitutive and IL-27-

induced IL-18BP in vitro secretion by cells isolated from patient’s ascites A98,

representative of three cases with similar results. B: The conditioned medium of IL-27-

stimulated A2780 cells containing 2ng/ml IL-18BP (BP+cm), significantly inhibits IL-18-

induced IFN-γ release by the NK-92 cells in a concentration (50, 25 or 12%v/v)-dependent

fashion (* P<0.05, ** P<0.001). As controls, conditioned medium of unstimulated cells

(ctrl cm) and IL-27-containing medium (IL-27+ cm) were used. C: Western-blot analysis of

tyrosine-phosphorylated STAT1 protein in A2780 cells and in cells isolated from three

different ascites unstimulated or stimulated for 10 min with IL-27 (20ng/ml); β-actin was

19

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

used as loading control. D: In the upper panel tumor-associated leukocytes show staining for

IL-27A, while tumor cell nests show no reactivity. Negative control of a contiguous section

is shown. Bar=100μm. Arrows indicate examples of negative tumor cell nests. E: EOC

Tissue array staining. Different EOC histotypes show expression of IL-27A in infiltrating

leukocytes (enlarged in the inset).

20

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1. Martin LP, Schilder RJ. Management of recurrent ovarian carcinoma: current status and future directions. Semin Oncol 2009;36:112-25.

2. Ferrini S, Biassoni R, Moretta A, Bruzzone M, Nicolin A, Moretta L. Clonal analysis of T lymphocytes isolated from ovarian carcinoma ascitic fluid. Phenotypic and functional characterization of T-cell clones capable of lysing autologous carcinoma cells. Int J Cancer 1985;36:337-43.

3. Nelson BH. The impact of T-cell immunity on ovarian cancer outcomes. Immunol Rev 2008;222:101-16.

4. Frumento G, Piazza T, Di Carlo E, Ferrini S. Targeting tumor-related immunosuppression for cancer immunotherapy. Metab Immune Disord Drug Targets 2006;6:233-7.

5. Preston CC, Goode EL, Hartmann LC, Kalli KR, Knutson KL. Immunity and immune suppression in human ovarian cancer. Immunotherapy 2011;3:539-56.

6. Nilsson MB, Langley RR, Fidler IJ. Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Res 2005;65:10794-800.

7. Leinster DA, Kulbe H, Everitt G, Thompson R, Perretti M, Gavins FN, et al. The peritoneal tumour microenvironment of high-grade serous ovarian cancer. J Pathol 2012;227:136-45.

8. Berek JS, Chung C, Kaldi K, Watson JM, Knox RM, Martinez-Maza O. Serum interleukin-6 levels correlate with disease status in patients with epithelial ovarian cancer. Am J Obstet Gynecol 1991;164:1038-43.

9. Duan Z, Feller AJ, Penson RT, Chabner BA, Seiden MV. Discovery of differentially expressed genes associated with paclitaxel resistance using cDNA array technology: analysis of interleukin (IL) 6, IL-8, and monocyte chemotactic protein 1 in the paclitaxel-resistant phenotype. Clin Cancer Res 1999;5:3445-53.

10. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004;10:942-9.

11. Lambeck AJ, Crijns AP, Leffers N, Sluiter WJ, ten Hoor KA, Braid M, et al. Serum cytokine profiling as a diagnostic and prognostic tool in ovarian cancer: a potential role for . Clin Cancer Res 2007;13:2385-91.

12. Le Page C, Ouellet V, Madore J, Hudson TJ, Tonin PN, Provencher DM, et al. From gene profiling to diagnostic markers: IL-18 and FGF-2 complement CA125 as serum-based markers in epithelial ovarian cancer. Int J Cancer 2006;118:1750-8.

13. Orengo AM, Fabbi M, Miglietta L, Andreani C, Bruzzone M, Puppo A, et al. Interleukin (IL)-18, a biomarker of human ovarian carcinoma, is predominantly released as biologically inactive precursor. Int J Cancer 2011;129:1116-25.

21

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

14. Okamura H, Tsutsui H, Komatsu T, Yutsudo M, Hakura A, Tanimoto T, et al. Cloning of a new cytokine that induces interferon-gamma. Nature 1995;378:88-91.

15. Takeda K, Tsutsui H, Yoshimoto T, Adachi O, Yoshida N, Kishimoto T, et al. Defective NK cell activity and Th1 response in IL-18-deficent mice. Immunity 1998;8:383-90.

16. Dinarello CA. IL-18: a Th1-inducing, proinflammatory cytokine and new member of the IL- 1 family. J Allergy Clin Immunol 1999;103:11-24.

17. Ghayur T, Banerjee S, Hugunin M, Butler D, Herzog L, Carter A, et al. Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature 1997;386:619-23.

18. Heuer JG, McClung CT, Hock RA. Neuroblastoma cells expressing mature IL-18, but not pro-IL-18, induce a strong and immediate antitumour immune response. J Immunother 1999;22:324-35.

19. Nakamori M, Iwahashi M, Nakamura M, Ueda K, Zhang X, Yamaue H. Intensification of antitumor effect by T helper 1-dominant adoptive immunogene therapy for advanced orthotopic colon cancer. Clin Cancer Res 2003;9:2357-65.

20. Wang ZY, Gaggero A, Rubartelli A, Rosso O, Miotti S, Mezzanzanica D, et al. Expression of interleukin-18 in human ovarian carcinoma and normal ovarian epithelium: evidence for defective processing in tumor cells. Int J Cancer 2002;98:873-8.

21. Novick D, Kim SH, Fantuzzi G, Reznikov LL, Dinarello CA, Rubinstein M. Interleukin-18 binding protein: a novel modulator of the Th1 cytokine response. Immunity 1999;10:127-36.

22. Kim SH, Eisenstein M, Reznikov L, Fantuzzi G, Novick D, Rubinstein M, et al. Structural requirements of six naturally occurring isoforms of the IL-18 binding protein to inhibit IL- 18. Proc Natl Acad Sci USA 2000;97:1190-5.

23. Veenstra KG, Jonak ZL, Trulli S, Gollob JA. IL-12 induces monocyte IL-18 binding protein expression via IFN-gamma. J Immunol 2002;168:2282-7.

24. Fujita K, Ewing CM, Isaacs WB, Pavlovich CP. Immunomodulatory IL-18 binding protein is produced by prostate cancer cells and its levels in urine and serum correlate with tumor status. Int J Cancer 2011;129:424-32.

25. Paulukat J, Bosmann M, Nold M, Garkisch S, Kämpfer H, Frank S, et al. Expression and release of IL-18 Binding Protein in Response to INF-γ. J Immunol 2001;167:7038-43.

26. Dinarello CA, Novick D, Rubinstein M, Lonnemann G. and interleukin 18 binding protein: possible role in immunosuppression of chronic renal failure. Blood Purif 2003;21:258-70.

27. Robertson MJ, Mier JW, Logan T, Atkins M, Koon H, Koch KM, et al. Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer. Clin Cancer Res 2006;12:4265-73.

22

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

28. Robertson MJ, Kirkwood JM, Logan TF, Koch KM, Kathman S, Kirby LC, et al. A dose- escalation study of recombinant human interleukin-18 using two different schedules of administration in patients with cancer. Clin Cancer Res 2008;14:3462-9.

29. Trinchieri G, Pflanz S, Kastelein RA. The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses. Immunity 2003;19:641-4.

30. Vignali DA, Kuchroo VK. IL-12 family cytokines: immunological playmakers. Nat Immunol 2012;13:722-8.

31. Kurman RJ, Shih IeM. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer-Shifting the paradigm. Hum Pathol 2011;42:918-31.

32. Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol. 2009;182:4499-506.

33. Wittmann M, Doble R, Bachmann M, Pfeilschifter J, Werfel T, Mühl H. IL-27 Regulates IL-18 Binding Protein in Skin Resident Cells. PLoS One. 2012;7:e38751.

34. Bachmann M, Paulukat J, Pfeilschifter J, Mühl H. Molecular mechanisms of IL-18BP regulation in DLD-1 cells: pivotal direct action of the STAT1/GAS axis on the promoter level. J Cell Mol Med 2009;13:1987-94.

35. Bufler P, Azam T, Gamboni-Robertson F, Reznikov LL, Kumar S, Dinarello CA, Kim SH. A complex of the IL-1 homologue IL-1F7b and IL-18-binding protein reduces IL-18 activity. Proc Natl Acad Sci U S A. 2002;99:13723-8.

36. Nold MF, Nold-Petry CA, Zepp JA, Palmer BE, Bufler P, Dinarello CA. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol. 2010;11:1014-22.

37. Carbone A, Vizio B, Novarino A, Mauri FA, Geuna M, Robino C, et al. IL-18 paradox in pancreatic carcinoma: elevated serum levels of free IL-18 are correlated with poor survival. J Immunother 2009;32:920-31.

38. Migliorini P, Anzilotti C, Pratesi F, Quattroni P, Bargagna M, Dinarello CA, et al. Serum and urinary levels of IL-18 and its inhibitor IL-18BP in systemic lupus erythematosus. Eur Cytokine Netw 2010;21:264-71.

39. Pagès F, Berger A, Henglein B, Piqueras B, Danel C, Zinzindohoue F, et al. Modulation of interleukin-18 expression in human colon carcinoma: consequences for tumour immune surveillance. Int J Cancer 1999;84:326-30.

40. Fantuzzi G, Reed D, Qi M, Scully S, Dinarello CA, Senaldi G. Role of interferon regulatory factor-1 in the regulation of IL-18 production and activity. Eur J Immunol. 2001;31:369-75.

41. Pflanz S, Timans JC, Cheung J, Rosales R, Kanzler H, Gilbert J, et al. IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4(+) T cells. Immunity 2002;16:779-90.

23

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

42. Lucas S, Ghilardi N, Li J, de Sauvage FJ. IL-27 regulates IL-12 responsiveness of naive CD4+ T cells through Stat1-dependent and -independent mechanisms. Proc Natl Acad Sci USA 2003;100:15047-52.

43. Shibata S, Tada Y, Kanda N, Nashiro K, Kamata M, Karakawa M, et al. Possible roles of IL-27 in the pathogenesis of psoriasis. J Invest Dermatol 2010;130:1034-9.

44. Batten M, Li J, Yi S, Kljavin NM, Danilenko DM, Lucas S, et al. limits autoimmune encephalomyelitis by suppressing the development of -producing T cells. Nat Immunol 2006;7:929-36.

45. Cocco C, Giuliani N, Di Carlo E, Ognio E, Storti P, Abeltino M, et al. Interleukin-27 acts as multifunctional antitumor agent in multiple myeloma. Clin Cancer Res 2010;16:4188-97.

46. Zorzoli A, Di Carlo E, Cocco C, Ognio E, Ribatti D, Ferretti E, et al. Interleukin-27 inhibits the growth of pediatric acute myeloid leukemia in NOD/SCID/Il2rg-/- mice. Clin Cancer Res 2012;18:1630-40.

47. Canale S, Cocco C, Frasson C, Seganfreddo E, Di Carlo E, Ognio E, et al. Interleukin-27 inhibits pediatric B-acute lymphoblastic leukemia cell spreading in a preclinical model. Leukemia. 2011;25:1815-24.

48. Wojno ED, Hosken N, Stumhofer JS, O'Hara AC, Mauldin E, Fang Q, et al. A role for IL-27 in limiting T regulatory cell populations. J Immunol 2011;187:266-73.

49. Awasthi A, Carrier Y, Peron JP, Bettelli E, Kamanaka M, Flavell RA, et al. A dominant function for interleukin 27 in generating -producing anti-inflammatory T cells. Nat Immunol 2007;8:1380-9.

50. Vidal-Vanaclocha F, Mendoza L, Telleria N, Salado C, Valcárcel M, Gallot N, et al. Clinical and experimental approaches to the pathophysiology of interleukin-18 in cancer progression. Cancer Metastasis Rev 2006;25:417-34.

24

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 1. Distribution by tumor characteristics for EOC patients with evidence of disease.  N. of cases N. of cases at diagnosis at relapse Total 48 7 Age at diagnosis >55 years 35 5 55 years 12 NA1 1 2 Stage2 I 15 II 7 III 22 3 IV 3 3 NA 1 1 Histotype Serous 30 4 Endometrioid 10 1 Mucinous 3 Others 3 1 NA 2 1 Grade 1 13 2 19 3 3 15 3 Borderline NA 1 1

1 NA not available 2 stage at diagnosis

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 19, 2013; DOI: 10.1158/1078-0432.CCR-13-0568 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

The IL-18 antagonist IL-18 Binding Protein is produced in the human ovarian cancer microenvironment

Grazia Carbotti, Gaia Barisione, Anna M. Orengo, et al.

Clin Cancer Res Published OnlineFirst July 19, 2013.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-13-0568

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2013/07/23/1078-0432.CCR-13-0568.DC1

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

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

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

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2013/07/26/1078-0432.CCR-13-0568. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 28, 2021. © 2013 American Association for Cancer Research.