Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer

Lucia De Monte, Michele Reni, Elena Tassi, Daniela Clavenna, Ilenia Papa, Helios Recalde, Marco Braga, Valerio Di Carlo, Claudio Doglioni, Maria Pia Protti, Lucia De Monte, Michele Reni, Elena Tassi, Daniela Clavenna, Ilenia Papa, Helios Recalde, Marco Braga, Valerio Di Carlo, Claudio Doglioni, Maria Pia Protti

Abstract

Pancreatic cancer is a very aggressive disease characterized by a marked desmoplasia with a predominant Th2 (GATA-3+) over Th1 (T-bet+) lymphoid infiltrate. We found that the ratio of GATA-3+/T-bet+ tumor-infiltrating lymphoid cells is an independent predictive marker of patient survival. Patients surgically treated for stage IB/III disease with a ratio inferior to the median value had a statistically significant prolonged overall survival, implying an active role for Th2 responses in disease progression. Thymic stromal lymphopoietin (TSLP), which favors Th2 cell polarization through myeloid dendritic cell (DC) conditioning, was secreted by cancer-associated fibroblasts (CAFs) after activation with tumor-derived tumor necrosis factor α and interleukin 1β. TSLP-containing supernatants from activated CAFs induced in vitro myeloid DCs to up-regulate the TSLP receptor (TSLPR), secrete Th2-attracting chemokines, and acquire TSLP-dependent Th2-polarizing capability in vitro. In vivo, Th2 chemoattractants were expressed in the tumor and in the stroma, and TSLPR-expressing DCs were present in the tumor stroma and in tumor-draining but not in nondraining lymph nodes. Collectively, this study identifies in pancreatic cancer a cross talk between tumor cells and CAFs, resulting in a TSLP-dependent induction of Th2-type inflammation which associates with reduced patient survival. Thus, blocking TSLP production by CAFs might help to improve prognosis in pancreatic cancer.

Figures

Figure 1.
Figure 1.
The ratio of G/T tumor-infiltrating lymphoid cells predicts survival after surgery in patients with stage IB/III pancreatic cancer. (A) Representative immunohistochemical analysis for lymphoid GATA-3 and T-bet staining in the tumor (n = 69; left) and normal pancreatic tissue from benign lesions (n = 3; right). The arrows indicate rare positive cells in normal tissue. Bars, 100 µm. (B) Percentage of GATA-3+ (circles) and T-bet+ (triangles) lymphoid cells for each of the analyzed tumor samples (n = 69). The values are significantly different and indicated as: ***, P < 0.001 (determined by paired one-tailed Student’s t test). (C) Waterfall plot of the G/T ratio for each tumor sample. The dashed line indicates the G/T ratio 5.2, which is the median value. (D) Kaplan-Meier curve for overall survival by median G/T ratio (5.2). Survival significantly decreased (P = 0.008) as a function of a G/T ratio ≥ 5.2.
Figure 2.
Figure 2.
TSLP is expressed by CAFs, and its secretion is induced by proinflammatory cytokines released by tumor cells. (A) TSLP mRNA expression in tumor and surrounding tissues, tumor cell lines, CAFs, and HDFs. Each dot represents a different surgical sample or a different cell line. (B) TSLP is expressed in vivo in the stroma. LCM (left) was used to collect stromal (top) and epithelial (bottom) tumor cells from surgical specimens and TSLP mRNA expression analyzed (right; representative of tumor samples from three patients). The mRNA expression levels were normalized to the expression of GAPDH. TSLP mRNA expression of Caco2 cell line was used as calibrator, as in Rimoldi et al. (2005). (C) TSLP protein secretion by CAFs, HDFs, and tumor cell lines treated with proinflammatory cytokines as single agent or in combination. Left, 20 ng/ml TNF; middle, 10 ng/ml IL-1β; right, TNF plus IL-1β. Each dot represents a different cell line. (D) TNF (top) and IL-1β (bottom) mRNA expression in tumor and the corresponding surrounding tissue (each dot and corresponding triangle represent surgical samples from single patients). (E) TNF (left) and IL-1β (right) mRNA expression in tumor epithelial and stromal cells collected by LCM (top; representative of tumor samples from three patients) and in isolated and in vitro–cultured paired tumor cell lines and CAFs from the same patient (bottom; representative of tumor samples from three patients). Inset magnifies IL-1β expression in CAFs. (F) TSLP protein secretion by CAFs treated with supernatant from tumor cell lines (tumor cells sup) in the absence and presence of anti-TNF, anti–IL-1β, and isotype control Abs (representative of experiments performed with three different tumor cell lines and three CAFs). Data in A–F are means of at least duplicate determinations ± SD. Responses significantly different in A, C, D, and F are indicated as: *, P < 0.05; **, 0.001< P < 0.01 (determined by paired or unpaired one-tailed Student’s t test).
Figure 3.
Figure 3.
Supernatant of TNF-treated CAF (CAF sup) activates in vitro myeloid DCs with features of TSLP-treated DCs. (A) FACS analysis of DCs after 24-h incubation with the indicated stimuli (representative of independent experiments, n = 3). Experiments were performed with supernatants from three different CAFs. Open histograms represent staining of DC activation markers; filled histograms represent the isotype control. (B) FACS analysis of TSLPR expression by DCs activated in the presence of medium plus TNF, supernatant of TNF-treated HDF (HDF sup; obtained from two different HDFs), and CAF sup (obtained from three different CAFs; representative of independent experiments n = 3). (C) FACS analysis of CD80 expression by DCs incubated with CAF sup (obtained from three different CAFs) in the absence and in the presence of an anti-TSLP Ab (representative of independent experiments, n = 4). Experiments with medium plus TNF and TSLP, in the absence and in the presence of the anti-TSLP Ab, are shown as negative and positive controls, respectively. (D) Chemokine production by DCs activated with the following stimuli: medium alone, 20 ng/ml TNF; 15 ng/ml TSLP; HDF sup and CAF sup (representative of three experiments performed with three different CAFs and two different HDFs). (E) CAF sup endows DCs with Th2 polarizing capability that depends on TSLP. CD4+CD45RA+ naive T cells were cultured with DCs previously activated with the indicated stimuli. At day 6, IFN-γ and IL-13 secreted by CD4+ T cells were tested by ELISA. When indicated, anti-TSLP and anti-TNF Abs were added in culture. Data are means of duplicate determinations ± SD and represent one of five experiments (performed with four different CAFs and two different HDFs). Release of IL-13 significantly lower in the presence of an anti-TSLP Ab are indicated as: *, P < 0.05; **, 0.001 < P < 0.01 (determined by unpaired one-tailed Student’s t test).
Figure 4.
Figure 4.
TSLP-conditioned DCs and Th2-attractant chemokines are present in vivo. (A and B) TSLP-conditioned DCs are present in the tumor and in draining but not in nondraining LNs. (A) Immunohistochemical analysis for CD11c+TSLPR+ cells in the tumor representative of tumor samples from 10 patients. Bars, 100 µm. (B) FACS analysis of CD11c+TSLPR+CD14− cells in draining (top) and corresponding nondraining (bottom) LNs representative of paired samples from four patients. Open histograms represent TSLPR staining; filled histograms represent the isotype control. Numbers indicate the percentage of gated (left) and positive (right) cells. (C and D) TARC/CCL17 and MDC/CCL22 mRNA expression in tumor and the corresponding surrounding tissue (C; each dot and corresponding triangle represent surgical samples from single patients), in tumor epithelial and stromal cells collected by LCM (D, top; representative of tumor samples from three patients), and in isolated and in vitro–cultured paired tumor cell lines and CAFs from the same patient (D, bottom; representative of tumor samples from three patients). Data in C and D are means of at least duplicate determinations ± SD. Responses significantly different are indicated as: *, P < 0.05 (determined by paired one-tailed Student’s t test).

References

    1. Arlt A., Vorndamm J., Müerköster S., Yu H., Schmidt W.E., Fölsch U.R., Schäfer H. 2002. Autocrine production of interleukin 1beta confers constitutive nuclear factor kappaB activity and chemoresistance in pancreatic carcinoma cell lines. Cancer Res. 62:910–916
    1. Atzeni F., Sarzi-Puttini P. 2009. Anti-cytokine antibodies for rheumatic diseases. Curr. Opin. Investig. Drugs. 10:1204–1211
    1. Bogiatzi S.I., Fernandez I., Bichet J.C., Marloie-Provost M.A., Volpe E., Sastre X., Soumelis V. 2007. Cutting edge: proinflammatory and Th2 cytokines synergize to induce thymic stromal lymphopoietin production by human skin keratinocytes. J. Immunol. 178:3373–3377
    1. Burris H.A., III, Moore M.J., Andersen J., Green M.R., Rothenberg M.L., Modiano M.R., Cripps M.C., Portenoy R.K., Storniolo A.M., Tarassoff P., et al. 1997. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J. Clin. Oncol. 15:2403–2413
    1. Curiel T.J., Coukos G., Zou L., Alvarez X., Cheng P., Mottram P., Evdemon-Hogan M., Conejo-Garcia J.R., Zhang L., Burow M., et al. 2004. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 10:942–949 10.1038/nm1093
    1. Dallal R.M., Christakos P., Lee K., Egawa S., Son Y.I., Lotze M.T. 2002. Paucity of dendritic cells in pancreatic cancer. Surgery. 131:135–138 10.1067/msy.2002.119937
    1. De Monte L., Sanvito F., Olivieri S., Viganò F., Doglioni C., Frasson M., Braga M., Bachi A., Dellabona P., Protti M.P., Alessio M. 2008. Serological immunoreactivity against colon cancer proteome varies upon disease progression. J. Proteome Res. 7:504–514 10.1021/pr070360m
    1. Dunn G.P., Old L.J., Schreiber R.D. 2004. The three Es of cancer immunoediting. Annu. Rev. Immunol. 22:329–360 10.1146/annurev.immunol.22.012703.104803
    1. Edwards M.J. 2008. Therapy directed against thymic stromal lymphopoietin. Drug News Perspect. 21:312–316 10.1358/dnp.2008.21.6.1246830
    1. Egberts J.H., Cloosters V., Noack A., Schniewind B., Thon L., Klose S., Kettler B., von Forstner C., Kneitz C., Tepel J., et al. 2008. Anti-tumor necrosis factor therapy inhibits pancreatic tumor growth and metastasis. Cancer Res. 68:1443–1450 10.1158/0008-5472.CAN-07-5704
    1. Erez N., Truitt M., Olson P., Arron S.T., Hanahan D. 2010. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 17:135–147 10.1016/j.ccr.2009.12.041
    1. Evans D.B., Charnsangavei C., Fernandez-del-Castillo C., Fong Y., Lauwers G.Y. 2002. Exocrine pancreas. AJCC Cancer Staging Manual. Greene F.L., Page D.L., Fleming I.D., Fritz A.G., Balch C.M., Haller D.G., Morrow M., Springer, New York: 157–164
    1. Fukunaga A., Miyamoto M., Cho Y., Murakami S., Kawarada Y., Oshikiri T., Kato K., Kurokawa T., Suzuoki M., Nakakubo Y., et al. 2004. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 28:e26–e31 10.1097/00006676-200401000-00023
    1. Glass G., Papin J.A., Mandell J.W. 2009. SIMPLE: a sequential immunoperoxidase labeling and erasing method. J. Histochem. Cytochem. 57:899–905 10.1369/jhc.2009.953612
    1. Grivennikov S.I., Greten F.R., Karin M. 2010. Immunity, inflammation, and cancer. Cell. 140:883–899 10.1016/j.cell.2010.01.025
    1. Harrison M.L., Obermueller E., Maisey N.R., Hoare S., Edmonds K., Li N.F., Chao D., Hall K., Lee C., Timotheadou E., et al. 2007. Tumor necrosis factor alpha as a new target for renal cell carcinoma: two sequential phase II trials of infliximab at standard and high dose. J. Clin. Oncol. 25:4542–4549 10.1200/JCO.2007.11.2136
    1. Hidalgo M. 2010. Pancreatic cancer. N. Engl. J. Med. 362:1605–1617 10.1056/NEJMra0901557
    1. Hingorani S.R., Petricoin E.F., Maitra A., Rajapakse V., King C., Jacobetz M.A., Ross S., Conrads T.P., Veenstra T.D., Hitt B.A., et al. 2003. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 4:437–450 10.1016/S1535-6108(03)00309-X
    1. Ito T., Wang Y.H., Duramad O., Hori T., Delespesse G.J., Watanabe N., Qin F.X., Yao Z., Cao W., Liu Y.J. 2005. TSLP-activated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand. J. Exp. Med. 202:1213–1223 10.1084/jem.20051135
    1. Joyce J.A., Pollard J.W. 2009. Microenvironmental regulation of metastasis. Nat. Rev. Cancer. 9:239–252 10.1038/nrc2618
    1. Kleeff J., Beckhove P., Esposito I., Herzig S., Huber P.E., Löhr J.M., Friess H. 2007. Pancreatic cancer microenvironment. Int. J. Cancer. 121:699–705 10.1002/ijc.22871
    1. Kurahara H., Shinchi H., Mataki Y., Maemura K., Noma H., Kubo F., Sakoda M., Ueno S., Natsugoe S., Takao S. 2009. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. J. Surg. Res. 10.1016/j.jss.2009.05.026
    1. Lachmann H.J., Kone-Paut I., Kuemmerle-Deschner J.B., Leslie K.S., Hachulla E., Quartier P., Gitton X., Widmer A., Patel N., Hawkins P.N.; Canakinumab in CAPS Study Group 2009. Use of canakinumab in the cryopyrin-associated periodic syndrome. N. Engl. J. Med. 360:2416–2425 10.1056/NEJMoa0810787
    1. Leach S.D. 2004. Mouse models of pancreatic cancer: the fur is finally flying! Cancer Cell. 5:7–11 10.1016/S1535-6108(03)00337-4
    1. Lee H.C., Ziegler S.F. 2007. Inducible expression of the proallergic cytokine thymic stromal lymphopoietin in airway epithelial cells is controlled by NFkappaB. Proc. Natl. Acad. Sci. USA. 104:914–919 10.1073/pnas.0607305104
    1. Liu Y.J., Soumelis V., Watanabe N., Ito T., Wang Y.H., Malefyt Rde.W., Omori M., Zhou B., Ziegler S.F. 2007. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu. Rev. Immunol. 25:193–219 10.1146/annurev.immunol.25.022106.141718
    1. Lu N., Wang Y.H., Wang Y.H., Arima K., Hanabuchi S., Liu Y.J. 2009. TSLP and IL-7 use two different mechanisms to regulate human CD4+ T cell homeostasis. J. Exp. Med. 206:2111–2119 10.1084/jem.20090153
    1. Lust J.A., Lacy M.Q., Zeldenrust S.R., Dispenzieri A., Gertz M.A., Witzig T.E., Kumar S., Hayman S.R., Russell S.J., Buadi F.K., et al. 2009. Induction of a chronic disease state in patients with smoldering or indolent multiple myeloma by targeting interleukin 1beta-induced interleukin 6 production and the myeloma proliferative component. Mayo Clin. Proc. 84:114–122 10.4065/84.2.114
    1. Mahadevan D., Von Hoff D.D. 2007. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol. Cancer Ther. 6:1186–1197 10.1158/1535-7163.MCT-06-0686
    1. Mantovani A., Allavena P., Sica A., Balkwill F. 2008. Cancer-related inflammation. Nature. 454:436–444 10.1038/nature07205
    1. Marturano J., Longhi R., Russo V., Protti M.P. 2008. Endosomal proteases influence the repertoire of MAGE-A3 epitopes recognized in vivo by CD4+ T cells. Cancer Res. 68:1555–1562 10.1158/0008-5472.CAN-07-5233
    1. Müerköster S., Wegehenkel K., Arlt A., Witt M., Sipos B., Kruse M.L., Sebens T., Klöppel G., Kalthoff H., Fölsch U.R., Schäfer H. 2004. Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin-1beta. Cancer Res. 64:1331–1337 10.1158/0008-5472.CAN-03-1860
    1. Recalde H.R. 1984. A simple method of obtaining monocytes in suspension. J. Immunol. Methods. 69:71–77 10.1016/0022-1759(84)90278-3
    1. Reche P.A., Soumelis V., Gorman D.M., Clifford T., Mr Liu, Travis M., Zurawski S.M., Johnston J., Liu Y.J., Spits H., et al. 2001. Human thymic stromal lymphopoietin preferentially stimulates myeloid cells. J. Immunol. 167:336–343
    1. Reni M., Passoni P., Bonetto E., Balzano G., Panucci M.G., Zerbi A., Ronzoni M., Staudacher C., Villa E., Di Carlo V. 2005. Final results of a prospective trial of a PEFG (Cisplatin, Epirubicin, 5-Fluorouracil, Gemcitabine) regimen followed by radiotherapy after curative surgery for pancreatic adenocarcinoma. Oncology. 68:239–245 10.1159/000086780
    1. Rimoldi M., Chieppa M., Salucci V., Avogadri F., Sonzogni A., Sampietro G.M., Nespoli A., Viale G., Allavena P., Rescigno M. 2005. Intestinal immune homeostasis is regulated by the crosstalk between epithelial cells and dendritic cells. Nat. Immunol. 6:507–514 10.1038/ni1192
    1. Slager E.H., Borghi M., van der Minne C.E., Aarnoudse C.A., Havenga M.J., Schrier P.I., Osanto S., Griffioen M. 2003. CD4+ Th2 cell recognition of HLA-DR-restricted epitopes derived from CAMEL: a tumor antigen translated in an alternative open reading frame. J. Immunol. 170:1490–1497
    1. Soumelis V., Reche P.A., Kanzler H., Yuan W., Edward G., Homey B., Gilliet M., Ho S., Antonenko S., Lauerma A., et al. 2002. Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nat. Immunol. 3:673–680 10.1038/ni805
    1. Tassi E., Gavazzi F., Albarello L., Senyukov V., Longhi R., Dellabona P., Doglioni C., Braga M., Di Carlo V., Protti M.P. 2008. Carcinoembryonic antigen-specific but not antiviral CD4+ T cell immunity is impaired in pancreatic carcinoma patients. J. Immunol. 181:6595–6603
    1. Tatsumi T., Kierstead L.S., Ranieri E., Gesualdo L., Schena F.P., Finke J.H., Bukowski R.M., Mueller-Berghaus J., Kirkwood J.M., Kwok W.W., Storkus W.J. 2002. Disease-associated bias in T helper type 1 (Th1)/Th2 CD4+ T cell responses against MAGE-6 in HLA-DRB10401+ patients with renal cell carcinoma or melanoma. J. Exp. Med. 196:619–628 10.1084/jem.20012142
    1. Tatsumi T., Herrem C.J., Olson W.C., Finke J.H., Bukowski R.M., Kinch M.S., Ranieri E., Storkus W.J. 2003. Disease stage variation in CD4+ and CD8+ T-cell reactivity to the receptor tyrosine kinase EphA2 in patients with renal cell carcinoma. Cancer Res. 63:4481–4489
    1. Tschopp J., Schroder K. 2010. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 10:210–215 10.1038/nri2725
    1. van den Berg W.B. 2000. Arguments for interleukin 1 as a target in chronic arthritis. Ann. Rheum. Dis. 59:i81–i84 10.1136/ard.59.suppl_1.i81
    1. Wynn T.A. 2004. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat. Rev. Immunol. 4:583–594 10.1038/nri1412
    1. Zhang K., Shan L., Rahman M.S., Unruh H., Halayko A.J., Gounni A.S. 2007. Constitutive and inducible thymic stromal lymphopoietin expression in human airway smooth muscle cells: role in chronic obstructive pulmonary disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 293:L375–L382 10.1152/ajplung.00045.2007

Source: PubMed

Подписаться