Intraoperative radiation therapy induces immune response activity after pancreatic surgery

Yun Sun Lee, Hyung Sun Kim, Yeona Cho, Ik Jae Lee, Hyo Jung Kim, Da Eun Lee, Hyeon Woong Kang, Joon Seong Park, Yun Sun Lee, Hyung Sun Kim, Yeona Cho, Ik Jae Lee, Hyo Jung Kim, Da Eun Lee, Hyeon Woong Kang, Joon Seong Park

Abstract

Background: Pancreatic cancer has highly aggressive features, such as local recurrence that leads to significantly high morbidity and mortality and recurrence after successful tumour resection. Intraoperative radiation therapy (IORT), which delivers targeted radiation to a tumour bed, is known to reduce local recurrence by directly killing tumour cells and modifying the tumour microenvironment.

Methods: Among 30 patients diagnosed with pancreatic cancer, 17 patients received IORT immediately after surgical resection. We investigated changes in the immune response induced by IORT by analysing the peritoneal fluid (PF) and blood of patients with and without IORT treatment after pancreatic cancer surgery. Further, we treated three pancreatic cell lines with PF to observe proliferation and activity changes.

Results: Levels of cytokines involved in the PI3K/SMAD pathway were increased in the PF of IORT-treated patients. Moreover, IORT-treated PF inhibited the growth, migration, and invasiveness of pancreatic cancer cells. Changes in lymphocyte populations in the blood of IORT-treated patients indicated an increased immune response.

Conclusions: Based on the characterisation and quantification of immune cells in the blood and cytokine levels in the PF, we conclude that IORT induced an anti-tumour effect by activating the immune response, which may prevent pancreatic cancer recurrence.

Clinical trial registration: NCT03273374 .

Keywords: Cytokine; Immune response; Intraoperative radiation therapy; Pancreatic cancer.

Conflict of interest statement

The authors declare no conflict of interest.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Intraoperative radiation therapy (IORT) leads to differential cytokine profile of the peritoneal fluid (PF) and affects PI3K/Smad signalling in pancreatic cancer cells. A Cytokine membrane array images. Each spot represents a cytokine hybridised in patient PF. B Heat map visualisation comparing cytokine array results. C. Cytokines significantly upregulated in the PF of IORT-treated patients (IORT n = 15, no IORT n = 11). D. Schematic overview of signalling pathways (PI3K-Akt, Smad2/3) in cancer cells associated with cytokines TGF-β, IFN-γ, and PDGF-BB. E. Western blot analysis of signalling-related protein xpression in Panc1 cells incubated with PF for 4 h. IORT: P3, P54, and P56; no IORT: P31, P33, and P42 (unpaired t-test; *p < 0.05 and **p ≤ 0.01)
Fig. 2
Fig. 2
Peritoneal fluid (PF) stimulates cancer cell proliferation. A. Mia PaCa-2, Panc1, and Aspc1 cell lines were incubated for 4 days in the presence of PF at the indicated concentrations. As a positive control, foetal bovine serum (FBS) was used at the same concentration as PF. Incubated cells were treated with WST-1 solution, and absorbance was detected at 450 nm. B. Cells were incubated in the presence of PF until the colonies were formed. Colonies were stained with crystal violet, and the percentage of colony areas were calculated using ImageJ software. Columns correspond to the mean of three independent experiments; bars indicate SD. Statistical analyses were performed using the unpaired t-test
Fig. 3
Fig. 3
Intraoperative radiation therapy (IORT)-treated peritoneal fluid (PF) decreased the invasiveness and wound healing ability of pancreatic cancer cells. A. Pancreatic cancer cell lines were tested for their invasiveness using Matrigel-coated Transwell chemotaxis assays. Random PF samples (7% in DMEM) were loaded in the bottom well of the transwell plate for pancreatic cancer cell treatment. Invaded cells were stained and analysed after 24 h. B. Pancreatic cancer cells cultured in 96-well plates were scratched using a wound maker and treated with PF for observing cell migration every 12 h
Fig. 4
Fig. 4
Changes in EMT marker expression in cancer cells following treatment with peritoneal fluid (PF) collected after intraoperative radiation therapy (IORT). A. qPCR was used to measure the expression levels of EMT-related genes encoding mesenchymal markers (vimentin, snail, and N-cadherin) and epithelial markers (E-cadherin). B. Western blotting analysis of the expression of indicated EMT markers. RNA and protein were extracted from cells after 4 days of treatment. Used PF were from IORT (P1, P43, P56, and P64) and no IORT (P26, P42, P45, and P62). The results were statistically analysed using the unpaired t-test, and significance was marked at *: p < 0.05 and **: p < 0.01 using three independent biological replicates
Fig. 5
Fig. 5
Changes in immune cell composition of PBMCs during the follow-up period of IORT-treated patients with pancreatic cancer. A. Representative flow cytometry plots illustrating the gating strategy used in the analysis of lymphocytes in the blood (left: CD3-CD56+ NK cell; middle: CD3 + CD4+ helper T cell and CD3 + CD8+ cytotoxic T cell; right: CD3-CD25 + FOXP3+ Treg cell). B. Rate of change in the indicated immune cells according to the blood collection time (baseline = post-operation day (POD) 1, normalised to each POD 1 value). C. Scheme of immune response by IORT treatment after pancreatic cancer surgery

References

    1. Kane S, Engelhart A, Guadagno J, Jones A, Usoro I, Brutcher E. Pancreatic ductal adenocarcinoma: characteristics of tumor microenvironment and barriers to treatment. J Adv Pract Oncol. 2020;11(7):693–698. doi: 10.6004/jadpro.2020.11.7.4.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34. doi: 10.3322/caac.21551.
    1. Paunesku T, Woloschak GE. Future directions of intraoperative radiation therapy: a brief review. Front Oncol. 2017;7:300. doi: 10.3389/fonc.2017.00300.
    1. Krempien R, Roeder F. Intraoperative radiation therapy (IORT) in pancreatic cancer. Radiat Oncol. 2017;12(1):8. doi: 10.1186/s13014-016-0753-0.
    1. Boldrini L, Cusumano D, Cellini F, Azario L, Mattiucci GC, Valentini V. Online adaptive magnetic resonance guided radiotherapy for pancreatic cancer: state of the art, pearls and pitfalls. Radiat Oncol. 2019;14(1):71. doi: 10.1186/s13014-019-1275-3.
    1. Heerkens HD, van Vulpen M, Erickson B, Reerink O, Intven MP, van den Berg CA, Molenaar IQ, Vleggaar FP, Meijer GJ. MRI guided stereotactic radiotherapy for locally advanced pancreatic cancer. Br J Radiol. 2018;91(1091):20170563. doi: 10.1259/bjr.20170563.
    1. Reese AS, Lu W, Regine WF. Utilization of intensity-modulated radiation therapy and image-guided radiation therapy in pancreatic cancer: is it beneficial? Semin Radiat Oncol. 2014;24(2):132–139. doi: 10.1016/j.semradonc.2013.11.003.
    1. Kinsella TJ, Sindelar WF. Intraoperative radiotherapy for pancreatic carcinoma. Experimental and clinical studies. Cancer. 1996;78(3 Suppl):598–604. doi: 10.1002/(SICI)1097-0142(19960801)78:3+<598::AID-CNCR4>;2-7.
    1. Crane CH, Beddar AS, Evans DB. The role of intraoperative radiotherapy in pancreatic cancer. Surg Oncol Clin N Am. 2003;12(4):965–977. doi: 10.1016/S1055-3207(03)00090-5.
    1. Pilar A, Gupta M, Ghosh Laskar S, Laskar S. Intraoperative radiotherapy: review of techniques and results. Ecancermedicalscience. 2017;11:750. doi: 10.3332/ecancer.2017.750.
    1. Demaria S, Formenti SC. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol. 2012;2:153. doi: 10.3389/fonc.2012.00153.
    1. Kroemer G, Galluzzi L, Zitvogel L. Immunological effects of chemotherapy in spontaneous breast cancers. Oncoimmunology. 2013;2(12):e27158. doi: 10.4161/onci.27158.
    1. Hellweg CE. The nuclear factor kappaB pathway: a link to the immune system in the radiation response. Cancer Lett. 2015;368(2):275–289. doi: 10.1016/j.canlet.2015.02.019.
    1. Shan YX, Jin SZ, Liu XD, Liu Y, Liu SZ. Ionizing radiation stimulates secretion of pro-inflammatory cytokines: dose-response relationship, mechanisms and implications. Radiat Environ Biophys. 2007;46(1):21–29. doi: 10.1007/s00411-006-0076-x.
    1. Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravata V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015;12(1):14. doi: 10.1186/s12950-015-0058-3.
    1. Kinoshita T, Uesaka K, Shimizu Y, Sakamoto H, Kimura W, Sunada S, Sunada S, Imaizumi T, Ozawa I, Okamoto A, et al. Effects of adjuvant intra-operative radiation therapy after curative resection in pancreatic cancer patients : Results of a randomized study by 11 institutions in Japan. J Clin Oncol. 2009;27(15_suppl):4622. doi: 10.1200/jco.2009.27.15_suppl.4622.
    1. Jin L, Shi N, Ruan S, Hou B, Zou Y, Zou X, Jin H, Jian Z. The role of intraoperative radiation therapy in resectable pancreatic cancer: a systematic review and meta-analysis. Radiat Oncol. 2020;15(1):76. doi: 10.1186/s13014-020-01511-9.
    1. Chen Y, Che X, Zhang J, Huang H, Zhao D, Tian Y, Li Y, Feng Q, Zhang Z, Jiang Q, Zhang S, Tang X, Huang X, Chu Y, Zhang J, Sun Y, Zhang Y, Wang C. Long-term results of intraoperative electron beam radiation therapy for nonmetastatic locally advanced pancreatic cancer: retrospective cohort study, 7-year experience with 247 patients at the National Cancer Center in China. Medicine (Baltimore) 2016;95(38):e4861. doi: 10.1097/MD.0000000000004861.
    1. Kulcenty K, Piotrowski I, Wroblewska JP, Wasiewicz J, AWM S. The Composition of Surgical Wound Fluids from Breast Cancer Patients is Affected by Intraoperative Radiotherapy Treatment and Depends on the Molecular Subtype of Breast Cancer. Cancers (Basel) 2019;12(1):11. doi: 10.3390/cancers12010011.
    1. Kim JW, Cho Y, Kim HS, Choi WH, Park JS, Lee IJ. A phase II study of intraoperative radiotherapy using a low-energy x-ray source for resectable pancreatic cancer: a study protocol. BMC Surg. 2019;19(1):31. doi: 10.1186/s12893-019-0492-x.
    1. Alfieri S, Morganti AG, Di Giorgio A, Valentini V, Bossola M, Trodella L, Cellini N, Doglietto GB. Improved survival and local control after intraoperative radiation therapy and postoperative radiotherapy: a multivariate analysis of 46 patients undergoing surgery for pancreatic head cancer. Arch Surg. 2001;136(3):343–347. doi: 10.1001/archsurg.136.3.343.
    1. Valeta-Magara A, Hatami R, Axelrod D, Roses DF, Guth A, Formenti SC, Schneider RJ. Pro-oncogenic cytokines and growth factors are differentially expressed in the post-surgical wound fluid from malignant compared to benign breast lesions. Springerplus. 2015;4(1):483. doi: 10.1186/s40064-015-1260-8.
    1. Walle T, Martinez Monge R, Cerwenka A, Ajona D, Melero I, Lecanda F. Radiation effects on antitumor immune responses: current perspectives and challenges. Ther Adv Med Oncol. 2018;10:1758834017742575. doi: 10.1177/1758834017742575.
    1. Heldin CH. Simultaneous induction of stimulatory and inhibitory signals by PDGF. FEBS Lett. 1997;410(1):17–21. doi: 10.1016/S0014-5793(97)00318-9.
    1. Fan H, Ma L, Fan B, Wu J, Yang Z, Wang L. Role of PDGFR-beta/PI3K/AKT signaling pathway in PDGF-BB induced myocardial fibrosis in rats. Am J Transl Res. 2014;6(6):714–723.
    1. Rodriguez-Ruiz ME, Rodriguez I, Mayorga L, Labiano T, Barbes B, Etxeberria I, Ponz-Sarvise M, Azpilikueta A, Bolanos E, Sanmamed MF, et al. TGFbeta blockade enhances radiotherapy Abscopal efficacy effects in combination with anti-PD1 and anti-CD137 Immunostimulatory monoclonal antibodies. Mol Cancer Ther. 2019;18(3):621–631. doi: 10.1158/1535-7163.MCT-18-0558.
    1. Dancea HC, Shareef MM, Ahmed MM. Role of radiation-induced TGF-beta signaling in Cancer therapy. Mol Cell Pharmacol. 2009;1(1):44–56. doi: 10.4255/mcpharmacol.09.06.
    1. Gerber SA, Sedlacek AL, Cron KR, Murphy SP, Frelinger JG, Lord EM. IFN-gamma mediates the antitumor effects of radiation therapy in a murine colon tumor. Am J Pathol. 2013;182(6):2345–2354. doi: 10.1016/j.ajpath.2013.02.041.
    1. Ivashkiv LB. IFNgamma: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat Rev Immunol. 2018;18(9):545–558. doi: 10.1038/s41577-018-0029-z.
    1. Rautela J, Huntington ND. IL-15 signaling in NK cell cancer immunotherapy. Curr Opin Immunol. 2017;44:1–6. doi: 10.1016/j.coi.2016.10.004.
    1. Ali AK, Nandagopal N, Lee SH. IL-15-PI3K-AKT-mTOR: a critical pathway in the life journey of natural killer cells. Front Immunol. 2015;6:355. doi: 10.3389/fimmu.2015.00355.
    1. Belletti B, Vaidya JS, D'Andrea S, Entschladen F, Roncadin M, Lovat F, Berton S, Perin T, Candiani E, Reccanello S, Veronesi A, Canzonieri V, Trovò MG, Zaenker KS, Colombatti A, Baldassarre G, Massarut S. Targeted intraoperative radiotherapy impairs the stimulation of breast cancer cell proliferation and invasion caused by surgical wounding. Clin Cancer Res. 2008;14(5):1325–1332. doi: 10.1158/1078-0432.CCR-07-4453.
    1. Cros J, Raffenne J, Couvelard A, Pote N. Tumor heterogeneity in pancreatic adenocarcinoma. Pathobiology. 2018;85(1–2):64–71. doi: 10.1159/000477773.
    1. Zhao M, Mishra L, Deng CX. The role of TGF-beta/SMAD4 signaling in cancer. Int J Biol Sci. 2018;14(2):111–123. doi: 10.7150/ijbs.23230.
    1. Singh AP, Arora S, Bhardwaj A, Srivastava SK, Kadakia MP, Wang B, Grizzle WE, Owen LB, Singh S. CXCL12/CXCR4 protein signaling axis induces sonic hedgehog expression in pancreatic cancer cells via extracellular regulated kinase- and Akt kinase-mediated activation of nuclear factor kappaB: implications for bidirectional tumor-stromal interactions. J Biol Chem. 2012;287(46):39115–39124. doi: 10.1074/jbc.M112.409581.
    1. Kulcenty K, Piotrowski I, Zaleska K, Wichtowski M, Wroblewska J, Murawa D, Suchorska WM. Wound fluids collected postoperatively from patients with breast cancer induce epithelial to mesenchymal transition but intraoperative radiotherapy impairs this effect by activating the radiation-induced bystander effect. Sci Rep. 2019;9(1):7891. doi: 10.1038/s41598-019-44412-y.
    1. Brown JM, Koong AC. High-dose single-fraction radiotherapy: exploiting a new biology? Int J Radiat Oncol Biol Phys. 2008;71(2):324–325. doi: 10.1016/j.ijrobp.2008.02.003.
    1. Herskind C, Wenz F, Giordano FA. Immunotherapy combined with large fractions of radiotherapy: stereotactic radiosurgery for brain metastases-implications for intraoperative radiotherapy after resection. Front Oncol. 2017;7:147. doi: 10.3389/fonc.2017.00147.
    1. Cecka F, Lovecek M, Jon B, Skalicky P, Subrt Z, Neoral C, Ferko A. Intra-abdominal drainage following pancreatic resection: a systematic review. World J Gastroenterol. 2015;21(40):11458–11468. doi: 10.3748/wjg.v21.i40.11458.
    1. Davidson TB, Yaghoobi M, Davidson BR, Gurusamy KS. Amylase in drain fluid for the diagnosis of pancreatic leak in post-pancreatic resection. Cochrane Database Syst Rev. 2017;4(4):CD012009. doi: 10.1002/14651858.CD012009.pub2.
    1. Hicks AM, Chou J, Capanu M, Lowery MA, Yu KH, O'Reilly EM. Pancreas adenocarcinoma: ascites, clinical manifestations, and management implications. Clin Colorectal Cancer. 2016;15(4):360–368. doi: 10.1016/j.clcc.2016.04.014.
    1. Kassis J, Klominek J, Kohn EC. Tumor microenvironment: what can effusions teach us? Diagn Cytopathol. 2005;33(5):316–319. doi: 10.1002/dc.20280.
    1. Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S, Muller R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front Oncol. 2017;7:24. doi: 10.3389/fonc.2017.00024.
    1. Wright EC, Connolly P, Vella M, Moug S. Peritoneal fluid biomarkers in the detection of colorectal anastomotic leaks: a systematic review. Int J Color Dis. 2017;32(7):935–945. doi: 10.1007/s00384-017-2799-3.

Source: PubMed

3
Abonneren