Programmed cell death ligand 1 (PD-L1, CD274) in cholangiocarcinoma - correlation with clinicopathological data and comparison of antibodies

Mark Kriegsmann, Stephanie Roessler, Katharina Kriegsmann, Marcus Renner, Rémi Longuespée, Thomas Albrecht, Moritz Loeffler, Stephan Singer, Arianeb Mehrabi, Monika Nadja Vogel, Anita Pathil, Bruno Köhler, Christoph Springfeld, Christian Rupp, Karl Heinz Weiss, Benjamin Goeppert, Mark Kriegsmann, Stephanie Roessler, Katharina Kriegsmann, Marcus Renner, Rémi Longuespée, Thomas Albrecht, Moritz Loeffler, Stephan Singer, Arianeb Mehrabi, Monika Nadja Vogel, Anita Pathil, Bruno Köhler, Christoph Springfeld, Christian Rupp, Karl Heinz Weiss, Benjamin Goeppert

Abstract

Background: Cholangiocarcinoma (CCA) may arise in the intra- or extrahepatic biliary tract and is associated with a poor prognosis. Despite recent advances, to date there is still no established targeted therapeutic approach available. Non-surgical therapeutic agents are urgently needed, as most patients are non-eligible to surgical resection. Anti-PD-L1 therapy prevents cancer cells from evading the immune system and has emerged as a new treatment option in several cancer entities. Recently, PD-L1 expression has been analyzed in comparably small CCA patient cohorts. However, a systematic validation of different PD-L1 antibodies has not been performed in CCA so far.

Methods: We stained a tissue microarray consisting of 170 patients, including 72 intrahepatic cholangiocarcinomas (iCCAs), 57 perihilar cholangiocarcinomas (pCCAs) and 41 distal cholangiocarcinomas (dCCAs) by immunohistochemistry and evaluated PD-L1 positivity in tumor and stromal cells. We analyzed three different PD-L1 antibodies (clones 28-8, SP142, and SP263) that are frequently used and recommended for predictive diagnostic testing in other cancer types.

Results: For PD-L1 antibody clone SP263, 5% of iCCAs, 4% of pCCAs and 3% of dCCAs exhibited PD-L1 expression on tumor cells, thereby showing the highest frequencies of PD-L1 positivity. Accordingly, highest PD-L1 positivity rates of stromal cells with 31% in iCCA, 40% in pCCA and 61% in dCCA were detected for clone SP263. Agreement of PD-L1 positivity in tumor cells was moderate for clone 28-8 and SP263 (κ = 0.44) and poor between 28-8 and SP142 (κ = 0.13), as well as SP142 and SP263 (κ = 0.11), respectively. Statistical analyses of PD-L1 expression (clone SP263) on tumor cells with clinicopathological data revealed a positive correlation with shortened overall survival in CCA patients.

Conclusions: Selection of appropriate PD-L1 antibodies and careful evaluation of immunohistochemical staining patterns have a significant impact on PD-L1 testing in CCA. Clinical trials are necessary to investigate the putative beneficial effects of PD-L1 targeted immunotherapy in CCA patients.

Keywords: 28–8; CD274; Cholangiocarcinoma; PD-L1; SP142; SP263.

Conflict of interest statement

Ethics approval and consent to participate

Tissues were used in accordance with the ethical regulations of the NCT tissue bank established by the local ethics committee (S-207/2015). Ethics approval, written and verbal consent to participate has been given by all participants.

Consent for publication

Consent for publication has been obtained (NCT; project # 2116 and S-207/2015).

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Examples of PD-L1 staining in a representative intrahepatic cholangiocarcinoma. The different staining characteristics of PD-L1 clones 28–8, SP142 and SP263 are displayed. In the first row, tumor cells show membranous positivity of few tumor cells with antibody clones 28–8 (a) and SP263 (c), but not with clone SP142 (b). In the second row, stromal inflammatory cells show membranous PD-L1 expression with all three antibody clones (d-f), while tumor cells are negative. In the third row, all samples are negative, both in tumor and stromal cells, and with all three PD-L1 antibodies (g-i). Original magnification: 200x, PD-L1 positive tumor cells are highlighted by black arrows, PD-L1 positive stromal cells are highlighted by black triangles
Fig. 2
Fig. 2
PD-L1 status in correlation with overall survival in cholangiocarcinoma patients. PD-L1 immunohistochemistry results using clone SP263 were correlated with overall survival of CCA patients. Kaplan-Meier curves show a trend of decreased overall survival in CCA patients with any PD-L1 positivity in tumor cells (p = 0.09;a). Significant decreased overall survival rates are seen in CCA patients with PD-L1 positivity in > 5% of tumor cells (p < 0.001; b). PD-L1 positivity in stromal cells has no impact on overall survival of CCA patients in none vs any (p = 0.89;c) or testing the cut-off of 5% (p = 0.69;d)
Fig. 3
Fig. 3
PD-L1 status in correlation with patient overall survival in cholangiocarcinoma subtypes. PD-L1 immunohistochemistry (clone SP263) results are correlated with overall survival of CCA patients, stratified by CCA subtypes. In intrahepatic CCA (iCCA), Kaplan-Meier curves show no significant overall patient survival difference in correlation with none vs any PD-L1 positivity in tumor cells (a), while significant decreased overall survival rates are seen in iCCA patients with PD-L1 positivity in > 5% of tumor cells (p = 0.024); (b)). In perihilar CCA (pCCA), Kaplan-Meier curves show no significant overall patient survival difference in correlation with none vs any PD-L1 positivity in tumor cells (c), while by trend decreased overall survival rates are in pCCA patients with PD-L1 positivity in > 5% of tumor cells (p = 0.060; (d)). PD-L1 positivity in distal CCA (dCCA) has no impact on overall patient survival in none vs any (e) or testing the cut-off of 5% (f)

References

    1. Ma W, Gilligan BM, Yuan J, Li T. Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy. J Hematol Oncol. 2016;9(1):47. doi: 10.1186/s13045-016-0277-y.
    1. Daud AI, Wolchok JD, Robert C, Hwu WJ, Weber JS, Ribas A, Hodi FS, Joshua AM, Kefford R, Hersey P, et al. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody Pembrolizumab in melanoma. J Clin Oncol. 2016;34(34):4102–4109. doi: 10.1200/JCO.2016.67.2477.
    1. Franklin C, Livingstone E, Roesch A, Schilling B, Schadendorf D. Immunotherapy in melanoma: recent advances and future directions. Eur J Surg Oncol. 2017;43(3):604–611. doi: 10.1016/j.ejso.2016.07.145.
    1. Yang Y, Pang Z, Ding N, Dong W, Ma W, Li Y, Du J, Liu Q. The efficacy and potential predictive factors of PD-1/PD-L1 blockades in epithelial carcinoma patients: a systematic review and meta analysis. Oncotarget. 2016;7(45):74350–74361.
    1. Shukuya T, Mori K, Amann JM, Bertino EM, Otterson GA, Shields PG, Morita S, Carbone DP. Relationship between overall survival and response or progression-free survival in advanced non-small cell lung Cancer patients treated with anti-PD-1/PD-L1 antibodies. J Thorac Oncol. 2016;11(11):1927–1939. doi: 10.1016/j.jtho.2016.07.017.
    1. Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung Cancer. N Engl J Med. 2016;375(19):1823–1833. doi: 10.1056/NEJMoa1606774.
    1. Beckermann KE, Johnson DB, Sosman JA. PD-1/PD-L1 blockade in renal cell cancer. Expert Rev Clin Immunol. 2017;13(1):77–84. doi: 10.1080/1744666X.2016.1214575.
    1. Donin NM, Lenis AT, Holden S, Drakaki A, Pantuck A, Belldegrun A, Chamie K. Immunotherapy for the treatment of urothelial carcinoma. J Urol. 2017;197(1):14–22. doi: 10.1016/j.juro.2016.02.3005.
    1. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016;100:88–98. doi: 10.1016/j.critrevonc.2016.02.001.
    1. Scheel AH, Dietel M, Heukamp LC, Johrens K, Kirchner T, Reu S, Ruschoff J, Schildhaus HU, Schirmacher P, Tiemann M, et al. Predictive PD-L1 immunohistochemistry for non-small cell lung cancer : current state of the art and experiences of the first German harmonization study. Pathologe. 2016;37(6):557–567. doi: 10.1007/s00292-016-0189-1.
    1. Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, et al. Expert consensus document: cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European network for the study of cholangiocarcinoma (ENS-CCA) Nat Rev Gastroenterol Hepatol. 2016;13(5):261–280. doi: 10.1038/nrgastro.2016.51.
    1. Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, Madhusudan S, Iveson T, Hughes S, Pereira SP, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362(14):1273–1281. doi: 10.1056/NEJMoa0908721.
    1. Razumilava N, Gores GJ. Building a staircase to precision medicine for biliary tract cancer. Nat Genet. 2015;47(9):967–968. doi: 10.1038/ng.3386.
    1. Rizvi S, Gores GJ. Emerging molecular therapeutic targets for cholangiocarcinoma. J Hepatol. 2017;67(3):632–644. doi: 10.1016/j.jhep.2017.03.026.
    1. Czink E, Kloor M, Goeppert B, Frohling S, Uhrig S, Weber TF, Meinel J, Sutter C, Weiss KH, Schirmacher P, et al. Successful immune checkpoint blockade in a patient with advanced stage microsatellite-unstable biliary tract cancer. Cold Spring Harb Mol Case Stud. 2017;3(5).
    1. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res. 2013;19(5):1021–1034. doi: 10.1158/1078-0432.CCR-12-2063.
    1. Pyo JS, Kang G, Kim JY. Prognostic role of PD-L1 in malignant solid tumors: a meta-analysis. Int J Biol Markers. 2017;32(1):e68–e74. doi: 10.5301/jbm.5000225.
    1. Hofman P, Ilie M, Lassalle S, Long E, Bence C, Butori C, Hofman V. PD1/PD-L1 immunohistochemistry in thoracic oncology: where are we? Ann Pathol. 2017;37(1):39–45. doi: 10.1016/j.annpat.2016.12.006.
    1. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. doi: 10.1056/NEJMoa1504030.
    1. Fontugne J, Augustin J, Pujals A, Compagnon P, Rousseau B, Luciani A, Tournigand C, Cherqui D, Azoulay D, Pawlotsky JM, et al. PD-L1 expression in perihilar and intrahepatic cholangiocarcinoma. Oncotarget. 2017;8(15):24644–24651. doi: 10.18632/oncotarget.15602.
    1. Gani F, Nagarajan N, Kim Y, Zhu Q, Luan L, Bhaijjee F, Anders RA, Pawlik TM. Program death 1 immune checkpoint and tumor microenvironment: implications for patients with intrahepatic cholangiocarcinoma. Ann Surg Oncol. 2016;23(8):2610–2617. doi: 10.1245/s10434-016-5101-y.
    1. Hirsch FR, McElhinny A, Stanforth D, Ranger-Moore J, Jansson M, Kulangara K, Richardson W, Towne P, Hanks D, Vennapusa B, et al. PD-L1 immunohistochemistry assays for lung Cancer: results from phase 1 of the blueprint PD-L1 IHC assay comparison project. J Thorac Oncol. 2017;12(2):208–222. doi: 10.1016/j.jtho.2016.11.2228.
    1. Sabbatino F, Villani V, Yearley JH, Deshpande V, Cai L, Konstantinidis IT, Moon C, Nota S, Wang Y, Al-Sukaini A, et al. PD-L1 and HLA class I antigen expression and clinical course of the disease in intrahepatic cholangiocarcinoma. Clin Cancer Res. 2016;22(2):470–478. doi: 10.1158/1078-0432.CCR-15-0715.
    1. Walter D, Herrmann E, Schnitzbauer AA, Zeuzem S, Hansmann ML, Peveling-Oberhag J, Hartmann S. PD-L1 expression in extrahepatic cholangiocarcinoma. Histopathology. 2017;71(3):383–392. doi: 10.1111/his.13238.
    1. Ma K, Wei X, Dong D, Wu Y, Geng Q, Li E. PD-L1 and PD-1 expression correlate with prognosis in extrahepatic cholangiocarcinoma. Oncol Lett. 2017;14(1):250–256. doi: 10.3892/ol.2017.6105.
    1. Sato Y, Kinoshita M, Takemura S, Tanaka S, Hamano G, Nakamori S, Fujikawa M, Sugawara Y, Yamamoto T, Arimoto A, et al. The PD-1/PD-L1 axis may be aberrantly activated in occupational cholangiocarcinoma. Pathol Int. 2017;67(3):163–170. doi: 10.1111/pin.12511.
    1. Sangkhamanon S, Jongpairat P, Sookprasert A, Wirasorn K, Titapun A, Pugkhem A, Ungareevittaya P, Chindaprasirt J. Programmed death-ligand 1 (PD-L1) expression associated with a high neutrophil/lymphocyte ratio in cholangiocarcinoma. Asian Pac J Cancer Prev. 2017;18(6):1671–1674.
    1. Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–1550. doi: 10.1016/S0140-6736(15)01281-7.

Source: PubMed

3
구독하다