Distinct PD-L1/PD1 Profiles and Clinical Implications in Intrahepatic Cholangiocarcinoma Patients with Different Risk Factors

Jia-Cheng Lu, Hai-Ying Zeng, Qi-Man Sun, Qing-Nan Meng, Xiao-Yong Huang, Peng-Fei Zhang, Xuan Yang, Rui Peng, Chao Gao, Chuan-Yuan Wei, Ying-Hao Shen, Jia-Bing Cai, Rui-Zhao Dong, Ying-Hong Shi, Hui-Chuan Sun, Yujiang G Shi, Jian Zhou, Jia Fan, Ai-Wu Ke, Liu-Xiao Yang, Guo-Ming Shi, Jia-Cheng Lu, Hai-Ying Zeng, Qi-Man Sun, Qing-Nan Meng, Xiao-Yong Huang, Peng-Fei Zhang, Xuan Yang, Rui Peng, Chao Gao, Chuan-Yuan Wei, Ying-Hao Shen, Jia-Bing Cai, Rui-Zhao Dong, Ying-Hong Shi, Hui-Chuan Sun, Yujiang G Shi, Jian Zhou, Jia Fan, Ai-Wu Ke, Liu-Xiao Yang, Guo-Ming Shi

Abstract

Rationale: PD1/PD-L1 immune checkpoint inhibitors have shown promising results for several malignancies. However, PD1/PD-L1 signaling and its therapeutic significance remains largely unknown in intrahepatic cholangiocarcinoma (ICC) cases with complex etiology. Methods: We investigated the expression and clinical significance of CD3 and PD1/PD-L1 in 320 ICC patients with different risk factors. In addition, we retrospectively analyzed 7 advanced ICC patients who were treated with PD1 inhibitor. Results: The cohort comprised 233 patients with HBV infection, 18 patients with hepatolithiasis, and 76 patients with undetermined risk factors. PD-L1 was mainly expressed in tumor cells, while CD3 and PD1 were expressed in infiltrating lymphocytes of tumor tissues. PD1/PD-L1 signals were activated in tumor tissues, and expression was positively correlated with HBV infection and lymph node invasion. More PD1+ T cells and higher PD-L1 expression were observed in tumor tissues of ICC patients with HBV infection compared to patients with hepatolithiasis or undetermined risk factors. More PD1+ T cells and/or high PD-L1 expression negatively impacted the prognosis of patients with HBV infection but not those with hepatolithiasis. Multivariate analysis showed PD1/PD-L1 expression was an independent indicator of ICC patient prognosis. Advanced ICC patients with HBV infection and less PD1+ T cells tended to have good response to anti-PD1 therapy. Conclusion: Hyperactivated PD1/PD-L1 signals in tumor tissues are a negative prognostic marker for ICCs after resection. HBV infection- and hepatolithiasis-related ICCs have distinct PD1/PD-L1 profiles. Further, PD1+ T cells could be used as a biomarker to predict prognosis and assay the efficiency of anti-PD1 immunotherapy in ICC patients with HBV infection.

Keywords: hepatitis B virus; hepatolithiasis; immune checkpoint blockage; intrahepatic cholangiocarcinoma; programmed cell death protein 1; programmed cell death protein ligand 1.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

Figures

Figure 1
Figure 1
PD1/PD-L1 expression in tumor tissues of 320 ICCs. (A) and (B) Two representative ICC tissues showing different levels of PD-L1 expression (200×). (C) Semi-quantitative analysis of PD-L1 staining in tumor tissues (T) and peritumor tissues (PT) (**p < 0.01, Wilcoxon signed-rank test). (D) and (E) Two representative ICC tissues with different numbers of PD1- and CD3-positive cells (200×). (F) Statistical analysis of PD1+ T cells in tumor tissues (T) and peritumor tissues (PT) (**p < 0.01, paired Student's t-test).
Figure 2
Figure 2
Prognostic implication of PD1/PD-L1 axis in ICC patients. (A) Kaplan-Meier estimate of overall survival in the whole cohort with different PD-L1 levels (log-rank test). (B) Kaplan-Meier estimate of cumulative recurrence in the whole cohort with different PD-L1 levels (log-rank test). (C) Kaplan-Meier estimate of overall survival in the whole cohort with different PD1 levels (log-rank test). (D) Kaplan-Meier estimate of cumulative recurrence in the whole cohort with different PD1 levels (log-rank test). (E) Kaplan-Meier estimate of overall survival in the whole cohort with combined PD1/PD-L1 levels (double-positive refers to PD1high/PD-L1high; single positive refers to PD1high/PD-L1low or PD1low/PD-L1high; double negative refers to PD1low/PD-L1low) (log-rank test). (F) Kaplan-Meier estimate of cumulative recurrence in the whole cohort with combined PD1/PD-L1 levels (double positive refers to PD1high/PD-L1high; single positive refers to PD1high/PD-L1low or PD1low/PD-L1high; double negative refers to PD1low/PD-L1low) (log-rank test).
Figure 3
Figure 3
Implication of PD1/PD-L1 expression in ICC patients with different risk factors. (A) Kaplan-Meier estimate of overall survival in the whole cohort with different risk factors (*p < 0.05, log-rank test). (B) Kaplan-Meier estimate of cumulative recurrence in the whole cohort with different risk factors (*p < 0.05, log-rank test). (C) Kaplan-Meier estimate of overall survival of ICC patients with only HBV infection, grouped by PD1 level (log-rank test). (D) Kaplan-Meier estimate of cumulative recurrence in ICC patients with only HBV infection, grouped by PD1 level (log-rank test). (E) Kaplan-Meier estimate of overall survival of ICC patients with only HBV infection, grouped by PD-L1 level (log-rank test). (F) Kaplan-Meier estimate of cumulative recurrence in ICC patients with only HBV infection, grouped by PD-L1 level (log-rank test). (G) Kaplan-Meier estimate of overall survival of ICC patients with only hepatolithiasis, grouped by PD1 level (log-rank test). (H) Kaplan-Meier estimate of cumulative recurrence in ICC patients with only hepatolithiasis, grouped by PD1 level (log-rank test).
Figure 4
Figure 4
CD3, PD1, and PD-L1 expression and tumor responses to anti-PD1 immunotherapy in 7 advanced ICC patients. (A) Representative ICC tissues stained for CD3, PD1, and PD-L1 (200×). (B) Percentage change in tumor burden after anti-PD1 immunotherapy.

References

    1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.
    1. Zhou J, Sun HC, Wang Z, Cong WM, Wang JH, Zeng MS. et al. Guidelines for Diagnosis and Treatment of Primary Liver Cancer in China (2017 Edition) Liver Cancer. 2018;7:235–60.
    1. Liu ZY, Zhou YM, Shi LH, Yin ZF. Risk factors of intrahepatic cholangiocarcinoma in patients with hepatolithiasis: a case-control study. Hepatobiliary Pancreat Dis Int. 2011;10:626–31.
    1. Wu J, Yang S, Xu K, Ding C, Zhou Y, Fu X. et al. Patterns and Trends of Liver Cancer Incidence Rates in Eastern and Southeastern Asian Countries (1983-2007) and Predictions to 2030. Gastroenterology. 2018;154:1719–28.e5.
    1. Mavros MN, Economopoulos KP, Alexiou VG, Pawlik TM. Treatment and Prognosis for Patients With Intrahepatic Cholangiocarcinoma: Systematic Review and Meta-analysis. JAMA Surg. 2014;149:565–74.
    1. Sirica AE, Gores GJ, Groopman JD, Selaru FM, Strazzabosco M, Wang XW, Intrahepatic Cholangiocarcinoma: Continuing Challenges and Translational Advances. Hepatology; 2018.
    1. de Jong MC, Nathan H, Sotiropoulos GC, Paul A, Alexandrescu S, Marques H. et al. Intrahepatic cholangiocarcinoma: an international multi-institutional analysis of prognostic factors and lymph node assessment. J Clin Oncol. 2011;29:3140–5.
    1. Finn OJ. Cancer immunology. N Engl J Med. 2008;358:2704–15.
    1. Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD. et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N Engl J Med. 2018;379:341–51.
    1. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD. et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509–20.
    1. Kansy BA, Concha-Benavente F, Srivastava RM, Jie HB, Shayan G, Lei Y. et al. PD-1 Status in CD8(+) T Cells Associates with Survival and Anti-PD-1 Therapeutic Outcomes in Head and Neck Cancer. Cancer Res. 2017;77:6353–64.
    1. Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359:582–7.
    1. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAS, Andrews MC. et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell. 2017;170:1120–33.e17.
    1. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.
    1. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8:467–77.
    1. Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336–47.
    1. Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, Nakajima Y. et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res. 2009;15:971–9.
    1. Solinas A, Calvisi DF. Programmed death ligand 1 expression in hepatocellular carcinoma: A prognostic marker and therapeutic target for liver cancer? Hepatology. 2016;64:1847–9.
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19:133–50.
    1. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? J Exp Med. 2016;213:2835–40.
    1. Sato Y, Kinoshita M, Takemura S, Tanaka S, Hamano G, Nakamori S. et al. The PD-1/PD-L1 axis may be aberrantly activated in occupational cholangiocarcinoma. Pathol Int. 2017;67:163–70.
    1. Ye Y, Zhou L, Xie X, Jiang G, Xie H, Zheng S. Interaction of B7-H1 on intrahepatic cholangiocarcinoma cells with PD-1 on tumor-infiltrating T cells as a mechanism of immune evasion. J Surg Oncol. 2009;100:500–4.
    1. Fontugne J, Augustin J, Pujals A, Compagnon P, Rousseau B, Luciani A. et al. PD-L1 expression in perihilar and intrahepatic cholangiocarcinoma. Oncotarget. 2017;8:24644–51.
    1. Gani F, Nagarajan N, Kim Y, Zhu Q, Luan L, Bhaijjee F. et al. Program Death 1 Immune Checkpoint and Tumor Microenvironment: Implications for Patients With Intrahepatic Cholangiocarcinoma. Ann Surg Oncol. 2016;23:2610–7.
    1. Khan SA, Thomas HC, Davidson BR, Taylor-Robinson SD. Cholangiocarcinoma. Lancet. 2005;366:1303–14.
    1. Bridgewater J, Galle PR, Khan SA, Llovet JM, Park JW, Patel T. et al. Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J Hepatol. 2014;60:1268–89.
    1. Ishak KG, Anthony PP, Sobin LH. Histological typing of tumours of the liver. Springer Science & Business Media; 2012.
    1. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D. et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol. 2018;19:940–52.
    1. Yang LX, Gao Q, Shi JY, Wang ZC, Zhang Y, Gao PT. et al. Mitogen-activated protein kinase kinase kinase 4 deficiency in intrahepatic cholangiocarcinoma leads to invasive growth and epithelial-mesenchymal transition. Hepatology. 2015;62:1804–16.
    1. Shi GM, Ke AW, Zhou J, Wang XY, Xu Y, Ding ZB. et al. CD151 modulates expression of matrix metalloproteinase 9 and promotes neoangiogenesis and progression of hepatocellular carcinoma. Hepatology. 2010;52:183–96.
    1. Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB. et al. A Prospective, Multi-institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non-Small Cell Lung Cancer. JAMA Oncol. 2017;3:1051–8.
    1. Tyson GL, El-Serag HB. Risk factors for cholangiocarcinoma. Hepatology. 2011;54:173–84.
    1. Su CH, Shyr YM, Lui WY, P'Eng FK. Hepatolithiasis associated with cholangiocarcinoma. Br J Surg. 1997;84:969–73.
    1. Calvisi DF. Inhibition of hepatitis B virus-associated liver cancer by antiplatelet therapy: a revolution in hepatocellular carcinoma prevention? Hepatology. 2013;57:848–50.
    1. Tang LSY, Covert E, Wilson E, Kottilil S. Chronic Hepatitis B Infection: A Review. JAMA. 2018;319:1802–13.
    1. Harada K, Zen Y, Kanemori Y, Chen TC, Chen MF, Yeh TS. et al. Human REG I gene is up-regulated in intrahepatic cholangiocarcinoma and its precursor lesions. Hepatology. 2001;33:1036–42.
    1. Labib PL, Goodchild G, Pereira SP. Molecular Pathogenesis of Cholangiocarcinoma. BMC Cancer. 2019;19:185.
    1. Bally AP, Austin JW, Boss JM. Genetic and Epigenetic Regulation of PD-1 Expression. J Immunol. 2016;196:2431–7.
    1. Lei Z, Xia Y, Si A, Wang K, Li J, Yan Z. et al. Antiviral therapy improves survival in patients with HBV infection and intrahepatic cholangiocarcinoma undergoing liver resection. J Hepatol. 2018;68:655–62.
    1. Barnes E. Unravelling the fate of functional PD1+ T cells in chronic viral hepatitis. J Clin Invest. 2018;128:573–6.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54.
    1. Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC. et al. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. 2009;137:682–90.
    1. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.
    1. Chew V, Lai L, Pan L, Lim CJ, Li J, Ong R. et al. Delineation of an immunosuppressive gradient in hepatocellular carcinoma using high-dimensional proteomic and transcriptomic analyses. Proc Natl Acad Sci U S A. 2017;114:E5900–e9.
    1. Fung J. Liver fluke infestation and cholangio-hepatitis. Br J Surg. 1961;48:404–15.
    1. Dey B, Kaushal G, Jacob SE, Barwad A, Pottakkat B. Pathogenesis and Management of Hepatolithiasis: A Report of Two Cases. J Clin Diagn Res. 2016;10:Pd11–3.
    1. Chisari FV, Ferrari C. Hepatitis B virus immunopathogenesis. Annu Rev Immunol. 1995;13:29–60.

Source: PubMed

3
구독하다