266
Views
0
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

PD-L1 is Fascinating but IDO Needs Attention in Non-HCV and Non-HBV-Associated Hepatocellular Carcinoma Patients

, ORCID Icon, , , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , , ORCID Icon, , & ORCID Icon show all
Pages 921-934 | Received 23 Feb 2023, Accepted 20 May 2023, Published online: 17 Jun 2023

References

  • Sim HW, Knox J. Hepatocellular carcinoma in the era of immunotherapy. Curr Probl Cancer. 2018;42:40–48. doi:10.1016/j.currproblcancer.2017.10.007
  • Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380:1450–1462. doi:10.1056/NEJMra1713263
  • Hafeez Bhatti AB, Dar FS, Waheed A, Shafique K, Sultan F, Shah NH. Hepatocellular carcinoma in Pakistan: national trends and global perspective. Gastroenterol Res Pract. 2016;2016:5942306. doi:10.1155/2016/5942306
  • Akinyemiju T, Abera S, Ahmed M, et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the global burden of disease study 2015. JAMA Oncol. 2017;3:1683–1691. doi:10.1001/jamaoncol.2017.3055
  • Hatanaka K, Kudo M, Fukunaga T, et al. Clinical characteristics of NonBNonC-HCC: comparison with HBV and HCV related HCC. Intervirology. 2007;50:24–31. doi:10.1159/000096309
  • Tateishi R, Okanoue T, Fujiwara N, et al. Clinical characteristics, treatment, and prognosis of non-B, non-C hepatocellular carcinoma: a large retrospective multicenter cohort study. J Gastroenterol. 2015;50:350–360. doi:10.1007/s00535-014-0973-8
  • Utsunomiya T, Shimada M, Kudo M, et al. Nationwide study of 4741 patients with non-B non-C hepatocellular carcinoma with special reference to the therapeutic impact. Ann Surg. 2014;259:336–345. doi:10.1097/SLA.0b013e31829291e9
  • Nagaoki Y, Hyogo H, Ando Y, et al. Increasing incidence of non-HBV-and non-HCV-related hepatocellular carcinoma: single-institution 20-year study. BMC Gastroenterol. 2021;21:306. doi:10.1186/s12876-021-01884-5
  • Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi:10.1038/nm.3394
  • Meng X, Huang Z, Teng F, Xing L, Yu J. Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy. Cancer Treat Rev. 2015;41:868–876. doi:10.1016/j.ctrv.2015.11.001
  • Katsuya Y, Fujita Y, Horinouchi H, Ohe Y, Watanabe SI, Tsuta K. Immunohistochemical status of PD-L1 in thymoma and thymic carcinoma. Lung Cancer. 2015;88:154–159. doi:10.1016/j.lungcan.2015.03.003
  • Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S. Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. 2007;56:1173–1182. doi:10.1007/s00262-006-0266-z
  • Wei R, Guo L, Wang Q, Miao J, Kwok HF, Lin Y. Targeting PD-L1 protein: translation, modification and transport. Curr Protein Pept Sci. 2019;20(1):82–91. doi:10.2174/1389203719666180928105632
  • Paterson AM, Brown KE, Keir ME, et al. The programmed death-1 ligand 1: B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011;187:1097–1105. doi:10.4049/jimmunol.1003496
  • Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54:307–314. doi:10.1007/s00262-004-0593-x
  • Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapyPD-L1 IHC as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–856. doi:10.1158/1535-7163.MCT-14-0983
  • Jung HI, Jeong D, Ji S, et al. Overexpression of PD-L1 and PD-L2 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Res Treat. 2017;49:246–254. doi:10.4143/crt.2016.066
  • Gao Q, Wang XY, Qiu SJ, et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res. 2009;15:971–979. doi:10.1158/1078-0432.CCR-08-1608
  • Rizzo A, Ricci AD, Di Federico A, et al. Predictive biomarkers for checkpoint inhibitor-based immunotherapy in hepatocellular carcinoma: where do we stand? Front Oncol. 2021;11:803133. doi:10.3389/fonc.2021.803133
  • Viscardi G, Tralongo AC, Massari F, et al. Comparative assessment of early mortality risk upon immune checkpoint inhibitors alone or in combination with other agents across solid malignancies: a systematic review and meta-analysis. Eur J Cancer. 2022;177:175–185. doi:10.1016/j.ejca.2022.09.031
  • Rizzo A, Cusmai A, Gadaleta-Caldarola G, Palmiotti G. Which role for predictors of response to immune checkpoint inhibitors in hepatocellular carcinoma? Expert Rev Gastroenterol Hepatol. 2022;16(4):333–339. doi:10.1080/17474124.2022.2064273
  • Di Federico A, Rizzo A, Carloni R, et al. Atezolizumab-bevacizumab plus Y-90 TARE for the treatment of hepatocellular carcinoma: preclinical rationale and ongoing clinical trials. Expert Opin Investig Drugs. 2022;31(4):361–369. doi:10.1080/13543784.2022.2009455
  • Yin X, Wu T, Lan Y, Yang W. Current progress of immune checkpoint inhibitors in the treatment of advanced hepatocellular carcinoma. Biosci Rep. 2022;42(2):BSR20212304. doi:10.1042/BSR20212304
  • Shrestha R, Prithviraj P, Anaka M, et al. Monitoring immune checkpoint regulators as predictive biomarkers in hepatocellular carcinoma. Front Oncol. 2018;8:269. doi:10.3389/fonc.2018.00269
  • Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214. doi:10.1016/j.cell.2015.03.030
  • Archilla-Ortega A, Domuro C, Martin-Liberal J, Muñoz P. Blockade of novel immune checkpoints and new therapeutic combinations to boost antitumor immunity. J Exp Clin Cancer Res. 2022;41:62. doi:10.1186/s13046-022-02264-x
  • Shiravand Y, Khodadadi F, Kashani SM, et al. Immune checkpoint inhibitors in cancer therapy. Curr Oncol. 2022;29:3044–3060. doi:10.3390/curroncol29050247
  • Song X, Si Q, Qi R, et al. Indoleamine 2, 3-dioxygenase 1: a promising therapeutic target in malignant tumor. Front Immunol. 2021;12:800630. doi:10.3389/fimmu.2021.800630
  • El-Fattah A, Eslam E. IDO/kynurenine pathway in cancer: possible therapeutic approaches. J Transl Med. 2022;20:347. doi:10.1186/s12967-022-03554-w
  • Lee GK, Park HJ, Macleod M, Chandler P, Munn DH, Mellor AL. Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology. 2002;107:452–460. doi:10.1046/j.1365-2567.2002.01526.x
  • Mellor AL, Munn DH. Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation? Immunol Today. 1999;20:469–473. doi:10.1016/s0167-5699(99)01520-0
  • Yu J, Du W, Yan F, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190:3783–3797. doi:10.4049/jimmunol.1201449
  • Brandacher G, Perathoner A, Ladurner R, et al. Prognostic value of indoleamine 2, 3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clin Cancer Res. 2006;12:1144–1151. doi:10.1158/1078-0432.CCR-05-1966
  • Okamoto A, Nikaido T, Ochiai K, et al. Indoleamine 2, 3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clin Cancer Res. 2005;11:6030–6039. doi:10.1158/1078-0432.CCR-04-2671
  • Liu H, Shen Z, Wang Z, et al. Increased expression of IDO associates with poor postoperative clinical outcome of patients with gastric adenocarcinoma. Sci Rep. 2016;6:21319. doi:10.1038/srep21319
  • Li S, Han X, Lyu N, et al. Mechanism and prognostic value of indoleamine 2, 3-dioxygenase 1 expressed in hepatocellular carcinoma. Cancer Sci. 2018;109:3726–3736. doi:10.1111/cas.13811
  • Asghar K, Farooq A, Zulfiqar B, Rashid MU. Indoleamine 2, 3-dioxygenase: as a potential prognostic marker and immunotherapeutic target for hepatocellular carcinoma. World J Gastroenterol. 2017;23:2286–2293. doi:10.3748/wjg.v23.i13.2286
  • Pan K, Wang H, Chen MS, et al. Expression and prognosis role of indoleamine 2, 3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134:1247–1253. doi:10.1007/s00432-008-0395-1
  • Li T, Yang Y, Hua X, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–161. doi:10.1016/j.canlet.2011.12.020
  • Lin L, Yang DH, Huang Y, et al. Relationship between the expressions of indoleamine 2, 3-dioxygenase in hepatocellular carcinoma and clinicopathological parameters. Zhonghua Yi Xue Za Zhi. 2013;93:2186–2190.
  • Asghar K, Brain J, Palmer JM, et al. Potential role of indoleamine 2, 3-dioxygenase in primary biliary cirrhosis. Oncol Lett. 2017;14:5497–5504. doi:10.3892/ol.2017.6834
  • Ilie M, Khambata-Ford S, Copie-Bergman C, et al. Use of the 22C3 anti–PD-L1 antibody to determine PD-L1 expression in multiple automated immunohistochemistry platforms. PLoS One. 2017;12:e0183023. doi:10.1371/journal.pone.0183023
  • Cooper WA, Tran T, Vilain RE, et al. PD-L1 expression is a favorable prognostic factor in early stage non-small cell carcinoma. Lung Cancer. 2015;89:181–188. doi:10.1016/j.lungcan.2015.05.007
  • Igarashi T, Teramoto K, Ishida M, Hanaoka J, Daigo Y. Scoring of PD-L1 expression intensity on pulmonary adenocarcinomas and the correlations with clinicopathological factors. ESMO Open. 2016;1:e000083. doi:10.1136/esmoopen-2016-000083
  • Toyoda H, Kariyama K, Hiraoka A, et al. Improved survival of viral hepatocellular carcinoma but not non-viral hepatocellular carcinoma from 2000 to 2020: a multi-centre cohort study of 6007 patients from high-volume academic centres in Japan. Aliment Pharmacol Ther. 2022;56:694–701. doi:10.1111/apt.17088
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. doi:10.1126/science.1203486
  • Foerster F, Gairing SJ, Ilyas SI, Galle PR. Emerging immunotherapy for HCC: a guide for hepatologists. Hepatology. 2022;75(6):1604–1626. doi:10.1002/hep.32447
  • Pardee AD, Butterfield LH. Immunotherapy of hepatocellular carcinoma: unique challenges and clinical opportunities. Oncoimmunology. 2012;1:48–55. doi:10.4161/onci.1.1.18344
  • van Doorn DJ, Takkenberg RB, Klümpen HJ. Immune checkpoint inhibitors in hepatocellular carcinoma: an overview. Pharmaceuticals. 2020;14:3. doi:10.3390/ph14010003
  • McDermott DF, Atkins MB. PD-1 as a potential target in cancer therapy. Cancer Med. 2013;2:662–673. doi:10.1002/cam4.106
  • Li X, Wang X, Gao P. Diabetes mellitus and risk of hepatocellular carcinoma. Biomed Res Int. 2017;2017:5202684. doi:10.1155/2017/5202684
  • Campbell PT, Newton CC, Freedman ND, et al. Body mass index, waist circumference, diabetes, and risk of liver cancer for US adults. Cancer Res. 2016;76:6076–6083. doi:10.1158/0008-5472.CAN-16-0787
  • Ambade A, Mandrekar P. Oxidative stress and inflammation: essential partners in alcoholic liver disease. Int J Hepatol. 2012;2012:853175. doi:10.1155/2012/853175
  • Azzalini L, Ferrer E, Ramalho LN, et al. Cigarette smoking exacerbates nonalcoholic fatty liver disease in obese rats. Hepatology. 2010;51:1567–1576. doi:10.1002/hep.23516
  • Liang F, Wang G-Z, Wang Y, et al. Tobacco carcinogen induces tryptophan metabolism and immune suppression via induction of indoleamine 2,3-dioxygenase 1. Signal Transduct Target Ther. 2022;7(1):311. doi:10.1038/s41392-022-01127-3
  • Feo F, Pascale RM. Multifocal hepatocellular carcinoma: intrahepatic metastasis or multicentric carcinogenesis? Ann Transl Med. 2015;3:4. doi:10.3978/j.issn.2305-5839.2014.12.08
  • Brown ZJ, Yu SJ, Heinrich B, et al. Indoleamine 2, 3-dioxygenase provides adaptive resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunol Immunother. 2018;67:1305–1315. doi:10.1007/s00262-018-2190-4