Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Epstein-Barr Virus | Primary research

Lymphocyte activating gene 3 protein expression in nasopharyngeal carcinoma is correlated with programmed cell death-1 and programmed cell death ligand-1, tumor-infiltrating lymphocytes

Authors: Fan Luo, Jiaxin Cao, Feiteng Lu, Kangmei Zeng, Wenjuan Ma, Yan Huang, Li Zhang, Hongyun Zhao

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Background

Immunotherapy has shown promising efficacy in patients with nasopharyngeal carcinoma (NPC). Lymphocyte activating 3 gene (LAG-3) represents a significant immune target, however, its relationship with NPC remains unclear. This study aimed to evaluate LAG-3 expression in NPC and its association with CD3+ tumor-infiltrating lymphocytes (TILs), Granzyme B (GZMB), programmed death ligand 1 (PD-L1), and programmed death 1 (PD-1) expression.

Methods

A total of 182 patients with NPC from Sun Yat-sen University Cancer Center, China, were included in this retrospective study. LAG-3 expression in 15 NPC cell lines and LAG-3, CD3+ TILs, GZMB, PD-L1 and PD-1 in clinical samples were estimated using immunohistochemistry. The Chi-square test was used to estimate the association between LAG-3, other biomarkers, and clinical characteristics. Survival analysis was performed using the Kaplan–Meier method and the Cox regression model.

Results

LAG-3 was negatively expressed in all of the 15 NPC cell lines, whereas, 147 patients with NPC (80.8%) exhibited high LAG-3 expression on TILs from tumor tissues. Male patients and those who were EBV-positive presented higher LAG-3 expression. Correlation analyses showed that LAG-3 expression was related to PD-1 expression on TILs, as well as, PD-L1 expression on tumor cells (TCs) and TILs. Both the univariate and multivariate Cox models indicated that pathological type III (P = 0.036), higher LAG-3 on TILs (P < 0.001), higher PD-L1 on TCs (P = 0.027), and higher PD-1 on TILs (P < 0.001) were associated with poorer disease-free survival (DFS). However, lower PD-L1 expression on TILs was related to superior DFS only in the univariate Cox analyses (P = 0.002).

Conclusion

Higher LAG-3 and PD-1 on TILs, and higher PD-L1 expression on TCs, and pathological type III were identified as independent risk factors for poorer DFS in NPC patients. Our data demonstrate that LAG-3 is a promising inhibitory receptor that may play an important role in anti-NPC therapy.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Henderson BE, Louie E, SooHoo Jing J, Buell P, Gardner MB. Risk factors associated with nasopharyngeal carcinoma. N Engl J Med. 1976;295(20):1101–6.PubMedCrossRef Henderson BE, Louie E, SooHoo Jing J, Buell P, Gardner MB. Risk factors associated with nasopharyngeal carcinoma. N Engl J Med. 1976;295(20):1101–6.PubMedCrossRef
3.
go back to reference Lee AW, Ng WT, Chan LL, Hung WM, Chan CC, Sze HC, Chan OS, Chang AT, Yeung RM. Evolution of treatment for nasopharyngeal cancer–success and setback in the intensity-modulated radiotherapy era. Radiother Oncol. 2014;110(3):377–84.PubMedCrossRef Lee AW, Ng WT, Chan LL, Hung WM, Chan CC, Sze HC, Chan OS, Chang AT, Yeung RM. Evolution of treatment for nasopharyngeal cancer–success and setback in the intensity-modulated radiotherapy era. Radiother Oncol. 2014;110(3):377–84.PubMedCrossRef
4.
go back to reference Zhang B, Mo Z, Du W, Wang Y, Liu L, Wei Y. Intensity-modulated radiation therapy versus 2D-RT or 3D-CRT for the treatment of nasopharyngeal carcinoma: a systematic review and meta-analysis. Oral Oncol. 2015;51(11):1041–6.PubMedCrossRef Zhang B, Mo Z, Du W, Wang Y, Liu L, Wei Y. Intensity-modulated radiation therapy versus 2D-RT or 3D-CRT for the treatment of nasopharyngeal carcinoma: a systematic review and meta-analysis. Oral Oncol. 2015;51(11):1041–6.PubMedCrossRef
5.
go back to reference Lee AW, Ma BB, Ng WT, Chan AT. Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol. 2015;33(29):3356–64.PubMedCrossRef Lee AW, Ma BB, Ng WT, Chan AT. Management of nasopharyngeal carcinoma: current practice and future perspective. J Clin Oncol. 2015;33(29):3356–64.PubMedCrossRef
6.
go back to reference Ma BBY, Lim WT, Goh BC, Hui EP, Lo KW, Pettinger A, Foster NR, Riess JW, Agulnik M, Chang AYC, et al. Antitumor Activity of nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an International, Multicenter Study of the Mayo Clinic Phase 2 Consortium (NCI-9742). J Clin Oncol. 2018;36(14):1412–8.PubMedPubMedCentralCrossRef Ma BBY, Lim WT, Goh BC, Hui EP, Lo KW, Pettinger A, Foster NR, Riess JW, Agulnik M, Chang AYC, et al. Antitumor Activity of nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an International, Multicenter Study of the Mayo Clinic Phase 2 Consortium (NCI-9742). J Clin Oncol. 2018;36(14):1412–8.PubMedPubMedCentralCrossRef
7.
go back to reference Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15(11):669–82.PubMedCrossRef Turley SJ, Cremasco V, Astarita JL. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat Rev Immunol. 2015;15(11):669–82.PubMedCrossRef
9.
go back to reference Hsu C, Lee SH, Ejadi S, Even C, Cohen RB, Le Tourneau C, Mehnert JM, Algazi A, van Brummelen EMJ, Saraf S, et al. Safety and antitumor activity of pembrolizumab in patients with programmed death-ligand 1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 Study. J Clin Oncol. 2017;35(36):4050–6.PubMedCrossRef Hsu C, Lee SH, Ejadi S, Even C, Cohen RB, Le Tourneau C, Mehnert JM, Algazi A, van Brummelen EMJ, Saraf S, et al. Safety and antitumor activity of pembrolizumab in patients with programmed death-ligand 1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 Study. J Clin Oncol. 2017;35(36):4050–6.PubMedCrossRef
10.
go back to reference Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy–inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res. 2012;18(24):6580–7.PubMedCrossRef Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy–inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res. 2012;18(24):6580–7.PubMedCrossRef
11.
go back to reference Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19(7):813–24.PubMedCrossRef Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007;19(7):813–24.PubMedCrossRef
12.
go back to reference Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMedCrossRef Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.PubMedCrossRef
13.
go back to reference Billon E, Finetti P, Bertucci A, Niccoli P, Birnbaum D, Mamessier E, Bertucci F. PDL1 expression is associated with longer postoperative, survival in adrenocortical carcinoma. Oncoimmunology. 2019;8(11):e1655362.PubMedPubMedCentralCrossRef Billon E, Finetti P, Bertucci A, Niccoli P, Birnbaum D, Mamessier E, Bertucci F. PDL1 expression is associated with longer postoperative, survival in adrenocortical carcinoma. Oncoimmunology. 2019;8(11):e1655362.PubMedPubMedCentralCrossRef
14.
go back to reference Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.PubMedPubMedCentralCrossRef Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.PubMedPubMedCentralCrossRef
15.
go back to reference Lee N, Zakka LR, Mihm MC Jr, Schatton T. Tumour-infiltrating lymphocytes in melanoma prognosis and cancer immunotherapy. Pathology. 2016;48(2):177–87.PubMedCrossRef Lee N, Zakka LR, Mihm MC Jr, Schatton T. Tumour-infiltrating lymphocytes in melanoma prognosis and cancer immunotherapy. Pathology. 2016;48(2):177–87.PubMedCrossRef
16.
go back to reference Rodriquenz MG, Roviello G, D'Angelo A, Lavacchi D, Roviello F, Polom K. MSI and EBV Positive gastric cancer's subgroups and their link with novel immunotherapy. J Clin Med. 2020. 2020;9(5): 1427.PubMedCentralCrossRef Rodriquenz MG, Roviello G, D'Angelo A, Lavacchi D, Roviello F, Polom K. MSI and EBV Positive gastric cancer's subgroups and their link with novel immunotherapy. J Clin Med. 2020. 2020;9(5): 1427.PubMedCentralCrossRef
17.
go back to reference Schalper KA, Kaftan E, Herbst RS. Predictive biomarkers for PD-1 axis therapies: the hidden treasure or a call for research. Clin Cancer Res. 2016;22(9):2102–4.PubMedPubMedCentralCrossRef Schalper KA, Kaftan E, Herbst RS. Predictive biomarkers for PD-1 axis therapies: the hidden treasure or a call for research. Clin Cancer Res. 2016;22(9):2102–4.PubMedPubMedCentralCrossRef
18.
go back to reference Chan OS, Kowanetz M, Ng WT, Koeppen H, Chan LK, Yeung RM, Wu H, Amler L, Mancao C. Characterization of PD-L1 expression and immune cell infiltration in nasopharyngeal cancer. Oral Oncol. 2017;67:52–60.PubMedCrossRef Chan OS, Kowanetz M, Ng WT, Koeppen H, Chan LK, Yeung RM, Wu H, Amler L, Mancao C. Characterization of PD-L1 expression and immune cell infiltration in nasopharyngeal cancer. Oral Oncol. 2017;67:52–60.PubMedCrossRef
19.
go back to reference Huang ZL, Liu S, Wang GN, Zheng SH, Ding SR, Tao YL, Chen C, Liu SR, Yang X, Chang H, et al. The prognostic significance of PD-L1 and PD-1 expression in patients with nasopharyngeal carcinoma: a systematic review and meta-analysis. Cancer Cell Int. 2019;19:141.PubMedPubMedCentralCrossRef Huang ZL, Liu S, Wang GN, Zheng SH, Ding SR, Tao YL, Chen C, Liu SR, Yang X, Chang H, et al. The prognostic significance of PD-L1 and PD-1 expression in patients with nasopharyngeal carcinoma: a systematic review and meta-analysis. Cancer Cell Int. 2019;19:141.PubMedPubMedCentralCrossRef
20.
go back to reference Zhang J, Fang W, Qin T, Yang Y, Hong S, Liang W, Ma Y, Zhao H, Huang Y, Xue C, et al. Co-expression of PD-1 and PD-L1 predicts poor outcome in nasopharyngeal carcinoma. Med Oncol. 2015;32(3):86.PubMedCrossRef Zhang J, Fang W, Qin T, Yang Y, Hong S, Liang W, Ma Y, Zhao H, Huang Y, Xue C, et al. Co-expression of PD-1 and PD-L1 predicts poor outcome in nasopharyngeal carcinoma. Med Oncol. 2015;32(3):86.PubMedCrossRef
21.
go back to reference Zhang Y, Kang S, Shen J, He J, Jiang L, Wang W, Guo Z, Peng G, Chen G, He J, et al. Prognostic significance of programmed cell death 1 (PD-1) or PD-1 ligand 1 (PD-L1) expression in epithelial-originated cancer: a meta-analysis. Medicine. 2015;94(6):e515.PubMedPubMedCentralCrossRef Zhang Y, Kang S, Shen J, He J, Jiang L, Wang W, Guo Z, Peng G, Chen G, He J, et al. Prognostic significance of programmed cell death 1 (PD-1) or PD-1 ligand 1 (PD-L1) expression in epithelial-originated cancer: a meta-analysis. Medicine. 2015;94(6):e515.PubMedPubMedCentralCrossRef
22.
go back to reference Zhu Q, Cai MY, Chen CL, Hu H, Lin HX, Li M, Weng DS, Zhao JJ, Guo L, Xia JC. Tumor cells PD-L1 expression as a favorable prognosis factor in nasopharyngeal carcinoma patients with pre-existing intratumor-infiltrating lymphocytes. Oncoimmunology. 2017;6(5):e1312240.PubMedPubMedCentralCrossRef Zhu Q, Cai MY, Chen CL, Hu H, Lin HX, Li M, Weng DS, Zhao JJ, Guo L, Xia JC. Tumor cells PD-L1 expression as a favorable prognosis factor in nasopharyngeal carcinoma patients with pre-existing intratumor-infiltrating lymphocytes. Oncoimmunology. 2017;6(5):e1312240.PubMedPubMedCentralCrossRef
23.
go back to reference Fang W, Yang Y, Ma Y, Hong S, Lin L, He X, Xiong J, Li P, Zhao H, Huang Y, et al. Camrelizumab (SHR-1210) alone or in combination with gemcitabine plus cisplatin for nasopharyngeal carcinoma: results from two single-arm, phase 1 trials. Lancet Oncol. 2018;19(10):1338–50.PubMedCrossRef Fang W, Yang Y, Ma Y, Hong S, Lin L, He X, Xiong J, Li P, Zhao H, Huang Y, et al. Camrelizumab (SHR-1210) alone or in combination with gemcitabine plus cisplatin for nasopharyngeal carcinoma: results from two single-arm, phase 1 trials. Lancet Oncol. 2018;19(10):1338–50.PubMedCrossRef
24.
go back to reference Prawira A, Oosting SF, Chen TW, Delos Santos KA, Saluja R, Wang L, Siu LL, Chan KKW, Hansen AR. Systemic therapies for recurrent or metastatic nasopharyngeal carcinoma: a systematic review. Br J Cancer. 2017;117(12):1743–52.PubMedPubMedCentralCrossRef Prawira A, Oosting SF, Chen TW, Delos Santos KA, Saluja R, Wang L, Siu LL, Chan KKW, Hansen AR. Systemic therapies for recurrent or metastatic nasopharyngeal carcinoma: a systematic review. Br J Cancer. 2017;117(12):1743–52.PubMedPubMedCentralCrossRef
25.
go back to reference He Y, Yu H, Rozeboom L, Rivard CJ, Ellison K, Dziadziuszko R, Suda K, Ren S, Wu C, Hou L, et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J Thorac Oncol. 2017;12(5):814–23.PubMedCrossRef He Y, Yu H, Rozeboom L, Rivard CJ, Ellison K, Dziadziuszko R, Suda K, Ren S, Wu C, Hou L, et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J Thorac Oncol. 2017;12(5):814–23.PubMedCrossRef
26.
go back to reference Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, Hercend T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393–405.PubMedCrossRef Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, Hercend T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393–405.PubMedCrossRef
27.
go back to reference Kisielow M, Kisielow J, Capoferri-Sollami G, Karjalainen K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur J Immunol. 2005;35(7):2081–8.PubMedCrossRef Kisielow M, Kisielow J, Capoferri-Sollami G, Karjalainen K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur J Immunol. 2005;35(7):2081–8.PubMedCrossRef
28.
go back to reference Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, De Marzo A, Anders R, Netto G, Getnet D, Bruno TC, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest. 2007;117(11):3383–92.PubMedPubMedCentralCrossRef Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, De Marzo A, Anders R, Netto G, Getnet D, Bruno TC, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest. 2007;117(11):3383–92.PubMedPubMedCentralCrossRef
29.
go back to reference Andreae S, Piras F, Burdin N, Triebel F. Maturation and activation of dendritic cells induced by lymphocyte activation gene-3 (CD223). J Immunol. 2002;168(8):3874–80.PubMedCrossRef Andreae S, Piras F, Burdin N, Triebel F. Maturation and activation of dendritic cells induced by lymphocyte activation gene-3 (CD223). J Immunol. 2002;168(8):3874–80.PubMedCrossRef
30.
go back to reference Workman CJ, Wang Y, El Kasmi KC, Pardoll DM, Murray PJ, Drake CG, Vignali DA. LAG-3 regulates plasmacytoid dendritic cell homeostasis. J Immunol. 2009;182(4):1885–91.PubMedCrossRef Workman CJ, Wang Y, El Kasmi KC, Pardoll DM, Murray PJ, Drake CG, Vignali DA. LAG-3 regulates plasmacytoid dendritic cell homeostasis. J Immunol. 2009;182(4):1885–91.PubMedCrossRef
31.
go back to reference Huard B, Mastrangeli R, Prigent P, Bruniquel D, Donini S, El-Tayar N, Maigret B, Dreano M, Triebel F. Characterization of the major histocompatibility complex class II binding site on LAG-3 protein. Proc Natl Acad Sci U S A. 1997;94(11):5744–9.PubMedPubMedCentralCrossRef Huard B, Mastrangeli R, Prigent P, Bruniquel D, Donini S, El-Tayar N, Maigret B, Dreano M, Triebel F. Characterization of the major histocompatibility complex class II binding site on LAG-3 protein. Proc Natl Acad Sci U S A. 1997;94(11):5744–9.PubMedPubMedCentralCrossRef
32.
go back to reference Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol. 1995;25(9):2718–21.PubMedCrossRef Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol. 1995;25(9):2718–21.PubMedCrossRef
33.
go back to reference Weber S, Karjalainen K. Mouse CD4 binds MHC class II with extremely low affinity. Int Immunol. 1993;5(6):695–8.PubMedCrossRef Weber S, Karjalainen K. Mouse CD4 binds MHC class II with extremely low affinity. Int Immunol. 1993;5(6):695–8.PubMedCrossRef
34.
go back to reference Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37.PubMedCrossRef Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10(1):29–37.PubMedCrossRef
35.
go back to reference Cappello P, Triebel F, Iezzi M, Caorsi C, Quaglino E, Lollini PL, Amici A, Di Carlo E, Musiani P, Giovarelli M, et al. LAG-3 enables DNA vaccination to persistently prevent mammary carcinogenesis in HER-2/neu transgenic BALB/c mice. Cancer Res. 2003;63(10):2518–25.PubMed Cappello P, Triebel F, Iezzi M, Caorsi C, Quaglino E, Lollini PL, Amici A, Di Carlo E, Musiani P, Giovarelli M, et al. LAG-3 enables DNA vaccination to persistently prevent mammary carcinogenesis in HER-2/neu transgenic BALB/c mice. Cancer Res. 2003;63(10):2518–25.PubMed
36.
go back to reference Chun T, Byun HJ, Chung HY, Chung YH. The effect of soluble LAG-3 (CD223) treatment in fetal thymic organ culture. Biotechnol Lett. 2004;26(17):1371–7.PubMedCrossRef Chun T, Byun HJ, Chung HY, Chung YH. The effect of soluble LAG-3 (CD223) treatment in fetal thymic organ culture. Biotechnol Lett. 2004;26(17):1371–7.PubMedCrossRef
37.
go back to reference Grosso JF, Goldberg MV, Getnet D, Bruno TC, Yen HR, Pyle KJ, Hipkiss E, Vignali DA, Pardoll DM, Drake CG. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J Immunol. 2009;182(11):6659–69.PubMedCrossRef Grosso JF, Goldberg MV, Getnet D, Bruno TC, Yen HR, Pyle KJ, Hipkiss E, Vignali DA, Pardoll DM, Drake CG. Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J Immunol. 2009;182(11):6659–69.PubMedCrossRef
38.
go back to reference Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21(4):503–13.PubMedCrossRef Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21(4):503–13.PubMedCrossRef
39.
go back to reference Johnson DB, Nixon MJ, Wang Y, Wang DY, Castellanos E, Estrada MV, Ericsson-Gonzalez PI, Cote CH, Salgado R, Sanchez V, et al. Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement. JCI Insight. 2018;3(24):e120360.PubMedCentralCrossRef Johnson DB, Nixon MJ, Wang Y, Wang DY, Castellanos E, Estrada MV, Ericsson-Gonzalez PI, Cote CH, Salgado R, Sanchez V, et al. Tumor-specific MHC-II expression drives a unique pattern of resistance to immunotherapy via LAG-3/FCRL6 engagement. JCI Insight. 2018;3(24):e120360.PubMedCentralCrossRef
40.
go back to reference Ohmura H, Yamaguchi K, Hanamura F, Ito M, Makiyama A, Uchino K, Shimokawa H, Tamura S, Esaki T, Mitsugi K, et al. OX40 and LAG3 are associated with better prognosis in advanced gastric cancer patients treated with anti-programmed death-1 antibody. Br J Cancer. 2020;122(10):1507–17.PubMedPubMedCentralCrossRef Ohmura H, Yamaguchi K, Hanamura F, Ito M, Makiyama A, Uchino K, Shimokawa H, Tamura S, Esaki T, Mitsugi K, et al. OX40 and LAG3 are associated with better prognosis in advanced gastric cancer patients treated with anti-programmed death-1 antibody. Br J Cancer. 2020;122(10):1507–17.PubMedPubMedCentralCrossRef
41.
go back to reference Sidaway P. Breast cancer: LAG3 expression indicates favourable outcomes. Nat Rev Clin Oncol. 2017;14(12):712.PubMed Sidaway P. Breast cancer: LAG3 expression indicates favourable outcomes. Nat Rev Clin Oncol. 2017;14(12):712.PubMed
42.
go back to reference Gameiro SF, Ghasemi F, Barrett JW, Koropatnick J, Nichols AC, Mymryk JS, Maleki Vareki S. Treatment-naïve HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology. 2018;7(10):e1498439.PubMedPubMedCentralCrossRef Gameiro SF, Ghasemi F, Barrett JW, Koropatnick J, Nichols AC, Mymryk JS, Maleki Vareki S. Treatment-naïve HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology. 2018;7(10):e1498439.PubMedPubMedCentralCrossRef
43.
go back to reference van der Zwan A, Bi K, Norwitz ER, Crespo ÂC, Claas FHJ, Strominger JL, Tilburgs T. Mixed signature of activation and dysfunction allows human decidual CD8(+) T cells to provide both tolerance and immunity. Proc Natl Acad Sci USA. 2018;115(2):385–90.PubMedCrossRef van der Zwan A, Bi K, Norwitz ER, Crespo ÂC, Claas FHJ, Strominger JL, Tilburgs T. Mixed signature of activation and dysfunction allows human decidual CD8(+) T cells to provide both tolerance and immunity. Proc Natl Acad Sci USA. 2018;115(2):385–90.PubMedCrossRef
44.
go back to reference Zhang Q, Chikina M, Szymczak-Workman AL, Horne W, Kolls JK, Vignali KM, Normolle D, Bettini M, Workman CJ, Vignali DAA. LAG3 limits regulatory T cell proliferation and function in autoimmune diabetes. Sci Immunol. 2017;2(9):eaah4569.PubMedPubMedCentralCrossRef Zhang Q, Chikina M, Szymczak-Workman AL, Horne W, Kolls JK, Vignali KM, Normolle D, Bettini M, Workman CJ, Vignali DAA. LAG3 limits regulatory T cell proliferation and function in autoimmune diabetes. Sci Immunol. 2017;2(9):eaah4569.PubMedPubMedCentralCrossRef
45.
go back to reference Dumic J, Dabelic S, Flögel M. Galectin-3: an open-ended story. Biochim Biophys Acta. 2006;1760(4):616–35.PubMedCrossRef Dumic J, Dabelic S, Flögel M. Galectin-3: an open-ended story. Biochim Biophys Acta. 2006;1760(4):616–35.PubMedCrossRef
46.
go back to reference Hsu MC, Hsiao JR, Chang KC, Wu YH, Su IJ, Jin YT, Chang Y. Increase of programmed death-1-expressing intratumoral CD8 T cells predicts a poor prognosis for nasopharyngeal carcinoma. Mod Pathol. 2010;23(10):1393–403.PubMedCrossRef Hsu MC, Hsiao JR, Chang KC, Wu YH, Su IJ, Jin YT, Chang Y. Increase of programmed death-1-expressing intratumoral CD8 T cells predicts a poor prognosis for nasopharyngeal carcinoma. Mod Pathol. 2010;23(10):1393–403.PubMedCrossRef
47.
go back to reference Al-Rajhi N, Soudy H, Ahmed SA, Elhassan T, Mohammed SF, Khoja HA, Ghebeh H. CD3+T-lymphocyte infiltration is an independent prognostic factor for advanced nasopharyngeal carcinoma. BMC Cancer. 2020;20(1):240.PubMedPubMedCentralCrossRef Al-Rajhi N, Soudy H, Ahmed SA, Elhassan T, Mohammed SF, Khoja HA, Ghebeh H. CD3+T-lymphocyte infiltration is an independent prognostic factor for advanced nasopharyngeal carcinoma. BMC Cancer. 2020;20(1):240.PubMedPubMedCentralCrossRef
48.
go back to reference Mhaidat NM, Al-azzam SI, Alzoubi KH, Khabour OF, Gharaibeh BF. Granzyme B gene polymorphisms, colorectal cancer risk, and metastasis. J Cancer Res Ther. 2014;10(3):587–90.PubMed Mhaidat NM, Al-azzam SI, Alzoubi KH, Khabour OF, Gharaibeh BF. Granzyme B gene polymorphisms, colorectal cancer risk, and metastasis. J Cancer Res Ther. 2014;10(3):587–90.PubMed
49.
go back to reference Wowk ME, Trapani JA. Cytotoxic activity of the lymphocyte toxin granzyme B. Microbes Infect. 2004;6(8):752–8.PubMedCrossRef Wowk ME, Trapani JA. Cytotoxic activity of the lymphocyte toxin granzyme B. Microbes Infect. 2004;6(8):752–8.PubMedCrossRef
50.
go back to reference Smyth MJ, McGuire MJ, Thia KY. Expression of recombinant human granzyme B. A processing and activation role for dipeptidyl peptidase I. J Immunol. 1995;154(12):6299–305.PubMedCrossRef Smyth MJ, McGuire MJ, Thia KY. Expression of recombinant human granzyme B. A processing and activation role for dipeptidyl peptidase I. J Immunol. 1995;154(12):6299–305.PubMedCrossRef
51.
go back to reference Ding W, Xu X, Qian Y, Xue W, Wang Y, Du J, Jin L, Tan Y. Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: a meta-analysis. Medicine. 2018;97(50):e13301.PubMedPubMedCentralCrossRef Ding W, Xu X, Qian Y, Xue W, Wang Y, Du J, Jin L, Tan Y. Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: a meta-analysis. Medicine. 2018;97(50):e13301.PubMedPubMedCentralCrossRef
52.
go back to reference Salama P, Phillips M, Platell C, Iacopetta B. Low expression of granzyme B in colorectal cancer is associated with signs of early metastastic invasion. Histopathology. 2011;59(2):207–15.PubMedCrossRef Salama P, Phillips M, Platell C, Iacopetta B. Low expression of granzyme B in colorectal cancer is associated with signs of early metastastic invasion. Histopathology. 2011;59(2):207–15.PubMedCrossRef
53.
go back to reference Wang Y, Dong T, Xuan Q, Zhao H, Qin L, Zhang Q. Lymphocyte-activation gene-3 expression and prognostic value in neoadjuvant-treated triple-negative breast cancer. J Breast Cancer. 2018;21(2):124–33.PubMedPubMedCentralCrossRef Wang Y, Dong T, Xuan Q, Zhao H, Qin L, Zhang Q. Lymphocyte-activation gene-3 expression and prognostic value in neoadjuvant-treated triple-negative breast cancer. J Breast Cancer. 2018;21(2):124–33.PubMedPubMedCentralCrossRef
54.
go back to reference Noh BJ, Kwak JY, Eom DW. Immune classification for the PD-L1 expression and tumour-infiltrating lymphocytes in colorectal adenocarcinoma. BMC Cancer. 2020;20(1):58.PubMedPubMedCentralCrossRef Noh BJ, Kwak JY, Eom DW. Immune classification for the PD-L1 expression and tumour-infiltrating lymphocytes in colorectal adenocarcinoma. BMC Cancer. 2020;20(1):58.PubMedPubMedCentralCrossRef
55.
go back to reference Zheng H, Zhou C, Lu X, Liu Q, Liu M, Chen G, Chen W, Wang S, Qiu Y. DJ-1 promotes survival of human colon cancer cells under hypoxia by modulating HIF-1α expression through the PI3K-AKT pathway. Cancer Manag Res. 2018;10:4615–29.PubMedPubMedCentralCrossRef Zheng H, Zhou C, Lu X, Liu Q, Liu M, Chen G, Chen W, Wang S, Qiu Y. DJ-1 promotes survival of human colon cancer cells under hypoxia by modulating HIF-1α expression through the PI3K-AKT pathway. Cancer Manag Res. 2018;10:4615–29.PubMedPubMedCentralCrossRef
56.
go back to reference Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10(21):7252–9.PubMedCrossRef Camp RL, Dolled-Filhart M, Rimm DL. X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 2004;10(21):7252–9.PubMedCrossRef
57.
go back to reference He Y, Rivard CJ, Rozeboom L, Yu H, Ellison K, Kowalewski A, Zhou C, Hirsch FR. Lymphocyte-activation gene-3, an important immune checkpoint in cancer. Cancer Sci. 2016;107(9):1193–7.PubMedPubMedCentralCrossRef He Y, Rivard CJ, Rozeboom L, Yu H, Ellison K, Kowalewski A, Zhou C, Hirsch FR. Lymphocyte-activation gene-3, an important immune checkpoint in cancer. Cancer Sci. 2016;107(9):1193–7.PubMedPubMedCentralCrossRef
58.
go back to reference Gandhi MK, Lambley E, Duraiswamy J, Dua U, Smith C, Elliott S, Gill D, Marlton P, Seymour J, Khanna R. Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. Blood. 2006;108(7):2280–9.PubMedCrossRef Gandhi MK, Lambley E, Duraiswamy J, Dua U, Smith C, Elliott S, Gill D, Marlton P, Seymour J, Khanna R. Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. Blood. 2006;108(7):2280–9.PubMedCrossRef
59.
go back to reference Martin MP, Borecki IB, Zhang Z, Nguyen L, Ma D, Gao X, Qi Y, Carrington M, Rader JS. HLA-Cw group 1 ligands for KIR increase susceptibility to invasive cervical cancer. Immunogenetics. 2010;62(11–12):761–5.PubMedPubMedCentralCrossRef Martin MP, Borecki IB, Zhang Z, Nguyen L, Ma D, Gao X, Qi Y, Carrington M, Rader JS. HLA-Cw group 1 ligands for KIR increase susceptibility to invasive cervical cancer. Immunogenetics. 2010;62(11–12):761–5.PubMedPubMedCentralCrossRef
60.
go back to reference Burova E, Hermann A, Dai J, Ullman E, Halasz G, Potocky T, Hong S, Liu M, Allbritton O, Woodruff A, et al. Preclinical development of the anti-LAG-3 antibody REGN3767: characterization and activity in combination with the anti-PD-1 antibody cemiplimab in human PD-1xLAG-3-knockin mice. Mol Cancer Ther. 2019;18(11):2051–62.PubMedCrossRef Burova E, Hermann A, Dai J, Ullman E, Halasz G, Potocky T, Hong S, Liu M, Allbritton O, Woodruff A, et al. Preclinical development of the anti-LAG-3 antibody REGN3767: characterization and activity in combination with the anti-PD-1 antibody cemiplimab in human PD-1xLAG-3-knockin mice. Mol Cancer Ther. 2019;18(11):2051–62.PubMedCrossRef
61.
go back to reference Ghosh S, Sharma G, Travers J, Kumar S, Choi J, Jun HT, Kehry M, Ramaswamy S, Jenkins D. TSR-033, a novel therapeutic antibody targeting LAG-3, enhances T-cell function and the activity of PD-1 blockade in vitro and in vivo. Mol Cancer Ther. 2019;18(3):632–41.PubMedCrossRef Ghosh S, Sharma G, Travers J, Kumar S, Choi J, Jun HT, Kehry M, Ramaswamy S, Jenkins D. TSR-033, a novel therapeutic antibody targeting LAG-3, enhances T-cell function and the activity of PD-1 blockade in vitro and in vivo. Mol Cancer Ther. 2019;18(3):632–41.PubMedCrossRef
62.
go back to reference Wan XX, Yi H, Qu JQ, He QY, Xiao ZQ. Integrated analysis of the differential cellular and EBV miRNA expression profiles in microdissected nasopharyngeal carcinoma and non-cancerous nasopharyngeal tissues. Oncol Rep. 2015;34(5):2585–601.PubMedCrossRef Wan XX, Yi H, Qu JQ, He QY, Xiao ZQ. Integrated analysis of the differential cellular and EBV miRNA expression profiles in microdissected nasopharyngeal carcinoma and non-cancerous nasopharyngeal tissues. Oncol Rep. 2015;34(5):2585–601.PubMedCrossRef
63.
go back to reference Müller E, Beleites E. The basaloid squamous cell carcinoma of the nasopharynx. Rhinology. 2000;38(4):208–11.PubMed Müller E, Beleites E. The basaloid squamous cell carcinoma of the nasopharynx. Rhinology. 2000;38(4):208–11.PubMed
64.
go back to reference Roy C, Choudhury KB, Basu S. Basaloid squamous cell carcinoma of nasopharynx: an extremely rare variety of tumour of nasopharynx. J Indian Med Assoc. 2011;109(5):343–4.PubMed Roy C, Choudhury KB, Basu S. Basaloid squamous cell carcinoma of nasopharynx: an extremely rare variety of tumour of nasopharynx. J Indian Med Assoc. 2011;109(5):343–4.PubMed
66.
go back to reference Okazaki T, Okazaki IM, Wang J, Sugiura D, Nakaki F, Yoshida T, Kato Y, Fagarasan S, Muramatsu M, Eto T, et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med. 2011;208(2):395–407.PubMedPubMedCentralCrossRef Okazaki T, Okazaki IM, Wang J, Sugiura D, Nakaki F, Yoshida T, Kato Y, Fagarasan S, Muramatsu M, Eto T, et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med. 2011;208(2):395–407.PubMedPubMedCentralCrossRef
67.
go back to reference Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, Eppolito C, Qian F, Lele S, Shrikant P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107(17):7875–80.PubMedPubMedCentralCrossRef Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, Eppolito C, Qian F, Lele S, Shrikant P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107(17):7875–80.PubMedPubMedCentralCrossRef
68.
go back to reference Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedCrossRef Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–27.PubMedCrossRef
69.
go back to reference Lucas CL, Workman CJ, Beyaz S, LoCascio S, Zhao G, Vignali DA, Sykes M. LAG-3, TGF-β, and cell-intrinsic PD-1 inhibitory pathways contribute to CD8 but not CD4 T-cell tolerance induced by allogeneic BMT with anti-CD40L. Blood. 2011;117(20):5532–40.PubMedPubMedCentralCrossRef Lucas CL, Workman CJ, Beyaz S, LoCascio S, Zhao G, Vignali DA, Sykes M. LAG-3, TGF-β, and cell-intrinsic PD-1 inhibitory pathways contribute to CD8 but not CD4 T-cell tolerance induced by allogeneic BMT with anti-CD40L. Blood. 2011;117(20):5532–40.PubMedPubMedCentralCrossRef
70.
go back to reference Richter K, Agnellini P, Oxenius A. On the role of the inhibitory receptor LAG-3 in acute and chronic LCMV infection. Int Immunol. 2010;22(1):13–23.PubMedCrossRef Richter K, Agnellini P, Oxenius A. On the role of the inhibitory receptor LAG-3 in acute and chronic LCMV infection. Int Immunol. 2010;22(1):13–23.PubMedCrossRef
71.
go back to reference Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, Guo B, Herbert DR, Bulfone A, Trentini F, et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med. 2013;19(6):739–46.PubMedCrossRef Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, Guo B, Herbert DR, Bulfone A, Trentini F, et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med. 2013;19(6):739–46.PubMedCrossRef
72.
go back to reference Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthelemy P, Porta C, George S, et al. Nivolumab plus Ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90.PubMedPubMedCentralCrossRef Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthelemy P, Porta C, George S, et al. Nivolumab plus Ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90.PubMedPubMedCentralCrossRef
73.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
74.
go back to reference Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, Minenza E, Linardou H, Burgers S, Salman P, et al. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378(22):2093–104.PubMedPubMedCentralCrossRef Hellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, Minenza E, Linardou H, Burgers S, Salman P, et al. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378(22):2093–104.PubMedPubMedCentralCrossRef
75.
go back to reference Antonia SJ, López-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17(7):883–95.PubMedCrossRef Antonia SJ, López-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17(7):883–95.PubMedCrossRef
76.
go back to reference Kotaskova J, Tichy B, Trbusek M, Francova HS, Kabathova J, Malcikova J, Doubek M, Brychtova Y, Mayer J, Pospisilova S. High expression of lymphocyte-activation gene 3 (LAG3) in chronic lymphocytic leukemia cells is associated with unmutated immunoglobulin variable heavy chain region (IGHV) gene and reduced treatment-free survival. J Mol Diagn. 2010;12(3):328–34.PubMedPubMedCentralCrossRef Kotaskova J, Tichy B, Trbusek M, Francova HS, Kabathova J, Malcikova J, Doubek M, Brychtova Y, Mayer J, Pospisilova S. High expression of lymphocyte-activation gene 3 (LAG3) in chronic lymphocytic leukemia cells is associated with unmutated immunoglobulin variable heavy chain region (IGHV) gene and reduced treatment-free survival. J Mol Diagn. 2010;12(3):328–34.PubMedPubMedCentralCrossRef
77.
go back to reference Gabrielson A, Wu Y, Wang H, Jiang J, Kallakury B, Gatalica Z, Reddy S, Kleiner D, Fishbein T, Johnson L, et al. Intratumoral CD3 and CD8 T-cell densities associated with relapse-free survival in HCC. Cancer Immunol Res. 2016;4(5):419–30.PubMedPubMedCentralCrossRef Gabrielson A, Wu Y, Wang H, Jiang J, Kallakury B, Gatalica Z, Reddy S, Kleiner D, Fishbein T, Johnson L, et al. Intratumoral CD3 and CD8 T-cell densities associated with relapse-free survival in HCC. Cancer Immunol Res. 2016;4(5):419–30.PubMedPubMedCentralCrossRef
Metadata
Title
Lymphocyte activating gene 3 protein expression in nasopharyngeal carcinoma is correlated with programmed cell death-1 and programmed cell death ligand-1, tumor-infiltrating lymphocytes
Authors
Fan Luo
Jiaxin Cao
Feiteng Lu
Kangmei Zeng
Wenjuan Ma
Yan Huang
Li Zhang
Hongyun Zhao
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02162-w

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine