1.
PD‐1 is an immune‐checkpoint regulator in T cells that transduces an inhibitory signal and inactivates T cells, while PD‐L1 is the PD‐1 ligand expressed in various cell types, including antigen‐ presenting cells (APCs) and cancer cells. 1 The PD‐L1/PD‐1 axis plays a key role in the immune escape of cancer cells, and inhibitory monoclonal antibodies (mAbs) against PD‐1 or PD‐L1 are currently used for treatment of a wide variety of cancer types. 2 These immune checkpoint inhibitors (ICIs) exhibit drastic therapeutic effects on a subset of patients and have revolutionized cancer therapy. However, the overall response rates are far from satisfactory due to intrinsic and acquired resistance. 3 Therefore, in order to reduce unnecessary treatment and improve the response rates, it is an urgent need to identify the appropriate markers that can discriminate responders and non‐responders to ICIs. So far, several markers associated with the positive response to PD‐1/PD‐L1 mAbs have been proposed, including, tumour infiltrated lymphocytes (TIL), tumour mutation burden (TMB), microsatellite instability, and PD‐L1 expression in tumors. 4 Among them, the PD‐L1 expression is used in clinic for several cancer types, and some immunohistochemistry (IHC) assays to quantify PD‐L1 expression in tumour tissues have been approved by the U.S. Food and Drug Administration (FDA). 5 However, in multiple clinical studies, PD‐L1 expression in tumours does not correlate with clinical responses to anti‐PD‐1/PD‐L1 therapy, 4 which is a puzzle in the field.
Recently, the puzzle seems to be resolved to some extent. PD‐L1 is highly glycosylated, and this post‐translational modification is critical for PD‐L1 protein stability and function. 6 , 7 PD‐L1 glycosylation is regulated by various oncogenic signalling pathways such as the epidermal growth factor receptor (EGFR) pathway, which inhibits phosphorylation of extracellular domain of PD‐L1 by GSK3β. 8 The phosphorylation by GSK3β hinders PD‐L1 from its glycosylation, leading to its ubiquitin‐mediated proteasome degradation. 6 In addition, another study also indicates that glycosylation of PD‐L1 interferes PD‐L1 protein detection by some traditional PD‐L1 antibodies that are designed to recognize its polypeptide antigens. 9 Human cancer cell lines or tissues section of several cancer types treated with a glycosidase have the higher signals of PD‐L1 in IHC staining than that of the untreated one, indicating that removal of N‐linked glycosylation of PD‐L1 enhances binding of traditional anti‐PD‐L1 mAb to PD‐L1. 9 Thus, it was proposed that inconsistent observations between PD‐L1 IHC staining and clinical responses may be due to the failure of accurate PD‐L1 detection in tumour tissues complicated by its glycosylation. Indeed, several studies have supported the notion that deglycosylation of tumour tissues improves predictive ability of PD‐L1 expression in tumours as a marker for anti‐PD‐1/PD‐L1 therapy. 10 , 11 , 12
It is worth noting that in 2019, based on the IMpassion130 trial, the U.S. FDA granted accelerated approval to atezolizumab, a humanized anti‐PD‐L1 monoclonal antibody, in combination with nab‐paclitaxel for treatment of advanced triple negative breast cancer (TNBC) patients, whose tumours show PD‐L1 positivity by IHC tests from immune cells. 13 However, in the following randomized Phase III clinical trial (IMpassion 131), the combination of atezolizumab and paclitaxel did not show significant clinical benefits compared to paclitaxel alone, 14 and the atezolizumab TNBC indication was withdrawn voluntarily by Roche in 2021. In a most recent study, nine TNBC patients, who were originally excluded for atezolizumab treatment based on criteria of the IMpassion130 trial, were treated with atezolizumab and the IHC of their tumor tissues were subjected to the deglycosylation process by glycosydase in order to better measure PD‐L1 level on the tumor cells. 12 In well consistent with the prediction, the PD‐L1 level on the surface of tumor cells after deglycosylation correlated very well to the response to atezolizumab treatment. 12 The study cohort is relatively small; however, the correlation is highly significant. This result suggests that the failure of the IMpassion 131 Phase III trial of atezolizumab is, at least in part, due to PD‐L1 glycosylation‐mediated incorrect readout of ICH staining. Thus, a larger cohort is warrant to develop further.
In addition, through mechanism studies, many druggable targets that are involved in the resistance to ICIs were identified and the combination therapy was shown to be able to increase therapeutic efficacy and/or reverse the resistance. 8 For instance, Tyro3 was shown to contribute to the resistance to ICI treatment through inhibition of ferroptosis that is required for T cell‐mediated cancer cell killing, providing a combination therapy of a Tyro3 inhibitor and ICIs to treat these types of resistant patients. 15 Several markers that were shown to increase therapeutic efficacy of ICIs by combination therapy from a subset of cancer cells through different kinds of mechanisms have also provided such type of marker‐guided effective therapy (MGET). 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 Thus, appropriate markers to stratify patients for different combination therapy may pave a way to increase therapeutic efficacy for ICIs.
In conclusion, PD‐L1 detection after deglycosylation by glycosidase pre‐treatment may improve the predictive value of PD‐L1 expression as a marker to select patients for ICI treatment (Figure 1). In addition, to increase therapeutic efficacy to benefit majority of patients, MGET continues to be needed to select right patients to be treated with right combination therapy. All these are worthy of validation by further clinical trials to enhance the therapeutic efficacy of ICIs.
FIGURE 1.

A potential strategy to improve PD‐L1 detection. TNBC tumour samples from patients are treated with glycosidase for deglycosylation, followed by immunohistochemistry (IHC) staining with anti‐PD‐L1 antibody. Compared to the conventional strategy without deglycosylation, it improves the detection of PD‐L1, thereby reducing false negative results. Thus, this strategy is expected to improve the predictive value of PD‐L1 expression as a marker to select patients for immune checkpoint inhibitors (ICI) treatment
CONFLICT OF INTEREST
Mien‐Chie Hung holds a patent on the methodology for de‐glycosylation staining.
ACKNOWLEDGEMENT
The work of the authors is supported by the Ministry of Science and Technology, Taiwan (MOST 110‐2314‐B‐039‐060 to H.Y., and MOST 110‐2639‐B‐039‐001‐ ASP to M.‐ C.H.), and the National Health Research Institute (NHRI‐EX110‐11012BI to S.‐C.W.); China Medical University Yingtsai Scholar Award (CMU109‐YT‐06 to H.Y.), and the institutional support to the Research Center for Cancer Biology of China Medical University.
Yamaguchi H, Wang S‐C, Hung M‐C. Prospects of the potential strategies to improve the efficacy of anti‐PD‐1/PD‐L1 therapy. Clin Transl Med. 2022;12:e803. 10.1002/ctm2.803
Contributor Information
Hirohito Yamaguchi, Email: hyamaguchi@mail.cmu.edu.tw.
Mien‐Chie Hung, Email: mhung@cmu.edu.tw.
REFERENCES
- 1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–245. [DOI] [PubMed] [Google Scholar]
- 2. Boussiotis VA. Molecular and biochemical aspects of the PD‐1 checkpoint pathway. N Engl J Med. 2016;375:1767–1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sharma P, Hu‐Lieskovan S, Wargo JA, Ribas A. Adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Grossman JE, Vasudevan D, Joyce CE, Hildago M. Is PD‐L1 a consistent biomarker for anti‐PD‐1 therapy? The model of balstilimab in a virally‐driven tumor. Oncogene. 2021;40:1393–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Prince EA, Sanzari JK, Pandya D, Huron D, Edwards R. Analytical concordance of PD‐L1 assays utilizing antibodies from FDA‐approved diagnostics in advanced cancers: A systematic literature review. JCO Precis Oncol. 2021;5:953–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Li C‐W, Lim S‐Oe, Xia W, et al. Glycosylation and stabilization of programmed death ligand‐1 suppresses T‐cell activity. Nat Commun. 2016;7:12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Li C‐W, Lim S‐Oe, Chung EM, et al. Eradication of triple‐negative breast cancer cells by targeting glycosylated PD‐L1. Cancer Cell. 2018;33:187–201. e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Yamaguchi H, Hsu J‐M, Yang W‐H, Hung M‐C. Mechanisms regulating PD‐L1 expression in cancers and associated opportunities for novel small‐molecule therapeutics. Nat Rev Clin Oncol. 2022. Online ahead of print. 10.1038/s41571-022-00601-9 [DOI] [PubMed] [Google Scholar]
- 9. Lee H‐H, Wang Y‐N, Xia W, et al. Removal of N‐linked glycosylation enhances PD‐L1 detection and predicts anti‐PD‐1/PD‐L1 therapeutic efficacy. Cancer Cell. 2019;36:168–178. e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Mei J, Xu J, Yang X, et al. A comparability study of natural and deglycosylated PD‐L1 levels in lung cancer: Evidence from immunohistochemical analysis. Mol Cancer. 2021;20:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Xu J, Yang X, Mao Y, et al. Removal of N‐linked glycosylation enhances PD‐L1 detection in colon cancer: Validation research based on immunohistochemistry analysis. Technol Cancer Res Treat. 2021;20:153303382110194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ou‐Yang F, Li C‐L, Chen C‐C, et al. De‐glycosylated membrane PD‐L1 in tumor tissues as a biomarker for responsiveness to atezolizumab (Tecentriq) in advanced breast cancer patients. Am J Cancer Res. 2022;12:123–137. [PMC free article] [PubMed] [Google Scholar]
- 13. Schmid P, Adams S, Rugo HS, et al. Atezolizumab and Nab‐Paclitaxel in advanced triple‐negative breast cancer. N Engl J Med. 2018;379:2108–2121. [DOI] [PubMed] [Google Scholar]
- 14. Miles D, Gligorov J, André F, et al. Primary results from IMpassion131, a double‐blind, placebo‐controlled, randomised phase III trial of first‐line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple‐negative breast cancer. Ann Oncol. 2021;32:994–1004. [DOI] [PubMed] [Google Scholar]
- 15. Jiang Z, Lim S‐O, Yan M, et al. TYRO3 induces anti‐PD‐1/PD‐L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J Clin Invest. 2021;131:e139434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Cha J‐H, Chan L‐C, Li C‐W, Hsu JL, Hung M‐C. Mechanisms controlling PD‐L1 expression in cancer. Mol Cell. 2019;76:359–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sun X, Li C‐W, Wang W‐J, et al. Inhibition of c‐MET upregulates PD‐L1 expression in lung adenocarcinoma. Am J Cancer Res. 2020;10:564–571. [PMC free article] [PubMed] [Google Scholar]
- 18. Chan L‐C, Li C‐W, Xia W, et al. IL‐6/JAK1 pathway drives PD‐L1 Y112 phosphorylation to promote cancer immune evasion. J Clin Invest. 2019;129:3324–3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Hsu J‐M, Xia W, Hsu Y‐H, et al. STT3‐dependent PD‐L1 accumulation on cancer stem cells promotes immune evasion. Nat Commun. 2018;9:1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Jiao S, Xia W, Yamaguchi H, et al. PARP inhibitor upregulates PD‐L1 expression and enhances cancer‐associated immunosuppression. Clin Cancer Res. 2017;23:3711–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Li H, Li C‐W, Li X, et al. MET inhibitors promote liver tumor evasion of the immune response by stabilizing PDL1. Gastroenterology. 2019;156:1849–1861. e1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Morel KL, Sheahan AV, Burkhart DL, et al. EZH2 inhibition activates a dsRNA‐STING‐interferon stress axis that potentiates response to PD‐1 checkpoint blockade in prostate cancer. Nat Cancer. 2021;2:444–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Zhang J, Bu X, Wang H, et al. Cyclin D‐CDK4 kinase destabilizes PD‐L1 via cullin 3‐SPOP to control cancer immune surveillance. Nature. 2018;553:91–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
