Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2024 May 18;23(1):108.
doi: 10.1186/s12943-024-02023-w.

Regulatory mechanisms of PD-1/PD-L1 in cancers

Affiliations
Review

Regulatory mechanisms of PD-1/PD-L1 in cancers

Xin Lin et al. Mol Cancer. .

Abstract

Immune evasion contributes to cancer growth and progression. Cancer cells have the ability to activate different immune checkpoint pathways that harbor immunosuppressive functions. The programmed death protein 1 (PD-1) and programmed cell death ligands (PD-Ls) are considered to be the major immune checkpoint molecules. The interaction of PD-1 and PD-L1 negatively regulates adaptive immune response mainly by inhibiting the activity of effector T cells while enhancing the function of immunosuppressive regulatory T cells (Tregs), largely contributing to the maintenance of immune homeostasis that prevents dysregulated immunity and harmful immune responses. However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by neutralizing antibodies restores T cells activity and enhances anti-tumor immunity, achieving remarkable success in cancer therapy. Therefore, the regulatory mechanisms of PD-1/PD-L1 in cancers have attracted an increasing attention. This article aims to provide a comprehensive review of the roles of the PD-1/PD-L1 signaling in human autoimmune diseases and cancers. We summarize all aspects of regulatory mechanisms underlying the expression and activity of PD-1 and PD-L1 in cancers, including genetic, epigenetic, post-transcriptional and post-translational regulatory mechanisms. In addition, we further summarize the progress in clinical research on the antitumor effects of targeting PD-1/PD-L1 antibodies alone and in combination with other therapeutic approaches, providing new strategies for finding new tumor markers and developing combined therapeutic approaches.

Keywords: Combination therapy; PD-1; PD-L1; Regulatory mechanism; Tumor immunity.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
The immune regulation mechanism of the PD-1/PD-L1 axis. Antigen-presenting cells (APCs) deliver tumor antigens to the T-cell receptor (TCR) via the major histocompatibility complex(MHC), and when the MHC-antigen complex specifically binds to the TCR, it triggers a series of signal transductions, including the phosphatidylinositol signaling and mitogen- activated protein kinases signaling pathways, thus activating the immune responses of effector T cells. Upon PD-L1 binding to PD-1, phosphorylation of tyrosine residues in the ITSM and ITIM domains of the PD-1 cytoplasmic region occurs, recruiting and activating SHP2. Subsequently, recruited SHP-2 mediates dephosphorylation of TCR-associated CD3 and ZAP70 signalosomes, while inhibiting CD28 co-stimulatory signals. This further attenuates downstream TCR signaling strength and cytokine secretion, such as IL-2, ultimately inhibiting the function of T cells. Figure created with BioRender
Fig. 2
Fig. 2
Role of PD-1/PD-L1 in transplantation and autoimmune diseases. During organ transplantation, PD-1 is highly expressed on the surface of infiltrating T cells in grafts. The negative regulatory signal mediated by PD-1/PD-L1 can inhibit the excessive activation of T cells, induce immune tolerance, and effectively reduce the immune rejection between the host and the donor after surgery. Blocking PD-1/PD-L1 promotes proliferation of infiltrating T cells in grafts, exacerbating post-transplant immune rejection reactions and inducing severe and persistent tissue damage. Similarly, the disruption of the balance between PD-1 and PD-L1 signals can lead to the occurrence of many autoimmune diseases, such as type 1 diabetes mellitus (T1DM), multiple sclerosis (MS), systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA). Figure created with BioRender
Fig. 3
Fig. 3
The regulation of PD-1/PD-L1 signaling on immune cells. a. The PD-1/PD-L1 pathway promotes the exhaustion and apoptosis of effector T cells. Tex cells are characterized by increased expression of highly inhibitory receptors, including PD-1, LAG3, and TIGIT, decreased cytokine production such as TNF, IL-2, and IFN-γ, metabolic alterations, and impaired proliferative capacity and survival. b. PD-1/PD-L1 promotes the generation and development of induced Tregs (iTregs) by reducing the phosphorylation of PI3K/Akt/mTOR and S6, while enhancing PTEN, thus enhancing the immune suppression functions of Treg cells and inducing immune tolerance. c. PD-1/PD-L1 can promote the polarization of tumor-associated macrophages (TAM) toward the M2 phenotype, releasing large amounts of fibroblast growth factor, VEGF, TNF-α, and other cytokines to promote angiogenesis and support immune suppression, invasion, and metastasis of cancer cells, accelerating cancer progression. d. PD-1 on NK cells binds to PD-L1 on cancer cells, inhibiting the degranulation and cytotoxic function of NK cells, decreasing their ability to kill tumor cells, and promoting tumor immune escape. The use of PD-1 and PD-L1 inhibitors may reactivate the anti-tumor immune response of the above immune cells. Figure created with BioRender
Fig. 4
Fig. 4
PD-1/PD-L1 promotes tumorigenesis and development. PD-1/PD-L1 signaling can alter cellular energy synthesis and metabolic pathways by disrupting aerobic glycolysis, thereby promoting fatty acid oxidation (FAO) as the main source of energy in T cells. Additionally, NAMPT, a component of NAD+ metabolism, can enhance PD-L1 expression in tumor cells through Stat1-dependent IFN-γ signaling, thereby inhibiting T cell function and remodeling the local tumor microenvironment, ultimately exerting significant effects on tumor metastasis, recurrence, and prognosis. Furthermore, the gut microbiota can also improve tumor progression and enhance T cell immune responses, suggesting its potential use in improving the efficacy of PD-1 blockade therapy. Figure created with BioRender
Fig. 5
Fig. 5
The effects of nuclear PD-L1 on the tumor itself. PD-L1 not only promotes tumor cells to evade immune surveillance but also facilitates tumor progression in an immune-independent manner. PD-L1, which is expressed at high levels in tumor cells, can enter the nucleus by binding to the nuclear entry protein KPNB1 and exert pro-cancer effects. Nuclear PD-L1 can also trigger the upregulation of immune checkpoint genes, including PD-L2 and VISTA, thereby enhancing the anti-tumor responses of PD-1 inhibition. Under hypoxic conditions, treatment with TNFα and CHX can promote the nuclear translocation of PD-L1, which then interacts with p-Stat3-Y705. Subsequently, p-Stat3-Y705 combines with the GSDMC promoter region, leading to the upregulation of GSDMC gene expression. Furthermore, GSDMC is cleaved and activated by caspase-8, triggering pyroptosis in cells and necrosis in the hypoxic areas of tumors. Figure created with BioRender
Fig. 6
Fig. 6
Genetic and transcriptional regulatory mechanisms of PD-L1 expression. At the gene level, tumors with PD-L1 gene amplification in chromosome 9p24.1 have a higher mutation load and a close correlation with increased PD-L1 expression. Epigenetic modifications of the PD-L1 promoter region, including DNA methylation, histone methylation, and acetylation, are also important in the regulation of PD-L1 expression. For example, TNF-α/TGFβ1 induces demethylation of the PD-L1 promoter by decreasing DNMT1 (DNA methyltransferase) levels, resulting in PD-L1 up-regulation and thus exerting immunosuppressive effects. At the transcriptional level, PD-L1 expression is primarily regulated by transcription factors, including STAT, MYC, NF-κB, IRF1, AP-1, and HIF-1α, as well as signaling pathway effector molecules such as MAPK/PI3K/Akt, JAK/STAT3, and EGFR/MAPK. Figure created with BioRender
Fig. 7
Fig. 7
Mechanisms of PD-L1 expression regulation at post-transcriptional and post-translational. Non-coding RNAs such as miR-34, miR-200, and miR-197 can suppress PD-L1 mRNA expression by directly binding to PD-L1 3′UTR. The m6A modification of PD-L1 mRNA is critical for regulating the expression and stability of PD-L1 and mediating tumor immune escape. For instance, demethylases such as FTO and ALKBH can remove the m6A modification on PD-L1 mRNA, increasing its stability and promoting high PD-L1 expression. The METTL3/IGF2BP3 axis also enhances the stability of PD-L1 mRNA by upregulating its m6A modification, further promoting tumor immune evasion. Additionally, post-translational modifications, including phosphorylation, ubiquitination, glycosylation, and palmitoylation can regulate PD-L1 protein expression by influencing its activity, stability, and membrane expression in cancer cells. Figure created with BioRender

Similar articles

Cited by

References

    1. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95. doi: 10.1002/j.1460-2075.1992.tb05481.x. - DOI - PMC - PubMed
    1. Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y et al. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol Baltim Md. 1950. 2002;169:5538–45. - PubMed
    1. Liang SC, Latchman YE, Buhlmann JE, Tomczak MF, Horwitz BH, Freeman GJ, et al. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur J Immunol. 2003;33:2706–16. doi: 10.1002/eji.200324228. - DOI - PubMed
    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the Pd-1 Immunoinhibitory receptor by a Novel B7 Family Member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34. doi: 10.1084/jem.192.7.1027. - DOI - PMC - PubMed
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. - DOI - PubMed