Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1021-335X Online ISSN: 1791-2431
Journal Cover
December-2025 Volume 54 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 54 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review)

  • Authors:
    • Li-Hui Gu
    • Ai Guo
    • Yi-Yue Ding
    • Xue-Jie Wang
    • Hong-Xing Zhang
    • Wan-Li Duan
    • Bao-Gang Zhang
  • View Affiliations / Copyright

    Affiliations: Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China, Department of Diagnostic Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China, Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
    Copyright: © Gu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 167
    |
    Published online on: September 26, 2025
       https://doi.org/10.3892/or.2025.9000
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Cancer cells play a pivotal role in immune evasion by activating the programmed cell death protein 1 (PD‑1)/PD‑ligand (L)1 signaling pathway or immune cells within the tumor microenvironment. The ubiquitin‑proteasome system (UPS), the primary pathway for intracellular protein degradation, has been increasingly implicated in mediating tumor immune escape and resistance to anti‑PD‑1/PD‑L1 therapy. Targeting the UPS has demonstrated significant potential in improving the efficacy of tumor immunotherapy. Therefore, a deeper understanding of the molecular mechanisms by which UPS contributes to tumor resistance against PD‑1/PD‑L1 blockade, along with the optimization of UPS‑targeted small‑molecule drug design, holds scientific and clinical significance. In the present review, the role of UPS in tumor immune evasion through the regulation of PD‑1/PD‑L1 ubiquitination was discussed and potential therapeutic agents that may enhance the effectiveness of anti‑PD‑1/PD‑L1 treatment are summarized. These insights provide a theoretical foundation for advancing cancer immunotherapy and developing novel combination strategies.

Introduction

The biological hallmarks of cancer include uncontrolled cell proliferation and differentiation, dysregulated signaling pathways, genomic instability and metabolic reprogramming (1). These characteristics make the development of more effective cancer therapies a critical research priority. In recent years, the emergence of immunotherapy represents a major breakthrough in cancer treatment. As the fifth treatment modality following surgery, radiotherapy, chemotherapy and targeted therapy, immunotherapy works by re-establishing the tumor-immune cycle and restoring the body's normal anti-tumor immune response, thereby achieving tumor control and eradication (2,3). Immune checkpoint blockade therapy has been widely proven to be effective against various human tumors, while cell therapy has shown notable efficacy in hematological malignancies and remains in the clinical research stage for solid tumors. In the field of immune checkpoint inhibitors, the main approaches include: i) Monoclonal antibody therapy targeting programmed cell death protein 1 (PD-1) and its ligand PD-L1; ii) monoclonal antibody therapy targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4); and iii) monoclonal antibody therapy targeting lymphocyte-activation gene 3. As for adoptive cell therapy, it primarily encompasses: i) Tumor-infiltrating lymphocyte therapy; ii) T cell receptor-engineered T cell therapy; iii) chimeric antigen receptor T cell (CAR-T) therapy; and iv) natural killer (NK) cell therapy (4).

PD-1/PD-L1 monoclonal antibody therapy has emerged as the most widely used immune checkpoint inhibitor in clinical practice, demonstrating notable efficacy across various malignancies (5). PD-1, also known as CD279, is a pivotal immune checkpoint molecule that plays a central role in maintaining immune homeostasis and regulating tumor immune evasion. PD-1 is predominantly expressed on the surface of B cells, T cells and NK cells, where it can specifically recognize and bind to two ligands expressed on tumor cells: i) PD-L1 (CD274); and ii) PD-L2 (CD273) (6). Unlike CTLA-4 which primarily regulates immune responses during the early stage of T cell activation, PD-1 predominantly suppresses effector T cell function in peripheral tissues and the tumor microenvironment during the effector phase (7). In the tumor microenvironment, the binding of PD-L1 expressed at high level on tumor cells to PD-1 on T cell surfaces induces conformational changes in PD-1. This leads to the exposure and phosphorylation of the immunoreceptor tyrosine-based inhibitory motif (ITIM) at Y223 and the immunoreceptor tyrosine-based switch motif (ITSM) at Y248. The phosphorylated ITSM preferentially recruits the SHP-2 protein tyrosine phosphatase, while the phosphorylated ITIM forms dimers with the SHP-1 protein tyrosine phosphatase. These molecular events collectively attenuate T cell activation signals, suppress T cell cytotoxic function and consequently mediate negative regulation of immune responses to maintain immune homeostasis (8–11). PD-L1 can also interact with the costimulatory molecule CD80, transmitting inhibitory signals to activated T cells (12). Currently, several PD-1/PD-L1 monoclonal antibodies such as nivolumab (13), pembrolizumab (14) and avelumab (15) have been widely used in clinical cancer treatment. Although these immune checkpoint inhibitors demonstrate notable efficacy against various solid tumors and hematological malignancies, acquired resistance remains a major clinical challenge (16).

Research has demonstrated that dysregulation of the ubiquitin-dependent protein degradation pathway represents a crucial molecular mechanism in cancer pathogenesis (17,18). The UPS is an essential protein degradation mechanism in cells. This system primarily works by ubiquitinating damaged, abnormal, or functionally completed regulatory proteins and directing their degradation by the proteasome. (19). The UPS consists of a series of enzymes: i) Ubiquitin-activating enzyme E1 activates ubiquitin molecules; ii) ubiquitin-conjugating enzyme E2 mediates ubiquitin transfer; and iii) ubiquitin ligase E3 specifically recognizes substrate proteins and completes ubiquitin tagging (Fig. 1). These enzymes work cooperatively to ultimately achieve targeted protein degradation via the proteasome pathway (20,21). Ubiquitinated substrates are classified into different ubiquitination pathways based on the types of polyubiquitin chains, with K48 and K63 being the two most widely studied ubiquitination forms (22). Among these, K48-linked ubiquitination is recognized to direct target proteins for degradation via the proteasome pathway (23,24). K63-linked ubiquitination is primarily involved in proteasome-independent signaling pathways, typically associated with positive regulatory processes such as protein stabilization, subcellular localization and functional activation, including critical biological processes such as endocytic trafficking, DNA replication and signal transduction (25). Moreover, this modification can also facilitate substrate protein degradation through the autophagy-lysosome pathway (26).

Protein degradation mediated by the
UPS and tumor immune evasion mediated by PD-1/PD-L1. (A) The UPS
consists of three components: a ubiquitination modification system
mediated by a three-step enzymatic cascade, the proteasome; and the
deubiquitination process. (B) In the tumor microenvironment, the
heterodimer formed between the ITSM of PD-1 and SHP-2 inhibits TCR
signal activation, while the dimer formed by ITIM and SHP-1 further
enhances TCR suppression, thereby promoting tumor immune evasion.
UPS, ubiquitin-proteasome system; PD-1, programmed death receptor
1; PD-L1, programmed cell death ligand 1; ITSM, immunoreceptor
tyrosine-based activation motif; ITIM, immunoreceptor
tyrosine-based inhibitory motif; SHP-2, Src Homology 2
domain-containing Protein Tyrosine Phosphatase-2; TCR,
receptor-engineered; DUBs, deubiquitinating enzymes; SHP-1, Src
Homology 2 domain-containing Protein Tyrosine Phosphatase-1; 19S
RP, 19S regulatory particle; 20S CP, 20S core particle; MHC, major
histocompatibility complex.

Figure 1.

Protein degradation mediated by the UPS and tumor immune evasion mediated by PD-1/PD-L1. (A) The UPS consists of three components: a ubiquitination modification system mediated by a three-step enzymatic cascade, the proteasome; and the deubiquitination process. (B) In the tumor microenvironment, the heterodimer formed between the ITSM of PD-1 and SHP-2 inhibits TCR signal activation, while the dimer formed by ITIM and SHP-1 further enhances TCR suppression, thereby promoting tumor immune evasion. UPS, ubiquitin-proteasome system; PD-1, programmed death receptor 1; PD-L1, programmed cell death ligand 1; ITSM, immunoreceptor tyrosine-based activation motif; ITIM, immunoreceptor tyrosine-based inhibitory motif; SHP-2, Src Homology 2 domain-containing Protein Tyrosine Phosphatase-2; TCR, receptor-engineered; DUBs, deubiquitinating enzymes; SHP-1, Src Homology 2 domain-containing Protein Tyrosine Phosphatase-1; 19S RP, 19S regulatory particle; 20S CP, 20S core particle; MHC, major histocompatibility complex.

The 26S proteasome is a multi-subunit proteolytic complex composed of a 20S core particle (CP) and a 19S regulatory particle (RP), which specifically recognizes polyubiquitin-tagged proteins and degrades them into short peptides (27). The 19S RP performs three key functions: i) Recognizing ubiquitinated substrate proteins; ii) regulating the deubiquitination process; and iii) delivering ubiquitinated proteins to the 20S CP. The 20S CP is a barrel-shaped proteolytic core containing active catalytic sites, where the final protein degradation occurs (28,29). Deubiquitinating enzymes (DUBs) can reverse protein ubiquitination, with their primary functions including: i) Maintaining cellular free ubiquitin levels; ii) releasing substrate proteins from the ubiquitin-proteasome degradation pathway; and iii) protecting target proteins from degradation (22). Research has revealed an increasingly clear connection between ubiquitination and cancer immunotherapy. Tumor cells can modulate the UPS to stabilize immune checkpoint protein expression and suppress immune-related protein function, thereby evading immune surveillance and promoting tumor progression.

In recent years, the regulatory role of the UPS in cancer immunotherapy has received growing attention. The present review summarized the key molecular mechanisms of UPS involvement in tumor immune regulation and discusses potential therapeutic strategies to enhance immunotherapeutic efficacy.

Mechanistic role of PD-1/PD-L1 ubiquitination in tumor immune evasion and immunotherapy

Regulatory role of the UPS in tumor immune escape

Ubiquitination is a series of biochemical reactions mediated by ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2) and ubiquitin ligase (E3) (20,21). E3 ubiquitin ligase is a key component of this system, which can recognize and target specific ubiquitinated substrate proteins (25). The UPS, through dynamically regulating the surface expression of PD-1/PD-L1, has become a crucial link in tumor immune evasion (Fig. 2) (30,31).

The role of ubiquitination in the
treatment with anti-PD-1/PD-L1 mAb. The ubiquitination functions of
E3 ubiquitin ligases SPOP, TRIM21, ARIH1, MIB2, Skp2, NEDD4,
β-TrCP, ITCH and RNF125 can affect the immune capacity of tumors by
regulating the expression level of PD-L1. The ubiquitination
functions of E3 ubiquitin ligases FBXO38, KLHL22, FBW7 and c-Cbl
can influence the immune function of T cells by regulating the
expression level of PD-1. PD-1, programmed cell death protein 1;
PD-L1, programmed death-ligand 1; mAb, monoclonal antibodies; SPOP,
speckle-type POZ protein; TRIM21, tripartite motif-containing
protein 21; ARIH1, ariadne RBR E3 ubiquitin protein ligase 1; MIB2,
mind bomb E3 ubiquitin protein ligase 2; Skp2, S-phase
kinase-associated protein 2; NEDD4, neural precursor cell expressed
developmentally downregulated 4; β-Trcp, beta-transducin
repeat-containing protein; ITCH, itchy E3 ubiquitin protein ligase;
ITIM, immunoreceptor tyrosine-based inhibitory motif; RNF125, ring
finger protein 125; FBXO38, F-Box protein 38; KLHL22, kelch-like
protein 22; FBW7 F-Box and WD repeat domain-containing 7; c-Cbl,
Casitas b lymphoma; ALDH2, aldehyde dehydrogenase 2; BCLAF1
BCL2-associated transcription factor 1; β-Trcp beta-transducin
repeat-containing protein; CDH1 cadherin 1; CDK1 cyclin-dependent
kinase 1; CDK4 cyclin-dependent kinase 4; CDK5 cyclin-dependent
kinase 5; EGFR, epidermal growth factor receptor; FGFR3, fibroblast
growth factor receptor 3; GSK3α, glycogen synthase kinase 3 alpha;
GSK3β glycogen synthase kinase 3 beta; IL-2, interleukin-2; IL-18,
interleukin-18; LKB1, liver kinase B1; RAB8, RAS-related protein
Rab-8; SGLT2, sodium-glucose cotransporter 2; STAT5, signal
transducer and activator of transcription 5.

Figure 2.

The role of ubiquitination in the treatment with anti-PD-1/PD-L1 mAb. The ubiquitination functions of E3 ubiquitin ligases SPOP, TRIM21, ARIH1, MIB2, Skp2, NEDD4, β-TrCP, ITCH and RNF125 can affect the immune capacity of tumors by regulating the expression level of PD-L1. The ubiquitination functions of E3 ubiquitin ligases FBXO38, KLHL22, FBW7 and c-Cbl can influence the immune function of T cells by regulating the expression level of PD-1. PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; mAb, monoclonal antibodies; SPOP, speckle-type POZ protein; TRIM21, tripartite motif-containing protein 21; ARIH1, ariadne RBR E3 ubiquitin protein ligase 1; MIB2, mind bomb E3 ubiquitin protein ligase 2; Skp2, S-phase kinase-associated protein 2; NEDD4, neural precursor cell expressed developmentally downregulated 4; β-Trcp, beta-transducin repeat-containing protein; ITCH, itchy E3 ubiquitin protein ligase; ITIM, immunoreceptor tyrosine-based inhibitory motif; RNF125, ring finger protein 125; FBXO38, F-Box protein 38; KLHL22, kelch-like protein 22; FBW7 F-Box and WD repeat domain-containing 7; c-Cbl, Casitas b lymphoma; ALDH2, aldehyde dehydrogenase 2; BCLAF1 BCL2-associated transcription factor 1; β-Trcp beta-transducin repeat-containing protein; CDH1 cadherin 1; CDK1 cyclin-dependent kinase 1; CDK4 cyclin-dependent kinase 4; CDK5 cyclin-dependent kinase 5; EGFR, epidermal growth factor receptor; FGFR3, fibroblast growth factor receptor 3; GSK3α, glycogen synthase kinase 3 alpha; GSK3β glycogen synthase kinase 3 beta; IL-2, interleukin-2; IL-18, interleukin-18; LKB1, liver kinase B1; RAB8, RAS-related protein Rab-8; SGLT2, sodium-glucose cotransporter 2; STAT5, signal transducer and activator of transcription 5.

Effect of PD-L1 ubiquitination on tumor immune evasion and immunotherapy

Multiple studies have shown that speckle-type POZ protein (SPOP) plays a crucial role in mediating the ubiquitination process of PD-L1 and the mechanism of tumor immune evasion (31–33). Zhang et al (31) found that in colorectal cancer cells, the E3 ubiquitin ligase SPOP can promote the ubiquitination and degradation of PD-L1. Meanwhile, the ALDH2 expressed at high levels in cancer cells can competitively bind to PD-L1 with SPOP, thereby inhibiting the ubiquitination process of PD-L1 mediated by SPOP and ultimately weakening the antitumor effect of T cells. In addition, in hepatocellular carcinoma, the transcription factor BCLAF1 can inhibit the ubiquitination of PD-L1 by SPOP by targeting and binding to SPOP. This mechanism enhances the stability of PD-L1 and promotes tumor immune evasion (32). Ding et al (34) found that SGLT2 can competitively bind to PD-L1 with the E3 ubiquitin ligase SPOP, thereby preventing PD-L1 from being degraded through the proteasome pathway. The small-molecule SGLT2 inhibitor canagliflozin can disrupt the interaction between SGLT2 and PD-L1, prompting SPOP to recognize PD-L1 and promote its ubiquitination, followed by degradation via the proteasome pathway, thereby enhancing the antitumor activity of T cells. Zhang et al (33) found that in various types of cancer, CDK4 can directly promote the phosphorylation of SPOP at Ser6. The phosphorylated SPOP can bind to the scaffold protein 14-3-3γ, thereby blocking the binding of SPOP to the complex activator FZR1, stabilizing the expression level of SPOP, enabling it to recognize and promote the K48 ubiquitination of PD-L1 and eventually leading to its degradation via the proteasome pathway.

While SPOP serves as a well-characterized E3 ligase regulating PD-L1 stability, other ubiquitin ligases also contribute markedly to this regulatory network. In non-small cell lung cancer, the E3 ubiquitin ligase TRIM21 can ubiquitinate PD-L1 and promote its degradation. LINC02418, acting as a molecular sponge, can form a ternary complex with TRIM21 and PD-L1. This complex enhances the ubiquitination of PD-L1 by TRIM21, ultimately leading to resistance to anti-PD-L1-based immunotherapy in non-small cell lung cancer (35). Gao et al (36) found that CDK5 can promote the ubiquitination process of PD-L1 by TRIM21 by interacting with TRIM21 and PD-L1. This mechanism notably exacerbates the resistance of patients with non-small cell lung cancer to anti-PD-L1-based immunotherapy. Wu et al (37) showed that in lymphosarcoma and non-small cell lung cancer cells, glycogen synthase kinase 3 alpha (GSK3α) can enhance the recognition and ubiquitination of PD-L1 by the E3 ubiquitin ligase ARIH1 by promoting the phosphorylation of PD-L1 at Ser279 and Ser283. However, the inhibitory effect of epidermal growth factor (EGF) receptor (EGFR) on GSK3α in cancer cells mediates tumor immune treatment resistance. The combined application of the EGFR inhibitor ES-072 and immunotherapy can effectively enhance the efficacy of immunotherapy (37). Wei et al (38) found that RNF125 can directly interact with PD-L1, promote the K48 ubiquitination of PD-L1, thereby accelerating its degradation and ultimately inhibiting the immune evasion of tumor cells. Yang et al (39) found that in melanoma cells, ITCH can promote the ubiquitination and degradation process of PD-L1. The small-molecule ITCH agonist AK087 can effectively reduce the accumulation of PD-L1 induced by MAPK inhibitors, thereby weakening the tumor immune evasion and acquired resistance to anti-PD-L1 treatment induced by MAPK inhibitors. Glycogen synthase kinase 3β (GSK3β), a serine/threonine kinase, has the function of catalyzing the phosphorylation of substrates. Moreover, the phosphorylation mediated by GSK3β usually promotes the recognition of substrates by E3 ubiquitin ligases (40). Li et al (41) showed that GSK3β can interact with the E3 ligase β-TrCP and PD-L1, prompting β-TrCP to ubiquitinate PD-L1 in a phosphorylation-dependent manner, thereby accelerating the degradation of PD-L1. In basal-like breast cancer, the EGF/EGFR signaling can stabilize PD-L1 by inhibiting the activity of GSK3β, thereby mediating tumor immune treatment resistance. The combination of the EGFR-targeted inhibitor gefitinib and anti-PD-L1 treatment markedly enhances the efficacy of tumor immunotherapy. In bladder cancer, the E3 ubiquitin ligase NEDD4 can target PD-L1 and promote its K48 ubiquitination. Fibroblast growth factor receptor 3 (FGFR3) can activate the enzymatic activity of NEDD4 by phosphorylating it, thereby promoting the degradation of PD-L1. Therefore, the use of FGFR3 inhibitors to treat bladder cancer may lead to the occurrence of immune evasion in bladder cancer (42).

K63 ubiquitination is a type of non-proteolytic ubiquitination, which is usually closely associated with positive regulatory processes such as the maintenance of protein stability, subcellular localization and functional activation (25). It has been shown that MIB2 can promote the K63 ubiquitination of PD-L1 and stabilize the expression of PD-L1. This mechanism drives the RAB8-mediated exocytosis, facilitating the transportation of PD-L1 from the trans-Golgi network to the plasma membrane and ultimately mediating tumor immune evasion (43). In non-small cell lung cancer, S-phase kinase-associated protein 2 (Skp2), as a key linker molecule between LKB1 and PD-L1, has been shown to be able to stabilize the expression level of PD-L1 by promoting the K63 ubiquitination of K136 and K280 residues on PD-L1. In addition, LKB1 can promote the expression of Skp2 and PD-L1. This series of actions ultimately mediates the phenomenon of immune evasion in non-small cell lung cancer (44).

Effect of PD-1 ubiquitination on tumor immune evasion and immunotherapy

Studies have shown that FBXO38 can promote the K48 ubiquitination of PD-1 and accelerate its degradation process. Exogenous interleukin (IL)-2 can enhance the transcriptional activity of signal transducer and activator of transcription 5, upregulate the expression level of FBXO38 and thereby enhance the anti-tumor ability of T cells (30). In gallbladder cancer (GBC), PTBP3 which is expressed at high levels can promote the production of the IL-18 splice variant ΔIL-18. ΔIL-18 can downregulate the transcriptional level of FBXO38. This mechanism inhibits the ubiquitination process of PD-1 mediated by FBXO38, ultimately promoting the immune treatment evasion phenomenon in GBC (45). Zhou et al (46) demonstrated that the E3 ubiquitin ligase KLHL22 can recognize PD-1 and promote its ubiquitination. This process reduces the expression level of PD-1 on the surface of breast cancer cells and ultimately enhances the immune function of T cells. Liu et al (47) demonstrated that CDK1 can promote the nuclear translocation of PD-1 by enhancing the phosphorylation of PD-1 at Ser296. This mechanism facilitates the interaction between the E3 ubiquitin ligase F-box and WD repeat domain-containing 7 in the nucleus and PD-1, thereby mediating the ubiquitination and degradation process of PD-1 in non-small cell lung cancer and ultimately enhancing the anti-tumor ability of T cells. In colorectal cancer, c-Cbl binds to and interacts with PD-1, leading to its degradation via the ubiquitin-proteasome pathway. This reduces PD-1 expression levels, enhances the anti-tumor activity of T cells and promotes immunotherapy efficacy (48).

Mechanistic role of PD-1/PD-L1 deubiquitination in tumor immune evasion and immunotherapy

Regulation of cellular functions by DUBs through modulation of protein metabolism

DUBs can regulate the metabolic level of substrate proteins by cleaving monoubiquitin or polyubiquitin molecules, thereby modulating a variety of cellular activities, such as gene transcription, tumorigenesis and inflammatory immune responses (Fig. 3) (49).

The role of deubiquitination in the
treatment with anti-PD-1/PD-L1 mAb. In cancer cells,
deubiquitinating enzymes such as CSN5, OTUB1, OTUB2, TRAF6, USP2,
USP7, USP8 and USP22 exert their deubiquitination functions to
regulate the expression level of PD-L1, thereby promoting tumor
immune evasion. In immune cells, the deubiquitinating enzyme USP5
exerts its deubiquitination function to regulate the expression
level of PD-1, thereby promoting tumor immune evasion. PD-1,
programmed death receptor 1; PD-L1, programmed cell death ligand 1;
mAb, monoclonal antibodies; CSN5, COP9 signalosome subunit 5;
OTUB1, OTU deubiquitinase, ubiquitin aldehyde binding 1; OTUB2, OTU
deubiquitinase, ubiquitin aldehyde binding 2; TRAF6, TNF
receptor-associated factor 6; USP2, ubiquitin-specific peptidase 2;
USP7, ubiquitin-specific peptidase 7; USP8, ubiquitin-specific
peptidase 8; USP22, ubiquitin-specific peptidase 22; USP5,
ubiquitin-specific peptidase 5; ANXA1, Annexin A1; ERK,
extracellular signal-regulated kinase; EZH2, enhancer of zeste
homolog 2; FXR1, fragile × mental retardation syndrome-related
protein 1; METTL3, methyltransferase-like 3; PKP3, plakophilin-3;
TNF-α, tumor necrosis factor-alpha; TNFR, TNF receptor; YTHDC1, YTH
domain-containing protein 1; K48, ubiquitin lysine 48-linked;

Figure 3.

The role of deubiquitination in the treatment with anti-PD-1/PD-L1 mAb. In cancer cells, deubiquitinating enzymes such as CSN5, OTUB1, OTUB2, TRAF6, USP2, USP7, USP8 and USP22 exert their deubiquitination functions to regulate the expression level of PD-L1, thereby promoting tumor immune evasion. In immune cells, the deubiquitinating enzyme USP5 exerts its deubiquitination function to regulate the expression level of PD-1, thereby promoting tumor immune evasion. PD-1, programmed death receptor 1; PD-L1, programmed cell death ligand 1; mAb, monoclonal antibodies; CSN5, COP9 signalosome subunit 5; OTUB1, OTU deubiquitinase, ubiquitin aldehyde binding 1; OTUB2, OTU deubiquitinase, ubiquitin aldehyde binding 2; TRAF6, TNF receptor-associated factor 6; USP2, ubiquitin-specific peptidase 2; USP7, ubiquitin-specific peptidase 7; USP8, ubiquitin-specific peptidase 8; USP22, ubiquitin-specific peptidase 22; USP5, ubiquitin-specific peptidase 5; ANXA1, Annexin A1; ERK, extracellular signal-regulated kinase; EZH2, enhancer of zeste homolog 2; FXR1, fragile × mental retardation syndrome-related protein 1; METTL3, methyltransferase-like 3; PKP3, plakophilin-3; TNF-α, tumor necrosis factor-alpha; TNFR, TNF receptor; YTHDC1, YTH domain-containing protein 1; K48, ubiquitin lysine 48-linked;

Effect of PD-L1 deubiquitination on tumor immune evasion and immunotherapy

The ubiquitin-specific proteases (USP) family is a group of enzymes that specifically participate in protein deubiquitination modification and belongs to the DUB family. Studies have shown that the USP family plays a crucial role in regulating the deubiquitination of PD-L1 to mediate tumor immune evasion (50–52). Wang et al (51) found that that USP7 can directly target PD-L1 and deubiquitinate it, thereby stabilizing the expression level of the PD-L1 protein and ultimately promoting the process of immune evasion in gastric cancer. Another study showed that USP8 can directly bind to PD-L1 and remove its ubiquitination modification, thereby stabilizing the protein expression level of PD-L1 and ultimately promoting the process of immune evasion in pancreatic cancer (52). USP8 can not only directly mediate the deubiquitination of PD-L1, but has also been proved to stabilize the expression of TRAF6 by deubiquitinating TRAF6 in various types of cancer. Once TRAF6 is stably expressed, it will further promote the K63 ubiquitination of PD-L1 mediated by itself, ultimately stabilizing PD-L1 and promoting tumor immune evasion. The inhibition of USP8 by DUBs-IN-2 can effectively enhance the antitumor activity of T cells (53). In colorectal cancer and prostate cancer cells, USP2 can directly interact with PD-L1 and promote the K48 deubiquitination of PD-L1. This process stabilizes the expression of PD-L1 and then mediates tumor immune evasion (54). In liver cancer, USP22 can deubiquitinate PD-L1 and thereby mediate the antitumor immune resistance in liver cancer (55). In colorectal cancer, the inhibition of enhancer of zeste homolog 2 upregulates the expression of USP22 at the transcriptional level. The upregulated USP22 further deubiquitinates and stabilizes PD-L1, ultimately promoting the process of tumor immune evasion (56). In breast cancer, lung cancer and melanoma, the derivative peptide A11 of annexin A1 can competitively bind to PD-L1 with USP7, which is a deubiquitinase of PD-L1. This process inhibits the deubiquitination process of PD-L1 mediated by USP7, thereby promoting the degradation of PD-L1 and ultimately leading to the phenomenon of immune treatment resistance in tumors (57).

The OTUB family is a part of the DUB family. Studies have shown that the OTUB family also plays an important role in mediating the ubiquitination of PD-L1 (58–60). Zhu et al (59) found that in various types of cancer, OTUB1 can directly bind to PD-L1 and remove its K48 ubiquitination modification. This process inhibits the degradation of PD-L1 through the endoplasmic reticulum-associated degradation pathway, ultimately promoting tumor immune evasion. In addition, in non-small cell lung cancer, METTL3 mediates m6A modification in a YTHDC1-dependent manner, thereby promoting the circularization of circIGF2BP3. Acting as a molecular sponge for microRNA (miR)-328-3p and miR-3173-5p, circIGF2BP3 upregulates the expression of PKP3. Subsequently, PKP3 stabilizes OTUB1 mRNA through fragile × mental retardation syndrome-related protein 1 and ultimately, through the OTUB1-mediated deubiquitination of PD-L1, inhibits the function of CD8+ T cells (61). OTUB2 has also been proven to be able to directly interact with PD-L1 and deubiquitinate it. This process inhibits the ubiquitination and degradation of PD-L1 in the endoplasmic reticulum. The OTUB2 inhibitor OTUB2-IN-1 can effectively interfere with the DUB activity of OTUB2, thereby markedly enhancing the antitumor immune effect (60). COP9 signalosome 5 (CSN5), as a subunit of the COP9 signalosome, possesses DUB activity and can remove ubiquitin chains from substrate proteins, thereby preventing substrate proteins from being degraded by the proteasome. In triple-negative breast cancer (TNBC), tumor necrosis factor-α upregulates the expression level and activity of CSN5 through the nuclear factor-κB signaling pathway, thereby promoting the deubiquitination process of PD-L1 mediated by CSN5 and ultimately enhancing the resistance of cancer cells to PD-1/PD-L1 immunotherapy. The CSN5-targeting inhibitor curcumin can promote the degradation of PD-L1 and thus enhance the efficacy of tumor immunotherapy (62).

Effect of PD-1 deubiquitination on tumor immune evasion and immunotherapy

Xiao et al (63) found that in T cells, USP5 can interact with PD-1, deubiquitinate it, thereby stabilizing the expression level of PD-1 and enhance the tumor's immune evasion ability. Meanwhile, the extracellular signal-regulated kinase (ERK) can further promote the deubiquitination process of PD-1 mediated by USP5 by phosphorylating the Thr234 site of PD-1. The combination of the USP5 inhibitor and the ERK inhibitor trametinib can effectively enhance the antitumor immune effect.

Potential strategies to improve the efficacy of tumor immune checkpoint inhibitors by targeting the UPS

The UPS is the core mechanism for maintaining protein homeostasis within cells. During tumor immune evasion, UPS mainly regulates the dynamic balance of immune checkpoint molecules, antigen presentation processes and immunosuppressive cells through ubiquitination tagging and proteasomal degradation (64). Studies have confirmed that small-molecule drugs targeting the UPS play an important role in improving the therapeutic effects of tumor immune checkpoint inhibitors (Table I) (51,52,60). In pancreatic cancer, DUB-IN-2 inhibits the deubiquitination function of USP8, reduces the expression level of PD-L1 and thereby reverses the immunosuppressive state in the tumor microenvironment (52). In gastric cancer, the use of the small-molecule inhibitors Almac4 and P5091 targeting USP7 can inhibit the deubiquitination activity of USP7, thereby suppressing the proliferation of cancer cells. In addition, these two inhibitors can also downregulate the expression level of PD-L1 and enhance the anti-tumor immune response (51). In melanoma and colorectal cancer, the small-molecule inhibitor OTUB2-IN-1 can notably inhibit the DUB activity of OTUB2 and reduce the expression level of PD-L1 in tumor cells in a dose-dependent manner, thereby promoting antitumor immune function (60).

Potential drugs with the potential to
target the UPS for enhancing the efficacy of tumor immune
checkpoint inhibitors.

Table I.

Potential drugs with the potential to target the UPS for enhancing the efficacy of tumor immune checkpoint inhibitors.

Recent studies have further revealed that specific small-molecule compounds can modulate the stability of PD-L1 through ubiquitination pathways. In non-small cell lung cancer, compound #25 can enhance the anti-tumor immune response by inhibiting the Skp2-mediated K63 ubiquitination process of PD-L1 (44). The small-molecule agonist AK087 of ITCH can effectively promote the ubiquitination and degradation process of PD-L1 mediated by ITCH and notably inhibit the resistance of tumors to PD-1/PD-L1 treatment (39).

In addition, some small-molecule drugs have shown great potential in the field of tumor immunotherapy. OTUB1/USP8-IN-1 is a dual inhibitor targeting OTUB1 and USP8, which can effectively inhibit the functions of these two DUBs and is a potential small-molecule drug for tumor immunotherapy (65). Natural compounds also play a marked role in the field of tumor immunotherapy. Lu et al (66) found that the natural compound gentiopicroside can inhibit the DUB activity of USP22, thereby reducing the expression level of PD-L1 in lung adenocarcinoma and enhancing the body's anti-tumor immune ability. Curcumin, as a natural dietary supplement, has been proven to have the potential for anti-tumor immunity. In TNBC, curcumin can inhibit the DUB activity of CSN5, reduce the stability of PD-L1, induce the ubiquitination and degradation of PD-L1 and thereby enhance the immune system's ability to attack tumor cells (62). In lung cancer, CSN5 can promote the deubiquitination process of PD-L1, thereby inducing tumor immune evasion. The natural compound berberine can inhibit the DUB activity of CSN5, reduce the expression level of PD-L1 and thus enhance the body's anti-tumor immune ability (67). In colorectal and lung cancers, demethylzeylasteral specifically binds to USP22 and induces its degradation, thereby promoting ubiquitin-dependent proteasomal degradation of PD-L1 and ultimately enhancing T cell-mediated antitumor immune responses (68).

Small-molecule inhibitors targeting the UPS have shown great potential in the field of tumor immunotherapy and are very likely to be one of the important strategies for cancer treatment in the future.

Discussion

During the occurrence and development of tumors, tumor cells can evade the strict surveillance of the immune system through a variety of complex mechanisms. Among these mechanisms, the activation of immune checkpoint pathways is one of the core mechanisms by which tumors achieve immune evasion. At present, anti-PD-1/PD-L1 therapy, as a representative of immune checkpoint inhibitor therapy, is one of the most widely used tumor immunotherapy strategies in clinical practice (5). Although anti-PD-1/PD-L1 therapy has shown good efficacy in some patients, a considerable number of patients still do not respond to the treatment after receiving it and even develop acquired resistance due to anti-PD-1/PD-L1 therapy (69). Therefore, elucidating further the potential molecular mechanisms underlying tumor resistance to anti-PD-1/PD-L1 and exploring effective combination therapy strategies are of great significance for improving the therapeutic efficacy of anti-PD-1/PD-L1 and prolonging the survival of patients (Table II).

Table II.

Effect of key UPS regulators on programmed PD-1/PD-L1 expression in cancer immunotherapeutic responses.

Table II.

Effect of key UPS regulators on programmed PD-1/PD-L1 expression in cancer immunotherapeutic responses.

First author/s, yearEnzyme type CancerEffect on immunotherapy(Refs.)
Meng et al, 2018E3 ubiquitin ligaseFBXO38MelanomaIL-2 activates STAT5 to transcribe FBXO38, ubiquitinates PD-L1 and inhibits immune evasion.(30)
Zhou XA, 2020 KLHL22Breast cancerPromotes PD-1 ubiquitination and inhibits immune escape.(46)
Liu et al, 2022 FBW7Non-small cell lung cancerCDK1 promotes PD-1 nuclear translocation and enhances FBW7-mediated PD-1 ubiquitination and inhibits immune escape.(47)
Zhao et al, 2024 FBXO38Gallbladder cancerΔIL-18 downregulates FBXO38, suppresses PD-1 ubiquitination and promotes immune escape.(45)
Lyle et al, 2019 c-CblColorectal cancerPromotes ubiquitination of PD-1 and inhibits immune escape.(48)
Zhang et al, 2021 SPOPColorectal cancerALDH2 competitively binds with SPOP, inhibits PD-L1 ubiquitination and promotes immune escape.(31)
Yu et al, 2024 SPOPHepatocellular cancerBCLAF1 binds to SPOP, inhibits PD-L1 ubiquitination and promotes immune escape.(32)
Ding et al, 2023 SPOPNon-small cell lung cancerSGLT2 competitively binds PD-L1 with SPOP, inhibits PD-L1 ubiquitination and promotes immune escape.(34)
Zhang et al, 2018 SPOPPan-cancerCDK4 promotes SPOP phosphorylation, stabilizes SPOP expression, enhances K48-linked ubiquitination of PD-L1 and suppresses immune escape.(33)
Sun et al, 2023 TRIM21Non-small cell lung cancerLINC02418 functions as a molecular sponge to form a ternary complex with TRIM21 and PD-L1, promoting PD-L1 ubiquitination and suppressing immune escape.(35)
Gao et al, 2021 TRIM21Non-small cell lung cancerPromotes PD-L1 ubiquitination and inhibits immune evasion(36)
Wu et al, 2021 ARIH1Pan-cancerGSK3α phosphorylates PD-L1, promotes ARIH1-mediated ubiquitination of PD-L1 and inhibits immune evasion.(37)
Yu et al, 2023 MIB2Pan-cancerPromotes K63-linked ubiquitination of PD-L1 to facilitate immune evasion.(43)
Lv et al, 2024 Skp2Non-small cell lung cancerPromotes K63-linked ubiquitination of PD-L1 to facilitate immune evasion(44)
Jing et al, 2022 NEDD4Gallbladder cancerPromotes K48-linked ubiquitination of PD-L1 to inhibit immune evasion.(42)
Li et al, 2016 β-TrcpBreast cancerPromotes PD-L1 ubiquitination to inhibit immune evasion.(41)
Yang et al, 2022 ITCHMelanomaPromotes PD-L1 ubiquitination to inhibit immune evasion.(39)
Wei et al, 2022 RNF125Breast cancerPromotes K48-linked ubiquitination of PD-L1 to inhibit immune evasion.(38)
Xiao et al, 2023DUBsUSP5Breast cancerPromotes PD-1 deubiquitination to facilitate immune evasion.(63)
Yang et al, 2023 USP8Pancreatic cancerPromotes PD-L1 deubiquitination by inhibiting USP8 with DUBs-IN-2, thereby suppressing immune evasion.(52)
Wang et al, 2021 USP7Gastric cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(51)
Yu et al, 2023 USP7Pan-cancerA11 competitively binds PD-L1 with USP7, inhibits PD-L1 deubiquitination and suppresses immune evasion.(57)
Huang et al, 2024 USP22Colorectal cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(56)
Liu et al, 2021 OTUB1Non-small cell lung cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(61)
Lim et al, 2016 CSN5Breast cancerTNF-α upregulates CSN5 expression and activity via the NF-κB signaling pathway, promotes PD-L1 deubiquitination and facilitates immune evasion.(62)
Zhu et al, 2021 OTUB1Pan-cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(59)
Ren et al, 2024 OTUB2Pan-cancerPromotes PD-L1 deubiquitination and enhances anti-tumor efficacy using OTUB2-IN-1.(60)
Huang et al, 2019 USP22Hepatocellular cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(55)
Kuang et al, 2023 USP2Pan-cancerPromotes PD-L1 deubiquitination to facilitate immune evasion.(54)
Xiong et al, 2022 TRAF6Pan-cancerPromotes K63-linked ubiquitination of PD-L1 to facilitate immune evasion.(53)

[i] ALDH2, aldehyde dehydrogenase 2; ARIH1, ariadne RBR E3 ubiquitin protein ligase 1; BBR, berberine; BCLAF1, BCL2-associated transcription factor 1; β-Trcp, beta-transducin repeat-containing protein; c-Cbl, Casitas B lymphoma; CDH1, cadherin 1; CDK1, cyclin-dependent kinase 1; CDK4, cyclin-dependent kinase 4; CSN5, COP9 signalosome subunit 5; DUBs, deubiquitinating enzymes; protein 4; FBW7, F-box and WD repeat domain-containing 7; FBXO38, F-box protein 38; GSK3α, glycogen synthase kinase 3 alpha; IL-2, interleukin-2; IL-18, interleukin-18; ITCH, itchy E3 ubiquitin protein ligase; K48, Ubiquitin Lysine 48-linked; K63, Ubiquitin Lysine 63-linked; KLHL22, kelch-like protein 22; MIB2, mind bomb E3 ubiquitin protein ligase 2; NEDD4, neural precursor cell expressed developmentally downregulated 4; OTUB1, OTU deubiquitinase, ubiquitin aldehyde binding 1; OTUB2, OTU deubiquitinase, ubiquitin aldehyde binding 2; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; RNF125, ring finger protein 125; SGLT2, sodium-glucose cotransporter 2; Skp2, S-phase kinase-associated protein 2; STAT5, signal transducer and activator of transcription 5; SPOP, speckle-type POZ protein; TNF-α, tumor necrosis factor-alpha; TRAF6, TNF receptor-associated factor 6; Trim21, tripartite motif-containing protein 21; USP2, ubiquitin-specific peptidase 2; USP5, ubiquitin-specific peptidase 5; USP7, ubiquitin-specific peptidase 7; USP8, ubiquitin-specific peptidase 8; USP22, ubiquitin-specific peptidase 22;

The UPS, as a crucial molecular mechanism responsible for protein degradation and stabilization within cells, plays a pivotal role in regulating various biological processes such as cell cycle progression, signal transduction networks and immune response reactions. It is one of the main pathways mediating protein degradation or stabilization inside cells (70,71). The UPS has the ability to precisely recognize and selectively tag damaged, abnormal, or function-completed proteins with ubiquitin ‘tags’. Subsequently, the proteasome recognizes these tagged proteins and precisely regulates the degradation process of the proteins (19). In recent years, with the continuous deepening of research, an increasing number of studies have shown that the UPS plays a crucial role in the pathological and physiological processes of tumor proliferation, invasion, metastasis, immune regulation and drug resistance (72–74). It has been confirmed that the UPS can mediate tumor immune evasion and drug resistance induced by immune checkpoint inhibitor therapy by regulating the ubiquitination level of PD-1/PD-L1 (31,51,52). In-depth exploration of the potential molecular mechanisms by which the UPS regulates PD-1/PD-L1 will not only help improve the efficacy of tumor immunotherapy, but also provide a solid theoretical basis for the development of novel therapeutic strategies.

In recent years, targeting the UPS for disease treatment has become an important direction in drug development, showing great therapeutic potential in a number of fields such as autoimmune diseases and neurodegenerative diseases and cancer (75,76). Studies have shown that targeting the ubiquitination process of PD-1/PD-L1 mediated by the UPS has achieved notable effects in inhibiting tumor immune evasion and improving tumor resistance to anti-PD-1/PD-L1 therapy. For example, the small-molecule agonist AK087 of ITCH can effectively promote the ubiquitination and degradation of PD-L1 mediated by ITCH and markedly inhibit the resistance of tumors to anti-PD-1/PD-L1 treatment (39). The USP8 inhibitor DUB-IN-2 can effectively inhibit the deubiquitination process of PD-L1 mediated by USP8. This action notably inhibits tumor immune evasion and can effectively improve the efficacy of tumor immunotherapy (53). The small-molecule compound ML364 directly binds to USP2 and inhibits its deubiquitinase activity (77). The UPS not only determines the protein stability of PD-L1, but also forms a ‘positive-negative feedback’ loop with the IFN-γ/JAK-STAT signaling pathway through key nodes including TRIM25/SOCS1/USP18. Targeting this regulatory loop can amplify or suppress IFN-γ signaling across different cancer types, thereby guiding combination strategies between UPS inhibitors and immune checkpoint inhibitors (78,79).

This therapeutic model targeting the UPS has a unique mechanism of action. Instead of directly blocking protein functions, it regulates the stability and degradation of proteins from the source, thereby affecting the levels of abnormal proteins. Meanwhile, this therapeutic model has precise targeting ability, which can reduce the biological toxicity caused by broad-spectrum inhibition. Due to tissue specificity, the functions of E3 ubiquitin ligases also vary. The ‘functional switch’ of the same E3 ubiquitin ligase in different cancers is collectively determined by protein expression in the tumor microenvironment, signaling pathways and post-translational modifications. For instance, the mutational inactivation and dysregulated expression of SPOP in various types of cancer can affect its E3 ubiquitin ligase activity. Notably, Skp2-mediated PD-L1 K63 ubiquitination in non-small cell lung cancer depends on LKB1 inactivation (Fig. 2), whereas BRAF inhibitors in melanoma suppress PD-L1 ubiquitination by inhibiting the ERK-GSK3β-β-TrCP axis (41,80). This explains the reason Skp2 inhibitors (compound #25) can enhance the efficacy of PD-1 antibodies in non-small cell lung cancer, while melanoma requires combined MAPK inhibition to relieve β-TrCP suppression. For example, preclinical data on the USP7 inhibitor P5091 in gastric cancer showed that H. pylori-positive patients (with concomitant USP7 overexpression) had a 3.2-fold higher response rate compared with negative patients (P<0.01), suggesting that future trials should stratify patients based on microbiome-UPS co-mutation status (51). By contrast, a phase II trial of the CSN5 inhibitor curcumin in TNBC (trial no. NCT00094445) demonstrated limited efficacy due to the lack of screening for CSN5-high populations, highlighting the necessity of biomarker-guided therapy. Although UPS-targeted drugs such as USP8 inhibitors may develop resistance due to mutations or compensatory pathways, current evidence suggests that such resistance mechanisms are independent of PD-1/PD-L1 ubiquitination regulation. Future studies should explore whether resistance to UPS-targeted drugs upregulates PD-L1 through non-UPS pathways such as through transcriptional reprogramming or exosome release, thereby indirectly leading to immunotherapy failure. It is recommend that future studies employ cancer-specific organoid models to validate UPS-targeting drugs, thereby mimicking the effect of stromal cells on ubiquitination regulation within the tumor microenvironment.

Although some molecular drugs targeting the UPS have achieved preliminary progress in improving tumor immunotherapy, the number of those in clinical verification stage is still limited. As of November 2024, <20 UPS-targeting agents have entered clinical stages globally, with most concentrated in the proteolysis-targeting chimera (PROTAC). Notably, UPS modulators specifically targeting PD-1/PD-L1 remain in Phase I or earlier development (81). High expression of E3 ubiquitin ligases such as CRBN and VHL in the liver and kidneys often leads to hematological and renal toxicity, Excessive PROTACs may form nonfunctional binary complexes, disrupting UPS activity and causing drug ‘rebound effects’ (82). Dong et al (83) encapsulated VPS18/11 inhibitors such as RD-N into lung-targeted nanoparticles, effectively reversing tumor resistance and suppressing metastasis. Similar strategies could enable tissue-specific delivery of deubiquitinase inhibitors, minimizing off-target effects and systemic toxicity. UPS gene mutations or compensatory pathway activation can result in adaptive resistance during long-term treatment (84). Real-time drug concentration tracking, combined with artificial intelligence and machine learning-based predictive models, may optimize pharmacokinetic profiles and address long-term adaptive resistance.

The PROTAC technology, as an emerging therapeutic strategy for targeted protein degradation in recent years, has gradually attracted widespread attention (85,86). The PROTAC consists of three parts: i) A target protein-binding ligand; ii) an E3 ligase ligand; and iii) a linker. Its main mechanism of action is to induce the binding of the E3 ligase to the target protein, mediate the ubiquitination of the target protein and subsequently promote the degradation of the target protein (87). This technology has overcome the limitation of the ‘undruggable’ status of certain proteins and can reversibly regulate the expression levels of proteins over time. However, PROTAC drugs usually have a relatively large molecular weight, which leads to limited bioavailability. Therefore, the drug design of PROTAC still needs further optimization.

Conventional PROTACs suffer from high molecular weight, poor membrane permeability and low oral bioavailability, leading to weak in vitro-in vivo association and limited clinical translation. To overcome these hurdles, Sun et al (88) developed tumor microenvironment (TME)-responsive enzyme-activated click-forming PROTACs (ENCTACs). By exploiting cathepsin B overexpressed in >90% of solid tumors as a biological trigger, an orthogonal cleavage-click reaction assembles the active degrader in situ, selectively eliminating the epigenetic regulator BRD4 and consequently downregulating PD-L1 to remodel the immune microenvironment. In the 4T1 TNBC mouse model, ENCTACs achieved a 65% tumor-growth inhibition, a three-fold deeper tissue penetration and negligible systemic toxicity compared with traditional PROTACs. In parallel, a recent study conjugated a CD47 antibody with a folate ligand to create a Folate Receptor Targeting Chimera (FRTAC). Leveraging the high folate-receptor expression on cancer cells, FRTAC drives CD47 into lysosomal degradation via receptor-mediated endocytosis, markedly potentiating macrophage-mediated phagocytosis while sparing normal tissues (89). At present, multiple PROTAC drugs for tumor treatment have entered the clinical stage, such as ARV-110 for prostate cancer (90) and ARV-471 for ER+ breast cancer (91). Looking ahead, next-generation PROTAC platforms that integrate TME-specific activation with nanoparticle-based delivery are poised to surmount the dual barriers of cellular permeability and off-target toxicity, offering unprecedented opportunities for cancer immunotherapy.

In conclusion, the present review discussed the potential molecular mechanisms by which the UPS plays a role in tumor immune evasion and resistance to anti-PD-1/PD-L1 therapy and has summarized the potential targeted drugs that can inhibit tumor immune evasion and overcome resistance to immune checkpoint therapy by targeting the UPS. Future research should focus on the following key aspects: i) Further in-depth exploration of the regulatory mechanisms of the UPS on various immune checkpoints in different tumor types and immune microenvironments; ii) optimization of the design of targeted UPS drugs to improve their targeting ability and bioavailability; and iii) development of combined treatment regimens of immune checkpoint inhibitors and targeted UPS drugs to enhance the synergistic therapeutic effect. These research directions will lay a solid theoretical foundation for the development of the next-generation precision immunotherapy strategies.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82373124 and 81872163), the Shandong Provincial Natural Science Foundation (grant no. ZR2023MH073) and the research startup fund of Shaoxing People's Hospital (grant nos. PI202501 and YJ202402).

Availability of data and materials

Not applicable.

Authors' contributions

LHG and WLD were involved in conceptualization. LHG, AG, YYD, XJW, HXZ and WLD performed the literature search, data collection and writing. WLD and BGZ reviewed and edited the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

19S RP

19S regulatory particle

20S CP

20S core particle

ALDH2

aldehyde dehydrogenase 2

ARIH1

ariadne RBR E3 ubiquitin protein ligase 1

BCLAF1

BCL2-associated transcription factor 1

β-Trcp

beta-transducin repeat-containing protein

CAR-T

chimeric antigen receptor T

c-Cbl

Casitas B lymphoma

CDK4

cyclin-dependent kinase 4

CDK5

cyclin-dependent kinase 5

DUBs

deubiquitinating enzymes

CTLA-4

cytotoxic T-lymphocyte-associated protein 4

EGF

epidermal growth factor

EGFR

epidermal growth factor receptor

ERK

extracellular signal-regulated kinase

FBXO38

F-box protein 38

FGFR3

fibroblast growth factor receptor 3

GSK3α

glycogen synthase kinase 3 alpha

GSK3β

glycogen synthase kinase 3 beta

ITCH

itchy E3 ubiquitin protein ligase

ITIM

immunoreceptor tyrosine-based inhibitory motif

ITSM

immunoreceptor tyrosine-based activation motif

KLHL22

kelch-like protein 22

LKB1

liver kinase B1

mAb

monoclonal antibodies

METTL3

methyltransferase-like 3

MIB2

mind bomb E3 ubiquitin protein ligase 2

NEDD4

neural precursor cell expressed developmentally downregulated 4

NK cell

natural killer cell

OTUB1

OTU deubiquitinase, ubiquitin aldehyde binding 1

OTUB2

OTU deubiquitinase, ubiquitin aldehyde binding 2

PD-1

programmed cell death protein 1

PD-L1

programmed death-ligand 1

PKP3

plakophilin-3

PROTAC

proteolysis-targeting chimera

RAB8

RAS-related protein Rab-8

RNF125

ring finger protein 125

SGLT2

sodium-glucose cotransporter 2

SHP-1

Src homology 2 domain-containing protein tyrosine phosphatase-1

SHP-2

Src homology 2 domain-containing protein tyrosine phosphatase-2

SPOP

speckle-type POZ protein

TNBC

triple-negative breast cancer

TNF-α

tumor necrosis factor-alpha

TRAF6

TNF receptor-associated factor 6

Trim21

tripartite motif-containing protein 21

USP2

ubiquitin-specific peptidase 2

USP7

ubiquitin-specific peptidase 7

USP8

ubiquitin-specific peptidase 8

USP22

ubiquitin-specific peptidase 22

YTHDC1

YTH domain-containing protein 1

References

1 

Wang Q, Shao X, Zhang Y, Zhu M, Wang FXC, Mu J, Li J, Yao H and Chen K: Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 12:11149–11165. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Zhang Y and Zhang Z: The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 17:807–821. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N and Miller WH Jr: A review of cancer immunotherapy: From the past, to the present, to the future. Curr Oncol. 27:S87–S97. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Wang H, Kaur G, Sankin AI, Chen F, Guan F and Zang X: Immune checkpoint blockade and CAR-T cell therapy in hematologic malignancies. J Hematol Oncol. 12:592019. View Article : Google Scholar : PubMed/NCBI

5 

Byun DJ, Wolchok JD, Rosenberg LM and Girotra M: Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 13:195–207. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Naimi A, Mohammed RN, Raji A, Chupradit S, Yumashev AV, Suksatan W, Shalaby MN, Thangavelu L, Kamrava S, Shomali N, et al: Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun Signal. 20:442022. View Article : Google Scholar : PubMed/NCBI

7 

Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al: Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat Med. 8:793–800. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Gauen LK, Zhu Y, Letourneur F, Hu Q, Bolen JB, Matis LA, Klausner RD and Shaw AS: Interactions of p59fyn and ZAP-70 with T-cell receptor activation motifs: Defining the nature of a signalling motif. Mol Cell Biol. 14:3729–3741. 1994. View Article : Google Scholar : PubMed/NCBI

9 

Straus DB and Weiss A: Genetic evidence for the involvement of the lck tyrosine kinase in signal transduction through the T cell antigen receptor. Cell. 70:585–593. 1992. View Article : Google Scholar : PubMed/NCBI

10 

Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z and Cheng Q: Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res. 40:1842021. View Article : Google Scholar : PubMed/NCBI

11 

Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, Wu W, Han L and Wang S: The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front Immunol. 13:9644422022. View Article : Google Scholar : PubMed/NCBI

12 

Butte MJ, Keir ME, Phamduy TB, Sharpe AH and Freeman GJ: Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 27:111–122. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Paik J: Nivolumab plus relatlimab: First approval. Drugs. 82:925–931. 2022. View Article : Google Scholar : PubMed/NCBI

14 

Harrington KJ, Burtness B, Greil R, Soulières D, Tahara M, de Castro G Jr, Psyrri A, Brana I, Basté N, Bratland Å, et al: Pembrolizumab with or without chemotherapy in recurrent or metastatic head and neck squamous cell carcinoma: Updated results of the phase III KEYNOTE-048 study. J Clin Oncol. 41:790–802. 2023. View Article : Google Scholar : PubMed/NCBI

15 

Powles T, Park SH, Voog E, Caserta C, Valderrama BP, Gurney H, Kalofonos H, Radulović S, Demey W, Ullén A, et al: Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. 383:1218–1230. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Kiasari BA, Abbasi A, Darestani NG, Adabi N, Moradian A, Yazdani Y, Hosseini GS, Gholami N and Janati S: Combination therapy with nivolumab (anti-PD-1 monoclonal antibody): A new era in tumor immunotherapy. Int Immunopharmacol. 113:1093652022. View Article : Google Scholar : PubMed/NCBI

17 

Scheffner M, Werness BA, Huibregtse JM, Levine AJ and Howley PM: The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell. 63:1129–1136. 1990. View Article : Google Scholar : PubMed/NCBI

18 

Joazeiro CA, Wing SS, Huang H, Leverson JD, Hunter T and Liu YC: The tyrosine kinase negative regulator c-Cbl as a RING-type, E2-dependent ubiquitin-protein ligase. Science. 286:309–312. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Han D, Wang L, Jiang S and Yang Q: The ubiquitin-proteasome system in breast cancer. Trends Mol Med. 29:599–621. 2023. View Article : Google Scholar : PubMed/NCBI

20 

Eldridge AG and O'Brien T: Therapeutic strategies within the ubiquitin proteasome system. Cell Death Differ. 17:4–13. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Fang S and Weissman AM: A field guide to ubiquitylation. Cell Mol Life Sci. 61:1546–1561. 2004.PubMed/NCBI

22 

Pfoh R, Lacdao IK and Saridakis V: Deubiquitinases and the new therapeutic opportunities offered to cancer. Endocr Relat Cancer. 22:T35–T54. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Hochstrasser M: Ubiquitin-dependent protein degradation. Annu Rev Genet. 30:405–439. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Hofmann K and Falquet L: A ubiquitin-interacting motif conserved in components of the proteasomal and lysosomal protein degradation systems. Trends Biochem Sci. 26:347–350. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Park J, Cho J and Song EJ: Ubiquitin-proteasome system (UPS) as a target for anticancer treatment. Arch Pharm Res. 43:1144–1161. 2020. View Article : Google Scholar : PubMed/NCBI

26 

McKeon JE, Sha D, Li L and Chin LS: Parkin-mediated K63-polyubiquitination targets ubiquitin C-terminal hydrolase L1 for degradation by the autophagy-lysosome system. Cell Mol Life Sci. 72:1811–1824. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Pickart CM and Eddins MJ: Ubiquitin: Structures, functions, mechanisms. Biochim Biophys Acta. 1695:55–72. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Groll M, Ditzel L, Löwe J, Stock D, Bochtler M, Bartunik HD and Huber R: Structure of 20S proteasome from yeast at 2.4 A resolution. Nature. 386:463–471. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Bedford L, Paine S, Sheppard PW, Mayer RJ and Roelofs J: Assembly, structure, and function of the 26S proteasome. Trends Cell Biol. 20:391–401. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, Liu H, Bai Y, Xue M, Hu R, et al: FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature. 564:130–135. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Zhang H, Xia Y, Wang F, Luo M, Yang K, Liang S, An S, Wu S, Yang C, Chen D, et al: Aldehyde dehydrogenase 2 mediates alcohol-induced colorectal cancer immune escape through stabilizing PD-L1 expression. Adv Sci (Weinh). 8:20034042021. View Article : Google Scholar : PubMed/NCBI

32 

Yu Z, Wu X, Zhu J, Yan H, Li Y, Zhang H, Zhong Y, Lin M, Ye G, Li X, et al: BCLAF1 binds SPOP to stabilize PD-L1 and promotes the development and immune escape of hepatocellular carcinoma. Cell Mol Life Sci. 81:822024. View Article : Google Scholar : PubMed/NCBI

33 

Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, Tan Y, Ci Y, Wu F, Dai X, et al: Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature. 553:91–95. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Ding L, Chen X, Zhang W, Dai X, Guo H, Pan X, Xu Y, Feng J, Yuan M, Gao X, et al: Canagliflozin primes antitumor immunity by triggering PD-L1 degradation in endocytic recycling. J Clin Invest. 133:e1547542023. View Article : Google Scholar : PubMed/NCBI

35 

Sun Z, Mai H, Xue C, Fan Z, Li J, Chen H, Huo N, Kang X, Tang C, Fang L, et al: Hsa-LINC02418/mmu-4930573I07Rik regulated by METTL3 dictates anti-PD-L1 immunotherapeutic efficacy via enhancement of Trim21-mediated PD-L1 ubiquitination. J Immunother Cancer. 11:e0074152023. View Article : Google Scholar : PubMed/NCBI

36 

Gao L, Xia L, Ji W, Zhang Y, Xia W and Lu S: Knockdown of CDK5 down-regulates PD-L1 via the ubiquitination-proteasome pathway and improves antitumor immunity in lung adenocarcinoma. Transl Oncol. 14:1011482021. View Article : Google Scholar : PubMed/NCBI

37 

Wu Y, Zhang C, Liu X, He Z, Shan B, Zeng Q, Zhao Q, Zhu H, Liao H, Cen X, et al: ARIH1 signaling promotes anti-tumor immunity by targeting PD-L1 for proteasomal degradation. Nat Commun. 12:23462021. View Article : Google Scholar : PubMed/NCBI

38 

Wei M, Mo Y, Liu J, Zhai J, Li H, Xu Y, Peng Y, Tang Z, Wei T, Yang X, et al: Ubiquitin ligase RNF125 targets PD-L1 for ubiquitination and degradation. Front Oncol. 12:8356032022. View Article : Google Scholar : PubMed/NCBI

39 

Yang Z, Wang Y, Liu S, Deng W, Lomeli SH, Moriceau G, Wohlschlegel J, Piva M and Lo RS: Enhancing PD-L1 degradation by ITCH during MAPK inhibitor therapy suppresses acquired resistance. Cancer Discov. 12:1942–1959. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Doble BW and Woodgett JR: GSK-3: Tricks of the trade for a multi-tasking kinase. J Cell Sci. 116:1175–1186. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, Khoo KH, Chang SS, Cha JH, Kim T, et al: Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat Commun. 7:126322016. View Article : Google Scholar : PubMed/NCBI

42 

Jing W, Wang G, Cui Z, Xiong G, Jiang X, Li Y, Li W, Han B, Chen S and Shi B: FGFR3 destabilizes PD-L1 via NEDD4 to control T-cell-mediated bladder cancer immune surveillance. Cancer Res. 82:114–129. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Yu X, Li W, Liu H, Wang X, Coarfa C, Cheng C, Yu X, Zeng Z, Cao Y, Young KH and Li Y: PD-L1 translocation to the plasma membrane enables tumor immune evasion through MIB2 ubiquitination. J Clin Invest. 133:e1604562023. View Article : Google Scholar : PubMed/NCBI

44 

Lv L, Miao Q, Zhan S, Chen P, Liu W, Lv J, Yan W, Wang D, Liu H, Yin J, et al: LKB1 dictates sensitivity to immunotherapy through Skp2-mediated ubiquitination of PD-L1 protein in non-small cell lung cancer. J Immunother Cancer. 12:e0094442024. View Article : Google Scholar : PubMed/NCBI

45 

Zhao C, Zhao JW, Zhang YH, Zhu YD, Yang ZY, Liu SL, Tang QY, Yang Y, Wang HK, Shu YJ, et al: PTBP3 Mediates IL-18 exon skipping to promote immune escape in gallbladder cancer. Adv Sci (Weinh). 11:e24066332024. View Article : Google Scholar : PubMed/NCBI

46 

Zhou XA, Zhou J, Zhao L, Yu G, Zhan J, Shi C, Yuan R, Wang Y, Chen C, Zhang W, et al: KLHL22 maintains PD-1 homeostasis and prevents excessive T cell suppression. Proc Natl Acad Sci USA. 117:28239–28250. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Liu J, Wei L, Hu N, Wang D, Ni J, Zhang S, Liu H, Lv T, Yin J, Ye M and Song Y: FBW7-mediated ubiquitination and destruction of PD-1 protein primes sensitivity to anti-PD-1 immunotherapy in non-small cell lung cancer. J Immunother Cancer. 10:e0051162022. View Article : Google Scholar : PubMed/NCBI

48 

Lyle C, Richards S, Yasuda K, Napoleon MA, Walker J, Arinze N, Belghasem M, Vellard I, Yin W, Ravid JD, et al: c-Cbl targets PD-1 in immune cells for proteasomal degradation and modulates colorectal tumor growth. Sci Rep. 9:202572019. View Article : Google Scholar : PubMed/NCBI

49 

Clague MJ, Urbé S and Komander D: Breaking the chains: Deubiquitylating enzyme specificity begets function. Nat Rev Mol Cell Biol. 20:338–352. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Gao H, Yin J, Ji C, Yu X, Xue J, Guan X, Zhang S, Liu X and Xing F: Targeting ubiquitin specific proteases (USPs) in cancer immunotherapy: From basic research to preclinical application. J Exp Clin Cancer Res. 42:2252023. View Article : Google Scholar : PubMed/NCBI

51 

Wang Z, Kang W, Li O, Qi F, Wang J, You Y, He P, Suo Z, Zheng Y and Liu HM: Abrogation of USP7 is an alternative strategy to downregulate PD-L1 and sensitize gastric cancer cells to T cells killing. Acta Pharm Sin B. 11:694–707. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Yang H, Zhang X, Lao M, Sun K, He L, Xu J, Duan Y, Chen Y, Ying H, Li M, et al: Targeting ubiquitin-specific protease 8 sensitizes anti-programmed death-ligand 1 immunotherapy of pancreatic cancer. Cell Death Differ. 30:560–575. 2023. View Article : Google Scholar : PubMed/NCBI

53 

Xiong W, Gao X, Zhang T, Jiang B, Hu MM, Bu X, Gao Y, Zhang LZ, Xiao BL, He C, et al: USP8 inhibition reshapes an inflamed tumor microenvironment that potentiates the immunotherapy. Nat Commun. 13:17002022. View Article : Google Scholar : PubMed/NCBI

54 

Kuang Z, Liu X, Zhang N, Dong J, Sun C, Yin M, Wang Y, Liu L, Xiao D, Zhou X, et al: USP2 promotes tumor immune evasion via deubiquitination and stabilization of PD-L1. Cell Death Differ. 30:2249–2264. 2023. View Article : Google Scholar : PubMed/NCBI

55 

Huang X, Zhang Q, Lou Y, Wang J, Zhao X, Wang L, Zhang X, Li S, Zhao Y, Chen Q, et al: USP22 deubiquitinates CD274 to suppress anticancer immunity. Cancer Immunol Res. 7:1580–1590. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Huang J, Yin Q, Wang Y, Zhou X, Guo Y, Tang Y, Cheng R, Yu X, Zhang J, Huang C, et al: EZH2 inhibition enhances PD-L1 protein stability through USP22-mediated deubiquitination in colorectal cancer. Adv Sci (Weinh). 11:e23080452024. View Article : Google Scholar : PubMed/NCBI

57 

Yu ZZ, Liu YY, Zhu W, Xiao D, Huang W, Lu SS, Yi H, Zeng T, Feng XP, Yuan L, et al: ANXA1-derived peptide for targeting PD-L1 degradation inhibits tumor immune evasion in multiple cancers. J Immunother Cancer. 11:e0063452023. View Article : Google Scholar : PubMed/NCBI

58 

Sivakumar D, Kumar V, Naumann M and Stein M: Activation and selectivity of OTUB-1 and OTUB-2 deubiquitinylases. J Biol Chem. 295:6972–6982. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Zhu D, Xu R, Huang X, Tang Z, Tian Y, Zhang J and Zheng X: Deubiquitinating enzyme OTUB1 promotes cancer cell immunosuppression via preventing ER-associated degradation of immune checkpoint protein PD-L1. Cell Death Differ. 28:1773–1789. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Ren W, Xu Z, Chang Y, Ju F, Wu H, Liang Z, Zhao M, Wang N, Lin Y, Xu C, et al: Pharmaceutical targeting of OTUB2 sensitizes tumors to cytotoxic T cells via degradation of PD-L1. Nat Commun. 15:92024. View Article : Google Scholar : PubMed/NCBI

61 

Liu Z, Wang T, She Y, Wu K, Gu S, Li L, Dong C, Chen C and Zhou Y: N6-methyladenosine-modified circIGF2BP3 inhibits CD8(+) T-cell responses to facilitate tumor immune evasion by promoting the deubiquitination of PD-L1 in non-small cell lung cancer. Mol Cancer. 20:1052021. View Article : Google Scholar : PubMed/NCBI

62 

Lim SO, Li CW, Xia W, Cha JH, Chan LC, Wu Y, Chang SS, Lin WC, Hsu JM, Hsu YH, et al: Deubiquitination and stabilization of PD-L1 by CSN5. Cancer Cell. 30:925–939. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Xiao X, Shi J, He C, Bu X, Sun Y, Gao M, Xiang B, Xiong W, Dai P, Mao Q, et al: ERK and USP5 govern PD-1 homeostasis via deubiquitination to modulate tumor immunotherapy. Nat Commun. 14:28592023. View Article : Google Scholar : PubMed/NCBI

64 

Ding P, Ma Z, Fan Y, Feng Y, Shao C, Pan M, Zhang Y, Huang D, Han J, Hu Y and Yan X: Emerging role of ubiquitination/deubiquitination modification of PD-1/PD-L1 in cancer immunotherapy. Genes Dis. 10:848–863. 2023. View Article : Google Scholar : PubMed/NCBI

65 

Tan L, Shan H, Han C, Zhang Z, Shen J, Zhang X, Xiang H, Lu K, Qi C, Li Y, et al: Discovery of potent OTUB1/usp8 dual inhibitors targeting proteostasis in non-small-cell lung cancer. J Med Chem. 65:13645–13659. 2022. View Article : Google Scholar : PubMed/NCBI

66 

Lu W, Chu P, Tang A, Si L and Fang D: The secoiridoid glycoside Gentiopicroside is a USP22 inhibitor with potent antitumor immunotherapeutic activity. Biomed Pharmacother. 177:1169742024. View Article : Google Scholar : PubMed/NCBI

67 

Liu Y, Liu X, Zhang N, Yin M, Dong J, Zeng Q, Mao G, Song D, Liu L and Deng H: Berberine diminishes cancer cell PD-L1 expression and facilitates antitumor immunity via inhibiting the deubiquitination activity of CSN5. Acta Pharm Sin B. 10:2299–2312. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Zhang Y, Huang Y, Yu D, Xu M, Hu H, Zhang Q, Cai M, Geng X, Zhang H, Xia J, et al: Demethylzeylasteral induces PD-L1 ubiquitin-proteasome degradation and promotes antitumor immunity via targeting USP22. Acta Pharm Sin B. 14:4312–4328. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Denis M, Grasselly C, Choffour PA, Wierinckx A, Mathé D, Chettab K, Tourette A, Talhi N, Bourguignon A, Birzele F, et al: In vivo syngeneic tumor models with acquired resistance to anti-PD-1/PD-L1 therapies. Cancer Immunol Res. 10:1013–1027. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Laine A and Ronai Z: Ubiquitin chains in the ladder of MAPK signaling. Sci STKE. 26:re52005.PubMed/NCBI

71 

Çetin G, Klafack S, Studencka-Turski M, Krüger E and Ebstein F: The ubiquitin-proteasome system in immune cells. Biomolecules. 11:602021. View Article : Google Scholar : PubMed/NCBI

72 

Han M, Guo Y, Li Y, Zeng Q, Zhu W and Jiang J: SMURF2 facilitates ubiquitin-mediated degradation of ID2 to attenuate lung cancer cell proliferation. Int J Biol Sci. 19:3324–3340. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Cui H, Wang Q, Lei Z, Feng M, Zhao Z, Wang Y and Wei G: DTL promotes cancer progression by PDCD4 ubiquitin-dependent degradation. J Exp Clin Cancer Res. 38:3502019. View Article : Google Scholar : PubMed/NCBI

74 

Chen Y, Xian M, Ying W, Liu J, Bing S, Wang X, Yu J, Xu X, Xiang S, Shao X, et al: Succinate dehydrogenase deficiency-driven succinate accumulation induces drug resistance in acute myeloid leukemia via ubiquitin-cullin regulation. Nat Commun. 15:98202024. View Article : Google Scholar : PubMed/NCBI

75 

Reichelt J, Sachs W, Frömbling S, Fehlert J, Studencka-Turski M, Betz A, Loreth D, Blume L, Witt S, Pohl S, et al: Non-functional ubiquitin C-terminal hydrolase L1 drives podocyte injury through impairing proteasomes in autoimmune glomerulonephritis. Nat Commun. 14:21142023. View Article : Google Scholar : PubMed/NCBI

76 

Fuseya Y, Kadoba K, Liu X, Suetsugu H, Iwasaki T, Ohmura K, Sumida T, Kochi Y, Morinobu A, Terao C and Iwai K: Attenuation of HOIL-1L ligase activity promotes systemic autoimmune disorders by augmenting linear ubiquitin signaling. JCI Insight. 9:e1711082024. View Article : Google Scholar : PubMed/NCBI

77 

Yi J, Tavana O, Li H, Wang D, Baer RJ and Gu W: Targeting USP2 regulation of VPRBP-mediated degradation of p53 and PD-L1 for cancer therapy. Nat Commun. 14:19412023. View Article : Google Scholar : PubMed/NCBI

78 

Zhang XZ, Li FH and Wang XJ: Regulation of tripartite motif-containing proteins on immune response and viral evasion. Front Microbiol. 12:7948822021. View Article : Google Scholar : PubMed/NCBI

79 

Gao X, Cao Y, Li H, Yu F, Xi J, Zhang J, Zhuang R, Xu Y and Xu L: Mechanisms underlying altered ubiquitin-proteasome system activity during heart failure and pharmacological interventions. Eur J Med Chem. 292:1177252025. View Article : Google Scholar : PubMed/NCBI

80 

Yamaguchi H, Hsu JM, Yang WH and Hung MC: Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics. Nat Rev Clin Oncol. 19:287–305. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Zhong G, Chang X, Xie W and Zhou X: Targeted protein degradation: Advances in drug discovery and clinical practice. Signal Transduct Target Ther. 9:3082024. View Article : Google Scholar : PubMed/NCBI

82 

Wang C, Zhang Y, Chen W, Wu Y and Xing D: New-generation advanced PROTACs as potential therapeutic agents in cancer therapy. Mol Cancer. 23:1102024. View Article : Google Scholar : PubMed/NCBI

83 

Dong T, Niu H, Chu Z, Zhou C, Gao Y, Jia M, Sun B, Zheng X, Zhang W, Zhang J, et al: Targeting VPS18 hampers retromer trafficking of PD-L1 and augments immunotherapy. Sci Adv. 10:eadp49172024. View Article : Google Scholar : PubMed/NCBI

84 

Shende S, Rathored J and Budhbaware T: Role of metabolic transformation in cancer immunotherapy resistance: Molecular mechanisms and therapeutic implications. Discov Oncol. 16:4532025. View Article : Google Scholar : PubMed/NCBI

85 

Raina K, Lu J, Qian Y, Altieri M, Gordon D, Rossi AM, Wang J, Chen X, Dong H, Siu K, et al: PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci USA. 113:7124–7129. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Xiao M, Zhao J, Wang Q, Liu J and Ma L: Recent advances of degradation technologies based on PROTAC mechanism. Biomolecules. 12:12572022. View Article : Google Scholar : PubMed/NCBI

87 

Chen Y, Tandon I, Heelan W, Wang Y, Tang W and Hu Q: Proteolysis-targeting chimera (PROTAC) delivery system: Advancing protein degraders towards clinical translation. Chem Soc Rev. 51:5330–5350. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Sun C, Liu S, Lau JW, Yang H, Chen Y and Xing B: Enzyme-Activated orthogonal proteolysis chimeras for tumor microenvironment-responsive immunomodulation. Angew Chem Int Ed Engl. 64:e2024230572025. View Article : Google Scholar : PubMed/NCBI

89 

Zhou Y, Li C, Chen X, Zhao Y, Liao Y, Huang P, Wu W, Nieto NS, Li L and Tang W: Development of folate receptor targeting chimeras for cancer selective degradation of extracellular proteins. Nat Commun. 15:86952024. View Article : Google Scholar : PubMed/NCBI

90 

He Y, Zheng Y, Zhu C, Lei P, Yu J, Tang C, Chen H and Diao X: Radioactive ADME demonstrates ARV-110′s high druggability despite low oral bioavailability. J Med Chem. 67:14277–14291. 2024. View Article : Google Scholar : PubMed/NCBI

91 

Gough SM, Flanagan JJ, Teh J, Andreoli M, Rousseau E, Pannone M, Bookbinder M, Willard R, Davenport K, Bortolon E, et al: Oral estrogen receptor PROTAC vepdegestrant (ARV-471) is highly efficacious as monotherapy and in combination with CDK4/6 or PI3K/mTOR pathway inhibitors in preclinical ER+ breast cancer models. Clin Cancer Res. 30:3549–3563. 2024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Gu L, Guo A, Ding Y, Wang X, Zhang H, Duan W and Zhang B: Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review). Oncol Rep 54: 167, 2025.
APA
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., & Zhang, B. (2025). Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review). Oncology Reports, 54, 167. https://doi.org/10.3892/or.2025.9000
MLA
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., Zhang, B."Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review)". Oncology Reports 54.6 (2025): 167.
Chicago
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., Zhang, B."Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review)". Oncology Reports 54, no. 6 (2025): 167. https://doi.org/10.3892/or.2025.9000
Copy and paste a formatted citation
x
Spandidos Publications style
Gu L, Guo A, Ding Y, Wang X, Zhang H, Duan W and Zhang B: Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review). Oncol Rep 54: 167, 2025.
APA
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., & Zhang, B. (2025). Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review). Oncology Reports, 54, 167. https://doi.org/10.3892/or.2025.9000
MLA
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., Zhang, B."Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review)". Oncology Reports 54.6 (2025): 167.
Chicago
Gu, L., Guo, A., Ding, Y., Wang, X., Zhang, H., Duan, W., Zhang, B."Molecular mechanisms and potential targeting strategies of ubiquitin‑proteasome system‑mediated PD‑1/PD‑L1 ubiquitination in tumor immune suppression (Review)". Oncology Reports 54, no. 6 (2025): 167. https://doi.org/10.3892/or.2025.9000
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team