Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Oct 1.
Published in final edited form as: Semin Cancer Biol. 2021 Apr 5;85:246–252. doi: 10.1016/j.semcancer.2021.04.002

Post-translational regulations of PD-L1/PD-1: Mechanisms and opportunities for combined immunotherapy

Xiaoming Dai 1,3, Yang Gao 1,2,3, Wenyi Wei 1,#
PMCID: PMC8490479  NIHMSID: NIHMS1691686  PMID: 33831533

Abstract

Antibodies targeting programmed cell death protein 1 (PD-1) or its ligand programmed death-ligand 1 (PD-L1) are profoundly changing the methods to treat cancers with long-term clinical benefits. Unlike conventional methods that directly target tumor cells, PD-L1/PD-1 blockade exerts anti-tumor effects largely through reactivating or normalizing cytotoxic T lymphocyte in the tumor microenvironment to combat cancer cells. However, only a small fraction of cancer patients responds well to PD-L1/PD-1 blockade and clinical outcomes have reached a bottleneck without substantial advances. Therefore, better understanding the molecular mechanisms underlying how PD-L1/PD-1 expression is regulated will provide new insights to improve the efficacy of current anti-PD-L1/PD-1 therapy. Here, we provide an update of current progress of PD-L1 and PD-1 post-translational regulations and highlight the mechanism-based combination therapy strategies for a better treatment of human cancer.

Keywords: PD-L1, PD-1, Immunotherapy, Posttranslational modification, Ubiquitination, PROTAC, Glycosylation, Phosphorylation, Acetylation, Palmitoylation

Introduction

In the last few decades, immunotherapy has become an essential part for treating various types of human cancer. The development of immune checkpoint blockade such as antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programed death 1/programmed death-ligand 1 (PD-1/PD-L1) are fundamentally reshaping the landscape of cancer therapy [1, 2]. Multiple antibodies targeting CTLA-4 or PD-1/PD-L1 have been approved by FDA to treat different types of cancer with long-term therapeutic benefits, which was rarely observed when using conventional therapies [3, 4].

PD-1 was discovered by Honjo’s group in 1992 as an apoptosis-associated gene [5]. Later, PD-1 was identified as a co-inhibitory receptor expressed mainly on immune cells, with major biological functions involved in the inhibition of immune responses [6-8]. Accumulating evidence highlights the pivotal function of PD-1 and its physiological ligands PD-L1 and programmed death-ligand 2 (PD-L2) in tumor immune microenvironment as well as the opportunities for cancer therapy [3, 8, 9]. Given that PD-L1 is more widely expressed than PD-L2 in both normal and tumor cells, later studies mainly focus on exploring the immune suppressive functions and mechanisms of PD-1/PD-L1 as well as how aberrant overexpression of PD-L1 in tumor cells allows tumor cells to escape immune surveillances [7]. Mechanistically, PD-L1 expression on the surface of tumor cells binds to PD-1 receptors on T cells, blocking the T cell proliferation and cytokine production (Fig. 1) [7, 8]. As such, antibodies targeting PD-1/PD-L1 are effective for many types of tumors because they enhance the anti-tumor activity of cytotoxic T lymphocytes (CTLs) [1, 3, 8]. However, a large proportion of cancer patients do not respond well to PD-L1/PD-1 blockade and clinical outcomes have reached a plateau without substantial advances [10, 11]. Hence, more studies are warranted to identify optimal therapeutic strategies that might improve the efficacy of cancer immunotherapy.

Figure 1. Mechanism of PD-1/PD-L1 blockade.

Figure 1.

The binding of TCR and MHC activates adaptive immune response. PD-L1 expressed on tumor cells interacts with its physiological receptor, PD-1 on T cell surface, preventing the signaling transduction of T cells to inhibit the immune response. Anti-PD-1 or anti-PD-L1 antibodies block the interaction of PD-1 and PD-L1, and abolish the inhibition of CD8+ T cell, thus enhancing the antitumor activity. TCR, T cell receptor; MHC, major histocompatibility complex.

Although initial work focused on genetic, transcriptional and post-transcriptional regulations of the PD-1/PD-L1 pathway [12-14], a large number of studies suggest that PD-L1 and PD-1 are also regulated by protein post-translational modification, which provides additional opportunities to manipulate immune system to eradicate tumors. In this review, we mainly summarize recent progressions on PD-L1/PD-1 regulations at protein levels and further emphasize the molecular mechanisms and the implicated therapeutic opportunities of these modifications for enhancing the efficacy of cancer immunotherapy.

Post-translational modifications of PD-L1 and the therapeutic opportunities

1. Ubiquitination and PROTACs of PD-L1

The ubiquitin-proteasome system is a crucial mechanism for intracellular protein degradation, which plays important roles in various cellular processes including immunity, inflammation and cancer [15-17]. The target protein can be covalently attached with polyubiquitin chain through the sequential action of three enzymes involving an E1 activating, an E2 conjugating and an E3 ligase enzyme. The polyubiquitinated protein will be subsequently recognized and degraded by the 26S proteasome complex [16, 17]. PD-L1 has been shown to be regulated by different E3 ubiquitin ligases, including but not limited to β-transducin repeat-containing protein (β-TRCP), speckle-type POZ protein (SPOP), HMG-CoA reductase degradation protein 1 (HRD1), and STIP1 homology and U-Box containing protein 1 (STUB1) (Fig. 2A). Specifically, SCFβ-TRCP promotes PD-L1 polyubiquitination and degradation following GSK3β-mediated phosphorylation of PD-L1 at the T180 and S184 residues [18, 19]. Our group identified Cullin 3SPOP as a physiological E3 ubiquitin ligase promoting PD-L1 polyubiquitination and degradation in a cell cycle dependent manner [20]. Mechanistically, cyclin-D-CDK4 stabilizes SPOP in part through phosphorylating SPOP at the S6 residue, leading to the inhibition of APC/Cdh1-mediated SPOP degradation. Furthermore, the combination of CDK4/6 kinase inhibitor palbociclib with anti-PD-1 immunotherapy dramatically enhanced the therapeutic efficacy in part by enhancing tumor regression and improving overall survival rates [20]. PD-L1 can also be targeted by the E3 ligase HRD1 through the ER-associated degradation (ERAD) pathway [21]. The phosphorylation of PD-L1 by AMPK at S195 blocks its ER-to-Golgi translocation, leading to PD-L1 degradation by the ERAD system [21]. As such, combination of AMPK agonist metformin with CTLA-4 blockade significantly enhances T lymphocyte infiltration and suppresses tumor growth in syngeneic mouse models [21]. Furthermore, STUB1 has been shown to polyubiquitinate and down-regulate PD-L1, which can be blocked by MARVEL transmembrane domain containing 4/6 (CMTM4/6) [22, 23]. However, whether targeting STUB1 or CMTM4/6 will pave a way to strength the immunotherapy is important to investigate in the future.

Fig. 2. Regulations of PD-L1 by polyubiquitination, deubiquitination and PROTACs.

Fig. 2.

A. PD-L1 can be polyubiquitinated by E3 ubiquitin ligases β-TRCP, SPOP, STUB1 and HRD1 and deubiquitinated by deubiquitinases CSN5, USP22, OTUB1 and USP9X. B. PROTACs of PD-L1. AbTAC of PD-L1 is a bispecific antibody which can target both PD-L1 and the E3 ligase RNF43 to induce the lysosomal degradation of PD-L1. LYTAC of PD-L1 is composed of an antibody specific to PD-L1 conjugated to a synthetic oligoglycopeptide ligand that binds CI-M6PR, a transmembrane glycoprotein responsible for trafficking proteins to lysosomes for degradation. PROTAC of PD-L1 is a novel resorcinol diphenyl ether-based PROTAC molecule which consists of BMS1198, a linker region and Pomalidomide.

Proteolysis-targeting chimeras (PROTACs) have recently arisen as novel therapeutic modalities to target traditionally “undruggable” proteins by hijacking the endogenous ubiquitin-proteasome system (UPS) to specifically degrade the protein of interest (POI) [24, 25]. PROTACs are ternary chemical complexes that usually consist of three functional parts, an E3 ligase-recruiting chemical ligand, a POI-binding chemical ligand and a linker [25, 26]. Typically, von Hippel–Lindau disease tumor suppressor (VHL) and Cereblon (CRBN) are the most commonly used endogenous E3 ligases in the PROTAC field. Chen et al. synthesized P22, a novel resorcinol diphenyl ether-based PROTAC molecule targeting the PD-1/PD-L1 pathway (Fig. 2B) [27]. The resorcinol diphenyl ether-based PROTAC molecule P22 uses pomalidomide as the chemical ligand of the Cullin 4CRBN E3 ubiquitin ligase [27]. P22 on one hand can inhibit the PD-1/ PD-L1 interaction, on the other hand, P22 can moderately reduce the protein levels of PD-L1 likely in a lysosome-dependent manner [27]. Besides the traditional inhibitor based PROTAC of PD-L1, Cotton and colleagues developed first antibody-based PROTACs (AbTACs) inducing the degradation of PD-L1 [28]. AbTACs are recombinant bispecific antibodies that recruit membrane-bound E3 ligases for the degradation of cell-surface proteins based on E3 ligase RNF43, a single-pass transmembrane E3 ligase. Cotton et al. synthesized a bispecific antibody which can target both PD-L1 and the E3 ligase RNF43 to induce the lysosomal degradation of PD-L1 (Fig. 2B) [28]. Moreover, the Bertozzi laboratory developed lysosome-targeting chimeras, termed LYTACs, which are composed of an antibody specific to the targeted protein conjugated to a synthetic oligoglycopeptide ligand that binds the cation-independent mannose-6-phosphate receptor (CI-M6PR), a transmembrane glycoprotein responsible for trafficking proteins to lysosomes for degradation [29]. Using this platform, Banik et al. successfully targeted PD-L1 for lysosomal degradation (Fig. 2B) [29]. Although several PROTACs have been developed for targeting degradation, they are only validated in cells. Thus, how these PROTACs work in vivo to retard tumorigenesis warrant further investigation.

2. Deubiquitination of PD-L1

As ubiquitination is a reversible process, the ubiquitination of PD-L1 has been reported to be reversed by several deubiquitinating enzymes (DUBs), namely COP9 signalosome 5 (CSN5), Ubiquitin Specific Peptidase 22 (USP22), OTU domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) and Ubiquitin Specific Peptidase 9 X-Linked (USP9X) (Fig. 2A). Lim and colleagues identified CSN5 as a DUB for PD-L1 deubiquitination. CSN5 removes the poly-ubiquitin chain on PD-L1, leading to the stabilization of PD-L1. Moreover, CSN5 natural inhibitor curcumin [30] destabilizes PD-L1 and enhances the therapeutic efficacy of CTLA-4 blockade therapy [31]. Except for CSN5, USP22 also can directly deubiquitinate PD-L1 and inhibit its proteasome degradation [32, 33]. Interestingly, USP22 also regulates PD-L1 protein level through modulating the CSN5-PD-L1 axis. Mechanistically USP22 interacts with CSN5 and stabilizes CSN5 in part through deubiquitination, which further inhibits the degradation of PD-L1 in cells [33]. USP9X deubiquitinates and stabilizes PD-L1 in oral squamous cell carcinoma (OSCC) [34]. On the other hand, OTUB1 was identified as another DUB for PD-L1 [35]. Mechanistically, OTUB1 removes K48-linked ubiquitin chains from the PD-L1 and inhibits the degradation of PD-L1 through the ERAD pathway [35]. Although multiple DUBs have been identified for antagonizing PD-L1 ubiquitination process, it remains largely unknown which DUB is more physiological involved in regulating PD-L1 ubiquitination and whether there is cellular context or tissue context dependent regulation of PD-L1 ubiquitination by different DUBs in different cancer types. More importantly, as the specific chemical inhibitors targeting DUBs are becoming a new method for cancer treatment [36], it will be very interesting in the future to evaluate the potential of DUBs inhibitor in combination with PD-1/PD-L1 blockade as more effective anti-cancer therapies.

3. N-glycosylation of PD-L1

Glycosylation including N-linked glycosylation and O-linked glycosylation is an enzyme-directed site-specific process of glycoconjugate formation, which plays important roles in regulating various human diseases [37, 38]. Accumulating evidence indicates that PD-L1 is glycosylated with heavy N-linked glycan moieties that regulates its protein stability and interaction with cognate receptor PD-1, thereby affecting anticancer immunotherapy (Fig. 3) [18, 39]. Although PD-L1 is N-linked glycosylated on N35/192/200/219, the N192/200/219 residues’ glycosylation regulates PD-L1 protein stability in part through suppressing GSK3β-β-TRCP-mediated PD-L1 polyubiquitination [18]. Moreover, proper glycosylation of PD-L1 is essential for its recognition by PD-1 in cells. Hence, B3GNT3-mediated poly-N-acetyllactosamine (poly-LacNAc) is required for PD-L1/PD-1 interaction [39]. Li and colleagues also generated glycosylation-specific PD-L1 antibodies (gPD-L1), which can efficiently block PD-L1/PD-1 interaction and subsequently promote PD-L1 internalization and degradation [39]. Furthermore, gPD-L1-ADC (antibody-drug conjugate) has potent anti-tumor activities in triple-negative breast cancer models [39]. STT3, the catalytically active subunit of oligosaccharyltransferase, increases PD-L1 glycosylation in cancer stem cells, leading to PD-L1 upregulation [40]. Etoposide, which can suppress the epithelial-mesenchymal transition (EMT) induced STT3 expression, enhances the therapeutic efficacy of T cell immunoglobulin mucin-3 (TIM-3) blockade therapy [40]. Chaperone Sigma1 and FKBP51s have also been reported to promote PD-L1 glycosylation and protein stability with unclear mechanisms [41, 42]. Pharmacologic inhibition of Sigma 1 with IPAG (1-(4-Iodophenyl)-3-(2-adamantyl) guanidine) reduced PD-L1 expression and activated T cells in vitro in prostate and triple-negative breast cancer models [42]. Inhibition of FKBP51s using selective inhibitor SAFit [43] reduced PD-L1 protein levels and subsequently induced peripheral blood mononuclear cells (PBMCs) death cocultured with glioma cells [41].

Fig 3. Regulation of PD-L1 by glycosylation.

Fig 3.

STT3 and B3GNT3 can glycosylate PD-L1, protecting PD-L1 from degradation. Chaperone Sigma1 and FKBP51s have also been reported to promote PD-L1 glycosylation.

4. Phosphorylation of PD-L1

Protein phosphorylation is a ubiquitous post-translational modification of proteins in which an amino acid residue is phosphorylated by a protein kinase by the addition of a covalently bound phosphate group [44]. Emerging evidence has shown that PD-L1 is subjected to phosphorylation at different Ser/Thr/Tyr residues that impact its protein stability and functions. Specifically, glycogen synthase kinase 3β (GSK3β) directly phosphorylates PD-L1 at the T180 and S184 sites, leading to the poly-ubiquitination by β-TRCP (Fig. 4) [18]. Phosphorylation of PD-L1 by GSK3β can be inactivated by the epidermal growth factor (EGF) signaling pathway, leading to inhibition of PD-L1’s degradation [18]. Based on this mechanism, EGFR inhibitor gefitinib destabilizes PD-L1 through activating GSK3β and enhances the therapeutic efficacy of PD-1 blockade in syngeneic mouse models [18]. In studies of liver cancer mice with orthotopic tumors grown from Hepa 1-6 cells, MET-mediated phosphorylation and activated GSK3β at the Y56 residue, leading to decreased expression of PD-L1 [45]. The combination of MET inhibitor with anti-PD-1 blockade significantly suppresses tumor growth and prolongs survival in hepatocellular carcinoma (HCC) mouse models [45]. Moreover, AMP activated kinase (AMPK) as an energy sensor directly phosphorylates PD-L1 at the S195 residue, resulting PD-L1 degradation in part through an ERAD pathway (Fig. 4) [21]. Based on these results, metformin that can activate AMPK elevates the efficacy of CTLA-4 blockade in different syngeneic mouse models [21]. Besides the Ser/Thr phosphorylation, PD-L1 can be phosphorylated on tyrosine [46]. Chan et al. reported that IL-6-activated Janus Kinase 1 (JAK1) phosphorylates PD-L1 at Y112 (Fig. 4) [46]. The phosphorylation of PD-L1 at Y112 recruits STT3 to promote PD-L1 glycosylation, which protects PD-L1 from proteolytic degradation [46]. Blocking IL-6/JAK1-mediated PD-L1 protein stability through targeting IL-6 by an IL-6 antibody induces synergistic T cell killing effects when combined with anti-TIM-3 therapy in a Hepa 1–6 liver cancer immunocompetent mouse model [46].

Fig. 4. Regulations of PD-L1 by phosphorylation, acetylation and palmitoylation.

Fig. 4.

PD-L1 can be phosphorylated by GSK3β, AMPK and JAK1. PD-L1 is acetylated at K263 by the acetyltransferase p300 and deacetylated by HDAC2. Moreover, ZDHHC3/9 promote the palmitoylation of PD-L1 at the C272 residue.

5. Acetylation of PD-L1

Non-histone protein acetylation has been demonstrated to play pivotal roles in affecting various physiological and pathological processes through modulating protein stability, protein-protein interaction, subcellular localization, and functional activity [47]. While an unbiased snapshot of key post-translational modification profiles for PD-L1 reported that PD-L1 could be acetylated and EGF treatment increased acetylation level of PD-L1, the detailed molecular mechanisms as well as the biological function of PD-L1 acetylation remains unknown [48]. Recently, our group uncovered that PD-L1 is acetylated on the K263 residual of the cytoplasmic domain, which is dynamically regulated by acetyltransferase p300 and deacetylase HDAC2 (Fig. 4) [49]. Furthermore, we revealed an unexpected PD-L1 translocation from membrane to the nucleus, a process that is largely dependent on the acetylation status of PD-L1 [49]. Through high throughput RNA-seq and ChIP-seq methods, we showed that PD-L1 per se, is essential for the expression of the immune-related genes that governs the anticancer immune response [49]. Moreover, we demonstrated that genetically or pharmacologically modulating PD-L1 acetylation blocks its nuclear translocation and consequently enhances the anti-tumor efficacy of the PD-1 blockade [49]. Interestingly, almost at the same time, several other groups also independently reported nuclear PD-L1 as emerging factor to affect cancer cell proliferation and necrosis [50-52]. However, whether and how PD-L1 acetylation plays important roles in these broad processes warrants further investigation. As the predictive marker for immunotherapy is urgently needed in clinic and high PD-L1 expression is widely used as a maker for patient selection, we also speculate whether nuclear PD-L1 or acetylation status of PD-L1 could serve as a useful biomarker for cancer immunotherapy in the future [53].

6. Palmitoylation of PD-L1

Palmitoylation is the covalent attachment of fatty acids to proteins which is generally done by proteins with the DHHC domain [54, 55]. Recently, three independent groups found that palmitoyltransferases ZDHHC3 or ZDHHC9 could induce PD-L1 palmitoylation at the cystine-272 site, which increases PD-L1 cell surface distribution by preventing its ubiquitination and degradation (Fig. 4) [56-58]. Genetically knockdown the palmitoyltransferase for PD-L1, or using a palmitoylation deficient C272A mutant version of PD-L1, remarkably reduced membrane PD-L1 levels in cancer cells, resulting in increased sensitivity to T-cell mediated cancer cell killing in an in vitro co-culture assay [56, 57]. Notably, Yao et al. designed a competitive inhibitor PD-PALM, which is a chimeric peptide comprising a cell-penetrating peptide and a peptide fragment from PD-L1 encompassing the Cys272 residue [57]. Treatment of cancer cells with PD-PALM significantly decreased the palmitoylation and expression of PD-L1 and suppressed tumorigenesis [57]. More importantly, in 4T1 syngeneic mouse breast cancer model, Zdhhc9 knockout potently inhibited 4T1 tumor growth and improved anti-mPD-1 therapeutic efficacy [56]. These results thus support the notion of interrupting PD-L1 palmitoylation to enhance the immunotherapy response.

Regulations of PD-L1 functions via exosomal secretion

Exosomes are small extracellular vesicles (EVs) that are membrane-enveloped particles produced by most cell types [59, 60]. Various protein post-translational modifications including ubiquitination, palmitoylation and acetylation have been previously suggested to be involved in regulation of exosome formation and protein transportation to the exosome [61-64]. Although PD-L1 can be primarily expressed on the surface of tumor cells, it is also found to exist in exosomes of various cancer types [65-68]. Exosomal PD-L1 displays the same extracellular domain topology as its cell surface counterpart, hence circulating PD-L1-positive exosomes can systemically inhibit anti-tumor immunity [65, 67]. Genetic ablation of exosomal PD-L1 or blockade of exosome secretion with inhibitors suppressed tumor growth via anti-tumor immunity in different cancer models [65-67, 69]. Immunotherapy requires biomarkers for providing personalized precision treatment and predicting tumor progression. Hence, exosomal PD-L1 is emerging as a non-invasive and readily available biomarker. In support of this point, the levels of exosomal PD-L1 in the blood are found to be relatively higher in different tumors such as melanoma, non-small cell lung cancer (NSCLC) [65, 70-72]. However, whether and how exosomal PD-L1 is regulated by post-translational modifications in a similar fashion as acetylation does in regulating PD-L1 nuclear translocation [49] warrants further investigation. Hence, therapeutic agents targeting the related protein modification and exosome pathway are attractive candidates for developing new combined immunotherapy.

Post-translational modifications of PD-1

Different from PD-L1, PD-1 mainly expresses on activated T and B cells [7]. Although the regulation of PD-1 on transcriptional levels has been extensively studied [12, 73], recent studies suggest that post-translational modifications also play important roles in the regulation of PD-1 expression as well as anti-cancer immunity [74, 75].

1. Ubiquitination of PD-1

Through mass spectrometry, Meng et al. identified the E3 ubiquitin ligase SCFFBXO38 as a binding partner of PD-1 and promoted K48-linked poly-ubiquitination of PD-1 at the K233 site, leading to subsequent degradation of PD-1 through the 26S proteasome (Fig. 5) [75]. Conditional knockout Fbxo38 in T cells led to faster tumor progression in mice owning to increased expression of PD-1 [75]. More importantly, IL-2 therapy could suppress tumor progression through rescuing Fbxo38 transcription and downregulating PD-1 protein levels [75]. Recently, Zhou et al. identified KLHL22, an adaptor of the Cul3-based E3 ligase, as another PD-1-interacting protein (Fig. 5) [76]. Biochemically, KLHL22 polyubiquitinates PD-1, leading to degradation of PD-1 before its transportation to the cell surface [76]. Treatment with 5-fluorouracil (5-FU) could increase PD-1 expression by inhibiting the transcription of KLHL22, which suggests that PD-1 expression is possibly responsible for the limited efficacy of 5-FU [76]. On the other hand, Casitas B-lineage lymphoma (c-Cbl) was reported to destabilize PD-1 in part through ubiquitination-proteasomal degradation depending on c-Cbl’s RING finger function (Fig. 5) [77]. In syngeneic colorectal cancer xenografts, immune cell infiltration was higher in c-Cbl+/− compared to c-Cbl+/+ mice and tumor-associated CD8+ T-lymphocytes and macrophages of c-Cbl+/− mice showed higher levels of PD-1 [77]. Although multiple E3 ligases have been reported to ubiquitinate PD-1, it remains unknown which E3 ligase is more physiologically relevant in governing PD-1 protein stability, aberrancy of which will lead to deregulated PD-1 signaling to impact tumorigenesis. Moreover, potential DUBs that targeting PD-1 are highly interested, which also require additional in-depth studies in the future.

Fig 5. Regulation of PD-1 by polyubiquitination.

Fig 5.

PD-1 can be polyubiquitinated by E3 ligases FBXO38, KLHL22, and c-Cbl, leading to PD-1 degradation through the 26S proteasome.

2. Glycosylation of PD-1

As a common modification in eukaryotic cells, glycosylation changes, including abnormal core fucosylation and increased N-glycan branching, have been observed in tumor cells [37, 38]. Similar to PD-L1 that has been reported to be heavily glycosylated, PD-1 is also glycosylated in cells [74, 78, 79]. PD-1’s molecular weight is about 14 kDa, when expressed in E. coli with no glycosylation, but shifts to 35–40 kDa, when expressed in 293T cells with glycosylation similar to that present in host cells [80]. Moreover, upon mutating four potential N-glycosylation sites, PD-1 exhibits substantially reduced molecular weight [80]. Notably, the binding affinity of PD-1 to Camrelizumab, a recently FDA approved PD-1-specific monoclonal antibody (mAb) [81], can be regulated by N-glycan composition of PD-1 [78]. Notably, Camrelizumab can strongly bind to glycosylated PD-1, but the binding to N58A mutant PD-1 that cannot be glycosylated on this site or non-glycosylated PD-1 proteins from E. coli is substantially decreased. These results suggest that glycosylation of PD-1 affects the activity of PD-1-specific mAbs [78].

Core-fucosylation is a kind of N-linked glycosylation in which an alpha-1,6 linked fucose is added to the innermost N-acetylglucosamine (GlcNAc) residue and plays important roles in tumorigenesis and immune escape [82, 83]. Through CRISPR-Cas9 screen, Okada et al. identified critical genes involved in the core fucosylation pathway as positive regulators of cell-surface PD-1 expression [74]. Among the genes identified, fucosyltransferase Fut8 is the only enzyme that catalyzes core fucosylation. Fut8 promotes PD-1-N-linked oligosaccharides at positions N49 and N74 to regulate cell-surface expression of PD-1 [74]. Notably, blocking the fucosylation using fucosylation inhibitor 2-fluoro-L-fucose (2F-Fuc) leads to better immune response in mouse models [74]. Mechanistically, loss of core fucosylation enhances the PD-1 ubiquitination and in turn leads to the degradation of PD-1 by the 26S proteasome [79].

Concluding Remarks

Although PD-1/PD-L1 immune checkpoint blockade exhibits promising clinical benefit, response rates are still lower than 40% in most cancer types [2-4]. The post-transcriptional regulations of PD-1/PD-L1 and the suggested combination therapy strategies thus provide new avenues to increase the efficacy of the PD-1/PD-L1 blockade. To this end, further in-depth investigation and elucidation of the new modifications will be a key step for improving the response for cancer therapy. In addition to find new regulation of PD-1/PD-L1, future work should also be put into developing preclinical/clinical models to translate the existing regulations into clinical benefit. Several novel PROTACs such as AbTACs [28] and LYTACs [29] have been developed to target PD-L1 for degradation, but their effects in preclinical models remain unknown and whether these PROTACs have better clinical outcomes than the original antibodies are needed in-depth exploration.

Compared with the post-translational modification of PD-L1, the modifications of PD-1 are also critical for anticancer immune response. Unlike PD-L1, the modulation of PD-1 at protein level is emerging and remains largely elusive. Although PD-1 can be polyubiquitinated by different E3 ligases such as FBXO38 [75], KLHL22 [76] and c-Cbl [76], it still remains unknown whether there is any DUB that can reverse PD-1’s polyubiquitination. It is also important to reveal which E3 ligase plays an important role in physiologically governing PD-1 ubiquitination and which E3 ligase is pathologically dysregulated to cause immune surveillance defects. Besides ubiquitination and glycosylation, whether is PD-1 regulated by other post-translational modifications such as phosphorylation, acetylation and palmitoylation? More importantly, how to translate these regulatory mechanisms into the enhancement of immunotherapy warrants further investigation.

Acknowledgements

We sincerely thank our colleagues for their critical comments and help. This research was supported by the NIH grants (R35CA253027 to W.W.). We apologize to those authors whose important work was not cited because of space limitation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest

W.W. is a co-founder and consultant for the ReKindle Therapeutics. Other authors declare no competing financial interests.

References:

  • [1].Pardoll DM, The blockade of immune checkpoints in cancer immunotherapy, Nature Reviews Cancer, 12 (2012) 252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Sharma P, Allison JP, The future of immune checkpoint therapy, Science, 348 (2015) 56–61. [DOI] [PubMed] [Google Scholar]
  • [3].Iwai Y, Hamanishi J, Chamoto K, Honjo T, Cancer immunotherapies targeting the PD-1 signaling pathway, J Biomed Sci, 24 (2017) 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Hargadon KM, Johnson CE, Williams CJ, Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors, Int Immunopharmacol, 62 (2018) 29–39. [DOI] [PubMed] [Google Scholar]
  • [5].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, 11 (1992) 3887–3895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T, Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes, Int Immunol, 8 (1996) 765–772. [DOI] [PubMed] [Google Scholar]
  • [7].Keir ME, Butte MJ, Freeman GJ, Sharpe AH, PD-1 and its ligands in tolerance and immunity, Annu Rev Immunol, 26 (2008) 677–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Sharpe AH, Pauken KE, The diverse functions of the PD1 inhibitory pathway, Nat Rev Immunol, 18(2018)153–167. [DOI] [PubMed] [Google Scholar]
  • [9].Schildberg Frank A., Klein Sarah R., Freeman Gordon J., Sharpe Arlene H., Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family, Immunity, 44 (2016) 955–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Pitt JM, Vetizou M, Daillere R, Roberti MP, Yamazaki T, Routy B, Lepage P, Boneca IG, Chamaillard M, Kroemer G, Zitvogel L, Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors, Immunity, 44 (2016) 1255–1269. [DOI] [PubMed] [Google Scholar]
  • [11].Yarchoan M, Hopkins A, Jaffee EM, Tumor Mutational Burden and Response Rate to PD-1 Inhibition, N Engl J Med, 377 (2017) 2500–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Bally AP, Austin JW, Boss JM, Genetic and Epigenetic Regulation of PD-1 Expression, J Immunol, 196 (2016) 2431–2437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Chen J, Jiang CC, Jin L, Zhang XD, Regulation of PD-L1: a novel role of pro-survival signalling in cancer, Ann Oncol, 27 (2016) 409–416. [DOI] [PubMed] [Google Scholar]
  • [14].Sun C, Mezzadra R, Schumacher TN, Regulation and Function of the PD-L1 Checkpoint, Immunity, 48 (2018) 434–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Liu J, Qian C, Cao X, Post-Translational Modification Control of Innate Immunity, Immunity, 45 (2016) 15–30. [DOI] [PubMed] [Google Scholar]
  • [16].Rape M, Ubiquitylation at the crossroads of development and disease, Nat Rev Mol Cell Biol, 19 (2018) 59–70. [DOI] [PubMed] [Google Scholar]
  • [17].Wang P, Dai X, Jiang W, Li Y, Wei W, RBR E3 ubiquitin ligases in tumorigenesis, Semin Cancer Biol, (2020). [DOI] [PubMed] [Google Scholar]
  • [18].Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, Khoo KH, Chang SS, Cha JH, Kim T, Hsu JL, Wu Y, Hsu JM, Yamaguchi H, Ding Q, Wang Y, Yao J, Lee CC, Wu HJ, Sahin AA, Allison JP, Yu D, Hortobagyi GN, Hung MC, Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity, Nat Commun, 7 (2016) 12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Deng L, Qian G, Zhang S, Zheng H, Fan S, Lesinski GB, Owonikoko TK, Ramalingam SS, Sun SY, Inhibition of mTOR complex 1/p70 S6 kinase signaling elevates PD-L1 levels in human cancer cells through enhancing protein stabilization accompanied with enhanced beta-TrCP degradation, Oncogene, 38 (2019) 6270–6282. [DOI] [PubMed] [Google Scholar]
  • [20].Zhang J, Bu X, Wang H, Zhu Y, Geng Y, Nihira NT, Tan Y, Ci Y, Wu F, Dai X, Guo J, Huang YH, Fan C, Ren S, Sun Y, Freeman GJ, Sicinski P, Wei W, Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance, Nature, 553 (2018) 91–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, Li CW, Kim T, Chang SS, Lee HH, Hsu JL, Wang HL, Kuo CW, Chang WC, Hadad S, Purdie CA, McCoy AM, Cai S, Tu Y, Litton JK, Mittendorf EA, Moulder SL, Symmans WF, Thompson AM, Piwnica-Worms H, Chen CH, Khoo KH, Hung MC, Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1, Mol Cell, 71 (2018) 606–620 e607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, Lam EYN, Henderson MA, Bell CC, Stolzenburg S, Gilan O, Bloor S, Noori T, Morgens DW, Bassik MC, Neeson PJ, Behren A, Darcy PK, Dawson SJ, Voskoboinik I, Trapani JA, Cebon J, Lehner PJ, Dawson MA, CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity, Nature, 549 (2017) 101–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Mezzadra R, Sun C, Jae LT, Gomez-Eerland R, de Vries E, Wu W, Logtenberg MEW, Slagter M, Rozeman EA, Hofland I, Broeks A, Horlings HM, Wessels LFA, Blank CU, Xiao Y, Heck AJR, Borst J, Brummelkamp TR, Schumacher TNM, Identification of CMTM6 and CMTM4 as PD-L1 protein regulators, Nature, 549 (2017) 106–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Sakamoto KM, Kim KB, Kumagai A, Mercurio F, Crews CM, Deshaies RJ, Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation, Proc Natl Acad Sci U S A, 98 (2001) 8554–8559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Liu J, Ma J, Liu Y, Xia J, Li Y, Wang ZP, Wei W, PROTACs: A novel strategy for cancer therapy, Semin Cancer Biol, 67 (2020) 171–179. [DOI] [PubMed] [Google Scholar]
  • [26].Pettersson M, Crews CM, PROteolysis TArgeting Chimeras (PROTACs) - Past, present and future, Drug Discov Today Technol, 31 (2019) 15–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Cheng B, Ren Y, Cao H, Chen J, Discovery of novel resorcinol diphenyl ether-based PROTAC-like molecules as dual inhibitors and degraders of PD-L1, Eur J Med Chem, 199 (2020) 112377. [DOI] [PubMed] [Google Scholar]
  • [28].Cotton AD, Nguyen DP, Gramespacher JA, Seiple IB, Wells JA, Development of Antibody-Based PROTACs for the Degradation of the Cell-Surface Immune Checkpoint Protein PD-L1, J Am Chem Soc, 143 (2021) 593–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Banik SM, Pedram K, Wisnovsky S, Ahn G, Riley NM, Bertozzi CR, Lysosome-targeting chimaeras for degradation of extracellular proteins, Nature, 584 (2020) 291–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Uhle S, Medalia O, Waldron R, Dumdey R, Henklein P, Bech-Otschir D, Huang X, Berse M, Sperling J, Schade R, Dubiel W, Protein kinase CK2 and protein kinase D are associated with the COP9 signalosome, EMBO J, 22 (2003) 1302–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Lim SO, Li CW, Xia W, Cha JH, Chan LC, Wu Y, Chang SS, Lin WC, Hsu JM, Hsu YH, Kim T, Chang WC, Hsu JL, Yamaguchi H, Ding Q, Wang Y, Yang Y, Chen CH, Sahin AA, Yu D, Hortobagyi GN, Hung MC, Deubiquitination and Stabilization of PD-L1 by CSN5, Cancer Cell, 30 (2016) 925–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Huang X, Zhang Q, Lou Y, Wang J, Zhao X, Wang L, Zhang X, Li S, Zhao Y, Chen Q, Liang T, Bai X, USP22 Deubiquitinates CD274 to Suppress Anticancer Immunity, Cancer Immunol Res, 7 (2019) 1580–1590. [DOI] [PubMed] [Google Scholar]
  • [33].Wang Y, Sun Q, Mu N, Sun X, Wang Y, Fan S, Su L, Liu X, The deubiquitinase USP22 regulates PD-L1 degradation in human cancer cells, Cell Commun Signal, 18 (2020) 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Jingjing W, Wenzheng G, Donghua W, Guangyu H, Aiping Z, Wenjuan W, Deubiquitination and stabilization of programmed cell death ligand 1 by ubiquitin-specific peptidase 9, X-linked in oral squamous cell carcinoma, Cancer Med, 7 (2018) 4004–4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Zhu D, Xu R, Huang X, Tang Z, Tian Y, Zhang J, Zheng X, Deubiquitinating enzyme OTUB1 promotes cancer cell immunosuppression via preventing ER-associated degradation of immune checkpoint protein PD-L1, Cell Death Differ, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Schauer NJ, Magin RS, Liu X, Doherty LM, Buhrlage SJ, Advances in Discovering Deubiquitinating Enzyme (DUB) Inhibitors, J Med Chem, 63 (2020) 2731–2750. [DOI] [PubMed] [Google Scholar]
  • [37].Schwarz F, Aebi M, Mechanisms and principles of N-linked protein glycosylation, Curr Opin Struct Biol, 21 (2011) 576–582. [DOI] [PubMed] [Google Scholar]
  • [38].Pinho SS, Reis CA, Glycosylation in cancer: mechanisms and clinical implications, Nat Rev Cancer, 15 (2015) 540–555. [DOI] [PubMed] [Google Scholar]
  • [39].Li CW, Lim SO, Chung EM, Kim YS, Park AH, Yao J, Cha JH, Xia W, Chan LC, Kim T, Chang SS, Lee HH, Chou CK, Liu YL, Yeh HC, Perillo EP, Dunn AK, Kuo CW, Khoo KH, Hsu JL, Wu Y, Hsu JM, Yamaguchi H, Huang TH, Sahin AA, Hortobagyi GN, Yoo SS, Hung MC, Eradication of Triple-Negative Breast Cancer Cells by Targeting Glycosylated PD-L1, Cancer Cell, 33 (2018) 187–201 e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Hsu JM, Xia W, Hsu YH, Chan LC, Yu WH, Cha JH, Chen CT, Liao HW, Kuo CW, Khoo KH, Hsu JL, Li CW, Lim SO, Chang SS, Chen YC, Ren GX, Hung MC, STT3-dependent PD-L1 accumulation on cancer stem cells promotes immune evasion, Nat Commun, 9 (2018) 1908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].D'Arrigo P, Russo M, Rea A, Tufano M, Guadagno E, Del ML Basso De Caro, Pacelli R, Hausch F, Staibano S, Ilardi G, Parisi S, Romano MF, Romano S, A regulatory role for the co-chaperone FKBP51s in PD-L1 expression in glioma, Oncotarget, 8 (2017) 68291–68304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Maher CM, Thomas JD, Haas DA, Longen CG, Oyer HM, Tong JY, Kim FJ, Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1, Mol Cancer Res, 16 (2018) 243–255. [DOI] [PubMed] [Google Scholar]
  • [43].Gaali S, Kirschner A, Cuboni S, Hartmann J, Kozany C, Balsevich G, Namendorf C, Fernandez-Vizarra P, Sippel C, Zannas AS, Draenert R, Binder EB, Almeida OF, Ruhter G, Uhr M, Schmidt MV, Touma C, Bracher A, Hausch F, Selective inhibitors of the FK506-binding protein 51 by induced fit, Nat Chem Biol, 11 (2015) 33–37. [DOI] [PubMed] [Google Scholar]
  • [44].Cohen P, The origins of protein phosphorylation, Nat Cell Biol, 4 (2002) E127–130. [DOI] [PubMed] [Google Scholar]
  • [45].Li H, Li CW, Li X, Ding Q, Guo L, Liu S, Liu C, Lai CC, Hsu JM, Dong Q, Xia W, Hsu JL, Yamaguchi H, Du Y, Lai YJ, Sun X, Koller PB, Ye Q, Hung MC, MET Inhibitors Promote Liver Tumor Evasion of the Immune Response by Stabilizing PDL1, Gastroenterology, 156 (2019) 1849–1861 e1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Chan LC, Li CW, Xia W, Hsu JM, Lee HH, Cha JH, Wang HL, Yang WH, Yen EY, Chang WC, Zha Z, Lim SO, Lai YJ, Liu C, Liu J, Dong Q, Yang Y, Sun L, Wei Y, Nie L, Hsu JL, Li H, Ye Q, Hassan MM, Amin HM, Kaseb AO, Lin X, Wang SC, Hung MC, IL-6/JAK1 pathway drives PD-L1 Y112 phosphorylation to promote cancer immune evasion, J Clin Invest, 129 (2019) 3324–3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Narita T, Weinert BT, Choudhary C, Functions and mechanisms of non-histone protein acetylation, Nat Rev Mol Cell Biol, 20 (2019) 156–174. [DOI] [PubMed] [Google Scholar]
  • [48].Horita H, Law A, Hong S, Middleton K, Identifying Regulatory Posttranslational Modifications of PD-L1: A Focus on Monoubiquitinaton, Neoplasia, 19 (2017) 346–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Gao Y, Nihira NT, Bu X, Chu C, Zhang J, Kolodziejczyk A, Fan Y, Chan NT, Ma L, Liu J, Wang D, Dai X, Liu H, Ono M, Nakanishi A, Inuzuka H, North BJ, Huang YH, Sharma S, Geng Y, Xu W, Liu XS, Li L, Miki Y, Sicinski P, Freeman GJ, Wei W, Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy, Nat Cell Biol, 22 (2020) 1064–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Du W, Zhu J, Zeng Y, Liu T, Zhang Y, Cai T, Fu Y, Zhang W, Zhang R, Liu Z, Huang JA, KPNB1-mediated nuclear translocation of PD-L1 promotes non-small cell lung cancer cell proliferation via the Gas6/MerTK signaling pathway, Cell Death Differ, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Hou J, Zhao R, Xia W, Chang CW, You Y, Hsu JM, Nie L, Chen Y, Wang YC, Liu C, Wang WJ, Wu Y, Ke B, Hsu JL, Huang K, Ye Z, Yang Y, Xia X, Li Y, Li CW, Shao B, Tainer JA, Hung MC, PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis, Nat Cell Biol, 22 (2020) 1264–1275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Yu J, Qin B, Moyer AM, Nowsheen S, Tu X, Dong H, Boughey JC, Goetz MP, Weinshilboum R, Lou Z, Wang L, Regulation of sister chromatid cohesion by nuclear PD-L1, Cell Res, 30 (2020) 590–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Walk EE, Yohe SL, Beckman A, Schade A, Zutter MM, Pfeifer J, Berry AB, C. College of American Pathologists Personalized Health Care, The Cancer Immunotherapy Biomarker Testing Landscape, Arch Pathol Lab Med, 144 (2020) 706–724. [DOI] [PubMed] [Google Scholar]
  • [54].Resh MD, Palmitoylation of proteins in cancer, Biochem Soc Trans, 45 (2017) 409–416. [DOI] [PubMed] [Google Scholar]
  • [55].De I, Sadhukhan S, Emerging Roles of DHHC-mediated Protein S-palmitoylation in Physiological and Pathophysiological Context, Eur J Cell Biol, 97 (2018) 319–338. [DOI] [PubMed] [Google Scholar]
  • [56].Yang Y, Hsu JM, Sun L, Chan LC, Li CW, Hsu JL, Wei Y, Xia W, Hou J, Qiu Y, Hung MC, Palmitoylation stabilizes PD-L1 to promote breast tumor growth, Cell Res, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Yao H, Lan J, Li C, Shi H, Brosseau J-P, Wang H, Lu H, Fang C, Zhang Y, Liang L, Zhou X, Wang C, Xue Y, Cui Y, Xu J, Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours, Nature Biomedical Engineering, (2019). [DOI] [PubMed] [Google Scholar]
  • [58].Shahid M, Kim M, Jin P, Zhou B, Wang Y, Yang W, You S, Kim J, S-Palmitoylation as a Functional Regulator of Proteins Associated with Cisplatin Resistance in Bladder Cancer, Int J Biol Sci, 16 (2020) 2490–2505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Kalluri R, The biology and function of exosomes in cancer, J Clin Invest, 126 (2016) 1208–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Tkach M, Thery C, Communication by Extracellular Vesicles: Where We Are and Where We Need to Go, Cell, 164 (2016) 1226–1232. [DOI] [PubMed] [Google Scholar]
  • [61].Moreno-Gonzalo O, Villarroya-Beltri C, Sanchez-Madrid F, Post-translational modifications of exosomal proteins, Front Immunol, 5 (2014) 383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Smith VL, Jackson L, Schorey JS, Ubiquitination as a Mechanism To Transport Soluble Mycobacterial and Eukaryotic Proteins to Exosomes, J Immunol, 195 (2015) 2722–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Li Z, Zhuang M, Zhang L, Zheng X, Yang P, Li Z, Acetylation modification regulates GRP78 secretion in colon cancer cells, Sci Rep, 6 (2016) 30406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Romancino DP, Buffa V, Caruso S, Ferrara I, Raccosta S, Notaro A, Campos Y, Noto R, Martorana V, Cupane A, Giallongo A, d'Azzo A, Manno M, Bongiovanni A, Palmitoylation is a post-translational modification of Alix regulating the membrane organization of exosome-like small extracellular vesicles, Biochim Biophys Acta Gen Subj, 1862 (2018) 2879–2887. [DOI] [PubMed] [Google Scholar]
  • [65].Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, Yu Z, Yang J, Wang B, Sun H, Xia H, Man Q, Zhong W, Antelo LF, Wu B, Xiong X, Liu X, Guan L, Li T, Liu S, Yang R, Lu Y, Dong L, McGettigan S, Somasundaram R, Radhakrishnan R, Mills G, Lu Y, Kim J, Chen YH, Dong H, Zhao Y, Karakousis GC, Mitchell TC, Schuchter LM, Herlyn M, Wherry EJ, Xu X, Guo W, Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response, Nature, 560 (2018) 382–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Yang Y, Li CW, Chan LC, Wei Y, Hsu JM, Xia W, Cha JH, Hou J, Hsu JL, Sun L, Hung MC, Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth, Cell Res, 28 (2018) 862–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, Montabana E, Lang UE, Fu Q, Fong L, Blelloch R, Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory, Cell, 177 (2019) 414–427 e413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Zhou K, Guo S, Li F, Sun Q, Liang G, Exosomal PD-L1: New Insights Into Tumor Immune Escape Mechanisms and Therapeutic Strategies, Front Cell Dev Biol, 8 (2020) 569219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Kim DH, Kim H, Choi YJ, Kim SY, Lee JE, Sung KJ, Sung YH, Pack CG, Jung MK, Han B, Kim K, Kim WS, Nam SJ, Choi CM, Yun M, Lee JC, Rho JK, Exosomal PD-L1 promotes tumor growth through immune escape in non-small cell lung cancer, Exp Mol Med, 51 (2019) 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Liu C, Zeng X, An Z, Yang Y, Eisenbaum M, Gu X, Jornet JM, Dy GK, Reid ME, Gan Q, Wu Y, Sensitive Detection of Exosomal Proteins via a Compact Surface Plasmon Resonance Biosensor for Cancer Diagnosis, ACS Sens, 3 (2018) 1471–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Li C, Li C, Zhi C, Liang W, Wang X, Chen X, Lv T, Shen Q, Song Y, Lin D, Liu H, Clinical significance of PD-L1 expression in serum-derived exosomes in NSCLC patients, J Transl Med, 17 (2019) 355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Huang M, Yang J, Wang T, Song J, Xia J, Wu L, Wang W, Wu Q, Zhu Z, Song Y, Yang C, Homogeneous, Low-volume, Efficient, and Sensitive Quantitation of Circulating Exosomal PD-L1 for Cancer Diagnosis and Immunotherapy Response Prediction, Angew Chem Int Ed Engl, 59 (2020) 4800–4805. [DOI] [PubMed] [Google Scholar]
  • [73].Yu X, Gao R, Li Y, Zeng C, Regulation of PD-1 in T cells for cancer immunotherapy, Eur J Pharmacol, 881 (2020) 173240. [DOI] [PubMed] [Google Scholar]
  • [74].Okada M, Chikuma S, Kondo T, Hibino S, Machiyama H, Yokosuka T, Nakano M, Yoshimura A, Blockage of Core Fucosylation Reduces Cell-Surface Expression of PD-1 and Promotes Anti-tumor Immune Responses of T Cells, Cell Rep, 20 (2017) 1017–1028. [DOI] [PubMed] [Google Scholar]
  • [75].Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, Liu H, Bai Y, Xue M, Hu R, Sun SC, Liu X, Zhou P, Huang X, Wei L, Yang W, Xu C, FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells, Nature, 564 (2018) 130–135. [DOI] [PubMed] [Google Scholar]
  • [76].Zhou XA, Zhou J, Zhao L, Yu G, Zhan J, Shi C, Yuan R, Wang Y, Chen C, Zhang W, Xu D, Ye Y, Wang W, Shen Z, Wang W, Wang J, KLHL22 maintains PD-1 homeostasis and prevents excessive T cell suppression, Proc Natl Acad Sci U S A, 117 (2020) 28239–28250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Lyle C, Richards S, Yasuda K, Napoleon MA, Walker J, Arinze N, Belghasem M, Vellard I, Yin W, Ravid JD, Zavaro E, Amraei R, Francis J, Phatak U, Rifkin IR, Rahimi N, Chitalia VC, c-Cbl targets PD-1 in immune cells for proteasomal degradation and modulates colorectal tumor growth, Sci Rep, 9 (2019) 20257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Liu K, Tan S, Jin W, Guan J, Wang Q, Sun H, Qi J, Yan J, Chai Y, Wang Z, Deng C, Gao GF, N-glycosylation of PD-1 promotes binding of camrelizumab, EMBO Rep, (2020) e51444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Zhang N, Li M, Xu X, Zhang Y, Liu Y, Zhao M, Li P, Chen J, Fukuda T, Gu J, Jin X, Li W, Loss of core fucosylation enhances the anticancer activity of cytotoxic T lymphocytes by increasing PD-1 degradation, Eur J Immunol, 50 (2020) 1820–1833. [DOI] [PubMed] [Google Scholar]
  • [80].Tan S, Zhang H, Chai Y, Song H, Tong Z, Wang Q, Qi J, Wong G, Zhu X, Liu WJ, Gao S, Wang Z, Shi Y, Yang F, Gao GF, Yan J, An unexpected N-terminal loop in PD-1 dominates binding by nivolumab, Nat Commun, 8 (2017) 14369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Markham A, Keam SJ, Camrelizumab: First Global Approval, Drugs, 79 (2019) 1355–1361. [DOI] [PubMed] [Google Scholar]
  • [82].Jia L, Zhang J, Ma T, Guo Y, Yu Y, Cui J, The Function of Fucosylation in Progression of Lung Cancer, Front Oncol, 8 (2018) 565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Keeley TS, Yang S, Lau E, The Diverse Contributions of Fucose Linkages in Cancer, Cancers (Basel), 11 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES