Programmed death-ligand 1 (PD-L1) is a transmembrane ligand for the programmed cell death protein 1 (PD-1), a receptor that inhibits T-cell activity. The PD-L1/PD-1 immune checkpoint axis has been successfully targeted to enhance antitumor immune responses. Tethering PD-L1 to the membrane spatially restricts its ability to inhibit immune responses, and it provides for the acute and reversible modulation of PD-L1 plasma membrane density by regulation of its trafficking. PD-L1 has functions that are independent of its role as a ligand for PD-1, and control of PD-L1 residence in different intracellular compartments might contribute to the regulation of those activities. Thus, control of PD-L1 trafficking is emerging as a key feature of its biology. Herein, we focus on current understating of PD-L1 trafficking and review current attempts to therapeutically target this process in cancer cells to enhance antitumor immunity.

Programmed death-ligand 1 (PD-L1) was first identified in a homology screen for the immune costimulatory molecules B7-1 and B7-2. Originally named B7 homolog 1 (B7-H1), it was later rechristened as PD-L1 emphasizing its ability to bind programmed cell death protein 1 (PD-1) on activated T cells (1, 2). The PD-L1/PD-1 interaction leads to the recruitment of the protein tyrosine phosphatases such as SHP2, which play a role in the dephosphorylation of key signaling mediators of T-cell activation (3–7). As an immune checkpoint, the PD-L1/PD-1 axis limits the intensity and duration of an immune response and maintains self-tolerance (8). Its dysregulation can lead to a wide array of diseases, such as lupuslike syndrome, type 1 diabetes mellitus, and loss of fetomaternal tolerance in mice (9).

Expression of PD-L1 protein is upregulated in response to different inflammatory cues (10). Cancer cells can coopt upregulation of PD-L1 expression to evade an effective antitumor response (11). In recent years, the PD-L1/PD-1 checkpoint axis has emerged as a therapeutic target, with eight mAbs specific for either PD-1 or PD-L1 approved by the FDA for clinical use in several cancers (12). While this has expanded the treatment options available for patients, for unknown reasons, most patients do not respond to immunotherapy targeting PD-L1/PD-1 (13). To better predict response rates and improve the efficacy of targeted anti–PD-L1 therapy, it is critical to have a detailed understanding of the regulation of PD-L1 at the transcriptional, translational, and posttranslational levels.

In this review, we discuss the transport of PD-L1 to and from the plasma membrane as a posttranslational mechanism that acutely regulates PD-L1 plasma membrane density and activity as a PD-1 ligand. We begin by briefly summarizing the transcriptional and translational regulation of PD-L1, topics that have been extensively reviewed elsewhere (14–16). We then describe the cellular distribution of PD-L1, the endocytic processes that mediate this distribution and how these may be exploited for cancer therapy.

Several inflammatory cytokines, including IFNγ, IFNα, TNFα, and multiple interleukins increase CD274 (PDL1) transcription (17, 18). The main transcriptional factors that regulate CD274 expression include IRF1, NFkB, and STAT3 (14). In addition to inflammatory cytokines, CD274 transcription has also been described to be upregulated in response to various cell stressors. For instance, during hypoxia, HIF1α and hedgehog signaling can induce PD-L1 expression (19, 20). Chemotherapy and radiotherapy have also been associated with enhanced PD-L1 expression (21, 22). Apart from cell-extrinsic stimuli, cell-intrinsic changes driven by oncogene signaling can also increase CD274 transcription. Driver oncogenes including those encoding mutant EGFR, BRAF, and ALK can enhance STAT3-mediated CD274 transcription via changes in c-Jun or AKT signaling (23–26). Oncogenic MYC has been shown to increase CD274 mRNA levels through direct binding to the CD274 promoter in several cancer models (27). CD274 transcription can also be epigenetically regulated by methylation of the promoter region and associated histones (28–30). Intriguingly, there is very little evidence indicating oncogene-driven CD274 transcription leads to increased steady state PD-L1 protein levels. In both melanoma and prostate cancer, no association was found between the presence of oncogenic mutations and PD-L1 protein level (31, 32).

Posttranscriptionally, PDL1 mRNA is subject to mRNA stability control. A number of miRNAs, such as miR-200 in non–small cell lung cancer (NSCLC) and miR-424(322) in ovarian cancer, have been shown to directly target the three prime untranslated region (3′-UTR) of PDL1 and enhance its turnover (33, 34). Moreover, PDL1 transcripts with aberrant 3′-UTR structure are associated with higher stability and enhanced PD-L1 protein levels (35). Expression of PD-L1 protein may also be more directly induced by recruiting polysomes to PDL1 mRNA, as happens in response to the activation of the PI3K/Akt/mTOR/S6K1 pathway (36, 37).

PD-L1 is a single-pass transmembrane protein with a 31 amino acid C-terminal cytoplasmic domain and a 220 amino acid ectodomain composed of IgV- and IgC-like domains (Fig. 1A; ref. 38). Because it is the PD-L1 IgV-like domain that binds PD-1 on T cells, PD-L1 must be properly translocated to the plasma membrane to engage PD-1 and exert its immunosuppressive effects (Fig. 1B; refs. 39–41). As a result, PD-L1 cellular compartmentalization is closely linked with its checkpoint activity.

Figure 1.

A, PD-L1 is a type I transmembrane protein with IgV- and IgC-like ectodomains, a transmembrane domain and a small cytoplasmic tail. B, PD-L1 binding to PD-1 leads to the phosphorylation of the tyrosine-based switch motif (ITSM) of the PD-1 intracellular domain. Phosphorylated ITSM recruits phosphatases such as the Src homology region 2 domain-containing phosphatases (SHP), which likely play a role in downregulating signaling pathways that lie downstream of CD28 and TCR molecules ligated by CD80 and antigenic peptide/MHC-I complex, respectively. The ZAP70, PI3K–AKT, and RAS pathways have been shown to be attenuated in response to PD-1 signaling. The specific protein targets of the phosphatase(s) action are not fully understood. The complex signaling pathways downstream of PD-1 have been extensively reviewed elsewhere (40, 41). TCR, T-cell receptor.

Figure 1.

A, PD-L1 is a type I transmembrane protein with IgV- and IgC-like ectodomains, a transmembrane domain and a small cytoplasmic tail. B, PD-L1 binding to PD-1 leads to the phosphorylation of the tyrosine-based switch motif (ITSM) of the PD-1 intracellular domain. Phosphorylated ITSM recruits phosphatases such as the Src homology region 2 domain-containing phosphatases (SHP), which likely play a role in downregulating signaling pathways that lie downstream of CD28 and TCR molecules ligated by CD80 and antigenic peptide/MHC-I complex, respectively. The ZAP70, PI3K–AKT, and RAS pathways have been shown to be attenuated in response to PD-1 signaling. The specific protein targets of the phosphatase(s) action are not fully understood. The complex signaling pathways downstream of PD-1 have been extensively reviewed elsewhere (40, 41). TCR, T-cell receptor.

Close modal

IHC staining of human tissue, using mAbs that recognize the PD-L1 extracellular domain, indicate a predominantly plasma membrane localization in malignant epithelial cells as well as a variety of stromal cells across multiple tumor types (42). It is difficult to evaluate the cellular distributions in IHC studies of fixed-formalin paraffin-embedded (FFPE) tissue because the processing required for antigen retrieval can alter staining patterns (43, 44).

Studies of cultured cells, which are not subject to the same technical constraints as FFPE studies, have demonstrated PD-L1 at the plasma membrane and in intracellular compartments of several cancer cell lines (45–47). Part of the intracellular pool of PD-L1 is newly synthesized protein in the endoplasmic reticulum (ER) and Golgi that is being modified into the mature protein along the secretory pathway (48, 49). In addition to the biosynthetic-secretory pathway, a perinuclear staining pattern in some of these cells suggests PD-L1 may also be present in the endosomal recycling compartment (45, 47, 50). This is supported by studies that show PD-L1 colocalization with markers of the endosomal recycling compartment, such as the transferrin receptor and/or Rab11 as well as analysis of the PD-L1 proximal proteome using APEX2 proximity mapping (47, 50–53).

Getting to the plasma membrane

PD-L1 is primarily N-glycosylated, through a process that begins in the ER with the transfer of oligosaccharides to specific asparagine residues (N35, N195, N200, and N219) of the PD-L1 extracellular domain in the ER lumen (54–56). The core glycan is then remodeled and further processed in the ER and Golgi apparatus where it acquires complex glycan modifications before PD-L1 is translocated to the plasma membrane. At the plasma membrane, PD-L1 is anchored by the electrostatic interaction between the positively charged amino acids of its cytoplasmic domain and the phospholipids of the plasma membrane (57). Aberrant glycosylation during PD-L1 maturation can lead to PD-L1 accumulation in the ER, and reduced translocation to the cell surface. Abnormally glycosylated PD-L1 can be removed through ER-associated degradation (ERAD; ref. 49).

Maintenance at the plasma membrane

At the cell surface, PD-L1 is constantly endocytosed and recycled (Fig. 2; refs. 47, 51). Recent APEX2 proximity mapping of the PD-L1 proximal proteome supports constitutive traffic of PD-L1 through the compartments of the general endosomal recycling system (47). This provides a mechanism for cells to rapidly, reversibly, and precisely regulate expression of PD-L1 on the plasma membrane (58). PD-L1 endocytosis appears to occur constitutively and does not require ligand binding. There is some evidence showing that PD-L1 is internalized through a dynamin- and clathrin-dependent process (59–61).

Figure 2.

PD-L1 trafficking. PD-L1 is trafficked from the ER and targeted to the plasma membrane. The dominant PD-L1 variant is inserted into the plasma membrane, whereas alternatively spliced variants that lack the transmembrane domain are released as soluble PD-L1 into the extracellular space. At the plasma membrane, PD-L1 is internalized in a dynamin-dependent process, and, in certain contexts, undergoes ARF6- and TRAPPC4-mediated recycling. PD-L1 can be polyubiquitinated by the E3 ligases SPOP and the GSK3α/β-recruited ARIH1 and βTRCP. Polyubiquitinated PD-L1 is destined for proteasomal degradation. Glycosylation by STT3 or B3GNT3 inhibits PD-L1 polyubiquitination and degradation. Similarly, the deubiquitinating enzymes CSN5 and USP22 stabilize PD-L1. PD-L1 can also be monoubiquitinated at the plasma membrane and targeted for lysosomal degradation. Palmitoylation of the PD-L1 intracellular domain by DHHC3 inhibits monoubiquitination. CMTM6 binds PD-L1 and blocks its ubiquitination by the E3 ligase STUB1. Monoubiquitinated PD-L1 is recognized by HRS (part of the ESCRT-0 complex) and sorted into the MVB lumen by the ESCRT accessory protein, ALIX. Rab27a mediates the fusion of MVBs to the plasma membrane, which results in the release of exosomal PD-L1. What determines the fate of the MVB, fusion with the plasma membrane or fusion with lysosomes is not understood. Cell surface proteases, ADAM-10 and ADAM-17 can cleave PD-L1 freeing a soluble PD-L1 fragment.

Figure 2.

PD-L1 trafficking. PD-L1 is trafficked from the ER and targeted to the plasma membrane. The dominant PD-L1 variant is inserted into the plasma membrane, whereas alternatively spliced variants that lack the transmembrane domain are released as soluble PD-L1 into the extracellular space. At the plasma membrane, PD-L1 is internalized in a dynamin-dependent process, and, in certain contexts, undergoes ARF6- and TRAPPC4-mediated recycling. PD-L1 can be polyubiquitinated by the E3 ligases SPOP and the GSK3α/β-recruited ARIH1 and βTRCP. Polyubiquitinated PD-L1 is destined for proteasomal degradation. Glycosylation by STT3 or B3GNT3 inhibits PD-L1 polyubiquitination and degradation. Similarly, the deubiquitinating enzymes CSN5 and USP22 stabilize PD-L1. PD-L1 can also be monoubiquitinated at the plasma membrane and targeted for lysosomal degradation. Palmitoylation of the PD-L1 intracellular domain by DHHC3 inhibits monoubiquitination. CMTM6 binds PD-L1 and blocks its ubiquitination by the E3 ligase STUB1. Monoubiquitinated PD-L1 is recognized by HRS (part of the ESCRT-0 complex) and sorted into the MVB lumen by the ESCRT accessory protein, ALIX. Rab27a mediates the fusion of MVBs to the plasma membrane, which results in the release of exosomal PD-L1. What determines the fate of the MVB, fusion with the plasma membrane or fusion with lysosomes is not understood. Cell surface proteases, ADAM-10 and ADAM-17 can cleave PD-L1 freeing a soluble PD-L1 fragment.

Close modal

Once internalized, a large portion of PD-L1 is recycled back in about 15 minutes, with about 70% of cycling PD-L1 found on the cell surface at equilibrium (45, 47, 51). Blocking recycling with primaquine rapidly depletes PD-L1 on the plasma membrane. PD-L1 recycling can be regulated by CKLF-like MARVEL transmembrane domain containing protein 6 (CMTM6), which is primarily located at the plasma membrane and in recycling endosomes, where it interacts with PD-L1 (51, 62). In CMTM6 knockout cells, PD-L1 is rapidly depleted from the cell surface compared with wild-type cells. However, in the presence of lysosomal inhibitors, the loss of surface PD-L1 is relatively small, suggesting that rerouting to the lysosome rather than slowed-down recycling is the main cause of reduced PD-L1 at the plasma membrane in the absence of CMTM6. The balance between recycling and lysosomal degradation is also central to the regulation of PD-L1 trafficking by trafficking protein particle complex (TRAPPC4) and Huntington-interacting protein 1-related (HIP1R; Fig. 2). TRAPPC4, like CMTM6, promotes PD-L1 recycling away from the lysosome (52). HIP1R, on the other hand, favors the degradative pathway and its ablation leads to increased distribution of PD-L1 from Rab7+ late endosomes to Rab11+ recycling endosomes (53). Distinct from this recycling–degradation modality, the small GTPase Arf6 has been shown to exert its effect solely on PD-L1 recycling (45). In platelet-derived growth factor–treated cells, ablating Arf6 or its downstream effector AMAP1 leads to reduced PD-L1 recycling and, consequently, lower cell surface levels of PD-L1 without changes in total protein.

Trafficking of soluble and exosomal PD-L1

In addition to the endomembrane system, PD-L1 has been found in the sera of patients and the supernatant of tumor cell lines and immune cells (63–65). Mechanisms by which PD-L1 could be secreted from cells include exosomal secretion, expression of truncated variants as well as proteolytic cleavage and shedding.

PD-L1+ exosomes are formed through an endosomal sorting complex required for transport (ESCRT)–dependent process, and PD-L1–containing exosomal fractions colocalize with ESCRT0, a complex required for the initial recognition of cargo destined for multivesicular bodies (MVB; Fig. 2; refs. 66, 67). Once PD-L1 is localized to the limiting membrane of MVBs, it is sorted into the intraluminal vesicles of MVBs through the action of the ESCRT accessory protein, ALG-2–interacting protein X (ALIX; ref. 68). The final fusion and release of PD-L1+ exosomes require the activity of Rab27A, a protein that is essential for the docking of MVBs at the plasma membrane (67, 68). PD-L1 on exosomes can inhibit T-cell activation in vitro and in the draining lymph node, promoting tumor progression in murine models of prostate cancer and melanoma (66, 67). Although PD-L1 has also been detected on microvesicles, the regulation of this process has not been investigated (66).

Distinct from its exosomal counterpart, soluble PD-L1 does not require membrane integration to be secreted (Fig. 2). Some of the soluble variants of PD-L1 result from alternative splicing that excludes the transmembrane domain encoded by exon 5. One such variant, identified in several cancers as well as normal placenta and myeloid cells, results from a read-through into intron 4 coupled with an in-frame stop codon and an alternative polyadenylation signal (69, 70). This variant appears to multimerize and lacks the ability to inhibit T cells both in vitro and in vivo. Instead, it was shown to act as a PD-1 antagonist, blocking the suppressive activity of cell-bound or exosome-bound PD-L1 (71). Other secreted splice variants, detected in melanoma cell lines and patient plasma, appear to result from aberrant splicing that leads to a premature stop codon well before exon 5 (72). In certain patients with NSCLC, two unique secreted PD-L1 variants that are associated with resistance to anti–PD-L1 therapy have been described previously (73). Both isoforms lack the transmembrane domain, and while one variant is devoid of the intracellular domain, the other maintains portions of the cytoplasmic domain (coded by exon 7) due to in-frame skipping of exons 5 and 6.

It is unlikely that all soluble PD-L1 variants arise due to alternative splicing. Several immune modulators can be released from the cell surface through the action of extracellular enzymes, and there is some evidence that PD-L1 may also be similarly shed (74). In vitro studies have shown that PD-L1 can be removed from the cell surface by matrix metallopeptidase 9 (MMP-9) and MMP-13 (75, 76). In addition, MMP inhibition results in reduced levels of soluble PD-L1 in cells engineered to overexpress PD-L1 (63). It is not clear whether these MMPs catalyze controlled proteolysis events that can result in a physiologically relevant PD-L1 fragment. In one study, the addition of MMP-13 to purified PD-L1 resulted in a reduction in full-size PD-L1 levels without the appearance of any discernable lower molecular weight variants, suggesting breakdown to nonfunctional peptides (77). On the other hand, in certain cancer cell lines, the cell surface proteases, ADAM metallopeptidase domain 10 (ADAM-10) and ADAM-17 have been shown to cleave PD-L1 at a yet unknown site(s) freeing a soluble 37 kDa PD-L1 fragment and an unstable, 18 kDa intracellular fragment. The soluble fragment appears to retain its ability to bind PD-1 and suppress CD8+ T-cell activity in vitro (78, 79).

PD-L1 degradation

Integral plasma membrane proteins, such as PD-L1, are not directly accessible to the proteasome. Instead, cells rely on ubiquitination and membrane trafficking events to target these proteins from the plasma membrane to the MVB for lysosomal fusion and degradation. Ubiquitination involves the attachment of ubiquitin, a 76 amino acid peptide, to lysine residues of substrate proteins in a multistep process (80). The final step of ubiquitin attachment is facilitated by enzymes known as E3 ubiquitin ligases, which recognize the substrate protein and facilitate the transfer of ubiquitin. Conversely, deubiquitinating enzymes catalyze the removal of ubiquitin from target proteins. Ubiquitin itself can be ubiquitinated at its seven lysine residues, resulting in an ubiquitin chain. A protein may be tagged by a single ubiquitin molecule (monoubiquitination) or an ubiquitin chain (polyubiquitination). The type of ubiquitination will influence the fate of the modified protein. For instance, membrane proteins destined for lysosomal degradation are usually marked by multiple monoubiquitination or Lysine-63–linked polyubiquitination (81).

Monoubiquitinated PD-L1 can be destined for lysosomal degradation in an ESCRT-dependent process (50). Although the specific site of monoubiquitination remains to be identified, it likely occurs on the lysines of the PD-L1 cytoplasmic domain, as palmitoylation at the cytoplasmic residue C272 inhibits monoubiquitination (Fig. 3; refs. 50, 82). Likewise, CMTM6, which is predominantly present within the plasma membrane, protects PD-L1 from degradation presumably by masking the STUB1 E3 ubiquitin ligase recognition site in the transmembrane-intracellular domain of PD-L1 (Fig. 2; refs. 51, 62). Another CMTM family member, CMTM4, similarly inhibits PD-L1 degradation (62). CMTM4 activity, unlike that of CMTM6, can be downregulated by AMPK, which disrupts the PD-L1–CMTM4 interaction by phosphorylating PD-L1 (83). PD-L1 may also be targeted to the lysosome in a ubiquitin-independent manner: in a number of cancer cell lines, HIP1R, which can recruit ALIX through its dileucine motif, can directly bind PD-L1 and direct it to MVBs for lysosomal degradation (Fig. 2; ref. 53). What determines the fate of the MVB, fusion with the plasma membrane or fusion with lysosomes, is not understood.

Figure 3.

Posttranslational modifications of PD-L1. PD-L1 is modified at several residues. Enzymes responsible for posttranslational modifications are shown with the functional consequence of the modification shown in red for those that promote PD-L1 degradation and green for those that have a stabilizing effect on PD-L1. PD-L1 glycosylation is necessary for proper maturation. It also stabilizes PD-L1 by antagonizing GSK3β-driven phosphorylation, which recruits βTRCP and leads to PD-L1 degradation. Phosphorylation by JAK1 and NEK2 promotes glycosylation while AMPK-mediated phosphorylation leads to aberrant glycosylation. Phosphorylation of the intracellular domain of PD-L1 by GSK3α recruits ARIH1, which mediates ubiquitination at K271/281 and promotes PD-L1 degradation. Conversely, phosphorylation by CK2 and palmitoylation by DHHC3 inhibits PD-L1 degradation. Acetylation at K263 maintains PD-L1 distribution in the cytosol by inhibiting nuclear translocation.

Figure 3.

Posttranslational modifications of PD-L1. PD-L1 is modified at several residues. Enzymes responsible for posttranslational modifications are shown with the functional consequence of the modification shown in red for those that promote PD-L1 degradation and green for those that have a stabilizing effect on PD-L1. PD-L1 glycosylation is necessary for proper maturation. It also stabilizes PD-L1 by antagonizing GSK3β-driven phosphorylation, which recruits βTRCP and leads to PD-L1 degradation. Phosphorylation by JAK1 and NEK2 promotes glycosylation while AMPK-mediated phosphorylation leads to aberrant glycosylation. Phosphorylation of the intracellular domain of PD-L1 by GSK3α recruits ARIH1, which mediates ubiquitination at K271/281 and promotes PD-L1 degradation. Conversely, phosphorylation by CK2 and palmitoylation by DHHC3 inhibits PD-L1 degradation. Acetylation at K263 maintains PD-L1 distribution in the cytosol by inhibiting nuclear translocation.

Close modal

Besides quality control, cells may target nascent proteins for proteasomal degradation as a way of regulating surface expression (81). This homoeostatic function is apparent in the fluctuation of PD-L1 protein level during cell-cycle progression observed in multiple human cancer cell lines: PD-L1 levels peak in M- and early G1-phases, followed by a rapid reduction in late G1- and S-phases (84). The mechanism involves cyclin D–CDK4, which is primarily active in the G1–S-phase. Cyclin D–CDK4 phosphorylates and stabilizes speckle-type POZ protein (SPOP), which is a cullin-3–based E3 ligase substrate-recruiting adaptor protein. In turn, SPOP mediates PD-L1 polyubiquitination and degradation. This interaction is dependent on the PD-L1 cytoplasmic domain, deletion of which disrupts binding of PD-L1 to SPOP and renders PD-L1 resistant to SPOP-meditated polyubiquitination.

Another example of PD-L1 regulation by proteasomal degradation involves the GSK kinases, which lie downstream of the PI3K/AKT signaling axis (Fig. 3). In response to EGFR inhibition, GSKα is activated and phosphorylates PD-L1 on the cytoplasmic tail at positions Ser279/283 (85). This leads to the recruitment of the E3 ubiquitin ligase ARIH1, which ubiquitinates and targets PD-L1 for degradation. GSK3β, which shares 97% catalytic domain sequence identity with GSKα, also phosphorylates PD-L1, but it does so on the extracellular domain at residues T180 and S184 (54). βTRCP, a subunit of the SCF E3 ligase complex, is then recruited to PD-L1. It is important to note that to access these PD-L1 ectodomain residues, which would be positioned in the ER lumen, GSK3β and βTRCP must localize either in the lumen or luminal face of the ER membrane. However, there is little to no evidence showing such a localization (86). It is rather more likely that these interactions occur only after selection and initial translocation of PD-L1 into the cytosol, which would then allow access to the GSK3β and βTRCP. While the signal for substrate recognition is not clear, there is some evidence suggesting glycosylation status may play a role.

Glycosylation of PD-L1 at N192, N200, and N219 prevents phosphorylation by GSK3β, and as a result blocks βTRCP recruitment, and proteasomal degradation (54). The enzyme(s) responsible for PD-L1 glycosylation, in the context of βTRCP-mediated degradation, have not been investigated. Nonetheless, the N-glycosyltransferases STT3 and B3GNT3 have been shown to glycosylate PD-L1 and protect it from degradation in separate studies using different cell types (55, 87). Modulating glycosyltransferase activity can be a way of regulating PD-L1 levels posttranslationally. For instance, EGF-induced B3GNT3 upregulation and IL6-induced STT3 recruitment can result in reduced PD-L1 degradation (55, 88). NIMA-related kinase 2 (NEK2), which phosphorylates PD-L1 at T194/T210, similarly promotes PD-L1 glycosylation and stability (89). Direct changes to deubiquitinating enzymes such as CSN5 and USP22 can also affect PD-L1 stability (90, 91).

The potential of endocytic trafficking either as a therapeutic target or a means of drug delivery has been extensively studied in several diseases (92, 93). With respect to targeting PD-L1, current treatment options have focused on blocking its interaction with PD-1 using PD-L1–specific mAbs (94). Although it is known that certain therapeutic PD-L1–specific mAbs can be internalized after binding PD-L1, mAb binding does not appear to alter PD-L1 trafficking (47, 95). There is, however, considerable information regarding ways of therapeutically targeting PD-L1 trafficking in the preclinical setting. Most of these have focused on inducing PD-L1 degradation by either targeting it to the lysosome or interfering with its maturation in the ER.

STM108, an antibody that binds and targets glycosylated PD-L1 to the lysosome, inhibits tumor growth in a syngeneic mouse model of triple-negative breast cancer in which the cancer cells have been made to express human PD-L1 (55). Lysosome-targeting peptides that bind PD-L1 and lead to its degradation are also being studied. For instance, a rationally designed peptide called PD-LYSO targets PD-L1 to the lysosome, whereas a cell-penetrating peptide called PD-PALM blocks PD-L1 palmitoylation leading to increased degradation in the lysosome (50, 53). Small molecules have also been used to reroute PD-L1 to the lysosome. An inhibitor of Sigma1 induces PD-L1 degradation via autophagy in triple-negative breast and androgen-independent prostate cancer cell lines (96). SA-49, an alpperine derivate, decreased the protein level of PD-L1 in NSCLC cells and the Lewis lung tumor model (46). However, its effect appears to result from a general increase in lysosomal activity, rather than a targeted removal of PD-L1. A similarly nonexclusive approach has been used to induce and target misfolded PD-L1 to ERAD by using SAFit 1 and 2 to inhibit FKBP5, a cochaperone with protein folding activity (97). Likewise, resveratrol inhibits enzymes that process glycosylated PD-L1 and promotes ER retention. It is not fully clear whether ERAD removes this population of PD-L1 (48). Modulating the activity of kinases such as AMPK, GSKα/β, and NEK2 that regulate PD-L1 stability is also being explored as a way to regulate PD-L1 levels (98).

Besides novel ways of targeting PD-L1, another approach has been to enhance the effectiveness of existing antibody treatment by modulating PD-L1 trafficking. Examples of this include the use of prochlorperazine (PCZ), a clinically available drug known to inhibit dynamin (59). By decreasing PD-L1 internalization and, consequently increasing surface expression, PCZ was shown to enhance the effect of the PD-L1–specific mAb avelumab in a model of colon cancer in mice. The drug tiplaxtinin, which inhibits PAI-1–mediated PD-L1 internalization, similarly synergizes with anti–PD-L1 therapy in a murine model of melanoma (60). It should be noted that both studies used anti–PD-L1 with an intact Fc region capable of inducing antibody-dependent cellular cytotoxicity (ADCC). Indeed, the improved efficacy of avelumab in the presence of PCZ is due to increased natural killer cell–mediated ADCC (99). It is not clear whether a similar approach would be useful for other PD-L1–specific antibodies such as durvalumab and atezolizumab, which lack a functional Fc region, and therefore do not induce ADCC. Combination of cyclin-dependent kinase (CDK) inhibitors with anti–PD-L1 therapy is also being extensively investigated. CDK 4/6 inhibitors such as palbociclib and abemaciclib have been shown to reduce tumor cell growth by cell-cycle arrest and positively impact T-cell activation, leading to increased memory T-cell phenotype and enhanced effector functions (84, 100, 101). However, CDK4/6 inhibition increases PD-L1 expression as previously discussed, therefore has limited efficacy alone (84). The increased PD-L1 can be offset by immune checkpoint inhibitor (ICI) therapy. Consistent with this, CDK inhibition has been shown to prime tumors for anti–PD-L1 therapy to markedly improve overall survival in several preclinical models (84, 100).

The clinical application of ICIs in treating cancer has vaulted the study of immune coinhibitors such as PD-L1 from the purview of basic immunology to the center stage of translational medicine. While there was, initially, great enthusiasm for ICIs directed against PD-L1, this has been tempered by low response rates and the associated suboptimal predictive value of PD-L1 expression. Consequently, there is a growing appreciation for exploring the basic biology of PD-L1 with the perspective that this may lead to a better understanding of the potential as well as the limitations of ICIs. Accordingly, a body of work focusing on mechanisms regulating PD-L1 expression at the transcript and protein level has emerged. Yet, the dynamic nature of an immune response from initiation to resolution makes it crucial to also investigate rapid regulatory mechanisms, such as changes in PD-L1 trafficking, which might play a role in modulating ICI activity.

As described in this review, a relatively small, but growing, number of studies have established PD-L1 as a constitutively recycling receptor that can be localized to different cellular compartments. What emerges from these findings is a model of posttranslational regulation centered around modulating total levels of PD-L1 through ubiquitin-mediated degradation in the lysosome and proteasome. A PD-L1 C-terminal degradation sequence appears to be crucial in determining the ubiquitination and, therefore, the half-life of PD-L1; CMTM4/6, glycosylation, and palmitoylation shield access to this putative degron(s), while cyclin D–CDK4 stabilizes the ubiquitin ligase that recognizes it. By controlling access to a native degradation sequence in such a manner, cells may rapidly upregulate or downregulate total PD-L1 levels (102). It is not clear whether the PD-L1 proteasomal degradation pathways that have been reported so far are distinct from the conventional quality control mechanism that operates in the ER. If PD-L1 can indeed be extracted from the plasma membrane or other intracellular compartments and targeted to the proteasome, it will be important to elucidate how this process unfolds, especially in relation to the conventional lysosomal pathway. Given that frequently used markers of PD-L1 proteasomal degradation such as K48-linked ubiquitination (103) and inhibition by the drug MG132 (104) can also affect MVB maturation and lysosomal degradation, it will be essential to use more refined methods to identify the degradative fate of PD-L1.

Besides regulating degradation, changes in protein trafficking can reversibly dictate protein function by translocating membrane proteins from intracellular stores to the plasma membrane. For instance, the immune checkpoint receptor, CTLA-4 can be translocated to the cell surface in response to T-cell receptor signaling at the immune synapse (105). PD-L1 has also been shown to accumulate at the immunological synapse formed by the interaction of PD-1+ T cells and activated dendritic cells (106).

It is plausible that PD-1/PD-L1 interactions immobilize PD-L1 at the plasma membrane and inhibit its endocytosis. This would then lead to a rapid accumulation at the plasma membrane, as exocytosis would still be maintained while PD-L1 molecules that would have otherwise been internalized and routed to degradative endolysosomal compartments would remain at the cell surface. In such a scenario, free PD-L1, owing to its constitutive endocytosis and lysosomal degradation, will not be abundantly available at the cell surface to be bound by either diagnostic or therapeutic antibodies. Yet, when bound by PD-1, PD-L1 would accumulate at the plasma membrane and exert its immune checkpoint function. This context-dependent PD-L1 surface expression may partially explain why there are low response rates to anti–PD-L1 therapy, and why certain tumors that are classified as PD-L1 using diagnostic antibodies are found to respond to anti–PD-1 therapy (13, 107). This is supported by preclinical studies that show an enhanced response to anti–PD-L1 therapy when PD-L1 surface expression is increased through the inhibition of PD-L1 endocytosis and/or degradation (59, 60, 84).

Finally, there is emerging evidence supporting PD-L1 having cell-intrinsic functions in addition to its role as a transmembrane ligand for PD1 in regulation of epithelial–mesenchymal transition, cell migration, mutant EGFR turnover, extracellular vesicle formation, resistance to chemotherapeutic agents as well as IFN-mediated cell toxicity (47, 51, 108–111). These functions may be dependent on subcellular PD-L1 localization, and therefore trafficking of PD-L1 may contribute to modulation of PD-L1–dependent biology beyond controlling PD-L1 density in the plasma membrane. In addition, therapeutic anti–PD-L1 treatment of EGFR-mutant cells but not EGFR wild-type cells, phenocopies the effect of PD-L1 knockout on cell migration and extracellular vesicle secretion (47), an intriguing finding considering EGFR-mutant tumors respond poorly to ICI therapy. Further studies are required to determine the full impact of PD-L1 cell-intrinsic biology and how ICI therapy affects these functions.

Much of the work discussed in this review has investigated PD-L1 trafficking in tumor cell lines. While immune cells such as dendritic cells also express PD-L1, we know very little about how PD-L1 is trafficked in these cells. Elucidating the basic mechanisms of PD-L1 trafficking in immune cell models is essential as it can be a steppingstone to addressing physiologically pertinent questions such as how PD-L1 trafficking is regulated by immune receptors and ligands that are present in the immune microenvironment. The relation between PD-L1 localization and its emerging tumor cell–intrinsic functions is another area that awaits investigation. Addressing these questions—using cell biological, biochemical, and advanced imaging tools that have already been refined to study protein trafficking—may open the door not only to new paradigms of immune checkpoint regulation, but also to the development of novel therapeutic strategies.

N.K. Altorki reports grants from AstraZeneca and Jansen; personal fees from AstraZeneca and Regeneron outside the submitted work. T.E. McGraw reports grants from NCI during the conduct of the study; nonfinancial support from Medimmune/AstraZeneca, and grants from Johnson and Johnson outside the submitted work. No disclosures were reported by the other authors.

N.K. Altorki and T.E. MsGraw are supported in part by NCI UG3 CA244697. E.Y. Lemma is supported by a diversity supplement to CA244697.

1.
Dong
H
,
Zhu
G
,
Tamada
K
,
Chen
L
.
B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion
.
Nat Med
1999
;
5
:
1365
9
.
2.
Freeman
GJ
,
Long
AJ
,
Iwai
Y
,
Bourque
K
,
Chernova
T
,
Nishimura
H
, et al
.
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation
.
J Exp Med
2000
;
192
:
1027
34
.
3.
Latchman
Y
,
Wood
CR
,
Chernova
T
,
Chaudhary
D
,
Borde
M
,
Chernova
I
, et al
.
PD-L2 is a second ligand for PD-1 and inhibits T cell activation
.
Nat Immunol
2001
;
2
:
261
8
.
4.
Chemnitz
JM
,
Parry
RV
,
Nichols
KE
,
June
CH
,
Riley
JL
.
SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation
.
J Immunol
2004
;
173
:
945
54
.
5.
Christofides
A
,
Katopodi
X-L
,
Cao
C
,
Karagkouni
D
,
Aliazis
K
,
Yenyuwadee
S
, et al
.
SHP-2 and PD-1-SHP-2 signaling regulate myeloid cell differentiation and antitumor responses
.
Nat Immunol
2023
;
24
:
55
68
.
6.
Patsoukis
N
,
Bardhan
K
,
Chatterjee
P
,
Sari
D
,
Liu
B
,
Bell
LN
, et al
.
PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation
.
Nat Commun
2015
;
6
:
6692
.
7.
Rudd
CE
,
Schneider
H
.
Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling
.
Nat Rev Immunol
2003
;
3
:
544
56
.
8.
Wei
SC
,
Duffy
CR
,
Allison
JP
.,
Fundamental mechanisms of immune checkpoint blockade therapy
.
Cancer Discov
2018
;
8
:
1069
86
.
9.
Okazaki
T
,
Honjo
T
.
PD-1 and PD-1 ligands: from discovery to clinical application
.
Int Immunol
2007
;
19
:
813
24
.
10.
Sun
C
,
Mezzadra
R
,
Schumacher
TN
.
Regulation and function of the PD-L1 checkpoint
.
Immunity
2018
;
48
:
434
52
.
11.
Cha
J-H
,
Chan
L-C
,
Li
C-W
,
Hsu
JL
,
Hung
M-C
.
Mechanisms controlling PD-L1 expression in cancer
.
Mol Cell
2019
;
76
:
359
70
.
12.
Upadhaya
S
,
Neftelinov
ST
,
Hodge
J
,
Campbell
J
.
Challenges and opportunities in the PD1/PDL1 inhibitor clinical trial landscape
.
Nat Rev Drug Discov
2022
;
21
:
482
3
.
13.
Sun
J-Y
,
Zhang
D
,
Wu
S
,
Xu
M
,
Zhou
X
,
Lu
X-J
, et al
.
Resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms, predictive factors, and future perspectives
.
Biomark Res
2020
;
8
:
35
.
14.
Zerdes
I
,
Matikas
A
,
Bergh
J
,
Rassidakis
GZ
,
Foukakis
T
.
Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations
.
Oncogene
2018
;
37
:
4639
61
.
15.
Wang
Y
,
Wang
H
,
Yao
H
,
Li
C
,
Fang
J-Y
,
Xu
J
.
Regulation of PD-L1: emerging routes for targeting tumor immune evasion
.
Front Pharmacol
2018
;
9
:
536
.
16.
Kornepati
AVR
,
Vadlamudi
RK
,
Curiel
TJ
.
Programmed death ligand 1 signals in cancer cells
.
Nat Rev Cancer
2022
;
22
:
174
89
.
17.
Chen
S
,
Crabill
GA
,
Pritchard
TS
,
McMiller
TL
,
Wei
P
,
Pardoll
DM
, et al
.
Mechanisms regulating PD-L1 expression on tumor and immune cells
.
J Immunother Cancer
2019
;
7
:
305
.
18.
Quandt
D
,
Jasinski-Bergner
S
,
Müller
U
,
Schulze
B
,
Seliger
B
.
Synergistic effects of IL-4 and TNFalpha on the induction of B7-H1 in renal cell carcinoma cells inhibiting allogeneic T cell proliferation
.
J Transl Med
2014
;
12
:
151
.
19.
Noman
MZ
,
Desantis
G
,
Janji
B
,
Hasmim
M
,
Karray
S
,
Dessen
P
, et al
.
PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation
.
J Exp Med
2014
;
211
:
781
90
.
20.
Onishi
H
,
Fujimura
A
,
Oyama
Y
,
Yamasaki
A
,
Imaizumi
A
,
Kawamoto
M
, et al
.
Hedgehog signaling regulates PDL-1 expression in cancer cells to induce anti-tumor activity by activated lymphocytes
.
Cell Immunol
2016
;
310
:
199
204
.
21.
Peng
J
,
Hamanishi
J
,
Matsumura
N
,
Abiko
K
,
Murat
K
,
Baba
T
, et al
.
Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-kappaB to foster an immunosuppressive tumor microenvironment in ovarian cancer
.
Cancer Res
2015
;
75
:
5034
45
.
22.
Dovedi
SJ
,
Adlard
AL
,
Lipowska-Bhalla
G
,
McKenna
C
,
Jones
S
,
Cheadle
EJ
, et al
.
Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade
.
Cancer Res
2014
;
74
:
5458
68
.
23.
Chen
N
,
Fang
W
,
Zhan
J
,
Hong
S
,
Tang
Y
,
Kang
S
, et al
.
Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation
.
J Thorac Oncol
2015
;
10
:
910
23
.
24.
Zhang
N
,
Zeng
Y
,
Du
W
,
Zhu
J
,
Shen
D
,
Liu
Z
, et al
.
The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer
.
Int J Oncol
2016
;
49
:
1360
8
.
25.
Lastwika
KJ
,
Wilson
W
,
Li
QK
,
Norris
J
,
Xu
H
,
Ghazarian
SR
, et al
.
Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer
.
Cancer Res
2016
;
76
:
227
38
.
26.
Feng
D
,
Qin
B
,
Pal
K
,
Sun
L
,
Dutta
S
,
Dong
H
, et al
.
BRAF(V600E)-induced, tumor intrinsic PD-L1 can regulate chemotherapy-induced apoptosis in human colon cancer cells and in tumor xenografts
.
Oncogene
2019
;
38
:
6752
66
.
27.
Casey
SC
,
Tong
L
,
Li
Y
,
Do
R
,
Walz
S
,
Fitzgerald
KN
, et al
.
MYC regulates the antitumor immune response through CD47 and PD-L1
.
Science
2016
;
352
:
227
31
.
28.
Micevic
G
,
Thakral
D
,
McGeary
M
,
Bosenberg
MW
.
PD-L1 methylation regulates PD-L1 expression and is associated with melanoma survival
.
Pigment Cell Melanoma Res
2019
;
32
:
435
40
.
29.
Asgarova
A
,
Asgarov
K
,
Godet
Y
,
Peixoto
P
,
Nadaradjane
A
,
Boyer-Guittaut
M
, et al
.
PD-L1 expression is regulated by both DNA methylation and NF-kB during EMT signaling in non-small cell lung carcinoma
.
Oncoimmunology
2018
;
7
:
e1423170
.
30.
Wang
YF
,
Liu
F
,
Sherwin
S
,
Farrelly
M
,
Yan
XG
,
Croft
A
, et al
.
Cooperativity of HOXA5 and STAT3 is critical for HDAC8 inhibition-mediated transcriptional activation of PD-L1 in human melanoma cells
.
J Invest Dermatol
2018
;
138
:
922
32
.
31.
Martin
AM
,
Nirschl
TR
,
Nirschl
CJ
,
Francica
BJ
,
Kochel
CM
,
van Bokhoven
A
, et al
.
Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance
.
Prostate Cancer Prostatic Dis
2015
;
18
:
325
32
.
32.
Atefi
M
,
Avramis
E
,
Lassen
A
,
Wong
DJL
,
Robert
L
,
Foulad
D
, et al
.
Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma
.
Clin Cancer Res
2014
;
20
:
3446
57
.
33.
Chen
L
,
Gibbons
DL
,
Goswami
S
,
Cortez
MA
,
Ahn
Y-H
,
Byers
LA
, et al
.
Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression
.
Nat Commun
2014
;
5
:
5241
.
34.
Xu
S
,
Tao
Z
,
Hai
B
,
Liang
H
,
Shi
Y
,
Wang
T
, et al
.
miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint
.
Nat Commun
2016
;
7
:
11406
.
35.
Kataoka
K
,
Shiraishi
Y
,
Takeda
Y
,
Sakata
S
,
Matsumoto
M
,
Nagano
S
, et al
.
Aberrant PD-L1 expression through 3'-UTR disruption in multiple cancers
.
Nature
2016
;
534
:
402
6
.
36.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
.
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
8
.
37.
Crane
CA
,
Panner
A
,
Murray
JC
,
Wilson
SP
,
Xu
H
,
Chen
L
, et al
.
PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer
.
Oncogene
2009
;
28
:
306
12
.
38.
UniProt: a worldwide hub of protein knowledge
.
Nucleic Acids Res
2019
;
47
:
D506
D515
.
39.
Freeman
GJ
.
Structures of PD-1 with its ligands: sideways and dancing cheek to cheek
.
Proc Natl Acad Sci U S A
2008
;
105
:
10275
6
.
40.
Sharpe
AH
,
Pauken
KE
.
The diverse functions of the PD1 inhibitory pathway
.
Nat Rev Immunol
2018
;
18
:
153
67
.
41.
Arasanz
H
,
Gato-Cañas
M
,
Zuazo
M
,
Ibañez-Vea
M
,
Breckpot
K
,
Kochan
G
, et al
.
PD1 signal transduction pathways in T cells
.
Oncotarget
2017
;
8
:
51936
45
.
42.
Parra
ER
,
Villalobos
P
,
Rodriguez-Canales
J
.
The multiple faces of programmed cell death ligand 1 expression in malignant and nonmalignant cells
.
Appl Immunohistochem Mol Morphol
2019
;
27
:
287
94
.
43.
Mahoney
KM
,
Sun
H
,
Liao
X
,
Hua
P
,
Callea
M
,
Greenfield
EA
, et al
.
PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells
.
Cancer Immunol Res
2015
;
3
:
1308
15
.
44.
Ezaki
T
.
Antigen retrieval on formaldehyde-fixed paraffin sections: its potential drawbacks and optimization for double immunostaining
.
Micron
2000
;
31
:
639
49
.
45.
Hashimoto
S
,
Furukawa
S
,
Hashimoto
A
,
Tsutaho
A
,
Fukao
A
,
Sakamura
Y
, et al
.
ARF6 and AMAP1 are major targets of KRAS and TP53 mutations to promote invasion, PD-L1 dynamics, and immune evasion of pancreatic cancer
.
Proc Natl Acad Sci U S A
2019
;
116
:
17450
9
.
46.
Zhang
N
,
Dou
Y
,
Liu
L
,
Zhang
X
,
Liu
X
,
Zeng
Q
, et al
.
SA-49, a novel aloperine derivative, induces MITF-dependent lysosomal degradation of PD-L1
.
EBioMedicine
2019
;
40
:
151
62
.
47.
Letian
A
,
Lemma
EY
,
Cavaliere
P
,
Dephoure
N
,
Altorki
NK
,
McGraw
TE
.
Proximity proteome mapping reveals PD-L1-dependent pathways disrupted by anti-PD-L1 antibody specifically in EGFR-mutant lung cancer cells
.
Cell Commun Signal
2023
;
21
:
58
.
48.
Verdura
S
,
Cuyàs
E
,
Cortada
E
,
Brunet
J
,
Lopez-Bonet
E
,
Martin-Castillo
B
, et al
.
Resveratrol targets PD-L1 glycosylation and dimerization to enhance antitumor T-cell immunity
.
Aging
2020
;
12
:
8
34
.
49.
Cha
J-H
,
Yang
W-H
,
Xia
W
,
Wei
Y
,
Chan
L-C
,
Lim
S-O
, et al
.
Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1
.
Mol Cell
2018
;
71
:
606
20
.
50.
Yao
H
,
Lan
J
,
Li
C
,
Shi
H
,
Brosseau
J-P
,
Wang
H
, et al
.
Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours
.
Nat Biomed Eng
2019
;
3
:
306
17
.
51.
Burr
ML
,
Sparbier
CE
,
Chan
Y-C
,
Williamson
JC
,
Woods
K
,
Beavis
PA
, et al
.
CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity
.
Nature
2017
;
549
:
101
5
.
52.
Ren
Y
,
Qian
Y
,
Ai
L
,
Xie
Y
,
Gao
Y
,
Zhuang
Z
, et al
.
TRAPPC4 regulates the intracellular trafficking of PD-L1 and antitumor immunity
.
Nat Commun
2021
;
12
:
5405
.
53.
Wang
H
,
Yao
H
,
Li
C
,
Shi
H
,
Lan
J
,
Li
Z
, et al
.
HIP1R targets PD-L1 to lysosomal degradation to alter T cell-mediated cytotoxicity
.
Nat Chem Biol
2019
;
15
:
42
50
.
54.
Li
C-W
,
Lim
S-O
,
Xia
W
,
Lee
H-H
,
Chan
L-C
,
Kuo
C-W
, et al
.
Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity
.
Nat Commun
2016
;
7
:
12632
.
55.
Li
C-W
,
Lim
S-O
,
Chung
EM
,
Kim
Y-S
,
Park
AH
,
Yao
J
, et al
.
Eradication of triple-negative breast cancer cells by targeting glycosylated PD-L1
.
Cancer Cell
2018
;
33
:
187
201
.
56.
Breitling
J
,
Aebi
M
.
N-linked protein glycosylation in the endoplasmic reticulum
.
Cold Spring Harb Perspect Biol
2013
;
5
:
a013359
.
57.
Wen
M
,
Cao
Y
,
Wu
B
,
Xiao
T
,
Cao
R
,
Wang
Q
, et al
.
PD-L1 degradation is regulated by electrostatic membrane association of its cytoplasmic domain
.
Nat Commun
2021
;
12
:
5106
.
58.
Maxfield
FR
,
McGraw
TE
.
Endocytic recycling
.
Nat Rev Mol Cell Biol
2004
;
5
:
121
32
.
59.
Chew
HY
,
De Lima
PO
,
Gonzalez Cruz
JL
,
Banushi
B
,
Echejoh
G
,
Hu
L
, et al
.
Endocytosis inhibition in humans to improve responses to ADCC-mediating antibodies
.
Cell
2020
;
180
:
895
914
.
60.
Tseng
Y-J
,
Lee
C-H
,
Chen
W-Y
,
Yang
J-L
,
Tzeng
H-T
.
Inhibition of PAI-1 blocks PD-L1 endocytosis and improves the response of melanoma cells to immune checkpoint blockade
.
J Invest Dermatol
2021
;
141
:
2690
8
.
61.
Gao
Y
,
Nihira
NT
,
Bu
X
,
Chu
C
,
Zhang
J
,
Kolodziejczyk
A
, et al
.
Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy
.
Nat Cell Biol
2020
;
22
:
1064
75
.
62.
Mezzadra
R
,
Sun
C
,
Jae
LT
,
Gomez-Eerland
R
,
de Vries
E
,
Wu
W
, et al
.
Identification of CMTM6 and CMTM4 as PD-L1 protein regulators
.
Nature
2017
;
549
:
106
10
.
63.
Chen
Y
,
Wang
Q
,
Shi
B
,
Xu
P
,
Hu
Z
,
Bai
L
, et al
.
Development of a sandwich ELISA for evaluating soluble PD-L1 (CD274) in human sera of different ages as well as supernatants of PD-L1+ cell lines
.
Cytokine
2011
;
56
:
231
8
.
64.
Tang
Y
,
Zhang
P
,
Wang
Y
,
Wang
J
,
Su
M
,
Wang
Y
, et al
.
The biogenesis, biology, and clinical significance of exosomal PD-L1 in cancer
.
Front Immunol
2020
;
11
:
604
.
65.
Shigemori
T
,
Toiyama
Y
,
Okugawa
Y
,
Yamamoto
A
,
Yin
C
,
Narumi
A
, et al
.
Soluble PD-L1 expression in circulation as a predictive marker for recurrence and prognosis in gastric cancer: direct comparison of the clinical burden between tissue and serum PD-L1 expression
.
Ann Surg Oncol
2019
;
26
:
876
83
.
66.
Chen
G
,
Huang
AC
,
Zhang
W
,
Zhang
G
,
Wu
M
,
Xu
W
, et al
.
Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response
.
Nature
2018
;
560
:
382
6
.
67.
Poggio
M
,
Hu
T
,
Pai
C-C
,
Chu
B
,
Belair
CD
,
Chang
A
, et al
.
Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory
.
Cell
2019
;
177
:
414
27
.
68.
Monypenny
J
,
Milewicz
H
,
Flores-Borja
F
,
Weitsman
G
,
Cheung
A
,
Chowdhury
R
, et al
.
ALIX regulates tumor-mediated immunosuppression by controlling EGFR activity and PD-L1 presentation
.
Cell Rep
2018
;
24
:
630
41
.
69.
Hassounah
NB
,
Malladi
VS
,
Huang
Y
,
Freeman
SS
,
Beauchamp
EM
,
Koyama
S
, et al
.
Identification and characterization of an alternative cancer-derived PD-L1 splice variant
.
Cancer Immunol Immunother
2019
;
68
:
407
20
.
70.
Mahoney
KM
,
Shukla
SA
,
Patsoukis
N
,
Chaudhri
A
,
Browne
EP
,
Arazi
A
, et al
.
A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression
.
Cancer Immunol Immunother
2019
;
68
:
421
32
.
71.
Ng
KW
,
Attig
J
,
Young
GR
,
Ottina
E
,
Papamichos
SI
,
Kotsianidis
I
, et al
.
Soluble PD-L1 generated by endogenous retroelement exaptation is a receptor antagonist
.
Elife
2019
;
8
:
e50256
.
72.
Zhou
J
,
Mahoney
KM
,
Giobbie-Hurder
A
,
Zhao
F
,
Lee
S
,
Liao
X
, et al
.
Soluble PD-L1 as a biomarker in malignant melanoma treated with checkpoint blockade
.
Cancer Immunol Res
2017
;
5
:
480
92
.
73.
Gong
B
,
Kiyotani
K
,
Sakata
S
,
Nagano
S
,
Kumehara
S
,
Baba
S
, et al
.
Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer
.
J Exp Med
2019
;
216
:
982
1000
.
74.
Gu
D
,
Ao
X
,
Yang
Y
,
Chen
Z
,
Xu
X
.
Soluble immune checkpoints in cancer: production, function and biological significance
.
J Immunother Cancer
2018
;
6
:
132
.
75.
Dezutter-Dambuyant
C
,
Durand
I
,
Alberti
L
,
Bendriss-Vermare
N
,
Valladeau-Guilemond
J
,
Duc
A
, et al
.
A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage
.
Oncoimmunology
2016
;
5
:
e1091146
.
76.
Zhao
F
,
Evans
K
,
Xiao
C
,
DeVito
N
,
Theivanthiran
B
,
Holtzhausen
A
, et al
.
Stromal fibroblasts mediate anti-PD-1 resistance via MMP-9 and dictate TGFbeta inhibitor sequencing in melanoma
.
Cancer Immunol Res
2018
;
6
:
1459
71
.
77.
Hira-Miyazawa
M
,
Nakamura
H
,
Hirai
M
,
Kobayashi
Y
,
Kitahara
H
,
Bou-Gharios
G
, et al
.
Regulation of programmed-death ligand in the human head and neck squamous cell carcinoma microenvironment is mediated through matrix metalloproteinase-mediated proteolytic cleavage
.
Int J Oncol
2018
;
52
:
379
88
.
78.
Romero
Y
,
Wise
R
,
Zolkiewska
A
.
Proteolytic processing of PD-L1 by ADAM proteases in breast cancer cells
.
Cancer Immunol Immunother
2020
;
69
:
43
55
.
79.
Orme
JJ
,
Jazieh
KA
,
Xie
T
,
Harrington
S
,
Liu
X
,
Ball
M
, et al
.
ADAM10 and ADAM17 cleave PD-L1 to mediate PD-(L)1 inhibitor resistance
.
Oncoimmunology
2020
;
9
:
1744980
.
80.
Swatek
KN
,
Komander
D
.
Ubiquitin modifications
.
Cell Res
2016
;
26
:
399
422
.
81.
MacGurn
JA
,
Hsu
PC
,
Emr
SD
.
Ubiquitin and membrane protein turnover: from cradle to grave
.
Annu Rev Biochem
2012
;
81
:
231
59
.
82.
Yang
Y
,
Hsu
J-M
,
Sun
L
,
Chan
L-C
,
Li
C-W
,
Hsu
JL
, et al
.
Palmitoylation stabilizes PD-L1 to promote breast tumor growth
.
Cell Res
2019
;
29
:
83
6
.
83.
Dai
X
,
Bu
X
,
Gao
Y
,
Guo
J
,
Hu
J
,
Jiang
C
, et al
.
Energy status dictates PD-L1 protein abundance and anti-tumor immunity to enable checkpoint blockade
.
Mol Cell
2021
;
81
:
2317
31
.
84.
Zhang
J
,
Bu
X
,
Wang
H
,
Zhu
Y
,
Geng
Y
,
Nihira
NT
, et al
.
Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance
.
Nature
2018
;
553
:
91
5
.
85.
Wu
Y
,
Zhang
C
,
Liu
X
,
He
Z
,
Shan
B
,
Zeng
Q
, et al
.
ARIH1 signaling promotes anti-tumor immunity by targeting PD-L1 for proteasomal degradation
.
Nat Commun
2021
;
12
:
2346
.
86.
Fuchs
SY
,
Spiegelman
VS
,
Kumar
KG
.
The many faces of beta-TrCP E3 ubiquitin ligases: reflections in the magic mirror of cancer
.
Oncogene
2004
;
23
:
2028
36
.
87.
Hsu
J-M
,
Xia
W
,
Hsu
Y-H
,
Chan
L-C
,
Yu
W-H
,
Cha
J-H
, et al
.
STT3-dependent PD-L1 accumulation on cancer stem cells promotes immune evasion
.
Nat Commun
2018
;
9
:
1908
.
88.
Chan
L-C
,
Li
C-W
,
Xia
W
,
Hsu
J-M
,
Lee
H-H
,
Cha
J-H
, et al
.
IL-6/JAK1 pathway drives PD-L1 Y112 phosphorylation to promote cancer immune evasion
.
J Clin Invest
2019
;
129
:
3324
38
.
89.
Zhang
X
,
Huang
X
,
Xu
J
,
Li
E
,
Lao
M
,
Tang
T
, et al
.
NEK2 inhibition triggers anti-pancreatic cancer immunity by targeting PD-L1
.
Nat Commun
2021
;
12
:
4536
.
90.
Lim
S-O
,
Li
C-W
,
Xia
W
,
Cha
J-H
,
Chan
L-C
,
Wu
Y
, et al
.
Deubiquitination and stabilization of PD-L1 by CSN5
.
Cancer Cell
2016
;
30
:
925
39
.
91.
Huang
X
,
Zhang
Q
,
Lou
Y
,
Wang
J
,
Zhao
X
,
Wang
L
, et al
.
USP22 deubiquitinates CD274 to suppress anticancer immunity
.
Cancer Immunol Res
2019
;
7
:
1580
90
.
92.
Stein
MP
,
Dong
J
,
Wandinger-Ness
A
.
Rab proteins and endocytic trafficking: potential targets for therapeutic intervention
.
Adv Drug Deliv Rev
2003
;
55
:
1421
37
.
93.
Bareford
LM
,
Swaan
PW
.
Endocytic mechanisms for targeted drug delivery
.
Adv Drug Deliv Rev
2007
;
59
:
748
58
.
94.
Yu
JX
,
Hodge
JP
,
Oliva
C
,
Neftelinov
ST
,
Hubbard-Lucey
VM
,
Tang
J
.
Trends in clinical development for PD-1/PD-L1 inhibitors
.
Nat Rev Drug Discov
2020
;
19
:
163
4
.
95.
Jin
H
,
D'Urso
V
,
Neuteboom
B
,
McKenna
SD
,
Schweickhardt
R
,
Gross
AW
, et al
.
Avelumab internalization by human circulating immune cells is mediated by both Fc gamma receptor and PD-L1 binding
.
Oncoimmunology
2021
;
10
:
1958590
.
96.
Maher
CM
,
Thomas
JD
,
Haas
DA
,
Longen
CG
,
Oyer
HM
,
Tong
JY
, et al
.
Small-molecule sigma1 modulator induces autophagic degradation of PD-L1
.
Mol Cancer Res
2018
;
16
:
243
55
.
97.
D'Arrigo
P
,
Russo
M
,
Rea
A
,
Tufano
M
,
Guadagno
E
,
Del Basso De Caro
ML
, et al
.
A regulatory role for the co-chaperone FKBP51s in PD-L1 expression in glioma
.
Oncotarget
2017
;
8
:
68291
304
.
98.
Yamaguchi
H
,
Hsu
J-M
,
Yang
W-H
,
Hung
M-C
.
Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics
.
Nat Rev Clin Oncol
2022
;
19
:
287
305
.
99.
Hamilton
G
,
Rath
B
.
Avelumab: combining immune checkpoint inhibition and antibody-dependent cytotoxicity
.
Expert Opin Biol Ther
2017
;
17
:
515
23
.
100.
Schaer
DA
,
Beckmann
RP
,
Dempsey
JA
,
Huber
L
,
Forest
A
,
Amaladas
N
, et al
.
The CDK4/6 inhibitor abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade
.
Cell Rep
2018
;
22
:
2978
94
.
101.
Ali
LR
,
Garrido-Castro
AC
,
Lenehan
PJ
,
Bollenrucher
N
,
Stump
CT
,
Dougan
M
, et al
.
PD-1 blockade and CDK4/6 inhibition augment nonoverlapping features of T cell activation in cancer
.
J Exp Med
2023
;
220
:
e20220729
.
102.
Lin
H-C
,
Yeh
C-W
,
Chen
Y-F
,
Lee
T-T
,
Hsieh
P-Y
,
Rusnac
DV
, et al
.
C-terminal end-directed protein elimination by CRL2 ubiquitin ligases
.
Mol Cell
2018
;
70
:
602
13
.
103.
Zhang
L
,
Xu
M
,
Scotti
E
,
Chen
ZJ
,
Tontonoz
P
.
Both K63 and K48 ubiquitin linkages signal lysosomal degradation of the LDL receptor
.
J Lipid Res
2013
;
54
:
1410
20
.
104.
van Kerkhof
P
,
dos Santos
CMA
,
Sachse
M
,
Klumperman
J
,
Bu
G
,
Strous
GJ
.
Proteasome inhibitors block a late step in lysosomal transport of selected membrane but not soluble proteins
.
Mol Biol Cell
2001
;
12
:
2556
66
.
105.
Valk
E
,
Rudd
CE
,
Schneider
H
.
CTLA-4 trafficking and surface expression
.
Trends Immunol
2008
;
29
:
272
9
.
106.
Pentcheva-Hoang
T
,
Chen
L
,
Pardoll
DM
,
Allison
JP
.
Programmed death-1 concentration at the immunological synapse is determined by ligand affinity and availability
.
Proc Natl Acad Sci U S A
2007
;
104
:
17765
70
.
107.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
.
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med
2015
;
372
:
320
30
.
108.
Chen
L
,
Xiong
Y
,
Li
J
,
Zheng
X
,
Zhou
Q
,
Turner
A
, et al
.
PD-L1 expression promotes epithelial to mesenchymal transition in human esophageal cancer
.
Cell Physiol Biochem
2017
;
42
:
2267
80
.
109.
Gato-Cañas
M
,
Zuazo
M
,
Arasanz
H
,
Ibañez-Vea
M
,
Lorenzo
L
,
Fernandez-Hinojal
G
, et al
.
PDL1 signals through conserved sequence motifs to overcome interferon-mediated cytotoxicity
.
Cell Rep
2017
;
20
:
1818
29
.
110.
Zhang
P
,
Ma
Y
,
Lv
C
,
Huang
M
,
Li
M
,
Dong
B
, et al
.
Upregulation of programmed cell death ligand 1 promotes resistance response in non-small-cell lung cancer patients treated with neo-adjuvant chemotherapy
.
Cancer Sci
2016
;
107
:
1563
71
.
111.
Yu
W
,
Hua
Y
,
Qiu
H
,
Hao
J
,
Zou
K
,
Li
Z
, et al
.
PD-L1 promotes tumor growth and progression by activating WIP and beta-catenin signaling pathways and predicts poor prognosis in lung cancer
.
Cell Death Dis
2020
;
11
:
506
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.