Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

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

Https

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

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Sep;10(9):e005116.
doi: 10.1136/jitc-2022-005116.

FBW7-mediated ubiquitination and destruction of PD-1 protein primes sensitivity to anti-PD-1 immunotherapy in non-small cell lung cancer

Affiliations

FBW7-mediated ubiquitination and destruction of PD-1 protein primes sensitivity to anti-PD-1 immunotherapy in non-small cell lung cancer

Jiaxin Liu et al. J Immunother Cancer. 2022 Sep.

Abstract

Background: Activation of the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway has been extensively described as a pivotal mechanism to escape immune surveillance and elicits suppressive effect on antitumor immunity. Blockade of the PD-1/PD-L1 interaction by checkpoint inhibitors has been shown to result in tumor shrinkage and prolong patient survival. However, regulatory machinery for PD-1/PD-L1 expression is largely unknown.

Methods: We used bioinformatic tools and biochemical methods to investigate the significance of F-box and WD repeat domain containing 7 (FBW7) in regulating PD-1 protein stability. By generating a panel of FBW7 and PD-1 encoding plasmids, we expressed FBW7 and PD-1 or their mutants to performed immunoprecipitation and immunoblotting assays. The efficacy of cotargeting FBW7 to enhance antitumor immunity was evaluated in C57BL/6J mice. These laboratory findings were further validated in tumor samples obtained from patients with non-small cell lung cancer (NSCLC).

Results: We identified FBW7 as a E3 ubiquitin ligase for PD-1 protein, in which FBW7 promotes the K48-linked polyubiquitination of PD-1 protein at Lys233 residue. Cotargeting FBW7 accelerates PD-1 protein degradation and enhances antitumor immunity in vivo. Moreover, we demonstrated that cyclin-dependent kinase 1-mediated phosphorylation of Ser261 residue primes PD-1 protein nucleus translocation and binding with FBW7. Higher expression of FBW7 characterizes a 'hot' tumor microenvironment and confers more favorable responses to PD-1 blockade therapy.

Conclusions: This study highlights the critical role of FBW7 in determining PD-1 protein stability. FBW7 ubiquitinates PD-1 in a phosphorylation-dependent manner, as a consequence, leading to PD-1 protein degradation and cytotoxic lymphocytes infiltrating the tumor microenvironment. Screening FBW7 status would predict clinical response to anti-PD-1 immunotherapy in patients with NSCLC, and targeting FBW7 is a promising strategy to enhance antitumor immunity.

Keywords: Immunotherapy; Lung Neoplasms; Programmed Cell Death 1 Receptor; Translational Medical Research; Tumor Escape.

PubMed Disclaimer

Conflict of interest statement

Competing interests: None declared.

Figures

Figure 1
Figure 1
FBW7 interacts with PD-1 protein. (A) Sequence alignment of the phosphodegron sequences recognized by FBW7 in c-Jun, c-Myc, cyclin E, MCL-1 and PD-1. Conserved degron sequences are shown in red. (B, C) Western blot analysis with antibodies specific for the indicated proteins of HEK293 cells transfected with Flag-tagged PD-1 and Myc-tagged FBW7 constructs. β-actin was used as equal loading control. (D) The primary PBMCs, THP-1 cells and MOLT-4 cells, which express endogenous PD-1 and FBW7, were precipitated with anti-PD-1/anti-FBW7 antibody or isotype IgG, and the resultant immunoprecipitates were subjected to immunoblotting. (E) The MOLT-4 and Jurkat cells were treated with increasing concentrations of oridonin and analyzed for PD-1 protein abundance by Western blot. (F) The THP-1 cells were transfected with indicated constructs or treated with increasing concentrations of oridonin. The expression of exogenous and endogenous FBW7 and PD-1 protein was determined by immunoblotting. (G) Primary T cells and MOLT-4 cells were treated with oridonin at final concentrations of 5 µM and 10 µM for 24 hours. After indicated treatment, cells were incubated with Alexa Fluor 647-conjugated anti-PD-1 antibody. Cell surface expression of PD-1 was assessed by flow cytometry. Representative flow cytometry images were shown. FBW7, F-box and WD repeat domain containing 7; PBMCs, peripheral blood mononuclear cells;DMSO, dimethyl sulfoxide.
Figure 2
Figure 2
FBW7 determines PD-1 protein stability. (A) HEK293 cells were transfected with Flag-tagged PD-1 together with indicated Myc-tagged FBW7 constructs. Twenty hours after transfection, cells were split into 6 cm dishes and treated with 25 µg/mL CHX. At the indicated time points, cell lysates were prepared and probed with the Flag-tag antibody. β-actin was used as equal loading control. (B) The THP-1 cells and MOLT-4 cells were treated with 10 µM oridonin or DMSO and evaluated for PD-1 protein half-time by CHX chase assay. (C) The THP-1 cells and MOLT-4 cells were infected with shRNA lentivirus against FBW7 or negative control (NC), and selected with puromycin to eliminate non-infected cells. The resultant cells were analyzed for FBW7 and PD-1 expression by Western blot. (D) The THP-1 cells and MOLT-4 cells stably expressing shRNA targeting FBW7 were treated with CHX and evaluated for PD-1 protein half-time. (E, F) The Flag-tagged PD-L1 and Myc-tagged FBW7 constructs were expressed in HEK293 cells and evaluated for PD-L1 protein half-time by CHX chase assay. CHX, cycloheximide; FBW7, F-box and WD repeat domain containing 7;DMSO, dimethyl sulfoxide.
Figure 3
Figure 3
FBW7 as a E3 uibiquitin ligase for PD-1 protein. (A) Western blot analysis of whole cell lysates (WCL) and immunoprecipitates derived from THP-1 and MOLT-4 cells treated with DMSO or oridonin (10 µM) for 24 hours, and cells were treated with 20 μM MG132 for additional 5 hours before harvesting. The ubiquitination status of endogenous PD-1 protein was determined by immunoprecipitation (IP). β-actin was used as equal loading control. (B) HEK293 cells were transfected with Flag PD-1 together with indicated Myc FBW7 construct. Thirty hours after transfection, cells were treated with 20 μM MG132 for additional 8 hours to block the proteasome pathway before cell collection. The cell lysates were subjected to IP and immunoblotting, respectively. (C) Immunoblotting analysis of Flag tag pull-down precipitates and WCL derived from HEK293 cells transfected with HA Ub, Myc FBW7 and indicated Flag PD-1 construct. Thirty hours after transfection, cells were treated with 20 μM MG132 for 8 hours before cell collection. (D) Myc-tagged FBW7, Flag-tagged PD-1, and His-tagged Ub WT, or Ub K48, or Ub K63 plasmids were expressed in HEK293 cells. PD-1 protein ubiquitination status was determined by Flag tag pull-down and anti-His immunoblotting. (E) Western blot analysis of WCL and immunoprecipitates derived from HEK293 cells transfected with Myc FBW7, HA Ub together with indicated Flag PD-1 construct. The ubiquitination status of Flag-tagged PD-1 protein was determined in the Flag tag pull-down precipitates by using anti-HA primary antibody. (F) The Flag-tagged PD-1 WT and its KR mutants were expressed in THP-1 cells. Thirty hours after transfection, cells were treated with or without MG132 before cell collection. The expression of Flag PD-1 protein was assessed by Western blot. FBW7, F-box and WD repeat domain containing 7; HA, hemagglutinin;DMSO, dimethyl sulfoxide.
Figure 4
Figure 4
The nucleus translocation of PD-1 protein is required for its interaction with FBW7. (A) Immunoblotting of WCL and immunoprecipitates from THP-1 and MOLT-4 cells treated with or without FBS for 6 hours. MG132 was added into the culture medium 5 hours before harvesting. The ubiquitination status of PD-1 was assessed by using an antibody against Ub. β-actin was used as equal loading control. (B) HEK293 cells were transfected with Myc-tagged FBW7 and Flg-tagged PD-1 constructs. The resultant cells, THP-1 and MOLT-4 cells were cultured with or without FBS for 6 hours. After indicated treatment, cell fraction assay was performed and the expression of FBW7 and PD-1 in the non-nucleus lysates and nucleus lysates was determined by immunoblotting. α-tubulin and Lamin A/C were used as equal loading control for non-nucleus protein and nucleus protein, respectively. (C) Representative immunofluorescent images of the PD-1 protein localization in THP-1 and MOLT-4 cells cultured with or without FBS. Scale bar=100 µm. (D) The EGFP-tagged PD-1 constructs were transiently expressed in HEK293 cells and the distribution of PD-1 protein was visualized under a fluorescent microscope. Representative fluorescent images were shown. Scale bar=20 µm. (E) Alignment analysis of putative NLS region of PD-1 protein across a panel species. The conserved NLS region is highlighted in red. (F) Immunoblotting with the anti-Flag tag precipitates and WCL derived from HEK293 cells expressing Myc FBW7, HA Ub and Flag PD-1 WT or Flag PD-1 ΔNLS constructs. MG132 was added for 8 hour before cell collection. FBW7, F-box and WD repeat domain containing 7; HA, hemagglutinin; NLS, nuclear localization signal; WCL, whole cell lysates; FBS, fetal bovine serum.
Figure 5
Figure 5
Phosphorylation of PD-1 prompts FBW7-mediated ubiquitination and protein destruction. (A) HEK293 cells were transfected with Myc FBW7, Flag PD-1 and HA Ub constructs. Thirty hours after transfection, cells were treated with the pan-CDK inhibitor Alvocidib (10 µM) or the selective CDK1 inhibitor Ro-3306 (10 µM), together with 20 μM MG132 for another 8 hours. The ubiquitination status and the binding with FBW7 were determined with anti-Flag immunoprecipitates. β-actin was used as equal loading control. (B) The THP-1 cells and MOLT-4 cells were transfected with indicated plasmids and treated with either siRNA targeting CDK1 or Ro-3306 (10 µM). The ubiquitination status of PD-1 and its binding with FBW7 were assessed as described. (C) The Flag-tagged PD-1 and its S261A/S261D mutant was expressed in HEK293 cells. Phosphorylation status of the serine residue was determined in the anti-Flag immunoprecipitates by using an anti-pSer antibody. The ubiquitination status of PD-1 and its binding with FBW7 were assessed as described. (D) The effect of Ser261 phosphorylation on PD-1 ubiquitination was evaluated in HEK293 cells. Cells were transfected with indicated plasmids and cultured in the presence or absence of FBS for 6 hours. The binding to nucleus cotranslocator importin α was assessed in the anti-Flag immunoprecipitates. FBW7, F-box and WD repeat domain containing 7; WCL, whole cell lysates;FBS, fetal bovine serum.
Figure 6
Figure 6
Cotargeting FBW7 enhances antitumor immunity in vivo. (A) The immune competent C57BL/6 J mice and the immune compromised nude mice were subcutaneously injected with LLC cells. The mice were treated with vehicle, oridonin, anti-PD-1 antibody or their combination. The tumor volume was routinely monitored. At the end of the experiment, the syngeneic tumor was carefully removed and photographed. NS, not significant; *p<0.05 vs vehicle. (B) Evaluation of cell apoptosis in the syngeneic tumor by TUNEL assay. Representative images were shown. Scale bar=100 µm. NS, not significant; *p<0.05 vs vehicle, **p<0.01 vs vehicle. (C) Representative images of IHC staining of Ki67, FBW7, PD-1 and CD8 in the syngeneic tumor. The enlarged window indicated the infiltration of CD8+CTLs in the tumor microenviroment. Scale bar=50 µm. (D) Measurement of hepatic and renal function in tumor bearing mice receiving indicated treatment. CTLs, cytotoxic lymphocytes; FBW7, F-box and WD repeat domain containing 7; LLC, Lewis lung cancer; AST, aspartate transaminase; ALT, alanine transaminase; BUN, blood urea nitrogen.
Figure 7
Figure 7
Screening FBW7 to predict response to anti-PD-1 immunotherapy in NSCLC. (A, B) A total number of 23 cases of surgical resected NSCLC were analyzed for FBW7 and PD-1 expression by IHC staining. Representative IHC images were shown. Scale bar=50 µm. Each NSCLC sample yielded an IHC H-score for FBW7 and PD-1, respectively. The correlation between FBW7 and PD-1 was determined by linear regression analysis.**p<0.01. (C) Evaluation of the tumor microenvironment in DCB (n=27) and NDB (n=21) patients. The panCK was used to distinguish cancer cells and non-cancerous cells. Representative IHC images for tumor nests (positive for panCK), FBW7, and infiltrating CD8+CTLs were shown. Scale bar=50 µm. ***p<0.001. CTLs, cytotoxic lymphocytes; DCB, durable clinical benefit; FBW7, F-box and WD repeat domain containing 7; NBD, no durable clinical benefit; NSCLC, non-small cell lung cancer; IHC, immunohistochemistry.
Figure 8
Figure 8
A schematic diagram of CDK1-mediated PD-1 and FBW7 interaction and their underlying implications for antitumor immunity. The PD-1 protein is primarily expressed in the cell membrane and FBW7 is expressed in the nucleus of immune cells. In the FBW7 OFF condition, PD-1 phosphorylation is suppressed by the CDK1 inhibitor and it could not undergo nucleus translocation. Thus, the PD-1 protein constitutively expresses in immune cells, which in return activates the PD-1/PD-L1 immune suppressive pathway and inhibits CTLs infiltration. Tumors harboring the FBW7low/PD-1high feature manifest as ‘cold tumor’ and are not sensitive to PD-1 blockade immunotherapy. In contrast, the CDK1-mediated phosphorylation of PD-1 protein at Ser261 residue triggers a rapid movement into the nucleus, where PD-1 binds to FBW7 and undergoes ubiquitination and destruction. Overexpression of FBW7 could enhance this interaction, as a consequence, leading to the suppression of PD-1/PD-L1 signaling (FBW7 ON). The CD8+CTLs infiltrates the FBW7high/PD-1low ‘hot tumor’, which confers increased responsiveness to anti-PD-1 immunotherapy. CTLs, cytotoxic lymphocytes; FBW7, F-box and WD repeat domain containing 7.

Similar articles

Cited by

References

    1. Salmaninejad A, Valilou SF, Shabgah AG, et al. . PD-1/PD-L1 pathway: basic biology and role in cancer immunotherapy. J Cell Physiol 2019;234:16824–37. 10.1002/jcp.28358 - DOI - PubMed
    1. Gong J, Chehrazi-Raffle A, Reddi S, et al. . Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer 2018;6:8. 10.1186/s40425-018-0316-z - DOI - PMC - PubMed
    1. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest 2015;125:3384–91. 10.1172/JCI80011 - DOI - PMC - PubMed
    1. Peranzoni E, Lemoine J, Vimeux L, et al. . Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A 2018;115:E4041–50. 10.1073/pnas.1720948115 - DOI - PMC - PubMed
    1. Xin Yu J, Hodge JP, Oliva C, et al. . Trends in clinical development for PD-1/PD-L1 inhibitors. Nat Rev Drug Discov 2020;19:163–4. 10.1038/d41573-019-00182-w - DOI - PubMed

Publication types

MeSH terms