High-Affinity Decoy PD-1 Mutant Screened from an Epitope-Specific Cell Library

Hao Liu, Chunxia Qiao, Naijing Hu, Zhihong Wang, Jing Wang, Jiannan Feng, Beifen Shen, Yuanfang Ma, Longlong Luo

PDF(1638 KB)
PDF(1638 KB)
Engineering ›› 2021, Vol. 7 ›› Issue (11) : 1557-1565. DOI: 10.1016/j.eng.2020.11.011

High-Affinity Decoy PD-1 Mutant Screened from an Epitope-Specific Cell Library

Author information +
History +

Abstract

Immunotherapy with anti-programmed cell death protein-1 (PD-1)/programmed cell death ligand-1 (PD-L1) monoclonal antibodies has become routine in the treatment of many kinds of human cancers, such as lung cancer, intestinal cancer, and melanoma. The PD-1/PD-L1 pathway inhibits T cell activation in the micro-environment, making it an attractive target against cancer. Wild-type (WT) PD-1 ectodomain has been shown to have difficulty blocking PD-1/PD-L1 mixture formation due to its low affinity. The present work uses three-dimensional (3D) crystal complex structures to analyze the interaction by which PD-1 binds to PD-L1 or PD-L2. It also reports on a theoretical study of the binding mode between PD-1 and its clinical antibody Opdivo. Based on the theoretical binding analysis of PD-1 and its ligands (i.e., PD-L1 and PD-L2) or antibody (Opdivo), a small-content, epitope-oriented mammalian cell library was established for PD-1. After three rounds of cell sorting, the decoy PD-1 mutant 463, which presented a higher affinity than WT PD-1 to the PD-L1 (the affinity has increased by almost three orders of magnitude) was screened out. It exhibited an inhibitory effect against PD-1 to prevent it from forming mixtures with PD-L1, which was similar to the effect of the commercial anti-PD-L1 antibody atezolizumab (ATE). The median effective concentration (EC50) value of the decoy mutant was 0.031 μg·mL−1 in comparison with 0.063 μg·mL−1 for ATE; both values were much lower than that of WT PD-1, at 2.571 μg·mL−1. The 463 decoy mutant reversed the inhibitory function of PD-1 in T cell activation; furthermore, 10 mg·kg−1 of 463 inhibited about 75% of tumor growth in vivo in a MC38 transgenic xenograft mice model, which was similar to the activity of ATE. More interestingly, an even lower dose of 463 (2 mg·kg−1) showed a better effect than 10 mg·kg−1 of WT PD-1. This work offers the decoy 463 with an improved curative effect, which holds potential to become a good option against PD-1/PD-L1-related cancers.

Keywords

Decoy PD-1 / PD-L1 / Mammalian cell library / Epitope-oriented

Cite this article

Download citation ▾
Hao Liu, Chunxia Qiao, Naijing Hu, Zhihong Wang, Jing Wang, Jiannan Feng, Beifen Shen, Yuanfang Ma, Longlong Luo. High-Affinity Decoy PD-1 Mutant Screened from an Epitope-Specific Cell Library. Engineering, 2021, 7(11): 1557‒1565 https://doi.org/10.1016/j.eng.2020.11.011

References

[[1]]
Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12(4):252–64.
[[2]]
Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour immunogenicity. Nat Rev Cancer 2012;12(4):307–13.
[[3]]
Klump KE, McGinnis JF. The role of reactive oxygen species in ocular malignancy. Adv Exp Med Biol 2014;801:655–9.
[[4]]
Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA 2002;99 (19):12293–7.
[[5]]
Riella LV, Paterson AM, Sharpe AH, Chandraker A. Role of the PD-1 pathway in the immune response. Am J Transplant 2012;12(10):2575–87.
[[6]]
Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 2010;236(1):219–42.
[[7]]
Hawkes EA, Grigg A, Chong G. Programmed cell death-1 inhibition in lymphoma. Lancet Oncol 2015;16(5):e234–45.
[[8]]
Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114(8):1537–44.
[[9]]
Muenst S, Soysal SD, Gao F, Obermann EC, Oertli D, Gillanders WE. The presence of programmed death 1 (PD-1)-positive tumor-infiltrating lymphocytes is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 2013;139(3):667–76.
[[10]]
Matsuzaki M, Nomizu T, Katagata N, Sakuma T, Momma T, Tachibana K, et al. A case of primary malignant lymphoma of the breast with an unusual ultrasound image. Fukushima J Med Sci 2010;56(2):145–50.
[[11]]
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(2):945–54.
[[12]]
Winograd R, Byrne KT, Evans RA, Odorizzi PM, Meyer ARL, Bajor DL, et al. Induction of T-cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma. Cancer Immunol Res 2015;3(4):399–411.
[[13]]
Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366(26):2455–65.
[[14]]
Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369(2):134–44.
[[15]]
Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366(26):2443–54.
[[16]]
Motzer RJ, Rini BI, McDermott DF, Redman BG, Kuzel TM, Harrison MR, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II. trail. J Clin Oncol 2015;33(13):1430–7.
[[17]]
Rizvi NA, Mazières J, Planchard D, Stinchcombe TE, Dy GK, Antonia SJ, et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol 2015;16 (3):257–65.
[[18]]
Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27(4):450–61.
[[19]]
Dömling A, Holak TA. Programmed death-1: therapeutic success after more than 100 years of cancer immunotherapy. Angew Chem Int Ed Engl 2014;53 (9):2286–8.
[[20]]
Homet Moreno B, Ribas A. Anti-programmed cell death protein-1/ligand-1 therapy in different cancers. Br J Cancer 2015;112(9):1421–7.
[[21]]
Mahoney KM, Freeman GJ, McDermott DF. The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma. Clin Ther 2015;37 (4):764–82.
[[22]]
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39(1):1–10.
[[23]]
Scott AM, Wolchok JD, Old LJ. Antibody therapy of cancer. Nat Rev Cancer 2012;12(4):278–87.
[[24]]
Sznol M, Chen L. Antagonist antibodies to PD-1 and B7–H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res 2013;19(5): 1021–34.
[[25]]
Lipson EJ, Forde PM, Hammers HJ, Emens LA, Taube JM, Topalian SL. Antagonists of PD-1 and PD-L1 in cancer treatment. Semin Oncol 2015;42 (4):587–600.
[[26]]
Cheng X, Veverka V, Radhakrishnan A, Waters LC, Muskett FW, Morgan SH, et al. Structure and interactions of the human programmed cell death 1 receptor. J Biol Chem 2013;288(17):11771–85.
[[27]]
Luo L, Wang S, Lang X, Zhou T, Geng J, Li X, et al. Selection and characterization of the novel anti-human PD-1 FV78 antibody from a targeted epitope mammalian cell-displayed antibody library. Cell Mol Immunol 2018;15 (2):146–57.
[[28]]
Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2011;208(6):1331.
[[29]]
Brahmer JR. PD-1-targeted immunotherapy: recent clinical findings. Clin Adv Hematol Oncol 2012;10(10):674–5.
[[30]]
Choueiri TK, Figueroa DJ, Fay AP, Signoretti S, Liu Y, Gagnon R, et al. Correlation of PD-L1 tumor expression and treatment outcomes in patients with renal cell carcinoma receiving sunitinib or pazopanib: results from COMPARZ, a randomized controlled trial. Clin Cancer Res 2015;21(5):1071–7.
[[31]]
Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, et al. MPDL3280A (antiPD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 2014;515(7528):558–62.
[[32]]
High-affinity PD-1 protein has potential. Cancer Discov 2015;5(11):1115–6.
[[33]]
Lin DY, Tanaka Y, Iwasaki M, Gittis AG, Su HP, Mikami B, et al. The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors. Proc Natl Acad Sci USA 2008;105(8):3011–6.
[[34]]
Freeman GJ. Structures of PD-1 with its ligands: sideways and dancing cheek to cheek. Proc Natl Acad Sci USA 2008;105(30):10275–6.
[[35]]
Lázár-Molnár E, Yan Q, Cao E, Ramagopal U, Nathenson SG, Almo SC. Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2. Proc Natl Acad Sci USA 2008;105(30):10483–8.
[[36]]
Zak KM, Kitel R, Przetocka S, Golik P, Guzik K, Musielak B, et al. Structure of the complex of human programmed death 1, PD-1, and its ligand PD-L1. Structure 2015;23(12):2341–8.
[[37]]
Yearley JH, Gibson C, Yu N, Moon C, Murphy E, Juco J, et al. PD-L2 expression in human tumors: relevance to anti-PD-1 therapy in cancer. Clin Cancer Res 2017;23(12):3158–67.
PDF(1638 KB)

Accesses

Citations

Detail

Sections
Recommended

/