Reversing T Cell Exhaustion by Converting Membrane PD-1 to Its Soluble form in Jurkat Cells; Applying The CRISPR/Cas9 Exon Skipping Strategy

Document Type : Original Article

Authors

1 Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran

2 Immunology, Asthma and Allergy Research Institute (IAARI), Tehran University of Medical Sciences, Tehran, Iran

3 Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran

4 Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran

Abstract

Objective: T-cells express two functional forms of the programmed cell death protein 1 (PD-1): membrane (mPD-1) and
soluble (sPD-1). The binding of mPD-1 and its ligand (PD-L1) on tumor cells could lead activated lymphocytes toward
exhaustion. Selective deletion of the transmembrane domain via alternative splicing of exon-3 in PD-1 mRNA could
generate sPD-1. Overexpression of sPD-1 could disrupt the mPD-1/PD-L1 interaction in tumor-specific T cells. We
investigated the effect of secreted sPD-1 from pooled engineered and non-engineered T cell supernatant on survival
and proliferation of lymphocytes in the tumor microenvironment (TME).
Materials and Methods: In this experimental study, we designed two sgRNA sequences upstream and downstream of
exon-3 in the PDCD1 gene. The lentiCRISPRv2 puro vector was used to clone the dual sgRNAs and produce lentiviral
particles to transduce Jurkat T cells. Analysis assays were used to clarify the change in PD-1 expression pattern in the
pooled (engineered and non-engineered) Jurkat cells. Co-culture conditions were established with PD-L1+ cancer cells
and lymphocytes.
Results: CRISPR/Cas9 could delete exon-3 of the PDCD1 gene in the engineered cells based on the tracking of indels
by decomposition (TIDE) and interference of CRISPR edit (ICE) sequencing analysis reports. Our results showed a
12% reduction in mPD-1 positive cell population after CRISPR manipulation and increment in sPD-1 concentration in
the supernatant. The increased sPD-1 confirmed its positive effect on proliferation of lymphocytes co-cultured with PDL1+
cancer cells. The survival percent of lymphocytes co-cultured with the pooled cells supernatant was 12.5% more
than the control.
Conclusion: The CRISPR/Cas9 exon skipping approach could be used in adoptive cell immunotherapies to change
PD-1 expression patterns and overcome exhaustion.

Keywords

Main Subjects


  1. Schietinger A, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 2014; 35(2): 51-60.
  2. Miggelbrink AM, Jackson JD, Lorrey SJ, Srinivasan ES, Waibl- Polania J, Wilkinson DS, et al. CD4 T-cell exhaustion: does it exist and what are its roles in cancer? Clin Cancer Res. 2021; 27(21): 5742-5752.
  3. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015; 15(8): 486-499.
  4. Bonorino C, Mognol G. Editorial: T cell exhaustion. Front Immunol. 2020; 11: 920.
  5. Guram K, Kim SS, Wu V, Sanders PD, Patel S, Schoenberger SP, et al. A threshold model for T-Cell activation in the era of checkpoint blockade immunotherapy. Front Immunol. 2019; 10: 491.
  6. Torphy RJ, Schulick RD, Zhu Y. Newly emerging immune checkpoints: promises for future cancer therapy. Int J Mol Sci. 2017; 18(12): 2642.
  7. Yan T, Yu L, Shangguan D, Li W, Liu N, Chen Y, et al. Advances in pharmacokinetics and pharmacodynamics of PD-1/PD-L1 inhibitors. Int Immunopharmacol. 2023; 115: 109638.
  8. Simon B, Harrer DC, Schuler-Thurner B, Schaft N, Schuler G, Dörrie J, et al. The siRNA-mediated downregulation of PD-1 alone or simultaneously with CTLA-4 shows enhanced in vitro CAR-T-cell functionality for further clinical development towards the potential use in immunotherapy of melanoma. Exp Dermatol. 2018; 27(7): 769-778.
  9. Beane JD, Lee G, Zheng Z, Mendel M, Abate-Daga D, Bharathan M, et al. Clinical scale zinc finger nuclease-mediated gene editing of PD-1 in tumor infiltrating lymphocytes for the treatment of metastatic melanoma. Mol Ther. 2015; 23(8): 1380-1390.
  10. Su S, Zou Z, Chen F, Ding N, Du J, Shao J, et al. CRISPR-Cas9- mediated disruption of PD-1 on human T cells for adoptive cellular therapies of EBV positive gastric cancer. Oncoimmunology. 2016; 6(1): e1249558.
  11. Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, et al. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017; 7(1): 737.
  12. Nakazawa T, Natsume A, Nishimura F, Morimoto T, Matsuda R, Nakamura M, et al. Effect of CRISPR/Cas9-mediated PD-1-disrupted primary human third-generation CAR-T cells targeting EGFRvIII on in vitro human glioblastoma cell growth. Cells. 2020; 9(4): 998.
  13. Ureña-Bailén G, Lamsfus-Calle A, Daniel-Moreno A, Raju J, Schlegel P, Seitz C, et al. CRISPR/Cas9 technology: towards a new generation of improved CAR-T cells for anticancer therapies. Brief Funct Genomics. 2020; 19(3): 191-200.
  14. Nielsen C, Ohm-Laursen L, Barington T, Husby S, Lillevang ST. Alternative splice variants of the human PD-1 gene. Cell Immunol. 2005; 235(2): 109-116.
  15. Zhu X, Lang J. Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. Oncotarget. 2017; 8(57): 97671-97682.
  16. Khan M, Zhao Z, Arooj S, Fu Y, Liao G. Soluble PD-1: predictive, prognostic, and therapeutic value for cancer immunotherapy. Front Immunol. 2020; 11: 587460.
  17. Song MY, Park SH, Nam HJ, Choi DH, Sung YC. Enhancement of vaccine-induced primary and memory CD8(+) T-cell responses by soluble PD-1. J Immunother. 2011; 34(3): 297-306.
  18. Bartee MY, Dunlap KM, Bartee E. Tumor-localized secretion of soluble PD1 enhances oncolytic virotherapy. Cancer Res. 2017; 77(11): 2952-2963
  19. Chamberlain CA, Bennett EP, Kverneland AH, Svane IM, DoniaM, Met Ö. Highly efficient PD-1-targeted CRISPR-Cas9 for tumorinfiltrating lymphocyte-based adoptive T cell therapy. Mol Ther Oncolytics. 2022; 24: 417-428.
  20. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020; 367(6481): eaba7365.
  21. Gapinske M, Luu A, Winter J, Woods WS, Kostan KA, Shiva N, et al. CRISPR-SKIP: programmable gene splicing with single base editors. Genome Biol. 2018; 19(1): 107.
  22. Mou H, Smith JL, Peng L, Yin H, Moore J, Zhang XO, et al. CRISPR/ Cas9-mediated genome editing induces exon skipping by alternative splicing or exon deletion. Genome Biol. 2017; 18(1): 108.
  23. Togashi Y, Mizuuchi H, Tomida S, Terashima M, Hayashi H, Nishio K, et al. MET gene exon 14 deletion created using the CRISPR/ Cas9 system enhances cellular growth and sensitivity to a MET inhibitor. Lung Cancer. 2015; 90(3): 590-597.
  24. Labun K, Montague TG, Krause M, Torres Cleuren YN, Tjeldnes H, Valen E. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 2019; 47(W1): W171- W174.
  25. Concordet JP, Haeussler M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 2018; 46(W1): W242-W245.
  26. Rajabzadeh A, Hamidieh AA, Rahbarizadeh F. Spinoculation and retronectin highly enhance the gene transduction efficiency of Mucin- 1-specific chimeric antigen receptor (CAR) in human primary T cells. BMC Mol Cell Biol. 2021; 22(1): 57.
  27. Brinkman EK, Chen T, Amendola M, van Steensel B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 2014; 42(22): e168.
  28. Nagasaki J, Togashi Y. A variety of ‘exhausted’ T cells in the tumor microenvironment. Int Immunol. 2022; 34(11): 563-570.
  29. Yu Y, Cui J. Present and future of cancer immunotherapy: a tumor microenvironmental perspective. Oncol Lett. 2018; 16(4): 4105- 4113.
  30. Balança CC, Salvioni A, Scarlata CM, Michelas M, Martinez- Gomez C, Gomez-Roca C, et al. PD-1 blockade restores helper activity of tumor-infiltrating, exhausted PD-1hiCD39+ CD4 T cells. JCI Insight. 2021; 6(2): e142513.
  31. Bloemberg D, Nguyen T, MacLean S, Zafer A, Gadoury C, Gurnani K, et al. A high-throughput method for characterizing novel chimeric antigen receptors in Jurkat cells. Mol Ther Methods Clin Dev. 2020; 16: 238-254.
  32. Yang W, Chen PW, Li H, Alizadeh H, Niederkorn JY. PD-L1: PD-1 interaction contributes to the functional suppression of T-cell responses to human uveal melanoma cells in vitro. Invest Ophthalmol Vis Sci. 2008; 49(6): 2518-2525.
  33. Niu M, Liu Y, Yi M, Jiao D, Wu K. Biological characteristics and clinical significance of soluble PD-1/PD-L1 and exosomal PD-L1 in cancer. Front Immunol. 2022; 13: 827921.
  34. He L, Zhang G, He Y, Zhu H, Zhang H, Feng Z. Blockade of B7-H1 with sPD-1 improves immunity against murine hepatocarcinoma. Anticancer Res. 2005; 25(5): 3309-3313.
  35. Qiu H, Liu S, Xie C, Long J, Feng Z. Regulating immunity and inhibiting tumor growth by the recombinant peptide sPD-1-CH50. Anticancer Res. 2009; 29(12): 5089-5094.
  36. Zhang A, Sun Y, Wang S, Du J, Gao X, Yuan Y, et al. Secretion of human soluble programmed cell death protein 1 by chimeric antigen receptor-modified T cells enhances anti-tumor efficacy. Cytotherapy. 2020; 22(12): 734-743.
  37. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008; 26: 677-704.
  38.