Cancer Res Treat.  2022 Apr;54(2):362-374. 10.4143/crt.2021.424.

Engineering a High-Affinity PD-1 Peptide for Optimized Immune Cell-Mediated Tumor Therapy

Affiliations
  • 1Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
  • 2Laboratory of Cancer and Stem Cell Biology, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou Higher Education Mega Center, Guangzhou, China
  • 3Guangzhou Yidai Pharmaceutical Co., Ltd., Guangzhou, Guangdong, China

Abstract

Purpose
The purpose of this study was to optimize a peptide (nABP284) that binds to programmed cell death protein 1 (PD-1) by a computer-based protocol in order to increase its affinity. Then, this study aimed to determine the inhibitory effects of this peptide on cancer immune escape by coculturing improving cytokine-induced killer (ICIK) cells with cancer cells.
Materials and Methods
nABP284 that binds to PD-1 was identified by phage display technology in our previous study. AutoDock and PyMOL were used to optimize the sequence of nABP284 to design a new peptide (nABPD1). Immunofluorescence was used to demonstrate that the peptides bound to PD-1. Surface plasmon resonance was used to measure the binding affinity of the peptides. The blocking effect of the peptides on PD-1 was evaluated by a neutralization experiment with human recombinant programmed death-ligand 1 (PD-L1) protein. The inhibition of activated lymphocytes by cancer cells was simulated by coculturing of human acute T lymphocytic leukemia cells (Jurkat T cells) with human tongue squamous cell carcinoma cells (Cal27 cells). The anticancer activities were determined by coculturing ICIK cells with Cal27 cells in vitro.
Results
A high-affinity peptide (nABPD1, KD=11.9 nM) for PD-1 was obtained by optimizing the nABP284 peptide (KD=11.8 μM). nABPD1 showed better efficacy than nABP284 in terms of increasing the secretion of interkeulin-2 by Jurkat T cells and enhancing the in vitro antitumor activity of ICIK cells.
Conclusion
nABPD1 possesses higher affinity for PD-1 than nABP284, which significantly enhances its ability to block the PD-1/PD-L1 interaction and to increase ICIK cell-mediated antitumor activity by armoring ICIK cells.

Keyword

PD-1; Peptide optimization; Affinity; Computer simulation; Immune checkpoint inhibitor; Improving cytokine-induced killer (ICIK) cells; Immunotherapy

Figure

  • Fig. 1 Optimization and improvement of the peptide. (A) Main binding regions between human programmed cell death protein 1 (hPD-1) and human programmed death-ligand 1 (hPD-L1). The key amino acid residues through which it binds programmed cell death protein 1 (PD-1) are labeled. The region including A121 K124 R125 in PD-L1 shows sequence homology with nABP284. (B) AutoDock simulation provided evidence for the hypothesis that the imidazole ring on histidine could bind to the cleft between Y68 and E136 in PD-1. (C) Sequence of nABP284 and nABPD1, nABPD1 was optimized from nABP284 by adding branched chains (SHHHRL) with three consecutive histidine residues (HHH) on the N-termini. (D) Structural formula of HHH.

  • Fig. 2 Binding affinity of the peptides. The binding affinity of nABP284 (A) and nABPD1 (B) to human programmed cell death protein 1 (PD-1) recombinant protein was analyzed by surface plasmon resonance, and the affinity of nABPD1 for PD-1 was significantly enhanced compared with that of nABP284.

  • Fig. 3 Binding of the peptides to stimulated Jurkat T cells. (A) Expression of programmed cell death protein 1 (PD-1) and binding of 10 μM nABP284 or nABPD1 labeled with FITC (green) on Jurkat T cells before and after treatment with PMA (phorbol 12-myristate 13-acetate; 50 ng/mL) and ionomycin (1 μg/mL) for 24 hours. After allowing the peptide to bind, the cells were incubated with an anti–PD-1 antibody (red), and nuclei were stained with DAPI (blue). Images were merged. (B, C) Flow cytometry analysis of the binding of nABP284 or nABPD1 labeled with FITC to Jurkat T cells before and after stimulation. The data are presented as the mean±standard error of three independent experiments and were analyzed by Student’s t test. ***p < 0.001.

  • Fig. 4 Competition of peptides for binding to programmed cell death protein 1 (PD-1). (A) Images of the binding of nABP284, nABPD1 and an anti–PD-1 antibody to PD-1 expressed on the surface of Jurkat cells stimulated with PMA (phorbol 12-myristate 13-acetate) and ionomycin for 24 hours were obtained by confocal microscopy. Scale bars=12 μm. (B) Image J was used to measure the mean fluorescence intensity to evaluate PD-1 binding on the surface of stimulated Jurkat cells that were blocked with nABP284, nABPD1, or a functional PD-1 antibody in advance. The data are presented as the mean±standard error of three independent experiments and were analyzed by one-way ANOVA. ***p < 0.001; ns, not significantly different.

  • Fig. 5 Neutralization of programmed death-ligand 1 (PD-L1) with the peptides. (A) The neutralization capacities of nABP284 and nABPD1 at concentrations of 4, 13.3, or 40 μM. (B) Flow cytometry showed that the binding of human PD-L1 recombinant protein to stimulated Jurkat cells overexpressing programmed cell death protein 1 was inhibited by increasing the concentrations of nABP284 or nABPD1. nABPD1 showed a stronger blocking effect than nABP284 at the same concentration. The IC50 values of nABP284 and nABPD1 were 9.610 and 2.585 μM, respectively. The concentration of PD-L1 was 2 μg/mL. The data are presented as the mean±standard error of three independent experiments and were analyzed by one-way ANOVA. *p < 0.05, **p < 0.01, ***p < 0.001; ns, not significantly different.

  • Fig. 6 In vitro analysis of peptide function. (A) Expression of programmed death-ligand 1 (PD-L1) on cal27 cells after 48 hours of stimulation with different concentrations of interferon γ (IFN-γ). (B) Coculture with Cal27 cells significantly reduced the amount of interleukin 2 (IL-2) secreted by Jurkat T cells, and this inhibitory effect was reversed by nABP284 or nABPD1. nABPD1 showed a better effect than nABP284. (C) The Cell Counting Assay (Cell Counting Kit-8) revealed that nABP284 and nABPD1 were not directly cytotoxic to stimulated Cal27 cells, Jurkat T cells, or improving cytokine-induced killer (ICIK) cells at a concentration of 64 μM after 24 hours of culture. (D) Venous blood was drawn from donators, and peripheral blood monocytes (PBMCs) were isolated from the venous blood by density gradient centrifugation with Ficoll. OKT3, IL-2, and IFN-γ were used as shown in the image above to stimulate PBMCs. These stimulated PBMCs were called ICIK cells. The expression of programmed death 1 (PD-1) on PBMCs and ICIK cells was analyzed by flow cytometry. (E) Compared with untreated group and nABP284 group, nABPD1 significantly enhanced the cytotoxicity of ICIK cells to Cal27 cells. The data are presented as the mean±standard error of three independent experiments and were analyzed by one-way ANOVA. *p < 0.05, ***p < 0.001; ns, not significantly different.


Reference

References

1. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019; 18:175–96.
Article
2. Yang Y. Cancer immunotherapy: harnessing the immune system to battle cancer. J Clin Invest. 2015; 125:3335–7.
Article
3. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008; 26:677–704.
Article
4. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015; 36:265–76.
Article
5. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015; 27:450–61.
Article
6. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015; 373:23–34.
Article
7. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015; 372:2521–32.
Article
8. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015; 372:2018–28.
Article
9. Morgensztern D, Herbst RS. Nivolumab and pembrolizumab for non-small cell lung cancer. Clin Cancer Res. 2016; 22:3713–7.
Article
10. Yang J, Hu L. Immunomodulators targeting the PD-1/PD-L1 protein-protein interaction: from antibodies to small molecules. Med Res Rev. 2019; 39:265–301.
Article
11. Ruoslahti E. Peptides as targeting elements and tissue penetration devices for nanoparticles. Adv Mater. 2012; 24:3747–56.
Article
12. Fosgerau K, Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discov Today. 2015; 20:122–8.
Article
13. 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; 23:2341–8.
Article
14. Lim H, Chun J, Jin X, Kim J, Yoon J, No KT. Investigation of protein-protein interactions and hot spot region between PD-1 and PD-L1 by fragment molecular orbital method. Sci Rep. 2019; 9:16727.
Article
15. Biesiada J, Porollo A, Velayutham P, Kouril M, Meller J. Survey of public domain software for docking simulations and virtual screening. Hum Genomics. 2011; 5:497–505.
Article
16. Martinez X, Krone M, Alharbi N, Rose AS, Laramee RS, O’Donoghue S, et al. Molecular graphics: bridging structural biologists and computer scientists. Structure. 2019; 27:1617–23.
Article
17. Pan K, Guan XX, Li YQ, Zhao JJ, Li JJ, Qiu HJ, et al. Clinical activity of adjuvant cytokine-induced killer cell immunotherapy in patients with post-mastectomy triple-negative breast cancer. Clin Cancer Res. 2014; 20:3003–11.
Article
18. Lee JH, Lee JH, Lim YS, Yeon JE, Song TJ, Yu SJ, et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology. 2015; 148:1383–91.
Article
19. Huang X, Zhang J, Li X, Huang H, Liu Y, Yu M, et al. Rescue of iCIKs transfer from PD-1/PD-L1 immune inhibition in patients with resectable tongue squamous cell carcinoma (TSCC). Int Immunopharmacol. 2018; 59:127–33.
Article
20. Jiang P, Zhang Y, Archibald SJ, Wang H. Adoptive cell transfer after chemotherapy enhances survival in patients with resectable HNSCC. Int Immunopharmacol. 2015; 28:208–14.
Article
21. Maute RL, Gordon SR, Mayer AT, McCracken MN, Natarajan A, Ring NG, et al. Engineering high-affinity PD-1 variants for optimized immunotherapy and immuno-PET imaging. Proc Natl Acad Sci U S A. 2015; 112:E6506–14.
Article
22. Saw PE, Song EW. Phage display screening of therapeutic peptide for cancer targeting and therapy. Protein Cell. 2019; 10:787–807.
Article
23. Johnston RJ, Su LJ, Pinckney J, Critton D, Boyer E, Krishnakumar A, et al. VISTA is an acidic pH-selective ligand for PSGL-1. Nature. 2019; 574:565–70.
Article
24. 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; 288:11771–85.
Article
25. Zorzi A, Middendorp SJ, Wilbs J, Deyle K, Heinis C. Acylated heptapeptide binds albumin with high affinity and application as tag furnishes long-acting peptides. Nat Commun. 2017; 8:16092.
Article
26. Yao JF, Yang H, Zhao YZ, Xue M. Metabolism of peptide drugs and strategies to improve their metabolic stability. Curr Drug Metab. 2018; 19:892–901.
Article
27. Tian Y, Yang D, Ye X, Li Z. Thioether-derived macrocycle for peptide secondary structure fixation. Chem Rec. 2017; 17:874–85.
Article
28. Ganesan P, Ramalingam R. Investigation of structural stability and functionality of homodimeric gramicidin towards peptide-based drug: a molecular simulation approach. J Cell Biochem. 2019; 120:4903–11.
Article
29. Estrella V, Chen T, Lloyd M, Wojtkowiak J, Cornnell HH, Ibrahim-Hashim A, et al. Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 2013; 73:1524–35.
Article
30. Gauthier L, Morel A, Anceriz N, Rossi B, Blanchard-Alvarez A, Grondin G, et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell. 2019; 177:1701–13.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr