Preparation and characterization of anti PD-L1 monoclonal antibody decorated poly lactic-co-glycolic acid nanoparticles containing linrodostat mesylate.

Document Type : Original Research

Authors

1 PhD Candidate of Immunology, Microbiology Department, School of Veterinary Medicine, Urmia University, Urmia, Iran

2 Professor of Immunology, Microbilogy Department, School of Veterinary Medicine, Urmia University, Urmia, Iran

3 Assistant Professor of Immunology, Microbilogy Department, School of Veterinary Medicine, Urmia University, Urmia, Iran

4 Associate Professor, Pathobiology Department, School of Veterinary Medicine, Urmia University, Urmia, Iran

5 Professor of Immunopharmacology, Utrecht University, Utrecht, Netherlands

Abstract
Nanotechnology mainly shows its inhibitory effect on the tumor microenvironment by modulating the immune suppression mechanism. Success in this field largely depends on the physicochemical characterization of nanoparticle vaccines. The goal of this study was to produce anti PD-L1 monoclonal antibody decorated nanoparticles containing linrodostat mesylate with desirable properties and to investigate their physicochemical characterization .

Nanoparticles were prepared using double emulsion-solvent_evaporation method. Size and morphology of the particles were investigated using the FESEM microscope method and polydispersity index and zeta potential of the particles using the DLS method, as well as release rate and encapsulation efficiency.

The research results showed that nanoparticles had a suitable uniform dispersion. In the group of nanoparticles containing linrodostat mesylate, the polydispersity index of particles was 0.06 and after the binding of anti-PDL1 monoclonal antibody was 0.24. All particles were spherical with a smooth surface. The ideal particle size for nanoparticles containing linrodostat mesylate was estimated to be 210.14 nm, and it was estimated to be about 270.35 nm after binding anti-PDL1 monoclonal antibody to nanoparticles. Binding of anti-PDL1 monoclonal antibody decreased the amount of encapsulated linrodostat mesylate. The release of linrodostat mesylate was biphasic, it has an initial phase with a steep slope and the next phase is a slow and controlled release.

The results showed that the vaccine based on nanoparticles produced by the double emulsion-solvent-evaporation method containing linrodostat mesylate and decorated with anti-PDL-1 monoclonal antibody has very suitable physicochemical characterization to be used as an immunotherapy vaccine.

Keywords

Subjects


1. Adusumilli PS, Cha E, Cornfeld M, Davis T, Diab A, Dubensky TW, et al. New Cancer Immunotherapy Agents in Development: A report from an associated program of the 31stAnnual Meeting of the Society for Immunotherapy of Cancer, 2016. J Immunother Cancer. 2017;5(1).
2. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell. 2017;168(4):707–23.
3. Devaud C, John LB, Westwood JA, Darcy PK, Kershaw MH. Immune modulation of the tumor microenvironment for enhancing cancer immunotherapy. Oncoimmunology. 2013;2(8).
4. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25(4):198–213.
5. Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 2016;27(8):1482–92.
6. Drew M. Pardoll. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev cancer. 2016;97(11):252–64.
7. Mohammadi P, Hesari M, Chalabi M, Salari F, Khademi F. An overview of immune checkpoint therapy in autoimmune diseases. Int Immunopharmacol. 2022;107.
8. D.S. S, A. R. The evolution of checkpoint blockade as a cancer therapy: What’s here, what’s next? Curr Opin Immunol [Internet]. 2015;33((Shin D.S.; Ribas A., aribas@mednet.ucla.edu) Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), Los Angeles, United States):23–35. Available from: http://www.embase.com/search/results?subaction=viewrecord&from=export&id=L601592513%5Cnhttp://dx.doi.org/10.1016/j.coi.2015.01.006%5Cnhttp://sfx.library.uu.nl/utrecht?sid=EMBASE&issn=18790372&id=doi:10.1016%2Fj.coi.2015.01.006&atitle=The+evolution+of+chec
9. 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.
10. Zou W, Wolchok JD, Chen L, Weiping Z, Jedd D. W, Lieping C. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy. Sci Transl Med. 2016;8(328):1–34.
11. Wang J, Yuan R, Song W, Sun J, Liu D, Li Z. PD-1, PD-L1 (B7-H1) and Tumor-Site Immune Modulation Therapy: The Historical Perspective. J Hematol Oncol. 2017;10(1).
12. Himmel ME, Saibil SD, Saltman AP. Immune checkpoint inhibitors in cancer immunotherapy. Cmaj. 2020;192(24):E651.
13. Couzin-Frankel J. Cancer immunotherapy. Science (80- ). 2013;342(6165):1432–3.
14. Holmgaard RB, Zamarin D, Munn DH, Wolchok JD, Allison JP. Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J Exp Med. 2013;210(7):1389–402.
15. Zhai L, Lauing KL, Chang AL, Dey M, Qian J, Cheng Y, et al. The role of IDO in brain tumor immunotherapy. J Neurooncol. 2015;123(3):395–403.
16. Sonpavde G, Necchi A, Gupta S, Steinberg GD, Gschwend JE, Van Der Heijden MS, et al. ENERGIZE: A Phase III study of neoadjuvant chemotherapy alone or with nivolumab with/without linrodostat mesylate for muscle-invasive bladder cancer. Futur Oncol. 2019;16(2):4359–68.
17. Wathoni N, Puluhulawa LE, Joni IM, Muchtaridi M, Mohammed AFA, Elamin KM, et al. Monoclonal antibody as a targeting mediator for nanoparticle targeted delivery system for lung cancer. Drug Deliv. 2022;29(1):2959–70.
18. Yoo J, Park C, Yi G, Lee D, Koo H. Active targeting strategies using biological ligands for nanoparticle drug delivery systems. Cancers (Basel). 2019;11(5).
19. Yu B, Tai HC, Xue W, Lee LJ, Lee RJ. Receptor-targeted nanocarriers for therapeutic delivery to cancer. Mol Membr Biol. 2010;27(7):286–98.
20. Le Naour J, Galluzzi L, Zitvogel L, Kroemer G, Vacchelli E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology. 2020;9(1).
21. Kanekiyo M, Wei CJ, Yassine HM, McTamney PM, Boyington JC, Whittle JRR, et al. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature. 2013;499(7456):102–6.
22. Alvi M, Yaqoob A, Rehman K, Shoaib SM, Akash MSH. PLGA-based nanoparticles for the treatment of cancer: current strategies and perspectives. AAPS Open. 2022;8(1).
23. Sinha VR, Trehan A. Biodegradable microspheres for protein delivery. J Control Release. 2003;90(3):261–80.
24. Sanaz Sheikhzadeh , Nowruz Delirezh RH. PREPARATION AND CHARACTERIZATION OF TUMOR CELL LYSATE AND POLY-IC LOADED POLY (LACTIC-CO-GLYCOLIC ACID) NANOPARTICLES AND EVALUATION OF THEIR ANTITUMOR EFFECTS IN MOUSE MODEL OF BREAST CANCER. Stud Med Sci. 2018;29(8):584.
25. Sheikhzadeh S, Delirezh N, Hobbenaghi R. Mannosylated polylactic-co-glycolic acid (MN-PLGA) nanoparticles induce potent anti-tumor immunity in murine model of breast cancer. Biomed Pharmacother. 2021;142.
26. Haddadi A, Hamdy S, Ghotbi Z, Samuel J, Lavasanifar A. Immunoadjuvant activity of the nanoparticles’ surface modified with mannan. Nanotechnology. 2014;25(35).
27. Marslin G, Revina AM, Khandelwal VKM, Balakumar K, Prakash J, Franklin G, et al. Delivery as nanoparticles reduces imatinib mesylate-induced cardiotoxicity and improves anticancer activity. Int J Nanomedicine. 2015;10:3163–70.
28. Rathee J, Kaur A, Kanwar R, Kaushik D, Kumar R, Salunke DB, et al. Polymeric Nanoparticles as a Promising Drug Delivery Platform for the Efficacious Delivery of Toll-Like Receptor 7/8 Agonist and IDO-Inhibitor. Colloids Surfaces A Physicochem Eng Asp. 2022;632.
29. Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30(6):507–19.
30. Kalbasi A, Ribas A. Antigen presentation keeps trending in immunotherapy resistance. Clin Cancer Res. 2018;24(14):3239–41.
31. Oyewumi MO, Kumar A, Cui Z. Nano-microparticles as immune adjuvants: Correlating particle sizes and the resultant immune responses. Expert Rev Vaccines. 2010;9(9):1095–107.
32. S. S, T.K.S. M, H.A.E. B, Y. C. Pharmaceutical aspects of intranasal delivery of vaccines using particulate systems. J Pharm Sci [Internet]. 2009;98(3):812–43. Available from: http://www.embase.com/search/results?subaction=viewrecord&from=export&id=L354354580%5Cnhttp://www3.interscience.wiley.com/cgi-bin/fulltext/120839833/PDFSTART%5Cnhttp://dx.doi.org/10.1002/jps.21493%5Cnhttp://sfx.metabib.ch/sfx_locater?sid=EMBASE&issn=00223
33. Fifis T, Gamvrellis A, Crimeen-Irwin B, Pietersz GA, Li J, Mottram PL, et al. Size-Dependent Immunogenicity: Therapeutic and Protective Properties of Nano-Vaccines against Tumors. J Immunol. 2004;173(5):3148–54.
34. Barua S, Mitragotri S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: A review of current status and future prospects. Nano Today. 2014;9(2):223–43.
35. Rice-Ficht AC, Arenas-Gamboa AM, Kahl-McDonagh MM, Ficht TA. Polymeric particles in vaccine delivery. Curr Opin Microbiol. 2010;13(1):106–12.
36. Silva AL, Rosalia RA, Varypataki E, Sibuea S, Ossendorp F, Jiskoot W. Poly-(lactic-co-glycolic-acid)-based particulate vaccines: Particle uptake by dendritic cells is a key parameter for immune activation. Vaccine. 2015;33(7):847–54.
37. Díez-Pascual AM. Surface Engineering of Nanomaterials with Polymers, Biomolecules, and Small Ligands for Nanomedicine. Materials (Basel). 2022;15(9).
38. Kim GJ, Nie S. Targeted cancer nanotherapy. Mater Today. 2005;8(8 SUPPL.):28–33.
39. Hamdy S, Molavi O, Ma Z, Haddadi A, Alshamsan A, Gobti Z, et al. Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+ T cell-mediated anti-tumor immunity. Vaccine. 2008;26(39):5046–57.
40. Hamdy S, Haddadi A, Hung RW, Lavasanifar A. Targeting dendritic cells with nano-particulate PLGA cancer vaccine formulations. Adv Drug Deliv Rev. 2011;63(10–11):943–55.
41. Cao J, Choi JS, Oshi MA, Lee J, Hasan N, Kim J, et al. Development of PLGA micro- and nanorods with high capacity of surface ligand conjugation for enhanced targeted delivery. Asian J Pharm Sci. 2019;14(1):86–94.
42. Joshi VB, Geary SM, Salem AK. Biodegradable particles as vaccine delivery systems: Size matters. AAPS J. 2013;15(1):85–94.
43. Foged C, Brodin B, Frokjaer S, Sundblad A. Particle size and surface charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm. 2005;298(2):315–22.
44. Mohammadi-Samani S, Taghipour B. PLGA micro and nanoparticles in delivery of peptides and proteins; Problems and approaches. Pharm Dev Technol. 2015;20(4):385–93.
45. Sahoo SK, Panyam J, Prabha S, Labhasetwar V. Residual polyvinyl alcohol associated with poly (D,L-lactide-co-glycolide) nanoparticles affects their physical properties and cellular uptake. J Control Release. 2002;82(1):105–14.
46. Swider E, Koshkina O, Tel J, Cruz LJ, de Vries IJM, Srinivas M. Customizing poly(lactic-co-glycolic acid) particles for biomedical applications. Acta Biomater. 2018;73:38–51.
47. Slimane M, Gaye I, Ghoul M, Chebil L. Mesoscale Modeling and Experimental Study of Quercetin Organization as Nanoparticles in the Poly-lactic- co-glycolic Acid/Water System under Different Conditions. Ind Eng Chem Res. 2020;59(10):4809–16.
48. Rasmussen MK, Pedersen JN, Marie R. Size and surface charge characterization of nanoparticles with a salt gradient. Nat Commun. 2020;11(1).
49. Limited MI. Zeta potential: An Introduction in 30 minutes. Zetasizer Nano Serles Tech Note MRK654-01 [Internet]. 2011;2:1–6. Available from: http://scholar.google.com/scholar?hl=en&btnG=Search&q=intitle:Zeta+Potential+An+Introduction+in+30+Minutes#0
50. Juttulapa M, Piriyaprasarth S, Sriamornsak P. Effect of ph on stability of oil-in-water emulsions stabilized by pectin-zein complexes. Adv Mater Res. 2013;747:127–30.
51. Govender T, Stolnik S, Garnett MC, Illum L, Davis SS. PLGA nanoparticles prepared by nanoprecipitation: Drug loading and release studies of a water soluble drug. J Control Release. 1999;57(2):171–85.
52. Kimura S, Egashira K, Nakano K, Iwata E, Miyagawa M, Tsujimoto H, et al. Local delivery of imatinib mesylate (STI571)-incorporated nanoparticle ex vivo suppresses vein graft neointima formation. Circulation. 2008;118(14 Suppl).