Development of specific nanobodies (VHH) for CD19 immuno-targeting of human B-lymphocytes

Document Type : Original Article

Authors

1 Department of Medical Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

2 Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

3 Department of Medical Biochemistry, Faculty of Applied Medical Sciences, Iran University of Medical Sciences,Tehran, Iran

Abstract

Objective(s): CD19 is a transmembrane glycoprotein of immunoglobulin superfamily. In order to treat lymphoma, monoclonal antibodies (mAb) can target different antigens, including CD19, CD20 and CD22 on the surface of B-cells. Along with biotechnology progress, a new generation of antibodies is introduced, with the purpose of eliminating the defects of the previous generation. Among the most developed one are nanobodies (Nb).  Nbs are a unique kind of camelid single domain antibody fragments with a broad range of medical applications. Unique physicochemical properties of Nbs have made them ideal candidates for therapeutic and diagnostic applications.
Materials and Methods: An immune gene library was created, and several CD19 specific Nbs were selected through antigen panning process, and their molecular properties as well as specificity, sensitivity, affinity and immunoreactivity against CD19 positive and negative cells were evaluated.
Results: The Nb library was prepared with 7.2 x107 members. We managed to isolate a panel of CD19-specific Nbs after the last round of selection with the affinity of isolated Nbs being estimated at the standard range of 15-35 nM.  Sequence analysis of positive clones was indicative of the fact that 12 variable sequences were confirmed. Of all these 12 clones, 2 clones with the greatest level signal in ELISA underwent subsequent analysis. Our sequencing results indicated high sequence homology (approximately 90%) between the Nb and Homa variable immunoglobulin domains.
Conclusion: Specific Nbs possess the potential to be used as novel therapeutic approaches in order to treat autoimmune diseases and B-cell lymphoma.

Keywords

Main Subjects


1. Zhou L-J, Ord DC, Hughes AL, Tedder TF. Structure and domain organization of the CD19 antigen of human, mouse, and guinea pig B lymphocytes. Conservation of the extensive cytoplasmic domain. J Immunol 1991;147:1424-1432.
2. Park J, editor CD19 CAR therapy for acute lymphoblastic leukemia2015: American Society of Clinical Oncology.
3. Fairfax KA, Tsantikos E, Figgett WA, Vincent FB, Quah PS, LePage M. BAFF-driven autoimmunity requires CD19 expression. J Autoimmun 2015;62:1-10.
4. Benjamini O, Jain P, Trinh L, Qiao W, Strom SS, Lerner S. Second cancers in patients with chronic lymphocytic leukemia who received frontline fludarabine, cyclophosphamide and rituximab therapy: distribution and clinical outcomes. Leuk Lymphoma. 2015;56:1643-1650.
5. Lugtenburg P, Brown PDN, Van der Holt B, D’Amore FA, Koene H, Berenschot H, editors. Randomized phase III study on the effect of early intensification of Rituximab in combination with 2-weekly CHOP chemotherapy flollowed by Rituximab or no maintenance in patients with diffuse large B-cell lymphoma: Results from a HOVON-Nordic Lymphoma Group Study. ASCO Annual Meeting; 2016.
6. Van der Kolk L, Grillo‐López A, Baars J, Hack C, Van Oers M. Complement activation plays a key role in the side‐effects of rituximab treatment. Br J Haematol 2001;115:807-811.
7. Hansen HJ, Qu Z, Goldenberg DM. Anti-CD19 antibodies. Google Patents; 2017.
8. Velasquez MP, Gottschalk S. Targeting CD19: the good, the bad, and CD81. Blood 2017;129:9-10.
9. Breton CS, Nahimana A, Aubry D, Macoin J, Moretti P, Bertschinger M. A novel anti-CD19 monoclonal antibody (GBR 401) with high killing activity against B cell malignancies. J HEMATOL ONCOL 2014;7:33.
10. Garfall AL, Maus MV, Hwang W-T, Lacey SF, Mahnke YD, Melenhorst JJ. Chimeric antigen receptor T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373(11):1040-7.
11. Katz B-Z, Herishanu Y. Therapeutic targeting of CD19 in hematological malignancies: past, present, future and beyond. Leuk Lymphoma 2014;55:999-1006.
12. De Meyer T, Muyldermans S, Depicker A. Nanobody-based products as research and diagnostic tools. Trends Biotechnol. 2014;32:263-270.
13. Honda T, Akahori Y, Kurosawa Y. Methods of constructing camel antibody libraries. Google Patents; 2008.
14. Harmsen M, De Haard H. Properties, production, and applications of camelid single-domain antibody fragments. Appl Microbiol Biotechnol. 2007;77:13-22.
15. Könning D, Zielonka S, Grzeschik J, Empting M, Valldorf B, Krah S. Camelid and shark single domain antibodies: structural features and therapeutic potential. Curr Opin Struct Biol. 2017;45:10-6.
16. Wang Y, Fan Z, Shao L, Kong X, Hou X, Tian D. Nanobody-derived nanobiotechnology tool kits for diverse biomedical and biotechnology applications. Int J Nanomedicine. 2016;11:3287.
17. van Lith SA, Roodink I, Verhoeff JJ, Mäkinen PI, Lappalainen JP, Ylä-Herttuala S. In vivo phage display screening for tumor vascular targets in glioblastoma identifies a llama nanobody against dynactin-1-p150 Glued. Oncotarget. 2016;7:71594-71607.
18. Veugelen S, Dewilde M, De Strooper B, Chávez-Gutiérrez L. Chapter Three-Screening and Characterization Strategies for Nanobodies Targeting Membrane Proteins. Methods Enzymol 2017;584:59-97.
19. Sharifzadeh Z, Rahbarizadeh F, Shokrgozar MA, Ahmadvand D, Mahboudi F, Jamnani FR. Development of oligoclonal nanobodies for targeting the tumor-associated glycoprotein 72 antigen. Mol Biotechnol 2013;54:590-601.
20. Ahmadvand D, Rasaee MJ, Rahbarizadeh F, Kontermann RE, Sheikholislami F. Cell selection and characterization of a novel human endothelial cell specific nanobody. Mol Immunol 2009;46:1814-1823.
21. Rahbarizadeh F, Rasaee MJ, Forouzandeh M, Allameh A-A. Over expression of anti-MUC1 single-domain antibody fragments in the yeast Pichia pastoris. Mol Immunol. 2006;43:426-435.
22. Hoogenboom HR, Lutgerink JT, Pelsers MM, Rousch MJ, Coote J, van Neer N Selection‐dominant and nonaccessible epitopes on cell‐surface receptors revealed by cell‐panning with a large phage antibody library. Eur J Biochem. 1999;260:774-784.
23. Jamnani FR, Rahbarizadeh F, Shokrgozar MA, Ahmadvand D, Mahboudi F, Sharifzadeh Z. Targeting high affinity and epitope-distinct oligoclonal nanobodies to HER2 over-expressing tumor cells. Exp Cell Res. 2012;318(10):1112-24.
24. Desper R, Gascuel O. Theoretical foundation of the balanced minimum evolution method of phylogenetic inference and its relationship to weighted least-squares tree fitting. Mol Biol Evol. 2004;21:587-598.
25. Gazarian T, Selisko B, Hérion P, Gazarian K. Isolation and structure–functional characterization of phage display library-derived mimotopes of noxiustoxin, a neurotoxin of the scorpion Centruroides noxius Hoffmann. Mol Immunol. 2000;37:755-766.
26. Kay BK, Winter J, McCafferty J. Phage display of peptides and proteins: a laboratory manual: Academic Press; 1996.
27. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248-254.
28. Dumoulin M, Conrath K, Van Meirhaeghe A, Meersman F, Heremans K, Frenken LG. Single‐domain antibody fragments with high conformational stability. Protein Sci  2002;11:500-515.
29. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. nature. 1970;227:680-685.
30. Engvall E. Enzyme immunoassay ELISA and EMIT. Methods Enzymol. 1980;70:419-439.
31. Rameh LE, Arvidsson A-k, Carraway KL, Couvillon AD, Rathbun G, Crompton A. A comparative analysis of the phosphoinositide binding specificity of pleckstrin homology domains. J Biol Chem. 1997;272:22059-22066.
32. Ghassabeh GH, Saerens D, Muyldermans S. Isolation of antigen-specific nanobodies. Antibody engineering 2010:251-266.
33. Beatty JD, Beatty BG, Vlahos WG. Measurement of monoclonal antibody affinity by non-competitive enzyme immunoassay. J Immunol Methods 1987;100:173-179.
34. Loken MR, Shah VO, Dattilio KL, Civin CI. Flowcytometric analysis of human bone marrow. II. Normal B lymphocyte development. Blood. 1987;70:1316-1324.
35. Nelson AL, Dhimolea E, Reichert JM. Development trends for human monoclonal antibody therapeutics. Nat Rev Drug Discov. 2010;9:767-774.
36. Morrow KJ, Liu C. Biosimilars of Monoclonal Antibodies: A Practical Guide to Manufacturing, Preclinical and Clinical Development: John Wiley & Sons; 2016.
37. Clackson T, Hoogenboom HR. Making antibody fragments using phage display libraries. Nature 1991;352:624.
38. Muyldermans S, Baral T, Retamozzo VC, De Baetselier P, De Genst E, Kinne J. Camelid immunoglobulins and nanobody technology. Vet Immunol Immunopathol 2009;128:178-183.
39. Van Audenhove I, Gettemans J. Nanobodies as versatile tools to understand, diagnose, visualize and treat cancer. EBioMedicine 2016;8:40-48.
40. Farajpour Z, Rahbarizadeh F, Kazemi B, Ahmadvand D. A nanobody directed to a functional epitope on VEGF, as a novel strategy for cancer treatment. Biochem Biophys Res Commun 2014;446(1):132-136.
41. Drive R, entitled DDEIaptsaora, B-Lymphocyte DoSCNVfCI-toH, disclose. TmhnbpainucfpeWhncoit, Co-author:, et al. Evaluation of a nanobody phage display library constructed from a Brucella-immunised camel. Vet Immunol Immunopathol. 2011;142:49-56.
42. Czuczman M, Grillo-Lopez A, White C, Saleh M, Gordon L, LoBuglio A. Treatment of patients with low-grade B-cell lymphoma with the combination of chimeric anti-CD20 monoclonal antibody and CHOP chemotherapy. J Clin Oncol 1999;17:268-276.
43. Reff ME, Carner K, Chambers K, Chinn P, Leonard J, Raab R. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood. 1994;83:435-445.
44. Schwemmlein M, Stieglmaier J, Kellner C, Peipp M, Saul D, Oduncu F, et al. A CD19-specific single-chain immunotoxin mediates potent apoptosis of B-lineage leukemic cells. Leukemia. 2007;21:1405-1412.
45. Hammer O, editor CD19 as an attractive target for antibody-based therapy. MAbs; 2012: Taylor & Francis.
46. Holliger P, Hudson PJ. Engineered antibody fragments and the rise of single domains. Nat Biotechnol 2005;23:1126-1136.
47. Adams GP, Schier R, McCall AM, Simmons HH, Horak EM, Alpaugh RK. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer Res 2001;61:4750-4755.
48. Henderson A, Ripley S, Heller M, Kieff E. Chromosome site for Epstein-Barr virus DNA in a Burkitt tumor cell line and in lymphocytes growth-transformed in vitro. Proc Natl Acad Sci. 1983;80:1987-1991.
49. Desmyterl A, Transuei TR, Ghahroudil MA, Thil M-HD, Poortmans F, Hamersz R. Crystal structure of a camel single—domain VH antibody fragment in complex. Nat Struct Biol 1996;3:803-811.
50. Vu KB, Ghahroudi MA, Wyns L, Muyldermans S. Comparison of llama VH sequences from conventional and heavy chain antibodies. Mol Immunol 1997;34:1121-1131.