Your browser doesn't support javascript.
loading
Construction and validation of a synthetic phage-displayed nanobody library.
Kim, Minju; Bai, Xuelian; Im, Hyewon; Yang, Jisoo; Kim, Youngju; Kim, Minjoo Mj; Oh, Yeonji; Jeon, Yuna; Kwon, Hayoung; Lee, Seunghyun; Lee, Chang-Han.
Affiliation
  • Kim M; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Bai X; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Im H; Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea.
  • Yang J; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Kim Y; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Kim MM; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Oh Y; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Jeon Y; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Kwon H; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Lee S; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.
  • Lee CH; Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea.
Korean J Physiol Pharmacol ; 28(5): 457-467, 2024 Sep 01.
Article in En | MEDLINE | ID: mdl-39198226
ABSTRACT
Nanobodies derived from camelids and sharks offer unique advantages in therapeutic applications due to their ability to bind to epitopes that were previously inaccessible. Traditional methods of nanobody development face challenges such as ethical concerns and antigen toxicity. Our study presents a synthetic, phagedisplayed nanobody library using trinucleotide-directed mutagenesis technology, which allows precise amino acid composition in complementarity-determining regions (CDRs), with a focus on CDR3 diversity. This approach avoids common problems such as frameshift mutations and stop codon insertions associated with other synthetic antibody library construction methods. By analyzing FDA-approved nanobodies and Protein Data Bank sequences, we designed sub-libraries with different CDR3 lengths and introduced amino acid substitutions to improve solubility. The validation of our library through the successful isolation of nanobodies against targets such as PD-1, ATXN1 and STAT3 demonstrates a versatile and ethical platform for the development of high specificity and affinity nanobodies and represents a significant advance in biotechnology.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Korean J Physiol Pharmacol Year: 2024 Document type: Article Country of publication:

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Korean J Physiol Pharmacol Year: 2024 Document type: Article Country of publication: