PEGylation improves the therapeutic potential of dimerized translationally controlled tumor protein blocking peptide in ovalbumin-induced mouse model of airway inflammation

Drug Deliv. 2022 Dec;29(1):2320-2329. doi: 10.1080/10717544.2022.2100511.

Abstract

Dimerized translationally controlled tumor protein (dTCTP) initiates a variety of allergic responses in mouse models and that dTCTP-binding peptide 2 (dTBP2) attenuates the allergic inflammation by targeting dTCTP. However, the usefulness of peptide-based drugs is often limited due to their short half-lives, rapid degradation, and high levels of clearance after systemic administration. In this study, we chemically conjugated dTBP2 with 10 kDa polyethylene glycol (PEG) to improve its therapeutic potential. N-terminal mono-PEGylated dTBP2 (PEG-dTBP2) was characterized by in vitro bioactivity assay, pharmacokinetics study, and in vivo efficacy. When compared to the unmodified dTBP2, PEG-dTBP2 reduced proinflammatory cytokine IL-8 secretion in human bronchial cells by 10 to 15% and increased plasma half-life by approximately 2.5-fold in mice. This study specifically demonstrated that PEG-dTBP2 shows higher inhibitory action against ovalbumin (OVA)-induced airway inflammation in mice compared to dTBP2. Importantly, PEG-dTBP2, when administered once at 1 mg/kg, significantly reduced the migration of inflammatory cells and the levels of cytokines in the bronchoalveolar lavage fluids as well as OVA-specific IgE levels in serum. In addition, the degree of goblet cell hyperplasia and mucus secretion were significantly attenuated in the PEG-dTBP2 group compared with the control group. These results suggest that PEG-dTBP2 can be considered a potential candidate drug for regulating allergic inflammation.

Keywords: PEGylation; allergic airway inflammation; dimerized TCTP-binding peptide 2 (dTBP2); dimerized translationally controlled tumor protein (dTCTP); histamine-releasing factor (HRF).

MeSH terms

  • Animals
  • Bronchoalveolar Lavage Fluid / chemistry
  • Disease Models, Animal
  • Humans
  • Inflammation* / drug therapy
  • Inflammation* / metabolism
  • Mice
  • Mice, Inbred BALB C
  • Ovalbumin / adverse effects
  • Peptides / therapeutic use
  • Tumor Protein, Translationally-Controlled 1*

Substances

  • Peptides
  • Tumor Protein, Translationally-Controlled 1
  • Ovalbumin

Grants and funding

This research was supported by the Bio and Medical Technology Development Program (2020M3E5E2036808) and (2021R1A2C2003629) by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) to K. Lee, and RP-Grant 2017 of Ewha Womans University to H-D Bae.