A novel high mobility group box 1 neutralizing chimeric antibody attenuates drug-induced liver injury and postinjury inflammation in mice



Lundback, P, Lea, JD, Sowinska, A, Ottosson, L, Furst, CM, Steen, J, Aulin, C, Clarke, JI, Kipar, A ORCID: 0000-0001-7289-3459, Klevenvall, L
et al (show 8 more authors) (2016) A novel high mobility group box 1 neutralizing chimeric antibody attenuates drug-induced liver injury and postinjury inflammation in mice. Hepatology, 64 (5). pp. 1699-1710.

Access the full-text of this item by clicking on the Open Access link.
[img] Text
s1-ln22936533-975314624-1939656818Hwf-1518847911IdV-119416326822936533PDF_HI0001.pdf - Author Accepted Manuscript

Download (3MB)

Abstract

Acetaminophen (APAP) overdoses are of major clinical concern. Growing evidence underlines a pathogenic contribution of sterile postinjury inflammation in APAP‐induced acute liver injury (APAP‐ALI) and justifies development of anti‐inflammatory therapies with therapeutic efficacy beyond the therapeutic window of the only current treatment option, N‐acetylcysteine (NAC). The inflammatory mediator, high mobility group box 1 (HMGB1), is a key regulator of a range of liver injury conditions and is elevated in clinical and preclinical APAP‐ALI. The anti‐HMGB1 antibody (m2G7) is therapeutically beneficial in multiple inflammatory conditions, and anti‐HMGB1 polyclonal antibody treatment improves survival in a model of APAP‐ALI. Herein, we developed and investigated the therapeutic efficacy of a partly humanized anti‐HMGB1 monoclonal antibody (mAb; h2G7) and identified its mechanism of action in preclinical APAP‐ALI. The mouse anti‐HMGB1 mAb (m2G7) was partly humanized (h2G7) by merging variable domains of m2G7 with human antibody‐Fc backbones. Effector function‐deficient variants of h2G7 were assessed in comparison with h2G7 in vitro and in preclinical APAP‐ALI. h2G7 retained identical antigen specificity and comparable affinity as m2G7. 2G7 treatments significantly attenuated APAP‐induced serum elevations of alanine aminotransferase and microRNA‐122 and completely abrogated markers of APAP‐induced inflammation (tumor necrosis factor, monocyte chemoattractant protein 1, and chemokine [C‐X‐C motif] ligand 1) with prolonged therapeutic efficacy as compared to NAC. Removal of complement and/or Fc receptor binding did not affect h2G7 efficacy. Conclusion: This is the first report describing the generation of a partly humanized HMGB1‐neutralizing antibody with validated therapeutic efficacy and with a prolonged therapeutic window, as compared to NAC, in APAP‐ALI. The therapeutic effect was mediated by HMGB1 neutralization and attenuation of postinjury inflammation. These results represent important progress toward clinical implementation of HMGB1‐specific therapy as a means to treat APAP‐ALI and other inflammatory conditions. (Hepatology 2016;64:1699‐1710).

Item Type: Article
Uncontrolled Keywords: Animals, Mice, Inbred C57BL, Mice, Inflammation, Acetaminophen, HMGB1 Protein, Analgesics, Non-Narcotic, Male, Antibodies, Neutralizing, Antipyretics, Chemical and Drug Induced Liver Injury
Depositing User: Symplectic Admin
Date Deposited: 28 Nov 2016 09:10
Last Modified: 23 Jan 2023 19:49
DOI: 10.1002/hep.28736
Open Access URL: https://aasldpubs.onlinelibrary.wiley.com/doi/full...
Related URLs:
URI: https://livrepository.liverpool.ac.uk/id/eprint/3004681