Article Text

Download PDFPDF

Original article
Humanisation of a claudin-1-specific monoclonal antibody for clinical prevention and cure of HCV infection without escape
  1. Che C Colpitts1,2,
  2. Rajiv G Tawar1,2,
  3. Laurent Mailly1,2,
  4. Christine Thumann1,2,
  5. Laura Heydmann1,2,
  6. Sarah C Durand1,2,
  7. Fei Xiao1,2,
  8. Eric Robinet1,2,
  9. Patrick Pessaux1,2,3,
  10. Mirjam B Zeisel1,2,
  11. Thomas F Baumert1,2,3
  1. 1Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
  2. 2University of Strasbourg, Strasbourg, France
  3. 3Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
  1. Correspondence to Professor Thomas F Baumert, Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 3 Rue Koeberlé, Nouvel Hôpital Civil, Strasbourg F-67000, France; Thomas.Baumert{at}unistra.fr

Abstract

Objective HCV infection is a leading cause of chronic liver disease and a major indication for liver transplantation. Although direct-acting antivirals (DAAs) have much improved the treatment of chronic HCV infection, alternative strategies are needed for patients with treatment failure. As an essential HCV entry factor, the tight junction protein claudin-1 (CLDN1) is a promising antiviral target. However, genotype-dependent escape via CLDN6 and CLDN9 has been described in some cell lines as a possible limitation facing CLDN1-targeted therapies. Here, we evaluated the clinical potential of therapeutic strategies targeting CLDN1.

Design We generated a humanised anti-CLDN1 monoclonal antibody (mAb) (H3L3) suitable for clinical development and characterised its anti-HCV activity using cell culture models, a large panel of primary human hepatocytes (PHH) from 12 different donors, and human liver chimeric mice.

Results H3L3 pan-genotypically inhibited HCV pseudoparticle entry into PHH, irrespective of donor. Escape was likely precluded by low surface expression of CLDN6 and CLDN9 on PHH. Co-treatment of a panel of PHH with a CLDN6-specific mAb did not enhance the antiviral effect of H3L3, confirming that CLDN6 does not function as an entry factor in PHH from multiple donors. H3L3 also inhibited DAA-resistant strains of HCV and synergised with current DAAs. Finally, H3L3 cured persistent HCV infection in human-liver chimeric uPA-SCID mice in monotherapy.

Conclusions Overall, these findings underscore the clinical potential of CLDN1-targeted therapies and describe the functional characterisation of a humanised anti-CLDN1 antibody suitable for further clinical development to complement existing therapeutic strategies for HCV.

  • HEPATITIS C
  • ANTIVIRAL THERAPY
  • TIGHT JUNCTION

This is an Open Access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Footnotes

  • CCC and RGT contributed equally as co-first authors.

  • Contributors CCC and RGT contributed equally. TFB initiated and supervised the study. CCC, RGT, LM, CT, LH, SCD, FX and ER performed the experiments. PP provided essential reagents. CCC, RGT, MBZ and TFB analysed data. CCC, RGT, MZB and TFB wrote the manuscript.

  • Funding This study was supported by funding from the European Union (FP7 HEPAMAB GAN 305600, ERC-2014-AdG-HEPCIR, EU H2020 HEPCAR and Interreg IV-Rhin Supérieur-FEDER-Hepato-Regio-Net 2012), the Agence Nationale de Recherche sur le SIDA (ANRS) (2012/318, 2013/108) the Direction Générale de l'Offre de Soins (A12027MS) and the US National Institutes of Health (1U19AI123862-01). This work has been published under the framework of the LABEX ANR-10-LABX-0028_HEPSYS and benefits from funding from the state managed by the French National Research Agency as part of the Investments for the Future programme. CCC is supported by fellowships from the Canadian Institutes of Health Research (201411MFE-338606-245517) and the Canadian Network on Hepatitis C.

  • Competing interests TFB is a co-inventor on a patent application on monoclonal anti-claudin1 antibodies for the inhibition of HCV infection (US patent no. 8,518,408; WO2010034812; PCT/EP 08 305 597 0).

  • Patient consent Obtained.

  • Ethics approval Strasbourg University Hospitals (CPP 10-17).

  • Provenance and peer review Not commissioned; externally peer reviewed.