1. Academic Validation
  2. A cytotoxic anti-IL-3Rα antibody targets key cells and cytokines implicated in systemic lupus erythematosus

A cytotoxic anti-IL-3Rα antibody targets key cells and cytokines implicated in systemic lupus erythematosus

  • JCI Insight. 2016 May 5;1(6):e86131. doi: 10.1172/jci.insight.86131.
Shereen Oon 1 2 3 Huy Huynh 4 Tsin Yee Tai 4 Milica Ng 4 Katherine Monaghan 4 Mark Biondo 4 Gino Vairo 4 Eugene Maraskovsky 4 Andrew D Nash 4 Ian P Wicks 1 2 3 Nicholas J Wilson 4
Affiliations

Affiliations

  • 1 Division of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
  • 2 Department of Rheumatology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.
  • 3 The University of Melbourne, Parkville, Victoria, Australia.
  • 4 CSL Limited, Parkville, Victoria, Australia.
Abstract

To date, the major target of biologic therapeutics in systemic lupus erythematosus (SLE) has been the B cell, which produces pathogenic autoantibodies. Recently, targeting type I IFN, which is elaborated by plasmacytoid dendritic cells (pDCs) in response to endosomal TLR7 and TLR9 stimulation by SLE immune complexes, has shown promising results. pDCs express high levels of the IL-3Rα chain (CD123), suggesting an alternative potential targeting strategy. We have developed an anti-CD123 monoclonal antibody, CSL362, and show here that it affects key cell types and cytokines that contribute to SLE. CSL362 potently depletes pDCs via antibody-dependent cell-mediated cytotoxicity, markedly reducing TLR7, TLR9, and SLE serum-induced IFN-α production and IFN-α-upregulated gene expression. The antibody also inhibits TLR7- and TLR9-induced plasmablast expansion by reducing IFN-α and IL-6 production. These effects are more pronounced than with IFN-α blockade alone, possibly because pDC depletion reduces production of other IFN subtypes, such as type III, as well as non-IFN proinflammatory cytokines, such as IL-6. In addition, CSL362 depletes basophils and inhibits IL-3 signaling. These effects were confirmed in cells derived from a heterogeneous population of SLE donors, various IFN-dependent autoimmune diseases, and healthy controls. We also demonstrate in vivo activity of CSL362 following its s.c. administration to cynomolgus monkeys. This spectrum of effects provides a preclinical rationale for the therapeutic evaluation of CSL362 in SLE.

Figures
Products