Molecular Therapy: Nucleic Acids (Sep 2022)

Identification and characterization of a MAPT-targeting locked nucleic acid antisense oligonucleotide therapeutic for tauopathies

  • Amy Easton,
  • Marianne L. Jensen,
  • Congwei Wang,
  • Peter H. Hagedorn,
  • Yuwen Li,
  • Michael Weed,
  • Jere E. Meredith,
  • Valerie Guss,
  • Kelli Jones,
  • Martin Gill,
  • Carol Krause,
  • Jeffrey M. Brown,
  • Lisa Hunihan,
  • Joanne Natale,
  • Alda Fernandes,
  • Yifeng Lu,
  • Joe Polino,
  • Mark Bookbinder,
  • Greg Cadelina,
  • Yulia Benitex,
  • Ramola Sane,
  • John Morrison,
  • Dieter Drexler,
  • Stephen E. Mercer,
  • Charlotte Bon,
  • Nikhil J. Pandya,
  • Ravi Jagasia,
  • Tai-Hsien Ou Yang,
  • Tania Distler,
  • Fiona Grüninger,
  • Michael Meldgaard,
  • Marco Terrigno,
  • John E. Macor,
  • Charles F. Albright,
  • James Loy,
  • Anja M. Hoeg,
  • Richard E. Olson,
  • Angela M. Cacace

Journal volume & issue
Vol. 29
pp. 625 – 642

Abstract

Read online

Tau is a microtubule-associated protein (MAPT, tau) implicated in the pathogenesis of tauopathies, a spectrum of neurodegenerative disorders characterized by accumulation of hyperphosphorylated, aggregated tau. Because tau pathology can be distinct across diseases, a pragmatic therapeutic approach may be to intervene at the level of the tau transcript, as it makes no assumptions to mechanisms of tau toxicity. Here we performed a large library screen of locked-nucleic-acid (LNA)-modified antisense oligonucleotides (ASOs), where careful tiling of the MAPT locus resulted in the identification of hot spots for activity in the 3′ UTR. Further modifications to the LNA design resulted in the generation of ASO-001933, which selectively and potently reduces tau in primary cultures from hTau mice, monkey, and human neurons. ASO-001933 was well tolerated and produced a robust, long-lasting reduction in tau protein in both mouse and cynomolgus monkey brain. In monkey, tau protein reduction was maintained in brain for 20 weeks post injection and corresponded with tau protein reduction in the cerebrospinal fluid (CSF). Our results demonstrate that LNA-ASOs exhibit excellent drug-like properties and sustained efficacy likely translating to infrequent, intrathecal dosing in patients. These data further support the development of LNA-ASOs against tau for the treatment of tauopathies.

Keywords