AAV‐mediated delivery of an anti‐BACE1 VHH alleviates pathology in an Alzheimer's disease model

Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood–brain b...

Full description

Saved in:
Bibliographic Details
Published inEMBO molecular medicine Vol. 14; no. 4; pp. e09824 - n/a
Main Authors Marino, Marika, Zhou, Lujia, Rincon, Melvin Y, Callaerts‐Vegh, Zsuzsanna, Verhaert, Jens, Wahis, Jérôme, Creemers, Eline, Yshii, Lidia, Wierda, Keimpe, Saito, Takashi, Marneffe, Catherine, Voytyuk, Iryna, Wouters, Yessica, Dewilde, Maarten, Duqué, Sandra I, Vincke, Cécile, Levites, Yona, Golde, Todd E, Saido, Takaomi C, Muyldermans, Serge, Liston, Adrian, De Strooper, Bart, Holt, Matthew G
Format Journal Article
LanguageEnglish
Published England EMBO Press 07.04.2022
John Wiley and Sons Inc
Springer Nature
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood–brain barrier (BBB) penetration. Here, we propose a gene transfer strategy based on BBB‐crossing adeno‐associated virus (AAV)‐based vectors to deliver VHH directly into the CNS. As a proof‐of‐concept, we explored the potential of AAV‐delivered VHH to inhibit BACE1, a well‐characterized target in Alzheimer’s disease. First, we generated a panel of VHHs targeting BACE1, one of which, VHH‐B9, shows high selectivity for BACE1 and efficacy in lowering BACE1 activity in vitro. We further demonstrate that a single systemic dose of AAV‐VHH‐B9 produces positive long‐term (12 months plus) effects on amyloid load, neuroinflammation, synaptic function, and cognitive performance, in the AppNL‐G‐F Alzheimer’s mouse model. These results constitute a novel therapeutic approach for neurodegenerative diseases, which is applicable to a range of CNS disease targets. Synopsis VHH and blood‐brain‐barrier (BBB)‐crossing AAV‐based vectors are combined to achieve highly‐specific, long‐term BACE1 inhibition in a mouse model of Alzheimer's disease (AD). A gene transfer strategy based on BBB‐crossing AAV vectors is developed to deliver VHH single domain antibodies directly into the CNS. VHH‐B9 is generated to target BACE1, an enzyme critical to Aβ generation in AD, and incorporated into an AAV‐PHP.B‐based vector. A single dose of AAV‐VHH resulted in long‐term VHH expression in the AppNL‐G‐F mouse model, with concomitant improvements in cognitive status, amyloidosis, neuroinflammation, and synaptic function. VHH and blood‐brain‐barrier (BBB)‐crossing AAV‐based vectors are combined to achieve highly‐specific, long‐term BACE1 inhibition in a mouse model of Alzheimer's disease (AD).
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
These authors contributed equally to this work
ISSN:1757-4676
1757-4684
DOI:10.15252/emmm.201809824