Cargando…

SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease

BACKGROUND: Alzheimer’s disease (AD) immunotherapy with antibodies targeting amyloid-β (Aβ) has been extensively explored in clinical trials. The aim of this study was to study the long-term brain distribution of two radiolabeled monoclonal Aβ antibody variants – RmAb158, the recombinant murine vers...

Descripción completa

Detalles Bibliográficos
Autores principales: Gustavsson, Tobias, Syvänen, Stina, O’Callaghan, Paul, Sehlin, Dag
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7504681/
https://www.ncbi.nlm.nih.gov/pubmed/32951598
http://dx.doi.org/10.1186/s40035-020-00214-1
_version_ 1783584679810564096
author Gustavsson, Tobias
Syvänen, Stina
O’Callaghan, Paul
Sehlin, Dag
author_facet Gustavsson, Tobias
Syvänen, Stina
O’Callaghan, Paul
Sehlin, Dag
author_sort Gustavsson, Tobias
collection PubMed
description BACKGROUND: Alzheimer’s disease (AD) immunotherapy with antibodies targeting amyloid-β (Aβ) has been extensively explored in clinical trials. The aim of this study was to study the long-term brain distribution of two radiolabeled monoclonal Aβ antibody variants – RmAb158, the recombinant murine version of BAN2401, which has recently demonstrated amyloid removal and reduced cognitive decline in AD patients, and the bispecific RmAb158-scFv8D3, which has been engineered for enhanced brain uptake via transferrin receptor-mediated transcytosis. METHODS: A single intravenous injection of iodine-125 ((125)I)-labeled RmAb158-scFv8D3 or RmAb158 was administered to AD transgenic mice (tg-ArcSwe). In vivo single-photon emission computed tomography was used to investigate brain retention and intrabrain distribution of the antibodies over a period of 4 weeks. Activity in blood and brain tissue was measured ex vivo and autoradiography was performed in combination with Aβ and CD31 immunostaining to investigate the intrabrain distribution of the antibodies and their interactions with Aβ. RESULTS: Despite faster blood clearance, [(125)I]RmAb158-scFv8D3 displayed higher brain exposure than [(125)I]RmAb158 throughout the study. The brain distribution of [(125)I]RmAb158-scFv8D3 was more uniform and coincided with parenchymal Aβ pathology, while [(125)I]RmAb158 displayed a more scattered distribution pattern and accumulated in central parts of the brain at later times. Ex vivo autoradiography indicated greater vascular escape and parenchymal Aβ interactions for [(125)I]RmAb158-scFv8D3, whereas [(125)I]RmAb158 displayed retention and Aβ interactions in lateral ventricles. CONCLUSIONS: The high brain uptake and uniform intrabrain distribution of RmAb158-scFv8D3 highlight the benefits of receptor-mediated transcytosis for antibody-based brain imaging. Moreover, it suggests that the alternative transport route of the bispecific antibody contributes to improved efficacy of brain-directed immunotherapy.
format Online
Article
Text
id pubmed-7504681
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-75046812020-09-23 SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease Gustavsson, Tobias Syvänen, Stina O’Callaghan, Paul Sehlin, Dag Transl Neurodegener Research BACKGROUND: Alzheimer’s disease (AD) immunotherapy with antibodies targeting amyloid-β (Aβ) has been extensively explored in clinical trials. The aim of this study was to study the long-term brain distribution of two radiolabeled monoclonal Aβ antibody variants – RmAb158, the recombinant murine version of BAN2401, which has recently demonstrated amyloid removal and reduced cognitive decline in AD patients, and the bispecific RmAb158-scFv8D3, which has been engineered for enhanced brain uptake via transferrin receptor-mediated transcytosis. METHODS: A single intravenous injection of iodine-125 ((125)I)-labeled RmAb158-scFv8D3 or RmAb158 was administered to AD transgenic mice (tg-ArcSwe). In vivo single-photon emission computed tomography was used to investigate brain retention and intrabrain distribution of the antibodies over a period of 4 weeks. Activity in blood and brain tissue was measured ex vivo and autoradiography was performed in combination with Aβ and CD31 immunostaining to investigate the intrabrain distribution of the antibodies and their interactions with Aβ. RESULTS: Despite faster blood clearance, [(125)I]RmAb158-scFv8D3 displayed higher brain exposure than [(125)I]RmAb158 throughout the study. The brain distribution of [(125)I]RmAb158-scFv8D3 was more uniform and coincided with parenchymal Aβ pathology, while [(125)I]RmAb158 displayed a more scattered distribution pattern and accumulated in central parts of the brain at later times. Ex vivo autoradiography indicated greater vascular escape and parenchymal Aβ interactions for [(125)I]RmAb158-scFv8D3, whereas [(125)I]RmAb158 displayed retention and Aβ interactions in lateral ventricles. CONCLUSIONS: The high brain uptake and uniform intrabrain distribution of RmAb158-scFv8D3 highlight the benefits of receptor-mediated transcytosis for antibody-based brain imaging. Moreover, it suggests that the alternative transport route of the bispecific antibody contributes to improved efficacy of brain-directed immunotherapy. BioMed Central 2020-09-21 /pmc/articles/PMC7504681/ /pubmed/32951598 http://dx.doi.org/10.1186/s40035-020-00214-1 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Gustavsson, Tobias
Syvänen, Stina
O’Callaghan, Paul
Sehlin, Dag
SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title_full SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title_fullStr SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title_full_unstemmed SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title_short SPECT imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of Alzheimer’s disease
title_sort spect imaging of distribution and retention of a brain-penetrating bispecific amyloid-β antibody in a mouse model of alzheimer’s disease
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7504681/
https://www.ncbi.nlm.nih.gov/pubmed/32951598
http://dx.doi.org/10.1186/s40035-020-00214-1
work_keys_str_mv AT gustavssontobias spectimagingofdistributionandretentionofabrainpenetratingbispecificamyloidbantibodyinamousemodelofalzheimersdisease
AT syvanenstina spectimagingofdistributionandretentionofabrainpenetratingbispecificamyloidbantibodyinamousemodelofalzheimersdisease
AT ocallaghanpaul spectimagingofdistributionandretentionofabrainpenetratingbispecificamyloidbantibodyinamousemodelofalzheimersdisease
AT sehlindag spectimagingofdistributionandretentionofabrainpenetratingbispecificamyloidbantibodyinamousemodelofalzheimersdisease