brain penetrating bifunctional erythropoietin–transferrin receptor antibody fusion protein for Alzheimer's disease

R Chang, A Al Maghribi, V Vanderpoel… - Molecular …, 2018 - ACS Publications
R Chang, A Al Maghribi, V Vanderpoel, V Vasilevko, DH Cribbs, R Boado, WM Pardridge
Molecular pharmaceutics, 2018ACS Publications
Erythropoietin (EPO), a glycoprotein cytokine essential to hematopoiesis, has
neuroprotective effects in rodent models of Alzheimer's disease (AD). However, high
therapeutic doses or invasive routes of administration of EPO are required to achieve
effective brain concentrations due to low blood–brain barrier (BBB) penetrability, and high
EPO doses result in hematopoietic side effects. These obstacles can be overcome by
engineering a BBB-penetrable analog of EPO, which is rapidly cleared from the blood, by …
Erythropoietin (EPO), a glycoprotein cytokine essential to hematopoiesis, has neuroprotective effects in rodent models of Alzheimer’s disease (AD). However, high therapeutic doses or invasive routes of administration of EPO are required to achieve effective brain concentrations due to low blood–brain barrier (BBB) penetrability, and high EPO doses result in hematopoietic side effects. These obstacles can be overcome by engineering a BBB-penetrable analog of EPO, which is rapidly cleared from the blood, by fusing EPO to a chimeric monoclonal antibody targeting the transferrin receptor (cTfRMAb), which acts as a molecular Trojan horse to ferry the EPO into the brain via the transvascular route. In the current study, we investigated the effects of the BBB-penetrable analog of EPO on AD pathology in a double transgenic mouse model of AD. Five and a half month old male APPswe/PSEN1dE9 (APP/PS1) transgenic mice were treated with saline (n = 10) or the BBB-penetrable EPO (n = 10) 3 days/week intraperitoneally for 8 weeks, compared to same-aged C57BL/6J wild-type mice treated with saline (n = 8) with identical regiment. At 9 weeks following treatment initiation, exploration and spatial memory were assessed with the open-field and Y-maze test, mice were sacrificed, and brains were evaluated for Aβ peptide load, synaptic loss, BBB disruption, microglial activation, and microhemorrhages. APP/PS1 mice treated with the BBB-penetrable cTfRMAb-EPO fusion protein had significantly lower cortical and hippocampal Aβ peptide number (p < 0.05) and immune-positive area (p < 0.05), a decrease in hippocampal synaptic loss (p < 0.05) and cortical microglial activation (p < 0.001), and improved spatial memory (p < 0.05) compared with APP/PS1 saline controls. BBB-penetrating EPO was not associated with microhemorrhage development. The cTfRMAb-EPO fusion protein offers therapeutic benefits by targeting multiple targets of AD pathogenesis and progression (Aβ load, synaptic loss, microglial activation) and improving spatial memory in the APP/PS1 mouse model of AD.
ACS Publications