Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 1.
Published in final edited form as: Neuromolecular Med. 2019 Jan 17;21(1):12–24. doi: 10.1007/s12017-019-08524-y

The promises and challenges of erythropoietin for treatment of Alzheimer’s disease

Jiahong Sun 1, Jan Michelle Martin 2, Victoria Vanderpoel 3, Rachita K Sumbria 1,4,*
PMCID: PMC6407698  NIHMSID: NIHMS1015777  PMID: 30656553

Abstract

Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder in the world, and intracellular neurofibrillary tangles and extracellular amyloid-beta protein deposits represent the major pathological hallmarks of the disease. Currently available treatments provide some symptomatic relief but fail to modify primary pathological processes that underlie the disease. Erythropoietin (EPO), a hematopoietic growth factor, acts primarily to stimulate erythroid cell production, and is clinically used to treat anemia. EPO has evolved as a therapeutic agent for neurodegeneration and has improved neurological outcomes and AD pathology in rodents. However, penetration of the blood-brain barrier (BBB) and negative hematopoietic effects are the two major challenges for the therapeutic development of EPO for chronic neurodegenerative diseases like AD. The transferrin receptors at the BBB, which are responsible for transporting transferrin-bound iron from the blood into the brain parenchyma, can be used to shuttle therapeutic molecules across the BBB. In this review, we discuss the role of EPO as a potential neurotherapeutic for AD, challenges associated with EPO development for AD, and targeting the BBB transferrin receptor for EPO brain delivery.

Keywords: erythropoietin, Alzheimer’s disease, transferrin receptor, blood-brain barrier, molecular Trojan horse

(I) Pathological mechanisms and recent insights of Alzheimer’s disease

Alzheimer’s disease (AD), a neurodegenerative disorder accounting for more than 60% of the 36.5 million dementia cases worldwide, is characterized by progressive memory loss, cognitive decline, and an inability to complete daily tasks such as driving, housekeeping, and keeping track of finances (Sosa-Ortiz et al. 2012). In 2017, the average cost of health care and long-term care for a patient with AD was $48,000/year. Furthermore, care provided by family members and unpaid caregivers was estimated at $232 billion (2018 Alzheimer’s disease facts and figures 2018). Since AD incidence in individuals ≥85 years old is expected to increase from 40% in 2001 to 62% in 2050 with the aging of the baby boomer population, the disease burden of AD in coming years will be immense (Hebert et al. 2001).

During early stages of AD, patients may exhibit either anosognosia, an inability to perceive their illness, or simply ignore their symptoms, attributing their memory loss to normal aging (Barrett et al. 2005). These factors greatly contribute to the delay in diagnosis and the underestimation of disease incidence and prevalence.

As the disease progresses, memory impairment is followed by motor and visuospatial deficits, with patients experiencing extrapyramidal symptoms, apraxia or seizures (Portet et al. 2009; Giannakopoulos et al. 1998; Vossel et al. 2017). In late-stage AD, patients may become disinhibited, withdrawn or mute, therefore requiring daily supervision while eating, dressing, and bathing (Sarazin et al. 2005). These patients begin to rely heavily on caregivers, leading to an increased incidence of caregiver burnout (Dauphinot et al. 2015). This burnout often persists for years, as the median time from diagnosis to death in AD patients is 3.8 years, with many dying of malnutrition or infection (James et al. 2014; Mitchell et al. 2009). Some patients may live with the disease much longer, causing a strain on financial and societal resources for decades.

Pathogenesis

Many hypotheses have been developed to elucidate AD pathogenesis; however, the amyloid cascade, cholinergic, and inflammatory hypotheses remain among the most heavily studied (J. Hardy 2009; De Felice et al. 2008; Eikelenboom et al. 2006; Bartus et al. 1982). An infectious origin of the disease has also gained traction in recent years (Mawanda and Wallace 2013).

The Amyloid Cascade Hypothesis

The leading model for AD progression is the amyloid cascade hypothesis, which posits that the deposition of β-amyloid (Aβ) is the initial pathological event in AD leading to the formation of senile plaques followed by neurofibrillary tangles (NFT). This leads to downstream neuronal cell death and ultimately dementia due to central nervous system (CNS) dysfunction (J. A. Hardy and Higgins 1992). Aβ is a protein consisting of 38–43 amino acids; it is formed proteolytically when amyloid precursor protein (APP) is cleaved by β- and γ-secretases via the amyloidogenic pathway (Haass and De Strooper 1999; Chow et al. 2010; Gremer et al. 2017). The role of APP in synaptic transmission helps explain its endogenous abundance within the brain (Priller et al. 2006). Two forms of Aβ that are abundantly found in the AD brain are Aβ40 and Aβ42, the latter being more abundant and exhibiting greater CNS toxicity (Potter et al. 2013; Wildburger et al. 2017). As Aβ accumulates within the brain, it forms insoluble neuritic plaques, which contain a central amyloid core surrounded by activated microglia (Hong et al. 2016). Furthermore, Aβ plaque accumulation induces hyperphosphorylation of tau protein resulting in further toxicity to the CNS (De Felice et al. 2008). Hyperphosphorylated tau is unable to bind and stabilize microtubules, and instead aggregates to form NFT, leading to downstream synaptic loss and impaired neuroregeneration (Z. Zhang et al. 2014). In concurrence with their involvement in AD pathology, Aβ and tau are often measured in the cerebrospinal fluid of patients for disease staging and prognostication (Braak et al. 2006; Thal et al. 2002), and a lot of research efforts are focused on reducing the burden of Aβ and NFT aggregation in the brain to stop AD progression (Panza et al. 2012).

Neurotransmitter Dysfunction and Synaptic Loss

Aβ and NFT disrupt normal synaptic transmission to neighboring cerebral areas (DeKosky and Scheff 1990). Neurotransmitters are often seen in abnormal amounts in AD patients (Lombardo and Maskos 2015; Wenk 2003; Kandimalla and Reddy 2017). For example, choline acetyltransferase, which is involved in the synthesis of acetylcholine, one of the most studied neurotransmitters in AD pathogenesis, is decreased in post-mortem AD brains (Slotkin et al. 1990). The excitatory neurotransmitter, glutamate, accumulates in AD brains, leading to increased N-methyl-D-aspartate (NMDA) receptor activation (Revett et al. 2013) and Aβ plaque formation (Dinamarca et al. 2012). Furthermore, Aβ plaque formation weakens synaptic inhibition via downregulation of gamma-Aminobutyric acid receptors, causing increased excitotoxicity in the CNS (Ulrich 2015). Serotonin receptors, which are involved in cognitive processing, are also downregulated in the cortical and hippocampal areas, contributing to cognitive decline in AD brains (Reynolds et al. 1995). Histamine, another neurotransmitter important in cognitive functioning, also plays a crucial role in the pathogenesis of AD (Kandimalla and Reddy 2017). Overall, improper synaptic communication has been linked to an imbalance in neurotransmitter levels, causing downstream synaptic loss and cognitive decline. Thus, drugs that work to replenish normal neurotransmitter levels have been developed for AD (Reisberg et al. 2003; Bartus et al. 1982).

Mitochondrial Dysfunction and Oxidative Stress

Mitochondrial dysfunction, a naturally occurring part of the aging process, is an early finding in AD pathogenesis. Aβ and tau aggregates impair mitochondrial function, causing oxidative stress within the cell that results in free radical damage (Eckert et al. 2012). This oxidative stress induces further Aβ accumulation and tau phosphorylation, causing a self-perpetuating cycle of mitochondrial dysfunction, oxidative damage, and plaque formation (Leuner et al. 2012; Melov et al. 2007).

Microglial Activation and Inflammation

The role of neuroinflammation in AD pathogenesis is now widely recognized. In the CNS, microglia are involved in synaptic remodeling and tissue surveillance. However, pathologic stimuli, including Aβ plaque accumulation, can result in sustained microglial activation, and in the AD brain, Aβ plaques and NFT are often found surrounded by activated microglia (Wang et al. 2015). Activated microglia are involved in Aβ plaque clearance; however, sustained microglial activation can perturb this clearance process. Activated microglia secrete pro-inflammatory cytokines like tumor necrosis factor α (TNF-α), and chemokines that further propagate an inflammatory response and neuronal degeneration in AD (Fan et al. 2015; Rogers et al. 1996; Heneka et al. 2015). Astrocytes, T-cells, complement proteins, and other acute phase reactants are also recruited to the sites of inflammation, causing an inflammatory storm that results in tissue damage. Moreover, widespread inflammation encourages free radical formation, inciting further damage (McGeer and McGeer 2003). Besides accelerating the progression of AD, growing evidence shows that inflammatory processes may drive AD pathology, thus providing a long window of opportunity for therapeutic intervention (Wyss-Coray 2006). Therapeutic strategies to mitigate neuroinflammation in AD, including the development of nonsteroidal anti-inflammatory drugs, peroxisome proliferator-activated receptor (PPAR)-γ agonists and cytokine inhibitors have been/or are under investigation (Ardura-Fabregat et al. 2017).

Genetic Risk Factors

Many genes play a role in the development of AD; however, the most heavily associated is the gene encoding apolipoprotein-E (Apo-E), a protein involved in cholesterol transport (Castellano et al. 2011). The Apo-E ε4 allele contributes to poor Aβ clearance from the brain and is a strong genetic risk factor for late-onset, sporadic AD. On the other hand, the Apo-E ε2 allele confers AD protective effects and is associated with increased age at onset (Castellano et al. 2011). Apart from Apo-E, increased APP expression greatly contributes to disease progression. Encoded by a gene on chromosome 21q21.3–22.05, higher levels of APP cause familial early-onset AD in patients with Down syndrome, owing to their extra copy of chromosome 21 (Schupf et al. 1998). Lastly, mutations in the genes encoding presenilins, neuronal proteins widely expressed in the CNS, are also associated with familial early-onset AD. Presenilin 1 and presenilin 2 found on chromosome 14q24.3 and 1q31–42 respectively, are critical components of the ɣ-secretase complex and are involved in the amyloidogenic cleavage of APP (Brunkan and Goate 2005; Rogaev et al. 1995). Other emerging genes associated with AD risk include genes involved in synapse turnover, clathrin-mediated endocytosis, amyloid clearance, and inflammation cascades (Karch and Goate 2015).

Current Treatment Options

Currently, there is no pharmacologic cure for AD, and drugs are prescribed to enhance cognition and treat AD symptoms. FDA-approved drugs for AD belong to two classes: acetylcholinesterase inhibitors and NMDA receptor antagonists. Acetylcholinesterase inhibitors currently approved for AD are donepezil, rivastigmine, and galantamine. These drugs act by increasing the amount of acetylcholine in the brain to reestablish proper synaptic communication (Bartus et al. 1982). However, these drugs only perform with modest efficacy and are approved for mild-to-moderate AD. The NMDA receptor antagonist memantine, on the other hand, works by blocking NMDA receptors to regulate glutamatergic neurotransmission and prevent synaptic loss, which is thought to improve cognition in AD (Danysz et al. 2000). AD patients taking memantine show improved functioning and decreased rate of decline upon cognition testing (van Marum 2009).

Apart from symptomatic treatment for AD, there are a number of anti-Aβ and anti-tau therapies in different stages of clinical development (Bachurin et al. 2017). AN1792 was the first active immunotherapy against Aβ42, however was discontinued due to reports of meningoencephalitis (Nicoll et al. 2003). Passive forms of immunotherapy against Aβ, including bapineuzumab and solanezumab, which act on soluble Aβ, performed poorly in Phase III clinical trials (Panza et al. 2014). Other anti-Aβ agents under development include antibodies that target aggregated Aβ, including aducanumab and gantenerumab. Aducanumab slowed cognitive decline and reduced Aβ plaques in patients with early-stage AD, and is currently being tested in Phase III trials of early-onset AD. Gantenerumab is currently in phase III trials for prodromal AD.

Additionally, β- and γ-secretase inhibitors that prevent the cleavage of APP to pathogenic Aβ, like verubecestat and semagacestat, have been investigated to reduce the burden of Aβ plaques in the brain; however, clinical trials were largely unsuccessful (Wan et al. 2009; Doody et al. 2013). Meanwhile, tau-based immunotherapies are currently in early stages of clinical development. RG7345, which is a humanized monoclonal antibody targeting the tau phosphoepitope pS422, caused an improvement in a transgenic mouse AD model but was discontinued after Phase I clinical trials (Collin et al. 2014; Sigurdsson 2018). Besides these approaches, drug candidates that target different neuronal receptors (e.g. serotonin receptor ligands), enzyme inhibitors (e.g. lipoprotein-associated phospholipase A2 inhibitor), neurotrophic drugs, and anti-inflammatory (e.g. etanercept) approaches, are currently under development (Bachurin et al. 2017).

(II) Erythropoietin and AD

Erythropoietin (EPO), a 30.4 kDa hematopoietic growth factor produced in the kidney, acts primarily to stimulate erythroid cell production by supporting the survival, proliferation and differentiation of erythroid progenitor cells (Jelkmann 2013). Recombinant human EPO is FDA approved and clinically used to treat anemia (Ng et al. 2003). In the past decade, over 60 studies have demonstrated the neuroprotective role of EPO in neurodegenerative conditions (Jelkmann 2005). As shown in Figure 1, EPO improves neurological outcome in experimental AD by reducing Aβ load, inflammation, oxidative stress, apoptosis and neuronal loss (Sargin et al. 2010). Overall, EPO has evolved as a therapeutic agent for CNS disorders given its robust non-hematopoietic neuroprotective effects.

Fig 1. The neuroprotective effect of EPO in AD.

Fig 1.

In experimental AD, EPO reduces Aβ toxicity, neuroinflammation and oxidative stress, and enhances neurogenesis and neurotrophic factor expression.

Expression of EPO and EPO receptor in the CNS

The EPO receptor (EPOR) is a highly conserved member of the cytokine receptor superfamily. EPOR expression in the brain is observed during development and adulthood in humans and other mammals (Marti et al. 1996). EPO expression is detected in neurons, astrocytes and pericytes, while EPOR is expressed in neurons, astrocytes and microglia (Ji 2016; Nagai et al. 2001). EPOR is prominently expressed in astrocytes, oligodendrocytes and certain neuronal populations in fetal brain during gestation (Jelkmann 2005), and brain EPOR expression is significantly reduced with development, indicating the vital role of EPO in brain maturation (Juul et al. 1998).

The expression of EPO and EPOR is controlled by hypoxia inducible factor-1 (HIF-1). This hypoxia-dependent transcription of EPO is regulated via the hypoxia response element in the 3’-flanking region of EPO gene that specifically binds to HIF-1 under hypoxic conditions (Haase 2013). Neuronal production of EPO is related to the oxygen tension of the medium in vitro, which is enhanced by hypoxia (Masuda et al. 1994). Further, astrocytic EPO expression is increased by more than 100-fold under hypoxic conditions (Marti et al. 1996). Radio-iodinated EPO studies in rats indicate that EPOR expression is localized to the hippocampal and cortical areas, which are most vulnerable to AD pathology and ischemic insult (Digicaylioglu et al. 1995). EPOR expression is also observed on the endothelial cells forming the blood-brain barrier (BBB), and EPOR is upregulated in the temporal cortex and hippocampus of patients with mild cognitive impairment and AD (Cornford and Hyman 2005; Assaraf et al. 2007).

Effect of EPO on AD pathology

Amyloid Toxicity

AD is pathologically characterized by the accumulation of Aβ and tau hyperphosphorylation in the hippocampus and cortex. Therefore, it is important for therapeutic AD agents to act on toxic Aβ buildup, association of newly cleaved Aβ fragments into oligomers and plaques, and tau phosphorylation. Intraperitoneal injections of EPO induce a 20% reduction of total Aβ load and a 59% reduction of Aβ40 protein level in mice; however, no significant decrease in Aβ42 load is observed after EPO treatment (Armand-Ugon et al. 2015). Tg2576 mice also exhibit a reduced amount of amyloid plaque after daily intraperitoneal injection of EPO for 5 days (S. T. Lee et al. 2012). Similarly, EPO reduces hippocampal Aβ accumulation in an intracerebroventricular streptozotocin-induced AD rat model (Samy et al. 2016). Intranasal-administration of EPO also decreases Aβ deposits in an APP/PS1 transgenic AD mouse model (Rodriguez Cruz et al. 2017). In vitro, EPO is reported to attenuate Aβ25–35-induced rat pheochromocytoma cell death by regulating anti-apoptotic protein expression (Ma et al. 2009). Further, EPO protects microglia mediated neuronal regeneration and Aβ clearance, in vitro (Shang et al. 2012). A recent study reported that EPO alleviates tau hyperphosphorylation via regulation of glycosynthase kinase-3β in a rodent AD model (Y. P. Li et al. 2015).

Mitochondrial Dysfunction and Oxidative Stress

Oxidative stress is linked to mitochondrial dysfunction in AD (Eckert et al. 2012), and EPO relieves oxidative stress by preventing the accumulation of lipid peroxidation in the hippocampus of AD mice (Maurice et al. 2013). In vitro, EPO treatment prior to Aβ25–35 exposure significantly elevates cell viability, decreases ROS production, and stabilizes mitochondrial membrane potential (G. Li et al. 2008). EPO further attenuates nitric oxide and staurosporine-induced oxidative toxicity in neonatal rat astrocytes by promoting the expression of heme oxygenase-1 (Diaz et al. 2005), and EPO mediated anti-oxidant effects reduce neuronal damage by activated microglia (Wenker et al. 2013).

A massive release of synaptic glutamate in brain regions susceptible to Aβ-toxicity leads to excitotoxicity and oxidative stress, resulting in neurotoxicity. EPO prevents glutamate-induced excitotoxicity and cell death by inhibiting mitochondrial calcium overload in cultured rat neurons (Morishita et al. 1997). EPO activates several kinase cascades to confer resistance against oxidative stress-induced apoptosis (G. Li et al. 2008). It has also been demonstrated that EPO inhibits Bax/Bcl complex-mediated apoptosis induced by Aβ (Rodriguez Cruz et al. 2017; Y. P. Li et al. 2015).

Neurotrophic Effects

The role of EPO as a neurotrophin and in neurogenesis has been well documented. Intracerebroventricular administration of EPO significantly increases brain-derived neurotrophic factor (BDNF) expression in the brain. EPO also induces BDNF production and long-term activation of its specific receptor, tyrosine receptor kinase B, resulting in EPO mediated neuroprotection against neurotoxin trimethyltin (Viviani et al. 2005). Notably, BDNF levels are reduced in AD, and exogenous BDNF offers protection against Aβ- and tau-related neurodegeneration (Jiao et al. 2016). The role of EPO-derived BDNF in AD, however, needs further investigation.

EPO augments choline acetyltransferase activity in mouse embryonic primary septal neurons and a cholinergic hybridoma cell line. Moreover, EPO promotes survival of septal cholinergic neurons in adult rats which have undergone fimbria-fornix transections (Konishi et al. 1993). Intraperitoneal administration of EPO spurs significant neurogenesis in the dentate gyrus in the streptozotocin-induced AD rat model; however, EPO does not change neurogenesis in the dentate gyrus of intact animals (Arabpoor et al. 2012). Tg2576 mice treated with EPO show increased hippocampal and cortical neurogenesis identified by 5-bromo-2-deoxyuridine fluorescent labeling, and increased synaptophysin expression. Further, EPO enhances endothelial proliferation in the brains of Tg2576 mice, indicating the positive role of EPO in angiogenesis (S. T. Lee et al. 2012). Overall, these studies indicate that EPO acts as a potent neurotrophic factor to influence differentiation, maintenance and regeneration in the CNS.

Neuroinflammation

Increasing evidence suggests that neuroinflammation in AD is implicated as a significant contributor to disease pathogenesis and progression. With respect to neuroinflammation, EPO prevents the Aβ25–35-induced increase in TNF-α and interleukin (IL) production in a rodent AD model (Maurice et al. 2013). Besides its effect in AD rodent models, EPO has been reported to decrease production of TNF-α, IL-6, and monocyte chemoattractant protein-1 in an ischemic stroke model (Villa et al. 2003). Moreover, EPO also diminishes inflammation and glial activation in a model of brain inflammation (Shang et al. 2011). These data collectively suggest that EPO act as a protective cytokine in inflammatory pathologies of neurodegeneration, including AD.

The effect of immunomodulatory peptides on EPO expression in cultured human neurons, microglia, astrocytes, and oligodendrocytes has also been investigated. EPO expression in human astrocytes was reduced by pro-inflammatory cytokines including IL-1β, IL-6 and TNF-α, whereas, EPOR expression was markedly increased in human neuronal cells by TNF-α (Nagai et al. 2001). EPOR and EPO expression may thus be regulated by pro-inflammatory cytokines in the CNS.

Cognitive Function

AD is the most common cause of senile dementia and is characterized by a progressive decline in cognitive function. Behavioral tests commonly used in animal AD studies attempt to mimic deficits in memory and cognition of AD patients. In a contextual fear-conditioning test, EPO improves associative learning memory in aged transgenic AD mice (S. T. Lee et al. 2012). In a step-down inhibitory avoidance test, which measures short-term retention of an aversive stimulus, intraperitoneal EPO treatment restores Aβ42-induced loss of step-down latency (Y. P. Li et al. 2015). Intranasal EPO alleviates place learning and spontaneous alteration deficits in the Morris Water Maze, which is used to measure spatial and long-term memory by recording escape latency, distance and velocity (Rodriguez Cruz et al. 2017). EPO administration also prevents deficits in alternation behavior in Y-maze and novel object recognition procedures in Aβ25–35 injected mice (Rodriguez Cruz et al. 2017; Maurice et al. 2013). Non-spatial long-term memory function deficits, tested by the passive avoidance procedure, are observed in AD and stroke models, and these are improved by EPO treatment (Esmaeili Tazangi et al. 2015; Sakanaka et al. 1998). Using place learning in water maze, hippocampus-dependent spatial memory is improved by EPO (Maurice et al. 2013). Similarly, bilateral injection of Aβ into the rat cortex leads to memory loss and synaptic plasticity defect, which are recovered by EPO (Esmaeili Tazangi et al. 2015).

(III) Limitation of EPO as a CNS-penetrating drug

Though EPO and EPOR have a role as an “endogenous neuroprotective system” in the brain, BBB penetration and negative hematopoietic effects are the two major challenges that have to be addressed before EPO can be developed as a neurotherapeutic. EPO is a 30.4 kDa polar macromolecule and is larger than the molecular weight cut-off for lipid mediated free diffusion across the BBB (Pardridge 2005; Konofagou et al. 2012). The finding that the EPOR is expressed at the brain capillaries suggested that peripheral EPO may cross the BBB to produce therapeutic effects in the brain (Brines et al. 2000). Further, systemically administered biotinylated-EPO was detected surrounding the brain capillaries, and this transport of biotinylated-EPO was eliminated by co-administration of unlabeled EPO (Brines et al. 2000). Based on these findings, it was suggested that EPO is transported across the BBB via a specific receptor-mediated transport system, however, the origin of the biotin label detected in these studies (biotin conjugated to EPO or free biotin) was not confirmed. The presumption that EPO enters the CNS is also largely based on measurement of systemically administered EPO in the cerebrospinal fluid (CSF) (Brettschneider et al. 2006). It should be noted that movement from blood into the CSF is controlled by the blood-CSF-barrier, and all molecules present in the blood enter the CSF at a rate that is inversely related to their molecular weight (Pardridge 2016). Follow-up brain uptake studies using radiolabeled EPO in rodents and primates showed that the rate of EPO entry into the brain was very slow and that EPO entry into the brain was via a non-specific mechanism (Boado et al. 2010; Banks et al. 2004). Using the capillary depletion technique, which separates the brain homogenate into vascular pellet and postvascular supernatant, the rate of EPO entry into the brain was found to be comparable to that of a marker of brain blood volume (human IgG1) showing the limited transport of EPO across the BBB (Boado et al. 2010).

EPO is administered systemically or intranasally in experimental AD (Esmaeili Tazangi et al. 2015; Maurice et al. 2013; S. T. Lee et al. 2012; Y. P. Li et al. 2015; Armand-Ugon et al. 2015; Samy et al. 2016; Arabpoor et al. 2012). Systemic routes, though less invasive than the intracranial or intracerebroventricular routes used to bypass the BBB, require high dosage to infiltrate the brain and therefore result in heightened peripheral toxicity risk. As a red blood cell growth factor, high systemic EPO doses induce negative hematopoietic side effects including increased hematocrit, arterial hypertension, cerebral convulsion, thromboembolism, iron deficiency, and influenza-like syndrome (Lapchak 2010; Lundby et al. 2007). Such hematopoietic side-effects associated with high systemic doses limit the applicability of the systemic route of administration for EPO delivery for chronic diseases like AD that require long term treatment.

EPO analogs with reduced erythropoietic activity and associated vascular complications are currently under investigation (King et al. 2007). Asialo-EPO, which is produced by the removal of sialic acid residues from EPO to yield a molecule with enhanced clearance has no effect on erythropoiesis but exerts neuroprotective effects in animal models of neural diseases (Erbayraktar et al. 2003; Mennini et al. 2006). Another non-erythropoietic EPO derivative, carbamylated-EPO (CEPO), which is produced by the modification of lysine residues to homocitrulline, produces its protective effects through the EPOR/β common receptor, and not the classical EPOR. In an AD mouse model, CEPO treatment improved memory and modulated the expression of molecules involved in neurotransmission. However, compared to EPO, CEPO did not decrease Aβ plaque burden and soluble Aβ40 (Armand-Ugon et al. 2015). The CEPO-Fc variant, which is engineered to prolong the half-life of CEPO, was also found to be protective in a rodent AD model (Hooshmandi et al. 2018). Notably, these non-erythropoietic variants of EPO are also macromolecules with limited BBB penetrability across the BBB, comparable to that of EPO (Torup and Leist 2006).

Another variant of EPO, neuro-EPO, which contains low sialic acid, has been designed. Neuro-EPO is devoid of hematopoietic effects and is administered intranasally. Neuro-EPO prevents oxidative damage, neuroinflammation, apoptosis and cognitive deficit in an AD animal model (Rodriguez Cruz et al. 2017; Maurice et al. 2013). Even though intranasal administration is advantageous over systemic administration with respect to systemic side effects, the efficacy of intranasal drug delivery decreases as molecular weight and hydrophilicity of the drug increase (Lu et al. 2014). Moreover, intranasal delivery relies on widespread drug diffusion from the olfactory CSF to brain entry sites, and drug diffusion decreases exponentially with distance (Lochhead and Thorne 2012; Pardridge 2012). Large diffusion distances in human brains compared with smaller rodent brains may limit the efficacy of the intranasal route of administration in humans. This is in contrast to the transvascular route across the BBB that results in widespread drug delivery throughout the brain because of the density and ubiquity of the cerebral vasculature (Pardridge 2005). Hence, modifying EPO for direct and specific penetration of the BBB via the transvascular route and minimal hematopoietic effects is needed for AD.

(IV) BBB Receptor Mediated Transport of EPO

Molecular Trojan horse (MTH) technology holds promise for efficiently transporting neurotherapeutics across the BBB (Pardridge 2006, 2017). MTH technology harnesses endogenous receptor mediated transcytosis (RMT) systems to ferry macromolecules into the brain. For this, a fusion protein of the neurotherapeutic and a MTH, which is an antibody against an endogenous receptor expressed on the luminal side of brain endothelial cells, is engineered. Systemic injection of the neurotherapeutic-MTH complex triggers its binding to the endogenous RMT receptor on the luminal side of the BBB, followed by subsequent endocytosis and release of the complex on the abluminal side (Pardridge 2012, 2009). This approach can therefore be used to shuttle therapeutic molecules across the BBB non-invasively to treat CNS pathology (Preston et al. 2014). The two widely studied MTH vehicles are antibodies against the human insulin receptor and transferrin receptor (TfR) (Jones and Shusta 2007; Pardridge 2006).

The TfR system is responsible for transporting transferrin-bound iron from blood across the brain endothelium into the brain parenchyma (Roberts et al. 1993; Jefferies et al. 1984; Fishman et al. 1987). TfR is a type II transmembrane protein and is expressed at both the luminal and abluminal surface of brain endothelial cells (Huwyler and Pardridge 1998). There are two TfR isoforms, TfR1 and TfR2, and the primary TfR isoform expressed at the BBB is TfR1 (J. Y. Li et al. 2001). Upon binding of the iron-transferrin complex on the luminal surface, TfR enters the brain endothelial cells via endocytosis. Once inside the brain endothelial cells, the remaining mechanism for iron entry into the brain is debatable. One proposed mechanism is transport of the iron-transferrin-TfR complex from the luminal to the abluminal membrane of brain endothelial cells followed by release of iron and transferrin into the brain interstitium with subsequent reverse transcytosis of iron-free transferrin into the blood; while another is dissociation of the iron from the transferrin-TfR complex within the acidified endosomes in the brain endothelial cells and transcytosis of the iron across the abluminal membrane to enter the brain (Roberts et al. 1993; H. J. Lee et al. 2000; Dautry-Varsat et al. 1983; Moos et al. 2007; Y. Zhang and Pardridge 2001).

The common mechanism of transcytosis of varying MTHs across the abluminal membrane is also debated, and it is clear that antibodies against the TfR may differ based on their affinity for the TfR. Both high- and low-affinity, as well as monovalent and bivalent TfR antibodies have been engineered, and these different antibodies for the same TfR may warrant different success rates of entry into the brain parenchyma (Niewoehner et al. 2014; Yu et al. 2011; Weber et al. 2018; Hultqvist et al. 2017; Chang et al. 2018). Some studies suggest that TfR antibodies with high affinity can be trapped within the brain vasculature due to their high affinity, which prevents their transportation across the abluminal side of brain capillaries (Yu et al. 2011). Reducing the affinity of the TfR antibody to enhance RMT across the BBB has been suggested as an alternative. It should however be noted that high injection doses are needed to produce a therapeutic effect while using low-affinity TfR antibodies, and such high doses result in effector function associated toxicity (acute-clinical signs and reticulocyte suppression) (Couch et al. 2013). Low therapeutic doses of high-affinity TfR antibodies on the other hand do not produce an immune-response or reticulocyte suppression (Pardridge et al. 2018). Overall, as the second most abundant protein on the brain endothelial cells compared to other RMT systems, TfR is considered an ideal candidate for MTH drug-delivery of therapeutics across the BBB (Bien-Ly et al. 2014; Pardridge 2015).

To facilitate non-invasive transvascular EPO delivery into the brain, human EPO was re-engineered as a TfR antibody-EPO fusion protein, designated as cTfRMAb-EPO (Figure 2). The cTfRMAb domain of the fusion protein that enables BBB penetration is a rat/mouse chimeric antibody that is specific to the mouse TfR and binds to the mouse TfR with high affinity (KD of 2.6 ± 0.3 nM) in a bivalent configuration (Boado et al. 2009). The EPO domain of the fusion protein binds to the mouse EPOR with an ED50 = 0.33 ± 0.04 nM to produce neuroprotection in brain behind the intact BBB (Zhou et al. 2010). Intravenous injection of radiolabeled cTfRMAb-EPO in mice results in a brain uptake of 2.0 ± 0.1% injected dose/g of brain and ~70% of the fusion protein is transcytosed across the BBB into the brain parenchyma measured using the capillary depletion technique (Zhou et al. 2010). The plasma clearance of the cTfRMAb-EPO fusion protein is > 10-fold higher than that of EPO due to the rapid peripheral TfR mediated clearance of the fusion protein (Kato et al. 1998). Such a pharmacokinetic profile of the cTfRMAb-EPO is desirable given that increased plasma clearance lowers the potential for peripheral hematopoietic side effects associated with the EPO domain of the fusion protein. In fact, desialylated variants of EPO have a reduced half-life and increased clearance, and offer the possibility to uncouple stimulation of erythropoiesis from tissue-protective effects of EPO (Erbayraktar et al. 2003; Torup and Leist 2006).

Fig 2. Schematic of routes of EPO brain delivery.

Fig 2.

Penetration of the BBB is a major challenge of EPO administration for CNS diseases including AD. One hypothesis is the existence of EPOR on the luminal side of the brain capillaries that is presumed to provide a route for circulating EPO to enter the brain. This route of EPO entry remains debatable. In the absence of such a receptor, brain delivery of EPO requires compromised BBB permeability. Alternatively, endogenous transferrin receptors (TfR), which are receptor-mediated transcytosis systems, and are highly expressed at the BBB can be harnessed to ferry EPO into the brain. A fusion protein of EPO and a chimeric monoclonal antibody against TfR has been engineered and designated, cTfRMAb-EPO. Upon binding to the BBB TfR, the cTfRMAb-EPO fusion protein is ferried into the brain to enable EPO delivery into the brain across the BBB.

Chronic 3 days a week intravenous treatment with the cTfRMAb-EPO fusion protein for 3 weeks offered neuroprotection in a mouse model of neural disease and resulted only in a 10% increase in hematocrit after 2 weeks of treatment with no further hematocrit increase (Zhou et al. 2011). The dose of the cTfRMAb-EPO fusion protein in the aforementioned study was 1mg/kg which is equivalent to an EPO dose of 200 μg/kg, since the cTfRMAb-EPO fusion protein is 20% EPO based on the amino-acid sequence of the fusion protein. Since the clearance of the cTfRMAb-EPO is > 10-fold faster than EPO, the equivalent EPO dose is 20 μg/kg per dose or 60 μg/kg per week. This is lower than the doses of EPO that are therapeutic in experimental models of AD (S. T. Lee et al. 2012; Armand-Ugon et al. 2015). In a recent study, chronic treatment with the cTfRMAb-EPO fusion protein for 8 weeks reduced Aβ pathology, microglial activation and synaptic loss in a transgenic mouse model of AD. cTfRMAb-EPO further improved performance in the modified Y-maze, showing that treatment with the BBB-penetrable EPO improved spatial memory in experimental AD (Chang et al. 2018). High-affinity humanized TfR antibodies targeting the human TfR have been engineered and their ability to act as efficient MTHs has been confirmed in primates (Pardridge et al. 2018; Yu et al. 2014). Species-specific TfR antibody-EPO fusion proteins can thus be engineered to translate these effects of the BBB-penetrable-EPO into other species. Overall, engineering EPO variants that target the BBB TfR to enable BBB penetration may provide a promising approach to further the development of EPO for chronic neurodegenerative conditions including AD.

Summary

EPO has evolved as a therapeutic agent for neurodegenerative conditions given its robust non-hematopoietic neuroprotective effects. However, penetration of the BBB and negative hematopoietic effects are the two major challenges for the development of EPO for chronic neurodegenerative diseases like AD. Brain drug delivery strategies, such as the MTH technology that harnesses endogenous RMT systems like the BBB TfR, hold promise for efficiently transporting EPO across the BBB for AD, while increasing systemic clearance to minimize hematopoietic side-effects.

Acknowledgement

This work was supported by a grant from The National Institute of Health, NIA R21AG055949 (to RKS).

Footnotes

Conflict of interest

None

References

  1. 2018 Alzheimer’s disease facts and figures (2018). Alzheimer’s & Dementia, 14(3), 367–429, doi: 10.1016/j.jalz.2018.02.001. [DOI] [Google Scholar]
  2. Arabpoor Z, Hamidi G, Rashidi B, Shabrang M, Alaei H, Sharifi MR, et al. (2012). Erythropoietin improves neuronal proliferation in dentate gyrus of hippocampal formation in an animal model of Alzheimer’s disease. Adv Biomed Res, 1, 50, doi: 10.4103/2277-9175.100157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ardura-Fabregat A, Boddeke E, Boza-Serrano A, Brioschi S, Castro-Gomez S, Ceyzeriat K, et al. (2017). Targeting Neuroinflammation to Treat Alzheimer’s Disease. CNS Drugs, 31(12), 1057–1082, doi: 10.1007/s40263-017-0483-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Armand-Ugon M, Aso E, Moreno J, Riera-Codina M, Sanchez A, Vegas E, et al. (2015). Memory Improvement in the AbetaPP/PS1 Mouse Model of Familial Alzheimer’s Disease Induced by Carbamylated-Erythropoietin is Accompanied by Modulation of Synaptic Genes. J Alzheimers Dis, 45(2), 407–421, doi: 10.3233/jad-150002. [DOI] [PubMed] [Google Scholar]
  5. Assaraf MI, Diaz Z, Liberman A, Miller WH Jr., Arvanitakis Z, Li Y, et al. (2007). Brain erythropoietin receptor expression in Alzheimer disease and mild cognitive impairment. J Neuropathol Exp Neurol, 66(5), 389–398, doi: 10.1097/nen.0b013e3180517b28. [DOI] [PubMed] [Google Scholar]
  6. Bachurin SO, Bovina EV, & Ustyugov AA (2017). Drugs in Clinical Trials for Alzheimer’s Disease: The Major Trends. Med Res Rev, 37(5), 1186–1225, doi: 10.1002/med.21434. [DOI] [PubMed] [Google Scholar]
  7. Banks WA, Jumbe NL, Farrell CL, Niehoff ML, & Heatherington AC (2004). Passage of erythropoietic agents across the blood-brain barrier: a comparison of human and murine erythropoietin and the analog darbepoetin alfa. Eur J Pharmacol, 505(1–3), 93–101, doi: 10.1016/j.ejphar.2004.10.035. [DOI] [PubMed] [Google Scholar]
  8. Barrett AM, Eslinger PJ, Ballentine NH, & Heilman KM (2005). Unawareness of cognitive deficit (cognitive anosognosia) in probable AD and control subjects. Neurology, 64(4), 693–699, doi: 10.1212/01.wnl.0000151959.64379.1b. [DOI] [PubMed] [Google Scholar]
  9. Bartus RT, Dean RL 3rd, Beer B, & Lippa AS (1982). The cholinergic hypothesis of geriatric memory dysfunction. Science, 217(4558), 408–414. [DOI] [PubMed] [Google Scholar]
  10. Bien-Ly N, Yu YJ, Bumbaca D, Elstrott J, Boswell CA, Zhang Y, et al. (2014). Transferrin receptor (TfR) trafficking determines brain uptake of TfR antibody affinity variants. J Exp Med, 211(2), 233–244, doi: 10.1084/jem.20131660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Boado RJ, Hui EK, Lu JZ, & Pardridge WM (2010). Drug targeting of erythropoietin across the primate blood-brain barrier with an IgG molecular Trojan horse. J Pharmacol Exp Ther, 333(3), 961–969, doi: 10.1124/jpet.109.165092. [DOI] [PubMed] [Google Scholar]
  12. Boado RJ, Zhang Y, Wang Y, & Pardridge WM (2009). Engineering and expression of a chimeric transferrin receptor monoclonal antibody for blood-brain barrier delivery in the mouse. Biotechnol Bioeng, 102(4), 1251–1258, doi: 10.1002/bit.22135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, & Del Tredici K (2006). Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol, 112(4), 389–404, doi: 10.1007/s00401-006-0127-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brettschneider J, Widl K, Ehrenreich H, Riepe M, & Tumani H (2006). Erythropoietin in the cerebrospinal fluid in neurodegenerative diseases. Neurosci Lett, 404(3), 347–351, doi: 10.1016/j.neulet.2006.06.011. [DOI] [PubMed] [Google Scholar]
  15. Brines ML, Ghezzi P, Keenan S, Agnello D, de Lanerolle NC, Cerami C, et al. (2000). Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Natl Acad Sci U S A, 97(19), 10526–10531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Brunkan AL, & Goate AM (2005). Presenilin function and gamma-secretase activity. J Neurochem, 93(4), 769–792, doi: 10.1111/j.1471-4159.2005.03099.x. [DOI] [PubMed] [Google Scholar]
  17. Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, et al. (2011). Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med, 3(89), 89ra57, doi: 10.1126/scitranslmed.3002156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chang R, Al Maghribi A, Vanderpoel V, Vasilevko V, Cribbs DH, Boado R, et al. (2018). Brain Penetrating Bifunctional Erythropoietin-Transferrin Receptor Antibody Fusion Protein for Alzheimer’s Disease. Mol Pharm, doi: 10.1021/acs.molpharmaceut.8b00594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chow VW, Mattson MP, Wong PC, & Gleichmann M (2010). An overview of APP processing enzymes and products. Neuromolecular Med, 12(1), 1–12, doi: 10.1007/s12017-009-8104-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Collin L, Bohrmann B, Gopfert U, Oroszlan-Szovik K, Ozmen L, & Gruninger F (2014). Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer’s disease. Brain, 137(Pt 10), 2834–2846, doi: 10.1093/brain/awu213. [DOI] [PubMed] [Google Scholar]
  21. Cornford EM, & Hyman S (2005). Localization of brain endothelial luminal and abluminal transporters with immunogold electron microscopy. NeuroRx, 2(1), 27–43, doi: 10.1602/neurorx.2.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Couch JA, Yu YJ, Zhang Y, Tarrant JM, Fuji RN, Meilandt WJ, et al. (2013). Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier. Sci Transl Med, 5(183), 183ra157, 181–112, doi: 10.1126/scitranslmed.3005338. [DOI] [PubMed] [Google Scholar]
  23. Danysz W, Parsons CG, Mobius HJ, Stoffler A, & Quack G (2000). Neuroprotective and symptomatological action of memantine relevant for Alzheimer’s disease--a unified glutamatergic hypothesis on the mechanism of action. Neurotox Res, 2(2–3), 85–97. [DOI] [PubMed] [Google Scholar]
  24. Dauphinot V, Delphin-Combe F, Mouchoux C, Dorey A, Bathsavanis A, Makaroff Z, et al. (2015). Risk factors of caregiver burden among patients with Alzheimer’s disease or related disorders: a cross-sectional study. J Alzheimers Dis, 44(3), 907–916, doi: 10.3233/jad-142337. [DOI] [PubMed] [Google Scholar]
  25. Dautry-Varsat A, Ciechanover A, & Lodish HF (1983). pH and the recycling of transferrin during receptor-mediated endocytosis. Proc Natl Acad Sci U S A, 80(8), 2258–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. De Felice FG, Wu D, Lambert MP, Fernandez SJ, Velasco PT, Lacor PN, et al. (2008). Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by A beta oligomers. Neurobiol Aging, 29(9), 1334–1347, doi: 10.1016/j.neurobiolaging.2007.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. DeKosky ST, & Scheff SW (1990). Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol, 27(5), 457–464, doi: 10.1002/ana.410270502. [DOI] [PubMed] [Google Scholar]
  28. Diaz Z, Assaraf MI, Miller WH Jr., & Schipper HM (2005). Astroglial cytoprotection by erythropoietin pre-conditioning: implications for ischemic and degenerative CNS disorders. J Neurochem, 93(2), 392–402, doi: 10.1111/j.1471-4159.2005.03038.x. [DOI] [PubMed] [Google Scholar]
  29. Digicaylioglu M, Bichet S, Marti HH, Wenger RH, Rivas LA, Bauer C, et al. (1995). Localization of specific erythropoietin binding sites in defined areas of the mouse brain. Proc Natl Acad Sci U S A, 92(9), 3717–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Dinamarca MC, Rios JA, & Inestrosa NC (2012). Postsynaptic Receptors for Amyloid-beta Oligomers as Mediators of Neuronal Damage in Alzheimer’s Disease. Front Physiol, 3, 464, doi: 10.3389/fphys.2012.00464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. (2013). A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med, 369(4), 341–350, doi: 10.1056/NEJMoa1210951. [DOI] [PubMed] [Google Scholar]
  32. Eckert GP, Renner K, Eckert SH, Eckmann J, Hagl S, Abdel-Kader RM, et al. (2012). Mitochondrial dysfunction--a pharmacological target in Alzheimer’s disease. Mol Neurobiol, 46(1), 136–150, doi: 10.1007/s12035-012-8271-z. [DOI] [PubMed] [Google Scholar]
  33. Eikelenboom P, Veerhuis R, Scheper W, Rozemuller AJ, van Gool WA, & Hoozemans JJ (2006). The significance of neuroinflammation in understanding Alzheimer’s disease. J Neural Transm (Vienna), 113(11), 1685–1695, doi: 10.1007/s00702-006-0575-6. [DOI] [PubMed] [Google Scholar]
  34. Erbayraktar S, Grasso G, Sfacteria A, Xie QW, Coleman T, Kreilgaard M, et al. (2003). Asialoerythropoietin is a nonerythropoietic cytokine with broad neuroprotective activity in vivo. Proc Natl Acad Sci U S A, 100(11), 6741–6746, doi: 10.1073/pnas.1031753100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Esmaeili Tazangi P, Moosavi SM, Shabani M, & Haghani M (2015). Erythropoietin improves synaptic plasticity and memory deficits by decrease of the neurotransmitter release probability in the rat model of Alzheimer’s disease. Pharmacol Biochem Behav, 130, 15–21, doi: 10.1016/j.pbb.2014.12.011. [DOI] [PubMed] [Google Scholar]
  36. Fan Z, Okello AA, Brooks DJ, & Edison P (2015). Longitudinal influence of microglial activation and amyloid on neuronal function in Alzheimer’s disease. Brain, 138(Pt 12), 3685–3698, doi: 10.1093/brain/awv288. [DOI] [PubMed] [Google Scholar]
  37. Fishman JB, Rubin JB, Handrahan JV, Connor JR, & Fine RE (1987). Receptor-mediated transcytosis of transferrin across the blood-brain barrier. J Neurosci Res, 18(2), 299–304, doi: 10.1002/jnr.490180206. [DOI] [PubMed] [Google Scholar]
  38. Giannakopoulos P, Duc M, Gold G, Hof PR, Michel JP, & Bouras C (1998). Pathologic correlates of apraxia in Alzheimer disease. Arch Neurol, 55(5), 689–695. [DOI] [PubMed] [Google Scholar]
  39. Gremer L, Scholzel D, Schenk C, Reinartz E, & Labahn J (2017). Fibril structure of amyloid-beta(1–42) by cryo-electron microscopy. 358(6359), 116–119, doi: 10.1126/science.aao2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Haase VH (2013). Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev, 27(1), 41–53, doi: 10.1016/j.blre.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Haass C, & De Strooper B (1999). The presenilins in Alzheimer’s disease--proteolysis holds the key. Science, 286(5441), 916–919. [DOI] [PubMed] [Google Scholar]
  42. Hardy J (2009). The amyloid hypothesis for Alzheimer’s disease: a critical reappraisal. J Neurochem, 110(4), 1129–1134, doi: 10.1111/j.1471-4159.2009.06181.x. [DOI] [PubMed] [Google Scholar]
  43. Hardy JA, & Higgins GA (1992). Alzheimer’s disease: the amyloid cascade hypothesis. Science, 256(5054), 184–185. [DOI] [PubMed] [Google Scholar]
  44. Hebert LE, Beckett LA, Scherr PA, & Evans DA (2001). Annual incidence of Alzheimer disease in the United States projected to the years 2000 through 2050. Alzheimer Dis Assoc Disord, 15(4), 169–173. [DOI] [PubMed] [Google Scholar]
  45. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. (2015). Neuroinflammation in Alzheimer’s disease. Lancet Neurol, 14(4), 388–405, doi: 10.1016/s1474-4422(15)70016-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, et al. (2016). Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science, 352(6286), 712–716, doi: 10.1126/science.aad8373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hooshmandi E, Motamedi F, Moosavi M, Katinger H, Zakeri Z, Zaringhalam J, et al. (2018). CEPO-Fc (An EPO Derivative) Protects Hippocampus Against Abeta-induced Memory Deterioration: A Behavioral and Molecular Study in a Rat Model of Abeta Toxicity. Neuroscience, 388, 405–417, doi: 10.1016/j.neuroscience.2018.08.001. [DOI] [PubMed] [Google Scholar]
  48. Hultqvist G, Syvanen S, Fang XT, Lannfelt L, & Sehlin D (2017). Bivalent Brain Shuttle Increases Antibody Uptake by Monovalent Binding to the Transferrin Receptor. Theranostics, 7(2), 308–318, doi: 10.7150/thno.17155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Huwyler J, & Pardridge WM (1998). Examination of blood-brain barrier transferrin receptor by confocal fluorescent microscopy of unfixed isolated rat brain capillaries. J Neurochem, 70(2), 883–886. [DOI] [PubMed] [Google Scholar]
  50. James BD, Leurgans SE, Hebert LE, Scherr PA, Yaffe K, & Bennett DA (2014). Contribution of Alzheimer disease to mortality in the United States. Neurology, 82(12), 1045–1050, doi: 10.1212/wnl.0000000000000240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, & Mason DY (1984). Transferrin receptor on endothelium of brain capillaries. Nature, 312(5990), 162–163. [DOI] [PubMed] [Google Scholar]
  52. Jelkmann W (2005). Effects of erythropoietin on brain function. Curr Pharm Biotechnol, 6(1), 65–79. [DOI] [PubMed] [Google Scholar]
  53. Jelkmann W (2013). Physiology and pharmacology of erythropoietin. Transfus Med Hemother, 40(5), 302–309, doi: 10.1159/000356193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ji P (2016). Pericytes: new EPO-producing cells in the brain. Blood, 128(21), 2483–2485, doi: 10.1182/blood-2016-10-743880. [DOI] [PubMed] [Google Scholar]
  55. Jiao SS, Shen LL, Zhu C, Bu XL, Liu YH, Liu CH, et al. (2016). Brain-derived neurotrophic factor protects against tau-related neurodegeneration of Alzheimer’s disease. Transl Psychiatry, 6(10), e907, doi: 10.1038/tp.2016.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Jones AR, & Shusta EV (2007). Blood-brain barrier transport of therapeutics via receptor-mediation. Pharm Res, 24(9), 1759–1771, doi: 10.1007/s11095-007-9379-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Juul SE, Anderson DK, Li Y, & Christensen RD (1998). Erythropoietin and erythropoietin receptor in the developing human central nervous system. Pediatr Res, 43(1), 40–49, doi: 10.1203/00006450-199804001-00243. [DOI] [PubMed] [Google Scholar]
  58. Kandimalla R, & Reddy PH (2017). Therapeutics of Neurotransmitters in Alzheimer’s Disease. J Alzheimers Dis, 57(4), 1049–1069, doi: 10.3233/jad-161118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Karch CM, & Goate AM (2015). Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry, 77(1), 43–51, doi: 10.1016/j.biopsych.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kato M, Miura K, Kamiyama H, Okazaki A, Kumaki K, Kato Y, et al. (1998). Pharmacokinetics of erythropoietin in genetically anemic mice. Drug Metab Dispos, 26(2), 126–131. [PubMed] [Google Scholar]
  61. King VR, Averill SA, Hewazy D, Priestley JV, Torup L, & Michael-Titus AT (2007). Erythropoietin and carbamylated erythropoietin are neuroprotective following spinal cord hemisection in the rat. Eur J Neurosci, 26(1), 90–100, doi: 10.1111/j.1460-9568.2007.05635.x. [DOI] [PubMed] [Google Scholar]
  62. Konishi Y, Chui DH, Hirose H, Kunishita T, & Tabira T (1993). Trophic effect of erythropoietin and other hematopoietic factors on central cholinergic neurons in vitro and in vivo. Brain Res, 609(1–2), 29–35. [DOI] [PubMed] [Google Scholar]
  63. Konofagou EE, Tung YS, Choi J, Deffieux T, Baseri B, & Vlachos F (2012). Ultrasound-induced blood-brain barrier opening. Curr Pharm Biotechnol, 13(7), 1332–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Lapchak PA (2010). Erythropoietin molecules to treat acute ischemic stroke: a translational dilemma! Expert Opin Investig Drugs, 19(10), 1179–1186, doi: 10.1517/13543784.2010.517954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lee HJ, Engelhardt B, Lesley J, Bickel U, & Pardridge WM (2000). Targeting rat anti-mouse transferrin receptor monoclonal antibodies through blood-brain barrier in mouse. J Pharmacol Exp Ther, 292(3), 1048–1052. [PubMed] [Google Scholar]
  66. Lee ST, Chu K, Park JE, Jung KH, Jeon D, Lim JY, et al. (2012). Erythropoietin improves memory function with reducing endothelial dysfunction and amyloid-beta burden in Alzheimer’s disease models. J Neurochem, 120(1), 115–124, doi: 10.1111/j.1471-4159.2011.07534.x. [DOI] [PubMed] [Google Scholar]
  67. Leuner K, Muller WE, & Reichert AS (2012). From mitochondrial dysfunction to amyloid beta formation: novel insights into the pathogenesis of Alzheimer’s disease. Mol Neurobiol, 46(1), 186–193, doi: 10.1007/s12035-012-8307-4. [DOI] [PubMed] [Google Scholar]
  68. Li G, Ma R, Huang C, Tang Q, Fu Q, Liu H, et al. (2008). Protective effect of erythropoietin on beta-amyloid-induced PC12 cell death through antioxidant mechanisms. Neurosci Lett, 442(2), 143–147, doi: 10.1016/j.neulet.2008.07.007. [DOI] [PubMed] [Google Scholar]
  69. Li JY, Boado RJ, & Pardridge WM (2001). Blood-brain barrier genomics. J Cereb Blood Flow Metab, 21(1), 61–68, doi: 10.1097/00004647-200101000-00008. [DOI] [PubMed] [Google Scholar]
  70. Li YP, Yang GJ, Jin L, Yang HM, Chen J, Chai GS, et al. (2015). Erythropoietin attenuates Alzheimer-like memory impairments and pathological changes induced by amyloid beta42 in mice. Brain Res, 1618, 159–167, doi: 10.1016/j.brainres.2015.05.031. [DOI] [PubMed] [Google Scholar]
  71. Lochhead JJ, & Thorne RG (2012). Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev, 64(7), 614–628, doi: 10.1016/j.addr.2011.11.002. [DOI] [PubMed] [Google Scholar]
  72. Lombardo S, & Maskos U (2015). Role of the nicotinic acetylcholine receptor in Alzheimer’s disease pathology and treatment. Neuropharmacology, 96(Pt B), 255–262, doi: 10.1016/j.neuropharm.2014.11.018. [DOI] [PubMed] [Google Scholar]
  73. Lu CT, Zhao YZ, Wong HL, Cai J, Peng L, & Tian XQ (2014). Current approaches to enhance CNS delivery of drugs across the brain barriers. Int J Nanomedicine, 9, 2241–2257, doi: 10.2147/ijn.s61288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lundby C, Thomsen JJ, Boushel R, Koskolou M, Warberg J, Calbet JA, et al. (2007). Erythropoietin treatment elevates haemoglobin concentration by increasing red cell volume and depressing plasma volume. J Physiol, 578(Pt 1), 309–314, doi: 10.1113/jphysiol.2006.122689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Ma R, Xiong N, Huang C, Tang Q, Hu B, Xiang J, et al. (2009). Erythropoietin protects PC12 cells from beta-amyloid(25–35)-induced apoptosis via PI3K/Akt signaling pathway. Neuropharmacology, 56(6–7), 1027–1034, doi: 10.1016/j.neuropharm.2009.02.006. [DOI] [PubMed] [Google Scholar]
  76. Marti HH, Wenger RH, Rivas LA, Straumann U, Digicaylioglu M, Henn V, et al. (1996). Erythropoietin gene expression in human, monkey and murine brain. Eur J Neurosci, 8(4), 666–676. [DOI] [PubMed] [Google Scholar]
  77. Masuda S, Okano M, Yamagishi K, Nagao M, Ueda M, & Sasaki R (1994). A novel site of erythropoietin production. Oxygen-dependent production in cultured rat astrocytes. J Biol Chem, 269(30), 19488–19493. [PubMed] [Google Scholar]
  78. Maurice T, Mustafa MH, Desrumaux C, Keller E, Naert G, de la CG-BM, et al. (2013). Intranasal formulation of erythropoietin (EPO) showed potent protective activity against amyloid toxicity in the Abeta(2)(5)(−)(3)(5) non-transgenic mouse model of Alzheimer’s disease. J Psychopharmacol, 27(11), 1044–1057, doi: 10.1177/0269881113494939. [DOI] [PubMed] [Google Scholar]
  79. Mawanda F, & Wallace R (2013). Can infections cause Alzheimer’s disease? Epidemiol Rev, 35, 161–180, doi: 10.1093/epirev/mxs007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. McGeer EG, & McGeer PL (2003). Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry, 27(5), 741–749, doi: 10.1016/s0278-5846(03)00124-6. [DOI] [PubMed] [Google Scholar]
  81. Melov S, Adlard PA, Morten K, Johnson F, Golden TR, Hinerfeld D, et al. (2007). Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS One, 2(6), e536, doi: 10.1371/journal.pone.0000536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Mennini T, De Paola M, Bigini P, Mastrotto C, Fumagalli E, Barbera S, et al. (2006). Nonhematopoietic erythropoietin derivatives prevent motoneuron degeneration in vitro and in vivo. Mol Med, 12(7–8), 153–160, doi: 10.2119/2006-00045.Mennini. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Mitchell SL, Teno JM, Kiely DK, Shaffer ML, Jones RN, Prigerson HG, et al. (2009). The clinical course of advanced dementia. N Engl J Med, 361(16), 1529–1538, doi: 10.1056/NEJMoa0902234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Moos T, Rosengren Nielsen T, Skjorringe T, & Morgan EH (2007). Iron trafficking inside the brain. J Neurochem, 103(5), 1730–1740, doi: 10.1111/j.1471-4159.2007.04976.x. [DOI] [PubMed] [Google Scholar]
  85. Morishita E, Masuda S, Nagao M, Yasuda Y, & Sasaki R (1997). Erythropoietin receptor is expressed in rat hippocampal and cerebral cortical neurons, and erythropoietin prevents in vitro glutamate-induced neuronal death. Neuroscience, 76(1), 105–116. [DOI] [PubMed] [Google Scholar]
  86. Nagai A, Nakagawa E, Choi HB, Hatori K, Kobayashi S, & Kim SU (2001). Erythropoietin and erythropoietin receptors in human CNS neurons, astrocytes, microglia, and oligodendrocytes grown in culture. J Neuropathol Exp Neurol, 60(4), 386–392. [DOI] [PubMed] [Google Scholar]
  87. Ng T, Marx G, Littlewood T, & Macdougall I (2003). Recombinant erythropoietin in clinical practice. Postgrad Med J, 79(933), 367–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, & Weller RO (2003). Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med, 9(4), 448–452, doi: 10.1038/nm840. [DOI] [PubMed] [Google Scholar]
  89. Niewoehner J, Bohrmann B, Collin L, Urich E, Sade H, Maier P, et al. (2014). Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron, 81(1), 49–60, doi: 10.1016/j.neuron.2013.10.061. [DOI] [PubMed] [Google Scholar]
  90. Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, et al. (2012). Immunotherapy for Alzheimer’s disease: from anti-beta-amyloid to tau-based immunization strategies. Immunotherapy, 4(2), 213–238, doi: 10.2217/imt.11.170. [DOI] [PubMed] [Google Scholar]
  91. Panza F, Logroscino G, Imbimbo BP, & Solfrizzi V (2014). Is there still any hope for amyloid-based immunotherapy for Alzheimer’s disease? Curr Opin Psychiatry, 27(2), 128–137, doi: 10.1097/yco.0000000000000041. [DOI] [PubMed] [Google Scholar]
  92. Pardridge WM (2005). The blood-brain barrier: bottleneck in brain drug development. NeuroRx, 2(1), 3–14, doi: 10.1602/neurorx.2.1.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Pardridge WM (2006). Molecular Trojan horses for blood-brain barrier drug delivery. Discov Med, 6(34), 139–143. [PubMed] [Google Scholar]
  94. Pardridge WM (2009). Alzheimer’s disease drug development and the problem of the blood-brain barrier. Alzheimers Dement, 5(5), 427–432, doi: 10.1016/j.jalz.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Pardridge WM (2012). Drug transport across the blood-brain barrier. J Cereb Blood Flow Metab, 32(11), 1959–1972, doi: 10.1038/jcbfm.2012.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Pardridge WM (2015). Blood-brain barrier drug delivery of IgG fusion proteins with a transferrin receptor monoclonal antibody. Expert Opin Drug Deliv, 12(2), 207–222, doi: 10.1517/17425247.2014.952627. [DOI] [PubMed] [Google Scholar]
  97. Pardridge WM (2016). CSF, blood-brain barrier, and brain drug delivery. Expert Opin Drug Deliv, 13(7), 963–975, doi: 10.1517/17425247.2016.1171315. [DOI] [PubMed] [Google Scholar]
  98. Pardridge WM (2017). Delivery of Biologics Across the Blood-Brain Barrier with Molecular Trojan Horse Technology. BioDrugs, 31(6), 503–519, doi: 10.1007/s40259-017-0248-z. [DOI] [PubMed] [Google Scholar]
  99. Pardridge WM, Boado RJ, Patrick DJ, Ka-Wai Hui E, & Lu JZ (2018). Blood-Brain Barrier Transport, Plasma Pharmacokinetics, and Neuropathology Following Chronic Treatment of the Rhesus Monkey with a Brain Penetrating Humanized Monoclonal Antibody Against the Human Transferrin Receptor. Mol Pharm, doi: 10.1021/acs.molpharmaceut.8b00730. [DOI] [PubMed] [Google Scholar]
  100. Portet F, Scarmeas N, Cosentino S, Helzner EP, & Stern Y (2009). Extrapyramidal signs before and after diagnosis of incident Alzheimer disease in a prospective population study. Arch Neurol, 66(9), 1120–1126, doi: 10.1001/archneurol.2009.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Potter R, Patterson BW, Elbert DL, Ovod V, Kasten T, Sigurdson W, et al. (2013). Increased in vivo amyloid-beta42 production, exchange, and loss in presenilin mutation carriers. Sci Transl Med, 5(189), 189ra177, doi: 10.1126/scitranslmed.3005615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Preston JE, Joan Abbott N, & Begley DJ (2014). Transcytosis of macromolecules at the blood-brain barrier. Adv Pharmacol, 71, 147–163, doi: 10.1016/bs.apha.2014.06.001. [DOI] [PubMed] [Google Scholar]
  103. Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, & Herms J (2006). Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci, 26(27), 7212–7221, doi: 10.1523/jneurosci.1450-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, & Mobius HJ (2003). Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med, 348(14), 1333–1341, doi: 10.1056/NEJMoa013128. [DOI] [PubMed] [Google Scholar]
  105. Revett TJ, Baker GB, Jhamandas J, & Kar S (2013). Glutamate system, amyloid ss peptides and tau protein: functional interrelationships and relevance to Alzheimer disease pathology. J Psychiatry Neurosci, 38(1), 6–23, doi: 10.1503/jpn.110190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Reynolds GP, Mason SL, Meldrum A, De Keczer S, Parnes H, Eglen RM, et al. (1995). 5-Hydroxytryptamine (5-HT)4 receptors in post mortem human brain tissue: distribution, pharmacology and effects of neurodegenerative diseases. Br J Pharmacol, 114(5), 993–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Roberts RL, Fine RE, & Sandra A (1993). Receptor-mediated endocytosis of transferrin at the blood-brain barrier. J Cell Sci, 104 ( Pt 2), 521–532. [DOI] [PubMed] [Google Scholar]
  108. Rodriguez Cruz Y, Strehaiano M, Rodriguez Obaya T, Garcia Rodriguez JC, & Maurice T (2017). An Intranasal Formulation of Erythropoietin (Neuro-EPO) Prevents Memory Deficits and Amyloid Toxicity in the APPSwe Transgenic Mouse Model of Alzheimer’s Disease. J Alzheimers Dis, 55(1), 231–248, doi: 10.3233/jad-160500. [DOI] [PubMed] [Google Scholar]
  109. Rogaev EI, Sherrington R, Rogaeva EA, Levesque G, Ikeda M, Liang Y, et al. (1995). Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature, 376(6543), 775–778, doi: 10.1038/376775a0. [DOI] [PubMed] [Google Scholar]
  110. Rogers J, Webster S, Lue LF, Brachova L, Civin WH, Emmerling M, et al. (1996). Inflammation and Alzheimer’s disease pathogenesis. Neurobiol Aging, 17(5), 681–686. [DOI] [PubMed] [Google Scholar]
  111. Sakanaka M, Wen TC, Matsuda S, Masuda S, Morishita E, Nagao M, et al. (1998). In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc Natl Acad Sci U S A, 95(8), 4635–4640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Samy DM, Ismail CA, Nassra RA, Zeitoun TM, & Nomair AM (2016). Downstream modulation of extrinsic apoptotic pathway in streptozotocin-induced Alzheimer’s dementia in rats: Erythropoietin versus curcumin. Eur J Pharmacol, 770, 52–60, doi: 10.1016/j.ejphar.2015.11.046. [DOI] [PubMed] [Google Scholar]
  113. Sarazin M, Stern Y, Berr C, Riba A, Albert M, Brandt J, et al. (2005). Neuropsychological predictors of dependency in patients with Alzheimer disease. Neurology, 64(6), 1027–1031, doi: 10.1212/01.wnl.0000154529.53488.30. [DOI] [PubMed] [Google Scholar]
  114. Sargin D, Friedrichs H, El-Kordi A, & Ehrenreich H (2010). Erythropoietin as neuroprotective and neuroregenerative treatment strategy: comprehensive overview of 12 years of preclinical and clinical research. Best Pract Res Clin Anaesthesiol, 24(4), 573–594, doi: 10.1016/j.bpa.2010.10.005. [DOI] [PubMed] [Google Scholar]
  115. Schupf N, Kapell D, Nightingale B, Rodriguez A, Tycko B, & Mayeux R (1998). Earlier onset of Alzheimer’s disease in men with Down syndrome. Neurology, 50(4), 991–995. [DOI] [PubMed] [Google Scholar]
  116. Shang YC, Chong ZZ, Wang S, & Maiese K (2011). Erythropoietin and Wnt1 govern pathways of mTOR, Apaf-1, and XIAP in inflammatory microglia. Curr Neurovasc Res, 8(4), 270–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Shang YC, Chong ZZ, Wang S, & Maiese K (2012). Prevention of beta-amyloid degeneration of microglia by erythropoietin depends on Wnt1, the PI 3-K/mTOR pathway, Bad, and Bcl-xL. Aging (Albany NY), 4(3), 187–201, doi: 10.18632/aging.100440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Sigurdsson EM (2018). Tau Immunotherapies for Alzheimer’s Disease and Related Tauopathies: Progress and Potential Pitfalls. J Alzheimers Dis, 64(s1), S555–s565, doi: 10.3233/jad-179937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Slotkin TA, Seidler FJ, Crain BJ, Bell JM, Bissette G, & Nemeroff CB (1990). Regulatory changes in presynaptic cholinergic function assessed in rapid autopsy material from patients with Alzheimer disease: implications for etiology and therapy. Proc Natl Acad Sci U S A, 87(7), 2452–2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Sosa-Ortiz AL, Acosta-Castillo I, & Prince MJ (2012). Epidemiology of dementias and Alzheimer’s disease. Arch Med Res, 43(8), 600–608, doi: 10.1016/j.arcmed.2012.11.003. [DOI] [PubMed] [Google Scholar]
  121. Thal DR, Rub U, Orantes M, & Braak H (2002). Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology, 58(12), 1791–1800. [DOI] [PubMed] [Google Scholar]
  122. Torup L, & Leist M (2006). Development of Non-Erythropoietic Erythropoietin Variants for Neuroprotection In Höke A (Ed.), Erythropoietin and the Nervous System: Novel Therapeutic Options for Neuroprotection (pp. 211–219). Boston, MA: Springer US. [Google Scholar]
  123. Ulrich D (2015). Amyloid-beta Impairs Synaptic Inhibition via GABA(A) Receptor Endocytosis. J Neurosci, 35(24), 9205–9210, doi: 10.1523/jneurosci.0950-15.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. van Marum RJ (2009). Update on the use of memantine in Alzheimer’s disease. Neuropsychiatr Dis Treat, 5, 237–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Villa P, Bigini P, Mennini T, Agnello D, Laragione T, Cagnotto A, et al. (2003). Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis. J Exp Med, 198(6), 971–975, doi: 10.1084/jem.20021067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Viviani B, Bartesaghi S, Corsini E, Villa P, Ghezzi P, Garau A, et al. (2005). Erythropoietin protects primary hippocampal neurons increasing the expression of brain-derived neurotrophic factor. J Neurochem, 93(2), 412–421, doi: 10.1111/j.1471-4159.2005.03033.x. [DOI] [PubMed] [Google Scholar]
  127. Vossel KA, Tartaglia MC, Nygaard HB, Zeman AZ, & Miller BL (2017). Epileptic activity in Alzheimer’s disease: causes and clinical relevance. Lancet Neurol, 16(4), 311–322, doi: 10.1016/s1474-4422(17)30044-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Wan HI, Jacobsen JS, Rutkowski JL, & Feuerstein GZ (2009). Translational medicine lessons from flurizan’s failure in Alzheimer’s disease (AD) trial: Implication for future drug discovery and development for AD. Clin Transl Sci, 2(3), 242–247, doi: 10.1111/j.1752-8062.2009.00121.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Wang WY, Tan MS, Yu JT, & Tan L (2015). Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med, 3(10), 136, doi: 10.3978/j.issn.2305-5839.2015.03.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Weber F, Bohrmann B, Niewoehner J, Fischer JAA, Rueger P, Tiefenthaler G, et al. (2018). Brain Shuttle Antibody for Alzheimer’s Disease with Attenuated Peripheral Effector Function due to an Inverted Binding Mode. Cell Rep, 22(1), 149–162, doi: 10.1016/j.celrep.2017.12.019. [DOI] [PubMed] [Google Scholar]
  131. Wenk GL (2003). Neuropathologic changes in Alzheimer’s disease. J Clin Psychiatry, 64 Suppl 9, 7–10. [PubMed] [Google Scholar]
  132. Wenker SD, Chamorro ME, Vittori DC, & Nesse AB (2013). Protective action of erythropoietin on neuronal damage induced by activated microglia. Febs j, 280(7), 1630–1642, doi: 10.1111/febs.12172. [DOI] [PubMed] [Google Scholar]
  133. Wildburger NC, Esparza TJ, & LeDuc RD (2017). Diversity of Amyloid-beta Proteoforms in the Alzheimer’s Disease Brain. 7(1), 9520, doi: 10.1038/s41598-017-10422-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Wyss-Coray T (2006). Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med, 12(9), 1005–1015, doi: 10.1038/nm1484. [DOI] [PubMed] [Google Scholar]
  135. Yu YJ, Atwal JK, Zhang Y, Tong RK, Wildsmith KR, Tan C, et al. (2014). Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman primates. Sci Transl Med, 6(261), 261ra154, doi: 10.1126/scitranslmed.3009835. [DOI] [PubMed] [Google Scholar]
  136. Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, et al. (2011). Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med, 3(84), 84ra44, doi: 10.1126/scitranslmed.3002230. [DOI] [PubMed] [Google Scholar]
  137. Zhang Y, & Pardridge WM (2001). Rapid transferrin efflux from brain to blood across the blood-brain barrier. J Neurochem, 76(5), 1597–1600. [DOI] [PubMed] [Google Scholar]
  138. Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, et al. (2014). Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat Med, 20(11), 1254–1262, doi: 10.1038/nm.3700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Zhou QH, Boado RJ, Lu JZ, Hui EK, & Pardridge WM (2010). Re-engineering erythropoietin as an IgG fusion protein that penetrates the blood-brain barrier in the mouse. Mol Pharm, 7(6), 2148–2155, doi: 10.1021/mp1001763. [DOI] [PubMed] [Google Scholar]
  140. Zhou QH, Hui EK, Lu JZ, Boado RJ, & Pardridge WM (2011). Brain penetrating IgG-erythropoietin fusion protein is neuroprotective following intravenous treatment in Parkinson’s disease in the mouse. Brain Res, 1382, 315–320, doi: 10.1016/j.brainres.2011.01.061. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES