You are on page 1of 13

Review

APOE and Alzheimer’s disease: advances in genetics,


pathophysiology, and therapeutic approaches
Alberto Serrano-Pozo, Sudeshna Das, Bradley T Hyman

Lancet Neurol 2021; 20: 68–80 The APOE ε4 allele remains the strongest genetic risk factor for sporadic Alzheimer’s disease and the APOE ε2
This online publication has allele the strongest genetic protective factor after multiple large scale genome-wide association studies and genome-
been corrected. The corrected wide association meta-analyses. However, no therapies directed at APOE are currently available. Although initial
version first appeared at
studies causally linked APOE with amyloid-β peptide aggregation and clearance, over the past 5 years our
thelancet.com/neurology on
January 20, 2021 understanding of APOE pathogenesis has expanded beyond amyloid-β peptide-centric mechanisms to tau
Department of Neurology,
neurofibrillary degeneration, microglia and astrocyte responses, and blood–brain barrier disruption. Because all
Massachusetts General these pathological processes can potentially contribute to cognitive impairment, it is important to use this new
Hospital, Boston, MA, USA knowledge to develop therapies directed at APOE. Several therapeutic approaches have been successful in mouse
(A Serrano-Pozo MD, S Das PhD,
models expressing human APOE alleles, including increasing or reducing APOE levels, enhancing its lipidation,
Prof B T Hyman MD);
Massachusetts Alzheimer’s blocking the interactions between APOE and amyloid-β peptide, and genetically switching APOE4 to APOE3 or
Disease Research Center, APOE2 isoforms, but translation to human clinical trials has proven challenging.
Charlestown, MA, USA
(A Serrano-Pozo, S Das,
Prof B T Hyman); and Harvard
Introduction to be effective in mouse models and hold promise for
Medical School, Boston, MA, Even after multiple large-scale genome-wide association trans­lation to human clinical trials. In this Review, we
USA (A Serrano-Pozo, S Das, studies (GWAS) and GWAS meta-analyses1, the ε4 allele of discuss the advances made in genetics, pathophysiol­ogy,
Prof B T Hyman) the APOE gene (compared with the most common ε3 and therapeutic approaches related to APOE and
Correspondence to: allele) continues to be the strongest genetic risk factor Alzheimer’s disease.
Prof Bradley T Hyman,
associated with sporadic Alzheimer’s disease since its
Massachusetts Alzheimer’s
Disease Research Center, discovery in 1993. Moreover, the relatively rare APOE ε2 Genetic discoveries related to APOE
Charlestown, MA 02129, USA allele remains by far the strongest genetic protective Over the past 3 years, human genetic studies have
bhyman@mgh.harvard.edu factor against sporadic Alzheimer’s disease (panel 1), suggested risk modifiers that mitigate or increase APOE
empha­ sising the importance of APOE’s role in ε4-associated Alzheimer’s disease risk, and identified
Alzheimer’s disease patho­genesis. Because Alzheimer’s haplo­­types with heterogeneous effects. Understanding the
disease is defined by the accumulation of two hallmark risk variation in APOE ε4 carriers has the potential to shed
pathological protein aggre­gates: amyloid-β pep­tide (Aβ) further light on APOE pathobiology and mechanisms of
plaques and neuro­ fibrillary tangles containing hyper­ resilience and resistance to Alzheimer’s disease, which
phosphorylated tau, one postulate is that APOE affects could have therapeutic value.
these lesions. Although solid evidence supports this view,
emerging advances are changing our understanding of APOE ε2 homozygosity
APOE involvement in Alzheimer’s disease. First, new In an analysis2 of a US cohort with approximately
genetic modifiers and the APOE local ancestry (ie, the 5000 neuropathologically confirmed Alzheimer’s disease
population-specific genetic variation in the APOE region) and control subjects, APOE ε2 homozygosity was associ­
have been associated with a differ­ential APOE ε4-linked ated with much lower odds of Alzheimer’s disease
increased risk of Alzheimer’s disease. Second, although than was APOE ε3 homozygosity (odds ratio [OR] 0·13
APOE modifica­ tion of Alzheimer’s disease risk has [95% CI 0·05–0·36]), and the APOE ε2ε3 genotype (0·39
been long attributed to its effects on Aβ, systematic [0·30–0·50]). The contrast of APOE ε2 homozygosity
neuropathological examina­tion of large autopsy cohorts versus APOE ε4 homozygosity was even greater (0·004
has suggested that the APOE genotype also correlates with [0·001–0·014]), and APOE ε2 was also associ­ ated with
the presence and severity of other proteinopathies, milder Alzheimer’s disease neuropathological changes (ie,
pointing to new causal links. Third, technological advances less widespread Aβ plaques and neuro­fibrillary tangles) in
in the past decade—including mouse models genetically this autopsy cohort. However, these excep­ tionally low
engineered to express human APOE alleles, virally Alzheimer’s disease ORs in APOE ε2 homozy­gotes were
mediated gene transfer, proteomics and trans­cripto­mics, not found in the larger clinically defined but neuropathol­
patient-derived human-induced pluripotent stem cells, ogically unconfirmed group (23 857 individ­uals; 10 430 with
plasma, CSF, PET, and MRI biomarkers—have impli­ probable Alzheimer’s disease and 13 426 cog­ n­
itively
cated APOE in other aspects of Alzheimer’s disease unimpaired), suggest­ ing a stronger pro­ tection against
pathophysiology, such as tau-induced neurodegen­era­tion, Alzheimer’s disease neuropathology.
microglial and astrocyte react­ ions (including neuro­
inflammation), and blood–brain barrier disruption. Last, APOE Christchurch mutation
although no APOE-based ther­apy is yet available, several A single case report27 described an approximately 70-year-
APOE-directed therapeutic approaches have been shown old Colombian woman who, despite carrying a fully

68 www.thelancet.com/neurology Vol 20 January 2021


Review

penetrant autosomal-dominant E280A mutation in


PSEN1, which is linked to familial Alzheimer’s dis­
ease, and abundant fibrillar Aβ deposits in her PET scan,
remained cognitively healthy well beyond her expected Panel 1: APOE basic facts
year of symp­tom onset (age 44 years). After whole exome • Two single nucleotide polymorphisms—rs429358 and
sequenc­ing, it was concluded that a rare homozygous rs7412—define the three alleles of APOE, located in
APOE ε3 Christchurch (R136S) mutation conferred her chromosome 19q13·2: ε2, ε3, and ε4. Relative to the most
resilience to Alzheimer’s disease. Mechanistically, the common APOEε3 homozygous genotype (reference
APOE3 R136S mutation appears to inhibit Aβ oligo­­ group), possessing one APOE ε4 allele increases the risk of
merisation, disrupt APOE binding to low-density lipo­ developing Alzheimer’s disease by approximately
protein receptor, and interfere with APOE affinity for 3·7 times and being homozygous for the APOE ε4 allele
heparan sul­ fate proteo­
glycans, which are involved in increases the risk up to 12 times, whereas carrying a single
toxic tau uptake by neurons, per­haps explaining the lower APOE ε2 allele reduces the risk by approximately 40%,
than average radioligand uptake observed in her tau and being homozygous for APOEε2 reduces the risk
PET scan.27 even further.2,3
• Besides Alzheimer’s disease risk, the APOE genotype
Other genetic modifiers mainly affects the age of onset of cognitive impairment,
A meta-analysis of 22 studies28 has revealed that with APOEε4 carriers having an earlier age of onset and
KLOTHO-VS heterozygosity—a polymorphism previously APOEε2 carriers a later age of onset than APOEε3
associated with longevity—might attenuate the increased homozygotes. By contrast, the effect of APOE genotype
Alzheimer’s disease risk associated with the APOE ε4 on the rate of cognitive decline after symptom onset
allele, because APOE ε4 carriers older than 60 years with remains controversial, with allele differences typically
KLOTHO-VS heterozygosity had a reduced Alzheimer’s considered not clinically relevant.4
disease risk (OR 0·75 [95% CI 0·67–0·84]; p=7·4×10­­-⁷), • APOE is a 299-amino acid (MW 34 kDa) secreted
reduced risk of conversion from mild cognitive impair­ glycoprotein that binds cholesterol and phospholipids
ment to dementia (hazard ratio [HR] 0·64 [95% CI through the C-terminus domain and to its receptors
0·44–0·94]; p=0·02], higher CSF Aβ levels, and lower through the N-terminus domain.5
Aβ PET burden; the results were significant specifically • The three APOE isoforms differ in two amino acid
in the group of individuals aged 60–80 years. A whole residues at positions 112 (Cys in APOE2 and APOE3, and
genome sequencing on a mainland Chinese cohort Arg in APOE4) and 158 (Cys in APOE2, and Arg in APOE3
identified nine potential causal variants in two genes and APOE4), and these polymorphisms cause significant
located in the vicinity of APOE, PVRL2 and APOC1,29 differences across APOE isoforms in both lipid binding
which increased the risk of developing Alzheimer’s disease properties (ie, APOE4 is hypolipidated compared with
independently of the APOE ε4 allele. The risk haplo­ APOE3 and APOE25,6) and receptor affinities.
types associated with these variants correlated with some • APOE transports lipids packed into HDL-like particles in
Alzheimer’s disease endophenotypes such as worse cogni­ the brain, or LDL particles in the peripheral blood. APOE
tion, more severe hippocampal atrophy, lower plasma main receptors in the brain are the LRP1, the LDL receptor,
Aβ levels, and higher brain APOE mRNA levels. Another the very LDL receptor, and the apolipoprotein E
analysis of whole genome sequencing data stratified receptor 2, all of which are also Aβ receptors.3,7–9,10
by APOE genotype identified three genes significantly • Lipidation of brain APOE is mediated by ATP-binding
associated with Alzheimer’s disease in APOE ε4 carriers cassette transporters A1 and G1.11–13
only: OR8G5 (p=4·67 × 10–7), IGHV3–7 (p=9·75 × 10–16), and • APOE directly interacts with amyloid β peptide,5,14–17 but
SLC24A3 (p=2·67 × 10–12).30 Conversely, a systematic review there is no solid in-vivo evidence of a direct interaction
investigating the genetic basis of resilience to Alzheimer’s between APOE and tau.18
disease among APOE ε4 homozygotes revealed that • Mouse models to study the effects of APOE isoforms
CASP7 (encoding caspase 7) rs10553596 and SERPINA3 on amyloid β peptide and tau include mice deficient in
(encoding α1-antichymotrypsin) rs4934-A/A polymorph­ APOE (Apoe knockout) and mice genetically engineered to
isms possibly reduce Alzheimer’s disease risk.31 replace the mouse Apoe with each of the human
APOE alleles (APOE-targeted replacement or knock-in),
Influence of race in APOE-linked Alzheimer’s disease risk crossed with either mice overexpressing one or more
An interaction between race and the effect of APOE geno­ familial Alzheimer’s disease-linked APP mutations—
type on Alzheimer’s disease risk has long been known, with or without one or more PSEN1 mutations
with African American and Hispanic APOE ε4 carriers (eg, APPV717F, 19,20 APPswePSEN1dE9,21,22 5xFAD,23
having a lower risk than white APOE ε4 carriers, and Asian APPPS1–21)21,24—or mice overexpressing frontotemporal
(ie, Japanese) carriers having the highest ORs.32–34 Studies lobar degeneration-tau-linked MAPT mutations
have found that the local ancestry of APOE (ie, the (eg, MAPTP301S).25,26
population-specific genetic variation within the APOE

www.thelancet.com/neurology Vol 20 January 2021 69


Review

Alzheimer’s disease (ie, mild cognitive impairment and


A Plaque Aβ plaque B Capillary CAA
core Endothelial cell mild-to-moderate dementia).39,40 The APOE ε4 allele has
Anti-APOE
ASOs also been associated with more severe tau pathology as
defined by Braak neurofibrillary tangles stages,2,41 and the
Astrocyte APOE ε2 allele with a lower Braak neurofibrillary tangles
Microglia stage,2,4 independently of their effects on Aβ plaques.
Pericyte Cross-sectional data on tau PET imaging examining
APOE effects on tau radioligand uptake after controlling
ABCA1
and RXR ABCA1 Anti-APOE for Aβ radioligand uptake are conflicting,42,43 but longi­
agonists antibodies Unlipidated tudinal combined tau and Aβ PET studies will elucidate
APOE4
C Lipidated this important question.
APOE4
Gene
APOE4 The APOE genotype can also affect the finding of
LRP1
therapy
TREM2 comorbid brain pathologies at autopsy. On one hand,
APOE2
Soluble tau APOE ε4 partly drives (together with ageing) the presence
Neuron Soluble Aβ of Aβ plaques and neurofibrillary tangles in indi­viduals
Neurofibrillary
Aβ fibrils
tangle with other primary neuropathological diag­noses such as
Synapse Tau fibrils
amyotrophic lateral sclerosis, primary tauopathies, and
Figure: Multifaceted effects of APOE in the brain and potential strategies to decrease APOE4 and increase Lewy body diseases.44 On the other hand, in individuals
APOE2 levels
In the healthy brain, APOE is expressed and secreted predominantly by astrocytes, and to a lesser extent by with Alzheimer’s disease the APOE ε4 allele appears
microglia. Most brain APOE is lipidated by the ATP-binding cassettes A1 (ABCA1) and G1 (ABCG1) and lipidated to correlate with the presence and severity of TDP-43
APOE is internalised via APOE receptors such low-density lipoprotein receptor-related protein 1 (LRP1), which is pathology,41,45 Lewy body diseases,46 and possibly cerebrovas­
expressed in astrocytes, neurons, vascular smooth muscle cells, endothelial cells, and pericytes. In the Alzheimer’s
cular disease,47 independently of its effects on Aβ plaques
disease brain, astrocytes and microglia react to (A) dense-core Aβ plaques,52 (B) cerebral amyloid angiopathy-
laden arteries and capillaries, and (C) neurofibrillary tangles, activating transcriptional programmes that include and neurofibrillary tangles. Lastly, APOE could be a genetic
APOE mRNA up-regulation in microglia53,54 and down-regulation in astrocytes53,55 and lead to altered lipid risk factor for neurodegenerative diseases other than
metabolism (not shown). APOE directly interacts with both soluble and fibrillar Aβ.3 Relative to APOE3 and Alzheimer’s disease. Indeed, APOE ε4 has been associated
APOE2, APOE4 promotes Aβ seeding and aggregation in oligomers and fibrils14,15,56 and reduces its clearance from
with Lewy body diseases, independently of the Aβ plaque
the interstitial fluid,20 potentially leading to Aβ deposition as dense-core (thioflavin-S positive) amyloid plaques
and cerebral amyloid angiopathy together with APOE.3 This evidence suggests that decreasing APOE (especially and neurofibrillary tangle burdens.48,49,50 Of note, APOE ε4
APOE4) expression or blocking the effects of APOE4 or enhancing the effects APOE2 would be beneficial has been associated with an earlier age of symptom onset
(dashed boxes). Experimental approaches to achieving these outcomes include lowering APOE4 levels with in patients with MAPT-linked or autopsy-proven fronto­
isoform-specific antisense oligonucleotides24 or antibodies,57–59 which could also target lipid-poor APOE associated
temporal lobar degeneration-tau indepen­ dently of its
with plaques.59 Alternatively, APOE4 could be switched to APOE3 or APOE2,60 or APOE2 could be added,6,22 with
gene therapy. Last, APOE lipidation could be enhanced with RXR61–63 and ABCA1 or ABCG113 agonists to improve effects on Aβ plaque burden,51 and with more severe neu­
APOE4 receptor-mediated internalisation and lower Aβ in the interstitial fluid. Dashed boxes illustrate the most ro­­­degen­eration at post-mortem examination in primary
promising therapeutic approaches. ASO=antisense oligonucleotides. Aβ=amyloid-β peptide. CAA=cerebral tauopathies.25 Paradoxically, APOE ε2 might increase the
amyloid angiopathy. TREM2=triggering receptor expressed in myeloid cells 2. RXR=retinoid X receptor. Adapted
from Servier Medical Art by Servier, which is licensed under a CC BY 3.0 licence.
risk of progressive supranuclear palsy,18 but results are
conflicting.46 The validation and expansion of PET imaging
and CSF biomarkers for other neuro­degenerative diseases
region), rather than global ancestry (ie, the population- will help to confirm these correlations between APOE
specific genetic variation in the entire genome) or genotype and non-Alzheimer’s pathologies.
environmental fac­ tors, explains these inter-racial differ­
ences in Alzheimer’s disease risk.35–38 Speci­ fically, an APOE pathophysiological mechanisms
African local ancestry region sur­rounding APOE underlies Although traditionally the APOE ε4 allele was represented
the smaller APOE ε4 allele effect on Alzheimer’s disease as a trigger of Aβ accumulation at the top of the sporadic
risk observed in African American and Caribbean Hispanic Alzheimer’s disease amyloid cascade, numerous new data
(from Puerto Rico) popula­ tions.35,36 Another study of show that the APOE alleles have differential downstream
809 indi­viduals identi­fied a poten­tially protective African effects in many other patho­physiological processes beyond
ances­tral haplo­type within APOE defined by the rs769449 Aβ metabolism (figure).
SNP,37 but this was not confirmed in a larger (7997 indi­
viduals) study.38 Cellular sources of APOE in brains with and without
Alzheimer’s disease
New pathological correlates of APOE genotype Brain and peripheral pools of APOE are independent from
The classic post-mortem neuropathological correlates of each other because liver transplantation changes APOE
the APOE genotype are a higher Aβ plaque burden and isoforms towards the donor’s in the recipient’s blood but
more severe cerebral amyloid angiopathy in APOE ε4 not CSF,64 and depleting APOE from hepatocytes—its
carriers, and a lower Aβ plaque burden in APOE ε2 main source cell type in the periphery—does not affect
carriers, relative to APOE ε3 homozygotes.4 These differ­ brain APOE (or Aβ) levels in mice.64 Understanding the
ential effects of APOE alleles have been confirmed by cell types expressing APOE in the brain is relevant because
Aβ PET imaging across preclinical and clinical stages of APOE is a secreted glycoprotein and could have autocrine

70 www.thelancet.com/neurology Vol 20 January 2021


Review

effects on the secreting cell, but also paracrine effects on Aβ metabolism remains debated. On one hand, it has been
neighbouring cells. Although astrocytes are the main proposed that APOE and Aβ direct interaction in the brain
source of APOE in the normal brain, in the Alzheimer’s extracellular space might be negligible in physiological
disease brain reactive astrocytes around Aβ plaques were conditions (ie, Aβ monomers and lipidated APOE), but
reported to be devoid of APOE, whereas Aβ plaque- that both APOE and Aβ can compete for the same
associated microglia express high levels of APOE66 receptors, namely LRP1,16 which is involved in Aβ clearance
(figure). Single nuclei RNA-sequencing studies in human by neurons,7 astrocytes,8 endothelial cells, vascular smooth
Alzheimer’s disease and control brains have confirmed a muscle cells,9 and pericytes.72 On the other hand, there is
down-regulation of APOE expression in reactive astro­ evidence supporting a direct interaction between APOE
cytes53,55 and an up-regulation in activated microglia.53,54,67 and oligomeric and fibrillar Aβ. First, APOE colocalises
Neuropathological studies also reported APOE staining in with synaptotoxic Aβ oligomers at the synapses in the
pyramidal neurons in neurodegenerating areas such as vicinity of Aβ plaques and leads to synapse loss in an
the hippocampus, but rare colocalisation between APOE isoform-dependent manner (APOE4 more than APOE3).55
and tangles, suggesting little direct interaction between Second, in-vivo experiments in which APOE expres­sion by
APOE and tau.3,67 The presence of APOE in pyramidal neu­ astro­cytes was conditionally deleted, or APOE expression
rons suggested the internalisation of APOE lipoparticles globally silenced with anti­sense oligonucleo­tides at differ­
from the interstitial space through the APOE receptor ent stages of Aβ deposition, have shown that APOE influ­
LRP1 (panel 1, figure),67 which is highly expressed in ences Aβ plaque burden mainly during the seeding phase
neurons among other cell types. Expression of APOE has of Aβ aggregation, but has a lesser effect during the
also been shown in vascular cells from the human brain, exponential growth phase (ie, when fibrillar Aβ deposits
speci­fically pericytes.68 are already formed).56,24 Third, a subtle dif­ference in tertiary
conformation across APOE isoforms (ie, closer N-terminus
Effects on Aβ and C-terminus in APOE4 vs APOE3 and more open in
The disparate effects of APOE isoforms on Alzheimer’s APOE2) could affect both the affinity of the Aβ and APOE
disease risk was attributed to a differential effect— interaction (higher for APOE4 vs APOE3 and APOE2) and
deleterious for APOE4 and protective for APOE2, with the APOE propensity to enzymatic cleavage in its hinge
respect to APOE3—on both Aβ plaque burden and cere­ region between the N-terminus and the C-terminus,
bral amyloid angiopathy severity.4 These well estab­lished rendering presumably toxic C-terminal frag­ ments (also
autopsy neuropathological correlates of APOE alleles higher for APOE4 vs APOE3 and APOE2).4,17,58
and the early observation that compact (dense-core, fib­
rillar, thioflavin-S-positive), but not diffuse (amorphous, Effects on tau
thioflavin-S-negative), Aβ plaques contain APOE,3 sup­ Unlike Aβ, there is little overlap between APOE-immuno­
ported the idea that APOE interacts with Aβ and pro­ reactive neurons and neurons that have neurofibrillary
motes its aggregation and deposition in insoluble fibrillar tangles.67 No direct interaction between APOE (primarily
deposits (figure).3 Indeed, genetic deletion and haplo­ secreted) and the microtubule-associated protein tau
insufficiency of APOE reduces dense-core Aβ plaque bur­ (primarily intraneuronal and axonal) has been shown in
den in various mouse models of cerebral β amyloidosis.21,69,70 vivo.30 However, studies25,26 in transgenic Apoe knockout or
Of note, APOE deficiency inhibits dif­fuse Aβ deposits in APOE knock-in mice overexpressing the P301S mutant
some of these models,69 but increases them in others,21 form of tau have shown that the human APOE isoforms
further reinforcing the requirement of APOE for plaque do affect tau downstream pathology. Specifically, APOE4
compaction. When these Aβ-plaque depositing mice were promotes tau-induced neurodegeneration and atrophy
crossed with APOE targeted replacement mice expressing compared with APOE3, whereas APOE2 is protective
human APOE alleles in place of the murine Apoe coding with respect to these outcomes.25 The mech­anism under­
sequence (knock-in mice; panel 1), APOE4 knock-in mice lying these effects is indirect, mediated by APOE effects
consistently exhibited higher Aβ plaque burden than did on microglia, rather than a direct interaction between
APOE3 knock-in mice, and these APOE3 knock-in mice APOE and tau; transcriptome profiling and cytokine
had higher Aβ plaque burden than did APOE2 knock- measures indicate that APOE4 microglia is primed
in mice,19,20,23 thus recapitulating the allele-specific differ­ towards a proinflam­ matory phenotype compared with
ences observed in human post­-mortem autopsy and Aβ APOE3, whereas APOE2 exhibits a more homeostatic
PET studies.4,39,40 pheno­type.25,75 Of note, LRP1 has been shown to be a
In-vitro and in-vivo studies have shown that, relative to receptor for tau uptake by neurons, and APOE affects the
APOE2 and APOE3, APOE4 promotes the seeding of ability of tau to bind LRP1 (figure), although in vitro all
Aβ peptide into Aβ oligomers, protofibrils, and fibrils,14,15,56 APOE isoforms reduced tau uptake to a similar extent.75
but also inhibits Aβ clearance from the brain prolong­ Additionally, knocking down neuronal LRP1 reduced
ing its half-life in the interstitial fluid20,56 and inhibiting neuronal tau spreading in mice, but some astrocytes took
its enzymatic degradation.71 The intimate mechanism up the human tau.75 Whether different receptors have
underlying these APOE isoform-driven differences in a role in tau uptake into different cell types remains

www.thelancet.com/neurology Vol 20 January 2021 71


Review

unknown. Moreover, since LRP1 is a recycling receptor haemor­ rhage and cortical superficial side­ rosis, cortical
delivered into endosomal or lysosomal compartments, it micro­­infarcts, and white matter ischaemic changes.
remains not clear how tau escapes to the cytoplasm to APOE ε4 effects on the blood–brain barrier were also
interact with endogenous tau in neurons or to accumulate shown in the first randomised clinical trials with anti-Aβ
as glial fibrillary tangles in astrocytes, but it is plausible mono­clonal antibodies,84 which reported a higher inci­
that APOE affects tau intra­ cellular trafficking in an dence of MRI findings (brain oedema, microbleeds, and
isoform-dependent manner.76 cortical super­ficial siderosis) in the treatment versus pla­
cebo groups—collectively termed amyloid-related imag­
Effects on glia ing abnor­malities.84 Only occasion­ ally symptomatic
Astrocytes and microglia are known to react to plaques, (eg, headaches, confusion, and sei­zures), these amyloid-
neurofibrillary tangles, and neurodegeneration. Although related imaging abnormalities are indicative of an
quantitation of reactive (GFAP+) astrocytes and activated increased blood–brain barrier per­ meability presumably
(IBA1+, CD68+) microglia per Aβ plaque in post-mortem caused by the antibody-mediated Aβ efflux from the brain
sections of the temporal neocortex has shown no dif­ paren­ chyma into the blood­ stream.84 Because amyloid-
ference between APOE ε4 carriers and non-carriers,52 related imaging abnor­malities are twice as probable in
transcripto­mic studies60,77 have reported that APOE influ­ APOE ε4 carriers, their occur­rence has been attributed to
ences glia reactions. Microglia from APOE4 knock-in a more severe pre-existing cerebral amyloid angio­pathy in
mice is primed towards a proinflammatory response APOE ε4 carriers vs APOE ε3 homozygotes.84 Support­ing
compared with those from APOE3 knock-in mice77 and this inter­ pretation, immu­ no­
therapy with an anti-Aβ
APOE ε4 microglia derived from human-induced pluri­ mono­clonal antibody is associ­ated with higher numbers
potent stem cells exhi­bit a proinflammatory gene expres­ of cerebral microbleeds in APPswePSEN1dE9 x APOE4
sion pro­gramme and impaired Aβ phagocytosis relative to knock-in mice versus APOE3 knock-in and APOE2 knock-
APOE ε3 microglia.60 These APOE-mediated differential in mice.85 A post-mortem quantitative neuropatho­logical
effects on microglia phenotype appear to be at least study86 on individuals who participated in a phase 2
partially mediated by the triggering receptor expressed on anti-Aβ active immuno­therapy trial (NCT00021723) also
mye­loid cells 2 (TREM2), which is another receptor for showed that Aβ plaque clearance is associated with a
both Aβ and APOE expressed by microglia.78 Loss of func­ redistribution of Aβ and APOE from plaques to vessels,
tion muta­tions in TREM2 (eg, R47H, R62H) have been and more severe cerebral amy­ loid angiopathy-related
associated with a 2–3 times increased risk of develop­ing vasculopathic changes. Pericytes, which are another
Alzheimer’s disease and with less compact Aβ plaques cellular source of APOE,68 are gain­ing atten­tion for their
that have more neuritic dys­ trophies, less coverage by implication in cere­bral amyloid angio­pathy patho­genesis
microglia, and less APOE con­ tent.79 The Aβ plaque (figure). Pericyte loss resulted in increased cerebral
features of Alzheimer’s disease mouse models deficient in amyloid angiopathy severity and Aβ plaques in a mouse
TREM2 or APOE are pheno­copies,21,79 suggesting that model of Aβ deposi­tion.87 Pericytes take up Aβ via LRP1
APOE and TREM2 are both involved in chemotaxis of in an APOE isoform-dependent manner, with APOE4
microglia towards plaques and that plaque-associated interfer­ing with this uptake com­ pared with APOE3.72
microglia has a neuroprotective role minimising neuritic Human APOE ε4 peri­cytes express higher levels of APOE
dystrophies. The transcriptomic changes associ­ated with mRNA and protein than APOE ε3 peri­cytes, result­ing in
the conver­sion from homeostatic to Alzheimer’s disease increased Aβ vascular accumu­lation.68 Pericytes exposed
micro­glia require both APOE and TREM2 because genetic to Aβ oligomers con­ strict capil­
laries via endothelin-1
deletion of either in Alzheimer’s disease transgenic mice receptor ETA activation, leading to reduced blood flow.88
precludes such transition,80,81 and TREM2 loss of func­ APOE4 can also increase blood–brain barrier per­
tion mutations partially abrogate the microglia trans­crip­ meability with respect to APOE3 in an Aβ-independent
tomic changes observed in the brains of patients with manner, as shown in APOE4 versus APOE3 knock-in
Alzheimer’s disease.54 Regarding APOE effects on astro­ mice89,90 and confirmed with dynamic contrast-enhanced
cytes, APOE ε4 human-induced pluripotent stem cell- MRI in the medial temporal lobe of cognitively healthy
derived astrocytes exhibit impaired choles­terol metab­­o­lism (clinical dementia rating score 0) and mildly impaired
and Aβ phago­cyto­sis,60 reduced neurotro­phic sup­port,82 (clinical dementia rating score 0·5) APOE ε4 carriers
and impaired synaptic pruning,83 relative to APOE ε3 and versus APOE ε3 homozygotes.47 The underlying proposed
APOE ε2 astrocytes. mech­anisms include the activa­tion of cyclophilin A, result­
ing in increased levels of MMP9 and pericyte injury,90
Effects on blood–brain barrier and disruption of the capillary base­ment membrane (ie,
Another area of growing interest is the effects of APOE ε4 collagen IV).89
allele on the blood–brain barrier. Traditionally, APOE ε4
had been associated with a more severe cerebral amyloid APOE-based therapeutic opportunities
angiopathy (figure), resulting in a higher risk of lobar Experimental in-vivo studies in Alzheimer’s disease mouse
intracerebral haemorrhage, but also focal sub­arachnoid models that have a human APOE knock-in background

72 www.thelancet.com/neurology Vol 20 January 2021


Review

have suggested promising approaches to amelio­ rate Increasing APOE levels and its lipidation
pheno­types related to Alzheimer’s disease (table 1). How­ Because brain APOE4 is less lipidated and stable than are
ever, there are only a few APOE-directed clinical trials APOE3 and APOE2,5,6 increasing brain APOE levels and
completed or underway (table 2), highlighting a lag in lipidation has been proposed as a therapeutic approach.
therapeutic translation for this target. Genetic deletion of ABCA1 results in poor APOE lipidation

Route Mouse model Age Duration Statistically significant results Comments


Increased APOE levels
Bexarotene61 Oral APPswe/PSEN1dE9 6 and 3, 7, Increased APOE, ABCA1, ABCG1, and Subsequent mouse studies by other
11 months 14, and HDL; reduced soluble or insoluble Aβ groups91–96 yielded mixed results on the
90 days and Aβ plaques; improved memory efficacy of bexarotene on Aβ phenotypes
Bexarotene61 Oral APPPS1‐21 7–8 months 20 days Increased APOE, ABCA1, ABCG1, Subsequent mouse studies by other
and HDL; reduced soluble and groups91 –93 yielded mixed results on the
insoluble Aβ, and Aβ plaques; efficacy of bexarotene on Aβ phenotypes
improved memory
Bexarotene61 Oral Tg2576 12–14 months 3 and Improved social behaviour, olfaction, Subsequent mouse studies by other
9 days and circuit connectivity groups91–93 yielded mixed results on the
efficacy of bexarotene on Aβ phenotypes

CS‐625313 Intraperitoneal APOE3 knock-in; APOE4 2·5 months 6 weeks APOE4 knock-in only: reduced Aβ and ABCA1 agonist derived from
knock-in phosphorylated tau; increased APOE C‐terminus, target engagement
ABCA1, APOE lipidation, APOER2, shown by CS‐6253+ astrocytes in
VGLUT1, and memory immunohistochemistry
Probucol97 Oral Wild-type male rats 26 months 30 days Increased hippocampus APOE, APOE Aged wild-type rats used as a model of
mRNA, HMGCoAR, LRP, and SNAP25; normal cognitive ageing
unchanged cholesterol, LDL receptor,
and synaptophysin; reduced glial
fibrillary acidic protein
Probucol98 Intraperitoneal Wild-type mice, 90 days 2 weeks Reduced plasma cholesterol; Acute intracerebroventricular injection of
intracerebroventricular improved memory and synapses Aβ in wild-type mice is not a faithful
aggregated Aβ 1–40 model of the Alzheimer’s disease scenario
of chronic Aβ accumulation
APOE mimetics
COG141099 Subcutaneous SwDI‐APP/NOS2 knock-out 9 months 3 months Reduced Aβ plaques, phosphorylated SwDI‐APP mice lacking nitric oxide
tau, and interleukin-6 mRNA; synthetase 2 exhibit endogenous tau
improved memory pathology and neuron loss besides
Aβ plaques
COG112100 Intraperitoneal Amyloid-β precursor 1 month 3 months Reduced phosphorylated tau and APP intracellular domain overexpressing
protein intracellular CD45+ and ionised calcium binding mice exhibit microglial activation but no
domain Tg (FeCγ 25 line) adapter molecule-1+ microglia; Aβ plaques
improved neurogenesis
CN‐105101 Subcutaneous APPPS1-21/APOE4 14–18 weeks 40 days Reduced soluble Aβ and Aβ plaques; Greater benefits in young mice
knock-in improved memory (14–18 weeks); for example, improved
CN‐105101 Subcutaneous APPPS1-21/APOE4 25–28 weeks 40 days Unchanged soluble Aβ; improved fear conditioning but not spatial memory
knock-in memory in older mice (25–28 weeks)

Blocking APOE and Aβ interaction


HJ6·3 anti‐APOE Intraperitoneal APPswe/PSEN1dE9 4 months 14 weeks Reduced soluble and insoluble Aβ, Preventative use prior to Aβ plaques
57
antibody Aβ plaques, interferon γ, and very effective compared with therapeutic
interleukin 1α; increased use, microglial activity modulation
CD45+ microglia suggested
HJ6·3 anti‐APOE Intraperitoneal APPswe/PSEN1dE9 7 months 21 weeks Reduced soluble and insoluble Aβ, Therapeutic use after Aβ plaque
antibody58 Aβ plaques, brain APOE, deposition also effective due to inhibition
CD45+ microglia, increased plasma of plaque formation and growth plus
Aβ; improved memory and removal of existing plaques
connectivity; unchanged cerebral
amyloid angiopathy, plasma APOE,
and plasma cholesterol
HAE‐4 anti‐APOE Intracerebroventricular APPPS1‐21/APOE4 2 months 6 weeks Reduced Aβ plaques; unchanged Additionally AAV2/8-mediated expression
antibody59 knock-in insoluble Aβ of HAE‐4 and HAE‐1 anti‐APOE
antibodies via intracerebroventricular
injection at birth reduced Aβ plaque and
insoluble Aβ at age 3·5‐months in a
microglia FcγR1‐dependent manner
(Table 1 continues on next page)

www.thelancet.com/neurology Vol 20 January 2021 73


Review

Route Mouse model Age Duration Statistically significant results Comments


(Continued from previous page)
HAE‐4 anti‐APOE Intraperitoneal APPPS1–21/APOE4 2 months 6 weeks Reduced insoluble Aβ and Aβ Additionally AAV2/8-mediated expression
antibody59 knock-in plaques; unchanged brain soluble of HAE‐4 and HAE‐1 anti‐APOE
and insoluble APOE, plasma APOE, antibodies via intracerebroventricular
and plasma Aβ injection at birth reduced Aβ plaque and
insoluble Aβ at age 3·5 months in a
microglia FcγR1‐dependent manner
Aβ12–28P102 Intraperitoneal APPswe/PSEN1dE9/APOE2 6 months 4 months Reduced soluble and insoluble Aβ, Greater cognitive benefit in APOE4 mice
knock-in; APPswe/ soluble and insoluble APOE, Aβ compared with APOE2 mice because only
PSEN1dE9/APOE4 knock-in plaques, dystrophic neurites, and vehicle‐treated APPswe/PSEN1dE9/
serum cholesterol in both mice; APOE4 knock-in mice (but not
improved memory (only APOE4 APPswe/PSEN1dE9/APOE2 knock-in
knock-in mice); unchanged mice) exhibited impaired cognition
serum APOE
Silencing APOE
Anti‐APOE antisense Intraperitoneal APPPS1–21/APOE3 and Birth 16 weeks Reduced soluble and insoluble Aβ, Reduced Aβ plaque burden when
oligonucleotides24 APPPS1–21/APOE4 Aβ plaques, dystrophic neurites, treated at birth but no change or
knock-in and soluble APOE; unchanged increased when treated at 6 weeks
insoluble APOE suggests preventative rather than
therapeutic use
Anti‐APOE antisense Intracerebroventricular APPPS1‐21/APOE3 and 6 weeks 10 weeks Reduced soluble APOE and Reduced Aβ plaque burden when
oligonucleotides24 APPPS1-21/APOE4 dystrophic neurites; unchanged treated at birth but no change or
knock-in soluble and insoluble Aβ, insoluble increased when treated at 6 weeks
APOE, and CD45+ microglia; suggests preventative rather than
unchanged or increased Aβ plaques therapeutic use
Switching APOE4 to APOE2
AAV4 and APOE2, Intraventricular APPswe/PSEN1dE9 7 months 2 months AAV4-APOE2 vs APOE3: reduced AAV4-mediated expression of APOE2
APOE3, or APOE422 and soluble and insoluble Aβ, Aβ mainly in choroid plexus and
5 months plaques, and dystrophic neurites, ependymal cells via intraventricular
and unchanged plasma Aβ; increased injection can improve Aβ measures in
synapses. AAV4‐APOE4 vs APOE3: aged mice (>12 months old) after
increased soluble and insoluble Aβ, Aβ plaque deposition
Aβ plaques, dystrophic neurites,
and plasma Aβ; reduced synapses
AAV4 and APOE2, Intraventricular Tg2576 16–18 months 3 months AAV4‐APOE2 vs APOE3: AAV4-mediated expression of APOE2
APOE3, or APOE422 reduced interstitial fluid Aβ and mainly in choroid plexus and
oligomeric Aβ, unchanged insoluble ependymal cells via intraventricular
Aβ. AAV4‐APOE4 vs APOE3: injection can improve Aβ measures in
increased interstitial fluid Aβ, aged mice (>12 months old) after
oligomeric Aβ, and insoluble Aβ Aβ plaque deposition
AAV8‐GFAP and Intracerebroventricular APOE3 knock-in and Birth 3 months AAV8-GFAP‐APOE2 vs APOE3: Results suggest that APOE4 is less
APOE2, APOE3, or APOE4 knock-in increased APOE and APOE lipidated and stable than APOE2, and
APOE46 lipidation; reduced mouse Aβ 40 that exogenous expression of APOE4 in
(trend in APOE4 knock-in only); APOE4 carriers could be deleterious by
unchanged APP‐full length, increasing endogenous Aβ, whereas
APP‐CTFs, ABCA1, ABCG1, and expression of APOE2 in APOE4 carriers
Apoe mRNA. AAV8‐GFAP‐APOE4 vs would have opposing beneficial effects
APOE3: increased mouse Aβ 40
(APOE4 knock-in mice only); reduced
APOE lipidation; unchanged APP‐FL,
APP‐CTFs, ABCA1, ABCG1, APOE and
Apoe mRNA.

APPswe=Swedish APP mutation (K670N/M671L). PSEN1dE9=presenilin-1 exon 9 deletion. APPPS1-21=Swedish APP mutation (K670N/M671L) and presenilin-1 L166P mutation. Tg2576=transgenic Swedish
(K670N/M671L) APP mutation overexpressing mice. APOER2=Apolipoprotein E receptor 2. SwDI-APP=Swedish (K670N/M671L), Dutch (E693Q) and Iowa (D694N) APP mutation. AAV4=adeno-associated virus
type-4. AAV8=Adeno-associated virus type-8. APP-FL=APP full length; APPswePSEN1dE9=Swedish APP mutation (K670N/M671L) and presenilin-1 exon 9 deletion; FcγR1=Fc gamma receptor-1.
AAV2/8=Adeno-associated virus type 2/8. APOER2=APOE receptor 2.

Table 1: Summary of APOE-directed therapeutic approaches tested in Alzheimer’s disease mouse models

and increased Aβ plaque burden,11 whereas ABCA1 over­ cause a rapid reduction of Aβ plaque burden and restora­
expression reduces Aβ deposition.12 ABCA1 and ABCG1 tion of cognitive functioning in Alzheimer’s disease
expression is induced by the stimulation of the reti­ mouse models by inducing ABCA1 and ABCG1 expres­
noid X receptor. Bexarotene is a US Food and Drug sion, enhancing APOE lipidation, and increasing APOE
Administration approved retinoid X receptor agonist for levels (table 1).61 This result was, at least partly, replicated
use in cutaneous T-cell lymphoma and was reported to by some investigators but not others, and led to examine
91–96

74 www.thelancet.com/neurology Vol 20 January 2021


Review

Rationale Status Design Phase Participants Dose, route, and Primary Secondary outcomes Results
duration outcome
Bexarotene62 Increase Completed Randomised 1b Young,healthy 225 mg twice daily Newly Fractional clearance Poor bexarotene
APOE levels double‐blind, participants (aged orally for 5 days generated Aβ rate of Aβ from CNS brain penetration
placebo‐ 21–49 years) with in CSF (stable (stable isotope (not detectable in
controlled trial APOE ε3 isotope labelling kinetics) >95% CSF samples),
homozygosity labelling 25% increase in CSF
kinetics) APOE levels, non
significant (p=0·054)
increase in newly
synthesised APOE,
no change in
Aβ synthesis or
clearance levels
Bexarotene63 Increase Completed Randomised, 2 Participants with 150 mg twice daily Aβ burden in Cognition (Mini Mental Significant reduction
APOE levels double-blind, moderate Alzheimer’s orally for 4 weeks amyloid PET State Examination, in Aβ burden only in
placebo‐ disease (Mini Mental imaging Alzheimer’s Disease APOE ε4 non‐carriers
controlled trial State Examination; Assessment which correlated
range 10–20) with Scale-Cognitive with increased serum
positive baseline subscale, clinical Aβ 42 levels,
amyloid PET scan dementia rating scale), increased triglyceride
behaviour plasma level in
(Neuropsychiatric bexarotene group,
Inventory), Activities of no efficacy in
Daily Living, serum clinical outcomes
Aβ 40/42
Probucol Increase Completed Open label, 1/2 Cognitively healthy Initial 600 mg once Plasma No outcomes reported Not available
(NCT02707458) APOE levels dose finding participants at risk of daily orally, then probucol, CSF (study not published)
trial Alzheimer’s disease individualised, 1 year and plasma
by family history of follow‐up APOE levels
CN‐105 APOE Recruiting Randomised, 2 Adults aged ≥60 who 0·1 mg/kg vs Safety CSF cytokine levels, Not available
(NCT03802396) mimetic double-blind, are having major 0·5 mg/kg vs change in cognition, (study ongoing)
placebo‐ surgery 1 mg/kg incidence of
controlled trial intravenously every postoperative delirium
6 h for 4 days and
6 weeks of follow‐up
Gene therapy Switch Recruiting Open label, 1 Symptomatic Single intracisternal Safety Maximum dose Not available
(AAVrh.10hPOE2 APOE4 to dose ranging participants (any injection 8 × 10¹⁰ tolerated (study is ongoing)
vector; APOE2 trial stage), APOEε4 genome copies per kg
NCT03634007) homozygotes, vs 2·5 × 10¹¹ genome
positive copies per kg vs
CSF biomarkers, or 8 × 10¹¹ genome
amyloid PET scan copies per kg, and
2 years of follow-up

Aβ=amyloid β.

Table 2: Early development of APOE-directed therapeutic approaches in human clinical trials

bexarotene for Alzheimer’s disease in human clinical APOE lipidation without chang­ing brain APOE levels, in
trials. A phase 1b proof-of-mechanism trial62 in young APOE4 but not APOE3 knock-in mice.13 These effects
(21–49 years) volunteers revealed poor penetration of correlated with a reduction of hippo­ campal Aβ and
bexarotene in the CNS according to its plasma versus CSF phosphorylated tau and improved learning and memory in
levels. Bexarotene was able to increase CSF APOE levels APOE4 knock-in mice.13
by 25%, although it had no effects on CSF Aβ levels as Probucol, the now abandoned non-statin lipid-lowering
measured by stable isotope labelling kinetics (table 2). In a drug, has been shown to counteract hippocampal synaptic
proof of concept, double-blind, placebo-controlled clinical loss and cognitive impairment in Aβ-injected wild-type
trial63 in 20 patients with moderate Alzheimer’s disease mice,98 increase APOE and LRP1 levels in the hippocampus
(Mini Mental State Examination range was 10–20; table 2), of aged rats97 and, while results from a phase 1/2 clini­
bexarotene 150 mg twice daily for 4 weeks was associated cal trial (NCT02707458) are awaited, might also increase
with a significant reduction in Aβ PET bur­ den and a CSF APOE levels in humans (table 2).103
proportional increase in serum Aβ42 levels but, contrary to
the prediction, only in APOE ε4 non-carriers. Blocking APOE and Aβ interaction
A derived from the C-terminus of APOE, CS-6253, has Another therapeutic strategy is to interfere with the APOE
been shown to increase ABCA1 levels and, subsequently, and Aβ interaction, because this is thought to stabilise

www.thelancet.com/neurology Vol 20 January 2021 75


Review

toxic oligomeric and fibrillar Aβ species existing within Lowering APOE levels
and around Aβ plaques.14,15,56,73 This strategy has been Lowering brain APOE levels has also been proposed as a
achieved in Alzheimer’s disease mouse models with both therapy because Apoe genetic deletion or haploinsuf­
mono­clonal anti-APOE antibodies and small mole­cules ficiency reduces Aβ deposition in mouse models of
that act as Aβ mimetics. cerebral β-amyloidosis69,70,21 and rescues neurodegenera­
Chronic intraperitoneal administration of an anti-APOE tion induced by tau in tauopathy mouse models.25 Addi­
monoclonal antibody (HJ6·3) to APPswePSEN1dE9 trans­ tionally, null mutations in the APOE gene do not seem to
genic mice led to a statistically significant reduction of have adverse effects on cognition in humans, although
insoluble Aβ levels and Aβ plaque burden, and APOE they are associ­ated with familial dysbetalipoproteinemia
levels in the brain, which correlated with improved learn­ (also known as type III hyperlipoproteinemia).104 One way
ing and memory and higher cortical network con­nectivity of reducing brain APOE levels is increasing the expres­
in the resting state.57,58 While the Aβ plaque reduction was sion of its receptors. Over-expression of LDLR reduced
larger when administered before plaque deposition, Aβ plaque deposition105 due to increased efflux of Aβ from
in older mice with substantial plaque deposition this the brain through the blood–brain barrier.10 A more specific
antibody appears to prevent the formation of new plaques approach is to silence APOE expression with specific anti­
and clear the smallest previously existing plaques by sense oligonucleotides. In APPPS1-21 x APOE3 knock-in
binding APOE within them.57,58 Of note, systemic treat­ mouse and APPPS1-21 x APOE4 knock-in mice, a reduction
ment with this anti-APOE antibody increased plasma in soluble APOE con­centrations by half with anti-APOE
Aβ levels, but did not have systemic (ie, unchanged plasma antisense oligo­nu­cleotides resulted in lower soluble and
cholesterol and APOE levels) or local (ie, cere­bral amy­loid insoluble Aβ levels and lower total and dense-core plaque
angiopathy did not worsen) adverse side-effects.57,58 burden when adminis­ tered intracerebro­ventricularly at
Another anti-APOE monoclonal antibody speci­fic for non- birth, but did not change much these Aβ measures
lipidated APOE (HAE-4) reduced the plaque burden in when applied at the onset of Aβ plaque deposition (ie,
APPPS1-21 x APOE4 knock-in mice through microglia- 6 weeks in this mouse model).24 How­ever, both treatments
mediated clearance, without affecting the levels of plasma resulted in fewer plaque-associated dystrophic neurites,
APOE, which is mostly lipidated.59 Therefore, anti-APOE suggesting less neuronal toxicity of existing plaques
immuno­therapy has promise for testing in future trials. and some beneficial effect of APOE reduction on micro­
Aβ12–28P, a small peptide corresponding to the APOE- glia and astrocyte responses to plaques, and lending
binding motif within Aβ except for a Val18Pro substitution, support for testing in patients with Alzheimer’s disease in
reduced soluble and insoluble Aβ levels and Aβ plaque clinical trials.
burden in APPswePSEN1dE9 x APOE4 knock-in mice and
APPswePSEN1dE9 x APOE3 knock-in mice and improved Genetic switch of APOE isoforms
memory deficits in APPswePSEN1dE9 x APOE4 knock-in Gene therapy has become a reality in several diseases,
mice.102 Moreover, Aβ12–28P reduced soluble and insoluble including neurodegenerative diseases such as spinal
APOE levels and the deposition of APOE into Aβ plaques. muscular atrophy. The application of CRISPR-Cas9 edit­
Of note, this improve­ ment was not due to an active ing technology to switch APOE alleles has been success­
immunisation effect, because these mice did not generate ful in a dish with neurons and glial cells derived from
antibodies against this Aβ fragment.102 Aβ12–28P efficacy human-induced pluripotent stem cells,60 but remains
remains to be tested in clinical trials. to be shown in APOE knock-in mice. However, the
application of gene therapy to express APOE ε2 and
APOE mimetics increase APOE2 levels in APOE ε4 carriers (or even
Another approach is to use APOE N-terminal frag­ APOE ε3 homozygotes) has become feasible and the first
ments that include its receptor-binding motif, so called phase 1 clinical trial with this approach has been initi­
APOE mimetics. Chronic subcutaneous administra­tion of ated (NCT03634007; table 2). In mice, intraventricular
CN-105, a pentapeptide corresponding to the recep­ tor transfer of human APOE alleles with an adeno-associated
binding face of APOE, reduced both soluble Aβ and Aβ virus type-4 leads to sustained expression of human
plaque burden and improved cognition in APPS1-21 x APOE in the choroid plexus and ependymal cell lining,
APOE4 knock-in mice before plaque deposi­tion, but not that diffuses to the brain paren­chyma reaching a con­
after this.101 CN-105 is being tested to pre­vent delirium after centration of 10% of mouse endog­enous APOE.22 Adeno-
major surgery in a phase 2 clinical trial (NCT03802396; associated virus type-4-mediated delivery and expres­sion
table 2). APOE mimetics spanning the APOE recep­ of APOE ε2 in 7-month-old APPswePSEN1dE9 mice
tor binding motif such as COG1410 (12 amino acids) and (ie, after Aβ plaque deposition) resulted in reduced
COG112 (34 amino acids) have also been shown to soluble and insoluble Aβ levels and enhanced plaque
ameliorate Aβ levels and Aβ plaque burden, tau hyper­ clearance, whereas delivery of APOE ε4 had the opposite
phosphorylation, and neuroinflammation in various effects.22 Plasma Aβ40 concentrations were decreased in
Alzheimer’s disease mouse models,99,100 but have not been the APOE ε3 and APOE ε4 treated mice versus APOE ε2
tested in human clinical trials. treated mice,22 suggesting a reduced efflux from brain

76 www.thelancet.com/neurology Vol 20 January 2021


Review

Panel 2: Areas of uncertainty and research priorities Search strategy and selection criteria
• Possible differential independent effects of APOE alleles We searched PubMed for articles in English published between
on Alzheimer’s disease progression through preclinical Jan 1, 1993, and May 15, 2020, using the search terms
and clinical stages with longitudinal multimodal imaging, “APOE AND Alzheimer’s disease”, “APOE AND blood-brain
CSF, and plasma or serum biomarkers barrier”, “APOE AND Lewy body disease”, “APOE AND
• Influence of genetic modifiers of APOE-linked Alzheimer’s alpha-synuclein”, “APOE AND TAR DNA-binding protein 43”,
disease risk, including the intimate mechanisms of local “APOE AND tau”, “APOE AND microglia”, “APOE AND
ancestry, interaction with longevity genetic astrocytes”, “APOE AND TREM2”, “APOE AND
polymorphisms such as KLOTHO-VS heterozygosity, and immunotherapy”, and “APOE AND gene therapy”. Only human,
APOE mutations such as R136S (Christchurch), mouse model, and human-induced pluripotent stem-cell
as plausible substrates of resistance or resilience to studies were reviewed. In-vitro studies using recombinant
Alzheimer’s disease APOE and synthetic or recombinant Aβ or tau species and
• Possible Aβ-independent mechanisms of APOE on tau in-cellulo studies using cultured cell lines or primary neuron,
seeding and propagation through neuronal circuits astrocyte, or microglial cultures were excluded. The final
• Influence of APOE genotype on other neurodegenerative reference list was generated on the basis of relevance and
proteinopathies, such as primary tauopathies originality with regard to the topics covered in this Review.
(eg, Pick´s disease, progressive supranuclear palsy, and
corticobasal degeneration), TDP-43 proteinopathies
(amyotrophic lateral sclerosis, frontotemporal lobar APOE4 levels and to increase APOE2 levels in the brain
degeneration-TDP-43), and α-synucleinopathies hold the greatest promise (table 2).6,22,24,57,58,59 Against this
(Parkinson’s disease, dementia with Lewy bodies, multiple remarkable momen­tum, there remains a paucity in trans­
system atrophy), as well as on other neurological lation of APOE-based therapies to human clinical trials,
disorders in which the blood–brain barrier or the immune especially when compared with the expedited cases of
system play a substantial role anti-Aβ and anti-tau immunotherapies (table 2).84,86 What
• Autocrine versus paracrine effects of APOE on each brain are the hurdles slowing APOE-based drug develop­ment
cell type (astrocytes, microglia, neurons, pro­grammes down? First, further development of small
oligodendrocytes, vascular smooth muscle cells, molecules that reliably change APOE4 conformation to
endothelial cells, and pericytes). APOE3 or APOE2 has proven to be difficult because the
• In-vivo applications of gene therapy, including genetic variable degree of lipidation of APOE might influence its
editing of APOE alleles with CRISPR-Cas9 technology and tertiary conformation.5 Second, the new data implicat­ing a
strategies for viral vector delivery to specific brain variety of non-Aβ and non-tau targets in APOE patho­
cell types physiology raises new questions, such as determining
what the best downstream therapeutic target should be
and monitoring the consequences of target engagement.
to plasma through the brain–blood barrier, relative to Third, there are some unique problems related to APOE
APOE ε2 treated mice. Monitoring of Aβ plaque growth separate peripheral (liver generated) and CNS pools and
by in-vivo multiphoton microscopy in living mice showed its inability to cross the blood–brain barrier.64,65 This means
a significant effect on plaque growth rate, slower in that affecting CNS APOE (levels, isoforms, or interac­
APOE ε2 treated mice and faster in APOE ε4 treated tions) will require drugs with adequate blood–brain
mice.22 Moreover, APOE ε2 ameliorated plaque-associated barrier penetration. Moreover, potential systemic off-target
dystrophic neurites and synapse loss, which were more adverse effects of some of these approaches should be
severe in APOE ε4 treated mice. Similarly, intracere­ carefully considered: rare APOE ε2 homozygotes and
broventricular AAV8-mediated astrocyte-specific expres­ Christchurch mutation carriers,106 and even more rare
sion of human APOE ε2 in APOE4 knock-in mice from individuals with homozygous APOE null mutations104
birth increased APOE lipidation and decreased endog­ suffer from type III hyperlipoproteinemia resulting in
enous murine Aβ, whereas APOE ε4 delivery had opposite accelerated atheroscler­osis; in fact, APOE2 knock-in and
deleterious effects.6 Apoe knockout mice are widely used to model athero­
sclerosis. Therefore, gene therapies to lower APOE levels
Conclusions and future directions or switch APOE4 to APOE2 should probably be targeted
New insights in genetic modifiers, neuropathological and specifically to the CNS (ie, via direct injection or with
gene expression correlates, and pathophysiological mech­ viral capsids that penetrate the blood–brain barrier and
anisms in different brain cell types are broadening our appropriate promoters), which poses its own challenges.
understanding of the implications of APOE in Alzheimer’s Notwithstanding all these barriers, the risk and pro­
disease and offering previously unforeseeable oppor­ tective profiles of APOE genotype in human popula­tions
tunities for therapeutic and preventive interventions. across the globe reinforce the robustness of the effects of
Because of this mounting evidence, strategies to lower subtle variations in this gene, and encourage the field

www.thelancet.com/neurology Vol 20 January 2021 77


Review

to redouble its efforts at further understanding the 15 Hori Y, Hashimoto T, Nomoto H, Hyman BT, Iwatsubo T. Role of
pathophysiology of APOE effects in Alzheimer’s disease apolipoprotein E in β-amyloidogenesis: isoform-specific effects on
protofibril to fibril conversion of aβ in vitro and brain Aβ deposition
(panel 2), and its attempts at translating that knowledge in vivo. J Biol Chem 2015; 290: 15163–74.
into therapeutics. 16 Verghese PB, Castellano JM, Garai K, et al. ApoE influences
amyloid-β (Aβ) clearance despite minimal apoE/Aβ association in
Contributors physiological conditions. Proc Natl Acad Sci USA 2013;
AS-P and SD did the literature search and wrote the manuscript. 110: E1807–16.
BTH reviewed and edited the manuscript. 17 Kara E, Marks JD, Roe AD, et al. A flow cytometry-based in vitro
Declaration of interests assay reveals that formation of apolipoprotein E (ApoE)-amyloid β
BTH receives research funds from AbbVie and F Prime; received complexes depends on ApoE isoform and cell type. J Biol Chem
consultant or science advisory boards honoraria from Arvinas, Biogen, 2018; 293: 13247–56.
Cell Signaling Technology, US Department of Justice, Dewpoint 18 Zhao N, Liu C-C, Van Ingelgom AJ, et al. APOE ε2 is associated
Therapeutics, and Novartis; has a family member who works for with increased tau pathology in primary tauopathy. Nat Commun
2018; 9: 4388.
Novartis and they own Novartis stocks; serves on the Board of Dewpoint
19 Holtzman DM, Bales KR, Tenkova T, et al. Apolipoprotein E
Therapeutics and owns stock; and is funded by the US National Institute
isoform-dependent amyloid deposition and neuritic degeneration in
on Aging (1R01AG047644–01) and the US National Institute of
a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA
Neurological Disorders and Stroke (U01NS111671–01). AS-P is funded by 2000; 97: 2892–97.
the US National Institute on Aging (K08AG064039) and the Alzheimer’s
20 Castellano JM, Kim J, Stewart FR, et al. Human apoE isoforms
Association (AACF-17–524184). SD is funded by the US National differentially regulate brain amyloid-β peptide clearance.
Institute on Aging (P30AG062421). Sci Transl Med 2011; 3: 89ra57.
Acknowledgments 21 Ulrich JD, Ulland TK, Mahan TE, et al. ApoE facilitates the
We want to thank Ayush Noori, Neurology Department, Massachusetts microglial response to amyloid plaque pathology. J Exp Med 2018;
General Hospital, Boston, MA, USA for his invaluable help preparing 215: 1047–58.
the figure. The funder of the study had no role in data interpretation or 22 Hudry E, Dashkoff J, Roe AD, et al. Gene transfer of human Apoe
writing of the report. isoforms results in differential modulation of amyloid deposition
and neurotoxicity in mouse brain. Sci Transl Med 2013; 5: 212ra161.
References 23 Youmans KL, Tai LM, Nwabuisi-Heath E, et al. APOE4-specific
1 Kunkle BW, Grenier-Boley B, Sims R, et al. Genetic meta-analysis of changes in Aβ accumulation in a new transgenic mouse model of
diagnosed Alzheimer’s disease identifies new risk loci and Alzheimer disease. J Biol Chem 2012; 287: 41774–86.
implicates Aβ, tau, immunity and lipid processing. Nat Genet 2019; 24 Huynh TV, Liao F, Francis CM, et al. Age-dependent effects of
51: 414–30. apoE reduction using antisense oligonucleotides in a model of
2 Reiman EM, Arboleda-Velasquez JF, Quiroz YT, et al. Exceptionally β-amyloidosis. Neuron 2017; 96: 1013–23.e4.
low likelihood of Alzheimer’s dementia in APOE2 homozygotes 25 Shi Y, Yamada K, Liddelow SA, et al. ApoE4 markedly exacerbates
from a 5,000-person neuropathological study. Nat Commun 2020; tau-mediated neurodegeneration in a mouse model of tauopathy.
11: 667. Nature 2017; 549: 523–27.
3 Rebeck GW, Reiter JS, Strickland DK, Hyman BT. Apolipoprotein E 26 Shi Y, Manis M, Long J, et al. Microglia drive APOE-dependent
in sporadic Alzheimer’s disease: allelic variation and receptor neurodegeneration in a tauopathy mouse model. J Exp Med 2019;
interactions. Neuron 1993; 11: 575–80. 216: 2546–61.
4 Serrano-Pozo A, Qian J, Monsell SE, Betensky RA, Hyman BT. 27 Arboleda-Velasquez JF, Lopera F, O’Hare M, et al. Resistance to
APOEε2 is associated with milder clinical and pathological autosomal dominant Alzheimer’s disease in an APOE3
Alzheimer disease. Ann Neurol 2015; 77: 917–29. Christchurch homozygote: a case report. Nat Med 2019; 25: 1680–83.
5 Jones PB, Adams KW, Rozkalne A, et al. Apolipoprotein E: isoform 28 Belloy ME, Napolioni V, Han SS, Le Guen Y, Greicius MD,
specific differences in tertiary structure and interaction with Alzheimer’s Disease Neuroimaging Initiative. Association of
amyloid-β in human Alzheimer brain. PLoS One 2011; 6: e14586. klotho-vs heterozygosity with risk of Alzheimer disease in
6 Hu J, Liu C-C, Chen X-F, Zhang Y-W, Xu H, Bu G. Opposing effects individuals who carry APOE4. JAMA Neurol 2020; 77: 849–62.
of viral mediated brain expression of apolipoprotein E2 (apoE2) and 29 Zhou X, Chen Y, Mok KY, et al. Non-coding variability at the APOE
apoE4 on apoE lipidation and Aβ metabolism in apoE4-targeted locus contributes to the Alzheimer’s risk. Nat Commun 2019;
replacement mice. Mol Neurodegener 2015; 10: 6. 10: 3310.
7 Kanekiyo T, Cirrito JR, Liu C-C, et al. Neuronal clearance of 30 Ma Y, Jun GR, Zhang X, et al. Analysis of whole-exome sequencing
amyloid-β by endocytic receptor LRP1. J Neurosci 2013; 33: 19276–83. data for Alzheimer disease stratified by APOE genotype.
8 Liu C-C, Hu J, Zhao N, et al. Astrocytic LRP1 mediates brain JAMA Neurol 2019; 76:1099–1108.
Aβ clearance and impacts amyloid deposition. J Neurosci 2017; 31 Huq AJ, Fransquet P, Laws SM, et al. Genetic resilience to
37: 4023–31. Alzheimer’s disease in APOE ε4 homozygotes: a systematic review.
9 Kanekiyo T, Liu C-C, Shinohara M, Li J, Bu G. LRP1 in brain Alzheimers Dement 2019; 15: 1612–23.
vascular smooth muscle cells mediates local clearance of 32 Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and
Alzheimer’s amyloid-β. J Neurosci 2012; 32: 16458–65. ethnicity on the association between apolipoprotein E genotype and
10 Castellano JM, Deane R, Gottesdiener AJ, et al. Low-density Alzheimer disease. A meta-analysis. JAMA 1997; 278: 1349–56.
lipoprotein receptor overexpression enhances the rate of 33 Tang MX, Stern Y, Marder K, et al. The APOE-epsilon4 allele and
brain-to-blood Aβ clearance in a mouse model of β-amyloidosis. the risk of Alzheimer disease among African Americans, whites,
Proc Natl Acad Sci USA 2012; 109: 15502–07. and Hispanics. JAMA 1998; 279: 751–55.
11 Wahrle SE, Jiang H, Parsadanian M, et al. Deletion of Abca1 34 Evans DA, Bennett DA, Wilson RS, et al. Incidence of Alzheimer
increases Aβ deposition in the PDAPP transgenic mouse model of disease in a biracial urban community: relation to apolipoprotein E
Alzheimer disease. J Biol Chem 2005; 280: 43236–42. allele status. Arch Neurol 2003; 60: 185–89.
12 Wahrle SE, Jiang H, Parsadanian M, et al. Overexpression of 35 Rajabli F, Feliciano BE, Celis K, et al. Ancestral origin of ApoE ε4
ABCA1 reduces amyloid deposition in the PDAPP mouse model of Alzheimer disease risk in Puerto Rican and African American
Alzheimer disease. J Clin Invest 2008; 118: 671–82. populations. PLoS Genet 2018; 14: e1007791.
13 Boehm-Cagan A, Bar R, Liraz O, Bielicki JK, Johansson JO, 36 Blue EE, Horimoto ARVR, Mukherjee S, Wijsman EM,
Michaelson DM. ABCA1 agonist eeverses the ApoE4-driven Thornton TA. Local ancestry at APOE modifies Alzheimer’s disease
cognitive and brain pathologies. J Alzheimers Dis 2016; 54: 1219–33. risk in Caribbean Hispanics. Alzheimers Dement 2019; 15: 1524–32.
14 Hashimoto T, Serrano-Pozo A, Hori Y, et al. Apolipoprotein E, 37 Babenko VN, Afonnikov DA, Ignatieva EV, Klimov AV, Gusev FE,
especially apolipoprotein E4, increases the oligomerization of Rogaev EI. Haplotype analysis of APOE intragenic SNPs.
amyloid β peptide. J Neurosci 2012; 32: 15181–92. BMC Neurosci 2018; 19 (suppl 1): 16.

78 www.thelancet.com/neurology Vol 20 January 2021


Review

38 Mezlini AM, Magdamo C, Merrill E, et al. Characterizing clinical 61 Cramer PE, Cirrito JR, Wesson DW, et al. ApoE-directed
and neuropathological traits of APOE haplotypes in African therapeutics rapidly clear β-amyloid and reverse deficits in
Americans and Europeans. J Alzheimers Dis JAD 2020; 78: 467–77. AD mouse models. Science 2012; 335: 1503–06.
39 Jansen WJ, Ossenkoppele R, Knol DL, et al. Prevalence of cerebral 62 Ghosal K, Haag M, Verghese PB, et al. A randomized controlled study
amyloid pathology in persons without dementia: a meta-analysis. to evaluate the effect of bexarotene on amyloid-β and apolipoprotein E
JAMA 2015; 313: 1924–38. metabolism in healthy subjects. Alzheimers Dement 2016; 2: 110–20.
40 Ossenkoppele R, Jansen WJ, Rabinovici GD, et al. Prevalence of 63 Cummings JL, Zhong K, Kinney JW, et al. Double-blind,
amyloid PET positivity in dementia syndromes: a meta-analysis. placebo-controlled, proof-of-concept trial of bexarotene Xin
JAMA 2015; 313: 1939–49. moderate Alzheimer’s disease. Alzheimers Res Ther 2016; 8: 4.
41 Wennberg AM, Tosakulwong N, Lesnick TG, et al. Association of 64 Linton MF, Gish R, Hubl ST, et al. Phenotypes of apolipoprotein B
apolipoprotein E ε4 with transactive response DNA-binding and apolipoprotein E after liver transplantation. J Clin Invest 1991;
protein 43. JAMA Neurol 2018; 75: 1347–54. 88: 270–81.
42 Ramanan VK, Castillo AM, Knopman DS, et al. Association of 65 Huynh TV, Wang C, Tran AC, et al. Lack of hepatic apoE does not
apolipoprotein E ε4, educational level, and sex with tau deposition influence early Aβ deposition: observations from a new APOE
and tau-mediated metabolic dysfunction in older adults. knock-in model. Mol Neurodegener 2019; 14: 37.
JAMA Netw Open 2019; 2: e1913909. 66 Uchihara T, Duyckaerts C, He Y, et al. ApoE immunoreactivity and
43 Therriault J, Benedet AL, Pascoal TA, et al. Association of microglial cells in Alzheimer’s disease brain. Neurosci Lett 1995;
apolipoprotein E ε4 with medial temporal tau independent of 195: 5–8.
amyloid-β. JAMA Neurol 2020; 77: 470–79. 67 Han SH, Hulette C, Saunders AM, et al. Apolipoprotein E is present
44 Robinson JL, Lee EB, Xie SX, et al. Neurodegenerative disease in hippocampal neurons without neurofibrillary tangles in
concomitant proteinopathies are prevalent, age-related and Alzheimer’s disease and in age-matched controls. Exp Neurol 1994;
APOE4-associated. Brain 2018; 141: 2181–93. 128: 13–26.
45 Yang H-S, Yu L, White CC, et al. Evaluation of TDP-43 68 Blanchard JW, Bula M, Davila-Velderrain J, et al. Reconstruction of
proteinopathy and hippocampal sclerosis in relation to APOE ε4 the human blood-brain barrier in vitro reveals a pathogenic
haplotype status: a community-based cohort study. Lancet Neurol mechanism of APOE4 in pericytes. Nat Med 2020; 26: 952–63.
2018; 17: 773–81. 69 Holtzman DM, Fagan AM, Mackey B, et al. Apolipoprotein E
46 Sabir MS, Blauwendraat C, Ahmed S, et al. Assessment of APOE facilitates neuritic and cerebrovascular plaque formation in an
in atypical parkinsonism syndromes. Neurobiol Dis 2019; Alzheimer’s disease model. Ann Neurol 2000; 47: 739–47.
127: 142–46. 70 Kim J, Jiang H, Park S, et al. Haploinsufficiency of human APOE
47 Montagne A, Nation DA, Sagare AP, et al. APOE4 leads to blood- reduces amyloid deposition in a mouse model of amyloid-β
brain barrier dysfunction predicting cognitive decline. Nature 2020; amyloidosis. J Neurosci 2011; 31: 18007–12.
581: 71–76. 71 Deane R, Sagare A, Hamm K, et al. ApoE isoform-specific
48 Tsuang D, Leverenz JB, Lopez OL, et al. APOE ε4 increases risk for disruption of amyloid β peptide clearance from mouse brain.
dementia in pure synucleinopathies. JAMA Neurol 2013; 70: 223–28. J Clin Invest 2008; 118: 4002–13.
49 Dickson DW, Heckman MG, Murray ME, et al. APOE ε4 is 72 Ma Q, Zhao Z, Sagare AP, et al. Blood-brain barrier-associated
associated with severity of Lewy body pathology independent of pericytes internalize and clear aggregated amyloid-β42 by
Alzheimer pathology. Neurology 2018; 91: e1182–95. LRP1-dependent apolipoprotein E isoform-specific mechanism.
50 Prokopenko I, Miyakawa G, Zheng B, et al. Alzheimer’s disease Mol Neurodegener 2018; 13: 57.
pathology explains association between dementia with Lewy bodies 73 Koffie RM, Hashimoto T, Tai H-C, et al. Apolipoprotein E4 effects in
and APOE-ε4/TOMM40 long poly-T repeat allele variants. Alzheimer’s disease are mediated by synaptotoxic oligomeric
Alzheimers Dement (N Y) 2019; 5: 814–24. amyloid-β. Brain 2012; 135: 2155–68.
51 Koriath C, Lashley T, Taylor W, et al. ApoE4 lowers age at onset in 74 Kara E, Marks JD, Fan Z, et al. Isoform- and cell type-specific structure
patients with frontotemporal dementia and tauopathy independent of apolipoprotein E lipoparticles as revealed by a novel Forster
of amyloid-β copathology. Alzheimers Dement (Amst) 2019; resonance energy transfer assay. J Biol Chem 2017; 292: 14720–29.
11: 277–80. 75 Rauch JN, Luna G, Guzman E, et al. LRP1 is a master regulator of
52 Serrano-Pozo A, Betensky RA, Frosch MP, Hyman BT. Plaque- tau uptake and spread. Nature 2020; 580: 381–85.
associated local toxicity increases over the clinical course of 76 Xian X, Pohlkamp T, Durakoglugil MS, et al. Reversal of
Alzheimer disease. Am J Pathol 2016; 186: 375–84. ApoE4-induced recycling block as a novel prevention approach for
53 Mathys H, Davila-Velderrain J, Peng Z, et al. Single-cell Alzheimer’s disease. eLife 2018; 7: e40048.
transcriptomic analysis of Alzheimer’s disease. Nature 2019; 77 Zhao N, Ren Y, Yamazaki Y, et al. Alzheimer’s risk factors age,
570: 332–37. APOE genotype, and sex drive distinct molecular pathways.
54 Zhou Y, Song WM, Andhey PS, et al. Human and mouse Neuron 2020; 6: 727–42.
single-nucleus transcriptomics reveal TREM2-dependent and 78 Atagi Y, Liu C-C, Painter MM, et al. Apolipoprotein E is a ligand for
TREM2-independent cellular responses in Alzheimer’s disease. triggering receptor expressed on myeloid cells 2 (TREM2).
Nat Med 2020; 26: 131–42. J Biol Chem 2015; 290: 26043–50.
55 Grubman A, Chew G, Ouyang JF, et al. A single-cell atlas of 79 Parhizkar S, Arzberger T, Brendel M, et al. Loss of TREM2 function
entorhinal cortex from individuals with Alzheimer’s disease reveals increases amyloid seeding but reduces plaque-associated ApoE.
cell-type-specific gene expression regulation. Nat Neurosci 2019; Nat Neurosci 2019; 22: 191–204.
22: 2087–97. 80 Keren-Shaul H, Spinrad A, Weiner A, et al. A unique microglia type
56 Liu C-C, Zhao N, Fu Y, et al. ApoE4 accelerates early seeding of associated with restricting development of Alzheimer’s disease.
amyloid pathology. Neuron 2017; 96: 1024–1032.e3. Cell 2017; 169: 1276–1290.
57 Kim J, Eltorai AEM, Jiang H, et al. Anti-apoE immunotherapy 81 Krasemann S, Madore C, Cialic R, et al. The TREM2-APOE pathway
inhibits amyloid accumulation in a transgenic mouse model of drives the transcriptional phenotype of dysfunctional microglia in
Aβ amyloidosis. J Exp Med 2012; 209: 2149–56. neurodegenerative diseases. Immunity 2017; 47: 566–581.
58 Liao F, Hori Y, Hudry E, et al. Anti-ApoE antibody given after plaque 82 Zhao J, Davis MD, Martens YA, et al. APOE ε4/ε4 diminishes
onset decreases Aβ accumulation and improves brain function in a neurotrophic function of human iPSC-derived astrocytes.
mouse model of Aβ amyloidosis. J Neurosci 2014; 34: 7281–92. Hum Mol Genet 2017; 26: 2690–700.
59 Liao F, Li A, Xiong M, et al. Targeting of nonlipidated, aggregated 83 Chung W-S, Verghese PB, Chakraborty C, et al. Novel allele-
apoE with antibodies inhibits amyloid accumulation. J Clin Invest dependent role for APOE in controlling the rate of synapse pruning
2018; 128: 2144–55. by astrocytes. Proc Natl Acad Sci USA 2016; 113: 10186–91.
60 Lin Y-T, Seo J, Gao F, et al. APOE4 causes widespread molecular 84 Sperling R, Salloway S, Brooks DJ, et al. Amyloid-related imaging
and cellular alterations associated with Alzheimer’s disease abnormalities in patients with Alzheimer’s disease treated with
phenotypes in human iPSC-derived brain cell types. Neuron 2018; bapineuzumab: a retrospective analysis. Lancet Neurol 2012;
98: 1141–1154. 11: 241–49.

www.thelancet.com/neurology Vol 20 January 2021 79


Review

85 Pankiewicz JE, Baquero-Buitrago J, Sanchez S, et al. APOE 97 Champagne D, Pearson D, Dea D, Rochford J, Poirier J.
genotype differentially modulates effects of anti-Aβ, passive The cholesterol-lowering drug probucol increases apolipoprotein E
immunization in APP transgenic mice. Mol Neurodegener 2017; production in the hippocampus of aged rats: implications for
12: 12. Alzheimer’s disease. Neuroscience 2003; 121: 99–110.
86 Sakai K, Boche D, Carare R, et al. Aβ immunotherapy for 98 Santos DB, Peres KC, Ribeiro RP, et al. Probucol, a lipid-lowering
Alzheimer’s disease: effects on apoE and cerebral vasculopathy. drug, prevents cognitive and hippocampal synaptic impairments
Acta Neuropathol 2014; 128: 777–89. induced by amyloid β peptide in mice. Exp Neurol 2012; 233: 767–75.
87 Sagare AP, Bell RD, Zhao Z, et al. Pericyte loss influences 99 Vitek MP, Christensen DJ, Wilcock D, et al. APOE-mimetic
Alzheimer-like neurodegeneration in mice. Nat Commun 2013; peptides reduce behavioral deficits, plaques and tangles in
4: 2932. Alzheimer’s disease transgenics. Neurodegener Dis 2012; 10: 122–26.
88 Nortley R, Korte N, Izquierdo P, et al. Amyloid β oligomers 100 Ghosal K, Stathopoulos A, Thomas D, Phenis D, Vitek MP,
constrict human capillaries in Alzheimer’s disease via signaling to Pimplikar SW. The apolipoprotein-E-mimetic COG112 protects
pericytes. Science 2019; 365: eaav9518. amyloid precursor protein intracellular domain-overexpressing
89 Yamazaki Y, Shinohara M, Yamazaki A, et al. ApoE animals from Alzheimer’s disease-like pathological features.
(Apolipoprotein E) in brain pericytes regulates endothelial function Neurodegener Dis 2013; 12: 51–58.
in an isoform-dependent manner by modulating basement 101 Krishnamurthy K, Cantillana V, Wang H, et al. ApoE mimetic
membrane components. Arterioscler Thromb Vasc Biol 2020; improves pathology and memory in a model of Alzheimer’s disease.
40: 128–44. Brain Res 2020; 1733: 146685.
90 Bell RD, Winkler EA, Singh I, et al. Apolipoprotein E controls 102 Pankiewicz JE, Guridi M, Kim J, et al. Blocking the apoE/Aβ
cerebrovascular integrity via cyclophilin A. Nature 2012; 485: 512–16. interaction ameliorates Aβ-related pathology in APOE ε2 and
91 Fitz NF, Cronican AA, Lefterov I, Koldamova R. Comment on ε4 targeted replacement Alzheimer model mice.
ApoE-directed therapeutics rapidly clear β-amyloid and reverse Acta Neuropathol Commun 2014; 2: 75.
deficits in AD mouse models. Science 2013; 340: 924. 103 Poirier J, Miron J, Picard C, et al. Apolipoprotein E and lipid
92 Muñoz-Cabrera JM, Sandoval-Hernández AG, Niño A, et al. homeostasis in the etiology and treatment of sporadic Alzheimer’s
Bexarotene therapy ameliorates behavioral deficits and induces disease. Neurobiol Aging 2014; 35 (suppl 2): S3–10.
functional and molecular changes in very-old Triple Transgenic 104 Mak ACY, Pullinger CR, Tang LF, et al. Effects of the absence of
Mice model of Alzheimer´s disease. PLoS One 2019; 14: e0223578. apolipoprotein E on lipoproteins, neurocognitive function, and
93 Tai LM, Koster KP, Luo J, et al. Amyloid-β pathology and APOE retinal function. JAMA Neurol 2014; 71: 1228–36.
genotype modulate retinoid X receptor agonist activity in vivo. 105 Kim J, Castellano JM, Jiang H, et al. Overexpression of low-density
J Biol Chem 2014; 289: 30238–55. lipoprotein receptor in the brain markedly inhibits amyloid
94 Price AR, Xu G, Siemienski ZB, et al. Comment on ApoE-directed deposition and increases extracellular A β clearance. Neuron 2009;
therapeutics rapidly clear β-amyloid and reverse deficits in AD 64: 632–44.
mouse models. Science 2013; 340: 924–d. 106 Pocovi M, Cenarro A, Civeira F, et al. Incomplete dominance of
95 Tesseur I, Lo AC, Roberfroid A, et al .Comment on ApoE-directed type III hyperlipoproteinemia is associated with the rare
therapeutics rapidly clear β-amyloid and reverse deficits in AD apolipoprotein E2 (Arg136-->Ser) variant in multigenerational
mouse models. Science 2013; 340: 924–d. pedigree studies. Atherosclerosis 1996; 122: 33–46.
96 Veeraraghavalu K, Zhang C, Miller S, et al. Comment on
ApoE-directed therapeutics rapidly clear β-amyloid and reverse © 2020 Elsevier Ltd. All rights reserved.
deficits in AD mouse models. Science 2013; 340: 924–d.

80 www.thelancet.com/neurology Vol 20 January 2021

You might also like