Open access peer-reviewed chapter

Passive Immunotherapy in Alzheimer’s Disease

Written By

Philip J. Dolan and Wagner Zago

Submitted: 06 November 2017 Reviewed: 07 March 2018 Published: 18 July 2018

DOI: 10.5772/intechopen.76299

From the Edited Volume

Alzheimer's Disease - The 21st Century Challenge

Edited by Jolanta Dorszewska and Wojciech Kozubski

Chapter metrics overview

1,246 Chapter Downloads

View Full Metrics

Abstract

The development of therapeutics for the treatment of Alzheimer’s disease (AD) has been challenged with a myriad of obstacles: an evolving and incomplete understanding of disease etiology and progression, challenges with early diagnosis, multifactorial genetic and environmental factors that contribute to patient variability, and the cost of conducting lengthy clinical trials. One approach that has garnered a significant amount of attention and resources for its potential as a disease modifying approach is passive immunotherapy directed at clearing amyloid-β (Aβ) species, a pathological hallmark of Alzheimer’s disease. While passive immunotherapeutic trials directed at Aβ have not yet demonstrated clinical benefit, they have prompted important advances in the application and understanding of biomarkers, patient selection, novel functional readouts, and safety monitoring. Application of these lessons has enabled more recent clinical trials to incorporate better trial designs and refine inclusion criteria to optimize patient population enrollment. In addition, new passive immunotherapy targets emerging in the clinic have emerged, as well as novel technologies to enhance future antibody therapeutics. Taken together, the advances in research and clinical science have prepared the passive immunotherapy field to advance emerging promising disease modifying treatments in AD.

Keywords

  • amyloid-β
  • tau
  • passive
  • immunotherapy
  • Alzheimer’s disease

1. Introduction

Alzheimer’s disease is a progressive neurodegenerative disease that clinically presents as a gradual onset of dementia, beginning with mild cognitive and functional deficits, leading eventually to an inability to carry out everyday tasks. Alzheimer’s disease and other dementias have a reported worldwide prevalence of approximately 42 million people, with an age-standardized rate of 761 per 100,000 [1]. Current therapeutics are limited to symptomatic approaches, such as acetylcholinesterase inhibitors and NMDA receptor (NMDAR) antagonists, which aim to enhance the function of unaffected neurocircuitry but do not target the underlying cause of the disease, thus there is a desperate need for approved disease modifying therapies.

Alzheimer’s disease is characterized by the dual pathological hallmarks of extracellular senile plaques and neurofibrillary tangles, composed of the amyloid-β (Aβ) peptide and tau protein, respectively. In addition, the primary familial forms of the disease are caused by mutations that directly affect Aβ homeostasis [2]. Due to both the pathological and genetic link to disease initiation, Aβ has been a prominent target for the development of disease-modifying therapeutics.

One such therapeutic approach is anti-Aβ immunotherapy. Active immunotherapy approaches utilize either the ability of the immune system to raise polyclonal antibodies against a therapeutic composed of an Aβ sequence-derived antigen and adjuvant, while passive immunotherapy approaches treat a patient with monoclonal antibodies with known antigen binding capabilities. While a large amount of research and development has been carried out regarding active immunotherapy towards AD targets [3], this chapter will focus on passive immunotherapy in AD, with the goal of describing what has been learned from past clinical studies, and what lessons may be applied to future efforts.

Advertisement

2. Aβ

2.1. Mechanisms of Aβ pathophysiology

The primary component of senile plaques is Aβ, a small peptide derived from the amyloid precursor protein (APP). In AD, Aβ is formed via sequential cleavage of APP by β-secretase [4] and the presenilin-1 (PS1) subunit of γ-secretase [5], respectively. This results in peptides of varying length, ranging from 38 to 43 amino acids [6], of which Aβ1–42 is the most amyloidogenic [7]. A central tenet in the understanding of causative factors of AD is the amyloid cascade hypothesis [8], which holds that the pathological increase of amyloidogenic Aβ in AD is a central initiating event in disease, that precedes and initiates a cascade of events that lead to other pathologies such as the formation of neurofibrillary tangles, inflammation, oxidative stress, neuronal dysfunction, and cell death [9]. While the amyloid cascade hypothesis has been challenged since first proposed [10, 11], there is abundant evidence from in vitro and in vivo studies confirming the significant role Aβ plays in inducing neurotoxicity, synaptic dysregulation, and pathology.

Degeneration of cultured neurons by treatment with aggregated forms of Aβ has been observed in multiple laboratories, and appears to correlate with extent of aggregation [7, 12]. Strong evidence indicates that soluble aggregated forms of Aβ might exert direct toxicity to neurons [13, 14, 15] through a variety of mechanisms, including (but not limited to) disruption of plasma membranes [16], dysregulation of mitochondrial function and dynamics via direct interaction [17], and excitotoxicity [18]. Confirming the centrality of Aβ’s role in neurotoxicity, myriad transgenic mouse models expressing mutant APP or APP/PS1 recapitulate many AD phenotypes, including plaque pathology, synaptic dysfunction, decreased cognition, neuroinflammation, and neuronal loss (reviewed in [19]).

One of the earliest mouse models of Aβ plaque deposition was the PDAPP mouse (Line109). These transgenic mice exhibit high human APP expression (>10-fold higher than endogenous levels), which is accompanied by extracellular Aβ plaque deposition, development of neuritic dystrophy, gliosis, and loss of synaptic and dendritic structures in the hippocampus [20]. The PDAPP mouse model was instrumental to demonstrate that therapies developed to clear Aβ deposits could potentially ameliorate functional deficits. Schenk and colleagues were the first to develop an active immunization approach using aggregated Aβ1–42 [21], which resulted in prevention of plaque formation in mice immunized before the development of pathology, and more importantly demonstrated that the induced polyclonal response can promote plaque clearance in aged PDAPP mice via phagocytosis by resident microglia. This breakthrough was later extended by administering the anti-N-terminal Aβ monoclonal antibody (mAb) 3D6 directly to PDAPP mice (passive immunotherapy); antibodies crossed the blood-brain barrier (BBB), localized to pathological features, and induced the opsonization and clearance of senile plaques in a microglia-dependent manner [22]. These preclinical findings validated Aβ-directed passive immunotherapy as a potential therapeutic strategy for AD.

2.2. Aβ passive immunotherapy in the clinic

The first Aβ immunotherapy clinical trial utilized active vaccination with Aβ1–42 (AN1792) and was halted during Phase IIa due to the appearance of meningoencephalitis, likely due to the infiltration of T-cells in the brain as a result of the presence of T-cell epitope(s) in the antigen, which contained the full-length Aβ1–42 peptide [23]. However, long-term follow-up indicated that patients that developed an immune response displayed modest but significant sparing of function, as assessed by the Disability Assessment for Dementia (DAD) and the Dependence scale [24]; in addition, autopsy of a patient immunized with AN1792 without meningoencephalitis displayed an absence of plaque pathology at autopsy and the presence of Aβ-reactive microglia, indicating that AN1792 was successful at engaging phagocytes to remove plaques [25].

Concerns for safety in active Aβ vaccination trials shifted most development efforts to passive immunotherapy, which carries less risk of an inflammatory response to drug. An overview of clinical Aβ antibody efforts described in the following text is listed in Table 1.

Name Epitope Most recent clinical phase References
First-generation Aβ passive immunotherapeutics
Bapineuzumab 1–6 PhIII (terminated) [22]
Solanezumab 16–26 PhIII [33, 41]
Ponezumab 35–40 PhIIa (terminated) [32, 42]
Second-generation Aβ passive immunotherapeutics
Crenezumab 16–26 (aggregate-selective) PhIII [41]
Gantenerumab 3–11, 18–27 PhIII [34]
BAN-2401 Protofibrils PhII [36]
Aducanumab N-terminus PhIII [37, 38]

Table 1.

Past and current Aβ antibody therapeutics.

2.2.1. First-generation Aβ passive immunotherapies

Bapineuzumab, directed at the N-terminus of Aβ, was the first monoclonal antibody therapy developed to target Aβ in AD. It was first tested in a phase I study in AD patients with single ascending doses ranging from 0.5 to 5 mg/kg administered every 13 weeks to evaluate safety, tolerability, and pharmacokinetics (PK) [26]. A significant safety finding of this study was the presence of vasogenic edema (VE) in the highest-dose cohort: 3/10 patients displayed these abnormalities, two of whom were asymptomatic. Due to the observation of VE at 5 mg/kg a dose regimen ranging from 0.15 to 2 mg/kg, administered every 13 weeks for 18 months was selected for the multiple ascending dose phase II trial [27]. In the phase II trial, study completers that received all 6 planned infusions displayed significant improvements in DAD score and the Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-cog), though this effect was not observed in the intent-to-treat population. VE was observed in ~10% of bapineuzumab treated patients (half of whom were asymptomatic), in comparison to 0% of the placebo group; the appearance of VE was dose-dependent and appeared early during the course of treatment. Interestingly, the majority (10/12) of VE cases occurred in carriers of the APOε4 allele, a risk factor for aggressive AD [28].

Two phase III trials for bapineuzumab were completed to evaluate efficacy in patients with mild to moderate AD who were either APOε4 carriers or non-carriers in separate trials, with a lower dose regimen in the carrier trial [29]. These trials did not meet the co-primary cognitive and functional endpoints, though CSF phospho-tau, a proposed biomarker of neurodegeneration in AD, did decrease in both studies and positron emission tomography-Pittsburgh B (PET-PIB) imaging revealed less amyloid pathology in the APOε4 carrier group treated with bapineuzumab compared to placebo. One important finding is that of the subgroup that underwent PET-PIB imaging, 6.5% of APOε4 carriers and 36.1% of non-carriers did not have detectable PET-PIB signal at trial entry, raising concerns about misdiagnosis and improper subject selection in the trials. While these studies did not succeed in meeting primary endpoints, they did provide information to guide future trials, particularly in understanding MRI abnormalities, such as VE and microhemorrhages.

During the course of the phase III trials, the observation that VE and microhemorrhages correlated with anti-amyloid dose levels was more pronounced in APOε4 carriers, and were normally transient and asymptomatic [30] led to the formation of an Alzheimer’s Association-led workgroup composed of industry and academic experts to advise the FDA on potential routes to monitor VE and microhemorrhages. The term amyloid-related imaging abnormalities (ARIA) was adopted to address the spectrum of MR imaging abnormalities observed with anti-amyloid therapies, spanning from sulcal effusion and vasogenic edema seen on FLAIR MRIs to hypointensities (hemosiderin deposits) on T2* MRI. The ARIA terminology was further subdivided to ARIA-E (sulcal effusion and edema) and ARIA-H (hemosiderin deposits) [31]. Recommendations from the workgroup included (a) standardization of technical and monitoring practices for MRI, (b) exclusion from trials of patients with preexisting ARIA-H, and (c) monitoring of symptoms potentially associated with ARIA. The adoption of these standards, and the understanding that ARIA is largely a short-lived treatment related effect inherent to many anti-amyloid therapies, opened the possibility of testing higher and more frequent drug administration regimens with appropriate patient safety monitoring.

In parallel with bapineuzumab, two additional anti-Aβ passive immunotherapies underwent contemporaneous clinical trials: Ponezumab, directed at the C-terminus of Aβ, underwent Phase I and IIa trials, but was discontinued after Phase IIa [32]. Solanezumab, directed at an internal epitope of Aβ and hypothesized to function by binding soluble species in the CNS and periphery, failed a phase III trial in mild AD patients [33], and a trial conducted in prodromal patients was discontinued. However, it is currently being tested in genetically-defined Alzheimer’s disease populations, with results expected in 2021 (clinicaltrials.gov; Identifier: NCT02008357).

2.2.2. Second-generation Aβ passive immunotherapies

Whereas the first generation of Aβ therapeutic mAbs differed in binding to distinct antibody domains (N-, mid-, and C-terminus), the second generation are intended to primarily bind specific conformations and aggregation states. Gantenerumab, currently in two phase III trials for mild and prodromal AD, binds a discontinuous epitope consisting of the N-terminus and an internal epitope, implying a unique conformational binding specificity (clinicaltrials.gov; Identifiers: NCT01224106, NCT02051608) [34]. Crenezumab, currently in phase II and phase III trials for autosomal dominant AD and prodromal-to-mild AD, respectively, is reported to selectively bind soluble and insoluble aggregates, but not monomers (clinicaltrials.gov; Identifiers: NCT01998841, NCT03114657) [35]. In contrast to other therapeutic mAbs, crenezumab is engineered on an IgG4 backbone to reduce effector function, and microglial-mediated phagocytosis of Aβ deposits is not anticipated. BAN-2401, is in clinical development in a large phase II study in early AD patients; is proposed to selectively bind Aβ protofibrils (clinicaltrials.gov; Identifier: NCT01767311) [36].

A promising antibody candidate from this group that is currently in the clinic is aducanumab. Aducanumab is a human mAb that selectively targets soluble aggregates and fibrils, and binds the N-terminus of Aβ. Preclinical studies demonstrate that the chimeric form of aducanumab peripherally administered to an APP transgenic mouse (a) crosses the BBB and binds to plaques (b) reduces calcium overload in neurons [37], and (c) reduces plaque burden in a dose-dependent manner [38]. An interim report from a double-blind, placebo controlled phase Ib study revealed a dose-dependent decrease of amyloid PET signal that corresponded with significant slowing of cognitive decline at 52 weeks at the highest dose level, 10 mg/kg [38]. While ARIA was reported at a similar frequency compared with previous trials, adherence to guidelines formalized by the Alzheimer’s Association ARIA working group [31] allowed for higher and more frequent dosing, potentially contributing to the positive results seen in these early studies. Aducanumab is currently in phase III trials in prodromal early AD patients, with endpoints and patient populations informed by the successful phase Ib study [39]. Interestingly, enrollment for these phase III clinical trials was recently increased by approximately 15% due to patient variability in the primary functional endpoint [40].

Advertisement

3. Tau

While most passive immunotherapy clinical trials in AD have been directed at Aβ, key discoveries regarding tau function and contribution to disease mechanisms have prompted significant efforts directed towards tau. Hyperphosphorylated and aggregated tau protein are the main component of neurofibrillary tangles (NFTs), which, together with Abeta plaques, are considered a primary hallmark in Alzheimer’s disease. Because of its intracellular localization, tau deposits have historically been thought to be unavailable to immunotherapeutic treatments. However, results outlined in this section indicate the potential for targeting tau through a passive immunotherapeutic approach.

3.1. Tau biology and pathophysiology

Since the discovery that NFTs are composed of the microtubule-associated protein tau [43, 44, 45], many efforts have been devoted to elucidating molecular mechanisms of tau pathophysiology. Tau is an intracellular microtubule binding protein, which is involved in the regulation of microtubule stability and dynamics. In the brain, tau exists principally as six different isoforms, which vary in the absence or presence of N-terminal acidic repeats and a microtubule repeat; these differences are due to the splicing in or out of exons 2, 3, and 10 [46]. In normal physiological situations, the specific ratio of tau isoforms is developmentally regulated, likely due to the changing needs of microtubule fluidity versus stability throughout development and maturity [47].

Tau is an intrinsically disordered, natively-unfolded protein [48] whose physiological function is tightly regulated by post-translational modifications—principally via phosphorylation, which regulates microtubule binding affinity [49, 50]. In the AD brain, tau aggregates to form hyperphosphorylated NFTs and inclusions, composed of paired-helical and straight filaments [51]. In contrast to the intrinsically disordered nature of monomeric tau in solution, these structures adopt an ordered structure composed of a β-sheet core comprised of central residues, surrounded by a disordered coat comprised of the C- and N-termini of the molecule [52]. In AD, the appearance of tau pathological features positively correlates with dementia and disease progression [53, 54], leading to the hypothesis that the formation of tau pathology is a primary causative agent in the development of AD.

While the stereotypic appearance and progression of tau pathology down the perforant pathway—the neurocircuit from the entorhinal cortex to the hippocampus—has been described [55, 56], the molecular mechanisms underpinning this observation had remained elusive. Neurons in the performant path have long been known to be selectively vulnerable to insult such as hyperactivity [57] and expression of AD-related presenilin mutations [58], but the discovery that, when injected into the brain parenchyma, tau from a mutant mouse could simulate the formation of tau aggregates in a previously healthy animal [59] allowed the possibility that this progression may be mediated by aggregated and misfolded forms of the protein. This was strikingly confirmed in mice with tau expression restricted to the entorhinal cortex: in these mice, tau pathology propagated from the region of expression to distant efferent neurons [60, 61], demonstrating that direct cell-cell contact was not required for propagation, and that the pathological signal could be spread trans-synaptically. The demonstration that tau itself was present in interstitial fluid [62], could be secreted from neurons [63], and passed between cells [64] and neurons [65] provided evidence that tau species themselves could be directly transmitted between neurons in vivo, providing a potential mechanistic basis for the propagation of tau pathology. Although tau and Aβ are likely associated with different pathophysiological processes in Alzheimer’s disease, the presence of pathogenic extracellular tau species could theoretically also be targeted by immunotherapeutic approaches, in this case by a different mechanism of action: interception/sequestration and prevention of cell-to-cell transmission.

3.2. Tau passive immunotherapy

An overview of preclinical and clinical tau antibody efforts described in the following text is listed in Table 2.

Name Epitope Most recent development phase References
MC1 7–9, 313–322 Preclinical [69, 70]
BIIB092/BMS986168 17–28 PhII (recruiting) [85, 88]
ABBV-8E12 25–30 PhI open label extension [81, 82]
Cis mAb Cis-pT231 Preclinical [74]
RO7105705 pSer409 PhII (recruiting) [71]
PHF1 pSer396/404 Preclinical [67, 69, 70]
TOMA Tau oligomer Preclinical [76]

Table 2.

Tau clinical and preclinical antibodies discussed in this chapter.

Pioneering tau immunotherapy studies demonstrated that immunization with phospho-tau peptides (phosphorylated at Ser396/404) in two different tau transgenic lines raised anti-tau antibodies, which immunohistochemically stained the brains of P301L-tau transgenic mice. In addition, active immunization resulted in reductions in tau pathology. The mice also displayed improved performance in motor tasks [66, 67]. Purified anti-tau antibodies from immunized mice were peripherally injected into naïve transgenic mice and localized to neurons in the brain displaying tau pathology, demonstrating their ability to cross the blood-brain barrier (BBB) and localize to their target. In a separate study performed by the same lab, passive administration of the mAb PHF1, directed at the Ser396/404 phosphoepitope, also resulted in reductions in tau pathology in mice compared to isotype control [68]. The findings from this series of studies were proposed to be due to two potential mechanisms: (a) antibody-mediated clearance of extracellular tau deposits and (b) intracellular uptake of tau antibodies. The efficacy of passive immunotherapy using PHF1, as well as the conformational antibody MC1, were also confirmed in independent labs [69, 70], bolstering early evidence of this novel promising therapeutic avenue.

An antibody targeting a different phosphoepitope, pSer409, also shows promise in preclinical models; however, conclusions regarding the mechanism of antibody function were considerably different than those proposed in the initial active and passive studies described in the prior paragraph. In this study, a highly selective mAb was able to bind tau phosphorylated at Ser409 and specifically bind AD brain tissue. The mAb was shown to neutralize oligomer-induced neurotoxicity; however, the neutralization activity of the antibody was reduced in mixed neuron-microglial cultures. Antibody engineered with reduced effector function (REF) maintained neutralization activity in mixed neuron-microglial cultures, while the wild-type anti-pSer409 antibody did not prevent neurotoxicity and in fact promoted the release of pro-inflammatory cytokines from microglia [71]. Both wild-type and REF variants of the antibody prevented the progression of tau pathology in the tau P301L mouse, leading the authors to conclude that phagocytic clearance of tau structures was not a contributing mechanism of action to efficacy in the transgenic mouse model. In addition, the lack of FcR message found in isolated neurons prompted the conclusion that receptor-mediated uptake did not occur. The antibody examined in this report has been developed into a therapeutic candidate, which is currently in clinical development (clinicaltrials.gov; Identifier: NCT03289143).

Additional studies have been conducted to identify and target post-translationally modified forms of tau to explore effects of antibody treatment. One compelling approach targets a unique structural isoform of tau induced by phosphorylation of tau at T231. Phosphorylation of tau at T231 occurs during disease progression; the prolyl isomerase Pin1 normally binds and converts the pT231/Proline motif from a toxic cis form to a soluble nontoxic trans form [72]. A mAb targeting cis but not trans pT231-tau detects pathology during mild cognitive impairment (MCI) [73]. In addition to AD, this post-translational signature (as well as others) appears in the brains of traumatic brain injury (TBI) patients. When administered peripherally in a murine TBI model carried out in tau transgenic mice, the cis-pT231 tau antibody prevented the spread of tauopathy and cortical LTP deficits, and improved performance in the elevated plus maze, which was correlated to TBI-induced disinhibition behavior in patients [74]. Another effort targeting disease-specific forms of tau is centered around developing antibodies that bind soluble oligomeric tau—hypothesized to be the most toxic form of the molecule [75]—and have minimal binding to monomeric or mature NFTs [76]. Tau oligomer-specific monoclonal antibodies (TOMAs) were dosed via intracerebroventricular (i.c.v.) infusion to tau P301L mice. Strikingly, a single i.c.v. injection reduced tau oligomers and histopathology, and rescued deficits in rotarod and spontaneous alternation tests. Examination of serum revealed oligomeric tau and antibody/antigen complexes, suggesting peripheral clearance as a mechanism of action [77].

Informed by studies indicating the potential for the propagation of tau pathology across cell membranes [64], as well as the demonstration of trans-synaptic transmission in vivo [60, 78], an independent effort to discover tau antibodies that interrupted cell-to-cell transmission yielded phosphorylation-independent antibodies that blocked uptake of tau aggregates to cultured cells [79, 80]. When administered to tau transgenic mice centrally via an Alzet minipump, these antibodies slowed the advance of tau pathology, as measured by immunohistochemical and biochemical means [79]. One of the efficacious antibodies used in this report, HJ8.5, was used in a peripheral administration model to further explore its potential as a therapeutic agent [81]. HJ8.5 is a high affinity anti-N terminal mAb that recognizes residues 25–30, which are present on all splice isoforms of tau. In this study, P301S tau transgenic mice were dosed intraperitoneally over a 3-month period with 10 or 50 mg/kg of HJ8.5. The high dose cohort displayed decreases of insoluble tau, AT8 staining, and thioflavin S staining. In addition, this cohort exhibited improvements in sensorimotor function compared to isotype control and low-dose cohorts. The preclinical efficacy profile, as well as the concordance of in vivo data with mechanistic in vitro studies, propelled the humanized analogue of this antibody into the clinic (clinicaltrials.gov; Identifier: NCT03391765) [82]. Interestingly, a separate effort focused on discovering antibodies and epitopes important for uptake and transmission determined that while N-terminal antibodies could indeed block uptake of recombinant and AD patient-derived tau, there were other epitopes with potentially more potent function, notably antibodies binding C-terminal to the acidic inserts [83].

A key component of the amyloid cascade hypothesis is that Aβ aggregation induces, either indirectly or directly, fibrillization of tau as well as other disease processes (reviewed in [84]). The finding that extracellular secreted and truncated forms of tau (termed eTau) could regulate Aβ levels demonstrated a potential upstream role of tau in relation to Aβ, complementary to the amyloid cascade hypothesis. In this study, secreted eTau was isolated from iPSC neurons derived from patients with AD; treatment of neurons with eTau displayed increases in secreted Aβ, and these increases could be prevented via application of eTau-binding antibodies such as MC1 and IPN002, which recognizes residues 17–28. Aβ levels were not affected by PHF1 antibody, as the PHF1 epitope is not present in eTau. This finding was recapitulated in transgenic P301L-tau mice; peripheral treatment with IPN002 resulted in reductions in Aβ in the interstitial fluid and cortical tissue [85]. These findings were recently confirmed by a different group using mAbs that target very similar N-terminal tau epitopes; in these studies, behavioral improvements as well as decreases in Aβ were noted in mice transgenic for mutant forms of presenilin, APP, and tau [86, 87]. IPN002 has been developed into a clinical therapeutic and is undergoing clinical trials as BIIB-092/BMS986168 (clinicaltrials.gov; Identifier: NCT03068468) [88].

Though the success of preclinical studies with tau antibodies has provided sufficient rationale to begin exploration in the clinic, a greater understanding of the full range of factors involved in tau toxicity and the mechanisms of action of tau passive immunotherapy are needed. These mechanisms may be different than those proposed for Aβ immunotherapy. There remain conflicting details from the studies presented here, such as the relative contribution of microglial-mediated phagocytosis, the relative importance of eTau-mediated Aβ production, the extent of trans-synaptic transmission in transgenic mice with widespread expression in the brain, and the optimal epitope to target. Gaining a clearer understanding of these factors continues are a current research focus.

Clinical trials with Aβ immunotherapies have demonstrated the importance of proper clinical diagnosis, patient selection, sensitive cognition tests, and effective biomarkers to monitor efficacy and disease progression. Though some general commonalities may exist in the clinical design of Aβ and tau passive immunotherapy trials, there are substantial differences in the targets and any potential clinical development approaches. In contrast to Aβ, there are a number of non-AD tauopathies such as progressive supranuclear palsy (PSP) [89] and frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) [90] that may provide alternative clinical development pathways to test novel tau-directed therapeutic approaches. In contrast to AD, these diseases present pathological signatures composed almost uniformly of tau and neurofibrillary tangles; in addition, FTDP-17 is an autosomal dominant disorder, genetically validating the causative role of tau. Diagnosis of these and other tauopathies have historically been made solely based on clinicopathology; due to the difficulty of diagnosis from to the overlap of symptomologies with other neurodegenerative disorders, as well as the lack of clear biomarkers, diagnosis is only confirmed at autopsy [91]. Modern tau PET imaging agents are currently under clinical investigation [92]; while early generations of tau PET tracers displayed nonspecificity and suboptimal binding and PK characteristics, the newest class of tracers display improved specificity, PK properties, and may allow for improved diagnosis in tauopathies as well as an ability to monitor tau pathology in AD clinical trials [93].

Advertisement

4. New targets and technologies

4.1. Targeting the immune system in AD

The vast majority of passive immunotherapeutic approaches in AD have targeted Aβ and tau; this is a natural outcome of the primacy of these proteins as the principal pathological hallmarks of the disease. The association of mutations of APP (and proteins that modulate its generation, such as presenilin-1) to familial AD, and the high degree of correlation between tau pathological development and cognition, strengthen the validity of these two proteins as important causative disease agents. However, new approaches, primarily targeting immunomodulatory proteins, are also currently under development.

The presence of neuroinflammatory processes and signatures in AD has been well established, but the exact role they play in disease etiology, or whether neuroinflammation has a primarily protective or harmful role, has not been clear (reviewed in [94]). Studies examining the complement cascade have helped to understand this duality. The synaptic pruning activity carried out by microglia is regulated by complement [95]. The initiating protein of the classical complement cascade, C1q, is enriched in the developing mouse CNS and localizes to synapses; genetic ablation of this protein results in misregulated innervation due to increased presence of synapses [96]. While C1q is normally downregulated after development, it is elevated in normal aging [97] and disease, including AD [98]. In a transgenic APP mouse, C1q localizes to synapses, and is required for pathological synapse loss. Treatment of C1q knockout mice with oligomeric Aβ displayed no synaptic loss, indicating that C1q is a required mediator of Aβ-induced toxicity. Interestingly, an anti-C1q antibody rescued Aβ-induced synaptotoxicity in vivo, and LTP impairment in situ, when compared to isotype control [99]. These data hinted at the promise of C1q immunotherapy to provide protective benefits by neutralizing a key mediator of Aβ-induced microglial overactivation, which results in synaptic loss. The anti-C1q antibody used in this study has been developed into a human therapeutic, and is beginning clinical trials (clinicaltrials.gov; Identifier: NCT03010046) [100].

The mounting evidence of involvement of the adaptive immune system in restraining the advance of AD pathology has opened the possibility of directing passive immunotherapies to the periphery, which considerably eases the challenge of achieving sufficient drug exposure in the CNS to affect pathology. Microglia resident in the brain are known to be recruited to sites of injury such as senile plaques, but the finding that peripherally-derived bone marrow stem cells are able to enter the CNS, and differentiate into microglia [101, 102], was the first direct evidence that repopulation and recruitment of microglia from the periphery was an active process. This finding was extended to AD mouse models with the finding that peripherally-recruited microglia are mobilized by Aβ, recruited to the site of senile plaques, and are able to clear plaques via phagocytosis [103]. The protective role of these immune cells in the presence of AD-like pathology was confirmed with the observation that (a) knocking out the chemokine receptor CCR2 in an APP-transgenic mouse resulted in decreased recruitment of monocytes to Aβ plaques [104], and (b) the specific ablation of bone-marrow derived cells via diphtheria-toxin receptor expression resulted in increased Aβ plaques [105]. Furthermore, increasing trafficking of macrophages by inhibiting the normally immunosuppressive regulatory T-cells through pharmacologic or genetic methods results in reduced Aβ pathology [106].

Elucidation of the biology of inhibitory signaling pathways and proteins such as Programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), known as immune checkpoints, led to the development of antibody therapeutics for use in cancer (reviewed in [107]). These therapies function by neutralizing immune checkpoints and activating T-cells, which prompts antitumor activity. The characterization of checkpoint signaling pathways, along with the findings that peripheral immune cells modulate AD-like pathology in a regulatory T-cell (Treg)-dependent manner, has prompted examination of the PD1/PD-L1 axis in AD. In a recent study, AD transgenic mice were treated with an anti-PD1 antibody to blockade the PD1/PD-L1 axis. Remarkably, checkpoint blockade in this model resulted in substantial rescue of performance in a behavioral assay of memory and cognition after a single dose, and mice exhibited decreases in Aβ pathology with only two dose administrations [108]. The effect on pathology was observed even in mice with profound amyloid burden. While the findings of a profound effect on functional measures after such a short dose regimen are very exciting, they should be taken with a note of caution. A follow-up study, carried out by three pharmaceutical companies using three transgenic models and numerous PD-L1 antibodies was attempted to recapitulate these results. Despite peripheral immune activation, in all instances neither reductions in Aβ pathology nor infiltration of peripheral monocytes were detected [109]. Further studies are needed to elucidate the potential of checkpoint modulation.

4.2. Increasing blood-brain barrier (BBB) penetrance for passive immunotherapeutics

A significant barrier in the development of passive immunotherapeutics for AD is the low percentage of circulating antibody that crosses the BBB. Animal studies have indicated that ~0.1–0.5% of IgG enters the CSF from the periphery [110, 111], which is borne out by preclinical [112] and clinical [113, 114] data obtained with antibodies tested for use in AD. This has led to trials with increasing amounts of antibody administered to patients ([82]; clinicaltrials.gov, Identifier NCT03318523) with the hope of delivering sufficient amounts of antibody to the CNS to achieve a clinical effect. There are, however, indications that concentrations of antibodies are higher in brain parenchyma than what is present in CSF. The chimeric form of aducanumab reported brain:plasma AUC ratios when tested in a transgenic APP model of 1.3% [38]. This is in agreement with the finding that the concentration of protein analyte present in the interstitial fluid is approximately 10-fold higher than in the ISF [62, 115]. This could be due to the rapid turnover of CSF volume [116] compared to ISF, longer elimination times of antibodies in brain parenchyma compared with CSF, or increased residence time due to target-mediated binding. Nevertheless, methods and technologies to increase BBB penetrance of biomolecules urgently need to be applied to antibodies and other proteins.

One of the more promising approaches to increase penetrance of protein therapeutics into the brain utilize endogenous receptors that transcytose between the brain and periphery, such as transferrin receptor (TfR) [117], insulin receptor [118], and LDL receptor-related protein 1 (LRP1) [119]. Protein engineering approaches feature fusion of the therapeutic molecule to proteins, ligands, or peptides that bind these receptors and facilitate transcytosis across the BBB (reviewed in [120]). One of the best understood receptor-mediated delivery systems is the use of TfR, though a similar path has been taken in the development of technologies that utilize insulin receptor. Increased brain uptake of transferrin/antibody fusion proteins were detected in rats [121], though the relatively large size (~80 kDa) of full-length transferrin make this impractical for biotherapeutic use. The detection of increased transcytosis of anti-TfR antibodies and antibody fragments [122, 123], and later advances in antibody generation technologies, enabled bispecific antibodies that bind TfR as well as target [124]. As understanding of the transcytotic properties of TfR binding moieties have increased, so has the understanding of how best to incorporate properties to ensure delivery to the brain. For example, reducing TfR affinity improves delivery, as a low affinity anti-TfR moiety will release from the receptor faster than a high affinity moiety [124]. As receptor-binding fusions enter the clinic, further questions regarding safety and distribution changes brought about by higher CNS concentrations will need to be continually addressed [125, 126]. Work continues to identify receptors that may be useful for increasing BBB concentrations of antibodies to allow engagement with wider range of drug targets [127, 128].

Advertisement

5. Conclusions and future perspectives

AD provides a monumentally challenging drug development landscape. The uncertainty about disease etiology, variability in patient genetics and disease progression, and difficulties in early diagnosis are all but a noncomprehensive list of hurdles to developing effective drugs. Though development of therapeutics to slow or halt AD disease progression, including passive immunotherapeutics, have not yet yielded clinical benefit, the prospect of applying lessons learned in the clinic towards validated targets such as Aβ and tau provides optimism for future success. In addition, our understanding of the mechanisms of other principal contributing factors to disease progression will provide a variety of new targets to explore. Combined with advances in drug technology to increase the availability of biomolecules in the CNS, these clinical and biological advances offer great promise around future success in treating AD.

Advertisement

Acknowledgments

There is a wealth of excellent studies in the areas of study covered by this chapter—unfortunately, we were unable to list them all. We thank the many researchers not recognized here who have contributed greatly to the field. We would also like to thank the thoughtful review and comments provided by Enchi Liu and Ellen Rose.

Advertisement

Conflict of interest

PJD and WZ are employees of Prothena Biosciences.

References

  1. 1. Group GBDNDC. Global, regional, and national burden of neurological disorders during 1990-2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurology. 2017;16(11):877-897
  2. 2. Wu L, Rosa-Neto P, Hsiung GY, Sadovnick AD, Masellis M, Black SE, et al. Early-onset familial Alzheimer’s disease (EOFAD). The Canadian Journal of Neurological Sciences. 2012;39(4):436-445
  3. 3. Pasquier F, Sadowsky C, Holstein A, Leterme Gle P, Peng Y, Jackson N, et al. Two phase 2 multiple ascending-dose studies of Vanutide Cridificar (ACC-001) and QS-21 adjuvant in mild-to-moderate Alzheimer’s disease. Journal of Alzheimer’s Disease. 2016;51(4):1131-1143
  4. 4. Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, et al. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286(5440):735-741
  5. 5. Wolfe MS, Xia W, Ostaszewski BL, Diehl TS, Kimberly WT, Selkoe DJ. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature. 1999;398(6727):513-517
  6. 6. Wiltfang J, Esselmann H, Bibl M, Smirnov A, Otto M, Paul S, et al. Highly conserved and disease-specific patterns of carboxyterminally truncated Abeta peptides 1-37/38/39 in addition to 1-40/42 in Alzheimer’s disease and in patients with chronic neuroinflammation. Journal of Neurochemistry. 2002;81(3):481-496
  7. 7. Pike CJ, Burdick D, Walencewicz AJ, Glabe CG, Cotman CW. Neurodegeneration induced by beta-amyloid peptides in vitro: The role of peptide assembly state. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 1993;13(4):1676-1687
  8. 8. Hardy JA, Higgins GA. Alzheimer’s disease: The amyloid cascade hypothesis. Science. 1992;256(5054):184-185
  9. 9. Butterfield DA, Griffin S, Munch G, Pasinetti GM. Amyloid beta-peptide and amyloid pathology are central to the oxidative stress and inflammatory cascades under which Alzheimer’s disease brain exists. Journal of Alzheimer’s Disease. 2002;4(3):193-201
  10. 10. Armstrong RA. A critical analysis of the ‘amyloid cascade hypothesis’. Folia Neuropathologica. 2014;52(3):211-225
  11. 11. Hardy J. The amyloid hypothesis for Alzheimer’s disease: A critical reappraisal. Journal of Neurochemistry. 2009;110(4):1129-1134
  12. 12. Nguyen L, Wright S, Lee M, Ren Z, Sauer JM, Hoffman W, et al. Quantifying amyloid beta (Abeta)-mediated changes in neuronal morphology in primary cultures: Implications for phenotypic screening. Journal of Biomolecular Screening. 2012;17(6):835-842
  13. 13. Demuro A, Mina E, Kayed R, Milton SC, Parker I, Glabe CG. Calcium dysregulation and membrane disruption as a ubiquitous neurotoxic mechanism of soluble amyloid oligomers. The Journal of Biological Chemistry. 2005;280(17):17294-17300
  14. 14. Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300(5618):486-489
  15. 15. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(11):6448-6453
  16. 16. McLaurin J, Chakrabartty A. Membrane disruption by Alzheimer beta-amyloid peptides mediated through specific binding to either phospholipids or gangliosides. Implications for neurotoxicity. The Journal of Biological Chemistry. 1996;271(43):26482-26489
  17. 17. Devi L, Prabhu BM, Galati DF, Avadhani NG, Anandatheerthavarada HK. Accumulation of amyloid precursor protein in the mitochondrial import channels of human Alzheimer’s disease brain is associated with mitochondrial dysfunction. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2006;26(35):9057-9068
  18. 18. Harkany T, Abraham I, Timmerman W, Laskay G, Toth B, Sasvari M, et al. Beta-amyloid neurotoxicity is mediated by a glutamate-triggered excitotoxic cascade in rat nucleus basalis. The European Journal of Neuroscience. 2000;12(8):2735-2745
  19. 19. Sasaguri H, Nilsson P, Hashimoto S, Nagata K, Saito T, De Strooper B, et al. APP mouse models for Alzheimer’s disease preclinical studies. The EMBO Journal. 2017;36(17):2473-2487
  20. 20. Schenk D, Games KD, McConlogue L. The potential utility of transgenic mice harboring beta-amyloid precursor protein. Neurobiology of Aging. 1995;16(4):711-713 discussion 5-8
  21. 21. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999;400(6740):173-177
  22. 22. Bard F, Cannon C, Barbour R, Burke RL, Games D, Grajeda H, et al. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nature Medicine. 2000;6(8):916-919
  23. 23. Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology. 2003;61(1):46-54
  24. 24. Vellas B, Black R, Thal LJ, Fox NC, Daniels M, McLennan G, et al. Long-term follow-up of patients immunized with AN1792: Reduced functional decline in antibody responders. Current Alzheimer Research. 2009;6(2):144-151
  25. 25. Masliah E, Hansen L, Adame A, Crews L, Bard F, Lee C, et al. Abeta vaccination effects on plaque pathology in the absence of encephalitis in Alzheimer disease. Neurology. 2005;64(1):129-131
  26. 26. Black RS, Sperling RA, Safirstein B, Motter RN, Pallay A, Nichols A, et al. A single ascending dose study of bapineuzumab in patients with Alzheimer disease. Alzheimer Disease and Associated Disorders. 2010;24(2):198-203
  27. 27. Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, et al. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73(24):2061-2070
  28. 28. Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurology. 2011;10(3):241-252
  29. 29. Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, Raskind M, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. The New England Journal of Medicine. 2014;370(4):322-333
  30. 30. Sperling R, Salloway S, Brooks DJ, Tampieri D, Barakos J, Fox NC, et al. Amyloid-related imaging abnormalities in patients with Alzheimer’s disease treated with bapineuzumab: A retrospective analysis. Lancet Neurology. 2012;11(3):241-249
  31. 31. Sperling RA, Jack CR Jr, Black SE, Frosch MP, Greenberg SM, Hyman BT, et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: Recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association. 2011;7(4):367-385
  32. 32. Landen JW, Andreasen N, Cronenberger CL, Schwartz PF, Borjesson-Hanson A, Ostlund H, et al. Ponezumab in mild-to-moderate Alzheimer’s disease: Randomized phase II PET-PIB study. Alzheimers Dement (N Y). 2017;3(3):393-401
  33. 33. Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, et al. Trial of Solanezumab for mild dementia due to Alzheimer’s disease. The New England Journal of Medicine. 2018;378(4):321-330
  34. 34. Bohrmann B, Baumann K, Benz J, Gerber F, Huber W, Knoflach F, et al. Gantenerumab: A novel human anti-Abeta antibody demonstrates sustained cerebral amyloid-beta binding and elicits cell-mediated removal of human amyloid-beta. Journal of Alzheimer’s Disease. 2012;28(1):49-69
  35. 35. Adolfsson O, Pihlgren M, Toni N, Varisco Y, Buccarello AL, Antoniello K, et al. An effector-reduced anti-beta-amyloid (Abeta) antibody with unique abeta binding properties promotes neuroprotection and glial engulfment of Abeta. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2012;32(28):9677-9689
  36. 36. Tucker S, Moller C, Tegerstedt K, Lord A, Laudon H, Sjodahl J, et al. The murine version of BAN2401 (mAb158) selectively reduces amyloid-beta protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. Journal of Alzheimer’s Disease. 2015;43(2):575-588
  37. 37. Kastanenka KV, Bussiere T, Shakerdge N, Qian F, Weinreb PH, Rhodes K, et al. Immunotherapy with Aducanumab restores calcium homeostasis in Tg2576 mice. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2016;36(50):12549-12558
  38. 38. Sevigny J, Chiao P, Bussiere T, Weinreb PH, Williams L, Maier M, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature. 2016;537(7618):50-56
  39. 39. Budd Haeberlein S, O’Gorman J, Chiao P, Bussiere T, von Rosenstiel P, Tian Y, et al. Clinical development of aducanumab, an anti-abeta human monoclonal antibody being investigated for the treatment of early Alzheimer’s disease. The Journal of Prevention of Alzheimer’s Disease. 2017;4(4):255-263
  40. 40. Pagliarulo N. Biogen Shared Fall after Unexpected ‘Variability’ in Alzheimer’s Studies. 2018. Available from: https://www.biopharmadive.com/news/biogen-shares-fall-after-unexpected-variability-in-alzheimers-studies/517193/2018 [Accessed 2018-03-04]
  41. 41. Crespi GA, Hermans SJ, Parker MW, Miles LA. Molecular basis for mid-region amyloid-beta capture by leading Alzheimer’s disease immunotherapies. Scientific Reports. 2015;5:9649
  42. 42. La Porte SL, Bollini SS, Lanz TA, Abdiche YN, Rusnak AS, Ho WH, et al. Structural basis of C-terminal beta-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer’s disease. Journal of Molecular Biology. 2012;421(4-5):525-536
  43. 43. Wolozin BL, Pruchnicki A, Dickson DW, Davies P. A neuronal antigen in the brains of Alzheimer patients. Science. 1986;232(4750):648-650
  44. 44. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, Wisniewski HM. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. The Journal of Biological Chemistry. 1986;261(13):6084-6089
  45. 45. Kosik KS, Joachim CL, Selkoe DJ. Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America. 1986;83(11):4044-4048
  46. 46. Goedert M, Spillantini MG, Potier MC, Ulrich J, Crowther RA. Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: Differential expression of tau protein mRNAs in human brain. The EMBO Journal. 1989;8(2):393-399
  47. 47. Lee G, Neve RL, Kosik KS. The microtubule binding domain of tau protein. Neuron. 1989;2(6):1615-1624
  48. 48. Jeganathan S, von Bergen M, Mandelkow EM, Mandelkow E. The natively unfolded character of tau and its aggregation to Alzheimer-like paired helical filaments. Biochemistry. 2008;47(40):10526-10539
  49. 49. Biernat J, Gustke N, Drewes G, Mandelkow EM, Mandelkow E. Phosphorylation of Ser262 strongly reduces binding of tau to microtubules: Distinction between PHF-like immunoreactivity and microtubule binding. Neuron. 1993;11(1):153-163
  50. 50. Gong CX, Liu F, Grundke-Iqbal I, Iqbal K. Post-translational modifications of tau protein in Alzheimer’s disease. Journal of Neural Transmission. 2005;112(6):813-838
  51. 51. Kidd M. Paired helical filaments in electron microscopy of Alzheimer’s disease. Nature. 1963;197:192-193
  52. 52. Fitzpatrick AWP, Falcon B, He S, Murzin AG, Murshudov G, Garringer HJ, et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature. 2017;547(7662):185-190
  53. 53. Wilcock GK, Esiri MM. Plaques, tangles and dementia. A quantitative study. Journal of the Neurological Sciences. 1982;56(2-3):343-356
  54. 54. Braak H, Braak E. Neuropathological staging of Alzheimer-related changes. Acta Neuropathologica. 1991;82(4):239-259
  55. 55. Thal DR, Holzer M, Rub U, Waldmann G, Gunzel S, Zedlick D, et al. Alzheimer-related tau-pathology in the perforant path target zone and in the hippocampal stratum oriens and radiatum correlates with onset and degree of dementia. Experimental Neurology. 2000;163(1):98-110
  56. 56. Garcia-Sierra F, Hauw JJ, Duyckaerts C, Wischik CM, Luna-Munoz J, Mena R. The extent of neurofibrillary pathology in perforant pathway neurons is the key determinant of dementia in the very old. Acta Neuropathologica. 2000;100(1):29-35
  57. 57. Scharfman HE, Schwartzkroin PA. Responses of cells of the rat fascia dentata to prolonged stimulation of the perforant path: Sensitivity of hilar cells and changes in granule cell excitability. Neuroscience. 1990;35(3):491-504
  58. 58. Lazarov O, Peterson LD, Peterson DA, Sisodia SS. Expression of a familial Alzheimer’s disease-linked presenilin-1 variant enhances perforant pathway lesion-induced neuronal loss in the entorhinal cortex. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2006;26(2):429-434
  59. 59. Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nature Cell Biology. 2009;11(7):909-913
  60. 60. de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73(4):685-697
  61. 61. Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland C, et al. Trans-synaptic spread of tau pathology in vivo. PLoS One. 2012;7(2):e31302
  62. 62. Yamada K, Cirrito JR, Stewart FR, Jiang H, Finn MB, Holmes BB, et al. In vivo microdialysis reveals age-dependent decrease of brain interstitial fluid tau levels in P301S human tau transgenic mice. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2011;31(37):13110-13117
  63. 63. Chai X, Dage JL, Citron M. Constitutive secretion of tau protein by an unconventional mechanism. Neurobiology of Disease. 2012;48(3):356-366
  64. 64. Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from the outside to the inside of a cell. The Journal of Biological Chemistry. 2009;284(19):12845-12852
  65. 65. Wu JW, Herman M, Liu L, Simoes S, Acker CM, Figueroa H, et al. Small misfolded tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons. The Journal of Biological Chemistry. 2013;288(3):1856-1870
  66. 66. Asuni AA, Boutajangout A, Quartermain D, Sigurdsson EM. Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2007;27(34):9115-9129
  67. 67. Boutajangout A, Quartermain D, Sigurdsson EM. Immunotherapy targeting pathological tau prevents cognitive decline in a new tangle mouse model. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2010;30(49):16559-16566
  68. 68. Boutajangout A, Ingadottir J, Davies P, Sigurdsson EM. Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. Journal of Neurochemistry. 2011;118(4):658-667
  69. 69. Chai X, Wu S, Murray TK, Kinley R, Cella CV, Sims H, et al. Passive immunization with anti-tau antibodies in two transgenic models: Reduction of tau pathology and delay of disease progression. The Journal of Biological Chemistry. 2011;286(39):34457-34467
  70. 70. d’Abramo C, Acker CM, Jimenez HT, Davies P. Tau passive immunotherapy in mutant P301L mice: Antibody affinity versus specificity. PLoS One. 2013;8(4):e62402
  71. 71. Lee SH, Le Pichon CE, Adolfsson O, Gafner V, Pihlgren M, Lin H, et al. Antibody-mediated targeting of tau in vivo does not require effector function and microglial engagement. Cell Reports. 2016;16(6):1690-1700
  72. 72. Lu PJ, Wulf G, Zhou XZ, Davies P, Lu KP. The prolyl isomerase Pin1 restores the function of Alzheimer-associated phosphorylated tau protein. Nature. 1999;399(6738):784-788
  73. 73. Nakamura K, Kosugi I, Lee DY, Hafner A, Sinclair DA, Ryo A, et al. Prolyl isomerase Pin1 regulates neuronal differentiation via beta-catenin. Molecular and Cellular Biology. 2012;32(15):2966-2978
  74. 74. Kondo A, Shahpasand K, Mannix R, Qiu J, Moncaster J, Chen CH, et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature. 2015;523(7561):431-436
  75. 75. Gadad BS, Britton GB, Rao KS. Targeting oligomers in neurodegenerative disorders: Lessons from alpha-synuclein, tau, and amyloid-beta peptide. Journal of Alzheimer’s Disease. 2011;24(Suppl 2):223-232
  76. 76. Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Kiritoshi T, Neugebauer V, et al. Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Scientific Reports. 2012;2:700
  77. 77. Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Gerson JE, Singh G, et al. Passive immunization with tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2014;34(12):4260-4272
  78. 78. Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(3):1024-1037
  79. 79. Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE, et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron. 2013;80(2):402-414
  80. 80. Funk KE, Mirbaha H, Jiang H, Holtzman DM, Diamond MI. Distinct therapeutic mechanisms of tau antibodies: Promoting microglial clearance versus blocking neuronal uptake. The Journal of Biological Chemistry. 2015;290(35):21652-21662
  81. 81. Yanamandra K, Jiang H, Mahan TE, Maloney SE, Wozniak DF, Diamond MI, et al. Anti-tau antibody reduces insoluble tau and decreases brain atrophy. Annals of Clinical Translational Neurology. 2015;2(3):278-288
  82. 82. West T, Hu Y, Verghese PB, Bateman RJ, Braunstein JB, Fogelman I, et al. Preclinical and clinical development of ABBV-8E12, a humanized anti-tau antibody, for treatment of Alzheimer’s disease and other Tauopathies. The Journal of Prevention of Alzheimer’s Disease. 2017;4(4):236-241
  83. 83. Nobuhara CK, DeVos SL, Commins C, Wegmann S, Moore BD, Roe AD, et al. Tau antibody targeting pathological species blocks neuronal uptake and interneuron propagation of tau in vitro. The American Journal of Pathology. 2017;187(6):1399-1412
  84. 84. Gotz J, Schild A, Hoerndli F, Pennanen L. Amyloid-induced neurofibrillary tangle formation in Alzheimer’s disease: Insight from transgenic mouse and tissue-culture models. International Journal of Developmental Neuroscience. 2004;22(7):453-465
  85. 85. Bright J, Hussain S, Dang V, Wright S, Cooper B, Byun T, et al. Human secreted tau increases amyloid-beta production. Neurobiology of Aging. 2015;36(2):693-709
  86. 86. Dai CL, Chen X, Kazim SF, Liu F, Gong CX, Grundke-Iqbal I, et al. Passive immunization targeting the N-terminal projection domain of tau decreases tau pathology and improves cognition in a transgenic mouse model of Alzheimer disease and tauopathies. Journal of Neural Transmission. 2015;122(4):607-617
  87. 87. Dai CL, Tung YC, Liu F, Gong CX, Iqbal K. Tau passive immunization inhibits not only tau but also Abeta pathology. Alzheimer’s Research & Therapy. 2017;9(1):1
  88. 88. Panza F, Solfrizzi V, Seripa D, Imbimbo BP, Lozupone M, Santamato A, et al. Tau-based therapeutics for Alzheimer’s disease: Active and passive immunotherapy. Immunotherapy. 2016;8(9):1119-1134
  89. 89. Roy S, Datta CK, Hirano A, Ghatak NR, Zimmerman HM. Electron microscopic study of neurofibrillary tangles in Steele-Richardson-Olszewski syndrome. Acta Neuropathologica. 1974;29(2):175-179
  90. 90. Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998;393(6686):702-705
  91. 91. Irwin DJ. Tauopathies as clinicopathological entities. Parkinsonism & Related Disorders. 2016;22(Suppl 1):S29-S33
  92. 92. Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: Early progress and future directions. Lancet Neurology. 2015;14(1):114-124
  93. 93. Hostetler ED, Walji AM, Zeng Z, Miller P, Bennacef I, Salinas C, et al. Preclinical characterization of 18F-MK-6240, a promising PET tracer for in vivo quantification of human neurofibrillary tangles. Journal of Nuclear Medicine. 2016;57(10):1599-1606
  94. 94. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, et al. Inflammation and Alzheimer’s disease. Neurobiology of Aging. 2000;21(3):383-421
  95. 95. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74(4):691-705
  96. 96. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131(6):1164-1178
  97. 97. Stephan AH, Madison DV, Mateos JM, Fraser DA, Lovelett EA, Coutellier L, et al. A dramatic increase of C1q protein in the CNS during normal aging. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(33):13460-13474
  98. 98. Stephan AH, Barres BA, Stevens B. The complement system: An unexpected role in synaptic pruning during development and disease. Annual Review of Neuroscience. 2012;35:369-389
  99. 99. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 2016;352(6286):712-716
  100. 100. Lansita JA, Mease KM, Qiu H, Yednock T, Sankaranarayanan S, Kramer S. Nonclinical development of ANX005: A humanized anti-C1q antibody for treatment of autoimmune and neurodegenerative diseases. International Journal of Toxicology. 2017;36(6):449-462
  101. 101. Hess DC, Abe T, Hill WD, Studdard AM, Carothers J, Masuya M, et al. Hematopoietic origin of microglial and perivascular cells in brain. Experimental Neurology. 2004;186(2):134-144
  102. 102. Simard AR, Rivest S. Bone marrow stem cells have the ability to populate the entire central nervous system into fully differentiated parenchymal microglia. The FASEB Journal. 2004;18(9):998-1000
  103. 103. Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron. 2006;49(4):489-502
  104. 104. El Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C, et al. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nature Medicine. 2007;13(4):432-438
  105. 105. Butovsky O, Kunis G, Koronyo-Hamaoui M, Schwartz M. Selective ablation of bone marrow-derived dendritic cells increases amyloid plaques in a mouse Alzheimer’s disease model. The European Journal of Neuroscience. 2007;26(2):413-416
  106. 106. Baruch K, Rosenzweig N, Kertser A, Deczkowska A, Sharif AM, Spinrad A, et al. Breaking immune tolerance by targeting Foxp3(+) regulatory T cells mitigates Alzheimer’s disease pathology. Nature Communications. 2015;6:7967
  107. 107. Kline J, Gajewski TF. Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer. Current Opinion in Investigational Drugs. 2010;11(12):1354-1359
  108. 108. Baruch K, Deczkowska A, Rosenzweig N, Tsitsou-Kampeli A, Sharif AM, Matcovitch-Natan O, et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nature Medicine. 2016;22(2):135-137
  109. 109. Latta-Mahieu M, Elmer B, Bretteville A, Wang Y, Lopez-Grancha M, Goniot P, et al. Systemic immune-checkpoint blockade with anti-PD1 antibodies does not alter cerebral amyloid-beta burden in several amyloid transgenic mouse models. Glia. 2018;66(3):492-504
  110. 110. Poduslo JF, Curran GL, Berg CT. Macromolecular permeability across the blood-nerve and blood-brain barriers. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(12):5705-5709
  111. 111. Levites Y, Smithson LA, Price RW, Dakin RS, Yuan B, Sierks MR, et al. Insights into the mechanisms of action of anti-Abeta antibodies in Alzheimer’s disease mouse models. The FASEB Journal. 2006;20(14):2576-2578
  112. 112. DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-a beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(15):8850-8855
  113. 113. Rubenstein JL, Combs D, Rosenberg J, Levy A, McDermott M, Damon L, et al. Rituximab therapy for CNS lymphomas: Targeting the leptomeningeal compartment. Blood. 2003;101(2):466-468
  114. 114. Blennow K, Zetterberg H, Rinne JO, Salloway S, Wei J, Black R, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Archives of Neurology. 2012;69(8):1002-1010
  115. 115. Seppala TT, Nerg O, Koivisto AM, Rummukainen J, Puli L, Zetterberg H, et al. CSF biomarkers for Alzheimer disease correlate with cortical brain biopsy findings. Neurology. 2012;78(20):1568-1575
  116. 116. Rubenstein E. Relationship of senescence of cerebrospinal fluid circulatory system to dementias of the aged. Lancet. 1998;351(9098):283-285
  117. 117. Skarlatos S, Yoshikawa T, Pardridge WM. Transport of [125I]transferrin through the rat blood-brain barrier. Brain Research. 1995;683(2):164-171
  118. 118. Duffy KR, Pardridge WM, Rosenfeld RG. Human blood-brain barrier insulin-like growth factor receptor. Metabolism. 1988;37(2):136-140
  119. 119. Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie J, et al. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. Journal of Neurochemistry. 2011;117(2):333-345
  120. 120. Pardridge WM. Delivery of biologics across the blood-brain barrier with molecular Trojan horse technology. BioDrugs. 2017;31(6):503-519
  121. 121. Shin SU, Friden P, Moran M, Olson T, Kang YS, Pardridge WM, et al. Transferrin-antibody fusion proteins are effective in brain targeting. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(7):2820-2824
  122. 122. Wu D, Pardridge WM. Pharmacokinetics and blood-brain barrier transport of an anti-transferrin receptor monoclonal antibody (OX26) in rats after chronic treatment with the antibody. Drug Metabolism and Disposition. 1998;26(9):937-939
  123. 123. Hackel BJ, Huang D, Bubolz JC, Wang XX, Shusta EV. Production of soluble and active transferrin receptor-targeting single-chain antibody using Saccharomyces cerevisiae. Pharmaceutical Research. 2006;23(4):790-797
  124. 124. Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Science Translational Medicine. 2011;3(84):84ra44
  125. 125. Couch JA, Yu YJ, Zhang Y, Tarrant JM, Fuji RN, Meilandt WJ, et al. Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier. Science Translational Medicine. 2013;5(183):183ra57, 1-12
  126. 126. Kanodia JS, Gadkar K, Bumbaca D, Zhang Y, Tong RK, Luk W, et al. Prospective design of anti-transferrin receptor bispecific antibodies for optimal delivery into the human brain. CPT: Pharmacometrics & Systems Pharmacology. 2016;5(5):283-291
  127. 127. Zuchero YJ, Chen X, Bien-Ly N, Bumbaca D, Tong RK, Gao X, et al. Discovery of novel blood-brain barrier targets to enhance brain uptake of therapeutic antibodies. Neuron. 2016;89(1):70-82
  128. 128. Thom G, Hatcher J, Hearn A, Paterson J, Rodrigo N, Beljean A, et al. Isolation of blood-brain barrier-crossing antibodies from a phage display library by competitive elution and their ability to penetrate the central nervous system. MAbs. 2018;10(2):304-314

Written By

Philip J. Dolan and Wagner Zago

Submitted: 06 November 2017 Reviewed: 07 March 2018 Published: 18 July 2018