16
REVIEW Emerging Immunotherapies for Parkinson Disease Samis M. A. Zella . Judith Metzdorf . Emine Ciftci . Friederike Ostendorf . Siegfried Muhlack . Ralf Gold . Lars To ¨nges Received: October 12, 2018 / Published online: December 11, 2018 Ó The Author(s) 2018 ABSTRACT Symptomatic treatment options for Parkinson disease have steadily improved, and individu- alized therapeutic approaches are becoming established for every stage of the disease. How- ever, disease-modifying therapy with a causal approach is still unavailable. The central cau- sative role of alpha-synuclein pathology, including its progressive spread to most areas of the CNS, has been widely recognized, and a strong involvement of immune responses has recently been discovered. New immunologic technologies have been shown to effectively prevent the progression of alpha-synuclein pathology in animal models. These approaches have recently been translated into the first human clinical trials, representing a novel starting point for the causal therapy of Parkin- son disease. In this review, the pathomechanistic role of alpha-synuclein and its influence on the surrounding cellular envi- ronment are analyzed with a strong focus on immune responses and neuroinflammation. The potential of novel immunotherapeutic approaches that reduce the burden of alpha- synuclein pathology in the CNS is critically evaluated, and currently ongoing human clini- cal trials are presented. The clinical develop- ment of these new immunotherapies is progressing rapidly and gives reason to hope that a causal therapy of Parkinson disease could be possible in the foreseeable future. Keywords: Alpha-synuclein; Antibody-based therapy; Immunotherapy; Neuroinflammation; Parkinson disease; Vaccination INTRODUCTION Parkinson disease (PD) is the most common neurodegenerative disorder after Alzheimer dis- ease (AD) and is neuropathologically character- ized by nigrostriatal dopaminergic degeneration and the presence of aggregated and misfolded alpha-synuclein (aSyn). In clinical examination, PD patients show motor deficits such as bradykinesia, rigidity, tremor and postural instability, which reflect the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and impairment of Enhanced digital features To view enhanced digital features for this article go to https://doi.org/10.6084/ m9.figshare.7378277. S. M. A. Zella J. Metzdorf E. Ciftci F. Ostendorf S. Muhlack R. Gold L. To ¨nges (&) Neurologische Klinik der Ruhr-Universita ¨t Bochum, St. Josef-Hospital, Bochum, Germany e-mail: [email protected] R. Gold L. To ¨nges Neurodegeneration Research, Protein Research Unit Ruhr (PURE), Ruhr University Bochum, Bochum, Germany Neurol Ther (2019) 8:29–44 https://doi.org/10.1007/s40120-018-0122-z

Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

Embed Size (px)

Citation preview

Page 1: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

REVIEW

Emerging Immunotherapies for Parkinson Disease

Samis M. A. Zella . Judith Metzdorf . Emine Ciftci . Friederike Ostendorf .

Siegfried Muhlack . Ralf Gold . Lars Tonges

Received: October 12, 2018 / Published online: December 11, 2018� The Author(s) 2018

ABSTRACT

Symptomatic treatment options for Parkinsondisease have steadily improved, and individu-alized therapeutic approaches are becomingestablished for every stage of the disease. How-ever, disease-modifying therapy with a causalapproach is still unavailable. The central cau-sative role of alpha-synuclein pathology,including its progressive spread to most areas ofthe CNS, has been widely recognized, and astrong involvement of immune responses hasrecently been discovered. New immunologictechnologies have been shown to effectivelyprevent the progression of alpha-synucleinpathology in animal models. These approacheshave recently been translated into the firsthuman clinical trials, representing a novelstarting point for the causal therapy of Parkin-son disease. In this review, the

pathomechanistic role of alpha-synuclein andits influence on the surrounding cellular envi-ronment are analyzed with a strong focus onimmune responses and neuroinflammation.The potential of novel immunotherapeuticapproaches that reduce the burden of alpha-synuclein pathology in the CNS is criticallyevaluated, and currently ongoing human clini-cal trials are presented. The clinical develop-ment of these new immunotherapies isprogressing rapidly and gives reason to hopethat a causal therapy of Parkinson disease couldbe possible in the foreseeable future.

Keywords: Alpha-synuclein; Antibody-basedtherapy; Immunotherapy; Neuroinflammation;Parkinson disease; Vaccination

INTRODUCTION

Parkinson disease (PD) is the most commonneurodegenerative disorder after Alzheimer dis-ease (AD) and is neuropathologically character-ized by nigrostriatal dopaminergic degenerationand the presence of aggregated and misfoldedalpha-synuclein (aSyn). In clinical examination,PD patients show motor deficits such asbradykinesia, rigidity, tremor and posturalinstability, which reflect the degeneration ofdopaminergic (DA) neurons in the substantianigra pars compacta (SNpc) and impairment of

Enhanced digital features To view enhanced digitalfeatures for this article go to https://doi.org/10.6084/m9.figshare.7378277.

S. M. A. Zella � J. Metzdorf � E. Ciftci � F. Ostendorf �S. Muhlack � R. Gold � L. Tonges (&)Neurologische Klinik der Ruhr-Universitat Bochum,St. Josef-Hospital, Bochum, Germanye-mail: [email protected]

R. Gold � L. TongesNeurodegeneration Research, Protein Research UnitRuhr (PURE), Ruhr University Bochum, Bochum,Germany

Neurol Ther (2019) 8:29–44

https://doi.org/10.1007/s40120-018-0122-z

Page 2: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

DA neurotransmission to basal ganglia motorcircuits. The accompanying heavy burden ofnon-motor symptoms such as autonomic failure,daytime sleepiness, cognitive deficits, or evenpsychiatric alterations indicates the involve-ment of other CNS neurotransmitter systems [1].

As the main neuropathologic culprit, aSynhas been identified in humans and was found tobe located in aSyn-immunopositive Lewy neu-rites and Lewy bodies. Several human post-mortem studies have demonstrated that notonly the nigrostriatal dopaminergic system isaffected in PD but that—depending on the(prodromal) disease stage—also Lewy pathologycan be early found in the peripheral autonomicnervous system, including neurons of theenteric plexus of the gastrointestinal tract, par-avertebral autonomic ganglia and sympatheticnerve fibers in the adrenal gland and heart andin cutaneous nerves. In subsequent diseasestages, the medulla, pons, midbrain, dien-cephalon, basal forebrain, amygdala, olfactorybulb, limbic cortex and finally higher orderassociation cortices can be involved, too [1–3].

Thus, the extent of aSyn pathology is not astable state but seems to progress in a prion-likecell-to-cell spreading to continuously involvefurther neuronal and non-neuronal cells—adisease propagation process that is also dis-cussed for other neurodegenerative diseases in asimilar way [4]. Importantly, aSyn-associatedneurodegeneration is accompanied by neu-roinflammatory features, which highlights therole of aSyn for the immune system and non-neuronal cells [5, 6]. As much as this spread ofthe disease in the CNS proves its aggressivecharacter, there are also possibilities for newtherapeutic approaches. A very obvious one is tointerfere with disease dissemination and elimi-nate excess or toxic compounds of aSyn that arelocated extracellularly and therefore are readilyaccessible to therapeutic approaches.

In this review, we will describe the propertiesof aSyn and its pathophysiologic implications, inparticular regarding its activation of cellularneuroinflammatory cascades. Thus, a clearrationale opens up to counteract this viciouscircle by controlling the spread of disease withthe help of immunologic, antibody-based tech-nologies. The potential of these active and

passive immunotherapies is presented from thefundamental scientific side as well as from recenthuman clinical data. Current knowledge is crit-ically evaluated with the goal to provide a timelyreview on immunotherapies for PD for both theclinician and basic scientist. This article is basedon previously conducted studies and does notcontain any studies with human participants oranimals performed by any of the authors.

ALPHA-SYNUCLEIN AND ITS ROLEIN THE PATHOGENESIS OF PD

Alpha-synuclein is a soluble and cytoplasmicprotein of 140-amino-acid (aa) length consist-ing of three domains: an amphipathic N-ter-minal region (1–65 aa), a non-amyloid-bcomponent (NAC) region (66–95 aa) and aC-terminal domain (96–140 aa) [7]. In the CNS,aSyn is predominately expressed in neurons ofthe thalamus, basal ganglia and substantia nigra[8] but also in the peripheral nervous system inblood cells and in platelets [9, 10].

In human neuropathology, aggregated aSynrepresents the main component of so-calledLewy bodies and Lewy neurites, which areintraneuronal filamentous inclusions and arelocated in perikarya or neurites, respectively, ofdegenerating DA neurons in PD patients[8, 11, 12]. In familial Parkinson disease, rarehuman genetic alterations occur mostly aspoint mutations in aSyn (A30P, E46 K, H50Q,G51D, A53E and A53T) or as gene duplicationsand triplications that cause autosomal-recessiveor -dominant forms of PD [13].

The function of aSyn in neurons is onlyincompletely understood, but it is known toplay an important role in neurotransmitterrelease, synaptic plasticity and synaptic vesiclerecycling [7, 14, 15]. aSyn is stored in presy-naptic nerve terminals and mediates synapticvesicle fusion with soluble NSF attachmentprotein (SNARE) complex assemblies and viadirect interaction with syntaxin-1, SNAP-25 andvesicle-associated membrane protein 2(VAMP2)/synaptobrevin-2 [15–17]. This enablessynaptic vesicle trafficking from the readyreleasable pool to the presynaptic active zonewhere the content of the synaptic vesicles is

30 Neurol Ther (2019) 8:29–44

Page 3: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

secreted. In case of aSyn accumulation, theneurotransmitter release, vesicle trafficking,recycling and SNARE complex stability are allaffected and thus severely impair synapticfunction of dopaminergic neurons [16–19].

Under physiologic conditions, wild-typeaSyn is a natively soluble unfolded monomer,which binds to curved membranes such assynaptic vesicles and regulates the above-men-tioned SNARE-complex chaperoning function.Under pathologic circumstances with changesin pH level or oxidative stress, aSyn can convertto insoluble and aggregated forms that areenriched in b-sheets and assemble into oligo-meric and fibrillar structures. These forms ofaSyn have a strong pathogenic potential andmay harm several physiologic functions of thecell [15, 20, 21].

Importantly, aSyn can be released fromintracellular compartments to the extracellularspace. From there it may propagate to otherneuronal or glial cells in which it elicits patho-genic cascades that impair their cellular func-tion. Concerning these aSyn propagationmechanisms, three main principles are known.First, in healthy neurons a non-classical ER-/Golgi-independent protein export pathway canbe used whereby aSyn can be directly integratedinto secretory vesicles and subsequentlyreleased by exocytosis. Second, aSyn vesiclescan be translocated into early endosomes andthen be released to the extracellular spacethrough recycling endosomes. As a third alter-native, early endosomal aSyn can be incorpo-rated to intraluminal vesicles that are part of theso-called multivesicular bodies (MVB). Thesecan undergo a degradative process after fusionwith lysosomes or directly secrete by fusionwith the plasma membrane [22]. Several in vitroand in vivo animal studies have shown thatespecially oligomeric aSyn directly confers toxiceffects to surrounding cells, whether they areneuronal or non-neuronal glial cells.

Concerning direct effects of aggregated aSynonto neuronal cells, several in vitro studies havebeen performed. Primary hippocampal neu-ronal cells readily internalized exogenous pre-formed fibrils of artificial recombinant aSyn ifprovided in the cell culture medium. In thecytoplasm, these exogenous fibrils recruited

endogenous aSyn and induced pathologic mis-folded aSyn, which was phosphorylated, ubiq-uitinated and aggregated to insoluble fibrils.Similarly to human PD, a LN-like pathologydeveloped, first located in axons, and was thenpropagated to form LB-like inclusions in the cellperikarya [23, 24]. The axonal protein accumu-lation was preceded by axonal transport alter-ations with an initial predominant anterogradedirection of aSyn, being similar to the amyloid-beta1-42 form but different from the processingof huntingtin protein fragments [25]. Accumu-lating pathologic aSyn then led to selectivedecreases in synaptic proteins, progressiveimpairments in neuronal excitability and con-nectivity and finally neuronal death. Interest-ingly, endogenous aSyn was sufficient for theformation of these aggregates, and overexpres-sion of wild-type or mutant aSyn was not nec-essarily required. A frequently observed uptakemechanism of aSyn could be attributed toabsorptive-mediated endocytosis [23, 24]. Morerecent findings have shown that aSyn uptake isin part also cell surface receptor mediated uti-lizing the protein product of lymphocyte-acti-vation gene 3 (LAG3) [26], which is expressednot only by neuronal cells but also microglia[27]. The underlying mechanism here seems tobe a temporary inhibition of the NMDA recep-tor making it obvious that aSyn-mediatedimpairment of cellular function is not onlyrestricted to dopaminergic neurons [28].

To evaluate direct aSyn toxicity in vivo, aroute of administration in animals has been thedirect injection of various aggregated seeds intothe substantia nigra of wild-type rats. Thisresulted in a progressive motor impairment andcell death with fibrils being the major toxicstrain. Here, ribbons caused a histopathologicphenotype similar to human PD neuropathol-ogy [29]. These findings indicate that distinctaSyn strains display differential seeding capaci-ties. Interestingly, aSyn assemblies also crossedthe blood-brain barrier and distributed to thecentral nervous system after intravenous injec-tion [29]. In another study, Luk et al. used wild-type mice that were unilaterally injected withrecombinant aSyn into the dorsal striatum. Atdifferent time points, an increasing spread ofLewy bodies was observed, and although aSyn

Neurol Ther (2019) 8:29–44 31

Page 4: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

was only injected into one hemisphere, afterseveral days Lewy bodies were also detected onthe contralateral hemisphere and in diversebrain regions including the hippocampus [30].Viral-mediated overexpression of wild-type andtransgenic aSyn injected into the substantianigra in mice severely damaged nigraldopaminergic cells and its axons. Importantly,also the intrinsic regenerative capacity ofdopaminergic neurons is significantly impairedif dopamine neurons suffer from aSyn overload[31].

Extracellular aSyn has a strong impact onglial cells and there can elicit a variety ofimmunologic responses. Glial cells are by far thelargest cell population, accounting for morethan 50% of all cells in the brain, and thuscomprise about 1000 billion cells [32]. Theydifferentiate in the CNS, where they are collec-tively referred to as neuroglia, consisting ofastroglia, microglia and oligodendroglia.

Astroglia account for by far the largest shareof glial cells and perform many tasks essential tothe survival of neurons. They ensure structuraland metabolic cerebral homeostasis, regulatesynaptic transmission, water transport andblood flow, and myelination and produce neu-rotrophic molecules such as GDNF [33]. Exper-iments with human ESC-derived astrocyteshave demonstrated that these cells activelytransferred aggregated aSyn to healthy astro-cytes via direct contact and tunneling nan-otubes in a propagating manner instead ofdegrading them [34]. The intracellularly accu-mulated aSyn affected the lysosomal machineryand induced mitochondrial damage. This eli-cited further internal proinflammatory respon-ses and caused cellular death [22, 35]. The aSyn-induced impairments of astrocyte functioncould be alleviated by an application of oligo-mer-selective anti-aSyn antibodies [36].

Microglial cells represent the innate immunesystem of the CNS. Precursor cells migrate intothe CNS already during the prenatal phase.Microglia account for approximately 10–20% ofthe total glia population [37]. The physiologicalfunction of CNS-resident microglia involves themaintenance of homeostasis through ongoingmonitoring of the CNS environment and, ifnecessary, phagocytosis or pinocytosis of

degradation products or cell debris [38]. Micro-glial cells have been shown to be able to directlyengulf aSyn attempting to clear it from theextracellular space. This response was stronglydependent on its aggregation state [39]. Distinctoligomeric forms but not monomers or fibrils ofaSyn interacted with microglial toll-like recep-tor 2 (TLR2) and activated the TLR2 signalingpathway [40] leading to increased production ofneurotoxic ROS [41].

To reduce increased levels of harmfullyaggregated extracellular but also endogenousaSyn, a clearing process of the protein has to beestablished. This can be achieved withimmunotherapies using vaccination strategiesor antibodies directed against aSyn [42]. Ifextracellular levels of aSyn decrease, the spreadto other neuronal and non-neuronal cells suchas astroglia or microglia will be reduced andultimately prevent pathologic inflammatoryactivation [42, 43]. Other aSyn degradativeprocesses are mediated through ubiquitination,lysosomal degradation by macroautophagy orchaperone-mediated autophagy (CMA) [7]. Inthe next section, we will present the currentstate of research in the first PD immunothera-pies, which all have the goal of a causal diseasemodification in humans. Findings from animalstudies and recent human clinical trials will becompared.

ALPHA-SYNUCLEIN AS TARGETFOR IMMUNIZATION STRATEGIES

Immunotherapies against aSyn currently appearto be a very promising therapeutic approach forthe modification of disease progression in thetreatment of PD patients with early stage disease[44, 45]. The aim of these therapies is to reducethe load of extracellular aSyn and thereby haltthe spread of aSyn in the brain. There are twoprincipal forms of anti-aSyn immunotherapy:active immunization or vaccination, whichemploy the immune system to self-generateantibodies against aSyn, and passive immu-nization, which is achieved by the administra-tion of antibodies directed against differentdomains of aSyn [46]. In the subsequent part wewill discuss recent data from active and passive

32 Neurol Ther (2019) 8:29–44

Page 5: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

immunization therapies in animal models andhuman clinical trials.

Active Immunization Strategies

Animal ModelsA seminal study in which an active immuniza-tion against aSyn was performed in animalsin vivo was published already in 2005. Masliahet al. used transgenic mice overexpressinghuman aSyn under control of the platelet-derived growth factor-b promoter (PDGF-b pro-moter). In this animal model, mice have aSynaggregations in neurons throughout variousregions of the brain (the cortex, hippocampusand olfactory system), thus mimicking humanLewy body neuropathology [47]. Mice wereimmunized with purified recombinant humanaSyn expressed in E. coli. The vaccination led toa production of antibodies with high affinity toaSyn. Immunized mice showed reduced aSynaggregation in neuronal cell bodies and synap-ses with decreased neurodegeneration [48, 49].The authors hypothesized an increased degra-dation of aggregated aSyn via lysosomal path-ways, which was supported by colocalization ofhuman aSyn and the lysosomal marker cathep-sin D. Furthermore, they observed only a rathermild activation of micro- and astroglia in vac-cinated mice [48, 49].

Ghochikyan et al. developed another vacci-nation approach. They used three differentfragments of aSyn fused to the epitope P30 fromtetanus toxin with the aim to generate anti-bodies against aSyn without eliciting a harmfulTH cell response. Wild-type mice were immu-nized with the different vaccines in a 2-weekinterval over 6 weeks. This led to a stronghumoral immune response whose aSyn anti-bodies recognized Lewy bodies and Lewy neu-rites in brain tissue of DLB patients. In the mice,no TH cell response to aSyn was detected [50].

A third active immunization approach wasthe use of short aSyn peptide fragments. Man-dler et al. immunized mice with a small peptidemimicking the C-terminal part of aSyn(110–130) (AFFITOPE vaccine AFF1) thatinduced a humoral response without eliciting aT cell autoimmune response. PDGF-aSyn and

mThy1-aSyn mice were immunized with AFF1twice a week to monthly over a 6-month period.Vaccinated mice showed an amelioration inmotor function (body suspension test) andlong-term memory (Water maze) as well asreduced neurodegeneration due to reducedaccumulation of aSyn oligomers in axons andsynapses. Furthermore, AFF1 treatment dimin-ished astro- and microgliosis in PDGF-b aSynmice and increased the production of antiin-flammatory cytokines such as Il-1Ra, Il-2 and Il-27 [51]. In a follow-up study, AFF1 was appliedto a mouse model of multiple system atrophy(MSA) in which aSyn is expressed specifically inoligodendrocytes under the control of themyelin basic protein promotor (MBP-aSynmice). AFF 1-treated mice showed a decreasedaccumulation of aSyn and a reduced demyeli-nation in the neocortex, striatum and corpuscallosum. Overall, neurodegeneration andmotor deficits were reduced as was diseasespreading from oligodendrocytes to astrocytes.Microglia activation was stronger than in con-trols, which was supposed to be important forthe effective clearance of aSyn [45].

A novel fourth vaccination strategy applied acombination of aSyn/Grp94 chaperone in amurine chronic MPTP model of PD. After directimmunization with aSyn/Grp94, an immuneprofile was observed in the peripheral systemthat consisted of a Th1-shifted aSyn-specificresponse accompanied by an immune-regula-tory/Th2-skewed general phenotype. In theCNS, a strong suppression of microglial activa-tion in the substantia nigra and striatum wasobserved [52]. This study indicates that remod-eling even of peripheral immunity by vaccina-tion could be a strong modifier of CNSneurodegenerative disease.

Human Clinical TrialsThe active anti-aSyn vaccination strategies haveso far only been studied in phase I human clin-ical trials. In the AFF008 study series, realized bythe Austrian company AFFiRiS, a synthetic aSynepitope called AFFITOPE� PD01A formulatedwith adjuvant was subcutaneously injected topatients with early PD. The study design wasrandomized, parallel group, single center, andthe study goals were the tolerability and safety as

Neurol Ther (2019) 8:29–44 33

Page 6: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

well as immunogenicity of repeated subcuta-neous (s.c.) administration of AFFITOPE�

PD01A. In four consecutive studies, AFFiRiS008,008E, 008A and 008AA, patients were random-ized to receive four immunizations of eitherAFFITOPE� PD01A low dose (15 lg) or high dose(75 lg) (study AFF008). At screening, the averagetime of PD after the first diagnosis was 2.6 years.Of the 32 patients enrolled, 24 received activetreatment, and 8 PD patients were left on stan-dard of care medication serving as an observa-tional comparison group. Twenty-one patientsin the PD01A treatment groups and five in theobservational group completed the entire seriesof studies. Patients were allowed to continuetheir standard of care PD medication. To extendthe first observation period, an additional12-month follow-up was added in the secondstudy (AFF008E). In the third study (AFF008A), abooster immunization was applied after re-ran-domizing patients from study AFF008E into twodifferent doses of boost agent (15 or 75 lgAFFITOPE� PD01A). In the fourth study(AFF008AA, ‘‘reboost study’’), a second boosterwith a fixed dose of 75 lg AFFITOPE� PD01A wasapplied to patients who had previously beenimmunized altogether five times.

Overall, both doses of AFFITOPE� PD01Awere well tolerated locally and systemically. Nostudy drug-related serious adverse events (SAE)or suspected unexpected serious adverse reac-tions (SUSAR) occurred. The majority of adverseevents (about 55%) consisted of local reactionsmostly being only mild and without dosedependency. AFFITOPE� PD01A elicited a clearimmune response against the peptide itself andcross-reactivity against the aSyn targeted epi-tope over time. The first boost immunizationproduced a significant effect on all analyzedtiters, resulting in the maximum titers observedin all studies. An immune response wasobserved in 19 of 22 (86%) of the vaccinatedpatients. In 12 of the 19 (63%) responders,specific serum antibodies were generated. Thesecond boost immunization had further stabi-lized the produced antibody titers. Altogether, asignificant increase in titers against PD01A wasobserved over time, which translated into ahumoral immune response against aSyn.Importantly, PD01-specific antibodies were also

detectable in cerebrospinal fluid and werereported to bind to both oligomeric and fibrillaraSyn compared with its monomers. There was atrend in reduction of oligomeric aSyn levels inplasma as well as in cerebrospinal fluid upontreatment with PD01A at week 26. Clinicalscores for PD were reported to be stable duringthe entire study period. However, because thestudy was not designed to evaluate clinicalefficacy, no further data were provided. A limi-tation of this study series is the lack of a double-blind design. So far, no peer-reviewed publica-tions are available [53–55].

In another vaccination study, a differentsynthetic aSyn epitope called AFFITOPE� PD03Awas used. In the phase 1 AFFiRiS011 study withrandomized, placebo-controlled, parallel-group,blinded, bi-centered design, aims were todemonstrate safety, tolerability and immuno-genicity in patients with early PD. At screening,the mean duration of PD after diagnosis wasbetween 1.6 and 2.3 years. Thirty-six patientswere randomized to a high dose of AFFITOPE�

PD03A (75 lg), low dose (15 lg) or placebogroup. Patients received fine injections, four forpriming every 4 weeks and the fifth as boostimmunization 9 months after the first immu-nization. Both doses of PD03A were locally andsystemically well tolerated. Fifty-nine percent ofadverse events consisted of only mild and dose-dependent local injection reactions. Theimmunogenicity profile in patients was judgedas encouraging as patients efficiently rose onespecific antibody response to aSyn [53, 54, 56].

In total, 98 patients have participated instudies investigating AFFITOPE� PD01A orPD03A and have been observed for up to48 months (AFFITOPE� PD01A) and 12 months(AFFITOPE� PD03A), respectively. Regardinglong-term safety, immunologic and clinicalparameters, the results are promising. Now, theclinical efficacy has to be demonstrated in phaseII studies.

Passive Immunization Strategies

Animal ModelsThe passive immunization approach basicallyconsists of a transfer of anti-aSyn-directed

34 Neurol Ther (2019) 8:29–44

Page 7: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

antibodies to the CNS. In a pioneering study, amonoclonal antibody directed against theC-terminus of aSyn (9E4) was applied in trans-genic mice overexpressing human wild-typeaSyn under control of the PDGF-b promoter[57]. The antibody 9E4 was injected intra-venously once a week over a 6-month periodwith a dose of 10 mg/kg bodyweight. Animproved performance in the water maze aswell as a reduced accumulation of calpain-cleaved aSyn in axons and synapses of corticaland hippocampal neurons was observedthroughout the study [57].

The antibody 9E4 was directed against theC-terminal region of aSyn on purpose becausethere is evidence that the cleavage of aSyn at theC-terminal region is involved in the formationof the toxic aSyn oligomers. Cleavage of fibrillaraSyn at the C-terminus by calpain I producesfragments that match to C-terminally cleavedfragments as found in human LBs [58]. Fur-thermore, the expression of C-terminal trun-cated aSyn in mice facilitates its oligomerizationand reduces dopamine levels in the striatum[59].

For this reason, another group applied a dif-ferent C-terminal antibody (Ab274) in the samemouse model with weekly intraperitonealinjections. After 4 weeks of treatment, an ame-lioration of behavioral deficits and neurode-generation was observed in the passivelyimmunized group. Additionally, an increasedlocalization of aSyn and AB274 in microglia wasfound. Here, the uptake of the Ab274/aSyncomplex by microglia was demonstrated to bemediated via the Fcc receptor and its transportto lysosomes in the microglial cell line (Bv2)[60].

In a third study, a transgenic Parkinsonmouse model was used in which wild-type aSynwas overexpressed under control of the mThy1promoter. This caused behavioral deficits andneuronal accumulation of aSyn in the thala-mus, basal ganglia, substantia nigra and brain-stem (Thy1 aSyn mice) [47, 61]. Theimmunization was performed with three C-ter-minally directed anti-aSyn antibodies, 1H7, 5C1and 5D12, once a week over a 6-week period.The application of 1H7 and 5C1 led to a sig-nificant reduction of cleaved aSyn in the cortex

and striatum resulting in an amelioration ofaxonal pathology and behavioral performance.Accompanying in vitro studies revealed areduction of cell-to-cell transmission, and theantibodies inhibited the cleavage of aSyn bycalpain-1. It was hypothesized that the stabi-lization of the C-terminal domain by 1H7 and5C1 could protect against cleavage and therebyreduce the amount of toxic aSyn aggregation[44].

N-terminally directed anti-aSyn antibodieswere also developed and have been applied in arat model of PD. The model was generated bynigral injection of a recombinant adeno-asso-ciated viral vector expressing human wild-typeaSyn under control of the CBA promoter.Intraperitoneal treatment with an anti-aSynantibody (AB1) every 2 weeks for a total of 3months resulted in a moderate but not signifi-cant amelioration of behavioral parameters androbust protection against the loss of nigraldopaminergic neurons. Overall, aSyn expres-sion and microglial activation were decreased inthe nigral region [62].

A second N-terminal anti-aSyn antibody(Syn303) was developed to specifically targetmisfolded aSyn in a mouse model with intras-triatal injections of synthetic preformed aSynfibrils that spread in the brain and cause motordeficits and dopaminergic cell loss. The weeklyintraperitoneal treatment with Syn303 up to26 weeks inhibited the spreading of aSynaggregates and improved behavioral deficits aswell as dopaminergic cell loss [63].

Because aSyn exerts the strongest toxiceffects in its aggregated conformational states,antibodies have been generated that specificallytarget oligomers and fibrils and avoid animpairment of physiologically needed aSyn.The Parkinson mouse model with expression ofthe human aSyn A30P mutant under the Thy-1promoter leads to the formation of aSyn fibrils,which cause dopaminergic neurodegenerationand behavioral and motor deficits [64]. Aweekly passive immunization of these micewith a monoclonal antibody specifically tar-geting the oligomeric/protofibrillar forms ofaSyn (antibody mAb47) for a total of 14 weeksled to a significant decrease of pathogenic aSynprotofibrils in the spinal cord without changing

Neurol Ther (2019) 8:29–44 35

Page 8: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

the level of monomeric forms after 1 year. Nodifferences in activation of microglia or astro-cytes were found [65]. The rapid uptake ofmAB47 is mediated by Fcc receptors as wasshown in H4 cells overexpressing aSyn [66].

In another approach, Thy1 aSyn mice wereimmunized weekly with specific antibodies(Syn-O1, Syn-O4 and Syn-F1) recognizing onlyoligomers and fibrils but not monomeric aSynover 3 months. All three led to an ameliorationof aSyn pathology and behavioral performanceby preventing the accumulation of aSyn fibrilsand related synaptic alterations. Furthermore,reduced astrogliosis and microglia activation inthe hippocampus was observed [67].

Taken together, targeting aSyn by passiveimmunization is a very promising disease-modifying therapy in Parkinson models in vivo.An overview of animal studies with passiveimmunization is provided in Table 1. Conse-quently, this approach has been translated intohuman clinical trials.

Human Clinical TrialsSeveral clinical trials have so far evaluated thesafety and tolerability of ascending doses of anintravenous anti-aSyn antibody infusion inhumans. The most advanced studies havealready reached the clinical phase II study leveland also involved PD patients.

The first aSyn-based therapeutic candidatefor PD—named PRX002—was developed byProthena and entered into clinical trials in2015. It is a humanized IgG1 monoclonal ver-sion of the before-mentioned C-terminallydirected murine monoclonal antibody 9E4. In afirst single ascending-dose study in healthyvolunteers, PRX002 demonstrated good safetyand tolerability if intravenous infusions of 0.3,1.0, 3.0, 10 or 30 mg/kg at maximum wereapplied. After a single intravenous infusion of30 mg/kg, PRX002 antibody levels in serum roseto 578 lg/ml and, after a single intravenousinfusion of 0.3 mg/kg, up to 7.6 lg/ml. Theaverage terminal half-life across all doses was18.2 days. Already within 1 h of PRX002administration a significant dose-dependentreduction in free serum aSyn was observed.Total aSyn (free plus bound) increased dosedependently, presumably because of the

expected change in kinetics following antibodybinding [68].

Recently, the data of a multiple ascending-dose trial with PRX002 in patients with mild-to-moderate idiopathic PD (Hoehn and Yahr stages1–3) were published. Here, participants wereenrolled into six ascending-dose cohorts andwere randomly assigned to receive PRX002(0.3 mg/kg, 1.0 mg/kg, 3.0 mg/kg, 10 mg/kg,30 mg/kg or 60 mg/kg) or placebo. Three intra-venous infusions of PRX002 or placebo wereapplied in 4-week intervals. Again, safety andtolerability data were favorable, and serumPRX002 levels increased in an approximatelydose-proportional manner. Serum levels of free-to-total aSyn were significantly reduced. Meanterminal elimination half-life of the antibodywas similar across all doses (10.2 days). Inaddition, cerebrospinal fluid (CSF) data weredemonstrated in this study. The mean cere-brospinal fluid PRX002 concentration increasedwith PRX002 dose and was approximately 0.3%relative to serum across all dose cohorts. How-ever, no change in the level of CSF aSyn couldbe demonstrated, which was attributed to therelatively low affinity of the antibody formonomeric, as opposed to aggregated, forms ofaSyn, which are much less abundant in CSF[69]. A phase 2, multinational study of PRX002/RO7046015 in recently diagnosed PD patientswas initiated in summer 2017 in collaborationwith Roche (PASADENA Study, ClinicalTrials.-gov identifier NCT03100149).

Another anti-aSyn-directed antibody thathas been evaluated in a human clinical trial isthe N-terminally directed BIIB054, realized byBiogen. It was developed by the Swiss biotechcompany Neurimmune and is a fully humanIgG1 monoclonal antibody. It has been isolatedfrom B-cell lines derived from neurologicallyhealthy individuals and is highly selective forthe aggregated form of aSyn as has been shownin tissue sections and extracts from PD and DLB,but not in unaffected brains. Therefore, it bindsmuch less of its monomeric form [70]. In arecently concluded phase 1, single ascending-dose study of BIIB054 in healthy volunteers, itwas well tolerated at single doses up to 90 mg/kg and had a serum half-life of 28 days. The CSFconcentrations achieved in healthy volunteers

36 Neurol Ther (2019) 8:29–44

Page 9: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

Table1

Passiveim

mun

izationstudiesin

Parkinsonanim

almodels

Target

site

ofaSyn

Antibod

yMod

eof

application

Frequency

of application

Treatment

duration

Disease

mod

eltype

Effecton

neuron

alcells

Effecton

non-

neuron

alcells

Behavioral

alteration

sReferences

C-terminal

9E4

i.v.

Weekly

4weeks

TransgenicPD

GFb

aSyn

mice

DA

cellloss

(30%

);

Microgliosis;

Cylindertest:

[57]

C-terminal

Ab274

i.p.

Weekly

4weeks

TransgenicPD

GFb

aSyn

mice

NeuN

cellloss;

IncreasedaSyn

clearanceby

microglia

Poletestandtotal

activity

:[60]

C-terminal

1H7

5C1

i.p.

Weekly

6months

TransgenicThy1aSyn

mice

DA

cellloss

(35%

);

Astro-and

microgliosis;

Probe/transversal

roun

dbeam

test:

[44]

N-terminal

Central

region

AB1

AB2

i.p.

2weekly

3months

NigralA

AV-CBA-aSyn

injectionin

wtrats

DA

andNeuN

cellloss;

Microgliosis;

Cylindertest:

[62]

N-terminal

Syn303

i.p.

Weekly

180days

Intrastriatalinjection

ofpreformed

fibrils

inwtmice

Blocked

a-Syn

spreading

DA

cellloss;

–Wirehang:

[63]

Protofibrils

mAB47

i.p.

Weekly

14weeks

TransgenicThy-1

A30Pmice

aSyn

protofibrils

inspinalcord

;

Microgliaand

astrocytes

$–

[64]

Oligom

ers

Oligom

ers

Fibrils

Syn-O1

Syn-O4

Syn-F1

i.p.

Weekly

3month

TransgenicThy1aSyn

mice

aSyn accumulation

;

NeuN

cellloss;

Microgliosisin

hippocam

pus

;

(O1,O4)

Totalactivity

in

open

fieldtest:

[67]

Neurol Ther (2019) 8:29–44 37

Page 10: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

Table2

Passiveim

mun

izationin

human

clinicaltrialsforParkinsondisease

Clin

ical

trial

no.

Antibod

ySp

onsor/collabo

rator

Stud

ytype

Stud

ydesign

Stud

ydu

ration

Outcomemeasures

Current

status/data

References

NCT02095171

PRX002(9E4)

humanized

IgG1

monoclonal

Prothena

Biosciences

andHoffm

ann-La

Roche

Phase

1Studytype:Single-center,

rand

omized,d

ouble-blind,

placebo-controlled,

single

ascend

ing-dose

studyin

healthyvolunteers

Cohorts:placebo,

0.3,

1.0,

3.0,

10or

30mg/kg

Treatment:onesingle

intravenousinfusion

ofPR

X002

12-W

eekfollow-up

period

after

administrationof

asingledose

ofthe

studydrug

Primary:safety,

tolerability,and

pharmacokineticsof

asingleintravenous

infusion

ofPR

X002

Secondary:

Immun

ogenicity

Studycompleted

inDec.2

014

Nodrug-related

safety

ortolerability

issues.D

ose-depend

enttarget

occupancyin

plasm,average

term

inalhalf-lifeacrossalldoses

18.2

days

Nogeneration

ofanti-PRX002

antibodies

[68]

NCT02157714

PRX002(9E4)/

RG7935

humanized

IgG1

monoclonal

Prothena

Biosciences

andHoffm

ann-La

Roche

Phase

1Studytype:Multicenter,

rand

omized,d

ouble-blind,

placebo-controlled,

multiple

ascend

ing-dose

trialin

patientswithmild

tomoderateidiopathicPD

(Hoehn

andYahrstages

1–3)

Cohorts:ascend

ing-dose

cohorts:placebo,

0.3mg/kg,

1.0,

3.0,

10,3

0or

60mg/kg

Treatment:3intravenous

infusionsevery4weeks

ofPR

X002or

placebo

8-Weektreatm

entand

16-weekfollow-up

period

after

administrationof

last

doseof

thestudydrug

Primary:safety

and

tolerabilityof

multiple

intravenousinfusions

ofPR

X002

Secondary:

pharmacokineticsand

immun

ogenicityof

singleandmultiple

infusions

Studycompleted

inSept.2

016

Nodrug-related

safety

ortolerability

issues.Noantidrug

antibodieswere

detected.S

erum

PRX002levels

increasedin

anapproxim

ately

dose-proportionalmanner;mean

term

inalhalf-lifesimilaracrossall

doses(10.2days).Rapid

dose-and

time-depend

entmeanreductions

from

baselin

evs.p

lacebo

infree

serum

a-synuclein

levelsof

upto

97%

[69]

NCT03100149

(PASA

DENA

Study)

PRX002/

RO7046015/

Prasinezum

abhumanized

IgG1

monoclonal

Hoffm

ann-LaRoche

andProthena

Biosciences

Phase

2Studytype:Multicenter,

rand

omized,d

ouble-blind,

placebo-controlledtrialin

patientswithmild

idiopathic

PD(H

oehn

andYahrstages

1–2)

Cohorts:Part1(year1):

placebo,low-dose(1500mg),

high-dose(4500mg)—Part2

(Year2):low-dose

(1500mg),h

igh-dose

(3500mgor

4500

mg

depend

ingon

body

weight)

Treatment:Part1:

4-weekly

intravenousinfusionsof

PRX002or

placebofor

52weeks,P

art2:

4-weekly

intravenousinfusionsof

PRX002foratotalof

52weeks

52-W

eektreatm

ent(part

1)withanother

52-weektreatm

ent

(Part2

)andfollow-up

period

after

administrationof

last

doseof

thestudydrug

Primary:clinicalefficacy

ofintravenous

PRX002vs.p

lacebo

(changein

MDS-

UPD

RSI–III)

Secondary:efficacyin

motor

subscales(M

DS-

UPD

RSIII),C

GI,

changesin

DaT

-SP

ECT,safety,

tolerability,

pharmacokineticsand

immun

ogenicity

Studycurrently

ongoing,estimated

studycompletiondate

Feb.

2021

38 Neurol Ther (2019) 8:29–44

Page 11: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

Table2

continued

Clin

ical

trial

no.

Antibod

ySp

onsor/collabo

rator

Stud

ytype

Stud

ydesign

Stud

ydu

ration

Outcomemeasures

Current

status/data

References

NCT02459886

BIIB054human

IgG1

monoclonal

Biogen

Phase

1Studytype:Multicenter,

rand

omized,d

ouble-blind,

placebo-controlledsingle-

ascend

ingdose

studyin

healthysubjectsandsubjects

withearlyParkinson’s

Disease

Cohorts:placebo,

1,5,

15,4

5,90

mg/kg

(onlyhealthy

subjects)

Treatment:onesingle

intravenousinfusion

ofBIIB054

16-W

eekfollow-up

period

after

administrationof

asingledose

ofthe

studydrug

Primary:safety,

tolerability,and

cognitiveassessmentof

asingleintravenous

infusion

ofBIIB054

Secondary:

pharmacokineticsand

immun

ogenicity

Studycompleted

inNov.2

017

Nodrug-related

safety

ortolerability

issues

except

foroneSA

Ein

the

highestdose

cohort

Serum

half-lifeof

28days

[71,

78]

NCT03318523

(SPA

RK

Study)

BIIB054human

IgG1

monoclonal

Biogen

Phase

2Studytype:Multicenter,

rand

omized,d

ouble-blind,

placebo-controlledtrialin

patientswithmild

idiopathic

PD(H

oehn

andYahrstages

1–2,

5)

Cohorts:placebo,

3mg/kg,

15mg/kg,4

5mg/kg

Treatment:4-weekly

intravenousinfusionsof

BIIB054foratotalof

72weeks

72-W

eektreatm

entand

12-weekfollow-up

period

after

administrationof

last

doseof

thestudydrug

Primary:dose-related

safety

ofBIIB054

Secondary:

pharmacokinetics,

pharmacodynam

icsand

immun

ogenicity

Studycurrently

ongoing,estimated

studycompletiondateApril2022

Neurol Ther (2019) 8:29–44 39

Page 12: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

were 0.2% of those seen in plasma [71]. Singledoses of BIIB054 up to 45 mg/kg were also welltolerated in PD patients, and the pharmacoki-netic profile was comparable to what had beenobserved in healthy volunteers. In PD patients,the CSF:plasma ratio of BIIB054 was 0.4% [71].Now, BIIB054 is being evaluated in a multina-tional phase 2 study with recently diagnosed PDpatients (SPARK Study, ClinicalTrials.gov iden-tifier NCT03318523).

Two other anti-aSyn monoclonal antibodiesare currently in the early stages of development.The monoclonal antibody MEDI1341 wasdeveloped by a collaboration between AstraZe-neca and Takeda. It is supposed to have only areduced immune effector function and is nowentering phase 1 testing in healthy volunteers(ClinicalTrials.gov identifier NCT03272165).Another monoclonal antibody targeted at aSynoligomers and protofibrils is BAN0805, whichwas developed in collaboration betweenBioarctic and Abbvie. For now, there is nopublicly available information regarding itsdevelopment status [72]. An overview of humanclinical trials with passive immunization isprovided in Table 2.

Potential risks associated with the use ofimmunotherapy in neurodegenerative diseaseshave to be differentiated between active orpassive immunization (see overview for AD in[73]). Active immunization therapies perform avaccination with antigenic material—a strategythat has so far mostly been used for the pre-vention of infectious diseases [74]. General sidereactions may consist of a local (e.g., sore arm)or systemic inflammatory reaction (e.g., low-grade fever) that disappears in a few days and inpart depends on the employed vaccinationadjuvant [75]. In an early AD vaccination study,there were cases of aseptic meningoencephalitisthat were probably of autoimmune origin beingmediated by an overwhelming Th1 helper cellstimulation [76]. Newer therapies includingaSyn now employ active vaccines that avoidTh1 helper cell stimulation [55]. In the currentphase I aSyn studies, there were no relevantsafety and tolerability issues.

Passive immunization approaches com-pletely bypass these issues and administerspecific antibodies. Here, systemic side reactions

with low-grade fever are possible, and there is arisk of allergenic reactions. Anti-amyloid-betatrials in AD have shown mixed safety results.There has been no increased risk of any adverseevent, serious adverse events or death with theexception of a nearly fivefold increase in so-called amyloid-related imaging abnormalities(ARIAs) [77]. These can present both as vaso-genic edema (ARIA-E) and cerebral microhem-orrhages (ARIA-H) but are supposedly associatedwith amyloid-beta targeting and therefore arenot expected in anti-aSyn therapies.

FUTURE PERSPECTIVESAND CONCLUSIONS

Through detailed studies on patients, but also inanimal models of PD, key neurodegenerativepathomechanisms of this disease have beendecrypted. In addition to the damage to neu-ronal cells, it is now obvious that also glial cellsare affected and that a strong neuroimmuno-logic interaction takes place with aSyn being thecentral player. From today’s perspective, amodulation of this cross-cell disease propaga-tion in PD appears possible by ‘‘targeting’’ ofaSyn.

Very elegant neuroimmunologic tools havebeen developed to inhibit disease spread byextracellular aSyn. The elimination of aSyn bytargeted antibody-based technologies withactive or passive immunization therefore seemsvery pragmatic. Certainly, technologic chal-lenges, such as overcoming the blood-brainbarrier or the targeted elimination of onlyaggregated aSyn, still need to be optimized.However, we are well on the way to providinganswers to these questions as several humanclinical trials with ambitious expectations arecurrently underway. The simultaneous applica-tion of antibody-based therapies in atypicalParkinson’s syndromes such as PSP or also Alz-heimer’s disease will in any case result inimportant basic insights. This will, it is our firmconviction, enable new technologic advances inthe immunotherapy for PD and bring aboutnew powerful options to modify diseaseprogression.

40 Neurol Ther (2019) 8:29–44

Page 13: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

ACKNOWLEDGEMENTS

Funding. No funding or sponsorship wasreceived for this study or publication of thisarticle.

Authorship. All named authors meet theInternational Committee of Medical JournalEditors (ICMJE) criteria for authorship for thisarticle, take responsibility for the integrity ofthe work as a whole and have given theirapproval for this version to be published.

Disclosures. Samis M.A. Zella, Judith Metz-dorf, Emine Ciftci, Friederike Ostendorf, Sieg-fried Muhlack, Ralf Gold and Lars Tonges havenothing to disclose.

Compliance with Ethics Guidelines. Thisarticle is based on previously conducted studiesand does not contain any studies with humanparticipants or animals performed by any of theauthors. I declare that the submitted work isentirely our own and is not under considerationfor publication elsewhere.

Open Access. This article is distributedunder the terms of the Creative CommonsAttribution-NonCommercial 4.0 InternationalLicense (http://creativecommons.org/licenses/by-nc/4.0/), which permits any non-commercial use, distribution, and reproductionin any medium, provided you give appropriatecredit to the original author(s) and the source,provide a link to the Creative Commons license,and indicate if changes were made.

REFERENCES

1. Poewe W, Seppi K, Tanner CM, Halliday GM,Brundin P, Volkmann J, et al. Parkinson disease.Nat Rev Dis Primers. 2017;3:17013.

2. Braak H, Del Tredici K, Rub U, De Vos RA, JansenSteur EN, Braak E. Staging of brain pathology rela-ted to sporadic Parkinson’s disease. NeurobiolAging. 2003;24(2):197–211.

3. Klingelhoefer L, Reichmann H. Pathogenesis ofParkinson disease—the gut-brain axis and environ-mental factorS. NAT REV NEUROL.2015;11(11):625–36.

4. Walsh DM, Selkoe DJ. A critical appraisal of thepathogenic protein spread hypothesis of neurode-generation. Nat Rev Neurosci. 2016;17(4):251–60.

5. Tonges L, Metzdorf J, Zella S. Parkinson’s diseaseand neuroinflammation—Cellular pathology,mechanisms and therapeuticoptions. FortschrNeurol Psychiatr. 2018;86(S 01):S10–20. https://doi.org/10.1055/s-0044-101608.

6. Chitnis T, Weiner HL. CNS inflammation andneurodegeneration. J Clin Invest.2017;127(10):3577–87.

7. Bendor JT, Logan TP, Edwards RH. The function ofalpha-synuclein. Neuron. 2013;79(6):1044–66.

8. Braak H, del Tredici K. Neuropathological staging ofbrain pathology in sporadic Parkinson’s disease:separating the wheat from the chaff. J ParkinsonsDis. 2017;7(S1):S71–85.

9. Barbour R, Kling K, Anderson JP, Banducci K, ColeT, Diep L, et al. Red blood cells are the major sourceof alpha-synuclein in blood. Neurodegener Dis.2008;5(2):55–9.

10. Doppler K, Jentschke HM, Schulmeyer L, Vadasz D,Janzen A, Luster M, et al. Dermal phospho-alpha-synuclein deposits confirm rem sleep behaviourdisorder as prodromal Parkinson’s disease. ActaNeuropathol. 2017;133(4):535–45.

11. Dauer W, Przedborski S. Parkinson’s disease: mech-anisms and models. Neuron. 2003;39(6):889–909.

12. Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, LeeVM, et al. Aggregation of alpha-synuclein in lewybodies of sporadic Parkinson’s disease and dementiawith lewy bodies. Am J Pathol. 1998;152(4):879–84.

13. Trinh J, Farrer M. Advances in the genetics ofParkinson disease. Nat Rev Neurol.2013;9(8):445–54.

14. Burre J, Sharma M, Tsetsenis T, Buchman V, Ether-ton MR, Sudhof TC. Alpha-synuclein promotessnare-complex assembly in vivo and in vitro. Sci-ence. 2010;329(5999):1663–7.

15. Wong YC, Krainc D. Alpha-synuclein toxicity inneurodegeneration: mechanism and therapeuticstrategies. Nat Med. 2017;23(2):1–13.

16. Bridi JC, Hirth F. Mechanisms, of alpha-synucleininduced synaptopathy in Parkinson’s disease. FrontNeurosci. 2018;12:80.

Neurol Ther (2019) 8:29–44 41

Page 14: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

17. Sudhof TC. A molecular machine for neurotrans-mitter release: synaptotagmin and beyond. NatMed. 2013;19(10):1227–31.

18. Lashuel HA, Overk CR, Oueslati A, Masliah E. The,many faces, of alpha-synuclein: from structure andtoxicity to therapeutic target. Nat Rev Neurosci.2013;14(1):38–48.

19. Sudhof TC. The presynaptic active zone. Neuron.2012;75(1):11–25.

20. Bergstrom AL, Kallunki P, Fog K. Development, ofpassive immunotherapies for synucleinopathies.Mov Disord. 2016;31(2):203–13.

21. Burre J, Sharma M, Sudhof TC. Definition of amolecular pathway mediating alpha-synucleinneurotoxicity. J Neurosci. 2015;35(13):5221–32.

22. Marques O, Outeiro TF. Alpha-synuclein: fromsecretion to dysfunction and death. Cell Death Dis.2012;3:E350.

23. Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA,Riddle DM, Stieber A, et al. Exogenous alpha-synu-clein fibrils induce lewy body pathology leading tosynaptic dysfunction and neuron death. Neuron.2011;72(1):57–71.

24. Volpicelli-Daley LA, Luk KC, Lee VM. Addition, ofexogenous alpha-synuclein preformed fibrils toprimary neuronal cultures to seed recruitment ofendogenous alpha-synuclein to lewy body and lewyneurite-like aggregates. Nat Protoc.2014;9(9):2135–46.

25. Brahic M, Bousset L, Bieri G, Melki R, Gitler AD.Axonal transport and secretion of fibrillar forms ofalpha-synuclein, ABETA42 peptide and httexon 1.Acta Neuropathol. 2016;131(4):539–48.

26. Mao X, Ou MT, Karuppagounder SS, Kam TI, Yin X,Xiong Y, et al. Pathological alpha-synuclein trans-mission initiated by binding lymphocyte-activationgene 3. Science. 2016;353(6307):aah3374.

27. Zhang Y, Chen K, Sloan SA, Bennett ML, ScholzeAR, O’Keeffe S, et al. An RNA-sequencing tran-scriptome and splicing database of glia, neurons,and vascular cells of the cerebral cortex. J Neurosci.2014;34(36):11929–47.

28. Diogenes MJ, Dias RB, Rombo DM, Vicente MirandaH, Maiolino F, Guerreiro P, et al. Extracellularalpha-synuclein oligomers modulate synaptictransmission and impair LTP via NMDA-receptoractivation. J Neurosci. 2012;32(34):11750–62.

29. Peelaerts W, Bousset L, van der Perren A, MoskalyukA, Pulizzi R, Giugliano M, et al. Alpha-synucleinstrains cause distinct synucleinopathies after local

and systemic administration. Nature.2015;522(7556):340–4.

30. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P,Trojanowski JQ, et al. Pathological alpha-synucleintransmission initiates Parkinson-like neurodegen-eration in nontransgenic mice. Science.2012;338(6109):949–53.

31. Tonges L, Szego EM, Hause P, Saal KA, Tatenhorst L,Koch JC, et al. Alpha-synuclein mutations impairaxonal regeneration in models of Parkinson’s dis-ease. Front Aging Neurosci. 2014;6:239.

32. Herculano-Houzel S. The human brain in numbers:a linearly scaled-up primate brain. Front HumNeurosci. 2009;3:31.

33. Sofroniew MV, Vinters HV. Astrocytes: biology andpathology. Acta Neuropathol. 2010;119(1):7–35.

34. Rostami J, Holmqvist S, Lindstrom V, Sigvardson J,Westermark GT, Ingelsson M, et al. Human astro-cytes transfer aggregated alpha-synuclein via tun-neling nanotubes. J Neurosci.2017;37(49):11835–53.

35. Dexter DT, Jenner P. Parkinson disease: frompathology to molecular disease mechanisms. FreeRadic Biol Med. 2013;62:132–44.

36. Gustafsson G, Lindstrom V, Rostami J, NordstromE, Lannfelt L, Bergstrom J, et al. Alpha-synucleinoligomer-selective antibodies reduce intracellularaccumulation and mitochondrial impairment inalpha-synuclein exposed astrocytes. J Neuroinflam-mation. 2017;14(1):241.

37. Soulet D, Rivest S. Microglia. Curr Biol.2008;18(12):R506–8.

38. Wolf SA, Boddeke HW, Kettenmann H. Microglia inphysiology and diseaSE. Annu Rev Physiol.2017;79:619–43.

39. Hoffmann A, Ettle B, Bruno A, Kulinich A, Hoff-mann AC, Von Wittgenstein J, et al. Alpha-synu-clein activates BV2 microglia dependent on itsaggregation state. Biochem Biophys Res Commun.2016;479(4):881–6.

40. Kim C, Ho DH, Suk JE, You S, Michael S, Kang J,et al. Neuron-released oligomeric alpha-synuclein isan endogenous agonist of TLR2 for paracrine acti-vation of microglia. Nat Commun. 2013;4:1562.

41. Hoenen C, Gustin A, Birck C, Kirchmeyer M,Beaume N, Felten P, et al. Alpha-synuclein proteinspromote pro-inflammatory cascades in microglia:stronger effects of the A53T mutant. PLoS One.2016;11(9):E0162717.

42 Neurol Ther (2019) 8:29–44

Page 15: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

42. Bergstrom AL, Kallunki P, Fog K. Development, ofpassive immunotherapies for synucleinopathies.Mov Disord. 2016;31(2):203–13.

43. Bruck D, Wenning GK, Stefanova N, Fellner L. Gliaand alpha-synuclein in neurodegeneration: a com-plex interaction. Neurobiol Dis. 2016;85:262–74.

44. Games D, Valera E, Spencer B, Rockenstein E, ManteM, Adame A, et al. Reducing C-TERMINAL-trun-cated alpha-synuclein by immunotherapy attenu-ates neurodegeneration and propagation inParkinson’s disease-like models. J Neurosci.2014;34(28):9441–54.

45. Mandler M, Valera E, Rockenstein E, Mante M,Weninger H, Patrick C, et al. Active immunizationagainst alpha-synuclein ameliorates the degenera-tive pathology and prevents demyelination in amodel of multiple system atrophy. Mol Neurode-gener. 2015;10:10.

46. Sardi SP, Cedarbaum JM, Brundin P. Targeted ther-apies for Parkinson’s disease: from genetics to theclinic. Mov Disord. 2018;33(5):684–96.

47. Rockenstein E, Mallory M, Hashimoto M, Song D,Shults CW, Lang I, et al. Differential neuropatho-logical alterations in transgenic mice expressingalpha-synuclein from the platelet-derived growthfactor and Thy-1 promoters. J Neurosci Res.2002;68(5):568–78.

48. Masliah E, Rockenstein E, Adame A, Alford M,Crews L, Hashimoto M, et al. Effects of alpha-synuclein immunization in a mouse model ofParkinson’s disease. Neuron. 2005;46(6):857–68.

49. Lobello K, Ryan JM, Liu E, Rippon G, Black R. Tar-geting Beta amyloid: a clinical review ofimmunotherapeutic approaches in Alzheimer’sdisease. Int J Alzheimers Dis. 2012;2012:628070.https://doi.org/10.1155/2012/628070.

50. Ghochikyan A, Petrushina I, Davtyan H, Hova-kimyan A, Saing T, Davtyan A, et al. Immuno-genicity of epitope vaccines targeting differenT Bcell antigenic determinants of human a-synuclein:feasibility study. Neurosci Lett. 2014;560:86–91.

51. Mandler M, Valera E, Rockenstein E, Weninger H,Patrick C, Adame A, et al. Next-generation activeimmunization approach for synucleinopathies:implications for parkinson’s disease clinical trials.Acta Neuropathol. 2014;127(6):861–79.

52. Villadiego J, Labrador-Garrido A, Franco JM, Leal-Lasarte M, De Genst EJ, Dobson CM, et al. Immu-nization with A-Synuclein/Grp94 reshapes periph-eral immunity and suppresses microgliosis in achronic Parkinsonism model. GLIA.2018;66(1):191–205.

53. Kingwell K. Zeroing in on neurodegenerative A-synuclein. Nat Rev Drug Discov. 2017;16(6):371–3.

54. Valera E, Masliah E. Immunotherapy for neurode-generative diseases: focus on A-synucleinopathies.Pharmacol Ther. 2013;138(3):311–22.

55. Affiris announces encouraging long-term data froma series of first-in-human studies using Affitope�

PD01A targeting oligomeric alpha-synuclein inearly Parkinson’s disease Patients [Press Release].2018.

56. Affiris announces top line results of first-in-humanclinical study using AFFITOPE�PD03A, Confirmingimmunogenicity and safety profile In Parkinson’sdisease patients [Press Release]. 2017. http://www.affiris.com/news/affiris-announces-top-line-results-of-first-in-human-clinical-study-using-affitope/.

57. Masliah E, Rockenstein E, Mante M, Crews L,Spencer B, Adame A, et al. Passive immunizationreduces behavioral and neuropathological deficitsin an alpha-synuclein transgenic model of lewybody disease. PLoS One. 2011;6(4):E19338.

58. Mishizen-Eberz AJ, Norris EH, Giasson BI, Hodara R,Ischiropoulos H, Lee VM, et al. Cleavage of alpha-synuclein by calpain: potential role in degradationof fibrillized and nitrated species of alpha-synu-clein. Biochemistry. 2005;44(21):7818–29.

59. Tofaris GK, Garcia Reitbock P, Humby T, Lam-bourne SL, O’Connell M, Ghetti B, et al. Patholog-ical changes in dopaminergic nerve cells of thesubstantia nigra and olfactory bulb in mice trans-genic for truncated human alpha-synuclein(1-120):implications for lewy body disorders. J Neurosci.2006;26(15):3942–50.

60. Bae EJ, Lee HJ, Rockenstein E, Ho DH, Park EB, YangNY, et al. Antibody-aided clearance of extracellularA-synuclein prevents cell-to-cell aggregate trans-mission. J Neurosci. 2012;32(39):13454–69.

61. Fleming SM, Salcedo J, Fernagut PO, Rockenstein E,Masliah E, Levine MS, et al. Early and progressivesensorimotor anomalies in mice overexpressingwild-type human alpha-synuclein. J Neurosci.2004;24(42):9434–40.

62. Shahaduzzaman M, Nash K, Hudson C, Sharif M,Grimmig B, Lin X, et al. anti-human A-synucleinN-terminal peptide antibody protects againstdopaminergic cell death and ameliorates behavioraldeficits in an aav-a-synuclein rat model of Parkin-son’s Disease. PLoS One. 2015;10(2):E0116841.

63. Tran HT, Chung CH, Iba M, Zhang B, TrojanowskiJQ, Luk KC, et al. A-synuclein immunotherapyblocks uptake and templated propagation of

Neurol Ther (2019) 8:29–44 43

Page 16: Emerging Immunotherapies for Parkinson Disease · disease have steadily improved, and individu-alized therapeutic approaches are becoming established for every stage of the disease

misfolded a-synuclein and neurodegeneration. CellRep. 2014;7(6):2054–65.

64. Kahle PJ, Neumann M, Ozmen L, Muller V, Jacob-sen H, Schindzielorz A, et al. Subcellular localiza-tion of wild-type and Parkinson’s disease-associatedmutant alpha-synuclein in human and transgenicmouse brain. J Neurosci. 2000;20(17):6365–73.

65. Lindstrom V, Fagerqvist T, Nordstrom E, Eriksson F,Lord A, Tucker S, et al. Immunotherapy targeting a-synuclein protofibrils reduced pathology iN (THY-1)-H[A30P] a-synuclein mice. Neurobiol Dis.2014;69:134–43.

66. Gustafsson G, Eriksson F, Moller C, Da Fonseca TL,Outeiro TF, LannfelT L, et al. Cellular uptake of a-synuclein oligomer-selective antibodies isenhanced by the extracellular presence of a-synu-clein and mediated via Fcc ReceptorS. Cell MolNeurobiol. 2017;37(1):121–31.

67. El-Agnaf O, Overk C, Rockenstein E, Mante M,Florio J, Adame A, et al. Differential effects ofimmunotherapy with antibodies targeting a-synu-clein oligomers and fibrils in a transgenic model ofsynucleinopathy. Neurobiol Dis. 2017;104:85–96.

68. Schenk DB, Koller M, Ness DK, Griffith SG,Grundman M, Zago W, et al. First-in-humanassessment of PRX002, an anti-alpha-synucleinmonoclonal antibody, in healthy volunteers. MovDisord. 2017;32(2):211–8.

69. Jankovic J, Goodman I, Safirstein B, Marmon TK,Schenk DB, Koller M et al. Safety and tolerability ofmultiple ascending doses of PRX002/RG7935, ananti-a-synuclein monoclonal antibody, in patientswith parkinson disease: a randomized clinical trial.JAMA Neurol. 2018;75(10):1206–14. https://doi.org/10.1001/jamaneurol.2018.1487.

70. Weihofen A, Patel H, Huy C, Liu C, Combaluzier I,Mueller-Steiner S, et al. Binding and functional

characterization of human-derived anti-alpha-synuclein antibody BIIB054. Neurodeg Dis. 2017;17(SUPPL 1)(8):59.

71. Brys M, Hung S, Fanning L, Penner N, Yang M,Welch M, et al. Randomized, double- blind, pla-cebo-controlled, single ascending dose study ofantialpha-synuclein antibody biib054 in patientswith Parkinson disease. Neurology. 2018;90(15 15SUPPLEMENT):S26.001.

72. Bioarctic enters into collaboration with abbvie forparkinson disease research [Press Release]. 2018.https://www.bioarctic.se/en/section/media/press-releases/

73. Wisniewski T, Goni F. Immunotherapeuticapproaches for Alzheimer’s disease. Neuron.2015;85(6):1162–76.

74. Zepp F. Principles of vaccination. Methods MolBiol. 2016;1403:57–84.

75. Guy B. The perfect mix: recent progress in adjuvantresearch. Nat Rev Microbiol. 2007;5(7):505–17.

76. Tabira T. Immunization therapy for alzheimer dis-ease: a comprehensive review of active immunizationstrategies. Tohoku J Exp Med. 2010;220(2):95–106.

77. Penninkilampi R, Brothers HM, Eslick GD. Safetyand efficacy of anti-amyloid-beta immunotherapyin Alzheimer’s disease: a systematic review andmeta-analysis. J Neuroimmune Pharmacol.2017;12(1):194–203.

78. Brys I, Halje P, Scheffer-Teixeira R, Varney M,Newman-Tancredi A, Petersson P. Neurophysiolog-ical effects in cortico-basal ganglia-thalamic circuitsof antidyskinetic treatment with 5-HT1A receptorbiased agonists. Exp Neurol. 2018;302:155–68.https://doi.org/10.1016/j.expneurol.2018.01.010.

44 Neurol Ther (2019) 8:29–44