You are on page 1of 19

REVIEWS

Therapeutic strategies for Parkinson


disease: beyond dopaminergic drugs
Delphine Charvin1*, Rossella Medori1, Robert A. Hauser2 and Olivier Rascol3
Abstract | Existing therapeutic strategies for managing Parkinson disease (PD), which focus on
addressing the loss of dopamine and dopaminergic function linked with degeneration of
dopaminergic neurons, are limited by side effects and lack of long-term efficacy. In recent
decades, research into PD pathophysiology and pharmacology has focused on understanding
and tackling the neurodegenerative processes and symptomology of PD. In this Review, we
discuss the challenges associated with the development of novel therapies for PD, highlighting
emerging agents that aim to target cell death, as well as new targets offering a symptomatic
approach to managing features and progression of the disease.

Bradykinesia The year 2017 marked the 200th anniversary of the first aetiology of PD because it appears to link familial and
Abnormal slowness of published description by James Parkinson of the disease sporadic forms of the disease. The presence of aggre‑
movement. that would later come to bear his name. In his seminal gates in patient brains suggests that the proteostasis
Autonomic dysfunction work ‘An essay on the shaking palsy’, Parkinson detailed of α-synuclein is disturbed in PD. Indeed, α-synuclein
Abnormal functioning of the the clinical features of what is now termed Parkinson exists in various conformations in a dynamic equilib‑
autonomic nervous system, disease (PD)1, although many decades were to pass rium that is modulated by many factors, including
affecting the functioning of the during which physicians could recognize the condi‑ oxidative stress, post-translational modifications and
heart, bladder, intestines, sweat
glands, pupils and blood vessels.
tion but considered it to be a disease that “the physi‑ concentrations of fatty acids, phospholipids and metal
Examples include constipation, cian can do little for by way of relief or cure”2. Indeed, ions, and a tight balance of these factors controls the
orthostatic hypotension, urinary the most important advance in the treatment of PD did levels and aggregation of α-synuclein9,10 (FIG. 2). A pro‑
incontinence and erectile not emerge until the early 1960s, when Birkmayer and gressive, age-related decline in proteolytic defence mech‑
dysfunction.
Hornykiewicz first documented the clinical benefit of anisms in the brain might play a role in the accumulation
Synucleinopathy levodopa (3,4‑dihydroxy-l‑phenylalanine), the precursor of α-synuclein11,12 and contribute to the role of ageing in
A neurodegenerative disease of dopamine, in patients with PD3,4 (FIG. 1). PD pathophysiology (BOX 3).
characterized by an excessive
PD is an age-related neurodegenerative disorder, PD remains an incurable neurological condition.
accumulation of α-synuclein.
Examples include PD, characterized by progressive and selective loss of dopa‑ Levodopa is a mainstay therapy and, to date, drug
dementia with Lewy bodies minergic neurons in the substantia nigra that contributes treatments used in the management of PD attempt to
and multiple system atrophy. to the cardinal motor symptoms of the disease: brady- compensate for the loss of dopamine and dopaminergic
kinesia, rigidity and resting tremor (BOX 1). In addition function. The shortcomings of such pharmacotherapies
to dopaminergic neuropathology, there is dysfunction include their diminished effectiveness over time; the
1
Prexton Therapeutics,
Geneva, Switzerland. in cholinergic, serotonergic, glutamatergic and noradr‑ development of medication-related complications such
2
Department of Neurology, energic pathways — some of which may be secondary as motor fluctuations and dyskinesia (levodopa-­induced
University of South Florida, to the deafferentation associated with dopamine system dyskinesia (LID)) and potential side effects such as
Tampa, FL, USA. dysfunction and/or dopaminergic neuronal death, with impulse control disorders, sleepiness or sudden-onset
3
Centre d’Investigation
Clinique CIC1436, Services
others being due to the same PD pathological process sleep; and dopamine dysregulation syndrome13,14. Moreover,
de Neurologie et de affecting dopaminergic and non-dopaminergic systems the disease continues to progress, and non-dopamine-­
Pharmacologie Clinique, (for a review of our current understanding of PD neuro‑ responsive symptoms such as cognitive dysfunction and
Réseau NS‑PARK/FCRIN et pathology, refer to5–8). People with PD also experience imbalance become more prominent and lead to long-
Centre COEN NeuroToul,
‘non-motor’ symptoms such as sleep disturbance, fatigue, term disability 15.
CHU de Toulouse, INSERM,
University of Toulouse 3, altered mood, cognitive changes, autonomic dysfunction In the past decade, researchers and clinicians have
Toulouse, France. and pain (BOX 2). pioneered a new area of discovery — looking beyond
*e-mail: delphine.charvin@ PD is classified as a synucleinopathy, as α-synuclein, a the progressive dopamine neuron loss in PD — to try
prextontherapeutics.com presynaptic neuronal protein, is a major constituent of to understand and tackle the neurodegenerative pro‑
doi:10.1038/nrd.2018.136 Lewy bodies, which are a pathological hallmark of PD. cesses and the symptomology of PD (FIG. 1). Two non-­
Published online 28 Sep 2018 Interestingly, α-synuclein has a unique importance in the dopaminergic medications have already been marketed

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

First POC clinical trial of


an A2A antagonist
First description Intravenous Introduction of the
of Lewy bodies L-DOPA Dopamine direct and indirect First POC clinical trial of Demonstration that the
agonists pathway model of the a cell-based therapy direct and indirect pathways
(apomorphine) basal ganglia circuitry are not alternatively but
First Trihexyphenidyl concomitantly active to
neurosurgical modulate motor function
First attempt
An Essay on the intervention of a cell-based Mutations in SNCA identified
Shaking Palsy is of the basal therapy for as the first genetic cause of PD First POC
published by ganglia to humans with clinical trial of
James Parkinson treat PD Amantadine PD Deep brain stimulation an mGlu5 NAM

1817 1872 1899 1913 1940 1960 1961 1962 1969 1970 1985 1988 1989 1990 1993 1995 1996 1997 1998 2001 2003 2008 2012 2014 2017

The shaking Evidence of Carbidopa and MAOB First POC First POC First phase I
palsy is striatal dopamine L-DOPA inhibitors clinical trial of clinical trial clinical trial
renamed PD deficiency in PD combination (selegiline) neurotrophic of a gene for immuno-
therapy factors therapy therapy in PD
Oral L-DOPA approved
by FDA
First suggestion that the COMT inhibitors α-Synuclein found to be First POC
substantia nigra could be (tolcapone) the main component of clinical trial of
the site of PD pathology Lewy bodies an mGlu4 PAM

First suggestion of First POC


Breakthrough in PD research cell-to-cell transmission clinical trial of a
Non-dopaminergic agents Dopaminergic agents of α-synuclein LRRK2 inhibitor

Figure 1 | History of Parkinson disease research and therapeutic advances. A2A, adenosine receptor type 2A; COMT, catechol-O‑methyltransferase;
l‑DOPA, levodopa; LRRK2, leucine-rich repeat serine/threonine-protein kinase 2; MAOB, monoamine oxidase type B; mGlu,Reviews
Nature metabotropic
| Drug Discovery
glutamate receptor; NAM, negative allosteric modulator; PAM, positive allosteric modulator; PD, Parkinson disease; POC, proof of concept.
Adapted from REF. , Springer Nature Limited.
239

for many years for the treatment of PD symptoms — disease-modifying drugs and novel symptomatic thera­
namely, the anti-muscarinic trihexyphenidyl16 and the pies. The mode of action, and the drug-specific preclin‑
anti-­glutamatergic amantadine17. Recent research has ical performances, of candidate treatments for PD are
Dyskinesia helped uncover some of the pathogenic mechanisms assessed, and some of the most promising biological tar‑
Abnormal involuntary
underlying PD, which include α-synuclein aggrega‑ gets and therapeutic agents in contemporary research
movements. Levodopa-induced
dyskinesia is associated with tion, mitochondrial dysfunction, endoplasmic reticu‑ are highlighted.
chronic levodopa treatment lum stress, impaired autophagy and the loss of calcium This Review does not purport to be systematic or
in patients with PD. It is homeostasis (BOX 3). Furthermore, studies have identified exhaustive, but is a narrative review aiming to provide
characterized by hyperkinetic a number of potential biomarkers, highlighted ways in an overview of the symptomatic and disease-modifying
movements including chorea,
athetosis and dystonia.
which existing drugs may be repurposed for PD, revealed approaches for the treatment of PD that have advanced to
alternative formulations of dopaminergic therapies to clinical studies and that are targeting non-­dopaminergic
Dopamine dysregulation improve their benefit/risk ratio and singled out novel tar‑ pathways. An extensive review of the pathogenetic
syndrome gets for pharmacological manipulation (BOX 3; TABLES 1,2). mechanisms of PD is beyond the scope of this article,
Dysfunction of the reward
A number of the resulting pharmaceutical candidates and readers are referred to BOX 3 and referenced reviews
system associated with
long-term dopaminergic have progressed and are now in clinical development. in each section for more information. One important
treatments. It is characterized New approaches to research are helping to change the objective of this Review is to analyse and understand
by addictive behaviours and paradigm of PD treatment — allowing a ‘rethinking’ of apparent translational discrepancies between preclinical
excessive use of dopaminergic the overall objectives of treatment and helping expand studies and the clinic. Thus, the preclinical research and
medication, with patients
taking quantities of medication
these to include innovative, dual symptomatic and approaches that are discussed here have been selected
well beyond the dose required complication-­preventive (or complication-limiting) and on the basis of their clinical status and availability of
to treat their motor disabilities. disease-modifying approaches. Today, the remaining published preclinical studies in animal models.
unmet medical needs include preventing or slowing dis‑
Disease modification
ease progression (disease modification), robust alleviation Disease-modifying therapies
Efficacy of a therapy in
stopping, slowing or delaying of symptoms throughout the day while avoiding motor Recent advances in understanding the mechanisms
disease progression, and complications and managing non-motor symptoms. contributing to PD pathophysiology (BOX 3) have initi‑
therefore the clinical decline, This Review provides an overview of the under‑ ated the investigation of therapeutic approaches beyond
through an effect on the lying rationale for, progress with and associated chal‑ targeting dopaminergic pathways. However, the discov‑
underlying pathological
process (either cellular
lenges in the development of novel treatments for PD ery of therapies to halt, slow or delay PD progression
protection or restoration in recent years — looking beyond levodopa and other (through either neuroprotection or restoration of neu‑
of cellular function). dopaminergic therapies — to include both potentially ronal function), and therefore the clinical decline, has

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Neuronal pathways and motor symptoms of Parkinson disease PD notes “for a disease modifying claim a two-step
procedure is foreseen, first a delay in disease progres‑
Neural circuits of the basal ganglia are critical for motor planning and action selection, sion should be shown, second an effect on the under‑
and they are directly linked to Parkinson disease (PD) motor symptoms. A model of this lying pathological process should be established”30.
circuitry has been described in past decades202,203 and is evolving to reflect improved Unfortunately, validated biomarkers reflecting progres‑
understanding of its function204–207 (FIG. 3). Two major neuronal pathways exert
sion of the disease are still lacking, therefore disease
opposing influences on the output of this circuitry, and dopamine acts as a
neuromodulator, reinforcing the direct, stimulatory pathway and inhibiting the indirect modification can be assessed only by a delay in symp‑
inhibitory pathway. It is the adequate balance and coordination between these two tom progression in PD clinical trials20,31,32. No definitive
pathways that is important for action selection and execution of appropriate biomarker of PD has yet been identified, but research is
movements202,204,205,208. In PD, the loss of dopaminergic control leads to an imbalance in very active in this field, involving increased application
favour of a hyperactive indirect pathway and a subsequent pathological global of proteomics, metabolomics and transcriptomics32–35.
inhibition of the motor cortex, leading to bradykinesia204,205,209,210. Thus, in order to Recently, free water volume in the posterior substantia
improve the management of PD motor symptoms, a novel promising therapeutic nigra was evaluated as an imaging marker of PD pro‑
strategy is to restore the relative balance between the direct and indirect pathways gression in a 4‑year follow‑up multicentre longitudinal
instead of acting only on one of the pathways (FIG. 3). study of de novo patients with PD from the Parkinson’s
Progression Markers Initiative (PPMI) cohort study 36.
This study showed a progressive increase of free water
proved difficult 18–21. Despite the various challenges and over 4 years in patients with PD, and the 1‑year and
recent clinical trial failures, promising novel therapeutic 2‑year increases predicted subsequent long-term pro‑
strategies are beginning to emerge. gression of motor symptoms as assessed on the Hoehn
and Yahr staging system. These results suggest that free
Challenges in drug development water is a valid outcome measure in future clinical trials
Part of the challenge in developing drugs aiming to modify of disease-modifying therapies.
disease is that the pathophysiology of PD is multifactorial Potential biomarkers for the diagnosis and progres‑
and still emerging (BOX 3). Compounding this are the dif‑ sion of PD neuropathology, including clinical (olfactory
ficulties of developing reliable animal models of PD pro‑ impairment and rapid eye movement sleep behaviour
gression and neurodegeneration as well as the challenges of disorder), biochemical (α-synuclein, protein DJ1,
designing and performing appropriate clinical trials. β‑glucocerebrosidase (GBA), amyloid-β (Aβ)/tau ratio
and homovanillic acid/xanthine ratios in cerebrospinal
Disease models should reproduce the progressive nature fluid (CSF); α-synuclein, protein DJ1, reactive oxygen
of PD. Appropriate experimental conditions mimicking species (ROS), uric acid/urate or glutathione/glutathione
PD neurodegeneration need to be selected because many S‑transferase ratios in blood; and α-synuclein in colonic
animal models, including the classical toxic models, fail and/or skin biopsy samples), genetic (GBA, PINK1,
to reproduce the progressive nature of PD22. Recently, SNCA, PARK2 (also known as PRKN), PARK7 and
several α-synuclein transgenic mice have been devel‑ LRRK2) and imaging biomarkers (dopamine transporter
oped, expressing wild-type or mutated α-synuclein, but imaging with single photon emission computed tomog‑
they fail to reproduce the combination of progressive and raphy (DaT-SPECT), deformation-based morphometry
specific dopamine cell loss, protein aggregation and man‑ MRI (DBM-MRI), electroencephalography and tran‑
ifestations of PD23,24. The progress made towards a better scranial sonography) have also been reported; some are
understanding of PD pathogenesis and recent advances promising but not yet fully validated32,37. Combining
in research techniques have opened up new possibili‑ different classes of biomarkers could also increase the
ties regarding disease modelling. Thus, very recently, predictability of disease progression in patients38.
novel disease models have been described to replicate
the progressive degeneration of the nigrostriatal path‑ Clinical trial design should allow the evaluation of clini-
way, accompanied by a progressive motor dysfunction, cal progression. To detect slowing of clinical progression
in rats and primates25–27. For example, adeno-­associated in PD, patients will require follow-up in controlled clini‑
virus (AAV) vector-mediated overexpression of mutated cal studies for a substantial period of time. One option is
α-synuclein (hybrid AAV2/9 A53T human α-synuclein) to evaluate such patients in the practically defined OFF
in the substantia nigra induced a progressive degenera‑ state after withholding their symptomatic dopaminergic
tion of the nigrostriatal pathway in both rats and mar‑ medications overnight 39,40 in an effort to evaluate clini‑
mosets and a progressive motor dysfunction in rats28. The cal progression of ‘untreated’ disease. However, it is not
development of similar models has recently been initiated clear that overnight withdrawal of dopaminergic medi‑
in macaques29. Although these disease models require fur‑ cations is sufficient to wash out symptomatic effects41,42.
ther predictive validity for disease-modifying drug devel‑ Therefore, most disease-modifying clinical trials today
opment, they may represent a considerable step towards evaluate early untreated patients with PD and monitor
the translational validation of new therapies. them off symptomatic therapy for up to 9–12 months.
The main difficulty with this design is that some patients
Biomarkers reflecting progression of the disease need will require symptomatic therapy before the designated
to be validated. For regulatory purposes, the European end point. In addition, there is no guarantee that a med‑
Medicines Agency (EMA) guidelines on clinical inves‑ ication that works in early disease will work in more
tigation of medicinal products in the treatment of advanced disease. Finally, evaluation of patients with

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Targeting non-motor symptoms of Parkinson disease and faster progression of motor and cognitive deficits
after an average of 2.7 years37. Long-term predictive
Many of the motor symptoms of Parkinson disease (PD) are preceded, often by validity of those criteria remains to be established.
several years, by non-motor symptoms (NMSs) that include hyposmia, sleep disorders, Ultimately, clinical subtyping of patients using base‑
depression and constipation. NMSs, including sleep impairment, pain syndromes and line clinical information should improve the stratifica‑
neuropsychiatric symptoms (depression, impulse control disorders, hallucinations,
tion of patients for a more efficient personalized clinical
psychosis and cognitive impairment progressing to dementia), increase with advancing
age and duration of PD211,212. The neuroanatomical and neuropharmacological bases trial. A precision or personalized medicine approach in
of non-motor abnormalities in PD remain largely undefined, although some are induced combination with genetic testing is commonplace in
by current dopaminergic therapies. patient selection for cancer clinical trials and has yielded
Given the major impact of NMSs on the quality of life for patients with PD, this numerous successes. Similarly, this approach is begin‑
represents an emerging unmet need in the management of patients with PD213,214 ning to be utilized in PD clinical trials and may improve
and another major development area for the future. outcomes, considering that cohorts may be better focused
Current therapies for NMSs in PD are limited. However, the 5‑HT2A inverse agonist on participants most likely to show a measurable change
pimavanserin has been approved in the United States for the treatment of dopami- over the course of study 46,47.
metic-induced psychosis in PD (TABLE 2). Evidence in support of pimavanserin for the
management of psychosis comes from a phase III placebo-controlled trial215. The
Recent clinical trial failures
primary outcome was antipsychotic benefit as assessed by the PD-adapted Scale for
the Assessment of Positive Symptoms (SAPS‑PD), and active treatment was Several disease-modifying strategies have recently
associated with a significant reduction in psychosis. Pimavanserin was well tolerated been tested in clinical trials but have largely proved
with no significant safety concerns or worsening of motor function. to be unsuccessful. Such approaches include antioxi‑
dant agents, dopamine agonists, monoamine oxidase
type B (MAOB) inhibitors, trophic factors and anti-­
inflammatory and gene-therapy-based approaches47.
early disease does not allow for an assessment of critical The examples we discuss below have been chosen as
symptoms that cause morbidity in late disease, including they allow comparison of the outcomes that have been
balance and cognitive dysfunction. described in animal models and the results observed in
Clinical trials aimed at demonstrating disease modi‑ human studies. The negative outcomes of these phase II
fication may rely on simultaneous assessment of changes and III types of trials highlight the difficulties of dem­
in progression of clinical symptoms plus imaging mark‑ onstrating an effect in PD cohorts, but they may help
ers (for example, DaT-SPECT) over time43. Validated progress our understanding of what should define dis‑
biomarkers and additional imaging techniques (for ease modification and how to move forward with thera­
example, synuclein imaging) are being sought to enable peutically innovative concepts.
better demonstration of disease modification.
Antioxidant agents. At both a cellular and a physiolog‑
Clinical trial protocols should better stratify and select ical level, oxidative stress is considered to be a potential
patients. PD is a multifactorial neurodegenerative dis‑ driver of PD progression (BOX 3; FIG. 2). Post-mortem
order, the aetiology of which remains largely unknown, brain tissues from patients with PD have shown accu‑
and patients present and progress in different ways8,44. mulation of oxidative damage markers, especially in the
Recently, the National Institutes of Health (NIH) estab‑ substantia nigra48. Mitochondrial dysfunction has also
lished disease subtype identification as one of the top been implicated in PD pathogenesis, partly through an
three clinical research priorities in PD45. Understanding increase in generation of ROS49 (BOX 3). Oxidative stress
the pathophysiological differences among individuals may increase mitochondrial DNA (mtDNA) mutations
and defining PD subtype populations may ultimately and accelerate α-synuclein aggregation50, parkin inac‑
allow for a better definition of the target population of tivation51,52 and proteasome dissociation53. Oxidative
each treatment strategy. Improved and, where necessary, stress can also affect dopamine release and affect cellu‑
targeted patient selection may be crucial for future trials lar self-defence mechanisms54,55. These are areas ripe for
assessing disease-modifying therapies. potential therapeutic manipulation.
Well-defined criteria have not yet been widely -Recently, the Parkinson Study Group attempted to
accepted to identify PD subtypes, but by using clinical, evaluate the disease-modifying potential of coenzyme
biochemical or genetic markers (or a combination), it Q10 (CoQ10)56 on the basis of the rationale that this
may be possible to improve patient stratification for agent might reduce the oxidative stress reported in PD.
inclusion of more homogeneous cohorts into future tri‑ CoQ10 is a component of the electron transport chain
als. Fereshtehnejad et al. recently used hierarchical clus‑ involved in ATP generation, and studies in a rodent
ter analysis of data collected from the PPMI to define model of PD suggested a mild neuroprotective effect
three clinical PD subtypes (‘mild motor-predominant’, of CoQ10 (REFS57–59). The phase III, placebo-controlled
‘intermediate’ and ‘diffuse malignant’), demonstrating clinical trial aimed to assess the impact of CoQ10 on
Unified Parkinson their validity in terms of both imaging (MRI morpho‑ disease progression in early PD by means of a change
Disease Rating Scale metry data) and CSF (Aβ levels and Aβ/tau ratio) bio‑ in total Unified Parkinson Disease Rating Scale (UPDRS)
(UPDRS). Used to assess markers37. Furthermore, they demonstrated that these score in 600 early untreated patients over 16 months
severity and impact of PD signs
and symptoms. It is made up of
subtypes have longitudinal validity with the diffuse or until dopaminergic therapy was required, which‑
sections (parts I–IV) assessing malignant group, demonstrating a more profound dopa‑ ever came first. However, after a prespecified futility
different aspects of the disease. minergic deficit, increased atrophy in PD brain networks criterion was reached, the study was terminated early

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Presynaptic neuron
Gene mutations and/or SNCA post-translational
modifications, toxins, neuroinflammation, etc.
siRNA or miRNA

Anti-inflammatories
SNCA synthesis
Altered SNCA
proteostasis

SNCA clearance SNCA monomers


Immunotherapy Antioxidants

Activation of autophagy,
proteasome, proteases
and chaperones
Mitochondrial Oxidative
SNCA oligomers dysfunction stress

Endoplasmic reticulum Impairment of


stress response homeostasis
Amyloid fibrils

Impairment of Cell death


Lewy body axonal transport

Immunotherapy Trans-synaptic transmission


and prion-like propagation

Figure 2 | Pathogenic mechanisms involved in Parkinson disease and associated therapeutic strategies. Schematic
diagram depicting interactions between major molecular pathways that are implicated inNature Reviews | Drug
the pathogenesis Discovery
of Parkinson
disease (PD) and possible therapeutic strategies (grey boxes) to combat cell death. Although α-synuclein (SNCA) is
depicted as a principal player in PD pathophysiology, cell death can be induced in its absence by any of the pathogenic
mechanisms outlined (blue boxes). Intracellular levels of SNCA are tightly regulated by the balance between the rates of
synthesis, clearance and aggregation of the protein. Abnormalities altering SNCA proteostasis, including gene mutations,
post-translational modifications, external toxins, neuroinflammation or oxidative stress, may increase intracellular SNCA
levels and induce its accumulation (yellow boxes). Accumulating SNCA monomers interact to form oligomers that grow to
form amyloid fibrils and Lewy bodies (purple boxes). Cellular damage induced by the toxic forms of SNCA includes
impairment of axonal transport, mitochondrial dysfunction and endoplasmic reticulum stress response, which ultimately
induce cell death. Dysfunction in these processes can also induce cell death in the absence of SNCA and thus may
precede and cause or contribute to SNCA dysfunction or follow as a consequence of SNCA toxicity and in turn feed back
and enhance its toxicity. Toxic forms of SNCA can be transferred between cells and induce disease spreading to other
brain regions. Targeted mechanisms to confer neuroprotection include decreasing SNCA synthesis using small interfering
RNA (siRNA) or microRNA (miRNA); increasing the clearance of SNCA by activating autophagy, the proteasome, proteases
or chaperones; or blocking SNCA aggregation and spreading using immunotherapy. In addition, anti-inflammatories and
antioxidants can be used to decrease cellular stress.

Methyl-4 phenyl 1,2,3,6 when no evidence of clinical benefit of the intervention with reported potential to control mitochondrial bio‑
tetrahydropyridine treatment was demonstrated56. Likewise, a long-term genesis and oxidative stress by inhibiting the activation
(MPTP). A neurotoxin that placebo-controlled study of creatine monohydrate was of microglia and astrocytes and the production of pro-­
causes irreversible loss of terminated after a median follow‑up of 4 years owing inflammatory cytokines and nitric oxide60. Preclinical
dopamine neurons and
parkinsonism in mice, monkeys
to futility. In this study, futility was defined as a failure studies performed with pioglitazone in Methyl-4 phenyl
and humans. The neurotoxicity to detect a difference in clinical decline, measured by 1,2,3,6 tetrahydropyridine (MPTP) monkeys have shown
of MPTP was first discovered a global statistical test that included five clinical status neuroprotection and improvements of motor symptoms
when a young man outcome measures. Here, creatine was assessed versus that were qualified as modest by the study authors61.
self-injected an illicit narcotic
placebo in 1,741 patients with early PD who had been In phase II, pioglitazone failed to show any disease-­
(desmethylprodine (MPPP))
containing MPTP as a major on dopaminergic treatment for 90 days to 2 years, and modifying activity in the subsequent 44‑week placebo-­
impurity. He developed PD patients were monitored for 5–8 years, during which controlled study in 210 patients with early PD on a
motor symptoms and time treating physicians used all available medications stable dosage of an MAOB inhibitor (rasagiline or sele‑
responded to levodopa to provide the best response19. giline) assessing UPDRS scores62. Moreover, the selected
therapy, and the autopsy
showed selective degeneration
Another antioxidant approach is to target peroxi‑ peripheral biomarkers for PD progression, including
of dopaminergic neurons of some proliferator-activated receptor-γ (PPARγ) with the leukocyte PPARγ coactivator 1α (PGC1α) and target
the substantia nigra. agonist pioglitazone, which belongs to a class of drugs gene expression, plasma interleukin 6 (IL‑6) as a marker

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of inflammation and urine 8‑hydroxydeoxyguanosine neurons, which resulted in robust improvements of


(8OHdG) as a marker of oxidative DNA damage, did not motor function65–67. However, several clinical trials failed
correlate with UPDRS changes62,63. to reproduce these beneficial effects but allowed for the
In the CoQ10 and pioglitazone examples, the neuro‑ progressive improvement of gene delivery protocols68,69.
protective effects described in preclinical animal models In a recent double-blind, randomized controlled trial,
were of mild amplitude. This finding may explain why gene delivery of NRTN was improved by utilizing an
these two compounds failed to produce significant benefit AAV2 vector that was genetically engineered to express
for patients in clinical trials. and secrete the human gene for NRTN (CERE‑120). This
agent was then injected directly into the putamen and
Gene therapy-based strategies. Many gene therapy substantia nigra of patients with advanced PD. However,
approaches aim to rescue degenerating neurons by this therapy was not shown to affect clinical outcomes in
delivering neurotrophic factors or to restore function the trial. The study end point was the change from base‑
by delivering key enzymes involved in or required for line to final visit at 15 months in the motor subscore of
dopamine synthesis and metabolism64. the UPDRS in OFF state, but no difference was observed
Early trials of disease-modifying gene therapy met between AAV2–NRTN-treated and sham-operated
with a number of obstacles. In preclinical studies using patients in this or any of the secondary end points of the
animal models of PD, injection of glial cell line-derived study 70. Human studies suggest that Ret, the receptor for
neurotrophic factor (GDNF) and neurturin (NRTN) were GDNF and NRTN, is downregulated in PD71, and this
found to enhance function and protect dopaminergic has been shown to result from α-synuclein accumulation

Box 3 | Emerging pathogenic mechanisms of Parkinson disease


Parkinson disease (PD) is an age-related neurodegenerative disorder, characterized by progressive and selective loss
of dopaminergic neurons in the substantia nigra and by Lewy body deposition in the midbrain, with widespread
involvement of other central nervous system structures and peripheral tissues. PD pathology is composite and due to
the action of genetic and environmental factors. The precise molecular mechanisms causing neuronal loss are still not
fully understood, but intensive research has helped uncover several pathways and mechanisms involved in PD
pathophysiology, establishing a vicious cycle in which these mechanisms exacerbate each other. The usual suspects
are the following:
• α-Synuclein aggregation. α-Synuclein exists in various conformations in a dynamic equilibrium that is modulated by
many factors, including oxidative stress, post-translational modifications and concentrations of fatty acids,
phospholipids and metal ions, and a tight balance of these factors controls the levels and aggregation of α-synuclein
(for a review of our current knowledge on α-synuclein, refer to10,216). α-Synuclein may acquire neurotoxic properties
during a pathogenetic process in which soluble monomers initially form oligomers then progressively combine to form
small protofibrils and further aggregate into large, insoluble fibrils forming Lewy bodies (FIG. 2).
• Prion-like cell‑to‑cell transmission of α-synuclein (possibly invading via the gut–brain axis). Several lines of
experimental evidence suggest that pathogenic misfolded α-synuclein is released into the extracellular space,
internalized by neighbouring neurons and seeds aggregation of endogenous α-synuclein once inside its new cellular
host217,218. Thus, initial α-synuclein misfolding in a small number of cells could progressively lead to the spread of
α-synuclein aggregates to multiple brain regions over years219. However, definitive proof of this intercellular
propagation has proved difficult to secure220.
• Mitochondrial dysfunction. Dysfunction of complex I (due in part to mitochondrial accumulation of α-synuclein),
change in membrane potential, impairment of mitochondrial quality control, disruption of calcium homeostasis and
enhanced release of cytochrome c (reviewed in detail in49,221,222).
• Oxidative stress, which is intimately linked to mitochondrial dysfunction and to which nigral dopaminergic neurons are
particularly vulnerable223–225. Mitochondrial dysfunction and increased oxidative stress can lead to functional
impairment of the lysosomal autophagy system, further demonstrating that several putative pathogenetic pathways in
PD are intimately linked226.
• Severe endoplasmic reticulum stress, resulting from the accumulation of misfolded or unfolded proteins and
contributing to neuronal death or apoptosis5,227.
• Impairment of autophagy, which may boost α-synuclein release and transfer from cell to cell228 and lead to the
accumulation of dysfunctional mitochondria229.
• Dysfunction of axonal transport, potentially due to the energy deficiency (due to mitochondrial dysfunction) and/or the
presence of α-synuclein aggregates becoming obstacles to normal axonal transport230,231.
• Motor circuit pathophysiology, with abnormal neural synchronization in the basal ganglia, and synaptic alterations,
in particular in corticostriatal plasticity, with both presynaptic and postsynaptic modifications232–234. Changes in
cerebellar circuits and in interactions between the cerebellum and the basal ganglia are also considered important in
the pathophysiology of PD235. The motor circuit pathophysiology results from dopamine loss but also from disturbances
in other neurotransmitters, in particular cholinergic and glutamatergic transmitters236,237.
• Neuroinflammation, with intense astrogliosis and microgliosis that may be associated with abnormal corticostriatal
plasticity. Although perhaps not the initial trigger, neuroinflammation is suspected to be an essential contributor to
PD pathogenesis238.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Non-dopaminergic therapies currently in development for PD modification


Compound and/or agent, Mechanism of action Phase of Clinical trial Refs
company development number
AXO-LENTI‑PD (OXB‑102; Triple gene therapy (Lenti‑TH– Phase I/IIa in ProSavin: 76,77,

ProSavin analogue with AADC–GTPCH) to restore striatal preparation • NCT01856439 79,80

increased level of transgene dopamine production • NCT00627588


expression), Oxford BioMedica
and Axovant Sciences
VY‑AADC, Voyager Gene therapy (AAV2–hAADC) Pivotal phase II/III • NCT01973543 81–85

Therapeutics to restore striatal dopamine ongoing • NCT03065192


production • NCT03562494
PRX002 (RO7046015), Passive immunotherapy targeting Phase II ongoing NCT03100149 88–91

ProThena and Roche α-synuclein


PD01A and PD03A, Affiris Active immunotherapy targeting Phase I • NCT01885494 88,92–94

α-synuclein completed • NCT01568099


• NCT02216188
• NCT02618941
• NCT02267434
BIIB054, Biogen and Passive immunotherapy targeting Phase II ongoing NCT03318523 95,96

Neurimmune aggregated α-synuclein


NPT200‑11, UCB and α-Synuclein aggregation Phase I NCT02606682 98

Neuropore Therapies modulator completed


Ambroxol, Lawson Health Increase glucocerebrosidase Phase II in • NCT02941822 120,122

Research Institute and Van activity preparation • NCT02941822


Andel Research Institute
LTI‑291, Allergan and Lysosomal Glucocerebrosidase activator Phase II ongoing NTR6960 123

Therapeutics (EudraCT
2017‑004086‑27)
Venglustat (GZ/SAR402671), Glucosylceramide synthase Phase IIa ongoing NCT02906020 124

Sanofi and Genzyme inhibitor in patients with PD


carrying a GBA mutation
Deferiprone, ApoPharma Iron chelator Phase II ongoing • NCT02655315 130

• NCT02728843
Exenatide, National Institute GLP1 agonist Phase I ongoing NCT03456687 39,133

of Neurological Disorders and


Stroke
Liraglutide, The Cure Parkinson’s GLP1 agonist Phase II ongoing NCT02953665 240

Trust and Novo Nordisk


DNL151 (backup of DNL201), LRRK2 inhibitor Phase I ongoing NA 141

Denali Therapeutics
KM‑819 (KR33493), Kainos FAF1 inhibitor to protect Phase I ongoing NCT03022799 144

Medicine dopaminergic neurons from


apoptosis
AAV2, adeno-associated virus type 2; FAF1, Fas-associated factor 1; GLP1, glucagon-like peptide 1; Lenti, lentiviral vector;
LRRK2, leucine-rich repeat serine/threonine-protein kinase 2; NA, not available; PD, Parkinson disease.

in rat models of α-synucleinopathy 71,72. Thus, it is pos‑ Emerging disease-modifying therapies


sible that, in the case of neurotrophic factors, benefits Despite the succession of past failures of disease-­
observed in MPTP monkeys do not translate into ben‑ modifying approaches in clinical trials, research
efits in patients because this model does not recapitu‑ efforts in the pursuit of neuroprotective strategies in
late the accumulation of α-synuclein found in PD. It is PD are still very intense. Increased understanding of
also possible that the patients enrolled in the clinical the pathogenetic mechanisms underlying PD (BOX 3)
trials had disease that was too advanced to benefit from have prompted the development of new therapies
neurotrophic factor therapy. Indeed, to limit misdiagno‑ and identified novel targets, including gene delivery
sis and to have clinical disease sufficiently advanced to of enzymes involved in dopamine metabolism, glu‑
observe benefit, inclusion criteria often include a mini‑ tathione replacement, iron chelators and glucagon-like
mum 5‑year duration of PD from diagnosis (average of peptide 1 (GLP1) agonists.
7.8 years in the NRTN-treated patient cohort)70. Notably, In the past 10 years, 24 loci have been identified
there might not be sufficient neuronal fibres remaining as risk factors for sporadic forms of PD73, and sev‑
for growth factors to be beneficial in these patients. eral drugs that target the related proteins are now in

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Non-dopaminergic therapies currently in development for the symptomatic treatment of PD


Compound and/or agent, company Mechanism of action Phase of development Clinical trial Refs
number
Motor fluctuations
Istradefylline (KW‑6002), Kyowa Adenosine receptor • Approved in Japan • NCT01968031 155–159

Hakko Kirin Pharma type 2A antagonist • Not approved by the • NCT02610231


FDA in February 2018
Dyskinesia
Amantadine ER (ADS‑5102 (Gocovri)), NMDA receptor • Approved in United NCT02136914 183,184

Adamas Pharmaceuticals antagonist States


• Phase III ongoing
Dipraglurant (ADX48621), Addex mGlu5 NAM Phase III in preparation NCT01336088 189–191

Therapeutics
Buspirone, Assistance Publique, 5‑HT1A and Phase III ongoing • NCT02617017 196,241

Hôpitaux de Paris, Oregon Health and α1-adrenergic receptor • NCT02803749


Science University and University of agonist • NCT02589340
Rochester
Eltoprazine, Amarantus BioScience 5‑HT1A/1B receptor Phase IIb in preparation NCT02439125 197,198

Holdings agonist
Motor fluctuations and dyskinesia
Foliglurax (PXT002331), Prexton mGlu4 PAM Phase IIa ongoing NCT03162874 192–194,

Therapeutics and Lundbeck 242,243

Other symptoms
Varenicline (gait and balance, excessive Partial agonist of • Phase II ongoing for • NCT01341080 244,245

daytime sleepiness), Rush University the α4β2 nicotinic gait and balance • NCT02473562
Medical Center and VU University acetylcholine receptor • Phase IV ongoing for
Medical Center sleep
Pimavanserin (formerly ACP‑103) 5‑HT2A receptor inverse Approved in United • NCT00550238 215,

(psychosis), Acadia Pharmaceuticals agonist States • NCT01174004 246–249

• NCT00477672
SYN120 (PD dementia), Acorda 5‑HT6/2A receptor Phase II ongoing NCT02258152 250

Therapeutics antagonist
5‑HT, serotonin; D1, dopamine receptor D1; FDA, US Food and Drug Administration; mGlu, metabotropic glutamate receptor; NAM,
negative allosteric modulator; NMDA, N‑methyl-d‑aspartate; PAM, positive allosteric modulator; PD, Parkinson disease.

early development. Some promising approaches aim MPTP macaques77. However, the study had no placebo
to address mutations in genes encoding α-synuclein, arm, and it is known that in PD, the placebo effect can
leucine-rich repeat serine/threonine-protein kinase 2 be significant, especially with a surgical intervention78.
(LRRK2), GBA and parkin47,74,75. Although the results were encouraging, some patients
displayed less than 50% improvement in the UPDRS
Gene therapy. Among the emerging and promising gene part III motor score and continued to require a sub‑
therapies for PD are novel agents that aim at delivering stantial daily dose of oral levodopa therapy to achieve
key proteins involved in dopamine metabolism directly maximal benefit. It was then concluded that higher lev‑
into the basal ganglia (TABLE 1). ProSavin (also known els of dopamine replacement might be required79 and
as OXB‑101) is a viral vector that offers a restorative a more potent vector construct (OXB‑102, now AXO-
approach by delivering three key enzymes involved in Lenti‑PD) has been developed to increase the level of
the synthesis of dopamine76. This triple gene therapy transgene expression79. A phase I/II dose escalation
has demonstrated strong improvement in parkinso‑ study is expected to be initiated by the end of 2018 in
nian motor symptoms and restoration of approximately patients with advanced PD80. In the meantime, longer-
50% of normal striatal dopamine concentrations in term safety and efficacy evaluation of ProSavin has been
MPTP macaques77. Therefore, ProSavin has been eval‑ pursued (NCT01856439).
uated in a small-scale phase I/II open-label study in 15 Another gene therapy approach currently being inves‑
patients with PD. Treatment was injected directly into tigated is the use of the recently developed VY‑AADC
the putamen and patients were monitored for a period vector, which provides AAV2–hAADC gene delivery
of 12 months. ProSavin was reported to be well toler‑ with the aim of restoring the loss of DOPA decarboxy‑
ated and offered patients significant dose-dependent lase (DDC; also known as AADC) in nigrostriatal dopa‑
improvements at 6 and 12 months in UPDRS motor mine neurons that occurs in advanced PD81. VY‑AADC
scores when compared with baseline78, which was con‑ is given as a single treatment that aims to enable neurons
sistent with the results of preclinical efficacy studies in of the putamen to express the AADC enzyme and thus

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

offer a durable therapy that enhances the conversion of mouse models of PD or dementia with Lewy bodies88,89.
levodopa to dopamine. A study in non-parkinsonian The tolerability and safety of a single intravenous infu‑
monkeys showed that MRI-guided infusion of sion of PRX002 was demonstrated in a phase I clinical
VY‑AADC into the midbrain was feasible and tolerable, trial in healthy volunteers90 and in a 12‑week phase Ib
without any assessment of efficacy at this stage82. After multiple-­ascending-dose study of PRX002 in patients
demonstration of the clinical safety of this approach83, a with early PD. This study demonstrated central nerv‑
phase Ib trial was initiated and is currently ongoing in ous system (CNS) penetration of PRX002 and dose-
15 patients with advanced PD (NCT01973543). Interim dependent and time-dependent reduction in levels of
results presented in 2017 at the annual meeting of the free serum α-synuclein, but effected no reduction in
American Academy of Neurology (AAN) and at the CSF α-­­synuclein91. However, no efficacy results have
American Association of Neurological Surgeons (AANS) been described as yet. A randomized, double-blind,
annual scientific meeting suggest that this treatment placebo-controlled phase II study was initiated by Roche
effects a durable, dose-dependent and time-dependent in July 2017 to evaluate the efficacy of monthly intra­
increase in AADC activity, as shown by positron emis‑ venous PRX002 over 52 weeks in patients with early PD.
sion tomography (PET) imaging using 18F-fluorodopa, The primary end point will be change from baseline in
which correlates with improvement of motor function MDS-UPDRS (a Movement Disorder Society (MDS)
(assessed by ON/OFF times and UPDRS part II and III revised version of the UPDRS) total score at week 52.
scores)84. Because of pulmonary embolism and related Neurodegeneration will be assessed by dopamine
heart arrhythmia reported in one treated subject, deep transporter imaging (DaT-SPECT) (NCT03100149).
vein thrombosis prophylaxis was added to this protocol, In parallel to the development of this passive
and a separate phase I study designed to further optimize immunotherapy, Affiris has developed two active
the intracranial delivery of VY‑AADC has been per‑ vaccines with short α-synuclein-mimicking pep‑
formed. The US Food and Drug Administration (FDA) tides — Affitopes PD01A and PD03A. This approach
has cleared investigational new drug (IND) application has shown efficacy in transgenic mouse models for
status and granted regenerative medicine advanced decreasing α-synuclein oligomers in axons and syn‑
therapy (RMAT) designation for a pivotal phase II/III apses and reducing loss of dopaminergic neurons88,92.
programme for VY‑AADC, which is currently ongo‑ In June 2017, top-line results of a 36‑week phase I clin‑
ing and is expected to conclude in late 2020/early 2021 ical study were announced at the 21st International
(NCT03562494)85. Congress of Parkinson’s Disease and Movement
The need for surgical injection of current gene thera­ Disorders93. It was reported that Affitope PD03A may
pies is likely to make these approaches unsuitable for be safe and well tolerated in patients with early PD and
patients with early stages of PD. Improving the modes indicates a dose-dependent immune response against
of delivery and rendering them less invasive will be key the peptide itself and cross-reactivity against the
to the advancement of gene therapy approaches in the α-synuclein targeted epitope. In March 2018, long-term
treatment of PD. Development of gene therapy that data were presented at the Advances in Alzheimer dis‑
directly affects progression of PD pathology and clinical ease and Parkinson disease Therapies (AAT-ADPD)
course might still be entertained as a possible treatment Focus Meeting, reporting that both tested doses of
for early PD in the future. PD03A induced peak titres 4 weeks after the third
injection, which were reactivated by the boost injection
α-Synuclein-targeted therapies. Common variants in at week 36 (REF.94). These results provide an encouraging
the gene encoding α-synuclein are believed to act as risk basis for further clinical development.
factors for sporadic forms of PD and, in all patients with Several unanswered questions remain regarding
PD, there is toxic aggregation of α-synuclein86 (FIG. 2). the application of immunotherapies for CNS disorders
Three approaches to therapeutically interfere with and chronic conditions such as PD, including whether
α-synuclein toxicity in PD pathology have been proposed: antibodies reach the brain compartment at sufficient
blocking α-synuclein spreading using immunotherapy; levels and are able to exert the desired effect on the tar‑
inhibiting α-synuclein aggregation; and increasing the get protein and for how long and with what amplitude.
clearance of α-synuclein by either decreasing its produc‑ Assessments of such antibodies in patients in the coming
tion or promoting its degradation6,10,87 (FIG. 2). years will bring a decisive part of the answer.
In the past decade, several research groups have Other agents targeting α-synuclein are also in clinical
ON/OFF worked extensively towards the development of anti-­ development. Among them, another passive immuno‑
Alternating periods of good α-synuclein antibodies or a α-synuclein vaccine as therapy agent, BIIB054, recognizes the aggregated form
(ON) and poor (OFF) control of potential disease-modifying treatments. Researchers of α-synuclein and a phase II study is underway 95,96. An
motor symptoms by levodopa
have developed a passive immunotherapy, PRX002 alternative strategy consists of preventing or decreasing
treatment. ON refers to time
when medication is providing (also known as RG7935 or RO7046015), a humanized α-synuclein aggregation using oligomer modulators,
benefit with regard to mobility, immuno­g lobulin G1 (IgG1) monoclonal antibody such as anle138b, which binds to a structure-dependent
slowness and stiffness. directed against the carboxy terminus of α-synuclein epitope and inhibits formation of pathological α-­synuclein
OFF refers to time when a with higher affinity and avidity for aggregated than oligomers in vitro and in vivo97. Although this compound
medication’s efficacy has worn
off and is no longer providing
monomeric forms. Its murine homologue 9E4 has been is still in preclinical validation in animal models26, another
benefit with regard to mobility, shown to reduce α-synuclein pathology and to improve oligomer modulator, NPT200‑11, which is designed to
slowness and stiffness. memory and motor functions in several transgenic alter α-synuclein interactions with membranes, thereby

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

reducing oligomer formation98,99, has been advancing Glutathione replacement. Reduced glutathione appears
through phase I trials. A single-­ascending-dose study was to be involved in scavenging ROS and cellular detoxifi‑
completed in early 2016 (REF.100). However, data from this cation, and this tripeptide appears to be depleted early
study are not available, and further plans for the devel‑ in the course of PD109. There has therefore been interest
opment of this compound have not been announced. in attempting glutathione replacement in patients with
Potential benefits of these agents are those of small mol‑ PD, although delivering sufficient levels of therapy to
ecules, with them being more likely to reach the brain the brain has been difficult thus far because of its short
than monoclonal antibodies, without the risk of being half-life of around 2.5 min in plasma and negligible oral
neutralized by host antibodies. However, these candi‑ bioavailability in humans110. Early-phase clinical studies
dates face the challenge of being able to achieve suffi‑ have failed to show appreciable benefits for treatment
cient levels in the right location in the brain to prevent — in terms of changes in UPDRS scores — as compared
α-synuclein misfolding. with placebo111,112. Although nasal delivery, as used in
Additional approaches aimed at targeting toxic these studies, has been proposed as a potentially useful
α-synuclein accumulation in PD are also being investi‑ approach for delivering glutathione to the brain113, it is
gated. Feasibility to reduce α-synuclein levels by using still unknown whether glutathione administration in its
gene-­silencing mechanisms (small interfering RNA current form allows glutathione to remain in the brain
(siRNA)) in the primate brain has been shown in squirrel for sufficient time and at sufficient levels to produce a
monkeys101. In this study, the siRNA molecule directed therapeutic effect. This may represent the biggest current
against α-synuclein was directly infused into the left challenge of this approach, and further dose–response
substantia nigra of non-parkinsonian monkeys and it and steady-state administration studies will be required
resulted in a 40% suppression of α-synuclein expression to optimize glutathione delivery before evaluating thera­
when compared with expression in the untreated intact peutic efficacy.
hemisphere. No assessment of antiparkinsonian efficacy
has been performed at this stage101. This approach may be Lysosomal pathway and GBA activators. Current esti‑
particularly attractive for patients who carry SNCA gene mates indicate that 5–10% of patients with PD carry
duplication or triplication. Recent reports of gene silenc‑ mutations in GBA that result in deficiency of the lysoso‑
ing tested in phase III for spinal muscular atrophy102–104, in mal enzyme114,115 (a condition named Gaucher disease),
phase I for Huntington disease105,106 and the first approval which makes GBA mutations the most common genetic
by the FDA of a siRNA therapy (patisiran, for the treat‑ risk factor for PD and an area of very active research.
ment of hereditary transthyretin-mediated amyloidosis) The clinical presentation of PD associated with GBA
are very encouraging for other disorders such as PD. mutations is almost identical to idiopathic PD, except
Approaches that are currently being investigated as for a slightly younger age of onset and a tendency for
possible therapies for PD but are further from clinical more cognitive impairment 116. Interestingly, there is
trials include strategies to modulate levels and activity a reciprocal relationship between GBA activity and
of chaperones such as heat shock proteins (for a review α-synuclein function: enhancing GBA activity may lead
see REF.107) and small molecules that enhance auto‑ to regulation or even attenuation of the formation of mis‑
phagy (see REF.108). Despite the strong interest in these folded oligomeric α-synuclein through glycosphingo­
novel anti-α-synuclein approaches, it remains to be seen lipid clearance117–119, supporting the development of GBA
whether enhancement of clearance mechanisms can augmentation therapies for PD.
prove effective in patients with PD, in whom clearance However, a key challenge facing GBA-related thera­
mechanisms may be so far deteriorated by the time clas‑ pies is the limited understanding of the precise mech‑
sical PD motor symptoms emerge that the potential to anism by which GBA mutations increase the risk of
provide meaningful clinical benefit is limited. synucleinopathy and accelerate disease progression.
Although promising, there are a number of chal‑ Thus, there may be fundamental differences between
lenges facing the development of therapies aimed at PD forms that are due to GBA mutations and those that
preventing α-synuclein aggregation and propagation75. are due to other causes, and uncertainty remains regard‑
First, there is no animal model that naturally replicates ing the extrapolation of results in a wider heterogeneous
α-synucleinopathy in humans, and α-synuclein has not population. Therefore, trials should first study patients
proved to correlate with neurodegeneration. Second, with GBA mutations, which is difficult owing to limited
there is currently no established method to assess target numbers of patients with PD having been screened for
engagement in the brain for anti-α-synuclein therapies. these mutations, followed by further studies to assess the
As a consequence, it is difficult, for example, to define response in a more heterogeneous patient population.
optimal dosing for clinical trials. Finally, it is not known Several phase II studies targeting GBA are cur‑
when during the natural history of PD the synucleinop‑ rently ongoing. Ambroxol is a small-molecule chap‑
athy appears and whether there is a ‘point of no return’ erone that has been shown to increase GBA activity
beyond which the damage to the neural system can no in vitro and in vivo and to decrease α-synuclein pro‑
longer be protected or revived. tein levels120. Ambroxol’s effects on neurodegener‑
Although several pressing knowledge gaps remain, ation were not assessed in animal models, but it is
these are exciting times for α-synuclein-targeted thera‑ expected that reducing α-synuclein levels might be
pies, with large clinical trials currently underway. These of therapeutic importance in PD. A phase II study is
studies will bring informative outcomes to the field. planned with ambroxol as a potential therapy in patients

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

with PD (NCT02941822) 121,122. LTI‑291 is another Exenatide is a licensed drug for the treatment of
GBA activator that is moving to a phase II study in T2DM. Exenatide is a GLP1 receptor agonist with
patients with PD carrying GBA mutations (NTR6960, anti-inflammatory and antioxidant effects that improves
EudraCT2017‑004086‑27) in the Netherlands123. glucose control by stimulating insulin release from the
Other strategies target the same lysosomal pathway. pancreas and inhibiting glucagon release133. Encouraging
Venglustat (GZ/SAR402671) is a glucosylceramide syn‑ pilot results have recently been reported with exenatide in
thase inhibitor, thus targeting the reduction of glyco­ PD39. Indeed, exenatide had positive effects on the prac‑
sphingolipid levels in patients with PD with a mutation tically defined off-medication state that were sustained
of GBA124. Venglustat is currently being assessed in a for 12 weeks after cessation of the exenatide treatment,
phase II study in patients with early-stage PD carrying which suggests disease modification39. However, method‑
a GBA mutation (NCT02906020)125. ological issues such as the small sample size (62 patients
Results of these ongoing phase II studies, and fol‑ with moderate PD), single-centre design, selection of
low-up studies, will be very informative to determine overnight washout UPDRS scores that may or may not
whether the lysosomal pathway may lead to novel treat‑ reflect ‘untreated’ disease and rapidity of the occurrence
ments for PD, with or without GBA mutations, despite of the between-group differences prevent robust conclu‑
the previous failure of afegostat (AT2101; also known as sions39,134. An additional clinical trial with exenatide is
plicera) in a phase II trial in 2009 (REF.126). currently ongoing with a small number of patients with
PD to assess the effects of a 1‑year treatment on qual‑
Iron chelators. In PD, excess iron is detected in the brain ity of life and depression, strength and motor function,
— primarily in the substantia nigra pars compacta, where cognition and functional and structural brain MRI135. A
dopaminergic neurons are exposed to high levels of ROS phase II trial is also currently ongoing with liraglutide,
produced by the metabolism of dopamine — and corre‑ an analogue of human GLP1 with longer half-life that
lates with disease severity 127. The underlying mechanism is licensed for T2DM, as a single-centre, double-blind,
of overdeposition of iron in PD brain remains unknown. placebo-controlled study with 57 patients with PD136.
However, iron chelation has been proposed for confer‑ Further larger studies are required in order to assess
ring neuroprotection in PD128,129. Recently, deferiprone, whether targeting GLP1 truly represents a durable and
a membrane-permeant chelator of labile iron, was shown meaningful neuroprotective effect of treatment in PD.
to dose-dependently relieve the oxidative damage gen‑
erated by MPTP in mice and to improve their motor LRRK2 inhibitors. Among the genes involved in PD, the
functions130. A subsequent 12‑month, single-centre, pilot LRRK2 gene is considered a key player 137. Mutations in
double-blind placebo-controlled trial with deferiprone LRRK2 in PD are associated with pathological kinase
was performed in 40 patients with early PD who were activity in cultured neurons — an observation that
already stabilized for dopamine regimens (the FAIR prompted interest in the development of drugs that
PARK I study). On the basis of a 6‑month delayed-start could act as LRRK2 kinase inhibitors. However, the
(DS) paradigm, early-start patients compared with DS development of agents targeting LRRK2 faces a number
patients responded significantly earlier and sustainably of challenges.
to treatment in both substantia nigra iron deposits, deter‑ A large number of LRRK2 kinase inhibitors have
mined by MRI, and UPDRS motor indicators of disease been studied in vitro, but an appropriate transgenic
progression130. Deferiprone is now being tested further in animal model in which to study these agents in vivo is
a phase II clinical trial in 338 patients with early PD over still lacking. Moreover, issues regarding the safety of
9 months (FAIR PARK II study, NCT02655315). Notably, LRRK2 inhibition emerged from preclinical studies
modifying iron metabolism in vivo in humans may reporting potential undesirable effects in the lung and
induce a number of undesirable adverse events, affecting kidneys138,139. There are also concerns that measuring the
haematological or other systemic parameters. This pos‑ effects of LRRK2 inhibition by assessing changes in phos‑
sibility is why the concept of a chelation strategy aimed phorylation of serine residues may not be an appropriate
at reducing oxidative damage associated with regional end point, that there are no central measures of inhibi‑
iron deposition without affecting circulating metals is tor target engagement that are suitable for human drug
appealing with drugs such as deferiprone. However, as trials and that there is no clear target patient population
for antioxidant strategies, iron chelation remains to be in which to study the effects of drugs with this mode of
validated in humans as a therapeutic strategy in PD. action137. In addition, it may be difficult at this time to
conduct large clinical trials that enrol only patients with
GLP1 agonists. Epidemiological and clinical data suggest PD with LRRK2 mutations as they only represent a small
that PD and type 2 diabetes mellitus (T2DM), both age- proportion of the PD population, and many patients with
related diseases, share similar dysregulated pathways, PD have not undergone genetic testing. However, in a
suggesting common underlying pathological mechanisms recent study, Di Maio and colleagues140 developed a novel
Type 2 diabetes mellitus (reviewed in REF.131). In its earliest stage, T2DM develops assay and showed that wild-type LRKK2 kinase activity
(T2DM). Type of diabetes that from insulin resistance, leading to a variety of detrimental was enhanced in dopaminergic neurons and in micro‑
is characterized by insulin effects on metabolism and inflammation. Similar dysreg‑ glia in post-mortem substantia nigra tissue from patients
resistance in appropriate
hepatic glucose production
ulation of glucose and energy metabolism also appears to with idiopathic PD. This suggests that LRKK2 inhibitors
and impaired insulin secretion. be an early event in PD132, and loss of insulin signalling in may be beneficial not only for mutation carriers but for
Onset is usually after 40 years. the brain may contribute to PD pathogenesis131. the broader sporadic PD population.

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The first small-molecule LRRK2 inhibitor, DNL201, side effects of dopaminergic agents (such as motor fluctu‑
reached clinical testing in 2017 and showed inhibition ations and dyskinesia, daytime sleepiness, nausea, ortho‑
of LRRK2 kinase activity in a healthy volunteer phase I static hypotension and impulse control disorders), thus
study 141. Another LRRK2 inhibitor, DNL151, is currently addressing the unmet needs of present PD therapies.
being assessed in healthy volunteers in the Netherlands in It should be noted that often many years before the
order to select the most promising molecule to be assessed onset of motor symptoms of PD, non-motor symptoms
in patients with PD carrying an LRRK2 mutation141. occur, which can ultimately have a major impact on
the quality of life of patients with PD, and represent an
FAF1 inhibitors. Mutations in the parkin gene (PARK2) emerging unmet need (BOX 2).
are associated with early-onset PD142. The discovery that
parkin normally acts as an inhibitor of the proapoptotic Adenosine receptor type 2A antagonists
Fas-associated factor 1 (FAF1) by promoting its degra‑ Adenosine receptor type 2A (A2A) is mainly expressed
dation recently led to the novel therapeutic approach of within the caudate and putamen nuclei of the basal gan‑
inhibiting FAF1 (REF.143). KM‑819 (also named KR33493), glia. Blockade of A2A in striato-pallidal neurons reduces
a small-molecule inhibitor for FAF1, is now entering a the postsynaptic effects of dopamine depletion and the
phase I clinical trial in Korea (NCT03022799) (TABLE 1). excessive firing of striato-pallidal neurons, thus sug‑
In MPTP mice, KM‑819 slightly increased striatal dopa‑ gesting a potential for A2A antagonists to improve motor
mine transporter activity as measured by PET studies, as deficits in PD153 (FIG. 3). Clinical trials to date have shown
described in a symposium abstract 144. variable efficacy to improve motor fluctuations154.
Istradefylline (KW‑6002) was the first A2A antago‑
Emerging symptomatic treatments nist evaluated as an adjunctive therapy with levodopa in
Despite the intense research focus on disease-modifying patients with PD with motor fluctuations. When char‑
therapies, traditional pharmacotherapeutic concepts are acterized in MPTP-lesioned macaques, istradefylline
still worthwhile because symptomatic therapy for con‑ increased ON time but did not improve PD scores and
trol of motor symptoms with dopaminergic and non-­ exacerbated dyskinesia155. This finding is in accordance
dopaminergic drugs is far from optimal. with phase II/III clinical findings in which istradefylline
The research of treatments to address motor symp‑ decreased OFF time but had no effect on UPDRS score
toms of PD is privileged by the growing knowledge on and increased dyskinesia156,157. Istradefylline was licensed
the functioning of the basal ganglia motor circuit and in Japan in 2013 for adjunctive treatment use in patients
the cause of PD motor symptoms (BOX 1) as well as the with PD experiencing wearing-off fluctuations, but the
availability of good animal models for this particular FDA has requested additional data for the United States.
indication. This understanding has led to the identifica‑ Expected timing of resubmission is in 2018, although the
tion of novel non-dopaminergic strategies that may be last global phase III study turned out to be negative158,159.
combined with dopaminergic treatments to improve the Preladenant was the second A2A antagonist evalu‑
motor functioning of patients with PD. ated in patients with PD. In MPTP-lesioned macaques,
The gold-standard model of PD motor symp‑ when added to a suboptimal dose of levodopa, prelade‑
toms is produced by administration of MPTP into old nant improved PD scores but could not reach the level
world non-human primate species (Macaca mulatta of efficacy of optimal levodopa. Preladenant produced
and Macaca fascicularis). With a close resemblance to a significant decrease in PD score in only half of the six
humans in physiology and brain anatomy, the PD motor MPTP macaques included in the study160. Again, this is in
features in macaques give an accurate model for PD accordance with the observations made in clinical trials:
clinical motor signs24,26,145,146. MPTP-lesioned primates after initial positive phase II results161, reduction of OFF
respond to dopaminergic drugs, including levodopa, time did not reach significance compared with placebo
and develop the same motor complications as patients in large phase III trials162. The phase III trial also failed to
with PD, including LID. This model has been widely and show the efficacy of the comparator therapy rasagiline,
successfully used to study the effects of drugs targeting thus rendering difficult the interpretation of the global
the symptoms of PD, including levodopa, dopamine ago‑ outcome. However, the development of preladenant for
nists147, MAOB inhibitors148 and many others149, as well PD has been abandoned.
as side effects including motor fluctuations and LID. A third A2A antagonist, tozadenant (SYN115), was
Consequently, the MPTP primate model and the use of recently evaluated in a phase III study (NCT02453386)
clinically relevant end points have been instrumental for after positive results in a phase IIb study 163. However,
evaluating the efficacy of symptomatic therapies. development of this agent has been terminated follow‑
Below, we review some of the emerging drugs being ing serious haematological adverse events resulting in
investigated for their efficacy in treating motor symptoms deaths164. Therefore, at the moment, there is no perspec‑
(TABLE 2). Some novel dopaminergic agents are currently tive for new A2A antagonists reaching the market in the
in development, but a full summary of such agents is near future.
outside the scope of this current Review, which focuses
on non-dopaminergic agents (for a review of current Agents targeting the glutamatergic system
dopaminergic agents developed for PD, see REFS6,150–152). Glutamate operates through two classes of receptors:
It is expected that non-dopaminergic agents may have ionotropic and metabotropic receptors. Ionotropic glu‑
improved tolerability profiles, being devoid of the classical tamate receptors (iGluRs) are directly coupled to the

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Movements opening of cation channels in the cellular membrane


of postsynaptic neurons. iGluRs are divided into three
subtypes, which are named according to their selective
agonists: N-methyl-d-aspartate (NMDA) receptors,
Cortex
α-amino‑3‑hydroxy‑5‑methyl‑4‑isoxazole propionic
Foliglurax acid (AMPA) receptors and kainate receptors. These
Dipraglurant
are non-­selective cation channels, and their activation
Amantadine ER always produces excitatory postsynaptic responses.
A2A Metabotropic glutamate (mGlu) receptors are G pro‑
mGlu4 Striatum mGlu4 tein-coupled receptors. They are localized both pre‑
synaptically and postsynaptically, and their role is to
mGlu5 A2A Istradefylline mGlu5
regulate the activity of the ionic channels or enzymes
producing second messengers via G proteins165. Unlike
NMDA D2 D1 NMDA
the excitatory iGluRs, mGlu receptors cause slower syn‑
aptic responses, modulating the efficacy of the synapses
A2A
by regulating either the postsynaptic channels and their
receptors or the presynaptic release or recapture of glu‑
D2 mGlu4
tamate166. Therefore, mGlu receptors play an important
GPe mGlu5 role in a variety of physiological processes such as long-
SNpc Thalamus term potentiation and long-term depression of synaptic
NMDA
transmission. To date, eight mGlu receptors have been
mGlu4 cloned and classified into three groups according to
their sequence homologies, pharmacological properties
and signal transduction mechanisms: group I (mGlu1
and mGlu5), II (mGlu2 and mGlu3) and III (mGlu4,
STN GPi mGlu6, mGlu7 and mGlu8)167,168.
mGlu5 mGlu4 mGlu5
SNr There is extensive evidence implicating gluta‑
NMDA NMDA mate in PD and in LID, and glutamate receptors may
represent therapeutic levers to restore basal ganglia
motor circuit functioning in these pathological condi‑
Inhibitory GABA transmission Hyperactive in PD tions169–173. Overactive glutamate transmission is a key
Excitatory glutamate transmission Hypoactive in PD feature underlying the pathophysiology of LID154,174,
Modulatory dopamine transmission Hypoactive in PD and/or leading to the concept that blocking glutamate trans‑
Therapeutic agents in development hyperactive in PD with LID
mission should be beneficial. This blockade could be
mGlu4 A2A obtained either by decreasing signalling downstream
Glutamate of excessive glutamate release, through inhibition
mGlu5 receptors Dopamine receptors
of postsynaptic NMDA or mGlu5 receptors, or by
NMDA
decreasing glutamate release itself through activa‑
tion of presynaptic mGlu4 or mGlu3 receptors and
Figure 3 | The basal ganglia motor circuit that controls movement, highlighting key restoring physiological neurotransmitter levels at the
targets and agents in the treatment of Parkinson disease.
NatureIn healthy| conditions,
Reviews Drug Discovery synapse.
the direct pathway, originating in dopamine D1 receptor (D1)-expressing striatal
medium-sized spiny projection neurons (MSNs), disinhibits the thalamo-cortical Amantadine ER. Amantadine, a non-selective NMDA
neurons through two successive inhibitory (GABAergic) transmissions: from the receptor antagonist, is commonly used to treat LID170,175.
putamen to the internal segment of the globus pallidus (GPi) and then to the thalamus.
This old drug has been used for nearly 50 years and is
The indirect pathway, originating in dopamine D2 receptor (D2)-expressing striatal
MSNs, maintains the thalamo-cortical inhibition through four successive transmissions: still the drug with the greatest antidyskinetic effect in
inhibitory (GABAergic) projections from the putamen to the external segment of the patients with PD176,177. However, its clinical utility may
globus pallidus (GPe) and then to the subthalamic nucleus (STN), an excitatory be limited by cardiovascular and psychiatric side effects
(glutamatergic) synapse between the STN and GPi/substantia nigra pars reticulata (SNr) such as insomnia, confusion and hallucinations178–180
and finally an inhibitory (GABAergic) synapse between the GPi/SNr and the thalamus. that may be directly due to inhibition of NMDA recep‑
Dopamine, which is released by nigro-striatal neurons, stimulates the direct pathway tors181,182. Amantadine ER (ADS‑5102 (Gocovri)), a
and inhibits the indirect pathway. In Parkinson disease (PD), progressive degeneration of novel extended-release formulation of amantadine,
the dopaminergic neurons in the substantia nigra pars compacta (SNpc) results in the was recently approved by the FDA for the treatment
loss of dopaminergic control in the striatum and leads to an imbalance in favour of a of LID in patients with PD receiving levodopa-based
hyperactive indirect pathway and a subsequent global inhibition of motor cortex areas.
therapy. Interestingly, in a phase III study, amantadine
This figure highlights some of the key receptors that are currently targeted for the
treatment of PD: metabotropic glutamate receptor 4 (mGlu4) and mGlu5 as well as ER reduced LID and slightly reduced OFF time whereas
NMDA (N-methyl-d-aspartate) glutamate receptors and adenosine receptor type 2A placebo increased it 183. However, the spectrum of
(A2A). Note that although the therapeutic agents in development are depicted only once adverse events was similar to immediate release forms
in the figure, they are expected to act everywhere their target is present in this circuit of amantadine, suggesting that some improvement can
(mGlu4 for foliglurax, mGlu5 for dipraglurant, NMDA for amantadine ER and A2A for still be pursued in the benefit/risk ratio of a treatment
istradefylline). LID, levodopa-induced dyskinesia. for LID183,184.

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Negative allosteric modulators of mGlu5. The three Positive allosteric modulators of mGlu4. The presyn‑
groups of mGlu receptors are widely expressed in aptic mGlu4 receptor is considered to be a promising
the basal ganglia (except mGlu6 receptors, which are candidate for drug targeting in PD given its role in
exclusively localized in the retina), where they act as modulating neurotransmitter release in the basal gan‑
key regulators of the motor circuit in both normal and glia motor circuit 173,185 (FIG. 2). Two candidate mGlu4
diseased conditions by modulating neuronal excitabil‑ positive allosteric modulators (PAMs) from a novel
ity, synaptic transmission and plasticity 166. Therefore, chemical series have recently been studied in rodent 192
targeting specific mGlu receptor subtypes by means of and non-human primate models193,194, with foliglurax
selective drugs could be a possible strategy for restoring (PXT002331) now advancing into phase II clinical
normal activity in the basal ganglia in PD, thus improv‑ development. Foliglurax has the potential to reduce
ing motor function. In this context, the development of both wearing-off and dyskinesia in patients with PD. In
subtype-­selective mGlu receptor agonists, antagonists models of MPTP-induced parkinsonism and LID, foli‑
and allosteric modulators has provided scientists with a glurax has shown robust and dose-dependent reversal
wide range of neuropharmacological tools that may lead of parkinsonian motor symptoms, reduction of wear‑
to effective treatments185. ing-off and a decrease of peak dyskinesia with a higher
Antagonism of postsynaptic mGlu5 could potentially efficacy and better tolerability than amantadine193,194.
decrease glutamate signalling downstream of excessive However, the efficacy of an mGlu4 PAM still remains to
glutamate release, an approach that could reduce LID186 be proved in patients. Results of the ongoing phase IIa
(FIG. 2). Mavoglurant (AFQ056) was the first mGlu5 study (NCT03162874), expected in 2019, will be very
negative allosteric modulator (NAM) evaluated in PD informative in this regard.
patients with LID. In an MPTP-lesioned macaque model
of LID, mavoglurant decreased peak dyskinesia while Other non-dopaminergic targets
maintaining the antiparkinsonian effects of levodopa187. Other non-dopaminergic targets are under investigation,
Mavoglurant was tested in two placebo-controlled the most advanced involving serotoninergic agents.
phase II studies in 31 patients with PD with moderate In advanced stages of PD, serotonin (5‑HT) terminals
to severe LID (study 1) and 28 patients with PD with take up levodopa and convert it to dopamine. The result‑
severe LID (study 2)188. In both studies, significant anti‑ ing non-physiological release of dopamine and abnormal
dyskinetic effects of treatment were observed. Treatment pulsatile stimulation of dopamine receptors within the
effects on PD motor symptoms were inconsistent striatum may participate in the development of LID195.
between the studies and adverse events were reported. Presynaptic 5‑HT1A receptor agonists can reduce dopa‑
The most common side effects were dizziness, worsen‑ mine release from these serotonergic striatal terminals
ing of dyskinesia when stopping treatment (rebound and thus potentially reduce dyskinesia. Several clinically
effect) and psychiatric disturbances (hallucinations, illu‑ available drugs that partly act as 5‑HT1A agonists have
sions and euphoric mood)188. Consequently, the clinical thus been assessed in patients with PD. Buspirone is a
development of mavoglurant for the treatment of LID combined 5‑HT1A and α1-adrenergic receptor agonist,
was discontinued. indicated for the management of anxiety disorders, with
Another agent of this class, dipraglurant (ADX48621), antidyskinetic effects that were first reported in the early
has recently been assessed in PD patients with LID. In an 1990s196. Buspirone is currently being assessed in three
MPTP macaque model, dipraglurant dose-­dependently clinical trials in PD patients with LID: as monotherapy
decreased LID189. There was no comparator in this study, in a phase III study (NCT02617017), in combination
such as amantadine for example. When assessed in a with a triptan in a phase II study (NCT02803749) and
phase II, placebo-controlled 4‑week study in 76 patients in combination with amantadine in a phase I study
with PD who exhibited moderate to severe LID, dipraglu‑ (NCT02589340).
rant did not show a significant effect on LID at study end Eltoprazine is a mixed 5‑HT1A and 5‑HT1B agonist
point (day 28) despite encouraging results at days 1 and that exerts antidyskinetic effects in rats by reducing
14 (REF.190). Side effects were reported with dipraglurant, striatal glutamate transmission197. A single oral treat‑
including dizziness, dyskinesia and hallucinations191, the ment with eltoprazine was tested in combination with
same as the other mGlu5 NAM, mavoglurant181,182. a suprathreshold dose of levodopa in a phase I/IIa
Overall, these studies support a potential antidys‑ study in 22 PD patients with LID198. Following levo‑
kinetic effect of mGlu5 NAM, validating the approach dopa challenge, eltoprazine significantly reduced LID
of blocking the excessive glutamate transmission in in this study. A phase IIb is currently in preparation
the basal ganglia to reduce LID. Both amantadine and and is expected to be initiated in the first half of 2019.
mGlu5 NAMs have postsynaptic targets, involving Among the symptoms of PD, cognitive impairment,
either direct or indirect NMDA blockade, which may dementia, gait (freezing) and balance (falls) problems
Allosteric modulators
Molecules that bind a site that be the cause of psychiatric side effects. Further stud‑ remain unmet needs. Currently there are no substantial
is different from the binding ies are needed to explore strategies to decrease the side hopes, although positive results have been reported in
site of the endogenous agonist. effects of this mode of action. Targeting presynaptic the past with cholinergic agents for falls199 and noradr‑
As modulators, they act only in receptors such as mGlu4 (currently being tested in a energic compounds for gait hypokinesia and freezing 200.
presence of the endogenous
agonist, either potentiating
proof‑of‑concept clinical study) and mGlu3 (still in Droxidopa, a prodrug of noradrenaline indicated in
(PAM) or reducing (NAM) the preclinical investigations) may be one avenue worth patients with orthostatic hypotension, was evaluated
receptor response. exploring. in a 10‑week phase III placebo-controlled study in

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

225 patients with PD with orthostatic hypotension201. Symptomatic treatments remain of interest to address
In this study, treatment with droxidopa reduced falls in the still high unmet needs of patients linked with, for
patients with PD. These findings should be confirmed example, the motor complications of levodopa treat‑
in more focused clinical evaluations in order to assess ments. Indeed, even when disease-modifying treatments
their clinical relevance. Additional references on non-­ become available, there is likely to be a continuing need
dopaminergic approaches for symptomatic control in for agents, with optimized tolerability, that alleviate
PD are reviewed in154,175. symptoms.
In the field of preclinical research, there is a need for
Outlook a better focus on the robustness of the effects of a new
Substantial progress has been made in the search for compound, confirmation of findings across different stud‑
and development of new agents to manage the clinical ies and models and comparison of the intensity of effects
features of PD, and a number of drug candidates are with those of known benchmark molecules. As described
poised ready to enter clinical study as treatments with in this Review, therapies that have shown a mild, non-­
disease-modifying potential. Although there is still much robust effect in preclinical animal models have then failed
to be learned about the pathophysiology of PD and the to demonstrate efficacy in phase II or III trials. Although
processes and chronology of neurodegeneration in this a high attrition rate is a feature of all drug development,
condition, with the identification of better disease bio‑ contemporary and forward-thinking research is doing
markers and treatment targets, there is much scope for much to advance endeavours to discover and develop
the development of improved therapies for PD. more effective drug entities for patients with PD.

1. Parkinson, J. An essay on the shaking palsy. 1817. 13. Bastide, M. F. et al. Pathophysiology of L‑DOPA- This article provides a review on the current animal
J. Neuropsychiatry Clin. Neurosci. 14, 223–236 induced motor and non-motor complications in models of PD and describes the transition from
(2002). Parkinson’s disease. Prog. Neurobiol. 132, 96–168 well-established animal models of PD
This essay is the first clear clinical description of (2015). symptomatology to models being developed to
the shaking palsy, or paralysis agitans, that we now 14. Moore, T. J., Glenmullen, J. & Mattison, D. R. Reports assess neuroprotective strategies.
refer to as PD. of pathological gambling, hypersexuality, and 27. Bourdenx, M. et al. Protein aggregation and
2. Donaldson, I. M. James Parkinson’s essay on the compulsive shopping associated with dopamine neurodegeneration in prototypical neurodegenerative
shaking palsy. J. R. Coll. Physicians Edinb. 45, 84–86 receptor agonist drugs. JAMA Intern. Med. 174, diseases: examples of amyloidopathies, tauopathies
(2015). 1930–1933 (2014). and synucleinopathies. Prog. Neurobiol. 155,
3. Birkmayer, W. & Hornykiewicz, O. The 15. Schrag, A., Jahanshahi, M. & Quinn, N. What 171–193 (2017).
L‑dihydroxyphenylalanine (L‑DOPA) effect in contributes to quality of life in patients with 28. Bourdenx, M. et al. Lack of additive role of ageing in
Parkinson’s syndrome in man: on the pathogenesis Parkinson’s disease? J. Neurol. Neurosurg. nigrostriatal neurodegeneration triggered by alpha-
and treatment of Parkinson akinesis [German]. Arch. Psychiatry 69, 308–312 (2000). synuclein overexpression. Acta Neuropathol.
Psychiatr. Nervenkr. Z. Gesamte Neurol. Psychiatr. 16. Izurieta-Sanchez, P., Sarre, S., Ebinger, G. & Commun. 3, 46 (2015).
203, 560–574 (1962). Michotte, Y. Effect of trihexyphenidyl, a non-selective 29. Koprich, J. B., Johnston, T. H., Reyes, G., Omana, V. &
4. Birkmayer, W. & Hornykiewicz, O. Additional antimuscarinic drug, on decarboxylation of L‑DOPA in Brotchie, J. M. Towards a non-human primate model
experimental studies on L‑DOPA in Parkinson’s hemi-Parkinson rats. Eur. J. Pharmacol. 353, 33–42 of alpha-synucleinopathy for development of
syndrome and reserpine parkinsonism [German]. (1998). therapeutics for Parkinson’s disease: optimization of
Arch. Psychiatr. Nervenkr. 206, 367–381 (1964). 17. Schwab, R. S., England, A. C. Jr., Poskanzer, D. C. & AAV1/2 delivery parameters to drive sustained
5. Zeng, X. S., Geng, W. S., Jia, J. J., Chen, L. & Young, R. R. Amantadine in the treatment of expression of alpha synuclein and dopaminergic
Zhang, P. P. Cellular and molecular basis of Parkinson’s disease. JAMA 208, 1168–1170 degeneration in macaque. PLOS ONE 11, e0167235
neurodegeneration in Parkinson disease. Front. Aging (1969). (2016).
Neurosci. 10, 109 (2018). 18. Athauda, D. & Foltynie, T. The glucagon-like peptide 1 30. European Agency for Evaluation of Medicinal
6. Poewe, W. et al. Parkinson disease. Nat. Rev. Dis. (GLP) receptor as a therapeutic target in Parkinson’s Products. Guideline on clinical investigation of
Primers 3, 17013 (2017). disease: mechanisms of action. Drug Discov. Today medicinal products in the treatment of Parkinson’s
This paper provides a comprehensive review on our 21, 802–818 (2016). disease. Eur. Neuropsychophamarcol. 9, 443–449
current understanding of the underlying pathology 19. Kieburtz, K. & Olanow, C. W. Advances in clinical (2012).
and molecular pathogenesis of PD. trials for movement disorders. Mov. Disord. 30, 31. Oertel, W. H. Recent advances in treating Parkinson’s
7. Obeso, J. A. et al. Past, present, and future of 1580–1587 (2015). disease. F1000Res 6, 260 (2017).
Parkinson’s disease: a special essay on the 200th 20. McGhee, D. J., Ritchie, C. W., Zajicek, J. P. & 32. Sauerbier, A., Qamar, M. A., Rajah, T. &
anniversary of the shaking palsy. Mov. Disord. 32, Counsell, C. E. A review of clinical trial designs used Chaudhuri, K. R. New concepts in the pathogenesis
1264–1310 (2017). to detect a disease-modifying effect of drug therapy in and presentation of Parkinson’s disease. Clin. Med.
Prepared to commemorate the bicentenary of PD, Alzheimer’s disease and Parkinson’s disease. BMC 16, 365–370 (2016).
this extensive review provides a historical Neurol. 16, 92 (2016). 33. Andersen, A. D., Binzer, M., Stenager, E. &
state‑of‑the-art account of what has been achieved, 21. Bartus, R. T., Weinberg, M. S. & Samulski, R. J. Gramsbergen, J. B. Cerebrospinal fluid biomarkers for
the current situation and how to progress towards Parkinson’s disease gene therapy: success by design Parkinson’s disease - a systematic review. Acta Neurol.
identifying treatments. meets failure by efficacy. Mol. Ther. 22, 487–497 Scand. 135, 34–56 (2017).
8. Michel, P. P., Hirsch, E. C. & Hunot, S. Understanding (2014). 34. Kalia, L. V. & Lang, A. E. Parkinson’s disease. Lancet
dopaminergic cell death pathways in Parkinson 22. Zeiss, C. J., Allore, H. G. & Beck, A. P. Established 386, 896–912 (2015).
disease. Neuron 90, 675–691 (2016). patterns of animal study design undermine 35. Saracchi, E., Fermi, S. & Brighina, L. Emerging
This paper provides a comprehensive review of the translation of disease-modifying therapies for candidate biomarkers for Parkinson’s disease: a
mechanisms that are suspected to participate in Parkinson’s disease. PLOS ONE 12, e0171790 review. Aging Dis. 5, 27–34 (2014).
dopaminergic cell death in PD. (2017). 36. Burciu, R. G. et al. Progression marker of Parkinson’s
9. Fink, A. L. The aggregation and fibrillation of alpha- 23. Magen, I. & Chesselet, M. F. Genetic mouse models of disease: a 4‑year multi-site imaging study. Brain 140,
synuclein. Acc. Chem. Res. 39, 628–634 (2006). Parkinson’s disease: the state of the art. Prog. Brain 2183–2192 (2017).
10. Lashuel, H. A., Overk, C. R., Oueslati, A. & Masliah, E. Res. 184, 53–87 (2010). Using MRI data from the PPMI study, results
The many faces of alpha-synuclein: from structure and 24. Bezard, E., Yue, Z., Kirik, D. & Spillantini, M. G. presented in this article support the potential of
toxicity to therapeutic target. Nat. Rev. Neurosci. 14, Animal models of Parkinson’s disease: limits and free water in the posterior substantia nigra as an
38–48 (2013). relevance to neuroprotection studies. Mov. Disord. imaging biomarker for PD progression.
This review provides an overview of the current 28, 61–70 (2013). 37. Fereshtehnejad, S. M., Zeighami, Y., Dagher, A. &
knowledge regarding the role of α-synuclein states 25. Dehay, B. & Fernagut, P. O. Alpha-synuclein-based Postuma, R. B. Clinical criteria for subtyping
in regulating its function under physiological and models of Parkinson’s disease. Rev. Neurol. 172, Parkinson’s disease: biomarkers and longitudinal
pathophysiological conditions. 371–378 (2016). progression. Brain 140, 1959–1976 (2017).
11. Kaushik, S. & Cuervo, A. M. Proteostasis and aging. 26. Ko, W. K. D. & Bezard, E. Experimental animal 38. Schrag, A., Siddiqui, U. F., Anastasiou, Z.,
Nat. Med. 21, 1406–1415 (2015). models of Parkinson’s disease: a transition from Weintraub, D. & Schott, J. M. Clinical variables and
12. Xilouri, M., Brekk, O. R. & Stefanis, L. α-Synuclein and assessing symptomatology to alpha-synuclein biomarkers in prediction of cognitive impairment in
protein degradation systems: a reciprocal relationship. targeted disease modification. Exp. Neurol. 298 patients with newly diagnosed Parkinson’s disease:
Mol. Neurobiol. 47, 537–551 (2013). (Suppl. B), 172–179 (2017). a cohort study. Lancet Neurol. 16, 66–75 (2017).

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

39. Athauda, D. et al. Exenatide once weekly versus 62. NINDS Exploratory Trials in Parkinson Disease (NET- 85. Voyager Therapeutics Inc. Voyager Therapeutics
placebo in Parkinson’s disease: a randomised, double- PD) FS-ZONE Investigators. Pioglitazone in early announces FDA Regenerative Medicine Advanced
blind, placebo-controlled trial. Lancet 390, Parkinson’s disease: a phase 2, multicentre, double- Therapy (RMAT) designation granted for VY‑AADC for
1664–1675 (2017). blind, randomised trial. Lancet Neurol. 14, 795–803 the treatment of Parkinson’s disease. Voyager
40. Aviles-Olmos, I. et al. Motor and cognitive advantages (2015). Therapeutics http://ir.voyagertherapeutics.com/
persist 12 months after exenatide exposure in 63. Simon, D. K. et al. Peripheral biomarkers of phoenix.zhtml?c=254026&p=irol-
Parkinson’s disease. J. Parkinsons Dis. 4, 337–344 Parkinson’s disease progression and pioglitazone newsArticle&ID=2355428 (2018).
(2014). effects. J. Parkinsons Dis. 5, 731–736 (2015). 86. Dickson, D. W. et al. Neuropathological assessment
41. Hauser, R. A. et al. Time course of loss of clinical 64. Blits, B. & Petry, H. Perspective on the road toward of Parkinson’s disease: refining the diagnostic criteria.
benefit following withdrawal of levodopa/carbidopa gene therapy for Parkinson’s disease. Front. Lancet Neurol. 8, 1150–1157 (2009).
and bromocriptine in early Parkinson’s disease. Mov. Neuroanat. 10, 128 (2016). 87. Olanow, C. W. & Kordower, J. H. Targeting alpha-
Disord. 15, 485–489 (2000). 65. Gash, D. M. et al. Functional recovery in parkinsonian synuclein as a therapy for Parkinson’s disease: the
42. Hauser, R. A. & Holford, N. H. Quantitative monkeys treated with GDNF. Nature 380, 252–255 battle begins. Mov. Disord. 32, 203–207 (2017).
description of loss of clinical benefit following (1996). 88. Masliah, E. et al. Passive immunization reduces
withdrawal of levodopa-carbidopa and bromocriptine 66. Emborg, M. E. et al. Response of aged parkinsonian behavioral and neuropathological deficits in an alpha-
in early Parkinson’s disease. Mov. Disord. 17, monkeys to in vivo gene transfer of GDNF. Neurobiol. synuclein transgenic model of Lewy body disease.
961–968 (2002). Dis. 36, 303–311 (2009). PLOS ONE 6, e19338 (2011).
43. Hauser, R. A. & Zesiewicz, T. A. Clinical trials aimed at 67. Kordower, J. H. et al. Neurodegeneration prevented 89. Games, D. et al. Reducing C‑terminal-truncated alpha-
detecting neuroprotection in Parkinson’s disease. by lentiviral vector delivery of GDNF in primate synuclein by immunotherapy attenuates
Neurology 66, S58–68 (2006). models of Parkinson’s disease. Science 290, neurodegeneration and propagation in Parkinson’s
44. Foltynie, T., Brayne, C. & Barker, R. A. The 767–773 (2000). disease-like models. J. Neurosci. 34, 9441–9454
heterogeneity of idiopathic Parkinson’s disease. 68. Nutt, J. G. et al. Randomized, double-blind trial of (2014).
J. Neurol. 249, 138–145 (2002). glial cell line-derived neurotrophic factor (GDNF) in 90. Schenk, D. B. et al. First‑in‑human assessment of
45. Sieber, B. A. et al. Prioritized research PD. Neurology 60, 69–73 (2003). PRX002, an anti-alpha-synuclein monoclonal
recommendations from the National Institute of 69. Lang, A. E. et al. Randomized controlled trial of antibody, in healthy volunteers. Mov. Disord. 32,
Neurological Disorders and Stroke Parkinson’s intraputamenal glial cell line-derived neurotrophic 211–218 (2017).
Disease 2014 conference. Ann. Neurol. 76, 469–472 factor infusion in Parkinson disease. Ann. Neurol. 59, 91. Jankovic, J. et al. Safety and tolerability of multiple
(2014). 459–466 (2006). ascending doses of PRX002/RG7935, an anti-α-
46. Athauda, D. & Foltynie, T. Challenges in detecting 70. Warren Olanow, C. et al. Gene delivery of neurturin to synuclein monoclonal antibody, in patients with
disease modification in Parkinson’s disease clinical putamen and substantia nigra in Parkinson disease: a Parkinson disease: a randomized clinical trial. JAMA
trials. Parkinsonism Relat. Disord. 32, 1–11 double-blind, randomized, controlled trial. Ann. Neurol. https://doi.org/10.1001/
(2016). Neurol. 78, 248–257 (2015). jamaneurol.2018.1487 (2018).
47. Lang, A. E. & Espay, A. J. Disease modification in 71. Decressac, M. et al. α-Synuclein-induced down- 92. Mandler, M. et al. Next-generation active
Parkinson’s disease: current approaches, challenges, regulation of Nurr1 disrupts GDNF signaling in nigral immunization approach for synucleinopathies:
and future considerations. Mov. Disord. 33, 660–677 dopamine neurons. Sci. Transl Med. 4, 163ra156 implications for Parkinson’s disease clinical trials. Acta
(2018). (2012). Neuropathol. 127, 861–879 (2014).
48. Jenner, P. & Olanow, C. W. Oxidative stress and the 72. Chung, C. Y., Koprich, J. B., Siddiqi, H. & Isacson, O. 93. AFFiRiS. AFFiRiS announces top line results of
pathogenesis of Parkinson’s disease. Neurology 47, Dynamic changes in presynaptic and axonal transport first‑in‑human clinical study using AFFITOPE®
S161–S170 (1996). proteins combined with striatal neuroinflammation PD03A, confirming immunogenicity and safety
49. Bose, A. & Beal, M. F. Mitochondrial dysfunction in precede dopaminergic neuronal loss in a rat model of profile in Parkinson’s disease patients. Cision PR
Parkinson’s disease. J. Neurochem. 139 (Suppl. 1), AAV alpha-synucleinopathy. J. Neurosci. 29, Newswire https://www.prnewswire.com/news-releases/
216–231 (2016). 3365–3373 (2009). affiris-announces-top-line-results-of-first-in-human-
This review provides an overview of the 73. Nalls, M. A. et al. Large-scale meta-analysis of clinical-study-using-affitope-pd03a-confirming-
mechanisms that can cause mitochondrial genome-wide association data identifies six new risk immunogenicity-and-safety-profile-in-parkinsons-
dysfunction in PD. loci for Parkinson’s disease. Nat. Genet. 46, 989–993 disease-patients-627025511.html (2017).
50. Cristovao, A. C. et al. NADPH oxidase 1 mediates (2014). 94. Poewe, W. et al. in AAT‑AD/PD Focus Meeting 2018
alpha-synucleinopathy in Parkinson’s disease. 74. West, A. B. Achieving neuroprotection with LRRK2 (Torino, Italy, 2018).
J. Neurosci. 32, 14465–14477 (2012). kinase inhibitors in Parkinson disease. Exp. Neurol. 95. Brys, M. et al. Randomized, double-blind, placebo-
51. Ko, H. S. et al. Phosphorylation by the c‑Abl protein 298, 236–245 (2017). controlled, single ascending dose study of anti-alpha-
tyrosine kinase inhibits parkin’s ubiquitination and 75. Sardi, S. P., Cedarbaum, J. M. & Brundin, P. Targeted synuclein antibody BIIB054 in patients with
protective function. Proc. Natl Acad. Sci. USA 107, therapies for Parkinson’s disease: from genetics to the Parkinson’s disease (S26.001). Neurology 90 (Suppl.
16691–16696 (2010). clinic. Mov. Disord. 33, 684–696 (2018). 15), S26.001 (2018).
52. Imam, S. Z. et al. Novel regulation of parkin function 76. Palfi, S. et al. Long-term safety and tolerability of 96. Weihofen, A. et al. in 13th International Conference on
through c‑Abl-mediated tyrosine phosphorylation: ProSavin, a lentiviral vector-based gene therapy for Alzheimer’s and Parkinson’s Diseases (Vienna,
implications for Parkinson’s disease. J. Neurosci. 31, Parkinson’s disease: a dose escalation, open-label, Austria, 2017).
157–163 (2011). phase 1/2 trial. Lancet 383, 1138–1146 (2014). 97. Wagner, J. et al. Anle138b: a novel oligomer modulator
53. Zhao, J., Yu, S., Zheng, Y., Yang, H. & Zhang, J. 77. Jarraya, B. et al. Dopamine gene therapy for for disease-modifying therapy of neurodegenerative
Oxidative modification and its implications for the Parkinson’s disease in a nonhuman primate without diseases such as prion and Parkinson’s disease. Acta
neurodegeneration of Parkinson’s disease. Mol. associated dyskinesia. Sci. Transl Med. 1, 2ra4 Neuropathol. 125, 795–813 (2013).
Neurobiol. 54, 1404–1418 (2017). (2009). 98. Szoke, B. et al. in Society for Neuroscience:
54. Cacabelos, R. Parkinson’s disease: from pathogenesis 78. Goetz, C. G. et al. Placebo response in Parkinson’s Neuroscience 2014 (Washington DC, 2014).
to pharmacogenomics. Int. J. Mol. Sci. 18, 551 disease: comparisons among 11 trials covering 99. Wrasidlo, W. et al. A de novo compound targeting
(2017). medical and surgical interventions. Mov. Disord. 23, alpha-synuclein improves deficits in models of
55. Hauser, D. N. & Hastings, T. G. Mitochondrial 690–699 (2008). Parkinson’s disease. Brain 139, 3217–3236 (2016).
dysfunction and oxidative stress in Parkinson’s disease 79. Stewart, H. J. et al. Optimizing transgene 100. US National Library of Medicine. ClinicalTrials.gov
and monogenic parkinsonism. Neurobiol. Dis. 51, configuration and protein fusions to maximize https://clinicaltrials.gov/ct2/show/NCT02606682
35–42 (2013). dopamine production for the gene therapy of (2016).
56. Parkinson Study Group QE3 Investigators. A Parkinson’s disease. Hum. Gene Ther. Clin. Dev. 27, 101. McCormack, A. L. et al. Alpha-synuclein suppression
randomized clinical trial of high-dosage coenzyme 100–110 (2016). by targeted small interfering RNA in the primate
Q10 in early Parkinson disease: no evidence of 80. Axovant Sciences Ltd. Axovant licenses investigational substantia nigra. PLOS ONE 5, e12122 (2010).
benefit. JAMA Neurol. 71, 543–552 (2014). gene therapy for Parkinson’s disease from Oxford 102. US Department of Health and Human Services. New
57. Yang, L. et al. Combination therapy with coenzyme BioMedica and announces key leadership team therapy addresses unmet medical need for rare disease.
Q10 and creatine produces additive neuroprotective addition. Axovant http://investors.axovant.com/ FDA.gov https://www.fda.gov/downloads/forindustry/
effects in models of Parkinson’s and Huntington’s node/7551/pdf (2018). developingproductsforrarediseasesconditions/
diseases. J. Neurochem. 109, 1427–1439 (2009). 81. Ciesielska, A. et al. Depletion of AADC activity in designatinghumanitarianusedeviceshuds/ucm597204.
58. Spindler, M., Beal, M. F. & Henchcliffe, C. Coenzyme caudate nucleus and putamen of Parkinson’s disease pdf (2016).
Q10 effects in neurodegenerative disease. patients; implications for ongoing AAV2‑AADC gene 103. Finkel, R. S. et al. Treatment of infantile-onset spinal
Neuropsychiatr. Dis. Treat. 5, 597–610 (2009). therapy trial. PLOS ONE 12, e0169965 (2017). muscular atrophy with nusinersen: a phase 2, open-
59. Cleren, C. et al. Therapeutic effects of coenzyme Q10 82. San Sebastian, W. et al. Safety and tolerability of MRI- label, dose-escalation study. Lancet 388, 3017–3026
(CoQ10) and reduced CoQ10 in the MPTP model of guided infusion of AAV2‑hAADC into the mid-brain of (2016).
Parkinsonism. J. Neurochem. 104, 1613–1621 non-human primate. Mol. Ther. Methods Clin. Dev. 1, 104. Finkel, R. S. et al. Nusinersen versus sham control in
(2008). 14049 (2014). infantile-onset spinal muscular atrophy. N. Engl.
60. Agarwal, S., Yadav, A. & Chaturvedi, R. K. Peroxisome 83. Mittermeyer, G. et al. Long-term evaluation of a J. Med. 377, 1723–1732 (2017).
proliferator-activated receptors (PPARs) as phase 1 study of AADC gene therapy for Parkinson’s 105. Ionis Pharmaceuticals Inc. Ionis Pharmaceuticals
therapeutic target in neurodegenerative disorders. disease. Hum. Gene Ther. 23, 377–381 (2012). licenses IONIS-HTT Rx to partner following successful
Biochem. Biophys. Res. Commun. 483, 1166–1177 84. Voyager Therapeutics Inc. Voyager Therapeutics phase 1/2a study in patients with Huntington’s
(2017). announces positive results from ongoing phase 1b disease. Ionis Pharmaceuticals http://ir.ionispharma.
61. Swanson, C. R. et al. The PPAR-gamma agonist trial of VY‑AADC01 for advanced Parkinson’s disease. com/node/23031/pdf (2017).
pioglitazone modulates inflammation and induces Voyager Therapeutics http://ir.voyagertherapeutics. 106. Wild, E. J. & Tabrizi, S. J. Therapies targeting DNA
neuroprotection in parkinsonian monkeys. com/phoenix.zhtml?c=254026&p=irol- and RNA in Huntington’s disease. Lancet Neurol. 16,
J. Neuroinflamm. 8, 91 (2011). newsArticle&ID=2298649 (2017). 837–847 (2017).

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

107. Gorenberg, E. L. & Chandra, S. S. The role of 129. Weinreb, O., Mandel, S., Youdim, M. B. & Amit, T. 154. Freitas, M. E. & Fox, S. H. Nondopaminergic
co‑chaperones in synaptic proteostasis and Targeting dysregulation of brain iron homeostasis in treatments for Parkinson’s disease: current and future
neurodegenerative disease. Front. Neurosci. 11, 248 Parkinson’s disease by iron chelators. Free Radic. Biol. prospects. Neurodegener. Dis. Manag. 6, 249–268
(2017). Med. 62, 52–64 (2013). (2016).
108. Moors, T. E. et al. Therapeutic potential of autophagy- 130. Devos, D. et al. Targeting chelatable iron as a 155. Ko, W. K. et al. An evaluation of istradefylline
enhancing agents in Parkinson’s disease. Mol. therapeutic modality in Parkinson’s disease. Antioxid. treatment on Parkinsonian motor and cognitive
Neurodegener. 12, 11 (2017). Redox Signal. 21, 195–210 (2014). deficits in
109. Sian, J. et al. Alterations in glutathione levels in 131. Athauda, D. & Foltynie, T. Insulin resistance and 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyridine
Parkinson’s disease and other neurodegenerative Parkinson’s disease: a new target for disease (MPTP)-treated macaque models.
disorders affecting basal ganglia. Ann. Neurol. 36, modification? Prog. Neurobiol. 145–146, 98–120 Neuropharmacology 110, 48–58 (2016).
348–355 (1994). (2016). 156. Kondo, T., Mizuno, Y. & Japanese Istradefylline Study
110. Witschi, A., Reddy, S., Stofer, B. & Lauterburg, B. H. This article reviews the evidence suggesting that Group. A long-term study of istradefylline safety and
The systemic availability of oral glutathione. Eur. PD and insulin resistance share common underlying efficacy in patients with Parkinson disease. Clin.
J. Clin. Pharmacol. 43, 667–669 (1992). pathological mechanisms. Neuropharmacol. 38, 41–46 (2015).
111. Hauser, R. A., Lyons, K. E., McClain, T., Carter, S. & 132. Dunn, L. et al. Dysregulation of glucose metabolism is 157. Hauser, R. A., Hubble, J. P., Truong, D. D. &
Perlmutter, D. Randomized, double-blind, pilot an early event in sporadic Parkinson’s disease. Istradefylline US‑001 Study Group. Randomized
evaluation of intravenous glutathione in Parkinson’s Neurobiol. Aging 35, 1111–1115 (2014). trial of the adenosine A(2A) receptor antagonist
disease. Mov. Disord. 24, 979–983 (2009). 133. Aksoy, D. et al. Neuroprotective effects of eexenatide istradefylline in advanced PD. Neurology 61,
112. Mischley, L. K., Lau, R. C., Shankland, E. G., in a rotenone-induced rat model of Parkinson’s 297–303 (2003).
Wilbur, T. K. & Padowski, J. M. Phase IIb study of disease. Am. J. Med. Sci. 354, 319–324 (2017). 158. Kyowa Hakko Kirin Co., Ltd. Kyowa Hakko Kirin
intranasal glutathione in Parkinson’s disease. 134. Athauda, D., Wyse, R., Brundin, P. & Foltynie, T. Is announces top-line results of global phase 3 trial of
J. Parkinsons Dis. 7, 289–299 (2017). exenatide a treatment for Parkinson’s disease? KW‑6002 (Istradefylline) for Parkinson’s disease.
113. Mischley, L. K. et al. Central nervous system uptake of J. Parkinsons Dis. 7, 451–458 (2017). Kyowa Kirin http://kyowa-kirin.com/news_
intranasal glutathione in Parkinson’s disease. NPJ 135. US National Library of Medicine. ClinicalTrials.gov releases/2016/pdf/e20161213_01.pdf (2016).
Parkinsons Dis. 2, 16002 (2016). https://clinicaltrials.gov/ct2/show/NCT03456687 159. Kyowa Hakko Kirin Co., Ltd. Kyowa Hakko Kirin
114. O’Regan, G., deSouza, R. M., Balestrino, R. & (2018). announces the intent to file Istradefylline (KW‑6002)
Schapira, A. H. Glucocerebrosidase mutations in 136. US National Library of Medicine. ClinicalTrials.gov for Parkinson’s disease with the US FDA. Kyowa Kirin
Parkinson disease. J. Parkinsons Dis. 7, 411–422 https://clinicaltrials.gov/ct2/show/NCT02953665 http://kyowa-kirin.com/news_releases/2017/pdf/
(2017). (2016). e20171017_01.pdf (2017).
115. Beavan, M. S. & Schapira, A. H. Glucocerebrosidase 137. Atashrazm, F. & Dzamko, N. LRRK2 inhibitors and 160. Hodgson, R. A. et al. Preladenant, a selective A(2A)
mutations and the pathogenesis of Parkinson disease. their potential in the treatment of Parkinson’s disease: receptor antagonist, is active in primate models of
Ann. Med. 45, 511–521 (2013). current perspectives. Clin. Pharmacol. 8, 177–189 movement disorders. Exp. Neurol. 225, 384–390
116. Migdalska-Richards, A. & Schapira, A. H. The (2016). (2010).
relationship between glucocerebrosidase mutations 138. Fuji, R. N. et al. Effect of selective LRRK2 kinase 161. Hauser, R. A. et al. Preladenant in patients with
and Parkinson disease. J. Neurochem. 139 (Suppl. 1), inhibition on nonhuman primate lung. Sci. Transl Med. Parkinson’s disease and motor fluctuations: a phase 2,
77–90 (2016). 7, 273ra215 (2015). double-blind, randomised trial. Lancet Neurol. 10,
117. Sardi, S. P. et al. Augmenting CNS glucocerebrosidase 139. Ness, D. et al. Leucine-rich repeat kinase 2 (LRRK2)- 221–229 (2011).
activity as a therapeutic strategy for parkinsonism and deficient rats exhibit renal tubule injury and 162. Hauser, R. A. et al. Preladenant as an adjunctive
other Gaucher-related synucleinopathies. Proc. Natl perturbations in metabolic and immunological therapy with levodopa in Parkinson disease: two
Acad. Sci. USA 110, 3537–3542 (2013). homeostasis. PLOS ONE 8, e66164 (2013). randomized clinical trials and lessons learned. JAMA
118. Zunke, F. et al. Reversible conformational conversion 140. Di Maio, R. et al. LRRK2 activation in idiopathic Neurol. 72, 1491–1500 (2015).
of alpha-synuclein into toxic assemblies by Parkinson’s disease. Sci. Transl Med. 10, eaar5429 163. Hauser, R. A. et al. Tozadenant (SYN115) in patients
glucosylceramide. Neuron 97, 92–107 (2017). (2018). with Parkinson’s disease who have motor fluctuations
119. Kim, S. et al. GBA1 deficiency negatively affects 141. Denali Therapeutics Inc. Denali Therapeutics on levodopa: a phase 2b, double-blind, randomised
physiological alpha-synuclein tetramers and related announces advancement and expansion of its LRRK2 trial. Lancet Neurol. 13, 767–776 (2014).
multimers. Proc. Natl Acad. Sci. USA 115, 798–803 inhibitor clinical program for Parkinson’s disease. 164. Acorda Therapeutics Inc. Acorda discontinues
(2018). Denali Therapeutics http://investors. tozadenant development program. Acord
120. Migdalska-Richards, A., Daly, L., Bezard, E. & denalitherapeutics.com/node/6496/pdf (2017). Therapeutics http://ir.acorda.com/investors/investor-
Schapira, A. H. Ambroxol effects in 142. Kitada, T. et al. Mutations in the parkin gene cause news/investor-news-details/2017/Acorda-Discontinues-
glucocerebrosidase and alpha-synuclein transgenic autosomal recessive juvenile parkinsonism. Nature Tozadenant-Development-Program/default.aspx
mice. Ann. Neurol. 80, 766–775 (2016). 392, 605–608 (1998). (2017).
121. Schapira, A. H., Chiasserini, D., Beccari, T. & 143. Sul, J. W. et al. Accumulation of the parkin substrate, 165. Conn, P. J. & Pin, J. P. Pharmacology and functions of
Parnetti, L. Glucocerebrosidase in Parkinson’s disease: FAF1, plays a key role in the dopaminergic metabotropic glutamate receptors. Annu. Rev.
insights into pathogenesis and prospects for neurodegeneration. Hum. Mol. Genet. 22, Pharmacol. Toxicol. 37, 205–237 (1997).
treatment. Mov. Disord. 31, 830–835 (2016). 1558–1573 (2013). 166. Nicoletti, F. et al. Metabotropic glutamate receptors:
122. Ambrosi, G. et al. Ambroxol-induced rescue of 144. Park, H. S. et al. Assessment of potential protective from the workbench to the bedside.
defective glucocerebrosidase is associated with effect on dopaminergic neuron by an Fas-associated Neuropharmacology 60, 1017–1041 (2011).
increased LIMP‑2 and saposin C levels in GBA1 factor 1 inhibitor treatment using [18F]FE‑PE2I PET in 167. Pin, J. P. & Acher, F. The metabotropic glutamate
mutant Parkinson’s disease cells. Neurobiol. Dis. 82, a Parkinson’s disease mouse model. J. Nuclear Med. receptors: structure, activation mechanism and
235–242 (2015). 58 (Suppl. 1), 13 (2017). pharmacology. Curr. Drug Targets CNS Neurol. Disord.
123. Allergan plc. Allergan enters Parkinson’s disease 145. Langston, J. W. The MPTP Story. J. Parkinsons Dis. 7 1, 297–317 (2002).
through option arrangement with Lysosomal (Suppl. 1), 11–19 (2017). 168. Schoepp, D. D., Jane, D. E. & Monn, J.
Therapeutics Inc. (LTI) for its potential first‑in‑class 146. Morissette, M. & Di Paolo, T. Non-human primate Pharmacological agents acting at subtypes of
breakthrough compounds. Cision PR Newswire https:// models of PD to test novel therapies. J. Neural metabotropic glutamate receptors.
www.prnewswire.com/news-releases/allergan-enters- Transm. 125, 291–324 (2017). Neuropharmacology 38, 1431–1476 (1999).
parkinsons-disease-through-option-arrangement-with- 147. Close, S. P., Elliott, P. J., Hayes, A. G. & Marriott, A. S. 169. Picconi, B. & Calabresi, P. in Levodopa-Induced
lysosomal-therapeutics-inc-lti-for-its-potential-first-in- Effects of classical and novel agents in a MPTP- Dyskinesia in Parkinson’s Disease (eds Fox, S. H. &
class-breakthrough-compounds-300387519.html induced reversible model of Parkinson’s disease. Brotchie, J. M.) 229–243 (Springer, London,
(2017). Psychopharmacology 102, 295–300 (1990). 2014).
124. Marshall, J. et al. CNS-accessible inhibitor of 148. Gregoire, L., Jourdain, V. A., Townsend, M., Roach, A. 170. Morin, N. & Di Paolo, T. Pharmacological treatments
glucosylceramide synthase for substrate reduction & Di Paolo, T. Safinamide reduces dyskinesias and inhibiting levodopa-induced dyskinesias in MPTP-
therapy of neuronopathic Gaucher disease. Mol. Ther. prolongs L‑DOPA antiparkinsonian effect in lesioned monkeys: brain glutamate biochemical
24, 1019–1029 (2016). parkinsonian monkeys. Parkinsonism Relat. Disord. correlates. Front. Neurol. 5, 144 (2014).
125. Sanofi Genzyme. Sanofi initiates phase 2 clinical trial to 19, 508–514 (2013). 171. Litim, N., Morissette, M. & Di Paolo, T. Metabotropic
evaluate therapy for genetic form of Parkinson’s 149. Fox, S. H. & Brotchie, J. M. The MPTP-lesioned non- glutamate receptors as therapeutic targets in
disease. Sanofi Genzyme https://news.genzyme.com/ human primate models of Parkinson’s disease. Past, Parkinson’s disease: an update from the last 5 years
press-release/sanofi-initiates-phase-2-clinical-trial- present, and future. 184, 133–157 (2010). of research. Neuropharmacology 115, 166–179
evaluate-therapy-genetic-form-parkinsons-disea (2017). 150. Ray Chaudhuri, K. et al. Non-oral dopaminergic (2017).
126. Amicus Therapeutics. Amicus Therapeutics announces therapies for Parkinson’s disease: current treatments 172. Amalric, M. Targeting metabotropic glutamate
preliminary results of phase 2 study with Plicera(TM) and the future. NPJ Parkinsons Dis. 2, 16023 (2016). receptors (mGluRs) in Parkinson’s disease. Curr. Opin.
for Gaucher disease. Amicus Therapeutics http://ir. 151. Pilleri, M. & Antonini, A. Novel levodopa formulations Pharmacol. 20, 29–34 (2015).
amicusrx.com/static-files/35813147-7c02-4311- in the treatment of Parkinson’s disease. Expert Rev. 173. Charvin, D. mGlu4 allosteric modulation for treating
b770-9279142ee4da (2009). Neurother. 14, 143–149 (2014). Parkinson’s disease. Neuropharmacology 135,
127. Belaidi, A. A. & Bush, A. I. Iron neurochemistry in 152. Tambasco, N., Romoli, M. & Calabresi, P. Levodopa in 308–315 (2018).
Alzheimer’s disease and Parkinson’s disease: targets Parkinson’s disease: current status and future 174. Duty, S. Targeting glutamate receptors to tackle the
for therapeutics. J. Neurochem. 139 (Suppl. 1), developments. Curr. Neuropharmacol. 16, pathogenesis, clinical symptoms and levodopa-
179–197 (2016). 1239–1252 (2017). induced dyskinesia associated with Parkinson’s
128. Kaur, D. et al. Genetic or pharmacological iron 153. Schwarzschild, M. A., Agnati, L., Fuxe, K., Chen, J. F. disease. CNS Drugs 26, 1017–1032 (2012).
chelation prevents MPTP-induced neurotoxicity & Morelli, M. Targeting adenosine A2A receptors in 175. Fox, S. H. Non-dopaminergic treatments for motor
in vivo: a novel therapy for Parkinson’s disease. Parkinson’s disease. Trends Neurosci. 29, 647–654 control in Parkinson’s disease. Drugs 73, 1405–1415
Neuron 37, 899–909 (2003). (2006). (2013).

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

176. Ory-Magne, F. et al. Withdrawing amantadine in 198. Svenningsson, P. et al. Eltoprazine counteracts 219. Braak, H. et al. Staging of brain pathology related to
dyskinetic patients with Parkinson disease: the L‑DOPA-induced dyskinesias in Parkinson’s disease: a sporadic Parkinson’s disease. Neurobiol. Aging 24,
AMANDYSK trial. Neurology 82, 300–307 (2014). dose-finding study. Brain 138, 963–973 (2015). 197–211 (2003).
177. Vijayakumar, D. & Jankovic, J. Drug-induced 199. Chung, K. A., Lobb, B. M., Nutt, J. G. & Horak, F. B. 220. Steiner, J. A., Quansah, E. & Brundin, P. The concept of
dyskinesia, part 1: treatment of levodopa-induced Effects of a central cholinesterase inhibitor on alpha-synuclein as a prion-like protein: ten years after.
dyskinesia. Drugs 76, 759–777 (2016). reducing falls in Parkinson disease. Neurology 75, Cell Tissue Res. 373, 161–173 (2018).
178. Vale, J. A. & Maclean, K. S. Amantadine-induced 1263–1269 (2010). 221. Park, J. S., Davis, R. L. & Sue, C. M. Mitochondrial
heart-failure. Lancet 1, 548 (1977). 200. Moreau, C. et al. Methylphenidate for gait dysfunction in Parkinson’s disease: new mechanistic
179. Flaherty, J. A. & Bellur, S. N. Mental side effects hypokinesia and freezing in patients with Parkinson’s insights and therapeutic perspectives. Curr. Neurol.
of amantadine therapy: its spectrum and disease undergoing subthalamic stimulation: a Neurosci. Rep. 18, 21 (2018).
characteristics in a normal population. J. Clin. multicentre, parallel, randomised, placebo-controlled This review focuses on recent advances in
Psychiatry 42, 344–345 (1981). trial. Lancet Neurol. 11, 589–596 (2012). understanding of the role that mitochondrial
180. Xu, W. J., Wei, N., Xu, Y. & Hu, S. H. Does amantadine 201. Hauser, R. A., Heritier, S., Rowse, G. J., Hewitt, L. A. & dysfunction plays in the pathogenesis of both
induce acute psychosis? A case report and literature Isaacson, S. H. Droxidopa and reduced falls in a trial sporadic and familial PD.
review. Neuropsychiatr. Dis. Treat. 12, 781–783 of Parkinson disease patients with neurogenic 222. Winklhofer, K. F. & Haass, C. Mitochondrial dysfunction
(2016). orthostatic hypotension. Clin. Neuropharmacol. 39, in Parkinson’s disease. Biochim. Biophys. Acta 1802,
181. Krystal, J. H. et al. Comparative and interactive 220–226 (2016). 29–44 (2010).
human psychopharmacologic effects of ketamine and 202. Albin, R. L., Young, A. B. & Penney, J. B. The functional 223. Pissadaki, E. K. & Bolam, J. P. The energy cost of action
amphetamine: implications for glutamatergic and anatomy of basal ganglia disorders. Trends Neurosci. potential propagation in dopamine neurons: clues to
dopaminergic model psychoses and cognitive function. 12, 366–375 (1989). susceptibility in Parkinson’s disease. Front. Comput.
Arch. Gen. Psychiatry 62, 985–994 (2005). 203. Graybiel, A. M. The basal ganglia. Curr. Biol. 10, Neurosci. 7, 13 (2013).
182. Blanpied, T. A., Clarke, R. J. & Johnson, J. W. R509–R511 (2000). 224. Bolam, J. P. & Pissadaki, E. K. Living on the edge with
Amantadine inhibits NMDA receptors by accelerating 204. Kravitz, A. V. et al. Regulation of parkinsonian motor too many mouths to feed: why dopamine neurons die.
channel closure during channel block. J. Neurosci. 25, behaviours by optogenetic control of basal ganglia Mov. Disord. 27, 1478–1483 (2012).
3312–3322 (2005). circuitry. Nature 466, 622–626 (2010). 225. Surmeier, D. J., Guzman, J. N., Sanchez-Padilla, J. &
183. Pahwa, R. et al. ADS‑5102 (amantadine) extended- 205. Cui, G. et al. Concurrent activation of striatal direct Schumacker, P. T. The role of calcium and mitochondrial
release capsules for levodopa-induced dyskinesia in and indirect pathways during action initiation. Nature oxidant stress in the loss of substantia nigra pars
Parkinson disease (EASE LID study): a randomized 494, 238–242 (2013). compacta dopaminergic neurons in Parkinson’s disease.
clinical trial. JAMA Neurol. 74, 941–949 (2017). This study provides the first definitive evidence Neuroscience 198, 221–231 (2011).
184. Pahwa, R. et al. Amantadine extended release for that direct and indirect pathway striatal neurons 226. Audano, M., Schneider, A. & Mitro, N. Mitochondria,
levodopa-induced dyskinesia in Parkinson’s disease are co‑activated during movement initiation and lysosomes and dysfunction: their meaning in
(EASED study). Mov. Disord. 30, 788–795 (2015). that an adequate coordination between both neurodegeneration. J. Neurochem. https://doi.
185. Gubellini, P., Iskhakova, L., Smith, Y. & Amalric, M. in pathways is important for the execution of org/10.1111/jnc.14471 (2018).
mGLU Receptors (The Receptors) 1st edn (eds appropriate movements. 227. Remondelli, P. & Renna, M. The endoplasmic reticulum
Ngomba, R. T., Di Giovanni, G., Battaglia, G., 206. Cahill, E., Salery, M., Vanhoutte, P. & Caboche, J. unfolded protein response in neurodegenerative
Nicoletti, F.) Vol. 31 33–57 (Humana Press, Cham, Convergence of dopamine and glutamate signaling disorders and its potential therapeutic significance.
2017). onto striatal ERK activation in response to drugs of Front. Mol. Neurosci. 10, 187 (2017).
186. Conn, P. J., Battaglia, G., Marino, M. J. & Nicoletti, F. abuse. Front. Pharmacol. 4, 172 (2014). 228. Minakaki, G. et al. Autophagy inhibition promotes
Metabotropic glutamate receptors in the basal ganglia 207. Cazorla, M., Kang, U. J. & Kellendonk, C. Balancing SNCA/alpha-synuclein release and transfer via
motor circuit. Nat. Rev. Neurosci. 6, 787–798 the basal ganglia circuitry: a possible new role for extracellular vesicles with a hybrid autophagosome-
(2005). dopamine D2 receptors in health and disease. Mov. exosome-like phenotype. Autophagy 14, 98–119
187. Gregoire, L. et al. The acute antiparkinsonian and Disord. 30, 895–903 (2015). (2018).
antidyskinetic effect of AFQ056, a novel metabotropic 208. Kim, J. et al. Inhibitory basal ganglia inputs induce 229. Gao, F. et al. Mitophagy in Parkinson’s disease:
glutamate receptor type 5 antagonist, in L‑DOPA- excitatory motor signals in the thalamus. Neuron 95, pathogenic and therapeutic implications. Front. Neurol.
treated parkinsonian monkeys. Parkinsonism Relat. 1181–1196 (2017). 8, 527 (2017).
Disord. 17, 270–276 (2011). 209. Bateup, H. S. et al. Distinct subclasses of medium 230. Tarrade, A. et al. A mutation of spastin is responsible
188. Berg, D. et al. AFQ056 treatment of levodopa- spiny neurons differentially regulate striatal motor for swellings and impairment of transport in a region of
induced dyskinesias: results of 2 randomized behaviors. Proc. Natl Acad. Sci. USA 107, axon characterized by changes in microtubule
controlled trials. Mov. Disord. 26, 1243–1250 14845–14850 (2010). composition. Hum. Mol. Genet. 15, 3544–3558
(2011). 210. Durieux, P. F. et al. D2R striatopallidal neurons inhibit (2006).
189. Bezard, E. et al. The mGluR5 negative allosteric both locomotor and drug reward processes. Nat. 231. Morfini, G. A. et al. Axonal transport defects in
modulator dipraglurant reduces dyskinesia in the Neurosci. 12, 393–395 (2009). neurodegenerative diseases. J. Neurosci. 29,
MPTP macaque model. Mov. Disord. 29, 1074–1079 211. Szewczyk-Krolikowski, K. et al. Functional connectivity 12776–12786 (2009).
(2014). in the basal ganglia network differentiates PD 232. Burre, J. The synaptic function of α-synuclein.
190. Rascol, O. et al. Use of metabotropic glutamate patients from controls. Neurology 83, 208–214 J. Parkinsons Dis. 5, 699–713 (2015).
5‑receptor antagonists for treatment of levodopa- (2014). 233. Guridi, J. & Alegre, M. Oscillatory activity in the basal
induced dyskinesias. Parkinsonism Relat. Disord. 20, 212. Rana, A. Q., Ahmed, U. S., Chaudry, Z. M. & Vasan, S. ganglia and deep brain stimulation. Mov. Disord. 32,
947–956 (2014). Parkinson’s disease: a review of non-motor symptoms. 64–69 (2017).
This article provides a review on the evidence Expert Rev. Neurother. 15, 549–562 (2015). 234. Picconi, B. et al. Loss of bidirectional striatal synaptic
supporting the development of mGlu5 NAMs for 213. Prakash, K. M., Nadkarni, N. V., Lye, W. K., plasticity in L‑DOPA-induced dyskinesia. Nat. Neurosci.
the treatment of LIDs. Yong, M. H. & Tan, E. K. The impact of non-motor 6, 501–506 (2003).
191. Tison, F. et al. A phase 2A trial of the novel symptoms on the quality of life of Parkinson’s disease 235. Caligiore, D. et al. Parkinson’s disease as a system-level
mGluR5‑negative allosteric modulator dipraglurant for patients: a longitudinal study. Eur. J. Neurol. 23, disorder. NPJ Parkinsons Dis. 2, 16025 (2016).
levodopa-induced dyskinesia in Parkinson’s disease. 854–860 (2016). 236. Blandini, F., Nappi, G., Tassorelli, C. & Martignoni, E.
Mov. Disord. 31, 1373–1380 (2016). 214. Erro, R. et al. The non-motor side of the honeymoon Functional changes of the basal ganglia circuitry in
192. Charvin, D. et al. Discovery, structure-activity period of Parkinson’s disease and its relationship with Parkinson’s disease. Prog. Neurobiol. 62, 63–88
relationship and anti-parkinsonian effect of a potent quality of life: a 4‑year longitudinal study. Eur. (2000).
and brain-penetrant chemical series of positive J. Neurol. 23, 1673–1679 (2016). 237. Mellone, M. & Gardoni, F. Glutamatergic mechanisms
allosteric modulators of metabotropic glutamate 215. Cummings, J. et al. Pimavanserin for patients with in L‑DOPA-induced dyskinesia and therapeutic
receptor 4. J. Med. Chem. 60, 8515–8537 (2017). Parkinson’s disease psychosis: a randomised, placebo- implications. J. Neural Transm. 125, 1225–1236
193. Charvin, D. et al. in 9th International Meeting On controlled phase 3 trial. Lancet 383, 533–540 (2018).
Metabotropic Glutamate Receptors (Sicily, Italy, 2017). (2014). 238. Kaur, K., Gill, J. S., Bansal, P. K. & Deshmukh, R.
194. Charvin, D. et al. An mGlu4 positive allosteric 216. Ghiglieri, V., Calabrese, V. & Calabresi, P. Alpha- Neuroinflammation - a major cause for striatal
modulator alleviates parkinsonism in primates. Mov. synuclein: from early synaptic dysfunction to dopaminergic degeneration in Parkinson’s disease.
Disord. https://doi.org/10.1002/mds.27462 (2018). neurodegeneration. Front. Neurol. 9, 295 (2018). J. Neurol. Sci. 381, 308–314 (2017).
This article provides a complete characterization This paper provides an outlook of seminal and 239. Przedborski, S. The two-century journey of Parkinson
and the first evidence that an mGlu4 PAM has both recent experimental studies that explore the roles disease research. Nat. Rev. Neurosci. 18, 251–259
antiparkinsonian and antidyskinetic efficacy in of α-synuclein. (2017).
primates 217. Angot, E., Steiner, J. A., Hansen, C., Li, J. Y. & 240. Foltynie, T. & Athauda, D. Glucagon-like peptides
195. Carta, M., Carlsson, T., Kirik, D. & Bjorklund, A. Brundin, P. Are synucleinopathies prion-like (GLP‑1) perspectives in synucleinopathies treatment.
Dopamine released from 5‑HT terminals is the cause disorders? Lancet Neurol. 9, 1128–1138 (2010). Mov. Disord. Clin. Pract. 5, 255–258 (2018).
of L‑DOPA-induced dyskinesia in parkinsonian rats. 218. Brundin, P., Melki, R. & Kopito, R. Prion-like 241. Eskow, K. L., Gupta, V., Alam, S., Park, J. Y. & Bishop, C.
Brain 130, 1819–1833 (2007). transmission of protein aggregates in The partial 5‑HT(1A) agonist buspirone reduces the
196. Kleedorfer, B., Lees, A. J. & Stern, G. M. Buspirone in neurodegenerative diseases. Nat. Rev. Mol. Cell Biol. expression and development of l‑DOPA-induced
the treatment of levodopa induced dyskinesias. 11, 301–307 (2010). dyskinesia in rats and improves L‑DOPA efficacy.
J. Neurol. Neurosurg. Psychiatry 54, 376–377 (1991). This article marks the start of a new research area Pharmacol. Biochem. Behav. 87, 306–314 (2007).
197. Paolone, G., Brugnoli, A., Arcuri, L., Mercatelli, D. & that is focused on the cell‑to‑cell propagation of 242. Prexton Therapeutics. Parkinson’s disease: Prexton
Morari, M. Eltoprazine prevents levodopa-induced protein aggregates in neurodegenerative diseases Therapeutics starts phase 1 clinical trial. Prexton
dyskinesias by reducing striatal glutamate and direct such as PD, Alzheimer disease or Huntington Therapeutics http://www.prextontherapeutics.com/
pathway activity. Mov. Disord. 30, 1728–1738 disease on the basis of observations of Lewy news/press_release/6-parkinson’s_disease__prexton_
(2015). bodies in grafted neurons in PD. therapeutics_starts_phase_1_clinical_trial.html (2016).

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrd


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

243. Prexton Therapeutics. Prexton announces initiation of 249. Stahl, S. M. Mechanism of action of pimavanserin in eResearch Technology, Inc., Lundbeck, Cynapsus, Sarepta
phase II clinical testing in Parkinson’s disease. Prexton Parkinson’s disease psychosis: targeting serotonin Therapeutics, Adamas Pharmaceuticals, Neurocrine
Therapeutics http://www.prextontherapeutics.com/ 5HT2A and 5HT2C receptors. CNS Spectr. 21, Biosciences, Back Bay Life Science, US WorldMeds, Biotie
news/press_release/9-prexton_announces_initiation_ 271–275 (2016). Therapies, MJFF, Neuropore Therapies, the US National
of_phase_ii_clinical_testing_in_parkinson’s_disease. 250. Weintraub, D. et al. in Thirtieth Annual Symposium on Institutes of Health, Projects in Knowledge, Prexton
html (2017). Etiology, Pathogenesis, and Treatment of Parkinson Therapeutics, Acorda Therapeutics, Vista Research, LifeMax,
244. Zesiewicz, T. A., Sullivan, K. L., Freeman, A. & Disease and Other Movement Disorders (Portland, Peerview Press, ClinicalMind Medical and Therapeutic
Juncos, J. L. Treatment of imbalance with varenicline USA, 2016). Communications, Sunovion Pharmaceuticals, Inc., the
Chantix(R): report of a patient with fragile X tremor/ Academy for Continued Healthcare Learning, Outcomes
ataxia syndrome. Acta Neurol. Scand. 119, 135–138
Acknowledgements Insights, Expert Connect, HealthLogix, Teva, Cowen and
D.C. thanks P. Hongaard for inspiring this Review.
(2009). Company, Pharma Two B, Ltd., Pfizer, RMEI Medical
245. Zhang, D. et al. Nicotinic receptor agonists reduce Competing interests Education for Better Outcomes, ClearView Healthcare
L‑DOPA-induced dyskinesias in a monkey model of D.C. has received scientific grants from the Michael J. Fox Partners, Health Advances, Kyowa Kirin Pharmaceutical
Parkinson’s disease. J. Pharmacol. Exp. Ther. 347, Foundation (MJFF). D.C. and R.M. are members of the Development, Ltd., Impax Laboratories, Quintiles, Pfizer,
225–234 (2013). Management Team of Prexton Therapeutics. O.R. has AbbVie, AstraZeneca, Eli Lilly & Company, Decision Resources
246. Hubbard, D., Hacksell, U. & McFarland, K. Behavioral received scientific grants from Agence Nationale de Group, Seagrove Partners, LLC, Intec Pharma, Ltd.,
effects of clozapine, pimavanserin, and quetiapine in la Recherche, CHU de Toulouse, France-Parkinson, Schlesinger Associates, Huron Consulting Group, Pennside
rodent models of Parkinson’s disease and Parkinson’s INSERM-DHOS Recherche Clinique Translationnelle, MJFF, Partners, Bracket, Phase Five Communications, LCN
disease psychosis: evaluation of therapeutic ratios. Programme Hospitalier de Recherche Clinique and the Consulting and the Windrose Consulting Group.

Behav. Pharmacol. 24, 628–632 (2013). European Commission (FP7, H2020). O.R. is a scientific
247. McFarland, K., Price, D. L. & Bonhaus, D. W. adviser or consultant for AbbVie, Adamas Pharmaceuticals, Publisher’s note
Pimavanserin, a 5‑HT2A inverse agonist, reverses Acorda Therapeutics, Addex Therapeutics, AlzProtect, Springer Nature remains neutral with regard to jurisdictional
psychosis-like behaviors in a rodent model of ApoPharma, AstraZeneca, Bial, Biogen, Britannia, Clevexel, claims in published maps and institutional affiliations.
Parkinson’s disease. Behav. Pharmacol. 22, 681–692 Cynapsus, INC Research, Lundbeck, Merck, MundiPharma,
(2011). Neuroderm, Novartis, Oxford Biomedica, Parexel, Pfizer,
248. Meltzer, H. Y. et al. Pimavanserin, a serotonin(2A) Prexton Therapeutics, Quintiles, Sanofi, Servier, Teva, UCB, RELATED LINKS
receptor inverse agonist, for the treatment of XenoPort and Zambon. R.A.H. reports consulting fees from ClinicalTrials.gov: https://clinicaltrials.gov/
parkinson’s disease psychosis. Neuropsycho­pharma­ Guidepoint Global, Gerson Lehrman Group, LCN Consulting, ALL LINKS ARE ACTIVE IN THE ONLINE PDF
cology 35, 881–892 (2010). Putnam Associates, the National Parkinson Foundation,

NATURE REVIEWS | DRUG DISCOVERY ADVANCE ONLINE PUBLICATION | 19


©
2
0
1
8
S
p
r
i
n
g
e
r
N
a
t
u
r
e
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like