You are on page 1of 14

Neuropharmacology 202 (2022) 108822

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

New therapeutic approaches to Parkinson’s disease targeting GBA, LRRK2


and Parkin
Konstantin Senkevich a, b, c, Uladzislau Rudakou a, b, d, Ziv Gan-Or a, b, d, *
a
The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
b
Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada
c
First Pavlov State Medical University of St. Petersburg, Saint-Petersburg, Russia
d
Department of Human Genetics, McGill University, Montréal, QC, Canada

A R T I C L E I N F O A B S T R A C T

Keywords: Parkinson’s disease (PD) is defined as a complex disorder with multifactorial pathogenesis, yet a more accurate
Parkinson’s disease definition could be that PD is not a single entity, but rather a mixture of different diseases with similar phe­
Genetic targets notypes. Attempts to classify subtypes of PD have been made based on clinical phenotypes or biomarkers.
Clinical trials
However, the most practical approach, at least for a portion of the patients, could be to classify patients based on
GBA
LRRK2
genes involved in PD. GBA and LRRK2 mutations are the most common genetic causes or risk factors of PD, and
PRKN PRKN is the most common cause of autosomal recessive form of PD. Patients carrying variants in GBA, LRRK2 or
PRKN differ in some of their clinical characteristics, pathology and biochemical parameters. Thus, these three
PD-associated genes are of special interest for drug development. Existing therapeutic approaches in PD are
strictly symptomatic, as numerous clinical trials aimed at modifying PD progression or providing neuro­
protection have failed over the last few decades. The lack of precision medicine approach in most of these trials
could be one of the reasons why they were not successful. In the current review we discuss novel therapeutic
approaches targeting GBA, LRRK2 and PRKN and discuss different aspects related to these genes and clinical
trials.

1. Introduction The most recent genome-wide association study (GWAS) calculated


the heritability of PD as 22% (Nalls et al., 2019), yet only a small pro­
Parkinson’s disease (PD) is a neurodegenerative disorder with portion of PD patients carry pathogenic variants with Mendelian in­
multifactorial pathogenesis and complex phenotypes, including various heritance. LRRK2 and GBA mutations are amongst the most common
motor and non-motor symptoms. Multiple lines of evidence suggest that genetic causes or risk factors of PD (Gan-Or et al., 2015a; Healy et al.,
instead of viewing PD as a single disorder, it should be considered a 2008; Ross et al., 2011). The frequency of LRRK2 variants in PD is
mixture of different diseases with similar phenotypes (Espay et al., 2020; 1–40% and GBA variants are found in 5–20% of PD patients, depending
Sauerbier et al., 2016). Many attempts have been made to classify PD on the population (Gan-Or et al., 2015a; Healy et al., 2008; Ross et al.,
into subtypes based on clinical presentation, neuroimaging and bio­ 2011). Other mutations in familial genes are relatively rare and repre­
markers (Campbell et al., 2020; Fereshtehnejad et al., 2017; Lawton sent up to 1–2% of all PD cases. Bi-allelic variants in PRKN are the most
et al., 2018; Qian and Huang, 2019; Thenganatt and Jankovic, 2014). common autosomal recessive cause of PD, and can explain about 8–15%
However, the most viable approach could be to classify patients based of early-onset PD (with age at onset, AAO, below 50 years) (Alcalay
on similar underlying disease mechanisms, potentially with similar et al., 2010a; Bonifati, 2014; Kilarski et al., 2012; Klein and West­
biomarkers, that lead to somewhat distinct phenotypes. One of the ways enberger, 2012). In this review, we only refer to bi-allelic PRKN muta­
this could be done is by using causative gene stratification, so that tion carriers, as the role of heterozygous PRKN mutations in PD is
treatment development could specifically target these genes (von Lin­ controversial (Clark et al., 2006; Lubbe et al., 2021; Yu et al., 2021).
stow et al., 2020). Patients carrying variants in LRRK2, GBA or PRKN may differ in some of

* Corresponding author. Department of Neurology and Neurosurgery McGill University 1033 Pine Avenue, West, Ludmer Pavilion, room 312, Montreal, QC, H3A
1A1, Canada.
E-mail address: ziv.gan-or@mcgill.ca (Z. Gan-Or).

https://doi.org/10.1016/j.neuropharm.2021.108822
Received 28 June 2021; Received in revised form 1 October 2021; Accepted 4 October 2021
Available online 7 October 2021
0028-3908/© 2021 Published by Elsevier Ltd.
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

their clinical characteristics and pathology, compared to sporadic PD Table 1


(Fig. 1; Table 1) (Alcalay et al., 2010b, 2015b; Kalia et al., 2015; Differences of clinical and pathological features of GBA, LRRK2 and PRKN
Schneider and Alcalay, 2017; Thaler et al., 2018). Patients with LRRK2 associated PD.
mutations have a more benign course of PD, as they progress slower and Clinical/Pathological GBA associated LRRK2 PRKN associated
have less non-motor symptoms than sporadic PD, whereas GBA carriers feature PD associated PD PD
have more malignant course with faster progression and more Lewy body pathology + +/− –
non-motor symptoms. Patients with biallelic PRKN mutations have an Early age at onset + – +
early onset, very slowly progressive PD, which responds well to rela­ Motor symptoms + – –
progression
tively low doses of levodopa (Alcalay et al., 2014; Piredda et al., 2020).
Cognitive deficit + – –
Moreover, α-synuclein pathology is only present in about 50% of LRRK2 Psychiatric symptoms + – +/−
mutation carriers and rarely found in PRKN mutation carriers with PD Olfactory dysfunction + – –
(Kalia et al., 2015; Schneider and Alcalay, 2017), in whom the neuro­
+ more prevalent than in sporadic Parkinson’s disease; - less prevalent than in
degenerative process is typically limited to the substantia nigra sporadic Parkinson’s disease; +/− conflicting reports.
(Schneider and Alcalay, 2017). Thus, these three relatively common
PD-associated genes lead to distinct PD subtypes and represent specific
mutations are also associated with other synucleinopathies such as de­
interest for drug development.
mentia with Lewy bodies and REM sleep behavior disorder (RBD) (Chia
Current therapeutic approaches in PD are symptomatic and do not
et al., 2021; Krohn et al., 2020b; Mata et al., 2008; Nalls et al., 2013), yet
involve disease modification. Moreover, most current drugs, whether in
it is still unclear whether GBA is associated with multiple system atro­
development or in clinical trials, target PD as a single clinical entity, and
phy, a more severe form of synucleinopathy (Table 2) (Mitsui et al.,
do not consider stratification by subtypes. This lack of stratification may
2015; Sklerov et al., 2017; Wernick et al., 2020).
be one of the reasons for our failure to develop disease modifying
GBA encodes the lysosomal enzyme, glucocerebrosidase (GCase),
therapies for PD. We suggest that our best chance to treat PD is by
responsible for the degradation of several glycolipids such as gluco­
identifying and treating specific subtypes of the disease. Out of all the
sylceramide (GlcCer) and glucosylsphyngosin (GlcSph). GCase is syn­
known genetic targets, GBA, LRRK2 and PRKN are likely the best can­
thesized in the endoplasmic reticulum (ER) (Do et al., 2019), and if
didates for drug development, although multiple challenges exist when
folded correctly, transported to the Golgi complex for processing and
designing the clinical trials for patients with mutations in these genes.
then to the lysosome by LIMP2 (Reczek et al., 2007).
There are several different hypotheses regarding the pathogenesis of
2. GBA in PD – genetic, enzymatic, clinical and therapeutic
GBA-associated PD, including loss of function, accumulation of GCase
considerations
substrates, alterations in the lysosomal membrane composition, ER
accumulation and ER stress due to misfolding, autophagy dysfunction,
2.1. GBA and PD
mitochondrial dysfunction, mitophagy dysfunction and neuro­
inflammation (Gan-Or et al., 2015b; Senkevich and Gan-Or, 2020). Most
Homozygous variants in GBA may cause a rare lysosomal storage
of these hypotheses are based on loss of function of GCase, which is
disorder, Gaucher disease (GD) (Stirnemann et al., 2017). Throughout
further supported by studies reporting reduced GCase activity in blood,
the 20th Century, scattered reports have been published on GD patients
CSF and brains of patients without GBA mutations (Alcalay et al., 2015a;
and their relatives (carriers of GBA variants) who developed PD (Miller
Gegg et al., 2012; Parnetti et al., 2014). One hypothesis suggests that the
et al., 1973; Neil et al., 1979; Soffer et al., 1980; Van Bogaert and
reduction of GCase activity may lead to the accumulation and oligo­
Froehlich, 1939). First genetic association studies of GBA mutations in
merization of α-synuclein due to its direct interaction with GlcCer
PD cohorts were reported in the early 2000s and were confirmed in a
(Mazzulli et al., 2011). Accumulated α-synuclein may block the trans­
large multicenter study (Aharon-Peretz et al., 2004; Lwin et al., 2004;
port of GCase from the ER to the lysosome, leading to further reduction
Sidransky et al., 2009), followed by numerous other studies. GBA
of GCase activity (Mazzulli et al., 2011). α-synuclein could also directly
interact with GCase in acid conditions further promoting its inhibition
(Yap et al., 2013). Other studies suggest that GlcCer metabolites such as
glucosylsphingosine (GlcSph) have higher potency to promote α-synu­
clein fibrils formation by direct interaction than GlcCer (Taguchi et al.,
2017). In human brains, there are conflicting evidence for accumulation
of sphingolipids (Gegg et al., 2015; Huebecker et al., 2019), which
question the suggested mechanisms involving GlcCer or GlcSph
accumulation.
Different GBA variants may have different effects on disease patho­
genesis. Some variants such as p.L444P and to a much lesser extent, p.

Table 2
Frequency of GBA variants in patients with synucleinopathies.
Synucleinopathy Frequency of all GBA References
variants

PD 5–20%* Gan-Or et al. (2015c)


DLB 8.1–32%* (Nalls et al., 2013; Shiner et al., 2021;
Tsuang et al., 2012)
MSA 10.8% Wernick et al. (2020)
RBD 9.5% Krohn et al. (2020b)

PD- Parkinson’s disease, DLB – dementia with Lewy bodies, MSA-multiple sys­
tem atrophy, RBD – REM sleep behaviour disorder. * high frequency; 20% in PD
Fig. 1. Parkinson’s disease (PD) genetic subtypes under PD as an umbrella term and 32% in DLB are due to inclusion of studies with Ashkenazi Jewish
on the example of PRKN, LRRK2 and GBA carriers. population.

2
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

N370S, may lead to ER retention (Fernandes et al., 2016; Ron and of cognitive decline in GBA-PD patients after deep brain stimulation
Horowitz, 2005; Thomas et al., 2021). This retention can lead to loss of (Lythe et al., 2017; Mangone et al., 2020).
function since GCase does not reach the lysosome, or it can lead to gain Classification of GBA variants can be taken from GD, where severe
of toxic function if retention leads to ER stress and cell death (Fernandes variants (e.g. p.L444P, c.84GG, p.V394L) lead to the neuronopathic
et al., 2016; Thomas et al., 2021). Truncating GBA mutations such as forms of GD (types 2 and 3) and mild variants (e.g. p.N370S, p.R496H)
c.84GG and IVS2+1 result in absolute absence of the protein (Beutler, lead to the non-neuronopathic form of GD (type 1). Several other GBA
2006), which argues for a loss of function mechanism. Other variants variants are associated with increased PD risk (p.T369M, p.E326K) but
may affect the function of the enzyme by changing the conformation of do not cause clinical GD (Berge-Seidl et al., 2017; Mallett et al., 2016). In
the active site (Toffoli et al., 2020). Given these distinctive effects of PD, severe GBA variants lead to a more severe PD phenotype compared
different GBA variants, drug development for GBA-associated PD should to mild variants (Cilia et al., 2016; Gan-Or et al., 2008; Thaler et al.,
take these differences into account, which may lead to mutation-specific 2018). Moreover, the phenotype of GBA-PD has a dose-dependent effect,
treatments in the future. as patients with homozygous or compound heterozygous variants may
have earlier AAO and more severe motor impairment and faster cogni­
2.2. GBA-PD biomarkers – current status tive decline (Thaler et al., 2017). It is therefore possible that the levels of
GCase activity are associated with PD severity, yet to further examine
Homozygous and compound heterozygous GBA variants causing GD this hypothesis, reliable lysosomal GCase activity measurement methods
lead to a significant reduction of GCase activity, making it an excellent need to be established.
biomarker for early GD detection (Ho et al., 1972). Similarly, the
accumulation of GCase substrate GlcCer and its deacetylated form, 2.4. GBA-targeted therapy
GlcSph, leads to the formation of enlarged macrophages, called Gaucher
cells, which are deposited in organs and tissues and cause GD symptoms 2.4.1. Substrate-reduction therapy
(Stirnemann et al., 2017). Thus, the level of GlcSph can serve as a One of the suggested strategies for treatment of GBA-PD is substrate
biomarker of GD progression and response to treatment (Dekker et al., reduction therapy, assuming that GlcCer and/or GlcSph may accumulate
2011; Rolfs et al., 2013). and be involved in the pathogenic process in GBA-PD (Sardi et al., 2017;
In PD, however, neither reduced GCase activity nor accumulation of Taguchi et al., 2017). Substrate reduction therapy, targeting excessive
GlcCer or GlcSph can be used as reliable biomarkers. While the average accumulation of GlcCer by glucosylceramide synthase inhibition, is
GCase activity has been repeatedly demonstrated to be reduced in blood, already being used in non-neuronopathic GD (eliglustat and miglustat)
cerebrospinal fluid (CSF), fibroblasts and brains of GBA variants carriers (Gary et al., 2018). Previous studies showed that miglustat may pene­
(Alcalay et al., 2015a; Gegg et al., 2012; Lerche et al.; Pchelina et al., trate the blood-brain barrier, yet it had no clinical effect on neuro­
2017), there is a big overlap between GCase activity levels in PD patients nopathic GD (Schiffmann et al., 2008; Treiber et al., 2007). Based on the
and controls with and without GBA variants (Alcalay et al., 2015a; Gegg findings suggesting that sphingolipids may induce and promote the
et al., 2012; Gündner et al., 2019; Moors et al., 2019). Similarly, con­ accumulation of α-synuclein, it was hypothesized that glucosylceramide
flicting evidence on GlcCer and GlcSph levels do not support these synthase inhibition could alleviate α-synuclein burden and PD symp­
substrates as potential biomarkers in GBA-PD (Gegg et al., 2015; Guedes toms (Sardi et al., 2017; Taguchi et al., 2017). Preclinical experiments
et al., 2017; Huebecker et al., 2019; Pchelina et al., 2018). Reduced were performed on murine models with transgenic α-synuclein p.A53T
GCase activity was also reported in blood and brains of a subgroup of overexpression and Gaucher model with homozygous p.D409V (Sardi
sporadic PD patients with age-dependent effect (Alcalay et al., 2015a; et al., 2017). This GBA variant has not been previously associated with
Huebecker et al., 2019; Muñoz et al., 2020; Rocha et al., 2015). While PD and was reported only in a single GD patient. In these models, the
this subgroup of patients may benefit from future GBA-targeted therapy, glucosylceramide synthase inhibitor GZ667161 demonstrated target
neither reduced GCase activity nor substrate levels are likely to be good engagement, reduction of α-synuclein accumulation and improvement
biomarkers. of cognitive symptoms (Sardi et al., 2017).
Previous studies have suggested that some immune markers may be These findings prompted the researchers to continue the clinical
associated with GBA-PD (Chahine et al., 2013; Miliukhina et al., 2020), development of the glucosylceramide synthase inhibitor GZ667161
whereas other studies showed no difference in immune biomarkers be­ analog venglustat (GZ/SAR402671) in humans (Peterschmitt et al.,
tween GBA-PD, sporadic PD and other subgroups of the disease (Galper 2021) (Table 3). Recently, three phase 1 studies on healthy volunteers
et al.; Thaler et al., 2021). As there are no reliable biomarkers for reported on the safety and tolerability of venglustat (Peterschmitt et al.,
GBA-PD and its progression, additional studies are needed to identify 2021). The safety profile allowed to start a phase 2 placebo-controlled
such biomarkers. study, MOVES-PD, including 221 GBA-PD patients (NCT02906020).
The study was divided into two stages. Safety, tolerability and target
2.3. Clinical features of GBA-PD engagement were demonstrated in GBA-PD patients during the first part
of the study, reported in 2019 (Fischer et al., 2019). The results of the
On the individual level, it is difficult to identify a GBA-PD patient second part of MOVES-PD study were recently reported in the
based on their clinical presentation, as their symptoms are overall AD/PD-2021 and MDS 2021 conferences (2021; J. Peterschmitt et al.,
typical. However, as a group, the disease course of GBA-PD patients is 2021). Target engagement was clearly demonstrated by a significant
more severe: they have earlier average AAO, higher frequencies of non- decrease (~75%) of GlcCer levels in the CSF as well as in plasma of
motor symptoms, and faster motor and non-motor progression patients receiving venglustat. The primary and secondary endpoints
compared to sporadic PD (Alcalay et al., 2012; Cilia et al., 2016; Creese were changes in MDS-UPDRS parts I, II and III and PD cognitive rating
et al., 2018; Liu et al., 2016; Maple-Grødem et al., 2021; Zhang et al., scale (PD-CRS) at 52 weeks. The primary motor endpoint, sum of
2018). Multiple studies have demonstrated faster cognitive decline and MDS-UPDRS parts II and III, was increased in the treatment group (7.29
higher frequency of psychiatric symptoms in GBA-PD (Alcalay et al., ± 1.36) compared to the placebo group (4.71 ± 1.27; p = 0.17),
2012; Cilia et al., 2016; Creese et al., 2018; Iwaki et al., 2019; Liu et al., meaning that those who received the treatment deteriorated faster.
2016). GBA-PD patients also have higher incidence of anxiety, depres­ PD-CRS total score was decreased in the venglustat treated group
sion and hallucinations as compared to non GBA carriers (Creese et al., (− 0.73 ± 1.36), demonstrating cognitive deterioration. Moreover, a
2018; Swan et al., 2016). A meta-analysis reported a higher occurrence higher rate of psychiatric disorders was reported in treatment group
of wearing-off and faster motor progression compared to non-carriers compared to placebo group. The progression of symptoms was reported
(Zhang et al., 2018). Moreover, recent studies suggested increased rate starting week 13 of treatment. These negative effect on motor symptoms

3
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

Table 3
Therapeutic approaches for GBA.
Type of Mechanism Drug name Registered clinical trial Cohort Phase Current status
treatment (clinicaltrials.gov)

Substrate Glucosylceramide synthase inhibition Venglustat (GZ/ NCT02906020 GBA-PD 2 Halted due to negative effect on
reduction SAR402671) clinical phenotype
therapy
Chaperone Inhibitory chaperone that facilitates Ambroxol NCT02941822 GBA-PD and 2 Positive outcome in terms of safety
transportation of GCase to the lysosome sporadic PD and target engagement
Ambroxol NCT02914366 PD-dementia 2 The study in process
Activator GCase activator BIA 28–6156/LTI- – GBA-PD 2 The study in process
291
Gene therapy Replacement of mutated GBA with WT PR001 NCT04127578 GBA-PD 1/2a Preliminary report. Recovery of
copy of the gene GCase activity. Possible severe
immune response

and cognition led to discontinuation of the development of venglustat suggested a dual role of ambroxol as a competitive and non-competitive
for GBA-PD. inhibitor in the GBA p.N370S model (Kopytova et al., 2021; Maegawa
Despite sparse clinical data and the lack of relevant preclinical et al., 2009). It is also possible that variants leading to conformational
models, venglustat demonstrated target engagement and significantly changes of the GCase active site could decrease ambroxol binding. This
decreased concentration of GlcCer in clinical trials. Nevertheless, dete­ might mean that ambroxol (or other chaperones) could be beneficial
rioration of PD symptoms was reported rather than improvement, sug­ only for specific GBA variants and not for others. This possibility should
gesting that substrate reduction therapy may not be a useful approach be further explored and considered in the future design of clinical trials.
for GBA-PD. There are many possible reasons for clinical trial failures, Isofagomine is a potent competitive inhibitor of GCase. Preclinical
and in this case, we should also consider that either the hypothesis was studies on isofagomine on PD and GD models in vitro and in vivo
wrong and there is no GlcCer accumulation, or that even if GlcCer demonstrated efficacy for GCase activation (Khanna et al., 2010; Korn­
accumulation exists, it does not contribute to PD progression. haber et al., 2008; Richter et al., 2014; Sanchez-Martinez et al., 2016;
Why did patients deteriorate in the venglustat trial? Sun et al., 2012). Despite showing some evidence for efficacy in pre­
Sphingolipids are very important components of cellular membranes clinical trials for GD, a phase 2 clinical trial in GD patients was halted
(Posse de Chaves and Sipione, 2010), and GlcCer has a central role in due to lack of efficacy (NCT00446550). Isofagomine has not been tested
sphingolipid homeostasis, as it is an intermediate metabolite and a in PD.
precursor of the vast majority of sphingolipids (Gault et al., 2010). Recently, a positive effect of the heat shock protein amplifier, ari­
GlcCer can be further converted to GlcSph through deacetylation and to moclomol, on GCase activity has been demonstrated in primary fibro­
LacCer by the addition of galactose (Gault et al., 2010; Olsen and Fær­ blasts and neuronal-like cells from GD patients (Fog et al., 2018).
geman, 2017). It is possible that by decreasing the levels of GlcCer in Arimoclomol is a well-known molecule and demonstrated some positive
neural tissue below normal values, sphingolipids metabolism is dis­ biochemical and clinical effects in phase 2/3 open-label clinical trials for
rupted, and neuronal membranes get damaged. In support of this hy­ Niemann-Pick Disease Type C and Amyotrophic Lateral Sclerosis (NC
pothesis, peripheral neuropathy was reported as an adverse effect of T00706147; NCT02612129). This molecule is currently considered for
substrate reduction therapy in GD (Ficicioglu, 2008; Giraldo et al., PD clinical trials. Other new molecular chaperones are currently being
2018). In addition, hand tremor is one of the known common adverse developed (Aflaki et al., 2016; Burbulla et al., 2019).
effects of miglustat (Giraldo et al., 2018). If similar adverse effects occur
with venglustat, it could have increased the MDS-UPDRS score results in 2.4.3. Ambroxol in clinical trials
the treatment group. Recently, an open-label, non-randomized clinical trial of ambroxol
has been completed (Mullin et al., 2020) (Table 3). This study included
2.4.2. GCase activators in development eight patients with and ten patients without GBA mutations. Dose
Almost 20 years ago, a novel strategy to increase GCase activity was constituted 1260 mg orally daily for 180 days with follow-up assessment
demonstrated in fibroblasts from GD patients with homozygous GBA p. at 270 days mark. Ambroxol concentration increased in CSF and was at a
N370S mutations (Sawkar et al., 2002). Chaperones are small molecules level of 11% of its blood level, demonstrating partial blood-brain barrier
that can cross the blood-brain barrier, bind to GCase in the ER and permeability. Target engagement was demonstrated by increased GCase
facilitate its folding and transport to the lysosome. Different groups have activity in blood, although in the CSF it was decreased. The authors
shown that such chaperones can potentially increase GCase activity in suggested that in the CSF, ambroxol may inhibit GCase but cannot
the lysosome (Fernandes et al., 2016; Thomas et al., 2021). In recent dissolve from it due to the absence of cellular structures and lysosomes.
years, multiple strategies to increase GCase activity have been proposed This hypothesis was supported by in vitro experiments in acellular con­
(Han et al., 2020; Jung et al., 2016). At least 14 small molecules have ditions (Mullin et al., 2020). A moderate increase of α-synuclein levels in
been assessed for efficacy for GCase activation in GD, and preclinical the CSF has also been reported, yet the nature of this increase and its
results on five molecules were reported in GBA-PD models (isofagomine, relevancy to PD is unclear. Furthermore, most of these changes occurred
S-181, ambroxol, NCGC607 and LTI-291) (Aflaki et al., 2016; Burbulla in the group of non-carriers of GBA variants. Some positive changes in
et al., 2019; Han et al., 2020; Mullin et al., 2020). motor and cognitive performance have also been reported in the study.
Ambroxol is a pH-dependent competitive GCase inhibitory chap­ However, due to the lack of placebo group or blind randomization, these
erone. Various studies suggest a different effect of ambroxol depending results are difficult to interpret and require replication in additional
on the mutation, with reduced effect of ambroxol in p.L444P carriers trials. A phase 2 clinical trial (NCT02914366) testing ambroxol for
(Ivanova et al., 2018; Khanna et al., 2010; Luan et al., 2013; Maegawa treatment of PD with dementia, regardless of GBA status, is currently
et al., 2009; Sanchez-Martinez et al., 2016). Yet, molecular docking ongoing (Silveira et al., 2019). While this study examines a more specific
studies showed similar binding effect of ambroxol to p.N370S and p. population (PD with dementia), it is not limited exclusively to carriers of
L444P (Thirumal Kumar et al., 2019). Currently, there is no definite GBA variants. Therefore, there is a reduced likelihood of success,
explanation for the different effects of ambroxol in different experiments because not all PD patients with dementia necessarily have reduced
and on different genotypes (Ivanova et al., 2018, 2021). Several studies GCase activity. Overall, the current results support the continuation of

4
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

clinical trials testing ambroxol and similar molecules in GBA-associated 2.6. Strategies and future directions
PD, yet with careful consideration of the participant’s genotype and the
trial design. To design a trial for GBA-PD in which the true effects of the treatment
will be visible, several important factors should be considered:
2.4.4. LTI-291 as a GCase activator
Preclinical studies and clinical trials on other GCase activators are 1) Targeting mutations that fit the specific properties of the treatment –
currently ongoing. Two phase 1 trials (single and multiple ascending as different mutations have different effects on GCase structure and
doses) on allosteric GCase modulator LTI-291 (currently BIA 28–6156/ function, these properties need to be considered when deciding
LTI-291) have been completed, demonstrating safety and partial blood- which mutation carriers should be enrolled into the trial. For
brain permeability of the drug (den Heijer et al.). The authors report that example, a chaperone may be beneficial for mutations that lead to ER
preclinical experiments demonstrated the efficacy of LTI-291 by retention, but it may not be useful for mutations in which GCase is
increasing GCase activity and decreasing GlcCer accumulation for GBA not synthesized at all, such as the c.84GG mutation. GCase activators
p.E326K and p.N370S in vitro and in vivo. The single ascending dose arm that work in the lysosome may be beneficial for mutations that allow
of the study included 40 healthy volunteers and the multiple ascending GCase to be transported to the lysosome, but may not be beneficial
dose arm included 39 middle-aged or elderly healthy subjects. In total, for mutations that are fully retained in the ER. Therefore, in each
five carriers of GBA variants were included in the study including four p. trial, the researchers should consider the known properties of their
T369M carriers and one p.E326K. No serious adverse events were re­ drug and of the specific mutation carriers that they enroll into the
ported in both studies. The authors did not identify significant changes trial, to make sure that the treatment fits the known mechanism of
in glycosphingolipid level in response to LTI-291 and GCase activity was the mutation.
not measured (den Heijer et al.). 2) Balancing the trial arms with similar GBA mutations – Several studies
have suggested that the rates of motor and non-motor progression in
2.5. Gene therapy targeting GBA carriers of severe GBA mutations are faster than among mild GBA
mutations or the GBA risk factors p.E326K and p.T369M. If the
There are several possible approaches for gene therapy in GBA-PD, treatment and placebo arms of a trial will not be balanced in terms of
the most obvious being gene therapy using a viral vector containing a the ratio of severe to mild mutations, it may lead to one of the arms of
normal copy of the GBA gene. In mouse models, direct administration of the trial to progress faster than the other, unrelated to the treatment.
the GBA adenoviral vector into several brain regions led to a decrease in Since motor and non-motor symptoms are typically the end-points of
α-synuclein accumulation in the substantia nigra and striatum, and to an trials, this imbalance may lead to false positive or false negative
increase in GCase activity (Rocha et al., 2015). Direct brain adminis­ results, depending on which arm harbors more severe mutation
tration of adenoviral vectors involves a number of risks and limitations, carriers.
and could be practically difficult in patients, who are very different from 3) Potential GBA modifiers may bias the trial – The penetrance and AAO
one another, unlike the inbred mice. An alternative approach is the of GBA-PD, as well as the activity of GCase, may be affected by ge­
systemic administration of a viral vector with GBA. In a mouse model, a netic variants in genes such as CTSB, TMEM175 and LRRK2 (Alcalay
similar effect was shown with an increase in GCase activity and a et al., 2015a; Krohn et al., 2020a; Sosero et al., 2021). Whether these
decrease in the concentration of α-synuclein in the brain (Morabito and other variants affect the rate of progression of GBA-PD has not
et al., 2017). Systemic administration of the vector in a mouse model for been studied, yet it is a reasonable possibility. Hence, to avoid po­
the neuropathic form of GD also demonstrated a decrease in neuro­ tential effects of these genetic variants on the rate of progression and
degeneration and an increase in lifespan (Massaro et al., 2020). In subsequently on trial results, these potential modifiers should also be
humans, systemic injection of a viral vector with a gene may lead to taken into account during the randomization process, to ensure that
off-target effects, meaning that the effect on other genes could be un­ they do not bias the results. Finally, it was shown that PD polygenic
expected (Zhang et al., 2015). risk score (PRS) may affect GBA-PD penetrance and age at onset, and
Despite these potential limitations, recently the first PD patients have that imbalance in PRS between trial arms may lead to false positive
received gene therapy with the GBA gene, in a Phase 1/2a clinical trial of or false negative results in clinical trials (Leonard et al., 2020). We
PRV-PD101 (PROPEL, NCT04127578) (Table 3). The preclinical data for therefore recommend that prior to randomization, these genetic data
this gene therapy, an in vitro study using rodent neuronal cell lines and are collected and considered in the randomization to trial arms. At
an in vivo study using rodent models, demonstrated increased GCase minimum, and much less preferable, post-hoc analyses using these
activity, decreased glycolipid substrate accumulation and improved data should be performed.
motor symptoms. The safety of the drug was also examined by injection 4) Combination of drugs – It is possible that to fully address the effects
to non-human primates (Sheehan et al., 2020). In the PROPEL study, the of specific mutations, combinations of drugs such as chaperone +
adenoviral vector (adeno-associated virus 9) containing GBA (PR001A) activator, will be required. One potential trial design for this com­
is administrated intracisternally. The PROPEL study design is a multi­ bination could include four arms: chaperone only, activator only,
center, randomized, double-blind, escalating dose controlled sham chaperone + activator and placebo. Such trial will be cost-effective
procedure. The study is conducted in patients with GBA-PD bearing and could answer multiple questions in a single trial.
pathogenic GBA mutations, although the definition of pathogenicity is 5) Overshooting the desired therapeutic effect – Since the lysosomal
not detailed in the protocol. In a recent company report, positive effect sphingolipid metabolism pathway is crucial for cell maintenance, it
of PR001 administration in two patients with GBA variants (Type 2 GD is possible that increasing GCase activity to levels that are too high
patient and GD patient with GBA-PD) was described. According to the may lead to undesired results. This may occur in gene therapy
report, both patients demonstrated increased level of GCase in CSF from approach, in GCase activators which may not only activate the
undetectable to normal, four and three months after the treatment mutated GCase, but also the wildtype, and in other enzymatic
respectively. The GBA-PD patient developed severe adverse event modulators.
related to immune-mediated response to the AAV9 viral vector three
months after the treatment. This led to the protocol amendment to One of the main challenges for the researchers designing trials will be
modify the immunosuppressive treatment (globenewswire.com, 2021). the identification and recruitment of GBA-PD patients. While GBA var­
iants are common in PD, they progress faster, which may be both an
advantage and a limitation. The main advantage is that in a disease that
progresses faster, it would be easier to see an effect in a shorter time

5
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

frame or with a smaller trial. The main limitation is that when designing vary. While the autopsies of the majority of p.G2019S carriers (65%)
a trial focused on PD patients at early stages and/or drug-naïve PD, in­ showed Lewy bodies (LB) containing neurons, most carriers of other
dividuals with GBA-PD will not likely stay at these stages for a long time, LRRK2 variants (70%) did not have that PD hallmark pathology (Kalia
and will therefore be more difficult to recruit. The high frequency and et al., 2015). This may suggest that LRRK2-PD represents a distinct
faster progression to dementia may also hamper recruitment, if these are neuropathological condition, as PD is pathologically characterized by
the exclusion criteria. GBA genotyping is a potential obstacle on its own, α-synuclein accumulation.
as the presence of the pseudogene GBAP1 makes the sequencing of GBA
challenging. Methods that properly distinguish between the gene and 3.2. LRRK2-PD clinical features
the pseudogene are therefore necessary for correct identification of all
GBA-PD patients. All these challenges should be carefully considered The clinical features of LRRK2-PD are often reported as indistin­
when designing a GBA-targeting trial. To facilitate these trials, we guishable from idiopathic PD (Obergasteiger et al., 2020; Zhao and
recommend that clinicians routinely screen for GBA variants in all their Dzamko, 2019), but accumulating evidence suggest that LRRK2-PD
patients, whether through research initiatives or through clinical ini­ patients have a milder disease phenotype. A recent longitudinal
tiatives such as PD GENEration (parkinson.org/PDGENEration) and assessment of pooled prospective data compared 155 LRRK2-PD patients
others. Since GBA mutations are common in RBD patients (Krohn et al., with 889 sporadic PD patients (Ortega et al., 2021). This study reported
2020b), and since they may be associated with faster conversion, tar­ possible slower rate of cognitive decline based on MoCA scores and
geting RBD patients with GBA mutations may be an attractive oppor­ slower motor disease progression based on MDS-UPDRS III scores, albeit
tunity, as they are at a much earlier stage of the disease. the results were not statistically significant (Ortega et al., 2021).
Another study used the longitudinal data from PPMI for 158 LRRK2-PD
3. Therapy targeting LRRK2 and 598 sporadic PD patients and reported 60% lower rate of non-motor
symptoms progression in carriers of LRRK2 mutations (Ahamadi et al.,
The importance of LRRK2 in PD emerged from linkage studies in 2021). Furthermore, LRRK2-PD was linked to lower likelihood of
families with autosomal-dominant, late-onset PD (Paisán-Ruı́;z et al., non-motor symptoms like olfactory disturbance (Marras et al., 2011;
2004; Zimprich et al., 2004). Seven mutations have been reported to Saunders-Pullman et al., 2011), depression (Piredda et al., 2020) and
cause PD: p.N1437H, p.R1441C/G/H, p.Y1699C, p.G2019S and p. RBD (Bencheikh et al., 2018; Saunders-Pullman et al., 2015). It is
I2020T (Ross et al., 2011), while additional variants such as p.R1628P interesting to note that unlike sporadic PD, LRRK2-PD seems to have no
(Zhang et al., 2017), p.G2385R (Xie et al., 2014) and p.M1646T (Sosero sex effect and prevalence in women is roughly similar to men (Gan-Or
et al., 2021) may increase the risk of PD. The most common pathogenic et al., 2015d; Kestenbaum and Alcalay, 2017). Also, homozygous car­
LRRK2 variant p.G2019S was reported in ~4% of familial PD cases and riers of p.G2019S were reported to be comparable to heterozygous
~1% in sporadic PD (Lesage et al., 2007), but it has a much lower or carriers when it came to AAO, cognitive performance, depression status
higher prevalence in some ethnicities. Due to founder effects, in and other non-motor symptoms (Kestenbaum and Alcalay, 2017).
Ashkenazi Jews and North African Arabs p.G2019S can be found in As mentioned before, LRRK2-PD patients have less abundant α-syn­
10–15% and ~39% respectively in sporadic PD patients and up to 26% uclein pathology (Kalia et al., 2015). GBA-PD and PD associated with
and 40% respectively in familial PD cases (Hassin-Baer et al., 2009; van SCNA multiplications are characterized by abundant α-synuclein pa­
Dijkman et al., 2018). The penetrance of LRRK2 variants is incomplete thology and more malign PD course (Cilia et al., 2016; Konno et al.,
and estimated to be from 25% to 43% in older adults (Lee et al., 2017). 2016; Schneider and Alcalay, 2017). It is therefore possible that the
lower rates of α-synuclein pathology in LRRK2-PD may be the reason (or
3.1. Mechanism and pathology of LRRK2-PD just an indicator) for the more benign clinical course observed in these
patients. It is also possible that other, α-synuclein independent mecha­
LRRK2 encodes a 2527 amino acid protein, leucine-rich repeat kinase nisms, are driving PD development in carriers of LRRK2 variants.
2, containing six independent domains. The enzymatic part of the pro­
tein consists of the Ras-like GTPase (ROC), C-terminal domain of ROC 3.3. LRRK2 inhibitors in development
(COR) and a kinase domain. Other domains, an N-terminal armadillo
repeat region (ARM), an ankyrin repeat (ANK) domain, a leucine-rich 3.3.1. Kinase inhibitors
repeat (LRR) domain and a C-terminal WD40 domain, play a role in The gain-of-function effect of LRRK2 mutations on the kinase activity
protein-protein binding (Bae and Lee, 2015). All the clearly pathogenic became an appealing target for therapeutic development because it is
LRRK2 variants are located in the catalytic domains and may induce usually easier to inhibit rather than to activate a protein. Additionally,
hyperactivation of the kinase domain (Bryant et al., 2021). Rab10 various kinase inhibitors have already been developed for use in
GTPase is one of the key substrates phosphorylated by LRRK2 and may oncology (Kannaiyan and Mahadevan, 2018). Therefore, the approach
be used as a biomarker of the kinase activity (Rideout et al., 2020). of kinase inhibition is not new, although it needs to be further improved
Studies using p.R1441C knock-in mutation in mice showed hyper­ in order to increase selectivity and penetrance of blood-brain barrier.
phosphorylation of Rab10, which was suppressed by LRRK2 inhibition There are several non-selective LRRK2 kinase inhibitor compounds
(Dhekne et al., 2018). This suggests that Rab10 is a physiological sub­ that have been so far only tested in preclinical models of PD, and which
strate of LRRK2 and that pathogenic variants do lead to hyper­ were recently discussed in detail, covering their efficacy and safety as­
phosphorylation in these models. Rab GTPase family regulates many pects (Wojewska and Kortholt, 2021). The safety concerns emerge from
important aspects of cell biology through their role in intracellular the fact that LRRK2 is expressed in lung and kidneys where LRRK2 in­
trafficking (Pfeffer, 2018). LRRK2 phosphorylates 14 different Rab hibitors might have detrimental effects (Baptista et al., 2013; Herzig
proteins (out of 50 that were tested), which are possibly physiological et al., 2011; Tong et al., 2012). These concerns were supported by the
substrates of LRRK2 (Steger et al., 2017). Various cellular pathways studies in rodents that showed that LRRK2 knockout animals or those
involving these Rab GTPases were proposed as potential mechanisms treated with LRRK2-inhibitors display pathological changes in kidneys
underlying PD pathology (thoroughly reviewed in (Bae and Lee, 2020)): or lungs (Andersen et al., 2018; Baptista et al., 2013; Hinkle et al., 2012).
disrupted ciliogenesis, reduced neuroprotective signaling, impaired However, the analysis of 1455 carriers of LRRK2 loss-of-function vari­
maintenance of lysosomal and mitochondrial homeostasis, disrupted ants showed that there is no significant association between suppressed
vesicle trafficking, promotion of α-synuclein propagation, inflammatory levels of LRRK2 and kidney or lung pathology (Whiffin et al., 2020).
response – just to name a few. Another study in nonhuman primates reported that the LRRK2 inhibitors
Pathological findings in patients with different LRRK2 variants may induced only minor changes in lung tissue which did not impair lung

6
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

function (Baptista et al., 2020). However, the short duration of the study 3.3.2. Antisense oligonucleotides
(2 weeks) raises questions on the generalizability of the safety conclu­ Antisense oligonucleotides (ASOs) are single-stranded DNA mole­
sions and their meaning for human patients who would be taking the cules that are designed to specifically bind to their target mRNA via base
drug for extended periods of time. Preclinical results regarding the pairing and inhibit or alter gene expression by causing steric hindrance,
safety effects associated with lungs and kidneys LRRK2 inhibition are inducing alternative splicing or triggering target degradation (Chery,
conflicting and may not be relevant for humans. Of note, the two 2016). ASOs are an alternative approach for inhibiting LRRK2 activity
non-selective LRRK2 kinase inhibitor compounds that reached clinical by decreasing the levels of translated protein or inducing splicing al­
trials (discussed later), did so without reporting preclinical studies. terations which will affect the catalytic core of the protein. Preclinical
Targeted inhibition of specific LRRK2 variants (i.e., p.G2019S) in studies of ASOs targeting LRRK2 reported some promising results
heterozygous patients might be a safer choice to avoid the potential side (Table 4). Decreased loss of dopaminergic neurons and improved grip
effects of systemic inhibition discussed earlier. A few preclinical studies strength was reported in murine models, along with decreased α-synu­
suggested efficacy of compounds EB-42486 and EB-42168 as potent clein aggregation in primary neurons of mice with and without the p.
inhibitors of LRRK2 p.G2019S (Bright et al., 2021; Garofalo et al., 2020). G2019S variant after intracerebral ventricular injections of ASOs (Zhao
However, EB-42486 was not able to cross the blood-brain-barrier and et al., 2017). It was noted that the treatment did not affect the gene
the ability of EB-42168 to cross it was not tested. Overall, targeted in­ expression in kidneys and lungs. Whether or not these findings are
hibition of specific LRRK2 variants is a viable approach for precision relevant in humans needs to be examined. Some in vitro studies also
medicine, although we still do not have enough preclinical data to start reported a protective effect of ASOs that induce skipping of exon 41,
clinical trials pursuing this target. containing the p.G2019S variant. These protective effects include
As of today, there are two LRRK2 kinase inhibitor compounds that endoplasmic reticulum Ca2+ homeostasis, which was normalized in
reached clinical trials, DNL151 and DNL201 (Table 4). In May 2021, it neurons with p.G2019S variant derived from induced pluripotent stem
was announced that DNL151 was selected to be advanced to late-stage cells (Korecka et al., 2019a) and mitophagy rate, presumably altered by
studies, testing it further in PD patients with LRRK2 gain-of-function p.G2019S, which was restored in PD-patient-derived fibroblasts car­
mutations and in patients with sporadic PD (denalitherapeutics.com, rying the p.G2019S variant (Korecka et al., 2019b). Additionally, there
2021). The DNL201 compound will be kept as a backup. DNL151 and was a decrease of Rab10 phosphorylation and normalization of auto­
DNL201 are orally available small molecules that can penetrate the phagosome maturation after the transgenic mice expressing either
blood-brain barrier. Both were tested in phase 1/1b randomized, human wildtype LRRK2 or p.G2019S were treated with ASO blocking
double-blind, placebo-controlled trials, as described below. The pre­ the 5’ splice site of exon 41 (Korecka et al., 2020). There is currently a
clinical data for DNL151 or DNL201 was not published by the company, Phase 1 randomized placebo-controlled clinical trial aiming to evaluate
but according to the reports from phases 1/1b of the clinical trial, the the safety and tolerability of LRRK2 ASO BIIB094 in 82 PD patients with
drug was well tolerated and considered safe. Moreover, DNL151 and without genetic variants (NCT03976349). The BIIB094 molecule,
demonstrated a favorable safety and tolerability profile, robust target delivered to the CNS via an intrathecal injection, is thought to lower
engagement, as well as significant change in biomarkers that indicate LRRK2 protein levels by binding mRNA and mediating its degradation
lysosomal function. Reduction of phosphorylated (pS935) LRRK2 and (ionispharma.com).
pT73 Rab10 levels imply possible normalization of kinase activity
(denalitherapeutics.com, 2021). DNL151 was tested in 36 PD patients, 3.3.3. GTPase modulators
with either LRRK2-PD or sporadic PD (NCT04056689) and in 184 The kinase domain is not the only target from the catalytic core of
healthy volunteers (NCT04557800). DNL201 met the objectives of LRRK2, as the ROC GTPase domain have also been suggested to play a
safety, tolerability, CSF exposure levels, target engagement, kinase in­ role in PD pathogenesis. It was pointed out that all pathogenic variants
hibition and lysosomal biomarkers in 122 healthy volunteers in the ROC domain increase the proportion of GTP-bound LRRK2, while
(NCT04551534) and in 29 PD patients with and without LRRK2 muta­ the protective variant p.R1398H decreases it (Nixon-Abell et al., 2016).
tions (NCT03710707) (denalitherapeutics.com, 2018, 2020). It is not yet clear if the observed relationship is due to changes in affinity
of the protein for GTP, decreased GTPase activity or due to indirect

Table 4
Therapeutic approaches for LRRK2.
Mechanism Drug Registered clinical trial Cohort Phase Current status
name (clinicaltrials.gov) o
r reference

LRRK2 kinase inhibitor DNL151 NCT04557800 Healthy 1/1b Completed with positive outcome in terms of safety and target
NCT04056689 volunteers/PD engagement.
Selected to be advanced to late stage studies.
DNL201 NCT04551534 Healthy 1/1b Completed with positive outcome in terms of safety and target
NCT03710707 volunteers/PD engagement.
Will be kept as backup for DNL151.
PFE-360 Baptista et al. (2020) – Preclinical Reported ~50% reduction in LRRK2 activity in the nonhuman primate
brain with minor or reversable changes in lungs.
GNE- Baptista et al. (2020) – Preclinical Reported that low doses were sufficient to completely inhibit brain LRRK2
7915 activity in macaque and to avoid major impairment of lung function.
MLi-2 Baptista et al. (2020) – Preclinical
p.G2019S selective LRRK2 EB- Bright et al. (2021) – Preclinical Ex vivo study reported selective p.G2019S LRRK2 inhibition measuring
kinase inhibitor 42168 Ser935 and The73 Rab10 biomarkers.
Antisense oligonucleotide BIIB094 NCT03976349 PD 1 Recruiting PD patients with and without genetic variants.
(inhibit translation)
Antisense oligonucleotide ASO 41- Korecka et al. (2020) – Preclinical Reported to decrease Rab10 phosphorylation and normalize
(Induces skipping of exon 1 autophagosome maturation in transgenic mice; normalize mitophagy rate
41) and endoplasmic reticulum Ca2+ homeostasis in vitro.
GTPase modulator FX2149 Li et al. (2015) – Preclinical Reported to decrease LRRK2 autophosphorylation, reduce
neuroinflammation, and decrease neuronal degeneration in vitro and in
murine model. More potent and brain-penetrant than earlier analogues.

7
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

inhibition of LRRK2 kinase activity. In vitro and in vivo studies testing either larger trials or longer trials will be required to observe a drug
GTP-binding inhibitors reported some decrease in LRRK2 autophos­ effect, unless the effect of the drug will be very large. However, a large
phorylation, reduced neuroinflammation and suppression of effect cannot be assumed in advance, even if in preclinical models it
LRRK2-linked neuronal degeneration (Li et al., 2014, 2015). Addition­ seems this way. Therefore, trials for LRRK2 inhibitors should probably
ally, what makes GTP-binding inhibitors an interesting approach, is the be designed to be larger and longer. The reduced frequency and slower
potential of great selectivity due to the fact that LRRK2 is one of only progression to cognitive decline, make it a less favorable endpoint in
four ROC GTPases in humans (Tomkins et al., 2018). We expect that LRRK2-targeting trials, which will probably have to focus on motor
additional GTPase modulators will be developed and progress into symptoms and other biomarkers. Such biomarkers may be proven
clinical trials in the future. crucial for the development of a therapeutic LRRK2 inhibitor, if the
clinical signal will be relatively weak, but only if there is clear evidence
3.4. Can LRRK2 inhibitors be used for sporadic PD and GBA-PD? from human studies that changes in biomarkers are strongly correlated
with clinical changes. One of the questions that may arise is whether the
Some genetic and experimental data suggest that LRRK2 may also be current biomarkers such as LRRK2 autophosphorylation, Rabs phos­
relevant in sporadic PD. For example, LRRK2 is one of the top genes phorylation, urine BMP and others are reliable enough to move forward
implicated in PD genome wide association studies, even after removing to clinical trials (Rideout et al., 2020). The first trials on LRRK2 in­
the effects of the p.G2019S mutation and other potentially pathogenic hibitors may provide the data needed to answer this question.
variants (Lake et al., 2021). This association may be driven by regula­ Another challenge, similar to GBA but more acute, is the challenge of
tory, non-coding variants in LRRK2, potentially affecting its expression recruitment. Since LRRK2 mutations are less common, recruitment
or activity (Lake et al., 2021). Experimental data from different in vitro might prove difficult. Same as for GBA, we recommend that all clinicians
and in vivo models suggest that LRRK2 is involved in mechanisms related will genotype LRRK2 mutations in all their patients to make them trial-
to sporadic PD, such as inflammation, α-synuclein accumulation and ready. There are multiple research- or clinical-based initiatives aiming
others (Kluss et al., 2019). Therefore, it is possible that LRRK2-targeting to genotype LRRK2 in multiple cohorts worldwide. Having trial-ready
treatment will be relevant for some sporadic PD patients, who may be PD populations will be crucial for developing LRRK2 trials, especially
genetically and biologically similar to LRRK2-PD. Identifying these due to the size and trial duration issues mentioned above.
specific individuals, perhaps by a combination of genetics and bio­ Less is known about LRRK2 modifiers, as opposed to GBA modifiers.
markers such as LRRK2 activity, substrate phosphorylation and others, A family and population-based study suggested that DNM3 may be a
could be crucial for treating sporadic PD patients with LRRK2 inhibitors. modifier of LRRK2-PD (Trinh et al., 2016), yet it was not replicated in a
Recently, experiments in cell models suggested that LRRK2 in­ follow-up study (Brown et al., 2021). More recently, a GWAS aimed to
hibitors may also be beneficial in GBA-PD. In these experiments, LRRK2 identify LRRK2 modifiers nominated variants in the CORO1C region as
mutations led to reduced GCase activity, and treatment with LRRK2 potential LRRK2 modifiers (Lai et al., 2020). Just like in GBA-PD,
inhibitors improved GCase activity (Ysselstein et al., 2019). However, polygenic risk score of PD also affected LRRK2 penetrance (Iwaki et al.,
multiple lines of human data contradict these results and do not support 2020). Although the effects of these variants on LRRK2-PD progression is
LRRK2 inhibitors as a viable treatment for GBA-PD. First, in a cohort of unknown, we suggest that in clinical trials targeting LRRK2, it will be
PD patients with the LRRK2 p.G2019S mutation, plasma GCase activity useful to match the treatment and placebo groups for CORO1C variants
was increased rather than decreased (Alcalay et al., 2015a). In carriers of and polygenic risk scores.
the LRRK2 p.M1646T risk variants, plasma GCase was increased as well
(Sosero et al., 2021). Carriers of both GBA and LRRK2 mutations provide 4. Therapy targeting PRKN
additional compelling evidence that LRRK2 mutations are not likely to
decrease GCase activity. If they were, we would expect that individuals PRKN or Parkin is one of the autosomal recessive genes that was
with both GBA and LRRK2 mutations will have a more severe pheno­ identified through gene mapping in juvenile-onset PD patients (Kitada
type, as they supposedly carry two mutations that reduce GCase activity. et al., 1998). Homozygous and compound heterozygous mutations in
However, data from three independent cohorts suggest the opposite PRKN cause early-onset PD (EOPD) (Brüggemann and Klein, 1993). A
(Omer et al., 2020; Ortega et al., 2021; Yahalom et al., 2019). In all three systematic review carried out to estimate the prevalence of biallelic
cohorts (two cross-sectional and one longitudinal), PD patients with PRKN variants in EOPD showed 4.3% in sporadic EOPD cases, up to
both LRRK2 and GBA mutations had milder phenotypes, similar to those 15.5% in familial cases of EOPD (Kilarski et al., 2012).
who carry only LRRK2 mutations. Lastly, the major differences in pa­ While the role of biallelic PRKN mutations in PD is well established,
thology and clinical course (Kalia et al., 2015; Schneider and Alcalay, the association of rare heterozygous variants with the disease is
2017; Srivatsal et al., 2015), the fact that GBA mutations may also lead controversial, as different studies yielded contradicting results (Kay
to RBD or dementia with Lewy bodies while LRRK2 mutations do not et al., 2010; Lesage et al., 2008). A recent study tried to elucidate this
(Creese et al., 2018; Honeycutt et al., 2019; Krohn et al., 2020b; Nalls link by assessing the rare heterozygous single nucleotide variations
et al., 2013; Ouled Amar Bencheikh et al., 2018; Pont-Sunyer et al., (SNVs) and copy number variations (CNVs) in a relatively large cohorts
2015), together with the previous data, all support a separate mecha­ of PD patients (Yu et al., 2021). The study reported no associations of
nisms for GBA-PD and LRRK2-PD, and may even suggest that LRRK2 rare heterozygous PRKN variants with PD risk, nor with the AAO.
mutations have a protective effect in GBA-PD. If this is true, LRRK2 in­ Another recent study suggested that the association of monoallelic PRKN
hibition in GBA-PD may even worsen their disease. We therefore variants could be inflated by cryptic biallelic variants (Lubbe et al.,
recommend, with the currently available data, not to pursue LRRK2 2021). Therefore, we currently cannot consider heterozygous carriage of
inhibition as a therapeutic strategy for GBA-PD. PRKN mutations as a risk factor of PD, or such carriers as candidates for
Parkin-targeting trials.
3.5. Challenges, strategies and future directions in LRRK2-PD clinical
trials 4.1. Mechanism, pathology and clinical features of PRKN-PD

Perhaps the main challenge in the development of LRRK2-targeting The parkin protein, encoded by PRKN, is an E3 ubiquitin protein
treatment is related to the clinical course and rate of progression of ligase that, which together with a kinase PINK1 plays a crucial role in
LRRK2-PD. Several studies have suggested that LRRK2-PD progresses mitophagy – removal of dysfunctional mitochondria through auto­
slower, and have less cognitive decline than sporadic PD or GBA-PD phagy, hence supporting healthy mitochondrial function (Narendra and
(Ortega et al., 2021; Yahalom et al., 2019). Since it progresses slower, Youle, 2011). One of the main triggers for mitophagy is the

8
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

accumulation of mitochondrial reactive oxygen species (ROS) and fail­ Table 5


ure of various antioxidant enzymes to adequately respond to the Therapeutic approaches for Parkin.
oxidative stress (Schofield and Schafer, 2021). Hence, disruption of this Mechanism Drug Registered Cohort Phase Current
important self degradation mechanism could lead to cytotoxic levels of name clinical trial status
ROS detrimental to dopaminergic neurons (Fujita et al., 2014; Kolodkin (clinicalt
rials.gov) o
et al., 2020). It was also suggested that by regulating mitophagy, Par­
r reference
kin/PINK1 have a role in the innate immunity modulation, and dis­
ruptions can lead to loss of dopaminergic neurons due to Parkin WO- Miller and - Preclinical Data
activators; 2018/ Muqit limited
neuroinflammatory response (Sliter et al., 2018). autoinhibitory 023029 (2019) to in vitro
Neuropathological studies, albeit only a few, showed that PD pa­ mechanism WO- assays.
tients carrying biallelic PRKN variants have an atypical neuropathology, disruptors 2010/
with mostly absent α-synuclein accumulations and LB (Schneider and 011839
Selective MF-094 Kluge et al. - Preclinical Data
Alcalay, 2017), as well as localized neurodegeneration limited only to
inhibitors of MF-095 (2018) limited
the substantia nigra and locus coeruleus (Dawson and Dawson, 2010). In ubiquitin- to in vitro
addition to early onset of disease, other clinical features of PRKN carriers specific assays
clearly differ from sporadic PD. The disease progression is much slower protease 30
(Alcalay et al., 2014), cognition is relatively preserved in advanced
disease and dementia is absent in most patients (Alcalay et al., 2014;
targeting drugs will become much larger. However, the current evidence
Piredda et al., 2020), and olfaction is mostly preserved (Wang et al.,
are conflicting, and recent large-scale analyses suggest that the previous
2017). Milder disease features observed in PRKN carriers could be
reported association between heterozygous PRKN variants and PD may
related to mostly absent α-synuclein pathology and pathology confined
be due to cryptic second mutations that have not been identified (Lubbe
to specific brain regions.
et al., 2021). Another issue is that even if the association was true, the
reported odds ratio is small, similar to those of top SNPs in PD GWAS.
4.2. PRKN targeting drugs in development
This would mean that for many of the PRKN heterozygous carriers,
having PD is likely not related to their PRKN genetic status, and they
At this point, no drug that specifically and directly targeting PRKN is
might not benefit from Parkin-targeting drugs. Including these patients
in clinical trials. However, there are multiple small molecules that are
in trials may therefore dilute the signal and in a case of a drug that could
going through preclinical development, mostly aimed at activation of
work, lead to false negative results. We therefore recommend at this
mitophagy. One of the evaluated approaches is the direct activation of
stage to focus only on biallelic PRKN-PD patients for clinical trials,
Parkin by disrupting its autoinhibitory mechanisms. Two small mole­
despite the aforementioned limitations of such trials.
cules (WO-2018/023029 and WO-2010/011839) showed this effect in
vitro but in vivo evidence are yet to be published (Miller and Muqit,
5. Conclusions
2019). Another proposed drug target is the ubiquitin-specific protease
30 (USP30), which removes ubiquitin from substrates of Parkin
Genetic studies have provided new and important insights into the
(Table 5) (Komander et al., 2009). Inhibiting this enzyme could poten­
pathogenesis of PD, and now allow for stratifying patients based on their
tially compensate for reduced Parkin activity. Two compounds (MF-094
genetic background towards clinical trials targeting specific genetic
and MF-095) showed effective and selective inhibition of USP30 in vitro
variants. Despite some major limitations discussed in this review, the
(Kluge et al., 2018). Additionally, there are multiple studies aimed at
most promising targets thus far are GBA, LRRK2 and PRKN, for which
supporting mitochondrial function by targeting the electron transport
specific treatments are in different stages of clinical development. Many
chain or by providing antioxidative effects but since these mechanisms
challenges still exist, including recruitment, biomarkers identification,
are not specific to PRKN-PD, they will not be discussed in this review.
trial design and others. Nevertheless, through international collabora­
tions and data sharing, we can enable large and properly designed
4.3. Challenges, strategies and future directions
clinical trials for PD patients who carry mutations in these specific
genes, towards future development of efficient treatments.
Some of the challenges we will face when designing a clinical trial
targeting Parkin are similar to those of LRRK2, but amplified. As biallelic
Financial disclosures
PRKN mutations carriers with PD are much rarer, recruiting cohorts that
will be large enough for meaningful clinical trials will be difficult and
ZGO received consultancy fees from Lysosomal Therapeutics Inc.
likely require years of preparations in multiple sites. As PRKN-PD can be
(LTI), Idorsia, Prevail Therapeutics, Inceptions Sciences (now Ventus),
considered as an independent, rare disease, smaller trials might be
Ono Therapeutics, Denali, Handl Therapeutics, Neuron23, Bial Biotech,
performed, with the hope of a large enough effect size of the drug that
Guidepoint, Lighthouse and Deerfield. Other authors have nothing to
will be detected in a smaller group during the trial. The rate of pro­
disclose.
gression of PRKN-PD and the overall conserved cognitive function also
pose major hurdles, since it will be difficult to obtain a significant signal
Acknowledgement
in both motor and cognitive domains as outcomes of the trials. For this
reason, identifying specific and reliable biomarkers for PRKN-PD pro­
ZGO is supported by the Fonds de recherche du Québec - Santé
gression may be crucial for future trials. While the slow progression of
(FRQS) Chercheurs-boursiers award, in collaboration with Parkinson
PRKN-PD is generally a disadvantage towards trial development, it may
Quebec, and is a William Dawson Scholar. KS is supported by a post-
also be conceived as an advantage, as more patients might be eligible to
doctoral fellowship from the Canada First Research Excellence Fund
enroll to the trial, as many are still defined at early stages of the disease.
(CFREF), awarded to McGill University for the Healthy Brains for
The development of international collaborations and registries of
Healthy Lives initiative (HBHL). Fig. 1 was created with BioRender.com.
monogenic form of PD will advance our ability to perform clinical trials
of PRKN-PD even with the above limitations (Klein et al., 2018). How­
ever, these trials should still be designed to be longer than typical trials.
An important issue to solve is the role of heterozygous PRKN variants
as risk factors in PD. If they are, then the population eligible for Parkin-

9
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

References Bonifati, V., 2014. Genetics of Parkinson’s disease–state of the art, 2013. Park. Relat.
Disord. 1 (20 Suppl), S23–28.
Bright, J.M., Carlisle, H.J., Toda, A.M., Murphy, M., Molitor, T.P., Wren, P., Andruska, K.
Peterschmitt, M.J., Crawford, N.P.S., Gaemers, S.J.M., Ji, A.J., Sharma, J., Pham, T.T.,
M., Liu, E., Barlow, C., 2021. Differential inhibition of LRRK2 in Parkinson’s disease
2021. Pharmacokinetics, pharmacodynamics, safety, and tolerability of oral
patient blood by a G2019S selective LRRK2 inhibitor. Movement disorders.
venglustat in healthy volunteers. Clin Pharmacol Drug Dev 10, 86–98.
Brown, E.E., Blauwendraat, C., Trinh, J., Rizig, M., Nalls, M.A., Leveille, E., Ruskey, J.A.,
Aflaki, E., Borger, D.K., Moaven, N., Stubblefield, B.K., Rogers, S.A., Patnaik, S.,
Jonvik, H., Tan, M.M.X., Bandres-Ciga, S., Hassin-Baer, S., Brockmann, K.,
Schoenen, F.J., Westbroek, W., Zheng, W., Sullivan, P., Fujiwara, H., Sidhu, R.,
Infante, J., Tolosa, E., Ezquerra, M., Ben Romdhan, S., Benmahdjoub, M., Arezki, M.,
Khaliq, Z.M., Lopez, G.J., Goldstein, D.S., Ory, D.S., Marugan, J., Sidransky, E., 2016.
Mhiri, C., Hardy, J., Singleton, A.B., Alcalay, R.N., Gasser, T., Grosset, D.G.,
A new glucocerebrosidase chaperone reduces α-synuclein and glycolipid levels in
Williams, N.M., Pittman, A., Gan-Or, Z., Fernandez-Santiago, R., Brice, A., Lesage, S.,
iPSC-derived dopaminergic neurons from patients with gaucher disease and
Farrer, M., Wood, N., Morris, H.R., 2021. Analysis of DNM3 and VAMP4 as genetic
parkinsonism. J. Neurosci. 36, 7441–7452.
modifiers of LRRK2 Parkinson’s disease. Neurobiol. Aging 97, 148 e117-148.e124.
Ahamadi, M., Mehrotra, N., Hanan, N., Yee, K.L., Gheyas, F., Anton, J., Bani, M.,
Brüggemann, N., Klein, C., 1993. Parkin type of early-onset Parkinson disease. In:
Boroojerdi, B., Smit, H., Weidemann, J., 2021. A disease progression model to
Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Mirzaa, G.,
quantify the non-motor symptoms of Parkinson’s disease in participants with
Amemiya, A. (Eds.), GeneReviews(®). University of Washington, Seattle Copyright ©
leucine-rich repeat kinase 2 mutation. Clinical Pharmacology & Therapeutics.
1993-2021, University of Washington, Seattle. GeneReviews Is a Registered
Aharon-Peretz, J., Rosenbaum, H., Gershoni-Baruch, R., 2004. Mutations in the
Trademark of the University of Washington, Seattle. All Rights Reserved. Seattle
glucocerebrosidase gene and Parkinson’s disease in Ashkenazi Jews. N. Engl. J. Med.
(WA).
351, 1972–1977.
Bryant, N., Malpeli, N., Ziaee, J., Blauwendraat, C., Liu, Z., Consortium, A.P., West, A.B.,
Alcalay, R.N., Caccappolo, E., Mejia-Santana, H., Tang, M.X., Rosado, L., Ross, B.M.,
2021. Identification of LRRK2 missense variants in the accelerating medicines
Verbitsky, M., Kisselev, S., Louis, E.D., Comella, C., Colcher, A., Jennings, D.,
partnership Parkinson’s disease cohort. Hum. Mol. Genet. 30, 454–466.
Nance, M.A., Bressman, S.B., Scott, W.K., Tanner, C., Mickel, S., Andrews, H.,
Burbulla, L.F., Jeon, S., Zheng, J., Song, P., Silverman, R.B., Krainc, D., 2019.
Waters, C., Fahn, S., Cote, L., Frucht, S., Ford, B., Rezak, M., Novak, K., Friedman, J.
A modulator of wild-type glucocerebrosidase improves pathogenic phenotypes in
H., Pfeiffer, R., Marsh, L., Hiner, B., Siderowf, A., Ottman, R., Marder, K., Clark, L.N.,
dopaminergic neuronal models of Parkinson’s disease. Sci. Transl. Med. 11.
2010a. Frequency of known mutations in early-onset Parkinson disease: implication
Campbell, M.C., Myers, P.S., Weigand, A.J., Foster, E.R., Cairns, N.J., Jackson, J.J.,
for genetic counseling: the consortium on risk for early onset Parkinson disease
Lessov-Schlaggar, C.N., Perlmutter, J.S., 2020. Parkinson disease clinical subtypes:
study. Arch. Neurol. 67, 1116–1122.
key features & clinical milestones. Annals of Clinical and Translational Neurology 7,
Alcalay, R.N., Mejia-Santana, H., Tang, M.X., Rakitin, B., Rosado, L., Ross, B.,
1272–1283.
Verbitsky, M., Kisselev, S., Louis, E.D., Comella, C.L., Colcher, A., Jennings, D.,
Chahine, L.M., Qiang, J., Ashbridge, E., Minger, J., Yearout, D., Horn, S., Colcher, A.,
Nance, M.A., Bressman, S., Scott, W.K., Tanner, C., Mickel, S.F., Andrews, H.F.,
Hurtig, H.I., Lee, V.M., Van Deerlin, V.M., 2013. Clinical and biochemical differences
Waters, C.H., Fahn, S., Cote, L.J., Frucht, S.J., Ford, B., Rezak, M., Novak, K.,
in patients having Parkinson disease with vs without GBA mutations. JAMA
Friedman, J.H., Pfeiffer, R., Marsh, L., Hiner, B., Siderowf, A., Ottman, R., Clark, L.
Neurology 70, 852–858.
N., Marder, K.S., Caccappolo, E., 2010b. Self-report of cognitive impairment and
Chery, J., 2016. RNA therapeutics: RNAi and antisense mechanisms and clinical
mini-mental state examination performance in PRKN, LRRK2, and GBA carriers with
applications. Postdoc J. 4, 35–50.
early onset Parkinson’s disease. Journal of clinical and experimental
Chia, R., Sabir, M.S., Bandres-Ciga, S., Saez-Atienzar, S., Reynolds, R.H., Gustavsson, E.,
neuropsychology 32, 775–779.
Walton, R.L., Ahmed, S., Viollet, C., Ding, J., Makarious, M.B., Diez-Fairen, M.,
Alcalay, R.N., Caccappolo, E., Mejia-Santana, H., Tang, M., Rosado, L., Orbe Reilly, M.,
Portley, M.K., Shah, Z., Abramzon, Y., Hernandez, D.G., Blauwendraat, C., Stone, D.
Ruiz, D., Ross, B., Verbitsky, M., Kisselev, S., Louis, E., Comella, C., Colcher, A.,
J., Eicher, J., Parkkinen, L., Ansorge, O., Clark, L., Honig, L.S., Marder, K.,
Jennings, D., Nance, M., Bressman, S., Scott, W.K., Tanner, C., Mickel, S.,
Lemstra, A., St George-Hyslop, P., Londos, E., Morgan, K., Lashley, T., Warner, T.T.,
Andrews, H., Waters, C., Fahn, S., Cote, L., Frucht, S., Ford, B., Rezak, M., Novak, K.,
Jaunmuktane, Z., Galasko, D., Santana, I., Tienari, P.J., Myllykangas, L., Oinas, M.,
Friedman, J.H., Pfeiffer, R., Marsh, L., Hiner, B., Siderowf, A., Payami, H., Molho, E.,
Cairns, N.J., Morris, J.C., Halliday, G.M., Van Deerlin, V.M., Trojanowski, J.Q.,
Factor, S., Ottman, R., Clark, L.N., Marder, K., 2012. Cognitive performance of GBA
Grassano, M., Calvo, A., Mora, G., Canosa, A., Floris, G., Bohannan, R.C., Brett, F.,
mutation carriers with early-onset PD: the CORE-PD study. Neurology 78,
Gan-Or, Z., Geiger, J.T., Moore, A., May, P., Krüger, R., Goldstein, D.S., Lopez, G.,
1434–1440.
Tayebi, N., Sidransky, E., Sotis, A.R., Sukumar, G., Alba, C., Lott, N., Martinez, E.M.,
Alcalay, R.N., Caccappolo, E., Mejia-Santana, H., Tang, M.X., Rosado, L., Orbe Reilly, M.,
Tuck, M., Singh, J., Bacikova, D., Zhang, X., Hupalo, D.N., Adeleye, A., Wilkerson, M.
Ruiz, D., Louis, E.D., Comella, C.L., Nance, M.A., Bressman, S.B., Scott, W.K.,
D., Pollard, H.B., Norcliffe-Kaufmann, L., Palma, J.-A., Kaufmann, H., Shakkottai, V.
Tanner, C.M., Mickel, S.F., Waters, C.H., Fahn, S., Cote, L.J., Frucht, S.J., Ford, B.,
G., Perkins, M., Newell, K.L., Gasser, T., Schulte, C., Landi, F., Salvi, E., Cusi, D.,
Rezak, M., Novak, K.E., Friedman, J.H., Pfeiffer, R.F., Marsh, L., Hiner, B.,
Masliah, E., Kim, R.C., Caraway, C.A., Monuki, E.S., Brunetti, M., Dawson, T.M.,
Payami, H., Molho, E., Factor, S.A., Nutt, J.G., Serrano, C., Arroyo, M., Ottman, R.,
Rosenthal, L.S., Albert, M.S., Pletnikova, O., Troncoso, J.C., Flanagan, M.E., Mao, Q.,
Pauciulo, M.W., Nichols, W.C., Clark, L.N., Marder, K.S., 2014. Cognitive and motor
Bigio, E.H., Rodríguez-Rodríguez, E., Infante, J., Lage, C., González-Aramburu, I.,
function in long-duration PARKIN-associated Parkinson disease. JAMA Neurol 71,
Sanchez-Juan, P., Ghetti, B., Keith, J., Black, S.E., Masellis, M., Rogaeva, E.,
62–67.
Duyckaerts, C., Brice, A., Lesage, S., Xiromerisiou, G., Barrett, M.J., Tilley, B.S.,
Alcalay, R.N., Levy, O.A., Waters, C.C., Fahn, S., Ford, B., Kuo, S.H., Mazzoni, P.,
Gentleman, S., Logroscino, G., Serrano, G.E., Beach, T.G., McKeith, I.G., Thomas, A.
Pauciulo, M.W., Nichols, W.C., Gan-Or, Z., Rouleau, G.A., Chung, W.K., Wolf, P.,
J., Attems, J., Morris, C.M., Palmer, L., Love, S., Troakes, C., Al-Sarraj, S., Hodges, A.
Oliva, P., Keutzer, J., Marder, K., Zhang, X., 2015a. Glucocerebrosidase activity in
K., Aarsland, D., Klein, G., Kaiser, S.M., Woltjer, R., Pastor, P., Bekris, L.M.,
Parkinson’s disease with and without GBA mutations. Brain 138, 2648–2658.
Leverenz, J.B., Besser, L.M., Kuzma, A., Renton, A.E., Goate, A., Bennett, D.A.,
Alcalay, R.N., Mejia-Santana, H., Mirelman, A., Saunders-Pullman, R., Raymond, D.,
Scherzer, C.R., Morris, H.R., Ferrari, R., Albani, D., Pickering-Brown, S., Faber, K.,
Palmese, C., Caccappolo, E., Ozelius, L., Orr-Urtreger, A., Clark, L., Giladi, N.,
Kukull, W.A., Morenas-Rodriguez, E., Lleó, A., Fortea, J., Alcolea, D., Clarimon, J.,
Bressman, S., Marder, K., Consortium, L.A.J., 2015b. Neuropsychological
Nalls, M.A., Ferrucci, L., Resnick, S.M., Tanaka, T., Foroud, T.M., Graff-Radford, N.
performance in LRRK2 G2019S carriers with Parkinson’s disease. Park. Relat.
R., Wszolek, Z.K., Ferman, T., Boeve, B.F., Hardy, J.A., Topol, E.J., Torkamani, A.,
Disord. 21, 106–110.
Singleton, A.B., Ryten, M., Dickson, D.W., Chiò, A., Ross, O.A., Gibbs, J.R.,
Andersen, M.A., Wegener, K.M., Larsen, S., Badolo, L., Smith, G.P., Jeggo, R., Jensen, P.
Dalgard, C.L., Traynor, B.J., Scholz, S.W., The American Genome, C., 2021. Genome
H., Sotty, F., Christensen, K.V., Thougaard, A., 2018. PFE-360-induced LRRK2
sequencing analysis identifies new loci associated with Lewy body dementia and
inhibition induces reversible, non-adverse renal changes in rats. Toxicology 395,
provides insights into its genetic architecture. Nat. Genet.
15–22.
Cilia, R., Tunesi, S., Marotta, G., Cereda, E., Siri, C., Tesei, S., Zecchinelli, A.L.,
Bae, J.R., Lee, B.D., 2015. Function and dysfunction of leucine-rich repeat kinase 2
Canesi, M., Mariani, C.B., Meucci, N., Sacilotto, G., Zini, M., Barichella, M.,
(LRRK2): Parkinson’s disease and beyond. BMB reports 48, 243.
Magnani, C., Duga, S., Asselta, R., Solda, G., Seresini, A., Seia, M., Pezzoli, G.,
Bae, E.-J., Lee, S.-J., 2020. The LRRK2-RAB axis in regulation of vesicle trafficking and
Goldwurm, S., 2016. Survival and dementia in GBA-associated Parkinson’s disease:
α-synuclein propagation. Biochim. Biophys. Acta (BBA) - Mol. Basis Dis. 1866,
the mutation matters. Ann. Neurol. 80, 662–673.
165632.
Clark, L.N., Afridi, S., Karlins, E., Wang, Y., Mejia-Santana, H., Harris, J., Louis, E.D.,
Baptista, M.A., Dave, K.D., Frasier, M.A., Sherer, T.B., Greeley, M., Beck, M.J., Varsho, J.
Cote, L.J., Andrews, H., Fahn, S., Waters, C., Ford, B., Frucht, S., Ottman, R.,
S., Parker, G.A., Moore, C., Churchill, M.J., 2013. Loss of leucine-rich repeat kinase 2
Marder, K., 2006. Case-control study of the parkin gene in early-onset Parkinson
(LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS
disease. Arch. Neurol. 63, 548–552.
One 8, e80705.
Creese, B., Bell, E., Johar, I., Francis, P., Ballard, C., Aarsland, D., 2018.
Baptista, M.A., Merchant, K., Barrett, T., Bhargava, S., Bryce, D.K., Ellis, J.M., Estrada, A.
Glucocerebrosidase mutations and neuropsychiatric phenotypes in Parkinson’s
A., Fell, M.J., Fiske, B.K., Fuji, R.N., 2020. LRRK2 inhibitors induce reversible
disease and Lewy body dementias: review and meta-analyses. Am J Med Genet B
changes in nonhuman primate lungs without measurable pulmonary deficits. Sci.
Neuropsychiatr Genet 177, 232–241.
Transl. Med. 12.
Dawson, T.M., Dawson, V.L., 2010. The role of parkin in familial and sporadic
Bencheikh, B.O.A., Ruskey, J.A., Arnulf, I., Dauvilliers, Y., Monaca, C.C., De Cock, V.C.,
Parkinson’s disease. Mov. Disord. 25, S32–S39.
Gagnon, J.-F., Spiegelman, D., Hu, M.T., Högl, B., 2018. LRRK2 protective haplotype
Dekker, N., van Dussen, L., Hollak, C.E., Overkleeft, H., Scheij, S., Ghauharali, K., van
and full sequencing study in REM sleep behavior disorder. Park. Relat. Disord. 52,
Breemen, M.J., Ferraz, M.J., Groener, J.E., Maas, M., Wijburg, F.A., Speijer, D.,
98–101.
Tylki-Szymanska, A., Mistry, P.K., Boot, R.G., Aerts, J.M., 2011. Elevated plasma
Berge-Seidl, V., Pihlstrøm, L., Maple-Grødem, J., Forsgren, L., Linder, J., Larsen, J.P.,
glucosylsphingosine in Gaucher disease: relation to phenotype, storage cell markers,
Tysnes, O.-B., Toft, M., 2017. The GBA variant E326K is associated with Parkinson’s
and therapeutic response. Blood 118, e118–127.
disease and explains a genome-wide association signal. Neurosci. Lett. 658, 48–52.
den Heijer, J. M., Kruithof, A. C., van Amerongen, G., de Kam, M. L., Thijssen, E.,
Beutler, E., 2006. Gaucher disease: multiple lessons from a single gene disorder. Acta
Grievink, H. W., Moerland, M., Walker, M., Been, K., Skerlj, R., Justman, C.,
Paediatr. Suppl. 95, 103–109.

10
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

Dudgeon, L., Lansbury, P., Cullen, V. C., Hilt, D. C., Groeneveld, G. J., A randomized Guedes, L.C., Chan, R.B., Gomes, M.A., Conceicao, V.A., Machado, R.B., Soares, T.,
single and multiple ascending dose study in healthy volunteers of LTI-291, a Xu, Y., Gaspar, P., Carriço, J.A., Alcalay, R.N., 2017. Serum lipid alterations in GBA-
centrally penetrant glucocerebrosidase activator. British Journal of Clinical associated Parkinson’s disease. Park. Relat. Disord. 44, 58–65.
Pharmacology n/a. Gündner, A.L., Duran-Pacheco, G., Zimmermann, S., Ruf, I., Moors, T., Baumann, K.,
denalitherapeuticscom, 2018. https://www.denalitherapeutics.com/investors/press-rele Jagasia, R., van de Berg, W.D.J., Kremer, T., 2019. Path mediation analysis reveals
ase?id=6741. GBA impacts Lewy body disease status by increasing α-synuclein levels. Neurobiol.
denalitherapeuticscom, 2020. https://www.denalitherapeutics.com/investors/press-rele Dis. 121, 205–213.
ase?id=7361. Han, T.-U., Sam, R., Sidransky, E., 2020. Small molecule chaperones for the treatment of
denalitherapeuticscom, 2021. https://www.denalitherapeutics.com/investors/press-rele gaucher disease and GBA1-associated Parkinson disease. Frontiers in Cell and
ase?id=8141&type=api. Developmental Biology 8.
Dhekne, H.S., Yanatori, I., Gomez, R.C., Tonelli, F., Diez, F., Schule, B., Steger, M., Hassin-Baer, S., Laitman, Y., Azizi, E., Molchadski, I., Galore-Haskel, G., Barak, F.,
Alessi, D.R., Pfeffer, S.R., 2018. A pathway for Parkinson’s Disease LRRK2 kinase to Cohen, O.S., Friedman, E., 2009. The leucine rich repeat kinase 2 (LRRK2) G2019S
block primary cilia and Sonic hedgehog signaling in the brain. Elife 7. substitution mutation. Association with Parkinson disease, malignant melanoma and
Do, J., McKinney, C., Sharma, P., Sidransky, E., 2019. Glucocerebrosidase and its prevalence in ethnic groups in Israel. J. Neurol. 256, 483–487.
relevance to Parkinson disease. Mol. Neurodegener. 14, 36. Healy, D.G., Falchi, M., O’Sullivan, S.S., Bonifati, V., Durr, A., Bressman, S., Brice, A.,
Espay, A.J., Kalia, L.V., Gan-Or, Z., Williams-Gray, C.H., Bedard, P.L., Rowe, S.M., Aasly, J., Zabetian, C.P., Goldwurm, S., Ferreira, J.J., Tolosa, E., Kay, D.M., Klein, C.,
Morgante, F., Fasano, A., Stecher, B., Kauffman, M.A., Farrer, M.J., Coffey, C.S., Williams, D.R., Marras, C., Lang, A.E., Wszolek, Z.K., Berciano, J., Schapira, A.H.,
Schwarzschild, M.A., Sherer, T., Postuma, R.B., Strafella, A.P., Singleton, A.B., Lynch, T., Bhatia, K.P., Gasser, T., Lees, A.J., Wood, N.W., 2008. Phenotype,
Barker, R.A., Kieburtz, K., Olanow, C.W., Lozano, A., Kordower, J.H., Cedarbaum, J. genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s
M., Brundin, P., Standaert, D.G., Lang, A.E., 2020. Disease modification and disease: a case-control study. Lancet Neurol. 7, 583–590.
biomarker development in Parkinson disease: revision or reconstruction? Neurology Herzig, M.C., Kolly, C., Persohn, E., Theil, D., Schweizer, T., Hafner, T., Stemmelen, C.,
94, 481–494. Troxler, T.J., Schmid, P., Danner, S., 2011. LRRK2 protein levels are determined by
Fereshtehnejad, S.M., Zeighami, Y., Dagher, A., Postuma, R.B., 2017. Clinical criteria for kinase function and are crucial for kidney and lung homeostasis in mice. Hum. Mol.
subtyping Parkinson’s disease: biomarkers and longitudinal progression. Brain 140, Genet. 20, 4209–4223.
1959–1976. Hinkle, K.M., Yue, M., Behrouz, B., Dächsel, J.C., Lincoln, S.J., Bowles, E.E., Beevers, J.
Fernandes, H.J., Hartfield, E.M., Christian, H.C., Emmanoulidou, E., Zheng, Y., Booth, H., E., Dugger, B., Winner, B., Prots, I., 2012. LRRK2 knockout mice have an intact
Bogetofte, H., Lang, C., Ryan, B.J., Sardi, S.P., Badger, J., Vowles, J., Evetts, S., dopaminergic system but display alterations in exploratory and motor co-ordination
Tofaris, G.K., Vekrellis, K., Talbot, K., Hu, M.T., James, W., Cowley, S.A., Wade- behaviors. Mol. Neurodegener. 7, 1–17.
Martins, R., 2016. ER stress and autophagic perturbations lead to elevated Ho, M., Seck, J., Schmidt, D., Veath, M., Johnson, W., Brady, R., O’Brien, J., 1972. Adult
extracellular α-synuclein in GBA-N370S Parkinson’s iPSC-derived dopamine Gaucher’s disease: kindred studies and demonstration of a deficiency of acid beta-
neurons. Stem Cell Reports 6, 342–356. glucosidase in cultured fibroblasts. Am. J. Hum. Genet. 24, 37.
Ficicioglu, C., 2008. Review of miglustat for clinical management in Gaucher disease Honeycutt, L., Montplaisir, J.Y., Gagnon, J.F., Ruskey, J., Pelletier, A., Gan-Or, Z.,
type 1. Ther Clin Risk Manag 4, 425–431. Postuma, R.B., 2019. Glucocerebrosidase mutations and phenoconversion of REM
Fischer, T., Gasser, T., Isaacson, S., Kulisevsky, J., Mir, P., Simuni, T., Wills, A.M., sleep behavior disorder to parkinsonism and dementia. Park. Relat. Disord. 65,
Guedes, L.C., Svenningsson, P., Waters, C., Ji, A., Li, J., Minini, P., Nembo, B., 230–233.
Sardi, S.P., Saubadu, S., Sharma, J., Peterschmitt, M.J., 2019. Safety, tolerability and Huebecker, M., Moloney, E.B., van der Spoel, A.C., Priestman, D.A., Isacson, O.,
pharmacokinetics of oral venglustat in Parkinson’s disease patients with a GBA Hallett, P.J., Platt, F.M., 2019. Reduced sphingolipid hydrolase activities, substrate
mutation (S4.002). Neurology 92. S4.002. accumulation and ganglioside decline in Parkinson’s disease. Mol. Neurodegener.
Fog, C.K., Zago, P., Malini, E., Solanko, L.M., Peruzzo, P., Bornaes, C., Magnoni, R., 14, 40.
Mehmedbasic, A., Petersen, N.H.T., Bembi, B., Aerts, J., Dardis, A., Kirkegaard, T., ionispharmacom. https://www.ionispharma.com/medicines/ionis-biib7/.
2018. The heat shock protein amplifier arimoclomol improves refolding, maturation Ivanova, M.M., Changsila, E., Turgut, A., Goker-Alpan, O., 2018. Individualized
and lysosomal activity of glucocerebrosidase. EBioMedicine 38, 142–153. screening for chaperone activity in Gaucher disease using multiple patient derived
Fujita, K.A., Ostaszewski, M., Matsuoka, Y., Ghosh, S., Glaab, E., Trefois, C., Crespo, I., primary cell lines. Am. J. Tourism Res. 10, 3750.
Perumal, T.M., Jurkowski, W., Antony, P.M., 2014. Integrating pathways of Ivanova, M., Dao, J., Kasaci, N., Adewale, B., Nazari, S., Noll, L., Fikry, J., Sanati, A.H.,
Parkinson’s disease in a molecular interaction map. Mol. Neurobiol. 49, 88–102. Goker-Alpan, O., 2021. Cellular and biochemical response to chaperone versus
Galper, J., Balwani, M., Fahn, S., Waters, C., Krohn, L., Gan-Or, Z., Dzamko, N., Alcalay, substrate reduction therapies in neuropathic Gaucher disease. bioRxiv. https://doi.
R. N., Cytokines and Gaucher Biomarkers In glucocerebrosidase carriers with and org/10.1101/2021.02.04.429713.
without Parkinson disease. Movement disorders n/a. Iwaki, H., Blauwendraat, C., Leonard, H.L., Kim, J.J., Liu, G., Maple-Grodem, J.,
Gan-Or, Z., Giladi, N., Rozovski, U., Shifrin, C., Rosner, S., Gurevich, T., Bar-Shira, A., Corvol, J.C., Pihlstrom, L., van Nimwegen, M., Hutten, S.J., Nguyen, K.H., Rick, J.,
Orr-Urtreger, A., 2008. Genotype-phenotype correlations between GBA mutations Eberly, S., Faghri, F., Auinger, P., Scott, K.M., Wijeyekoon, R., Van Deerlin, V.M.,
and Parkinson disease risk and onset. Neurology 70, 2277–2283. Hernandez, D.G., Gibbs, J.R., Chitrala, K.N., Day-Williams, A.G., Brice, A., Alves, G.,
Gan-Or, Z., Amshalom, I., Kilarski, L.L., Bar-Shira, A., Gana-Weisz, M., Mirelman, A., Noyce, A.J., Tysnes, O.B., Evans, J.R., Breen, D.P., Estrada, K., Wegel, C.E.,
Marder, K., Bressman, S., Giladi, N., Orr-Urtreger, A., 2015a. Differential effects of Danjou, F., Simon, D.K., Andreassen, O., Ravina, B., Toft, M., Heutink, P., Bloem, B.
severe vs mild GBA mutations on Parkinson disease. Neurology 84, 880–887. R., Weintraub, D., Barker, R.A., Williams-Gray, C.H., van de Warrenburg, B.P., Van
Gan-Or, Z., Dion, P.A., Rouleau, G.A., 2015b. Genetic perspective on the role of the Hilten, J.J., Scherzer, C.R., Singleton, A.B., Nalls, M.A., 2019. Genomewide
autophagy-lysosome pathway in Parkinson disease. Autophagy 11, 1443–1457. association study of Parkinson’s disease clinical biomarkers in 12 longitudinal
Gan-Or, Z., Girard, S.L., Noreau, A., Leblond, C.S., Gagnon, J.F., Arnulf, I., Mirarchi, C., patients’ cohorts. Mov. Disord. 34, 1839–1850.
Dauvilliers, Y., Desautels, A., Mitterling, T., Cochen De Cock, V., Frauscher, B., Iwaki, H., Blauwendraat, C., Makarious, M.B., Bandrés-Ciga, S., Leonard, H.L., Gibbs, J.
Monaca, C., Hogl, B., Dion, P.A., Postuma, R.B., Montplaisir, J.Y., Rouleau, G.A., R., Hernandez, D.G., Scholz, S.W., Faghri, F., Consortium, I.P.s.D.G., 2020.
2015c. Parkinson’s disease genetic loci in rapid eye movement sleep behavior Penetrance of Parkinson’s disease in LRRK2 p. G2019S carriers is modified by a
disorder. J. Mol. Neurosci. 56, 617–622. polygenic risk score. Mov. Disord. 35, 774–780.
Gan-Or, Z., Leblond, C.S., Mallett, V., Orr-Urtreger, A., Dion, P.A., Rouleau, G.A., 2015d. Jung, O., Patnaik, S., Marugan, J., Sidransky, E., Westbroek, W., 2016. Progress and
LRRK2 mutations in Parkinson disease; a sex effect or lack thereof? A meta-analysis. potential of non-inhibitory small molecule chaperones for the treatment of Gaucher
Park. Relat. Disord. 21, 778–782. disease and its implications for Parkinson disease. Expert Rev. Proteomics 13,
Garofalo, A.W., Bright, J., De Lombaert, S., Toda, A.M., Zobel, K., Andreotti, D., 471–479.
Beato, C., Bernardi, S., Budassi, F., Caberlotto, L., 2020. Selective inhibitors of Kalia, L.V., Lang, A.E., Hazrati, L.-N., Fujioka, S., Wszolek, Z.K., Dickson, D.W., Ross, O.
G2019S-LRRK2 kinase activity. J. Med. Chem. 63, 14821–14839. A., Van Deerlin, V.M., Trojanowski, J.Q., Hurtig, H.I., Alcalay, R.N., Marder, K.S.,
Gary, S.E., Ryan, E., Steward, A.M., Sidransky, E., 2018. Recent advances in the diagnosis Clark, L.N., Gaig, C., Tolosa, E., Ruiz-Martínez, J., Marti-Masso, J.F., Ferrer, I., López
and management of Gaucher disease. Expet Rev. Endocrinol. Metabol. 13, 107–118. de Munain, A., Goldman, S.M., Schüle, B., Langston, J.W., Aasly, J.O., Giordana, M.
Gault, C.R., Obeid, L.M., Hannun, Y.A., 2010. An overview of sphingolipid metabolism: T., Bonifati, V., Puschmann, A., Canesi, M., Pezzoli, G., Maues De Paula, A.,
from synthesis to breakdown. Adv. Exp. Med. Biol. 688, 1–23. Hasegawa, K., Duyckaerts, C., Brice, A., Stoessl, A.J., Marras, C., 2015. Clinical
Gegg, M.E., Burke, D., Heales, S.J., Cooper, J.M., Hardy, J., Wood, N.W., Schapira, A.H., correlations with Lewy body pathology in LRRK2-related Parkinson DiseaseLewy
2012. Glucocerebrosidase deficiency in substantia nigra of Parkinson disease brains. body pathology in LRRK2-related Parkinson DiseaseLewy body pathology in LRRK2-
Ann. Neurol. 72, 455–463. related Parkinson disease. JAMA Neurology 72, 100–105.
Gegg, M.E., Sweet, L., Wang, B.H., Shihabuddin, L.S., Sardi, S.P., Schapira, A.H., 2015. Kannaiyan, R., Mahadevan, D., 2018. A comprehensive review of protein kinase
No evidence for substrate accumulation in Parkinson brains with GBA mutations. inhibitors for cancer therapy. Expert Rev. Anticancer Ther. 18, 1249–1270.
Mov. Disord. 30, 1085–1089. Kay, D.M., Stevens, C.F., Hamza, T.H., Montimurro, J.S., Zabetian, C.P., Factor, S.A.,
Giraldo, P., Andrade-Campos, M., Alfonso, P., Irun, P., Atutxa, K., Acedo, A., Barez, A., Samii, A., Griffith, A., Roberts, J.W., Molho, E.S., Higgins, D.S., Gancher, S.,
Blanes, M., Diaz-Morant, V., Fernández-Galán, M.A., Franco, R., Gil-Cortes, C., Moses, L., Zareparsi, S., Poorkaj, P., Bird, T., Nutt, J., Schellenberg, G.D., Payami, H.,
Giner, V., Ibañez, A., Latre, P., Loyola, I., Luño, E., Hernández-Martin, R., Medrano- 2010. A comprehensive analysis of deletions, multiplications, and copy number
Engay, B., Puerta, J., Roig, I., de la Serna, J., Salamero, O., Villalón, L., Pocovi, M., variations in PARK2. Neurology 75, 1189–1194.
2018. Twelve years of experience with miglustat in the treatment of type 1 Gaucher Kestenbaum, M., Alcalay, R.N., 2017. Clinical features of LRRK2 carriers with
disease: the Spanish ZAGAL project. Blood Cell Mol. Dis. 68, 173–179. Parkinson’s disease. Leucine-Rich Repeat Kinase 2 (LRRK2), 31–48.
globenewswirecom, 2021. https://www.globenewswire.com/news-release/2020/0 Khanna, R., Benjamin, E.R., Pellegrino, L., Schilling, A., Rigat, B.A., Soska, R., Nafar, H.,
8/11/2076218/0/en/Prevail-Therapeutics-Reports-Second-Quarter-2020-Financial- Ranes, B.E., Feng, J., Lun, Y., Powe, A.C., Palling, D.J., Wustman, B.A.,
Results-and-Business-Highlights.html. Schiffmann, R., Mahuran, D.J., Lockhart, D.J., Valenzano, K.J., 2010. The

11
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

pharmacological chaperone isofagomine increases the activity of the Gaucher variability and potential effects on clinical trial outcomes: perspectives in
disease L444P mutant form of beta-glucosidase. FEBS J. 277, 1618–1638. Parkinson’s disease. J. Med. Genet. 57, 331–338.
Kilarski, L.L., Pearson, J.P., Newsway, V., Majounie, E., Knipe, M.D., Misbahuddin, A., Lerche, S., Schulte, C., Wurster, I., Machetanz, G., Roeben, B., Zimmermann, M.,
Chinnery, P.F., Burn, D.J., Clarke, C.E., Marion, M.H., Lewthwaite, A.J., Nicholl, D. Deuschle, C., Hauser, A.-K., Böhringer, J., Krägeloh-Mann, I., Waniek, K., Lachmann,
J., Wood, N.W., Morrison, K.E., Williams-Gray, C.H., Evans, J.R., Sawcer, S.J., I., Petterson, X.-M. T., Chiang, R., Park, H., Wang, B., Liepelt-Scarfone, I., Maetzler,
Barker, R.A., Wickremaratchi, M.M., Ben-Shlomo, Y., Williams, N.M., Morris, H.R., W., Galasko, D., Scherzer, C. R., Gasser, T., Mielke, M. M., Hutten, S. J.,
2012. Systematic review and UK-based study of PARK2 (parkin), PINK1, PARK7 (DJ- Mollenhauer, B., Sardi, S. P., Berg, D., Brockmann, K., The mutation matters: CSF
1) and LRRK2 in early-onset Parkinson’s disease. Mov. Disord. 27, 1522–1529. profiles of GCase, sphingolipids, α-synuclein in PDGBA. Mov. Disord. n/a.
Kitada, T., Asakawa, S., Hattori, N., Matsumine, H., Yamamura, Y., Minoshima, S., Lesage, S., Janin, S., Lohmann, E., Leutenegger, A.L., Leclere, L., Viallet, F., Pollak, P.,
Yokochi, M., Mizuno, Y., Shimizu, N., 1998. Mutations in the parkin gene cause Durif, F., Thobois, S., Layet, V., Vidailhet, M., Agid, Y., Dürr, A., Brice, A., Bonnet, A.
autosomal recessive juvenile parkinsonism. Nature 392, 605–608. M., Borg, M., Broussolle, E., Damier, P., Destée, A., Martinez, M., Penet, C.,
Klein, C., Westenberger, A., 2012. Genetics of Parkinson’s disease. Cold spring harb Rasco, O., Tison, F., Tranchan, C., Vérin, M., 2007. LRRK2 exon 41 mutations in
perspect med 2, a008888. sporadic Parkinson disease in Europeans. Arch. Neurol. 64, 425–430.
Klein, C., Hattori, N., Marras, C., 2018. MDSGene: closing data gaps in genotype- Lesage, S., Lohmann, E., Tison, F., Durif, F., Dürr, A., Brice, A., F.P.s.D.G. S. Group, 2008.
phenotype correlations of monogenic Parkinson’s disease. J. Parkinsons Dis. 8, Rare heterozygous parkin variants in French early-onset Parkinson disease patients
S25–S30. and controls. J. Med. Genet. 45, 43–46.
Kluge, A.F., Lagu, B.R., Maiti, P., Jaleel, M., Webb, M., Malhotra, J., Mallat, A., Li, T., Yang, D., Zhong, S., Thomas, J.M., Xue, F., Liu, J., Kong, L., Voulalas, P.,
Srinivas, P.A., Thompson, J.E., 2018. Novel highly selective inhibitors of ubiquitin Hassan, H.E., Park, J.-S., 2014. Novel LRRK2 GTP-binding inhibitors reduced
specific protease 30 (USP30) accelerate mitophagy. Bioorg. Med. Chem. Lett 28, degeneration in Parkinson’s disease cell and mouse models. Hum. Mol. Genet. 23,
2655–2659. 6212–6222.
Kluss, J.H., Mamais, A., Cookson, M.R., 2019. LRRK2 links genetic and sporadic Li, T., He, X., Thomas, J.M., Yang, D., Zhong, S., Xue, F., Smith, W.W., 2015. A novel
Parkinson’s disease. Biochem. Soc. Trans. 47, 651–661. GTP-binding inhibitor, FX2149, attenuates LRRK2 toxicity in Parkinson’s disease
Kolodkin, A.N., Sharma, R.P., Colangelo, A.M., Ignatenko, A., Martorana, F., Jennen, D., models. PLoS One 10, e0122461.
Briedé, J.J., Brady, N., Barberis, M., Mondeel, T.D., 2020. ROS networks: designs, Liu, G., Boot, B., Locascio, J.J., Jansen, I.E., Winder-Rhodes, S., Eberly, S., Elbaz, A.,
aging, Parkinson’s disease and precision therapies. NPJ systems biology and Brice, A., Ravina, B., van Hilten, J.J., Cormier-Dequaire, F., Corvol, J.C., Barker, R.
applications 6, 1–20. A., Heutink, P., Marinus, J., Williams-Gray, C.H., Scherzer, C.R., 2016. Specifically
Komander, D., Clague, M.J., Urbé, S., 2009. Breaking the chains: structure and function neuropathic Gaucher’s mutations accelerate cognitive decline in Parkinson’s. Ann.
of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 10, 550–563. Neurol. 80, 674–685.
Konno, T., Ross, O.A., Puschmann, A., Dickson, D.W., Wszolek, Z.K., 2016. Autosomal Luan, Z., Li, L., Higaki, K., Nanba, E., Suzuki, Y., Ohno, K., 2013. The chaperone activity
dominant Parkinson’s disease caused by SNCA duplications. Park. Relat. Disord. 1 and toxicity of ambroxol on Gaucher cells and normal mice. Brain Dev. 35, 317–322.
(22 Suppl), S1–6. Lubbe, S.J., Bustos, B.I., Hu, J., Krainc, D., Joseph, T., Hehir, J., Tan, M., Zhang, W.,
Kopytova, A.E., Rychkov, G.N., Nikolaev, M.A., Baydakova, G.V., Cheblokov, A.A., Escott-Price, V., Williams, N.M., Blauwendraat, C., Singleton, A.B., Morris, H.R., f.I.
Senkevich, K.A., Bogdanova, D.A., Bolshakova, O.I., Miliukhina, I.V., Bezrukikh, V. P.s.D.G. Consortium, 2021. Assessing the relationship between monoallelic PRKN
A., Salogub, G.N., Sarantseva, S.V., Usenko, T.C., Zakharova, E.Y., Emelyanov, A.K., mutations and Parkinson’s risk. Hum. Mol. Genet. 30, 78–86.
Pchelina, S.N., 2021. Ambroxol increases glucocerebrosidase (GCase) activity and Lwin, A., Orvisky, E., Goker-Alpan, O., LaMarca, M.E., Sidransky, E., 2004.
restores GCase translocation in primary patient-derived macrophages in Gaucher Glucocerebrosidase mutations in subjects with parkinsonism. Mol. Genet. Metabol.
disease and Parkinsonism. Park. Relat. Disord. 84, 112–121. 81, 70–73.
Korecka, J.A., Talbot, S., Osborn, T.M., de Leeuw, S.M., Levy, S.A., Ferrari, E.J., Lythe, V., Athauda, D., Foley, J., Mencacci, N.E., Jahanshahi, M., Cipolotti, L., Hyam, J.,
Moskites, A., Atkinson, E., Jodelka, F.M., Hinrich, A.J., Hastings, M.L., Woolf, C.J., Zrinzo, L., Hariz, M., Hardy, J., 2017. GBA-associated Parkinson’s disease:
Hallett, P.J., Isacson, O., 2019a. Neurite collapse and altered ER Ca(2+) control in progression in a deep brain stimulation cohort. J. Parkinsons Dis. 7, 635–644.
human Parkinson disease patient iPSC-derived neurons with LRRK2 G2019S Maegawa, G.H., Tropak, M.B., Buttner, J.D., Rigat, B.A., Fuller, M., Pandit, D., Tang, L.,
mutation. Stem Cell Reports 12, 29–41. Kornhaber, G.J., Hamuro, Y., Clarke, J.T., 2009. Identification and characterization
Korecka, J.A., Thomas, R., Christensen, D.P., Hinrich, A.J., Ferrari, E.J., Levy, S.A., of ambroxol as an enzyme enhancement agent for Gaucher disease. J. Biol. Chem.
Hastings, M.L., Hallett, P.J., Isacson, O., 2019b. Mitochondrial clearance and 284, 23502–23516.
maturation of autophagosomes are compromised in LRRK2 G2019S familial Mallett, V., Ross, J.P., Alcalay, R.N., Ambalavanan, A., Sidransky, E., Dion, P.A.,
Parkinson’s disease patient fibroblasts. Hum. Mol. Genet. 28, 3232–3243. Rouleau, G.A., Gan-Or, Z., 2016. GBA p. T369M substitution in Parkinson disease:
Korecka, J.A., Thomas, R., Hinrich, A.J., Moskites, A.M., Macbain, Z.K., Hallett, P.J., polymorphism or association? A meta-analysis. Neurology Genetics 2.
Isacson, O., Hastings, M.L., 2020. Splice-switching antisense oligonucleotides reduce Mangone, G., Bekadar, S., Cormier-Dequaire, F., Tahiri, K., Welaratne, A., Czernecki, V.,
LRRK2 kinase activity in human LRRK2 transgenic mice. Mol. Ther. Nucleic Acids Pineau, F., Karachi, C., Castrioto, A., Durif, F., 2020. Early cognitive decline after
21, 623–635. bilateral subthalamic deep brain stimulation in Parkinson’s disease patients with
Kornhaber, G.J., Tropak, M.B., Maegawa, G.H., Tuske, S.J., Coales, S.J., Mahuran, D.J., GBA mutations. Park. Relat. Disord. 76, 56–62.
Hamuro, Y., 2008. Isofagomine induced stabilization of glucocerebrosidase. Maple-Grødem, J., Dalen, I., Tysnes, O.B., Macleod, A.D., Forsgren, L., Counsell, C.E.,
Chembiochem 9, 2643–2649. Alves, G., 2021. Association of GBA genotype with motor and functional decline in
Krohn, L., Öztürk, T.N., Vanderperre, B., Ouled Amar Bencheikh, B., Ruskey, J.A., patients with newly diagnosed Parkinson disease. Neurology 96, e1036–e1044.
Laurent, S.B., Spiegelman, D., Postuma, R.B., Arnulf, I., Hu, M.T., 2020a. Genetic, Marras, C., Schüle, B., Munhoz, R.P., Rogaeva, E., Langston, J.W., Kasten, M.,
structural, and functional evidence link TMEM175 to synucleinopathies. Ann. Meaney, C., Klein, C., Wadia, P.M., Lim, S.Y., Chuang, R.S., Zadikof, C., Steeves, T.,
Neurol. 87, 139–153. Prakash, K.M., de Bie, R.M., Adeli, G., Thomsen, T., Johansen, K.K., Teive, H.A.,
Krohn, L., Ruskey, J.A., Rudakou, U., Leveille, E., Asayesh, F., Hu, M.T.M., Arnulf, I., Asante, A., Reginold, W., Lang, A.E., 2011. Phenotype in parkinsonian and
Dauvilliers, Y., Högl, B., Stefani, A., Monaca, C.C., Abril, B., Plazzi, G., Antelmi, E., nonparkinsonian LRRK2 G2019S mutation carriers. Neurology 77, 325–333.
Ferini-Strambi, L., Heidbreder, A., Boeve, B.F., Espay, A.J., De Cock, V.C., Massaro, G., Hughes, M.P., Whaler, S.M., Wallom, K.-L., Priestman, D.A., Platt, F.M.,
Mollenhauer, B., Sixel-Döring, F., Trenkwalder, C., Sonka, K., Kemlink, D., Waddington, S.N., Rahim, A.A., 2020. Systemic AAV9 gene therapy using the
Figorilli, M., Puligheddu, M., Dijkstra, F., Viaene, M., Oertel, W., Toffoli, M., Gigli, G. synapsin I promoter rescues a mouse model of neuronopathic Gaucher disease but
L., Valente, M., Gagnon, J.F., Desautels, A., Montplaisir, J.Y., Postuma, R.B., with limited cross-correction potential to astrocytes. Hum. Mol. Genet. 29,
Rouleau, G.A., Gan-Or, Z., 2020b. GBA variants in REM sleep behavior disorder: a 1933–1949.
multicenter study. Neurology 95, e1008–e1016. Mata, I.F., Samii, A., Schneer, S.H., Roberts, J.W., Griffith, A., Leis, B.C., Schellenberg, G.
Lai, D., Alipanahi, B., Fontanillas, P., Schwantes-An, T.-H., Aasly, J., Alcalay, R.N., D., Sidransky, E., Bird, T.D., Leverenz, J.B., Tsuang, D., Zabetian, C.P., 2008.
Beecham, G.W., Berg, D., Bressman, S., Brice, A., 2020. Genome-wide association Glucocerebrosidase gene mutations: a risk factor for Lewy body disorders. Arch.
studies of LRRK2 modifiers of Parkinson’s disease. medRxiv. Neurol. 65, 379–382.
Lake, J., Reed, X., Langston, R.G., Nalls, M.A., Gan-Or, Z., Cookson, M.R., Singleton, A.B., Mazzulli, J.R., Xu, Y.-H., Sun, Y., Knight, A.L., McLean, P.J., Caldwell, G.A.,
Blauwendraat, C., Leonard, H.L., 2021. <em>LRRK2</em> coding variants and the Sidransky, E., Grabowski, G.A., Krainc, D., 2011. Gaucher disease
risk of Parkinson’s disease. medRxiv. https://doi.org/10.1101/ glucocerebrosidase and α-synuclein form a bidirectional pathogenic loop in
2021.04.22.21255928. synucleinopathies. Cell 146, 37–52.
Lawton, M., Ben-Shlomo, Y., May, M.T., Baig, F., Barber, T.R., Klein, J.C., Swallow, D.M. Miliukhina, I.V., Usenko, T.S., Senkevich, K.A., Nikolaev, M.A., Timofeeva, A.A.,
A., Malek, N., Grosset, K.A., Bajaj, N., Barker, R.A., Williams, N., Burn, D.J., Agapova, E.A., Semenov, A.V., Lubimova, N.E., Totolyan, A.A., Pchelina, S.N., 2020.
Foltynie, T., Morris, H.R., Wood, N.W., Grosset, D.G., Hu, M.T.M., 2018. Developing Plasma cytokines profile in patients with Parkinson’s disease associated with
and validating Parkinson’s disease subtypes and their motor and cognitive mutations in GBA gene. Bull. Exp. Biol. Med. 168, 423–426.
progression. Journal of Neurology. Neurosurgery & Psychiatry 89, 1279–1287. Miller, S., Muqit, M.M., 2019. Therapeutic approaches to enhance PINK1/Parkin
Lee, A.J., Wang, Y., Alcalay, R.N., Mejia-Santana, H., Saunders-Pullman, R., mediated mitophagy for the treatment of Parkinson’s disease. Neurosci. Lett. 705,
Bressman, S., Corvol, J.C., Brice, A., Lesage, S., Mangone, G., Tolosa, E., Pont- 7–13.
Sunyer, C., Vilas, D., Schüle, B., Kausar, F., Foroud, T., Berg, D., Brockmann, K., Miller, J., McCLUER, R., Kanfer, J.N., 1973. Gaucher’s disease: neurologic disorder in
Goldwurm, S., Siri, C., Asselta, R., Ruiz-Martinez, J., Mondragón, E., Marras, C., adult siblings. Ann. Intern. Med. 78, 883–887.
Ghate, T., Giladi, N., Mirelman, A., Marder, K., 2017. Penetrance estimate of LRRK2 Mitsui, J., Matsukawa, T., Sasaki, H., Yabe, I., Matsushima, M., Durr, A., Brice, A.,
p.G2019S mutation in individuals of non-Ashkenazi Jewish ancestry. Mov. Disord. Takashima, H., Kikuchi, A., Aoki, M., Ishiura, H., Yasuda, T., Date, H., Ahsan, B.,
32, 1432–1438. Iwata, A., Goto, J., Ichikawa, Y., Nakahara, Y., Momose, Y., Takahashi, Y., Hara, K.,
Leonard, H., Blauwendraat, C., Krohn, L., Faghri, F., Iwaki, H., Ferguson, G., Day- Kakita, A., Yamada, M., Takahashi, H., Onodera, O., Nishizawa, M., Watanabe, H.,
Williams, A.G., Stone, D.J., Singleton, A.B., Nalls, M.A., Gan-Or, Z., 2020. Genetic Ito, M., Sobue, G., Ishikawa, K., Mizusawa, H., Kanai, K., Hattori, T., Kuwabara, S.,

12
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

Arai, K., Koyano, S., Kuroiwa, Y., Hasegawa, K., Yuasa, T., Yasui, K., Nakashima, K., Pchelina, S., Emelyanov, A., Baydakova, G., Andoskin, P., Senkevich, K., Nikolaev, M.,
Ito, H., Izumi, Y., Kaji, R., Kato, T., Kusunoki, S., Osaki, Y., Horiuchi, M., Kondo, T., Miliukhina, I., Yakimovskii, A., Timofeeva, A., Fedotova, E., Abramycheva, N.,
Murayama, S., Hattori, N., Yamamoto, M., Murata, M., Satake, W., Toda, T., Filla, A., Usenko, T., Kulabukhova, D., Lavrinova, A., Kopytova, A., Garaeva, L., Nuzhnyi, E.,
Klockgether, T., Wullner, U., Nicholson, G., Gilman, S., Tanner, C.M., Kukull, W.A., Illarioshkin, S., Zakharova, E., 2017. Oligomeric alpha-synuclein and
Stern, M.B., Lee, V.M., Trojanowski, J.Q., Masliah, E., Low, P.A., Sandroni, P., glucocerebrosidase activity levels in GBA-associated Parkinson’s disease. Neurosci.
Ozelius, L.J., Foroud, T., Tsuji, S., 2015. Variants associated with Gaucher disease in Lett. 636, 70–76.
multiple system atrophy. Ann Clin Transl Neurol 2, 417–426. Pchelina, S., Baydakova, G., Nikolaev, M., Senkevich, K., Emelyanov, A., Kopytova, A.,
Moors, T.E., Paciotti, S., Ingrassia, A., Quadri, M., Breedveld, G., Tasegian, A., Miliukhina, I., Yakimovskii, A., Timofeeva, A., Berkovich, O., Fedotova, E.,
Chiasserini, D., Eusebi, P., Duran-Pacheco, G., Kremer, T., Calabresi, P., Bonifati, V., Illarioshkin, S., Zakharova, E., 2018. Blood lysosphingolipids accumulation in
Parnetti, L., Beccari, T., van de Berg, W.D.J., 2019. Characterization of brain patients with Parkinson’s disease with glucocerebrosidase 1 mutations. Mov. Disord.
lysosomal activities in GBA-related and sporadic Parkinson’s disease and dementia 33, 1325–1330.
with Lewy bodies. Mol. Neurobiol. 56, 1344–1355. Peterschmitt, J., Alcalay, N.G.R., Cutter, G., Höglinger, G., Schapira, A., Simuni, C.S.T.,
Morabito, G., Giannelli, S.G., Ordazzo, G., Bido, S., Castoldi, V., Indrigo, M., Cabassi, T., Gurevich, T., Gasser, T., Pacchetti, C., Minini, K.M.P., Sardi, S., 2021. Effect of
Cattaneo, S., Luoni, M., Cancellieri, C., Sessa, A., Bacigaluppi, M., Taverna, S., venglustat by GBA mutation severity in patients with Parkinson’s disease. Mov.
Leocani, L., Lanciego, J.L., Broccoli, V., 2017. AAV-PHP.B-Mediated global-scale Disord. 36, 190.
expression in the mouse nervous system enables GBA1 gene therapy for wide Pfeffer, S.R., 2018. LRRK2 and Rab GTPases. Biochem. Soc. Trans. 46, 1707–1712.
protection from synucleinopathy. Mol. Ther. 25, 2727–2742. Piredda, R., Desmarais, P., Masellis, M., Gasca-Salas, C., 2020. Cognitive and psychiatric
Mullin, S., Smith, L., Lee, K., D’Souza, G., Woodgate, P., Elflein, J., Hällqvist, J., symptoms in genetically determined Parkinson’s disease: a systematic review. Eur. J.
Toffoli, M., Streeter, A., Hosking, J., Heywood, W.E., Khengar, R., Campbell, P., Neurol. 27, 229–234.
Hehir, J., Cable, S., Mills, K., Zetterberg, H., Limousin, P., Libri, V., Foltynie, T., Pont-Sunyer, C., Iranzo, A., Gaig, C., Fernández-Arcos, A., Vilas, D., Valldeoriola, F.,
Schapira, A.H.V., 2020. Ambroxol for the treatment of patients with Parkinson Compta, Y., Fernández-Santiago, R., Fernández, M., Bayés, A., Calopa, M.,
disease with and without glucocerebrosidase gene mutations: a nonrandomized, Casquero, P., de Fàbregues, O., Jaumà, S., Puente, V., Salamero, M., José Martí, M.,
noncontrolled trial. JAMA Neurology 77, 427–434. Santamaría, J., Tolosa, E., 2015. Sleep Disorders in Parkinsonian and
Muñoz, S.S., Petersen, D., Marlet, F.R., Kücükköse, E., Galvagnion, C., 2020. The Nonparkinsonian LRRK2 Mutation Carriers. PLoS One 10, e0132368.
interplay between GCase, α-synuclein and lipids in human models of Parkinson’s Posse de Chaves, E., Sipione, S., 2010. Sphingolipids and gangliosides of the nervous
disease. Biophys. Chem. 106534. system in membrane function and dysfunction. FEBS Lett. 584, 1748–1759.
Nalls, M.A., Duran, R., Lopez, G., Kurzawa-Akanbi, M., McKeith, I.G., Chinnery, P.F., Qian, E., Huang, Y., 2019. Subtyping of Parkinson’s disease - where are we up to? Aging
Morris, C.M., Theuns, J., Crosiers, D., Cras, P., Engelborghs, S., De Deyn, P.P., Van Dis 10, 1130–1139.
Broeckhoven, C., Mann, D.M., Snowden, J., Pickering-Brown, S., Halliwell, N., Reczek, D., Schwake, M., Schröder, J., Hughes, H., Blanz, J., Jin, X., Brondyk, W., Van
Davidson, Y., Gibbons, L., Harris, J., Sheerin, U.M., Bras, J., Hardy, J., Clark, L., Patten, S., Edmunds, T., Saftig, P., 2007. LIMP-2 is a receptor for lysosomal mannose-
Marder, K., Honig, L.S., Berg, D., Maetzler, W., Brockmann, K., Gasser, T., 6-phosphate-independent targeting of β-glucocerebrosidase. Cell 131, 770–783.
Novellino, F., Quattrone, A., Annesi, G., De Marco, E.V., Rogaeva, E., Masellis, M., Richter, F., Fleming, S.M., Watson, M., Lemesre, V., Pellegrino, L., Ranes, B., Zhu, C.,
Black, S.E., Bilbao, J.M., Foroud, T., Ghetti, B., Nichols, W.C., Pankratz, N., Mortazavi, F., Mulligan, C.K., Sioshansi, P.C., Hean, S., De La Rosa, K., Khanna, R.,
Halliday, G., Lesage, S., Klebe, S., Durr, A., Duyckaerts, C., Brice, A., Giasson, B.I., Flanagan, J., Lockhart, D.J., Wustman, B.A., Clark, S.W., Chesselet, M.F., 2014.
Trojanowski, J.Q., Hurtig, H.I., Tayebi, N., Landazabal, C., Knight, M.A., Keller, M., A GCase chaperone improves motor function in a mouse model of synucleinopathy.
Singleton, A.B., Wolfsberg, T.G., Sidransky, E., 2013. A multicenter study of Neurotherapeutics 11, 840–856.
glucocerebrosidase mutations in dementia with Lewy bodies. JAMA Neurol 70, Rideout, H.J., Chartier-Harlin, M.-C., Fell, M.J., Hirst, W.D., Huntwork-Rodriguez, S.,
727–735. Leyns, C.E., Mabrouk, O.S., Taymans, J.-M., 2020. The current state-of-the art of
Nalls, M.A., Blauwendraat, C., Vallerga, C.L., Heilbron, K., Bandres-Ciga, S., Chang, D., LRRK2-based biomarker assay development in Parkinson’s disease. Front. Neurosci.
Tan, M., Kia, D.A., Noyce, A.J., Xue, A., Bras, J., Young, E., von Coelln, R., Simon- 14, 865.
Sanchez, J., Schulte, C., Sharma, M., Krohn, L., Pihlstrom, L., Siitonen, A., Iwaki, H., Rocha, E.M., Smith, G.A., Park, E., Cao, H., Brown, E., Hallett, P., Isacson, O., 2015.
Leonard, H., Faghri, F., Gibbs, J.R., Hernandez, D.G., Scholz, S.W., Botia, J.A., Progressive decline of glucocerebrosidase in aging and Parkinson’s disease. Ann Clin
Martinez, M., Corvol, J.C., Lesage, S., Jankovic, J., Shulman, L.M., Sutherland, M., Transl Neurol 2, 433–438.
Tienari, P., Majamaa, K., Toft, M., Andreassen, O.A., Bangale, T., Brice, A., Yang, J., Rolfs, A., Giese, A.K., Grittner, U., Mascher, D., Elstein, D., Zimran, A., Böttcher, T.,
Gan-Or, Z., Gasser, T., Heutink, P., Shulman, J.M., Wood, N.W., Hinds, D.A., Lukas, J., Hübner, R., Gölnitz, U., Röhle, A., Dudesek, A., Meyer, W., Wittstock, M.,
Hardy, J.A., Morris, H.R., Gratten, J., Visscher, P.M., Graham, R.R., Singleton, A.B., Mascher, H., 2013. Glucosylsphingosine is a highly sensitive and specific biomarker
Me Research, T., System Genomics of Parkinson’s Disease, C., International for primary diagnostic and follow-up monitoring in Gaucher disease in a non-Jewish,
Parkinson’s Disease Genomics, C., 2019. Identification of novel risk loci, causal Caucasian cohort of Gaucher disease patients. PLoS One 8, e79732.
insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide Ron, I., Horowitz, M., 2005. ER retention and degradation as the molecular basis
association studies. Lancet Neurol. 18, 1091–1102. underlying Gaucher disease heterogeneity. Hum. Mol. Genet. 14, 2387–2398.
Narendra, D.P., Youle, R.J., 2011. Targeting mitochondrial dysfunction: role for PINK1 Ross, O.A., Soto-Ortolaza, A.I., Heckman, M.G., Aasly, J.O., Abahuni, N., Annesi, G.,
and Parkin in mitochondrial quality control. Antioxidants Redox Signal. 14, Bacon, J.A., Bardien, S., Bozi, M., Brice, A., Brighina, L., Van Broeckhoven, C.,
1929–1938. Carr, J., Chartier-Harlin, M.C., Dardiotis, E., Dickson, D.W., Diehl, N.N., Elbaz, A.,
Neil, J.F., Glew, R.H., Peters, S.P., 1979. Familial psychosis and diverse neurologic Ferrarese, C., Ferraris, A., Fiske, B., Gibson, J.M., Gibson, R., Hadjigeorgiou, G.M.,
abnormalities in adult-onset Gaucher’s disease. Arch. Neurol. 36, 95–99. Hattori, N., Ioannidis, J.P., Jasinska-Myga, B., Jeon, B.S., Kim, Y.J., Klein, C.,
Nixon-Abell, J., Berwick, D.C., Harvey, K., 2016. L’RRK de Triomphe: a solution for Kruger, R., Kyratzi, E., Lesage, S., Lin, C.H., Lynch, T., Maraganore, D.M., Mellick, G.
LRRK2 GTPase activity? Biochem. Soc. Trans. 44, 1625–1634. D., Mutez, E., Nilsson, C., Opala, G., Park, S.S., Puschmann, A., Quattrone, A.,
Obergasteiger, J., Frapporti, G., Lamonaca, G., Pizzi, S., Picard, A., Lavdas, A.A., Sharma, M., Silburn, P.A., Sohn, Y.H., Stefanis, L., Tadic, V., Theuns, J.,
Pischedda, F., Piccoli, G., Hilfiker, S., Lobbestael, E., 2020. Kinase inhibition of Tomiyama, H., Uitti, R.J., Valente, E.M., van de Loo, S., Vassilatis, D.K., Vilarino-
G2019S-LRRK2 enhances autolysosome formation and function to reduce Guell, C., White, L.R., Wirdefeldt, K., Wszolek, Z.K., Wu, R.M., Farrer, M.J., 2011.
endogenous alpha-synuclein intracellular inclusions. Cell death discovery 6, 1–13. Association of LRRK2 exonic variants with susceptibility to Parkinson’s disease: a
Olsen, A.S.B., Færgeman, N.J., 2017. Sphingolipids: membrane microdomains in brain case-control study. Lancet Neurol. 10, 898–908.
development, function and neurological diseases. Open Biol 7. Sanchez-Martinez, A., Beavan, M., Gegg, M.E., Chau, K.-Y., Whitworth, A.J., Schapira, A.
Omer, N., Giladi, N., Gurevich, T., Bar-Shira, A., Gana-Weisz, M., Goldstein, O., H.V., 2016. Parkinson disease-linked GBA mutation effects reversed by molecular
Kestenbaum, M., Cedarbaum, J.M., Orr-Urtreger, A., Mirelman, A., Thaler, A., 2020. chaperones in human cell and fly models. Sci. Rep. 6, 31380.
A possible modifying effect of the G2019S mutation in the LRRK2 gene on GBA Sardi, S.P., Viel, C., Clarke, J., Treleaven, C.M., Richards, A.M., Park, H., Olszewski, M.
Parkinson’s disease. Mov. Disord. 35, 1249–1253. A., Dodge, J.C., Marshall, J., Makino, E., Wang, B., Sidman, R.L., Cheng, S.H.,
Ortega, R.A., Wang, C., Raymond, D., Bryant, N., Scherzer, C.R., Thaler, A., Alcalay, R.N., Shihabuddin, L.S., 2017. Glucosylceramide synthase inhibition alleviates aberrations
West, A.B., Mirelman, A., Kuras, Y., 2021. Association of dual LRRK2 G2019S and in synucleinopathy models. Proc. Natl. Acad. Sci. Unit. States Am. 114, 2699–2704.
GBA variations with Parkinson disease progression. JAMA Network Open 4, Sauerbier, A., Jenner, P., Todorova, A., Chaudhuri, K.R., 2016. Non motor subtypes and
e215845 e215845. Parkinson’s disease. Park. Relat. Disord. 1 (22 Suppl), S41–46.
Ouled Amar Bencheikh, B., Ruskey, J.A., Arnulf, I., Dauvilliers, Y., Monaca, C.C., De Saunders-Pullman, R., Stanley, K., Wang, C., San Luciano, M., Shanker, V., Hunt, A.,
Cock, V.C., Gagnon, J.F., Spiegelman, D., Hu, M.T.M., Hogl, B., Stefani, A., Ferini- Severt, L., Raymond, D., Ozelius, L.J., Lipton, R.B., Bressman, S.B., 2011. Olfactory
Strambi, L., Plazzi, G., Antelmi, E., Young, P., Heidbreder, A., Mollenhauer, B., Sixel- dysfunction in LRRK2 G2019S mutation carriers. Neurology 77, 319–324.
Doring, F., Trenkwalder, C., Oertel, W., Montplaisir, J.Y., Postuma, R.B., Rouleau, G. Saunders-Pullman, R., Alcalay, R.N., Mirelman, A., Wang, C., Luciano, M.S., Ortega, R.A.,
A., Gan-Or, Z., 2018. LRRK2 protective haplotype and full sequencing study in REM Glickman, A., Raymond, D., Mejia-Santana, H., Doan, N., Johannes, B.,
sleep behavior disorder. Park. Relat. Disord. 52, 98–101. Yasinovsky, K., Ozelius, L., Clark, L., Orr-Utreger, A., Marder, K., Giladi, N.,
Paisán-Ruız, C., Jain, S., Evans, E.W., Gilks, W.P., Simón, J., Van Der Brug, M., De Bressman, S.B., Consortium, t. A. L, 2015. REM sleep behavior disorder, as assessed
Munain, A.L., Aparicio, S., Gil, A.M.n., Khan, N., 2004. Cloning of the gene by questionnaire, in G2019S LRRK2 mutation PD and carriers. Mov. Disord. 30,
containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 44, 1834–1839.
595–600. Sawkar, A.R., Cheng, W.-C., Beutler, E., Wong, C.-H., Balch, W.E., Kelly, J.W., 2002.
Parnetti, L., Chiasserini, D., Persichetti, E., Eusebi, P., Varghese, S., Qureshi, M.M., Chemical chaperones increase the cellular activity of N370S β-glucosidase: a
Dardis, A., Deganuto, M., De Carlo, C., Castrioto, A., Balducci, C., Paciotti, S., therapeutic strategy for Gaucher disease. Proc. Natl. Acad. Sci. Unit. States Am. 99,
Tambasco, N., Bembi, B., Bonanni, L., Onofrj, M., Rossi, A., Beccari, T., El-Agnaf, O., 15428–15433.
Calabresi, P., 2014. Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in
Parkinson’s disease. Mov. Disord. 29, 1019–1027.

13
K. Senkevich et al. Neuropharmacology 202 (2022) 108822

Schiffmann, R., Fitzgibbon, E.J., Harris, C., DeVile, C., Davies, E.H., Abel, L., van glucocerebrosidase activity associated with reduced levels of the trafficking receptor
Schaik, I.N., Benko, W., Timmons, M., Ries, M., Vellodi, A., 2008. Randomized, LIMP2. Mol. Brain 14, 16.
controlled trial of miglustat in Gaucher’s disease type 3. Ann. Neurol. 64, 514–522. Toffoli, M., Smith, L., Schapira, A.H.V., 2020. The biochemical basis of interactions
Schneider, S.A., Alcalay, R.N., 2017. Neuropathology of genetic synucleinopathies with between Glucocerebrosidase and alpha-synuclein in GBA1 mutation carriers.
parkinsonism: review of the literature. Mov. Disord. 32, 1504–1523. J. Neurochem. 154, 11–24.
Schofield, J.H., Schafer, Z.T., 2021. Mitochondrial reactive oxygen species and Tomkins, J.E., Dihanich, S., Beilina, A., Ferrari, R., Ilacqua, N., Cookson, M.R., Lewis, P.
mitophagy: a complex and nuanced relationship. Antioxidants Redox Signal. 34, A., Manzoni, C., 2018. Comparative protein interaction network analysis identifies
517–530. shared and distinct functions for the human ROCO proteins. Proteomics 18,
Senkevich, K., Gan-Or, Z., 2020. Autophagy lysosomal pathway dysfunction in 1700444.
Parkinson’s disease; evidence from human genetics. Park. Relat. Disord. 73, 60–71. Tong, Y., Giaime, E., Yamaguchi, H., Ichimura, T., Liu, Y., Si, H., Cai, H., Bonventre, J.V.,
Sheehan, P., Heckman, L.D., Fenn, T., Wong, L.C., Nelson, S., Garimalla, S., Haller, J., Shen, J., 2012. Loss of leucine-rich repeat kinase 2 causes age-dependent bi-phasic
Daily, J., Politi, J., Dai, Y., Hefti, F., Abeliovich, A., 2020. PR001 gene therapy alterations of the autophagy pathway. Mol. Neurodegener. 7, 1–16.
improved phenotypes in models of Parkinson’s disease with GBA1 mutation. Treiber, A., Morand, O., Clozel, M., 2007. The pharmacokinetics and tissue distribution
Alzheimer’s & Dementia 16, e043614. of the glucosylceramide synthase inhibitor miglustat in the rat. Xenobiotica 37,
Shiner, T., Mirelman, A., Rosenblum, Y., Kavé, G., Weisz, M.G., Bar-Shira, A., 298–314.
Goldstein, O., Thaler, A., Gurevich, T., Orr-Urtreger, A., Giladi, N., Bregman, N., Trinh, J., Gustavsson, E.K., Vilariño-Güell, C., Bortnick, S., Latourelle, J., McKenzie, M.
2021. The effect of GBA mutations and APOE polymorphisms on dementia with B., Tu, C.S., Nosova, E., Khinda, J., Milnerwood, A., 2016. DNM3 and genetic
Lewy bodies in Ashkenazi Jews. J Alzheimers Dis 80, 1221–1229. modifiers of age of onset in LRRK2 Gly2019Ser parkinsonism: a genome-wide
Sidransky, E., Nalls, M.A., Aasly, J.O., Aharon-Peretz, J., Annesi, G., Barbosa, E.R., Bar- linkage and association study. Lancet Neurol. 15, 1248–1256.
Shira, A., Berg, D., Bras, J., Brice, A., 2009. Multicenter analysis of Tsuang, D., Leverenz, J.B., Lopez, O.L., Hamilton, R.L., Bennett, D.A., Schneider, J.A.,
glucocerebrosidase mutations in Parkinson’s disease. N. Engl. J. Med. 361, Buchman, A.S., Larson, E.B., Crane, P.K., Kaye, J.A., Kramer, P., Woltjer, R.,
1651–1661. Kukull, W., Nelson, P.T., Jicha, G.A., Neltner, J.H., Galasko, D., Masliah, E.,
Silveira, C.R.A., MacKinley, J., Coleman, K., Li, Z., Finger, E., Bartha, R., Morrow, S.A., Trojanowski, J.Q., Schellenberg, G.D., Yearout, D., Huston, H., Fritts-Penniman, A.,
Wells, J., Borrie, M., Tirona, R.G., Rupar, C.A., Zou, G., Hegele, R.A., Mahuran, D., Mata, I.F., Wan, J.Y., Edwards, K.L., Montine, T.J., Zabetian, C.P., 2012. GBA
MacDonald, P., Jenkins, M.E., Jog, M., Pasternak, S.H., 2019. Ambroxol as a novel mutations increase risk for Lewy body disease with and without Alzheimer disease
disease-modifying treatment for Parkinson’s disease dementia: protocol for a single- pathology. Neurology 79, 1944–1950.
centre, randomized, double-blind, placebo-controlled trial. BMC Neurol. 19, 20. Van Bogaert, L., Froehlich, A., 1939. Un cas de maladie de Gaucher de la dulte avec
Sklerov, M., Kang, U.J., Liong, C., Clark, L., Marder, K., Pauciulo, M., Nichols, W.C., syndrome de Raynaud, pigmentation, et rigidite du type extrapyr-ajidal aux
Chung, W.K., Honig, L.S., Cortes, E., Vonsattel, J.P., Alcalay, R.N., 2017. Frequency membres inferieurs. Ann. Med. 45, 57–70.
of GBA variants in autopsy-proven multiple system atrophy. Mov Disord Clin Pract 4, van Dijkman, S.C., de Jager, N.C.B., Rauwé, W.M., Danhof, M., Della Pasqua, O., 2018.
574–581. Effect of age-related factors on the pharmacokinetics of lamotrigine and potential
Sliter, D.A., Martinez, J., Hao, L., Chen, X., Sun, N., Fischer, T.D., Burman, J.L., Li, Y., implications for maintenance dose optimisation in future clinical trials. Clin.
Zhang, Z., Narendra, D.P., 2018. Parkin and PINK1 mitigate STING-induced Pharmacokinet. 57, 1039–1053.
inflammation. Nature 561, 258–262. Venglustat in Parkinson’s disease patients with a GBA mutation: results from Part 2 of the
Soffer, D., Yamanaka, T., Wenger, D.A., Suzuki, K., Suzuki, K., 1980. Central nervous phase 2 MOVES-PD trial. In: The 15th International Conference for Alzheimer’s &
system involvement in adult-onset Gaucher’s disease. Acta Neuropathol. 49, 1–6. Parkinson’s Diseases (Virtual), 1937.
Sosero, Y.L., Yu, E., Krohn, L., Rudakou, U., Mufti, K., Ruskey, J.A., Asayesh, F., von Linstow, C.U., Gan-Or, Z., Brundin, P., 2020. Precision medicine in Parkinson’s
Laurent, S.B., Spiegelman, D., Fahn, S., Waters, C., Sardi, S.P., Bandres-Ciga, S., disease patients with LRRK2 and GBA risk variants – let’s get even more personal.
Alcalay, R.N., Gan-Or, Z., Senkevich, K., 2021. LRRK2 p.M1646T is associated with Transl. Neurodegener. 9, 39.
glucocerebrosidase activity and with Parkinson’s disease. Neurobiol Aging. Wang, Y., Wu, J.J., Liu, F.T., Chen, K., Chen, C., Luo, S.S., Wang, Y.X., Li, D.k., Guan, R.
Srivatsal, S., Cholerton, B., Leverenz, J.B., Wszolek, Z.K., Uitti, R.J., Dickson, D.W., Y., Yang, Y.J., 2017. Olfaction in Parkin carriers in Chinese patients with Parkinson
Weintraub, D., Trojanowski, J.Q., Van Deerlin, V.M., Quinn, J.F., Chung, K.A., disease. Brain and behavior 7, e00680.
Peterson, A.L., Factor, S.A., Wood-Siverio, C., Goldman, J.G., Stebbins, G.T., Wernick, A.I., Walton, R.L., Koga, S., Soto-Beasley, A.I., Heckman, M.G., Gan-Or, Z.,
Bernard, B., Ritz, B., Rausch, R., Espay, A.J., Revilla, F.J., Devoto, J., Rosenthal, L.S., Ren, Y., Rademakers, R., Uitti, R.J., Wszolek, Z.K., Cheshire, W.P., Dickson, D.W.,
Dawson, T.M., Albert, M.S., Mata, I.F., Hu, S.-C., Montine, K.S., Johnson, C., Ross, O.A., 2020. GBA variation and susceptibility to multiple system atrophy. Park.
Montine, T.J., Edwards, K.L., Zhang, J., Zabetian, C.P., 2015. Cognitive profile of Relat. Disord. 77, 64–69.
LRRK2-related Parkinson’s disease. Movement disorders. official journal of the Whiffin, N., Armean, I.M., Kleinman, A., Marshall, J.L., Minikel, E.V., Goodrich, J.K.,
Movement Disorder Society 30, 728–733. Quaife, N.M., Cole, J.B., Wang, Q., Karczewski, K.J., 2020. The effect of LRRK2 loss-
Steger, M., Diez, F., Dhekne, H.S., Lis, P., Nirujogi, R.S., Karayel, O., Tonelli, F., of-function variants in humans. Nat. Med. 26, 869–877.
Martinez, T.N., Lorentzen, E., Pfeffer, S.R., 2017. Systematic proteomic analysis of Wojewska, D.N., Kortholt, A., 2021. LRRK2 targeting strategies as potential treatment of
LRRK2-mediated Rab GTPase phosphorylation establishes a connection to Parkinson’s disease. Biomolecules 11, 1101.
ciliogenesis. Elife 6, e31012. Xie, C.-l., Pan, J.-L., Wang, W.-W., Zhang, Y., Zhang, S.-f., Gan, J., Liu, Z.-G., 2014. The
Stirnemann, J., Belmatoug, N., Camou, F., Serratrice, C., Froissart, R., Caillaud, C., association between the LRRK2 G2385R variant and the risk of Parkinson’s disease: a
Levade, T., Astudillo, L., Serratrice, J., Brassier, A., 2017. A review of Gaucher meta-analysis based on 23 case–control studies. Neurol. Sci. 35, 1495–1504.
disease pathophysiology, clinical presentation and treatments. Int. J. Mol. Sci. 18, Yahalom, G., Greenbaum, L., Israeli-Korn, S., Fay-Karmon, T., Livneh, V., Ruskey, J.A.,
441. Ronciere, L., Alam, A., Gan-Or, Z., Hassin-Baer, S., 2019. Carriers of both GBA and
Sun, Y., Liou, B., Xu, Y.-H., Quinn, B., Zhang, W., Hamler, R., Setchell, K.D., LRRK2 mutations, compared to carriers of either, in Parkinson’s disease: risk
Grabowski, G.A., 2012. Ex vivo and in vivo effects of isofagomine on acid estimates and genotype-phenotype correlations. Park. Relat. Disord. 62, 179–184.
β-glucosidase variants and substrate levels in Gaucher disease. J. Biol. Chem. 287, Yap, T.L., Gruschus, J.M., Velayati, A., Sidransky, E., Lee, J.C., 2013. Saposin C protects
4275–4287. glucocerebrosidase against α-synuclein inhibition. Biochemistry 52, 7161–7163.
Swan, M., Doan, N., Ortega, R.A., Barrett, M., Nichols, W., Ozelius, L., Soto-Valencia, J., Ysselstein, D., Nguyen, M., Young, T.J., Severino, A., Schwake, M., Merchant, K.,
Boschung, S., Deik, A., Sarva, H., 2016. Neuropsychiatric characteristics of GBA- Krainc, D., 2019. LRRK2 kinase activity regulates lysosomal glucocerebrosidase in
associated Parkinson disease. Journal of the neurological sciences 370, 63–69. neurons derived from Parkinson’s disease patients. Nat. Commun. 10, 5570.
Taguchi, Y.V., Liu, J., Ruan, J., Pacheco, J., Zhang, X., Abbasi, J., Keutzer, J., Mistry, P. Yu, E., Rudakou, U., Krohn, L., Mufti, K., Ruskey, J.A., Asayesh, F., Estiar, M.A.,
K., Chandra, S.S., 2017. Glucosylsphingosine promotes α-synuclein pathology in Spiegelman, D., Surface, M., Fahn, S., Waters, C.H., Greenbaum, L., Espay, A.J.,
mutant GBA-associated Parkinson’s disease. J. Neurosci. 37, 9617–9631. Dauvilliers, Y., Dupré, N., Rouleau, G.A., Hassin-Baer, S., Fon, E.A., Alcalay, R.N.,
Thaler, A., Gurevich, T., Bar Shira, A., Gana Weisz, M., Ash, E., Shiner, T., Orr- Gan-Or, Z., 2021. Analysis of heterozygous PRKN variants and copy-number
Urtreger, A., Giladi, N., Mirelman, A., 2017. A "dose" effect of mutations in the GBA variations in Parkinson’s disease. Mov. Disord. 36, 178–187.
gene on Parkinson’s disease phenotype. Park. Relat. Disord. 36, 47–51. Zhang, X.-H., Tee, L.Y., Wang, X.-G., Huang, Q.-S., Yang, S.-H., 2015. Off-target effects in
Thaler, A., Bregman, N., Gurevich, T., Shiner, T., Dror, Y., Zmira, O., Gan-Or, Z., Bar- CRISPR/Cas9-mediated genome engineering. Mol. Ther. Nucleic Acids 4, e264.
Shira, A., Gana-Weisz, M., Orr-Urtreger, A., Giladi, N., Mirelman, A., 2018. Zhang, Y., Sun, Q., Yi, M., Zhou, X., Guo, J., Xu, Q., Tang, B., Yan, X., 2017. Genetic
Parkinson’s disease phenotype is influenced by the severity of the mutations in the analysis of LRRK2 R1628P in Parkinson’s disease in Asian populations. Parkinson’s
GBA gene. Park. Relat. Disord. 55, 45–49. Disease 2017.
Thaler, A., Omer, N., Giladi, N., Gurevich, T., Bar-Shira, A., Gana-Weisz, M., Zhang, Y., Shu, L., Zhou, X., Pan, H., Xu, Q., Guo, J., Tang, B., Sun, Q., 2018. A meta-
Goldstein, O., Kestenbaum, M., Shirvan, J.C., Cedarbaum, J.M., Orr-Urtreger, A., analysis of GBA-related clinical symptoms in Parkinson’s disease. Parkinson’s Dis.
Regev, K., Shenhar-Tsarfaty, S., Mirelman, A., 2021. Mutations in GBA and LRRK2 2018, 3136415.
are not associated with increased inflammatory markers. J Parkinsons dis. Zhao, Y., Dzamko, N., 2019. Recent developments in LRRK2-targeted therapy for
Thenganatt, M.A., Jankovic, J., 2014. Parkinson disease subtypes. JAMA Neurol 71, Parkinson’s disease. Drugs 79, 1037–1051.
499–504. Zhao, H.T., John, N., Delic, V., Ikeda-Lee, K., Kim, A., Weihofen, A., Swayze, E.E.,
Thirumal Kumar, D., Iyer, S., Christy, J.P., Siva, R., Tayubi, I.A., George Priya Doss, C., Kordasiewicz, H.B., West, A.B., Volpicelli-Daley, L.A., 2017. LRRK2 antisense
Zayed, H., 2019. A comparative computational approach toward pharmacological oligonucleotides ameliorate α-synuclein inclusion formation in a Parkinson’s disease
chaperones (NN-DNJ and ambroxol) on N370S and L444P mutations causing mouse model. Mol. Ther. Nucleic Acids 8, 508–519.
Gaucher’s disease. Adv Protein Chem Struct Biol 114, 315–339. Zimprich, A., Biskup, S., Leitner, P., Lichtner, P., Farrer, M., Lincoln, S., Kachergus, J.,
Thomas, R., Moloney, E.B., Macbain, Z.K., Hallett, P.J., Isacson, O., 2021. Fibroblasts Hulihan, M., Uitti, R.J., Calne, D.B., 2004. Mutations in LRRK2 cause autosomal-
from idiopathic Parkinson’s disease exhibit deficiency of lysosomal dominant parkinsonism with pleomorphic pathology. Neuron 44, 601–607.

14

You might also like