O-Glcnacase Inhibitors As Potential Therapeutics For The Treatment of Alzheimer'S Disease and Related Tauopathies: Analysis of The Patent Literature

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 39

Expert Opinion on Therapeutic Patents

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ietp20

O-GlcNAcase inhibitors as potential therapeutics


for the treatment of Alzheimer’s disease and
related tauopathies: analysis of the patent
literature

Jose M. Bartolomé-Nebreda, Andrés A. Trabanco, Adriana Ingrid Velter &


Peter Buijnsters

To cite this article: Jose M. Bartolomé-Nebreda, Andrés A. Trabanco, Adriana Ingrid Velter &
Peter Buijnsters (2021): O-GlcNAcase inhibitors as potential therapeutics for the treatment of
Alzheimer’s disease and related tauopathies: analysis of the patent literature, Expert Opinion on
Therapeutic Patents, DOI: 10.1080/13543776.2021.1947242

To link to this article: https://doi.org/10.1080/13543776.2021.1947242

Published online: 08 Jul 2021.

Submit your article to this journal

Article views: 73

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ietp20
EXPERT OPINION ON THERAPEUTIC PATENTS
https://doi.org/10.1080/13543776.2021.1947242

REVIEW

O-GlcNAcase inhibitors as potential therapeutics for the treatment of Alzheimer’s


disease and related tauopathies: analysis of the patent literature
Jose M. Bartolomé-Nebredaa, Andrés A. Trabancoa, Adriana Ingrid Velterb and Peter Buijnstersb
a
A Division of Janssen-Cilag SA, Discovery Chemistry Department, Discovery, Product Development & Supply, Janssen Research and Development,
Toledo, Spain; bA Division of Janssen Pharmaceutica NV, Discovery Chemistry Department, Discovery, Product Development & Supply, Janssen
Research and Development, Beerse, Belgium

ABSTRACT ARTICLE HISTORY


Introduction: O-GlcNAcylation is a highly abundant post-translational modification of multiple proteins, Received 05 March 2021
including the microtubule-binding protein tau, governed by just two enzymes’ concerted action Accepted 21 June 2021
O-GlcNAc transferase OGT and the hydrolase OGA. It is an approach to reduce abnormal tau hyperpho­
KEYWORDS
sphorylation and aggregation in Alzheimer’s disease (AD) and related tauopathies based on the ability
O-GlcNAcase; OGA; tau;
of O-GlcNAcylation competing with tau phosphorylation, thus minimizing aggregation. The preclinical Alzheimer’s disease;
validation confirmed OGA inhibitors’ efficacy in different transgenic tau mice models. Only three other tauopathies
OGA inhibitors have advanced into clinical trials thus far.
Areas covered: 2008–2020 patent literature on OGA inhibitors.
Expert opinion: Neurodegenerative disorders and AD specifically represent an enormous challenge
since no effective treatments are available. Promising preclinical data has prompted considerable
interest in searching for OGA inhibitors as a potential treatment for neurodegenerative disorders.
Efforts from different companies have yielded a diverse set of chemotypes. OGA is a highly ubiquitous
enzyme with many client proteins, generated data confirms a promising benign profile for OGA
inhibition in healthy volunteers. Additionally, OGA PET tracers’ existence will be critical for proper
dose selection for future PoC Phase II studies, which will proof the true potential of OGA inhibition for
the treatment of AD and other tauopathies.

1. Introduction preparations in 1994 [27]. Then in 1998, it was identified as


antigen five expressed by meningiomas (MGEA5) [28]. Full-
O-GlcNAcylation is a post-translational modification (PTM),
length OGA is a 103 kDa ubiquitously expressed enzyme. It
described for the first time by Torres and Hart in 1984 [1,2].
is soluble and highly conserved in eukaryotes with more than
It consists of the covalent connection of a single sugar
4000 client proteins [29]. Two different isoforms of OGA have
residue, O-linked β-N-acetyl glucosamine (O-GlcNAc), onto
been described so far [30]: long OGA, present in the nucleus
serine and threonine side-chain hydroxyl moieties of
and the cytoplasm, and a shorter isoform (sOGA) mainly found
nuclear, cytoplasmic and, to a lesser extent, mitochondrial
in the nucleus [31]. The catalytic activity of OGA is in the
proteins [3–5]. This PTM is implicated in regulating several
N-terminal part. It belongs to the glycoside hydrolase family
vital cellular processes: gene regulation [6], signal transduc­
84 (GH84) [29]. The C-terminus contains a putative histone
tion [7,8], cell cycle management [9] or proteasomal degra­
acetyltransferase (HAT) domain connected through an intrin­
dation[10]. Dysregulation in O-GlcNAcylation is associated
sically disordered stalk region [32,33]. OGA catalyzes the
with diseases, i.e. insulin resistance [11,12], obesity [13],
hydrolytic cleavage of O-GlcNAc residues from client proteins
diabetes [14,15], cardiovascular diseases [16], cancer [17,18]
through a two-step mechanism. The substrate acetamido
and neurodegeneration [19,20].
group [34] assists and acts as an intramolecular nucleophile
Akin to other PTMs, such as phosphorylation,
attacking the anomeric position. This process involves the
O-GlcNAcylation is a non-canonical, highly dynamic and rever­
participation of two aspartic (Asp) acid residues in the active
sible process [21,22]. The two concerted actions of the
site (Asp 174 and Asp175) and takes place through the forma­
enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA)
tion of a bicyclic oxazolidine-containing intermediate [35].
are responsible for O-GlcNAcylation homeostasis. OGT installs
The first OGA inhibitor used for in vitro studies was the
the O-GlcNAc residue on proteins using UDP-GlcNAc as
natural product streptozotocin (STZ, 1, Figure 2) [36,37]. STZ is
a donor. [23–25] OGA, a hydrolase, is accountable for its
a modest micromolar inhibitor in vitro with limited applicabil­
hydrolytic removal (Figure 1). OGA, initially described as hex­
ity due to structure-related toxicities unrelated to its ability to
osaminidase C [26], was isolated and purified from spleen
inhibit OGA [38]. PUGNAc [39] (2, Figure 2) is a significantly

CONTACT Peter Buijnsters pbuijnst@its.jnj.com Discovery Chemistry; Discovery, Product Development & Supply, Janssen Research and Development,
Turnhoutseweg 30, 2340 Beerse, Belgium
© 2021 Informa UK Limited, trading as Taylor & Francis Group
2 J. M. BARTOLOMÉ-NEBREDA ET AL.

a transition state mimic of the planar oxocarbenium ion-like


Article highlights OGA transition state, which could explain this compound’s
high potency[43]. Noteworthy is the publication of iminocycli­
● O-GlcNAcylation is a highly abundant post-translational modification of
multiple proteins. It includes the concerted action of O-GlcNAc transfer­ tol derivative 8, a potent, orally available and brain-penetrant
ase OGT and the hydrolase OGA. OGA-inhibitor. [44]
● In Alzheimer’s disease (AD) and related tauopathies, tau gets hyperpho­ X-ray crystallography aided the identification and optimiza­
sphorylated and aggregates into neurofibrillary tangles. The extent, the
spreading pattern and the concentration of neurofibrillary tangles cor­ tion of various prototypical OGA-inhibitors mentioned above.
relate with neurodegeneration severity. In vitro, increased However, they used different bacterial OGA orthologs,
O-GlcNAcylation reduces tau aggregation’s extent and speed, and whereas human OGA crystallography remained elusive until
O-GlcNAcylation can compete with phosphorylation.
● Extensive preclinical validation, using relevant tau transgenic models, very recently. Three independent laboratories reported, almost
has confirmed robust in vivo efficacy for the highly potent and brain simultaneously, X-ray-derived structures of other human OGA
penetrant OGA tool compound Thiamet G. constructs complexed with different inhibitors, including
● OGA inhibition has emerged as a novel potential therapeutic target for
treating AD and related neurodegenerative diseases. Thiamet G [45–47]. This will undoubtedly spur on the rational
● Three OGA inhibitors have entered clinical trials: MK-8719 from Merck/ design of novel classes of inhibitors.
Alectos, ASN-120,290 from Asceneuron S.A. and LY-3,372,689 from Eli OGT and OGA are expressed ubiquitously in the human body.
Lilly.
● Released data suggests a benign side effect profile for MK-8719 and One of the highest expressing areas for both enzymes is the brain
ASN-120,290 and confirmed target engagement in the brain for MK- [4,28] especially hippocampal and the cortical regions [48–50]. In
8719. addition, several brain-specific proteins are O-GlcNAcylated [51],
● Phase II PoC studies are still pending to undoubtedly support the great
promises that OGA inhibition holds for treating AD and related thereby suggesting a prominent relevant role for GlcNAcylation in
tauopathies. regulating multiple brain processes[52]. The hexosaminidase path­
way (HBP, Figure 1) [53]converts 3–5% of the body glucose to UDP-
This box summarizes key points contained in the article.
GlcNAc. Moreover, dysregulation of brain glucose metabolism by
decreasing O-GlcNAcylation of proteins might play a role in the
pathogenesis of neurodegenerative diseases such as AD[54]..
Moreover, the formation of neurofibrillary tangles (NFTs) as
more potent in vitro inhibitor than STZ. However, its limited a result of abnormal hyperphosphorylation and aggregation of tau-
selectivity versus lysosomal hexosaminidases [35] has also proteins is one of AD’s main hallmarks. Recent longitudinal studies
hampered its wider use. Off-target inhibition of the lysosomal in AD patients confirmed that tau aggregation, spreading pattern,
hexosaminidases is undesirable due to the association of these and concentration of NFTs correlate with neurodegeneration, brain
enzymes with the lysosomal storage disorders Tay-Sachs and atrophy, and cognitive symptom evolution, suggesting a pivotal
Sandhoff diseases. Figure 2 depicts a series of fused thiazoline- role for tau in disease progression [55,56]. Aggregates of hyperpho­
containing OGA inhibitors designed and inspired by oxazoline sphorylated tau are also a common feature of other neurodegen­
intermediate 3. NAG-Thiazoline [40] (4, Figure 2), the first erative diseases, collectively termed tauopathies. They comprise
member of the series, is a potent OGA inhibitor but lacks progressive supranuclear palsy (PSP), frontotemporal dementia
adequate selectivity versus lysosomal hexosaminidases [35]. (FD) or corticobasal syndrome (CBS), among others [57]. Tau is
The introduction of larger substituents on the thiazoline core one of the client proteins of OGA[58]. Postmortem studies of AD
resulted in a significantly improved selectivity in NButGT [41] brain extracts revealed a reduction of O-GlcNAcylated tau levels
(5, Figure 2), especially in Thiamet G [42] (6, Figure 2). Thiamet and an almost entire lack of O-GlcNAcylation in aggregates.
G is a highly potent and selective inhibitor that additionally [19,55,59] In a related way, in vitro studies have shown how
possesses the ability to penetrate the blood-brain barrier and O-GlcNAcylation reduces tau aggregation’s degree and speed in
is currently the most widely used OGA tool compound. cells [60,61].
Figure 2 depicts a structurally unrelated OGA inhibitor, Phosphorylation and O-GlcNAcylation occur on identical
GlcNAcstatin C [43] (7), a highly potent and selective tetrahy­ residues on client proteins suggesting a reciprocal interplay
dro-imidazopyridine-containing, picomolar inhibitor. The imi­ between them. Thus, O-GlcNAcylation could directly compete
dazopyridine core of GlcNAcstatin C is proposed to act as with phosphorylation on the identical residues and indirectly

Figure 1. Schematic representation of the O-GlcNAcylation cycle.


EXPERT OPINION ON THERAPEUTIC PATENTS 3

affect neighboring or proximal sites [62,63]. This evidence


supports a potential role for the upregulation of tau
O-GlcNAcylation via OGA inhibition with small molecules for
the possible treatment of AD and related tauopathies[64].
Suitable OGA inhibitor tool compounds played an essential
role in the preclinical validation of the hypothesis, as men­
tioned above. Sub-chronic administration of Thiamet G [43] in
rodents revealed that OGA inhibition is well-tolerated.
Prolonged OGA inhibition in transgenic mice expressing pro- Figure 3. Structure of the Merck/Alectos clinical asset MK-8719 and the Eli Lilly
aggregation tau-mutants (JNPL3 [65,66] and Tg4510 [67–70]) PET tracer [18F]-LSN3316612.
showed an increase in tau O-GlcNAcylation, decreasing tau
aggregation and NFTs. In P301L transgenic mice, it reduced
Treatment with MK-8719 increased the O-GlcNAc level in per­
the severity of breathing defects and enhanced survival [71].
ipheral blood mononuclear cells (PBMCs) and displacement of
Nurtured by these promising results, several pharmaceutical
[18F]-MK-8553, a positron emission tomography (PET) ligand of
companies have embarked on drug discovery programs to
undisclosed structure, in the brain. Asceneuron S.A. has also
identify OGA inhibitors that could have the potential to pro­
reported phase I data on ASN-120,290 (formerly ASN-561); its
gress into clinical trials to confirm in humans the therapeutic
molecular structure is unknown, and the compound is being
potential of this approach for the treatment of AD and related
developed for PSP treatment. Sub-chronic administration of
tauopathies [72]. A close analogue of Thiamet G, MK-8719,
ASN-120,290 to P301S mice proved to be well tolerated and
compound 9 (Figure 3) has an improved brain penetration
increased tau O-GlcNAcylation and significantly reduced
and a favorable preclinical toxicological profile. Therefore,
abnormally hyperphosphorylated tau [76,77]. In the clinic,
Merck/Alectos have moved MK-8719 [73] (9, Figure 3) into
ASN-120,290 was safe and taken by healthy young and elderly
phase I clinical trials for PSP treatment. Thus, single ascending
volunteers following single oral doses up to 1000 mg and after
doses (SAD) of MK-8719 up to 1200 mg have shown good
multiple oral doses up to 500 mg BID. To support dose selec­
pharmacokinetics, and the compound was safe and well-
tion for PSP efficacy trials, Asceneuron S.A. has also reported
tolerated with no noticeable adverse effects [74,75]. In vivo,
the start of an additional clinical trial aimed to evaluate target
target engagement was confirmed in blood and the brain.
engagement of ASN120,290 in the brain using a proprietary

Figure 2. Structures of some prototypical OGA inhibitor tool compounds.


4 J. M. BARTOLOMÉ-NEBREDA ET AL.

PET ligand of undisclosed structure [78]. Finally, Eli Lilly also and research collaboration agreement with Merck Sharp &
claims in its pipeline to have both an OGA inhibitor (LY- Dohme Inc., published multiple patent applications around
3,372,689) [79] of an unknown structure [80] as well as a PET several chemotypes between 2008 and 2014. Among those,
ligand ([18F]-LSN3316612) [81] undergoing Phase 1 clinical they published six patent applications disclosing substituted
trials for AD. However, no data has been released yet. octahydroindolizines, piperidines and pyrrolidines. The first
Following the high promise as potential therapeutic agents held application from 2010 described 51 compounds, all compris­
by OGA inhibitors, supported by the extensive preclinical validation ing a (1S,6S,7 R,8 R,8aR)-6-aminooctahydroindolizine-1,7,8-triol
described above, this review will summarize the existing OGA small scaffold[82]. Figure 4 depicts some specific derivatives (11–14)
molecule patent landscape focusing onAD and related tauopathies. from the patent application. It contains substituted amines,
amides and ureas, of which the R-groups are linear, substi­
tuted small alkyl or alkenyl radicals. Simultaneously, the patent
2. Patent review of OGA inhibitors application disclosed three unsubstituted benzylic derivatives,
2.1. Simon Fraser University/Alectos Therapeutics Inc. and it revealed hOGA enzymatic activity for compound 11
and Merck, Sharp & Dohme Corp only.
The next case, also from 2010, disclosed 30 2-(amino­
Simon Fraser University first, followed by Alectos Therapeutics methyl)-pyrrolidine-3,4-diol derivatives defined by the
(a spin-out company from Simon Fraser University) in a license

Figure 4. Representative aminooctahydroindolizine OGA inhibitors from Simon Fraser University.

Figure 5. Representative substituted pyrrolidine OGA inhibitors from Simon Fraser University.

Figure 6. Representative substituted pyrrolidine OGA inhibitors from Simon Fraser University, Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 5

Figure 7. Representative substituted pyrrolidine OGA inhibitors from Simon Fraser University, Alectos Therapeutics and Merck Sharp & Dohme.

Figure 8. Representative substituted piperidine-3-carboxamide OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.

Figure 9. Representative substituted piperidine-2-carboxamide OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
6 J. M. BARTOLOMÉ-NEBREDA ET AL.

Markush formula depicted in Figure 5[83]. It describes mainly The subsequent patent application (WO032188) reported
acetamides such as compound 15 with 0.85 µM enzymatic more chemical diversity (Figure 7). The exemplified com­
activity for hOGA. Other variations include various alkyl groups pounds, however, differ in the position the amide is attached
on the pyrrolidine N1 atom, e.g. 16–19, and different groups to the 2-position of the pyrrolidine core, i.e. CH2N(CO)alkyl or
at C5 position such as compounds 20 and 21. (CH2)C(O)N-alkyl.
In 2014, two patent applications, WO032185 [84] and Figure 7 depicts several representative examples that
WO032188[85], described various substituted pyrrolidine ana­ demonstrate this difference (30–35). The most promising bio­
logues. The first publication disclosed 21 substituted pyrroli­ logical activity resides in compounds with the three-carbon
dines, and Figure 6 depicts that all the compounds in that tether when comparing close analogues (33, 34, 38). This
document have no substituents at the C2 position. The pyrro­ patent publication reveals apart from hOGA biological data
lidines contain F-atoms at the C4-atom and the C5 methylene also intrinsic permeability for selected molecules (compounds
group (compounds 22–24). Most likely, the presence of the 31 and 32). Interestingly, the chirality has a marked influence
F-atoms helps modulate the basicity of the pyrrolidine on the pharmacological activity, i.e. the 3S-hydroxy derivative
N-atom. Most of the molecules comprise a 3-carbon tether (39) is approximately seven times more active than the
that links a (hetero)aromatic group to the pyrrolidine 3 R analogue (33). Again, several examples have F-atoms
(Figure 6). Furthermore, most compounds include an aceta­ installed at different positions in the products like in the
mide or a propionamide at the N3-atom. However, the docu­ previous application; however, without biological activity.
ment revealed biological activity for three compounds 27–29 The subsequent two patent applications disclosed piperi­
only. dine derivatives. These compounds revealed a similar substi­
tution pattern, and the various groups decorating them are
very similar to the pyrrolidine derivatives. In WO032187, the
C2-atom is not substituted, and the R-groups have
a comparable scope as the corresponding pyrrolidines
(WO032185). Compound 40 (Figure 8) has an inhibitory activ­
ity of 2578 nM against hOGA [86].
The 2-substituted piperidine derivatives described in
WO032184 resemble the pyrrolidine analogues of WO032187.
Albeit, all the 56 exemplified compounds are 2-methyl or
2-ethylcarboxamides. Figure 9 shows some representative
examples, and derivative 41 has a remarkable difference in
Figure 10. Representative tetrahydro-4 H-cyclopenta[d]oxazole and thiazole-
4,5-diol OGA inhibitors from Simon Fraser University. biological activity compared to molecule 42, 165 nM versus

Figure 11. Representative hexahydrobenzo[d]oxazole-4,5-diol OGA inhibitors from Simon Fraser University, Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 7

Figure 12. Representative 2-(dimethylamino)-1-hydroxyethyl-3a,4,5,6,7,7a-hexahydrobenzo[d]oxazole-4,5-diol OGA inhibitors from Alectos Therapeutics and Merck
Sharp & Dohme.

3297 nM, respectively. Furthermore, adding more lipophilicity 4 H-cyclopenta[d]thiazole (Figure 10) comprising alkyl,
as in compounds 43–45 results in interesting SAR. Moreover, it branched alkyl, cycloalkyl, aryl or heteroaryl groups. Still, in
resulted in a single-digit nanomolar inhibitor 43, but it does most cases, this includes N(Me)2, and biological data is
not improve the ligand efficiency (LE) compared to compound reported for one molecule (46) only.
41, i.e. LE amounts to 0.62 and 0.37, respectively [87]. The next application disclosed (WO140640) [89] com­
Eight patent applications revealed Thiamet G (6) analogues. pounds that comprise hexahydrobenzo[d]thiazole and hex­
They described differences in the central core scaffold: (1) the ahydrobenzo[d]oxazole derivatives (33 compounds), in
size of the two interconnected rings, (2) the nature of the analogy to the previous document, replacing the O-atom
heteroaromatic 5-membered moiety and (3) the various of the pyrane ring of Thiamet G with a carbon atom.
(cyclo)alkyl substituents. The first document (WO012107) [88] Figure 11 depicts some exciting examples. Interestingly,
described 21 exemplified compounds that possess instead of the synthesized examples in this document reveal different
a tetrahydro-5 H-pyrano[3,2-d]thiazole moiety (Thiatmet G) or substituents than the previous document, e.g. 4-methoxy­
a tetrahydro-4 H-cyclopenta[d]oxazole core a tetrahydro- benzylamino, azetidin-1-yl (52), mono (54), di, or

Figure 13. Representative 6-substituted 2-(azetidin-1-yl)-4-fluoro-3a,4,5,6,7,7a-hexahydrobenzo[d]oxazol-5-ol OGA inhibitors from Alectos Therapeutics and Merck
Sharp & Dohme.

Figure 14. Representative 2-substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
8 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 15. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.

trifluoroethylamino, methyl, ethyl compared to the last pub­ The thiazole ring’s inversion has a marked effect on the
lication. The enzymatic activity of the compounds 48, 50, enzymatic activity, Ki 0.5 nM (51, 3aR/7aS) versus 484 nM
51, and 53 against hOGA is in the sub-nanomolar range.

Figure 16. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 9

(52: 3aS, 7aR), which appears a crucial feature of these structure of 32 molecules and the biological data and intrinsic
derivatives. permeability of seven compounds. Introducing fluorine atoms
The inventors explored this specific chemotype in the fol­ at different positions on the molecule reduces the topological
lowing two applications [90,91]. Figure 12 shows representa­ polar surface area (TPSA). In this way, the inventors envisaged
tive examples which mainly differ at the 6-position of the enhancing the intrinsic and blood-brain permeability,
hexahydrobenzo[d]oxazole or thiazole ring system. The (6S) a recurring theme in the patent applications from Alectos
configuration of the hydroxyethyl group is the preferred one, Therapeutics/Merck Sharp and Dohme.
i.e. the primary hOGA potency increased 10- or 18-fold for Alectos replaced the six-membered cyclohexane ring with
compounds 56 and 58 and 57 and 55, respectively. a tetrahydropyran moiety (WO061927, WO061971 and
Inversion of the 7a and the 3a position from 3aS/7aS to 3aR/ WO129651) [92–94]. They disclosed 21 substituted tetrahy­
7aR results in a 1680-fold decrease in primary potency, 57 dro-3aH-pyrano[3,2-d]thiazoles, and Figure 14 reveals various
versus 59. exemplified molecules. Enlarging the ring size and adding
Figure 13 depicts representative azetidine-1-yl derivatives (polar) substituents on the cyclic group at the two-position
(60–63) of the same scaffold; however, they do not meet the of the thiazole ring resulted in a decrease in inhibitory potency
previous publication’s activity. This document revealed the against hOGA viz. molecules 64–68.
Figure 15 depicts a further elaboration of this tetrahydro-
3aH-pyrano[3,2-d]thiazoles scaffold at the two and the pen­
dant five positions.
The application disclosed the synthesis of 57 compounds
and the biological data of 18 derivatives. The inhibitory activ­
ity against hOGA is in the 0.6–10 nM range, and the various
groups at the pendant five-position have little effect on the
potency, which is clear from Figure 15 (69–74). The most
active compound is derivative 71 (0.6 nM, LE = 0.67).
Changing the group’s nature at position five of the scaffold
into a carbamate did not significantly improve the in vitro
biological activity; compounds 75 and 76. However, the car­
bamate moiety negatively impacted the cellular activity by
almost twenty-fold versus the in vitro activity.
In another application [95], the same scaffold is explored
further by modifying the pyrane ring, i.e. omitting various
hydroxyl functionalities and adding fluorine atoms at different
positions and with specific configurations. Figure 16 shows
some representative examples, and noteworthy are the exam­
Figure 17. Representative close analogues of Thiamet G as OGA inhibitors from ples that possess improved intrinsic permeability resulting in
Alectos Therapeutics and Merck Sharp & Dohme.

Figure 18. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
10 J. M. BARTOLOMÉ-NEBREDA ET AL.

improved cellular activity. The omission of hydroxyl function­ [3,2-d]thiazole-6,7-diol. Changes in the configuration of
alities, the addition of Me-groups, and the replacement of a single hydroxyl from 6S to 6 R while keeping the other
hydroxy groups with fluorine atoms result in improved proper­ stereo centers constant, resulted in a decrease of activity,
ties. Primary activity data enhances by removing the ability of albeit compound 86 is not a matched pair of Thiamet
H-bond donating capability by adding an alkyl group. This, G. Then, adding another methyl group to the amino group
however, did not increase the cell-based activity viz. com­ increased the enzymatic activity 8.5-fold viz. compound 86;
pounds 77 and 79. Compounds that lack the 7-OH group 243,3 nM versus compound 87; 28.6 nM.
are less potent than the corresponding substituted ones, The in vitro activity of the matched pair of Thiamet G,
either with a 7-OH or a 7-F moiety. Replacing hydroxyl groups molecule 88, which has a 7S instead of 7 R configuration,
with F-atoms reduces TPSA, enhances the intrinsic permeabil­ decreased about 10-fold. Remarkably, changing the
ity, and improves the cellular activity, e.g. molecules 80–82 6 R,7 R to 6S, 7S like in compound 89, a direct analogue of
with molecule 82, the most interesting one disclosed in this 86, resulted in the best hOGA enzymatic activity of 0.7 nM in
patent application. Noteworthy is the configurational impact this publication[96].
of the six-position at the tetrahydropyran ring. The (6S)-OH is They disclosed in another application [97] molecules with
about 30-fold more active than the (6 R)-OH, compare mole­ a sulfur atom instead of oxygen or a carbon atom, i.e. tetra­
cule 80 with 83, 5.7 nM and 164 nM, respectively. hydro-5 H-thiopyrano[3,2-d]thiazole ring system. Figure 18
Figure 17 depicts four close analogues of Thiamet G. Subtle depicts representative examples and the biological data of 6
changes in the stereochemical nature of the hydroxyl groups compounds. Incorporation of a sulfur atom resulted in an
at the 6- or 7-position had a significant effect on the in vitro increase of the TPSA compared to an oxygen atom; however,
activity. The full stereochemical descriptors and name of the intrinsic permeability is the reverse order viz. molecule 92;
Thiamet G (90, Figure 18) is (3aR,5 R,6S,7 R,7aR)-2-(ethyla­ 3.7 × 10−6 cm/s versus molecule 90 (Thiamet G); <1.0x10−6
mino)-5-(hydroxymethyl)-3a,6,7,7a-tetrahydro-5 H-pyrano cm/s. The cellular activity, however, is correlating with the

Figure 19. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 11

Figure 20. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.

Figure 21. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.

Figure 22. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
12 J. M. BARTOLOMÉ-NEBREDA ET AL.

primary potency; 90, Ki (hOGA) = 0.4 nM, EC50 = 13 nM versus the cell-based assay than some of the compounds illustrated
92, Ki(hOGA) = 3.0 nM, EC50 = 139 nM, approximately ten-fold in Figure 19.
difference. Another application revealed an exploration of the
The following two applications disclosed tetrahydro- 2-amino group viz. small alkylic groups, amides and ureas
5 H-pyrano[3,2-d]thiazole derivatives, and Figure 19 depicts [101]. The publication described the synthesis of thirty-one
various representative examples [98,99]. The R1 and R2 groups derivatives and the biological activity of 8 examples.
comprise small alkylic moieties such as Me, Et, Pr, cBu, cPent, Figure 21 depicts representative molecules, and the
methoxy(methyl)amino and cyclopropyl methyl groups. These in vitro activity is lower than other derivatives from the
derivatives possess excellent in vitro enzymatic activity in the same scaffold (114–116).
sub-nanomolar range, i.e. 0.12–16.3 nM and the SAR among In another application, WO000084, the Alectos and Merck
the different sub-series is relatively flat. The in vitro potency Sharp & Dohme researchers disclosed 80 compounds, of
correlates well with the cellular activity with a 4- to 30-fold which 17 derivatives revealed interesting SAR. Changing
difference in many examples. An exception is compound 101, the stereochemical nature from (7 R) to (7S) improved the
with a Ki of 0.82 nM and an EC50 of 62.7 nM in the cell-based in vitro potency moderately; 3.6-fold, 1,1 nM versus 0.3 nM,
assay. molecule 117 versus molecule 118, respectively (Figure 22).
Another publication of Alectos Therapeutics and Merck The in vitro activity decreased 886-fold when the hydro­
Sharp & Dohme overlapped with the previous applications, xyethyl group’s stereochemistry inverted from (1S) to (2 R);
albeit with some slightly different derivatives with various compare compounds 117 to 119; 0.3 nM versus 266 nM,
fluorine-containing groups, and Figure 20 depicts some fasci­ respectively. Moreover, changing the hydroxyl group from
nating examples[100]. These very potent inhibitors in the (6 R) to (6S) at the tetrahydro-5 H-pyrano[3,2-d]thiazole core
enzymatic hOGA assay did not translate in high potency in decreased the activity 510-fold; viz. compounds 113 to 120,

Figure 23. Representative radiolabeled substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 13

Figure 24. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.

Figure 25. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
14 J. M. BARTOLOMÉ-NEBREDA ET AL.

1.1 nM versus 562 nM, respectively. The omission of an However, using the data from the application WO169576,
F atom at the 7-position resulted in a 23-fold decrease of different compounds could have been selected because deri­
activity, viz. compound 122 compared to derivative 117. In vatives 129–131 (Figure 23) combine the best values for the
addition to this, removing one hydroxyl group at the 1-posi­ in vitro, cellular, and permeability data reason for the initial
tion also diminished the activity 140-fold, i.e. compound selection remains, therefore, elusive.
117 versus derivative 121 (Figure 22). In February 2014, Alectos published an application contain­
Two applications, WO166654 and WO169576, revealed ing Thiamet G derivatives revealing remarkable differences
O-Glc-NAcase imaging agents labeled with [101]C isotopes between diastereomers [104]. The chirality of the flexible
[102,103]. They disclosed compounds that possess elaborate group attached at the sugar moiety has a significant effect
substitution patterns on the hydroxyl group with linear alkyl on the enzymatic hOGA activity. Figure 24 depicts representa­
groups or (hetero)aromatic moieties. Substitution patterns tive examples.
encompass fluorine atoms, small cycloalkyl groups or more Inversion of the chirality increased the enzymatic activity
polar moieties, whereas the aromatic groups possess small significantly. The difference between the diastereomeric
substituents, at different positions, such as methyl, fluorine, mono-methyl derivatives is smaller than for the dimethyl
methoxy. Both applications do not disclose the PET tracers’ ones; compound 134 versus 135, and derivatives 136/137
data (compounds 123–125). The corresponding non- versus 138/139 (Figure 24). Interestingly, the additional
radiolabeled compounds 126–128 combine moderate to methyl group decreased the PgP efflux ratio by about 7-fold.
good in vitro activity and cellular potency with an intrinsic The intrinsic permeability was for the matched pairs within
permeability exceeding 20 × 10−6 cm/s (Figure 23). a similar range; changing the PSA was in this case, decisive for
less efflux; compare compound 134 (Papp = 18.8x10−6 cm/s,

Figure 26. Representative substituted tetrahydro-3aH-pyrano[3,2-d]thiazole OGA inhibitors from Simon Fraser University.

Figure 27. Representative substituted piperidin-1-yl-methyl-(thiazol-2-yl)acetamide OGA inhibitors from Alectos Therapeutics and Merck Sharp & Dohme.
EXPERT OPINION ON THERAPEUTIC PATENTS 15

Figure 28. The resemblance of piperidin-1-yl-methyl-(thiazol-2-yl)acetamide OGA inhibitor from Alectos Therapeutics and Merck Sharp & Dohme and the PET-tracer
of Eli Lilly.

PgP efflux = 21) to 137 (Papp = 28x10−6 cm/s, PgP secondary and tertiary amides at the same position resulted
efflux = 3.5). in inactive compounds.
This application also revealed compounds possessing smal­ Figure 26 represents the same central scaffold’s continua­
ler groups, such as depicted in Figure 25. These derivatives tion, namely, the tetrahydro-5 H-pyrano[3,2-d]thiazole core.
displayed noteworthy behavior in the enzymatic activity upon The application disclosed 97 examples, out of which three
small molecular changes. Changing the methyl group to an have biological data (compounds 145–148)[105]. However,
ethyl one decreased the primary activity 30-fold (compounds the focus is on the in vivo evaluation of two compounds
140 versus 141). Linear moieties such as an alkyne of a cyano Thiamet G (NAG-AE) and NAG-Bt (Figure 26). Increasing
functionality resulted in less active compounds, i.e. 837 nM doses of NAG-Bt via intravenous (one time 0–250 mg/kg) tail
and 601 nM for derivatives 142 and 144. Interestingly, chan­ vein injection or oral administration (100 mg/kg/day for five
ging the CN-group into a CH2CN group decreased the activity days) elevated O-GlcNAc levels in the brain and muscle,
more than 550-fold, albeit no matched pairs, 1.08 nM versus respectively. IV administration of 50 mg/kg NAG-EA increased
601 nM, compounds 143 and 144, respectively. Installing O-GlcNAc levels in cardiac tissue.

Figure 29. Representative Thiamet G analogues from Merck GMBH.


16 J. M. BARTOLOMÉ-NEBREDA ET AL.

Furthermore, NAG-Bt and Thiamet G blocked phosphoryla­ compounds have a suitable safety profile for the studies
tion sites which are involved in the toxic self-assembly of tau. performed.
Moreover, both compounds reduced neurofibrillary tangle for­ In WO106254 Alectos Therapeutics and Merck Sharp &
mation in transgenic P301LJNPL3 mice and the tau phosphor­ Dohme disclosed compounds based on an N-(thiazol-2-yl)acet­
ylation levels, albeit through different dosing regimens and amide scaffold[106]. The publication revealed the enzymatic
doses; NAG-Bt at 100 mg/kg/day via food (week 1–15) and hOGA data of 54 compounds and interesting SAR. Figure 27
then 1000 mg/kg/day via drinking water (week 16–32); depicts some compelling examples of this class of hOGA
Thiamet G at 500 mg/kg/day via drinking water (week 16– inhibitors. Compounds 148–155 reflect the effect on the enzy­
32). An eight to nine months repeat-dose toxicology in vivo matic activity of the spatial orientation of the double bond to
evaluation of both NAG-Bt and Thiamet G revealed that both the chiral center at the piperidine. The absolute S,

Figure 30. Representative N-substituted phenylpiperidine OGA inhibitors from Merck GMBH.

Figure 31. Representative 4-substituted piperidin-1-yl-methyl-(thiazol-2-yl)acetamide OGA inhibitors from Merck GMBH.
EXPERT OPINION ON THERAPEUTIC PATENTS 17

Z-configuration resulted in the highest primary activity for 2.2. Merck GMBH
multiple compounds disclosed in this publication.
In February 2013, scientists at Merck Patent GMBH published
This document revealed another exciting compound, and
an application that disclosed 105 Thiamet G analogues as
Figure 28 shows the similarity of this compound 156 and the
hOGA inhibitors[107]. All the analogues posess the central
Eli Lily PET-tracer [18F]-LSN3316612.
pyrano[3,2-d][1,3]thiazole core (Figure 29). They encompass
variations introduced at the 2-amino substituent of the

Figure 32. R of N-4 substituents on the N1-arylpiperazine core from Asceneuron S.A.

Figure 33. Examples of N1 substituents on the N4-(1-arylethyl)-piperazine core from Asceneuron S.A.
18 J. M. BARTOLOMÉ-NEBREDA ET AL.

dihydrothiazole ring, at the hydroxyl substituents and the O-GlcNAcylated cellular protein levels were measured using
5-position of the tetrahydropyranyl ring. This patent reported CTD110.3 antibody and determined by the ELISA technique.
the enzymatic and cellular activity of the OGA inhibitors. The Four compounds demonstrated hOGA inhibitory activity
cellular assays utilized rat B35, rat PC-12 and human Sh-SY5Y below 100 nM in both enzymatic and cellular assays, e.g.
cells, expressing endogenous OGA levels. Increases in 157, 159, 160 and 161 (Figure 29). All of them contained

Figure 34. Representative dihydrobenzofurane OGA inhibitors from Asceneuron S.A.

Figure 35. Examples of piperazine OGA inhibitors from Asceneuron S.A.


EXPERT OPINION ON THERAPEUTIC PATENTS 19

a 2-aminoethyl substituent on the dihydrothiazole ring. responsible for the loss in potency since 3-F piperidine analo­
Substituents at the 5- position of the tetrahydropyranyl ring gue 170 maintained the hOGA enzymatic activity.
included chloromethyl (156), 1-hydroxy-3-arylpropyl (158) and Replacement of the piperidine core by piperazine resulted in
methyltriazolyl analogues, e.g. 159–161. Interestingly, the decreased enzymatic and cellular OGA inhibitory activity
more sterically hindered trifluoromethylphenyl ethanol analo­ (Figures 32, 171).
gue 160 did not show any inhibitory effect in the cellular
assay. Most likely, the topological polar surface area of this
derivative, 150 Å2 for compound 160 vs 130 Å2 for derivative 2.3. Asceneuron S.A
159, hampered the cellular permeability. In contrast, the
Asceneuron S.A., a spin-off of Merck Serono’s Alzheimer’s drug
hydroxy substituent at C5 of the tetrahydropyranyl ring of discovery portfolio and research group in Switzerland[109],
molecule 158 could be involved in internal H-bonding, thus
filed 12 patents, in which they disclosed a variety of 1,4-dis­
displaying a reduced TPSA (119 Å2) and therefore improved
ubstituted piperidyl or piperazinyl analogues as glycosidase
cell permeation. inhibitors. Thus, in 2016, Asceneuron S.A. revealed a new ser­
In 2014, researchers at Merck GMBH filed a second applica­
ies of glycosidase inhibitors, most of which featured an N1-aryl
tion disclosing 4-substituted piperidin-1-yl-methyl-(thiazol-
-N4-(1-arylethyl)-piperazine common central scaffold[110]. This
2-yl)acetamide derivatives as glycosidase inhibitors[108]. application disclosed 202 compounds, of which 63 demon­
A total of 65 compounds were exemplified, out of which 26
strated potent OGA enzymatic activity, with hOGA IC50
analogues revealed hOGA IC50 < 200 nM in the enzymatic
< 50 nM. Of these compounds, the preferred N-4 substitution
inhibition assay, whereas four analogues demonstrated an seemed to be bicyclic heterocycles, either exclusively aro­
EC50 < 200 nM cell inhibition assay utilizing B35 rat neuroblas­
matic, such as quinoxaline (172), or benzothiazole (173), or
toma cells. Most examples carried a common piperidine core,
containing partially saturated rings such as dihydrobenzofur­
N-substituted by a 5-methylenethiazol-2-acetamide moiety anes, benzoxolane or benzodioxane, as exemplified in
(Figures 30, 162). Interestingly, methylation of the acetamide
Figure 32, molecules 174–176.
moiety in this compound, or its replacement with an amine or
Among the preferred substituents on the piperazine scaf­
sulfonamide moiety, derivatives 163–165, lead to a significant fold, selected examples include N1-substituted pyrimidines
loss of enzymatic activity. Aliphatic substituents at 4-position
(177–179, Figure 33) and pyridines or bicyclic heterocycles,
of the piperidine core, such as benzyl, phenethyl or cyclohexyl
e.g. tetrahydrothiazolopyridines 180–181.
derivatives 166–168 (Figure 31), were beneficial for OGA inhi­ The publication claimed that the preferred compounds in
bitory activity in both assays. The introduction of a quaternary
this patent, 182–185, depicted in Figure 34, demonstrated
center at the 4-position of the piperidine core was detrimental
acceptable drug-like properties, such as metabolic stability in
for activity, as illustrated by derivative 169. In this case, it liver microsomes, stability toward oxidation or cellular perme­
seemed that steric rather than electronic effects were
ability. Furthermore, these compounds showed high biological

Figure 36. Representative piperazine OGA inhibitors from Asceneuron S.A.


20 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 37. Examples of 2,3-dihydrobenzofuran-6-ethylpiperazine OGA inhibitors from Asceneuron S.A.

Figure 38. Representative [1-(benzothiazol-5-yl)ethyl]piperazine OGA inhibitors from Asceneuron S.A.

activity, as determined by the level of OGlcNAcylation of the achieved excellent hOGA enzymatic similar to the tetrahy­
total proteins measured in rodent brain extracts. drothiazolo[5,4-c]pyridine analogue 182.
Later patents from Asceneuron S.A. disclosed additional Patents filed in the same year disclosed synthetic methods
analogues of the piperazine derivatives described above for the preparation of enantiomerically enriched or pure (S)-
[111]. Thus applications from August 31st, 2017 (WO144633 [1-(benzodioxol-5-yl)ethyl]piperazine analogues, with com­
and WO144639) claimed 287 compounds with the N1-aryl-N4 pounds like 173 (Figure 32), 177 (Figure 33), 182 and 185
-(1-arylethyl)-piperazine as their common central scaffold, with (Figure 34) being among the specific examples[112]. Other
many heterocycles disclosed as N1- and N4- substituents. patents from 2017 claim acid addition salts and polymorphs
From the exemplified analogues, 56 compounds demon­ of the piperazine analogues discussed above[113].
strated hOGA enzymatic activities at IC50 < 50 nM. Most of Furthermore, the physicochemical data for compound 185,
these more potent analogues contained dihydrobenzofuranes both as free base and acid salts, are provided, such as visual
as aromatic N4-substituents, combined with various N1- aqueous solubility, DSC thermogram, X-ray powder diffraction
piperazine substituents, either monoaryls or bicyclic hetero­ pattern, Raman spectra, STA thermogram, NMR spectra.
cycles (Figure 34). In August 2018, scientists at Asceneuron S.A. disclosed two
Figure 35 depicts other N4-substituents that demonstrated additional patents of N,N’-1,4-disubstituted piperazines[114].
high potency in the enzymatic hOGA assay, including quinox­ The first patent (WO153507) exemplified 21 compounds con­
alines, benzothiazoles and naphthyridines, and compounds taining a common 2,3-dihydrobenzofuran-6-ethylpiperazine
186–193. The tetrahydropyrido[4,3-d]pyrimidine analogues substructure substituted with mono-and bicyclic heterocycles
EXPERT OPINION ON THERAPEUTIC PATENTS 21

Figure 39. Representative annulated piperidine OGA inhibitors from Asceneuron S.A.

Figure 40. Representative pyrrolidine OGA inhibitors from Asceneuron S.A.

at the N-1 position, among which phenyl, thiadiazoles and In a more recent patent from February 2019, researchers at
pyrimidines. Nine compounds demonstrated IC50 < 50 nM in Asceneuron S.A. reported glycosidase inhibitors in which the
the OGA inhibition assay, and Figure 36 and Figure 37 depicts central piperidine or piperazine core was annulated into
representative derivatives. They claimed that introducing a bicyclic ring system, demonstrated in Figure 39[116]. The
a sulfoximine moiety, as in compound 201, was beneficial publication reported potency ranges for 21 compounds out of
for decreasing the plasma protein binding (PPB) in this series. 162 exemplified analogues only. Of these 21 one molecules,
The second patent application [115] exemplified 71 sul­ seven analogues showed hOGA activity between 50–200 nM
foximine analogues of molecule 201. Apart from the in the enzymatic inhibition assay. The 4,5,6,7-tetrahydro-
potency range, the respective free fractions in mouse 1 H-pyrazolo[4,3-c]pyridine derivatives demonstrated moder­
plasma were also reported. The exemplified compounds ate enzymatic activity, as shown in Figure 39. Benzyl substitu­
preserved the piperazinyl central core; however, they ents attached to the central pyrazole core, as in molecule 210,
described a broader range of substituents at both N1 and showed similar a potency range as the compounds having
N4 of the piperazine moiety. These 45 compounds demon­ aromatic substituents directly connected to the pyrazole cen­
strated IC50 < 50 nM in the hOGA enzymatic assay. These tral core (209).
included examples such as 202–204 (Figure 38), having In February 2020, scientists at Asceneuron S.A. disclosed
a benzothiazolyl substituent at N4. three different series OGA inhibitors containing pyrrolidines,
22 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 41. Representative tetrahydro-benzoazepines OGA inhibitors from Asceneuron S.A.

tetrahydro-benzoazepines and spirocyclic piperidines as (Figure 41). Other groups with promising enzymatic activity
a central core. Thus, a first patent described 465 compounds are partially saturated bicyclic heterocycles, such as 2,3-dihy­
having a central pyrrolidine core[117]. The document dis­ drobenzofuranes or 2,3-dihydrofuro[3,2-b]pyridines, e.g. 223
closed the enzymatic activity for 94 compounds, of which and 6,6-bicyclic heterocycles, quinaxolines, e.g. 224.
four analogues demonstrated IC50 < 50 nM in the hOGA Changing the spatial orientation of the two parts of the mole­
enzymatic inhibition assay. The majority of the exemplified cule resulted in a decrease in activity; compare IC50 < 50 nM
compounds contained a pyrrolidine central core with versus IC50 = 200–100 nM, derivatives 221 (2,3,4,5-tetrahydro-
a 2,3-dihydrobenzo[b][1,4]dioxine moiety or 2,3-dihydrofur­ 1 H-benzo[d]azepine) and molecule 222 (2,3,4,5-tetrahydro-
ane at the 2-position of the pyrrolidine, e.g. 211–216 in 1 H-benzo[c]azepine), respectively. Changing the position of
Figure 40. The central core connected various aromatic a Cl-atom to the aromatic ring of the tetrahydro-1 H-benzo[d]
groups via a methylene linker, and Figure 40 depicts some azepine scaffold led to a decrease in enzymatic hOGA activity;
representative examples. The pyrazol-4-yl-pyridine (212, the 6-chloro-7-methoxy derivative versus 7-chloro-8-methoxy,
212) and pyridine-2-yl-pyrimidine derivatives (213) demon­ compounds 218 and 219, respectively.
strated IC50 values below 50 nM. Replacing one aromatic In the third patent [119] from February 2020 were disclosed
ring with an aliphatic substituent, as in 214, or fusing the 226 compounds containing spirocyclic piperidines as the cen­
aromatic ring into a bicyclic heterocycle, as in 215, led to tral core. The patent application revealed pharmacological
a decrease in potency. Moreover, changing the orientation data for 58 compounds, of which seven demonstrated IC50
of the aromatic moieties connected to the N atom of the < 50 nM in the enzymatic hOGA assay. The majority of the
pyrrolidine ring led to a significant decrease in potency, i.e. exemplified compounds contained a 1,3,8-triazaspiro[4,5]
molecule 216. decane-2,4 dione core, connected at the N-8 piperidine with
In a second patent from February 2020, scientists at thiazoles, benzothiazoles or dihydrobenzofuranes via
Asceneuron S.A. disclosed 2,3,4,5-tetrahydro-1 H-benzo[d]aze­ a methylene linker. Many substituents were introduced at
pine derivatives as potent OGA inhibitors[118]. The document the N-3 position of the imidazolidine-2,4-dione, including
revealed 363 exemplified compounds and the in vitro data of alkyl chains, substituted phenyl and heteroaromatic rings.
58 derivatives. Of these, 33 compounds were potent deriva­ Among these, aromatic substituents linked either directly or
tives, demonstrating IC50 < 50 nM activities in the hOGA via a methylene linker to N-3 were beneficial for potency, e.g.
enzymatic assay. Particularly, the 2,3,4,5-tetrahydro- 226 and 227 versus 225, depicted in Figure 42. Additional
1 H-benzo[d]azepine scaffold, connected at N-1 position via substitution at N-1 decreased the potency, as exemplified by
a methylene linker to aromatic groups, were interesting com­ molecule 228. Variations of the central core included spiro
binations. It included 5,6- bicyclic heterocycles, especially [indoline-3,4ʹ-piperidin]-2-one, diazaspiro[4.5] decane,
benzo[d]thiazole derivatives, e.g. molecules 217–221
EXPERT OPINION ON THERAPEUTIC PATENTS 23

Figure 42. Representative spirocyclic piperidine OGA inhibitors from Asceneuron S.A.

2,8-diazaspiro[4.5]decan-1-one and 1,3,8-triazaspiro[4.5]decan- 2.4. Janssen Pharmaceuticals


2-one derivatives 229–232, and led to a decrease in potency.
Janssen published 11 patent applications that disclosed three
Asceneuron S.A. published a patent application in
main OGA chemotypes, i.e. saturated cyclic amines, spirocyclic
August 2020, and it described 165 fused tetrahydro-azepine
diamines and azaindoles. Thus, three patent applications from
derivatives as OGA inhibitors[120]. The document disclosed
2018 described various 5- and 6-membered heterocyclic
the pharmacological data for 17 compounds. The tetrahydro-
amine series as OGA inhibitors. The first application discloses
azepine scaffold was fused with structurally diverse saturated
64 examples of 5- and 6-membered cyclic amines substituted
and aromatic 5-membered rings and eleven OGA inhibitors
at N-1 either with a heteroaryl sulfonyl or, in most cases,
with IC50 values below the 50 nM range. Figure 43 shows
benzyl-like radicals; the 2-acetamidothiazole-5-yl radical is
interesting examples. The N-6 position of the tetrahydroaze­
the most frequently used moiety[121]. All examples described
pine ring was connected via a methylene linker to aromatic
in this patent application present a 6,5-heterobicyclic substi­
substituents such as benzothiazole 233, N-(5-methylthiazol-
tuent attached through the 5-membered ring to the C-3 posi­
2-yl)acetamide 234 and dihydrobenzofurane 235. Changing
tion of the central heterocyclic scaffold, in most cases,
the way the heteroaromatic moiety is linked to the 7-mem­
a piperidine or pyrrolidine ring. Figure 44 depicts representa­
bered rind and thus the orientation of this aromatic group
tive examples 237–240. A second publication disclosed
decreased the hOGA potency; compare molecules 234
6.5-heteroaromatic bicyclic systems direct-linked or connected
(2-methyl-2,4,5,6,7,8-hexahydropyrazolo[3,4-d]azepine) and
via a spacer to the C-3 position of piperidine, morpholine or
235 2-methyl-2,4,5,6,7,8-hexahydropyrazolo[4,3-c]azepine
piperazine as OGA inhibitors[122]. Out of the 109 examples,
albeit no matched pair.
100 derivatives contain the 2-acetamidothiazole group
24 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 43. Representative fused tetrahydro-azepine OGA inhibitors from Asceneuron S.A.

Figure 44. Representative 6,5-heterobicyclic OGA inhibitors from Janssen.


EXPERT OPINION ON THERAPEUTIC PATENTS 25

Figure 45. Representative monoheterocyclic OGA inhibitors from Janssen.

attached to the N-1 atom of the central core. The patent activities in the hOGA in vitro assay and cell-based assay for
application disclosed enzymatic OGA activity data for all com­ a subset of compounds; the most potent derivative is 249.
pounds and cell-based assay data for representative examples. Additional representatives containing a benzothiazole (250) or
Figure 44 shows representative examples (241–244) from this benzodioxane (251) N-1 substituents are also shown in
application. Figure 46. More recently, Janssen scientists have reported
Interestingly, sulfonamide examples described containing a more extensive characterization of one of the compounds
a morpholine (243) or piperazine (244) central scaffold dis­ disclosed in this patent application, confirming it as a potent
played good inhibitory values in the OGA enzymatic assay metabolically stable and orally bioavailable OGA inhibitor with
while not showing activity in the cell-based assay at the high­ modest central penetration[125].
est concentration tested. In 2019 Janssen scientists published nine new patent
A third application described 171 examples of piperidines applications disclosing novel families of OGA inhibitors. On
and pyrrolidines substituted at C-3 by a monocyclic hetero­ December 26th, five applications revealed mostly piperi­
cycle[123]. The application described substituted N1-linked dines and pyrrolidines substituted at position C-3 with
heteroaryl sulfonyl or benzyl-like radicals. The groups attached a great variety of aromatic heterocycles and bearing at
at the C-3 position of the piperidine or pyrrolidine, via a direct N-1 fully and partially saturated heteroaromatic rings con­
bond or a methylene spacer, are monocyclic aromatic nected through a methylene spacer optionally substituted
heterocycles. with a small alkyl group [126–130]. Thus, an application
The 2-acetamidothiazole-5-yl and 2,6-dimethylpyridin-4-yl disclosed 198 examples where the central scaffold is
radicals are the most frequently described heterocycles in a piperidine ring. The piperidine ring was substituted at
substituents at N-1 and C-3 positions. Compounds 245–248 the C-3 position by a 6-membered heteroaryl substituent
in Figure 45 are among the most potent examples from this via a linker of variable length (0–2 atoms All examples
patent application. from this patent application contain a partially saturated
The Janssen scientist reported novel spirocyclic diamines heterobicyclic ring connected through a methylene linker
with OGA inhibitory activity in 2018[124]. This patent applica­ to the N-1 position of the central scaffold. Figure 47
tion disclosed 69 examples where a 2-acetamidothiazole-5-yl depicts compounds (253–256) displaying suitable OGA
group connected via a methylene linker to a 2,7-diazaspiro inhibitory enzymatic activities in the 1.3–10 nM range.
[4,4]nonane, which is the common substructure to 45 mole­ Moreover, the patent application disclosed ex vivo recep­
cules. Sixty-seven derivatives have a six-membered monocyc­ tor occupancy data of the compounds 255 and 257. Oral
lic heteroarene directly linked to the N-2 position of the spiro- administration of 25 mg/kg of compound 257 achieved
bicyclic central scaffold. They disclosed a set of eleven N-1 nearly full occupancy (Occ. 93%) in the forebrain at the
alternative bicyclic systems – including benzodioxane, pyrido­ 24 h post-dose time point.
dioxane, quinoline, indazole and benzothiazole – to the acet­ Piperidine OGA inhibitors containing 6,6-bicyclic aromatic
amidothiazole group. Figure 46 displays the inhibitory heterocycles at N-1 position were published in a patent
26 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 46. Representative spirocyclic diamine OGA inhibitors from Janssen.

Figure 47. Representative piperidine OGA inhibitors containing a partially saturated heterobicyclic N-1 substituent from Janssen.
EXPERT OPINION ON THERAPEUTIC PATENTS 27

Figure 48. Representative piperidine OGA inhibitors containing an aromatic 6,6-heterobicycle as N-1 substituent from Janssen.

Figure 49. Representative piperidine OGA inhibitors containing an aromatic 6,5-heterobicycle as N-1 substituent from Janssen.

application disclosing ten examples. All compounds described general pyrrolidine-containing examples are less potent than
have a 2,6-dimethyl-4-(piperidin-3-ylmethyl)pyridine as their corresponding piperidine matched-pairs. Pyrazole, pyri­
a common substructure connected to a quinoline, naphthyr­ dine and thiophene, combined with a 2,6-dimethyl-4-(piper­
idine or quinoxaline ring via a methylene spacer. The most idin-3-ylmethyl)pyridine, are among the heterocycles leading
potent examples from this patent application are compounds to more potent OGA inhibitors in the set of 165 examples
257 and 258 (Figure 48) containing a 1,5-naphthyridine and disclosed in this case. Figure 50 depicts representative exam­
1,8-naphthyridine ring, respectively. ples 263–266.
The Janssen researchers disclosed in 2019 109 OGA inhibi­ Janssen scientists also published a patent application dis­
tors containing piperidine and pyrrolidine scaffolds. As in pre­ closing 129 pyrrolidines bearing a partially saturated hetero­
vious patent applications, these piperidine and pyrrolidine bicyclic ring connected through a methylene linker to the N-1
scaffolds were substituted at the C-3 position with position. In all examples from this case, the methylene linker is
a monocyclic heteroaromatic ring connect via a 0–2 atom also substituted with a methyl group. In most compounds,
spacer. The methylene spacer connects a 6,5-bicyclic aromatic a methylene or oxygen spacer links the C-3 position of the
heterocycle to the N-1 of the central scaffold in all examples. pyrrolidine to a 6-membered heteroaromatic ring. Examples
Representative compounds showing suitable inhibitory activ­ displaying good OGA inhibitory activity are compounds 267–
ities in the OGA enzymatic and cellular assay are compounds 270 in Figure 51.
259–262 (Figure 49). Derivative 262 showed negligible occu­ Piperidines substituted at the C-4 position by
pancy of the OGA enzyme 24 hours post-dose following a 6-membered heteroaromatic ring connected via a 1- or
25 mg/kg oral administration. 2-atom spacer were published on December 26th in a patent
Monocyclic aromatic heterocycles were also reported as application describing 169 examples[131]. In most cases, they
suitable substituents at N-1 in a patent application disclosing comprise substituted pyridine and pyrimidine rings linked
a series of C-3 substituted piperidines and pyrrolidines. In through an oxygen atom to the C-4 position of the piperidine
28 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 50. Representative piperidine OGA inhibitors containing an aromatic monocycle as N-1 substituent from Janssen.

Figure 51. Representative pyrrolidine OGA inhibitors containing a partially saturated heterobicyclic N-1 substituent from Janssen.

ring. This application also disclosed fully unsaturated aromatic Finally, two additional patent applications from Janssen
6,5-bicycles and partially saturated hetero bicycles connected disclosed substituted pyrrolo[3,2-c]pyridine compounds as
via a methylene linker to the N-1 atom of the piperidine core; OGA inhibitors on 26 December 2019. In both publications,
in many examples, the methylene linker contained a methyl the pyrrolo[3,2-c]pyridine core contains an aniline or an
group. Figure 52 displays potent OGA inhibitors from this amino-heterocycle at the C-4 position of the central scaffold.
patent application (271–274). They used an ex-vivo protocol Many of the examples comprise an ortho-ortho disubstitution.
to assess the forebrain’s occupancy after 24 h post- In the first patent application, 110 out of the 114 examples
administration orally of compounds 271, 273 and 274. given have at the N-1 position an N,N’-dimethylacetamide
Gratifyingly, compounds 271 and 273 demonstrated full occu­ function[132]. The derivatives show, in general, very high
pancy of the target at 25 mg/kg dose. activities in the OGA enzymatic and cellular assays.
EXPERT OPINION ON THERAPEUTIC PATENTS 29

Figure 52. Representative 4-substituted piperidines OGA inhibitors from Janssen.

Compounds 275–278 are the most potent examples disclosed In December 2019 a third application published [136] covering
in this application (Figure 53). The second application revealed a small set of 4 6-[1-[4-(5-methyl-1,3,4-oxadiazol-2yl)-1-piperidyl]
209 compounds[133]. Substituents attached to the N-1 posi­ ethyl]-2,3-dihydrofuro[2,3-b]piridine derivatives. The compounds
tion of the pyrrolo[3,2-c]pyridine are, in most cases, lipophilic only differ in the presence or absence of a fluorine atom at
alkyl groups optionally substituted with fluorine atoms. Also, position 5 of the 2,3-dihydrofuro[2,3-b]pyridine motif, and both
substituted alkyl chains with cyclic ethers, such as oxetane, possible individual enantiomers are claimed (Figure 55, 286 and
were exemplified. Compounds in this patent application are 287). Whilst the presence or absence of the fluorine substitution
potent OGA inhibitors in the enzymatic assay, with 94 exam­ has an almost negligible impact on the OGA inhibitory potency,
ples displaying a pIC50 > 8. Representative compounds 279– both (-) enantiomers show significantly increased potency (~
283 from this patent application (Figure 53) are among the 1 nM) vs the corresponding (+) enantiomers.
most potent OGA inhibitors described by Janssen to date. In February 2020 another Eli-Lilly application [137] claimed
a N-[5-[[(2S,4S)-4-(5-cyanopyrazin-2-yl)oxy-2-methyl-l-piperidyl]
methyl]-4-fluoro-thiazol-2-yl acetamide (Figure 56, 288) and its
2.5. Eli Lilly and Company diastereomers, as OGA inhibitors. This compound is a close ana­
Researchers from Eli Lilly published six patent applications logue of 284 and 285 (Figure 54) that contain a cyanopyrazine as
between 2018–2020. Thus, in August 2018[134], they claimed left-hand side heteroaryl instead of a 5-methyl-1,2,4-oxadiazole.
N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-1,2,4-oxadiazol- The biological data reported molecule 288 as a sub-nanomolar
3-yl)methoxy]-1-piperidyl]methyl]thiazol-2-yl]acetamide 245 OGA inhibitor (IC50 = 0.343 nM) in an enzymatic assay.
(Figure 54), and its diastereomers, as OGA inhibitors. They Eli Lilly scientists claimed six 6-fluoro-2-methylbenzo[d]
measured the activity of molecule 245 in a biochemical thyazol-yl analogues in another patent application in
assay that utilized fluorescein di-N-acetyl-β-N-acetyl-D gluco­ March 2020 [138]. Instead of central piperidine, common
saminide as a fluorogenic OGA substrate. It amounted to an to all previous Eli Lilly’s application, the reported com­
IC50 of 2.36 nM. The whole-cell assay, which utilized TRex-293 pounds contain a 2-methylpyrrolidine as the central scaf­
cells expressing P301S-1N4R tau protein, revealed an inhibi­ fold. As a left-hand side motif, all the said compounds
tory OGA activity of 245 of 21.9 nM (Figure 49). possess an acetylpyrrolidine fused to either a pyrimidine
A second patent published in November 2018 [135] or a pyridine (different isomers) heterobicyclic system con­
claimed a close analogue of 284. Instead of the 5-methyl- nected to the central pyrrolidine through an oxygen linker.
1,2,4-oxadiazol-3-yl derivative 284, they disclosed a 5-methyl- The described compounds show OGA inhibitory activity
1,3,4-oxadiazol-2-yl group (Figure 54, 285), and its diastereo­ below 1 nM in an enzymatic assay with exemplary com­
mers, as OGA inhibitors. This compound demonstrated OGA pound 289 as the most potent molecule.
inhibition in the enzymatic and cellular assays, having an IC50 Finally, in a patent from 2020 (WO 2020/028141) [139],
= 2.13 nM and 22.6 nM, respectively. researchers from Eli Lilly claimed that combination therapy
30 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 53. Representative pyrrolo[3,2-c]pyridine OGA inhibitors from Janssen.

Figure 54. Preferred Eli Lilly OGA inhibitors.

of compounds 284 or 285 with an engineered anti-tau recombinant full-length hOGA enzyme and 4-MUGlcNac sub­
antibody described in the patent might result in the reduc­ strate. The OGA inhibitors contained di- and tri-substituted
tion of tau pathology in-vivo. cyclic amines as the common central core, having the 2-acet­
amidothiazole substituent connected via a methylene linker at
the N-atom of the pyrrolidine or piperidine ring. A flexible
methylene function at the C-3 or C-5 position, dependent on
2.6. Biogen MA INC
the group’s priority, links various heteroaromatic groups to the
In September 2019, researchers at Biogen filed a patent appli­ cyclic amines. Figure 57 shows the selected analogues. Several
cation in which they disclosed 175 OGA inhibitors[140], vary­ 5- and 6- membered (hetero)aromatics and bicyclic hetero­
ing from sub-nanomolar (<1 nM) to micromolar (>20 µM) cycles result in excellent in vitro hOGA activity, i.e. molecules
ranges in an enzymatic biochemical assay which utilized 290, 291, 297, 298, and 299. Introducing an additional bulky
EXPERT OPINION ON THERAPEUTIC PATENTS 31

Figure 55. Representative 2,3-dihydrofuro[2,3-b]pyridine OGA inhibitors from Eli Lilly.

Figure 56. Selected OGA inhibitors from Eli Lilly.

Figure 57. Selected examples of OGA inhibitors from Biogen.


32 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 58. Selected pyrrolidine OGA inhibitors from Biogen.

substituent at the C-2, as in 292, lead to a decrease in potency, of the rat hepatic blood flow Qh) are reported for a large
IC50 = 270 nM, whereas a methyl group (compound 293) had subset of examples. In this application, the described OGA
a 29-fold improve in enzymatic activity. The addition of two inhibitors contain either a 3-heteroaryloxy-piperidine or pyr­
fluorine atoms to the 3- or 4-position of the piperidine ring rolidine as central core connected via a methylene linker to
significantly impacted the enzymatic activity, viz. molecule the same 2-acetamidothiazole motif as in the previous appli­
295, IC50 = 12 nM, molecule 294, IC50 = 450 nM, respectively. cation. Figure 58 shows representative molecules and rele­
The most active compounds from the patent application are vant SAR. Thus, of the compounds containing
the disubstituted pyrrolidines 298 and 299 (Figure 57). a 5-methoxypyridine, a substituent highly represented, com­
Biogen described, in a second application, published in pound 300 is among the most potent, with low efflux poten­
March 2020, 166 new OGA inhibitors[141]. The compounds tial and moderate metabolic stability. A methyl at position 2
encompass highly potent hOGA inhibitors with several exam­ of the pyrrolidine and a specific stereochemistry seem crucial
ples showing sub-nanomolar (<1 nM) enzymatic activity and for potent OGA inhibitory activity. The deletion of the
single-digit nanomolar activity in a cell-based OGA-tau MSD 2-methyl or changes in the pyrrolidine stereochemistry
assay. Additionally, MDR1-MDCK efflux ratio (B-A/A-B) and resulted in a significant drop in potency, i.e. compounds
metabolic stability in rat liver microsomes (expressed as % 301, 302 and 303. Whilst replacing the pyrrolidine with
EXPERT OPINION ON THERAPEUTIC PATENTS 33

Figure 59. Selected OGA inhibitors from Biogen.

Figure 60. Selected 4-heteroaryloxy-2-methylpyrrolidine OGA inhibitors from Biogen.

Figure 61. Selected 4-heteroaryloxy-2-methylpyrrolidine OGA inhibitors from Biogen.

piperidine or introducing a fluorine atom on the 2-acetami­ Finally, an analogue of compound 300, a 13C radiolabeled
dothiazol had an almost negligible impact on the com­ on the pyrimidine, is described, and this modification also
pounds’ activity, those modifications resulted in doesn’t affect the molecule’s properties.
a significant decrease in metabolic stability (304 and 305, Biogen claimed, in a subsequent application published
respectively). Additionally, several deuterated analogues of in June 2020, similar compounds to the previous ones. The
300 are reported, although this modification had no overall central piperidine or pyrrolidine was replaced by morpho­
effect on the molecules’ properties, e.g. molecule 306. line, piperazine, oxazepane or diazepane[142]. The right-
34 J. M. BARTOLOMÉ-NEBREDA ET AL.

Figure 62. Selected examples of pyrrolidine derivatives as OGA inhibitors from Summit Corporation.

hand side 2-acetamidothiazole motif and the left-hand side 4-heteroaryloxy-2-methylpyrrolidine connected again
aryl or heteroaryl, both connected to the central core by through a methylene spacer to a 2-acetamidothiazole. In
small linkers, are maintained. Figure 59 details the most this case, the document revealed that the left-hand side
active compound of this application, compound 308, and comprised many diverse 5,6 and 6,6 bicyclic heteroaryl sys­
relevant SAR. Thus, the introduction of a second methyl tems. The claimed compounds are highly potent OGA inhi­
substituent on the pyridine in 270 decreased potency by bitors in an enzymatic biochemical assay, and more than
6-fold versus 309. Replacement of the morpholine by two-thirds of the described compounds show sub-
piperazine or methyl piperazine reduced the in vitro activ­ nanomolar (< 1 nM) potencies. Figure 60 shows representa­
ity 5-fold, e.g. molecules 310 and 311. The additional tive compounds from this application (315–317).
deletion of the methyl at the piperazine position Finally, the last patent application from Biogen published
decreased the in vitro potency significantly. Central ring in September 2020 [144] and describes 43 azetidinyl-
expansion to either oxazepane or diazepane also reduced containing OGA inhibitors. In contrast to the previous
the enzymatic activity, with 314 being the most promising cases, using a smaller azetidine instead of piperidine or
analogue but still 10-fold less potent than 308. Finally, the pyrrolidine as central core resulted in an overall slightly
right-hand side spacer lengthening also resulted in less potent OGA inhibition, and the reported potencies
reduced potency (314). range between 10 and 1500 nM. A 2-acetamidothiazol is
Another application described 174 Bicyclic ethers, pub­ constant as a substituent on the azetidine nitrogen, and
lished in August 2020 [143]. Although Biogen researchers different left-hand side heteroaryls (both monocyclic or
claim both piperidine and pyrrolidine as a central scaffold, bicyclic) are connected to the 3-position of the azetidine
all the exemplified compounds contain a (2S,4 R)- core through a 1 or 2 atom linker of varying nature.
EXPERT OPINION ON THERAPEUTIC PATENTS 35

Figure 61 depicts the most potent compounds 318–320. In from Asceneuron S.A. and LY-3,372,689 from Eli Lilly) that
analogy with some of the previous cases, introducing have progressed in Phase I clinical trials so far. Although
a methyl substituent at position 2 of the azetidine core OGT and OGA manage the O-GlcNAcylation cycle, and OGA
seems to impact potency (i.e. 318 versus 320) positively. is a highly ubiquitous enzyme with many client proteins,
all the data confirms a promising benign profile for OGA
inhibition after single and multiple ascending doses in
2.7. Summit corporation PLC healthy volunteers. Finally, suitable OGA PET tracers have
In September 2012, scientists at Summit Corporation PLC pub­ already enabled the confirmation for target engagement in
lished an application in which they claimed pyrrolidine deriva­ healthy volunteers’ brains. It will undoubtedly be of crucial
tives as selective OGA inhibitors[145]. They disclosed 128 support for proper dose selection for future PoC Phase II
compounds. They measured the enzymatic hOGA activity, and studies that, in the end, are essential to shed light on the
the selectivity against h-β-hexosaminidases (hHEXA/B) was great promises held by OGA inhibition for the manage­
reported for 72 compounds. These compounds demonstrated ment of AD and other tauopathies.
inhibition of hOGA with IC50 values ranging from < 3 nM to
224 µM, whereas selectivity vs hHEXA/B varied from 0.02 to > Funding
65,000 fold. Figure 62 reveals some of the preferred examples, i.e.
321–324. They all contain a common (2 R,3S,4 R,5 R)-3,4-dihy­ This paper was not funded.
droxy-5-(hydroxymethyl)pyrrolidin-2-yl)-N-methylacetamide
core. The cellular activities were also reported for a small selec­
tion of compounds. In some cases, a large cell shift was observed, Declaration of interest
most likely due to a suboptimal cell permeability, e.g. molecule All authors are employees of Janssen Research and Development. The
322, cell shift > 85x. authors have no other relevant affiliations or financial involvement with
any organization or entity with a financial interest in or financial conflict
with the subject matter or materials discussed in the manuscript. This
3. Conclusion includes employment, consultancies, honoraria, stock ownership or
options, expert testimony, grants or patents received or pending, or
We reviewed the existing patent literature between March 2008 royalties.
and March 2021 on OGA inhibitors. Sixty-five patent applications
from different companies comprising large pharma and biotech
have been summarized and relevant chemotypes extracted. Reviewer disclosures
These include both carbohydrate derivatives as well as non- A reviewer on this manuscript has disclosed that they are an employee of
sugar-based structures. The high number of patent applications Alectos Therapeutics, which is engaged in developing OGA inhibitors as
and the companies engaged in this research area confirm that therapeutics. All other peer reviewers on this manuscript have no relevant
OGA inhibition is a fascinating and promising novel target in AD financial or other relationships to disclose.
and related neurodegenerative disorders.

References
4. Expert opinion
Papers of special note have been highlighted as either of interest (•) or of
Neurodegenerative disorders in general, and AD in parti­ considerable interest (••) to readers.
cular, represent an enormous challenge for an increasingly 1. Torres CR, Hart GW. Topography and polypeptide distribution of
aging population in terms of a social and economic bur­ terminal N-acetylglucosamine residues on the surfaces of intact.
Evidence for O-linked GlcNAclymphocytes. J Biol Chem. 1984;259
den, especially since there are no available treatments that (5):3308–3317.
can significantly impact these diseases’ progression. The 2. Holt GD, Hart GW. The subcellular distribution of terminal
availability of high-quality tool compounds suggested N-acetylglucosamine moieties. Localization of a novel
a role for O-GlcNAcylation in neurodegenerative disorders protein-saccharide linkage, O-linked GlcNAc. J Biol Chem.
1986;261(17):8049–8057.
such as AD. It could prevent the accumulation of hyper­
3. Wells L, Vosseller K, Hart GW. Glycosylation of nucleocytoplasmic
phosphorylated tau protein in the brain, which has proteins: signal transduction and O-GlcNAc. Science. 2001;291
prompted enormous interest in searching OGA inhibitors (5512):2376–2378.
amenable for human testing to confirm its potential. This 4. Gao Y, Wells L, Comer FI, et al. Dynamic O-glycosylation of nuclear
interest has resulted in multiple companies engaged in and cytosolic proteins: cloning and characterization of a neutral,
cytosolic beta-N-acetylglucosaminidase from human brain. J Biol
research programs that have yielded a diverse set of che­
Chem. 2001;276(13):9838–9845.
motypes, both carbohydrate and non-carbohydrate 5. Wells L, Gao Y, Mahoney JA, et al. Dynamic O-glycosylation of
derived, suggesting that OGA inhibition is suitable for nuclear and cytosolic proteins: further characterization of the
small molecule intervention. Additionally, this diversity of nucleocytoplasmic beta-N-acetylglucosaminidase, OGlcNAcase.
chemical matter could be of great help to circumvent J Biol Chem. 2002;277(3):1755–1761.
eventual structure-based toxicities that could hamper the 6. Hanover JA, Krause MW, Love DC. Bittersweet memories: linking
metabolism to epigenetics through O-GlcNAcylation. Nat Rev Mol
validation of the role of OGA inhibition in humans. These Cell Biol. 2012;13(5):312–321.
efforts have culminated in identifying three OGA inhibitor 7. Bond MR, Hanover JA. A little sugar goes a long way: the cell
clinical assets (MK-8719 from Merck/Alectos, ASN-120290 biology of O-Glc NAc. J Cell Biol. 2015;208(7):869–880.
36 J. M. BARTOLOMÉ-NEBREDA ET AL.

8. Müller R, Jenny A, Stanley P. The EGF repeat-specific O-GlcNAc- 31. Comtesse N, Maldener E, Meese E. Identification of a nuclear var­
transferase Eogt interacts with notch signaling and pyrimidine iant of MGEA5, a cytoplasmic hyaluronidase and a
metabolism pathways in Drosophila. PLoS ONE. 2013;8(5):e62835. beta-N-acetylglucosaminidase. Biochem Biophys Res Commun.
9. Drougat L, Olivier-Van Stichelen S, Mortuaire M, et al. 2001;283(3):634–640.
Characterization of O-GlcNAc cycling and proteomic identification 32. Schimpl M, Schüttelkopf AW, Borodkin VS, et al. Human OGA binds
of differentially O-GlcNAcylated proteins during G1/S transition. substrates in a conserved peptide recognition groove. Biochem J.
Biochim Biophys Acta. 2012;1820 (12); 1839–1848. 2010;432(1–7):2010.
10. Zachara NE, Hart GW. O-GlcNAc modification: a nutritional sensor 33. Rao FV, Dorfmueller HC, Villa F, et al. Insights into the mechanism
that modulates proteasome function. Trends Cell Biol. 2004;14 and inhibition of eukaryotic O-GlcNAc hydrolysis. EMBO J. 2006;25
(5):218–221. (7):1569–1578.
11. Vosseller K, Wells L, Lane MD, et al. Elevated nucleocytoplasmic 34. Macauley MS, Whitworth GE, Debowski AW, et al. O-GlcNAcase uses
glycosylation by O-GlcNAc results in insulin resistance associated substrate-assisted catalysis: kinetic analysis and development of
with defects in Akt activation in 3T3–L1 adipocytes. Proc Natl Acad highly selective mechanism-inspired inhibitors. J Biol Chem.
Sci U S A. 2002;99(8):5313–5318. 2005;280(27):25313–25322.
12. Arias EB, Kim J, Cartee GD. Prolonged incubation in PUGNAc results 35. Cetinbas N, Macauley MS, Stubbs KA, et al. Identification of Asp 174
in increased protein O-linked glycosylation and insulin resistance in and Asp 175 as the key catalytic residues of human O-GlcNAcase
rat skeletal muscle. Diabetes. 2004;53(4):921–930. by functional analysis of site-directed mutants. Biochemistry.
13. Lagerlöf O, Slocomb JE, Hong I, et al. The nutrient sensor OGT in 2006;45(11):3835–3844.
PVN neurons regulates feeding. Science. 2016;351 36. Herr R, Jahnke HK, Argoudelis AD. Structure of streptozotocin. J Am
(6279):1293–1296. Chem Soc. 1967;89(18):4808–4809.
14. Lehman DM, Fu D-J, Freeman AB, et al. A single nucleotide poly­ 37. Roos MD, Xie W, Su K. et al. Streptozotocin, an analog of
morphism in MGEA5 encoding O-GlcNAc-selective N-acetyl-beta-D- N-acetylglucosamine, blocks the removal of O-GlcNAc from intra­
Glucosaminidase is associated with Type 2 diabetes in Mexican cellular proteins. Proc Assoc Am Physicians. 1998;110(5):422–432.
Americans. Diabetes. 2005;54(4):1214–1221. 38. Gao Y, Parker GJ, Hart GW. Streptozotocin-induced beta-cell death
15. Ma J, Hart GW. Protein O-GlcNAcylation in diabetes and diabetic is independent of its inhibition of O-GlcNAcase in pancreatic Min6
complications. Expert Rev Proteomics. 2013;10(4):365–380. cells. Arch Biochem Biophys. 2000;383(2):296–302.
16. Zachara NE. The roles of O-linked β-N-acetylglucosamine in cardi­ 39. Horsch M, Hoesch L, Vasella A, et al. N-acetylglucosaminono-
ovascular physiology and disease. Am J Physiol Heart Circ Physiol. 1,5-lactone oxime and the corresponding (phenylcarbamoyl)
2012;302(10):H1905–H1918. oxime. Novel and potent inhibitors of
17. Ferrer CM, Sodi VL, Reginato MJ. O-GlcNacylation in cancer biology: beta-N-acetylglucosaminidase. Eur J Biochem. 1991;197(3):815–818.
linking metabolism and signaling. J Mol Biol. 2016;428 40. Vocadlo DJ, Gao Z, Gao Z, et al. NAG-thiazoline, An N-Acetyl-β-
(16):3282–3294. hexosaminidase inhibitor that implicates acetamido participation.
18. Singh JP, Zhang K, Wu J, et al. O-GlcNAc signaling in cancer J Am Chem Soc. 1996;118(28):6804–6805.
metabolism and epigenetics. Cancer Lett. 2015;356(2):244–250. 41. Whitworth GE, Macauley MS, Stubbs KA, et al. Analysis of PUGNAc
19. Yuzwa SA, Vocadlo DJ. OGlcNAc and neurodegeneration: biochem­ and NAG-thiazoline as transition state analogues for human
ical mechanism and potential roles in Alzheimer’s disease and O-GlcNAcase: mechanistic and structural insights into inhibitor
beyond. J Chem Soc Rev. 2014;43(19):6839–6858. selectivity and transition state poise. J Am Chem Soc. 2007;129
20. Ryan P, Xu M, Davey AK, et al. O-GlcNAc modification protects (3):635–644.
against protein misfolding and aggregation in neurodegenerative 42. Yuzwa SA, Macauley MS, Heinonen JE, et al. A potent
disease. ACS Chem Neurosci. 2019;10(5):2209–2221. mechanism-inspired O-GlcNAcase inhibitor that blocks phosphor­
21. Roquemore EP, Chevrier MR, Cotter RJ, et al. Dynamic ylation of tau in vivo. Nat Chem Biol. 2008;4(8):483.
O-GlcNAcylation of the small heat shock protein αB-Crystallin†. 43. Dorfmueller HC, Borodkin VS, Schimpl M, et al. GlcNAcstatin:
Biochemistry. 1996;35(11):3578–3586. a picomolar, selective O-GlcNAcase inhibitor that modulates intra­
22. Chou CF, Smith AJ, Omary MB. Characterization and dynamics of cellular O-GlcNAcylation levels. J Am Chem Soc. 2006;128
O-linked glycosylation of human cytokeratin 8 and 18. J Biol Chem. (51):16484–16485.
1992;267(6):3901–3906. 44. Bergeron-Brlek M, Goodwin-Tindall J, Cekic N, et al. A convenient
23. Haltiwanger RS, Holt GD, Hart GW. Enzymatic addition of O-GlcNAc to approach to stereoisomeric iminocyclitols: generation of potent
nuclear and cytoplasmic proteins. Identification of a uridine diphospho- brain-permeable OGA inhibitors. Angew Chem Int Ed. 2015;54
N-acetylglucosamine: peptideβ-N-acetylglucosaminyltransferase. J Biol (51):15429–15433.
Chem. 1990;265(5):2563–2568. 45. Elsen NL, Patel SB, Ford RE, et al. Insights into activity and inhibi­
24. Kreppel L, Blomberg M, Hart G. Dynamic glycosylation of nuclear tion from the crystal structure of human OGlcNAcase. Nat Chem
and cytosolic proteins. Cloning and characterization of a unique Biol. 2017;13(6):613–615.
O-GlcNAc transferase with multiple tetra-tricopeptide repeats. 46. Roth C, Chan S, Offen WA, et al. Structural and functional insight
J Biol Chem. 1997;272(14):9308–9315. into human O-GlcNAcase. Nat Chem Biol. 2017;13(6):610–612.
25. Lubas W, Frank D, Krause M, et al. O-Linked GlcNAc transferase is 47. Li B, Li H, Lu L, et al. Structures of human OGlcNAcase and its
a conserved nucleocytoplasmic protein containing tetratricopep­ complexes reveal a new substrate recognition mode. Nat Struct
tide repeats. J Biol Chem. 1997;272(14):9316–9324. Mol Biol. 2017;24(4):362–369.
26. Braidman I, Carroll M, Dance N, et al. Characterisation of human 48. Akimoto Y, Comer FI, Cole RN, et al. Localization of the O-GlcNAc
N-acetyl-β-hexosaminadase C. FEBS Lett. 1974;41(2):181–184. transferase and O-GlcNAc- modified proteins in rat cerebellar
27. Dong DL, Hart GW. Purification and characterization of an OglcNAc cortex. Brain Res. 2003;966(2):194–205.
selective N-acetyl-β-D-glucosaminidase from rat spleen cytosol. 49. Liu K, Paterson AJ, Zhang F, et al. Accumulation of protein
J Biol Chem. 1994;269(30):19321–19330. O-GlcNAc modification inhibits proteasomes in the brain and coin­
28. Heckel D, Comtesse N, Brass N, et al. Novel immunogenic antigen cides with neuronal apoptosis in brain areas with high O-GlcNAc
homologous to hyaluronidase in meningioma. Hum Mol Genet. metabolism. J Neurochem. 2004;89(4):1044–1055.
1998;7(12); 1859–1872. 50. Liu Y, Li X, Yu Y, et al. Developmental regulation of protein
29. Ma J, Hart GW. O-GlcNAc profiling: from proteins to proteomes. O-GlcNAcylation, O-GlcNAc transferase, and O-GlcNAcase in mam­
Clin Proteomics. 2014;11(1):8. malian brain. PLoS ONE. 2012;7(8):e43724.
30. Vocadlo DJ. O-GlcNAc processing enzymes: catalytic mechanisms, 51. Khidekel N, Ficarro SB, Peters EC, et al. Exploring the O-GlcNAc
substrate specificity, and enzyme regulation. Curr Opin Chem Biol. proteome: direct identification of O-GlcNAc-modified proteins from
2012;16(5–6):488–497. the brain. Proc Natl Acad Sci U.S.A. 2004;101(36):13132–13137.
EXPERT OPINION ON THERAPEUTIC PATENTS 37

52. Lagerlöf O. O-GlcNAc cycling in the developing, adult and geriatric a novel therapeutic for the treatment of tauopathies. Alzheimer´s &
brain. J Bioenerg Biomembr. 2018;50:241–261. Dementia. 2016;12:261.
53. Marshall S, Bacote V, Traxinger RR. Discovery of a metabolic path­ 75. [cited 2020 Feb 5]. Available from:http://alectos.com/content/pipe
way mediating glucose-induced desensitization of the glucose line/.
transport system. Role of hexosamine biosynthesis in the induction 76. Quattropani A, Hantson J, Hantson J, et al. Pharmacological inter­
of insulin resistance. J Biol Chem. 1991;266(8):4706–4712. vention with the novel o-GlcNacase inhibitor ASN-561 reduces
54. Liu F, Iqbal K, Grundke-Iqbal I, et al. O-GlcNAcylation regulates pathological tau in transgenic mice. Alzheimers Dement. 2015;11
phosphorylation of tau: a mechanism involved in Alzheimer’s dis­ (7S_Part_5):227.
ease. Proc Natl Acad Sci U.S.A. 2004;101(29):10804–10809. 77. [cited 2020 Feb 12]. Available from:https://www.asceneuron.com/
55. Jack CR, Knopman DS, Jagust WJ, et al. Update on hypothetical pipeline.
model of Alzheimer’s disease biomarkers. Lancet Neurol. 2013;12 78. [cited 2020 Feb 12]. Available from:https://www.asceneuron.com/
(2):207–216. news.
56. Iqbal K, Liu F, Gong C-X. Tau and neurodegenerative disease: the 79. [cited 2020 Feb 12]. Available from:https://clinicaltrials.gov/.
story so far. Nat Rev Neurol. 2016;12:15–27. 80. [cited 2020 Feb 12]. Available from:https://www.lilly.com/discov
57. Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative ery/pipeline.
tauopathies. Annu Rev Neurosci. 2001;24(1):1121–1159. 81. Paul S, Haskali MB, Liow JS, et al. Evaluation of a PET radioligand to
58. Arnold CS, Johnson GVW, Cole RN, et al. The image O-GlcNAcase in brain and periphery of Rhesus monkey and
microtubule-associated protein tau is extensively modified with Knock-Out Mouse. J Nucl Med. 2019;60(1):129–134.
O-linked N-acetylglucosamine. J Biol Chem. 1996;271 82. Vocadlo DJ, Mceachern EJ, Stubbs K, Selective glycosidase inhibi­
(46):28741–28744. tors and uses thereof. WO012106 (2010).
59. Liu F, Shi J, Tanimukai H, et al. Reduced O-GlcNAcylation links lower 83. Vocadlo DJ, Mceachern EJ, Selective glycosidase inhibitors and uses
brain glucose metabolism and tau pathology in Alzheimer’s dis­ thereof. WO037207 (2010).
ease. Brain. 2009;132(7):1820–1832. 84. Mceachern EJ, Sun J, Vocadlo DJ, et al. Glycosidase inhibitors and
60. Yuzwa SA, Vocadlo DJ. O-GlcNAc and neurodegeneration: bio­ uses thereof. WO032185 (2014).
chemical mechanisms and potential roles in Alzheimer’s disease 85. Mceachern EJ, Sun J, Vocadlo DJ, et al. Glycosidase inhibitors and
and beyond. Chem Soc Rev. 2014;43(19):6839–6858. uses thereof. WO032188 (2014).
61. Yuzwa SA, Shan X, Macauley MS, et al. Increasing O-GlcNAc slows 86. Mceachern EJ, Vocadlo DJ, Zhou Y, et al. Glycosidase inhibitors and
neurodegeneration and stabilizes tau against aggregation. Nat uses thereof WO032187 (2014).
Chem Biol. 2012;8(4):393–399. 87. The formula LE = (pIC50 x 1.37)/heavy atom count, was used.
62. Hart GW, Slawson C, Ramirez-Correa G, et al. Cross talk between 88. Vocadlo DJ, Mceachern EJ, Selective glycosydase inhibitors and
O-GlcNAcylation and phosphorylation: roles in signaling, transcrip­ uses thereof, WO012107 (2010).
tion, and chronic disease. Annu Rev Biochem. 2011;80(1):825–858. 89. Tong-Shang LI, Mceachern EJ, Vocadlo DJ, et al. Selective glycosy­
63. van der Laarse SAM, Leney AC, Heck AJR. Crosstalk between phos­ dase inhibitors and uses thereof, WO140640 (2011).
phorylation and O-Glc NA cylation: friend or foe. FEBS J. 2018;285 90. Li T-S, Mceachern EJ, Vocadlo DJ, et al. Selective glycosydase
(17):3152–3167. inhibitors and uses thereof, WO000085 (2013).
64. Di Domenico F, Lanzillotta C, Tramutola A. Therapeutic potential of 91. Selnick HG, Liu K, Mceachern EJ, et al. Selective glycosydase inhi­
rescuing protein O-GlcNAcylation in tau-related pathologies. Expert bitors and uses thereof, WO025452 (2013).
Rev Neurother. 2019;19(1):1–3. 92. Coburn C, Liu K, Mceachern EJ, et al. Selective glycosydase inhibi­
65. Yuzwa SA, Shan X, Macauley MS, et al. Increasing O-GlNAc slows tors and uses thereof, WO061927 (2012).
neurodegeneration and stabilizes tau against aggregation. Nat 93. Coburn C, Liu K, Mceachern EJ, et al. Selective glycosydase inhibi­
Chem Biol. 2012;8(4):393–399. tors and uses thereof, WO061971 (2012).
66. Yuzwa SA, Macauley MS, Heinonen JE, et al. Apotent 94. Kaul R, Mceachern EJ, Vocadlo DJ, et al. Selective glycosidase
mechanism-inspired O-GlcNAcase inhibitor that blocks phosphor­ inhibitors and uses thereof, WO129651 (2012).
ylation of tau in vivo. Nat Chem Biol. 2008;4(8):483–490. 95. Kaul R, Mceachern EJ, Mu C, et al. Selective glycosydase inhibi­
67. Graham DL, Gray AJ, Joyce JA, et al. Increased O-GlcNAcylation tors and uses thereof, WO083435 (2012). Data differs from the
reduces pathological tau without affecting its normal phosphoryla­ recently published work by the Alectos Therapeutics and Merck
tion in a mouse model of tauopathy. Neuropharmacology. Sharp and Dohme scientists in. J Med Chem. 2019;62
2014;79:307–313. (22):10062–10097.
68. Hastings NB, Wang X, Song L, et al. Inhibition of O-GlcNacase leads 96. Tong-Shuang LI, Mceachern EJ, Vocadlo DJ, et al. Selective glycosi­
to elevation of O-GlcNac tau and reduction of tauopathy and dase inhibitors and uses thereof, WO000086 (2013).
cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener. 97. Kaul R, Mceachern EJ, Vocadlo DJ, et al. Glycosidase inhibitors and
2017;12(1):39–55. uses thereof, WO067003 (2014).
69. Wang X, Smith K, Pearson M, et al. Early intervention of Tau 98. Liu K, Mceachern EJ, Mu C, et al. Selective glycosidase inhibitors
Pathology prevents behavioral changes in the rTg4510 mouse and uses thereof, WO061972 (2012).
model of tauopathy. PLoS ONE. 2018;13(4):e0195486. 99. Chang J, Liu K, Mceachern EJ et al. Pyrano[3,2-d]thiazol derivatives
70. Wang X, Li W, Marcus J, et al. MK-8719, a novel and selective and uses thereof as elective glycosidase inhibitors, WO062157
O-GlcNAcase inhibitor that reduces the formation of pathological (2012).
tau and ameliorates neurodegeneration in a mouse model of 100. Chang J, Liu K, Mceachern EJ, et al. Selective glycosidase inhibitors
tauopathy. J Pharmacol Exp Ther. 2020;374(2):252–263. and uses thereof, WO064680 (2012).
71. Borghgraef P, Menuet C, Theunis C, et al. Increasing brain protein 101. Mceachern EJ, Vocadlo DJ, Zhou Y, et al. Selective glycosidase
O-GlcNAcylation mitigates breathing defects and mortality of tau inhibitors and uses thereof, WO126091 (2012).
P301L mice. PLoS ONE. 2013;8(12):e84442. 102. Selnick HG, Li W, Hostetler E, et al. Permeable glycosidase inhibitors
72. Medina M. An overview on the clinical development of tau-based and uses thereof, WO168576 (2013).
therapeutics. Int J Mol Sci. 2018;19(4):1160–1173. 103. Hg S, Li W, Hostetler E, et al. Permeable glycosidase inhibitors and
73. Selnick HG, Hess JF, Tang C, et al. Discovery of MK-8719, a potent uses thereof, WO166654 (2013).
O-GlcNAcase inhibitor as a potential treatment for tauopathies. 104. Selnick HG, Liu K, Kaul R et al. Selective glycosidase inhibitors and
J Med Chem. 2019;62(22):10062–10097. uses thereof, WO100934 (2014).
74. Smith SM, Struyk A, Jonathan D, et al. Early clinical results and 105. Vocadlo D, Mceachern EJ, Withworth G, Selective glycosidase inhi­
preclinical validation of the O-GlNAcase (OGA) inhibitor MK-8719 as bitors and uses thereof, WO025170 (2008).
38 J. M. BARTOLOMÉ-NEBREDA ET AL.

106. Mceachern EJ, Selnick HG, Zhou Y, Glycosidase inhibitors and uses 126. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Martínez Viturro CM,
thereof, WO106254 (2017). et al. Bicyclic OGA inhibitor compounds. WO243526 (2019).
107. Donnely M, Qiu H, Yu H et al., Pyrano[3,2-d][1,3]thiazole as glyco­ 127. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Martínez Viturro CM
sidase inhibitors, WO028715 (2013). OGA inhibitor compounds. WO243525 (2019).
108. Yu H, Liu-Bujalski L, Johnson TL, Glycosidase inhibitors, WO159234 128. Bartolomé-Nebreda JM, Trabanco-Suárez AA, De Lucas-Olivares AI,
(2014). et al. OGA inhibitor compounds. WO243528 (2019).
109. [cited 2021 May 04]. Available from:https://www.asceneuron.com/ 129. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Delgado-Jiménez F,
mission-history. . et al. OGA inhibitor compounds. WO243531 (2019).
110. Quattropanni A, Kulkarni S, Giri AG, Glycosidase inhibitors, 130. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Martínez Viturro CM,
WO030443 (2016). et al. Bicyclic OGA inhibitor compounds. WO243527 (2019).
111. Quattropanni A, Kulkarni S, Giri AG, Glycosidase inhibitors, 131. Bartolomé-Nebreda JM, Trabanco-Suárez AA, De Lucas-Olivares AI,
WO144633 (2017); (b) Quattropanni A, Kulkarni S, Giri AG, et al. OGA inhibitor compounds. WO243530 (2019).
Glycosidase inhibitors, WO144639 (2017). 132. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Leenaerts JE, et al.
112. Quattropanni A, Kulkarni S, Giri AG, et al. Process for the separation OGA inhibitor compounds. WO243533 (2019).
of enantiomers of piperazine derivatives, IN21006637 (2016). 133. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Tresadern GJ, et al.
113. Quattropanni A, Kulkarni S, Giri AG, et al., Acid addition salts of OGA inhibitor compounds. WO243535 (2019).
piperazine derivatives, WO144637 (2017). 134. Dreyfus NJF, Lindsay-Scott PJ, N-[4-Fluoro-5-[[(2S,4S)-2-methyl-
114. Quattropanni A, Kulkarni S, Giri AG, Substituted dihydrobenzofur­ 4-[(5-methyl-1,2,4-oxadiazol-3-yl)methoxy]-1-piperidyl]methyl]thia­
ane glycosidase inhibitors, WO153507 (2018). zol-2-yl]acetamide as OGA inhibitor. WO140299 (2018).
115. Quattropanni A, Kulkarni S, Giri AG, Sulfoximine glycosidase 135. NJF D, Lindsay-Scott PJ, Rathmell RE, 5-Methyl-1,3,4-oxadiazol-2-yl
inhibitors, WO153508 (2018). compounds. WO217588 (2018).
116. Quattropanni A, Kulkarni S, Giri AG, Annulated glycosidase 136. NJF D, Faller A, 2,3-Dihydrofuro[2,3-b]pyridine compounds.
inhibitors, WO037861 (2019). WO245907 (2019).
117. Quatroppani A, Wishart G, Kulkarni S et al. Pyrrolidine glycosidase 137. Dreyfus NJF, Lindsay-Scott PJ, 5-Methyl-4-fluoro-thiazol-2-yl
inhibitors. WO039027 (2020). compounds. WO028115 (2020).
118. Quatroppani A, Wishart G, Kulkarni S, et al. Tetrahydro- 138. Dreyfus NJF, Lopez JE, Winneroski LL, et al. 6-Fluoro-
benzoazepine glycosidase inhibitors. WO039028 (2020). 2-methylbenzo[b]thiazol-5-yl compounds. WO068530 (2020).
119. Quatroppani A, Kulkarni S, Rakesh P, et al. Spirocompounds as 139. Hayashi ML, Irizzary MC, Nuthall H, Combination Therapy.
glycosidase inhibitors. WO039029 (2020). WO028141 (2020).
120. Quatroppani A, Kulkarni S, Rakesh P Fused glycosidase inhibitors. 140. Genung N, Guckian KM, Vessels J, et al. O-Glycoprotein-2-acetamido-
WO169804 (2020). 2-deoxy-3-D-glycopyranosidase inhibitors. WO178191 (2019).
121. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Martínez Viturro CM, 141. Genung N, Guckian KM, Vessels J, et al. O-Glycoprotein-2-acet­
et al. Bicyclic OGA inhibitor compounds. WO109198 (2018). amido-2-deoxy-3-D-glycopyranosidase inhibitors. WO117961
122. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Alcázar-Vaca MJ (2020).
[1,2,4]Triazolo[1,5-a]pyrimidinyl derivatives substituted with piper­ 142. Genung N, Guckian KM, Vessels J, et al. Morpholinyl, piperazinyl,
idine, morpholine or piperazine as OGA inhibitors and their oxazepanyl and diazepanyl O-glycoprotein-2-acetamido-2-deoxy-
preparation. WO154133 (2018). 3-D-glycopyranosidase inhibitors. WO061150 (2020).
123. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Alcázar-Vaca MJ, et al. 143. Genung N, Guckian KM, Vessels J, et al. Bicyclic ether
Monocyclic OGA inhibitor compounds. WO109202 (2018). O-glycoprotein-2-acetamido-2-deoxy-3-D-glycopyranosidase
124. Bartolomé-Nebreda JM, Trabanco-Suárez AA, Martínez Viturro CM, inhibitors. WO163193 (2020).
et al. Bicyclic OGA inhibitor compounds. WO141984 (2018). 144. Genung N, Guckian KM, Vessels J, et al. Azetidinyl O-glycoprotein-
125. Martínez-Viturro CM, Trabanco AA, Royes J, et al. Diazaspirononane 2-acetamido-2-deoxy-3-D-glycopyranosidase inhibitors. WO185593
Nonsaccharide Inhibitors of O-GlcNAcase (OGA) for the Treatment (2020).
of Neurodegenerative Disorders. J Med Chem. 2020;63 145. Storer R, Tinsley JM, Wilson FX et al. Pyrrolidine derivatives as
(22):14017–14044. selective glycosidase inhibitors and uses thereof. WO117219 (2012).

You might also like