Subscriber access provided by READING UNIV
Perspective
Novel Phosphodiesterase Inhibitors for Cognitive Improvement in Alzheimer’s Disease Yinuo Wu, Zhe Li, Yiyou Huang, Deyan Wu, and Hai-Bin Luo J. Med. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jmedchem.7b01370 • Publication Date (Web): 24 Jan 2018 Downloaded from http://pubs.acs.org on January 24, 2018
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
Journal of Medicinal Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
Novel Phosphodiesterase Inhibitors for Cognitive Improvement in
2
Alzheimer’s Disease
3
Yinuo Wu, Zhe Li, Yiyou Huang, Deyan Wu, and Hai-Bin Luo*
4
School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
5
Abstract: :
6
Alzheimer’s disease (AD) is one of the greatest public health challenges. Phosphodiesterases (PDEs)
7
are a super enzyme family responsible for the hydrolysis of two second messengers cyclic adenosine
8
monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Since several PDE
9
subfamilies are highly expressed in the human brain, the inhibition of PDEs is involved in
10
neurodegenerative processes by regulating the concentration of cAMP and/or cGMP. Currently,
11
PDEs are considered as promising targets for the treatment of AD since many PDE inhibitors have
12
exhibited remarkable cognitive improvement effects in preclinical studies and over fifteen of them
13
have been subjected to clinical trials. The aim of this review is to summarize the outstanding
14
progress that has been made by PDE inhibitors as anti-AD agents with encouraging results in
15
preclinical studies and clinical trials. The binding affinity, pharmacokinetics, underlying mechanisms,
16
and limitations of these PDE inhibitors in the treatment of AD are also reviewed and discussed.
17 18
1. Introduction
19
Alzheimer's disease (AD) is the most common type of dementia, which is characterized by
20
progressive memory loss, a decline in language skills, and some other neurodegenerative disorders.1
21
According to the data provided by the Alzheimer's Association (USA), approximately 46.8 million
22
people worldwide are suffering from AD. The morbidity and mortality rates of patients with AD are ACS Paragon Plus Environment 1
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
high, especially for elderly individuals over the age of 60. The global societal economic cost for AD
2
patients was estimated at $818 billion in 2015 and is expected to rise to $1 trillion in 2018 and $2
3
trillion in 2030.2 Thus, AD has become one of the greatest public health challenges across the world
4
and severely impacts individuals and their families.
5
Currently, the clinically available anti-AD drugs are three acetylcholine inhibitors (donepezil,
6
rivastigmine, and galantamine) and one N-methyl-D-aspartate receptor (NMDA) antagonist
7
(memantine),3 and these drugs improve only cognition and the degree of dementia in AD patients but
8
do not reverse the progression of AD. Thus, the need to develop novel anti-AD drugs is extremely
9
compelling. While the pathogenic mechanism of AD is highly complicated and still not fully
10
understood, several hypotheses about the pathophysiology of AD, including low levels of
11
acetylcholine, deposition of β-amyloid (Aβ) plaques, aggregation of Tau-protein, oxidative stress,
12
and the accumulation of biometals, have been proposed to facilitate drug development.4 Recently, the
13
Aβ pathological hypothesis has attracted great interest from many studies, as Aβ-plaques and
14
neurofibrillary tangles are two major hallmarks in the brains of AD patients. However, several
15
promising candidates targeting Aβ, such as bapineuzumab and solanuzumab, have failed in Phase III
16
clinical trials.5 Although limited information was obtained for the failure, one important possible
17
reason is that treatment of symptomatic dementia such as β-amyloid (Aβ) plaques was too late to
18
delay the progression of the AD.6 Furthermore, approximately 20-25% of patients have been
19
observed with the assistance of positron emission tomography imaging to be without Aβ burden.7
20
Thus, the discovery of anti-AD agents, including non-Aβ-directed therapies, still has a long way to
21
go.
22
Phosphodiesterases (PDEs) are a super enzyme family that are responsible for the hydrolysis of
23
two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine ACS Paragon Plus Environment 2
Page 2 of 61
Page 3 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
monophosphate (cGMP).8 The PDE superfamily is coded by 21 identified genes and can be divided
2
into 11 subtypes (PDE1-11). Among these subtypes, PDEs 4, 7, and 8 specifically hydrolyze cAMP,
3
whereas PDEs 5, 6, and 9 specifically hydrolyze cGMP. The remaining PDE families are capable of
4
degrading both cGMP and cAMP.9 As cAMP and cGMP are essential in cellular signaling and
5
function, PDE inhibitors are used to regulate many biological processes by increasing the cellular
6
levels of cAMP and cGMP. Thus far, several PDE inhibitors have been approved as therapeutics for
7
the treatment of erectile dysfunction, pulmonary hypertension, chronic obstructive pulmonary
8
disease, and heart failure (Table 1).10 The most successful example of this drug class is sildenafil, a
9
PDE5 inhibitor that is used for the treatment of male erectile dysfunction (Viagra) and pulmonary
10
hypertension (Revatio).
11
PDEs are highly expressed in the human brain, and their inhibitors regulate neurodegenerative
12
processes by increasing the concentrations of cAMP and cGMP in brain tissue (Figure 1).11 In
13
general, cAMP is synthesized from ATP by adenylate cyclase (AC), activates protein kinase A (PKA)
14
and phosphorylates cAMP-responsive element binding protein (CREB), which has been considered a
15
molecular switch required for learning and memory. Several preclinical and clinical studies have
16
demonstrated that impaired CREB phosphorylation plays an important role in neurodegenerative
17
disorders, especially AD.12 Decreasing the level of cAMP may cause a decreased concentration of
18
CREB, which affects the transcription of genes related to synaptic plasticity and survival, such as
19
brain-derived neurotrophic factor (BDNF), and resulting in the loss of synaptic plasticity and in
20
memory decline in AD.13 Similarly, cGMP, which is derived from GTP by guanylyl cyclase (GC),
21
could be activated by nitric oxide (NO) via the NO/cGMP pathway, which activates protein kinase G
22
(PKG) and subsequent CREB phosphorylation, thus increasing the level of the antiapoptotic protein
23
Bcl-2.14,15 ACS Paragon Plus Environment 3
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Since the first preclinical AD study on the PDE4 inhibitor, rolipram, which indicated that the
2
inhibition of PDEs might have promising effects on the improvement of the memory deficits in
3
AD,15 several PDE inhibitors have exhibited remarkable effects in animal models related to AD,
4
including the Morris water maze, passive avoidance, and object recognition tasks (Table 2). In view
5
of the encouraging results obtained in preclinical studies, several PDE inhibitors have been subjected
6
to clinical trials for the treatment of cognitive disorders in AD patients (Table 3). Most of these
7
inhibitors exhibited outstanding safety and tolerability profiles in Phase I studies. Furthermore, over
8
ten PDE inhibitors have been subjected to Phase II/III/IV clinical trials. Thus, this review mainly
9
focuses on the outstanding progress that has been made in the application of PDE inhibitors for
10
memory enhancement effects for anti-AD purposes in preclinical or clinical trials.
11
Table 1. PDEs inhibitors approved on the market. Compound Vinpocetine
Drug name Calan®, Cavinton®
Target PDE1
Clinical use Parkinson' s disease Alzheimer disease
Cilostazol
Amrinone Lactate
Pletal®, Pletaal® Inocor®
PDE3 PDE3
Anti-platelet, intermittent claudication Heart failure
ilrinone Lactate
Primacor®
PDE3
Congestive heart failure
Roflumilast Apremilast Crisaborole Sildenafil Citrate
Daxas® Daliresp® Otezla® Eucrisa® Viagra®
PDE4 PDE4 PDE4 PDE5
Tadalafil
Cialis®
PDE5
Vardenafil Hydrochloride Avanafil
Vivanza®
PDE5
Chronic obstructive pulmonary disease Psoriasis and psoriatic arthritis Allergic dermatitis Erectile dysfunction, Pulmonary Arterial Hypertension Erectile dysfunction, Pulmonary Arterial Hypertension Erectile Dysfunction
Zepeed®, Stendra®
PDE5
Erectile Dysfunction
,
12 13 14
2. PDE inhibitors studied for the treatment of AD 2.1 . PDE1 inhibitors ACS Paragon Plus Environment 4
Page 4 of 61
Page 5 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
PDE1 is a Ca2+/calmodulin-dependent PDE family with three isoforms (PDE1A, 1B, and 1C)
2
that hydrolyzes both cGMP and cAMP.17 PDE1B and PDE1C are the two major isoforms existing in
3
humans, while PDE1A has relatively low expression. Each isoform is also expressed in a
4
tissue-specific manner. In the caudate nucleus and the nucleus accumbens, for example, PDE1B is
5
the most abundant isoform among all PDE species, and the level of PDE1B is 10- and 100-fold
6
higher than that of PDE1C and PDE1A, respectively. In the cortex and hippocampus, the levels of
7
PDE1B and PDE1C are almost the same. In the periphery, PDE1C is the major isoform of PDE1 and
8
mainly exists in the heart, bladder, and lungs (Figure S1).18 Its specific distribution in the brain and
9
its distinctive regulation by intracellular calcium and calmodulin make PDE1 an attractive target to
10
improve cognitive impairments in neurodegenerative diseases, such as AD, schizophrenia, and
11
Parkinson’s disease.19
,
ACS Paragon Plus Environment 5
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2
Figure 1: The relation of PDE inhibition and the cAMP or cGMP signaling pathway in the brain.
3
PDE: phosphodiesterase; AC: adenyl cyclase; GC: guanylyl cyclase; ATP: adenosine triphosphate;
4
GTP: guanosine triphosphate; PKA: protein kinase A; PKG: protein kinase G; P: phosphorylated;
5
CREB: cAMP response element binding protein.
6
ACS Paragon Plus Environment 6
Page 6 of 61
Page 7 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1 2
Figure 2. Current PDE1 inhibitors for the treatment of AD.
3 4
Vinpocetine (1), a derivative of the alkaloid vincamine, shows weak inhibition against PDE1
5
(IC50: 30 µM) and has been widely used for the treatment of cognitive dysfunction (Figure 2).20, 21 In
6
streptozotocin (STZ)-induced rat models, which closely simulate the clinical and pathologic features
7
of sporadic AD22 (Table 2), chronic treatment with 1 significantly improved learning and memory
8
abilities in the Morris water maze and passive avoidance tests. Furthermore, the levels of
9
malondialdehyde (MDA) and nitrite decreased, whereas that of glutathione (GSH) increased. The
10
concentrations of acetylcholinesterase and lactate dehydrogenase were also regulated by 1. These
11
results suggested that its effect on memory impairment might be related to oxidative stress and
12
cholinergic function mechanisms. Pharmacokinetics studies of patients with cerebrovascular
13
disorders demonstrated that 1 is able to pass through the blood-brain barrier and reach the central
14
nervous system.23 A clinical study of patients with mild to moderate organic psychosyndromes,
15
including primary dementia, was carried out in 1991 to evaluate the efficacy and tolerance of orally
16
administering 1. The result showed that 1 could efficiently enhance the cognitive performance of
17
these patients.24
18 19
Table 2. PDEs inhibitors studied in the animal models of Alzheimer's disease. ACS Paragon Plus Environment 7
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Compounds
Subject
Model
Page 8 of 61
Results
(No, its target)
Vinpocetine
Streptozotocin injected rats 22
(1, PDE1)
Morris water maze, 10 mg/kg,
Improve memory,
i.c.v.;
Reduce oxidative–nitritive stress,
Passive avoidance, 10 mg/kg, i.c.v. Modulate cholinergic functions,
Prevent Neuronal damage.
ITI-214
Rats 26
(2, PDE1)
BAY-60-7550
Chronic stress-induced mice 36
(4, PDE2)
Rats and C57BL/6J mice 31
Object recognition, 0.3-3.0 mg/kg,
Improve acquisition, consolidation, and
i.e.
retrieval memory.
Morris water maze, 3.0 mg/kg, i.p.; Reverse cognitive impairment via
Novel object recognition and
neuroplasticity-related
location task, 3.0 mg/kg, i.p.
NMDAR-CaMKII-cGMP/cAMP signaling.
Object recognition and social
Improve memory,
memory task, 1.0 mg/kg, p.o.
Enhance LTP,
Reversed MK801-induced deficits. Aged rats 33
APP/PS1 mice 35
Cilostazol
Aβ25–35-injected mice
Object recognition task, 0.3 mg/kg, Improve cognition and memory through
i.p.
enhancing the nNOS activity.
Object location test, 0.3 mg/kg,
Improve memory without anxiety,
p.o.;
depressive-like behavior or
Y- maze, 0.3 mg/kg, p.o.;
hypothalamusepituitaryeadrenal axis
Forced swim test, 0.3 mg/kg, p.o.;
regulation;
Elevated zero maze, 0.3 mg/kg,
No changes on the level of Aβ, pCREB ,
p.o.
BDNF or presynaptic density.
Y-maze, 30 mg/kg, p.o. 54;
Improve memory possibly due to the
ACS Paragon Plus Environment 8
Page 9 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
(8, PDE3)
Passive avoidance, 30 mg/kg, p.o.
prevention of oxidative damage51 or by
54
decreasing Aβ accumulation by the reduction
;
Morris water maze, 10 mg/kg, i.p.55 of Aβ accumulation and tau phosphorylation.52
Rolipram
APP/PS1 mice 66
Morris water maze, 0.03 mg/kg,
Improve memory.
s.c.
(9, PDE4)
Radial-arm water maze, 0.03
mg/kg, s.c. Aβ25–35 or Aβ40 infused rats68, 71 Morris water maze, 0.5 mg/kg, i.p.; Improve memory; Passive avoidance, 0.5 and 1.25
Reduce oxidative–nitritive stress; Upregulate
mg/kg, i.p.
thioredoxin;
Inhibit iNOS/NO pathway.
Streptozotocin injected and
Morris water maze, 0.05 mg/kg and Improve memory probably due to its
aged mice72
0.1 mg/kg, i.p.
anti-cholinesterase, anti-amyloid,
anti-oxidative and anti-inflammatory effects Iron-impaired aged rat73
Object recognition, 0.01-0.1
Improve memory.
mg/kg, i.p.
Roflumilast
Mice 75
(10, PDE4)
Object location, 0.03 mg/kg, s.c.;
Improve memory,
Y-maze, 0.1 mg/kg, s.c.
Increased emetic-like properties at a dose
Xylazine/ketamine induced
100 times the memory-enhancing dose.
anaesthesia. Scopolamine-impaired rats 75
Object recognition, 0.1 mg/kg, i.p.
No improvement on memory when given
alone;
ACS Paragon Plus Environment 9
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 61
Fully restored memory deficit when given in
combination with donepezil (0.1 mg/kg,
p.o.).
HT-0712
Aged mice 82
(11, PDE4)
MK-0952
Rats 84
(12, PDE4)
Fear conditioning, 0.1 mg/kg, i.p.;
Enhance long-term memory;
Morris water maze, 0.15 mg/kg,
Facilitate expression of CREB-regulated
i.p.
genes in aged hippocampus.
Object recognition, 0.01mg/kg,
Improve long-term recognition, memory and
i.p.;
cognition
Water maze delayed matching to
position test, 0.3mg/kg, i.p.
FFPM
APP/PS1 mice91
(14, PDE4)
GEBR-7b
Mice96
(15, PDE4)
Morris water maze, 0.5mg/kg, p.o.
Improve memory probably due to stimulation
Passive avoidance, 0.5mg/kg, p.o.
of the cAMP/PKA/CREB/BDNF pathway
Xylazine/ketamine induced
and anti-inflammatory effects;
anaesthesia.
Little emetic potential.
Object location and recognition,
Improve memory at doses without
0.003 mg/kg, s.c.
emesis-like behavior.
Xylazine/ketamine induced
anaesthesia.
D159687
Scopolamine-injected mice 98
Y-maze, 0.001mg/kg, i.v.
Improve memory at doses without
emesis-like behavior.
(17, PDE4) Mice 98
Object recognition , 0.01mg/kg, i.v.
Xylazine/ketamine induced
anaesthesia.
ACS Paragon Plus Environment 10
Page 11 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
Sildenafil
Tg2576 mice107
(19, PDE5)
SAMP8 mice108
Morris water maze, 15 mg/kg, i.p.;
Reverse memory deficits by regulating the
Fear conditioning, 15 mg/kg, i.p.
Akt/GSK3b and p25/CDK5 pathways.
Morris water maze, 7.5 mg/kg, i.p.
Ameliorate age-dependent cognitive
impairments,
Reduce the Tau phosphorylation. APP/PS1 mice103,104
Fear conditioning, 3 mg/kg, i.p.
Improve memory by a long-lasting reduction
Reversal Morris water maze, 3
of Aβ levels.
mg/kg, i.p.
Object recognition, 10 mg/kg, i.p.
Restore cognitive deficits by the regulation
of PKG/pCREB signaling, anti-inflammatory
response and reduction of Aβ levels. SAMP8 and SAMR1 mice 109
Passive avoidance, 7.5 mg/kg, i.p.
Improve memory by reducing APP
processing, Aβ levels and tau
hyperphosphorylation.
Tadalafil
Aged J20-mice114
Morris water maze, 15 mg/kg, v.o.
(20, PDE5)
S14
Reduce Tau phosphorylation but not Aβ. APP/PS1 mice 116
Radial arm water maze, 3 mg/kg,
Improve memory.
i.p.
(23, PDE7)
BAY 73-6691
Improve memory;
Scopolamine-injected rats127
T-maze, 10 mg/kg, p.o.
(24, PDE9)
Passive avoidance, 10 mg/kg, p.o.
ACS Paragon Plus Environment 11
Improve long-term and short-term memory.
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Rats128
Social recognition, 0.3 mg/kg, p.o.
Page 12 of 61
Improve both early and late LTP.
Object recognition, 0.1 mg/kg, p.o. Transform early into late LTP. Tg2576 mice129
Object location, 5 mg/kg, p.o.
Improve memory;
Restore Aβ induced impairment of long-term
potentiation.
PF-04447943
Aβ25–35-injected mice130
Morris water maze, 1 mg/kg, i.p.
Scopolamine-injected rats131
Conditioned avoidance attention, 3 Improve learning and memory.
Improve memory.
mg/kg, i.p.
(25, PDE9)
Object recognition,1 mg/kg, p.o. Mice131
Social recognition, 1 mg/kg, p.o.
Y-maze, 1 mg/kg, p.o.
1 2 3
ACS Paragon Plus Environment 12
Improve cognitive performance.
Page 13 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Journal of Medicinal Chemistry
Table 3. PDEs inhibitors in clinical trials for the treatment of Alzheimer's disease or related diseases. Compound
Target Condition
ITI-214 (2)
PDE1 Schizophrenia
Phase I, 2013, Terminated
NCT01900522
PF-0999
PDE2 Schizophrenia
Phase I, 2011, completed
NCT01429740
Healthy Volunteers
Phase I, 2012, completed
NCT01530529
PDE2 Healthy Volunteers
Phase I, 2015, completed
NCT02584569
Healthy Volunteers
Phase I, 2015, completed
NCT02461160
Phase IV, 2011, completed
NCT01409564
(PF-05180999, 6) TAK-915
Cilostazol (8)
Current status
PDE3 Alzheimer's Disease
Mild Cognitive Impairment Phase II, 2015, recruiting
Roflumilast (10)
PDE4 Dementia
Clinical trial registry no.
NCT02491268
Phase II, 2011, completed
NCT01433666
Phase I,2014, completed
NCT02051335
Preclinical, 2016, recruiting
NCT02835716
Phase II, 2013, completed
NCT02013310
Healthy Elderly Volunteers Phase I, 2010, terminated
NCT01215552
Memory Impairment, Alzheimer's Disease Alzheimer Disease HT-0712 (11)
PDE4 Age-Associated Memory Impairment
MK-0952 (12)
PDE4 Alzheimer's Disease
Phase II, 2006, terminated
NCT00362024
Etazolate (13)
PDE4 Alzheimer's Disease
Phase II, 2009, completed
NCT00880412
PDE4 Alzheimer Disease
Phase I, 2017, ongoing, but
NCT03030105
(EHT0202)
BPN14770 (18)
not recruiting participants.
Sildenafil (19)
Alzheimer's Disease
Phase I, 2016, completed
NCT02840279
Alzheimer's Disease
Phase I, 2016, completed
NCT02648672
Phase IV, 2007, completed
NCT00455715
Phase IV, 2013
NCT01941732
Phase II, 2007, terminated
NCT02162979
PDE5 Schizophrenia Parkinson's Disease Parkinson's Disease
due to not enough subjects. PDE5 Dementia, Vascular
Phase II, 2017, recruting.
NCT02450253
PF-04447943 (25) PDE9 Alzheimer's Disease
Phase II, 2009, completed
NCT00930059
Tadalafil
ACS Paragon Plus Environment 13
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Alzheimer's Disease
Phase I, 2009, completed
NCT00988598
Healthy
Phase I, 2010, completed
NCT01097876
PDE9 Alzheimer's Disease
Phase II, 2014, recruiting
NCT02240693
Alzheimer's Disease
Phase II, 2014, recruiting
NCT02337907
PF-02545920 (31) PDE10 Huntington's Disease
Phase II, 2015, terminated
NCT02342548
Huntington's Disease
Phase II, 2014, completed
NCT02197130
BI-409306
1
Page 14 of 61
TAK-063 (32)
PDE10 Schizophrenia
Phase II, 2015, terminated
NCT02477020
OMS643762
PDE10 Schizophrenia
Phase II, 2013, terminated
NCT01952132
EVP-6308
PDE10 Schizophrenia
Phase I, 2014, completed
NCT02037074
a
Information from www.clinicaltrials.gov.
2 3
ITI-214 (2), a PDE1 inhibitor reported by Intra-Cellular Therapies in 2012, had an IC50 value of
4
0.058 nM against PDE1 and an excellent selectivity (>1000-fold) across other PDEs (Figure
5
2).25 The good pharmacokinetic properties of 2 made it suitable as a CNS drug candidate. In a novel
6
object recognition test in rats, with a single oral dose of 0.1-10 mg/kg, 2 could significantly enhance
7
memory performance during acquisition and consolidation without changing exploratory behavior or
8
basal locomotor activity. Furthermore, its administration did not change or have an effect on the
9
antipsychotic activity or pharmacokinetic profile of risperidone in the conditioned avoidance
10
response test.26 Currently, 2 has already completed four Phase I studies (Table 3), including a single
11
rising dose study in normal healthy volunteers and a multiple rising dose study once daily.27 Its
12
safety and tolerability in healthy volunteers and patients with schizophrenia have also been evaluated.
13
Although this trial has been terminated due to a business decision, the obtained results proved that 2
14
might work as an efficient candidate for the treatment of cognitive dysfunction as seen in AD,
15
schizophrenia, and Parkinson’s disease.
16
Most recently, a thienotriazolopyrimidinone PDE1 inhibitor, DNS-0056 (3), with good ACS Paragon Plus Environment 14
Page 15 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
pharmacokinetic properties and brain penetration has been developed (Figure 2).28 In a rat model of
2
recognition memory, 3 significantly increases long-term memory without altering exploratory
3
behavior in rats.
4 5
2.2 PDE2 inhibitors
6
Similar to PDE1, PDE2 is another subfamily that hydrolyzes both cAMP and cGMP. Unlike
7
other PDE families, in which activating cGMP signals could decrease the level of local cAMP, the
8
inhibition of PDE2 selectively abolishes the negative effects of cGMP on cAMP.29 PDE2A is the
9
only isoform of PDE2 that is highly expressed in most brain regions, including the caudate, nucleus
10
accumbens, cortex, and hippocampus (Figure S2).18 In most peripheral tissues, except the spleen, the
11
level of PDE2 is relatively low. This spatial distribution makes PDE2 inhibitors ideal as therapeutic
12
agents against cognitive disorder diseases but with less cardiovascular and other side effects that
13
commonly exist in other PDE inhibitors.30
14
15 16 17
Figure 3. Current PDE2 inhibitors for the treatment of AD.
18 19
BAY-60-7550 (4) is a highly selective PDE2 inhibitor developed by Bayer with an IC50 of 4.7
20
nM and up to 100-fold selectivity across other PDEs (Figure 3).30 Its poor pharmacokinetic ACS Paragon Plus Environment 15
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 61
1
properties have limited its clinical use. Thus, this compound has usually worked as a tool medicine to
2
investigate the potential therapeutic utility of PDE2 inhibitors. The inhibition of PDE2A by 4
3
enhanced the long-term potentiation of synaptic transmission without altering basal synaptic
4
transmission. In addition, 4 improved memory performance in social and object recognition memory
5
tasks and rescued deficits of spontaneous alternation in the T-maze induced by the
6
N-methyl-D-aspartate (NMDA) antagonist MK801.31 Further study of object location and
7
recognition tasks indicated that its role in memory improvement was probably due to the
8
enhancement of memory consolidation instead of attention.32 In memory-impaired, aged rats, 4
9
enhanced the impaired learning and recognition memory for objects at different ages and was
10
accompanied by increased basal constitutive nitric oxide synthase activity in the hippocampus and
11
striatum.33 Another study discovered that the inhibition of PDE2 in the hydrolysis of both cGMP and
12
cAMP using 4 improved spatial object memory consolidation in both early and late stages, whereas
13
the vardenafil-induced inhibition of PDE5 in the hydrolysis of cGMP only enhanced the early stage,
14
and the rolipram-induced inhibition of PDE4 in the hydrolysis of cAMP enhanced only the late
15
stage.34 In APP/PS1 mice, chronic treatment with 4 improved memory performance without causing
16
anxiety, indicating that signaling pathways other than the NO/cAMP/CREB pathway might be
17
responsible for its cognition-enhancing effects.35 Most recently, another study demonstrated that 4
18
reduced stress-induced activation of an extracellularly regulated protein kinase and attenuated the
19
loss
20
neuroplasticity-related NMDAR-CaMKII-cGMP/cAMP signaling pathway.36
of
stress-induced
transcription
factors
and
plasticity-related
proteins
via
the
21
Pharmaceutical study in rats revealed that the brain penetration of BAY 60-7550 was poor. The
22
administration of BAY 60-7550 at the dose of 10 mg/kg only resulted in a brain concentration of 3.6
23
ng/g.37 Thus, the main reason for the good memory improvement effects obtained by BAY 60-7550 ACS Paragon Plus Environment 16
Page 17 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
is probably due to the highest expression of PDE2 in brain, in where low brain concentrations of
2
PDE2 inhibitor might be sufficient to active the cAMP and cGMP signal, causing signal
3
amplification for memory improvement.
4
ND-7001 (5), developed by the Neuro3d company, showed an IC50 of 50 nM against PDE2 and
5
good selectivity over PDE3 and PDE4 (Figure 3).38 Its safety, tolerability, and pharmacokinetics
6
were evaluated in Phase I trials, which demonstrated that it was safe and well tolerated at high doses
7
and produced no sedation, memory disturbance or withdrawal effects.39 However, no further
8
development has been reported since 2010.
9
PF-999 (6, also named as PF-05180999), developed by Pfizer, showed remarkable inhibitory
10
activity against PDE2 (IC50: 2.3 nM) and selectivity over other PDEs, as well as a reasonable free
11
brain/plasma ratio in rats (Figure 3).40 PF-999 was found to increase the level of cGMP in the
12
cerebrospinal fluid of rats, attenuate ketamine-induced memory deficits and reverse spatial learning
13
and memory in scopolamine-induced models.39 In 2015, a study that explored the primarily
14
presynaptic mechanism of PDE2A inhibition was also performed by using PF-999. The results
15
showed that the inhibition of PDE2 might be involved in short-term synaptic plasticity by
16
modulating the hydrolysis of cAMP to accommodate changes in cGMP levels associated with
17
presynaptic short-term plasticity.41 Currently, 6 has already completed two Phase I clinical trials: one
18
trial evaluated its safety, tolerability, and pharmacokinetics in healthy subjects, and the other
19
evaluated the relative bioavailability of a modified-release formulation at 30 mg and 120 mg in
20
patients with schizophrenia.42-44 However, no further information has been reported since 2012.
21
Recently, compound 71 (7), developed by Pfizer, showed an IC50 of 2 nM against PDE2 and
22
selectivity of up to 4000-fold over other PDEs (Figure 3).45 7 increased the level of cGMP in rodent
23
brain regions expressing the highest levels of the PDE2A enzyme. In the radial arm maze, 7 ACS Paragon Plus Environment 17
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
significantly attenuated memory impairments induced by ketamine in rats. Furthermore, 7 reversed a
2
maximum of 80% of the MK-801-induced local field potential disruption at a steady-state Cbu of 11
3
nM. These results indicated selective PDE2A inhibition in brain-potentiated, pharmacologically
4
induced NMDA hypofunction in vivo and that it is suitable to be used in treating neurological and
5
neuropsychiatric disorders associated with NMDA hypofunction, such as schizophrenia.
6
TAK-915, developed by Takeda with no structural or other information disclosed, has already
7
finished two phase I clinical trials of healthy participants in 2015. One trial served to examine the
8
degree and duration of brain PDE2A enzyme occupancy/target engagement after its administration in
9
order to provide evidence of dosing and a schedule for future clinical studies of schizophrenia.46 The
10
other trial was conducted to characterize its safety, tolerability, and plasma pharmacokinetic
11
properties when administered as single and multiple oral suspension doses at escalating dose levels
12
in healthy participants, including elderly participants.47
13 14
2.3 PDE3 inhibitors
15
Similar to PDE1 and PDE2, PDE3 is another subfamily responsible for hydrolyzing both cAMP
16
and cGMP and has two isoforms: PDE3A and PDE3B. In the brain, the expression of PDE3A and
17
PDE3B is relatively low and is mainly in the cerebellum. In the periphery, the expression of PDE3A
18
is mainly in the heart, while the expression of PDE3B is mostly in the lungs and liver (Figure S3).18
19
PDE3A variants have differential expression in cardiovascular tissues. In intracellular, PDE3B is
20
predominantly membrane-associated and localized to endoplasmic reticulum and microsomal
21
fractions. Furthermore, although the concentration of PDE3 is low in brain compared with other PDE
22
subfamilies, the inhibition of PDE3 might be sufficient to active the cAMP and cGMP signal,
23
causing signal amplification for memory improvement.8, 48 ACS Paragon Plus Environment 18
Page 18 of 61
Page 19 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
AD and vascular dementia are two major types of dementia in elderly persons. A number of
2
elderly people may suffer from both conditions. Cerebral white matter lesions have been
3
demonstrated to be one important risk factor for transforming mild cognitive impairment to AD and
4
are the main reason for the age-dependent coexistence of AD and cerebrovascular disease.49, 50 The
5
neurovascular dysfunction of aged AD is associated with cerebral ischemia and the accumulation of
6
Aβ.51, 52
7
8 9
Figure 4. Selective PDE3 inhibitor, cilostazol, may be used to treat cerebrovascular diseases and AD
10
Cilostazol (8) is a selective PDE3 inhibitor (IC50: 0.2 µM) that has been clinically used as an
11
antiplatelet drug for the prevention of cerebrovascular disease (Figure 4).53 Recently, therapy using 8
12
directed at both vascular and neurodegenerative aspects of dementia has been considered a more
13
suitable approach for elderly patients with AD. In an Aβ25-35-injected mouse model of AD, repeated
14
administration of 8 significantly attenuated impairment of spontaneous alternation and reversed the
15
shortening of step-down latency induced by Aβ25-35. The increased level of MDA was completely
16
prevented, indicating that 8 may reverse oxidative damage induced by Aβ25-35.54 Another study
17
demonstrated that 8 significantly attenuated Aβ accumulation and improved spatial learning and
18
memory. The level of Apolipoprotein E (ApoE), a protein associated with Alzheimer's
19
neurofibrillary tangles and Aβ protein, was decreased, resulting in reduced Aβ aggregation.55
20
A clinical study of 10 patients with moderate AD proved that 8 ameliorated cognitive decline ACS Paragon Plus Environment 19
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 20 of 61
1
efficiently when co-administered with donepezil after an average follow-up period. However, this
2
combinatorial therapy had no effect on patients with moderate or severe dementia.56,
3
retrospective analysis revealed that 8 could significantly improve memory in patients with mild
4
cognitive impairment, but no significant effect was observed in patients with normal cognitive
5
function or dementia.58 The clinical effects of monotherapy with galantamine or 8 added to
6
combinatorial therapy in AD patients were also evaluated in a clinical study. Galantamine or 8
7
monotherapy increased the cognitive, affective, and activities of daily living functions in AD patients,
8
while the combination of galantamine and 8 resulted in a better therapeutic effect than the
9
monotherapy.59 Most recently, a clinical study was performed in a large Asian population to
10
investigate the potential risk and benefit of using 8 to reduce the risk of dementia (9148 participants
11
free of dementia at 40 years or older). Patients given 8 had a significantly decreased risk of incident
12
dementia compared with patients without the drug. Notably, the use of 8 had a dose-dependent
13
association with the reduced rate of dementia emergence. Subgroup analysis identified a decrease in
14
dementia in 8 users with diagnosed ischemic heart disease and cerebrovascular disease.60
57
A
15
Two clinical trials of 8 related to AD or dementia were registered on the clinicaltrials.gov. A
16
Phase IV study was performed by Seoul National University Hospital in 2011 to examine the added
17
effect of 8 with donepezil treatment using cognitive tasks and PET imaging in mild to moderate AD
18
patients with subcortical white matter hyperintensities (WMHI).61 A Phase II trial in patients with
19
mild cognitive impairment was performed by the National Cerebral and Cardiovascular Center in
20
2015 to evaluate whether 8 could prevent the conversion from mild cognitive impairment to
21
dementia.62
22 23
2.4 PDE4 inhibitors ACS Paragon Plus Environment 20
Page 21 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
PDE4 is a subfamily that hydrolyzes cAMP only. The four isoforms of PDE4 (PDE4A, PDE4B,
2
PDE4C, and PDE4D) are widely expressed in the CNS and have been found to remain present in the
3
aged and Alzheimer’s brain.63 PDE4B is highly expressed in most areas of brain, except in the dorsal
4
root ganglia, where the levels of all the PDE4 isoforms are low. The levels of PDE4A and PDE4B
5
are equally high in the cortex, but PDE4A is two- to four-fold lower in other brain regions. PDE4C
6
has very low expression in the brain. PDE4D has relatively high expression in the frontal cortex but
7
is 3- to 10-fold lower than PDE4B in all CNS tissues. In the periphery, PDE4B and PDE4D are two
8
major isoforms (Figure S4).18
9
Currently, three PDE4 inhibitors, namely, roflumilast, crisaborole, and apremilast, have been
10
approved on the market for the treatment of chronic obstructive pulmonary disease (COPD), atopic
11
dermatitis, and psoriatic arthritis, respectively.64 In general, PDE4 inhibitors have been widely
12
explored for the treatment of AD.65 However, most of the current PDE4 inhibitors may cause emetic
13
side effects, limiting their usage in CNS diseases.
ACS Paragon Plus Environment 21
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3
Figure 5. Current PDE4 inhibitors for the treatment of AD.
4 5
Rolipram (9) is the first generation of PDE4 inhibitors with an IC50 of 230 nM (Figure 5).
6
Compared with the approved drugs roflumilast and apremilast, rolipram has good brain penetration,
7
or an ability to pass the blood-brain barrier. The concentration of 9 in the brain is twice as high as that
8
in the plasma.65 However, low-dose administration of rolipram causes emesis. Currently, rolipram is
9
widely used to investigate the underlying mechanisms of PDE4 inhibitors in AD.
10
The neuroprotective effect of rolipram in AD models was first demonstrated on
11
hippocampal-dependent memory tasks in 1998, which suggested that the inhibition of PDE4 might
12
decrease the threshold for generating long-lasting long-term potentiation (LTP) and increase ACS Paragon Plus Environment 22
Page 22 of 61
Page 23 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
behavioral memory through the cAMP pathway.16 Gong and coworkers demonstrated that 9
2
improved cognitive deficits in both LTP and contextual learning in APP/PS1 mouse models. Notably,
3
its effect on the improvement of LTP and basal synaptic transmission, as well as working, reference,
4
and associative memory deficits, lasts at least two months after treatment.66 The loss of dendritic
5
spines and dystrophic neurites existed in the hippocampus of APP/PS1 transgenic mice and AD
6
patients. Gong also demonstrated that 9 reversed spine density to a normal level in APP/PS1 mice
7
and aged mice. The changes in dendritic structure and function caused by Aβ peptides were also
8
reversed by rolipram.67 In Aβ25–35- or Aβ1-40-injected rats, chronic administration or repeated
9
treatment with 9 reversed memory impairment, while acute treatment with 9 did not. Furthermore,
10
the decreased levels of Pcreb and Bcl-2 and the increased level of NF-κB p65 and Bax in the
11
hippocampus were regulated by 9 in a dose-dependent manner.68 These results indicated that the
12
attenuation of neuronal inflammation and apoptosis mediated by cAMP/CREB signaling might be
13
involved in the process, which is in accordance with a previous study showing that 9 reversed
14
scopolamine-induced and time-dependent memory in object recognition tasks by elevating cAMP
15
levels.69,70 Most recently, it has been proven that 10 significantly improved learning and memory
16
ability in streptozotocin-induced and naturally aged mice.71 Its potential for treatment of memory
17
dysfunctions can probably be attributed to its anti-cholinesterase, anti-amyloid, anti-oxidative, and
18
anti-inflammatory actions and other effects.72, 73
19
Roflumilast (10, Daliresp or Daxas) was the first selective PDE4 inhibitor approved by the FDA
20
for the treatment of COPD in 2011 (Figure 5). Compared with 9, 10 showed a significantly better
21
inhibitory affinity against PDE4 with decreased emetic properties and may be suitable as a candidate
22
drug for the treatment of AD.74 A preclinical study demonstrated that both 10 and 9 have positive
23
effects on memory improvement in an object location task in male C57BL/6NCrl mice. In the ACS Paragon Plus Environment 23
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Y-maze test, 11 improved spatial memory performance, while 9 did not. More importantly, 10
2
produced emetic-like effects at a dose 100 times that of the memory-enhancing dose, indicating that
3
it is relatively safe in AD treatment. A single administration of donepezil or 10 did not improve
4
memory, while combining efficacious doses of both fully reversed scopolamine-induced memory
5
deficits in object recognition tasks.75 Its beneficial effect on memory and its non-emetic properties
6
promoted 10 to clinical trials of AD. Currently, 10 has already finished one Phase II study as a
7
translational cognition enhancer76 and one Phase I study to evaluate whether co-administration of 10
8
and donepezil could attenuate scopolamine-induced cognitive impairment.77 A preclinical trial is
9
underway to evaluate its effect on preventing the onset of AD in high-risk individuals.78 Although
10
the IC50 of 11 is almost 1000 times higher than that of 9 in vitro, the effects obtained in memory
11
improvement experiments by roflumilast are almost the same or even less than those of rolipram.
12
The difference has been attributed to the relatively poor ability of roflumilast to penetrate the
13
blood-brain barrier. Recent studies demonstrated that 10 efficiently reversed cognitive impairment
14
induced by hypertension in rodents, even at low doses.79,80
15
HT-0712 (11) was developed as a novel PDE4 inhibitor by Bourtchouladze and Scott (Figure 5).
16
Both 11 and 9 ameliorated long-term memory in a mouse model of Rubinstein-Taybi syndrome.81,82
17
Currently, 11 has already completed a Phase II trial to evaluate its efficacy in improving memory and
18
cognitive performance in subjects with age-associated memory impairment (AAMI) and had positive
19
results.83
20
MK-0952 (12), developed by Merck, shows an IC50 of 0.6 nM against PDE4 (Figure 5).84 A
21
Phase II clinical trial with 12 was performed to determine its effect on improving cognitive
22
impairments in patients in mild to moderate stages of AD.85
23
Etazolate (13, also named as EHT0202) is a PDE4 inhibitor and a GABA-A receptor ACS Paragon Plus Environment 24
Page 24 of 61
Page 25 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
modulator (Figure 5).86 Its underlying mechanism of memory improvement in AD might be
2
cross-linked.87 Currently, 13 has already finished one Phase II trial to evaluate its safety and
3
tolerability as an adjunctive therapy to acetylcholinesterase inhibitors in mild to moderate AD.88 The
4
results showed that 13 was safe and generally well tolerated. Dose-dependent numbers of early
5
withdrawal and central nervous system-related adverse events were observed.89
6
FFPM (14) is a novel PDE4 inhibitor that was developed by Ke and coworkers and that has an
7
IC50 of 26 nM and good selectivity over other PDEs (Figure 5).90 Its effects on learning and memory
8
were investigated by Xu and coworkers using the APP/PS1 mouse model of AD.91 After 3 weeks of
9
treatment, the learning and memory abilities of APP/PS1 transgenic mice were significantly
10
improved, as measured by the Morris water maze test and the step-down passive avoidance task. 14
11
had no significant effect on the duration of xylazine/ketamine anesthesia in mice, indicating that 14
12
might not cause emesis during the treatment of AD. Furthermore, 14 penetrated the blood-brain
13
barrier quickly after oral administration, with a half-life in plasma of 1.5 h.
14
PDE4D has been demonstrated to be the main subtype involved in the process of memory
15
consolidation and LTP. The inhibition of PDE4 by 9 in PDE4D-deficient mice did not alter memory
16
performance.92 Thus, selective PDE4D inhibitors may have beneficial effects on cognition
17
enhancement. However, the inhibition of PDE4D is also the main reason for the emetic side effects
18
of a PDE4 inhibitor, as it may mimic the pharmacological actions of α2-adrenoceptor antagonists.93,
19
94
20
GEBR-7b (15) is a selective PDE4D inhibitor, showing low inhibitory activities towards
21
PDE4A4, PDE4B2, and PDE4C2 (Figure 5).95 An in vivo study demonstrated that 16 improved
22
spatial and object recognition memory in late-phase consolidation processes in object recognition
23
and location tests. The level of cAMP in the hippocampus was increased without affecting the level ACS Paragon Plus Environment 25
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
of Aβ. The effect of 15 on memory was 3- to 10-fold more potent than that of 9. In the
2
xylazine/ketamine test, no emetic effect was observed in mice, even at doses of 30 times higher than
3
those in the object location test for behavioral performance improvement. The greatly reduced emetic
4
effect of 15 is probably due to the relatively low doses required to improve memory.96 Most recently,
5
the development of 15 led to a new molecule, 8a (16), which showed good PDE4D3 selective
6
inhibition and the ability to cross the blood-brain barrier.97 The brain/plasma ratio of 16 is 0.8,
7
while those of roflumilast and rolipram are approximately 1 and 2, respectively. In the object
8
recognition task, administration of 16 at a dose of 0.003 mg/kg significantly enhanced long-term
9
memory performance and fully reversed the scopolamine induced short-term memory deficit
10
without causing any emetic-like behavior. These results offered another strategy for the discovery
11
of PDE4 inhibitors with non-emesis behavior for the treatment of AD.
12
Recent studies demonstrated that allosteric modulators of PDE4D that do not completely inhibit
13
enzymatic activity may enhance memory with reduced emetic effects. Unlike the usual PDE4
14
inhibitors, these PDE4D allosteric modulators form an allosteric binding to the specific
15
phenylalanine in primates in the UCR2 region of PDE4D, resulting in its high affinity and selectivity.
16
In the scopolamine-impaired mouse model, the PDE4D allosteric modulator D159687 (17), with an
17
IC50 of 27 nM against PDE4D and good selectivity, efficiently improved cognitive memory in novel
18
object recognition and Y-maze tests (Figure 5). Compared with 9, 17 showed 100-fold less emetic in
19
S. murinus, 3,000-fold less in the beagle dog, and 500-fold less in monkey. Thus, 17 may work
20
efficiently to improve memory with reduced emetic potential. The total brain/plasma AUC ratio for
21
D159687 was above 1 across both rodents and primates.98
22
Another PDE4D allosteric modulator, BPN14770 (18), had IC50 values of 8 and 130 nM against
23
human and mouse PDE4D, respectively (Figure 5). This modulator also enhanced the long-term ACS Paragon Plus Environment 26
Page 26 of 61
Page 27 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
potential in hippocampal slices.99 In 2016, two clinical trials of 18 were conducted for the evaluation
2
of its safety, tolerability, and pharmacokinetic profile in different subjects.100, 101 Another Phase I
3
trial to assess its effect on reversing scopolamine-induced cognitive impairment in healthy volunteers
4
is in progress, with donepezil used as the positive control.102
5 6
Figure 6. Current PDE5 inhibitors for the treatment of AD.
7 8
2.5 PDE5 inhibitors
9
Phosphodiesterase-5 (PDE5) is a subfamily that hydrolyzes cGMP and has one isoform, PDE5A.
10
Compared with other PDE subfamilies, the expression of PDE5A in the brain is relatively low. The
11
levels of PDE5A mRNA are highest in the periphery, bladder, and lungs (Figure S5).18 However,
12
several studies have demonstrated that PDE5 inhibitors have a potential therapeutic effect on the
13
treatment of AD through stimulation of nitric oxide (NO)/cGMP signaling by elevating the level of
14
cGMP. Currently, several PDE5 inhibitors, such as sildenafil and tadalafil, have been approved by
15
the FDA for the treatment of erectile dysfunction and pulmonary arterial hypertension.103
16
Sildenafil (19) has shown an IC50 of 2.2 nM against PDE5A and moderate selectivity across
17
other PDEs (Figure 6).104 Its pharmaceutical properties, such as easily crossing the blood brain
18
barrier and lower toxicity, indicate that 19 is suitable as a candidate drug for the investigation of the ACS Paragon Plus Environment 27
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
effect and mechanism of PDE5 inhibitors in neurodegenerative processes.104-109 19 produced an
2
immediate and long-lasting improvement of synaptic function, CREB phosphorylation, and memory
3
in an APP/PS1 mouse model.105 In the same study, PDE5 inhibitor tadalafil and a highly selective
4
PDE1 inhibitor IC354 (IC50: 80 nM) were also tested. Tadalafil (1 mg/kg, i.p.) did not improve either
5
contextual fear conditioning or spatial working memory in APP/PS1 mice and IC354 had no effect
6
on the LTP amplitude in hippocampal slices. Further study proved that 19 regulates the level of Aβ
7
possibly by modifying their production, metabolism, or clearance. The increased CREB
8
phosphorylation might be due to the anti-inflammatory effect of 19 via the cGMP/PKG/pCREB
9
signaling pathway.106 Currently, 19 has already finished one Phase IV trial for schizophrenia and one
10
Phase IV trial for Parkinson's disease.110,111 However, the clinical trial for AD has not been
11
performed yet. It is worth mentioning that the selectivity of 19 for PDE1 and PDE6 is 180 and 12,
12
respectively, and may cause mild vasodilatory effects and transiently disturb vision.
13
As is mentioned before, there are direct links between AD pathology with cerebrovascular
14
disease. Compared to healthy controls, patients with AD usually have reduced cerebral blood flow
15
(CBF), increased cerebrovascular resistance, and reduced cerebral metabolic rate. A clinical study on
16
AD patients was performed in 2017, demonstrating that 19 could significantly improve the cerebral
17
blood flow and oxygen consumption at a single dose of 50 mg. The cerebrovascular reactivity was
18
decreased. This result indicated that the memory effect of PDE5 inhibitors may possibly be due to
19
the increase in cerebral blood flow mediated by PDE5 expressed in endothelial brain tissue.112
20
Tadalafil (20) is an efficient PDE5 inhibitor developed by Eli Lilly (Figure 6). Compared
21
with 19, 20 has shown better selectivity against PDE6 and a longer half-life.113 These properties
22
suggest that 20 might be more suitable than 19 as an anti-AD drug. However, the administration
23
of 20 (1 mg/kg, i.p.) did not improve either contextual fear conditioning or spatial working ACS Paragon Plus Environment 28
Page 28 of 61
Page 29 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
memory in APP/PS1 mice, while 19 did.105 The main reason for this result has been attributed to
2
the fact that 19 can cross the BBB, while 20 cannot. A further study in 2013 proved that 12% of
3
tadalafil found in the bloodstream crosses the BBB and reaches the brain. Chronic treatment with
4
20 leads its accumulation in the brain of the J20 transgenic mouse model of AD, which reverses
5
memory deficits. 20 is even more effective than 19 in the improvement of memory performance. The
6
level of hyperphosphorylated Tau protein decreased via activation of the pAkt/GSK3 pathway.
7
Neither 20 nor 19 decreased the level of Aβ plaques, indicating that cognition enhancement by
8
PDE5 inhibitors may occur without gross improvement of Aβ pathology.114 Currently, a phase II
9
clinical trial of tadalafil is underway in patients with cerebral small vessel disease, to identify
10
whether tadalafil improves blood flow in deep brain tissue and potentially improves cognitive
11
function 115
12
A quinoline compound, 7a (21), exhibited a higher inhibitory affinity (IC50 = 0.27 nM) and
13
better selectivity than 19, vardenafil, and 20 (Figure 6). In the APP/PS1 mouse model of AD, 21
14
crossed the blood brain barrier readily and increased the level of cGMP in the mouse hippocampus,
15
thus restoring synaptic plasticity and memory damage caused by the elevation of Aβ42.116
16
The pathogenic mechanism of AD is complicated. A combination of multiple targets involved
17
in AD has been considered more appropriate than individual ones. Histone deacetylase inhibitors
18
(HDACIs) are potential modulators of cognitive impairment in AD. Administration of both the
19
pan-HDACI vorinostat and PDE5 inhibitor 20 in aged Tg2576 mice effectively reversed the
20
cognitive deficits and increased the reduced dendritic spine density in hippocampal neurons. The
21
co-administration of these two drugs gave better and longer-lasting effects than each drug alone.117
22
Based on this result, CM-414 (22), an inhibitor acting on both PDE5 and HDAC was developed with
23
remarkable IC50 values against PDE5 and moderate inhibition against HDAC class I (Figure 6).118 22 ACS Paragon Plus Environment 29
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
was able to cross the blood-brain barrier, inducing AcH3K9 acetylation and CREB phosphorylation
2
in the hippocampus and rescuing LTP in APP/PS1 mice. Chronic treatment of Tg2576 mice with 22
3
decreased the level of Aβ and tau phosphorylation in the brain and increased the inactive form of
4
GSK3β. Both the decrease in dendritic spine density in hippocampal neurons and the cognitive
5
deficits were reversed. Thus, 22 may act as an efficient PDE5/HDAC dual inhibitor with a good
6
safety profile, and it may be worth exploration in the clinical trials for AD patients.119 O N N H
S
23 (S14)
7 8
IC50 (PDE7A): 4.7 M IC50 (PDE7B): 8.8 M MW:254.3
Figure 7. Current PDE7 inhibitor for the treatment of AD.
9 10
2.6 PDE7 inhibitors
11
PDE7 is a cAMP-specific subfamily composed of two isoforms, PDE7A and PDE7B.
12
Compared with other PDEs, the expression of PDE7 is relatively low in all tissues. The expression of
13
PDE7B is relatively higher than that of PDE7A in the CNS, such as in the caudate, nucleus
14
accumbens, cortical tissues, and hippocampus. In the periphery, the level of PDE7A is higher than or
15
equal to that of PDE7B in the heart, lungs, etc. (Figure S6).18 Moreover, in situ hybridization
16
demonstrated that the expression of PDE7B remained unchanged and that PDE7A was reduced in the
17
hippocampal regions of advanced AD patients.120 Several PDE7 inhibitors have been recently
18
reported to be potential new agents for the treatment of brain diseases. However, investigation of
19
PDE7 inhibitors for the treatment of AD is still limited.121
20
A quinazoline compound, S14 (23), was identified by the CODES program with IC50 values of ACS Paragon Plus Environment 30
Page 30 of 61
Page 31 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
4.7 and 8.8 µM against PDE7A and PDE7B, respectively, as well as good selectivity over the other
2
five PDEs (Figure 7).122,123 In the APP/PS1 mouse, its administration reduced behavioral impairment
3
in the T-maze via the cAMP/CREB signaling pathway. Moreover, 23 reduced the accumulation of
4
Aβ deposits and modulated brain astrocyte distribution. Significant decreases in tau phosphorylation,
5
cell death and proapoptotic protein expression were also observed.124
6
7 8
Figure 8. Current PDE9 inhibitors for the treatment of AD.
9 10
2.7 PDE9 inhibitors
11
PDE9 is a subfamily that hydrolyzes cGMP only. Among the 11 PDE subfamilies, PDE9 has
12
the highest binding affinity for cGMP (Km: 70 nM) with only isoform PDE9A.125 The expression of
13
PDE9A occurs in most human tissues (Figure S8). In the CNS, the highest levels of PDE9A is in the
14
caudate nucleus and cerebellum. In the periphery, the highest level of PDE9 occurs in the bladder.
15
PDE9 has been regarded as an important target since several efficient inhibitors are in clinical trials
16
for the treatment of AD. ACS Paragon Plus Environment 31
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
In 2005, the first selective PDE9 inhibitor, BAY 73-6691 (24), was reported by Bayer. 24
2
showed an IC50 of 55 nM against PDE9A and moderate selectivity over PDE1 (Figure 8).126 A
3
program was performed to investigate its potential treatment effect on neurological diseases,
4
including AD, but was terminated in 2004 with no reason disclosed. Currently, 24 is often used as a
5
tool in medicine to investigate the underlying mechanism of PDE9A inhibition in AD.127-130 24
6
enhanced the ability of acquisition, consolidation, and retention of LTP in social and object
7
recognition tasks in rodents, improving the scopolamine-induced passive avoidance deficit and the
8
MK-801-induced short-term memory deficits.127 Compared with donepezil, which only increased
9
early LTP, 24 improved both early and late LTP, even transforming early into late LTP. These
10
results indicated that PDE9 inhibition might have better therapeutic effects on AD patients than
11
donepezil.128 In APP transgenic Tg2576 mice with Alzheimer’s plaque pathology, 24 restored Aβ42
12
oligomer-induced LTP and improved memory performance.129,130
13
PF-04447943 (25) is a highly selective and brain penetrant PDE9A inhibitor that was developed
14
by Pfizer in 2011, with an IC50 of 8.3 nM against PDE9A and above 100-fold selectivity over other
15
PDEs (Figure 8).131 Compared with other current AD drugs, which only ameliorate learning and
16
memory function, 25 was able to reverse scopolamine-induced deficits in both the Morris water
17
maze and novel object recognition tests.132 In the Y-maze spatial recognition memory, social
18
recognition memory and scopolamine-deficit novel object recognition tasks, 25 significantly
19
improved cognitive performance.133 In the Tg2576 mouse model of amyloid precursor protein (APP)
20
overexpression, 25 regulated the dendritic spine density of hippocampal neurons.134A preclinical test
21
proved that 25 has excellent tolerability, exposure, and a long half-life of 19-31 h. Currently, 25 has
22
already completed six Phase I trials and one Phase II trial for AD. In the six Phase I trials, its safety,
23
tolerability, and blood level after multiple doses were evaluated in different subjects with good ACS Paragon Plus Environment 32
Page 32 of 61
Page 33 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
results. One Phase I trial was performed to evaluate its safety when given in combination with
2
donepezil in AD patients and to evaluate the absorption and distribution of both it and donepezil.135
3
Its efficacy, safety, and pharmacokinetics were investigated in a phase II trial in subjects with mild to
4
moderate probable AD.136 However, its administration over two weeks did not improve cognitive
5
behavior or cause global changes with statistically significant differences compared with a placebo.
6
BI-409306 was developed by Boehringer Ingelheim for the treatment of cognitive disorder
7
diseases, such as AD and schizophrenia (IC50: 52 nM). Currently, BI-409306 has already
8
completed fifteen Phase I trials. The safety, tolerability, and pharmacokinetics of BI-409306
9
were investigated in different subjects, including healthy males, CYP2C19-genotyped persons,
10
and patients with AD. The results showed that BI-409306 was safe and well tolerated in most
11
subjects. The most commonly experienced adverse effects were headache and photopsia, with
12
mild to moderate intensity, which might be caused by the modulation of PDE9A of some
13
processes in the retina. Furthermore, in a study aiming to assess its exposure in cerebrospinal
14
fluid, BI-409306 was found to cross the blood-CSF barrier and be rapidly absorbed in plasma
15
and distributed in CSF. The level of cGMP in CSF increased in a dose-dependent manner.
16
Currently, two phase II trials for AD are in progress.137,138 One trial aims to investigate its effect
17
on cognitive impairment in AD patients in comparison with a placebo or donepezil. The other
18
trial aims to explore the effect of different doses on the treatment of AD. IMR-687(26) and
19
PF-4181366, developed by H Lundbeck company and Pfizer, respectively were evaluated in
20
preclinical and clinical trials of AD, but no progress has been reported.
21
A series of PDE9 inhibitors was reported by Luo’s group with the assistance of structure-based
22
design and computational docking (Figure 8).139 Compound 27 has an IC50 of 21 nM against
23
PDE9A and >150-fold selectivity over other PDEs. Further structural modification of compound 27 ACS Paragon Plus Environment 33
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 34 of 61
1
led to compound 3r (28), with an improved IC50 of 0.6 nM. However, these two compounds were
2
designed for the treatment of diabetes, and thus, their physicochemical properties might be beyond
3
the range of CNS drugs.140 Compound C33 (29), with an IC50 value of 16 nM against PDE9 and
4
moderate selectivity over other PDEs, could significantly improve memory and cognitive impairment
5
induced by scopolamine or Aβ25-35.141 In 2015, Luo’s group used a combinatorial method to discover
6
novel PDE9 inhibitors with new scaffolds rather than pyrazolopyrimidinones from the SPECS
7
database.142 Fifteen hits out of 29 molecules, with five novel scaffolds, were identified as PDE9
8
inhibitors. Further structural modification of compound AG-690/40135604 (IC50 = 8.0 µM) led to a
9
new compound, 30, with an improved inhibitory affinity of 2.1 µM. The five novel scaffolds
10
discovered could be used for the rational design of PDE9 inhibitors with higher affinities. N N
F N
OMe
O Me
N
O
N Me O
N N
N
N
H N
OMe
N
O
Br OMe
N
11 12
31 (PF-02545920) IC50 (PDE10A): 0.37 nM MW:392.5
N N 32 (TAK-063) IC50 (PDE10A): 0.3 nM MW:428.4
33 (OMS-824) IC50 (PDE10A): not disclosed MW:487.1
Figure 9. Current PDE10 inhibitors for the treatment of AD and other diseases.
13 14
2.8 PDE10 inhibitors
15
PDE10A is a dual-specificity subfamily that hydrolyzes cAMP (Km = 0.05 µM) and cGMP (Km
16
= 3 µM).143 The highest expression of PDE10A in the brain is in the caudate nucleus, and it is also
17
the most prevalent PDE species in this tissue, together with PDE1B. The level of PDE10A is
18
relatively high in the nucleus accumbens. In other parts of the brain and the peripheral tissues
19
examined, the level of PDE10 mRNA could be detected but was very low (Figure S8).18 Furthermore, ACS Paragon Plus Environment 34
Page 35 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
the level of PDE10A decreased in the striatum of patients with Huntington's disease and immediately
2
preceded the onset of motor symptoms.144 Currently, PDE10 is considered a promising target for
3
CNS diseases, especially for schizophrenia and Huntington's disease.
4
PF-02545920 (31, MP-10) was developed by Pfizer (Figure 9).145 After its safety, tolerability,
5
and pharmacokinetics in different subjects were identified in five Phase I trials, its efficacy in the
6
treatment of schizophrenia was evaluated in three Phase II trials. However, 31 failed to reach its
7
primary efficacy end-point in a trial with schizophrenia patients in 2012, and thus, its condition was
8
changed to Huntington’s disease, which was completed in 2014.146 Another Phase II trial in 2015
9
sought to investigate its long-term safety, tolerability, and efficacy in subjects with Huntington's
10
disease. However, this study was terminated in 2016 due to no significant difference being observed
11
in the primary endpoint between 31 and placebo.147
12
TAK-063 (32), developed by Takeda by using structure-based drug design (SBDD) techniques,
13
has an IC50 of 0.30 nM against PDE10 (Figure 9). Its selectivity over other PDEs could be up to
14
15000-fold. Its pharmacokinetics were promising, including high brain penetration in mice.148 In vivo
15
studies have demonstrated that 32 produced dose-dependent, antipsychotic-like effects in
16
METH-induced hyperactivity and prepulse inhibition in rodents.149 Furthermore, 32 attenuated both
17
phencyclidine-induced working memory deficits in a Y-maze test in mice and MK-801-induced
18
working memory deficits in an eight-arm radial maze task in rats.150 Recently, 32 has been evaluated
19
in a Phase II trial for the treatment of acutely exacerbated schizophrenia.151
20
OMS-824 (33, OMS643762), developed by Omeros Corporation, has finished one Phase II trial
21
that evaluated its safety, tolerability, and pharmacokinetics in psychiatrically stable schizophrenia
22
patients (Figure 9).152 Another Phase I trial of 33 in subjects with Huntington's disease has been
23
suspended with no reason disclosed.153 Another PDE10 inhibitor, EVP-6308, has finished a Phase II ACS Paragon Plus Environment 35
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
trial in subjects with schizophrenia who are on a stable anti-psychotic regimen.154
2 3
3. Other PDE subfamilies and their application.
4
PDE6 is an enzyme-specific hydrolyzing cGMP that is divided into three subtypes: PDE6A, 6B
5
and 6C. The retina is the only human tissue with high levels of PDE6A expression. PDE6 has not
6
been detected in the CNS.18 Currently, the purification of PDE6 enzyme is still a challenge, and no
7
selective PDE6 inhibitor has been reported yet.155 The structural, biochemical, and pharmacologic
8
properties of PDE6 are closely related to PDE5. PDE5 inhibitors, such as sildenafil, vardenafil, and
9
tadalafil, have exhibited low selectivity towards PDE6, which may disturb the cGMP signaling
10
pathway in the retina during treatment for other diseases and may cause side effects, including visual
11
disturbances, blurry vision, and light sensitivity.156
12
PDE8 specifically hydrolyzes cAMP, which is coded by two genes: PDE8A and PDE8B.
13
PDE8A exists throughout the brain and peripheral organs at low levels, while PDE8B is mainly
14
expressed in the brain and thyroid.18 Based on its expression, PDE8 has been considered a target for
15
the treatment of thyroid dysfunction and modulation of steroidogenesis in the testes and adrenal
16
glands.157-159 Furthermore, the overexpression of PDE8B, as well as PDE7, has been observed in the
17
brains of Alzheimer’s patients, indicating that the inhibition of PDE8B was able to enhance memory
18
function by the activation of the cAMP signal pathway. However, only a few PDE8-selective
19
inhibitors are available,160 limiting the usage of PDE8.
20
PDE11A is the only isoform of PDE11 that hydrolyzes both cAMP and cGMP. PDE11A is
21
expressed at relatively low levels in most tissues, especially in the cortex and the hippocampus.18
22
Studies of PDE11 are few, and the biological roles of PDE11 are poorly understood due to the lack
23
of selective inhibitors. ACS Paragon Plus Environment 36
Page 36 of 61
Page 37 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Journal of Medicinal Chemistry
4. Prospective
2
In light of the recent high-profile Phase III failures of several Aβ-targeting agents, such as
3
bapineuzumab and solanuzumab, there has been a trend to refocus on the discovery of novel
4
non-Aβ-directed, anti-AD drugs rather than acetylcholine inhibitors and NMDA antagonists, which
5
still has a long way to go.
6
Over the last twenty decades, great efforts have been made to understand the roles of PDEs and
7
to discover effective inhibitors for cognitive improvement in AD. Compared with other hot research
8
areas related to the deposition of Aβ or aggregation of Tau protein, PDE inhibitors hydrolyse two
9
important second messengers (cAMP/cGMP), which can be used before the formation of amyloid
10
plaques and neurofibrillary tangles, regulating central nervous system function such as
11
emotion-related learning, memory, and neural regeneration. Alterations in the levels of cAMP and
12
cGMP are due to the accumulation of cyclase and the degradation of PDE regulation. The regulation
13
of the concentration of cAMP/cGMP may cause a makes PDEs promising drug targets. Until now,
14
there have been many reports of PDE inhibitors with outstanding and remarkable effects on memory
15
in AD in preclinical (Table 2) or clinical (Table 3) trials. The role of PDE inhibitors in improving
16
learning and memory is gaining more and more data, but the underlying mechanisms are still not
17
fully understood. Sufficient evidence has demonstrated that the cAMP-mediated PKA/CREB or
18
cGMP-mediated PKG/CREB signaling pathways are involved in this process. In addition, enhanced
19
cAMP/cGMP concentrations by several PDE inhibitors can significantly improve both early and late
20
LTP, even transforming early into late LTP, which demonstrates that the inhibition of PDEs might
21
have better therapeutic effects on AD patients than acetylcholine inhibitors and suggests a possible
22
mechanism for the use of PDE inhibitors in the treatment of AD. This finding may lead to the
23
development of drugs for treating cognitive dysfunctions, especially in AD.
24
As candidates for the treatment of AD, PDE inhibitors may cause several adverse reactions,
25
which greatly limits their clinical applications. For example, most current PDE4 inhibitor including
26
Roflumilast may cause nausea, emesis, diarrhea, and headache, due to the inhibition of PDE4D ACS Paragon Plus Environment 37
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
subtype. PDE5 inhibitor Sildenafil may cause visual abnormality as it inhibit PDE6 at the same time.
2
Solutions for avoiding these side effects should be further studied through effective methods. Many
3
studies used structure-based molecular design methods to discover highly selective PDE inhibitors.
4
In addition, the discovery of novel allosteric modulators93-97 based on the allosteric site of PDEs
5
could significantly improve their selectivity and decrease the likelihood of side effects. Such
6
allosteric modulators will provide a better basis for the development of future anti-AD drugs in the
7
field of PDE inhibitors, but their role needs to be examined in further preclinical or clinical
8
evaluations.
9 10
■ ASSOCIATED CONTENT
11
Supporting Information
12
The Supporting Information is available free of charge on the ACS Publications website.
13 14
■ AUTHOR INFORMATION
15
Corresponding Authors
16
* E-mail:
[email protected]; Fax: +86-20-3994 3000 (H.B.L.).
17
ORCID
18
Yinuo Wu: 0000-0003-3071-5333
19
Hai-Bin Luo: 0000-0002-2163-0509
20 21
Funding
22
This work was supported by the National Key R&D Program of China (2017YFB0202600), Natural
23
Science Foundation of China (21402243, 21572279, 81522041, 81373258, and 81602968), Science
24
Foundation of Guangdong Province (2014A020210009 and 2016A030310144), Guangdong
ACS Paragon Plus Environment 38
Page 38 of 61
Page 39 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
Province Higher Vocational Colleges & Schools Pearl River Scholar Funded Scheme (2016), and
2
Foundation of Traditional Chinese medicine Bureau of Guangdong Province (20171049).
3 4
Notes
5
The authors declare no competing financial interest.
6 7
Biographies
8
Yinuo WU is currently an assistant professor in the School of Pharmaceutical Science at Sun Yat-sen
9
University (SYSU). She received her PhD degree at SYSU in 2011 and subsequently went to Hong
10
Kong Polytechnic University as a postdoctoral fellow. Her research encompasses discovery of novel
11
PDEs inhibitors and their applications in the treatment of neurological disorders.
12
Zhe LI is currently an assistant professor in the School of Pharmaceutical Science at Sun Yat-sen
13
University (SYSU). He received his PhD degree in Medicinal Chemistry from SYSU in 2017.
14
During his PhD study, he went to Prof. Chang-Guo Zhan’s group as an exchange student in
15
University of Kentucky for two years. His research in Medicinal Chemistry focuses on the
16
development of drug design methods and the applications of these methods in the development of
17
novel PDEs inhibitors.
18
Yi-You HUANG is currently pursuing his Ph.D in the School of Pharmaceutical Science at Sun
19
Yat-sen University under the supervision of Prof. Hai-Bin Luo. He went to Prof. Hengming Ke’s
20
group as a visiting scholar in the University of North Carolina at Chapel Hill from 2017.04 to
21
2017.10. His research focuses in structural biology and their applications on the design of novel PDE
22
inhibitors.
23
Deyan WU is currently an assistant professor in the School of Pharmaceutical Science at Sun
24
Yat-sen University (SYSU). He received her PhD degree at East China University of Science &
25
Technology in 2014 and subsequently worked in ChemPartner as a senior researcher. After one-year
26
research, he joined Sun Yat-sen University as an assistant professor. His research currently ACS Paragon Plus Environment 39
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 40 of 61
1
specialized in medicinal chemistry and total synthesis of natural products, focusing on the research
2
and development of PDEs inhibitors for Treatment of Pulmonary Arterial Hypertension (PAH),
3
erectile dysfunction (ED), chronic obstructive pulmonary disease (COPD), and structural
4
modification of active natural products as novel PDEs inhibitors.
5
Hai-Bin LUO is currently a full professor and the associate dean of the School of Pharmaceutical
6
Science at Sun Yat-sen University (SYSU). He received his bachelor degree from Xiamen university
7
in 1999 and PhD degree from Hong Kong Baptist university in 2005. After one-year postdoctoral
8
research, he joined Sun Yat-sen University as an associate professor. His research currently
9
encompasses the rational design and discovery of novel PDEs inhibitors and their applications in the
10
treatment of several diseases.
11 12
References
13
(1)
neurodegenerative diseases. Lancet Neurol. 2017, 16, 323-332.
14 15
Stopschinski, B. E.; Diamond, M. I. The prion model for progression and diversity of
(2)
Alzheimer's
Association
(USA).
World
Alzheimer
Report,
2015,
http://news.medlive.cn/uploadfile/20150902/14411602338388.pdf.
16 17
(3)
Goedert, M.; Spillantini, M. G. A century of Alzheimer's disease. Science 2006, 314, 777-781.
18
(4)
Huang, Y.; Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 2012, 148, 1204-1222.
19 20
(5)
2016, 540, 15-16.
21 22
Abbott, A.; Dolgin, E. Failed Alzheimer's trial does not kill leading theory of disease. Nature
(6)
Mehta, D.; Jackson, R.; Paul, G.; Shi, J.; Sabbagh, M. Why do trials for Alzheimer’s disease
23
drugs keep failing? A discontinued drug perspective for 2010-2015. Expert Opin. Inv. Drug.
24
2017, 26, 735-739.
25 26
(7)
Mosconi, L.; Berti, V.; Glodzik, L.; Pupi, A.; De Santi, S.; de Leon, M. J. Pre-clinical detection of Alzheimer's disease using FDG-PET, with or without amyloid imaging. J. Alzheimer's Dis. ACS Paragon Plus Environment 40
Page 41 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
2010, 20, 843-854.
1 2
(8)
clinical use. Pharmacol. Rev. 2006, 58, 488-520.
3 4
Bender, A. T.; Beavo, J. A. Cyclic nucleotide phosphodiesterases: molecular regulation to
(9)
Maurice, D. H.; Ke, H.; Ahmad, F.; Wang, Y.; Chung, J.; Manganiello, V. C. Advances in
5
targeting cyclic nucleotide phosphodiesterases. Nat. Rev. Drug Discov. 2014, 13, 290-314.
6
(10) Azevedo, M. F.; Faucz, F.R.; Bimpaki, E.; Horvath, A.; Levy, I.; de Alexandre, R. B.; Ahmad,
7
F.; Manganiello, V.; Stratakis, C. A. Clinical and molecular genetics of the phosphodiesterases
8
(PDEs). Endoc. Rev. 2014, 35, 195-233.
9
(11) Wang, Z.; Zhang, Y.; Zhang, H. T.; Li, Y. F. Phosphodiesterase: an interface connecting
10
cognitive deficits to neuropsychiatric and neurodegenerative diseases. Curr. Pharm. Design
11
2015, 21, 303-316.
12 13
(12) Lu, Y. F.; Hawkins, R. D. Ryanodine receptors contribute to cGMP-induced late-phase LTP and CREB phosphorylation in the hippocampus. J. Neurophysiol. 2002, 88, 1270-1278.
14
(13) Scott Bitner, R. Cyclic AMP response element-binding protein (CREB) phosphorylation: a
15
mechanistic marker in the development of memory enhancing Alzheimer’s disease therapeutics.
16
Biochem. Pharmacol. 2011, 83, 705-714.
17 18
(14) Domek-Lopacinska, K. U.; Strosznajder, J. B. Cyclic GMP and nitric oxide synthase in aging and Alzheimer’s disease. Mol. Neurobiol. 2010, 41, 129-137.
19
(15) Ciani, E.; Guidi, S.; Bartesaghi, R.; Contestabile, A. Nitric oxide regulates cGMP-dependent
20
cAMP-responsive element binding protein phosphorylation and Bcl-2 expression in cerebellar
21
neurons: implication for a survival role of nitric oxide. J. Neurochem. 2002, 82, 1282-1289.
22
(16) Barad, M.; Bourtchouladze, R.; Winder, D. G.; Golan, H.; Kandel, E. Rolipram, a type
23
IV-specific phosphodiesterase inhibitor, facilitates the establishment of long-lasting LTP and
24
improves memory. Proc. Natl. Acad. Sci. U.S.A. 1998, 95, 15020-15025.
25
(17) Sharma, R. K.; Das, S. B.; Lakshmikuttyamma, A.; Selvakumar, P.; Shrivastav, A. Regulation
26
of calmodulin-stimulated cyclic nucleotide phosphodiesterase (PDE1): Review. Int. J. Mol.
ACS Paragon Plus Environment 41
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Page 42 of 61
Med. 2006, 18, 95-105.
2
(18) Lakics, V.; Karran, E. H.; Boess, F. G. Quantitative comparison of phosphodiesterase mRNA
3
distribution in human brain and peripheral tissues. Neuropharmacology 2010, 59, 367-374.
4
(19) Wennogle, L. Phosphodiesterase 1: a unique therapeutic target with multiple potential
5
indications. Chin. J. Pharmacol. Toxicol. 2017, 31, 452.
6
(20) Bonoczk, P.; Gulyas, B.; Adam-ViziNemes, A.; Karpati, E.; Kiass, B.; Kapas, M.; Szantay, C.;
7
Koncz, I.; Zelles, T.; Vas, A. Role of sodium channel inhibition in neuroprotection: effect of
8
vinpocetine. Brain Res. Bull. 2000, 53, 245-254.
9
(21) Vas, A.; Gulyas, B. Eburnamine derivatives and the brain. Med. Res. Ev. 2005, 25, 737-757.
10
(22) Deshmukh, R.; Sharma, V.; Mehan, S.; Sharma, N.; Bedi, K. L. Amelioration of
11
intracerebroventricular streptozotocin induced cognitive dysfunction and oxidative stress by
12
vinpocetine - a PDE1 inhibitor. Eur. J. Pharmacol. 2009, 620, 49-56.
13
(23) Polgar, M.; Vereczkey, L.; Nyary, I. Pharmacokinetics of vinpocetine and its metabolite,
14
apovincaminic acid, in plasma and cerebrospinal fluid after intravenous infusion. J. Pharm.
15
Biomed. Anal. 1985, 3, 131-139.
16
(24) Hindmarch, I.; Fuchs, H. H.; Erzigkeit, H. Efficacy and tolerance of vinpocetine in ambulant
17
patients
suffering
from
mild
18
Psychopharmacol. 1991, 6, 31-43.
to
moderate
organic
psychosyndromes.
Int.
Clin.
19
(25) Li, P.; Zheng, H.; Zhao, J.; Zhang, L.; Yao, W.; Zhu, H.; Beard, J. D.; Ida, K.; Lane, W.; Snell,
20
G.; Sogabe, S.; Heyser, C. J.; Snyder, G. L.; Hendrick, J. P.; Vanover, K. E.; Davis, R. E.;
21
Wennogle, L. P. Discovery of potent and selective inhibitors of phosphodiesterase 1 for the
22
treatment of cognitive impairment associated with neurodegenerative and neuropsychiatric
23
diseases. J. Med. Chem. 2016, 59, 1149-1164.
24
(26) Snyder, G. L.; Prickaerts, J.; Wadenberg, M. L.; Zhang, L.; Zheng, H.; Yao, W.; Akkerman, S.;
25
Zhu, H.; Hendrick, J. P.; Vanover, K. E.; Davis, R.; Li, P.; Mates, S.; Wennogle, L. P.
26
Preclinical profile of ITI-214, an inhibitor of phosphodiesterase 1, for enhancement of memory
ACS Paragon Plus Environment 42
Page 43 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3
Journal of Medicinal Chemistry
performance in rats. Psychopharmacology 2016, 233, 3113-3124. (27) Clinical
trial
study
found
in
clinical
trials
for
ITI-214:
NCT01900522,
2017,
https://clinicaltrials.gov/ct2/show/NCT01900522?term=NCT01900522&rank=1)
4
(28) Dyck, B.; Branstetter, B.; Gharbaoui, T.; Hudson, A. R.; Breitenbucher, J. G.; Gomez, L.;
5
Cedervall, E. P. Discovery of selective phosphodiesterase 1 inhibitors with memory enhancing
6
properties. J. Med. Chem. 2017, 60, 3472-3483.
7
(29) Stangherlin, A.; Gesellchen, F.; Zoccarato, A.; Terrin, A.; Fields, L. A.; Berrera, M.; Surdo, N.
8
C.; Craig, M. A.; Smith, G.; Hamilton, G.; Zaccolo, M., cGMP signals modulate cAMP levels
9
in a compartment-specific manner to regulate catecholamine-dependent signaling in cardiac
10
myocytes. Circ. Res. 2011, 108, 929-939.
11
(30) Boss, F. G.; Hendrix, M.; Konig, G.; Niewohner, U.; Schlemmer, K.-H.; Schreiber, R.; Van Der
12
Staay, F. J.; Schauss, D. Selective Phosphodiesterase 2 Inhibitors Used as Medicaments for
13
Improving Cognition. 2002, WO2002009713A2.
14
(31) Boess, F. G.; Hendrix, M.; van der Staay, F. J.; Erb, C.; Schreiber, R.; van Staveren, W.; de
15
Vente, J.; Prickaerts, J.; Blokland, A.; Koenig, G. Inhibition of phosphodiesterase 2 increases
16
neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology 2004, 47,
17
1081-1092.
18
(32) Rutten, K.; Prickaerts, J.; Hendrix, M.; van der Staay, F. J.; Sik, A.; Blokland, A.
19
Time-dependent involvement of cAMP and cGMP in consolidation of object memory: studies
20
using selective phosphodiesterase type 2, 4 and 5 inhibitors. Eur. J. Pharmacol. 2007, 558,
21
107-112.
22
(33) Domek-Lopacinska, K.; Strosznajder, J. B. The effect of selective inhibition of cyclic GMP
23
hydrolyzing phosphodiesterases 2 and 5 on learning and memory processes and nitric oxide
24
synthase activity in brain during aging. Brain Res. 2008, 1216, 68-77.
25
(34) Rutten, K.; Van Donkelaar, E. L.; Ferrington, L.; Blokland, A.; Bollen, E.; Steinbusch, H. W.;
26
Kelly, P. A.; Prickaerts, J. H. Phosphodiesterase inhibitors enhance object memory independent
ACS Paragon Plus Environment 43
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
of cerebral blood flow and glucose utilization in rats. Neuropsychopharmacology 2009, 34,
2
1914-1925.
3
(35) Sierksma, A. S.; Rutten, K.; Sydlik, S.; Rostamian, S.; Steinbusch, H. W.; van den Hove, D. L.;
4
Prickaerts, J. Chronic phosphodiesterase type 2 inhibition improves memory in the
5
APPswe/PS1dE9 mouse model of Alzheimer's disease. Neuropharmacology 2013, 64,
6
124-136.
7
(36) Xu, Y.; Pan, J.; Sun, J.; Ding, L.; Ruan, L.; Reed, M.; Yu, X.; Klabnik, J.; Lin, D.; Li, J.; Chen,
8
L.; Zhang, C.; Zhang, H.; O'Donnell, J. M. Inhibition of phosphodiesterase 2 reverses impaired
9
cognition and neuronal remodeling caused by chronic stress. Neurobio. Aging 2015, 36,
10
955-970.
11
(37) Reneerkens, O. A.; Rutten, K.; Bollen, E.; Hage, T.; Blokland, A.; Steinbusch, H. W.;
12
Prickaerts, J. Inhibition of phoshodiesterase type 2 or type 10 reverses object memory deficits
13
induced by scopolamine or MK-801. Behavioural Brain Res. 2013, 236, 16-22.
14
(38) Abarghaz, M.; Biondi, S.; Duranton, J.; Limanton, E.; Mondadori, C.; Wagner, P. Benzo-1,
15
4-diazepin-2-one Derivatives as Phosphodiesterase 2 Inhibitors, Preparation and Therapeutic
16
Use Thereof. 2005, WO2005063723.
17 18 19 20
(39) Gomez, L.; Breitenbucher, J. G. PDE2 inhibition: potential for the treatment of cognitive disorders. Bioorg. Med. Chem. Lett. 2013, 23, 6522-6527. (40) Helal, C. J.; Chappie, T. A.; Humphrey, J. M.; Verhoest, P. R.; Yang, E. Imidazo[5, 1-f][1, 2, 4] triazines for the Treatment of Neurological Disorders. 2012, US 20120214791.
21
(41) Fernandez-Fernandez, D.; Rosenbrock, H.; Kroker, K. S. Inhibition of PDE2A, but not PDE9A,
22
modulates presynaptic short-term plasticity measured by paired-pulse facilitation in the CA1
23
region of the hippocampus. Synapse 2015, 69, 484-496.
24
(42) Helal, C. J. Identification of a brain penetrant, highly selective phosphodiesterase 2A inhibitor
25
for the treatment of cognitive impairment associated with schizophrenia (CIAS) Abstracts of
26
Papers, 244th ACS National Meeting & Exposition, Philadelphia, PA, United States, August
ACS Paragon Plus Environment 44
Page 44 of 61
Page 45 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2
Journal of Medicinal Chemistry
19-23, 2012. (43) Clinical
trial
study
found
in
clinical
trials
for
PF-05180999:
NCT01429740,
3
https://clinicaltrials.gov/ct2/show/NCT01429740?term=PF-05180999&rank=2 (accessed July
4
24, 2017).
5
(44) Clinical
trial
study
found
in
clinical
trials
for
PF-05180999:
NCT01530529,
6
https://clinicaltrials.gov/ct2/show/NCT01530529?term=PF-05180999&rank=3 (accessed July
7
24, 2017).
8
(45) Helal, C. J.; Arnold, E. P.; Boyden, T. L.; Chang, C.; Chappie, T. A.; Fennell, K. F.; Forman, M.
9
D.; Hajos, M.; Harms, J. F.; Hoffman, W. E.; Humphrey, J. M.; Kang, Z.; Kleiman, R. J.;
10
Kormos, B. L.; Lee, C. W.; Lu, J.; Maklad, N.; McDowell, L.; Mente, S.; O'Connor, R. E.;
11
Pandit, J.; Piotrowski, M.; Schmidt, A. W.; Schmidt, C. J.; Ueno, H.; Verhoest, P. R.; Yang, E.
12
X. Application of structure-based design and parallel chemistry to identify a potent, selective,
13
and brain penetrant phosphodiesterase 2A inhibitor. J. Med. Chem. 2017, 60, 5673-5698.
14
(46) Clinical
trial
study
found
in
clinical
trials
for
TAK-915:
NCT02584569,
15
https://clinicaltrials.gov/ct2/show/NCT02584569?term=TAK-915&rank=1 (accessed July 24,
16
2017).
17
(47) Clinical
trial
study
found
in
clinical
trials
for
TAK-915:
NCT02461160,
18
https://clinicaltrials.gov/ct2/show/NCT02461160?term=NCT02461160&rank=1 (accessed July
19
24, 2017).
20 21
(48) Movsesian, M. Novel approaches to targeting PDE3 in cardiovascular disease. Pharmacol.
Therapeut. 2016, 163, 74-81.
22
(49) Tokuchi, R.; Hishikawa, N.; Kurata, T.; Sato, K.; Kono, S.; Yamashita, T.; Deguchi, K.; Abe, K.
23
Clinical and demographic predictors of mild cognitive impairment for converting to
24
Alzheimer's disease and reverting to normal cognition. J. Neurol. Sci. 2014, 346, 288-292.
25
(50) Jellinger, K. A.; Attems, J. Prevalence of dementia disorders in the oldest-old: an autopsy study.
26
Acta. Neuropathol. 2010, 119, 421-433.
ACS Paragon Plus Environment 45
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3 4 5 6
(51) Tanzi, R. E.; Moir, R. D.; Wagner, S. L. Clearance of Alzheimer's Abeta peptide: the many roads to perdition. Neuron 2004, 43, 605-608. (52) Kalaria, R. N. The role of cerebral ischemia in Alzheimer’s disease. Neurobiol. Aging 2000, 21, 321-330. (53) Hong, K.W.; Lee, J. H.; Kima, K.Y.; Park, S.Y.; Lee, W. S. Cilostazol: therapeutic potential against focal cerebral ischemic damage. Curr Pharm Des. 2006, 12, 565-573.
7
(54) Hiramatsu, M.; Takiguchi, O.; Nishiyama, A.; Mori, H. Cilostazol prevents amyloid beta
8
peptide(25-35)-induced memory impairment and oxidative stress in mice. Brit. J. Pharmacol.
9
2010, 161, 1899-1912.
10
(55) Park, S. H.; Kim, J. H.; Bae, S. S.; Hong, K. W.; Lee, D. S.; Leem, J. Y.; Choi, B. T.; Shin, H.
11
K. Protective effect of the phosphodiesterase III inhibitor cilostazol on amyloid beta-induced
12
cognitive deficits associated with decreased amyloid beta accumulation. Biochem. Bioph. Res.
13
Commun. 2011, 408, 602-608.
14
(56) Arai, H.; Takahashi, T. A combination therapy of donepezil and cilostazol for patients with
15
moderate Alzheimer disease: pilot follow-up study. Am. J. Geriatr. Psychiatry. 2009, 17,
16
353-354.
17
(57) Ihara, M.; Nishino, M.; Taguchi, A.; Yamamoto, Y.; Hattori, Y.; Saito, S.; Takahashi1, Y.; Tsuji,
18
M.; Kasahara, Y.; Takata, Y.; Okada, M. Cilostazol add-on therapy in patients with mild
19
dementia receiving donepezil: a retrospective study. PLoS One 2014, 9, e89516.
20
(58) Taguchi, A.; Takata, Y.; Ihara, M.; Kasahara, Y.; Tsuji, M.; Nishino, M.; Stern, D.; Okada, M.
21
Cilostazol improves cognitive function in patients with mild cognitive impairment: a
22
retrospective analysis. Psychogeriatrics 2013, 13, 164-169.
23
(59) Hishikawa, N.; Fukui, Y.; Sato, K.; Ohta, Y.; Yamashita, T.; Abe, K. Comprehensive effects of
24
galantamine and cilostazol combination therapy on patients with Alzheimer's disease with
25
asymptomatic lacunar infarction. Geriatr. Gerontol. Int. 2016, 17(10),1384-1391..
26
(60) Tai, S. Y.; Chien, C. Y.; Chang, Y. H.; Yang, Y. H. Cilostazol use is associated with reduced risk
ACS Paragon Plus Environment 46
Page 46 of 61
Page 47 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
of
2
10.1007/s13311-017-0512-4.
3
dementia:
(61) Clinical
trial
a
nationwide
study
4
https://clinicaltrials.gov/
5
(accessed July 24, 2017).
6
(62) Clinical
trial
study
found
cohort
in
clinical
study.
Neurotherapeutics
trials
for
ct2/show/NCT01409564?term
found
in
clinical
trials
for
Cilostazol:
2017,
NCT01409564,
=Cilostazol&draw=3
Cilostazol:
&rank=12
NCT02491268,
7
https://clinicaltrials.gov/ct2/show/NCT02491268?term=Cilostazol&draw=3&rank=37
8
(accessed July 24, 2017).
9
doi:
(63) McLachlan, C. S.; Chen, M. L.; Lynex, C. N.; Goh, D. L.; Brenner, S.; Tay, S. K.
Changes in
10
PDE4D isoforms in the hippocampus of a patient with advanced Alzheimer disease, Arch.
11
Neurol. 2007, 64, 456-457.
12
(64) Spina, D. PDE4 inhibitors: current status. Br. J. Pharmacol.2008, 155, 308-315.
13
(65) Krause, W.; Kühne, G. Pharmacokinetics of rolipram in the rhesus and cynomolgus monkeys,
14
the rat and the rabbit. Studies on species differences. Xenobiotica 1998, 18, 561-571.
15
(66) Gong, B.; Vitolo, O. V.; Trinchese, F.; Liu, S.; Shelanski, M.; Arancio, O. Persistent
16
improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram
17
treatment. J. Clin. Invest. 2004, 114, 1624-1634.
18
(67) Smith, D. L.; Pozueta, J.; Gong, B.; Arancio, O.; Shelanski, M. Reversal of long-term dendritic
19
spine alterations in Alzheimer disease models. Proc. Natl. Acad. Sci. U.S.A. 2009, 106,
20
16877-16882.
21
(68) Cheng, Y. F.; Wang, C.; Lin, H. B.; Li, Y. F.; Huang, Y.; Xu, J. P.; Zhang, H. T. Inhibition of
22
phosphodiesterase-4 reverses memory deficits produced by Aβ25–35 or Aβ1-40 peptide in rats.
23
Psychopharmacology 2010, 212, 181-191.
24
(69) Wang, C.; Yang, X. M.; Zhuo, Y. Y.; Zhou, H.; Lin, H. B.; Cheng, Y. F.; Xu, J. P.; Zhang, H. T.
25
The phosphodiesterase-4 inhibitor rolipram reverses Aβ-induced cognitive impairment and
26
neuroinflammatory and apoptotic responses in rats. Int. J. Neuropsychoph. 2012, 15, 749-766.
ACS Paragon Plus Environment 47
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 48 of 61
1
(70) Rutten, K.; Prickaerts, J.; Blokland, A. Rolipram reverses scopolamine-induced and
2
time-dependent memory deficits in object recognition by different mechanisms of action.
3
Neurobiol. Learn. Mem. 2006, 85, 132-138.
4
(71) Zhuo, Y.; Guo, H.; Cheng, Y.; Wang, C.; Wang, C.; Wu, J.; Zou, Z.; Gan, D.; Li, Y.; Xu, J.
5
Inhibition of phosphodiesterase-4 reverses the cognitive dysfunction and oxidative stress
6
induced by Abeta25-35 in rats. Metab. Brain Dis. 2016, 31, 779-791.
7
(72) Kumar, A.; Singh, N. Inhibitor of phosphodiestearse-4 improves memory deficits, oxidative
8
stress, neuroinflammation and neuropathological alterations in mouse models of dementia of
9
Alzheimer’s Type. Biomed. Pharmacother. 2017, 88, 698-707.
10
(73) de Lima, M. N.; Presti-Torres, J.; Garcia, V. A.; Guimaraes, M. R.; Scalco, F. S.; Roesler, R.;
11
Schroder, N. Amelioration of recognition memory impairment associated with iron loading or
12
aging by the type 4-specific phosphodiesterase inhibitor rolipram in rats. Neuropharmacology
13
2008, 55, 788-792.
14
(74) Rabe, K. F.; Bateman, E. D.; O'Donnell, D.; Witte, S.; Bredenbröker, D.; Bethke, T. D.
15
Roflumilast-an oral anti-inflammatory treatment for chronic obstructive pulmonary disease: a
16
randomised controlled trial. The Lancet 2005, 366, 563-571.
17
(75) Vanmierlo, T.; Creemers, P.; Akkerman, S.; van Duinen, M.; Sambeth, A.; De Vry, J.;Uzd, T.;
18
Bloklandc, A.; Prickaerts, J. The PDE4 inhibitor roflumilast improves memory in rodents at
19
non-emetic doses. Behav. Brain Res. 2016, 303, 26-33.
20
(76) Clinical
trial
study
found
in
clinical
trials
for
roflumilast,
NCT01433666,
21
https://clinicaltrials.gov/ct2/show/NCT01433666?term=NCT01433666&rank=1 (accessed July
22
24, 2017).
23
(77) Clinical
trial
study
found
in
clinical
trials
for
roflumilast,
NCT02051335,
24
https://clinicaltrials.gov/ct2/show/NCT02051335?term=NCT02051335&rank=1 (accessed July
25
24, 2017).
26
(78) Clinical
trial
study
found
in
clinical
trials
ACS Paragon Plus Environment 48
for
roflumilast,
NCT02835716,
Page 49 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
https://clinicaltrials.gov/ct2/show/NCT02835716?term=NCT02835716&rank=1(accessed July
2
24, 2017).
3
(79) Jabaris, S. S.; Sumathy, H.; Girish, R.; Narayanan, S.; Sugumar, M.; Saravana Babu, C.;
4
Thanikachalam, S.; Thanikachalam, M. Phosphodiesterase-4 inhibitors ameliorates cognitive
5
deficits in deoxycorticosterone acetate induced hypertensive rats via cAMP/ CREB signaling
6
system. Brain Res. 2015, 1622, 279-291.
7
(80) Jabaris, S. G.; Sumathy, H.; Kumar, R. S.; Narayanan, S.; Thanikachalam, S.; Babu, C. S.
8
Effects of rolipram and roflumilast, phosphodiesterase-4 inhibitors, on hypertension-induced
9
defects in memory function in rats. Eur. J. Pharmacol. 2015, 746, 138-147.
10
(81) Bourtchouladze, R.; Lidge, R.; Catapano, R.; Stanley, J.; Gossweiler, S.; Romashko, D.; Scott,
11
R.; Tully, T., A mouse model of Rubinstein-Taybi syndrome: defective long-term memory is
12
ameliorated by inhibitors of phosphodiesterase 4. Proc. Natl. Acad. Sci. U. S. A. 2003, 100,
13
10518-10522.
14
(82) Peters, M.; Bletsch, M.; Stanley, J.; Wheeler, D.; Scott, R.; Tully, T. The PDE4 inhibitor
15
HT-0712 improves hippocampus-dependent memory in aged mice. Neuropsychopharmacology
16
2014, 39, 2938-2948.
17
(83) Clinical
trial
study
found
in
clinical
trials
for
HT-0712,
NCT02013310,
18
https://clinicaltrials.gov/ct2/show/NCT02013310?term=NCT02013310&rank=1 (accessed July
19
24, 2017).
20
(84) Gallant, M.; Aspiotis, R.; Day, S.; Dias, R.; Dube, D.; Dube, L.; Friesen, R. W.; Girard, M.;
21
Guay, D.; Hamel, P.; Huang, Z.; Lacombe, P.; Laliberte, S.; Levesque, J. F.; Liu, S.; Macdonald,
22
D.; Mancini, J.; Nicholson, D. W.; Styhler, A.; Townson, K.; Waters, K.; Young, R. N.; Girard,
23
Y. Discovery of MK-0952, a selective PDE4 inhibitor for the treatment of long-term memory
24
loss and mild cognitive impairment. Bioorg. Med. Chem. Lett. 2010, 20, 6387-6393.
25 26
(85) Clinical
trial
study
found
in
clinical
trials
for
MK-0952,
NCT00362024,
https://clinicaltrials.gov/ct2/show/NCT00362024?term=MK-0952&rank=1 (accessed July 24,
ACS Paragon Plus Environment 49
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2 3
Page 50 of 61
2017). (86) Drott, J.; Desire, L.; Drouin, D.; Pando, M.; Haun, F. Etazolate improves performance in a foraging and homing task in aged rats. Eur. J. Pharmacol. 2010, 634, 95-100.
4
(87) Mattson, M. P. Secreted forms of β-amyloid precursor protein modulate dendrite outgrowth
5
and calcium responses to glutamate in cultured embryonic hippocampal neurons. Dev.
6
Neurobiol. 1994, 25, 439-450.
7
(88) Clinical
trial
study
found
in
clinical
trials
for
Etazolate,
NCT00880412,
8
https://clinicaltrials.gov/ct2/results?cond=&term=NCT00880412&cntry1=&state1=&Search=S
9
earch (accessed July 24, 2017).
10
(89) Vellas, B.; Sol, O. J.; Snyder, P.; Ousset, P. J.; Haddad, R.; Maurin, M.; Lemarie, J. C.; Desire,
11
L.; P. Pando, M. EHT0202 in Alzheimer's disease: a 3-month, randomized, placebo-controlled,
12
double-blind study. Curr. Alzheimer Res. 2011, 8, 203-212.
13
(90) Zheng, S.; Kaur, G.; Wang, H.; Li, M.; Macnaughtan, M.; Yang, X.; Reid, S.; Prestegard, J.;
14
Wang, B.; Ke, H. Design, synthesis, and structure-activity relationship, molecular modeling,
15
and NMR studies of a series of phenyl alkyl ketones as highly potent and selective
16
phosphodiesterase-4 inhibitors. J. Med. Chem. 2008, 51, 7673-7688.
17
(91) Guo, H.; Cheng, Y.; Wang, C.; Wu, J.; Zou, Z.; Niu, B.;Yu, H.; Wang, H.; Xu, J. FFPM, a
18
PDE4 inhibitor, reverses learning and memory deficits in APP/PS1 transgenic mice via
19
cAMP/PKA/CREB signaling and anti-inflammatory effects. Neuropharmacology 2017, 116,
20
260-269.
21
(92) Li, Y. F.; Cheng, Y. F.; Huang, Y.; Conti, M.; Wilson, S. P.; O'Donnell, J. M.; Zhang, H. T.
22
Phosphodiesterase-4D knock-out and RNA interference-mediated knock-down enhance
23
memory and increase hippocampal neurogenesis via increased cAMP signaling. J. Neurosci.
24
2011, 31, 172-183.
25
(93) Robichaud, A.; Stamatiou, P. B.; Jin, S. L. C.; Lachance, N.; MacDonald, D.; Laliberté, F.; Liu,
26
S.; Huang, Z.; Conti, M.; Chan, C.-C. Deletion of phosphodiesterase 4D in mice shortens
ACS Paragon Plus Environment 50
Page 51 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
α2-adrenoceptor-mediated anesthesia, a behavioral correlate of emesis. J. Clin. Invest. 2002,
2
110, 1045-1052.
3
(94) Robichaud, A.; Savoie, C.; Stamatiou, P. B.; Lachance, N.; Jolicoeur, P.; Rasori, R.; Chan, C.-C.
4
Assessing the emetic potential of PDE4 inhibitors in rats. Brit. J. Pharmacol. 2002,135,
5
113-118.
6
(95) Bruno, O.; Romussi, A.; Spallarossa, A.; Brullo, C.; Schenone, S.; Bondavalli, F.; Vanthuyne,
7
N.; Roussel, C. New selective phosphodiesterase 4D inhibitors differently acting on long, short,
8
and supershort isoforms. J. Med. Chem. 2009, 52, 6546-6557.
9
(96) Bruno, O.; Fedele, E.; Prickaerts, J.; Parker, L. A.; Canepa, E.; Brullo, C.; Cavallero, A.;
10
Gardella, E.; Balbi, A.; Domenicotti, C.; Bollen, E.; Gijselaers, H. J.; Vanmierlo, T.; Erb, K.;
11
Limebeer, C. L.; Argellati, F.; Marinari, U. M.; Pronzato, M. A.; Ricciarelli, R. GEBR-7b, a
12
novel PDE4D selective inhibitor that improves memory in rodents at non-emetic doses. Brit. J.
13
Pharmacol. 2011, 164, 2054-2063.
14
(97) Brullo, C.; Ricciarelli, R.; Prickaerts, J.; Arancio, O.; Massa, M.; Rotolo, C.; van Hagen, B. T.
15
New insights into selective PDE4D inhibitors: 3-(Cyclopentyloxy)-4-methoxybenzaldehyde
16
O-(2-(2, 6-dimethylmorpholino)-2-oxoethyl) oxime (GEBR-7b) structural development and
17
promising activities to restore memory impairment. Eur. J. Med. Chem. 2016, 124, 82-102.
18
(98) Burgin, A. B.; Magnusson, O. T.; Singh, J.; Witte, P.; Staker, B. L.; Bjornsson, J. M.;
19
Thorsteinsdottir, M.; Hrafnsdottir, S.; Hagen, T.; Kiselyov, A. S.; Stewart, L. J.; Gurney, M. E.
20
Design of phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing cognition with
21
improved safety. Nat. Biotechnol. 2010, 28, 63-70.
22 23 24
(99) Zhang, C.; Xu, Y.; Gurney, M.; O’Donnell, J. Role of phosphodiesterase 4D in memory and cognition. The FASEB Journal 2017, 31, 662-664. (100) Clinical
trial
study
found
in
clinical
trials
for
BPN14770,
NCT02648672,
25
https://clinicaltrials.gov/ct2/show/NCT02648672?term=BPN14770&rank=1 (accessed July 24,
26
2017).
ACS Paragon Plus Environment 51
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
(101) Clinical
trial
study
found
in
clinical
trials
Page 52 of 61
for
BPN14770,
NCT02840279,
2
https://clinicaltrials.gov/ct2/show/NCT02840279?term=BPN14770&rank=2 (accessed July 24,
3
2017).
4
(102) Clinical
trial
study
found
in
clinical
trials
for
BPN14770,
NCT03030105,
5
https://clinicaltrials.gov/ct2/show/NCT03030105?term=BPN14770&rank=3(accessed July 24,
6
2017).
7 8
(103) Wright, P. J. Comparison of phosphodiesterase type 5 (PDE5) inhibitors. Int. J. Clin. Pract. 2006, 60, 967-975.
9
(104) Wang, H.; Liu, Y.; Huai, Q.; Cai, J.; Zoraghi, R.; Francis, S. H.; Corbin, J. D.; Robinson, H.;
10
Xin, Z.; Lin, G.; Ke, H. Multiple conformations of phosphodiesterase-5: implications for
11
enzyme function and drug development. J. Biol. Chem. 2006, 281, 21469-21479.
12
(105) Puzzo, D.; Staniszewski, A.;Deng, S. X.;Privitera, L.; Leznik, E.; Liu, S.; Zhang, H; Feng, Y.;
13
Palmeri, A.; Landry, D. W.; Arancio, O. Phosphodiesterase 5 inhibition improves synaptic
14
function, memory, and amyloid-β load in an Alzheimer's disease mouse model. J. Neurosci.
15
2009, 29, 8075-8086.
16
(106) Zhang, J.; Guo, J.; Zhao, X.; Chen, Z.; Wang, G.;Liu, A.; Wang,Q.; Zhou, W.; Xue, Y.; Wang,
17
C. Phosphodiesterase-5 inhibitor sildenafil prevents neuroinflammation, lowers beta-amyloid
18
levels and improves cognitive performance in APP/PS1 transgenic mice. Behav. Brain Res.
19
2013, 250, 230-237.
20
(107) Cuadrado-Tejedor, M.; Hervias, I.; Ricobaraza, A.; Puerta, E.; Pérez-Roldán, J. M.;
21
García-Barroso, C.; Franco, R.; Aguirre, N.; García-Osta, A. sildenafil restores cognitive
22
function without affecting β-amyloid burden in a mouse model of Alzheimer's disease. Brit. J.
23
Pharmacol. 2011, 164, 2029-2041.
24
(108) Orejana, L.; Barros-Minones, L.; Jordan, J.; Puerta, E.; Aguirre, N. Sildenafil ameliorates
25
cognitive deficits and tau pathology in a senescence-accelerated mouse model. Neurobio.
26
Aging 2012, 33, 625 e11-e20.
ACS Paragon Plus Environment 52
Page 53 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Journal of Medicinal Chemistry
(109)
Orejana, L.; Barros-Minones, L.; Jordan, J.; Cedazo-Minguez, A.; Tordera, R. M.; Aguirre,
2
N.; Puerta, E. Sildenafil decreases BACE1 and cathepsin B levels and reduces APP
3
Amyloidogenic processing in the SAMP8 Mouse. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70,
4
675-685.
5
(110) Clinical
trial
study
found
in
clinical
trials
for
sildenafil,
NCT00455715,
6
https://clinicaltrials.gov/ct2/show/NCT00455715?term=sildenafil&draw=3&rank=12 (accessed
7
July 24, 2017).
8 9 10
(111) Clinical
trial
study
found
in
clinical
trials
for
sildenafil,
NCT01941732,
https://clinicaltrials.gov/ct2/show/NCT01941732?term=sildenafil&draw=3&rank=28 (accessed July 24, 2017).
11
(112) Sheng, M.; Lu, H.; Liu, P.; Li, Y.; Ravi, H.; Peng, S. L.; Dia-Arrastia,R.; Devous, M. D.;
12
Womack, K. B. Sildenafil improves vascular and metabolic function in patients with
13
Alzheimer’s Disease. J. Alzheimer's Dis. 2017, 60, 1351-1364.
14
(113) Forgue, S. T.; Patterson, B. E.; Bedding, A. W.; Payne, C. D.; Phillips, D. L.; Wrishko, R. E.;
15
Mitchell, M. I. Tadalafil pharmacokinetics in healthy subjects. Brit. J. Clin. Pharmaco. 2006,
16
61, 280-288.
17
(114) Garcia-Barroso, C.; Ricobaraza, A.; Pascual-Lucas, M.; Unceta, N.; Rico, A. J.; Goicolea, M.
18
A.; Salles, J.; Lanciego, J. L.; Oyarzabal, J.; Franco, R.; Cuadrado-Tejedor, M.; Garcia-Osta, A.
19
Tadalafil crosses the blood-brain barrier and reverses cognitive dysfunction in a mouse model
20
of AD. Neuropharmacology 2013, 64, 114-123.
21
(115) Clinical
trial
study
found
in
clinical
trials
for
sildenafil,
NCT02450253,
22
https://clinicaltrials.gov/ct2/show/NCT02450253?term=tadalafil&cond=vascular+dementia&ra
23
nk=1 (accessed Dec. 18, 2017).
24
(116) Fiorito, J.; Saeed, F.; Zhang, H.; Staniszewski, A.; Feng, Y.; Francis, Y. I.; Rao, S.; Thakkar, D.
25
M.; Deng, S. X.; Landry, D. W.; Arancio, O. Synthesis of quinoline derivatives: discovery of a
26
potent and selective phosphodiesterase 5 inhibitor for the treatment of Alzheimer's disease. Eur.
ACS Paragon Plus Environment 53
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
J. Med. Chem. 2013, 60, 285-294.
2
(117) Cuadrado-Tejedor, M.; Garcia-Barroso, C.; Sanzhez-Arias, J.; Mederos, S.; Rabal, O.; Ugarte,
3
A.;Franco, R.; Pascual-Lucas1, M.; Segura, V.; Perea, G.; Oyarzabal, J. Concomitant histone
4
deacetylase and phosphodiesterase 5 inhibition synergistically prevents the disruption in
5
synaptic plasticity and it reverses cognitive impairment in a mouse model of Alzheimer’s
6
disease. Clin. Epigenetics 2015, 7, 108.
7
(118) Rabal, O.; Sánchez-Arias, J. A.; Cuadrado-Tejedor, M.; De Miguel, I.; Pérez-González, M.;
8
Garcia-Barroso, C.; Ugarte, A.; Estella-Hermoso de Mendoza, A.; Sáez, E.; Espelosin, M.;
9
Ursua, S.; Tan, H.; Wu, W.; Xu, M.; Garcia-Osta, A.; Oyarzabal, J. Design, synthesis, and
10
biological evaluation of first-in-class dual acting histone deacetylases (HDACs) and
11
phosphodiesterase 5 (PDE5) inhibitors for the treatment of Alzheimer’s disease. J. Med. Chem.
12
2016, 59, 8967-9004.
13
(119) Cuadrado-Tejedor, M.; Garcia-Barroso, C.; Sanchez-Arias, J. A.; Rabal, O.; Perez-Gonzalez,
14
M.; Mederos, S.; Ugarte, A.; Franco, R.; Segura, V.; Perea, G.; Oyarzabal, J.; Garcia-Osta, A. A
15
first-in-class small-molecule that acts as a dual inhibitor of HDAC and PDE5 and that rescues
16
hippocampal synaptic impairment in Alzheimer's Disease mice. Neuropsychopharmacology
17
2017, 42, 524-539.
18
(120) Perez-Torres, S.; Cortes, R.; Tolnay, M.; Probst, A.; Palacios, J. M.; Mengod, G. Alterations on
19
phosphodiesterase type 7 and 8 isozyme mRNA expression in Alzheimer’s disease brains
20
examined by in situ hybridization. Exp. Neurol. 2003, 182, 322-334
21 22
(121) Gil, C.; Campillo, N. E.; Perez, D. I.; Martinez, A. PDE7 inhibitors as new drugs for neurological and inflammatory disorders. Expert Opin. Ther. Pat. 2008, 18, 1127-1139.
23
(122) Castro, A.; Jerez, M. J.; Gil, C.; Calderon, F.; Domenech, T.; Nueda, A.; Martinez, A. CODES,
24
a novel procedure for ligand-based virtual screening: PDE7 inhibitors as an application
25
example. Eur. J. Med. Chem. 2008, 43, 1349-1359.
26
(123) Paterniti, I.; Mazzon, E.; Gil, C.; Impellizzeri, D.; Palomo, V.; Redondo, M.; Perez, D. I.;
ACS Paragon Plus Environment 54
Page 54 of 61
Page 55 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
Esposito, E.; Martinez, A.; Cuzzocrea, S. PDE 7 inhibitors: new potential drugs for the therapy
2
of spinal cord injury. PloS one 2011, 6, e15937.
3
(124) Perez-Gonzalez, R.; Pascual, C.; Antequera, D.; Bolos, M.; Redondo, M.; Perez, D. I.;
4
Perez-Grijalba, V.; Krzyzanowska, A.; Sarasa, M.; Gil, C.; Ferrer, I.; Martinez, A.; Carro, E.
5
Phosphodiesterase 7 inhibitor reduced cognitive impairment and pathological hallmarks in a
6
mouse model of Alzheimer's disease. Neurobio. Aging 2013, 34, 2133-2145.
7 8
(125) Soderling, S. H.; Bayuga, S. J.; Beavo, J. A. Identification and characterization of a novel family of cyclic nucleotide phosphodiesterases. J. Biol. Chem. 1998, 273, 15553-15558.
9
(126) Wunder, F.; Tersteegen, A.; Rebmann, A.; Erb, C.; Fahrig, T.; Hendrix, M. Characterization of
10
the first potent and selective PDE9 inhibitor using a cGMP reporter cell line. Mol. Pharmacol.
11
2005, 68, 1775-1781.
12
(127) van der Staay, F. J.; Rutten, K.; Barfacker, L.; Devry, J.; Erb, C.; Heckroth, H.; Karthaus, D.;
13
Tersteegen, A.; van Kampen, M.; Blokland, A.; Prickaerts, J.; Reymann, K. G.; Schroder, U. H.;
14
Hendrix, M. The novel selective PDE9 inhibitor BAY 73-6691 improves learning and memory
15
in rodents. Neuropharmacology 2008, 55, 908-918.
16
(128) Kroker, K. S.; Rast, G.; Giovannini, R.; Marti, A.; Dorner-Ciossek, C.; Rosenbrock, H.
17
Inhibition of acetylcholinesterase and phosphodiesterase-9A has differential effects on
18
hippocampal early and late LTP. Neuropharmacology 2012, 62, 1964-1974.
19
(129) Kroker, K. S.; Mathis, C.; Marti, A.; Cassel, J. C.; Rosenbrock, H.; Dorner-Ciossek, C. PDE9A
20
inhibition rescues amyloid beta-induced deficits in synaptic plasticity and cognition. Neurobio.
21
Aging 2014, 35, 2072-2078.
22
(130) Li, J.; Liu, C. N.; Wei, N.; Li, X. D.; Liu, Y. Y.; Yang, R.; Jia, Y. J. Protective effects of BAY
23
73-6691, a selective inhibitor of phosphodiesterase 9, on amyloid-beta peptides-induced
24
oxidative stress in in-vivo and in-vitro models of Alzheimer's disease. Brain Res. 2016, 1642,
25
327-335.
26
(131) Verhoest, P. R.; Fonseca, K. R.; Hou, X.; Proulx-Lafrance, C.; Corman, M.; Helal, C. J.;
ACS Paragon Plus Environment 55
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 56 of 61
1
Claffey, M. M.; Tuttle, J. B.; Coffman, K. J.; Liu, S.; Nelson, F.; Kleiman, R. J.; Menniti, F. S.;
2
Schmidt,
3
6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,
4
5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a selective brain penetrant
5
PDE9A inhibitor for the treatment of cognitive disorders. J. Med. Chem. 2012, 55, 9045-9054.
C.
J.;
Vanase-Frawley,
M.;
Liras,
S.
Design
and
discovery
of
6
(132) Vardigan, J. D.; Converso, A.; Hutson, P. H.; Uslaner, J. M. The selective phosphodiesterase 9
7
(PDE9) inhibitor PF-04447943 attenuates a scopolamine-induced deficit in a novel rodent
8
attention task. J. Neurogenet. 2011, 25, 120-126.
9
(133) Hutson, P. H.; Finger, E. N.; Magliaro, B. C.; Smith, S. M.; Converso, A.; Sanderson, P. E.;
10
Mullins, D.; Hyde, L. A.; Eschle, B. K.; Turnbull, Z.; Sloan, H.; Guzzi, M.; Zhang, X.; Wang,
11
A.; Rindgen, D.; Mazzola, R.; Vivian, J. A.; Eddins, D.; Uslaner, J. M.; Bednar, R.; Gambone,
12
C.; Le-Mair, W.; Marino, M. J.; Sachs, N.; Xu, G.; Parmentier-Batteur, S. The selective
13
phosphodiesterase 9 (PDE9) inhibitor PF-04447943 (6-[(3S,4S)-4-methyl-1 -(pyrimidin-2
14
-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrim
15
idin-4-one) enhances synaptic plasticity and cognitive function in rodents. Neuropharmacology
16
2011, 61, 665-676.
17
(134) Kleiman, R. J.; Lanz, T. A.; Finley, J. E.; Bove, S. E.; Majchrzak, M. J.; Becker, S. L.;
18
Carvajal-Gonzales, S.; Kuhn, A. M.; Wood, K. M.; Mariga, A.; Nelson, F. R.; Verhoest, P. R.;
19
Seymour, P. A.; Stephenson, D. T., Dendritic spine density deficits in the hippocampal CA1
20
region of young Tg2576 mice are ameliorated with the PDE9A inhibitor PF-04447943.
21
Alzheimer's Dement. 2010, 6, S563-S564.
22
(135) Clinical
trial
study
found
in
clinical
trials
for
PF-04447943,
NCT00988598,
23
https://clinicaltrials.gov/ct2/show/NCT00988598?term=PF-04447943&rank=6 (accessed July
24
24, 2017).
25 26
(136) Clinical
trial
study
found
in
clinical
trials
for
PF-04447943,
NCT00930059,
https://clinicaltrials.gov/ct2/show/NCT00930059?term=PF-04447943&rank=3 (accessed July
ACS Paragon Plus Environment 56
Page 57 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2
Journal of Medicinal Chemistry
24, 2017). (137) Clinical
trial
study
found
in
clinical
trials
for
BI-409306,
NCT02337907,
3
https://clinicaltrials.gov/ct2/show/NCT02337907?term=BI-409306&draw=3&rank=12
4
(accessed July 24, 2017).
5
(138) Clinical
trial
study
found
in
clinical
trials
for
BI-409306,
NCT02240693,
6
https://clinicaltrials.gov/ct2/show/NCT02240693?term=BI-409306&draw=3&rank=13
7
(accessed July 24, 2017).
8 9
(139) Meng, F.; Hou, J.; Shao, Y.-X.; Wu, P.-Y.; Huang, M.; Zhu, X.; Cai, Y.; Li, Z.; Xu, J.; Liu, P.; Luo,
H.-B.;
Wan,
Y.;
Ke,
H.
Structure-based
discovery
of
highly
selective
10
phosphodiesterase-9A inhibitors and implications for inhibitor design. J. Med. Chem. 2012, 55,
11
8549-8558.
12
(140) Shao, Y. X.; Huang, M.; Cui, W.; Feng, L. J.; Wu, Y.; Cai, Y.; Li, Z.; Zhu, X.; Liu, P.; Wan, Y.;
13
Ke, H.; Luo, H.-B. Discovery of a phosphodiesterase 9A inhibitor as a potential hypoglycemic
14
agent. J. Med. Chem. 2014, 57, 10304-10313.
15
(141) Huang, M.; Shao, Y.; Hou, J.; Cui, W.; Liang, B.; Huang, Y.; Li, Z.; Wu, Y.; Zhu, X.; Liu, P.;
16
Wan, Y.; Ke, H.; Luo, H.-B. Structural asymmetry of phosphodiesterase-9A and a unique
17
pocket for selective binding of a potent enantiomeric inhibitor. Mol. Pharmacol. 2015, 88,
18
836-845.
19
(142) Li, Z.; Lu, X.; Feng, L.-J.; Gu, Y.; Li, X.; Wu, Y.; Luo, H.-B. Molecular dynamics-based
20
discovery of novel phosphodiesterase-9A inhibitors with non-pyrazolopyrimidinone scaffolds.
21
Mol. BioSyst. 2015, 11, 115-125.
22 23
(143) Soderling, S. H.; Bayuga, S. J.; Beavo J. A. Isolation and characterization of a dual-substrate phosphodiesterase gene family: PDE10A. Proc. Natl. Acad. Sci. U.S.A. 1999, 96, 7071-7076.
24
(144) Hebb, A. L.; Robertson, H. A.; Denovan-Wright, E. M. Striatal phosphodiesterase mRNA and
25
protein levels are reduced in Huntington’s disease transgenic mice prior to the onset of motor
26
symptoms. Neuroscience 2004, 123, 967-981.
ACS Paragon Plus Environment 57
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 58 of 61
1
(145) Verhoest, P. R.; Chapin, D. S.; Corman, M.; Fonseca, K.; Harms, J. F.; Hou, X.; Marr, E. S.;
2
Menniti, F. S.; Nelson, F.; O’Connor, R.; Pandit, J.; Proulx-LaFrance, C.; Schmidt, A. W.;
3
Schmidt, C. J.; Suiciak, J. A.; Liras, S. Discovery of a novel class of phosphodiesterase 10A
4
inhibitors and identification of clinical candidate 2-[4-(1-Methyl-4-pyridin-4-yl-1H-pyrazol-3-
5
yl)-phenoxymethyl]-quinoline (PF-2545920) for the treatment of Schizophrenia. J. Med. Chem.
6
2009, 52, 5188-5196.
7
(146) Clinical
trial
study
found
in
clinical
trials
for
PF-02545920,
NCT02197130,
8
https://clinicaltrials.gov/ct2/show/NCT02197130?term=PF-02545920&draw=1&rank=6,
9
(accessed July 24, 2017).
10
(147) Clinical
trial
study
found
in
clinical
trials
for
PF-02545920,
NCT02342548,
11
https://clinicaltrials.gov/ct2/show/NCT02342548?term=PF-02545920&draw=1&rank=4,
12
(accessed July 24, 2017).
13
(148) Kunitomo, J.; Yoshikawa, M.; Fushimi, M.; Kawada, A.; Quinn, J. F.; Oki, H.; Kokubo, H.;
14
Kondo, M.; Nakashima, K.; Kamiguchi, N.; Suzuki, K.; Kimura, H.; Taniguchi, T. Discovery
15
of 1-[2-fluoro-4-(1H-pyrazol-1-yl)phenyl]-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-
16
4(1H)-one (TAK-063), a highly potent, selective, and orally active phosphodiesterase 10A
17
(PDE10A) inhibitor. J. Med. Chem. 2014, 57, 9627-9643.
18
(149) Suzuki, K.; Harada, A.; Suzuki, H.; Miyamoto, M.; Kimura, H. TAK-063, a PDE10A inhibitor
19
with balanced activation of direct and indirect pathways, provides potent antipsychotic-like
20
effects in multiple paradigms. Neuropsychopharmacol 2016, 41, 2252-2262.
21
(150) Shiraishi, E.; Suzuki, K.; Harada, A.; Suzuki, N.; Kimura, H. The phosphodiesterase 10A
22
selective inhibitor TAK-063 improves cognitive functions associated with Schizophrenia in
23
rodent models. J. Pharmacol. Exp. Ther. 2016, 356, 587-595.
24
(151) Clinical
trial
study
found
in
clinical
trials
for
TAK-063,
NCT02477020,
25
https://clinicaltrials.gov/ct2/show/NCT02477020?term=NCT02477020&rank=1 (accessed July
26
24, 2017).
ACS Paragon Plus Environment 58
Page 59 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Journal of Medicinal Chemistry
1
(152) Leblond, B.; Donello, J. E.; Chauvignac, C.; Taverne, T.; Beausoleil, E.; Casagrande, A. S.;
2
Yang, R. PDE10 Inhibitors and Related Compositions and Methods. 2011, WO 2011112828
3
A1.
4
(153) Clinical
trial
study
found
in
clinical
trials
for
OMS643762,
NCT01952132,
5
https://clinicaltrials.gov/ct2/show/NCT01952132?term=NCT01952132&rank=1 (accessed July
6
24, 2017).
7
(154) Clinical
trial
study
found
in
clinical
trials
for
EVP-6308,
NCT02037074,
8
https://clinicaltrials.gov/ct2/show/NCT02037074?term=NCT02037074&rank=1(accessed July
9
24, 2017).
10 11
(155) Cote, R. H. Characteristics of photoreceptor PDE (PDE6): similarities and differences to PDE5.
Int. J. Impot. Res. 2004, 16, S28–S33.
12
(156) Boolell, M.; Allen, M. J.; Ballard, S. A. Sildenafil, an inhibitor of phosphodiesterase type 5,
13
enhances nitric oxide mediated relaxation of human corpus cavernosum. Int. J. Impot. Res.
14
1996, 8, 103.
15
(157) Amanuma, M.; Yuasa, K.; Sasaki, T.; Comparison of enzymatic characterization and gene
16
organization of cyclic nucleotide phosphodiesterase 8 family in humans. Cell Signal 2003, 15,
17
565-574.
18
(158) Tsai, L. C.; Shimizu-Albergine, M.; Beavo, J. A.; The high-affinity cAMP-specific
19
phosphodiesterase 8B controls steroidogenesis in the mouse adrenal gland. Mol. Pharmacol.
20
2011, 79, 639-648.
21 22 23 24
(159) Shimizu-Albergine, M.; Tsai, L. C.; Patrucco, E. cAMP-specific phosphodiesterases 8A and 8B, essential regulators of Leydig cell steroidogenesis. Mol. Pharmacol. 2012, 81, 556-566. (160) Vang, A. G.; Ben-Sasson, S. Z.; Dong, H. PDE8 regulates rapid Teff cell adhesion and proliferation independent of ICER. PLoS One 2010, 5, e12011.
25 26 27
ACS Paragon Plus Environment 59
Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1 2
Figure captions
3
Figure 1: The relation of PDE inhibition and the cAMP or cGMP signaling pathway in the
4
brain. PDE: phosphodiesterase; AC: adenyl cyclase; GC: guanylyl cyclase; ATP: adenosine
5
triphosphate; GTP: guanosine triphosphate; PKA: protein kinase A; PKG: protein kinase G;
6
P: phosphorylated; CREB: cAMP response element binding protein.
7
Figure 2. Current PDE1 inhibitors for the treatment of AD.
8
Figure 3. Current PDE2 inhibitors for the treatment of AD
9
Figure 4. Selective PDE3 inhibitor, cilostazol, may be used to treat cerebrovascular diseases
10
and AD
11
Figure 5. Current PDE4 inhibitors for the treatment of AD.
12
Figure 6. Current PDE5 inhibitors for the treatment of AD.
13
Figure 7. Current PDE7 inhibitor for the treatment of AD.
14
Figure 8. Current PDE9 inhibitors for the treatment of AD.
15
Figure 9. Current PDE10 inhibitors for the treatment of AD and other diseases.
16 17
ACS Paragon Plus Environment 60
Page 60 of 61
Page 61 of 61 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Journal of Medicinal Chemistry
Table of Contents Graphic
2 3
ACS Paragon Plus Environment 61