You are on page 1of 24

pubs.acs.

org/jmc Perspective

Advances in the Development of Phosphodiesterase‑4 Inhibitors


Ting Peng,# Baowen Qi,# Jun He,# Hengming Ke,* and Jianyou Shi*

Cite This: https://dx.doi.org/10.1021/acs.jmedchem.9b02170 Read Online

ACCESS Metrics & More Article Recommendations


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via CARLETON UNIV on July 17, 2020 at 09:13:26 (UTC).

ABSTRACT: Cyclic nucleotide phosphodiesterase 4 (PDE4) specifically hydrolyzes cyclic adenosine monophosphate (cAMP) and
plays vital roles in biological processes such as cancer development. To date, PDE4 inhibitors have been widely studied as
therapeutics for the treatment of various diseases such as chronic obstructive pulmonary disease, and many of them have progressed
to clinical trials or have been approved as drugs. Herein, we review the advances in the development of PDE4 inhibitors in the past
decade and will focus on their pharmacophores, PDE4 subfamily selectivity, and therapeutic potential. Hopefully, this analysis will
lead to a strategy for development of novel therapeutics targeting PDE4.

1. INTRODUCTION mRNA splicing, which are grouped into 11 families based on


their sequence similarity and biological properties. PDE4, -7,
Cyclic nucleotide phosphodiesterases (PDEs) are a family of
and -8 are cAMP-specific, while PDE5, -6, and -9 specifically
enzymes that uniquely hydrolyze cyclic adenosine mono-
hydrolyze cGMP. PDE1, -2, -3, -10, and -11 show dual activity
phosphate (cAMP) and cyclic guanosine monophosphate
on hydrolysis of both cAMP and cGMP.5
(cGMP) into 5′-AMP and 5′-GMP. The cAMP and cGMP
signaling systems regulate the functions of many physiological
processes, such as central nervous, cardiovascular, and immune 2. OVERVIEW OF PDE4
systems.1−4 Under normal physiological conditions, the intra- PDE4, the most diverse family of PDEs, is abundantly expressed
cellular levels of cAMP and cGMP are determined by the rates of in most cells6 and prefers to hydrolyze cAMP with the
cyclic nucleotide production and decomposition and main- micromolar Km values. PDE4 molecules are involved in a variety
tained in a dynamic equilibrium between synthesis and of physiological processes, including brain function, monocyte
hydrolysis. There are two ways to increase intracellular and macrophage activation, neutrophil infiltration, vascular
cAMP/cGMP concentrations: (a) increasing cAMP/cGMP smooth muscle proliferation, and myocardial contractility
synthesis by stimulating adenylyl/guanylyl cyclases through (Figure 1). PDE4 has been reported to be a target for various
external stimuli such as hormones and neurotransmitters and inflammatory diseases, such as asthma, chronic obstructive
(b) preventing cAMP/cGMP hydrolysis by inhibiting PDEs. pulmonary disease (COPD), and rheumatoid arthritis. In
Since many diseases are associated with low intracellular levels of addition, PDE4 plays important roles in the development of
cAMP or cGMP, inhibition of PDEs would be a preferred cardiovascular diseases, autoimmune diseases, and cancers.7−9
strategy for drug development. PDEs are involved in the
regulation of many physiological and metabolic processes, Received: December 28, 2019
including inflammation, ion channel signaling, cell differ- Published: April 7, 2020
entiation and apoptosis, muscle contraction, lipogenesis,
glycogen synthesis, and gluconeogenesis.1−4 To date, 21 PDE
genes have been identified in the human genome. These genes
express more than 100 PDE isoforms in human tissues through

© XXXX American Chemical Society https://dx.doi.org/10.1021/acs.jmedchem.9b02170


A J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 1. Schematic presentation of PDE4 and cAMP signaling pathway. The following abbreviations are used: β-AR, β-arrestin; GPCRs, G-protein-
coupled receptors; Gs, heterotrimeric guanine nucleotide binding protein; AC, adenylyl cyclase; AKAPs, A-kinase-anchoring proteins; PKA, protein
kinase A; EPAC, exchange protein activated by cAMP; CREB, cAMP response element binding protein; C3G, cyanidin-3-glucoside; Rap1, Ras-
proximate-1; MEK1/2, mitogen-activated protein kinase; ERK1/2, extracellular signal-regulated kinase 1/2; NF-κB, nuclear factor κ-light-chain-
enhancer of activated B-cells; MAPK, mitogen-activated protein kinase; mTORC1, mammalian target of rapamycin complex 1.

In recent years, involvement of PDE4 in regulation of the central


nervous system has received substantial attention.10
PDE4 comprises four subtypes: PDE4A, PDE4B, PDE4C,
and PDE4D, which are located on chromosomes 19p13.2, 1p31,
19p13.11, and 5q12, respectively.11 Each subtype of PDE4 genes
can express 3−11 proteins, resulting in at least 25 different PDE4
isoforms. These PDE4 proteins are located in different cell
compartments and regulated by kinases, playing critical roles in
cell functions (Figure 1). PDE4 molecules exist in long, short,
and supershort forms according to their molecular sizes (Figure
2). The long form of PDE4 has two upstream conserved regions
(UCR1 and UCR2) in the N-terminal region, while the short
form contains only UCR2 and the supershort form has a
truncated UCR2. UCR1 consists of approximate 60 amino acids,
while UCR2 has approximate 80 amino acids. All PDE4 Figure 2. Domain organization of PDE4. UCR: Upstream conserved
molecules have a highly conserved catalytic domain at the C- region.
terminus, which contains 300−350 amino acids.11
X-ray structures of PDE4 molecules have shown that the PDE4s make the discovery of PDE4 subtype-selective inhibitors
active site can be divided into three subpockets (Figure 3): a quite challenging.
divalent metal pocket that interacts with the phosphate moiety The UCRs are the unique motifs of the PDE4 family and play
of cAMP; two Q pockets that form hydrogen bonds and an important role in the regulation of PDE4 activity.14 The
hydrophobic interactions with inhibitors; and a solvated pocket phosphorylation of the UCRs by protein kinase A (PKA) and
(S pocket).12,13 PDE4 inhibitors occupy this active site with a extracellular signal-regulated kinase (ERK) regulates PDE4
variety of interactions, including hydrophobic interactions with dimerization and catalytic activities. UCR1 contains a conserved
the conserved phenylalanine and isoleucine, and hydrogen PKA phosphorylation site in PDE4 subfamilies. The activity of
bonds with the invariant glutamine. The highly conserved PDE4 is regulated by long-term transcriptional regulation or
sequences and structural homology of the catalytic domains of short-term posttranslational modifications. In long-term regu-
B https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

3. PDE4 AS A THERAPEUTIC TARGET FOR


TREATMENT OF HUMAN DISEASES
Members of four subfamilies, PDE4A, PDE4B, PDE4C, and
PDE4D are expressed in various tissues and cellular compart-
ments and involved in many physiological processes, including
airway immunity, asthma, signaling of CNS, cardiovascular
function, inflammatory processes, cell adhesion, and metabolic
processes.7,10,17−19 As the key enzyme hydrolyzing cAMP in
immune cells, PDE4 is one of the main targets of anti-
inflammatory drugs.6 PDE4 is involved in the regulation of the
activation and recruitment of inflammatory cells and the release
of excessive or unregulated cytokines in many allergic and
autoimmune diseases. For example, lipopolysaccharide (LPS)
selectively induces PDE4B2 mRNA expression in human
circulating monocytes.20 PDE4 inhibitors inhibit LPS-mediated
secretion of tumor necrosis factor α (TNF-α) in peripheral
monocytes,21 indicating their important roles in anti-inflamma-
Figure 3. Active site of PDE4D2. Dotted lines represent hydrogen tion. In addition, PDE4B and PDE4D, but not PDE4A, play
bonds. Three inhibitors are shown: compound 1 (green), compound important regulatory roles in neutrophil function, such as
14 (yellow), and compound 15 (salmon).
inhibiting the release of a variety of proinflammatory
mediators22 and delaying neutrophil apoptosis.23 In addition,
PDE4 inhibitors caused airway smooth muscle relaxation,
lation, an increase of cAMP by PDE inhibition conjugates with suggesting their potential values for the treatment of airway
diseases.
activation of adenylyl cyclase (AC) by hormone stimulation. At present, four PDE4 inhibitors have been approved for the
Studies have shown that this regulation is highly specific and is a treatment of human diseases (Figure 4 and Table 1): roflumilast
result of increased gene expression. In short-term regulation, (1) for COPD,24 crisaborole (2) for atopic dermatitis,25
increase of cAMP levels activates PKA, which in turn apremilast (3) for psoriatic arthritis,26 and ibudilast (4) for
phosphorylates specific serine residues in UCR1 of the long the rare child disease Krabbe by USA and for bronchial asthma
forms of PDE4. As a result, the activity of PDE4 increases by Japanese authority.27 In addition, the dual selective PDE3/4
rapidly, and the hydrolysis of cAMP causes the cAMP level to inhibitor RPL554 (5) is in phase II clinical trials for the
treatment of COPD, and the PDE4 inhibitor BPN14770 (6) is
return to equilibrium. In addition, PDE4 activity is regulated by in phase II clinical trials for the treatment of Alzheimer’s disease
several proteins, including arrestin, Src family tyrosine protein and fragile X syndrome (Table 1).
kinases, A-kinase anchoring proteins (AKAPs), and receptor for In recent years, studies on the medical applications of PDE4
activated C kinase 1 (RACK1).14−16 inhibitors have shifted to their uses in treatment of cognitive

Figure 4. Chemical structures of PDE4 inhibitors in clinic trials or in market.

C https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Table 1. PDE4 Inhibitors in Clinic Trials or in Market


ClinicalTrials.gov
drug, owner condition or disease status identifiersa
Roflumilast (1, Daxas, Daliresp), Severe chronic obstructive pulmonary disease Approved by EU, June 2010, and USA, March 2011
AstraZeneca Asthma Phase I December 2019 to March 2021 NCT04108377
Asthma Phase II, August 2018 to April 2020 NCT03532490
Bronchiectasis Phase II, July 2019 to December 2020 NCT03988816
Lymphoma, B-cell Phase I, May 2018 to December 2020 NCT03458546
Bronchiectasis Early Phase I October 1, 2019 to November 2020 NCT04090294
Crisaborole (2), Pfizer Atopic dermatitis ointment Approved by USA Dec 14, 2016
Morphea Phase II, September 2018 to December 2019 NCT03351114
Seborrheic dermatitis Phase IV September 1, 2018 to September 2020 NCT03567980
Atopic dermatitis eczema Phase IV September 2019 to July 2020 NCT04023084
Apremilast (3, CC-10004, Otezla), Celgene Psoriatic arthritis Approved by USA, 2014
Acne conglobata Phase II October 9, 2019 to September 2020 NCT04161456
Dermatomyositis, adult type Phase II June 12, 2018 to April 2020 NCT03529955
Lichen planus of vulva female genital disease Phase II August 27, 2019 to December 1, 2020 NCT03656666
Behcet syndrome Phase III, December 2014 to March 2021 NCT02307513
Nummular and dermatitis eczema Phase II, July 2017 to March 2020 NCT03160248
Psoriasis, psoriatic nail and vulgaris Phase IV January 2017 to December 2019 NCT03022617
Alcohol use disorder Phase II, November 2017 to December 2019 NCT03175549
Recurrent aphthous stomatitis Phase I, October 2018 to December 2019 NCT03690544
Dermatomyositis, adult type Phase II, June 2018 to April 2020 NCT03529955
Lichen planus of vulva female genital disease Phase II, January 2019 to December 2020 NCT03656666
Ibudilast (4, MN-166), MediciNova Krabbe disease Approved by USA, June 2016
poststroke complications and bronchial Approved by Japan and Korea, 1989
asthma
Methamphetamine dependence Phase II, May 2019 to January 2022 NCT03341078
Alcohol use disorder Phase II, July 2018 to June 2020 NCT03489850
Alcohol use disorder Phase II, October 2018 to September 2021 NCT03594435
Glioblastoma Phase I/II, December 2018 to December 2020 NCT03782415
RPL554 (5), Verona Pharma COPD Phase II, May 2019 to October 2019 NCT03937479
BPN14770 (6), Tetra Discovery Partners Alzheimer’s disease Phase II, April 2019 to June 2020 NCT03817684
Fragile X syndrome Phase II, June 2018 to December 2019 NCT03569631
a
Data collected from https://clinicaltrials.gov [Last accessed December 1, 2019].

Figure 5. PDE4 inhibitors in the nervous system.

disorders. PDE4A, -4B, and -4D are expressed in the central relieve cognitive impairment.29 PDE4D is an important
nervous system, while PDE4C is mainly expressed in the regulatory factor in depression, learning, and memory
peripheral nervous system. PDE4A plays an important role in dysfunctions.30,31 The PDE4D gene is highly expressed in the
anxiety and emotional memory.28 PDE4B is involved in hippocampus, and PDE4D gene-deficient mice exhibit long-
dopamine and stress-related signaling and may thus potentially term potentiation that significantly increases learning ability,
D https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 6. Active site of PDE4. Dotted lines represent hydrogen bonds. Residues are numbered using the PDE4B2B sequence. (A) Surface presentation
on binding of compound 1 (yellow sticks) to PDE4B (green sticks) and PDE4D (white sticks). P1−P3 and CORE denote four potential
pharmacophores of PDE4 inhibitors. (B) Coverage of the PDE4 active site by a UCR2 helix (yellow ribbons). (C) Interaction of compound 1 with the
UCR helix.

memory, and nerve regeneration.31,32 The potential applicability nervous system may result in new platforms for the treatment of
of PDE4 inhibitors in improving learning and memory is well neurological diseases.
supported experimentally. For example, compound 14 improves In addition, regulation of the cellular cAMP level via PDE4
learning and memory by inhibiting PDE4 activity and inhibition is directly related to pathological functions of tumors,
influencing cAMP-mediated signaling pathways such as the such as tumor cell migration and proliferation, as well as tumor
cAMP/PKA/CREB and EPAC/ERK signaling pathways vascularization and development.8,49 Genetic studies showed
(EPAC, exchange protein directly activated by cAMP).33 The that PDE4D is related to the expression of the thiol purine gene
intervention and regulation of PDE4 provide new therapeutic set, leading to an investigation of PDE4-based purine drugs for
strategies for the treatment of neurological diseases such as the treatment of various tumors. Moreover, PDE4B was
Parkinson’s disease (PD), Alzheimer’s disease (AD), and reported to regulate glucocorticoid reactivity of B-cell
cerebral ischemia. PDE4 inhibitors relieve AD through lymphoma.50 PDE4 has been shown to have the highest
reduction of neuroinflammation and benefit memory consol- cAMP hydrolysis activity in 41 out of 60 tumor cell lines,51
highlighting its potential for the treatment of cancers.
idation.34 Although the etiology of PD has not been completely
revealed, involvement of PDE4 in regulation of oxidative stress
in neurodegeneration of PD is obvious.35,36 4. SUBFAMILY SELECTIVE PDE4 INHIBITORS AND
Many PDE4 inhibitors have been developed for the treatment EMESIS
of neurological diseases; for example, HT0712 (7, Figure 5), Although PDE4 has been identified as a therapeutic target, the
etazolate (8, Figure 5), compound 6 (Figure 4), and MK-0952 clinical development of inhibitors has been hampered by dose-
(9, Figure 5) were tested in clinical trials. Compound 7, also limiting side effects, most notably, nausea and vomiting, which
known as IPL-455903, enhanced memory and promoted motor were observed with the first-generation PDE4 inhibitors such as
function recovery after cerebral ischemia by stimulating the compound 14 and the second-generation inhibitors such as
generation of synapses in the cortex37 and showed the positive compound 15. The early studies suggested that the emesis
results in the phase II clinical trial of age-associated memory results from the inhibition of PDE4D. Knocking out PDE4B, but
impairment (AAMI).38 Etazolate (9, EHT-0202), as a PDE4 not PDE4A/PDE4D, significantly inhibited the synthesis of
inhibitor and a GABA-A receptor modulator, increased TNF-α in LPS-mediated monocyte and peritoneal macro-
expression of the α-secretase enzymes and APP alpha phages. In mouse allergic asthma models, Th2 cell function was
secretion.39 Compound 9 was generally tolerated in clinical regulated by PDE4B rather than PDE4A/PDE4D.52 In addition,
trials for the treatment of the early Alzheimer’s disease but a study on lighting reflection of rodents showed that the
showed dose-dependent and central nervous system-related inhibition of PDE4D but not PDE4B may be the cause of the
emesis observed in the study of nonsubfamily selective PDE4
adverse events.40 Compound 6, a PDE4D allosteric modulator,
inhibitors.53 This is shown by several PDE4B selective inhibitors
enhanced the long-term potential in hippocampal slices and is
such as compound 38 that has potent anti-inflammatory effects
currently in phase II clinic trials for treatment of the early and reduced emetic effects.54 However, recent studies implied
Alzheimer’s disease and fragile X syndrome (Table 1).41 MK- that inhibition of PDE4D may not be a key factor to cause
0952 was tested in a phase II clinic trial for its effect on vomiting.55
improving cognitive impairment in patients with mild-to- Structural superposition between the catalytic domains of
moderate AD.42 Compound (S)-Zl-n-91 (10, Figure 5) has PDE4B and PDE4D showed that the active site residues for the
the IC50 values of 12 nM for PDE4D and 20 nM for PDE4B and binding of PDE4 inhibitors are identical and have the same
showed potent neuroprotection activities in ICR mice.43,44 In conformation, including the invariant glutamine and phenyl-
addition, GEBR PDE4 inhibitors such as GEBR-20b (11, Figure alanine (Q443 and F446 of PDE4B2B, Figure 6A) that are two
5) facilitated long-term potentiation and improved memory basic residues for binding of substrates and inhibitors of PDEs.
without emetic-like behavior in rodent tests.45−48 Thus, in- The only minor difference between the active sites of PDE4B
depth studies on the specific functions and regulatory and PDE4D is the conformation of Met431 of PDE4B (Figure
mechanisms of PDE4s and their inhibitors in the central 6A). Thus, it will be difficult to design PDE4 subfamily selective
E https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

inhibitors against only the catalytic domain of PDE4. The crystal BPN14470 that is currently in phase II clinic trials for treatment
structures of PDE4 plus UCR2 domain55,56 showed that an of Alzheimer’s disease and fragile X syndrome (Table 1).52
UCR2 helix completely covered the active site of PDE4 (Figure
6). The sequence variation in UCR2 among the PDE4 5. SURVEY OF PDE4 INHIBITORS
subfamilies may allow the design of PDE4 subfamily selective 5.a. Catechol Ether-Based PDE4 Inhibitors. Catechol
inhibitors. ether simulates the hydrophobic properties of the adenine
On basis of the fact that a tyrosine in the UCR2 region of portion of cAMP and, thus, has been used as a scaffold for many
PDE4A/B/C (Tyr274 of PDE4B3) corresponds to a phenyl- PDE4 inhibitors,62,63 such as the first-generation drug rolipram
alanine in PDE4D, a PDE4D-selective allosteric modulator was (14, Figure 8) and more advanced roflumilast (1) analogs. The
designed and showed good therapeutic efficacy and reduced crystal structures (PDB code 1XMY, Figure 9) showed that the
emesis.55 This PDE4 allosteric modulator interacts with both catechol ether-containing compounds form a hydrogen bond
the active site and a helix of UCR2, implying a possibility of with the invariant glutamine across the PDE families and also
development of a PDE4 subfamily selective inhibitor. The π−π stacking against a conserved phenylalanine (Figure 3).
crystal structure of PDE4D in complex with inhibitor PMNPQ Compound 14 is sandwiched by the hydrophobic clamp made
(12, Figure 7) showed that Phe196 of PDE4D3 from the UCR2 up of conserved phenylalanine and isoleucine on the two sides
(Figures 3 and 9). These interactions, hydrogen bonding with
the invariant glutamine and stacking against the phenylalanine,
occurred in almost all the inhibitors-PDEs crystal structures and
thus form the basic affinities of the PDE inhibitors.62
The dialkoxyphenyl attachments of the compounds in this
category are often the methoxy and cyclopentyloxy groups, such
as in compound 14 and cilomilast (15, Figure 8). The methoxy
group oriented into the small Q1 pocket that is formed by
Tyr233, Asn395, Thr407, and Tyr403 of PDE4B. The larger
hydrophobic cyclopentyl moiety occupied the Q2 pocket formed
by Phe414, Met411, Met431, and Phe446 of PDE4B.12
Compound 14 has been extensively studied for the treatment
of various diseases, including asthma, but shows severe side
effects including dizziness, headache, nausea, and vomiting.6 It is
often used as a positive control for studies of PDE4 inhibitors.
Figure 7. PDE4B/4D subfamily selective inhibitors 12 and 13 and Compound 15 (Figure 8), developed by GlaxoSmithKline, is a
binding of 12 to PDE4B (cyan sticks and ribbons, PDB code 3G45) and selective PDE4 inhibitor with an IC50 of 95 nM. Compound 15
PDE4D (green sticks, PDB code 3G58). Tyr274 of PDE4B (a was tested for the treatment of COPD but caused severe adverse
phenylalanine in PDE4D) from UCR helix determines the subfamily reactions, including nausea, vomiting, headache, abdominal
selectivity, while the residues in the active sites show great conservation pain, diarrhea, and indigestion.64 In the phase III clinical trial on
in both sequence and conformation. asthma, the FEV1 values of the patients treated with compound
15 were <0.03 L, indicating lack of clinical efficacy on the
improvement of lung function.65
helix, which corresponds to Tyr274 of PDE4B3, plays a key role Tetomilast (16, Figure 8), a PDE4 inhibitor with an IC50 value
in ligand binding.57 By utilization of this difference, several of 74 nM, suppresses the expression of TNFα and IL-12 in LPS-
PDE4D selective inhibitors have been developed, such as stimulated human monocytes and the production of interferon γ
D159687 (13, Figure 7) and compound 6 (Figure 5), which and IL-10 in CD4 lymphocytes at the transcriptional level.66 In a
have a 100-fold selectivity between PDE4D and PDE4B.55,58−60 phase II clinical trial of 246 patients with COPD, the 12 week
In particular, compound 6, the first PDE4D selective inhibitor, treatment with compound 16 (50 mg) improved peak FEV1 to
entered phase II clinical trials for the treatment of Alzheimer’s 50 mL and forced vital capacity (FVC) to 130 mL. However, this
disease and fragile X syndrome.59 In addition, both PDE4B and clinical trial was terminated in 2017.67
PDE4D negative allosteric modulators are shown to play Compound 3 (Figure 4) is an oral and selective PDE4
important roles in neurological diseases such as AD.58,60,61 On inhibitor with IC50 of 74 nM and was studied for the treatment of
the basis of the difference of tyrosine/phenylalanine, the PDE4B autoimmune diseases. Compound 3 acts on many types of cells
allosteric modulators can be designed and showed the potential and shows a wide range of anti-inflammatory activities such as
to reduce neuroinflammation via inhibition of the production of inhibition on production of TNF-α, IL-12, and IL-23 and
microglia from cerebral amyloid, while the PDE4D allosteric responses of natural killer cells and keratinocytes. Clinical data
modulator enhances memory consolidation.58−61 show that compound 3 significantly improved psoriasis of nails,
The UCR2-catalytic domain interactions block substrate scalp, and palmar (hands and feet). Compound 3 was approved
entry into the active site or product release, and therefore the by the U.S. FDA in 2014 for the treatment of active psoriatic
activity of different splice variants of PDE4 can be modulated arthritis and moderate to severe plaque psoriasis by oral
differently. This study opens a new avenue for the design of small administration. Additional clinical trials on the treatment of
molecule inhibitors specifically targeting PDE4 specific splice other diseases are underway (Table 1), including cardiovascular
variants. Since the PDE4 allosteric modulators interact with diseases, parapsoriasis, palmoplantar psoriasis, psoriasis of the
both UCR2 and the catalytic domain, they may preferentially scalp, and psoriasis vulgaris.68
inhibit a certain form of PDE4 subfamilies and also have less Replacement of the phenyl ring of compound 3 with a
chance to interact with nontargeted proteins. As a result, they thiophene yields compound 17 (Figure 8) that has an IC50 value
may be less toxic than other PDE4 inhibitors, as shown by of 26 nM.69 Expansion of the acetyamide group of compound 3
F https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 8. Chemical structures of catechol ether-based PDE4 inhibitors.

increased mental illness (such as insomnia, anxiety, and


depression). In addition, compound 1 was studied for the
treatment of asthma, noncystic fibrosis bronchiectasis, obesity,
and lymphoma B-cells (Table 1).
Compound 1 has a remarkable inhibitory affinity toward
PDE4B and PDE4D, with IC50 values of 0.84 nM and 0.68 nM,
respectively.12 The X-ray crystal structures (PDB code 1XMU,
Figure 11) showed that the difluoromethoxy group of
compound 1 interacts with the Q1 pocket and the cyclo-
propylmethoxy group occupies the Q2 pocket. The 3,5-
dichloropyridyl group extends into the metal binding pocket
Figure 9. X-ray crystal structure of PDE4B-compound 14 (PDB code to form a hydrogen bond with the water molecule.12
1XMY). Replacement of the methyl cyclopropane of compound 1 with
longer-chain aminofuran leads to compound 20 (Figure 10).
led to a series of highly selective PDE4 inhibitors (18, n = 3−9, Compound 20 showed IC50 of 5.4 and 18 nM for PDE4B1 and
Figure 8) with IC50 values of 3−8 nM.70 Compound 19 (IC50 = PDE4D2, respectively.73 Compounds 21, 22, and 23 (Figure
40 nM, Figure 8) and its analogs are benzodiazepinone-derived 10) were designed by modification of the amide linker between
catechol ether with amine-containing chains.71,72 the dichloropyridine and catechol ether of roflumilst and
In short, it can be summarized as follows: (1) The two showed subnanomolar inhibitory activities against PDE4.74
substituents on the catechol ether occupy the hydrophobic Q1 CHF6001 (23, IC50 = 26 pM, Figure 10) explored the chemical
and Q2 pockets. (2) The hydrophilic group orients to the space of the solvent-exposed region of the enzyme and inhibited
solvation region and hence effectively enhances the affinity of the release of the proinflammatory cytokine TNF-α from LPS-
the compounds. (3) The interactions with water molecules in stimulated PBMCs.75 The phase II clinical trial of compound 23
metal ion pocket significantly increase the inhibitory capacity of on 1130 COPD patients was completed in January 2018 and
the compounds. showed no significant emesis at tested doses.76 However, the
5.b. Compound 1 Analogs. Compound 1 (Figure 4), the clinical results of the phase III trial have not been revealed yet.
first marketed PDE4 inhibitor, was approved for the treatment of Piclamilast (24, Figure 10) combines the structural features of
severe COPD-associated chronic bronchitis in Europe in 2010 compound 14 and compound 1 and showed IC50 values of 41
and in the USA in 2011. Clinical studies showed that compound and 21 pM for PDE4B and PDE4D, respectively. The
1 reduced inflammation and fibrosis in lung, enhanced mucosal cyclopentyl ether binds to the Q2 pocket, while the methoxy
clearance, and remodeled the airways. Adverse reactions were group occupies the Q1 pocket (PDB code 1XM4, Figure 11b).
mainly diarrhea, weight loss, nausea, atrial fibrillation, and The 3,5-dichloropyridyl group forms hydrophobic interactions
G https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 10. Chemical structures of compound 1 analogs.

Figure 11. X-ray crystal structure of PDE4. (A) Compound 1 bound to PDE4B (PDB code 1XMU). (B) Compound 24 bound to PDE4B (PDB code
1XM4).

with Met347, Leu393, and Phe414 and extends into the metal pockets are filled respectively with the small and large
pocket to form a hydrogen bond with the water molecule attachments of the catechol ether; and (3) inappropriate
(Figure 11).12 The replacement of the central phenyl group of modification of the amide linker reduced the affinity of
compound 24 with chromenone yields compound 25 (Figure inhibitors, such as in the cases of compounds 25, 26, and 27
10) that has an IC50 of 5 nM against PDE4B.77 (Figure 12).
Compound 26 (LEO-29102, Figure 10) was designed by 5.c. Quinoline-Based PDE4 Inhibitors. GSK256066 (28,
modification of dimethyl catechol ether and dichloropyridine of Figure 13) has an IC50 value of 3.2 pM for PDE4 and high
compound 1.78 By introduction of a soft spot in the 2′-position, selectivity over other PDEs and is an inhaled PDE4 inhibitor for
no cases of emesis were observed in the clinical study and may
the treatment of asthma.80,81 A large number of studies showed
thus work as a soft drug for the treatment of AD. Compound 26
that compound 28, delivered orally or by inhalation, reduced
was tested in a phase II clinical trial for the treatment of eczema,
FVC and inhibited allergen-mediated eosinophilia, airway
atopic dermatitis and psoriasis, but no recent progress has been
reported. The introduction of catechol pyrimidine core, which is hyperresponsiveness and remodeling, mucin production, and
a potential bioisostere of adenosine and also mimics the goblet cell hyperplasia in CD rats or other rodent models.80
nucleotide part of the cAMP, yielded a series of novel Compound 28 significantly inhibited early and late allergic
compounds, such as compound 27 (Figure 10) that shows an responses by 26.2−34.3% but limitedly decreased the FEV1 and
IC50 of 15 nM against PDE4B.79 weighted FEV1 values by 40.9% and 57.2%, respectively. This
In short, we can conclude that (1) the 3,5-dichloropyridine compound underwent phase II clinical trials for asthma, COPD,
moiety interacts with Mg2+-bound water; (2) the Q1 and Q2 and allergic rhinitis. Clinical trials were completed, but further
H https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 14. X-ray crystal structure of compound 28 bound to PDE4B


(PDB code 3GWT).

was highly efficacious with ED50 = 0.1 mg/kg following oral


Figure 12. Schematic presentation of compound 1 interaction with administration.85
PDE4. The binding mode of quinoline inhibitors to PDE4 is
summarized as follows (Figure 15): (1) the methoxy and
development of compound 28 is not evident since the
compound no longer appears in the GSK product pipeline.
Compound 29 (Figure 13) has a 2-trifluoromethyl-8-
methoxyquinoline core and exhibited high PDE4 inhibitory
potency (IC50 = 0.01 nM).82 Derivatizing the amide group
afforded compound 30 (Figure 13), whose xinafoate salt
inhibited inflammatory responses by 69% at 0.02 mg kg−1
day−1 in rat model, and monkey PK studies of 30 showed no
emesis (AUC = 31 ng·h/mL at 10 mg/kg po).83,84 Optimization
of both the 4-benzylcarboxamide and 5-α-aminoethyl groups
leads to an orally active, highly selective compound with
picomolar potency against PDE4. For example, compound 31
(IC50 = 60 pM, Figure 13) is a very potent PDE4 inhibitor with
high selectivity over PDE10 (15 800-fold) and has an IC50 of 0.6
nM for the inhibition of TNF-α release in a cell-based assay.85 At
an oral dose of 10 mg/kg, compound 31 was found to have Cmax
= 960 nM and 24% bioavailability in a rat PK study. In an in vivo
rat LPS-induced pulmonary inflammation model, compound 31 Figure 15. Interactions of compounds 29−31 with PDE4.

Figure 13. Chemical structures of quinolone analogs.

I https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 16. Chemical structures of PDE4 inhibitors with a fused bicyclic ring.

difluoromethyl groups form hydrophobic interactions with the With the combination of dimethoxybenzene, indole, and
Q1 and Q2 pockets of PDE4, respectively; (2) the quinoline ring dihydropyridine, compound 37 (Figure 16) was found to be a
stacks against the phenylalanine via π−π interactions; (3) R2 potent PDE4 inhibitor with IC50 values of 0.54 and 0.65 nM for
interacts with the S pocket, and thus its size influences the PDE4B and PDE4D, respectively.91 The docking of 37 to
activity. PDE4B/PDE4D showed that its indole moiety has two possible
NVP-ABE-171 (32, Figure 13) has the IC50 values of 602, 34, orientations in PDE4D and forms hydrogen bonds with Glu396
1230, and 1.5 nM for four subtypes of PDE4 (A, B, C, D) and or Glu505, whereas 37 exhibits a completely different
was tested for treatment of asthma and chronic obstructive orientation in PDE4B and forms a hydrogen bond with
pneumonia.86,87 Compound 32 inhibited the activation of Ser442. The dimethoxy groups of 37 interact with the metal
airway neutrophils with an ED50 value of 3 mg/kg in BALB/c ions in PDE4B, and the indole moiety forms hydrogen bonds to
mice and of 0.2 mg/kg in the Norway rat model. It also inhibited Gln443.
ovalbumin-induced airway eosinophil activation with an ED50 of GSK356278 (38, Figure 16), a selective and brain-penetrable
0.1 mg·kg−1. However, the relatively poor pharmacokinetic PDE4B inhibitor with the pIC50 value 8.8, showed a therapeutic
parameters (especially solubility) of compound 32 hinder its index superior to both compound 14 and compound 1 and had
further development. Replacement of the benzoxadiazole of 32 beneficial effects on anxiety and cognition in primates without
with fluorobenzene leads to compound 33 (Figure 13), which emesis or other dose-limiting side effects.92 However,
compound 38 was apparently discontinued after phase I safety
exhibited 400-fold improvement of water solubility, good
and tolerability studies in 2012.
pharmacokinetics, and no significant nausea and emesis.
High throughput screening led to the discovery of
YM976 (34,Figure 13) is a pyrimidine derivative and
pyrazolopyridine-containing compound 39 (Figure 16), a
structurally different from the other PDE4 inhibitors such as
potent and selective PDE4 inhibitor. Compound 39 inhibits
compound 14.87 YM976 inhibited PDE4 with an IC50 value of LPS-induced TNF-α production and shows an encouraging PK
2.2 nM, inhibited cell infiltration into the pleural cavity with an profile in rat, indicating its suitability for oral administration.93
oral ED30 of 9.1 mg/kg in a rat carrageenan-induced pleurisy The structure of PDE4B-39 showed that the pyrazolo[3,4-
model, and produced no emesis at doses of up to 10 mg/kg.88 b]pyridine moiety forms a hydrogen bond with Gln443, and the
5.d. PDE4 Inhibitors with a Scaffold of Fused Bicyclic N-ethyl substituent occupies the lipophilic pocket, similar to the
Rings. Several compounds with a scaffold of the fused bicyclic pattern of compound 14 binding (PDB code 3D3P, Figure 17).
ring show excellent PDE4 inhibitory activities. Ronomilast Exploration of the SAR on the amide fragment of 39 led to the
(ELB353) (35, Figure 16) is a potent oral PDE4 inhibitor with discovery of compound 40 (Figure 16), which has a few tens of
an IC50 of 3 nM and good pharmacokinetic properties in Lewis picomolar affinity with PDE4 and subnanomolar inhibition on
rat or ferrets.89 In the preclinical and phase I trials, compound 35 LPS-induced TNFα production in human peripheral blood
demonstrated efficacy for the treatment of inflammation-related mononuclear cells.94
diseases and no severe adverse events in humans. AWD12-281 Modification of the pyrazolopyridine scaffold led to
(36, GSK842470, Figure 16) is an inhaled PDE4 inhibitor with preparation of compound 41 (IC50 = 26 nM, Figure 16).95
an IC50 of 9.7 nM and an EC50 of 110 nM for TNF-α release. It Molecular docking of compound 41 to the crystal structure of
was tested in phase II clinical trials for the treatment of COPD, PDE4B showed that interactions of the amide, 3-methoxyaryl,
asthma, and allergic rhinitis.90 and sulfone groups with PDE4B completely coincide with those
J https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

0.21 nM) and LPS-induced pulmonary neutrophilia in vivo in


mice by 41% at a dose of 1.0 mg/kg (i.t.).98 The crystal structure
of PDE4B-45 showed that its pyrimidine ring forms π−π
stacking against Phe446 of PDE4B, while the neopentyl amide
moiety occupies a lipophilic Q1 pocket.
Compound 46 (Figure 19), with a 2-arylpyrimidine core,
showed anti-inflammatory therapeutic potential and a wider
safety margin than compound 15.99 The central pyrimidine ring
of 46 is sandwiched by Phe618 and Ile582 of PDE4B and forms a
hydrogen bond with Gln615. The aryl group extends into the
metal pocket and forms a hydrogen bond with the water
Figure 17. X-ray crystal structure of 39 bound to PDE4B (PDB code molecule bound to Mg2+, while the ethyl and methyl
3D3P). substituents fit the Q1 pocket. In the mouse model, 46 showed
a strong antidepressant effect in mouse models of forced swim
of compound 28 (PDB code 3GWT, Figure 14). A nitrogen in and tail suspension.99
the heterocyclic ring and the amide form hydrogen bonds with Compound 47 (Figure 19) had an IC50 of 7 nM for PDE4 and
Gln443 through a water molecule, in addition to a hydrogen an EC50 of 0.01 mg/kg for TNF-α inhibition in a mouse sepsis
bond between the amide of 39 and Asn395.95 Cyclization of the model and reduced ear thickness by 40% in a psoriasis model.100
primary amide at the 5-position of the pyrazolo[1,5-a]- Introduction of a pyrido[3,2-d]pyrimidine substituent resulted
pyrimidine yielded compound 42 (Figure 16) and improved in compounds such as 48 (Figure 19) with IC50 = 0.4 nM against
the affinity by 200-fold (IC50 = 0.03 nM). The amide of 42 PDE4.101
contacted Asn395 and displaced a water molecule.95 5.f. Pyridazinone-Based PDE4 Inhibitors. Zardaverine
We can summarize the above discussion as follows (Figure (49 Figure 20) interacts with the catalytic site via hydrogen
18). (1) The pyridine core inserts into the hydrophobic clamp bonds between (1) the catechol oxygen and the side chain of
invariant Gln369 of PDE4D2 and (2) the pyridazinone oxygen
and the nitrogen atom of His160. The pyridazinone moiety of
compound 49 is surrounded by residues Leu319, Ser320, and
Asn321 (PDB code 1XOR, Figure 21).12
Tetrahydrophthalazinone derivatives such as compound 50
(IC50 = 4 nM, Figure 20) can be considered as an analog of
compound 49, but molecular modeling revealed that the cis-
fused cyclohexane ring occupies a different region from that of
compound 49. Therefore, it was assumed that the steric
interactions of these rings with the residues in the binding site of
PDE4 play an important role in enzymatic inhibition. On the
other hand, substitution of the pyridazinones with pyrazoles/
1,2-diazepin-7-ones or N-substitution on compound 49 led to a
series of new derivatives such as 51 (Figure 20) that has pIC50 of
Figure 18. Interactions of compounds 38−42. 9.45.102
Optimization of the pyridazinone branch resulted in
and forms a hydrogen bond with Gln443 of PDE4B to increase compound 52 (Figure 20).103 The X-ray crystal structure of
the binding strength. (2) The amide branch forms a hydrogen human PDE4D in complex with 52 demonstrated that the
bond with Asn321 of PDE4D2 in the active site while interacting pyridazinone core sits in the hydrophobic clamp and forms a
with the small hydrophobic pocket (Q1). The inhibitory activity hydrogen bond with Glu369. The ethyl branch at the 6-position
could be significantly increased if the amide branch is a lactam. of the benzene ring of 52 fits in the lipophilic pockets, while the
(3) The substituent at the 2- or 3-position of pyridine fills the benzoate extends into the outer solvated space. Replacement of
large hydrophobic pocket (Q2). (4) The aryl fragment at the 4- the acetyl carbonyl group with a urea link between 3,5-
position of pyridine interacts with ion pocket to increase its dichloropyridine and pyridazinone led to compounds 53 and 54
inhibitory activity. (Figure 20), which were shown to be potent, long-acting, and
5.e. Pyrimidine-Based PDE4 Inhibitors. Compound 43 potentially safe inhaled PDE4 inhibitors.103,104 The combina-
(Figure 19) was identified as an orally active PDE4B-selective torial assembly of functional linkers led to compounds such as
inhibitor and had IC50 values of 5.5 nM and 440 nM for PDE4B 55, in which a phenyl ring is connected to the C4 pyridazine
and PDE4D, respectively.96 Its therapeutic index, which was atom.105
calculated based on its inhibitory effect on LPS inhalation- 5.g. Methoxy Heteroaromatic-Based PDE4 Inhibitors.
induced neutrophilia and gastric emptying, was similar to that of Compound 4 (AV-411 or MN-166, 4, Figure 4), an anti-
compound 1 in mice. Replacements of the sulfide with a sulfone inflammatory drug approved in Japan and Korea, inhibited
and of the fluorine with chlorine of 43 afforded highly selective human PDE4A-D with IC50 values of 54, 65, 239, and 166 nM,
PDE4B inhibitor 44 (Figure 19), which had an IC50 of 3 nM for respectively, as well as weakly inhibiting other PDE families.106
PDE4B and 433-fold selectivity over PDE4D.97 Changing the Compound 4 was shown to be a bronchodilator and vasodilator,
sulfonyl moiety to a lactam and the carboxylic acid to an N- had neuroprotective effects, and was used for oral treatment of
neopentylacetamide afforded compound 45 (Figure 19), which asthma, stroke, and topical treatment of allergic conjunctivi-
has an IC50 of 7.3 nM against PDE4B and potently inhibited tis.107 In addition, phase II clinical trials are underway for
LPS-induced TNF-α production in mouse splenocytes (IC50 = treatments of progressive multiple sclerosis, neuroinflammation,
K https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 19. Chemical structures of pyrimidine-based PDE4 inhibitors.

Figure 20. Chemical structures of pyridazinone-based PDE4 inhibitors.

amyotrophic lateral sclerosis as an adjunct to riluzole, chronic the PDE4 catalytic domain suggested that the amide linker is
migraine, and alcohol use disorder (Table 1). Compound 4 was largely responsible for the improved activity.108,109 The nitrogen
approved as an orphan drug in the U.S. for the treatment of at position 1 of the pyrazolopyridine interacted with the
progressive Krabbe disease in 2015 and for the treatment of invariant glutamine, and the pyridine ring extended into the
poststroke complications and bronchial asthma in Japan and metal binding pocket to form hydrogen bonds with water.
Korea in 1989. Lotamilast (E6005, RVT-501, 57, Figure 22) is a selective
Installation of a methoxyl substituent and 3,5-dichloropyr- PDE4 inhibitor with an IC50 of 2.8 nM and suppresses the
idine onto the heterocycle of compound 4 leads to compound production of various cytokines in human lymphocytes and
56 (Figure 22) that showed dramatic improvement in PDE4 monocytes.110 Topical application of 0.03% compound 57 to
inhibition (IC50 of 0.27 nM). The docking of compound 56 to the rostral back significantly inhibited the activity of the
L https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 21. X-ray crystal structure of PDE4D2 (PDB code 1XOR) in complex with compound 49.

Figure 22. Chemical structures of methoxy heteroaromatics PDE4 inhibitors.

cutaneous nerves and increased the cAMP concentration in


mice with chronic dermatitis.111 Lotamilast completed phase II
clinical trials sponsored by Dermavant Sciences Inc. for the
treatment of atopic dermatitis in July, 2017.
Oglemilast (58, Figure 22) is an oral PDE4 inhibitor for
asthma and rheumatoid arthritis and exhibited an IC50 value of
1.4 nM for PDE4 and showed high selectivity over other PDEs.
Compound 58 inhibited LPS-induced TNF-α production (IC50
= 190 nM) and blocked LPS-induced pulmonary neutrophilia in
rats (ID50 = 1.45 mg/kg) without obvious side effects.53
However, the phase IIb clinical data were discouraging and
showed no evidence of clinical effects.112 Revamilast (59, Figure Figure 23. Chemical structures of triazine-based PDE4 inhibitors.
22) was launched by Glenmark as a PDE4 inhibitor after the
failure of compound 58 and showed an IC50 of 2.7 nM for PDE4
and high selectivity over other PDEs. In 2011, Glenmark
globally announced the initiation of a phase IIb trial of
compound 59 on chronic inflammatory disorders such as
asthma and rheumatoid arthritis. However, the phase IIb data
were discouraging and the trial was discontinued.
5.h. Triazine-Based PDE4 Inhibitors. Compound 60, a
1,3,5-substituted triazine derivative (Figure 23), was identified
by screening a compound library and inhibited PDE4A with an
IC50 value of 245 nM. Structure-based design leads to
compound 61 (PDB code 4KP6, Figure 23), a potent and
orally active PDE4 inhibitor with an IC50 value of 1 nM against
PDE4A, -4B, and -4D.113 The crystal structure (Figure 24)
showed that the triazine core was located in the hydrophobic
clamp and that the substituents occupied the Q1 and Q2 pockets.
The triazine ring forms hydrogen bonds directly with Gln443
Figure 24. Diagram of the interactions between PDE4B and 61 (PDB
and indirectly with Asn395, Tyr233, and Asp392 via a water
code 4KP6).
molecule, in addition to π−π stacking interactions with Phe446
and Phe414 of PDE4B.
5.i. Cyclohexanediamide-Based PDE4 Inhibitors. A hexanediamide, such as compound 62 (Figure 25), have been
class of pyridopyrimidinedione derivatives containing cyclo- reported.114,115 Compound 62 inhibited PDE4B2 with a pIC50
M https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 25. Chemical structures of cyclohexanediamide-based PDE4 inhibitors.

of 9.9. UK-500,001 (63, Figure 25), developed by Pfizer for as an important mechanism.117 As the key regulator of the cAMP
inhaled delivery, has IC50 values of 0.38−1.9 nM for PDE4A, -B, signaling pathways in physiological processes, PDE4 probably
and -D and strongly inhibited TNF-α production (IC50 = 0.13 serves as a cellular receptor for many natural products and
nM). However, the phase II clinical data of its use against asthma traditional herbal medicines to exert their beneficial effects on
showed that this compound does not effectively improve human health.117 Nonselective inhibition of PDEs may be a
allergen-induced FEV1 but shows the typical side effects of general pattern for functions of many natural products and
PDE4 inhibitors. Therefore, Pfizer ceased further development traditional herbal medicines. A well-known example of such
of compound 63. AstraZeneca obtained several patents for 2- natural products is theophylline (65, Figure 27), which is a
phenoxypyridin-3-ylamide derivatives, such as compound 64 nonselective inhibitor of PDEs with IC50 values of 50−100 μM
(pIC50 = 11, Figure 25).116 These compounds contain a that is a first generation respiratory drug for the treatment of
cyclohexanediamide motif similar to that of compound 63 and asthma and COPD.118,119
show pIC50 values between 10 and 11. Another example is the superfamily of flavonoids that are
In summary, for the conservation of the binding patterns, abundant and key bioactive components of our daily foods and
PDE4 inhibitors can be divided into four pharmacophores for have been thought to exert their nutritional effects via the
design: P1−P3 and CORE (Figures 6A and 26). The CORE nonselective inhibition of PDEs.120 Many flavonoids with a
chromone scaffold (quercetin (66), genistein, luteolin, daidzein,
hesperidin, prunetin, diosmetin, biochanin A, apigenin, and
myricetin), which are nutrition supplements involved in
regulation of various biological processes, are nonselective
inhibitors of PDEs with affinity in the tens of micromolar
range.121 A recent study showed that the cocktail of antileukemia
drug dasatinib and natural product compound 66 (Figure 27)
prolongs the lifespan of mice by 36%.122 The mechanism for the
cocktail to increase lifespan is not clear, but perhaps the PDE4-
cAMP signaling pathway plays a critical role since compound 66
is a well-known PDE4 inhibitor.121
The catechol ether, a characteristic motif of PDE4 inhibitors,
also appears in some of the natural products. For example,
mesembrenone (67, Figure 27), which is an alkaloid isolated
from the South African plant Sceletium tortuosum and has
antianxiety and antidepressant effects, is an inhibitor of PDE4
with an IC50 value of 0.47 μM but not other PDEs.123
Figure 26. Superposition of PDE4 inhibitors, which are colored with
Compound 67 is also a selective inhibitor of the serotonin
green (compound 1), yellow (GEBR20b, 11), cyan (compound 24),
salmon (coumarin), orange (compound 46), and pink (naph). transporter (SERT) with Ki = 27 nM to inhibit reuptake of
neurotransmitter serotonin.124
While most natural products nonselectively inhibit PDEs or
contributes the basic affinity for all inhibitors and fits to the even simultaneously target other proteins, some of them showed
hydrophobic clamp of the PDE4 active site. Pharmacophores moderate or high selectivity among PDE families. The most
P1−P3 respectively oriented to subpockets of Q1 and Q2 and the potent component 68 (Figure 27) out of 13 compounds that
solvent region. were isolated from Gaultheria yunnanensis has an IC50 of 245 nM
for PDE4D and moderate selectivity over other PDEs.125
6. NATURAL PRODUCTS AS PDE4 INHIBITORS Compounds, which were extracted from the Chinese herb
Natural products have served as original sources of current drugs “Sang-Bai-Pi” (Morus alba L.) and have been used for the
and attracted a great deal of attention for their benefits for treatment of coughing and inflammatory diseases, also show
human health in the past years. While functions of natural relatively good selectivity for PDE4 over other PDEs.126
products may be achieved via interactions with multiple targets, Moracin M (69, Figure 27) has IC50 values of 2.9, 4.5, >40,
regulation on the second messenger cAMP has been recognized and >100 μM for PDE4D2, PDE4B2, PDE5A1, and PDE9A2,
N https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 27. Chemical structures of natural products that inhibit PDE4.

respectively. Muracin A (70, Figure 27), a prenylated flavonoid, In short, although natural products may target multiple
shows the highest affinity for PDE4 (IC50 of 5.4 nM for proteins, PDE4 must be an important receptor for their
PDE4D2) and outstanding selectivity over other PDEs.127 Thus, functions. For their different skeletons and potentially different
PDE4 may be the receptor mediating the functions of the binding patterns, natural products may be an abundant resource
moracin compounds. for the development of PDE4 selective inhibitors.
The natural product selaginpulvilin K (71) is a highly selective
inhibitor of PDE4D with an IC50 of 11 nM (Figure 27).128 The 7. DUAL PDE4 INHIBITORS
cocrystal structure of PDE4D-71 (PDB code 5WQA) revealed
that 71 shallowly binds to the active site pocket and forms a For the important roles of the cAMP and cGMP signaling in
hydrogen bond with the side chain of Gln369, while a benzene various physiological processes1−4 and the existence of multiple
ring of 71 contacts Phe372 with π−π stacking (Figure 28).128 PDEs in certain tissues such as seven PDE families in the
heart,129 simultaneous inhibition of multiple PDE families may
produce synergistic effects of physiological processes. In
addition, simultaneous regulation on the cAMP/cGMP signal-
ing pathways and the key proteins in the physiological processes
such as cancer development might produce additive or even
synergistic effect. On the basis of these considerations, dual
selective PDE4 inhibitors that target PDE4 and other PDEs or
that conjugate with other functional proteins have been
developed in recent decades.
7.a. Dual Inhibitors of PDE4 and Other PDEs. Dual
inhibition of PDE3/PDE4 and PDE4/PDE7 may synergistically
suppress inflammatory diseases such as COPD. However,
because of the conservation of the active site of the PDE
isozymes, the PDE4 dual inhibitors may also inhibit other PDEs,
resulting in unpredictable beneficial or adverse effects.
A survey of nonselective PDE inhibitors containing
pyrazolopyridine-pyridazinone motifs led to the development
Figure 28. Diagram of the binding of compound 71 (yellow sticks) of dual PDE3/4 selective inhibitors for the treatment of
PDE4D (PDB code 5WQA). The dotted line represents hydrogen respiratory diseases. These dual PDE3/4 inhibitors often show
bond. Compound 1 is shown as green sticks. The binding residues of both bronchodilatory and anti-inflammatory activities, thus
PDE4D2 are shown as cyan and white sticks, respectively, for the having advantages over the singly selective inhibitors. For
structures of PDE4D-compound 71 and PDE4D-compound 1. example, KCA-1490 (72, Figure 29) displayed IC50 values of
360 and 42 nM against PDE3 and PDE4, respectively.137
Compounds 73 and 74 (Figure 29) are orally active and have
These interactions are comparable with those of synthetic PDE4 well-balanced dual PDE3/4-inhibitory activities. However, most
inhibitors. However, compound 71 shows more extensive of these agents caused adverse reactions in the gastrointestinal
interactions with some PDE4 residues that have rarely been system.138,139 In the past, at least five dual PDE3/4 inhibitors,
reported in the literature, such as Ser208, Pro356, and Ile376 compound 49 (Figure 20), pumafentrine (75, Figure 29),
(Figure 28). benzafentrine (76, Figure 29), tolafentrine (77, Figure 29), and
O https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 29. Chemical structures of dual PDE inhibitors 72−83.

compound 5 (Figure 4, Table 1), entered clinical trials, but most nM, Figure 29) binds to PDE4 through its pyrazolopyridine
of them, except compound 5, have been discontinued.140 group in a pattern similar to that of compound 28.143
Compound 5 is a long-acting dual PDE3/PDE4 inhibitor Compound 81 (Figure 29) was identified by yeast-based
developed by Verona Pharma as a bronchodilator and anti- screening and displayed dual PDE4/PDE7 inhibition at the
inflammatory agent for the treatment of asthma and COPD. micromolar level: 64−90% inhibition of PDE4 and 98%
Compound 5 triggers significant bronchiectasis in asthmatics, inhibition of PDE7 at 2 μM concentration of the compound.144
which is even detectable 7 h after administration, and induces A novel butanehydrazide derivative of purine-2,6-dione
bronchial protection against methacholine. For patients with (compound 82,Figure 29) appeared to be a dual PDE4/7
allergic rhinitis, a single dose of compound 5 (18 μg/kg) inhibitor (PDE4 IC50 = 1.4 μM, PDE7 IC50 = 3.2 μM).145
reduced inflammatory cells (eosinophils and neutrophils) 7 and In addition, a number of pyrazolopyridine derivatives showed
24 h after the allergen challenge. In the phase II clinical trial, a inhibitory activities on PDE4/PDE10/JNK (Jun N-terminal
single dose of nebulized compound 5 (18 μg/kg) showed a rapid kinase) with binding affinity in the micromolar range. For
bronchodilator response and a >15% increase in FEV1 in COPD example, compound 83 (Figure 29) showed IC50 values of 1.2,
patients and was tolerated without significant side effects. In 7.4, 0.3, and 1.2 μM against PDE4, PDE10, JNK2, and JNK3,
addition, a phase II clinical trial on compound 5 for the respectively. These compounds were considered to be useful for
treatment of cystic fibrosis is in progress. treating neurological diseases such as Parkinson’s disease and
Compound 78 (Figure 29), which contains a compound 49 neuropathic pain, and some were tested in a chronic constriction
fragment, was claimed as a dual PDE4/PDE7 inhibitor with an injury model in rats.146
IC50 of 12 nM for PDE4, but its exact number for PDE7 was not 7.b. Dual PDE4 Inhibitors in Conjugation with Other
reported.141 Compound 79 (Figure 29) is a moderate dual Proteins. Successful examples of the dual PDE4 inhibitors are
inhibitor with IC50 values of 2.9 μM and 1.4 μM against PDE4 conjugated with β2 agonists, M3 antagonists, and selective
and PDE7.142 In a series of spiroisoxazoline-containing dual serotonin reuptake inhibitors (SSRIs). These inhibitors are
inhibitors, compound 80 (PDE4 IC50 = 5 nM, PDE7 IC50 = 55 designed by covalently grafting two pharmacophores together
P https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 30. Chemical structures of dual PDE4 inhibitors 84−93.

with a flexible linker, and each pharmacophore targets its own disubstituted tetrahydrofuran of an SSRI inhibitor and inhibited
protein in the anticipation of a synergistic effect. serotonin reuptake with an IC50 value of 127 nM and PDE4D3
Compound 85 (Figure 30), which combines compound 28 with Ki of 2.0 nM. This combination showed synergistic
(Figure 13) and carmoterol (84,Figure 30), showed activities of antidepressant effects.133
both β2 agonist and PDE4 inhibitor with IC50 values of 8 and 5 The combination of pyrazolopyridine of PDE4 inhibitor 39
nM, respectively.130 The combination of the β2 agonist with M3 antagonists yielded a series of compounds with the
formoterol (86,Figure 30) and the PDE4 inhibitor 55 (Figure capacity of dual PDE4 inhibition and M3 antagonism, which
20) yielded compound 87 (Figure 30) that possessed moderate have IC50 values of 0.1−2511 nM for PDE4B and 10 nM to 33
inhibitory activity against PDE4B2 (IC50 = 278 nM) and μM for M3 antagonist. The best compound 90 (Figure 30)
excellent relaxant effects on tracheal tissue precontracted by shows potency for the treatment of respiratory and allergic
histamine (pEC50 = 9.3).131 By exploration of the composition diseases, such as COPD, asthma, rhinitis, and inflammatory
and length of the linker, a new series of hybrids that combined diseases.134 Compound 91 (Figure 30), a carbamate derivative,
formoterol (86) and the PDE4 inhibitor phthalazinone (50, showed both activities of PDE4 inhibition (IC50 ≈ 100 nM) and
Figure 20) were investigated as potent agents for the treatment muscarinic M3 receptor antagonism (IC50 ≈ 100 nM), but its
of asthma and COPD. The most potent one in this class is therapeutic effects need further study.135
compound 88 (Figure 30), which exhibited both a β agonist In addition, a series of orally bioavailable dual inhibitors of
effect (EC50 = 1.05 nM, pEC50 = 9.0) and PDE4B2 inhibition epoxide hydrolase (sEH)/PDE4 were developed for the
(IC50 = 92 nM).132 treatment of inflammatory pain.136 Unlike other dual inhibitors
Compound 89 (Figure 30), a dual PDE4/SSRI inhibitor, that were prepared by simple grafting, compound 92 (Figure 30)
grafted the PDE4 inhibitor compound 50 to the 2,5- was designed by linking the benzyl amide moiety of the sEH
Q https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

inhibitor to a methoxybenzene fragment (compound 14 on human diseases, modulates the PDE4/3-cAMP signaling
simulation) and showed highly potent and dual inhibition of pathway for part of its function.159 Another example is the recent
PDE4 and sEH with IC50 values of 8.1 nM and 2.1 nM, report that the cocktail of the antileukemia drug dasatinib and
respectively. Compound 93 (Figure 30) is a triple p38α MAPK/ the natural product quercetin prolongs the lifespan of mice by
JNK3/PDE4 inhibitor with IC50 values of 500, 700, and 200 nM, 36%,122 which may be achieved via the regulation of the PDE4-
respectively, and demonstrated desirable pharmacokinetic cAMP signaling pathway since compound 66 is a well-known
properties and low plasma protein binding in vitro and in vivo PDE4 inhibitor.121 Thus, simultaneous inhibition of multiple
even at the nanomolar doses. PDE families would be a promising direction for discovery of
PDE inhibitors and is worthy of further exploration.
8. PERSPECTIVE
While PDE4 inhibitors have been extensively investigated as
therapeutics for treatment of human diseases such as COPD,
■ AUTHOR INFORMATION
Corresponding Authors
only a few of them have been approved for practical use, due to Hengming Ke − Department of Biochemistry and Biophysics, and
side effects. It was thought that the side effects of nausea and Lineberger Comprehensive Cancer Center, University of North
emesis are caused by the poor selectivity of the PDE4 inhibitors Carolina, Chapel Hill, North Carolina 27599, United States;
between subfamilies of PDE4B and PDE4D. However, this Email: hengming_ke@med.unc.edu
argument is not supported by the recent studies of PDE4D Jianyou Shi − Personalized Drug Therapy Key Laboratory of
inhibitors (see the discussion in section 4). Sichuan Province, Sichuan Academy of Medical Science &
A statistical survey shows that the four approved PDE4 Sichuan Provincial People’s Hospital, School of Medicine of
inhibitors for the treatment of human diseases in market have University of Electronic Science and Technology of China,
only nanomolar affinity with PDE4, but the compounds that Chengdu 610072, China; orcid.org/0000-0002-1026-0282;
have picomolar affinity never progressed beyond phase II clinical Phone: 86-18908001802; Email: shijianyoude@126.com
trials. For example, Merck’s compounds 29 (IC50 = 10 pM
against PDE4B)147 and 31 (IC50 = 60 pM against PDE4B) and Authors
compound 42 (IC50 of 30 pM for PDE4B)148 have never been Ting Peng − Personalized Drug Therapy Key Laboratory of
reported to enter clinical trials. The tightest-bound compound Sichuan Province, Sichuan Academy of Medical Science &
28 (IC50 = 3.2 pM against PDE4) was tested in a dozen phase II Sichuan Provincial People’s Hospital, School of Medicine of
clinical trials on treatment of various disease but does not seem University of Electronic Science and Technology of China,
to advance to further trials. The failure of these very tight Chengdu 610072, China
binding PDE4 inhibitors might be associated with their side Baowen Qi − Center for Nanomedicine and Department of
effects. A possible reason to explain the failure might be Anesthesiology, Brigham and Women’s Hospital, Harvard
inhibition of nontargeted cellular proteins, which causes Medical School, Boston, Massachusetts 02115, United States
disturbance on normal physiological functions of cAMP Jun He − Cancer Center, Collaborative Innovation Center for
signaling. Besides, we believe that the cellular cAMP level Biotherapy, West China Hospital, Sichuan University, Sichuan
needs to be maintained in an equilibrium for cellular functions. 610041, China
Deep inhibition of the PDE4 activity may cause redundant Complete contact information is available at:
accumulation of cAMP and thus disturb normal physiological https://pubs.acs.org/10.1021/acs.jmedchem.9b02170
processes. This hypothesis of cellular balance of cAMP levels is
supported by the case of the first generation PDE4 inhibitor Author Contributions
rolipram (compound 14) that was shown to have two binding #
T.P., B.Q., and J.H. contributed equally to this manuscript.
sites with high and low binding affinity states (HARBS/LARBS) Notes
against PDE4.149 The binding of compound 14 to the high
The authors declare no competing financial interest.
affinity site showed antidepressant and anti-inflammatory
activities but was also correlated with side effects such as Biographies
vomiting and gastric acid secretion.150,151 Ting Peng obtained her Bachelor’s degree in Pharmacy at Zhengzhou
At present, several dual selective PDE4 inhibitors have been University, China, in 2016. She is now a graduate student at the
reported to show synergistic effects on human health, implying University of Electronic Science and Technology of China. Her
that simultaneous inhibition on multiple PDEs produces research interests involve the design and synthesis of small molecules
synergistic therapeutic effects. This argument is supported in for the treatment of tumor and neurological and inflammatory diseases.
theory by the facts that both cAMP and cGMP signaling Baowen Qi received his Ph.D. from the University of Montreal in
pathways play critical roles in physiological processes and Pharmaceutical Science in 2016 and is now a Postdoctoral Fellow at
disease development.152−158 Inhibition of PDE4 and other Brigham and Women Hospital, Harvard Medical School. His research
PDEs may increase levels of cAMP, cGMP, or both in cells and focuses on the development and formulation of novel therapeutic
thus boost physiological processes. This assumption of potential agents for the treatment of various diseases.
synergistic effects from simultaneous inhibition of multiple
PDEs is also supported by several experimental observations. Jun He, a Research Associate, received his Ph.D. in Pharmacy from
First, theophylline (compound 65), the first generation Sichuan University in 2011 and now works at the State Key Laboratory
antiasthma drug, nonselectively inhibits most PDEs with a few of Biotherapy of Sichuan University in China. His research focuses on
tens micromolar affinity and presumably functions via inhibition small molecule drug discovery and computer-aided drug design for the
of multiple PDEs. Second, many natural products that are treatment of diseases.
nutritional supplements to benefit human health have been Hengming Ke received his Ph.D. in Biophysics from Harvard
shown to be nonselective PDE inhibitors. For example, University in 1989 and is now a Professor at the University of North
resveratrol, a natural phenol that shows several beneficial effects Carolina at Chapel Hill. His research focuses on the crystal structures of

R https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

medically important proteins and rational drug design for the treatment (7) Bedioune, I.; Bobin, P.; Karam, S.; Lindner, M.; Mika, D.;
of diseases. Lechene, P.; Leroy, J.; Fischmeister, R.; Vandecasteele, G. Cyclic
nucleotide phosphodiesterases: role in the heart and therapeutic
Jianyou Shi received his Ph.D. in Medicinal Chemistry from Sichuan
perspectives. Biol. Aujourd'hui 2016, 210, 127−138.
University in 2010. After 3 years of postdoctoral training, he joined a (8) Peng, T.; Gong, J.; Jin, Y.; Zhou, Y.; Tong, R.; Wei, X.; Bai, L.; Shi,
group at the University of North Carolina at Chapel Hill in 2016, and he J. Inhibitors of phosphodiesterase as cancer therapeutics. Eur. J. Med.
worked in this group for 2 years. He is currently working at the Chem. 2018, 150, 742−756.
University of Electronic Science and Technology as a professor. His (9) Sakkas, L. I.; Mavropoulos, A.; Bogdanos, D. P. Phosphodiesterase
major scientific interests focus on the discovery of small molecules 4 inhibitors in immune-mediated diseases: mode of action, clinical
targeting oncogenesis, neurological, and inflammatory diseases. applications, current and future perspectives. Curr. Med. Chem. 2017,

■ ACKNOWLEDGMENTS
The authors are thankful for the financial support from the
24, 3054−3067.
(10) Hansen, R. T.; Zhang, H.-T. The past, present, and future of
phosphodiesterase-4 modulation for age-induced memory loss.
Advances in neurobiology 2017, 17, 169−199.
Sichuan Science and Technology Department Project (Grant (11) Bolger, G.; Michaeli, T.; Martins, T.; St John, T.; Steiner, B.;
2017JQ0038) and the Sichuan Provincial People’s Hospital Rodgers, L.; Riggs, M.; Wigler, M.; Ferguson, K. A family of human
Clinical Research and Transformation Fund Key Project (Grant phosphodiesterases homologous to the dunce learning and memory
2016LZ03).


gene product of drosophila melanogaster are potential targets for
antidepressant drugs. Mol. Cell. Biol. 1993, 13, 6558−6571.
ABBREVIATIONS USED (12) Card, G. L.; England, B. P.; Suzuki, Y.; Fong, D.; Powell, B.; Lee,
PDE, phosphodiesterase; cAMP, cyclic adenosine mono- B.; Luu, C.; Tabrizizad, M.; Gillette, S.; Ibrahim, P. N.; Artis, D. R.;
phosphate; cGMP, cyclic guanosine monophosphate; COPD, Bollag, G.; Milburn, M. V.; Kim, S. H.; Schlessinger, J.; Zhang, K. Y. J.
chronic obstructive pulmonary disease; β-AR, β-arrestin; GPCR, Structural basis for the activity of drugs that inhibit phosphodiesterases.
G-protein-coupled receptor; Gs, heterotrimeric guanine nucleo- Structure 2004, 12, 2233−2247.
tide binding protein; AC, adenylyl cyclase; AKAP, A-kinase- (13) Xu, R. X.; Hassell, A. M.; Vanderwall, D.; Lambert, M. H.;
anchoring protein; PKA, protein kinase A; EPAC, exchange Holmes, W. D.; Luther, M. A.; Rocque, W. J.; Milburn, M. V.; Zhao, Y.;
Ke, H.; Nolte, R. T. Atomic structure of PDE4: insights into
protein activated by cAMP; CREB, cAMP response element
phosphodiesterase mechanism and specificity. Science 2000, 288,
binding protein; C3G, cyanidin-3-glucoside; Rap1, Ras-
1822−1825.
proximate-1; MEK1/2, mitogen-activated protein kinase; (14) Conti, M.; Richter, W.; Mehats, C.; Livera, G.; Park, J.-Y.; Jin, C.
ERK1/2, extracellular signal-regulated kinase 1/2; NF-κB, Cyclic AMP-specific PDE4 phosphodiesterases as critical components
nuclear factor κ-light-chain-enhancer of activated B-cells; of cyclic AMP signaling. J. Biol. Chem. 2003, 278, 5493−5496.
MAPK, mitogen-activated protein kinase; mTORC1, mamma- (15) Dodge, K. L.; Khouangsathiene, S.; Kapiloff, M. S.; Mouton, R.;
lian target of rapamycin complex 1; UCR, upstream conserved Hill, E. V.; Houslay, M. D.; Langeberg, L. K.; Scott, J. D. MAKAP
regions; RACK1, receptor for activated C kinase 1; LPS, assembles a protein kinase A/PDE4 phosphodiesterase cAMP signaling
lipopolysaccharide; TNFα, tumor necrosis factor α; PD, module. EMBO J. 2001, 20, 1921−1930.
Parkinson’s disease; AD, Alzheimer’s disease; AAMI, age- (16) Taskén, K. A.; Collas, P.; Kemmner, W. A.; Witczak, O.; Conti,
associated memory impairment; IL, interleukin; CD4, cluster M.; Taskén, K. Phosphodiesterase 4D and protein kinase a type II
of differentiation 4; FEV1, forced expiratory volume; FVC, constitute a signaling unit in the centrosomal area. J. Biol. Chem. 2001,
forced vital capacity; FST, forced swim test; TST, tailsus- 276, 21999−22002.
pension test; NAH, N-acylhydrazone; NK, natural killer cell; (17) Garcia, A. M.; Martinez, A.; Gil, C. Enhancing cAMP levels as
SSRI, selective serotonin reuptake inhibitor; sEH, epoxide strategy for the treatment of neuropsychiatric disorders. Curr. Top. Med.
hydrolase; BALF, bronchoalveolar lavage fluid; JNK, Jun N- Chem. 2016, 16, 3527−3535.
terminal kinase. (18) Fertig, B. A.; Baillie, G. S. PDE4-mediated cAMP signalling. J.


Cardiovasc. Dev. Dis. 2018, 5, 8.
(19) Wu, C.; Rajagopalan, S. Phosphodiesterase-4 inhibition as a
REFERENCES
therapeutic strategy for metabolic disorders. Obes. Rev. 2016, 17, 429−
(1) Maurice, D. H.; Ke, H.; Ahmad, F.; Wang, Y.; Chung, J.; 441.
Manganiello, V. C. Advances in targeting cyclic nucleotide (20) Wang, P.; Wu, P.; Ohleth, K. M.; Egan, R. W.; Billah, M. M.
phosphodiesterases. Nat. Rev. Drug Discovery 2014, 13, 290−314. Phosphodiesterase 4B2 is the predominant phosphodiesterase species
(2) Conti, M.; Beavo, J. Biochemistry and physiology of cyclic and undergoes differential regulation of gene expression in human
nucleotide phosphodiesterases: essential components in cyclic
monocytes and neutrophils. Mol. Pharmacol. 1999, 56, 170−174.
nucleotide signaling. Annu. Rev. Biochem. 2007, 76, 481−511.
(21) Jin, S. L. C.; Conti, M. Induction of the cyclic nucleotide
(3) Francis, S. H.; Blount, M. A.; Corbin, J. D. Mammalian cyclic
phosphodiesterase PDE4B is essential for LPS-activated TNF-α
nucleotide phosphodiesterases: molecular mechanisms and physio-
logical functions. Physiol. Rev. 2011, 91, 651−690. responses. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 7628−7633.
(4) Baillie, G. S.; Tejeda, G. S.; Kelly, M. P. Therapeutic targeting of (22) Jones, N. A.; Boswell-Smith, V.; Lever, R.; Page, C. P. The effect
3′,5′-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat. of selective phosphodiesterase isoenzyme inhibition on neutrophil
Rev. Drug Discovery 2019, 18, 770−796. function in vitro. Pulm. Pharmacol. Ther. 2005, 18, 93−101.
(5) Keravis, T.; Lugnier, C. Cyclic nucleotide phosphodiesterase (23) Daguès, N.; Pawlowski, V.; Sobry, C.; Hanton, G.; Borde, F.;
(PDE) isozymes as targets of the intracellular signalling network: Soler, S.; Freslon, J. L.; Chevalier, S. Investigation of the molecular
benefits of PDE inhibitors in various diseases and perspectives for mechanisms preceding PDE4 inhibitor-induced vasculopathy in rats:
future therapeutic developments. Br. J. Pharmacol. 2012, 165, 1288− tissue inhibitor of metalloproteinase 1, a potential predictive biomarker.
1305. Toxicol. Sci. 2007, 100, 238−247.
(6) Eskandari, N.; Mirmosayyeb, O.; Bordbari, G.; Bastan, R.; Yousefi, (24) Cazzola, M.; Ora, J.; Puxeddu, E. Dual bronchodilation and
Z.; Andalib, A. A short review on structure and role of cyclic-3′,5′- exacerbations of COPD. J. Thorac. Dis. 2016, 8, 2383−2386.
adenosine monophosphate-specific phosphodiesterase 4 as a treatment (25) Woo, T. E.; Kuzel, P. Crisaborole 2% ointment (eucrisa) for
tool. J. Res. Pharm. Pract. 2015, 4, 175−181. atopic dermatitis. Skin Ther. Lett. 2019, 24, 4−6.

S https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

(26) Dozier, L.; Bartos, G.; Kerdel, F. Apremilast and psoriasis in the (44) Feng, X.; Wang, H.; Ye, M.; Xu, X. T.; Xu, Y.; Yang, W.; Zhang,
real world: a retrospective case series. J. Am. Acad. Dermatol. 2019, H. T.; Song, G.; Ke, H. Identification of a PDE4-specific pocket for the
DOI: 10.1016/j.jaad.2019.10.009. design of selective inhibitors. Biochemistry 2018, 57, 4518−4525.
(27) Rolan, P.; Hutchinson, M.; Johnson, K. Ibudilast: a review of its (45) Bruno, O.; Fedele, E.; Prickaerts, J.; Parker, L. A.; Canepa, E.;
pharmacology, efficacy and safety in respiratory and neurological Brullo, C.; Cavallero, A.; Gardella, E.; Balbi, A.; Domenicotti, C.;
disease. Expert Opin. Pharmacother. 2009, 10, 2897−2904. Bollen, E.; Gijselaers, H. J.; Vanmierlo, T.; Erb, K.; Limebeer, C. L.;
(28) Hansen, R. T., 3rd; Conti, M.; Zhang, H. T. Mice deficient in Argellati, F.; Marinari, U. M.; Pronzato, M. A.; Ricciarelli, R. GEBR-7b,
phosphodiesterase-4A display anxiogenic-like behavior. Psychopharma- a novel PDE4D selective inhibitor that improves memory in rodents at
cology 2014, 231, 2941−2954. non-emetic doses. Br. J. Pharmacol. 2011, 164, 2054−2063.
(29) Millar, J.; Pickard, B.; Mackie, S.; James, R.; Christie, S.; (46) Bruno, O.; Romussi, A.; Spallarossa, A.; Brullo, C.; Schenone, S.;
Buchanan, S.; Malloy, M.; Chubb, J.; Huston, E.; Baillie, G.; Thomson, Bondavalli, F.; Vanthuyne, N.; Roussel, C. New selective phosphodies-
P.; Hill, E.; Brandon, N.; Rain, J.; Camargo, L.; Whiting, P.; Houslay, terase 4D inhibitors differently acting on long, short, and supershort
M.; Blackwood, D.; Muir, W.; Porteous, D. DISC1 and PDE4B are isoforms. J. Med. Chem. 2009, 52, 6546−6557.
interacting genetic factors in schizophrenia that regulate cAMP (47) Ricciarelli, R.; Brullo, C.; Prickaerts, J.; Arancio, O.; Villa, C.;
signaling. Science 2005, 310, 1187−1191. Rebosio, C.; Calcagno, E.; Balbi, M.; van Hagen, B. T.; Argyrousi, E. K.;
(30) Zhang, C.; Cheng, Y.; Wang, H.; Wang, C.; Wilson, S. P.; Xu, J.; Zhang, H.; Pronzato, M. A.; Bruno, O.; Fedele, E. Memory-enhancing
Zhang, H. T. RNA interference-mediated knockdown of long-form effects of GEBR-32a, a new PDE4D inhibitor holding promise for the
phosphodiesterase-4D (PDE4D) enzyme reverses amyloid-I2̂ 42- treatment of Alzheimer’s disease. Sci. Rep. 2017, 7, 46320.
induced memory deficits in mice. J. Alzheimer's Dis. 2013, 38, 269−280. (48) Prosdocimi, T.; Mollica, L.; Donini, S.; Semrau, M. S.; Lucarelli,
(31) Li, Y. F.; Cheng, Y. F.; Huang, Y.; Conti, M.; Wilson, S. P.; A. P.; Aiolfi, E.; Cavalli, A.; Storici, P.; Alfei, S.; Brullo, C.; Bruno, O.;
O’Donnell, J. M.; Zhang, H. T. Phosphodiesterase-4D knockout and Parisini, E. Molecular bases of PDE4D inhibition by memory-
RNAi-mediated knockdown enhance memory and increase hippo- enhancing GEBR library compounds. Biochemistry 2018, 57, 2876−
campal neurogenesis via increased cAMP signaling. J. Neurosci. 2011, 2888.
31, 172−183. (49) Cooney, J. D.; Aguiar, R. C. T. Phosphodiesterase 4 inhibitors
(32) Rutten, K.; Misner, D. L.; Works, M.; Blokland, A.; Novak, T. J.; have wide-ranging activity in B-cell malignancies. Blood 2016, 128,
Santarelli, L.; Wallace, T. L. Enhanced long-term potentiation and 2886−2890.
impaired learning in phosphodiesterase 4D-knockout (PDE4D) mice. (50) Kim, S. W.; Rai, D.; Aguiar, R. C. Gene set enrichment analysis
unveils the mechanism for the phosphodiesterase 4B control of
Eur. J. Neurosci. 2008, 28, 625−632.
(33) Gong, B.; Vitolo, O. V.; Trinchese, F.; Liu, S.; Shelanski, M.; glucocorticoid response in B-cell lymphoma. Clin. Cancer Res. 2011, 17,
6723−6732.
Arancio, O. Persistent improvement in synaptic and cognitive functions
(51) Marko, D.; Pahlke, G.; Merz, K. H.; Eisenbrand, G. Cyclic 3′,5′-
in an Alzheimer mouse model after rolipram treatment. J. Clin. Invest.
nucleotide phosphodiesterases: potential targets for anticancer therapy.
2004, 114, 1624−1634.
Chem. Res. Toxicol. 2000, 13, 944−948.
(34) Gurney, M. E.; D’Amato, E. C.; Burgin, A. B. Phosphodiesterase-
(52) Jin, S. L.; Goya, S.; Nakae, S.; Wang, D.; Bruss, M.; Hou, C.;
4 (PDE4) molecular pharmacology and Alzheimer’s disease. Neuro-
Umetsu, D.; Conti, M. Phosphodiesterase 4B is essential for T(H)2-cell
therapeutics 2015, 12, 49−56.
function and development of airway hyperresponsiveness in allergic
(35) Jiang, T. F.; Sun, Q.; Chen, S. D. Oxidative stress: a major
asthma. J. Allergy Clin. Immunol. 2010, 126, 1252−1259.
pathogenesis and potential therapeutic target of antioxidative agents in (53) Robichaud, A.; Stamatiou, P. B.; Jin, S. L.; Lachance, N.;
Parkinson’s disease and Alzheimer’s disease. Prog. Neurobiol. 2016, 147, Macdonald, D.; Laliberté, F.; Liu, S.; Huang, Z.; Conti, M.; Chan, C. C.
1−19. Deletion of phosphodiesterase 4D in mice shortens alpha(2)-
(36) Subramaniam, S. R.; Chesselet, M. F. Mitochondrial dysfunction adrenoceptor-mediated anesthesia, a behavioral correlate of emesis. J.
and oxidative stress in Parkinson’s disease. Prog. Neurobiol. 2013, 106, Clin. Invest. 2002, 110, 1045−1052.
17−32. (54) Giovannoni, M. P.; Cesari, N.; Graziano, A.; Vergelli, C.;
(37) MacDonald, E.; Van der Lee, H.; Pocock, D.; Cole, C.; Thomas, Biancalani, C.; Biagini, P.; Dal Piaz, V. Synthesis of pyrrolo[2,3-
N.; VandenBerg, P. M.; Bourtchouladze, R.; Kleim, J. A. Novel d]pyridazinones as potent, subtype selective PDE4 inhibitors. J. Enzyme
phosphodiesterase type 4 inhibitor, HT-0712, enhances rehabilitation- Inhib. Med. Chem. 2007, 22, 309−318.
dependent motor recovery and cortical reorganization after focal (55) Burgin, A. B.; Magnusson, O. T.; Singh, J.; Witte, P.; Staker, B. L.;
cortical ischemia. Neurorehab. Neural. Re. 2007, 21, 486−496. Bjornsson, J. M.; Thorsteinsdottir, M.; Hrafnsdottir, S.; Hagen, T.;
(38) Clinical trials for HT-0712. https://www.clinicaltrials.gov/ct2/ Kiselyov, A. S.; Stewart, L. J.; Gurney, M. E. Design of
show/NCT02013310?term=HT-0712&rank=2 (accessed Aug 2, phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing
2019). cognition with improved safety. Nat. Biotechnol. 2010, 28, 63−72.
(39) Vellas, B.; Sol, O.; Snyder, P. J.; Ousset, P. J.; Haddad, R.; Maurin, (56) Cedervall, P.; Aulabaugh, A.; Geoghegan, K. F.; McLellan, T. J.;
M.; Lemarie, J. C.; Desire, L.; Pando, M. P.; Pando, M. P. EHT0202 in Pandit, J. Engineered stabilization and structural analysis of the
Alzheimer’s disease: a 3-month, randomized, placebo-controlled, autoinhibited conformation of PDE4. Proc. Natl. Acad. Sci. U. S. A.
double-blind study. Curr. Alzheimer Res. 2011, 8, 203−212. 2015, 112, 1414−1422.
(40) Clinical trials for etazolate. https://www.clinicaltrials.gov/ct2/ (57) Fox, D., 3rd; Burgin, A. B.; Gurney, M. E. Structural basis for the
show/NCT00880412?term=Etazolate&rank=1 (accessed Aug 2, design of selective phosphodiesterase 4B inhibitors. Cell. Signalling
2019). 2014, 26, 657−663.
(41) Clinical trials for BPN14770. https://www.clinicaltrials.gov/ct2/ (58) Zhang, C.; Xu, Y.; Chowdhary, A.; Fox, D., 3rd; Gurney, M. E.;
show/NCT03817684?term=BPN14770&rank=1 (accessed Aug 2, Zhang, H. T.; Auerbach, B. D.; Salvi, R. J.; Yang, M.; Li, G.; O’Donnell,
2019). J. M. Memory enhancing effects of BPN14770, an allosteric inhibitor of
(42) Clinical trials for MK0952. https://clinicaltrials.gov/ct2/show/ phosphodiesterase-4D, in wild-type and humanized mice. Neuro-
NCT00362024?term=MK-0952 (accessed Aug 2, 2019). psychopharmacology 2018, 43, 2299−2309.
(43) Zheng, S.; Kaur, G.; Wang, H.; Li, M.; Macnaughtan, M.; Yang, (59) Gurney, M. E.; Nugent, R. A.; Mo, X.; Sindac, J. A.; Hagen, T. J.;
X.; Reid, S.; Prestegard, J.; Wang, B.; Ke, H. Design, synthesis, and Fox, D., 3rd; O’Donnell, J. M.; Zhang, C.; Xu, Y.; Zhang, H. T.; Groppi,
structure-activity relationship, molecular modeling, and NMR studies V. E.; Bailie, M.; White, R. E.; Romero, D. L.; Vellekoop, A. S.; Walker,
of a series of phenyl alkyl ketones as highly potent and selective J. R.; Surman, M. D.; Zhu, L.; Campbell, R. F. Design and synthesis of
phosphodiesterase-4 inhibitors. J. Med. Chem. 2008, 51, 7673−7688. selective phosphodiesterase 4D (PDE4D) allosteric inhibitors for the

T https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

treatment of fragile X syndrome and other brain disorders. J. Med. early clinical development of 2-{6-2-(3,5-dichloro-4-pyridyl)acetyl-2,3-
Chem. 2019, 62, 4884−4901. dimethoxyphenoxy}-N-propylac etamide (LEO 29102), a soft-drug
(60) Titus, D. J.; Wilson, N. M.; Alcazar, O.; Calixte, D. A.; Dietrich, inhibitor of phosphodiesterase 4 for topical treatment of atopic
W. D.; Gurney, M. E.; Atkins, C. M. A negative allosteric modulator of dermatitis. J. Med. Chem. 2014, 57, 5893−5903.
PDE4D enhances learning after traumatic brain injury. Neurobiol. (79) Purushothaman, B.; Arumugam, P.; Kulsi, G.; Song, J. M. Design,
Learn. Mem. 2018, 148, 38−49. synthesis, and biological evaluation of novel catecholopyrimidine based
(61) Sutcliffe, J. S.; Beaumont, V.; Watson, J. M.; Chew, C. S.; Beconi, PDE4 inhibitor for the treatment of atopic dermatitis. Eur. J. Med. Chem.
M.; Hutcheson, D. M.; Dominguez, C.; Munoz-Sanjuan, I. Efficacy of 2018, 145, 673−690.
selective PDE4D negative allosteric modulators in the object retrieval (80) Tralau-Stewart, C. J.; Williamson, R. A.; Nials, A. T.; Gascoigne,
task in female cynomolgus monkeys (Macaca fascicularis). PLoS One M.; Dawson, J.; Hart, G. J.; Angell, A. D. R.; Solanke, Y. E.; Lucas, F. S.;
2014, 9, No. e102449. Wiseman, J.; Ward, P.; Ranshaw, L. E.; Knowles, R. G. GSK256066, an
(62) Ke, H.; Wang, H. Crystal structures of phosphodiesterases and exceptionally high-affinity and selective inhibitor of phosphodiesterase
implications on substrate specificity and inhibitor selectivity. Curr. Top. 4 suitable for administration by inhalation: in vitro, kinetic, and in vivo
Med. Chem. 2007, 7, 391−403. characterization. J. Pharmacol. Exp. Ther. 2011, 337, 145−154.
(63) Wang, H.; Robinson, H.; Ke, H. The molecular basis for different (81) Singh, D.; Petavy, F.; Macdonald, A. J.; Lazaar, A. L.; O’Connor,
recognition of substrates by phosphodiesterase families 4 and 10. J. Mol. B. J. The inhaled phosphodiesterase 4 inhibitor GSK256066 reduces
Biol. 2007, 371, 302−307. allergen challenge responses in asthma. Respir. Res. 2010, 11, 26.
(64) Giembycz, M. A. An update and appraisal of the cilomilast Phase (82) Henriksson, K.; Lisius, A.; Sjo, P.; Storm, P. Novel 5,6-
III clinical development programme for chronic obstructive pulmonary Dihydropyrazolo[3,4-E][L,4]Diazepin-4 (IH)-One Derivatives for the
disease. Br. J. Clin. Pharmacol. 2006, 62, 138−152. Treatment of Asthma and Chronic Obstructive Pulmonary Disease.
(65) Giembycz, M. A. Can the anti-inflammatory potential of PDE4 Patent WO2007040435, 2007.
inhibitors be realized: guarded optimism or wishful thinking? Br. J. (83) Palle, V. P.; Balachandran, A.-N. I. I. S.; Gupta, N.; Kukreja, G.;
Pharmacol. 2008, 155, 288−290. Khera, M. K.; Baregama, L. K.; Mandadapu, R.; Ray, A.; Dastidar, S. G.
(66) Ichikawa, H.; Okamoto, S.; Kamada, N.; Nagamoto, H.; Phosphodiesterase Inhibitors. Patent WO2005051931, 2005.
Kitazume, M. T.; Kobayashi, T.; Chinen, H.; Hisamatsu, T.; Hibi, T. (84) Ting, P. C.; Lee, J. F.; Kuang, R.; Cao, J.; Gu, D.; Huang, Y.; Liu,
Tetomilast suppressed production of proinflammatory cytokines from Z.; Aslanian, R. G.; Feng, K.-I.; Prelusky, D.; Lamca, J.; House, A.;
human monocytes and ameliorated chronic colitis in IL-10-deficient Phillips, J. E.; Wang, P.; Wu, P.; Lundell, D.; Chapman, R. W.; Celly, C.
mice. Inflamm. Bowel Dis. 2008, 14, 1483−1490. S. Discovery of oral and inhaled PDE4 inhibitors. Bioorg. Med. Chem.
(67) Clinical trials for tetomilast. https://www.clinicaltrials.gov/ct2/ Lett. 2013, 23, 5528−5532.
results?cond=&term=Tetomilast&cntry=&state=&city=&dist= (ac- (85) Kuang, R.; Shue, H.-J.; Xiao, L.; Blythin, D. J.; Shih, N.-Y.; Chen,
cessed Aug 2, 2019). X.; Gu, D.; Schwerdt, J.; Lin, L.; Ting, P. C.; Cao, J.; Aslanian, R.;
(68) Clinical trials for apremilast. https://www.clinicaltrials.gov/ct2/ Piwinski, J. J.; Prelusky, D.; Wu, P.; Zhang, J.; Zhang, X.; Celly, C. S.;
results?cond=&term=Apremilast&cntry=&state=&city=&dist= (ac- Billah, M.; Wang, P. Discovery of oxazole-based PDE4 inhibitors with
cessed Aug 2, 2019). picomolar potency. Bioorg. Med. Chem. Lett. 2012, 22, 2594−2597.
(69) Zhang, H.; Zeng, G.; Gao, Y. Thiophene Derivatives. Patent (86) Press, N. J.; Taylor, R. J.; Fullerton, J. D.; Tranter, P.; McCarthy,
WO2010130224, 2010. C.; Keller, T. H.; Arnold, N.; Beer, D.; Brown, L.; Cheung, R.; Christie,
(70) Riggs-Sauthire, J.; Duarte Franco, J.; Allums-Donald, S. J.; Denholm, A.; Haberthuer, S.; Hatto, J. D. I.; Keenan, M.; Mercer, M.
Oligomer-Containing Apremilast Moiety Compounds. Patent K.; Oakman, H.; Sahri, H.; Tuffnell, A. R.; Tweed, M.; Trifilieff, A.
WO2012083153, 2012. Discovery and optimization of 4-(8-(3-Fluorophenyl)-1,7-naphthyr-
(71) Abarghaz, M.; Oumouch, S.; Lagouge, Y. 5-Substituted-1,4- idin-6-yl)transcyclohexanecarboxylic acid, an improved PDE4 inhibitor
Benzodiazepines as Phosphodiesterase Inhibitors. Patent for the Treatment of chronic obstructive pulmonary disease (COPD). J.
WO2009037302, 2009. Med. Chem. 2015, 58, 6747−6752.
(72) Sakaguchi, T.; Emi, H. Ointment with Excellent Formulation (87) Press, N. J.; Taylor, R. J.; Fullerton, J. D.; Tranter, P.; McCarthy,
Stability. Patent WO2012133492, 2012. C.; Keller, T. H.; Arnold, N.; Beer, D.; Brown, L.; Cheung, R.; Christie,
(73) Boland, S.; Alen, J.; Bourin, A.; Castermans, K.; Boumans, N.; J.; Denholm, A.; Haberthuer, S.; Hatto, J. D. I.; Keenan, M.; Mercer, M.
Panitti, L.; Vanormelingen, J.; Leysen, D.; Defert, O. Novel Roflumilast K.; Oakman, H.; Sahri, H.; Tuffnell, A. R.; Tweed, M.; Tyler, J. W.;
analogs as soft PDE4 inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, Wagner, T.; Fozard, J. R.; Trifilieff, A. Solubility-driven optimization of
4594−4597. phosphodiesterase-4 inhibitors leading to a clinical candidate. J. Med.
(74) Delcanale, M.; Amari, G.; Armani, E. Derivatives of 1-Phenyl-2- Chem. 2012, 55, 7472−7479.
Pyridinyl Alkyl Alcohols as Phosphodiesterase Inhibitors. Patent (88) Aoki, M.; Kobayashi, M.; Ishikawa, J.; Saita, Y.; Terai, Y.;
WO2009018909, 2009. Takayama, K.; Miyata, K.; Yamada, T. A novel phosphodiesterase type
(75) Moretto, N.; Caruso, P.; Bosco, R.; Marchini, G.; Pastore, F.; 4 inhibitor, YM976 (4-(3-chlorophenyl)-1,7-diethylpyrido 2,3-d
Armani, E.; Amari, G.; Rizzi, A.; Ghidini, E.; De Fanti, R.; Capaldi, C.; pyrimidin-2(1H)-one), with little emetogenic activity. J. Pharmacol.
Carzaniga, L.; Hirsch, E.; Buccellati, C.; Sala, A.; Carnini, C.; Exp. Ther. 2000, 295, 255−260.
Patacchini, R.; Delcanale, M.; Civelli, M.; Villetti, G.; Facchinetti, F. (89) Kuss, H.; Hoppmann, J.; Egerland, U.; Hoefgen, N.; Kronbach,
CHF6001 I: a novel highly potent and selective phosphodiesterase 4 T.; Rundfeldt, C. ELB353, a new selective PDE4-inhibitor with a high
inhibitor with robust anti-inflammatory activity and suitable for topical safety margin and anti-inflammatory effects in rat and ferret lungs.
pulmonary administration. J. Pharmacol. Exp. Ther. 2015, 352, 559− Inflammation Res. 2005, 54, S135−S136.
567. (90) Draheim, R.; Egerland, U.; Rundfeldt, C. Anti-inflammatory
(76) Clinical trials for CHF6001. https://www.clinicaltrials.gov/ct2/ potential of the selective phosphodiesterase 4 inhibitor N-(3,5-
results?term=CHF6001&draw=1&rank=8#rowId7 (accessed Aug 2, dichloro-pyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indole-3-yl]-
2019). glyoxylic acid amide (AWD 12-281), in human cell preparations. J.
(77) Govek, S. P.; Beauregard, C.; Gamache, D. A.; Hellberg, M. R.; Pharmacol. Exp. Ther. 2004, 308, 555−563.
Noble, S. A.; Shiau, A. K.; Thomas, D. J.; Yanni, J. M. (91) Poondra, R. R.; Nallamelli, R. V.; Meda, C. L.; Srinivas, B. N.;
Heteroarylalkoxysubstituted Quinolone Inhibitors of PDE4. Patent Grover, A.; Muttabathula, J.; Voleti, S. R.; Sridhar, B.; Pal, M.; Parsa, K.
WO2011093924, 2011. V. Discovery of novel 1,4-dihydropyridine-based PDE4 inhibitors.
(78) Felding, J.; Sorensen, M. D.; Poulsen, T. D.; Larsen, J.; Bioorg. Med. Chem. Lett. 2013, 23, 1104−1109.
Andersson, C.; Refer, P.; Engell, K.; Ladefoged, L. G.; Thormann, T.; (92) Rutter, A. R.; Poffe, A.; Cavallini, P.; Davis, T. G.; Schneck, J.;
Vinggaard, A. M.; Hegardt, P.; Sohoel, A.; Nielsen, S. F. Discovery and Negri, M.; Vicentini, E.; Montanari, D.; Arban, R.; Gray, F. A.; Davies,

U https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

C. H.; Wren, P. B. GSK356278, a potent, selective, brain-penetrant (105) Abdel-Rahman, H. M.; Abdel-Aziz, M.; Tinsley, H. N.; Gary, B.
Phosphodiesterase 4 Inhibitor that demonstrates anxiolytic and D.; Canzoneri, J. C.; Piazza, G. A. Design and synthesis of substituted
cognition-enhancing effects without inducing side effects in preclinical pyridazinone-1-acetylhydrazones as novel phosphodiesterase 4 inhib-
species. J. Pharmacol. Exp. Ther. 2014, 350, 153−163. itors. Arch. Pharm. 2016, 349, 104−111.
(93) Hamblin, J. N.; Angell, T. D. R.; Ballantine, S. P.; Cook, C. M.; (106) Gibson, L. C. D.; Hastings, S. F.; Mcphee, I.; Clayton, R. A.;
Cooper, A. W. J.; Dawson, J.; Delves, C. J.; Jones, P. S.; Lindvall, M.; Darroch, C. E.; Mackenzie, A.; Mackenzie, F. L.; Nagasawa, M.;
Lucas, F. S.; Mitchell, C. J.; Neu, M. Y.; Ranshaw, L. E.; Solanke, Y. E.; Stevens, P. A.; Mackenzie, S. J. The inhibitory profile of Ibudilast
Somers, D. O.; Wiseman, J. O. Pyrazolopyridines as a novel structural against the human phosphodiesterase enzyme family. Eur. J. Pharmacol.
class of potent and selective PDE4 inhibitors. Bioorg. Med. Chem. Lett. 2006, 538, 39−42.
2008, 18, 4237−4241. (107) Jin, S. L.; Ding, S. L.; Lin, S. C. Phosphodiesterase 4 and its
(94) Mitchell, C. J.; Ballantine, S. P.; Coe, D. M.; Cook, C. M.; Delves, inhibitors in inflammatory diseases. Biomed. J. 2012, 35, 197−210.
C. J.; Dowle, M. D.; Edlin, C. D.; Hamblin, J. N.; Holman, S.; Johnson, (108) Kojima, A.; Takita, S.; Sumiya, T.; Ochiai, K.; Iwase, K.; Kishi,
M. R.; Jones, P. S.; Keeling, S. E.; Kranz, M.; Lindvall, M.; Lucas, F. S.; T.; Ohinata, A.; Yageta, Y.; Yasue, T.; Kohno, Y. Phosphodiesterase
Neu, M.; Solanke, Y. E.; Somers, D. O.; Trivedi, N. A.; Wiseman, J. O. inhibitors. Part 6: design, synthesis, and structure-activity relationships
Pyrazolopyridines as potent PDE4B inhibitors: 5-heterocycle SAR. of PDE4-inhibitory pyrazolo 1,5-a pyridines with anti-inflammatory
Bioorg. Med. Chem. Lett. 2010, 20, 5803−5806. activity. Bioorg. Med. Chem. Lett. 2013, 23, 5311−5316.
(95) Le Roux, J.; Leriche, C.; Chamiot-Clerc, P.; Feutrill, J.; Halley, F.; (109) Kohno, Y.; Sumiya, T.; Takita, S.; Kojima, A. Heterocyclic
Papin, D.; Derimay, N.; Mugler, C.; Grepin, C.; Schio, L. Preparation Biaryl Derivative, and PDE4 Inhibitor Comprising Same as Active
and optimization of pyrazolo 1,5-a pyrimidines as new potent PDE4 Ingredient. Patent WO2010035745, 2010.
inhibitors. Bioorg. Med. Chem. Lett. 2016, 26, 454−459. (110) Ishii, N.; Shirato, M.; Wakita, H.; Miyazaki, K.; Takase, Y.;
(96) Suzuki, O.; Mizukami, K.; Etori, M.; Sogawa, Y.; Takagi, N.; Asano, O.; Kusano, K.; Yamamoto, E.; Inoue, C.; Hishinuma, I.
Tsuchida, H.; Morimoto, K.; Goto, T.; Yoshino, T.; Mikkaichi, T.; Antipruritic effect of the topical phosphodiesterase 4 inhibitor E6005
Hirahara, K.; Nakamura, S.; Maeda, H. Evaluation of the therapeutic ameliorates skin lesions in a mouse atopic dermatitis model. J.
index of a novel phosphodiesterase 4B-selective inhibitor over Pharmacol. Exp. Ther. 2013, 346, 105−112.
phosphodiesterase 4D in mice. J. Pharmacol. Sci. 2013, 123, 219−226. (111) Andoh, T.; Yoshida, T.; Kuraishi, Y. Topical E6005, a novel
(97) Goto, T.; Shiina, A.; Murata, T.; Tomii, M.; Yamazaki, T.; phosphodiesterase 4 inhibitor, attenuates spontaneous itch-related
Yoshida, K.-i.; Yoshino, T.; Suzuki, O.; Sogawa, Y.; Mizukami, K.; responses in mice with chronic atopy-like dermatitis. Exp. Dermatol.
Takagi, N.; Yoshitomi, T.; Etori, M.; Tsuchida, H.; Mikkaichi, T.; 2014, 23, 359−361.
Nakao, N.; Takahashi, M.; Takahashi, H.; Sasaki, S. Identification of the (112) Clinical trials for oglemilast. https://www.clinicaltrials.gov/
5,5-dioxo-7,8-dihydro-6H-thiopyrano 3,2-d pyrimidine derivatives as ct2/results?cond=&term=Oglemilast&cntry=&state=&city=&dist=
(accessed Aug 2, 2019).
highly selective PDE4B inhibitors. Bioorg. Med. Chem. Lett. 2014, 24,
(113) Gewald, R.; Grunwald, C.; Egerland, U. Discovery of triazines as
893−899.
potent, selective and orally active PDE4 inhibitors. Bioorg. Med. Chem.
(98) Goto, T.; Shiina, A.; Yoshino, T.; Mizukami, K.; Hirahara, K.;
Lett. 2013, 23, 4308−4314.
Suzuki, O.; Sogawa, Y.; Takahashi, T.; Mikkaichi, T.; Nakao, N.;
(114) Bonnert, R. V.; Humphries, T.; Hunt, S. F.; Sanganee, H. J.
Takahashi, M.; Hasegawa, M.; Sasaki, S. Synthesis and biological
Chemical Compounds 635: Pyrudopyrimidinediones as PDE4
evaluation of 5-carbamoyl-2-phenylpyrimidine derivatives as novel and
Inhibitors. Patent WO2008084236, 2008.
potent PDE4 inhibitors. Bioorg. Med. Chem. 2013, 21, 7025−7037. (115) Hartopp, P.; Johnson, T.; Sanganee, H. J. Combination
(99) Hagen, T. J.; Mo, X.; Burgin, A. B.; Fox, D., III; Zhang, Z.;
Comprising 6-fluoro-N-((1S,4S)-4-(6-fluoro-2,4-dioxo-1-(4’-(pipera-
Gurney, M. E. Discovery of triazines as selective PDE4B versus PDE4D zin-1-ylmethyl)biphenyl-3-yl)-1,2-dihydropyrido[2,3-d]pyrimidin-
inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, 4031−4034. 3(4H)-yl)cyclohexyl) imidazo[1,2-a]pyridine-2-carboxamide or A Salt.
(100) Srinivas, V. A. S. S.; Tadiparthi, R.; Sharma, G. V. R.; Patent WO2010004319, 2010.
Sappanimuthu, T.; Bhakiaraj, D. P.; Kachhadia, V.; Narasimhan, K.; (116) Andrews, G.; Cox, R. J.; De Savi, C.; Meghani, P.; Sanganee, H.
Thara, S. N.; Rajagopal, S.; Narayanan, S.; Parameswaran, V.; J.; Warner, D. J. Phenoxypyridinylamide Derivatives and their Use in
Janarthanam, V.; Narayanam, M.; Gadde, S.; Ramachandran, U.; the Treatment of PDE4 Mediated Disease States. Patent
Balasubramanian, G.; Surendran, N.; Narayanan, S.; Vishwakarma, L. WO2009144494, 2009.
S.; Saxena, S.; Reddy, G. O. Novel Heterocycles. Patent (117) Sengupta, S.; Mehta, G. Natural products as modulators of the
WO2009095773, 2009. cyclic-AMP pathway: evaluation and synthesis of lead compounds. Org.
(101) De Jonghe, S. C. A.; Dolusic, E.; Gao, L.-j.; Herdewijn, P. A. M. Biomol. Chem. 2018, 16, 6372−6390.
M.; Pfleiderer, W. E. Pyrido(3,2-d)Pyrimidines and Pharmaceutical (118) Rabe, K. F.; Magnussen, H.; Dent, G. Theophylline and
Compositions Useful for Medical Treatment. Patent US20090264415, selective PDE inhibitors as bronchodilators and smooth muscle
2009. relaxants. Eur. Respir. J. 1995, 8, 637−642.
(102) Menge, W. M. P. B.; Sterk, G. J.; Hatzelmann, A.; Barsig, J.; (119) Boswell-Smith, V.; Cazzola, M.; Page, C. P. Are phosphodies-
Marx, D.; Kley, H.; Christiaans, J. A. M.; Kley, H. P. New 2-(Piperidin- terase 4 inhibitors just more theophylline? J. Allergy Clin. Immunol.
4-yl)-4,5-Dihydro-2H-Pyridazin-3-one Derivatives Useful for Treat- 2006, 117, 1237−1243.
ment of e.g. Airway Disorders, Rheumatoid Arthritis, Osteoarthritis, (120) Rathee, P.; Chaudhary, H.; Rathee, S.; Rathee, D.; Kumar, V.;
Multiple Sclerosis and Graft Versus Host Reaction. Patent Kohli, K. Mechanism of action of flavonoids as anti-inflammatory
WO2005075456, 2005. agents: a review. Inflammation Allergy: Drug Targets 2009, 8, 229−235.
(103) Gracia, J.; Buil, M. A.; Castro, J.; Eichhorn, P.; Ferrer, M.; (121) Ko, W. C.; Shih, C. M.; Lai, Y. H.; Chen, J. H.; Huang, H. L.
Gavalda, A.; Hernandez, B.; Segarra, V.; Lehner, M. D.; Moreno, I.; Inhibitory effects of flavonoids on phosphodiesterase isozymes from
Pages, L.; Roberts, R. S.; Serrat, J.; Sevilla, S.; Taltavull, J.; Andres, M.; guinea pig and their structure-activity relationships. Biochem.
Cabedo, J.; Vilella, D.; Calama, E.; Carcasona, C.; Miralpeix, M. Pharmacol. 2004, 68, 2087−2094.
Biphenyl Pyridazinone derivatives as inhaled PDE4 inhibitors: (122) Xu, M.; Pirtskhalava, T.; Farr, J. N.; Weigand, B. M.; Palmer, A.
structural biology and structure-activity relationships. J. Med. Chem. K.; Weivoda, M. M.; Inman, C. L.; Ogrodnik, M. B.; Hachfeld, C. M.;
2016, 59, 10479−10497. Fraser, D. G.; Onken, J. L.; Johnson, K. O.; Verzosa, G. C.; Langhi, L. G.
(104) Gracia Ferrer, J.; Buil Albero, M. A.; Moreno Mollo, I. M.; Pages P.; Weigl, M.; Giorgadze, N.; LeBrasseur, N. K.; Miller, J. D.; Jurk, D.;
Santacana, L. M.; Roberts, R. S.; Sevilla Gome, S.; Taltavull Moll, J. (3- Singh, R. J.; Allison, D. B.; Ejima, K.; Hubbard, G. B.; Ikeno, Y.; Cubro,
Oxo)Pyridazin-4-ylurea Derivatives as PDE4 Inhibitors. Patent H.; Garovic, V. D.; Hou, X.; Weroha, S. J.; Robbins, P. D.;
WO2010069504, 2010. Niedernhofer, L. J.; Khosla, S.; Tchkonia, T.; Kirkland, J. L. Senolytics

V https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

improve physical function and increase lifespan in old age. Nat. Med. heteroaromatic-4,4-dimethylpyrazolones. Bioorg. Med. Chem. Lett.
2018, 24, 1246−1256. 2012, 22, 5833−5838.
(123) Harvey, A.; Gericke, N.; Viljoen, A. Use of Pharmaceutical (140) Abbott-Banner, K. H.; Page, C. P. Dual PDE3/4 and PDE4
Compositions Containing Mesembrenone. Patent WO2010106494, inhibitors: novel treatments for COPD and other inflammatory airway
2010. diseases. Basic Clin. Pharmacol. Toxicol. 2014, 114, 365−376.
(124) Harvey, A. L.; Young, L. C.; Viljoen, A. M.; Gericke, N. P. (141) Pelcman, B.; Krog-Jensen, C.; No, K.; Sanin, A. Pyrimidinones
Pharmacological actions of the south African medicinal and functional for Use as Medicaments. Patent WO2011114103, 2011.
food plant Sceletium tortuosum and its principal alkaloids. J. Ethno- (142) Pelcman, B.; Yee, J.; Gee, K.; Mackenzie, L. F.; Zhou, Y.; Han,
pharmacol. 2011, 137, 1124−1129. K. Isochromenones Useful in the Treatment of Inflammation. Patent
(125) Cai, Y. H.; Guo, Y.; Li, Z.; Wu, D.; Li, X.; Zhang, H.; Yang, J.; Lu, WO2010076564, 2010.
H.; Sun, Z.; Luo, H.-B.; Yin, S.; Wu, Y. Discovery and modelling studies (143) Rudra, S.; Gupta, N.; Chandrakant, K. G.; Jain, T.; Voleti, S. R.;
of natural ingredients from Gaultheria yunnanensis (FRANCH.) against Ray, A.; Dastidar, S. G.; Vijaykrishnan, L. Phosphodiesterase Inhibitors.
phosphodiesterase-4. Eur. J. Med. Chem. 2016, 114, 134−140. Patent WO2010046791, 2010.
(126) Chen, S. K.; Zhao, P.; Shao, Y. X.; Li, Z.; Zhang, C.; Liu, P.; He, (144) Hoffman, C. S.; Ivey, F. D.; Wyman Petri, A. Inhibitors of Cyclic
X.; Luo, H.-B.; Hu, X. Moracin M from Morus alba L. is a natural AMP Phosphodiesterases. Patent US20100227853, 2010.
phosphodiesterase-4 inhibitor. Bioorg. Med. Chem. Lett. 2012, 22, (145) Chlon-Rzepa, G.; Jankowska, A.; Slusarczyk, M.; Swierczek, A.;
3261−3264. Pociecha, K.; Wyska, E.; Bucki, A.; Gawalska, A.; Kolaczkowski, M.;
(127) Guo, Y. Q.; Tang, G. H.; Lou, L. L.; Li, W.; Zhang, B.; Liu, B.; Pawlowski, M. Novel butanehydrazide derivatives of purine-2,6-dione
Yin, S. Prenylated flavonoids as potent phosphodiesterase-4 inhibitors as dual PDE4/7 inhibitors with potential anti-inflammatory activity:
from Morus alba: Isolation, modification, and structure-activity design, synthesis and biological evaluation. Eur. J. Med. Chem. 2018,
relationship study. Eur. J. Med. Chem. 2018, 144, 758−766. 146, 381−394.
(128) Huang, Y.; Liu, X.; Wu, D.; Tang, G.; Lai, Z.; Zheng, X.; Yin, S.; (146) Gaeta, F. C. A.; Johnson, K. W.; Gross, M. I.; Ledeboer, A.
Luo, H.-B. The discovery, complex crystal structure, and recognition Substituted Pyrazolo[1,5-a]pyridine Compounds Having Multi-Target
mechanism of a novel natural PDE4 inhibitor from Selaginella pulvinata. Activity. Patent WO2010144416, 2010.
Biochem. Pharmacol. 2017, 130, 51−59. (147) Kuang, R.; Blythin, D.; Shih, N. Y.; Shue, H. J.; Chen, X.; Cao, J.;
(129) Kokkonen, K.; Kass, D. A. Nanodomain regulation of cardiac Gu, D.; Huang, Y.; Schwerdt, J. H.; Ting, P. C. Substituted 2-quinolyl-
cyclic nucleotide signaling by phosphodiesterases. Annu. Rev. oxazoles useful as PDE4 inhibitors. Patent WO2005116009, 2009.
Pharmacol. Toxicol. 2017, 57, 455−479. (148) Le Roux, J.; Leriche, C.; Chamiot-Clerc, P.; Feutrill, J.; Halley,
(130) Tannheimer, S. L.; Sorensen, E. A.; Cui, Z.-H.; Kim, M.; Patel, F.; Papin, D.; Derimay, N.; Mugler, C.; Grepin, C.; Schio, L.
L.; Baker, W. R.; Phillips, G. B.; Wright, C. D.; Salmon, M. The in vitro Preparation and optimization of pyrazolo[1,5-a]pyrimidines as new
pharmacology of GS-5759, a novel bifunctional phosphodiesterase 4 potent PDE4 inhibitors. Bioorg. Med. Chem. Lett. 2016, 26, 454−459.
(149) Barnette, M. S.; Grous, M.; Cieslinski, L. B.; Burman, M.;
inhibitor and long acting beta(2)-adrenoceptor agonist. J. Pharmacol.
Christensen, S. B.; Torphy, T. J. Inhibitors of phosphodiesterase IV
Exp. Ther. 2014, 349, 85−93.
(PDE IV) increase acid secretion in rabbit isolated gastric glands:
(131) Shan, W. J.; Huang, L.; Zhou, Q.; Jiang, H. L.; Luo, Z. H.; Lai, K.
correlation between function and interaction with a high-affinity
F.; Li, X. S. Dual beta(2)-adrenoceptor agonists-PDE4 inhibitors for the
rolipram binding site. J. Pharmacol. Exp. Ther. 1995, 273, 1396−1402.
treatment of asthma and COPD. Bioorg. Med. Chem. Lett. 2012, 22,
(150) Schmiechen, R.; Schneider, H. H.; Wachtel, H. Close
1523−1526.
correlation between behavioural response and binding in vivo for
(132) Huang, L.; Shan, W.; Zhou, Q.; Xie, J.; Lai, K.; Li, X. Design,
inhibitors of the rolipram-sensitive phosphodiesterase. Psychopharma-
synthesis and evaluation of dual pharmacology beta(2)-adrenoceptor
cology 1990, 102, 17−20.
agonists and PDE4 inhibitors. Bioorg. Med. Chem. Lett. 2014, 24, 249− (151) Jacobitz, S.; McLaughlin, M. M.; Livi, G. P.; Burman, M.;
253. Torphy, T. J. Mapping the functional domains of human recombinant
(133) Cashman, J. R.; Voelker, T.; Zhang, H. T.; O’Donnell, J. M. phosphodiesterase 4A: structural requirements for catalytic activity and
Dual inhibitors of phosphodiesterase-4 and serotonin reuptake. J. Med. rolipram binding. Mol. Pharmacol. 1996, 50, 891−899.
Chem. 2009, 52, 1530−1539. (152) Nunes, A. R.; Holmes, A. P.; Conde, S. V.; Gauda, E. B.;
(134) Callahan, J.F.; Wan, Z.; Yan, H. Dual Pharmacophores-PDE4- Monteiro, E. C. Revisiting cAMP signaling in the carotid body. Front.
Muscarinic Antagonistics. Patent WO2009100170, 2009. Physiol. 2014, 5, 406.
(135) Elisabetta, A.; Gabriele, A.; Carmelida, C.; Charles, B.-G. (153) Prysyazhna, O.; Eaton, P. Redox regulation of cGMP-
Carbamate Derivatives which are both Phosphodiesterase 4 (PDE4) dependent protein kinase Ialpha in the cardiovascular system. Front.
Enzyme Inhibitors and Muscarinic M3 Receptor Antagonists. Patent Pharmacol. 2015, 6, 139.
WO2015185650, 2015. (154) Inserte, J.; Garcia-Dorado, D. The cGMP/PKG pathway as a
(136) Blocher, R.; Wagner, K. M.; Gopireddy, R. R.; Harris, T. R.; Wu, common mediator of cardioprotection: translatability and mechanism.
H.; Barnych, B.; Hwang, S. H.; Xiang, Y. K.; Proschak, E.; Morisseau, Br. J. Pharmacol. 2015, 172, 1996−2009.
C.; Hammock, B. D. Orally available soluble epoxide hydrolase/ (155) Froese, A.; Nikolaev, V. O. Imaging alterations of
phosphodiesterase 4 dual inhibitor treats inflammatory pain. J. Med. cardiomyocyte cAMP microdomains in disease. Front. Pharmacol.
Chem. 2018, 61, 3541−3550. 2015, 6, 172.
(137) Kono, Y.; Ochiai, K.; Takita, T.; Kojima, A. 2-Alkyl-6- (156) Kokkonen, K.; Kass, D. A. Nanodomain regulation of cardiac
(pyrazolopyridin-4-yl)pyridazinone Derivative and PDE Inhibitor cyclic nucleotide signaling by phosphodiesterases. Annu. Rev.
Using them as Active Ingredient. Patent JP2010013354, 2010. Pharmacol. Toxicol. 2017, 57, 455−479.
(138) Ochiai, K.; Takita, S.; Eiraku, T.; Kojima, A.; Iwase, K.; Kishi, (157) Boularan, C.; Gales, C. Cardiac cAMP: production, hydrolysis,
T.; Fukuchi, K.; Yasue, T.; Adams, D. R.; Allcock, R. W.; Jiang, Z.; modulation and detection. Front. Pharmacol. 2015, 6, 203.
Kohno, Y. Phosphodiesterase inhibitors. Part 3: design, synthesis and (158) Koga, Y.; Tsurumaki, H.; Aoki-Saito, H.; Sato, M.; Yatomi, M.;
structure-activity relationships of dual PDE3/4-inhibitory fused Takehara, K.; Hisada, T. Roles of cyclic AMP response element binding
bicyclic heteroaromatic-dihydropyridazinones with anti-inflammatory activation in the ERK1/2 and p38 MAPK signalling pathway in central
and bronchodilatory activity. Bioorg. Med. Chem. 2012, 20, 1644−1658. nervous system, cardiovascular system, osteoclast differentiation and
(139) Ochiai, K.; Takita, S.; Kojima, A.; Eiraku, T.; Ando, N.; Iwase, mucin and cytokine production. Int. J. Mol. Sci. 2019, 20, 1346.
K.; Kishi, T.; Ohinata, A.; Yageta, Y.; Yasue, T.; Adams, D. R.; Kohno, Y. (159) Park, S. J.; Ahmad, F.; Philp, A.; Baar, K.; Williams, T.; Luo, H.;
Phosphodiesterase inhibitors. Part 4: design, synthesis and structure- Ke, H.; Rehmann, H.; Taussig, R.; Brown, A. L.; Kim, M. K.; Beaven, M.
activity relationships of dual PDE3/4-inhibitory fused bicyclic A.; Burgin, A. B.; Manganiello, V.; Chung, J. H. Resveratrol ameliorates

W https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

aging-related metabolic phenotypes by inhibiting cAMP phosphodies-


terases. Cell 2012, 148, 421−433.

X https://dx.doi.org/10.1021/acs.jmedchem.9b02170
J. Med. Chem. XXXX, XXX, XXX−XXX

You might also like