Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

Review

cAMP-specific phosphodiesterase inhibitors: promising drugs for inflammatory and neurological diseases

1.

Introduction

2.

PDE4 inhibitors

3.

PDE7 inhibitors

Ana Martinez & Carmen Gil†

4.

PDE8 inhibitors

Centro de Investigaciones Biolo´gicas (CSIC), Madrid, Spain

5.

Expert opinion

Introduction: PDEs are key enzymes in the adenosine and guanosine cyclic nucleotides (cAMP and cGMP) signaling cascade. Their inhibition increases cyclic nucleotide levels inside the cell. Thus, pharmacological modulation of PDE activity can have profound effects on the function of cells and organ systems throughout the body. Areas covered: Among the large PDE families, only PDE4, PDE7 and PDE8 are cAMP-specific hydrolyzing enzymes. cAMP is an important second messenger not only by its involvement in a vast number of physiological processes but also by activation of protein kinase A, exchange protein activated by cAMP (Epac) and cAMP response element-binding (CREB) or cyclic nucleotide-gated channels. Clearly, such enzymes represent ideal drug targets for the pharmacological treatment of many pathologies. The discovery and development of small molecules targeting cAMP-specific PDEs reported in the last 5 years is the focus of the present review. Expert opinion: The first PDE4 inhibitors recently reached the market, having avoided, by different strategies, their dose-limiting side effects (after more than two decades of drug development). Meanwhile, new cAMP-specific PDE7 and PDE8 inhibitors emerged as effective and safe drugs for severe unmet diseases. The therapeutic potential of these inhibitors will be tested in the near future, as many of these drug candidates are ready to start clinical trials. Keywords: cAMP, PDE inhibitors, PDE4, PDE7, PDE8 Expert Opin. Ther. Patents [Early Online]

1.

Introduction

The cAMP signaling system was discovered more than half a century ago [1]. This nucleotide is able to convert the signal of a large variety of extracellular stimuli into specific cellular responses. In fact, cAMP is a second messenger that regulates many diverse cellular functions, including gene transcription, cell migration, mitochondrial homeostasis, cell proliferation and cell death [2]. cAMP is generated from ATP through the action of adenylyl cyclase, whereas PDEs hydrolyze this second messenger to its inactive 5¢-monophosphate. The only way to inactivate cAMP is to degrade it through the action of PDEs. These enzymes are able to degrade not only cAMP but also cGMP. Up to now, eleven human PDE families are known. They present different selectivity for cAMP or cGMP and possess different intracellular distributions and catalytic/regulatory properties [3,4]. Among the large PDE families, only PDE4, PDE7 and PDE8 are cAMP-specific, whereas PDE5, PDE6 and PDE9 are specific for cGMP. The remaining PDE families hydrolyze both cyclic nucleotides [5]. As the imbalance of cAMP in inflammatory cells can lead to several inflammatory disorders, great efforts in the last two decades have 10.1517/13543776.2014.968127 © 2014 Informa UK, Ltd. ISSN 1354-3776, e-ISSN 1744-7674 All rights reserved: reproduction in whole or in part not permitted

1

A. Martinez & C. Gil

PDE7 and PDE8 inhibitors and their potential as drug candidates for human unmet diseases.

Article highlights. .

. .

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

.

.

Pharmacological modulation of cAMP levels through PDE activity is a good therapeutic strategy to alleviate several diseases in which inflammation plays a critical role. Only PDE4, PDE7 and PDE8 are cAMP-specific hydrolyzing enzymes. Although the therapeutic potential of PDE4 inhibitors has been widely reported, the dose-limiting side effects, such as nausea and emesis, have significantly delayed their clinical success. New research strategies, such as targeting PDE7 and/or PDE8, are in progress to develop compounds able to increase cellular cAMP levels but with a reduced side-effect profile. Development of PDE7 and PDE8 inhibitors alone or in combination with PDE4 inhibitors may provide innovative pharmacological therapies for severe unmet diseases.

This box summarizes key points contained in the article.

been devoted to the discovery and development of PDE4 inhibitors as pharmacological agents [6]. Small molecules targeting PDE4, such as rolipram, have shown promising results in preclinical models of different inflammatory diseases, but success in the clinical development of PDE4 inhibitors has been limited due to a number of dose-limiting side effects, such as nausea and emesis [7]. The mechanism by which PDE4 inhibition results in these adverse gastrointestinal effects is not fully understood but probably includes both central and peripheral sites of action. In fact, the human area postrema and other nuclei related to the emetic reflex express PDE4B and 4D [8]. Moreover, the emetic effect of PDE4 inhibitors is caused, at least in part, via binding to the high-affinity rolipram binding site (HARBS) in the brain in vivo [9]. Roflumilast, the first marketed PDE4 inhibitor, was approved in Europe in 2010 and in the USA in 2011 as an oral add-on treatment for chronic obstructive pulmonary disease (COPD) patients [10]. Since then, other PDE4 inhibitors have progressed in clinical development (reviewed in [11]). A second oral small-molecule inhibitor of PDE4, apremilast, has been recently approved in the USA for the treatment of active psoriatic arthritis [12,13], while the PDE4 inhibitor ASP9831 failed in Phase II trials due to the lack of efficacy in non-alcoholic steatohepatitis [14]. In addition, new research strategies are in progress to develop compounds able to increase cellular cAMP levels with a reduced side-effect profile. These include development of subtypeselective and non-brain penetrant PDE4 inhibitors, compounds with a reduced affinity for HARBS, or allosteric enzyme inhibitors [15]. Another strategy is the inhibition of different cAMP-specific PDEs, such as PDE7 and/or PDE8 [16,17], the new emerging targets in the field. This review will be focused specifically on cAMP-specific PDE4, 2

2.

PDE4 inhibitors

The PDE4 inhibitor family has attracted considerable attention in the last decade, because of their potential therapeutic uses in a range of major disease areas where inflammation plays a severe pathological role [18-21]. PDE4 enzymes specifically hydrolyze cAMP and are encoded by four distinct genes (PDE4A--PDE4D). Their isoforms show distinct and specific cell-type patterns of expression and distribution, and they play major regulatory roles in many cell types and tissues, including leukocytes, airway and vascular smooth muscle, vascular endothelium and the brain [5,22]. The involvement of PDE4 in pathological processes associated with these tissues suggests a great potential for pharmacological intervention in a large variety of disorders, such as inflammatory and neurological pathologies, through modulation of cAMP levels [23-27]. In fact, a number of PDE4 inhibitors have reached clinical trials for the treatment of inflammatory diseases [22,28]. Despite the failure of most of them because of side effects, such as emesis and nausea, roflumilast was approved in 2011 for the treatment of COPD [29] and apremilast received its approval in 2014 as the first target-specific oral psoriasis drugs (Figure 1) [30,31]. Although both are pan-inhibitors of PDE4A--4D, they have demonstrated that it is possible to achieve an improved therapeutic window. The improved tolerability of roflumilast is due to a suitable pharmacokinetic profile without a sharp Cmax [32], while the reason why apremilast has reduced emetic effects is not yet disclosed [33]. All these facts have maintained the interest in the development of new drugs targeting PDE4, and several small molecules have been patented over the past 5 years as PDE4 inhibitors. The inventions cover the preparation thereof, composition of them, combinations and therapeutic uses thereof. Most of them are claimed to be good anti-inflammatory agents based on the well-known relationship between cAMP levels and inflammatory processes. The range of diseases is huge, and due to the recently approval of roflumilast as treatment for COPD, most of the recently disclosed patents are focused on airway diseases. The avoidance of gastrointestinal effects is the main issue in this second generation of PDE4 inhibitors, and administration via inhalation has been one of the strategies used [26]. As representative examples of the new chemical structures (Figure 2), Chiesi Farmaceutici produced different series of 1-phenyl-2-pyridinyl alkyl alcohols (I [34], II [35] and III [36]) for the treatment of different pulmonary diseases after inhaled administration [37]. The most advanced candidate of these series, named CHF 6001, has reached clinical trial Phase II for asthma and COPD treatment [38]. CHF 6001 is a novel selective PDE4 inhibitor, optimized for inhaled delivery to improve efficacy and tolerability. The anti-inflammatory activity of CHF 6001 has been proven in vitro and in several

Expert Opin. Ther. Patents (2014) 24(12)

cAMP-specific phosphodiesterase inhibitors

Cl

N

Cl

O O

N H

F

O

F

Roflumilast OCH3 OEt

O N

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

Me

NH O

O

O S O Me

Apremilast

Figure 1. PDE4 inhibitors on the market.

animal models of airway inflammation, showing higher potency than roflumilast and more targeted anti-inflammatory effects than corticosteroids in COPD models [37]. Moreover, these compounds can also be used as coadjuvant in leukemia treatment [39]. Almirall recently claimed that heterocyclic compounds, such as 7,8-dihydro-1,6-naphthyrydin-5(6H)-one derivatives (IV [40]), are potent PDE4 inhibitors with IC50 values in the subnanomolar range and have the ability to decrease TNF-a levels. Other heterocyclic compounds, such as 5substituted-1,4-benzodiazepines (V [41]), benzo-fused heterocycles (VI [42], VII [43], VIII [44]), triazolopyridazines (IX [45]), triazolopyridines (X [46]), pyrimidinone derivatives (XI [47]) or biaryls (XII [48]) used alone or in combination with other known drugs, have also been recently claimed to be effective in the treatment of a variety of disorders involving inflammatory states, such as asthma, COPD, rheumatoid arthritis, autoimmune pathology and neurological disorders, such as depression, Parkinson’s and Alzheimer’s disease. However, nothing has been reported about the potential emetic side effects even when CNS diseases are claimed as the target for these compounds. Emesis has been shown to be a dose-limiting side effect. Thus, the selective PDE4D inhibitor, GEBR-7b, is able to improve memory in rodents at non-emetic doses due to the relatively low doses required to improve memory [49]. Moreover, allosteric PDE4D modulators have reduced potential to cause emesis because they do not completely inhibit enzyme activity while maintaining efficacy [15]. Another potential alternative used to overcome the doselimiting side effects of PDE4 inhibitors is to develop new chemotypes that target other members of the cAMPdependent family, such as PDE7, which is also distributed in almost all pro-inflammatory and immune cells [50]. This is the strategy claimed by Ranbaxy Laboratories Limited to design their nitrogen heterocycle dual inhibitors of PDE4 and PDE7 (XIII [51]). It is expected that these dual inhibitors may achieve a higher therapeutic index and thereby exhibit a

lower propensity to cause adverse side effects that are characteristic when targeting PDE4 alone [52]. Another alternative to overcome the PDE4 emetogenic side effects is the development of soft drugs that are inactivated early in the blood for the local treatment of inflammatory diseases, such as the drugs disclosed by Amakem NV for the treatment of dry eye disease (XIV [53]). Finally, it is worth mentioning that in addition to Chiesi Farmaceutici’s inhibitor, other PDE4 inhibitors have entered clinical development in the last 5 years (Figure 3). Two compounds from GlaxoSmithKline, GSK356278 and GSK25 6066, are in clinical trials Phase I and II, respectively. GSK356278 [54,55], which inhibits PDE4A, PDE4B and PDE4D enzyme activity with a pIC50 of 8.6, 8.8 and 8.7, respectively, and binds the high-affinity rolipram-binding site, has shown anxiolytic effects in common marmosets and enhances performance in a model of executive function in cynomolgus macaques with no adverse effects at the doses tested. This therapeutic profile, together with the good data in clinical Phase I, supports further evaluation of GSK3 56278 in patients for depression, anxiety and Huntington’s disease [56]. Although studies have now been completed, results are not yet available. Regarding GSK256066, the safety and tolerability of this intranasally administered PDE4 inhibitor has been reported [57], while its efficacy in COPD and seasonal allergic rhinitis patients is under study [58]. Merck also has one PDE4 inhibitor in clinical development. The 8-biarylnaphthyridinone called MK0952 was discovered with the aim of penetrating into the brain through the blood--brain barrier [59]. This compound showed benefits in animal models for long-term memory and mild cognitive impairment and is currently in clinical trials for mild-tomoderate Alzheimer’s disease [60]. This study finished in 2013 but the results are not yet available. Dart NeuroScience LLC has conducted clinical trials of a PDE4 inhibitor named HT-0712 [61]. As the preclinical data in a model of focal ischemia showed a cortical reorganization [62] and improvement in hippocampus-dependent memory in aged mice after HT-0712 administration [63], a Phase IIa study is being carried out in elderly subjects with age-associated memory impairment [61]. Topical administration of PDE4 inhibitors for the treatment of dermatological diseases, such as atopic dermatitis, is also being tested on humans [64]. That is the case of Anacor, which is the sponsor of the clinical development of AN2728, a novel boron-containing small molecule that reduces the production of TNF-a by inhibition of PDE4. Other cytokines, including IL-12 and IL-23, which are proteins believed to be involved in the inflammation process and immune response, presented reduced levels after treatment with AN2728 [65]. Because AN2728 has demonstrated safety in 18 clinical trials, it is expected to become a safe and effective treatment option for patients who suffer from atopic dermatitis and that it could potentially be combined with topical corticosteroids and vitamin D analogs for patients with

Expert Opin. Ther. Patents (2014) 24(12)

3

A. Martinez & C. Gil

R1 K

R2

R3

A Z

O

O

Cl

(O)n

N

R

O

F

O

Cl

R3

N H

R′

B

A 3′

R

R4 4′

R

N H

R5

R4

N R1

N

X

O

R2

R1

III

N

R

Z

R4 R1O

II R3

N

O

O R2O

R1O

1

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

N R3

I

R2

Z O

R2O

O

F

19

R3 R5

n

R3

X

N

Y

R4

IV

(R1)n

R′′

V

R2

R2

X

Y

VI

VII O R1

3

R

O

m OR1

R1

R

R2O R

O

N

X

m

O

X

X N

N

Y

N

A

X

VIII

N

N

R2 (R6)n

1

R3

2

N

R2

(R7)n

N

3

R

OR4

A

IX

X

XI

OR 1 OR 2 OR 3 Z1 Z2 Z3

Y

N

XII

O W

N

A n

OR2

NH OR1

R4

R3

O

O R3

R6 N

Z4

X

O R5 NR1R2 N

R′3

R4 O

XIII

XIV

Figure 2. Patented PDE4 inhibitors during the period 2009 -- 2013.

mild-to-moderate psoriasis. The Phase III study in mild-tomoderate atopic dermatitis started in March 2014, and it is expected to produce the final results in the first half of 2015 [66]. 3.

PDE7 inhibitors

The second cAMP-specific PDE discovered was PDE7 [67]. This isoenzyme is encoded by two genes, PDE7A and PDE7B. High mRNA concentrations of both PDE7A and PDE7B are found in rat brains and in numerous peripheral 4

tissues, although the distribution of these enzymes at the protein level has not been reported [68]. Within the brain, PDE7A mRNA is abundant in the olfactory bulb, hippocampus and several brain-stem nuclei [69]. The highest concentrations of PDE7B transcripts in the brain are found in the cerebellum, dentate gyrus of the hippocampus and the striatum [70,71]. There is very little information regarding the physiological functions that are regulated by PDE7, although its transcriptional activation through the dopamine receptor D1 is well documented [72]. It has been shown that PDE7 is involved in pro-inflammatory processes and is necessary for the

Expert Opin. Ther. Patents (2014) 24(12)

cAMP-specific phosphodiesterase inhibitors

OCH3 Me

N

N

Me S

O N N

HN

Et N N

O

O

S

NH2 N

HN

O O

N

Me

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

GSK256066

O

O

Me NH

N H N

Me

Me

GSK356278

O

O

N

OH B O

N

O

O OCH3

MK-0952

F

COOH

HT-0712

AN2728

Figure 3. PDE4 inhibitors in clinical trials.

induction of T-cell proliferation [73]. Moreover, an increase in PDE7B expression in chronic lymphocytic leukemia has been described [74]. In a previous comprehensive review article, a number of chemically diverse PDE7 inhibitors were shown and the development of inhibitors of PDE7 was reported as a new approach to be explored for the treatment of neurological and inflammatory disorders due to the increasing levels of cAMP [75]. In the last 5 years, several methods have been developed for the discovery of PDE7 inhibitors [16,76,77] because great interest in them emerged as a way to overcome the side effects of PDE4 inhibitors [78]. These new compounds provide useful pharmacological tools for the assessment of the physiological and pathological function of PDE7 in different cellular and animal models. Several new and diverse chemical structures have been recently reported, such as thioxoquinazolines [79], quinazolines [80] and pyrimidine derivatives [81]. Moreover, some of them are emerging as valuable clinical candidates [82], while the potential benefit of their concomitant administration with a PDE4 inhibitor has been recently reported [83] (see also Ranbaxy patent [51]). The pharmacological profile of one heterocyclic smallmolecule inhibitor of PDE7 called S14, which belongs to a new family of quinazoline derivatives patented by CSIC (Figure 4) (XV [82]), has been well studied, mainly for neurological disorders. First, a detailed theoretical and experimental study of the mechanism of PDE7 inhibition by the quinazoline S14 reveals that it is a specific and allosteric modulator [84], providing a subtle modulation of cAMP homeostasis with great beneficial effects in animal models. S14 conferred significant neuronal protection against different insults both in the human dopaminergic cell line SH-SY5Y and in primary rat

mesencephalic cultures. S14 treatment reduced microglial activation and improved motor function in the lipopolysaccharide rat model of Parkinson’s disease (PD) [85] and in a model of spinal cord injury [86]. The efficacy of S14 is abolished by blocking the cAMP signaling pathways that operate through cAMP-dependent protein kinase A. These results show that inhibition of the PDE7 enzyme leads to dopaminergic neuronal protection and, therefore, its inhibitors may exert useful therapeutic actions in patients with PD by modifying the course of the neurodegenerative process [87]. Additionally, the neuroprotective effects of S14 are present in a model of Alzheimer’s disease, and thus, S14 is a good candidate for further development with the aim of reaching clinical trials [88]. More recently, the adequate CNS penetration of a different PDE7 inhibitor belonging to the imidazopyridazinone family in mice allowed it to be tested in the MPTP-induced PD model and haloperidol-induced catalepsy model, probing the differential pharmacology of PDE7 in the striatal pathway [89] and confirming the utility of this new class of drugs for the treatment of PD [87]. In fact, Omeros has claimed the use of several diverse PDE7 inhibitors for the treatment of different movement disorders, such as PD, among others [90]. It is noteworthy that cAMP is involved in the differentiation of several cell lines or stem cells into dopamine neurons [91], which is considered a promising strategy for cell replacement therapy in PD. Recently, it has been shown that the quinazoline PDE7 inhibitor S14 is able to promote differentiation of stem cells isolated from the adult rat hippocampus and from the subventricular zone into tyrosine hydroxylasepositive mature neurons [92]. Furthermore, S14 promotes new neurogenesis in the substantia nigra after the administration of 6-OHDA.

Expert Opin. Ther. Patents (2014) 24(12)

5

A. Martinez & C. Gil

X R

N

A N R2

XV

R2 S N

R1 Y 3

R

N

R1

N

XVI

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

Figure 4. Quinazoline- and iminothiadiazole-like PDE7 inhibitors.

Finally, CSIC has also claimed other different chemotype for targeting PDE7: the 5-imino-thiadiazole family (Figure 4) (XVI [93]). These compounds produce a decrease in the inflammatory reaction in a spinal cord injury model [86] and induce neuronal in vivo neuroprotection [94]. The enhancement of oligodendrocyte precursor survival and their differentiation promoted by PDE7 inhibitor treatment is promising. These important properties may facilitate the therapeutic remyelination strategies for the treatment of multiple sclerosis [95]. Moreover, the therapeutic potential of PDE7 inhibitors for multiple sclerosis has been unequivocally assessed in murine experimental allergic encephalomyelitis models with treatment with chemically diverse PDE7 inhibitors [96-98]. This fact opens a new and promising therapeutic strategy for the future treatment of this neurological disease. Recently, crosstalk between PDE7 and glycogen synthase kinase 3, a key target in neurological pathologies, has been described, thus increasing the potential of these cAMPspecific PDE inhibitors in the treatment of many unmet diseases [99]. Overall, inhibition of PDE7 is a good strategy to modulate cAMP with its beneficial effects in dopaminergic cell protection and decreased neuroinflammatory activation, and also because of the lack of gastrointestinal side effects [100,101], such as those produced by PDE4 inhibitors. This makes the PDE7 inhibitors good drug candidates for CNS diseases and especially for PD and multiple sclerosis.

of PDE8B could be an interesting therapeutic intervention to enhance memory and motor function [115]. In addition, overexpression of brain protein levels of PDE8 has been recently associated with age, confirming the pharmacological interest of its inhibitors for the treatment of age-associated diseases [116]. However, only a few PDE8-selective inhibitors are available, and thus, there is an urgent need not only to explore the physiological and pathological role of PDE8 but also to develop them for clinical trials of human disease. The particular structures of these PDEs suggest that specific drugs for PDE8 might be developed [17]. In fact, the recent development of a new PDE8 inhibitor by Pfizer, named PF-4957325 [117], has helped to identify PDE8 as a novel target for suppression of effector T-cell functions due to the important role of the PDE8 family in regulating cAMP signaling in these cells. Until the discovery of this new inhibitor, only dipyridamole was used experimentally to study the cellular roles of PDE8, even though it is a nonselective inhibitor. Due to the lack of PDE8 inhibitors and the potential of this target, high-throughput screening (HTS) campaigns were carried out by Pfizer with the aim of discovering new hits that will allow an in-depth study of the PDE8 function and further development of its inhibitors (Figure 5). Of particular interest were the hits found based on tetrahydroisoquinoline [118] or triazolopyrimidine [119] scaffolds, which have been the starting points of medicinal chemistry programs to increase potency and selectivity [120]. The utility of PDE8 inhibitors alone or in combination with PDE4 or PDE7 inhibitors has also been claimed to treat inflammation and immune-related disorders by the University of Connecticut [121], and a cell-based HTS was developed to detect PDE8 inhibitors and PDE4/PDE8 inhibitors combination. By using this methodology, the dual-inhibitor BC8-15was discovered and supported the proposal that this combination might be a good therapeutic candidate to elevate steroidogenesis in Leydig cells [122]. 5.

4.

PDE8A and PDE8B exhibit a high-affinity cAMP-specific PDE activity that was not inhibited by various PDE inhibitors, including IBMX (3-isobutyl-1-methylxanthine) [102-104]. PDE8A is expressed in Leydig cells in the testes [105] and also in cardiomyocytes [106], whereas PDE8B is found in the thyroid gland [107], brain [107,108] and adrenal gland [109]. The biological function of PDE8 is still unknown, although based on its pattern of expression it is possible to speculate an important physiological role in different processes, including testosterone production, thyroid function or cognitive disorders. Based on this, a PDE8 inhibitor could be useful as a treatment of thyroid dysfunction [110] and modulation of steroidogenesis in the testes and adrenal glands [111-113]. Moreover, as overexpression of PDE8B mRNA is observed in the brains of Alzheimer’s patients [114], partial inactivation 6

Expert opinion

PDE8 inhibitors Enhancing intracellular cAMP levels may be a good therapeutic strategy to alleviate several diseases in which inflammation plays a critical role, such as some pulmonary, dermatological and severe neurological diseases. cAMP-specific PDEs, PDE4, PDE7 and PDE8, are the enzymes responsible for cAMP degradation, and their inhibition produces an increase in intracellular cAMP levels. They are druggable targets, and their inhibitors have great therapeutic potential for several unmet disorders [123]. PDE4 inhibitors were the first compounds in clinical development after effective preclinical results, but the emetic side effect found in humans stopped or delayed the success of these clinical trials. However, roflumilast and apremilast have been approved in the last 2 years for the treatment of COPD and psoriasis. These drugs target PDE4 with an improved therapeutic window. Although the mechanism by which PDE4 inhibitors result in these side

Expert Opin. Ther. Patents (2014) 24(12)

cAMP-specific phosphodiesterase inhibitors

OH

NH2

F3C

N N

N

N

N N

N

N

N

HO N

N

N

NH

N

N S

PF-04957325

N

N

OH

N

N

O

N

O OH

OH

Dipyridamole

BC8-15

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

Figure 5. PDE8 inhibitors.

effects is not fully understood, several strategies to dissociate the beneficial and detrimental effects of PDE4 inhibitors have led to some degree of success, and the second generation of PDE4 inhibitors is on the way with better pharmacokinetics profiles. Different administration patterns, such as inhalation or topical administration, are being developed for PDE4 inhibitors. Moreover, compounds that simultaneously target other cAMP-specific PDEs, such as PDE7 or PDE8, are being testing to obtain new effective drugs with a better therapeutic window [78]. Some of these novel dual PDE7/ PDE4 inhibitors have been claimed by different groups [124], while a new antisense therapy targeting these two PDEs has shown efficacy in decreasing lung inflammation in mice [125,126]. In the meantime, PDE7 inhibitors emerged as a promising family of new anti-inflammatory drugs. They have proven efficacy in animal models of multiple sclerosis, Parkinson’s and Alzheimer’s diseases, while they have shown a decrease in the inflammatory reaction of a spinal cord injury model and stroke. Some companies, such as Omeros and Araclon, are involved in active development of PDE7 inhibitors to reach clinical trials. They are looking for innovative drugs for neurodegenerative diseases [82] and/or movement disorders [90]. PDE7 inhibitors have emerged as potent and effective drugs in preclinical models, offering good candidates to be future drugs with an apparent lack of emetic side effects. Finally, PDE8 inhibitors can be considered as a new opportunity to be explored as valuable drug candidates for neurological diseases, including Alzheimer’s disease and also

pathologies related to male fertility. Research has just started in this field, and the near future will see the results. In summary, enhancing cAMP intracellular levels that restore the endogenous homeostasis of this important second messenger that is lost in different unmet diseases is a good therapeutic approach for human diseases. As the only way to inactivate cAMP is to degrade it through the action of PDEs, cAMP-PDEs have emerged as promising therapeutic targets for inflammatory diseases as their inhibitors are innovative drugs to treat a great variety of pathologies. Different expression of these targets throughout the body may modulate the therapeutic potential of their inhibitors. While some PDE4 inhibitors have recently reached the market, only human clinical trials will confirm the great hope in the new PDE7 and PDE8 inhibitors as effective drugs for severe unmet diseases. Moreover, based on the promising preclinical studies reviewed here, we are confident in the clinical success of PDE7 inhibitors.

Declaration of interest The authors were supported by Financial support from MINECO and FEDER funds (EU program) (project nos. SAF2012-33600 and IPT-2012-0762-300000). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Expert Opin. Ther. Patents (2014) 24(12)

7

A. Martinez & C. Gil

Bibliography

12.

Poole RM, Ballantyne AD. Apremilast: first global approval. Drugs 2014;74:825-37

23.

1.

Rall TW, Sutherland EW. Formation of a cyclic adenine ribonucleotide by tissue particles. J Biol Chem 1958;232:1065-76

13.

Page CP, Spina D. Phosphodiesterase inhibitors in the treatment of inflammatory diseases. Handb Exp Pharmacol 2011;391-414

2.

Lefkimmiatis K, Zaccolo M. cAMP signaling in subcellular compartments. Pharmacol Ther 2014;doi:10.1016/j. pharmthera.2014.03.008

Varada S, Tintle SJ, Gottlieb AB. Apremilast for the treatment of psoriatic arthritis. Expert Rev Clin Pharmacol 2014;7:239-50

24.

14.

Ratziu V, Bedossa P, Francque SM, et al. Lack of efficacy of an inhibitor of PDE4 in phase 1 and 2 trials of patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2014;12:1724-30

Garcia-Osta A, Cuadrado-Tejedor M, Garcia-Barroso C, et al. Phosphodiesterases as therapeutic targets for Alzheimer’s disease. ACS Chem Neurosci 2012;3:832-44

25.

Richter W, Menniti FS, Zhang HT, et al. PDE4 as a target for cognition enhancement. Expert Opin Ther Targets 2013;17:1011-27

26.

Lipworth BJ. Phosphodiesterase-4 inhibitors for asthma and chronic obstructive pulmonary disease. Lancet 2005;365:167-75

27.

Kumar N, Goldminz AM, Kim N, et al. Phosphodiesterase 4-targeted treatments for autoimmune diseases. BMC Med 2013;11:96

28.

Tenor H, Hatzelmann A, Beume R, et al. Pharmacology, clinical efficacy, and tolerability of phosphodiesterase-4 inhibitors: impact of human pharmacokinetics. Handb Exp Pharmacol 2011;85-119

29.

Giembycz MA, Field SK. Roflumilast: first phosphodiesterase 4 inhibitor approved for treatment of COPD. Drug Des Devel Ther 2010;4:147-58

30.

Harrison C. Trial watch: PDE4 inhibitor leads wave of target-specific oral psoriasis drugs. Nat Rev Drug Discov 2013;12:335

31.

Schafer PH, Day RM. Novel systemic drugs for psoriasis: mechanism of action for apremilast, a specific inhibitor of PDE4. J Am Acad Dermatol 2013;68:1041-2

32.

Bethke TD, Bohmer GM, Hermann R, et al. Dose-proportional intraindividual single- and repeated-dose pharmacokinetics of roflumilast, an oral, once-daily phosphodiesterase 4 inhibitor. J Clin Pharmacol 2007;47:26-36

33.

Darout E, Menhaji-Klotz E, Chappie TA. PDE4: recent medicinal chemisty strategies to mitigate adverse effects. In: Liras S, Bell AS, editors. Phosphodiesterase and their inhibitors. Wiley-VCH; Weinheim, Germany: 2014. 45-64

3.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

therapeutic target. Drug Discov Today 2005;10:1503-19

Papers of special note have been highlighted as either of interest () or of considerable interest () to readers.

4.

5.

Francis SH, Blount MA, Corbin JD. Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol Rev 2011;91:651-90 Johansson EM, Reyes-Irisarri E, Mengod G. Comparison of cAMPspecific phosphodiesterase mRNAs distribution in mouse and rat brain. Neurosci Lett 2012;525:1-6 Conti M, Jin SL. The molecular biology of cyclic nucleotide phosphodiesterases. Prog Nucleic Acid Res Mol Biol 1999;63:1-38

6.

Pages L, Gavalda A, Lehner MD. PDE4 inhibitors: a review of current developments (2005 - 2009). Expert Opin Ther Pat 2009;19:1501-19

7.

Press NJ, Banner KH. PDE4 inhibitors a review of the current field. Prog Med Chem 2009;47:37-74

8.

Mori F, Perez-Torres S, De Caro R, et al. The human area postrema and other nuclei related to the emetic reflex express cAMP phosphodiesterases 4B and 4D. J Chem Neuroanat 2010;40:36-42

15.

..

16.

17.

Burgin AB, Magnusson OT, Singh J, et al. Design of phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing cognition with improved safety. Nat Biotechnol 2010;28:63-70 Excellent paper that describes the design of PDE4D allosteric modulators which reduced potential to cause emesis. Safavi M, Baeeri M, Abdollahi M. New methods for the discovery and synthesis of PDE7 inhibitors as new drugs for neurological and inflammatory disorders. Expert Opin Drug Discov 2013;8:733-51 Wang H, Yan Z, Yang S, et al. Kinetic and structural studies of phosphodiesterase-8A and implication on the inhibitor selectivity. Biochemistry 2008;47:12760-8

18.

Jeffery P. Phosphodiesterase 4-selective inhibition: novel therapy for the inflammation of COPD. Pulm Pharmacol Ther 2005;18:9-17

Hirose R, Manabe H, Nonaka H, et al. Correlation between emetic effect of phosphodiesterase 4 inhibitors and their occupation of the high-affinity rolipram binding site in Suncus murinus brain. Eur J Pharmacol 2007;573:93-9

19.

Keshavarzian A, Mutlu E, Guzman JP, et al. Phosphodiesterase 4 inhibitors and inflammatory bowel disease: emerging therapies in inflammatory bowel disease. Expert Opin Investig Drugs 2007;16:1489-506

10.

Pinner NA, Hamilton LA, Hughes A. Roflumilast: a phosphodiesterase-4 inhibitor for the treatment of severe chronic obstructive pulmonary disease. Clin Ther 2012;34:56-66

20.

Jin SL, Ding SL, Lin SC. Phosphodiesterase 4 and its inhibitors in inflammatory diseases. Chang Gung Med J 2012;35:197-210

21.

11.

Gavalda A, Roberts RS. Phosphodiesterase-4 inhibitors: a review of current developments (2010 - 2012). Expert Opin Ther Pat 2013;23:997-1016 An useful review to understand the progress in the development of PDE4 inhibitors up to 2012.

Matera MG, Page C, Cazzola M. PDE inhibitors currently in early clinical trials for the treatment of asthma. Expert Opin Investig Drugs 2014;23:1267-75 An useful review which describes the PDE inhibitors that have been evaluated for the treatment of asthma.

9.

..

8

.

22.

Houslay MD, Schafer P, Zhang KY. Keynote review: phosphodiesterase-4 as a

Expert Opin. Ther. Patents (2014) 24(12)

cAMP-specific phosphodiesterase inhibitors

34.

35.

36.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

37.

38.

39.

40.

41.

42.

43.

44.

45.

Chiesi Farmaceutici S.p.A. Preparation of benzoic acid phenylpyridinyl ethyl ester derivatives for use as phosphodiesterase inhibitors. WO2010089107A1; 2010

46.

Chiesi Farmaceutici S.p.A. Derivatives of 1-phenyl-2-pyridinyl alkyl alcohols as phosphodiesterase inhibitors and their preparation. WO2012168226A1; 2012

47.

Chiesi Farmaceutici S.p.A. Derivatives of 1-phenyl-2-pyridinyl alkyl alcohols as phosphodiesterase inhibitors and their preparation. US20130079313A1; 2013 Armani E, Amari G, Rizzi A, et al. Novel class of benzoic acid ester derivatives as potent PDE4 inhibitors for inhaled administration in the treatment of respiratory diseases. J Med Chem 2014;57:793-816 Clinical trials for CHF6001. Available from: http://clinicaltrials.gov/ct2/results? term=chiesi+PDE4&Search=Search [Last accessed 24 July 2014] Chiesi Farmaceutici S.p.A. Potentiation induced by PDE4 inhibitors in the treatment of leukemia. WO2013087749A1; 2013 Almirall, S.A. Preparation of new 7,8dihydro-1,6-naphthyridin-5(6H)-one derivatives as PDE4 inhibitors. EP2380890A1; 2011 Via Pharmaceuticals, Inc. Benzodiazepine derivatives as phosphodiesterase inhibitors and their preparation, pharmaceutical compositions and use in the treatment of inflammation. WO2009037302A1; 2009 Matrix Laboratories Ltd. Novel heterocyclic compounds, pharmaceutical compositions containing them and processes for their preparation. WO2009115874A2; 2009 Orchid Research Laboratories Ltd. Azolyldibenzofuran derivatives as phosphodiesterase inhibitors and their preparation and use for the treatment of PDE-4-mediated diseases. WO2010084402A2; 2010 Leo Pharma A/S. Preparation of benzodioxole or benzodioxepine heterocyclic compounds as phosphodiesterase inhibitors. WO2011160632A1; 2011 US Department of Health and Human Service. Preparation of triazolopyridazines, triazolothiadiazines, and related heterocycles as phosphodiesterase inhibitors for therapy. WO2009089027A1; 2009

Leo Pharma A/S. Preparation of triazolopyridines as phosphodiesterase inhibitors for treatment of dermal diseases. WO2010069322A1; 2010 Biolipox AB. Preparation of pyrimidinone derivatives as PDE4 and PDE7 inhibitors useful in treatment of inflammatory and other diseases. WO2010029299A1; 2010

48.

LEO Pharma A/S. Biaryl derivatives as phosphodiesterase inhibitors and their preparation and use in the treatment of diseases. WO2011134468A1; 2011

49.

Bruno O, Fedele E, Prickaerts J, et al. GEBR-7b, a novel PDE4D selective inhibitor that improves memory in rodents at non-emetic doses. Br J Pharmacol 2011;164:2054-63

50.

Lee R, Wolda S, Moon E, et al. PDE7A is expressed in human Blymphocytes and is up-regulated by elevation of intracellular cAMP. Cell Signal 2002;14:277-84

51.

Ranbaxy Laboratories Ltd. Preparation of pyrazolo[3,4-b]pyridine compounds as phosphodiesterase inhibitors for treating immune disorders, CNS disorders, and other diseases. WO2010046791A1; 2010

52.

Vijayakrishnan L, Rudra S, Eapen MS, et al. Small-molecule inhibitors of PDEIV and -VII in the treatment of respiratory diseases and chronic inflammation. Expert Opin Investig Drugs 2007;16:1585-99

53.

Amakem NV. Preparation of pyridine derivatives as PDE4 inhibitors. WO2013021021A1; 2013

54.

Guercio G, Castoldi D, Giubellina N, et al. Overall synthesis of GSK356278: quick delivery of a PDE4 inhibitor using a fit-for-purpose approach. Org Process Res Dev 2010;14:1153-61

55.

Rutter AR, Poffe A, Cavallini P, et al. GSK356278, a potent, selective, brain penetrant PDE4 inhibitor that demonstrates anxiolytic and cognition enhancing effects without inducing emesis in preclinical species. J Pharmacol Exp Ther 2014;350:153-63

56.

Clinical trials for GSK356278. Available from: http://clinicaltrials.gov/ct2/results? term=GSK356278&Search=Search [Last accessed 24 July 2014]

57.

Watz H, Mistry SJ, Lazaar AL, et al. Safety and tolerability of the inhaled phosphodiesterase 4 inhibitor

Expert Opin. Ther. Patents (2014) 24(12)

GSK256066 in moderate COPD. Pulm Pharmacol Ther 2013;26:588-95 58.

Clinical trials for GSK256066. Available from: http://clinicaltrials.gov/ct2/results? term=GSK256066%3B&Search=Search [Last accessed 24 July 2014]

59.

Gallant M, Aspiotis R, Day S, et al. Discovery of MK-0952, a selective PDE4 inhibitor for the treatment of long-term memory loss and mild cognitive impairment. Bioorg Med Chem Lett 2010;20:6387-93

60.

Clinical trials for MK0952. Available from: http://clinicaltrials.gov/ct2/results? term=MK0952&Search=Search [Last accessed 24 July 2014]

61.

Clinical trials for HT-0712. Available from: http://clinicaltrials.gov/ct2/results? term=HT-0712&Search=Search [Last accessed 24 July 2014]

62.

MacDonald E, Van der Lee H, Pocock D, et al. A novel phosphodiesterase type 4 inhibitor, HT0712, enhances rehabilitation-dependent motor recovery and cortical reorganization after focal cortical ischemia. Neurorehabil Neural Repair 2007;21:486-96

63.

Peters M, Bletsch M, Stanley J, et al. The PDE4 inhibitor HT-0712 improves hippocampus-dependent memory in aged mice. Neuropsychopharmacology 2014;doi 10.1038/npp.2014.154

64.

Nazarian R, Weinberg JM. AN-2728, a PDE4 inhibitor for the potential topical treatment of psoriasis and atopic dermatitis. Curr Opin Investig Drugs 2009;10:1236-42

65.

Freund YR, Akama T, Alley MR, et al. Boron-based phosphodiesterase inhibitors show novel binding of boron to PDE4 bimetal center. FEBS Lett 2012;586:3410-14

66.

Clinical trials for AN-2728. Available from: http://clinicaltrials.gov/ct2/results? term=AN-2728&Search=Search [Last accessed 24 July 2014]

67.

Michaeli T, Bloom TJ, Martins T, et al. Isolation and characterization of a previously undetected human cAMP phosphodiesterase by complementation of cAMP phosphodiesterase-deficient Saccharomyces cerevisiae. J Biol Chem 1993;268:12925-32

68.

Hoffmann R, Abdel’Al S, Engels P. Differential distribution of rat PDE-7

9

A. Martinez & C. Gil

mRNA in embryonic and adult rat brain. Cell Biochem Biophys 1998;28:103-13 69.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

70.

71.

72.

73.

74.

75.

.

76.

77.

10

78.

Giembycz MA. Life after PDE4: overcoming adverse events with dualspecificity phosphodiesterase inhibitors. Curr Opin Pharmacol 2005;5:238-44 An useful review which describes the alternatives for PDE4 inhibitors.

88.

Perez-Gonzalez R, Pascual C, Antequera D, et al. Phosphodiesterase 7 inhibitor reduced cognitive impairment and pathological hallmarks in a mouse model of Alzheimer’s disease. Neurobiol Aging 2013;34:2133-45

Miro X, Perez-Torres S, Palacios JM, et al. Differential distribution of cAMPspecific phosphodiesterase 7A mRNA in rat brain and peripheral organs. Synapse 2001;40:201-14

79.

89.

Sasaki T, Kotera J, Omori K. Novel alternative splice variants of rat phosphodiesterase 7B showing unique tissue-specific expression and phosphorylation. Biochem J 2002;361:211-20

Castan˜o T, Wang H, Campillo NE, et al. Synthesis, structural analysis, and biological evaluation of thioxoquinazoline derivatives as phosphodiesterase 7 inhibitors. ChemMedChem 2009;4:866-76

Banerjee A, Patil S, Pawar MY, et al. Imidazopyridazinones as novel PDE7 inhibitors: SAR and in vivo studies in Parkinson’s disease model. Bioorg Med Chem Lett 2012;22:6286-91

80.

90.

Reyes-Irisarri E, Perez-Torres S, Mengod G. Neuronal expression of cAMP-specific phosphodiesterase 7B mRNA in the rat brain. Neuroscience 2005;132:1173-85

Sanchez AI, Martinez-Barrasa V, Burgos C, et al. Synthesis and evaluation of quinazoline derivatives as phosphodiesterase 7 inhibitors. Bioorg Med Chem 2013;21:2370-8

Omeros Corp. The use of PDE7 inhibitors for the treatment of movement disorders. WO2008119057A2; 2008

91.

81.

Guo J, Watson A, Kempson J, et al. Identification of potent pyrimidine inhibitors of phosphodiesterase 7 (PDE7) and their ability to inhibit T cell proliferation. Bioorg Med Chem Lett 2009;19:1935-8

Tremblay RG, Sikorska M, Sandhu JK, et al. Differentiation of mouse Neuro 2A cells into dopamine neurons. J Neurosci Methods 2010;186:60-7

92.

Morales-Garcı´a JA, Perez-Castillo A, Redondo M, et al. The PDE7 inhibitor S14 induces neurogenesis in vitro and in a rodent model of Parkinson disease. Neurodeg Dis 2013;11:Supp1

93.

CSIC. 5-Imino-1,2,4-thiadiazoles derivatives useful for the treatment of neurodegenerative diseases. WO2011039403; 2011

94.

Susin C, Morales-Garcia JA, Aguilar-Morante D, et al. The new iminothiadiazole derivative VP1.14 ameliorates hippocampal damage after an excitotoxic injury. J Neurochem 2012;122:1193-202

95.

Morales-Garcia JA, Redondo M, Alonso-Gil S, et al. Phosphodiesterase 7 inhibition preserves dopaminergic neurons in cellular and rodent models of Parkinson disease. PLoS One 2011;6:e17240 A paper that reports the efficacy of a PDE7 inhibitor in a Parkinson’s disease model.

Medina-Rodriguez EM, Arenzana FJ, Pastor J, et al. Inhibition of endogenous phosphodiesterase 7 promotes oligodendrocyte precursor differentiation and survival. Cell Mol Life Sci 2013;70:3449-62

96.

Paterniti I, Mazzon E, Gil C, et al. PDE 7 inhibitors: new potential drugs for the therapy of spinal cord injury. PLoS One 2011;6:e15937

Gonzalez-Garcia C, Bravo B, Ballester A, et al. Comparative assessment of PDE 4 and 7 inhibitors as therapeutic agents in experimental autoimmune encephalomyelitis. Br J Pharmacol 2013;170:602-13

97.

Redondo M, Brea J, Perez DI, et al. Effect of phosphodiesterase 7 (PDE7) inhibitors in experimental autoimmune encephalomyelitis mice. Discovery of a new chemically diverse family of compounds. J Med Chem 2012;55:3274-84

98.

Redondo M, Palomo V, Brea J, et al. Identification in silico and experimental validation of novel phosphodiesterase

Sasaki T, Kotera J, Omori K. Transcriptional activation of phosphodiesterase 7B1 by dopamine D1 receptor stimulation through the cyclic AMP/cyclic AMP-dependent protein kinase/cyclic AMP-response element binding protein pathway in primary striatal neurons. J Neurochem 2004;89:474-83 Nakata A, Ogawa K, Sasaki T, et al. Potential role of phosphodiesterase 7 in human T cell function: comparative effects of two phosphodiesterase inhibitors. Clin Exp Immunol 2002;128:460-6 Zhang L, Murray F, Zahno A, et al. Cyclic nucleotide phosphodiesterase profiling reveals increased expression of phosphodiesterase 7B in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2008;105:19532-7 Gil C, Campillo NE, Perez DI, et al. Phosphodiesterase 7 (PDE7) inhibitors as new drugs for neurological and inflammatory disorders. Expert Opin Ther Pat 2008;18:1127-39 An useful review which describes the potential of PDE7 inhibitors for the treatment of neurological and inflammatory disorders. Alaamery MA, Wyman AR, Ivey FD, et al. New classes of PDE7 inhibitors identified by a fission yeast-based HTS. J Biomol Screen 2010;15:359-67 Castro A, Jerez MJ, Gil C, et al. CODES, a novel procedure for ligandbased virtual screening: PDE7 inhibitors as an application example. Eur J Med Chem 2008;43:1349-59

.

82.

CSIC. Use of quinazoline derivatives for neurodegenerative diseases. WO2010133742; 2010

83.

Mokry J, Joskova M, Mokra D, et al. Effects of selective inhibition of PDE4 and PDE7 on airway reactivity and cough in healthy and ovalbuminsensitized guinea pigs. Adv Exp Med Biol 2013;756:57-64

84.

85.

.

86.

87.

Redondo M, Soteras I, Brea J, et al. Unraveling phosphodiesterase surfaces. Identification of phosphodiesterase 7 allosteric modulation cavities. Eur J Med Chem 2013;70:781-8

Martinez A, Gil C. Phosphodiesterase inhibitors as a new therapeutic approach for the treatment of Parkinson’s disease. In: Martinez A, Gil C, editors. Emerging drugs and targets for Parkinson’s disease. The Royal Society of Chemistry; Cambridge, UK: 2013. p. 294-307

Expert Opin. Ther. Patents (2014) 24(12)

cAMP-specific phosphodiesterase inhibitors

7 inhibitors with efficacy in experimental autoimmune encephalomyelitis mice. ACS Chem Neurosci 2012;3:793-803 99.

Expert Opin. Ther. Patents Downloaded from informahealthcare.com by Universitat de Girona on 10/14/14 For personal use only.

100.

..

101.

..

102.

103.

104.

105.

106.

107.

Morales-Garcia JA, Palomo V, Redondo M, et al. Crosstalk between phosphodiesterase 7 and glycogen synthase kinase-3: two relevant therapeutic targets for neurological disorders. ACS Chem Neurosci 2014;5:194-204 Yamamoto S, Sugahara S, Naito R, et al. The effects of a novel phosphodiesterase 7A and -4 dual inhibitor, YM-393059, on T-cell-related cytokine production in vitro and in vivo. Eur J Pharmacol 2006;541:106-14 A paper that reports the lack of emetic effect of a dual PDE7/PDE4 inhibitor in a surrogate model. Garcı´a AM, Brea J, Morales-Garcı´a JA, et al. Modulation of cAMP specific PDE without emetogenic activity: new sulfidelike PDE7 inhibitors. J Med Chem 2014;In press A paper that reports the lack of emetic effect of PDE7 inhibitors in a surrogate model. Fisher DA, Smith JF, Pillar JS, et al. Isolation and characterization of PDE8A, a novel human cAMP-specific phosphodiesterase. Biochem Biophys Res Commun 1998;246:570-7 Soderling SH, Bayuga SJ, Beavo JA. Cloning and characterization of a cAMPspecific cyclic nucleotide phosphodiesterase. Proc Natl Acad Sci USA 1998;95:8991-6 Hayashi M, Matsushima K, Ohashi H, et al. Molecular cloning and characterization of human PDE8B, a novel thyroid-specific isozyme of 3’,5’cyclic nucleotide phosphodiesterase. Biochem Biophys Res Commun 1998;250:751-6 Vasta V, Shimizu-Albergine M, Beavo JA. Modulation of Leydig cell function by cyclic nucleotide phosphodiesterase 8A. Proc Natl Acad Sci USA 2006;103:19925-30 Patrucco E, Albergine MS, Santana LF, et al. Phosphodiesterase 8A (PDE8A) regulates excitation-contraction coupling in ventricular myocytes. J Mol Cell Cardiol 2010;49:330-3 Hayashi M, Shimada Y, Nishimura Y, et al. Genomic organization, chromosomal localization, and alternative splicing of the human phosphodiesterase

8B gene. Biochem Biophys Res Commun 2002;297:1253-8 108.

109.

110.

Appenzeller S, Schirmacher A, Halfter H, et al. Autosomal-dominant striatal degeneration is caused by a mutation in the phosphodiesterase 8B gene. Am J Hum Genet 2010;86:83-7 Horvath A, Giatzakis C, Tsang K, et al. A cAMP-specific phosphodiesterase (PDE8B) that is mutated in adrenal hyperplasia is expressed widely in human and mouse tissues: a novel PDE8B isoform in human adrenal cortex. Eur J Hum Genet 2008;16:1245-53 Gamanuma M, Yuasa K, Sasaki T, et al. Comparison of enzymatic characterization and gene organization of cyclic nucleotide phosphodiesterase 8 family in humans. Cell Signal 2003;15:565-74

111.

Tsai LC, Shimizu-Albergine M, Beavo JA. The high-affinity cAMPspecific phosphodiesterase 8B controls steroidogenesis in the mouse adrenal gland. Mol Pharmacol 2011;79:639-48

112.

Tsai LC, Beavo JA. Regulation of adrenal steroidogenesis by the high-affinity phosphodiesterase 8 family. Horm Metab Res 2012;44:790-4

113.

114.

115.

116.

117.

.

Shimizu-Albergine M, Tsai LC, Patrucco E, et al. cAMP-specific phosphodiesterases 8A and 8B, essential regulators of Leydig cell steroidogenesis. Mol Pharmacol 2012;81:556-66 Perez-Torres S, Cortes R, Tolnay M, et al. Alterations on phosphodiesterase type 7 and 8 isozyme mRNA expression in Alzheimer’s disease brains examined by in situ hybridization. Exp Neurol 2003;182:322-34 Tsai LC, Chan GC, Nangle SN, et al. Inactivation of Pde8b enhances memory, motor performance, and protects against age-induced motor coordination decay. Genes Brain Behav 2012;11:837-47 Kelly MP, Adamowicz W, Bove S, et al. Select 3’,5’-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cell Signal 2014;26:383-97 Vang AG, Ben-Sasson SZ, Dong H, et al. PDE8 regulates rapid Teff cell adhesion and proliferation independent of ICER. PLoS One 2010;5:e12011 A paper that reports the first PDE8 selective inhibitor.

Expert Opin. Ther. Patents (2014) 24(12)

118. DeNinno MP, Wright SW, Visser MS, et al. 1,5-Substituted nipecotic amides: selective PDE8 inhibitors displaying diastereomer-dependent microsomal stability. Bioorg Med Chem Lett 2011;21:3095-8 119. Pfizer Inc. Substituted triazolopyrimidines as PDE8 inhibitors. WO2011058478A1; 2011 120. DeNinno MP, Wright SW, Etienne JB, et al. Discovery of triazolopyrimidinebased PDE8B inhibitors: exceptionally ligand-efficient and lipophilic ligandefficient compounds for the treatment of diabetes. Bioorg Med Chem Lett 2012;22:5721-6 121. The University of Connecticut. Methods using phosphodiesterase 8 inhibitors for the treatment of inflammation. US20090105281A1; 2009 122. Demirbas D, Wyman AR, Shimizu-Albergine M, et al. A yeastbased chemical screen identifies a PDE inhibitor that elevates steroidogenesis in mouse Leydig cells via PDE8 and PDE4 inhibition. PLoS One 2013;8:e71279 123. Maurice DH, Ke H, Ahmad F, et al. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 2014;13:290-314 .. Excellent review of the phosphodiesterase’s field. 124. CSIC. Compound that is a dual inhibitor of enzymes PDE7 and/or PDE4, pharmaceutical compositions and uses thereof. WO2008113881A1; 2008 125. Fortin M, D’Anjou H, Higgins ME, et al. A multi-target antisense approach against PDE4 and PDE7 reduces smokeinduced lung inflammation in mice. Respir Res 2009;10:39 126. Guimond A, Viau E, Aube P, et al. Advantageous toxicity profile of inhaled antisense oligonucleotides following chronic dosing in non-human primates. Pulm Pharmacol Ther 2008;21:845-54

Affiliation

Ana Martinez & Carmen Gil† Author for correspondence Centro de Investigaciones Biolo´gicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain E-mail: [email protected]

11

cAMP-specific phosphodiesterase inhibitors: promising drugs for inflammatory and neurological diseases.

PDEs are key enzymes in the adenosine and guanosine cyclic nucleotides (cAMP and cGMP) signaling cascade. Their inhibition increases cyclic nucleotide...
570KB Sizes 0 Downloads 5 Views