1
|
Moua T, Baqir M, Ryu JH. What Is on the Horizon for Treatments in Idiopathic Pulmonary Fibrosis? J Clin Med 2024; 13:6304. [PMID: 39518443 PMCID: PMC11546700 DOI: 10.3390/jcm13216304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and often fatal lung disease most commonly encountered in older individuals. Several decades of research have contributed to a better understanding of its pathogenesis, though only two drugs thus far have shown treatment efficacy, i.e., by slowing the decline of lung function. The pathogenesis of IPF remains incompletely understood and involves multiple complex interactions and mechanisms working in tandem or separately to result in unchecked deposition of extracellular matrix components and collagen characteristic of the disease. These mechanisms include aberrant response to injury in the alveolar epithelium, inappropriate communication between epithelial cells and mesenchymal cells, imbalances between oxidative injury and tissue repair, recruitment of inflammatory pathways that induce fibrosis, and cell senescence leading to sustained activation and proliferation of fibroblasts and myofibroblasts. Targeted approaches to each of these mechanistic pathways have led to recent clinical studies evaluating the safety and efficacy of several agents. This review highlights selected concepts in the pathogenesis of IPF as a rationale for understanding current or future therapeutic approaches, followed by a review of several selected agents and their recent or active clinical studies. Current novel therapies include approaches to attenuating or modifying specific cellular or signaling processes in the fibrotic pathway, modifying inflammatory and metabolic derangements, and minimizing inappropriate cell senescence.
Collapse
Affiliation(s)
- Teng Moua
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA; (M.B.); (J.H.R.)
| | | | | |
Collapse
|
2
|
Nguyen HO, Tiberio L, Facchinetti F, Ripari G, Violi V, Villetti G, Salvi V, Bosisio D. Modulation of Human Dendritic Cell Functions by Phosphodiesterase-4 Inhibitors: Potential Relevance for the Treatment of Respiratory Diseases. Pharmaceutics 2023; 15:2254. [PMID: 37765223 PMCID: PMC10535230 DOI: 10.3390/pharmaceutics15092254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Inhibitors of phosphodiesterase-4 (PDE4) are small-molecule drugs that, by increasing the intracellular levels of cAMP in immune cells, elicit a broad spectrum of anti-inflammatory effects. As such, PDE4 inhibitors are actively studied as therapeutic options in a variety of human diseases characterized by an underlying inflammatory pathogenesis. Dendritic cells (DCs) are checkpoints of the inflammatory and immune responses, being responsible for both activation and dampening depending on their activation status. This review shows evidence that PDE4 inhibitors modulate inflammatory DC activation by decreasing the secretion of inflammatory and Th1/Th17-polarizing cytokines, although preserving the expression of costimulatory molecules and the CD4+ T cell-activating potential. In addition, DCs activated in the presence of PDE4 inhibitors induce a preferential Th2 skewing of effector T cells, retain the secretion of Th2-attracting chemokines and increase the production of T cell regulatory mediators, such as IDO1, TSP-1, VEGF-A and Amphiregulin. Finally, PDE4 inhibitors selectively induce the expression of the surface molecule CD141/Thrombomodulin/BDCA-3. The result of such fine-tuning is immunomodulatory DCs that are distinct from those induced by classical anti-inflammatory drugs, such as corticosteroids. The possible implications for the treatment of respiratory disorders (such as COPD, asthma and COVID-19) by PDE4 inhibitors will be discussed.
Collapse
Affiliation(s)
- Hoang Oanh Nguyen
- ImmunoConcEpT, CNRS UMR 5164, University of Bordeaux, 33000 Bordeaux, France;
| | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Fabrizio Facchinetti
- Department of Experimental Pharmacology and Translational Science, Corporate Pre-Clinical R&D, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy; (F.F.); (G.V.)
| | - Giulia Ripari
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Valentina Violi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Gino Villetti
- Department of Experimental Pharmacology and Translational Science, Corporate Pre-Clinical R&D, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy; (F.F.); (G.V.)
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (L.T.); (G.R.); (V.V.)
| |
Collapse
|
3
|
Hiyama H, Arichika N, Okada M, Koyama N, Tahara T, Haruta J. Pharmacological Profile of Difamilast, a Novel Selective Phosphodiesterase 4 Inhibitor, for Topical Treatment of Atopic Dermatitis. J Pharmacol Exp Ther 2023; 386:45-55. [PMID: 37041087 DOI: 10.1124/jpet.123.001609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/10/2023] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
PDE4 inhibitors are expected to be anti-inflammatory agents based on their mechanism of action, but the application of this drug class is limited by a narrow therapeutic window due to adverse effects associated with gastrointestinal function. Difamilast, a novel selective phosphodiesterase 4 (PDE4) inhibitor, demonstrated significant efficacy without adverse reactions such as nausea and diarrhea in patients with atopic dermatitis (AD) and was recently approved in Japan. In this study, we investigated the pharmacological and pharmacokinetic properties of difamilast to provide nonclinical data to help understand the clinical effects. Difamilast selectively inhibited recombinant human PDE4 activity in assays. The IC50 of difamilast against PDE4B, a PDE4 subtype that plays an important role in the inflammatory response, was 0.0112 μM, representing a 6.6-fold decrease compared with the IC50 against PDE4D (0.0738 μM), a subtype that can trigger emesis. Difamilast inhibited TNF-α production in human (IC50 = 0.0109 μM) and mouse (IC50 = 0.0035 μM) peripheral blood mononuclear cells and improved skin inflammation in a mouse model of chronic allergic contact dermatitis. These effects of difamilast on TNF-α production and dermatitis were superior to those of other topical PDE4 inhibitors: CP-80633, cipamfylline, and crisaborole. In pharmacokinetic studies using miniature pigs and rats, the concentrations of difamilast in the blood and brain after topical application were not sufficient to support pharmacological activity. This nonclinical study contributes to explain the efficacy and safety of difamilast with a sufficient therapeutic window in the clinical trials. SIGNIFICANCE STATEMENT: This is the first report on the nonclinical pharmacological profile of difamilast ointment, a novel topical PDE4 inhibitor that demonstrated utility in clinical trials in patients with atopic dermatitis. Difamilast, which has high PDE4 selectivity (especially for the PDE4B subtype), ameliorated chronic allergic contact dermatitis in mice after topical application, with a pharmacokinetic profile in animals that suggests few systemic side effects; thus, difamilast is a promising new therapeutic treatment for atopic dermatitis.
Collapse
Affiliation(s)
- Hidetaka Hiyama
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| | - Naoya Arichika
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| | - Minoru Okada
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| | - Noriyuki Koyama
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| | - Tomonori Tahara
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| | - Junpei Haruta
- Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Hyogo, Japan (H.H., N.A., M.O., J.H.) and Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan (N.K., T.T.)
| |
Collapse
|
4
|
Yang X, Long F, Jia W, Zhang M, Su G, Liao M, Zeng Z, Chen W, Chen J. Artesunate inhibits PDE4 leading to intracellular cAMP accumulation, reduced ERK/MAPK signaling, and blockade of influenza A virus vRNP nuclear export. Antiviral Res 2023; 215:105635. [PMID: 37192683 DOI: 10.1016/j.antiviral.2023.105635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023]
Abstract
Influenza A viruses (IAV) have been a major cause of mortality. Given the potential for future deadly pandemics, effective drugs are needed for the treatment of severe influenzas, such as those caused by H5N1 IAV. The anti-malaria drugs artemisinin and its derivates, including artesunate (AS), have been reported to have broad antiviral activities. Here, we showed AS's antiviral activity against H5N1, H1N1, H3N2 and oseltamivir-resistant influenza A(H1N1)virus in vitro. Moreover, we showed that AS treatment significantly protected mice from lethal challenges with H1N1 and H5N1 IAV. Strikingly, the combination of AS and peramivir treatment significantly improved survival outcomes compared to their monotherapy with either AS or peramivir. Furthermore, we demonstrated mechanistically that AS affected the later stages of IAV replication and limited nuclear export of viral ribonucleoprotein (vRNP) complexes. In A549 cells, we demonstrated for the first time that AS treatment induced cAMP accumulation via inhibiting PDE4, and consequently reduced ERK phosphorylation and blocked IAV vRNP export, and thus suppressed IAV replication. These AS's effects were reversed by the pre-treatment with a cAMP inhibitor SQ22536. Our findings suggest that AS could serve as a novel IAV inhibitor by interfering vRNP nuclear export to prevent and treat IAV infection.
Collapse
Affiliation(s)
- Xia Yang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Feixiang Long
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Weixin Jia
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Mingxin Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Guanming Su
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zhenling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, Guangzhou, 510642, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
5
|
Spatiotemporal Changes in the Gene Expression Spectrum of the β2 Adrenergic Receptor Signaling Pathway in the Lungs of Rhesus Monkeys. Lung 2021; 199:73-82. [PMID: 33512584 PMCID: PMC7870609 DOI: 10.1007/s00408-021-00420-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/11/2021] [Indexed: 11/16/2022]
Abstract
Objective β2 adrenergic receptor (ADRB2) agonists mainly participate in regulation of airway function through the ADRB2-G protein-adenylyl cyclase (AC) signaling pathway; however, the key genes associated with this pathway and the spatiotemporal changes in the expression spectrum of some of their subtypes remain unclear, resulting in an insufficient theoretical basis for formulating the dose and method of drug administration for neonates. Methods We performed sampling at different developmental time points in rhesus monkeys, including the embryo stage, neonatal stage, and adolescence. The MiSeq platform was used for sequencing of key genes and some of their subtypes in the ADRB2 signaling pathway in lung tissues, and target gene expression was normalized and calculated according to reads per kilobase million. Results At different lung-developmental stages, we observed expression of phenylethanolamine N-methyltransferase (PNMT), ADRB2, AC, AKAP and EPAC subtypes (except AC8, AKAP4/5), and various phosphodiesterase (PDE) subtypes (PDE3, PDE4, PDE7, and PDE8), with persistently high expression of AC6, PDE4B, and AKAP(1/2/8/9/12/13, and EZR) maintained throughout the lung-developmental process, PNMT, ADRB2, AC(4/6), PDE4B, and AKAP(1/2/8/9/12/13, EZR, and MAP2)were highly expressed at the neonatal stage. Conclusion During normal lung development in rhesus monkeys, key genes associated with ADRB2–G protein–AC signaling and some of their subtypes are almost all expressed at the neonatal stage, suggesting that this signaling pathway plays a role in this developmental stage. Additionally, AC6, PDE4B, and AKAP(1/2/8/9/12/13, and EZR) showed persistently high expression during the entire lung-developmental process, which provides a reference for the development and utilization of key gene subtypes in this pathway.
Collapse
|
6
|
Morales-Garcia JA, Alonso-Gil S, Santos Á, Perez-Castillo A. Phosphodiesterase 7 Regulation in Cellular and Rodent Models of Parkinson's Disease. Mol Neurobiol 2019; 57:806-822. [PMID: 31473904 DOI: 10.1007/s12035-019-01745-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/19/2019] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is characterized by a loss of dopaminergic neurons in the ventral midbrain. This disease is diagnosed when around 50% of these neurons have already died; consequently, therapeutic treatments start too late. Therefore, an urgent need exists to find new targets involved in the onset and progression of the disease. Phosphodiesterase 7 (PDE7) is a key enzyme involved in the degradation of intracellular levels of cyclic adenosine 3', 5'-monophosphate in different cell types; however, little is known regarding its role in neurodegenerative diseases, and specifically in Parkinson's disease. We have previously shown that chemical as well as genetic inhibition of this enzyme results in neuroprotection and anti-inflammatory activity in different models of neurodegenerative disorders, including Parkinson's disease. Here, we have used in vitro and in vivo models of Parkinson's disease to study the regulation of PDE7 protein levels. Our results show that PDE7 is upregulated after an injury both in the human dopaminergic cell line SH-SY5Y and in primary rat mesencephalic cultures and after lipopolysaccharide or 6-hidroxydopamine injection in the Substantia nigra pars compacta of adult mice. PDE7 increase takes place mainly in degenerating dopaminergic neurons and in microglia cells. This enhanced expression appears to be direct since 6-hydroxydopamine and lipopolysaccharide increase the expression of a 962-bp fragment of its promoter. Taking together, these results reveal an essential function for PDE7 in the pathways leading to neurodegeneration and inflammatory-mediated brain damage and suggest novel roles for PDE7 in neurodegenerative diseases, specifically in PD, opening the door for new therapeutic interventions.
Collapse
Affiliation(s)
- Jose A Morales-Garcia
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier, 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo, 5, 28031, Madrid, Spain.
- Departamento de Biología Celular, Facultad de Medicina, UCM, Avda. Complutense s/n, 28040, Madrid, Spain.
| | - Sandra Alonso-Gil
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo, 5, 28031, Madrid, Spain
| | - Ángel Santos
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo, 5, 28031, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, UCM, Avda. Complutense s/n, 28040, Madrid, Spain
| | - Ana Perez-Castillo
- Instituto de Investigaciones Biomédicas (CSIC-UAM), Arturo Duperier, 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo, 5, 28031, Madrid, Spain.
| |
Collapse
|
7
|
Tran RQ, Jacoby SA, Roberts KE, Swann WA, Harris NW, Dinh LP, Denison EL, Yet L. Synthesis of 3-aryl-2-phosphinoimidazo[1,2- a]pyridine ligands for use in palladium-catalyzed cross-coupling reactions. RSC Adv 2019; 9:17778-17782. [PMID: 35520553 PMCID: PMC9064587 DOI: 10.1039/c9ra02200g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
3-Aryl-2-phosphinoimidazo[1,2-a]pyridine ligands were synthesized from 2-aminopyridine via two complementary routes. The first synthetic route involves the copper-catalyzed iodine-mediated cyclizations of 2-aminopyridine with arylacetylenes followed by palladium-catalyzed cross-coupling reactions with phosphines. The second synthetic route requires the preparation of 2,3-diiodoimidazo[1,2-a]pyridine or 2-iodo-3-bromoimidazo[1,2-a]pyridine from 2-aminopyridine followed by palladium-catalyzed Suzuki/phosphination or a phosphination/Suzuki cross-coupling reactions sequence, respectively. Preliminary model studies on the Suzuki synthesis of sterically-hindered biaryl and Buchwald-Hartwig amination compounds are presented with these ligands.
Collapse
Affiliation(s)
- Ryan Q Tran
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Seth A Jacoby
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Kaitlyn E Roberts
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - William A Swann
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Nekoda W Harris
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Long P Dinh
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Emily L Denison
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| | - Larry Yet
- University of South Alabama, Department of Chemistry Mobile AL 36618 USA
| |
Collapse
|
8
|
Sengupta S, Mehta G. Natural products as modulators of the cyclic-AMP pathway: evaluation and synthesis of lead compounds. Org Biomol Chem 2019; 16:6372-6390. [PMID: 30140804 DOI: 10.1039/c8ob01388h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is now well recognized that the normal cellular response in mammalian cells is critically regulated by the cyclic-AMP (cAMP) pathway through the appropriate balance of adenylyl cyclase (AC) and phosphodiesterase-4 (PDE4) activities. Dysfunctions in the cAMP pathway have major implications in various diseases like CNS disorders, inflammation and cardiac syndromes and, hence, the modulation of cAMP signalling through appropriate intervention of AC/PDE4 activities has emerged as a promising new drug discovery strategy of current interest. In this context, synthetic small molecules have had limited success so far and therefore parallel efforts on natural product leads have been actively pursued. The early promise of using the diterpene forskolin and its semi-synthetic analogs as AC activators has given way to new leads in the last decade from novel natural products like the marine sesterterpenoids alotaketals and ansellones and the 9,9'-diarylfluorenone cored selaginpulvilins, etc. and their synthesis has drawn much attention. This review captures these contemporary developments, particularly total synthesis campaigns and structure-guided analog design in the context of AC and PDE-4 modulating attributes and the scope for future possibilities.
Collapse
Affiliation(s)
- Saumitra Sengupta
- School of Chemistry, University of Hyderabad, Gachibowli, Hyderabad - 5000 046, Telengana, India.
| | | |
Collapse
|
9
|
Ntontsi P, Detta A, Bakakos P, Loukides S, Hillas G. Experimental and investigational phosphodiesterase inhibitors in development for asthma. Expert Opin Investig Drugs 2019; 28:261-266. [PMID: 30678501 DOI: 10.1080/13543784.2019.1571582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Severe, inadequately-controlled asthma remains a clinical challenge. For this reason, clinical trials and preclinical experimental studies on novel agents as an add-on therapies continue emerge. Phosphodiesterases (PDEs) are enzymes that regulate the function of immune cells by hydrolyzing cyclic guanosine monophosphate/cGMP and cyclic adenosine monophosphate/cAMP. PDEs are divided into subfamilies [PDE3, PDE4, PDE5 and PDE7] which are mainly found in the respiratory tract. Inhibitors of PDEs have already been approved for COPD and pulmonary hypertension. AREAS COVERED The role of PDE inhibitors in asthma treatment and the possible mechanism of action via their anti-inflammatory and/or bronchodilating effect are discussed. EXPERT OPINION Novel PDE inhibitors exhibiting fewer adverse events may have a role as add-on therapies in asthma treatment in the future. More clinical trials are necessary to prove their efficacy and evaluate their safety profile before approval by regulatory bodies is granted.
Collapse
Affiliation(s)
- Polyxeni Ntontsi
- a 2nd Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Attikon Hospital , Athens , Greece
| | - Aggeliki Detta
- b 1st Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Sotiria Chest Hospital , Athens , Greece
| | - Petros Bakakos
- b 1st Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Sotiria Chest Hospital , Athens , Greece
| | - Stelios Loukides
- a 2nd Respiratory Medicine Department , National and Kapodistrian University of Athens, Medical School, Attikon Hospital , Athens , Greece
| | - Georgios Hillas
- c 5th Pulmonary Department , "Sotiria" Chest Diseases Hospital , Athens , Greece
| |
Collapse
|
10
|
Huang J, Fu CX, Yang XY, Cui C, Yang S, Kuang Y, Guo CX, Hu P, Pei Q, Yang GP. Pharmacokinetics of single- and multiple-dose roflumilast: an open-label, three-way crossover study in healthy Chinese volunteers. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:4047-4057. [PMID: 30538429 PMCID: PMC6263297 DOI: 10.2147/dddt.s178862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Purpose To determine the pharmacokinetic properties of the common tablet of roflumilast administered in single and multiple oral doses in Chinese subjects. Subjects and methods Both the single- and multiple-dose studies included 12 adults (6 males and 6 females). In this single-center, open-label study, single doses of 0.25, 0.375, and 0.5 mg were administered using a randomized, three-way crossover design, and then, the 0.375 mg dose was continued for 11 days once daily. The pharmacokinetic parameters for roflumilast and roflumilast N-oxide were determined and the safety evaluation included adverse events assessed by monitoring, physical examination, vital sign tests, and clinical laboratory tests. Results After every single dose, the time to the maximum concentration (Cmax) of roflumilast (Tmax) was 0.25–2.0 hours; thereafter, the concentration declined, with a mean half-life (t1/2) of 19.7–20.9 hours over the range of 0.25–0.50 mg. As for roflumilast N-oxide, the mean t1/2 was 23.2–26.2 hours. The area under curve from the beginning to 24 hours (AUC0–24 h), the AUC until infinity (AUCinf), and the Cmax of roflumilast and roflumilast N-oxide increased in a dose-proportional manner. After multiple doses, the accumulation index (Rac) on the 11th day of the steady state was ~1.63 for roflumilast and 3.20 for roflumilast N-oxide. No significant sex differences were observed in the pharmacokinetic parameters of roflumilast and roflumilast N-oxide. In addition, there were no serious adverse events across the trial. Conclusion Roflumilast was safe and well-tolerated in healthy volunteers, and a linear increase in its Cmax and AUC values was observed at doses ranging from 0.25 to 0.50 mg.
Collapse
Affiliation(s)
- Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Cheng-Xiao Fu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Xiao-Yan Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Chan Cui
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Shuang Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Yun Kuang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| | - Pei Hu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Beijing 100032, People's Republic of China
| | - Qi Pei
- Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, , .,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Guo-Ping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China, .,Center for Clinical Drug Evaluation, Central South University, Changsha, Hunan 410013, People's Republic of China, ,
| |
Collapse
|
11
|
Namazi Sarvestani N, Saberi Firouzi S, Falak R, Karimi MY, Davoodzadeh Gholami M, Rangbar A, Hosseini A. Phosphodiesterase 4 and 7 inhibitors produce protective effects against high glucose-induced neurotoxicity in PC12 cells via modulation of the oxidative stress, apoptosis and inflammation pathways. Metab Brain Dis 2018; 33:1293-1306. [PMID: 29713919 DOI: 10.1007/s11011-018-0241-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/13/2018] [Indexed: 12/13/2022]
Abstract
Diabetic neuropathy (DN) is the most common diabetic complication. It is estimated diabetic population will increase to 592 million by the year 2035. This is while at least 50-60% of all diabetic patients will suffer from neuropathy in their lifetime. Oxidative stress, mitochondrial dysfunction, apoptosis, and inflammation are crucial pathways in development and progression of DN. Since there is also no selective and effective therapeutic agent to prevent or treat high glucose (HG)-induced neuronal cell injury, it is crucial to explore tools by which one can reduce factors related to these pathways. Phosphodiesterase 4 and 7 (PDE 4 and 7) regulate oxidative damage, neurodegenaration, and inflammatory responses through modulation of cyclic adenosine monophosphate (cAMP) level, and thus can be as important drug targets for regulating DN. The aim of this study was to evaluate the protective effects of inhibitors of PDE 4 and 7, named rolipram and BRL5048, on HG-induced neurotoxicity in PC12 cells as an in vitro cellular model for DN and determine the possible mechanisms for theirs effects. We report that the PC12 cells pre-treatment with rolipram (2 μM) and/or BRL5048 (0.2 μM) for 60 min and then exposing the cells to HG (4.5 g/L for 72 h) or normal glucose (NG) (1 g/L for 72 h) condition show: (1) significant attenuation in ROS, MDA and TNF-a levels, Bax/Bcl-2 ratio, expression of caspase 3 and UCP2 proteins; (2) significant increase in viability, GSH/GSSG ratio, MMP and ATP levels. All these data together led us to propose PDE 4 and 7 inhibitors, and specifically, rolipram and BRL5048, as potential drugs candidate to be further studied for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Nazanin Namazi Sarvestani
- Department of Animal Biology, School of Biology, Department of Science, University of Tehran, Tehran, Iran
| | - Saeedeh Saberi Firouzi
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Akram Rangbar
- Department of Toxicology and Pharmacology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Peng Y, Li Y, Tian Y, Ao G. PDE4a predicts poor prognosis and promotes metastasis by inducing epithelial-mesenchymal transition in hepatocellular carcinoma. J Cancer 2018; 9:2389-2396. [PMID: 30026835 PMCID: PMC6036708 DOI: 10.7150/jca.24079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Phosphodiesterases (PDEs) was found to be involved in a variety of cancer pathologies by modulating the degradation of levels of cAMP/cGMP. However, the prognostic significance and biological effect of PDE4a in hepatocellular carcinoma (HCC) have not been understood completely. In the present study, PDE4a expression was detected in a cohort of HCC and matched adjacent liver tissues (n = 210) by immunohistochemistry staining and Western immunoblotting assay, And in vitro experiments were conducted to determine the effect of PDE4a on metastatic capacity of HCC cells. The data here displayed that the majority of HCC patients had higher PDE4a expression in tumor tissues compared to matched adjacent liver tissues and enhanced PDE4a expression in tumor tissues was associated positively with HBV infection, liver cirrhosis, higher serum AFP level, advanced TNM stage, vascular embolus, intrahepatic metastases and portal vein tumor thrombus (PVTT). Survival analyses suggested that higher PDE4a was indicated the poor prognosis of HCCs after liver resection. Ectopic expression of PDE4a in Huh7 cells leaded to significant repression of E-cadherin and up-regulated the expression of N-cadherin and Vimentin, and facilitated migration and invasion abilities. Silencing PDE4a in MHCC97h cells acquired the opposite results. Taken together, PDE4a triggered EMT in HCC cells and acted as a predictive factor candidate and a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yanghong Peng
- Department of Radiology, the 309th Hospital of Chinese People's Liberation Army, Beijing 100091, P.R. China
| | - Yijun Li
- Department of Gastroenterology, Xi'an Central Hospital, Xian 710010, P.R. China
| | - Yu Tian
- Department of Gastroenterology, the 6th Affiliated Hospital of Xinjiang Medical University, Urumchi 830002, P.R. China
| | - Guokun Ao
- Department of Radiology, the 309th Hospital of Chinese People's Liberation Army, Beijing 100091, P.R. China
| |
Collapse
|
13
|
Chamseddine AN, Cabrero M, Wei Y, Ganan-Gomez I, Colla S, Takahashi K, Yang H, Bohannan ZS, Garcia-Manero G. PDE4 Differential Expression Is a Potential Prognostic Factor and Therapeutic Target in Patients With Myelodysplastic Syndrome and Chronic Myelomonocytic Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 16 Suppl:S67-73. [PMID: 27521329 DOI: 10.1016/j.clml.2016.02.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND (OR PURPOSE) Inflammation has an essential role in the pathogenesis of myelodysplastic syndromes (MDS). Its expression is controlled by phosphodiesterase 4 (PDE4). Thus, PDE4 inhibitors might be useful therapeutic targets for MDS. PATIENTS (OR MATERIALS) AND METHODS We evaluated the expression of each isoform of PDE4 (A, B, C, and D) using transcriptomic profiling and examined the potential impact on the outcome of patients with MDS in terms of survival and response to hypomethylating agents. Total RNA was extracted from CD34(+) bone marrow hematopoietic cells from healthy individuals (n = 10) and patients with MDS (n = 24) or chronic myelomonocytic leukemia (n = 19). RESULTS The study cohort had a median follow-up period of 21.2 months (range, 0.2-68 months) and a median overall survival of 17.6 months (95% confidence interval, 9.6-25.6). The main finding of the present study was that PDE4 mean expression was generally higher in patients with MDS than in healthy individuals. Also, upregulated PDE4 expression seemed to have a possible negative effect on survival (P > .05). Moreover, lower, compared with higher, mean PDE4A and PDE4C expression is indicative of a response to a hypomethylating agent (0.09 and 0.03 vs. 0.54 and 0.49, respectively; P > .05). CONCLUSION These results should be confirmed in a larger patient cohort. PDE4 expression could be an effective potential prognostic factor and therapeutic target for patients with MDS and chronic myelomonocytic leukemia. The role of PDE4 inhibitors should be explored in vitro against MDS cell lines and in preclinical mouse models of MDS.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Monica Cabrero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yue Wei
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Irene Ganan-Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hui Yang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zachary S Bohannan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
14
|
Fioni A, Selg E, Cenacchi V, Acevedo F, Brogin G, Gerde P, Puccini P. Investigation of Lung Pharmacokinetic of the Novel PDE4 Inhibitor CHF6001 in Preclinical Models: Evaluation of the PreciseInhale Technology. J Aerosol Med Pulm Drug Deliv 2017; 31:61-70. [PMID: 28768120 DOI: 10.1089/jamp.2017.1369] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Preclinical evaluation of new chemical entities (NCEs) designed to be administered by inhalation route requires lung administration to rodents, especially in the discovery phase. Different administration methods have been used until now, but more efforts are required to obtain controlled and reproducible lung deposition when only small amounts of neat powder material are available. METHODS The PreciseInhale platform used in the present study enables well-controlled powder aerosol exposures with only small amounts of micronized neat material, providing data on inhalation pharmacokinetic (PK) of NCEs at a very early stage. The DustGun aerosol technology uses compressed air to generate a respirable aerosol from milligram-amounts of powder that is delivered to one animal at a time. The new methodology was used to investigate the inhalation PK and lung retention in the rat of the novel Chiesi PDE4 inhibitor CHF6001 in three exposure models of the PreciseInhale platform: nose-only, intratracheally intubated rat, and the isolated, ventilated, and perfused rat lung. Results were compared with data from two other pulmonary delivery systems commonly used in preclinical studies: liquid instillation and powder insufflation. RESULTS Administration of micronized CHF6001 using the PreciseInhale system yielded lung exposures in the same range as the other tested devices, but the reproducibility in lung deposition was improved. The initial amount of CHF6001 in lungs at the first sampling time point was close to the predetermined target dose. Tracheal deposition with PreciseInhale (0.36 ± 0.22 μg) was significantly less than with other tested delivery systems: PennCentury (23.7 ± 3.2 μg) and Airjet (25.6 ± 7.2 μg). CONCLUSIONS The PreciseInhale platform enabled the administration of CHF6001 powder with good accuracy and reproducibility, with low tracheal deposition. The new platform can be used at an early discovery stage to obtain inhalatory PK data for respirable aerosols of neat NCE powder without excipients and with minimal use of dry powder formulation work.
Collapse
Affiliation(s)
| | - Ewa Selg
- 2 Inhalation Sciences Sweden AB , Stockholm, Sweden
| | | | | | | | - Per Gerde
- 2 Inhalation Sciences Sweden AB , Stockholm, Sweden .,3 Division of Physiology, Institute of Environmental Medicine, Karolinska Institutet , Stockholm, Sweden
| | | |
Collapse
|
15
|
Fontana E, Cenacchi V, Pivetti F, Pignatti A, Pazzi T, Bondanza L, Pazzi M. Synthesis of 14 C- and 2 H-labelled CHF6001: A new potent PDE4 inhibitor. J Labelled Comp Radiopharm 2017; 60:577-585. [PMID: 28763109 DOI: 10.1002/jlcr.3537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 11/11/2022]
Abstract
An 8-step preparation of 14 C-labelled CHF6001, a potent phosphodiesterase 4 inhibitor in the treatment of respiratory diseases, is described. An overall yield of approximately 9% was obtained starting from copper[14 C]cyanide. The synthesis of a stable labelled version of CHF6001 is also reported using the commercially available trideuterated bromomethylcyclopropane as starting material.
Collapse
Affiliation(s)
- E Fontana
- Isotope Chemistry, Accelera S.r.l., Nerviano (MI), Italy
| | - V Cenacchi
- Pharmacokinetics Biochemistry and Metabolism, Corporate Pre-Clinical R&D Chiesi Farmaceutici SpA, Parma, Italy
| | - F Pivetti
- Chemical Process Development, Chiesi Farmaceutici SpA, Parma, Italy
| | - A Pignatti
- Isotope Chemistry, Accelera S.r.l., Nerviano (MI), Italy
| | - T Pazzi
- Chimete S.r.l., Tortona (Al), Italy
| | | | - M Pazzi
- Chimete S.r.l., Tortona (Al), Italy
| |
Collapse
|
16
|
Zhou J, Iwasaki S, Yamakage M. Time- and Dose-Dependent Effects of Desflurane in Sensitized Airways. Anesth Analg 2017; 124:465-471. [PMID: 28067710 DOI: 10.1213/ane.0000000000001754] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Although the bronchodilatory actions of volatile anesthetics, such as halothane, isoflurane, and sevoflurane, have been well documented in previous studies, the properties of desflurane remain controversial. The aim of this study was to investigate the effects of desflurane at different concentrations and durations in an ovalbumin-sensitized guinea pig model of airway hyper-responsiveness. METHODS Ovalbumin-sensitized animals (n = 176) were randomly assigned to 5 groups according to the minimum alveolar concentration (MAC) of desflurane they received: 0.0, 0.5, 1.0, 1.5, and 2.0 MAC. Total lung resistance in vivo, airway smooth muscle tension in vitro, and intracellular cyclic adenosine monophosphate (AMP) levels were measured to evaluate the effects of desflurane. RESULTS In 5 sensitized groups, total lung resistance increased from baseline to peak at approximately 8 minutes and then decreased slowly until about 17 minutes with extended administration of desflurane. Desflurane dose-dependently increased total lung resistance with or without incremental doses of acetylcholine and reduced muscle tension with increasing concentrations of carbacholine. Cyclic AMP levels were increased by desflurane: at the 60-minute time point, cyclic AMP concentrations (means ± SD) with 0.5 MAC (1.96 ± 0.40) and 1.0 MAC (2.11 ± 0.50) desflurane were higher than those at the 8-minute time point (1.11 ± 0.23 and 1.32 ± 0.32). CONCLUSIONS Desflurane exerted time- and dose-dependent effects and could be used at 0.5 and 1.0 MAC concentrations without significant bronchoconstriction in ovalbumin-sensitized guinea pigs. Cyclic AMP-mediated airway smooth muscle relaxation might be one mechanism by which desflurane induces bronchodilation.
Collapse
Affiliation(s)
- Jing Zhou
- From the *Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China; and †Department of Anesthesiology, Sapporo Medical University, Sapporo, Japan
| | | | | |
Collapse
|
17
|
Martin N, Reid PT. The potential role of phosphodiesterase inhibitors in the management of asthma. ACTA ACUST UNITED AC 2016; 5:207-17. [PMID: 16696590 DOI: 10.2165/00151829-200605030-00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Asthma is a chronic inflammatory condition characterised by reversible airflow obstruction and airway hyperreactivity. The course of the illness may be punctuated by exacerbations resulting in deterioration in quality of life and, in some cases, days lost from school or work. That asthma is common and increasingly prevalent magnifies the importance of any potential economic costs, and promoting asthma control represents an important public health agenda. While lifestyle changes represent a valuable contribution in some patients, the majority of asthmatic patients require therapeutic intervention. The recognition of the role of inflammation in the pathogenesis of asthma has led to an emphasis on regular anti-inflammatory therapy, of which inhaled corticosteroid treatment remains the most superior. In selected patients, further improvements in asthma control may be gained by the addition of regular inhaled long-acting beta(2)-adrenoceptor agonists or oral leukotriene receptor antagonists to inhaled corticosteroid therapy. However, a significant minority of patients with asthma remain poorly controlled despite appropriate treatment, suggesting that additional corticosteroid nonresponsive inflammatory pathways may be operative. Furthermore, some patients with asthma display an accelerated decline in lung function, suggesting that active airway re-modeling is occurring. Such observations have focused attention on the potential to develop new therapies which complement existing treatments by targeting additional inflammatory pathways. The central role of phosphodiesterase (PDE), and in particular the PDE4 enzyme, in the regulation of key inflammatory cells believed to be important in asthma - including eosinophils, lymphocytes, neutrophils and airway smooth muscle - suggests that drugs designed to target this enzyme will have the potential to deliver both bronchodilation and modulate the asthmatic inflammatory response. In vivo studies on individual inflammatory cells suggest that the effects are likely to be favorable in asthma, and animal study models have provided proof of concept; however, first-generation PDE inhibitors have been poorly tolerated due to adverse effects. The development of second-generation agents such as cilomilast and roflumilast heralds a further opportunity to test the potential of these agents, although to date only a limited amount of data from human studies has been published, making it difficult to draw firm conclusions.
Collapse
Affiliation(s)
- Neil Martin
- Respiratory Medicine Unit, Western General Hospital, Edinburgh, Scotland
| | | |
Collapse
|
18
|
Tripuraneni NS, Azam MA. A combination of pharmacophore modeling, atom-based 3D-QSAR, molecular docking and molecular dynamics simulation studies on PDE4 enzyme inhibitors. J Biomol Struct Dyn 2016; 34:2481-92. [PMID: 26587754 DOI: 10.1080/07391102.2015.1119732] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Phosphodiesterases 4 enzyme is an attractive target for the design of anti-inflammatory and bronchodilator agents. In the present study, pharmacophore and atom-based 3D-QSAR studies were carried out for pyrazolopyridine and quinoline derivatives using Schrödinger suite 2014-3. A four-point pharmacophore model was developed using 74 molecules having pIC50 ranging from 10.1 to 4.5. The best four feature model consists of one hydrogen bond acceptor, two aromatic rings, and one hydrophobic group. The pharmacophore hypothesis yielded a statistically significant 3D-QSAR model, with a high correlation coefficient (R(2 )= .9949), cross validation coefficient (Q(2 )= .7291), and Pearson-r (.9107) at six component partial least square factor. The external validation indicated that our QSAR model possessed high predictive power with R(2) value of .88. The generated model was further validated by enrichment studies using the decoy test. Molecular docking, free energy calculation, and molecular dynamics (MD) simulation studies have been performed to explore the putative binding modes of these ligands. A 10-ns MD simulation confirmed the docking results of both stability of the 1XMU-ligand complex and the presumed active conformation. Outcomes of the present study provide insight in designing novel molecules with better PDE4 inhibitory activity.
Collapse
Affiliation(s)
- Naga Srinivas Tripuraneni
- a Department of Pharmaceutical Chemistry , J.S.S. College of Pharmacy (Constituent College of JSS University, Mysore) , Ooty, Udhagamandalam 643001 Tamil Nadu , India
| | - Mohammed Afzal Azam
- a Department of Pharmaceutical Chemistry , J.S.S. College of Pharmacy (Constituent College of JSS University, Mysore) , Ooty, Udhagamandalam 643001 Tamil Nadu , India
| |
Collapse
|
19
|
Tripuraneni NS, Azam MA. Pharmacophore modeling, 3D-QSAR and docking study of 2-phenylpyrimidine analogues as selective PDE4B inhibitors. J Theor Biol 2016; 394:117-126. [PMID: 26804643 DOI: 10.1016/j.jtbi.2016.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 12/30/2015] [Accepted: 01/04/2016] [Indexed: 12/19/2022]
Abstract
Pharmacophore modeling, molecular docking, and molecular dynamics (MD) simulation studies have been performed, to explore the putative binding modes of 2-phenylpyrimidine series as PDE4B selective inhibitors. A five point pharmacophore model was developed using 87 molecules having pIC50 ranging from 8.52 to 5.07. The pharmacophore hypothesis yielded a statistically significant 3D-QSAR model, with a high correlation coefficient (R(2)=0.918), cross validation coefficient (Q(2)=0.852), and F value (175) at 4 component PLS factor. The external validation indicated that our QSAR model possessed high predictive power (R(2)=0.70). The generated model was further validated by enrichment studies using the decoy test. To evaluate the effectiveness of docking protocol in flexible docking, we have selected crystallographic bound compound to validate our docking procedure as evident from root mean square deviation. A 10ns molecular dynamics simulation confirmed the docking results of both stability of the 1XMU-ligand complex and the presumed active conformation. Further, similar orientation was observed between the superposition of the conformations of 85 after MD simulation and best XP-docking pose; MD simulation and 3D-QSAR pose; best XP-docking and 3D-QSAR poses. Outcomes of the present study provide insight in designing novel molecules with better PDE4B selective inhibitory activity.
Collapse
Affiliation(s)
- Naga Srinivas Tripuraneni
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty, Udhagamandalam 643001, Tamil Nadu, India; Constituent College of JSS University, Mysore, India
| | - Mohammed Afzal Azam
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ooty, Udhagamandalam 643001, Tamil Nadu, India; Constituent College of JSS University, Mysore, India.
| |
Collapse
|
20
|
Li J, Zhou N, Liu W, Li J, Feng Y, Wang X, Wu C, Bao J. Discover natural compounds as potential phosphodiesterase-4B inhibitors via computational approaches. J Biomol Struct Dyn 2016; 34:1101-12. [PMID: 26159554 DOI: 10.1080/07391102.2015.1070749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
cAMP, intracellular cyclic adenosine monophosphate, is a ubiquitous second messenger that plays a key role in many physiological processes. PDE4B which can reduce the cAMP level by hydrolyzing cAMP to 5'-AMP has become a therapeutic target for the treatment of human diseases such as respiratory disorders, inflammation diseases, neurological and psychiatric disorders. However, the use of currently available PDE4B inhibitors is restricted due to serious side effects caused by targeting PDE4D. Hence, we are attempting to find out subfamily-selective PDE4B inhibitors from natural products, using computer-aided approaches such as virtual screening, docking, and molecular dynamics simulation. Finally, four potential PDE4B-selective inhibitors (ZINC67912770, ZINC67912780, ZINC72320169, and ZINC28882432) were found. Compared to the reference drug (roflumilast), they scored better during the virtual screening process. Binding free energy for them was -317.51, -239.44, -215.52, and -165.77 kJ/mol, better than -129.05 kJ/mol of roflumilast. The pharmacophore model of the four candidate inhibitors comprised six features, including one hydrogen bond donor, four hydrogen bond acceptors, and one aromatic ring feature. It is expected that our study will pave the way for the design of potent PDE4B-selective inhibitors of new drugs to treat a wide variety of diseases such as asthma, COPD, psoriasis, depression, etc.
Collapse
Affiliation(s)
- Jing Li
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Nan Zhou
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Wen Liu
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Jianzong Li
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Yu Feng
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Xiaoyun Wang
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Chuanfang Wu
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China
| | - Jinku Bao
- a College of Life Sciences & Key Laboratory for Bio-resources of Ministry of Education , Sichuan University , Chengdu 610064 , China.,b State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital , Sichuan University , Chengdu 610041 , China.,c State Key Laboratory of Oral Diseases , West China College of Stomatology, Sichuan University , Chengdu 610041 , China
| |
Collapse
|
21
|
Subbian S, Tsenova L, Holloway J, Peixoto B, O'Brien P, Dartois V, Khetani V, Zeldis JB, Kaplan G. Adjunctive Phosphodiesterase-4 Inhibitor Therapy Improves Antibiotic Response to Pulmonary Tuberculosis in a Rabbit Model. EBioMedicine 2016; 4:104-14. [PMID: 26981575 PMCID: PMC4776074 DOI: 10.1016/j.ebiom.2016.01.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/06/2016] [Accepted: 01/11/2016] [Indexed: 12/14/2022] Open
Abstract
Objectives Adjunctive host-directed therapy is emerging as a new potential approach to improve the outcome of conventional antimicrobial treatment for tuberculosis (TB). We tested the ability of a phosphodiesterase-4 inhibitor (PDE4i) CC-11050, co-administered with the first-line anti-TB drug isoniazid (INH), to accelerate bacillary killing and reduce chronic inflammation in the lungs of rabbits with experimental Mycobacterium tuberculosis (Mtb) infection. Methods A rabbit model of pulmonary TB that recapitulates the pathologic manifestations seen in humans was used. Rabbits were infected with virulent Mtb by aerosol exposure and treated for eight weeks with INH with or without CC-11050, starting at four weeks post infection. The effect of CC-11050 treatment on disease severity, pathology, bacillary load, T cell proliferation and global lung transcriptome profiles were analyzed. Results Significant improvement in bacillary clearance and reduced lung pathology and fibrosis were noted in the rabbits treated for eight weeks with INH + CC-11050, compared to those treated with INH or CC-11050 only. In addition, expression of host genes associated with tissue remodeling, tumor necrosis factor alpha (TNF-α) regulation, macrophage activation and lung inflammation networks was dampened in CC-11050-treated, compared to the untreated rabbits. Conclusions Adjunctive CC-11050 therapy significantly improves the response of rabbits with experimental pulmonary TB to INH treatment. We propose that CC-11050 may be a promising candidate for host directed therapy of patients with pulmonary TB, reducing the duration and improving clinical outcome of antibiotic treatment. CC-11050 is an anti-inflammatory molecule targeting host phosphodiesterase-4. CC-11050 plus isoniazid therapy significantly reduced bacillary load and pathology in a rabbit model pulmonary tuberculosis. CC-11050 can be a promising candidate for adjunctive host directed therapy of patients with active pulmonary tuberculosis.
In 2013, tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) killed 1.5 million people worldwide. Current antibiotic therapy for tuberculosis is ineffective in eliminating the infecting bacilli and/or disease pathology such as lung fibrosis. Therefore, alternate approaches are urgently needed to control the TB epidemic. In this study, using a rabbit model of pulmonary TB, which closely mimics the human disease, we tested the hypothesis that reducing the host inflammatory response during Mtb infection would improve the outcome of antibiotic treatment; we show that adjunctive phosphodiesterase-4 inhibition therapy with isoniazid improves bacterial clearance and lung pathology.
Collapse
Affiliation(s)
- Selvakumar Subbian
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Liana Tsenova
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA; Department of Biological Sciences, NYC College of Technology, Brooklyn, NY, USA
| | - Jennifer Holloway
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Blas Peixoto
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Paul O'Brien
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Véronique Dartois
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | | | | | - Gilla Kaplan
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| |
Collapse
|
22
|
Pharmacophore modeling, 3D-QSAR, and docking study of pyrozolo[1,5-a]pyridine/4,4-dimethylpyrazolone analogues as PDE4 selective inhibitors. J Mol Model 2015; 21:289. [DOI: 10.1007/s00894-015-2837-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/09/2015] [Indexed: 01/26/2023]
|
23
|
Mulhall AM, Droege CA, Ernst NE, Panos RJ, Zafar MA. Phosphodiesterase 4 inhibitors for the treatment of chronic obstructive pulmonary disease: a review of current and developing drugs. Expert Opin Investig Drugs 2015; 24:1597-611. [PMID: 26419847 DOI: 10.1517/13543784.2015.1094054] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Phosphodiesterase (PDE) inhibitors modulate lung inflammation and cause bronchodilation by increasing intracellular cyclic adenosine 3', 5'-monophosphate in airway smooth muscle and inflammatory cells. Roflumilast is the only approved PDE-4 inhibitor (PDE4I) for use in chronic obstructive pulmonary disease (COPD). Its beneficial clinical effects occur preferentially in patients with chronic bronchitis and frequent COPD exacerbations. Use of roflumilast as adjunctive or alternate therapy to other COPD medications reduces exacerbations and modestly improves lung function. AREAS COVERED This article reviews the current role of PDE4I in COPD treatment emphasizing roflumilast's clinical efficacy and adverse effects. This article also reviews developing PDE4Is in early clinical trials and in preclinical studies. EXPERT OPINION After decades of research in drug development, PDE4Is are a welcomed addition to the COPD therapeutic armamentarium. In its current clinical role, the salubrious clinical effects of PDE4I in reducing exacerbations and stabilizing the frequent exacerbator phenotype have to be cautiously balanced with numerous adverse effects. Developing drugs may provide similar or better clinical benefits while minimizing adverse effects by changing the mode of drug delivery to inhaled formulations, combining dual PDE isoenzyme inhibitors (PDE1/4I and PDE3/4I) and by forming hybrid molecules with other bronchodilators (muscarinic receptor antagonist/PDE4I and β2-agonist/PDE4I).
Collapse
Affiliation(s)
- Aaron M Mulhall
- a 1 University of Cincinnati Medical Center, Division of Pulmonary and Critical Care Medicine , Cincinnati, USA .,b 2 Division of Pulmonary and Critical Care Medicine, Veterans Affairs Medical Center , Cincinnati, USA
| | - Christopher A Droege
- c 3 University of Cincinnati Medical Center, Department of Pharmacy Services , Cincinnati, USA
| | - Neil E Ernst
- c 3 University of Cincinnati Medical Center, Department of Pharmacy Services , Cincinnati, USA
| | - Ralph J Panos
- a 1 University of Cincinnati Medical Center, Division of Pulmonary and Critical Care Medicine , Cincinnati, USA .,b 2 Division of Pulmonary and Critical Care Medicine, Veterans Affairs Medical Center , Cincinnati, USA
| | - Muhammad A Zafar
- a 1 University of Cincinnati Medical Center, Division of Pulmonary and Critical Care Medicine , Cincinnati, USA .,b 2 Division of Pulmonary and Critical Care Medicine, Veterans Affairs Medical Center , Cincinnati, USA
| |
Collapse
|
24
|
Abstract
Atrophy occurs in specific muscles with inactivity (for example, during plaster cast immobilization) or denervation (for example, in patients with spinal cord injuries). Muscle wasting occurs systemically in older people (a condition known as sarcopenia); as a physiological response to fasting or malnutrition; and in many diseases, including chronic obstructive pulmonary disorder, cancer-associated cachexia, diabetes, renal failure, cardiac failure, Cushing syndrome, sepsis, burns and trauma. The rapid loss of muscle mass and strength primarily results from excessive protein breakdown, which is often accompanied by reduced protein synthesis. This loss of muscle function can lead to reduced quality of life, increased morbidity and mortality. Exercise is the only accepted approach to prevent or slow atrophy. However, several promising therapeutic agents are in development, and major advances in our understanding of the cellular mechanisms that regulate the protein balance in muscle include the identification of several cytokines, particularly myostatin, and a common transcriptional programme that promotes muscle wasting. Here, we discuss these new insights and the rationally designed therapies that are emerging to combat muscle wasting.
Collapse
|
25
|
Cenacchi V, Battaglia R, Cinato F, Riccardi B, Spinabelli D, Brogin G, Puccini P, Pezzetta D. In vitro and in vivo metabolism of CHF 6001, a selective phosphodiesterase (PDE4) inhibitor. Xenobiotica 2015; 45:693-710. [PMID: 25733029 DOI: 10.3109/00498254.2015.1014945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. The metabolism of CHF 6001, a novel PDE4 inhibitor, was determined in vitro in mouse, rat, dog, monkey and human microsomes and hepatocytes and in vivo in plasma, urine, feces and bile of rats after intravenous and intratracheal administration. 2. The behavior of CHF 6001 in microsomes and hepatocytes changed across species. CYP3A4/5 isoenzymes were identified to be the primary enzymes responsible for the metabolism of CHF 6001 in human liver microsomes. 3. In the rat, CHF 6001 was found extensively metabolized in urine, feces and bile, but not in plasma, where CHF 6001 was the main compound present. The metabolite profiles were different in the four biological matrices from both qualitative and quantitative point of view. 4. CHF 6001 was metabolized through hydrolysis with the formation of the alcohol CHF 5956, loss of a chlorine atom, loss of the N-oxide, hydroxylation, loss of the cyclopropylmethyl group in the alcohol moiety, conjugation with glucuronic acid, glutathione and cysteine-glycine. 5. The major metabolite present in the bile was isolated and characterized by nuclear magnetic resonance analysis. It derived from CHF 6001 through contraction of the pyridine-N-oxide ring to N-hydroxy pyrrole and conjugation with glucuronic acid.
Collapse
|
26
|
Moretto N, Caruso P, Bosco R, Marchini G, Pastore F, Armani E, Amari G, Rizzi A, Ghidini E, De Fanti R, Capaldi C, Carzaniga L, Hirsch E, Buccellati C, Sala A, Carnini C, Patacchini R, Delcanale M, Civelli M, Villetti G, Facchinetti F. CHF6001 I: a novel highly potent and selective phosphodiesterase 4 inhibitor with robust anti-inflammatory activity and suitable for topical pulmonary administration. J Pharmacol Exp Ther 2015; 352:559-67. [PMID: 25576075 DOI: 10.1124/jpet.114.220541] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This study examined the pharmacologic characterization of CHF6001 [(S)-3,5-dichloro-4-(2-(3-(cyclopropylmethoxy)-4-(difluoromethoxy)phenyl)-2-(3-(cyclopropylmethoxy)-4-(methylsulfonamido)benzoyloxy)ethyl)pyridine 1-oxide], a novel phosphodiesterase (PDE)4 inhibitor designed for treating pulmonary inflammatory diseases via inhaled administration. CHF6001 was 7- and 923-fold more potent than roflumilast and cilomilast, respectively, in inhibiting PDE4 enzymatic activity (IC50 = 0.026 ± 0.006 nM). CHF6001 inhibited PDE4 isoforms A-D with equal potency, showed an elevated ratio of high-affinity rolipram binding site versus low-affinity rolipram binding site (i.e., >40) and displayed >20,000-fold selectivity versus PDE4 compared with a panel of PDEs. CHF6001 effectively inhibited (subnanomolar IC50 values) the release of tumor necrosis factor-α from human peripheral blood mononuclear cells, human acute monocytic leukemia cell line macrophages (THP-1), and rodent macrophages (RAW264.7 and NR8383). Moreover, CHF6001 potently inhibited the activation of oxidative burst in neutrophils and eosinophils, neutrophil chemotaxis, and the release of interferon-γ from CD4(+) T cells. In all these functional assays, CHF6001 was more potent than previously described PDE4 inhibitors, including roflumilast, UK-500,001 [2-(3,4-difluorophenoxy)-5-fluoro-N-((1S,4S)-4-(2-hydroxy-5-methylbenzamido)cyclohexyl)nicotinamide], and cilomilast, and it was comparable to GSK256066 [6-((3-(dimethylcarbamoyl)phenyl)sulfonyl)-4-((3-methoxyphenyl)amino)-8-methylquinoline-3-carboxamide]. When administered intratracheally to rats as a micronized dry powder, CHF6001 inhibited liposaccharide-induced pulmonary neutrophilia (ED50 = 0.205 μmol/kg) and leukocyte infiltration (ED50 = 0.188 μmol/kg) with an efficacy comparable to a high dose of budesonide (1 μmol/kg i.p.). In sum, CHF6001 has the potential to be an effective topical treatment of conditions associated with pulmonary inflammation, including asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Nadia Moretto
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Paola Caruso
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Raffaella Bosco
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Gessica Marchini
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Fiorella Pastore
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Elisabetta Armani
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Gabriele Amari
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Andrea Rizzi
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Eleonora Ghidini
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Renato De Fanti
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Carmelida Capaldi
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Laura Carzaniga
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Emilio Hirsch
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Carola Buccellati
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Angelo Sala
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Chiara Carnini
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Riccardo Patacchini
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Maurizio Delcanale
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Maurizio Civelli
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Gino Villetti
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| | - Fabrizio Facchinetti
- Corporate Pre-Clinical R & D, Chiesi Farmaceutici S.p.A., Parma, Italy (N.M., P.C., R.B., G.M., F.P., E.A., G.A., A.R., E.G., R.D.F., Ca.C., L.C., Ch.C., R.P. M.D., M.C., G.V., F.F.); Molecular Biotechnology Center, University of Turin, Turin, Italy (E.H.); and Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy (C.B., A.S.)
| |
Collapse
|
27
|
Villetti G, Carnini C, Battipaglia L, Preynat L, Bolzoni PT, Bassani F, Caruso P, Bergamaschi M, Pisano AR, Puviani V, Stellari FF, Cenacchi V, Volta R, Bertacche V, Mileo V, Bagnacani V, Moretti E, Puccini P, Catinella S, Facchinetti F, Sala A, Civelli M. CHF6001 II: a novel phosphodiesterase 4 inhibitor, suitable for topical pulmonary administration--in vivo preclinical pharmacology profile defines a potent anti-inflammatory compound with a wide therapeutic window. J Pharmacol Exp Ther 2015; 352:568-78. [PMID: 25576073 DOI: 10.1124/jpet.114.220558] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
CHF6001 [(S)-3,5-dichloro-4-(2-(3-(cyclopropylmethoxy)-4-(difluoromethoxy)phenyl)-2-(3-(cyclopropylmethoxy)-4-(methylsulfonamido)benzoyloxy)ethyl)pyridine 1-oxide] is a novel phosphodiesterase 4 (PDE4) inhibitor designed for use in pulmonary diseases by inhaled administration. Intratracheal administration of CHF6001 to ovalbumin-sensitized Brown-Norway rats suppressed the antigen-induced decline of lung functions (ED50 = 0.1 µmol/kg) and antigen-induced eosinophilia (ED50 = 0.03 µmol/kg) when administered (0.09 μmol/kg) up to 24 hours before antigen challenge, in agreement with CHF6001-sustained lung concentrations up to 72 hours after intratracheal treatment (mean residence time 26 hours). Intranasal, once daily administration of CHF6001 inhibited neutrophil infiltration observed after 11 days of tobacco smoke exposure in mice, both upon prophylactic (0.15-0.45 µmol/kg per day) or interventional (0.045-0.45 µmol/kg per day) treatment. CHF6001 was ineffective in reversing ketamine/xylazine-induced anesthesia (a surrogate of emesis in rat) up to 5 µmol/kg administered intratracheally, a dose 50- to 150-fold higher than anti-inflammatory ED50 observed in rats. When given topically to ferrets, no emesis and nausea were evident up to 10 to 20 µmol/kg, respectively, whereas the PDE4 inhibitor GSK-256066 (6-[3-(dimethylcarbamoyl)phenyl]sulfonyl-4-(3-methoxyanilino)-8-methylquinoline-3-carboxamide) induced nausea at 1 µmol/kg intratracheally. A 14-day inhalation toxicology study in rats showed a no-observed-adverse-effect level dose of 4.4 µmol/kg per day for CHF6001, lower than the 0.015 μmol/kg per day for GSK-256066. CHF6001 was found effective and extremely well tolerated upon topical administration in relevant animal models, and may represent a step forward in PDE4 inhibition for the treatment of asthma and chronic obstructive respiratory disease.
Collapse
Affiliation(s)
- Gino Villetti
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Chiara Carnini
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Loredana Battipaglia
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Laurent Preynat
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Pier Tonino Bolzoni
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Franco Bassani
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Paola Caruso
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Marco Bergamaschi
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Anna Rita Pisano
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Veronica Puviani
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Fabio Franco Stellari
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Valentina Cenacchi
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Roberta Volta
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Vittorio Bertacche
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Valentina Mileo
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Valentina Bagnacani
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Elisa Moretti
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Paola Puccini
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Silvia Catinella
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Fabrizio Facchinetti
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Angelo Sala
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| | - Maurizio Civelli
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy (G.V., C.C., L.B., L.P., P.T.B., F.B., P.C., M.B., A.R.P., V.P., F.F.S., V.C., R.V., Vi.B., V.M., Va.B., E.M., P.P, S.C., F.F., M.C.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (A.S.); and Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy (A.S.)
| |
Collapse
|
28
|
Lomas O, Zaccolo M. Phosphodiesterases maintain signaling fidelity via compartmentalization of cyclic nucleotides. Physiology (Bethesda) 2014; 29:141-9. [PMID: 24583770 PMCID: PMC3949206 DOI: 10.1152/physiol.00040.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Novel technological advances have improved our understanding of how cyclic nucleotides are able to convey signals faithfully between cellular compartments. Phosphodiesterases play a crucial role in shaping these signals in health and disease. The concept of compartmentalization is guiding the search for therapies that have the potential to offer greater efficacy and tolerability compared with current treatments.
Collapse
Affiliation(s)
- Oliver Lomas
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom; and
| | | |
Collapse
|
29
|
Phosphodiesterase Inhibitors for Chronic Obstructive Pulmonary Disease: What Does the Future Hold? Drugs 2014; 74:1983-92. [DOI: 10.1007/s40265-014-0303-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Phosphodiesterase 4 inhibitor roflumilast improves the bronchodilative effect of sevoflurane in sensitized airways. Anesthesiology 2014; 120:1152-9. [PMID: 24503814 DOI: 10.1097/aln.0000000000000160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Although phosphodiesterase 4 inhibitors and the volatile anesthetic sevoflurane are known to have independent bronchodilator properties, the combined administration of these two agents may have the potential to exert an additive or synergistic bronchodilator effect. The authors tested this hypothesis and investigated the common site of this combined relaxation effect in a model of airway hyperresponsiveness with ovalbumin-sensitized guinea pigs. METHODS Ovalbumin-sensitized animals (n = 138) were randomized into six groups: sensitized, sevoflurane, rolipram1.0, roflumilast1.0, sevoflurane/rolipram1.0, and sevoflurane/roflumilast1.0. Total lung resistance in vivo, airway smooth muscle tension in vitro, and intracellular cyclic adenosine monophosphate levels were measured to evaluate the relaxation effect. RESULTS Among the six sensitized groups, total lung resistance was higher in the order of sensitized > sevoflurane > rolipram 1.0 > roflumilast1.0 > sevoflurane/rolipram1.0 > sevoflurane/roflumilast1.0, with an increase in acetylcholine concentration. Compared with the other five groups, the muscle tensions in the sevoflurane/roflumilast1.0 group were significantly lower at carbacholine doses of 10, 10, and 10 M; the cyclic adenosine monophosphate concentrations (means ± SD) in the sevoflurane/rolipram1.0 (1.61 ± 0.34) and sevoflurane/roflumilast1.0 (1.50 ± 0.20) groups were higher than that in the sensitized (0.52 ± 0.15) and sevoflurane (1.12 ± 0.32) groups. CONCLUSIONS The combined use of phosphodiesterase 4 inhibitors with the volatile anesthetic sevoflurane had an additive bronchodilator effect in ovalbumin-sensitized guinea pigs. The concurrent increase in cyclic adenosine monophosphate levels in sensitized airway smooth muscle might be a mechanism of this combined relaxation effect.
Collapse
|
31
|
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. [PMID: 24865624 DOI: 10.1517/13543784.2014.921157] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION PDE inhibitors could be useful in the treatment of asthma because of their bronchodilator and/or anti-inflammatory activities. Recently, some selective PDE3, PDE4 and PDE3/4 inhibitors have been shown to have beneficial effects in patients with asthma suggesting that such drugs may offer novel therapeutic options for the treatment of this disease. AREAS COVERED The authors describe the main PDE families that could be involved in asthma as well as the PDE inhibitors that have been evaluated for the treatment of asthma. EXPERT OPINION Although the potential therapeutic utility of PDE inhibitors has been demonstrated in various animal models of asthma, their clinical efficacy have been restricted by the dose-limiting side effects; no PDE inhibitor has yet been approved for the treatment of patients with asthma. Although new PDE inhibitors have been synthesised, most data are from cellular and tissue-level studies with human trials still on the horizon. Apparently, only CHF 6001, an inhaled PDE4 inhibitor, and RPL554, a dual PDE3/4 inhibitor, are still under clinical development. Further data from these new drugs are eagerly anticipated to better understand where these drugs might stand in the future treatment of asthma.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Second University of Naples, Department of Experimental Medicine, Unit of Pharmacology , Naples , Italy
| | | | | |
Collapse
|
32
|
The role of macrophages in obstructive airways disease: chronic obstructive pulmonary disease and asthma. Cytokine 2013; 64:613-25. [PMID: 24084332 DOI: 10.1016/j.cyto.2013.09.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/04/2013] [Accepted: 09/06/2013] [Indexed: 12/15/2022]
Abstract
Macrophages are a major cellular component of the innate immune system, and play an important role in the recognition of microbes, particulates, and immunogens and to the regulation of inflammatory responses. In the lung, macrophages react with soluble proteins that bind microbial products in order to remove pathogens and particles and to maintain the sterility of the airway tract. Chronic obstructive pulmonary disease and asthma are both obstructive airway diseases that involve chronic inflammation of the respiratory tract which contributes to disease progression. In the case of COPD, there is increasing evidence that lung macrophages orchestrate inflammation through the release of chemokines that attract neutrophils, monocytes and T cells and the release of several proteases. On the other hand, in asthma, it seems that alveolar macrophages are inappropriately activated and are implicated in the development and progression of the disease. In this review we summarize the current basic and clinical research studies which highlight the role of macrophages in asthma and COPD.
Collapse
|
33
|
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. [DOI: 10.1517/17460441.2013.787986] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Ochiai K, Takita S, Kojima A, Eiraku T, Iwase K, Kishi T, Ohinata A, Yageta Y, Yasue T, Adams DR, Kohno Y. Phosphodiesterase inhibitors. Part 5: Hybrid PDE3/4 inhibitors as dual bronchorelaxant/anti-inflammatory agents for inhaled administration. Bioorg Med Chem Lett 2013. [DOI: 10.1016/j.bmcl.2012.08.121] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Kobayashi M, Kubo S, Shiraki K, Iwata M, Hirano Y, Ohtsu Y, Takahashi K, Shimizu Y. Therapeutic potential of ASP3258, a selective phosphodiesterase 4 inhibitor, on chronic eosinophilic airway inflammation. Pharmacology 2012; 90:223-32. [PMID: 23038661 DOI: 10.1159/000342380] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/31/2012] [Indexed: 11/19/2022]
Abstract
We investigated and compared the pharmacological effects of a PDE4 inhibitor ASP3258 (3-[4-(3-chlorophenyl)-1-ethyl-7-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl] propanoic acid), with those of roflumilast, the most clinically advanced PDE4 inhibitor known. ASP3258 inhibited human PDE4A, 4B, 4C, and 4D with respective IC(50) values of 0.036, 0.050, 0.45, and 0.035 nmol/l, all approximately 3-6 times more potent than roflumilast. ASP3258 inhibited LPS-induced TNF-α production and PHA-induced IL-5 production in human whole blood cells with respective IC(50) values of 110 and 100 nmol/l, both approximately 10 times less potent than roflumilast. Repeatedly administered ASP3258 and roflumilast both suppressed chronic airway eosinophilia induced by repeated exposure to ovalbumin in Brown Norway rats with respective ED(50) values of 0.092 and 0.17 mg/kg. We also evaluated the toxicological profiles of ASP3258. Although PDE4 inhibitors induce emesis by mimicking the pharmacological action of an α(2)-adrenoceptor antagonist, repeated administration of ASP3258 (3 mg/kg) had no such inhibitory effect on rats anesthetized with α(2) - adrenoceptor agonist. PDE4 inhibitors are also known to induce vascular injury in rats. Although repeatedly administered ASP3258 (3 and 10 mg/kg) significantly increased plasma fibrinogen, a biomarker for toxicity, 1 mg/kg of ASP3258 did not. These results suggest that ASP3258 is an attractive PDE4 inhibitor for treating chronic eosinophilic airway inflammation due to asthma.
Collapse
Affiliation(s)
- Miki Kobayashi
- Drug Discovery Research, Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Lee J, Komatsu K, Lee BC, Lim JH, Jono H, Xu H, Kai H, Zhang ZJ, Yan C, Li JD. Phosphodiesterase 4B mediates extracellular signal-regulated kinase-dependent up-regulation of mucin MUC5AC protein by Streptococcus pneumoniae by inhibiting cAMP-protein kinase A-dependent MKP-1 phosphatase pathway. J Biol Chem 2012; 287:22799-811. [PMID: 22610099 DOI: 10.1074/jbc.m111.337378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Otitis media (OM) is the most common childhood bacterial infection and the major cause of conductive hearing loss in children. Mucus overproduction is a hallmark of OM. Streptococcus pneumoniae is the most common gram-positive bacterial pathogen causing OM. Among many mucin genes, MUC5AC has been found to be greatly up-regulated in the middle ear mucosa of human patients with OM. We previously reported that S. pneumoniae up-regulates MUC5AC expression in a MAPK ERK-dependent manner. We also found that MAPK phosphatase-1 (MKP-1) negatively regulates S. pneumoniae-induced ERK-dependent MUC5AC up-regulation. Therapeutic strategies for up-regulating the expression of negative regulators such as MKP-1 may have significant therapeutic potential for treating mucus overproduction in OM. However, the underlying molecular mechanism by which MKP-1 expression is negatively regulated during S. pneumoniae infection is unknown. In this study we show that phosphodiesterase 4B (PDE4B) mediates S. pneumoniae-induced MUC5AC up-regulation by inhibiting the expression of a negative regulator MKP-1, which in turn leads to enhanced MAPK ERK activation and subsequent up-regulation of MUC5AC. PDE4B inhibits MKP-1 expression in a cAMP-PKA-dependent manner. PDE4-specific inhibitor rolipram inhibits S. pneumoniae-induced MUC5AC up-regulation both in vitro and in vivo. Moreover, we show that PDE4B plays a critical role in MUC5AC induction. Finally, topical and post-infection administration of rolipram into the middle ear potently inhibited S. pneumoniae-induced MUC5AC up-regulation. Collectively, these data demonstrate that PDE4B mediates ERK-dependent up-regulation of mucin MUC5AC by S. pneumoniae by inhibiting cAMP-PKA-dependent MKP-1 pathway. This study may lead to novel therapeutic strategy for inhibiting mucus overproduction.
Collapse
Affiliation(s)
- Jiyun Lee
- Center for Inflammation, Immunity, and Infection and Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Al-Tawashi A, Jung SY, Liu D, Su B, Qin J. Protein implicated in nonsyndromic mental retardation regulates protein kinase A (PKA) activity. J Biol Chem 2012; 287:14644-58. [PMID: 22375002 PMCID: PMC3340277 DOI: 10.1074/jbc.m111.261875] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mutation of the coiled-coil and C2 domain-containing 1A (CC2D1A) gene, which encodes a C2 domain and DM14 domain-containing protein, has been linked to severe autosomal recessive nonsyndromic mental retardation. Using a mouse model that produces a truncated form of CC2D1A that lacks the C2 domain and three of the four DM14 domains, we show that CC2D1A is important for neuronal differentiation and brain development. CC2D1A mutant neurons are hypersensitive to stress and have a reduced capacity to form dendrites and synapses in culture. At the biochemical level, CC2D1A transduces signals to the cyclic adenosine 3′,5′-monophosphate (cAMP)-protein kinase A (PKA) pathway during neuronal cell differentiation. PKA activity is compromised, and the translocation of its catalytic subunit to the nucleus is also defective in CC2D1A mutant cells. Consistently, phosphorylation of the PKA target cAMP-responsive element-binding protein, at serine 133, is nearly abolished in CC2D1A mutant cells. The defects in cAMP/PKA signaling were observed in fibroblast, macrophage, and neuronal primary cells derived from the CC2D1A KO mice. CC2D1A associates with the cAMP-PKA complex following forskolin treatment and accumulates in vesicles or on the plasma membrane in wild-type cells, suggesting that CC2D1A may recruit the PKA complex to the membrane to facilitate signal transduction. Together, our data show that CC2D1A is an important regulator of the cAMP/PKA signaling pathway, which may be the underlying cause for impaired mental function in nonsyndromic mental retardation patients with CC2D1A mutation.
Collapse
Affiliation(s)
- Azza Al-Tawashi
- Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | |
Collapse
|
38
|
Matera MG, Calzetta L, Segreti A, Cazzola M. Emerging drugs for chronic obstructive pulmonary disease. Expert Opin Emerg Drugs 2012; 17:61-82. [DOI: 10.1517/14728214.2012.660917] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
39
|
Kobayashi M, Kubo S, Hirano Y, Kobayashi S, Takahashi K, Shimizu Y. Anti-asthmatic effect of ASP3258, a novel phosphodiesterase 4 inhibitor. Int Immunopharmacol 2011; 12:50-8. [PMID: 22036845 DOI: 10.1016/j.intimp.2011.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 10/07/2011] [Accepted: 10/11/2011] [Indexed: 12/15/2022]
Abstract
ASP3258 is a potent and selective PDE4 inhibitor and exerts a wide-range of anti-inflammatory effects with low emetic potential, a major adverse effect of PDE4 inhibitors. Here, we investigated the anti-asthmatic potency of ASP3258 as compared with those of two representative PDE4 inhibitors: roflumilast and cilomilast. Orally administered ASP3258, roflumilast, and cilomilast all inhibited ovalbumin (OVA)-induced eosinophil infiltration into the airway of sensitized Brown Norway rats with ED(50) values of 0.81, 0.46, and 4.4 mg/kg, respectively. Histological examination also revealed a decreasing trend in inflammatory cell infiltration into the lung following ASP3258 administration. In vitro investigation of bronchodilatory activities showed that these compounds (10(-8)-10(-6) M) concentration-dependently inhibited OVA-induced contraction of trachea isolated from sensitized guinea pigs but had no effect on spasmogen-precontracted tracheal tension prepared from non-sensitized guinea pigs up to 10(-6) M. In vivo experiments using sensitized guinea pigs showed that these orally administered compounds inhibited OVA-induced increases in airway resistance with ED(50) values of 2.2, 0.35, and 12 mg/kg, respectively. Further, orally administered ASP3258 (0.1 and 1 mg/kg), roflumilast (0.1 and 1 mg/kg), and cilomilast (10 mg/kg) significantly suppressed airway hyperresponsiveness caused by OVA exposure. ASP3258's potent inhibition of antigen-induced bronchoconstriction and airway hyperresponsiveness, two characteristic symptoms of bronchial asthma, suggests that this compound will be useful in treating asthma.
Collapse
Affiliation(s)
- Miki Kobayashi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan.
| | | | | | | | | | | |
Collapse
|
40
|
Field SK. Roflumilast, a Novel Phosphodiesterase 4 Inhibitor, for COPD Patients with a History of Exacerbations. Clin Med Insights Circ Respir Pulm Med 2011; 5:57-70. [PMID: 22084617 PMCID: PMC3212861 DOI: 10.4137/ccrpm.s7049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Acute exacerbations of COPD (AECOPD) are major clinical events. They are associated with a more rapid decline in lung function, poorer quality of life scores, and an increased risk of dying. Exacerbations that require hospitalization have particular significance. Approximately 40% of the AECOPD patients who require hospitalization will die in the subsequent year. Since many AECOPD require hospitalization, they account for most of the expense of caring for COPD patients. Treatment with long-acting bronchodilators and combination inhaled corticosteroid/long-acting bronchodilator inhalers reduces but does not eliminate AECOPD. Roflumilast, a selective phosphodiesterase 4 (PDE4) inhibitor, is an anti-inflammatory medication that improves lung function in patients with COPD. In patients with more severe airway obstruction, clinical features of chronic bronchitis, and a history of AECOPD, roflumilast reduces the frequency of AECOPD when given in combination with short-acting bronchodilators, long-acting bronchodilators, or inhaled corticosteroids. It is generally well tolerated but the most common adverse effects include diarrhea, nausea, weight loss, and headaches. In clinical trials, patients treated with roflumilast experienced weight loss that averaged just over 2 kg but was primarily due to the loss of fat tissue. Weight loss was least in underweight patients and obese patients experienced the greatest weight loss. An unexpected benefit of treatment with roflumilast was that fasting blood glucose and hemoglobin A1c levels improved in patients with comorbid type 2 diabetes mellitus. Roflumilast, the first selective PDE4 inhibitor to be marketed, is a promising drug for the management of COPD patients with more severe disease.
Collapse
Affiliation(s)
- Stephen K. Field
- Division of Respiratory Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
41
|
Page CP, Spina D. Phosphodiesterase inhibitors in the treatment of inflammatory diseases. Handb Exp Pharmacol 2011:391-414. [PMID: 21695650 DOI: 10.1007/978-3-642-17969-3_17] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Phosphodiesterase 4 (PDE4) belongs to a family of enzymes which catalyzes the breakdown of 3, 5'-adenosine cyclic monophosphate (cAMP) and is ubiquitously expressed in inflammatory cells. There is little evidence that inflammatory diseases are caused by increased expression of this isoenzyme, although human inflammatory cell activity can be suppressed by selective PDE4 inhibitors. Consequently, there is intense interest in the development of selective PDE4 inhibitors for the treatment of a range of inflammatory diseases, including asthma, chronic obstructive pulmonary disease (COPD), inflammatory bowel disease, and psoriasis. Recent clinical trials with roflumilast in COPD have confirmed the therapeutic potential of targeting PDE4 and recently roflumilast has been approved for marketing in Europe and the USA, although side effects such as gastrointestinal disturbances, particularly nausea and emesis as well as headache and weight loss, may limit the use of this drug class, at least when administered by the oral route. However, a number of strategies are currently being pursued in attempts to improve clinical efficacy and reduce side effects of PDE4 inhibitors, including delivery via the inhaled route, development of nonemetic PDE4 inhibitors, mixed PDE inhibitors, and/or antisense biologicals targeted toward PDE4.
Collapse
Affiliation(s)
- C P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, Franklin Wilkins Building, London SE1 9NH, UK.
| | | |
Collapse
|
42
|
Lira EC, Gonçalves DA, Parreiras-E-Silva LT, Zanon NM, Kettelhut IC, Navegantes LC. Phosphodiesterase-4 inhibition reduces proteolysis and atrogenes expression in rat skeletal muscles. Muscle Nerve 2011; 44:371-81. [DOI: 10.1002/mus.22066] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
43
|
de Mey C, Nassr N, Lahu G. No relevant cardiac, pharmacokinetic or safety interactions between roflumilast and inhaled formoterol in healthy subjects: an open-label, randomised, actively controlled study. BMC CLINICAL PHARMACOLOGY 2011; 11:7. [PMID: 21631929 PMCID: PMC3127977 DOI: 10.1186/1472-6904-11-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 06/01/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND Roflumilast is an oral, selective phosphodiesterase 4 inhibitor with anti-inflammatory effects in chronic obstructive pulmonary disease (COPD). The addition of roflumilast to long-acting bronchodilators improves lung function in patients with moderate-to-severe COPD. The present study investigated drug-drug interaction effects between inhaled formoterol and oral roflumilast. METHODS This was a single-centre (investigational clinic), open, randomised, multiple-dose, parallel-group study. In Regimen A, healthy men were treated with roflumilast (500 μg tablet once daily; Day 2-18) and concomitant formoterol (24 μg twice daily; Day 12-18). In Regimen B, healthy men were treated with formoterol (24 μg twice daily; Day 2-18) and concomitant roflumilast (500 μg once daily; Day 9-18). Steady-state plasma pharmacokinetics of roflumilast, roflumilast N-oxide and/or formoterol (Cmax and AUC0-τ) as well as pharmacodynamics - blood pressure, transthoracic impedance cardiography (ZCG), 12-lead digital electrocardiography, peripheral blood eosinophils, and serum glucose and potassium concentrations - were evaluated through Day 1 (baseline), Day 8 (Regimen B: formoterol alone) or Day 11 (Regimen A: roflumilast alone), and Day 18 (Regimen A and B: roflumilast plus formoterol). Blood and urine samples were taken for safety assessment at screening, pharmacokinetic profiling days and Day 19. Adverse events were monitored throughout the study. RESULTS Of the 27 subjects enrolled, 24 were evaluable (12 in each regimen). No relevant pharmacokinetic interactions occurred. Neither roflumilast nor formoterol were associated with significant changes in cardiovascular parameters as measured by ZCG, and these parameters were not affected during concomitant administration. Formoterol was associated with a slight increase in heart rate and a corresponding shortening of the QT interval, without changes in the heart rate-corrected QTc interval. There were small effects on the other pharmacodynamic assessments when roflumilast and formoterol were administered individually, but no interactions or safety concerns were seen after concomitant administration. No severe or serious adverse events were reported, and no adverse events led to premature study discontinuation. CONCLUSIONS No clinically relevant pharmacokinetic or pharmacodynamic interactions were found when oral roflumilast was administered concomitantly with inhaled formoterol, including no effect on cardiac repolarisation. Roflumilast was well tolerated.
Collapse
Affiliation(s)
- Christian de Mey
- ACPS - Applied Clinical Pharmacology Services, Mainz-Kastel, Germany
| | | | | |
Collapse
|
44
|
Huennemeyer A, Nassr N, Bredenbröker D, Lahu G. Supra-therapeutic doses of roflumilast have no effect on cardiac repolarization in healthy subjects. Expert Opin Drug Saf 2011; 10:509-19. [DOI: 10.1517/14740338.2011.581659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Rabe KF. Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease. Br J Pharmacol 2011; 163:53-67. [PMID: 21232047 PMCID: PMC3085868 DOI: 10.1111/j.1476-5381.2011.01218.x] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/22/2010] [Accepted: 12/28/2010] [Indexed: 01/20/2023] Open
Abstract
Phosphodiesterase 4 (PDE4) is a member of the PDE enzyme superfamily that inactivates cyclic adenosine monophosphate and cyclic guanosine monophosphate, and is the main PDE isoenzyme occurring in cells involved in inflammatory airway disease such as chronic obstructive pulmonary disease (COPD). COPD is a preventable and treatable disease and is characterized by airflow obstruction that is not fully reversible. Chronic progressive symptoms, particularly dyspnoea, chronic bronchitis and impaired overall health are worse in those who have frequent, acute episodes of symptom exacerbation. Although several experimental PDE4 inhibitors are in clinical development, roflumilast, a highly selective PDE4 inhibitor, is the first in its class to be licensed, and has recently been approved in several countries for oral, once-daily treatment of severe COPD. Clinical trials have demonstrated that roflumilast improves lung function and reduces exacerbation frequency in COPD. Furthermore, its unique mode of action may offer the potential to target the inflammatory processes underlying COPD. Roflumilast is effective when used concomitantly with all forms of bronchodilator and even in patients treated with inhaled corticosteroids. Roflumilast thus represents an important addition to current therapeutic options for COPD patients with chronic bronchitis, including those who remain symptomatic despite treatment. This article reviews the current status of PDE4 inhibitors, focusing on the pharmacokinetics, efficacy and safety of roflumilast. In particular, it provides an overview of the effects of roflumilast on lung function and exacerbations, glucose homoeostasis and weight loss, and the concomitant use of long-acting beta(2)-adrenergic receptor agonists and short-acting muscarinic receptor antagonists.
Collapse
Affiliation(s)
- Klaus F Rabe
- Department of Medicine, University Kiel, Germany.
| |
Collapse
|
46
|
Kobayashi M, Kubo S, Iwata M, Ohtsu Y, Takahashi K, Shimizu Y. ASP3258, an orally active potent phosphodiesterase 4 inhibitor with low emetic activity. Int Immunopharmacol 2011; 11:732-9. [PMID: 21315169 DOI: 10.1016/j.intimp.2011.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 01/05/2011] [Accepted: 01/21/2011] [Indexed: 10/18/2022]
Abstract
We investigated the pharmacology of a novel phosphodiesterase (PDE) 4 inhibitor, ASP3258 (3-[4-(3-chlorophenyl)-1-ethyl-7-methyl-2-oxo-1,2-dihydro-1,8-naphthyridin-3-yl] propanoic acid), comparing its potency with that of the most advanced PDE4 inhibitors, roflumilast and cilomilast. PDE4 inhibition by ASP3258 (IC(50)=0.28nM) was as potent as that achieved with roflumilast. ASP3258 inhibited lipopolysaccharide-induced tumor necrosis factor (TNF)-α production in rat whole blood cells (IC(50)=8.8 nM) and rat alveolar macrophages (IC(50)=2.6 nM). Orally administered ASP3258, roflumilast, and cilomilast dose-dependently inhibited production of interleukin-4, TNF-α, and cysteinyl leukotrienes, as well as leukocyte infiltration in bronchoalveolar lavage fluid from the airways of ovalbumin-sensitized Brown Norway rats, and these compounds showed almost complete inhibition at doses of 3, 3, and 30 mg/kg, respectively. PDE4 inhibitors induce emesis by mimicking the pharmacological action of α(2)-adrenoceptor antagonist. However, orally administered roflumilast (3mg/kg) and cilomilast (10mg/kg), but not ASP3258 (3mg/kg), inhibited α(2)-adrenoceptor agonist-induced anesthesia in rats and induced emesis in ferrets. Although ASP3258 (3mg/kg) inhibited airway inflammation completely, it had no emetic activity. As such, this compound may be useful in treating airway inflammatory diseases such as asthma and COPD.
Collapse
Affiliation(s)
- Miki Kobayashi
- Pharmacology Research Labs, Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Paterniti I, Mazzon E, Gil C, Impellizzeri D, Palomo V, Redondo M, Perez DI, Esposito E, Martinez A, Cuzzocrea S. PDE 7 inhibitors: new potential drugs for the therapy of spinal cord injury. PLoS One 2011; 6:e15937. [PMID: 21297958 PMCID: PMC3031524 DOI: 10.1371/journal.pone.0015937] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 11/30/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Primary traumatic mechanical injury to the spinal cord (SCI) causes the death of a number of neurons that to date can neither be recovered nor regenerated. During the last years our group has been involved in the design, synthesis and evaluation of PDE7 inhibitors as new innovative drugs for several neurological disorders. Our working hypothesis is based on two different facts. Firstly, neuroinflammation is modulated by cAMP levels, thus the key role for phosphodiesterases (PDEs), which hydrolyze cAMP, is undoubtedly demonstrated. On the other hand, PDE7 is expressed simultaneously on leukocytes and on the brain, highlighting the potential crucial role of PDE7 as drug target for neuroinflammation. METHODOLOGY/PRINCIPAL FINDINGS Here we present two chemically diverse families of PDE7 inhibitors, designed using computational techniques such as virtual screening and neuronal networks. We report their biological profile and their efficacy in an experimental SCI model induced by the application of vascular clips (force of 24 g) to the dura via a four-level T5-T8 laminectomy. We have selected two candidates, namely S14 and VP1.15, as PDE7 inhibitors. These compounds increase cAMP production both in macrophage and neuronal cell lines. Regarding drug-like properties, compounds were able to cross the blood brain barrier using parallel artificial membranes (PAMPA) methodology. SCI in mice resulted in severe trauma characterized by edema, neutrophil infiltration, and production of a range of inflammatory mediators, tissue damage, and apoptosis. Treatment of the mice with S14 and VP1.15, two PDE7 inhibitors, significantly reduced the degree of spinal cord inflammation, tissue injury (histological score), and TNF-α, IL-6, COX-2 and iNOS expression. CONCLUSIONS/SIGNIFICANCE All these data together led us to propose PDE7 inhibitors, and specifically S14 and VP1.15, as potential drug candidates to be further studied for the treatment of SCI.
Collapse
Affiliation(s)
- Irene Paterniti
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | - Carmen Gil
- Instituto de Quimica Médica-CSIC, Madrid, Spain
| | - Daniela Impellizzeri
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | | | | | | | - Emanuela Esposito
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| | | | - Salvatore Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| |
Collapse
|
48
|
Song JS, Rho HJ, Park JS, Kim MS, Lee BH, Seo JW, Jeon DJ, Cheon HG, Ahn SH, Kwon KI, Bae MA. Preclinical pharmacokinetics of PDE-310, a novel PDE4 inhibitor. Drug Metab Pharmacokinet 2010; 26:192-200. [PMID: 21206135 DOI: 10.2133/dmpk.dmpk-10-rg-065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A novel phosphodiesterase-4 inhibitor, 2-aryl-7(3',4'-dialkoxyphenyl)-pyrazolo[1,5-alpha] pyrimidine (PDE-310), has been synthesized for the treatment of respiratory diseases. We conducted in vitro and in vivo studies to characterize the pharmacokinetics of PDE-310. The high liver microsomal stability and low inhibitory potency against CYP isoforms of PDE-310 suggested a low first-pass effect and high bioavailability. PDE-310 exhibited high Caco-2 cell permeability in the absorptive direction (apparent permeability coefficients, ∼20 × 10(-6) cm/s), with higher transport in the secretory direction, giving efflux ratios of 3.9 and 2.6 at 5 and 10 µM, respectively. In addition, the high efflux ratio and improved absorption on treatment with efflux transporter inhibitors such as verapamil and MK-571 indicated the involvement of P-gp, BCRP and MRP2 in intestinal transport. PDE-310 bound strongly to human plasma proteins, whereas significantly more PDE-310 (27-34%) was free in rat plasma. Following intravenous administration, nonlinear elimination of PDE-310 was observed at the tested dose ranges (K(m), 0.87 µg/mL; V(max), 0.3 mg·h(-1)·kg(-1)). Following oral PDE-310 administration, dose-normalized AUC and T(max) increased significantly in a dose-dependent manner. PDE-310 exhibited high oral bioavailability (>70%) and was distributed well to various tissues except brain and testis.
Collapse
Affiliation(s)
- Jin Sook Song
- Drug Discovery Platform Technology Team, Division of Bio-organic Science, Korea Research Institute of Chemical Technology, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lahu G, Hünnemeyer A, Diletti E, Elmlinger M, Ruth P, Zech K, McCracken N, Facius A. Population pharmacokinetic modelling of roflumilast and roflumilast N-oxide by total phosphodiesterase-4 inhibitory activity and development of a population pharmacodynamic-adverse event model. Clin Pharmacokinet 2010; 49:589-606. [PMID: 20690782 DOI: 10.2165/11536600-000000000-00000] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Roflumilast is an oral, selective phosphodiesterase (PDE)-4 inhibitor in development for the treatment of chronic obstructive pulmonary disease (COPD). Both roflumilast and its metabolite roflumilast N-oxide have anti-inflammatory properties that contribute to overall pharmacological activity. OBJECTIVES To model the pharmacokinetics of roflumilast and roflumilast N-oxide, evaluate the influence of potential covariates, use the total PDE4 inhibitory activity (tPDE4i) concept to estimate the combined inhibition of PDE4 by roflumilast and roflumilast N-oxide, and use individual estimates of tPDE4i to predict the occurrence of adverse events (AEs) in patients with moderate-to-severe COPD. METHODS We modelled exposure to roflumilast and roflumilast N-oxide (21 studies provided the index dataset and five separate studies provided the validation dataset), extended the models to COPD (using data from two studies) and assessed the robustness of the parameter estimates. A parametric bootstrap estimation was used to quantify tPDE4i in subpopulations. We established logistic regression models for each AE occurring in >2% of patients in a placebo-controlled trial that achieved a p-value of <0.2 in a permutation test. The exposure variables were the area under the plasma concentration-time curve (AUC) of roflumilast, the AUC of roflumilast N-oxide and tPDE4i. Individual AUC values were estimated from population models. RESULTS Roflumilast pharmacokinetics were modelled with a two-compartment model with first-order absorption including a lag time. A one-compartment model with zero-order absorption was used for roflumilast N-oxide. The final models displayed good descriptive and predictive performance with no appreciable systematic trends versus time, dose or study. Posterior predictive checks and robustness analysis showed that the models adequately described the pharmacokinetic parameters and the covariate effects on disposition. For roflumilast, the covariates of sex, smoking and race influenced clearance; and food influenced the absorption rate constant and lag time. For roflumilast N-oxide, age, sex and smoking influenced clearance; age, sex and race influenced the fraction metabolized; bodyweight influenced the apparent volume of distribution; and food influenced the apparent duration of formation. The COPD covariate increased the central volume of distribution of roflumilast by 184% and reduced its clearance by 39%; it also reduced the estimated volume of distribution of roflumilast N-oxide by 21% and reduced its clearance by 7.9%. Compared with the reference population (male, non-smoking, White, healthy, 40-year-old subjects), the relative geometric mean [95% CI] tPDE4i was higher in patients with COPD (12.6% [-6.6, 35.6]), women (19.3% [8.2, 31.6]), Black subjects (42.1% [16.4, 73.4]), Hispanic subjects (28.2% [4.1, 57.9]) and older subjects (e.g. 8.3% [-11.2, 32.2] in 60-year-olds), and was lower in smokers (-19.1% [-34.0, -0.7]). Among all possible subgroups in this analysis, the subgroup with maximal tPDE4i comprised elderly, Black, female, non-smoking, COPD patients (tPDE4i 217% [95% CI 107, 437] compared with the value in the reference population). The probability of a patient with tPDE4i at the population geometric mean [95% CI] was 13.0% [7.5, 18.5] for developing diarrhoea, 6.0% [2.6, 9.4] for nausea and 5.1% [1.9, 8.6] for headache. CONCLUSIONS Covariate effects have a limited impact on tPDE4i. There was a general association between tPDE4i and the occurrence of common AEs in patients with COPD.
Collapse
Affiliation(s)
- Gezim Lahu
- Department of Pharmacometrics and Pharmacokinetics, Nycomed GmbH, Byk-Gulden-Strasse 2, Konstanz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Giembycz MA, Field SK. Roflumilast: first phosphodiesterase 4 inhibitor approved for treatment of COPD. DRUG DESIGN DEVELOPMENT AND THERAPY 2010; 4:147-58. [PMID: 20689641 PMCID: PMC2915539 DOI: 10.2147/dddt.s7667] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Indexed: 01/28/2023]
Abstract
In April 2010, the European Medicines Agency Committee for Medicinal Products for Human Use recommended approval of roflumilast, a selective phosphodiesterase 4 inhibitor, for the “maintenance treatment of severe chronic obstructive pulmonary disease (COPD, FEV1 postbronchodilator less than 50% predicted) associated with chronic bronchitis in adult patients with a history of frequent exacerbations as add-on to bronchodilator treatment”. This decision was based, in part, on the results of several large, international, multicenter, randomized, placebo-controlled trials of either six or 12 months’ duration that had been undertaken in COPD patients. Roflumilast 500 μg daily improved lung function and reduced exacerbations in patients with more severe COPD, especially those with chronic bronchitis, frequent exacerbations, or who required frequent rescue inhaler therapy in the placebo-controlled trials. It also improved lung function and reduced exacerbations in patients with moderately severe COPD treated with salmeterol or tiotropium. Advantages of roflumilast over inhaler therapy are that it is an oral tablet and only needs to be taken once daily. While taking roflumilast, the most common adverse effects patients experienced were gastrointestinal upset and headache. Weight loss, averaging 2.2 kg, occurred in patients treated with roflumilast. Patients taking roflumilast were more likely to drop out of the trials than patients in the control groups. Patients who discontinued therapy usually did so during the first few weeks and were more likely to have experienced gastrointestinal side effects. Roflumilast is the first selective phosphodiesterase 4 inhibitor and will offer physicians another treatment option for patients with more severe COPD.
Collapse
Affiliation(s)
- Mark A Giembycz
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Alberta, Canada
| | | |
Collapse
|