1
|
Liu H, Wang Q, Huang Y, Deng J, Xie X, Zhu J, Yuan Y, He YM, Huang YY, Luo HB, He X. Discovery of novel PDE4 inhibitors targeting the M-pocket from natural mangostanin with improved safety for the treatment of Inflammatory Bowel Diseases. Eur J Med Chem 2022; 242:114631. [PMID: 35985255 DOI: 10.1016/j.ejmech.2022.114631] [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: 05/15/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 11/28/2022]
Abstract
Inflammatory Bowel Diseases (IBDs) are chronic disorders with iterative intestinal mucosal inflammation which remain unmet medical needs. PDE4 inhibitors were reported to be novel anti-IBD agents, but their clinical use was hampered by side effects such as emesis and nausea. Herein, structure-based discovery of natural mangostanin (1) targeting the M-pocket resulted in the novel and potent PDE4 inhibitor 22d (IC50 = 3.5 nM) and favorable physico-chemical properties. X-Ray study revealed that 22d interacted tightly with the M-pocket and maintained the key interactions between PDE4 and roflumilast. Worthy to note that compounds 22d and our previously reported 4e and 18a, originating from mangostanin, all caused no emesis on beagle dogs at the oral dose of 10 mg/kg, confirming the safety superiority of scaffold in mangostanin derivatives over that in positive roflumilast. Finally, administration of 22d (5.0 mg/kg, twice-daily) exhibited comparable anti-IBD effects to the positive control dipyridamole (25.0 mg/kg, twice-daily) in the dextran sulfate sodium (DSS)-induced IBD mice model, indicating its potential as a novel anti-IBD agent.
Collapse
Affiliation(s)
- Haobai Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Quan Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yue Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Jinhui Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Xi Xie
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Jiaqi Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yijun Yuan
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China; Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya, 572000, China
| | - Yue-Ming He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yi-You Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China; Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya, 572000, China.
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China; Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya, 572000, China.
| | - Xixin He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Estimation of the lipophilicity of purine-2,6-dione-based TRPA1 antagonists and PDE4/7 inhibitors with analgesic activity. Bioorg Med Chem Lett 2021; 49:128318. [PMID: 34391892 DOI: 10.1016/j.bmcl.2021.128318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/03/2021] [Accepted: 08/08/2021] [Indexed: 11/22/2022]
Abstract
Lipophilicity is one of the principal QSAR parameters which influences among others the pharmacodynamics and pharmacokinetic properties of a drug candidates. In this paper, the lipophilicity of 14 amide derivatives of 1,3-dimethyl-2,6-dioxopurin-7-yl-alkylcarboxylic acids as multifunctional TRPA1 channel antagonists and phosphodiesterase 4/7 inhibitors with analgesic activity were investigated, using reversed-phase thin-layer chromatography method. It was observed that the retention behavior of the analyzed compounds was dependent on their structural features i.e. an aliphatic linker length, a kind of substituent at 8 position of purine-2,6-dione scaffold as well as on a substitution in a phenyl group. The experimental parameters (RM0) were compared with computationally calculated partition coefficient values by Principal Component Analysis (PCA). To verify the influence of lipophilic parameter of the investigated compounds on their biological activity the Kruskal-Wallis test was performed. The lowest lipophilicity was observed for the compounds with weak PDE4/7 inhibitory potency. The differences between the lipophilicity of potent inhibitors and inactive compounds were statistically significant. It was found that the presence of more lipophilic propoxy- or butoxy- substituents as well as the elongation of the aliphatic chain to propylene one between the purine-2,6-dione core and amide group were preferable for desired multifunctional activity.
Collapse
|
3
|
Jankowska A, Pawłowski M, Chłoń-Rzepa G. Diabetic Theory in Anti-Alzheimer's Drug Research and Development. Part 2: Therapeutic Potential of cAMP-Specific Phosphodiesterase Inhibitors. Curr Med Chem 2021; 28:3535-3553. [PMID: 32940168 DOI: 10.2174/0929867327666200917125857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent age-related neurodegenerative disease that affects the cognition, behavior, and daily activities of individuals. Studies indicate that this disease is characterized by several pathological mechanisms, including the accumulation of amyloid-beta peptide, hyperphosphorylation of tau protein, impairment of cholinergic neurotransmission, and increase in inflammatory responses within the central nervous system. Chronic neuroinflammation associated with AD is closely related to disturbances in metabolic processes, including insulin release and glucose metabolism. As AD is also called type III diabetes, diverse compounds having antidiabetic effects have been investigated as potential drugs for its symptomatic and disease-modifying treatment. In addition to insulin and oral antidiabetic drugs, scientific attention has been paid to cyclic-3',5'-adenosine monophosphate (cAMP)-specific phosphodiesterase (PDE) inhibitors that can modulate the concentration of glucose and related hormones and exert beneficial effects on memory, mood, and emotional processing. In this review, we present the most recent reports focusing on the involvement of cAMP-specific PDE4, PDE7, and PDE8 in glycemic and inflammatory response controls as well as the potential utility of the PDE inhibitors in the treatment of AD. Besides the results of in vitro and in vivo studies, the review also presents recent reports from clinical trials.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| | - Maciej Pawłowski
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| |
Collapse
|
4
|
Amin KM, Hegazy GH, George RF, Ibrahim NR, Mohamed NM. Design, synthesis, and pharmacological characterization of some 2-substituted-3-phenyl-quinazolin-4(3H)-one derivatives as phosphodiesterase inhibitors. Arch Pharm (Weinheim) 2021; 354:e2100051. [PMID: 33977557 DOI: 10.1002/ardp.202100051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022]
Abstract
Some 3-phenyl-quinazolin-4(3H)-one-2-thioethers (3a-e, 5a,b, 7a-e, 9a-d, 10a-d, and 12) along with 2-aminoquinazoline derivatives 13a-c were prepared and screened for their in vitro phosphodiesterase (PDE) inhibitory activity. Some compounds such as 7d,e, 9a,b,d, 10a,d, and 13b exhibited promising activity as compared with the non-selective PDE inhibitor IBMX. This inhibitory activity was validated by molecular docking in the active site of PDE7A and PDE4 to investigate their selectivity. Furthermore, the most active compound 10d (IC50 = 1.15 μM) was tested in vivo using behavioral tests. Compound 10d was able to pass the blood-brain barrier and improve scopolamine-induced cognitive deficits. Therefore, this core can be considered as a promising scaffold for further optimization to obtain new compounds with better PDE7A selective inhibition.
Collapse
Affiliation(s)
- Kamilia M Amin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Gehan H Hegazy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham F George
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nahla R Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nada M Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo, Egypt
| |
Collapse
|
5
|
Schröder S, Scheunemann M, Wenzel B, Brust P. Challenges on Cyclic Nucleotide Phosphodiesterases Imaging with Positron Emission Tomography: Novel Radioligands and (Pre-)Clinical Insights since 2016. Int J Mol Sci 2021; 22:ijms22083832. [PMID: 33917199 PMCID: PMC8068090 DOI: 10.3390/ijms22083832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) represent one of the key targets in the research field of intracellular signaling related to the second messenger molecules cyclic adenosine monophosphate (cAMP) and/or cyclic guanosine monophosphate (cGMP). Hence, non-invasive imaging of this enzyme class by positron emission tomography (PET) using appropriate isoform-selective PDE radioligands is gaining importance. This methodology enables the in vivo diagnosis and staging of numerous diseases associated with altered PDE density or activity in the periphery and the central nervous system as well as the translational evaluation of novel PDE inhibitors as therapeutics. In this follow-up review, we summarize the efforts in the development of novel PDE radioligands and highlight (pre-)clinical insights from PET studies using already known PDE radioligands since 2016.
Collapse
Affiliation(s)
- Susann Schröder
- Department of Research and Development, ROTOP Pharmaka Ltd., 01328 Dresden, Germany
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
- Correspondence: ; Tel.: +49-341-234-179-4631
| | - Matthias Scheunemann
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| | - Barbara Wenzel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| |
Collapse
|
6
|
Ručilová V, Świerczek A, Vanda D, Funk P, Lemrová B, Gawalska A, Bucki A, Nowak B, Zadrożna M, Pociecha K, Soural M, Wyska E, Pawłowski M, Chłoń-Rzepa G, Zajdel P. New imidazopyridines with phosphodiesterase 4 and 7 inhibitory activity and their efficacy in animal models of inflammatory and autoimmune diseases. Eur J Med Chem 2021; 209:112854. [PMID: 33022582 DOI: 10.1016/j.ejmech.2020.112854] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/02/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022]
Abstract
Herein, we describe the rapid synthesis of a focused library of trisubstituted imidazo[4,5-b]pyridines and imidazo[4,5-c]pyridines from 2,4-dichloro-3-nitropyridine using the combination of solution-phase/solid-phase chemistry as new potential anti-inflammatory agents in the treatment of autoimmune diseases. Structure-activity relationship studies, followed by the structure optimization, provided hit compounds (17 and 28) which inhibited phosphodiesterase 4 (PDE4) with IC50 values comparable to rolipram and displayed different inhibitory potency against phosphodiesterase 7 (PDE7). Among them, compound 17 showed a beneficial effect in all the studied animal models of inflammatory and autoimmune diseases (concanavalin A-induced hepatitis, lipopolysaccharide-induced endotoxemia, collagen-induced arthritis, and MOG35-55-induced encephalomyelitis). In addition, compound 17 showed a favorable pharmacokinetic profile after intraperitoneal administration; it was characterized by a fast absorption from the peritoneal cavity and a relatively long terminal half-life in rats. It was found to penetrate brain barrier in mice. The performed experiments sheds light on the impact of PDE7A inhibition for the efficacy of PDE4 inhibitors in these disease conditions.
Collapse
Affiliation(s)
- Veronika Ručilová
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00, Olomouc, Czech Republic; Palacký University, Faculty of Science, Department of Organic Chemistry, 17 listopadu 12, 771 46, Olomouc, Czech Republic
| | - Artur Świerczek
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics and Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - David Vanda
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00, Olomouc, Czech Republic; Palacký University, Faculty of Science, Department of Organic Chemistry, 17 listopadu 12, 771 46, Olomouc, Czech Republic
| | - Petr Funk
- Palacký University, Faculty of Science, Department of Organic Chemistry, 17 listopadu 12, 771 46, Olomouc, Czech Republic
| | - Barbora Lemrová
- Palacký University, Faculty of Science, Department of Organic Chemistry, 17 listopadu 12, 771 46, Olomouc, Czech Republic
| | - Alicja Gawalska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Adam Bucki
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Barbara Nowak
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacobiology, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Monika Zadrożna
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacobiology, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Krzysztof Pociecha
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics and Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Miroslav Soural
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 779 00, Olomouc, Czech Republic; Palacký University, Faculty of Science, Department of Organic Chemistry, 17 listopadu 12, 771 46, Olomouc, Czech Republic
| | - Elżbieta Wyska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics and Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Maciej Pawłowski
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Paweł Zajdel
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland.
| |
Collapse
|
7
|
Sharma H, Lather V, Grewal AS, Pandita D. Synthesis, Anti-inflammatory Activity and Docking Studies of Some Newer 1,3-Thiazolidine-2,4-dione Derivatives as Dual Inhibitors of PDE4 and PDE7. Curr Comput Aided Drug Des 2019; 15:225-234. [PMID: 30280674 DOI: 10.2174/1573409914666181003151528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/17/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Phosphodiesterase 4 (PDE4) and phosphodiesterase 7 (PDE7), PDE superfamily members, increase inflammatory processes in immunomodulatory as well as pro-inflammatory cells via breakdown of cyclic adenosine monophosphate. Dual inhibitors of PDE4 and PDE7 are a novel class of drug candidates which can regulate pro-inflammatory as well as T-cell function and can be particularly advantageous in the treatment of a wide-ranging disorders associated with the immune system as well as inflammatory diseases with fewer unwanted adverse effects. OBJECTIVE The current research work was planned to design and synthesize some newer substituted 1,3- thiazolidine-2,4-dione derivatives as dual inhibitors of PDE4 and PDE7 followed by evaluation of their anti-inflammatory activity and in silico docking studies. METHODS A new series of substituted 1,3-thiazolidine-2,4-dione derivatives was synthesized followed by evaluation of their anti-inflammatory activity in animal models. In silico docking studies were performed for the evaluation of the binding pattern of synthesized derivatives in the binding site of both PDE4 and PDE7 proteins. RESULTS Amongst the newly synthesized derivatives, compounds 5 and 12 showed higher antiinflammatory activity in the animal model. The results of in vivo animal studies were found to be in concordance with the results of molecular docking studies. CONCLUSION These newly synthesized derivatives can act as the lead molecules for the design of safe and therapeutically effective agents for various inflammatory diseases acting via inhibition of both PDE4 and PDE7.
Collapse
Affiliation(s)
- Himanshu Sharma
- Jan Nayak Ch. Devi Lal Memorial College of Pharmacy, Sirsa 125055, Haryana, India
| | - Viney Lather
- Jan Nayak Ch. Devi Lal Memorial College of Pharmacy, Sirsa 125055, Haryana, India.,Amity Institute of Pharmacy, Amity University, Noida 201303, Uttar Pradesh, India
| | - Ajmer Singh Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Patiala 140401, Punjab, India
| | - Deepti Pandita
- Jan Nayak Ch. Devi Lal Memorial College of Pharmacy, Sirsa 125055, Haryana, India.,Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida 201303, Uttar Pradesh, India
| |
Collapse
|
8
|
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
|
9
|
Wu Y, Li Z, Huang YY, Wu D, Luo HB. Novel Phosphodiesterase Inhibitors for Cognitive Improvement in Alzheimer's Disease. J Med Chem 2018; 61:5467-5483. [PMID: 29363967 DOI: 10.1021/acs.jmedchem.7b01370] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is one of the greatest public health challenges. Phosphodiesterases (PDEs) are a superenzyme family responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Since several PDE subfamilies are highly expressed in the human brain, the inhibition of PDEs is involved in neurodegenerative processes by regulating the concentration of cAMP and/or cGMP. Currently, PDEs are considered as promising targets for the treatment of AD since many PDE inhibitors have exhibited remarkable cognitive improvement effects in preclinical studies and over 15 of them have been subjected to clinical trials. The aim of this review is to summarize the outstanding progress that has been made by PDE inhibitors as anti-AD agents with encouraging results in preclinical studies and clinical trials. The binding affinity, pharmacokinetics, underlying mechanisms, and limitations of these PDE inhibitors in the treatment of AD are also reviewed and discussed.
Collapse
Affiliation(s)
- Yinuo Wu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Zhe Li
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Yi-You Huang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Deyan Wu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| |
Collapse
|
10
|
Martín-Álvarez R, Paúl-Fernández N, Palomo V, Gil C, Martínez A, Mengod G. A preliminary investigation of phoshodiesterase 7 inhibitor VP3.15 as therapeutic agent for the treatment of experimental autoimmune encephalomyelitis mice. J Chem Neuroanat 2017; 80:27-36. [DOI: 10.1016/j.jchemneu.2016.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/10/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
|
11
|
Novel Radioligands for Cyclic Nucleotide Phosphodiesterase Imaging with Positron Emission Tomography: An Update on Developments Since 2012. Molecules 2016; 21:molecules21050650. [PMID: 27213312 PMCID: PMC6273803 DOI: 10.3390/molecules21050650] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/19/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a class of intracellular enzymes that inactivate the secondary messenger molecules, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Thus, PDEs regulate the signaling cascades mediated by these cyclic nucleotides and affect fundamental intracellular processes. Pharmacological inhibition of PDE activity is a promising strategy for treatment of several diseases. However, the role of the different PDEs in related pathologies is not completely clarified yet. PDE-specific radioligands enable non-invasive visualization and quantification of these enzymes by positron emission tomography (PET) in vivo and provide an important translational tool for elucidation of the relationship between altered expression of PDEs and pathophysiological effects as well as (pre-)clinical evaluation of novel PDE inhibitors developed as therapeutics. Herein we present an overview of novel PDE radioligands for PET published since 2012.
Collapse
|
12
|
Thomae D, Servaes S, Vazquez N, Wyffels L, Dedeurwaerdere S, Van der Veken P, Joossens J, Augustyns K, Stroobants S, Staelens S. Synthesis and preclinical evaluation of an 18F labeled PDE7 inhibitor for PET neuroimaging. Nucl Med Biol 2015; 42:975-81. [PMID: 26330158 DOI: 10.1016/j.nucmedbio.2015.07.007] [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] [Received: 06/12/2015] [Revised: 07/14/2015] [Accepted: 07/21/2015] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Phosphodiesterase 7 (PDE7) hydrolyzes selectively cyclic adenosine monophosphate (cAMP) which is an intracellular second messenger. PDE7 is expressed by 2 genes which are both present in the brain. To date there is no radiotracer for PDE7 imaging described and detection of PDE7 has only been performed by ex vivo techniques. In this report we describe the radiosynthesis of a novel fluorine-18 labeled radiotracer for PDE7 as well as the in vivo evaluation in mice to verify whether it has potential for imaging of PDE7 in the brain. METHODS We have synthesized a potent fluorinated PDE7 inhibitor, [(18)F]MICA-003 (PDE7 IC(50)=17 nM) and the corresponding tosylate precursor for radiolabeling. [(18)F]MICA-003 was injected in C57BL/6J mice (n=5) and in vivo images were acquired by μPET imaging. Radiometabolite analysis in plasma and brain was performed to determine the stability of the radioligand. RESULTS [(18)F]MICA-003 was synthesized by direct fluorination of the tosylate and produced in high decay corrected radiochemical yield (40%), high radiochemical purity (>98%) and high specific activity (86-497 GBq/μmol). μPET imaging revealed that [(18)F]MICA-003 crosses the blood brain barrier and has a homogenous distribution over the brain which washes out after the initial peak uptake. [(18)F]MICA-003 was quickly metabolized in plasma with 8.9%±0.59% of intact [(18)F]MICA-003 remaining at 5m in post injection. We observed the formation of three distinct radiometabolites of which the main radiometabolite was also detected in the brain in a proportion of 25.7±2.57% at this same time point. CONCLUSION We have described the synthesis and in vivo evaluation of a novel radioligand for PDE7 imaging. Despite high uptake in the brain and favorable kinetics in vivo, the presence of a brain penetrant radiometabolite makes [(18)F]MICA-003 unfavorable for the accurate quantification of PDE7 and more stable spiroquinazolinones analogs are in development.
Collapse
Affiliation(s)
- David Thomae
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium; Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Stijn Servaes
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Naiara Vazquez
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium; Department of Translational Neurosciences, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium; University Hospital Antwerp, Department of Nuclear Medicine, Edegem, Belgium
| | | | | | - Jurgen Joossens
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium; Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
13
|
Umar T, Hoda N. Selective inhibitors of phosphodiesterases: therapeutic promise for neurodegenerative disorders. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00419e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PDE inhibitors: significant contributors to the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tarana Umar
- Department of Chemistry
- Jamia Millia Islamia
- Central University
- New Delhi
- 110025 India
| | - Nasimul Hoda
- Department of Chemistry
- Jamia Millia Islamia
- Central University
- New Delhi
- 110025 India
| |
Collapse
|
14
|
García AM, Brea J, Morales-García JA, Perez DI, González A, Alonso-Gil S, Gracia-Rubio I, Ros-Simó C, Conde S, Cadavid MI, Loza MI, Perez-Castillo A, Valverde O, Martinez A, Gil C. Modulation of cAMP-specific PDE without emetogenic activity: new sulfide-like PDE7 inhibitors. J Med Chem 2014; 57:8590-607. [PMID: 25264825 DOI: 10.1021/jm501090m] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A forward chemical genetic approach was followed to discover new targets and lead compounds for Parkinson's disease (PD) treatment. By analysis of the cell protection produced by some small molecules, a diphenyl sulfide compound was revealed to be a new phosphodiesterase 7 (PDE7) inhibitor and identified as a new hit. This result allows us to confirm the utility of PDE7 inhibitors as a potential pharmacological treatment of PD. On the basis of these data, a diverse family of diphenyl sulfides has been developed and pharmacologically evaluated in the present work. Moreover, to gain insight into the safety of PDE7 inhibitors for human chronic treatment, we evaluated the new compounds in a surrogate emesis model, showing nonemetic effects.
Collapse
Affiliation(s)
- Ana M García
- Centro de Investigaciones Biológicas (CSIC) , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Martinez A, Gil C. cAMP-specific phosphodiesterase inhibitors: promising drugs for inflammatory and neurological diseases. Expert Opin Ther Pat 2014; 24:1311-21. [PMID: 25284693 DOI: 10.1517/13543776.2014.968127] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION PDEs are key enzymes in the adenosine and guanosine cyclic nucleotides (cAMP and cGMP) signaling cascade. Their inhibition increases cyclic nucleotide levels inside the cell. Thus, pharmacological modulation of PDE activity can have profound effects on the function of cells and organ systems throughout the body. AREAS COVERED Among the large PDE families, only PDE4, PDE7 and PDE8 are cAMP-specific hydrolyzing enzymes. cAMP is an important second messenger not only by its involvement in a vast number of physiological processes but also by activation of protein kinase A, exchange protein activated by cAMP (Epac) and cAMP response element-binding (CREB) or cyclic nucleotide-gated channels. Clearly, such enzymes represent ideal drug targets for the pharmacological treatment of many pathologies. The discovery and development of small molecules targeting cAMP-specific PDEs reported in the last 5 years is the focus of the present review. EXPERT OPINION The first PDE4 inhibitors recently reached the market, having avoided, by different strategies, their dose-limiting side effects (after more than two decades of drug development). Meanwhile, new cAMP-specific PDE7 and PDE8 inhibitors emerged as effective and safe drugs for severe unmet diseases. The therapeutic potential of these inhibitors will be tested in the near future, as many of these drug candidates are ready to start clinical trials.
Collapse
Affiliation(s)
- Ana Martinez
- Centro de Investigaciones Biológicas (CSIC) , Ramiro de Maeztu 9, 28040 Madrid , Spain
| | | |
Collapse
|
16
|
Sánchez AI, Meneses R, Mínguez JM, Núñez A, Castillo RR, Filace F, Burgos C, Vaquero JJ, Álvarez-Builla J, Cortés-Cabrera A, Gago F, Terricabras E, Segarra V. Microwave-assisted synthesis of potent PDE7 inhibitors containing a thienopyrimidin-4-amine scaffold. Org Biomol Chem 2014; 12:4233-42. [DOI: 10.1039/c4ob00175c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thienopyrimidin-4-amines have been synthesized, evaluated and modelled as phosphodiesterase inhibitors.
Collapse
Affiliation(s)
- Ana I. Sánchez
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Ricardo Meneses
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - José M. Mínguez
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Araceli Núñez
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Rafael R. Castillo
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Fabiana Filace
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Carolina Burgos
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Juan J. Vaquero
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Julio Álvarez-Builla
- Departamento de Química Orgánica
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Alvaro Cortés-Cabrera
- Departamento de Ciencias Biomédicas
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | - Federico Gago
- Departamento de Ciencias Biomédicas
- Universidad de Alcalá
- E-28871 Alcalá de Henares, Spain
| | | | - Víctor Segarra
- Almirall-Prodesfarma
- 08980 Sant Feliu de Llobregat, Spain
| |
Collapse
|
17
|
Martinez A, Gil C. Phosphodiesterase Inhibitors as a New Therapeutic Approach for the Treatment of Parkinson’s Disease. EMERGING DRUGS AND TARGETS FOR PARKINSON’S DISEASE 2013. [DOI: 10.1039/9781849737357-00294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Phosphodiesterases (PDEs) are expressed in different brain areas including the striatum. PDEs have recently emerged as important drug targets for central nervous system disorders, including Parkinson’s disease (PD). Levels of cyclic adenosine monophosphate (cAMP) control many cellular signaling pathways and are crucial for the dopamine signal, which is disturbed in PD due to the progressive loss of dopaminergic neurons. PDEs play a key role in cAMP homeostasis, as they are the enzymes responsible for its degradation. Moreover, beyond dopamine neurotransmission, cAMP is involved in many other cellular processes, such as neuroinflammation and neuronal plasticity. This enhances the value of PDEs as promising pharmacological targets for neurological disorders. Furthermore, cAMP‐PDE inhibitors with drug profiles may be used in the near future as disease‐modifying drugs for the treatment of PD. A concise review of the main roles of cAMP‐PDEs expressed in the striatum and the potential of their inhibitors in different animal models of PD is described in this chapter.
Collapse
|
18
|
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]
|
19
|
Perez-Gonzalez R, Pascual C, Antequera D, Bolos M, Redondo M, Perez DI, Pérez-Grijalba V, Krzyzanowska A, Sarasa M, Gil C, Ferrer I, Martinez A, Carro E. Phosphodiesterase 7 inhibitor reduced cognitive impairment and pathological hallmarks in a mouse model of Alzheimer's disease. Neurobiol Aging 2013; 34:2133-45. [PMID: 23582662 DOI: 10.1016/j.neurobiolaging.2013.03.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/10/2013] [Accepted: 03/11/2013] [Indexed: 01/12/2023]
Abstract
Elevated levels of amyloid beta (Aβ) peptide, hyperphosphorylation of tau protein, and inflammation are pathological hallmarks in Alzheimer's disease (AD). Phosphodiesterase 7 (PDE7) regulates the inflammatory response through the cyclic adenosine monophosphate signaling cascade, and thus plays a central role in AD. The aim of this study was to evaluate the efficacy of an inhibitor of PDE7, named S14, in a mouse model of AD. We report that APP/Ps1 mice treated daily for 4 weeks with S14 show: (1) significant attenuation in behavioral impairment; (2) decreased brain Aβ deposition; (3) enhanced astrocyte-mediated Aβ degradation; and (4) decreased tau phosphorylation. These effects are mediated via the cyclic adenosine monophosphate/cyclic adenosine monophosphate response element-binding protein signaling pathway, and inactivation of glycogen synthase kinase (GSK)3. Our data support the use of PDE7 inhibitors, and specifically S14, as effective therapeutic agents for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rocio Perez-Gonzalez
- Neuroscience Group, Instituto de Investigacion Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sánchez AI, Martínez-Barrasa V, Burgos C, Vaquero JJ, Alvarez-Builla J, Terricabras E, Segarra V. Synthesis and evaluation of quinazoline derivatives as phosphodiesterase 7 inhibitors. Bioorg Med Chem 2013; 21:2370-2378. [DOI: 10.1016/j.bmc.2013.01.067] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/24/2013] [Indexed: 11/28/2022]
|
21
|
Jerez MJ, Jerez M, González-García C, Ballester S, Castro A. Combined use of pharmacophoric models together with drug metabolism and genotoxicity "in silico" studies in the hit finding process. J Comput Aided Mol Des 2013; 27:79-90. [PMID: 23296989 DOI: 10.1007/s10822-012-9627-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/15/2012] [Indexed: 01/23/2023]
Abstract
In this study we propose a virtual screening strategy based on the generation of a pharmacophore hypothesis, followed by an in silico evaluation of some ADME-TOX properties with the aim to apply it to the hit finding process and, specifically, to characterize new chemical entities with potential to control inflammatory processes mediated by T lymphocytes such as multiple sclerosis, systemic lupus erithematosus or rheumatoid arthritis. As a result, three compounds with completely novel scaffolds were selected as final hits for future hit-to-lead optimization due to their anti-inflammatory profile. The biological results showed that the selected compounds increased the intracellular cAMP levels and inhibited cell proliferation in T lymphocytes. Moreover, two of these compounds were able to increase the production of IL-4, an immunoregulatory cytokine involved in the selective deviation of T helper (Th) immune response Th type 2 (Th2), which has been proved to have anti-inflammatory properties in several animal models for autoimmune pathologies as multiple sclerosis or rheumatoid arthritis. Thus our pharmacological strategy has shown to be useful to find molecules with biological activity to control immune responses involved in many inflammatory disorders. Such promising data suggested that this in silico strategy might be useful as hit finding process for future drug development.
Collapse
Affiliation(s)
- Ma José Jerez
- Instituto de Química Médica-CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | | | | | | | | |
Collapse
|
22
|
Redondo M, Palomo V, Brea J, Pérez DI, Martín-Álvarez R, Pérez C, Paúl-Fernández N, Conde S, Cadavid MI, Loza MI, Mengod G, Martínez A, Gil C, Campillo NE. Identification in silico and experimental validation of novel phosphodiesterase 7 inhibitors with efficacy in experimental autoimmune encephalomyelitis mice. ACS Chem Neurosci 2012; 3:793-803. [PMID: 23077723 DOI: 10.1021/cn300105c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/08/2012] [Indexed: 12/27/2022] Open
Abstract
A neural network model has been developed to predict the inhibitory capacity of any chemical structure to be a phosphodiesterase 7 (PDE7) inhibitor, a new promising kind of drugs for the treatment of neurological disorders. The numerical definition of the structures was achieved using CODES program. Through the validation of this neural network model, a novel family of 5-imino-1,2,4-thiadiazoles (ITDZs) has been identified as inhibitors of PDE7. Experimental extensive biological studies have demonstrated the ability of ITDZs to inhibit PDE7 and to increase intracellular levels of cAMP. Among them, the derivative 15 showed a high in vitro potency with desirable pharmacokinetic profile (safe genotoxicity and blood brain barrier penetration). Administration of ITDZ 15 in an experimental autoimmune encephalomyelitis (EAE) mouse model results in a significant attenuation of clinical symptoms, showing the potential of ITDZs, especially compound 15, for the effective treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Miriam Redondo
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - Valle Palomo
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - José Brea
- Instituto de Farmacia
Industrial,
Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Universitario Sur s/n, 15782 Santiago de Compostela, Spain
| | - Daniel I. Pérez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - Rocío Martín-Álvarez
- Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS, CIBERNED),
Rosselló 161, 08036 Barcelona, Spain
| | - Concepción Pérez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - Nuria Paúl-Fernández
- Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS, CIBERNED),
Rosselló 161, 08036 Barcelona, Spain
| | - Santiago Conde
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - María Isabel Cadavid
- Instituto de Farmacia
Industrial,
Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Universitario Sur s/n, 15782 Santiago de Compostela, Spain
| | - María Isabel Loza
- Instituto de Farmacia
Industrial,
Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Universitario Sur s/n, 15782 Santiago de Compostela, Spain
| | - Guadalupe Mengod
- Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS, CIBERNED),
Rosselló 161, 08036 Barcelona, Spain
| | - Ana Martínez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - Carmen Gil
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| | - Nuria E. Campillo
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid,
Spain
| |
Collapse
|
23
|
Imidazopyridazinones as novel PDE7 inhibitors: SAR and in vivo studies in Parkinson’s disease model. Bioorg Med Chem Lett 2012; 22:6286-91. [DOI: 10.1016/j.bmcl.2012.07.077] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/02/2012] [Accepted: 07/24/2012] [Indexed: 12/13/2022]
|
24
|
González M, Cerecetto H. Quinoxaline derivatives: a patent review (2006--present). Expert Opin Ther Pat 2012; 22:1289-302. [PMID: 22971178 DOI: 10.1517/13543776.2012.724677] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Quinoxaline scaffold is included in a large number of therapeutic agents because of its physicochemical properties that make the difference between them and the carbono analogue, naphthalene. AREAS COVERED This review of patented products presents the quinoxaline heterocycle as part of the structural patent claims from a medicinal chemistry perspective. EXPERT OPINION We centred our discussion in the various drug patent applications of the quinoxaline and its derivatives. The applications are based firstly in the specific enzyme target with very low development in the disease treatment. Only for cancer and antimicrobial agents they were specifically determined but little is mentioned in order to insight in the last development activities.
Collapse
Affiliation(s)
- Mercedes González
- Grupo de Química Medicinal, Laboratorio de Química Orgánica-Instituto de Química Biológica-Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo 11400, Uruguay.
| | | |
Collapse
|
25
|
Banerjee A, Yadav PS, Bajpai M, Sangana RR, Gullapalli S, Gudi GS, Gharat LA. Isothiazole and isoxazole fused pyrimidones as PDE7 inhibitors: SAR and pharmacokinetic evaluation. Bioorg Med Chem Lett 2012; 22:3223-8. [DOI: 10.1016/j.bmcl.2012.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 02/28/2012] [Accepted: 03/07/2012] [Indexed: 01/01/2023]
|
26
|
Pérez DI, Pistolozzi M, Palomo V, Redondo M, Fortugno C, Gil C, Felix G, Martinez A, Bertucci C. 5-Imino-1,2-4-thiadiazoles and quinazolines derivatives as glycogen synthase kinase 3β (GSK-3β) and phosphodiesterase 7 (PDE7) inhibitors: Determination of blood–brain barrier penetration and binding to human serum albumin. Eur J Pharm Sci 2012; 45:677-84. [DOI: 10.1016/j.ejps.2012.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 01/11/2012] [Indexed: 10/14/2022]
|
27
|
Redondo M, Brea J, Perez DI, Soteras I, Val C, Perez C, Morales-García JA, Alonso-Gil S, Paul-Fernandez N, Martin-Alvarez R, Cadavid MI, Loza MI, Perez-Castillo A, Mengod G, Campillo NE, Martinez A, Gil C. Effect of phosphodiesterase 7 (PDE7) inhibitors in experimental autoimmune encephalomyelitis mice. Discovery of a new chemically diverse family of compounds. J Med Chem 2012; 55:3274-84. [PMID: 22385507 DOI: 10.1021/jm201720d] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Phosphodiesterase (PDE) 7 is involved in proinflammatory processes, being widely expressed both on lymphocytes and on certain brain regions. Specific inhibitors of PDE7 have been recently reported as potential new drugs for the treatment of neurological disorders because of their ability to increase intracellular levels of cAMP and thus to modulate the inflammatory process, as a neuroprotective well-established strategy. Multiple sclerosis is an unmet disease in which pathologies on the immune system, T-cells, and specific neural cells are involved simultaneously. Therefore, PDE7 inhibitors able to interfere with all these targets may represent an innovative therapy for this pathology. Here, we report a new chemically diverse family of heterocyclic PDE7 inhibitors, discovered and optimized by using molecular modeling studies, able to increase cAMP levels in cells, decrease inflammatory activation on primary neural cultures, and also attenuate the clinical symptoms in the experimental autoimmune encephalomyelitis (EAE) mouse model. These results led us to propose the use of PDE7 inhibitors as innovative therapeutic agents for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Miriam Redondo
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Redondo M, Zarruk JG, Ceballos P, Pérez DI, Pérez C, Perez-Castillo A, Moro MA, Brea J, Val C, Cadavid MI, Loza MI, Campillo NE, Martínez A, Gil C. Neuroprotective efficacy of quinazoline type phosphodiesterase 7 inhibitors in cellular cultures and experimental stroke model. Eur J Med Chem 2012; 47:175-85. [DOI: 10.1016/j.ejmech.2011.10.040] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 10/18/2011] [Accepted: 10/21/2011] [Indexed: 12/31/2022]
|
29
|
Gewald R, Rueger C, Grunwald C, Egerland U, Hoefgen N. Synthesis and structure–activity relationship studies of dihydronaphthyridinediones as a novel structural class of potent and selective PDE7 inhibitors. Bioorg Med Chem Lett 2011; 21:6652-6. [DOI: 10.1016/j.bmcl.2011.09.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/14/2011] [Accepted: 09/16/2011] [Indexed: 10/17/2022]
|
30
|
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
|
31
|
Giembycz MA, Newton R. Harnessing the clinical efficacy of phosphodiesterase 4 inhibitors in inflammatory lung diseases: dual-selective phosphodiesterase inhibitors and novel combination therapies. Handb Exp Pharmacol 2011:415-446. [PMID: 21695651 DOI: 10.1007/978-3-642-17969-3_18] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Phosphodiesterase (PDE) 4 inhibitors have been in development as a novel anti-inflammatory therapy for more than 20 years, with asthma and chronic obstructive pulmonary disease (COPD) being primary indications. Despite initial optimism, only one selective PDE4 inhibitor, roflumilast (Daxas (®)), has been approved for use in humans and available in Canada and the European Union in 2011 for the treatment of a specific population of patients with severe COPD. In many other cases, the development of PDE4 inhibitors of various structural classes has been discontinued due to lack of efficacy and/or dose-limiting adverse events. Indeed, for many of these compounds, it is likely that the maximum tolerated dose is either subtherapeutic or at the very bottom of the efficacy dose-response curve. Thus, a significant ongoing challenge that faces the pharmaceutical industry is to synthesize compounds with therapeutic ratios that are superior to roflumilast. Several strategies are being considered, but clinically effective compounds with an optimal pharmacophore have not, thus far, been reported. In this chapter, alternative means of harnessing the clinical efficacy of PDE4 inhibitors are described. These concepts are based on the assumption that additive or synergistic anti-inflammatory effects can be produced with inhibitors that target either two or more PDE families or with a PDE4 inhibitor in combination with other anti-inflammatory drugs such as a glucocorticoid.
Collapse
Affiliation(s)
- Mark A Giembycz
- Airways Inflammation Research Group, Departments of Physiology and Pharmacology, Institute of Infection, Immunity and Inflammation, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| | | |
Collapse
|
32
|
Bales KR, Plath N, Svenstrup N, Menniti FS. Phosphodiesterase Inhibition to Target the Synaptic Dysfunction in Alzheimer’s Disease. TOPICS IN MEDICINAL CHEMISTRY 2010. [DOI: 10.1007/7355_2010_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
33
|
Castaño T, Wang H, Campillo NE, Ballester S, González-García C, Hernández J, Pérez C, Cuenca J, Pérez-Castillo A, Martínez A, Huertas O, Gelpí JL, Luque FJ, Ke H, Gil C. Synthesis, structural analysis, and biological evaluation of thioxoquinazoline derivatives as phosphodiesterase 7 inhibitors. ChemMedChem 2009; 4:866-76. [PMID: 19350606 PMCID: PMC2952885 DOI: 10.1002/cmdc.200900043] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Indexed: 01/21/2023]
Abstract
PDE7 inhibitors regulate pro-inflammatory and immune T-cell functions, and are a potentially novel class of drugs especially useful in the treatment of a wide variety of immune and inflammatory disorders. Starting from our lead family of thioxoquinazolines, we designed, synthesized, and characterized a novel series of thioxoquinazoline derivatives. Many of these compounds showed inhibitory potencies at sub-micromolar levels against the catalytic domain of PDE7A1 and at the micromolar level against PDE4D2. Cell-based studies showed that these compounds not only increased intracellular cAMP levels, but also had interesting anti-inflammatory properties within a therapeutic window. The in silico data predict that these compounds are capable of the crossing the blood-brain barrier. The X-ray crystal structure of the PDE7A1 catalytic domain in complex with compound 15 at a resolution of 2.4 A demonstrated that hydrophobic interactions at the active site pocket are a key feature. This structure, together with molecular modeling, provides insight into the selectivity of the PDE inhibitors and a template for the discovery of new PDE7 or PDE7/PDE4 dual inhibitors.
Collapse
Affiliation(s)
- Tania Castaño
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid (Spain)
| | - Huanchen Wang
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260 (USA)
| | - Nuria E. Campillo
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid (Spain)
| | - Sara Ballester
- Unidad de Regulación Génica, CNM Instituto de Salud Carlos III, Madrid (Spain)
| | | | - Javier Hernández
- Unidad de Regulación Génica, CNM Instituto de Salud Carlos III, Madrid (Spain)
| | - Concepción Pérez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid (Spain)
| | - Jimena Cuenca
- Instituto de Investigaciones Biomédicas (CSIC-UAM) Arturo Duperier 4, 28029 Madrid (Spain)
| | - Ana Pérez-Castillo
- Instituto de Investigaciones Biomédicas (CSIC-UAM) Arturo Duperier 4, 28029 Madrid (Spain)
| | - Ana Martínez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid (Spain)
| | - Oscar Huertas
- Departamento de Fisicoquímica e Instituto de Biomedicina (IBUB) Facultad de Farmacia, Universidad de Barcelona, Avda. Diagonal 643, 08028 Barcelona (Spain)
| | - José Luis Gelpí
- Departamento de Bioquímica y Biología Molecular Facultad de Bioquímica, Universidad de Barcelona Avda. Diagonal 643, 08028 Barcelona (Spain)
| | - F. Javier Luque
- Departamento de Fisicoquímica e Instituto de Biomedicina (IBUB) Facultad de Farmacia, Universidad de Barcelona, Avda. Diagonal 643, 08028 Barcelona (Spain)
| | - Hengming Ke
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599-7260 (USA)
| | - Carmen Gil
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid (Spain)
| |
Collapse
|