1
|
Mota É, Bompierre S, Betolngar D, Castro LRV, Vincent P. Pivotal role of phosphodiesterase 10A in the integration of dopamine signals in mice striatal D1 and D2 medium-sized spiny neurones. Br J Pharmacol 2021; 178:4873-4890. [PMID: 34399440 DOI: 10.1111/bph.15664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/21/2021] [Accepted: 06/17/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Dopamine in the striatum plays a crucial role in reward processes and action selection. Dopamine signals are transduced by D1 and D2 dopamine receptors which trigger mirror effects through the cAMP/PKA signalling cascade in D1 and D2 medium-sized spiny neurones (MSNs). Phosphodiesterases (PDEs), which determine the profile of cAMP signals, are highly expressed in MSNs, but their respective roles in dopamine signal integration remain poorly understood. EXPERIMENTAL APPROACH We used genetically-encoded FRET biosensors to monitor at the single cell level the functional contribution of PDE2A, PDE4 and PDE10A in the changes of the cAMP/PKA response to transient and continuous dopamine in mouse striatal brain slices. KEY RESULTS We found that PDE2A, PDE4 and PDE10A operate on the moderate to high cAMP levels elicited by D1 or A2A receptor stimulation. In contrast, only PDE10A is able to reduce cAMP down to baseline in both type of neurones, leading to the dephosphorylation of PKA substrates. CONCLUSION AND IMPLICATIONS In both MSN types, PDE10A inhibition blunts the responsiveness to dopamine, whereas PDE2A or PDE4 inhibition reinforces dopamine action.
Collapse
Affiliation(s)
- Élia Mota
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, UMR 8256, Paris, France.,Now at Novel Human Genetics Research Unit, GSK R&D, Stevenage, UK
| | - Ségolène Bompierre
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, UMR 8256, Paris, France
| | - Dahdjim Betolngar
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, UMR 8256, Paris, France
| | - Liliana R V Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, UMR 8256, Paris, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, UMR 8256, Paris, France
| |
Collapse
|
2
|
Moszczyński-Pętkowski R, Majer J, Borkowska M, Bojarski Ł, Janowska S, Matłoka M, Stefaniak F, Smuga D, Bazydło K, Dubiel K, Wieczorek M. Synthesis and characterization of novel classes of PDE10A inhibitors - 1H-1,3-benzodiazoles and imidazo[1,2-a]pyrimidines. Eur J Med Chem 2018; 155:96-116. [PMID: 29870883 DOI: 10.1016/j.ejmech.2018.05.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 10/14/2022]
Abstract
New compounds containing [1,2,4]triazolo [1,5-a]pyridine (I), pyrazolo [1,5-a]pyridine (II), 1H-1,3-benzodiazole (III) and imidazo [1,2-a]pyrimidine (IV) backbones were designed and synthesized for PDE10A interaction. Among these compounds, 1H-1,3-benzodiazoles and imidazo [1,2-a]pyrimidines showed the highest affinity for PDE10A enzyme as well as good metabolic stability. Both classes of compounds were identified as selective and potent PDE10A enzyme inhibitors.
Collapse
Affiliation(s)
| | - Jakub Majer
- Celon Pharma S.A., Medicinal Chemistry Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Małgorzata Borkowska
- Celon Pharma S.A., Medicinal Chemistry Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Łukasz Bojarski
- Celon Pharma S.A., Innovative Drugs Research and Development Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Sylwia Janowska
- Celon Pharma S.A., Innovative Drugs Research and Development Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Mikołaj Matłoka
- Celon Pharma S.A., Innovative Drugs Research and Development Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Filip Stefaniak
- Celon Pharma S.A., Medicinal Chemistry Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Damian Smuga
- Celon Pharma S.A., Medicinal Chemistry Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Katarzyna Bazydło
- Celon Pharma S.A., Innovative Drugs Research and Development Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Krzysztof Dubiel
- Celon Pharma S.A., Medicinal Chemistry Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| | - Maciej Wieczorek
- Celon Pharma S.A., Innovative Drugs Research and Development Department, Mokra 41a, Kiełpin, 05-092, Łomianki, Poland
| |
Collapse
|
3
|
Peng T, Gong J, Jin Y, Zhou Y, Tong R, Wei X, Bai L, Shi J. Inhibitors of phosphodiesterase as cancer therapeutics. Eur J Med Chem 2018; 150:742-756. [PMID: 29574203 DOI: 10.1016/j.ejmech.2018.03.046] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/26/2018] [Accepted: 03/16/2018] [Indexed: 01/05/2023]
Abstract
Phosphodiesterases (PDEs) are a class of enzymes that hydrolyze cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which is involved in many physiological processes including visual transduction, cell proliferation and differentiation, cell-cycle regulation, gene expression, inflammation, apoptosis, and metabolic function. PDEs are composed of 11 different families and each family contains different subtypes. The distribution, expression, regulation mode and sensitivity to inhibitors of each subtype are different, and they are involved in cancer, inflammation, asthma, depression, erectile dysfunction and other pathological processes of development. A large number of studies have shown that PDEs play an important role in the development of tumors by affecting the intracellular level of cAMP and/or cGMP and PDEs could become diagnostic markers or therapeutic targets. This review will give a brief overview of the expression and regulation of PDE families in the process of tumorigenesis and their anti-tumor inhibitors, which may guide the design of novel therapeutic drugs targeting PDEs for anticancer agent.
Collapse
Affiliation(s)
- Ting Peng
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun Gong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yongzhe Jin
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanping Zhou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xin Wei
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
4
|
Castro L, Yapo C, Vincent P. [Physiopathology of cAMP/PKA signaling in neurons]. Biol Aujourdhui 2017; 210:191-203. [PMID: 28327278 DOI: 10.1051/jbio/2017005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Indexed: 11/15/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) and the cyclic-AMP dependent protein kinase (PKA) regulate a plethora of cellular functions in virtually all eukaryotic cells. In neurons, the cAMP/PKA signaling cascade controls a number of biological properties such as axonal growth, synaptic transmission, regulation of excitability or long term changes in the nucleus. Genetically-encoded optical biosensors for cAMP or PKA considerably improved our understanding of these processes by providing a real-time measurement in living neurons. In this review, we describe the recent progresses made in the creation of biosensors for cAMP or PKA activity. These biosensors revealed profound differences in the amplitude of the cAMP signal evoked by neuromodulators between various neuronal preparations. These responses can be resolved at the level of individual neurons, also revealing differences related to the neuronal type. At the subcellular level, biosensors reported different signal dynamics in domains like dendrites, cell body, nucleus and axon. Combining this imaging approach with pharmacology or genetical models points at phosphodiesterases and phosphatases as critical regulatory proteins. Biosensor imaging will certainly help understand the mechanism of action of current drugs as well as help in devising novel therapeutic strategies for neuropsychiatric diseases.
Collapse
|
5
|
Development of highly potent phosphodiesterase 10A (PDE10A) inhibitors: Synthesis and in vitro evaluation of 1,8-dipyridinyl- and 1-pyridinyl-substituted imidazo[1,5-a]quinoxalines. Eur J Med Chem 2015; 107:97-108. [PMID: 26562545 DOI: 10.1016/j.ejmech.2015.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022]
Abstract
Herein we report the synthesis of fluorinated inhibitors of phosphodiesterase 10A (PDE10A) which can be used potentially as lead structure for the development of a (18)F-labeled PDE10A imaging agent for positron emission tomography. The use of ortho-fluoropyridines as residues could potentially enable the introduction of (18)F through nucleophilic substitution for radiolabeling purposes. 2-Fluoropyridines are introduced by a Suzuki coupling at different positions of the molecule. The reference compounds, 1,8-dipyridinylimidazo[1,5-a]quinoxalines and 1-pyridinylimidazo[1,5-a]quinoxalines, show inhibitory potencies at best in the subnanomolar range and selectivity factors greater than 38 against other PDE's. 1,8-Dipyridinylimidazo[1,5-a]quinoxalines are more potent inhibitors than 1-pyridinylimidazo[1,5-a]quinoxalines. Using 2-fluoro-3-pyridinyl as residue provided the most potent inhibitors 16 (IC50 = 0.12 nM), 17 (IC50 = 0.048 nM) and 32 (IC50 = 0.037 nM).
Collapse
|
6
|
Vlasceanu A, Jessing M, Kilburn JP. BN/CC isosterism in borazaronaphthalenes towards phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem 2015; 23:4453-4461. [DOI: 10.1016/j.bmc.2015.06.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/01/2015] [Accepted: 06/05/2015] [Indexed: 10/23/2022]
|
7
|
Kjellgren ER, Glue OES, Reinholdt P, Meyer JE, Kongsted J, Poongavanam V. A comparative study of binding affinities for 6,7-dimethoxy-4-pyrrolidylquinazolines as phosphodiesterase 10A inhibitors using the linear interaction energy method. J Mol Graph Model 2015; 61:44-52. [PMID: 26188794 DOI: 10.1016/j.jmgm.2015.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/05/2015] [Accepted: 06/20/2015] [Indexed: 01/29/2023]
Abstract
The linear interaction energy (LIE) method was used to estimate the free energies of binding for a set of 27 pyrrolidylquinazoline derivatives as phosphodiesterase 10A inhibitors. Twenty-six X-ray crystal structures of phosphodiesterase 10A and two sampling methods, minimization and Hybrid Monte Carlo, were used to assess the affinity models based on the linear interaction energies. The best model was obtained based on the parameters α=0.16 and β=0.04, which represent non-polar and polar interactions, respectively, with a root mean square error (RMSE) of 0.42kcal/mol (R(2)=0.71) and 0.52kcal/mol (R(2)=0.86) for the training and test sets, respectively. In addition, the applicability domain of the model was investigated. After validation of the models, the best model was subsequently used in a virtual screening process, which resulted in a set of optimized compounds. The models developed in this study could be useful as filter for virtual screening and lead optimization processes for phosphodiesterase 10A drug developments.
Collapse
Affiliation(s)
- Erik Rosendahl Kjellgren
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Oliver Emil Skytte Glue
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Peter Reinholdt
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Julie Egeskov Meyer
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | | |
Collapse
|
8
|
Bartolomé-Nebreda JM, Alonso de Diego SA, Artola M, Delgado F, Delgado Ó, Martín-Martín ML, Martínez-Viturro CM, Pena MÁ, Tong HM, Van Gool M, Alonso JM, Fontana A, Macdonald GJ, Megens A, Langlois X, Somers M, Vanhoof G, Conde-Ceide S. Identification of a Novel Orally Bioavailable Phosphodiesterase 10A (PDE10A) Inhibitor with Efficacy in Animal Models of Schizophrenia. J Med Chem 2015; 58:978-93. [DOI: 10.1021/jm501651a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- José Manuel Bartolomé-Nebreda
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Sergio A. Alonso de Diego
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Marta Artola
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Francisca Delgado
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Óscar Delgado
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - María Luz Martín-Martín
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Carlos M. Martínez-Viturro
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Miguel Ángel Pena
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Han Min Tong
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Michiel Van Gool
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - José Manuel Alonso
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Alberto Fontana
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Gregor J. Macdonald
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Anton Megens
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Xavier Langlois
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Marijke Somers
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Greet Vanhoof
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Susana Conde-Ceide
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Chemistry, Janssen Research & Development, Calle Jarama 75, Polígono Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Neuroscience Biology, ⊥Discovery Sciences ADME/Tox, and #Discovery Sciences Cellular Pharmacology, Janssen Research & Development, Turnhoutseweg 30, B-2340, Beerse, Belgium
| |
Collapse
|
9
|
Dore A, Asproni B, Scampuddu A, Pinna GA, Christoffersen CT, Langgård M, Kehler J. Synthesis and SAR study of novel tricyclic pyrazoles as potent phosphodiesterase 10A inhibitors. Eur J Med Chem 2014; 84:181-93. [DOI: 10.1016/j.ejmech.2014.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 06/17/2014] [Accepted: 07/06/2014] [Indexed: 10/25/2022]
|
10
|
Hu E, Chen N, Bourbeau MP, Harrington PE, Biswas K, Kunz RK, Andrews KL, Chmait S, Zhao X, Davis C, Ma J, Shi J, Lester-Zeiner D, Danao J, Able J, Cueva M, Talreja S, Kornecook T, Chen H, Porter A, Hungate R, Treanor J, Allen JR. Discovery of clinical candidate 1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)ethanone (AMG 579), a potent, selective, and efficacious inhibitor of phosphodiesterase 10A (PDE10A). J Med Chem 2014; 57:6632-41. [PMID: 25062128 DOI: 10.1021/jm500713j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We report the identification of a PDE10A clinical candidate by optimizing potency and in vivo efficacy of promising keto-benzimidazole leads 1 and 2. Significant increase in biochemical potency was observed when the saturated rings on morpholine 1 and N-acetyl piperazine 2 were changed by a single atom to tetrahydropyran 3 and N-acetyl piperidine 5. A second single atom modification from pyrazines 3 and 5 to pyridines 4 and 6 improved the inhibitory activity of 4 but not 6. In the in vivo LC-MS/MS target occupancy (TO) study at 10 mg/kg, 3, 5, and 6 achieved 86-91% occupancy of PDE10A in the brain. Furthermore, both CNS TO and efficacy in PCP-LMA behavioral model were observed in a dose dependent manner. With superior in vivo TO, in vivo efficacy and in vivo PK profiles in multiple preclinical species, compound 5 (AMG 579) was advanced as our PDE10A clinical candidate.
Collapse
Affiliation(s)
- Essa Hu
- Department of Medicinal Chemistry, ‡Department of Molecular Structure and Characterization, §Department of Pharmacokinetics and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc. , One Amgen Center Drive, Thousand Oaks, California 93012-1799, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Das S, Shelke DE, Harde RL, Avhad VB, Khairatkar-Joshi N, Gullapalli S, Gupta PK, Gandhi MN, Bhateja DK, Bajpai M, Joshi AA, Marathe MY, Gudi GS, Jadhav SB, Mahat MYA, Thomas A. Design, synthesis and pharmacological evaluation of novel polycyclic heteroarene ethers as PDE10A inhibitors: Part II. Bioorg Med Chem Lett 2014; 24:3238-42. [DOI: 10.1016/j.bmcl.2014.06.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/03/2014] [Accepted: 06/10/2014] [Indexed: 01/02/2023]
|
12
|
Megens AAHP, Hendrickx HMR, Mahieu MMA, Wellens ALY, de Boer P, Vanhoof G. PDE10A inhibitors stimulate or suppress motor behavior dependent on the relative activation state of the direct and indirect striatal output pathways. Pharmacol Res Perspect 2014; 2:e00057. [PMID: 25505601 PMCID: PMC4186443 DOI: 10.1002/prp2.57] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/09/2014] [Indexed: 11/09/2022] Open
Abstract
The enzyme phosphodiesterase 10A (PDE10A) regulates the activity of striatal, medium spiny neurons (MSNs), which are divided into a behaviorally stimulating, Gs-coupled D1 receptor-expressing “direct” pathway and a behaviorally suppressant, Gi-coupled D2 receptor-expressing “indirect” pathway. Activating both pathways, PDE10A inhibitors (PDE10AIs) combine functional characteristics of D2 antagonists and D1 agonists. While the effects of PDE10AIs on spontaneous and stimulated behavior have been extensively reported, the present study investigates their effects on suppressed behavior under various conditions of reduced dopaminergic neurotransmission: blockade of D1 receptors with SCH-23390, blockade of D2 receptors with haloperidol, or depletion of dopamine with RO-4-1284 or reserpine. In rats, PDE10AIs displayed relatively low cataleptic activity per se. After blocking D1 receptors, however, they induced pronounced catalepsy at low doses close to those required for inhibition of apomorphine-induced behavior; slightly higher doses resulted in behavioral stimulant effects, counteracting the catalepsy. PDE10AIs also counteracted catalepsy and related behaviors induced by D2 receptor blockade or dopamine depletion; catalepsy was replaced by behavioral stimulant effects under the latter but not the former condition. Similar interactions were observed at the level of locomotion in mice. At doses close to those inhibiting d-amphetamine-induced hyperlocomotion, PDE10AIs reversed hypolocomotion induced by D1 receptor blockade or dopamine depletion but not hypolocomotion induced by D2 receptor blockade. It is concluded that PDE10AIs stimulate or inhibit motor behavior dependent on the relative activation state of the direct and indirect striatal output pathways.
Collapse
Affiliation(s)
- Anton A H P Megens
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Herman M R Hendrickx
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Michel M A Mahieu
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Annemie L Y Wellens
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Peter de Boer
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Greet Vanhoof
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| |
Collapse
|
13
|
Bartolomé-Nebreda JM, Delgado F, Martín-Martín ML, Martínez-Viturro CM, Pastor J, Tong HM, Iturrino L, Macdonald GJ, Sanderson W, Megens A, Langlois X, Somers M, Vanhoof G, Conde-Ceide S. Discovery of a Potent, Selective, and Orally Active Phosphodiesterase 10A Inhibitor for the Potential Treatment of Schizophrenia. J Med Chem 2014; 57:4196-212. [DOI: 10.1021/jm500073h] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- José Manuel Bartolomé-Nebreda
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Francisca Delgado
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - María Luz Martín-Martín
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Carlos M. Martínez-Viturro
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Joaquín Pastor
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Han Min Tong
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Laura Iturrino
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Gregor J. Macdonald
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Wendy Sanderson
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Anton Megens
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Xavier Langlois
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Marijke Somers
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Greet Vanhoof
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| | - Susana Conde-Ceide
- Neuroscience Medicinal Chemistry and ‡Discovery Sciences Analytical Sciences, Janssen Research & Development, Calle Jarama 75, Polígono
Industrial, Toledo 45007, Spain
- Neuroscience Medicinal Chemistry, ∥Discovery Sciences
Molecular Informatics, ⊥Neuroscience Biology, #Discovery Sciences ADME/Tox, and ∇Discovery Sciences
Translational Sciences, Janssen Research & Development, Turnhoutseweg
30, B-2340, Beerse, Belgium
| |
Collapse
|
14
|
Design, synthesis and pharmacological evaluation of novel polycyclic heteroarene ethers as PDE10A inhibitors: Part I. Bioorg Med Chem Lett 2014; 24:2073-8. [DOI: 10.1016/j.bmcl.2014.03.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/06/2014] [Accepted: 03/18/2014] [Indexed: 01/26/2023]
|
15
|
Megens AAHP, Hendrickx HMR, Hens KA, Fonteyn I, Langlois X, Lenaerts I, Somers MVF, de Boer P, Vanhoof G. Pharmacology of JNJ-42314415, a centrally active phosphodiesterase 10A (PDE10A) inhibitor: a comparison of PDE10A inhibitors with D2 receptor blockers as potential antipsychotic drugs. J Pharmacol Exp Ther 2014; 349:138-54. [PMID: 24421319 DOI: 10.1124/jpet.113.211904] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The new phosphodiesterase 10A inhibitor (PDE10AI) JNJ-42314415 [3-[6-(2-methoxyethyl)pyridin-3-yl]-2-methyl-8-morpholin-4-ylimidazo[1,2-a]pyrazine] was compared with three reference PDE10AIs and eight dopamine 2 (D(2)) receptor blockers. Despite displaying relatively low PDE10A activity in vitro, JNJ-42314415 was found to be a relatively potent and specific PDE10AI in vivo. The compound was devoid of effects on prolactin release and of receptor interactions associated with other commonly observed adverse effects of available antipsychotics. Similar to D(2) receptor blockers, the tested PDE10AIs antagonized stimulant-induced behavior and inhibited conditioned avoidance behavior; these effects were observed at doses close to the ED(50) for striatal PDE10A occupancy. Relative to the ED(50) for inhibition of apomorphine-induced stereotypy, PDE10AIs blocked conditioned avoidance behavior and behaviors induced by nondopaminergic stimulants (phencyclidine, scopolamine) more efficiently than did D(2) receptor blockers; however, they blocked behaviors induced by dopaminergic stimulants (apomorphine, d-amphetamine) less efficiently. PDE10AIs also induced less pronounced catalepsy than D(2) receptor blockers. The effects of PDE10A inhibition against dopaminergic stimulants and on catalepsy were potentiated by the D(1) antagonist SCH-23390 (8-chloro-3-methyl-5-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-ol), suggesting that enhancement of D(1) receptor-mediated neurotransmission contributes to the behavioral profile of PDE10AIs. By reducing dopamine D(2) and concomitantly potentiating dopamine D(1) receptor-mediated neurotransmission, PDE10AIs may show antipsychotic activity with an improved side-effect profile relative to D(2) receptor blockers. However, the clinical implications of this dual mechanism must be further explored.
Collapse
Affiliation(s)
- Anton A H P Megens
- Janssen Research & Development, a Division of Janssen Pharmaceutica, NV Beerse, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Uthayathas S, Masilamoni GJ, Shaffer CL, Schmidt CJ, Menniti FS, Papa SM. Phosphodiesterase 10A inhibitor MP-10 effects in primates: comparison with risperidone and mechanistic implications. Neuropharmacology 2014; 77:257-67. [PMID: 24490227 PMCID: PMC3934827 DOI: 10.1016/j.neuropharm.2013.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Phosphodiesterase 10A (PDE10A) is highly expressed in striatal medium spiny neurons of both the direct and indirect output pathways. Similar to dopamine D₂ receptor antagonists acting on indirect pathway neurons, PDE10A inhibitors have shown behavioral effects in rodent models that predict antipsychotic efficacy. These findings have supported the clinical investigation of PDE10A inhibitors as a new treatment for schizophrenia. However, PDE10A inhibitors and D₂ antagonists differ in effects on direct pathway and other neurons of the basal ganglia, indicating that these two drug classes may have divergent antipsychotic efficacy and side effect profile. In the present study, we compare the behavioral effects of the selective PDE10A inhibitor MP-10 to those of the clinical standard D₂ antagonist risperidone in rhesus monkeys using a standardized motor disability scale for parkinsonian primates and a newly designed "Drug Effects on Nervous System" scale to assess non-motor effects. Behavioral effects of MP-10 correlated with its plasma levels and its regulation of metabolic activity in striatal and cortical regions as measured by FDG-PET imaging. While MP-10 and risperidone broadly impacted similar behavioral domains in the primate, their effects had a different underlying basis. MP-10-treated animals retained the ability to respond but did not engage tasks, whereas risperidone-treated animals retained the motivation to respond but were unable to perform the intended actions. These findings are discussed in light of what is currently known about the modulation of striatal circuitry by these two classes of compounds, and provide insight into interpreting emerging clinical data with PDE10A inhibitors for the treatment of psychotic symptoms.
Collapse
Affiliation(s)
- Subramaniam Uthayathas
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Gunasingh J. Masilamoni
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Christopher L. Shaffer
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research and Development, Pfizer Inc., Cambridge, MA
| | - Christopher J. Schmidt
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, MA
| | | | - Stella M. Papa
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA
- Department of Neurology, Emory University, Atlanta, GA
| |
Collapse
|
17
|
Hu E, Kunz RK, Chen N, Rumfelt S, Siegmund A, Andrews K, Chmait S, Zhao S, Davis C, Chen H, Lester-Zeiner D, Ma J, Biorn C, Shi J, Porter A, Treanor J, Allen JR. Design, Optimization, and Biological Evaluation of Novel Keto-Benzimidazoles as Potent and Selective Inhibitors of Phosphodiesterase 10A (PDE10A). J Med Chem 2013; 56:8781-92. [DOI: 10.1021/jm401234w] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Essa Hu
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Roxanne K. Kunz
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Ning Chen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Shannon Rumfelt
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Aaron Siegmund
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Kristin Andrews
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Samer Chmait
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Sharon Zhao
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Carl Davis
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Hang Chen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Dianna Lester-Zeiner
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Ji Ma
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Christopher Biorn
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Jianxia Shi
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Amy Porter
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - James Treanor
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Jennifer R. Allen
- Department of Medicinal Chemistry, ‡Department of Molecular
Structure, §Department of Pharmacokinetics
and Drug Metabolism, ∥Department of Neuroscience, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
- Department of Neuroscience and #Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
18
|
Kilburn JP, Kehler J, Langgård M, Erichsen MN, Leth-Petersen S, Larsen M, Christoffersen CT, Nielsen J. N-Methylanilide and N-methylbenzamide derivatives as phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem 2013; 21:6053-62. [DOI: 10.1016/j.bmc.2013.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 10/26/2022]
|
19
|
Li J, Jin H, Zhou H, Rothfuss J, Tu Z. Synthesis and in vitro biological evaluation of pyrazole group-containing analogues for PDE10A. MEDCHEMCOMM 2013; 4:443-449. [PMID: 23585921 DOI: 10.1039/c2md20239e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Twenty eight new analogues were synthesized by optimizing the structure of MP-10 and their in vitro binding affinities towards PDE10A, PDE3A/B, and PDE4A/B were determined. Among these new analogues, 10a, 10b, 10d, 11a, 11b and 11d are very potent towards PDE10A and have IC50 values of 0.40 ± 0.02, 0.28 ± 0.06, 1.82 ± 0.25, 0.24 ± 0.05, 0.36 ± 0.03 and 1.78 ± 0.03 nM respectively; these six compounds displayed high selectivity for PDE10A versus PDE3A/3B/4A/4B. The promising compounds will be further validated in vivo to identify PDE10A imaging tracers.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA; ; Tel: +1-314-362-8487
| | | | | | | | | |
Collapse
|
20
|
|
21
|
Hu E, Kunz RK, Rumfelt S, Andrews KL, Li C, Hitchcock SA, Lindstrom M, Treanor J. Use of structure based design to increase selectivity of pyridyl-cinnoline phosphodiesterase 10A (PDE10A) inhibitors against phosphodiesterase 3 (PDE3). Bioorg Med Chem Lett 2012; 22:6938-42. [DOI: 10.1016/j.bmcl.2012.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 09/01/2012] [Accepted: 09/04/2012] [Indexed: 12/31/2022]
|
22
|
Hu E, Ma J, Biorn C, Lester-Zeiner D, Cho R, Rumfelt S, Kunz RK, Nixey T, Michelsen K, Miller S, Shi J, Wong J, Hill Della Puppa G, Able J, Talreja S, Hwang DR, Hitchcock SA, Porter A, Immke D, Allen JR, Treanor J, Chen H. Rapid identification of a novel small molecule phosphodiesterase 10A (PDE10A) tracer. J Med Chem 2012; 55:4776-87. [PMID: 22548439 DOI: 10.1021/jm3002372] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A radiolabeled tracer for imaging therapeutic targets in the brain is a valuable tool for lead optimization in CNS drug discovery and for dose selection in clinical development. We report the rapid identification of a novel phosphodiesterase 10A (PDE10A) tracer candidate using a LC-MS/MS technology. This structurally distinct PDE10A tracer, AMG-7980 (5), has been shown to have good uptake in the striatum (1.2% ID/g tissue), high specificity (striatum/thalamus ratio of 10), and saturable binding in vivo. The PDE10A affinity (K(D)) and PDE10A target density (B(max)) were determined to be 0.94 nM and 2.3 pmol/mg protein, respectively, using [(3)H]5 on rat striatum homogenate. Autoradiography on rat brain sections indicated that the tracer signal was consistent with known PDE10A expression pattern. The specific binding of [(3)H]5 to rat brain was blocked by another structurally distinct, published PDE10A inhibitor, MP-10. Lastly, our tracer was used to measure in vivo PDE10A target occupancy of a PDE10A inhibitor in rats using LC-MS/MS technology.
Collapse
Affiliation(s)
- Essa Hu
- Department of Small Molecule Chemistry, Amgen Inc. , One Amgen Center Drive, Thousand Oaks, California 91320-1799, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Discovery of potent, selective, and metabolically stable 4-(pyridin-3-yl)cinnolines as novel phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem Lett 2012; 22:2262-5. [DOI: 10.1016/j.bmcl.2012.01.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/19/2012] [Accepted: 01/23/2012] [Indexed: 11/23/2022]
|
24
|
Radiosynthesis and Radiotracer Properties of a 7-(2-[18F]Fluoroethoxy)-6-methoxypyrrolidinylquinazoline for Imaging of Phosphodiesterase 10A with PET. Pharmaceuticals (Basel) 2012; 5:169-88. [PMID: 24288087 PMCID: PMC3763632 DOI: 10.3390/ph5020169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 01/18/2012] [Accepted: 01/19/2012] [Indexed: 12/02/2022] Open
Abstract
Phosphodiesterase 10A (PDE10A) is a key enzyme of intracellular signal transduction which is involved in the regulation of neurotransmission. The molecular imaging of PDE10A by PET is expected to allow a better understanding of physiological and pathological processes related to PDE10A expression and function in the brain. The aim of this study was to develop a new 18F-labeled PDE10A ligand based on a 6,7-dimethoxy-4-pyrrolidinylquinazoline and to evaluate its properties in biodistribution studies. Nucleophilic substitution of the 7-tosyloxy-analogue led to the 7-[18F]fluoroethoxy-derivative [18F]IV with radiochemical yields of 25% ± 9% (n = 9), high radiochemical purity of ≥99% and specific activities of 110–1,100 GBq/μmol. [18F]IV showed moderate PDE10A affinity (KD,PDE10A = 14 nM) and high metabolic stability in the brain of female CD-1 mice, wherein the radioligand entered rapidly with a peak uptake of 2.3% ID/g in striatum at 5 min p.i. However, ex vivo autoradiographic and in vivo blocking studies revealed no target specific accumulation and demonstrated [18F]IV to be inapplicable for imaging PDE10A with PET.
Collapse
|
25
|
Pyrazoloquinolines as PDE10A inhibitors: Discovery of a tool compound. Bioorg Med Chem Lett 2012; 22:1335-9. [DOI: 10.1016/j.bmcl.2011.12.080] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 12/13/2011] [Accepted: 12/15/2011] [Indexed: 11/17/2022]
|
26
|
The discovery of potent, selective, and orally active pyrazoloquinolines as PDE10A inhibitors for the treatment of Schizophrenia. Bioorg Med Chem Lett 2012; 22:1019-22. [DOI: 10.1016/j.bmcl.2011.11.127] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 01/06/2023]
|
27
|
Kehler J, Ritzen A, Langgård M, Petersen SL, Farah MM, Bundgaard C, Christoffersen CT, Nielsen J, Kilburn JP. Triazoloquinazolines as a novel class of phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem Lett 2011; 21:3738-42. [DOI: 10.1016/j.bmcl.2011.04.067] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 04/12/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
|
28
|
Asproni B, Murineddu G, Pau A, Pinna GA, Langgård M, Christoffersen CT, Nielsen J, Kehler J. Synthesis and SAR study of new phenylimidazole-pyrazolo[1,5-c]quinazolines as potent phosphodiesterase 10A inhibitors. Bioorg Med Chem 2011; 19:642-9. [DOI: 10.1016/j.bmc.2010.10.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 10/11/2010] [Accepted: 10/14/2010] [Indexed: 01/21/2023]
|
29
|
Celen S, Koole M, De Angelis M, Sannen I, Chitneni SK, Alcazar J, Dedeurwaerdere S, Moechars D, Schmidt M, Verbruggen A, Langlois X, Van Laere K, Andrés JI, Bormans G. Preclinical evaluation of 18F-JNJ41510417 as a radioligand for PET imaging of phosphodiesterase-10A in the brain. J Nucl Med 2010; 51:1584-91. [PMID: 20847170 DOI: 10.2967/jnumed.110.077040] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Phosphodiesterases are enzymes that inactivate the intracellular second messengers 3',5'-cyclic adenosine-monophosphate and/or cyclic guanosine-monophosphate. Of all 11 known phosphodiesterase families, phosphodiesterase-10A (PDE10A) has the most restricted distribution, with high expression in the striatum. PDE10A inhibitors are pursued as drugs for treatment of neuropsychiatric disorders. We have synthesized and evaluated (18)F-JNJ41510417 as a selective and high-affinity radioligand for in vivo brain imaging of PDE10A using PET. METHODS The biodistribution of (18)F-JNJ41510417 was evaluated in rats. Rat plasma and perfused brain homogenates were analyzed by high-performance liquid chromatography to quantify radiometabolites. Dynamic small-animal PET was performed in rats and in wild-type and PDE10A knock-out mice and compared with ex vivo autoradiography. Blocking and displacement experiments were performed using the nonradioactive analog and other selective PDE10A inhibitors. RESULTS Tissue distribution studies showed predominant hepatobiliary excretion, sufficient brain uptake (0.56 ± 0.00 percentage injected dose at 2 min after tracer injection), and continuous accumulation of the tracer in the striatum over time; rapid washout of nonspecific binding from other brain regions was observed. Polar radiometabolites were detected in plasma and brain tissue. Dynamic small-animal PET showed continuous tracer accumulation in the striatum, with rapid decline in the cortex and cerebellum. Pretreatment and chase experiments with PDE10A inhibitors showed that the tracer binding to PDE10A was specific and reversible. Imaging in PDE10A knock-out and wild-type mice further confirmed that binding in the striatum was specific for PDE10A. CONCLUSION Experiments in rats and PDE10A knock-out mice indicate that (18)F-JNJ41510417 binds specifically and reversibly to PDE10A in the striatum, suggesting that this new fluorinated quinoline derivative is a promising candidate for in vivo imaging of PDE10A using PET.
Collapse
Affiliation(s)
- Sofie Celen
- Laboratory for Radiopharmacy, Faculty of Pharmaceutical Sciences, K.U. Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Höfgen N, Stange H, Schindler R, Lankau HJ, Grunwald C, Langen B, Egerland U, Tremmel P, Pangalos MN, Marquis KL, Hage T, Harrison BL, Malamas MS, Brandon NJ, Kronbach T. Discovery of Imidazo[1,5-a]pyrido[3,2-e]pyrazines as a New Class of Phosphodiesterase 10A Inhibitiors. J Med Chem 2010; 53:4399-411. [DOI: 10.1021/jm1002793] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Norbert Höfgen
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Hans Stange
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Rudolf Schindler
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | | | | | - Barbara Langen
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Ute Egerland
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Peter Tremmel
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Menelas N. Pangalos
- Pfizer Neuroscience Princeton, 865 Ridge Road, Monmouth Junction, New Jersey 08852
| | - Karen L. Marquis
- Pfizer Neuroscience Princeton, 865 Ridge Road, Monmouth Junction, New Jersey 08852
| | - Thorsten Hage
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| | - Boyd L. Harrison
- Pfizer Neuroscience Princeton, 865 Ridge Road, Monmouth Junction, New Jersey 08852
| | - Michael S. Malamas
- Pfizer Neuroscience Princeton, 865 Ridge Road, Monmouth Junction, New Jersey 08852
| | - Nicholas J. Brandon
- Pfizer Neuroscience Princeton, 865 Ridge Road, Monmouth Junction, New Jersey 08852
| | - Thomas Kronbach
- Biotie Therapies GmbH, Meissner Strasse 191, 01445 Radebeul, Germany
| |
Collapse
|
31
|
Tu Z, Xu J, Jones LA, Li S, Mach RH. Carbon-11 labeled papaverine as a PET tracer for imaging PDE10A: radiosynthesis, in vitro and in vivo evaluation. Nucl Med Biol 2010; 37:509-16. [PMID: 20447563 PMCID: PMC2866165 DOI: 10.1016/j.nucmedbio.2009.12.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 12/06/2009] [Accepted: 12/30/2009] [Indexed: 11/23/2022]
Abstract
Papaverine, 1-(3,4-dimethoxybenzyl)-6,7-dimethoxyisoquinoline, a specific inhibitor of phosphodiesterase (PDE) 10A with IC(50) values of 36 nM for PDE10A, 1,300 nM for PDE3A and 320 nM for PDE4D, has served as a useful pharmaceutical tool to study the physiological role of PDE10A. Here, we report the radiosynthesis of [(11)C]papaverine and the in vitro and in vivo evaluation of [(11)C]papaverine as a potential positron emission tomography (PET) radiotracer for imaging PDE10A in the central nervous system (CNS). The radiosynthesis of papaverine with (11)C was achieved by O-methylation of the corresponding des-methyl precursor with [(11)C]methyl iodide. [(11)C]papaverine was obtained with approximately 70% radiochemical yield and a specific activity >10 Ci/mumol. In vitro autoradiography studies of rat and monkey brain sections revealed selective binding of [(11)C]papaverine to PDE10A enriched regions: the striatum of rat brain and the caudate and putamen of rhesus monkey brain. The biodistribution of [(11)C]papaverine in rats at 5 min demonstrated an initially higher accumulation in striatum than in other brain regions, however the washout was rapid. MicroPET imaging studies in rhesus macaques similarly displayed initial specific uptake in the striatum with very rapid clearance of [(11)C]papaverine from brain. Our initial evaluation suggests that despite papaverine's utility for in vitro studies and as a pharmaceutical tool, [(11)C]papaverine is not an ideal radioligand for clinical imaging of PDE10A in the CNS. Analogs of papaverine having a higher potency for inhibiting PDE10A and improved pharmacokinetic properties will be necessary for imaging this enzyme with PET.
Collapse
Affiliation(s)
- Zhude Tu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
32
|
Deninno MP, Andrews M, Bell AS, Chen Y, Eller-Zarbo C, Eshelby N, Etienne JB, Moore DE, Palmer MJ, Visser MS, Yu LJ, Zavadoski WJ, Michael Gibbs E. The discovery of potent, selective, and orally bioavailable PDE9 inhibitors as potential hypoglycemic agents. Bioorg Med Chem Lett 2009; 19:2537-41. [PMID: 19339180 DOI: 10.1016/j.bmcl.2009.03.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/05/2009] [Accepted: 03/09/2009] [Indexed: 11/24/2022]
Abstract
Starting from a non-selective pyrazolo-pyrimidone lead, the sequential use of parallel medicinal chemistry and directed synthesis led to the discovery of potent, highly selective, and orally bioavailable PDE9 inhibitors. The availability of these tools allowed for a thorough evaluation of the therapeutic potential of PDE9 inhibition.
Collapse
Affiliation(s)
- Michael P Deninno
- Pfizer Global Research and Development, Groton Laboratories, Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Marino MJ, Knutsen LJS, Williams M. Emerging Opportunities for Antipsychotic Drug Discovery in the Postgenomic Era. J Med Chem 2008; 51:1077-107. [PMID: 18198826 DOI: 10.1021/jm701094q] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Michael J. Marino
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| | - Lars J. S. Knutsen
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| | - Michael Williams
- Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380
| |
Collapse
|
34
|
Brandon NJ, Rotella DP. Chapter 1 Potential CNS Applications for Phosphodiesterase Enzyme Inhibitors. ANNUAL REPORTS IN MEDICINAL CHEMISTRY VOLUME 42 2007. [DOI: 10.1016/s0065-7743(07)42001-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|