440
Views
17
CrossRef citations to date
0
Altmetric
Review

Towards selective phosphodiesterase 2A (PDE2A) inhibitors: a patent review (2010 - present)

, &
Pages 933-946 | Received 29 Feb 2016, Accepted 16 Jun 2016, Published online: 30 Jun 2016

References

  • Rall TW, Sutherland EW. Formation of a cyclic adenine ribonucleotide by tissue particles. J Biol Chem. 1958;232:1065–1076.
  • Rall TW, Sutherland EW. Fractionation and characterization of a cyclic adenine ribonucleotide formed by tissue particulates. J Biol Chem. 1958;232:1077–1091.
  • Smith M, Drummond GI, Khorana HG. Cyclic phosphates. IV. Ribonucleotide 3‘,5’-cyclic phosphates. A general method of synthesis and some properties. J Am Chem Soc. 1979;83:698–706.
  • Goldberg ND, Walseth TF, Stephenson JH, et al. 18O-labelling of guanosine monophosphate upon hydrolysis of cyclic guanosine 3‘,5’-monophosphate by phosphodiesterase. J Biol Chem. 1980;255:10344–10347.
  • Burgers PMJ, Eckstein F, Hunneman DH, et al. Stereochemistry of hydrolysis of adenosine 3‘,5’-cyclic phosphorodithioate by the cyclic phosphodiesterase from beef heart. J Biol Chem. 1979;254:9959–9961.
  • Maurice DH, Ke H, Ahmad F, et al. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discovery. 2014;13:290–314.
  • Lugnier C. Cyclic nucleotide phosphodiesterase (PDE) superfamily: a new target for the development of specific therapeutic agents. Pharmacol Ther. 2006;109:366–398.
  • Beavo JA, Hardman JG, Sutherland EW. Hydrolysis of cyclic guanosine and adenosine 3‘,5’-monophosphates by rat and bovine tissues. J Biol Chem. 1970;245:5649–5655.
  • Le Trong H, Beier N, Sonnenburg WK, et al. Amino acid sequence of the cyclic GMP stimulated cyclic nucleotide phosphodiesterase from bovine heart. Biochemistry. 1990;29:10280–10288.
  • Sonnenburg WK, Mullaney PJ, Beavo JA. Molecular cloning of a cyclic GMP-stimulated cyclic nucleotide phosphodiesterase cDNA. Identification and distribution of isozyme variants. J Biol Chem. 1991;266:17655–17661.
  • Yang Q, Paskind M, Bolger G, et al. A novel cyclic GMP stimulated phosphodiesterase from rat brain. Biochem Biophys Res Commun. 1994;205:1850–1858.
  • Rosman GJ, Martins TJ, Sonneburg WK, et al. Isolation and characterization of human cDNAs encoding a cGMP stimulated 3‘,5’-cyclic nucleotide phosphodiesterase. Gene. 1997;191:89–95.
  • Martins TJ, Mumby MC, Beavo JA. Purification and characterization of a cyclic GMP-stimulated cyclic nucleotide phosphodiesterase from bovine tissues. J Biol Chem. 1982;254:1973–1979.
  • Martins TJ. Cyclic GMP-stimulated cyclic nucleotide phosphodiesterase: purification from bovine tissues and characterization [ Ph.D. thesis]. University of Washington; 1984:43–64.
  • Beavo JA, Hardman JG, Sutherland EW. Stimulation of adenine 3‘,5’-monophosphate hydrolysis by guanosine 3‘,5’-monophosphate. J Biol Chem. 1971;246:3841–3846.
  • Erneux C, Couchie D, Dumont JE, et al. Specificity of cyclic GMP activation of a multi-substrate cyclic nucleotide phosphodiesterase from rat liver. Eur J Biochem. 1981;115:503–510.
  • Stroop SD, Beavo JA. Structure and function studies of the cGMP-stimulated phosphodiesterase. J Biol Chem. 1991;266:23802–23809.
  • Wu AY, Tang X-B, Martinez SE, et al. Molecular determinants for cyclic nucleotide binding to the regulatory domains of phosphodiesterase 2A. J Biol Chem. 2004;279:37928–37938.
  • Pandit J, Forman MD, Fennell KF, et al. Mechanism for the allosteric regulation of phosphodiesterase 2A deduced from the X-ray structure of a near full-length construct. Proc Natl Acad Sci USA. 2009;106:18225–18230.
  • Mehats C, Andersen CB, Filopanti M, et al. Cylic nucleotide phosphodiesterases and their role in endocrine cell signalling. Trends Endocrinol Metab. 2002;13:29–35.
  • Stephenson DT, Coskran TM, Wilhelms MB, et al. Immunohistochemical localization of phosphodiesterase 2A in multiple mammalian species. J Histochem Cytocochem. 2009;75:933–949.
  • The human protein atlas. [cited 2016 June 6]. Available from: http://www.proteinatlas.org/ENSG00000186642-PDE2A/tissue.
  • Human proteome map. [cited 2016 June 6]. Available from: http://www.humanproteomemap.org/.
  • Lakics V, Karran EH, Boess FG. Quantitative comparison of phosphodiesterase mRNA distribution in human brain and peripheral tissues. Neuropharmacology. 2010;59:367–374.
  • Reyes-Irisarri E, Herzog Y, Eyal I, et al. Cognitex supplementation of the cGMP-specific phosphodiesterases 2 and 9 in normal and Alzheimer’s disease human brains. Eur J Neurosci. 2007;25:3332–3338.
  • Reneerkens OAH, Rutten K, Steinbusch WWM, et al. Selective phosphodiesterase inhibitors: a promising target for cognition enhancement. Psychopharmacology. 2009;202:419–443.
  • Zhang C, Yu Y, Ruan L, et al. The role of phosphodiesterase 2 in the central nervous and peripheral systems. Curr Pharm Des. 2015;21:274–290.
  • Xu Y, Pan J, Chen L, et al. Phosphodiesterase-2 inhibitor reverses corticosterone-induced neurotoxicity and related behavioral changes via cGMP/PKG dependent pathway. J Neuropsychopharmacol. 2013;16:835–847.
  • Masood A, Huang Y, Hajjhussein H, et al. Anxiolytic effects of phosphodiesterase-2 inhibitors associated with increased cGMP signaling. J Pharmacol Exp Ther. 2009;331:690–699.
  • Plummer MS, Cornicelli J, Roark H, et al. Discovery of potent, selective, bioavailable phosphodiesterase 2 (PDE2) inhibitors active in an osteoarthritis pain model. Part I. Transformation of selective phosphodiesterase 4 (PDE4) inhibitors into selective PDE2 inhibitors. Bioorg Med Chem Lett. 2013;23:3438–3442.
  • Plummer MS, Cornicelli J, Roark H, et al. Discovery of potent, selective, bioavailable phosphodiesterase 2 (PDE2) inhibitors active in an osteoarthritis pain model. Part II. Optimization studies and demonstration of in vivo efficacy. Bioorg Med Chem Lett. 2013;23:3443–3447.
  • Mseeh F, Colman RF, Colman RW. Inactivation of platelet PDE2 by and affinity label: 8-[(4-bromo-2,3-dioxobutylthio]cAMP. Thromb Res. 2000;98:395–401.
  • Surapisitchat J, Jeon K-I, Yan C, et al. Differential regulation of endothelial cell permeability by cGMP via phosphodiesterases 2 and 3. Circulation. 2007;101:811–818.
  • Zhang Y, Daaka Y. PDE2 promotes angiogenesis through EP4 and PKA Cγ pathway. Blood. 2011;118:5355–5364.
  • Favot L, Keravis T, Lugnier C. Modulation of VEGF-induced endothelial cell cycle protein expression through cyclic AMP hydrolysis by PDE2 and PDE4. Thromb Haemost. 2004;92:634–645.
  • Wilson D, Tutulan-Cunita A, Jung W, et al. Deletion of the high-affinity cAMP phosphodiesterase encoded by PDE2 affects stress responses and virulence in Candida albicans. Mol Microbiol. 2007;65:841–856.
  • Bayer Aktiengesellschaft, Preparation of imidazotriazinones as phosphodiesetrase II inhibitors. WO 050078. 2002.
  • Rutten K, Prickaerts J, Hendrix M, et al. Time-dependent involvement of cAMP and cGMP in consolidation of object memory: studies using selective phosphodiesterase type 2, 4 and 5 inhibitors. Eur J Pharm. 2007;558:107–112.
  • Boess FG, Hendrix M, van der Staay F-J, et al. Inhibition of phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology. 2004;47:1081–1092.
  • Prickaerts J, De Vente J, Honig W, et al. A. cGMP, but not cAMP, in rat hippocampus is involved in early stages of object memory consolidation. Eur J Pharmacol. 2002;436:83–87.
  • Rodefer JS, Saland SK, Eckrich SJ. Selective phosphodiesterase inhibitors improve performance on the ED/ID cognitive task in rats. Neuropharmacology. 2012;62:1182–1190.
  • Reneerkens OA, Rutten K, Bollen E, et al. Inhibition of phoshodiesterase type 2 or type 10 reverses object memory deficits induced by scopolamine or MK-801. Behav Brain Res. 2013;236:16–22.
  • Xu Y, Pan J, Sun J, et al. Inhibition of phosphodiesterarse 2 reverses impaired cognition and neuronal remodeling caused by chronic stress. Neurobiol Aging. 2015;36:955–970.
  • Ding L, Zhang C, Masood A, et al. Protective effects of phosphodiesterase 2 inhibitor on depression- and anxiety-like behaviors: involvement of antioxidant and anti-apoptotic mechanisms. Behav Brain Res. 2014;268:150–158.
  • Reneerkens OAH, Sambeth A, Blokland A, et al. PDE2 and PDE10, but not PDE5, inhibition affect auditory information processing in rats. Behav Brain Res. 2013;250:251–256.
  • Zhu J, Yang Q, Dai D, et al. X-ray crystal structure of phosphodiesterase 2 in complex with a highly selective, nanomolar inhibitor reveals a binding-induced pocket important for selectivity. J Am Chem Soc. 2013;135:11708–11711.
  • Altana Pharma AG, Preparation of purin-6-one derivatives for use in pharmaceutical compositions for the treatment of neoangiogenesis related disorders. WO089953. 2004.
  • Altana Pharma AG, Preparation of triazolophthalazines as PDE2 inhibitors. WO024640. 2006).
  • Altana Pharma AG, Triazolophthalazines as phosphodiesterase inhibitors. WO072612. 2006.
  • Altana Pharma AG, Preparation of 6-anilinotriazolophthalazines as PDE2 inhibitors for medical uses. WO072615. 2006.
  • Neuro3d, Université Louis Pasteur, Centre National de la Recherche Scientifique, Forenap, Cyclic nucleotide phosphodiesterase inhibitors, preparation and uses. W041258. 2004.
  • Neuro3d, Benzo[1,4]diazepin-2-one derivatives as phosphodiesterase PDE2 inhibitors, preparation and therapeutic use thereof. EP1548011, 2005.
  • Neuro3d, Benzodiazepine derivatives, their preparation and therapeutic use thereof. EP1749824. 2007.
  • Pfizer Products Inc., Oxindole derivatives and their use as phosphodiesterase type 2 inhibitors. WO041957. 2005.
  • Cambers RJ, Abrams K, Garceau NY, et al. A new chemical tool for exploring the physiological function of the PDE2 isozyme. Bioorg Med Chem Lett. 2006;16:307–310.
  • Pfizer Products Inc., Pyrido[2,3-d]pyrimidine-2,4-diamines as PDE 2 inhibitors. WO061497. 2005.
  • Biotie Therapies GMBH and Wyeth, Imidazo[5,1-c][1,2,4]benzotriazine derivatives as inhibitors of phosphodiesterases. WO054260. 2010.
  • Biotie Therapies GMBH and Wyeth. Triazine derivatives as inhibitors of phosphodiesterases. WO054253. 2010.
  • Schröder S, Wenzel B, Deuther-Conrad W, et al. Synthesis, 18F-radiolabelling and biological characterization of novel fluoroalkylated triazine derivatives for in vivo imaging of phosphodiesterase 2A in brain via positron emission tomography. Molecules. 2015;20:9591–9615.
  • Boehringer Ingelheim International GMBH. [1,2,4]Triazolo[4,3-a]quinoxaline derivatives as inhibitors of phosphodiesterases. WO104293. 2012.
  • Malamas MS, Stange H, Schindler R, et al. Novel triazines as potent and selective phosphodiesterase 10A inhibitors. Bioorg Med Chem Lett. 2012;22:5876–5884.
  • Verhoest PR, Chapin DS, Corman M, et al. Discovery of a novel class of phosphodiesterase 10A inhibitors and identification of clinical candidate 2-[4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)-phenoxymethyl]-quinoline (PF-2545920) for the treatment of schizophrenia. J Med Chem. 2009;52:5188–5196.
  • Boehringer Ingelheim International GMBH, 4-Methyl-2,3,5,9,9b-pentaaza-cyclopenta[a]naphthalenes. WO019979. 2014.
  • Dart Neuroscience, LLC, Substituted [l,2,4]triazolo[l,5-a]pyrimidin-7-yl compounds as PDE2 inhibitors. WO164508.2015.
  • Janssen Pharmaceutica NV, 1-Aryl-4-methyl-[1,2,4]triazolo[4,3-a]quinoxaline derivatives. WO000924. 2013.
  • Janssen Pharmaceutica NV, Combinations comprising PDE2 inhibitors such as 1-aryl-4-methyl-[1,2,4]triazolo[4,3-a]quinoxaline compounds and PDE10 inhibitors for use in the treatment of neurological or metabolic disorders. WO001314. 2014.
  • Chappie TA, Humphrey JM, Allen MP, et al. Discovery of a series of 6,7-dimethoxy-4-pyrrolidylquinazoline PDE10A inhibitors. J Med Chem. 2007;50:182–185.
  • Schmidt CJ, Chapin DS, Cianfrogna J, et al. Preclinical characterization of selective phosphodiesterase 10A inhibitors: a new therapeutic approach to the treatment of schizophrenia. J Pharmacol Exp Ther. 2008;325:681–690.
  • Siuciak JA, McCarthy SA, Chapin DS, et al. Genetic deletion of the striatum-enriched phosphodiesterase PDE10A: evidence for altered striatal function. Neuropharmacology. 2006;51:374–385.
  • Janssen Pharmaceutica NV. Imidazo[1,2-b]pyridazine derivatives and their use as PDE10 inhibitors. WO051342. 2011.
  • Janssen Pharmaceutica NV. Imidazo[1,2-a]pyrazine derivatives and their use for the prevention or treatment of neurological, psychiatric and metabolic disorders and diseases. WO110545. 2011.
  • Andrés J-I, Buijnsters P, De Angelis M, et al. Discovery of a new series of [1,2,4]triazolo[4,3-a]quinoxalines as dual phosphodiesterase 2/phosphodiesterase 10 (PDE2/PDE10) inhibitors. Bioorg Med Chem Lett. 2013;23:785–790.
  • Buijnsters P, De Angelis M, Langlois X, et al. Structure-based design of a potent, selective, and brain penetrating PDE2 inhibitor with demonstrated target engagement. ACS Med Chem Lett. 2014;5:1049–1053.
  • Rombouts FJR, Tresadern G, Buijnsters P, et al. Pyrido[4,3-e][1,2,4]triazolo[4,3-a]pyrazines as selective, brain penetrant phosphodiesterase 2 (PDE2) inhibitors. ACS Med Chem Lett. 2015;6:282−86.
  • H. Lundbeck A/S. Triazolopyrazine derivatives and their use for treating neurological and psychiatric disorders. WO034755. 2013.
  • H. Lundbeck A/S. Substituted triazolopyrazines and uses thereof. WO034758. 2013.
  • H. Lundbeck A/S. Pyridine compounds and uses thereof. WO034761. 2013.
  • H. Lundbeck A/S. [l,2,4]Triazolo[4,3-A]quinoxalines as dual PDE2/PDE10 inhibitors. WO139983. 2014.
  • Redrobe JP, Jørgensen M, Christoffersen CT, et al. In vitro and in vivo characterisation of Lu AF64280, a novel, brain penetrant phosphodiesterase (PDE) 2A inhibitor: potential relevance to cognitive deficits in schizophrenia. Psychopharmacology. 2014;213:3151–3167.
  • Redrobe JP, Rasmussen LK, Christoffersen CT, et al. Characterisation of Lu AF33241: a novel, brain-penetrant, dual inhibitor of phosphodiesterase (PDE) 2A and PDE10A. Eur J Pharmacol. 2015;761:79–85.
  • Pfizer Incorporated. Imidazo[5,1-f][1,2,4]triazines for the treatment of neurological disorders. US0214791. 2012.
  • Zhang L, Villalobos A, Beck EM, et al. Design and selection parameters to accelerate the discovery of novel central nervous system positron emission tomography (PET) ligands and their application in the development of a novel phosphodiesterase 2A PET ligand. J Med Chem. 2013;56:4568–4579.
  • Helal CJ. Identification of a brain penetrant, highly selective phosphodiesterase 2A inhibitor for the treatment of cognitive impairment associated with schizophrenia (CIAS). Abstract of Papers, 244th ACS National Meeting & Exposition; 2012 Aug 19–23; Philadelphia, PA.
  • Pfizer Incorporated. Pyrazolo[3,4-d]pyrimidine compounds and their use as PDE2 inhibitors and/or CPY3A4 inhibitors. WO168817. 2012.
  • Takeda Pharmaceutical Company Limited. Heterocyclic compound. WO010732. 2014.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.