Drug Res (Stuttg) 2020; 70(06): 243-256
DOI: 10.1055/a-1146-2996
Original Article

Synthesis and Structure Activity Relationships of Chalcone based Benzocycloalkanone Derivatives as Adenosine A1 and/or A2A Receptor Antagonists

Helena D. Janse van Rensburg
1   Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
,
Lesetja J. Legoabe
1   Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
,
Gisella Terre’Blanche
1   Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
2   Pharmaceutical Chemistry, School of Pharmacy, North-West University, Potchefstroom, South Africa
› Author Affiliations
Funding: This work is based on the research supported in part by the National Research Foundation (NRF) of South Africa (grant number 111814) and the North-West University (NWU).

Abstract

Adenosine A1 and/or A2A receptor antagonists hold promise for the potential treatment of neurological conditions, such as Parkinson’s disease. Herein, a total of seventeen benzocycloalkanone derivatives were synthesised and evaluated for affinity towards adenosine receptors (A1 and A2A AR). The obtained results allowed for the conclusion that affinity and/or selectivity of the 2-benzylidene-1-indanone and -tetralone derivatives toward A1 and/or A2A ARs may be modulated by the nature of the substituents (either -OH, -OCH3 or morpholine) attached at position C4 of the 1-indanone core and C5 of the 1-tetralone core as well as the meta (C3’) and/or para (C4’) position(s) on ring B. Several compounds (2ab, 3bc and 4ab) possessed affinity for the A1 and/or A2A AR below 10 µM. Additionally, compounds 2a, 3b and 4a were A1 AR antagonists. These results, once again, confirmed the importance of C4 methoxy-group substitution on ring A in combination with meta (C3’) and/or para (C4’) hydroxyl-group substitution ring B of the 2-benzylidene-1-indanone scaffold leading to drug-like compounds 1h and 1j with affinity in the nanomolar-range.

Supporting Information



Publication History

Received: 02 March 2020

Accepted: 23 March 2020

Article published online:
29 April 2020

© Georg Thieme Verlag KG
Stuttgart · New York

 
  • References

  • 1 de Lera Ruiz M, Lim Y-H, Zheng J. Adenosine A2A receptor as a drug discovery target. J Med Chem 2013; 57: 3623-3650
  • 2 Ltd KHKC. Approval for manufacturing and marketing of NOURIAST tablets 20mg, a novel antiparkinsonian agent. In: March 25 2013
  • 3 Takahashi M, Fujita M, Asai N. et al. Safety and effectiveness of istradefylline in patients with Parkinson’s disease: Interim analysis of a post-marketing surveillance study in Japan. Expert opinion on pharmacotherapy 2018; 19: 1635-1642
  • 4 White Oak, MD: FDA. FDA approves new add-on drug to treat off episodes in adults with Parkinson’s disease [press release]. In: August 27, 2019
  • 5 Aarsland D, Påhlhagen S, Ballard CG. et al. Depression in Parkinson disease—epidemiology, mechanisms and management. Nature Reviews. Neurology 2012; 8: 35
  • 6 Nagayama H, Kano O, Murakami H. et al. Effect of istradefylline on mood disorders in Parkinson's disease. J Neurol Sci 2019; 396: 78-83
  • 7 Kitta T, Yabe I, Kanno Y. et al. Long-term outcome of adenosine A2A receptor antagonist on lower urinary tract symptoms in male parkinson disease patients. Clinical Neuropharmacology 2018; 41: 98
  • 8 Matsuura K, Kajikawa H, Tabei K-i. et al. The effectiveness of istradefylline for the treatment of gait deficits and sleepiness in patients with Parkinson’s disease. Neurosci Lett 2018; 662: 158-161
  • 9 Franco R, Navarro G. Adenosine A2A receptor antagonists in neurodegenerative diseases: Huge potential and huge challenges. Frontiers in Psychiatry 2018; 9: 68
  • 10 Ikeda K, Kurokawa M, Aoyama S. et al. Neuroprotection by adenosine A2A receptor blockade in experimental models of Parkinson's disease. J Neurochem 2002; 80: 262-270
  • 11 Kalda A, Yu L, Oztas E. et al. Novel neuroprotection by caffeine and adenosine A2A receptor antagonists in animal models of Parkinsonʼs disease. J Neurol Sci 2006; 248: 9-15
  • 12 Mathew B, Parambi DG, Mathew GE. et al. Emerging therapeutic potentials of dual-acting MAO and AChE inhibitors in Alzheimerʼs and Parkinson's diseases. Archiv der Pharmazie. 2019: e1900177. DOI: https://doi.org/10.1002/ardp.201900177
  • 13 Sollevi A. Cardiovascular effects of adenosine in man; possible clinical implications. Progress in Neurobiology 1986; 27: 319-349
  • 14 Shook BC, Rassnick S, Osborne MC. et al. In vivo characterization of a dual adenosine A2A/A1 receptor antagonist in animal models of Parkinson’s disease. J Med Chem 2010; 53: 8104-8115
  • 15 Mihara T, Mihara K, Yarimizu J. et al. Pharmacological characterization of a novel, potent adenosine A1 and A2A receptor dual antagonist, 5-[5-amino-3-(4-fluorophenyl) pyrazin-2-yl]-1-isopropylpyridine-2 (1H)-one (ASP5854), in models of Parkinson's disease and cognition. J Pharmacol Exp Ther 2007; 323: 708-719
  • 16 Atack JR, Shook BC, Rassnick S. et al. JNJ-40255293, a novel adenosine A2A/A1 antagonist with efficacy in preclinical models of Parkinson’s disease. ACS Chemical Neuroscience 2014; 5: 1005-1019
  • 17 Turek M, Szczęsna D, Koprowski M. et al. Synthesis of 1-indanones with a broad range of biological activity. Beilstein Journal of Organic Chemistry 2017; 13: 451
  • 18 Patil SA, Patil R, Patil SA. Recent developments in biological activities of indanones. European Journal of Medicinal chemistry 2017; 138: 182-198
  • 19 Sugimoto H, Iimura Y, Yamanishi Y. et al. Synthesis and structure-activity relationships of acetylcholinesterase inhibitors: 1-benzyl-4-[(5, 6-dimethoxy-1-oxoindan-2-yl) methyl] piperidine hydrochloride and related compounds. J Med Chem 1995; 38: 4821-4829
  • 20 Shintani EY, Uchida KM. Donepezil: An anticholinesterase inhibitor for Alzheimer’s disease. American Journal of Health-system Pharmacy 1997; 54: 2805-2810
  • 21 Menezes JC. Arylidene indanone scaffold: medicinal chemistry and structure–activity relationship view. RSC advances 2017; 7: 9357-9372
  • 22 Perjési P, Perjéssy A, Kolehmainen E. et al. E-2-Benzylidenebenzocycloalkanones III. Studies on transmission of substituent effects on IR carbonyl stretching frequencies and 13C NMR chemical shifts of E-2-(X-benzylidene)-1-indanones. Comparison with the IR data of E-2-(X-benzylidene)-1-indanones,-tetralones, and-benzosuberones. J Mol Struct 2004; 697: 41-47
  • 23 Janse van Rensburg HD, Legoabe LJ, Terre'Blanche G. et al. Methoxy substituted 2-benzylidene-1-indanone derivatives as A1 and/or A2A AR antagonists for the potential treatment of neurological conditions. Medchemcomm 2019; 10: 300-309
  • 24 Janse van Rensburg HD, Legoabe LJ, Terre'Blanche G. et al. 2-benzylidene-1-indanone analogues as dual adenosine A1/A2a receptor antagonists for the potential treatment of neurological conditions. Drug Research 2019; 69: 382-391
  • 25 Legoabe LJ, Van der Walt MM, Terre'Blanche G. Evaluation of 2-benzylidene-1-tetralone derivatives as antagonists of A1 and A2A adenosine receptors. Chemical Biology & Drug Design 2018; 91: 234-244
  • 26 Janse van Rensburg H, Terre'Blanche G, van der Walt M. et al. 5-Substituted 2-benzylidene-1-tetralone analogues as A1 and/or A2A antagonists for the potential treatment of neurological conditions. Bioorg Chem 2017; 74: 251-259
  • 27 Zhuang C, Zhang W, Sheng C. et al. Chalcone: A privileged structure in medicinal chemistry. Chemical reviews 2017; 117: 7762-7810
  • 28 Mathew B, Parambi DGT, Uddin M. et al. Perspective Design of Chalcones for the Management of CNS Disorders: A Mini-Review. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders). 2019 DOI: doi:10.2174/1871527318666190610111246
  • 29 Bruns RF, Fergus JH, Badger EW. et al. Binding of the A 1-selective adenosine antagonist 8-cyclopentyl-1, 3-dipropylxanthine to rat brain membranes. Naunyn-Schmiedeberg's Archives of Pharmacology 1987; 335: 59-63
  • 30 Bruns RF, Watson IA. Rules for identifying potentially reactive or promiscuous compounds. J Med Chem 2012; 55: 9763-9772
  • 31 Van der Walt MM, Terre’Blanche G. 1, 3, 7-Triethyl-substituted xanthines—possess nanomolar affinity for the adenosine A1 receptor. Bioorganic & Medicinal Chemistry 2015; 23: 6641-6649
  • 32 Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976; 72: 248-254
  • 33 Lohse MJ, Klotz K-N, Lindenborn-Fotinos J. et al. 8-Cyclopentyl-1, 3-dipropylxanthine (DPCPX)-a selective high affinity antagonist radioligand for A 1 adenosine receptors. Naunyn-Schmiedebergʼs archives of pharmacology 1987; 336: 204-210
  • 34 Van der Werten EM, Hartog-Witte HR, Roelen HC. et al. 8-Substituted adenosine and theophylline-7-riboside analogues as potential partial agonists for the adenosine A1 receptor. European Journal of Pharmacology: Molecular Pharmacology 1995; 290: 189-199
  • 35 Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports 2017; 7: 42717
  • 36 Delmedico MK. Methods of treating or preventing psoriasis, and/or alzheimerʼs disease using indane acetic acid derivatives. In: Google Patents. 2012
  • 37 Amakali KT, Legoabe LJ, Petzer A. et al. Synthesis and evaluation of 2-benzylidene-1-tetralone derivatives for monoamine oxidase inhibitory activity. Central Nervous System Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Central Nervous System Agents) 2018; 18: 136-149
  • 38 Amakali KT, Legoabe LJ, Petzer A. et al. Synthesis and in vitro evaluation of 2-heteroarylidene-1-tetralone derivatives as monoamine oxidase inhibitors. Drug Research 2018; 68: 687-695
  • 39 Hallgas B, Dobos Z, Ősz E. et al. Characterization of lipophilicity and antiproliferative activity of E-2-arylmethylene-1-tetralones and their heteroanalogues. Journal of Chromatography B 2005; 819: 283-291
  • 40 Bayer H, Batzl C, Hartman RW. et al. New aromatase inhibitors. Synthesis and biological activity of pyridyl-substituted tetralone derivatives. J Med Chem 1991; 34: 2685-2691
  • 41 Mathew B, Baek SC, Thomas Parambi DG. et al. Potent and highly selective dual-targeting monoamine oxidase-B inhibitors: Fluorinated chalcones of morpholine versus imidazole. Archiv der Pharmazie 2019; 352: 1800309
  • 42 Shook BC, Rassnick S, Wallace N. et al. Design and characterization of optimized adenosine A2A/A1 receptor antagonists for the treatment of Parkinsonʼs disease. Journal of Medicinal Chemistry 2012; 55: 1402-1417
  • 43 der Wenden Van E, Hartog-Witte HR, Roelen H. et al. 8-substituted adenosine and theophylline-7-riboside analogues as potential partial agonists for the adenosine A1 receptor. Eur J Pharmacol 1995; 290: 189-199
  • 44 Gütschow M, Schlenk M, Gäb Jr. et al. Benzothiazinones: A novel class of adenosine receptor antagonists structurally unrelated to xanthine and adenine derivatives. J Med Chem 2012; 55: 3331-3341
  • 45 Lipinski CA, Lombardo F, Dominy BW. et al. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Del Rev 1997; 23: 3-25
  • 46 Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem 1999; 1: 55-68
  • 47 Veber DF, Johnson SR, Cheng H-Y. et al. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem 2002; 45: 2615-2623
  • 48 Egan WJ, Merz KM, Baldwin JJ. Prediction of drug absorption using multivariate statistics. J Med Chem 2000; 43: 3867-3877
  • 49 Muegge I, Heald SL, Brittelli D. Simple selection criteria for drug-like chemical matter. J Med Chem 2001; 44: 1841-1846
  • 50 Teague SJ, Davis AM, Leeson PD. et al. The design of leadlike combinatorial libraries. Angewandte Chemie International Edition 1999; 38: 3743-3748
  • 51 Hann MM, Keserü GM. Finding the sweet spot: the role of nature and nurture in medicinal chemistry. Nature Reviews Drug Discovery 2012; 11: 355