334
Views
0
CrossRef citations to date
0
Altmetric
Research Articles

Update of Recently (2016–2020) Designed Azepine Analogs and Related Heterocyclic Compounds with Potent Pharmacological Activities

Pages 2250-2268 | Received 09 Apr 2021, Accepted 06 Feb 2022, Published online: 24 Feb 2022

References

  • C. Kunick, C. Bleeker, C. Prühs, F. Totzke, C. Schächtele, M. H. G. Kubbutat, and A. Link, “Matrix Compare Analysis Discriminates Subtle Structural Differences in a Family of Novel Antiproliferative Agents, Diaryl-3-Hydroxy-2,3,3a,10a-Tetrahydrobenzo[b]Cycylopenta[e]Azepine-4,10(1H,5H)-Diones,” Bioorganic & Medicinal Chemistry Letters 16, no. 8 (2006): 2148–53. DOI: 10.1016/j.bmcl.2006.01.071.
  • T. Tsunoda, A. Yamazaki, T. Mase, and S. Sakamoto, “A Scalable Process for the Synthesis of 2-Methyl-1,4,5,6-Tetrahydroimidazo[4,5- d ][1]Benzazepine Monohydrate and 4-[(Biphenyl-2-Ylcarbonyl)Amino]Benzoic Acid: Two New Key Intermediates for the Synthesis of the AVP Antagonist Conivaptan Hydrochloride,” Organic Process Research & Development 9, no. 5 (2005): 593–8. DOI: 10.1021/op050061n.
  • M. Seto, N. Miyamoto, K. Aikawa, Y. Aramaki, N. Kanzaki, Y. Iizawa, M. Baba, and M. Shiraishi, “Orally Active CCR5 Antagonists as anti-HIV-1 Agents. Part 3: Synthesis and Biological Activities of 1-Benzazepine Derivatives Containing a Sulfoxide Moiety,” Bioorganic & Medicinal Chemistry 13, no. 2 (2005): 363–86. DOI: 10.1016/j.bmc.2004.10.021.
  • N. A. El-Sayed, F. M. Awadallah, N. A. Ibrahim, and M. T. El-Saadi, “Synthesis, anti-Inflammatory and Ulcerogenicity Studies of Some Substituted Pyrimido[1,6-a]Azepine Derivatives,” European Journal of Medicinal Chemistry 45, no. 7 (2010): 3147–54. DOI: 10.1016/j.ejmech.2010.04.005.
  • J. Benes, A. Parada, A. A. Figueiredo, P. C. Alves, A. P. Freitas, D. A. Learmonth, R. A. Cunha, J. Garrett, and P. Soares-da-Silva, “Anticonvulsant and Sodium Channel-Blocking Properties of Novel 10,11-dihydro-5H-dibenz[b,f]azepine-5-carboxamide derivatives,” Journal of Medicinal Chemistry 42, no. 14 (1999): 2582–7. DOI: 10.1021/jm980627g.
  • A. Link, and C. Kunick, “ d-Fused [1]benzazepines with selective in vitro antitumor activity: synthesis and structure-activity relationships,” Journal of Medicinal Chemistry 41, no. 8 (1998): 1299–305. DOI: 10.1021/jm970675l.
  • W. Ouyang, J. Rao, Y. Li, X. Liu, Y. Huo, Q. Chen, and X. Li, “Recent Achievements in the Rhodium‐Catalyzed Concise Construction of Medium N‐Heterocycles, Azepines and Azocines,” Advanced Synthesis & Catalysis 362, no. 24 (2020): 5576–600. DOI: 10.1002/adsc.202001040.
  • J. R. Heys, and S. G. Senderoff, “A New Entry into C7-Oxygenated Tetrahydro-1H-3-Benzazepines; Efficient Labeling with Carbon-14 in the Benzo Ring,” The Journal of Organic Chemistry 54, no. 19 (1989): 4702–6. DOI: 10.1021/jo00280a050.
  • K. Ersmark, I. Feierberg, S. Bjelic, E. Hamelink, F. Hackett, M. J. Blackman, J. Hultén, B. Samuelsson, J. Åqvist, and A. Hallberg, “Potent Inhibitors of the Plasmodium falciparum Enzymes Plasmepsin I and II Devoid of Cathepsin D Inhibitory Activity,” Journal of Medicinal Chemistry 47, no. 1 (2004): 110–22. DOI: 10.1021/jm030933g.
  • T. Spangenberg, “Azepanyl - Derivatives and Pharmaceutical Compositions Comprising the Same with Antiparasitic Activity” (US 9994577 B2, 2018).
  • L. de, F. Diniz, J. A. Urbina, I. M. de Andrade, A. L. Mazzeti, T. A. F. Martins, I. S. Caldas, A. Talvani, I. Ribeiro, and M. T. Bahia, “Benznidazole and Posaconazole in Experimental Chagas Disease: Positive Interaction in Concomitant and Sequential Treatments,” PLoS Neglected Tropical Diseases 7, no. 8 (2013): e2367. DOI: 10.1371/journal.pntd.0002367.
  • P. J. Hotez, E. Dumonteil, M. J. Heffernan, and M. E. Bottazzi, “Innovation for the ‘Bottom 100 Million’: Eliminating Neglected Tropical Diseases in the Americas,” in Hot Topics in Infection and Immunity in Children IX. Advances in Experimental Medicine and Biology, edited by N. Curtis, A. Fin, and A. Pollard, Vol. 764 (2013), 1–12. DOI: 10.1007/978-1-4614-4726-9_1.
  • S. Sundar, and J. Chakravarty, “An Update on Pharmacotherapy for leishmaniasis,” Expert Opin Pharmacother 16, no. 2 (2015): 237–52. DOI: 10.1517/14656566.2015.973850.
  • M. Knockaert, K. Wieking, S. Schmitt, M. Leost, K. M. Grant, J. C. Mottram, C. Kunick, and L. Meijer, “Intracellular Targets of Paullones. Identification following Affinity Purification on Immobilized Inhibitor,” The Journal of Biological Chemistry 277, no. 28 (2002): 25493–501. DOI: 10.1074/jbc.M202651200.
  • A. Palma, A. F. Yépes, S. M. Leal, C. A. Coronado, and P. Escobar, “Synthesis and in Vitro Activity of New Tetrahydronaphtho[1,2-b]Azepine Derivatives against Trypanosoma cruzi and Leishmania chagasi Parasites,” Bioorganic & Medicinal Chemistry Letters 19, no. 8 (2009): 2360–3. DOI: 10.1016/j.bmcl.2008.05.013.
  • A. F. Yépes, A. Bahsas, P. Escobar, J. Cobo, A. Palma, J. C. Garro Martinez, and R. Enriz, “Synthesis, anti-Parasitic Activity and QSAR Study of a New Library of Polysubstituted Tetrahydronaphtho[1,2-b]Azepines,” Medicinal Chemistry Research 27, no. 10 (2018): 2239–64. DOI: 10.1007/s00044-018-2232-7.
  • J. Fouche, and A. Leger, “German Patent, 2031 (1971) 236,” Chem. Abstr 74, no. 1971: 76346.
  • S. K. Maddili, R. Chowrasia, V. K. Kannekanti, and H. Gandham, “1,3-Di-n-Butylimidazolium Tribromide [BBim]Br3: An Efficient Recyclable Catalyst Mediated Synthesis of N-Substituted Azepines and their Biological Evaluation-Interaction Study with Human Serum Albumin,” Journal of Photochemistry and Photobiology. B, Biology 178 (2018): 101–7. DOI: 10.1016/j.jphotobiol.2017.10.028.
  • A. Sharma, D. Kishore, and B. Singh, “An Expedient Method for the Synthesis of 1,2,4-Triazolo-Fused 1,5-Benzodiazepine, 1,5-Benzoxazepine, and 1,5-Benzothiazepine Scaffolds: A Novel Seven-Membered Ring System of Biological Interest,” Journal of Heterocyclic Chemistry 55, no. 3 (2018): 586–92. DOI: 10.1002/jhet.3060.
  • S.A. Demchenko, O.E. Yadlovskyi, L.S. Bobkova, А.M. Demchenko, Synthesis and Antiviral Activity of Derivatives of 1-(41-Chlorophenyl)-3-Aryl-3-Hydroxy-2,5,6,7,8,9- Hexahydro-3Н-Imidazo[1,2-а]Azepinium for Flu A H1N1 California/07/2009 and Flu A H3N2 Brisbane/10/2007, Pharmacology and Drug Toxicology 6 (2018) 44–51. http://nbuv.gov.ua/UJRN/ flt _2018_6_7
  • S. A. Demchenko, T. A. Bukhtiarova, L. S. Bobkova, and A. M. Demchenko, “Synthesis and Antiviral Activity of Derivatives of 1- (2,3-Dihydrobenzo | 1,4 | Dioxan-6-yl) -3-Aryl-3-Hydroxy-2,5,6,7,8,9- 3H-Imidazo | 1,2-a | Azepine and Amizon Substances,” Pharmacology and Drug Toxicology 1 (2018): 26–31.
  • P. Bonaventure, C. Dugovic, M. Kramer, P. de Boer, J. Singh, S. Wilson, K. Bertelsen, J. Di, J. Shelton, L. Aluisio, et al, “Translational Evaluation of JNJ-18038683, a 5-Hydroxytryptamine Type 7 Receptor Antagonist, on Rapid Eye Movement Sleep and in Major Depressive Disorder,” Journal of Pharmacology and Experimental Therapeutics 342, no. 2 (2012): 429–40. DOI: 10.1124/jpet.112.193995.
  • P. J. Lovell, S. M. Bromidge, S. Dabbs, D. M. Duckworth, I. T. Forbes, A. J. Jennings, F. D. King, D. N. Middlemiss, S. K. Rahman, D. V .Saunders, et al, “ A novel, Potent, and Selective 5-HT(7) Antagonist: (R)-3-(2-(2-(4-Methylpiperidin-1-yl)Ethyl)Pyrrolidine-1-Sulfonyl) Phen ol (SB-269970)),” Journal of Medicinal Chemistry 43, no. 3 (2000): 342–5. DOI: 10.1021/jm991151j.
  • C. A. Dvorak, D. A. Rudolph, D. Nepomuceno, L. Dvorak, B. Lord, I. Fraser, P. Bonaventure, T. Lovenberg, and N. I. Carruthers, “Discovery and SAR Studies of 2-Alkyl-3-Phenyl-2,4,5,6,7,8-Hexahydropyrazolo[3,4-d]Azepines as 5-HT7/2 Inhibitors Leading to the Identification of a Clinical Candidate,” Bioorganic & Medicinal Chemistry Letters 31 (2021): 127669. DOI: 10.1016/j.bmcl.2020.127669.
  • F. G. Ashby, V. Valentin, and S. von Meer, “Differential Effects of Dopamine-Directed Treatments on Cognition,” Neuropsychiatric Disease and Treatment 29 (2015): 1859–75. DOI: 10.2147/NDT.S65875.
  • Q. Mu, K. Johnson, P. S. Morgan, E. L. Grenesko, C. E. Molnar, B. Anderson, Z. Nahas, F. A. Kozel, S. Kose, M. Knable, et al, “A Single 20 mg Dose of the Full D1 Dopamine Agonist Dihydrexidine (DAR-0100) Increases Prefrontal Perfusion in Schizophrenia,” Schizophrenia Research 94, no. 1-3 (2007): 332–41. DOI: 10.1016/j.schres.2007.03.033.
  • N. H. Mutschler, and J. Bergman, “Effects of Chronic Administration of the D1 Receptor Partial Agonist SKF 77434 on Cocaine Self-Administration in Rhesus Monkeys,” Psychopharmacology 160, no. 4 (2002): 362–70. DOI: 10.1007/s00213-001-0976-z.
  • R. Giri, I. Alberts, and W. W. Harding, “Synthesis, Pharmacological Evaluations, and Molecular Docking Studies on a New 1,3,4,11b-Tetrahydro-1H-Fluoreno[9,1-cd]Azepine Framework: Rigidification of D1 Receptor Selective 1-Phenylbenzazepines and Discovery of a New 5-HT6 Receptor Scaffold,” Chemical Biology & Drug Design 96, no. 2 (2020): 825–35. DOI: 10.1111/cbdd.13691.
  • S. Celanire, L. Tang, N. J. Liverton, and P. Jones, “Tetrahydropyrazolo [ 3,4-b) Azepine Derivatives and their use as Allosteric Modulators of Metabotropic Glutamate Receptors” (US9765091B2, 2011).
  • H. E. Scharfman, “The Neurobiology of Epilepsy,” Current Neurology and Neuroscience Reports 7, no. 4 (2007): 348–54. DOI: 10.1007/s11910-007-0053-z.
  • C. E. Stafstrom, “Epilepsy: A Review of Selected Clinical Syndromes and Advances in Basic Science,” Journal of Cerebral Blood Flow and Metabolism : official Journal of the International Society of Cerebral Blood Flow and Metabolism 26, no. 8 (2006): 983–1004. DOI: 10.1038/sj.jcbfm.9600265.
  • M. J. Brodie, “Monotherapy Trials: Prerequisite Data,” Epilepsy Research 45, no. 1-3 (2001): 61–4. DOI: 10.1016/S0920-1211(01.)00218-2.
  • A. Reimers, “New Antiepileptic Drugs and Women,” Seizure 23, no. 8 (2014): 585–91. DOI: 10.1016/j.seizure.2014.05.004.
  • H. Kaur, B. Kumar, and B. Medhi, “Antiepileptic Drugs in Development Pipeline: A Recent Update,” ENeurologicalSci 4 (2016): 42–51. DOI: 10.1016/j.ensci.2016.06.003.
  • S. N. Sirakanyan, D. Spinelli, A. Geronikaki, and A. A. Hovakimyan, “On the Reactivity of Pyrido[3′,2′:4,5]Furo(Thieno)[3,2-d]Pyrimidin-7(8)-Ones with Some Alkyl Mono- and di-Halides: synthesis of New Heterocyclic Systems Containing Thiazolo[3,2-a]Pyrimidine and Pyrimido[2,1-b]Thiazine Moiety,” Tetrahedron 71, no. 40 (2015): 7638–46. DOI: 10.1016/j.tet.2015.07.069.
  • S. N. Sirakanyan, A. A. Ovakimyan, A. S. Noravyan, N. S. Minasyan, I. A. Dzhagatspanyan, I. M. Nazaryan, and A. G. Akopyan, “Synthesis and Neurotropic Activity of 8-Amino Derivatives of Condensed Thieno[3,2-d]- and Furo[3,2-d]Pyrimidines,” Pharmaceutical Chemistry Journal 47, no. 12 (2014): 655–9. DOI: 10.1007/s11094-014-1026-6.
  • S. N. Sirakanyan, A. Geronikaki, D. Spinelli, R. G. Paronikyan, I. A. Dzhagatspanyan, I. M. Nazaryan, A. H. Akopyan, and A. A. Hovakimyan, “Pyridofuropyrrolo[1,2-a]Pyrimidines and Pyridofuropyrimido[1,2-a]Azepines: new Chemical Entities (NCE) with Anticonvulsive and Psychotropic Properties,” RSC Advances 6, no. 38 (2016): 32234–44. DOI: 10.1039/C6RA02581A.
  • G. Guerrini, A. Costanzo, G. Ciciani, F. Bruni, S. Selleri, C. Costagli, F. Besnard, B. Costa, C. Martini, G. de Siena, et al, “Benzodiazepine Receptor Ligands. 8: Synthesis and Pharmacological Evaluation of New Pyrazolo[5,1-c] [1,2,4]Benzotriazine 5-Oxide 3- and 8-Disubstituted: High Affinity Ligands Endowed with Inverse-Agonist Pharmacological Efficacy,” Bioorganic & Medicinal Chemistry 14, no. 3 (2006): 758–75. DOI: 10.1016/j.bmc.2005.08.058.
  • R. Kumar, and Y. C. Joshi, “Synthesis and Antimicrobial, Antifungal and Anthelmintic Activities of 3h-1,5-Benzodiazepine Derivatives,” Journal of the Serbian Chemical Society 73, no. 10 (2008): 937–43. DOI: 10.2298/JSC0810937K.
  • S. J. R. Rajarao, B. Platt, S. J. Sukoff, Q. Lin, C. N. Bender, B. W. Nieuwenhuijsen, R. H. Ring, L. E. Schechter, S. Rosenzweig-Lipson, and C. E. Beyer, “Anxiolytic-like Activity of the Non-Selective Galanin Receptor Agonist, Galnon,” Neuropeptides 41, no. 5 (2007): 307–20. DOI: 10.1016/j.npep.2007.05.001.
  • Y.-P. Shao, R.-B. Han, H.-F. Wu, and F.-Y. Piao, “Synthesis and Anticonvulsant Activity of Some Novel 7-(Benzylamino) -1H-Benzo[b][1,4]Diazepine-2,4(3H, 5H)-Dione Derivatives,” Medicinal Chemistry Research 27, no. 2 (2018): 642–52. DOI: 10.1007/s00044-017-2089-1.
  • F.-Y. Piao, Y.-Z. Xie, W.-B. Zhang, W. Zhang, and R.-B. Han, “High-Yield Method for the Preparation of 1,3,4,5-Tetrahydro-7-Methoxy-2 H -1-Benzazepin-2-One with Excellent Regio- and Stereoselectivity,” Synthetic Communications 43, no. 14 (2013): 1920–30. DOI: 10.1080/00397911.2012.680571.
  • C.-X. Wei, W. Zhang, Z.-S. Quan, R.-B. Han, R.-S. Jiang, and F.-Y. Piao, “Synthesis, Anticonvulsant Evaluation of 2,3,4,5-Tetrahydro-7-Alkoxy-1H-2-Benzazepin-1-Ones,” Letters in Drug Design & Discovery 6, no. 7 (2009): 548–53. DOI: 10.2174/157018009789108231.
  • M. G. Craske, and M. B. Stein, “Anxiety,” The Lancet 388, no. 10063 (2016): 3048–59. DOI: 10.1016/S0140-6736.(16)30381-6.
  • R. B. Lydiard, “The Role of GABA in Anxiety Disorders,” Journal of Clinical Psychiatry 64 (2003): 21–27.
  • D. K. Mahapatra, D. Das, R. S. Shivhare, and K. M. Dadure, “Transforming Murrayanine-Chalcone into Corresponding 3H-Benzo[b] [1,4] Diazepine Derivatives: Accessing the anti-Anxiety Effect by Inhibition of Locomotor Activity,” Acta Scientific Pharmaceutical Sciences 2 (2018): 40–4.
  • J. R. Atack, “GABAA Receptor Subtype-Selective Modulators. I. α2/α3-Selective Agonists as Non-Sedating Anxiolytics,” Current Topics in Medicinal Chemistry 11 (2011): 1176–202. DOI: 10.2174/156802611795371350.
  • S. A. Demchenko, O. E. Yadlovskyi, O. v Yudina, I. I. Tubaltseva, YuA. Fedchenkova, L. S. Bobkova, and АM. Demchenko, “Synthesis and Anxiolytic Activity of 3-Aryl-1-(41methoxyphenyl)-1-(6,7,8,9-Tetrahydro5h -[1,2,4] Triazolo[4,3-a]Azepine-3-yl-Methyl)- Urea Derivatives,” Pharmacology and Drug Toxicology 14 (2020): 88–96. DOI: 10.33250/14.02.088.
  • S. A. Hrnack, and F. A. Barber, “Managing the Pain of Knee Osteoarthritis,” The Physician and Sportsmedicine 42, no. 3 (2014): 63–70. DOI: 10.3810/psm.2014.09.2077.
  • P. Pirlamarla, and R. M. Bond, “FDA Labeling of NSAIDs: Review of Nonsteroidal anti-Inflammatory Drugs in Cardiovascular Disease,” Trends in Cardiovascular Medicine 26, no. 8 (2016): 675–80. DOI: 10.1016/j.tcm.2016.04.011.
  • P. Rao, and E. E. Knaus, “Evolution of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs): Cyclooxygenase (COX) Inhibition and Beyond,” Journal of Pharmacy & Pharmaceutical Sciences : a Publication of the Canadian Society for Pharmaceutical Sciences, Societe Canadienne Des Sciences Pharmaceutiques 11, no. 2 (2008): 81s–110s. DOI: 10.18433/J3T886.
  • S. A. Demchenko, H. O. Yeromina, L. O. Perekhoda, T. A. Bukhtiarova, L. S. Bobkova, and A. M. Demchenko, “Synthesis and Anаlgеsic Properties of (3-Allyl-4-Aryl-3H-Thiazol-2-Ylidene)-[4-(6,7,8,9-Tetrahydro-5H-[1,2,4]Triazolo[4,3-a]Azepin-3-yl)Phenyl]Amine Derivatives,” Farmatsevtychnyi Zhurnal 1, no. 1 (2018): 67–73. DOI: 10.32352/0367-3057.1.17.09.
  • S. Demchenko, N. Chalenko, T. Bukhtiarova, N. Seredynska, L. Bobkova, and O. Yadlovsky, “Synthesis and Analgesic Activity of 6,7,8,9-Tetrahydro-5H- [1,2,4] Triazolo [4,3-a]Azepines,” Pharmacology and Drug Toxicology 60 (2018): 25–31.
  • C. Guo, L. Wang, X. Li, S. Wang, X. Yu, K. Xu, Y. Zhao, J. Luo, X. Li, B. Jiang, et al, “Discovery of Novel Bromophenol-Thiosemicarbazone Hybrids as Potent Selective Inhibitors of Poly(ADP-ribose) Polymerase-1 (PARP-1) for Use in Cancer,” Journal of Medicinal Chemistry 62, no. 6 (2019): 3051–67. DOI: 10.1021/acs.jmedchem.8b01946.
  • L.-W. Wang, H.-J. Su, S.-Z. Yang, S.-J. Won, and C.-N. Lin, “New Alkaloids and A Tetraflavonoid from Cephalotaxus wilsoniana,” Journal of Natural Products 67, no. 7 (2004): 1182–5. DOI: 10.1021/np0498657.
  • H. Morita, M. Yoshinaga, and J. Kobayashi, “Cephalezomines G, H, J, K, L, and M, New Alkaloids from Cephalotaxus Harringtonia Var. nana,” Tetrahedron 58, no. 27 (2002): 5489–95. DOI: 10.1016/S0040-4020(02.)00521-5.
  • S. Li, X. Li, T. Zhang, M. O. Kamara, J. Liang, J. Zhu, and F. Meng, “Design, Synthesis and Biological Evaluation of Homoerythrina Alkaloid Derivatives Bearing a Triazole Moiety as PARP-1 Inhibitors and as Potential Antitumor Drugs,” Bioorganic Chemistry 94 (2020): 103385. DOI: 10.1016/j.bioorg.2019.103385.
  • R. H. Nelson, “Hyperlipidemia as a Risk Factor for Cardiovascular Disease,” Prim Care 40, no. 1 (2013): 195–211. DOI: 10.1016/j.pop.2012.11.003.
  • S. E.-S. Barakat, M. A A. El-Zahabi, A. A. Abdel-Rahman, A. H. Bayomi, H. E. Ali, M. A. Amin, M. A. El-Zahabi, and S. Barakat, “Synthesis of Certain New Dibenz[c,e]Azepine-5,7-Diones of Expected Antihyperlipidemic Activity,” Journal of King Abdulaziz University-Medical Sciences 14, no. 4 (2007): 3–17. DOI: 10.4197/Med.14-4.1.
  • M. Verma, A. K. Gupta, and R. N. Thakur, “Synthesis and Antihyperlipidemic Activity of Dibenz[c,e]Azepine-5,7-Dione Derivatives in Triton WR-1339-Induced Hyperlipidemic Rats,” Journal of Drug Delivery and Therapeutics 8, no. 2 (2018): 129–35. DOI: 10.22270/jddt.v8i2.1684.
  • Y. Ohsawa, and N. Hirasawa, “The Antagonism of Histamine H1 and H4 Receptors Ameliorates Chronic Allergic Dermatitis via anti-Pruritic and Anti-Inflammatory Effects in NC/Nga Mice,” Allergy 67, no. 8 (2012): 1014–22. DOI: 10.1111/j.1398-9995.2012.02854.x.
  • T. Somma, L. Cinci, G. Formicola, A. Pini, R. Thurmond, M. Ennis, D. Bani, and E. Masini, “A Selective Antagonist of Histamine H4 Receptors Prevents Antigen-Induced Airway Inflammation and Bronchoconstriction in guinea Pigs: Involvement of Lipocortin-1,” British Journal of Pharmacology 170, no. 1 (2013): 200–13. DOI: 10.1111/bph.12264.
  • I. Berrebi-Bertrand, X. Billot, T. Calmels, M. Capet, S. Krief, O. Labeeuw, J.-M. Lecomte, N. Levoin, X. Ligneau, P. Robert, and J.-C. Schwartz, “6,11-Dihydro-5h-benzodimidazo1,2- aiazepines Derivatives as Histamine h4 Receptor Ligands” (US 9.255,101 B2, 2016).
  • B. E. Blass, “Triazolo-Azepine Inhibitors of γ-Secretase and Their Methods of Use,” ACS Medicinal Chemistry Letters 10, no. 9 (2019): 1243–4. DOI: 10.1021/acsmedchemlett.9b00327.
  • S. P. Kollur, J. O. Castro, J. Frau, and D. Glossman-Mitnik, “Chemical Synthesis, Spectroscopic Studies, Chemical Reactivity Properties and Bioactivity Scores of an Azepin-Based Molecule,” Journal of Molecular Structure 1180 (2019): 300–6. DOI: 10.1016/j.molstruc.2018.11.061.

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.