Medicinal chemistry of indole derivatives: Current to future therapeutic prospectives

Bioorganic Chemistry - Tập 89 - Trang 103021 - 2019
Archana Kumari1, Rajesh K. Singh2
1Rayat-Bahra Institute of Pharmacy, Dist. Hoshiarpur, 146104, Punjab, India
2Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126 Punjab, India

Tóm tắt

Từ khóa


Tài liệu tham khảo

Kaushik, 2013, Biomedical importance of indoles, Molecules, 18, 6620, 10.3390/molecules18066620

Taber, 2011, Indole synthesis: a review and proposed classification, Tetrahedron, 67, 7195, 10.1016/j.tet.2011.06.040

Baeyer, 1886, Ueber die Reduction aromatischer Verbindungen mittelst Zinkstaub [On the reduction of aromatic compounds by means of zinc dust], Annalen der Chemie und Pharmacie, Eur. J. Organic Chem., 140, 295

Barluenga, 2011, Five-membered heterocycles: indole and related systems, Mod. Heterocycl. Chem., 377, 10.1002/9783527637737.ch5

P.M. Dewick. Essentials of organic chemistry: for students of pharmacy, medicinal chemistry and biological chemistry.

James, 1959, Indole-3-aldehyde, Organic Syntheses, 39, 30, 10.15227/orgsyn.039.0030

Heaney, 1974, 1-Benzylindole, Organic Syntheses, 54

Bergman, 1992, Efficient synthesis of 2-chloro-, 2-bromo-, and 2-iodoindole, J. Org. Chem., 57, 2495, 10.1021/jo00034a058

Katritzky, 1995, Facile synthesis of 2-substituted indoles and indolo[3,2-b] carbazoles from 2-(benzotriazol-1-ylmethyl) indole, J. Org. Chem., 60, 3401, 10.1021/jo00116a026

Ximenes, 2001, The oxidation of indole derivatives catalysed by horseradish peroxidase is highly chemiluminescent, Arch. Biochem. Biophys., 387, 173, 10.1006/abbi.2000.2228

Lynch, 2002, Intramolecular amidofuran cycloadditions across an indole pi-bond: an efficient approach to the aspidosperma and strychnos ABCE core, Org. Lett., 4, 4643, 10.1021/ol027024q

Sharma, 2010, Biological importance of the indole nucleus in recent years: A comprehensive review, J. Heterocyclic Chem., 47, 491, 10.1002/jhet.349

Chadha, 2017, Indoles as therapeutics of interest in medicinal chemistry: Bird's eye view, Eur. J. Med. Chem., 134, 159, 10.1016/j.ejmech.2017.04.003

Baudin, 1986, Synthesis of indoles from N-aryl-1-alkenylsulphinamides, Tetrahedron Lett., 27, 837, 10.1016/S0040-4039(00)84114-3

Fischer, 1883, Ueber die hydrazine der brenztraubens€aure, Berichte Dtsch. Chem. Ges., 16, 2241, 10.1002/cber.188301602141

Reissert, 1897, Einwirkung und natrium€athylat auf nitrotoluole. Synthese nitrirter phenyl brenz traubens€auren, Berichte Dtsch, Chem. Ges., 30, 1030, 10.1002/cber.189703001200

Bayer, 1869, Synthese des indoles [Synthesis of indoles], Berichte der Deutschen chemischen Gesellschaft., 2, 679, 10.1002/cber.186900201268

Larock, 1991, Synthesis of indoles via palladium-catalyzed heteroannulation of internal alkynes, J. Am. Chem. Soc., 113, 6689, 10.1021/ja00017a059

Bartoli, 1989, The reaction of vinyl Grignard reagents with 2-substituted nitroarenes: a new approach to the synthesis of 7-substituted indoles, Tetrahedron Lett., 30, 2129, 10.1016/S0040-4039(01)93730-X

Madelung, 1912, Über eine neue darstellungsweise für substituierte indole, i Berichte. Dtsch. Chem. Ges., 45, 1128, 10.1002/cber.191204501160

Fukuyama, 1994, A novel tin-mediated indole synthesis, J. Am. Chem. Soc., 116, 3127, 10.1021/ja00086a054

A.D. Batcho, W. Leimgruber, U.S. Patent 3,732,245 and U.S. Patent 3,976,639.

Zeidan, 2017, Palladium-catalyzed synthesis of 2-cyanoindoles from 2-gem-dihalovinylanilines, Org. Lett., 19, 5058, 10.1021/acs.orglett.7b02244

Shi, 2017, A C-H activation-based strategy for N-amino azaheterocycle synthesis, Org. Lett., 19, 4359, 10.1021/acs.orglett.7b02066

Yu, 2017, The development of a palladium-catalyzed tandem addition/cyclization for the construction of indole skeletons, J. Org. Chem., 82, 3631, 10.1021/acs.joc.7b00148

Wanga, 2017, Iron-catalyzed c3-formylation of indoles with formaldehyde and aqueous ammonia under air, Synlett., 28, 2670, 10.1055/s-0036-1589079

Miao, 2016, Cyclic anti-azacarboxylation of 2-alkynyl anilines with carbon dioxide, Org. Lett., 18, 2556, 10.1021/acs.orglett.6b00884

Hu, 2016, Rhodium (iii)-catalyzed cascade cyclization/electrophilic amidation for the synthesis of 3-amidoindoles and 3-amidofurans, Org. Lett., 18, 2058, 10.1021/acs.orglett.6b00689

Mizukami, 2016, Rhodium-catalyzed cyclization of 2-ethynylanilines in the presence of isocyanates: approach toward indole-3-carboxamides, Org. Lett., 18, 748, 10.1021/acs.orglett.6b00007

Wu, 2016, Synthesis of indoles from 2-vinylanilines with pifa or tfa and quinones, Synlett., 27

Sayyad, 2015, A synthetic route to 2-alkyl indoles via thiophenol-mediated ring-opening of N-tosylaziridines followed by copper powder-mediated C-N cyclization/aromatization, J. Org. Chem., 80, 12659, 10.1021/acs.joc.5b02251

Ortgies, 2015, Selenium-catalyzed oxidative C(sp(2))-H amination of alkenes exemplified in the expedient synthesis of (aza-)indoles, Org. Lett., 17, 2748, 10.1021/acs.orglett.5b01156

Li, 2015, Metal-free synthesis of indole via NIS-mediated cascade C-N bond formation/aromatization, J. Org. Chem., 80, 3841, 10.1021/acs.joc.5b00090

Michalska, 2015, Simple and mild synthesis of indoles via hydroamination reaction catalysed by NHC–gold complexes: looking for optimized conditions, Synlett, 10.1055/s-0035-1560976

Nallagonda, 2014, Synthesis of functionalized indoles via palladium-catalyzed aerobic oxidative cycloisomerizationof o-allylanilines, Org. Lett., 16, 4786, 10.1021/ol502244g

Jiao, 2014, Regioselective direct C-H alkylation of NH indoles and pyrroles by a Palladium/Nonbornene-cocatalysed process, Synthesis, 46, 0035

Hamid, 2017, Indole alkaloids from plants as potential leads for antidepressant drugs: A mini-review, Front Pharmacol., 8, 96, 10.3389/fphar.2017.00096

Shahab, 2009, Indole acetic acid production and enhanced plant growth promotion by indigenous PSBs, Afr. J. Agri. Res., 4, 1312

Murch, 2000, Tryptophan is a precursor for melatonin and serotonin biosynthesis in In vitro regenerated St. John's wort (Hypericum perforatum L. cv. Anthos) plants, Plant Cell Reports, 19, 698, 10.1007/s002990000206

Takayama, 2004, Chemistry and pharmacology of analgesic indole alkaloids from the rubiaceous plant, Mitragynaspeciosa, Chem. Pharm. Bull. (Tokyo), 52, 916, 10.1248/cpb.52.916

Andreas Moser (1998) Pharmacology of endogenous neurotoxins: a handbook. Braun-Brumfield. P. 131.

Brown, 2013, Ibogaine in the treatment of substance dependence, Curr. Drug. Abuse. Rev., 6, 3, 10.2174/15672050113109990001

Colović, 2013, Curr. Neuro. Pharmacol., 11, 315, 10.2174/1570159X11311030006

Negård, 2015, Links between genetic groups, indole alkaloids and ecology within the grass-parasitic Claviceps purpurea species complex, Toxins (Basel), 7, 1431, 10.3390/toxins7051431

Moudi, 2013, Vinca alkaloids, Int. J. Prev. Med., 4, 1231

Kumar, 2015, Structural characterization of monoterpene indole alkaloids in ethanolic extract of Rauwolfia species by liquid chromatography with quadrupole time of flight mass spectrometry, J. Pharm. Anal., 6, 363, 10.1016/j.jpha.2016.04.008

Liu, 2015, Antibacterial monoterpenoid indole alkaloids from Alstonia scholaris cultivated in temperate zone, Fitoterapia, 105, 160, 10.1016/j.fitote.2015.06.019

Shao, 2015, Cytotoxic indole alkaloids from the fruits of Melodinus cochinchinensis, Phytochemistry, 116, 367, 10.1016/j.phytochem.2015.02.028

Netz, 2015, Marine indole alkaloids, Mar. Drugs., 13, 4814, 10.3390/md13084814

Liu, 2017, Tulongicin and antibacterial triindole alkaloid from a deep water topsentia sp. Sponge, J. Nat. Prod., 80, 2556, 10.1021/acs.jnatprod.7b00452

Desai, 2016, Synthesis biological evaluation and molecular docking study of some novel indole and pyridine based 1.3.4-oxadiazole derivatives as potential antitubercular agents, Bioorg. Med. Chem. Lett., 26, 1776, 10.1016/j.bmcl.2016.02.043

Sakhuja, 2011, Design and synthesis of spiro [indole-thiazolidine] spiro [indole-pyrans] as antimicrobial agents, Bioorg. Med. Chem. Lett., 21, 5465, 10.1016/j.bmcl.2011.06.121

Atterhög, 1976, Experience with pindolol, a beta receptor blocker, in the treatment of hypertension, Am. J. Med., 60, 872, 10.1016/0002-9343(76)90907-4

London, 2004, REASON Project Investigators, Mechanism (s) of selective systolic blood pressure reduction after a low dose combination of perindopril/indapamide in hypertensive subjects: comparison with atenolol, J. Am. Coll. Cardiol., 43, 92, 10.1016/j.jacc.2003.07.039

Rezaiefar, 2002, Blood pressure and secondary prevention of strokes. How low should we go? Randomised trial of a perindopril-based blood-pressure lowering regimen among 6,105 individuals with previous stroke or transient ischaemic attack, Can. Fam. Physician, 48, 1625

Snyman, 2009, Perindopril: do randomised, controlled trials support an ACE inhibitor class effect? A meta-analysis of clinical trials, Cardiovasc. J. Afr., 20, 127

Køber, 1995, A clinical trial of the angiotensin-converting-enzyme inhibitor trandolapril in patients with left ventricular dysfunction after myocardial infarction, Trandolapril Cardiac Evaluation (TRACE) Study Group, N. Engl. J. Med., 333, 1670, 10.1056/NEJM199512213332503

Packer, 2001, Effect of carvedilol on survival in severe chronic heart failure, N. Engl. J. Med., 344, 1651, 10.1056/NEJM200105313442201

Chen, 2011, Recent advances in DAPYs and related analogues as HIV-1 NNRTIs, Curr. Med. Chem., 18, 359, 10.2174/092986711794839142

Jacobi, 1980, On the pharmacodynamics of acemetacin (author's transl), Arzneimittelforschung, 30, 1348

Dwivedi, 2015, Chem Biol. Drug. Des., 86, 1471, 10.1111/cbdd.12613

Vitaku, 2014, Analysis of the structural diversity, substitution patterns, and frequency of nitrogen heterocycles among U.S. FDA approved pharmaceuticals, J. Med. Chem., 57, 10257, 10.1021/jm501100b

Patil, 2017, Medicinal applications of (benz) imidazole- and indole-based macrocycles, Chem. Biol. Drug. Des., 89, 639, 10.1111/cbdd.12802

Goyal, 2018, Benzofuran and Indole: Promising Scaffolds for Drug Development in Alzheimer's Disease, Chem. Med. Chem., 13, 1275, 10.1002/cmdc.201800156

Zhang, 2015, A review on recent developments of indole-containing antiviral agents, Eur. J. Med. Chem, 89, 421, 10.1016/j.ejmech.2014.10.065

Dadashpour, 2018, Indole in the target-based design of anticancer agents: A versatile scaffold with diverse mechanisms, Eur. J. Med. Chem., 150, 9, 10.1016/j.ejmech.2018.02.065

Bariwal, 2018, Recent advances in spirocyclization of indole derivatives, Chem. Soc. Rev., 47, 3831, 10.1039/C7CS00508C

Chen, 2018, Recent progress in the synthesis of phosphorus-containing indole derivatives, Org. Biomol. Chem., 16, 7544, 10.1039/C8OB02100G

Ziarani, 2018, Recent advances in the application of indoles in multicomponent reactions, RSC Adv., 8, 12069, 10.1039/C7RA13321A

Singh, 2018, Recent progress in biological activities of indole and indole alkaloids, Mini Rev. Med. Chem., 18, 9

Sravanthi, 2016, Indoles-A promising scaffold for drug development, Eur. J. Pharm. Sci., 91, 1, 10.1016/j.ejps.2016.05.025

World Health Organization (WHO), Cancer Fact sheet http://www.who.int/news-room/fact-sheets/detail/cancer. Retrieved on 21-03-18.

Janani, 2015, PPAR gamma gene—a review, Diabetes Metab Syndr., 9, 46, 10.1016/j.dsx.2014.09.015

Nanjan, 2018, Thiazolidinediones as antidiabetic agents: a critical review, Bioorg Chem., 77, 548, 10.1016/j.bioorg.2018.02.009

Corigliano, 2018, Indole and 2,4-Thiazolidinedione conjugates as potential anticancer modulators, Peer J., 6, 5386, 10.7717/peerj.5386

Üstündağ, 2016, Indole-based hydrazide-hydrazones and 4-thiazolidinones: synthesis and evaluation as antitubercular and anticancer agents, J. Enzyme Inhib. Med. Chem., 31, 369

Demurtas, 2019, Indole derivatives as multifunctional drugs: Synthesis and evaluation of antioxidant, photoprotective and antiproliferative activity of indole hydrazones, Bioorg Chem., 85, 568, 10.1016/j.bioorg.2019.02.007

Prakash, 2018, Novel indole derivatives as potential anticancer agents: Design, synthesis and biological screening, Med. Chem. Res., 27, 321, 10.1007/s00044-017-2065-9

Tocco, 2017, Solvent-free addition of indole to aldehydes: unexpected synthesis of novel 1-[1-(1h-indol-3-yl) alkyl]-1h-indoles and preliminary evaluation of their cytotoxicity in hepatocarcinoma cells, Molecules, 22, 10.3390/molecules22101747

Romagnoli, 2017, Design, synthesis and biological evaluation of 3-substituted-2-oxindole hybrid derivatives as novel anticancer agents, Eur. J. Med. Chem., 134, 258, 10.1016/j.ejmech.2017.03.089

Honore, 2005, Understanding microtubule dynamics for improved cancer therapy, Cell. Mol. Life Sci., 62, 3039, 10.1007/s00018-005-5330-x

Lu, 2014, Design, synthesis and biological evaluation of stable colchicines binding site tubulin inhibitors as potential anticancer agents, J. Med. Chem., 57, 7355, 10.1021/jm500764v

Mirzaei, 2017, New indole-based chalconoids as tubulin-targeting antiproliferative agents, Bioorg. Chem., 75, 86, 10.1016/j.bioorg.2017.09.005

Lafayette, 2017, Synthesis of novel indole derivatives as promising DNA binding agents and evaluation of antitumor and antitopoisomerase I activities, Eur. J. Med. Chem., 136, 511, 10.1016/j.ejmech.2017.05.012

Zhou, 2016, Synthesis and cytotoxic activity of novel hexahydropyrrolo [2,3-b] indole imidazolium salts, Bioorg. Med. Chem. Lett., 26, 460, 10.1016/j.bmcl.2015.11.092

Chang, 2017, Novel indolizino[8,7-b] indole hybrids as anti-small cell lung cancer agents: regioselective modulation of topoisomerase II inhibitory and DNA cross-linking activities, Eur. J. Med. Chem., 127, 235, 10.1016/j.ejmech.2016.12.046

Bakherad, 2019, Anti-cancer anti-oxidant and molecular docking studies of thiosemicarbazone indole-based derivatives, Res. Chem. Intermediat., 45, 2827, 10.1007/s11164-019-03765-9

El-Shariefa, 2019, Design synthesis, molecular docking and biological activity evaluation of some novel indole derivatives as potent anticancer active agents and apoptosis inducers, Bioorg. Chem., 85, 399, 10.1016/j.bioorg.2019.01.016

Jiang, 2016, Synthesis and anticancer activity of 1-(1H-indol-3-yl)-2-(4-diarylmethylpiperazine-1-yl) ethane-1, 2-dione derivatives, J. Chem., 10.1155/2016/4617454

Yan, 2016, Synthesis, evaluation, and mechanism study of novel indole-chalcone derivatives exerting effective antitumor activity through microtubule destabilization in vitro and in vivo, J. Med. Chem., 59, 5264, 10.1021/acs.jmedchem.6b00021

Takahashi, 2006, Hypothesis: the antitumor activities of statins may be mediated by IL-18, J. Leukocyte Biol., 80, 215, 10.1189/jlb.0406245

Kumar, 2016, Cu-Catalyzed ligand-free synthesis of rosuvastatin based novel indole derivatives as potential anticancer agents, RSC Adv., 6, 100487, 10.1039/C6RA20148B

Hu, 2016, Synthesis, structure-activity relationship studies and biological evaluation of novel 2,5-disubstituted indole derivatives as anticancer agents, Chem. Biol. Drug. Des., 88, 766, 10.1111/cbdd.12808

Choppara, 2015, Synthesis characterization and cytotoxic investigations of novel bis(indole) analogues besides antimicrobial study, Arab. J. Chem., 10.1016/j.arabjc.2015.05.015

Kamath, 2015, Some new indole-coumarin hybrids; Synthesis, anticancer and Bcl-2 docking studies, Bioorg. Chem., 63, 101, 10.1016/j.bioorg.2015.10.001

Wang, 2014, Design, synthesis and biological evaluation of a series of pyrano chalcone derivatives containing indole moiety as novel anti-tubulin agents, Bioorg. Med. Chem., 22, 2060, 10.1016/j.bmc.2014.02.028

Kumar, 2014, Synthesis and identification of α-cyano bis (indolyl) chalcones as novel anticancer agents, Bioorg. Med. Chem. Lett., 24, 5170, 10.1016/j.bmcl.2014.09.085

Gali, 2015, One pot multicomponent synthesis of indole incorporated thiazolylcoumarins and their antibacterial, anticancer and DNA cleavage studies, Bioorg. Med. Chem. Lett., 25, 106, 10.1016/j.bmcl.2014.10.100

Pingaew, 2013, Synthesis and structure-activity relationship of mono-indole-, bis-indole-, and tris-indole-based sulfonamides as potential anticancer agents, Mol. Divers., 595–604

World Health Organization (WHO). Epilepsy, http://www.who.int/news-room/fact-sheets/detail/epilepsy, Retrieved on 20-04-2018.

Saini, 2015, Central nervous system activities of indole derivatives: An overview, Cent. Nerv. Syst. Agents Med. Chem., 16, 19, 10.2174/1871524915666150608103224

Swathi, 2015, Synthesis and antiepileptic activity of schiff's bases of dialkylamino alkoxy isatin derivatives, Adv. Exp. Med. Biol., 822, 119, 10.1007/978-3-319-08927-0_13

Osyanin, 2015, Synthesis of 4-(1H-Azol-1-ylmethyl) benzo hydrazides and Their Acyclic and Heterocyclic Derivatives, Russian J. Gen. Chem., 75, 111, 10.1007/s11176-005-0180-7

Madhira, 2017, Synthesis and evaluation of some novel N, N-dialkylaminoalkoxy-2-oxo-indole-3-ylidene benzohydrazides as anticonvulsant agents, IOSR-JPBS, 12, 84, 10.9790/3008-1202028493

Raju, 2016, Synthesis, characterization and anticonvulsant activity of novel benzyl-6-chloro indole carboxylate derivatives, J. Basic Appl. Res., 2, 437

Abdel-Aziz, 2009, Synthesis of novel pyrazole derivativeseir antidepressant and anticonvulsant activities, Eur. J. Med. Chem., 44, 3480, 10.1016/j.ejmech.2009.01.032

Khan, 2016, The therapeutic voyage of pyrazole and its analogs: A review, Eur. J. Med. Chem., 120, 170, 10.1016/j.ejmech.2016.04.077

Karrouchi, 2018, Synthesis and pharmacological activities of pyrazole derivatives: a review, Molecules, 23, 134, 10.3390/molecules23010134

Patil, 2013, Synthesis, characterization and screening for antidepressant and anticonvulsant activity of 4, 5-dihydropyrazole bearing indole derivatives, Arab. J. Chem., 9, 588, 10.1016/j.arabjc.2013.08.027

Zhen, 2015, Synthesis, potential anticonvulsant and antidepressant effects of 2-(5-methyl-2,3-dioxoindolin-1-yl)acetamide derivatives, Acta. Pharm. Sin. B., 5, 343, 10.1016/j.apsb.2015.01.008

Yar, 2014, Design and synthesis of new dual binding site cholinesterase inhibitors: in vitro inhibition studies with in silico docking, Lett. Drug Des. Discov., 11, 331, 10.2174/15701808113106660078

Ahuja, 2014, Anticonvulsant evaluation of clubbed indole-1,2,4-triazine derivatives: a synthetic approach, Eur. J. Med. Chem., 80, 509, 10.1016/j.ejmech.2014.04.043

Khan, 2013, Indole derivatives with anticonvulsant activity against two seizure models, Pharmacophore, 3, 55

Kumar, 2011, Synthesis and evaluation of some newer indole derivatives as anticonvulsant agents, Int. J. Pharm. Bio. Arch., 2, 744

Tabbi, 2016, Synthesis of novel thiazolylpyrazoline derivatives and evaluation of their antimicrobial activities and cytotoxicities, Turk. J. Chem., 40, 641, 10.3906/kim-1512-12

World Health Organization (WHO), http://www.who. int/ mediacentre/ news/ releases/ 2018/antibiotic-resistance-found/en/ Retrieved on 30-08-2018.

Sanna, 2018, Synthesis and biological evaluation of novel indole-derived thioureas, Molecules, 23, 2554, 10.3390/molecules23102554

Hafez, 2018, Synthesis and antimicrobial activity of new thiazolidine-based heterocycles as rhodanine, J. Heterocyclic Chem., 55, 685, 10.1002/jhet.3087

Abo-Ashour, 2018, Novel indole-thiazolidinone conjugates: Design, synthesis and whole-cell phenotypic evaluation as a novel class of antimicrobial agents, Eur. J. Med. Chem., 160, 49, 10.1016/j.ejmech.2018.10.008

Sayed, 2018, Synthesis of some heterocyclic compounds derived from indole as antimicrobial agents, Synth. Comm., 48, 413, 10.1080/00397911.2017.1403627

Gani, 2017, Chemo selective synthesis and evaluation of antimicrobial activity of novel 5-hydroxyindole derivatives, Der. Pharmacia Sinica, 8, 1

Mane, 2017, Design, synthesis, and antimicrobial activity of novel 5-substituted indole-2-carboxamide derivatives, Res. Chem. Intermediat., 43, 1253, 10.1007/s11164-016-2696-3

Badgujar, 2018, Synthesis, antimicrobial and antioxidant activity of pyrazole-based sulfonamide derivatives, Ind. J. Microbiol., 58, 93, 10.1007/s12088-017-0689-6

Faria, 2017, Recently reported biological activities of pyrazole compounds, Bioorg. Med. Chem., 25, 5891, 10.1016/j.bmc.2017.09.035

Skocibusic, 2018, Novel imidazole aldoximes with broad-spectrum antimicrobial potency against multidrug-resistant gram-negative bacteria, Molecules, 23, 1212, 10.3390/molecules23051212

Quazi, 2017, Synthesis and antimicrobial activity of indole derivative bearing the pyrazole moiety, IJPSR, 8, 1145

Rajaraman, 2017, Synthesis, molecular structure, DFT studies and antimicrobial activities of some novel 3-(1-(3,4-dimethoxyphenethyl)-4,5-diphenyl-1H-imidazol-2-yl)-1H-indole derivatives and its molecular docking studies, J. Mol. Str., 1127, 597, 10.1016/j.molstruc.2016.08.021

Yadav, 2016, Synthesis of some novel indole derivatives as potential antibacterial, antifungal and antimalarial agents, IOSR. J. Pharm., 6, 27

Popiołek, 2010, Hydrazide-hydrazones as potential antimicrobial agents: overview of the literature since, Med. Chem. Res., 26, 287

Neeraj, 2015, Antimicrobial potential of hydrazide-hydrazone derivatives: a review, Int. J. Pharm. Clinical Res., 7, 154

Shirinzadeh, 2011, Antimicrobial evaluation of indole-containing hydrazone derivatives, Z. Naturforsch C., 66, 340, 10.1515/znc-2011-7-804

Nassar, 2010, Synthesis, (in vitro) antitumor and antimicrobial activity of some pyrazoline, pyridine, and pyrimidine derivatives linked to indole moiety, J. Am. Sci., 6, 338

Zhang, 2012, Synthesis and fungicidal activity of novel pimprinine analogues, Eur. J. Med. Chem., 53, 283, 10.1016/j.ejmech.2012.04.012

Zhang, 2013, Synthesis and antifungal activity of 3-(1,3,4-oxadiazol-5-yl)-indoles and 3-(1,3,4-oxadiazol-5-yl)methyl-indoles, Eur. J. Med. Chem., 63, 22, 10.1016/j.ejmech.2013.01.038

Zhang, 2015, Synthesis and antifungal activity of novel streptochlorin analogues, Eur. J. Med. Chem., 92, 776, 10.1016/j.ejmech.2015.01.043

Zhang, 2017, Synthesis and antifungal activity of novel indole-replaced streptochlorin analogues, Eur. J. Med. Chem., 27, 669, 10.1016/j.ejmech.2016.12.001

Jia, 2018, An efficient synthesis and antifungal evaluation of natural product streptochlorin and its analogues, Fitoterapia, 125, 106, 10.1016/j.fitote.2017.12.017

Kaplancikli, 2005, Synthesis and antituberculosis activity of new 3-alkylsulfanyl-1, 2, 4- triazole derivatives, J. Enzyme Inhib. Med. Chem., 20, 179, 10.1080/14756360500043471

World Health Organization (WHO), http://www.who.int/en/news-room/fact-sheets / tail/tuberculosis. Retrieved 20-04-2018.

World Health Organization (WHO), http:// www. who. int/ tb/ publications / global_ report/en/. World Health Organization. Retrieved on 20-04-2018.

CDC. Reported tuberculosis in the United States, 2016. Atlanta, GA: US Department of Health and Human Services, CDC, 2017.

Trott, 2010, Auto Dock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading, J. Comput. Chem., 31, 455, 10.1002/jcc.21334

Khan, 2017, A facile synthesis of novel indole derivatives as potential antitubercular agents, J. Taib. Uni. Sci., 11, 910, 10.1016/j.jtusci.2016.09.002

Rohde, 2016, Synthesis and antitubercular activity of 1,2,4-trisubstitued piperazines, Bioorg. Med. Chem. Lett., 26, 2206, 10.1016/j.bmcl.2016.03.063

Patel, 2014, Design, synthesis and antitubercular evaluation of novel series of N-[4-(piperazin-1-yl) phenyl]cinnamamide derivatives, Eur. J. Med. Chem., 75, 43, 10.1016/j.ejmech.2014.01.024

Naidu, 2016, Sekhar, Seeking potent anti-tubercular agents: Design, synthesis, anti-tubercular activity and docking study of various (triazoles/indole)-piperazin-1-yl/1, 4-diazepan-1-yl) benzo [d] isoxazole derivatives, Bioorg. Med. Chem. Lett., 26, 2245, 10.1016/j.bmcl.2016.03.059

Stec, 2016, Indole-2-carboxamide-based mmpl3 inhibitors show exceptional antitubercular activity in an animal model of tuberculosis infection, J. Med. Chem., 59, 6232, 10.1021/acs.jmedchem.6b00415

Schrödinger, LLC, New York, NY, 2015.

Khan, 2018, Porous CuO catalyzed green synthesis of some novel 3-alkylated indoles as potent antitubercular agents, J. Saudi Chem. Soc., 22, 6, 10.1016/j.jscs.2016.03.009

Mandewale, 2015, Synthesis and biological evaluation of new hydrazone derivatives of quinoline and their Cu(II) and Zn(II) complexes against Mycobacterium tuberculosis, Bioinorg. Chem. Appl., 201

Thomas, 2014, Novel mannich bases of 4-thiazolidinone derivatives as antitubercular agents, IJRPC., 4, 351

Mustapha, 2017, A review on quinoline hydrazone derivatives as a new class of potent antitubercular and anticancer agents, Beni-Suef Univ. J. Basic Appl. Sci., 6, 354, 10.1016/j.bjbas.2017.07.005

Kondreddi, 2013, Design, synthesis, and biological evaluation of indole-2-carboxamides: a promising class of antituberculosis agents, J. Med. Chem., 56, 8849, 10.1021/jm4012774

Tehrani, 2014, Synthesis and antimycobacterial activity of novel thiadiazolylhydrazones of 1-substituted indole-3-carboxaldehydes, Chem. Biol. Drug Des., 83, 224, 10.1111/cbdd.12230

Selvaretnam, 2016, A review of concurrent infections of malaria and dengue in Asia, Asian Pacific J. Trop. Biomedicine, 6, 633, 10.1016/j.apjtb.2016.05.008

World Health Organization (WHO), http:// apps. who. int/ iris/ bitstream/ handle/ 10665/259492/9789241565523-eng.pdf;jsessionid=D585F2C066C6B97CEA476DF1B59DC6A D?sequence=1 Retrieved on 30-08-2018.

Santos, 2015, Exploring the 3-piperidin-4-yl-1H-indole scaffold as a novel antimalarial chemotype, Eur J. Med. Chem., 102, 320, 10.1016/j.ejmech.2015.07.047

Schuck, 2014, Synthetic indole and melatonin derivatives exhibit antimalarial activity on the cell cycle of the human malaria parasite Plasmodium falciparum, Eur. J. Med. Chem., 78, 375, 10.1016/j.ejmech.2014.03.055

Singh, 2014, A brief history of quinoline as antimalarial agents, Int. J. Pharm. Sci. Rev. Res., 25, 295

Golden, 2015, Quinoline-based antimalarial drugs: a novel class of autophagy inhibitors, Neurosurg. Focus., 38, 10.3171/2014.12.FOCUS14748

Teguh, 2013, Novel conjugated quinoline-indoles compromise plasmodium falciparum mitochondrial function and show promising antimalarial activity, J. Med. Chem., 56, 6200, 10.1021/jm400656s

Bharate, 2013, Meridianin G and its analogs as antimalarial agents, Med. Chem. Comm., 4, 1042, 10.1039/c3md00097d

Koren, 2018, new treatments in diabetes mellitus - an update, Harefuah, 157, 318

World Health Organization (WHO), http:// apps. who. int/iris/ bitstream/ handle/ 10665/ 272433/9789241550284-eng.pdf?ua=1 Retrieved on 30-08-2018.

Kavitha, 2017, Synthesis, characterization and biological evaluation of novel 2,5 substituted-1,3,4 oxadiazole derivatives, Saudi Pharm. J., 25, 337, 10.1016/j.jsps.2016.07.004

Kinfe, 2013, Evaluation of the Influence of thio semicarbazone-triazole hybrids on genes implicated in lipid oxidation and accumulation as potential anti-obesity agents, Bioorg. Med. Chem. Lett., 23, 5275, 10.1016/j.bmcl.2013.08.028

Rajan, 2018, Novel indole and triazole based hybrid molecules exhibit potent anti-adipogenic and antidyslipidemic activity by activating Wnt3a/β-catenin pathway, Eur. J. Med. Chem., 143, 1345, 10.1016/j.ejmech.2017.10.034

Nazir, 2018, New indole-based hybrid oxadiazole scaffolds with N-substituted acetamides: As potent anti-diabetic agents, Bioorg. Chem., 81, 253, 10.1016/j.bioorg.2018.08.010

Lonkala Srividya, 2017, Antidiabetic activity of 1-(4-chloro benzy lidene)-5-(2-oxoindolin-3-ylidene) thiocarbohydrazone in chick model, Asian, J. Biol. Sci., 10, 126

Taha, 2017, Synthesis α-glucosidase inhibitory activity and insilico study of tris- indole hybrid scaffold with oxadiazole ring: As potential leads for the management of type-II diabetes mellitus, Bioorg. Chem., 74, 30, 10.1016/j.bioorg.2017.07.009

Nomura, 2014, Novel indole-N-glucoside, TA-1887 as a sodium glucose cotransporter 2 inhibitor for treatment of type 2 diabetes, ACS. Med. Chem. Lett., 5, 51, 10.1021/ml400339b

Xu, 2012, Design, synthesis and biological evaluation of thiazole-and indole-based derivatives for the treatment of type II diabetes, Eur. J. Med. Chem., 52, 70, 10.1016/j.ejmech.2012.03.006

https://www.hiv.gov/hiv-basics/overview/data-and-trends/statistics. Retrieved on 22-04-2018.

Scuotto, 2016, Discovery of novel multi-target indole-based derivatives as potent and selective inhibitors of chikungunya virus replication, Bioorg. Med. Chem., 25, 327, 10.1016/j.bmc.2016.10.037

Jiang, 2006, Design synthesis and biological evaluations of novel oxindoles as HIV-1 non-nucleoside reverse transcriptase inhibitors. Part I, Bioorg. Med. Chem. Lett., 16, 2109, 10.1016/j.bmcl.2006.01.066

Chen, 2017, Expanding indole diversity: direct 1-step synthesis of 1,2-fused indoles and spiroindolines from 2-halo anilines for fast SAR antiviral elucidation against tobacco mosaic virus (TMV), Mol. Divers., 21, 61, 10.1007/s11030-016-9697-4

Musella, 2016, Identification of an indole-based derivative as potent and selective Varicella-zoster virus (VZV) inhibitor, Eur. J. Med. Chem., 124, 773, 10.1016/j.ejmech.2016.09.014

Hamad, 2010, Amino acid derivatives, part 4: synthesis and anti-HIV activity of new naphthalene derivatives, Arch. Pharm (Weinheim), 343, 397, 10.1002/ardp.200900293

Giapieri, 2009, Antiviral activity of indole derivatives, Antiviral Res., 83, 179, 10.1016/j.antiviral.2009.05.001

Soumya, 2011, Opportunistic diseases as a consequence of HIV/AIDS, J. AIDS Clinic. Res., 2, 133

https://data.unicef.org/topic/hivaids/global-regional-trends/. Retrieved on 24-04-2018.

Dousson, 2016, Discovery of the Aryl-phospho-indole IDX899, a Highly Potent Anti-HIV Non-nucleoside Reverse Transcriptase Inhibitor, J. Med. Chem., 59, 1891, 10.1021/acs.jmedchem.5b01430

Ravichandran, 2016, Exploring the structural insights of Indole-7-carboxamides as anti-HIV agents, FARMACIA, 64, 745

Lu, 2015, Design, synthesis, and biological evaluation of 1-(thiophen-2-yl)-9H-pyrido [3,4-b]indole derivatives as anti-HIV agents, Chem. Biol. Drug Des., 85, 722, 10.1111/cbdd.12456

Jiang, 2014, Design, synthesis, and biological evaluation of novel trifluoromethyl indoles as potent HIV-1 NNRTIs with an improved drug resistance profile, Org. Biomol. Chem., 12, 3446, 10.1039/C3OB42186D

Ferro, 2014, Synthesis and biological evaluation of novel antiviral agents as protein-protein interaction inhibitors, J. Enzyme Inhib. Med. Chem., 29, 237, 10.3109/14756366.2013.766609

Hassam, 2012, Novel Cyclopropyl-Indole Derivatives as HIV Non-Nucleoside Reverse Transcriptase Inhibitors, ACS Med. Chem. Lett., 3, 470, 10.1021/ml3000462

Balupuri, 2014, In silico study on indole derivatives as anti HIV-1 agents: a combined docking, molecular dynamics and 3D-QSAR study, Arch. Pharm. Res., 37, 1001, 10.1007/s12272-013-0313-1

Yeung, 2013, Inhibitors of HIV-1 attachment. Part 7: Indole-7-carboxamides as potent and orally bioavailable antiviral agents, Bioorg. Med. Chem. Lett., 23, 198, 10.1016/j.bmcl.2012.10.115

La Regina, 2012, New nitrogen containing substituents at the indole-2-carboxamide yield high potent and broad spectrum indolylarylsulfone HIV-1 non-nucleoside reverse transcriptase inhibitors, J. Med. Chem., 55, 6634, 10.1021/jm300477h

La Regina, 2011, Indolylarylsulfones as HIV-1 nonnucleoside reverse transcriptase inhibitors: new cyclic substituents at indole-2-carboxamide, J. Med. Chem., 54, 1587, 10.1021/jm101614j

Lamie, 2016, Novel N-substituted indole Schiff bases as dual inhibitors of cyclooxygenase-2 and 5-lipoxygenase enzymes: Synthesis, biological activities in vitro and docking study, Eur. J. Med. Chem., 123, 803, 10.1016/j.ejmech.2016.08.013

Mukthung, 2018, Capsaicin derivatives containing indole and nitroindole for improved anti-inflammatory activity, Naresuan Univ. J.: Sci. Technol. (NUJST), 26, 157

Bhat, 2018, indole derivatives as cyclooxygenase inhibitors: synthesis biological evaluation and docking studies, Molecules, 23, 10.3390/molecules23061250

Shaker, 2018, Design, synthesis, and biological evaluation of 2-(4-(methylsulfonyl) phenyl) indole derivatives with promising COX-2 inhibitory activity, J. Appl. Pharma. Sci., 8, 001, 10.7324/JAPS.2018.81101

Torres, 2004, Cyclooxygenase-mediated generation of free radicals during hypoxia in the cerebral cortex of newborn piglets, Neurochem. Res., 29, 1825, 10.1023/B:NERE.0000042208.20730.23

Mayo, 2005, Anti-inflammatory actions of melatonin and its metabolites, N1-acetyl-N2-formyl-5-methoxy kynura mine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK), in macrophages, J. Neuroimmunol., 165, 139, 10.1016/j.jneuroim.2005.05.002

Abdellatif, 2016, 3-Methyl-2-phenyl-1-substituted-indole derivatives as indomethacin analogs: design, synthesis and biological evaluation as potential anti-inflammatory and analgesic agents, J. Enzyme Inhib. Med. Chem., 31, 318, 10.3109/14756366.2015.1022174

Shroff, 2017, Newer substituted indolyl-pyrazoline derivatives as anti- inflammatory agents, Br. Biomed. Bull., 5, 298

Fatahala, 2017, Synthesis and structure activity relationship of some indole derivatives as potential anti-inflammatory agents, Acta. Chim. Slov., 64, 865, 10.17344/acsi.2017.3481

Liu, 2016, G, Liang, Design, synthesis, and structure-activity relationship study of novel indole-2-carboxamide derivatives as anti-inflammatory agents for the treatment of sepsis, J. Med. Chem., 59, 4637, 10.1021/acs.jmedchem.5b02006

Özdemir, 2015, İ, Synthesis and evaluation of new indole-based chalcones as potential antiinflammatory agents, Eur. J. Med. Chem., 89, 304, 10.1016/j.ejmech.2014.10.056

Matsuzawa, 2005, ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity, Nat. Immunol., 6, 587, 10.1038/ni1200

Shaveta, 2014, Rational design, synthesis and evaluation of chromone-indole and chromone-pyrazole based conjugates: Identification of a lead for anti-inflammatory drug, Eur. J. Med. Chem., 77, 185, 10.1016/j.ejmech.2014.03.003

Sharath, 2013, Synthesis of novel indole based scaffolds holding pyrazole ring as anti-inflammatory and antioxidant agents, J. Pharm. Res., 6, 785

Varaprasad, 2012, Synthesis and biological evaluation of 2-aryl-3-isoxazolinyl-indole derivatives as anti-inflammatory agents, IOSRJ. Appl. Chem., 2, 44, 10.9790/5736-0234449

Guerra, 2011, Anti-inflammatory and antinociceptive activities of indole-imidazolidine derivatives, Int. Immunopharmacol., 11, 1816, 10.1016/j.intimp.2011.07.010

Chandra, 2010, Synthesis and anti-inflammatory activity of indole derivatives, Int. J. Chem. Tech. Res., 2, 762

Huang, 2017

World Health Organization (WHO). http:// www. who. int/ mental_ health/ management/depression/en/. Retrieved on 25-05-2018.

D.J. Brody, L.A. Pratt, J.P. Hughes, Prevalance of depression among adults aged 20 and over united states, NCHS Data Brief (303), 1-8 (2018) 2013-2016.

Thomas, 2016, synthesis and biological evaluation of schiff’s bases and 2-azetidinones of isonocotinyl hydrazone as potential antidepressant and ionotropic agents, Arab. J. Chem., 9, 79, 10.1016/j.arabjc.2011.02.015

Kerzare, 2018, Synthesis, characterization, antidepressant activity and docking studies of some novel indole bearing azetidinone derivatives, Ind. J. Pharm. Edu. Res., 52, 110, 10.5530/ijper.52.1.13

Avinash, 2018, Derivatives of 4,5-dihydro (1H) pyrazoles as possible MAO-A inhibitors in depression and anxiety disorders: synthesis, biological evaluation and molecular modeling studies, Med. Chem. Res., 27, 1485, 10.1007/s00044-018-2167-z

P.O. Patil, S.B. Bari, Synthesis and antidepressant activity of some new 5-(1H-indol-3-yl)-3-(substituted aryl)-4,5-dihydroisoxazoline derivatives, J. Chem. 2013, Article ID 637205, http://doi.org/10.1155/2013/637205.

Di Matteo, 2003, Biochemical and therapeutic effects of antioxidants in the treatment of Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, CNS Neurol. Disord., 2, 95, 10.2174/1568007033482959

Rao, 2002, Role of oxidative stress and antioxidants in neurodegenerative diseases, Nutr. Neurosci., 5, 291, 10.1080/1028415021000033767

Gurer-Orhan, 2016, Novel indole-based melatonin analogues: Evaluation of antioxidant activity and protective effect against amyloid β-induced damage, Bioorg. Med. Chem., 24, 1658, 10.1016/j.bmc.2016.02.039

Silveira, 2013, Synthesis and antioxidant activity of new C-3 sulfenyl indoles, Tetrahedron Letters, 54, 4926, 10.1016/j.tetlet.2013.07.004

Baytas, 2012, Synthesis and antioxidant and antimicrobial evaluation of novel 4-substituted-1H-1,2,4-triazole derivatives, Turk. J. Chem., 36, 867

Suzen, 2007, Antioxidant activities of synthetic indole derivatives and possible activity mechanisms, Top. Heterocycl. Chem., 11, 145, 10.1007/7081_2007_074

Ölgen, 2011, Evaluation of novel amino methyl indole derivatives as Src kinase inhibitors and antioxidant agents, Chemotherapy, 57, 1, 10.1159/000317764

Eren, 2010, Novel indole-based analogs of melatonin: synthesis and in vitro antioxidant activity studies, Molecules, 15, 2187, 10.3390/molecules15042187

Estevão, 2010, Antioxidant activity of unexplored indole derivatives: synthesis and screening, Eur. J. Med. Chem., 45, 4869, 10.1016/j.ejmech.2010.07.059

Pallavi, 2017, Hepatitis: a review on current and future scenario, J. In Silico In Vitro Pharmacol., 3, 15

What is hepatitis. http://origin.who.int/features/qa/76/en. World Health Organization (WHO). Retrieved on 30-08-2018.

http://www.who.int/hepatitis/en/. World Health Organization (WHO). Retrieved on 30-08-2018.

Pyenson, 2018, Newly diagnosed hepatitisC inthe us commercially insured population beforeand after the2012 implementation of expanded screening guidelines, Am. Health Drug Benefits, 11, 30

Zoidis, 2015, Novel indole-flutamide heterocycles with activity against influenza PA endonuclease and hepatitis C virus, Med. Chem. Commun., 7, 447, 10.1039/C5MD00439J

Han, 2016, The discovery of indole derivatives as novel hepatitis C virus inhibitors, Eur. J. Med. Chem., 116, 147, 10.1016/j.ejmech.2016.03.062

Andreev, 2015, Discovery of the 2-phenyl-4, 5, 6, 7-tetrahydro-1H-indole as a novel anti-hepatitis C virus targeting scaffold, Eur. J. Med. Chem., 96, 250, 10.1016/j.ejmech.2015.04.022

Zhang, 2016, Discovery of 2-(4-sulfonamidophenyl)-indole 3-carboxamides as potent and selective inhibitors with broad hepatitis C virus genotype activity targeting HCV NS4B, Bioorg. Med. Chem. Lett., 26, 594, 10.1016/j.bmcl.2015.11.065

Varun, 2014, Novel indole derivatives as hepatitis C virus NS5B polymerase inhibitors: Pharmacophore modeling and 3D QSAR studies, Bangladesh, J. Pharmacol., 9, 290

Jin, 2014, Chemical genetics-based discovery of indole derivatives as HCV NS5B polymerase inhibitors, Eur. J. Med. Chem., 75, 413, 10.1016/j.ejmech.2014.01.062

Mitropoulos, 2010, New World cutaneous leishmaniasis: updated review of current and future diagnosis and treatment, J. Am. Acad. Dermatol., 63, 309, 10.1016/j.jaad.2009.06.088

Oryan, 2016, Worldwide risk factors in leishmaniasis, Asian Pac. J. Trop. Med., 9, 925, 10.1016/j.apjtm.2016.06.021

Porwal, 2017, gem-Dithioacetylated indole derivatives as novel antileishmanial agents, Bioorg. Med. Chem. Lett., 27, 4643, 10.1016/j.bmcl.2017.09.018

Félix, 2016, Antileishmanial activity of new thipheneindole hybrids: Junior, Design, synthesis, biological and cytotoxic evaluation, and chemometric studies, Bioorg. Med. Chem., 24, 3972, 10.1016/j.bmc.2016.04.057

Sharma, 2014, Triazino indole-quinoline hybrid: a novel approach to antileishmanial agents, Bioorg. Med. Chem. Lett., 24, 298, 10.1016/j.bmcl.2013.11.018

Bharate, 2013, Discovery of 3,3'-diindolylmethanes as potent antileishmanial agents, Eur. J. Med. Chem., 63, 435, 10.1016/j.ejmech.2013.02.024

Singh, 2012, Synthesis of N-(1-methyl-1H-indol-3-yl) methyleneamines and 3,3-diaryl-4-(1-methyl-1H-indol-3-yl)azetidin-2-ones as potential antileishmanial agents, Bioorg. Med. Chem. Lett., 22, 5704, 10.1016/j.bmcl.2012.06.081

Chini, 2009, CD38 as a regulator of cellular NAD: a novel potential pharmacological target for metabolic conditions, Curr. Pharm. Des., 15, 57, 10.2174/138161209787185788

Wu, 2013, Synthesis and activity of novel indole derivatives as inhibitors of CD38, Acta. Pharmaceutica. Sinica. B., 3, 245, 10.1016/j.apsb.2013.05.003

Chang, 2009, An updated review of tyrosinase inhibitors, Int. J. Mol. Sci., 10, 2440, 10.3390/ijms10062440

Ferro, 2016, Searching for indole derivatives as potential mushroom tyrosinase inhibitors, J. Enzyme Inhib. Med. Chem., 31, 398

Moss, 1948, The inhibition of lysozyme activity, American J. Digestive Dis., 15, 412, 10.1007/BF03001591

Shinitzky, 1966, Inhibition of lysozyme by imidazole and indole derivatives, Arch. Biochem. Biophys., 116, 332, 10.1016/0003-9861(66)90039-7

Girish, 2009, Hyaluronidase inhibitors: a biological and therapeutic perspective, Curr. Med. Chem., 16, 2261, 10.2174/092986709788453078

Olgen, 2010, Investigation of aminoethyl indole derivatives as hyaluronidase inhibitors, Z. Naturforsch. C., 65, 445, 10.1515/znc-2010-7-805

Olgen, 2007, New potent indole derivatives as hyaluronidase inhibitors, Chem. Biol. Drug. Des., 70, 547, 10.1111/j.1747-0285.2007.00590.x

Čolović, 2013, Acetylcholinesterase inhibitors: pharmacology and toxicology, Curr. Neuropharmacol., 11, 315, 10.2174/1570159X11311030006

Parveen, 2018, Noropacursane: A new nortriterpenoid from the methanolic extract of Carissa opaca, Rec. Nat. Prod., 12, 512, 10.25135/rnp.42.17.09.153

Mohammadi-Farani, 2013, Synthesis, docking and acetylcholinesterase inhibitory assessment of 2-(2-(4-Benzylpiperazin-1-yl)ethyl)isoindoline-1,3-dione derivatives with potential anti-Alzheimer effects, DARU, 21, 47, 10.1186/2008-2231-21-47

Norrby, 1995, Evidence of a dual role of endogenous histamine in angiogenesis, Int. J. Exp. Pathol., 76, 87

Jr Santillan, 2010, Indole-and benzothiophene-based histamine H3 antagonists, Bioorg. Med. Chem. Lett., 20, 6226, 10.1016/j.bmcl.2010.08.103

Madasu, 2012, Synthesis of compounds related to the anti-migraine drug eletriptan hydrobromide, Beilstein. J. Org. Chem., 8, 1400, 10.3762/bjoc.8.162

Swathi, 2010, Design, synthesis and biological evaluation of 5-[2(3)-dialkylaminoalkoxy] indole 2, 3-diones as new antihistamine agents, J. Chem. Pharm. Res., 2, 220

Agarwal, 2010, Antifertility effects of herbs: Need for responsible reporting, J. Ayurveda Integr. Med., 1, 129, 10.4103/0975-9476.65092

Bhowal, 2008, Synthesis and assessment of fertility-regulating potential of 2-(2''-chloroacetamidobenzyl)-3-(3'-indolyl) quinoline in adult rats as a male contraceptive agent, Contraception, 77, 214, 10.1016/j.contraception.2007.09.014

Collins-Burow, 2000, Estrogenic and anti-estrogenic activities of flavonoid phytochemicals through estrogen receptor binding-dependent and-independent mechanisms, Nutr. Cancer., 38, 229, 10.1207/S15327914NC382_13

Ji, 2005, Benzothieno [3,2-b]indole derivatives as potent selective estrogen receptor modulators, Bioorg. Med. Chem. Lett., 15, 2891, 10.1016/j.bmcl.2005.03.111

Cassier, 2013, A phase II trial of panobinostat in patients with advanced pretreated soft tissue sarcoma. A study from the French Sarcoma Group, Br. J. Cancer., 109, 909, 10.1038/bjc.2013.442

Ellis, 2009, The histone deacetylase inhibitors LAQ824 and LBH589 do not require death receptor signaling or a functional apoptosome to mediate tumor cell death or therapeutic efficacy, Blood, 114, 380, 10.1182/blood-2008-10-182758

Balasubramanian, 2008, A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas, Leukemia, 22, 1026, 10.1038/leu.2008.9

Cabral, 2013, Targeted therapy of spontaneous murine pancreatic tumors by polymeric micelles prolongs survival and prevents peritoneal metastasis, Proc. Natl. Acad. Sci. USA, 110, 11397, 10.1073/pnas.1301348110

Chen, 2016, The novel EZH2 inhibitor, GSK126, suppresses cell migration and angiogenesis via down-regulating VEGF-A, Cancer Chemother. Pharmacol., 77, 757, 10.1007/s00280-016-2990-1

Knutson, 2012, A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells, Nat. Chem. Biol., 8, 890, 10.1038/nchembio.1084

https://pubchem.ncbi.nlm.nih.gov/compound/21769#section=Top. Retrieved on 30-08-2018.

https://pubchem.ncbi.nlm.nih.gov/compound/24474#section=Top. Retrieved on 30-07-2018.

Foote, 2013, Discovery of 4-{4-[(3R)-3-Methylmorpholin-4-yl]-6-[1-(methylsulfonyl) cyclopropyl] pyrimidin-2-yl}-1H-indole (AZ20): A potent and selective inhibitor of ATR protein kinase with monotherapy in vivo antitumor activity, J. Med. Chem., 56, 2125, 10.1021/jm301859s

Martínez-Fernández, 2015, EZH2 in bladder cancer, a promising therapeutic target, Int. J. Mol. Sci., 16, 27107, 10.3390/ijms161126000

Vaswani, 2016, J. Med. Chem., 59, 9928, 10.1021/acs.jmedchem.6b01315

Bradley, 2014, EZH2 inhibitor efficacy in Non-Hodgkin’s lymphoma does not require suppression of H3K27 monomethylation, Chem. Biol., 21, 1463, 10.1016/j.chembiol.2014.09.017

Tanaka, 2015, Inhibitors of emerging epigenetic targets for cancer therapy: a patent review (2010–2014), Pharm. Pat. Anal., 4, 261, 10.4155/ppa.15.16

Kudo, 2018, Orantinib versus placebo combined with transcatheter arterial chemo embolisation in patients with unresectable hepatocellular carcinoma (ORIENTAL): a randomised, double-blind, placebo-controlled, multicentre, phase 3 study, Lancet Gastroenterol. Hepatol., 3, 37, 10.1016/S2468-1253(17)30290-X

Scagliotti, 2012, International, randomized, placebo-controlled, double-blind phase III study of motesanib plus carboplatin/paclitaxel in patients with advanced nonsquamous non-small-cell lung cancer, J. Clin. Oncol., 30, 2829, 10.1200/JCO.2011.41.4987

Infante, 2012, Safety, pharmacokinetic, and pharmacodynamic phase I dose-escalation trial of PF-00562271, an inhibitor of focal adhesion kinase, in advanced solid tumors, J. Clin. Oncol., 30, 1527, 10.1200/JCO.2011.38.9346

Friman, 2011, Sotrastaurin, a novel small molecule inhibiting protein-kinase C: randomized phase II study in renal transplant recipients, Am. J. Transplant., 11, 1444, 10.1111/j.1600-6143.2011.03538.x

Parikh, 2010, Phase II study of obatoclax mesylate (GX15-070), a small-molecule BCL-2 family antagonist, for patients with myelofibrosis, Clin. Lymphoma Myeloma Leuk., 10, 285, 10.3816/CLML.2010.n.059

Jones, 2013, The novel anticancer agent JNJ-26854165 induces cell death through inhibition of cholesterol transport and degradation of ABCA, J. Pharmacol. Exp. Ther., 346, 381, 10.1124/jpet.113.204958

Condon, 2014, a smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies, J. Med. Chem., 57, 3666, 10.1021/jm500176w

Axten, 2012, Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl) phenyl] acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), J. Med. Chem., 55, 7193, 10.1021/jm300713s

Axten, 2013, Discovery of GSK2656157: an optimized perk inhibitor selected for preclinical development, ACS. Med. Chem. Lett., 4, 964, 10.1021/ml400228e

Crump, 2016, Randamised double blind, phase III trial of enzastaurin versus placebo in patients achieving remission after first-line therapy for high-risk diffuse large b-cell lymphoma, J. Clin. Oncol., 34, 2484, 10.1200/JCO.2015.65.7171

Koivunen, 2004, Protein kinase C alpha/beta inhibitor Go6976 promotes formation of cell junctions and inhibits invasion of urinary bladder carcinoma cells, Cancer Res., 64, 5693, 10.1158/0008-5472.CAN-03-3511

Ihnen, 2013, Therapeutic potential of the poly (ADP-ribose) polymerase inhibitor rucaparib for the treatment of sporadic human ovarian cancer, Mol. Cancer Ther., 12, 1002, 10.1158/1535-7163.MCT-12-0813

Weng, 2008, Indole-3-carbinol as a chemopreventive and anti-cancer agent, Cancer Lett., 262, 153, 10.1016/j.canlet.2008.01.033

Katayama, 2013, Cytotoxic activity of tivantinib (ARQ 197) is not due solely to c-MET inhibition, Cancer Res., 73, 3087, 10.1158/0008-5472.CAN-12-3256

Yun, 2014, Novel 2,4-dianilino-5-fluoro pyrimidine derivatives possessing ALK inhibitory activities, Arch. Pharm. Res., 37, 873, 10.1007/s12272-013-0247-7

Huang, 2014, Discovery of highly potent p53-MDM2 antagonists and structural basis for anti-acute myeloid leukemia activities, ACS. Chem. Biol., 9, 802, 10.1021/cb400728e

Christensen, 2014, Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor, Cancer Cell, 26, 909, 10.1016/j.ccell.2014.10.019

Rosser, 1994, In vitro protein-binding characteristics of atevirdine and its N-dealkylated metabolite, Antiviral Res., 25, 193, 10.1016/0166-3542(94)90003-5

Sgadari, 2011, Pharmacological management of Kaposi's sarcoma, Expert Opin. Pharmacother., 12, 1669, 10.1517/14656566.2011.577066

Ashkenazi, 1983, Effects of LM 5008, a selective inhibitor of 5-hydroxytryptamine uptake, on blood pressure and responses to sympathomimetic amines, Br. J. Pharmacol., 79, 915, 10.1111/j.1476-5381.1983.tb10536.x

Sánchez, 2000, The selective sigma2 ligand Lu 28–179 has an antidepressant-like profile in the rat chronic mild stress model of depression, Behav Pharmacol., 11, 117, 10.1097/00008877-200004000-00003

Wetzel, 1994, Roxindole, a dopamine autoreceptor agonist, in the treatment of positive and negative schizophrenic symptoms, Am. J. Psychiatry, 151, 1499, 10.1176/ajp.151.10.1499

Zala, 2012, Safety and efficacy of GSK2248761, a next-generation non nucleoside reverse transcriptase inhibitor, in treatment-naive HIV-1-infected subjects, Antimicrob. Agents Chemother., 56, 2570, 10.1128/AAC.05597-11

Geiger, 2016, The STAT3 pathway as a therapeutic target in head and neck cancer: Barriers and innovations, Oral Oncol., 56, 84, 10.1016/j.oraloncology.2015.11.022

Beaulieu, 1941, Discovery of the first thumb pocket 1 NS5B polymerase inhibitor (BILB with demonstrated antiviral activity in patients chronically infected with genotype 1 hepatitis C virus (HCV), J. Med. Chem., 55, 7650

Lu, 2014, Mechanism of inhibition for BMS-791325, a novel non-nucleoside inhibitor of hepatitis C virus NS5B polymerase, J. Biol. Chem., 289, 33456, 10.1074/jbc.M114.613653

Coburn, 2013, Discovery of MK- 8742: an HCV NS5A inhibitor with broad genotype activity, Chem. Med. Chem., 8, 1930, 10.1002/cmdc.201300343

Devogelaere, 2012, TMC647055, a potent non nucleoside hepatitis C virus NS5B polymerase inhibitor with cross-genotypic coverage, Antimicrob. Agents Chemother., 56, 4676, 10.1128/AAC.00245-12

Cao, 2010, YQ36: A novel bisindolylmaleimide analogue induces KB/VCR cell death, J. Biomed. Biotechnol., 2009