RECENT DEVELOPMENTS AND MULTIPLE BIOLOGICAL ACTIVITIES AVAILABLE WITH 1, 8-NAPHTHYRIDINE DERIVATIVES: A REVIEW

Authors

  • Vinod Kumar Gurjar Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur 495009, C.G., India
  • Dilipkumar Pal Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Koni, Bilaspur 495009, C.G., India

DOI:

https://doi.org/10.22159/ijpps.2019v11i1.30429

Keywords:

1, 8-Naphthyridine (NPTR), Quinolone, Antimicrobial activity, Anticancer activity, Antitumor, Anti-inflammatory activity, Antimalarial antihypertensive, Antiplatelet activity, Nil

Abstract

Within the wide range of nitrogen-containing heterocyclic compounds, the derivatives of 1,8-naphthyridine (NPTR) have gained a rising interest due to their reported versatile biological activities. The derivatives of NPTR scaffold are found to invite special interest from researchers nowadays on the significance of their manifestations of multiple attractive pharmacological activities which establish them as an effective and versatile tool in pharmaceutical chemistry and drug discovery. The diverse biological activities mainly include anti-inflammatory, antimicrobial, antiviral, anticancer, antihypertensive and analgesic activities. Novel NPTR scaffold has emerged its potency to treat neurological diseases like depression and Alzheimer's disease. Further these agents possess different inhibitory activities, such as anti-HIV, anti-osteoporotic, αvβ3 antagonism, antimalarial, platelet aggregation, anti-oxidant, anti-allergic, gastric antisecretory, anticonvulsant, epidermal growth factor receptor (EGFR) inhibition, protein kinase inhibition, ionotropic properties, β3 antagonism, phosphodiesterase 4 (PDE 4) inhibitions, adenosine receptor agonistic activity, adrenoceptors antagonism and DNA stabilizing activity, etc. In this review, we highlight the updates of different 1,8-naphthyridine derivatives and explain the key data available in the context of various biological activities of NPTR derivatives available from the literature. This may direct opportunity in researches in the synthesis of novel medicinal agents and the development of new heterocycles for modification of existing biological actions as well as evaluation of other possible pharmacological activities.

Downloads

Download data is not yet available.

References

Rani P, Pal DK, Hegde RR, Hashim SR. Synthesis, characterization and pharmacological evaluation of substituted phenoxy acetamide derivatives. Hem Ind 2015;69:405–15.

Reissert A. About di-(γ-amidopropyl) acetic acid (diamino.1.7. heptan methylester.4) and their inner condensations product which octohydro.1.8. naphtyridin. Berichte Der Deutschen Chemischen Gesellschaft 1893;26:2137–44.

Ikekawa N. Studies on naphthyridines. I. Synthesis of 1, 6-naphthyridine. Chem Pharm Bull 1958;6:263–9.

Ikekawa N. Studies on naphthyridines. II. Synthesis of 2, 7-naphthyridine. Chem Pharm Bull 1958;6:269–72.

Ikekawa N. Studies on naphthyridines. III. Syntheses of 2, 10-diazaanthracene and 1, 7-naphthyridine. Chem Pharm Bull 1958;6:401–4.

Giacomello G, Gualtieri F, Riccieri FM, Stein ML. Synthesis of 2, 6-naphthyridine. Tetrahedron Lett 1965;6:1117–21.

Tan R, Taurins A. Synthesis of 2, 6-naphthyridine and some of its derivatives. Tetrahedron Lett 1965;6:2737–44.

Lesher GY, Froelich EJ, Gruett MD, Bailey JH, Brundag RP. 1, 8-Naphthyridine derivatives. A new class of chemotherapeutic agents. J Med Chem 1962;5:1063–5.

Allen CFH. The naphthyridines. Chem Rev 1950;47:275–305.

Litvinov VP, Roman SV, Dyachenko VD. Naphthyridines structure, physicochemical properties and general methods of synthesis. Rus Chem Rev 2000;69:201–20.

Litvinov VP. Chemistry and biological activities of 1, 8-naphthyridines. Rus Chem Rev 2004;73:637–69.

Litvinov VP. Advances in the chemistry of naphthyridines. Adv Heterocycl Chem 2006;91:189–300.

Fadda AA, El-Hadidy SA, Elattar KM. Advances in 1, 8-naphthyridines chemistry. Synth Comm Rev 2015;45:2765–801.

Noravyan AS, Paronikyan EG, Vartanyan SA. Synthesis and pharmacological properties of naphthyridines (review). Pharm Chem J 1985;19:439–48.

Shen LL, Pernet AG. Mechanism of inhibition of DNA gyrase by analogues of nalidixic acid: the target of the drugs is DNA. Proc Nat Acad Sci 1985;82:307–11.

Muhammad NU, Bature M, Nuhu T, Nafiu A. Fluoroquinolone resistance pattern among the pathogens causing urinary tract infection in a tertiary care hospital in Kanchipuram district, Tamil Nadu, India. Asian J Pharm Clin Res 2017;10:292–4.

Vincent J, Venitz J, Teng R, Baris BA, Willavize SA, Polzer RJ, et al. Pharmacokinetics and safety of trovafloxacin in healthy male volunteers following administration of single intravenous doses of the prodrug, alatrofloxacin. J Antimicrob Chemother 1997;39 Suppl B:75–80.

Qureshi ZP, Vazquez ES, Monguio RR, Stevenson KB, Szeinbach SL. Market withdrawal of new molecular entities approved in the United States from 1980 to 2009. Pharmacoepidemiol Drug Saf 2011;20:772–7.

Mizuki Y, Fujiwara I, Yamaguchi T, Sekine Y. Structure-related inhibitory effect of antimicrobial enoxacin and derivatives on theophylline metabolism by rat liver microsomes. Antimicrob Agents Chemother 1996;40:1875–80.

Hall IH, Schwab UE, Ward ES, Ives TJ. Effects of alatrofloxacin, the parental prodrug of trovafloxacin, on phagocytic, anti-inflammatory and immunomodulation events of human THP-1 monocytes. Biomed Pharmacother 2003;57:359–65.

Paim CS, Nogueira DR, Mitjans M, Lopez DR, Perez JL, Steppe M, et al. Biological safety studies of gemifloxacin mesylate and related substances Photochem Photobiol Sci 2013;12:805–12.

Hoch U, Lynch J, Kashimoto YSS, Kajikawa F, Furutani Y, Silverman JA. Voreloxin, formerly SNS-595, has potent activity against a broad panel of cancer cell lines and in vivo tumor models. Cancer Chemother Pharmacol 2009;64:53–65.

Evanchik MJ, Allen D, Yoburn JC, Silverman JA, Hoch U. Metabolism of (+)-1,4-dihydro-7-(trans-3-methoxy-4-methylamino-1-pyrrolidinyl)-4-oxo-1-(2-thiazol-yl)-1, 8-naphthyridine-3-carboxylic acid (Voreloxin; formerly SNS-595), a novel replication-dependent DNA-damaging agent. Drug Metab Dispos 2009;37:594–601.

Advani RH, Hurwitz HI, Gordon MS, Ebbinghaus SW, Mendelson DS, Wakelee HA, et al. Voreloxin, a first-in-class anticancer quinolone derivative, in relapsed/refractory solid tumors: a report on two dosing schedules. Clin Cancer Res 2010;16:2167–75.

Hawtin RE, Stockett DE, Byl JAW, McDowell RS, Tan N, Arkin MR, et al. Voreloxin is an anticancer quinolone derivative that intercalates DNA and poisons topoisomerase II. PLoS One 2010;5:e10186.

Scatena CD, Kumer JL, Arbitrario JP, Howlett AR, Hawtin RE, Fox JA, et al. Voreloxin, a first-in-class anticancer quinolone derivative, acts synergistically with cytarabine in vitro and induces bone marrow aplasia in vivo. Cancer Chemother Pharmacol 2010;66:881–8.

Freeman C, Keane N, Swords R, Giles F. Vosaroxin: a new valuable tool with the potential to replace anthracyclines in the treatment of AML? Expert Opin Pharmacother 2013;14:1417–27.

Pal DK, Mandal M, Senthilkumar GP, Padhiari A. Antibacterial activity of methanol extract of Cuscuta reflexa Roxb. Stem and Corchorus olitorius Linn. Seed Fitoterapia 2006;77:589–91.

Mohanta TK, Patra JK, Rath SK, Pal DK, Thatoi HN. Evaluation of antimicrobial activity and phytochemical screening of oils and nuts of Semicarpus anacardium L. f. Sci Res Essays 2007;2:486–90.

Nimse SB, Pal DK, Mazumder A, Mazumder R. Synthesis of cinnamanilide derivatives and their antioxidant and antimicrobial activity. J Chem 2015. http://dx.doi.org/10.1155/2015/208910

Gohil JD, Patel HB, Patel MP. Synthesis and evaluation of new chromene based [1, 8] naphthyridines derivatives as potential antimicrobial agents. RSC Adv 2016;6:74726–33.

Ghalia S, Thanaa M. An in silico study of novel fluoroquinolones as inhibitors of DNA gyrase of Staphylococcus aureus. Int J Pharm Pharm Sci 2016;8:67–75.

Arayne MS, Sultana N, Haroon U, Mesaik MA, Asif M. Synthesis and biological evaluations of enoxacin carboxamide derivatives. Arch Pharm Res 2009;32:967–74.

Lv K, Liu ML, Feng LS, Sun LY, Sun YX, Wei ZQ, et al. Synthesis and antibacterial activity of naphthyridone derivatives containing mono/difluoro-methyloxime pyrrolidine scaffolds. Eur J Med Chem 2012;47:619–25.

Feng L, Lv K, Liu M, Wang S, Zhao J, You X, et al. Synthesis and in vitro antibacterial activity of gemifloxacin derivatives containing a substituted benzyloxime moiety. Eur J Med Chem 2012;55:125–36.

Aggarwal N, Kumar R, Dureja P, Khurana JM. Synthesis of novel nalidixic acid-based 1, 3, 4-thiadiazole and 1, 3, 4-oxadiazole derivatives as potent antibacterial agents. Chem Biol Drug Des 2012;79:384–97.

Fadla TA, Radwan AA, Abdelaziz HA, Baseeruddin M, Attiaa MI, Kadi A. Novel Schiff bases of indoline-2, 3-dione and nalidixic acid hydrazide: synthesis, in vitro antimycobacterial and in silico mycobacterium tuberculosis (MTB) DNA gyrase inhibitory activity. Dig J Nanomater Bios 2012;7:327–36.

Jalal A, Badawneh M. Synthesis and antitubercular activity of piperidine and morpholine 1, 8-naphthyridine analogues. Int J Pharm Pharm Sci 2016;8:252–7.

Kumble D, Geetha MP, Asha FP. Application of metal complexes of Schiff bases as an antimicrobial drug: a review of recent works. Int J Curr Pharm Res 2017;9:27–30.

Singh R, Debnath A, Masram DT, Rathore D. Synthesis and biological activities of selected quinolone-metal complexes. Res J Chem Sci 2013;3:83–94.

Zakya M, Sayed MYE, Megharbe SME, Taleb SA, Refat MS. Complexes of nalidixic acid with some vital metal ions: synthesis, chemical structure elucidation, and antimicrobial evaluation. Rus J Gen Chem 2013;83:2488–501.

Debnath A, Mogha NK, Masram DT. Metal complex of the first-generation quinolone antimicrobial drug nalidixic acid: structure and it's a biological evaluation. Appl Biochem Biotechnol 2015;175:2659–67.

Saleh MY, Ayoub AI. Synthesis of new derivatives of 2-chloro-3-formyl-1, 8‐naphthyridine. Eur J Chem 2014;5:475–80.

Gao LZ, Xie YS, Li T, Huang WL, Hu GQ. Synthesis and antibacterial activity of novel [1,2,4]triazolo [3,4-h][1, 8]naphthyridine-7-carboxylic acid derivatives. Chin Chem Lett 2015;26:149–51.

Acosta P, Butassi E, Insuasty B, Ortiz A, Abonia R, Zacchino SA, et al. Microwave-assisted synthesis of novel pyrazolo[3,4-g][1, 8]naphthyridine-5-amine with potential antifungal and antitumor activity. Molecules 2015;20:8499–520.

Donalisio M, Massari S, Argenziano M, Manfroni G, Cagno V, Civra A, et al. Ethyl 1, 8-naphthyridone-3-carboxylates downregulate human papillomavirus-16 E6 and E7 oncogene expression. J Med Chem 2014;57:5649–63.

Sakram B, Ashok K, Rambabu S, Sonyanaik B, Ravi D. A novel and efficient synthesis of 3-iodo substituted 1,8-naphthyridines by electrophilic cyclization of 2-amino nicotinaldehyde and their antimicrobial activity. Russ J Gen Chem 2017;87:1794–9.

Valadbeigi E, Ghodsi S. Synthesis, and study of some new quinolone derivatives containing a 3-acetyl coumarin for their antibacterial and antifungal activities. Iran J Pharm Res 2017;16:554–64.

Gençer HK, Levent S, Acar U, Ozkay Y, Ilgın S. New 1, 4-dihydro [1,8]naphthyridine derivatives as DNA gyrase inhibitors. Bioorg Med Chem Lett 2017;27:1162–8.

Sherlock MH, Kaminski JJ, Tom WC, Lee JF, Wong S, Kreutner W, et al. Antiallergy agents. 1. Substituted 1, 8-naphthyridine-2(1H)-ones as inhibitors of SRS-A release. J Med Chem 1988;31:2108–21.

Nishikawa Y, Shind T, Ishi K, Nakamura H, Kon T, Uno H, et al. Synthesis and antiallergic activity of N-[4-(4’diphenylmethyl-1-piperazinyl) butyl-1, 4. dihydro-4-oxo-pyridine-3-carboxamides. Chem Pharm Bull 1989;37:1256–9.

Saccomanni G, Badawneh M, Adinolfi B, Calderone V, Cavallini T, Ferrarini PL, et al. Synthesis and β-blocking activity of (R, S)-(E)-oxime ethers of 2,3-dihydro-1, 8-naphthyridine and 2,3dihydrothiopyrano [2,3-b] pyridine: identification of β-3-antagonists. Bioorg Med Chem 2003;11:4921–31.

Badawneh M, Ferrarini PL, Calderone V, Manera C, Martinotti E, Mori C, et al. Synthesis and evaluation of antihypertensive activity of 1, 8-naphthyridine derivatives. Part X. Eur J Med Chem 2001;36:925–34.

Jalal A, Jamal A, Badawneh M. Characterization of the biochemical effects of new 1, 8-naphthyridine derivatives, β-receptor antagonists, in ventricular myocytes. Arch Pharm Pharm Med Chem 2003;336:285–92.

Sannigrahi S, Mazumder UK, Pal DK, Mishra SL. Terpenoids of methanol extract of C. infortunatum exhibit anticancer activity against Ehrlich’s ascites carcinoma (EAC) in mice. Pharm Biol 2012;50:304–9.

Rani P, Pal DK, Hegde RR, Hashim SR. Acetamides: chemotherapeutic agents for inflammation-associated cancers. J Chemother 2016;28:255–65.

Sannigrahi S, Mazumder UK, Pal DK, Mondal A, Mishra SL, Roy S. Flavonoids of Enhydra fluctuans exhibit anticancer activity against ehrlich’s ascites carcinoma in mice. Nat Prod Comm 2010;5:1239–42.

Atanasova M, Ilieva S, Galabov B. QSAR analysis of 1, 4-dihydro-4-oxo-1-(2-thiazolyl)-1, 8-naphthyridines with anticancer activity. Eur J Med Chem 2007;42:1184–92.

Eweas AF, Khalifa NM, Ismail NS, Al-Omar MA, Soliman AMM. Synthesis, molecular docking of novel 1, 8-naphthyridine derivatives and their cytotoxic activity against HePG2 cell lines. Med Chem Res 2014;23:76–86.

Ahmed NS, Alfooty KO, Khalifah SS. An efficient sonochemical synthesis of novel Schiff’s bases, thiazolidine, and pyrazolidine incorporating 1, 8-naphthyridine moiety and their cytotoxic activity against HePG2 cell lines. Sci World J 2014;25:587059.

Ahmed NS, Alfooty KO, Khalifah SS. Synthesis of 1, 8-Naphthyridine derivatives under ultrasound irradiation and cytotoxic activity against HePG2 cell lines. J Chem 2014;1:1–8.

Elansary AK, Moneer AA, Kadry HH, Gedawy EM. Synthesis and antitumor activity of certain pyrido [2,3-d ] pyrimidine and 1, 8-naphthyridine derivatives. J Chem Res 2014;38:147–53.

Fu L, Feng X, Wang JJ, Xun Z, Hu JD, Zhang JJ, et al. Efficient synthesis and evaluation of antitumor activities of novel functionalized 1, 8-naphthyridine derivatives. ACS Comb Sci 2015;17:24–31.

Graf M, Gothe Y, Metzler Nolte N, Sünkel K. Cyclometalated iridium(III) and rhodium(III) complexes containing naphthyridine ligands: synthesis, characterization and biological studies. Z Anorg Allg Chem 2017;643:306–10.

Jia XD, Wang S, Wang MH, Liu ML, Xia GM, Liu XJ, et al. Synthesis and in vitro antitumor activity of novel naphthyridine derivatives. Chin Chem Lett 2017;28:235–9.

Behalo MS, Mele G. Synthesis, and evaluation of pyrido[2,3-d]pyrimidine and 1,8-naphthyridine derivatives as potential antitumor agents. J Heterocyclic Chem 2017;54:295–300.

Tang Q, Duan Y, Wang L, Wang M, O'Yang Y, Wang C, et al. Synthesis and antiproliferative activity of pyrrolo[2,3-b]pyridine derivatives bearing the 1,8-naphthyridine-2-one moiety. Eur J Med Chem 2018;143:266–75.

Melha SA. Synthesis and biological evaluation of some novel 1, 8-naphthyridine derivatives. Acta Chim Slov 2017;64:919–30.

Pal DK, Mazumder UK. Isolation of compound and studies on CNS depressant activities of Mikania scandens with special emphasis to brain biogenic amines in mice. Indian J Exp Biol 2014;52:1186–94.

Negus B, Markocic S. A surgical antiemetic protocol-implementation and evaluation. Acute Pain 2003;5:63–8.

Gautam BK, Jindal A, Dhar AK, Mahesh R. Antidepressant-like activity of 2-(4-phenylpiperazine-1-yl)-1, 8-naphthyridine-3-carboxylic acid (7a), a 5-HT3 receptor antagonist in behaviour based rodent models: evidence for the involvement of serotonergic system. Pharmacol Biochem Behav 2013;109:91–7.

Mahesh R, Dhar AK, Jindal A, Bhatt S. 2-(4-substituted piperazine-1-yl)-1, 8-naphthyridine-3-carboxylic acids: novel 5-HT3 receptor antagonists with anxiolytic-like activity in rodent behavioral models. Can J Physiol Pharmacol 2013;91:848–54.

Mahesh R, Dhar AK, Jindal A, Bhatt S. Design, synthesis and evaluation of antidepressant activity of novel 2-methoxy 1, 8-naphthyridine 3-carboxamides as 5-HT3 receptor antagonists. Chem Biol Drug Des 2014;83:583–91.

Dhar AK, Mahesh R, Jindal A, Devadoss T. Design, synthesis and pharmacological evaluation of novel 2-(4-substituted piperazine-1-yl)-1, 8-naphthyridine-3-carboxylic acids as 5-HT3 receptor antagonists for the management of depression. Chem Biol Drug Des 2014;84:721–31.

Dhar AK, Mahesh R, Jindal A, Bhatt S. Piperazine analogs of naphthyridine-3-carboxamides and Indole-2-carboxamides: novel 5-HT3 receptor antagonists with antidepressant-like activity. Arch Pharm Chem Life Sci 2015;348:34–45.

Pal DK, Panda C, Sinhababu S, Dutta A, Bhattacharya S. Evaluation of psychopharmacological effects of petroleum ether extract of Cuscuta reflexa Roxb stem in mice. Acta Polon Pharm Drug Res 2003;60:481–6.

Jagerovic N, Fernandez CF, Goya P. CB1 cannabinoid antagonists: structure-activity relationships and potential therapeutic applications. Curr Top Med Chem 2008;8:205–30.

Duggan CBM, Debenham JS, Walsh TF, Yan L, Huo P, Wang J, et al. Dihydro-pyrano [2,3-b] pyridines and tetrahydro-1, 8-naphthyridines as CB1 receptor inverse agonists: synthesis, SAR, and biological evaluation. Bioorg Med Chem Lett 2010;20:3750–4.

Manera C, Benetti V, Castelli MP, Cavallini T, Lazzarotti S, Pibiri F, et al. Design, synthesis, and biological evaluation of new 1, 8-naphthyridine-4 (1H)-on-3-carboxamide and quinoline-4 (1H)-on-3-carboxamide derivatives as CB2 selective agonists. J Med Chem 2006;49:5947–57.

Manera C, Saccomanni G, Malfitano AM, Bertini S, Castelli F, Laezza C, et al. Rational design, synthesis and anti-proliferative properties of new CB2 selective cannabinoid receptor ligands: an investigation of the 1, 8-naphthyridine-2 (1H)-one scaffold. Eur J Med Chem 2012;52:284–94.

Lucchesi V, Hurst DP, Shore DM, Bertini S, Ehrmann BM, Allara M, et al. CB2-selective cannabinoid receptor ligands: synthesis, pharmacological evaluation, and molecular modeling investigation of 1, 8-naphthyridine-2 (1H)-one-3-carboxamides. J Med Chem 2014;57:8777–91.

Manera C, Saccomanni G, Adinolfi B, Benetti V, Ligresti A, Cascio MG, et al. Rational design, synthesis, and pharmacological properties of new 1, 8-naphthyridine-2(1H)-on-3-carboxamide derivatives as highly selective cannabinoid-2 receptor agonists. J Med Chem 2009;52:3644–51.

Saccomanni G, Pascali G, Carlo SD, Panetta D, Simone MD, Bertini S, et al. Design, synthesis and preliminary evaluation of [18]F-labelled 1, 8-naphthyridine and quinoline-2-one-3-carboxamide derivatives for PET imaging of CB2 cannabinoid receptor. Bioorg Med Chem Lett 2015;25:2532–5.

Manera C, Malfitano AM, Parkkari T, Lucchesi V, Carpi S, Fogli S, et al. New quinolone and 1, 8-naphthyridine-3-carboxamides as selective CB2 receptor agonists with anticancer and immunomodulatory activity. Eur J Med Chem 2015;97:10–8.

Malfitano AM, Laezza C, Saccomanni G, Tuccinardi T, Manera C, Martinelli A, et al. Immune-modulation and properties of absorption and blood brain barrier permeability of 1, 8-naphthyridine derivatives. J Neuroimmune Pharmacol 2013;8:1077–86.

Malfitano AM, Laezza C, Alessandro AD, Procaccini C, Saccomanni G, Tuccinardi T, et al. Effects on immune cells of a new 1, 8-naphthyridine-2-one derivative and its analogues as selective CB2 agonists: implications in multiple sclerosis. PLOS One 2013;8:1–9.

Malfitano AM, Laezza C, Bertini S, Marasco D, Tuccinardi T, Bifulco M, et al. Immunomodulatory properties of 1,2-dihydro-4-hydroxy-2-oxo-1,8-naphthyridine-3-carboxamide derivative VL15. Biochimie 2017;135:173–80.

Pommier Y, Johnson AA, Marchand C. Integrase inhibitors to treat HIV/AIDS. Nat Rev Drug Disc 2005;4:236–48.

Serrao E, Odde S, Ramkumar K, Neamati N. Raltegravir, elvitegravir, and metoogravir: the birth of "me-too" HIV-1 integrase inhibitors. Retrovirology 2009;6:33.

Nagasawa JY, Song J, Chen H, Kim HW, Blazel J, Ouk S, et al. 6-benzylamino 4-oxo-1,4-dihydro-1, 8-naphthyridines and 4-oxo-1,4-dihydroquinolines as HIV integrase inhibitors. Bioorg Med Chem Lett 2011;21:760–3.

Zhao XZ, Smith SJ, Metifiot M, Johnson BC, Marchand C, Pommier P, et al. Bicyclic 1-hydroxy-2-oxo-1,2-dihydropyridine-3-carboxamide-containing HIV-1 integrase inhibitors having high antiviral potency against cells harboring raltegravir-resistant integrase mutants. J Med Chem 2014;57:1573–82.

Zhao XZ, Smith SJ, Metifiot M, Marchand C, Boyer PL, Pommier Y, et al. 4-Amino-1-hydroxy-2-oxo-1, 8-naphthyridine-containing compounds having high potency against raltegravir-resistant integrase mutants of HIV-1. J Med Chem 2014;57:5190–202.

Massari S, Daelemans D, Barreca ML, Knezevich A, Sabatini S, Cecchetti V, et al. A 1, 8-naphthyridone derivative targets the HIV-1 tat-mediated transcription and potently inhibits the HIV-1 replication. J Med Chem 2010;53:641–8.

Tabarrini O, Massari S, Sancineto L, Daelemans D, Sabatini S, Manfroni G, et al. Structural investigation of the naphthyridone scaffold: identification of a 1,6-naphthyridone derivative with potent and selective anti-HIV activity. Chem Med Chem 2011;6:1249–57.

Rani P, Pal DK, Hegde RR, Hashim SR. Anticancer, anti-inflammatory, analgesic activities of synthesized 2-(substituted phenoxy) acetamide derivatives. Biomed Int 2014;386473:1–9.

Rani P, Pal DK, Hegde RR, Hashim SR. Leuckart synthesis and studies on anticancer, anti-inflammatory and analgesic activities of novel acetamide derivatives. Anticancer Agents Med Chem 2016;16:898–906.

Pal DK, Sannigrahi S, Mazumder UK. Analgesic and anticonvulsant effects of isolated saponin from the leaves of Clerodendrum infortunatum Linn. in mice. Indian J Exp Biol 2009;47:743–7.

Sun XY, Wei CX, Chai KY, Piao HR, Quan ZS. Synthesis and anti-inflammatory activity evaluation of novel 7-alkoxy-1-amino-4,5-dihydro[1,2,4]triazole[4,3-a]quinolines. Arch Pharm Chem Life Sci 2008;341:288–93.

Madaan A, Kumar V, Verma R, Singh AT, Jain SK, Jaggi M. Anti-inflammatory activity of a naphthyridine derivative (7-chloro-6-fluoro-N-(2-hydroxy-3-oxo-1-phenyl-3-(phenylamino) propyl)-4-oxo-1-(prop-2-yn-1-yl)-1, 4-dihydro-1, 8-naphthyridine-3-carboxamide) possessing in vitro anticancer potential. Int Immunopharmacol 2013;15:606–13.

Braccio MD, Grossi G, Alfei S, Ballabeni V, Tognolini M, Flammini L, et al. 1, 8-Naphthyridines IX. Potent anti-inflammatory and/or analgesic activity of a new group of substituted 5-amino [1, 2, 4] triazolo [4, 3-a] [1, 8] naphthyridine-6-carboxamides, of some their mannich base derivatives and of one novel substituted 5-amino-10-oxo-10Hpyrimido [1, 2-a] [1, 8] naphthyridine-6-carboxamide derivative. Eur J Med Chem 2014;86:394–405.

Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat Rev Drug Discovery 2006;5:247–64

Manera C, Betti L, Cavallini T, Giannaccini G, Martinelli A, Ortore G, et al. 1, 8-Naphthyridine-4-one derivatives as new ligands of A2A adenosine receptors, Bioorg Med Chem Lett 2015;15:4604–10.

Ferrarini PL, Mori C, Manera C, Martinelli A, Mori F, Saccomanni G, et al. A novel class of highly potent and selective A1 adenosine antagonists: structure-affinity profile of a series of 1, 8-naphthyridine derivatives. J Med Chem 2000;43:2814–23.

Sandhya A, Gomathi K. Alzheimer’s disease therapeutic approaches. Asian J Pharm Clin Res 2018;11:17–24.

Silva D, Chioua M, Samadi A, Carreiras MC, Jimeno ML, Mendes E, et al. Synthesis and pharmacological assessment of diversely substituted pyrazolo [3, 4-b] quinoline, and benzo [b] pyrazolo [4, 3-g] [1, 8] naphthyridine derivatives. Eur J Med Chem 2011; 46:4676–81.

Klenc J, Lipowska M, Taylor AT. Identification of lead compounds for [99]mTc and [18]F GPR91 radiotracers. Bioorg Med Chem Lett 2015;25:2335–9.

Chandra P, Sachan N, Pal DK. Protective effect of Dalbergia sissoo Roxb. Ex Dc (family Fabaceae) leaves against experimentally induced diarrhea and peristalsis in mice. Toxicol Ind Health 2013;20:1–7.

Sacchan N, Chandra P, Pal DK. Evaluation of gastroprotective potential of Dalbergia sissoo (family: Fabaceae) in experimental rats. Int J Indigen Med Plants 2015;48:1778–85.

Sacchan N, Chandra P, Pal DK. Assessment of the gastroprotective potential of Dalonix revia (Boj-Ex Sooa) Raf. Against ethanol and cold rextrin stressed-induced ulcer in rats. Trop J Pharm Res 2015;14:1063–70.

Santilli AA, Scotese AC, Bauer RF, Bell SC. 2-Oxo-1, 8-naphthyridine-3-carboxylic acid derivatives with potent gastric antisecretory properties. J Med Chem 1987;30:2270–7.

Sekhar KVGC, Rao VS, Conrad WD, Reddy AS, Brust P, Kumar MMK. Design, synthesis, and preliminary in vitro and in vivo pharmacological evaluation of 2-{4-[4-(2,5-disubstitutedthiazol-4-yl) phenylethyl] piperazine-1-yl}-1, 8-naphthyridine-3-carbonitriles as atypical antipsychotic agents. J Enzyme Inhib Med Chem 2011;26:561–8.

Ferrarini PL, Mori C, Badawneh M, Franconi F, Manera C, Miceli M, et al. Synthesis and antiplatelet activity of some 3-phenyl-1, 8-naphthyridine derivatives. Il Farmaco 2000;55:603–10.

Ferrarini PL, Badawneh M, Franconi F, Manera C, Miceli M, Mori C, et al. Synthesis and antiplatelet activity of some 2, 7-di (N-cycloamino)-3-phenyl-1, 8-naphthyridine derivatives. Il Farmaco 2001;56:311–8.

Naik TR, Naik HS, Naik HR, Bindu PJ, Harish BG, Krishna V. Synthesis, DNA binding, docking and photocleavage studies of novel benzo [b] [1, 8] naphthyridines. Med Chem 2009;5:411–8.

Dhamodharan V, Harikrishna S, Jagadeeswaran C, Halder K, Pradeepkumar PI. Selective G-quadruplex DNA stabilizing agents based on bisquinolinium and bispyridinium derivatives of 1, 8-naphthyridine. J Org Chem 2012;77:229–42.

Ban H, Muraoka M, Ioriya K, Ohashi N. Synthesis and biological activity of novel 4-phenyl-1, 8-naphthyridine-2 (1H)-on-3-yl ureas: Potent acyl-CoA: cholesterol acyltransferase inhibitor with improved aqueous solubility. Bioorg Med Chem Lett 2006;16:44–8.

Kanouni T, Dong Q, Abelovski B, Wallace MB. Synthetic approaches to 1, 8-naphthyridine-2, 5-dione compounds. Tetrahedron Lett 2011;52:477–9.

Takayama K, Iwata M, Hisamichi H, Okamoto Y, Aoki M, Niwa A. Synthetic studies on selective type 4 phosphodiesterase (PDE 4) inhibitors. 1. structure-activity relationships and pharmacological evaluation of 1, 8-naphthyridine-2(1H)-one derivatives. Chem Pharm Bull 2002;50:1050–9.

Rodan SB, Rodan GA. Integrin function in osteoclasts. J Endocrinol 1997;154:S47–56.

Hartner FW, Hsiao Y, Eng KK, Rivera NR, Palucki M, Tan L, et al. Methods for the synthesis of 5, 6, 7, 8-Tetrahydro-1, 8-naphthyridine fragments for αVβ3 integrin antagonists. J Org Chem 2004;69:8723–30.

Nagarajan SR, Lu HF, Gasiecki AF, Khanna IK, Parikh MD, Desai BN, et al. Discovery of+(2-{4-[2-(5,6,7,8-tetrahydro-1, 8-naphthyridine-2-yl) ethoxy] phenyl}-cyclopropyl) acetic acid as potent and selective αvβ3 inhibitor: design, synthesis, and optimization. Bioorg Med Chem 2007;15:3390–412.

Al-Salem HS, Hegazy GH, El-Taher KE, El-Messery SM, Al-Obaid AM, El-Subbagh HI. Synthesis, anticonvulsant activity and molecular modeling study of some new hydrazinecarbothioamide, benzenesulfonohydrazide, and phenacylacetohydrazide analogues of 4 (3H)-quinazolinone. Bioorg Med Chem Lett 2015;25:1490–9.

Stuk TL, Assink BK, Bates RC, Erdman DT, Fedij V, Jennings SM, et al. An efficient and cost-effective synthesis of pagoclone. Org Proc Res Dev 2003;7:851–5.

Lorrio S, Romero A, Gonzalez Lafuente L, Lajarin Cuesta R, Martinez Sanz FJ, Estrada M, et al. PP2A ligand ITH12246 protects against memory impairment and focal cerebral ischemia in mice. ACS Chem Neurosci 2013;4:1267–77.

Wang H, Wang S, Cheng L, Chen L, Wang Y, Qing J, et al. Discovery of imidazo [1, 2-α] [1, 8] naphthyridine derivatives as a potential HCV entry inhibitor. ACS Med Chem Lett 2015;6:977–81.

Nimse SB, Pal DK. Free radicals, natural antioxidants, and their reaction mechanisms. RSC Adv 2015;5:27986–8006.

Sannigrahi S, Mazumder UK, Pal DK, Parida S, Jain S. Antioxidant potential of crude extract and its different fraction of methanol extract of Enhydra fluctuans Lour. Iranian J Pharm Res 2010;9:75–82.

Nam TG, Rector CL, Kim HY, Sonnen AF, Meyer R, Nau WM, et al. Tetrahydro-1, 8-naphthyridineol analogues of α-Tocopherol as antioxidants in lipid membranes and low-density lipoproteins. J Am Chem Soc 2007;129:10211–9.

Yu A, Wang J, Xue X, Wang Y. Theoretical study of the peripheral disulfide bridge substituent effects on the antioxidant properties of naphthyridine diol derivatives. J Phy Chem A 2010;114:1008–16.

Aljamal JA, Badawneh M. Antilipolytic effects of 1,8-naphthyridine derivatives β-adrenoceptor antagonists in rat white adipocytes. Chem Biol Drug Des 2017;90:119–27.

Madaan A, Verma R, Kumar V, Singh AT, Jain SK, Jaggi M. 8-Naphthyridine derivatives: a review of multiple biological activities. Arch Pharm Chem Life Sci 2015;348:1–24.

Shimamoto T, Inoue H, Hayashi Y. 1-Cycloalkyl-1,8-naphthyridine-4-one derivative as type iv phosphodiesterase inhibitor. US patent 6331548; 2001.

Bachand B, Nguyen-Ba N, Siddiqui A. Antiviral methods using [1,8] naphthyridine derivatives. US patent 6340690; 2002.

Vaillancourt VA, Thorarensen A. 4-Oxo-1,4 dihydro[1,8]naphthyridine 3-carboxamides as antiviral agents. US patent 6451811; 2002.

Yamin W, David EG, Qingjie L, Sidney XL, William HB, Donglei L, et al. 1, 8-Naphthyridine derivatives as antidiabetics. WIPO patent 03/027112; 2003.

Bachand B, Nguyen Ba N, Siddiqui A. 1,8-Naphthyridine derivatives having antiviral activity. US patent US 6605614; 2003.

Yamin W, William B, David G, Qingjie L, Sidney L, Donglei L, et al. 1,8-Naphthyridine derivatives and their use to treat diabetes and related disorders. US patent 20040014751A1; 2001.

Muraoka M, Ohnuma S, Ban H. 1, 2-Dihydro-2-oxo-1, 8-naphthyridine derivative. US patent 7067528B2; 2006.

Yamin W, William B, David G, Qingjie L, Sidney L, Donglei L, et al. 1,8-Naphthyridine derivatives and their use to treat diabetes and related disorders. US patent 7109196B2; 2006.

Jeffrey PW, Michael S, Terence M. Heterocyclic substituted 1,4-dihydro-4ox9-1,8-naphthyridine analogs. US patent 7163948 B2; 2007.

Deodialsingh G, Jeffrey JH, Shankaran K. Novel 1,8-naphthyridine compounds. US patent 20100056563A1; 2010.

Fabio R, Di F, Micheli Y. 1,8-Naphthyridines as CRF antagonists. EP patent EP1695974B1; 2010.

Miller WH, Newlander KA, Seefeld MA, Uzinskas IN, DeWolf Jr WE, Jakas DR. Fab I inhibitors. US patent US7790716B2; 2010.

Rainer H. 1,6-Naphthyridine and 1,8-naphthyridine derivatives and their use to treat diabetes and related disorders. WIPO patent WO2005091857A3; 2006.

Adelman DC, Evanchik MJ, Sudhakar A, Jacobs JW, Silverman JA. 1,8-Naphthyridine compounds for the treatment of cancer. US patent 8124773B2; 2012.

Jonczyk A, Dorsch D, Zenke F, Amendt C. Hetarylaminonaphthyridines. US patent US 2012/0316166A1; 2012.

Alam A, Biscarrat S, Blanc I, Bono F, Duclos O, McCort G. Derivatives of 7-alkynyl-1,8-naphthyridones, preparation method thereof and 2011‘use of same in therapeutics. US Patent US 8470847B2; 2013.

Adelman DC, Evanchik MJ, Sudhakar A, Jacobs JW, Silverman JA. 1,8-Naphthyridine compounds for the treatment of cancer. US Patent US 8765954B2; 2014.

Jonczyk A, Dorsch D, Zenke F, Amendt C. Hetaryl-[1,8]naphthyridine derivatives. US patent US 8912216; 2012.

Choi DR, Yang J, Yoon SH, Pyun SJ, Kim SH, Seong SK, et al. R-7-(3-aminomethyl-4-methoxyimino-3-methyl-pyrrolidine-1-yl)-1-cyclopropyl-6--fluoro-4-oxo-1,4-dihydo-[1,8]naphthyridine-3-carboxylic acid and an l-aspartic acid salt, the process for the preparation thereof and a pharmaceutical composition comprising the same for antimicrobial. US patent US 8952164B2; 2015.

Glenn M. Methods of using (+)-1,4-dihydro-7-[(3s,4s)-3-methoxy-4-(methylamino)-1-pyrrolidinyl]-4-ox-o-1-(2-thiazolyl)-1,8-naphthyridine-3-carboxylic acid for treatment of antecedent hematologic disorders. US patent 20150250775; 2015.

Published

01-01-2019

How to Cite

Gurjar, V. K., and D. Pal. “RECENT DEVELOPMENTS AND MULTIPLE BIOLOGICAL ACTIVITIES AVAILABLE WITH 1, 8-NAPHTHYRIDINE DERIVATIVES: A REVIEW”. International Journal of Pharmacy and Pharmaceutical Sciences, vol. 11, no. 1, Jan. 2019, pp. 17-37, doi:10.22159/ijpps.2019v11i1.30429.

Issue

Section

Review Article(s)