International Journal of Pharma and Bio Sciences
 
 
    ISSN 0975-6299
www.ijpbs.net


REVIEW ARTICLE
Int J Pharm Bio Sci Volume 12 issue 4, October - December, Pages:40-55

Review of Chemical, Pharmacological Activities of Naphthyridine Derivatives (1970 To 2020)

Seshaiah Krishnan Sridhar, Sreerama Rajasekhar, A. Ikram Khan, Karnam Charan Sai, M.D. Kusuma Priya and Vinod Kumar
DOI: http://dx.doi.org/10.22376/ijpbs.2021.12.4.p40-55
Abstract:

: Naphthyridine is the class of aromatic heterocyclic compounds which are subgroup of diazanaphthalene. To our knowledge and search from this work perspective, six naphthyridine drugs were  found around (enoxacin, nalidixic acid, trovafloxacin, alatrofloxacin, gemifloxacin and pyronaridine) that are therapeutically approved; these drugs are useful in the management of bacterial infections and pyronaridine is useful in malarial therapy. There were also several patents of naphthyridines as potential drugs and intermediates. We also came across some of naphthyridine derivatives that are in the clinical trial phases. Even though naphthyridine derivatives are versatile molecules that exhibit a variety of pharmacological activities and biological effects; in our extensive literature search, we did not encounter comprehensive review reports on naphthyridine derivatives. There were review reports available about some of the drugs (naphthyridines) clinically approved and under trials, these were mostly oriented towards a single chemical entity. This lead to conceptualize the present compilation of the most significant research literature available on naphthyridine derivatives. This compilation intends to be a major single source of information of several chemical derivatives of naphthyridine that are screened for pharmacological and biological activities. Naphthyridines derivatives exhibit various biological and pharmacological activities which includes antimicrobial (antibacterial, antifungal, antiviral & antimalarial), anti-leishmanial, immunomodulatory, analgesic, anti-inflammatory, anti-alzheimer, anti-vertigo, anticonvulsant, cardiovascular (antiarrhythmic, antihypertensive, α-receptor antagonist & β-blocker), anticancer, diuretic, gastric anti-secretary, bile acid transport inhibitory, antioxidant and phytocytotoxicity activity. In this review article, we have attempted to compile the most significant reports of biological and pharmacological activities of naphthyridine derivatives available literature from 1970 to 2020.

Keywords: Naphthyridine, Antibacterial, Antiviral, Anticancer, Cardiac
Full HTML:

REFERENCES

 

Wang BZ, Zhou JP, Zhou Y, Luo JS, Yang JJ, Chi SMi. Crystal structure of N-(7-di-bromomethyl-5-methyl-1,8-naphthyridin-2-yl)benzamide-pyrrolidine-2,5-dione (1/1). Acta Crystallogr E Cryst Commun. 2017;73(1):1-3. doi: 10.1107/S2056989016019034.

2.       Allen CF. The naphthyridines. Chem Rev. 1950;47(2):275-305. doi: 10.1021/cr60147a004, PMID 24538878.

3.       Reinhardt J, Schmedeberg N, Spanka C. Naphthyridine derivatives as suppressors of non-sense mutations. EP315229A1, 2017.

4.       Hu L, Wang X, Wang Y, Yang Y, Zeng C. Disubstituted octahydro-1,6- naphthyridine compound, and preparation method and application thereof. CN106588922A, 2017.

5.       Huang W. Naphthyridine compounds, medical combinations and use thereof. US2018099962A1, 2018.

6.       Adelman D, Aekin M, Hyde J, Silverman J, Walker D, Wright J. Methods of using(+)-1,4-dihydro-7-[(3s,4s)-3-methoxy-4-9methylamino-1-pyrrolidinyl]-4-oxo-1-2-thiazolyl-1,8-naphthyridine-3-carboxylic acid for treatment of cancer. US2018098975A1, 2018.

7.       Ahn YG, Jung SH, Kim TW, Lee MS. Novel 26-naphthyridine 2-oxide derivatives and use thereof. KR20190076339A, 2019.

8.       Chen C, Lei C, Wang F. Naphthyridine bipyrrole compound and preparation method thereof. CN110016028A, 2019.

9.       Lu H. Method for preparing medical intermediate 5,10-diarylpyrido. 2020;4(1, 6)naphthyridine derivative. CN11393440A.

10.     Chen X, He Q, Li Y, Liang W, Yang Z, Zheng W. 2-methyl-1,8-naphthyridine compound and preparation method and application therefor. WO2020253415A1, 2020.

11.     Pandeya SN, Smitha S, Jyoti M, Sridhar SK. Biological activities of isatin and its derivatives. Acta Pharm. 2005;55(1):27-46. PMID 15907222.

12.     Sridhar SK, Britoraj S, Rajasekhar S, Sundaraseelan J. Review of chemical, pharmacological, biological activities of isatin and its derivatives. J Sci Technol. 2020; 5(3);1 (1877 to 2002):225-47.

13.     Sridhar SK, Britoraj S, Rajasekhar S, Jothieswari D, Sundaraseelan J. Review of chemical, pharmacological, biological activities of isatin and its derivatives. J Sci Technol. 2020: 5(4);2 (2002-2020):288-307.

14.     Leonard JT, Gangadhar R, Gnanasam SK, Ramachandran S, Saravanan M, Sridhar SK. Synthesis and pharmacological activities of 1,8-naphthyridine derivatives. Biol Pharm Bull. 2002;25(6):798-802. doi: 10.1248/bpb.25.798, PMID 12081151.

15.     Santilli AA, Scotese AC, Yurchenco JA. Synthesis and antibacterial evaluation of 1,2,3,4-tetrahydro-4-oxo-1,8-naphthyridine-3-carboxylic acid esters, carbonitriles, and carboxamides. J Med Chem. 1975;18(10):1038-41. doi: 10.1021/jm00244a021, PMID 1099201.

16.     Nishigaki S, Mizushima N, Senga K. Synthetic antibacterials VI. 7-[1-Substituted 2-(5-nitro-2-furyl)-vinyl]-1,8-naphthyridines. Chem Pharm Bull (Tokyo). 1976;24(7):1658-61. doi: 10.1248/cpb.24.1658, PMID 975427.

17.     Suzuki N. Synthesis of antimicrobial agents. V. Synthesis and antimicrobial activities of some heterocylic condensed 1,8-naphthyridine derivatives. Chem Pharm Bull (Tokyo). 1980;28(3):761-8. doi: 10.1248/cpb.28.761, PMID 7389031.

18.     Tani J, Mushika Y, Yamaguchi T. Studies on biologically active halogenated compounds. III. Synthesis and antibacterial activity of 7-fluoromethyl-1,8-naphthyridine and quinoline derivatives. Chem Pharm Bull. 1982;30(10):3517-29. doi: 10.1248/cpb.30.3517.

19.     Kouno K, Inoue M, Mitsuhashi S. In vitro and in vivo antibacterial activity of AT-2266. Antimicrob Agents Chemother. 1983;24(1):78-84. doi: 10.1128/AAC.24.1.78, PMID 6226242.

20.     Hayakawa I, Suzuki N, Suzuki K, Tanaka Y. Synthesis of antimicrobial agents. VI. Studies on the synthesis of furo[3,2-b][1,8]naphthyridine derivatives. Chem Pharm Bull (Tokyo). 1984;32(12):4914-22. doi: 10.1248/cpb.32.4914, PMID 6534540.

21.     Nishimura Y, Matsumoto J. Pyridonecarboxylic acids as antibacterial agents. 9. Synthesis and antibacterial activity of 1-substituted 6-fluoro-1,4-dihydro-4-oxo-7-(4-pyridyl)-1,8-naphthyridine-3- carboxylic acids. J Med Chem. 1987;30(9):1622-6. doi: 10.1021/jm00392a017, PMID 3114492.

22.     King A, Bethune L, Phillips I. The in-vitro activity of tosufloxacin, a new fluorinated quinolone, compared with that of ciprofloxacin and temafloxacin. J Antimicrob Chemother. 1991;28(5):719-25. doi: 10.1093/jac/28.5.719, PMID 1663930.

23.     Cohen MA, Huband MD, Mailloux GB, Yoder SL, Roland GE, Domagala JM, Heifetz CL. In vitro antibacterial activities of PD 131628, a new 1,8-naphthyridine anti-infective agent. Antimicrob Agents Chemother. 1991;35(1):141-6. doi: 10.1128/AAC.35.1.141, PMID 2014970.

24.     Lewin CS. Antibacterial activity of a 1,8-naphthyridine quinolone, PD 131628. J Med Microbiol. 1992;36(5):353-7. doi: 10.1099/00222615-36-5-353, PMID 1316970.

25.     Gooding BB, Jones RN. In vitro antimicrobial activity of CP-99,219, a novel azabicyclo-naphthyridone. Antimicrob Agents Chemother. 1993;37(2):349-53. doi: 10.1128/AAC.37.2.349, PMID 8043036.

26.     Child J, Andrews J, Boswell F, Brenwald N, Wise R. The in-vitro activity of CP 99,219, a new naphthyridone antimicrobial agent: a comparison with fluoroquinolone agents. J Antimicrob Chemother. 1995;35(6):869-76. doi: 10.1093/jac/35.6.869, PMID 7559198.

27.     Ferrarini PL, Manera C, Mori C, Badawneh M, Saccomanni G. Synthesis and evaluation of antimycobacterial activity of 4-phenyl-1,8-naphthyridine derivatives. Farmaco. 1998;53(12):741-6. doi: 10.1016/s0014-827x(98)00094-9, PMID 10230055.

28.     Badawneh M, Bellini L, Cavallini T, Al jamal J, Manera C, Saccomanni G, Ferrarini PL. Synthesis of 3- or 4-phenyl-1,8-naphthyridine derivatives and evaluation of antimycobacterial and antimicrobial activity. Farmaco. 2003;58(9):859-66. doi: 10.1016/s0014-827x(03)00144-7, PMID 13679180.

29.     Chennam KP, Ravi M, Ushaiah B, Srinu V, Eslavath RK, Devi ChS. Synthesis, characterization, DNA interactions, DNA cleavage, radical scavenging activity, antibacterial, anti-proliferative and docking studies of new transition metal complexes. J Fluoresc. 2016;26(1):189-205. doi: 10.1007/s10895-015-1701-3, PMID 26545354.

30.     Gençer HK, Levent S, Acar Çevik U, Özkay Y, Ilg?n S. New 1,4-dihydro[1,8]naphthyridine derivatives as DNA gyrase inhibitors. Bioorg Med Chem Lett. 2017;27(5):1162-8. doi: 10.1016/j.bmcl.2017.01.073, PMID 28174104.

31.     Mohammed AAM, Suaifan GARY, Shehadeh MB, Okechukwu PN. Design, synthesis, and biological evaluation of 1,8-naphthyridine glucosamine conjugates as antimicrobial agents. Drug Dev Res. 2019;80(1):179-86. doi: 10.1002/ddr.21508, PMID 30570767.

32.     Gurjar VK, Pal D, Mazumder A, Mazumder R. Synthesis, biological evaluation and molecular docking studies of novel 1,8-naphthyridine-3-carboxylic acid derivatives as potential antimicrobial agents (Part-1). Ind J Pharm Sci. 2020; 82:37-42. doi: 10.36468/pharmaceutical-sciences.621.

33.     Taghavi-Moghadam S, Kwong CD, Secrist JA, Khan SI, Clark AM. The synthesis and biological evaluation of alkyl and benzyl naphthyridinium analogs of eupolauridine as potential antimicrobial and cytotoxic agents. Bioorg Med Chem. 2016;24(23):6119-30. doi: 10.1016/j.bmc.2016.02.028, PMID 27769668.

34.     Huang S, Qing J, Wang S, Wang H, Zhang L, Tang Y. Design and synthesis of imidazo[1,2-α][1,8]naphthyridine derivatives as anti-HCV agents via direct C-H arylation. Org Biomol Chem. 2014;12(15):2344-8. doi: 10.1039/c3ob42525h, PMID 24595428.

35.     Falardeau G, Chan L, Stefanac T, May S, Jin H, Lavallée JF. Substituted 1,6-naphthyridines as human cytomegalovirus inhibitors: conformational requirements. Bioorg Med Chem Lett. 2000;10(24):2769-70. doi: 10.1016/s0960-894x(00)00568-0, PMID 11133087.

36.     Melamed JY, Egbertson MS, Varga S, Vacca JP, Moyer G, Gabryelski L, Felock PJ, Stillmock KA, Witmer MV, Schleif W, Hazuda DJ, Leonard Y, Jin L, Ellis JD, Young SD. Synthesis of 5-(1-H or 1-alkyl-5-oxopyrrolidin-3-yl)-8-hydroxy-[1,6]-naphthyridine-7-carboxamide inhibitors of HIV-1 integrase. Bioorg Med Chem Lett. 2008;18(19):5307-10. doi: 10.1016/j.bmcl.2008.08.038, PMID 18774711.

37.     Barlin GB, Tan W. Potential antimalarials. I. 1,8-Naphthyridines. Aust J Chem. 1984;37(5):1065-73. doi: 10.1071/CH9841065.

38.     Barlin GB, Tan W. Potential antimalarials. III. N4-Substituted 7-bromo-1,5-naphthyridin-4-amines. Aust J Chem. 1985;38(3):459-65. doi: 10.1071/CH9850459.

39.     Kandepedu N, Gonzàlez CD, Eedubilli S, Taylor D, Brunschwig C, Gibhard L, Njoroge M, Lawrence N, Paquet T, Eyermann CJ, Spangenberg T, Basarab GS. Street LJ, Chibale K. Identification, characterization, and optimization of 2,8-disubstituted-1,5-naphthyridines as novel Plasmodium falciparum phosphatidylinositol-4-kinase inhibitors with in vivo efficacy in a humanized mouse model of malaria. J Med Chem. 2018;61(13):5692-703.

40.     Tejería A, Pérez-Pertejo Y, Reguera RM, Balaña-Fouce R, Alonso C, Fuertes M, González M, Rubiales G, Palacios F. Antileishmanial effect of new indeno-1,5-naphthyridines, selective inhibitors of Leishmania infantum type IB DNA topoisomerase. Eur J Med Chem. 2016;124:740-9. doi: 10.1016/j.ejmech.2016.09.017, PMID 27639365.

41.     Malfitano AM, Laezza C, Bertini S, Marasco D, Tuccinardi T, Bifulco M, Manera C. Immunomodulatory properties of 1,2-dihydro-4-hydroxy-2-oxo-1,8-naphthyridine-3-carboxamide derivative VL15. Biochimie. 2017;135:173-80. doi: 10.1016/j.biochi.2017.02.009, PMID 28219703.

42.     Hinschberger A, Butt S, Lelong V, Boulouard M, Dumuis A, Dauphin F, Bureau R, Pfeiffer B, Renard P, Rault S. New benzo[h][1,6]naphthyridine and azepino[3,2-c]quinoline derivatives as selective antagonists of 5-HT4 receptors: binding profile and pharmacological characterization. J Med Chem. 2003;46(1):138-47. doi: 10.1021/jm020954v, PMID 12502367.

43.     Kuroda T, Suzuki F, Tamura T, Ohmori K, Hosoe H. A novel synthesis and potent antiinflammatory activity of 4-hydroxy-2(1H)-oxo-1-phenyl-1,8-naphthyridine-3-carboxamides. J Med Chem. 1992;35(6):1130-6. doi: 10.1021/jm00084a019, PMID 1552506.

44.     Anderson VE, Zaniewski RP, Kaczmarek FS, Gootz TS, Osheroff N. Effects of anti-inflammatory [1, 2, 4]triazolo[4,3-a] [1,8]naphthyridine derivatives on human stimulated PMN and endothelial cells: an in vitro study. J Inflam. 2006;3(4):1476-92.

45.     Zengin Kurt B. Synthesis and anticholinesterase activity of novel non-hepatotoxic naphthyridine-11-amine derivatives. Mol Divers. 2019;23(3):625-38. doi: 10.1007/s11030-018-9897-1, PMID 625638.

46.     Almansour AI, Suresh Kumar R, Arumugam N, Basiri A, Kia Y, Ashraf Ali M. An expedient synthesis, acetylcholinesterase inhibitory activity, and molecular modelling study of highly functionalized hexahydro-1,6-naphthyridines. BioMed Res Int. 2015;2015:1-9. doi: 10.1155/2015/965987, PMID 965987.

47.     Fiorito J, Vendome J, Saeed F, Staniszewski A, Zhang H, Yan S, Deng SX, Arancio O, Landry DW. Identification of a novel 1, 2, 3, 4-tetrahydrobenzo[b][1,6]naphthyridine analogue as a potent phosphodiesterase-5 inhibitor with improved aqueous solubility for the treatment of Alzheimer’s disease. J Med Chem. 2017;60(21):8858-75. doi: 10.1021/acs.jmedchem.7b00979, PMID 28985058.

48.     Lovell MA, Lynn BC, Fister S, Bradley-Whitman M, Murphy MP, Beckett TL, Norris CM. A novel small molecule modulator of amyloid pathology. J Alzheimers Dis. 2016;53(1):273-87. doi: 10.3233/JAD-151160, PMID 27163808.

49.     Shiozawa A, Ichikawa Y, Ishikawa M, Kogo Y, Kurashige S, Miyazaki H, Yamanaka H, Sakamoto T. Antivertigo agents. II. Structure–activity relationships of 6-substituted 5,6,7,8-tetrahydro-1,6-naphthyridines. Chem Pharm Bull (Tokyo). 1984;32(3):995-1005. doi: 10.1248/cpb.32.995, PMID 6744495.

50.     Paronikyan EG, Sirakanyan SN, Noravyan AS, Asatryan TO, Markaryan KZ, Aleksanyan RA. Synthesis and antiarrhythmic activity of 7-benzyl(ethyl)-1-hydroxy-4-carbamoyl-3-oxo-5, 6-dihydro-8H-2,7-naphthyridines. Pharm Chem J. 1996;30(6):365-67. doi: 10.1007/BF02219320.

51.     Da Settimo A, Ferrarini PL, Mori C, Primofiore G, Subissi A. Synthesis of 1,8-naphthyridine derivatives. Potential antihypertensive agents. II. Farmaco Sci. 1986;41(11):827-38. PMID 2879740.

52.     Ferrarini PL, Mori C, Calderone V, Calzolari L, Nieri P, Saccomanni G, Martinotti E. Synthesis of 1,8-naphthyridine derivatives: potential antihypertensive agents – Part VIII. Eur J Med Chem. 1999;34(6):505-13. doi: 10.1016/S0223-5234(99)80099-3.

53.     Ferrarini PL, Mori C, Badawneh M, Calderone V, Calzolari L, Loffredo T, Saccomanni G. Synthesis of 1,8-naphthyridine derivatives: potential antihypertensive agents - Part VII. Eur J Med Chem. 1988;33(5):383-97.

54.     Clark RD, Repke DB, Kilpatrick AT, Brown CM, MacKinnon AC, Clague RU, Spedding M. (8a alpha,12a alpha,13a alpha)-5,8,8a,9,10,11,12,12a,13,13a-decahydro- 3-methoxy-12-(methylsulfonyl)-6H-isoquino[2,1-g][1,6]naphthyridi ne, a potent and highly selective alpha 2-adrenoceptor antagonist. J Med Chem. 1989;32(9):2034-6. doi: 10.1021/jm00129a002, PMID 2570150.

55.     Al-Jamal JA, Badawneh M. Characterization of the biochemical effects of new 1,8-naphthyridine derivatives, beta-receptor antagonists, in ventricular myocytes. Arch Pharm (Weinheim). 2003;336(6-7):285-92. doi: 10.1002/ardp.200300781, PMID 12953216.

56.     Manera C, Malfitano AM, Parkkari T, Lucchesi V, Carpi S, Fogli S, Bertini S, Laezza C, Ligresti A, Saccomanni G, Savinainen JR, Ciaglia E, Pisanti S, Gazzerro P, Di Marzo V, Nieri P, Macchia M, Bifulco M. New quinolone- and 1,8-naphthyridine-3-carboxamides as selective CB2 receptor agonists with anticancer and immuno-modulatory activity. Eur J Med Chem. 2015;97:10-8. doi: 10.1016/j.ejmech.2015.04.034, PMID 25935384.

57.     Kong Q, Lv J, Yan S, Chang KJ, Wang G. A novel naphthyridine derivative, 3u, induces necroptosis at low concentrations and apoptosis at high concentrations in human melanoma A375 Cells. Int J Mol Sci. 2018;19(10):2975. doi: 10.3390/ijms19102975, PMID 30274263.

58.     Sinha BK, Sato RI. Synthesis and biological activities of 10-substituted benzo[b][1,5]naphthyridines. J Pharm Sci. 1978;67(3):407-9. doi: 10.1002/jps.2600670337, PMID 347048.

59.     Alonso C, Fuertes M, González M, Rubiales G, Tesauro C, Knudsen BR, Palacios F. Synthesis and biological evaluation of indeno[1,5]naphthyridines as topoisomerase I (TopI) inhibitors with antiproliferative activity. Eur J Med Chem. 2016;115:179-90. doi: 10.1016/j.ejmech.2016.03.031, PMID 27017547.

60.     Li Y, Liang J, Siu T, Hu E, Rossi MA, Barnett SF, Defeo-Jones D, Jones RE, Robinson RG, Leander K, Huber HE, Mittal S, Cosford N, Prasit P. Allosteric inhibitors of Akt1 and Akt2: discovery of [1,2,4]triazolo[3,4-f][1,6]naphthyridines with potent and balanced activity. Bioorg Med Chem Lett. 2009;19(3):834-6. doi: 10.1016/j.bmcl.2008.12.017, PMID 19097777.

61.     Wang Y, Xu ZL, Ai J, Peng X, Lin JP, Ji YC, Geng MY, Long YQ. Investigation on the 1,6-naphthyridine motif: discovery and SAR study of 1H-imidazo[4,5-h][1,6]naphthyridin-2(3H)-one-based c-Met kinase inhibitors. Org Biomol Chem. 2013;11(9):1545-62. doi: 10.1039/c2ob26710a, PMID 23188156.

62.     Montoir D, Barillé-Nion S, Tonnerre A, Juin P, Duflos M, Bazin MA. Novel 1,6-naphthyridin-2(1H)-ones as potential anticancer agents targeting Hsp90. Eur J Med Chem. 2016;119:17-33. doi: 10.1016/j.ejmech.2016.04.050, PMID 27153346.

63.     Arepalli SK, Lee C, Sim S, Lee K, Jo H, Jun KY, Kwon Y, Kang JS, Jung JK, Lee H. Development of 13H-benzo[f]chromeno[4,3-b][1,7]naphthyridines and their salts as potent cytotoxic agents and topoisomerase I/IIα inhibitors. Bioorg Med Chem. 2018;26(18):5181-93. doi: 10.1016/j.bmc.2018.09.019, PMID 30253887.

64.     Hawes EM, Gorecki DK, Gedir RG. 2,3-Disubstituted 1,8-naphthyridines as potential diuretic agents. 2. 5,7-Dimethyl derivatives. J Med Chem. 1977;20(6):838-41. doi: 10.1021/jm00216a021, PMID 874960.

65.     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(12):2270-7. doi: 10.1021/jm00395a015, PMID 3681897.

66.     Liu H, Pang G, Ren J, Zhao Y, Wang J. A novel class of apical sodium-dependent bile salt transporter inhibitors: 1-(2,4-bifluorophenyl)-7-dialkylamino-1,8-naphthyridine-3-carbox-amides. Acta Pharm Sin B. 2017;7(2):223-29. doi: 10.1016/j.apsb.2016.11.005, PMID 28303230.

67.     Frasson I, Spanò V, Di Martino S, Nadai M, Doria F, Parrino B, Carbone A, Cascioferro SM, Diana P, Cirrincione G, Freccero M, Barraja P, Richter SN, Montalbano A. Synthesis and photocytotoxic activity of [1,2,3]triazolo[4,5-h][1,6]naphthyridines and [1,3]oxazolo[5,4-h][1,6]naphthyridines. Eur J Med Chem. 2019;162:176-93. doi: 10.1016/j.ejmech.2018.10.071, PMID 30445266.

 

 

[Download PDF]
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy
Pharmaceutical Fields
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy Pharmaceutics
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy Novel drug delivery system
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy Nanotechnology
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy Pharmacology
Welcome to IJPBS,Pharmaceutics, Novel, drug, delivery, system, Nanotechnology, Pharmacology, Pharmacognosy Pharmacognosy
© Copyright 2009-2015 IJPBS, India. All rights reserved. Specialized online journals by ubijournal. Website by Ubitech Solutions
         Home I Contact I Terms & Conditions