ArticlePDF Available

Synergy Potential of Ursolic Acid-Based Hybrid Molecules

Authors:
  • Rajkiya Engineering College,Sonbhadra
  • Rajkiya Engineering College, Sonbhadra, U.P., India

Abstract

Background Ursolic acid (UA, 3β-hydroxy-urs-12-en-28-oic acid), a pentacyclic triterpenoid from various medicinal plants, has been blessed with proven biological effects, including anti-inflammatory, anticancer, antidiabetic, antioxidant and antibacterial, but its bioavailability and solubility limit its clinical application. Objective Synthesis of UA-based hybrid molecules to explore their antibacterial and synergy potential in combination with azithromycin (AZT) for the treatment of multidrug-resistant (MDR) bacterial infections. Methods Hybrid molecules of UA with menthol, eugenol, and nalidixic acid (NAL) along with some other ester derivatives were synthesized, and evaluated for their antibacterial and synergy potential in combination with AZT against the clinical isolate of Escherichia coli in terms of their minimum inhibitory concentration (MIC), fold reduction in MIC, fractional inhibitory concentration index (FICI) and type of interaction. In silico screening of pharmacokinetic parameters, docking affinity against efflux pump proteins AcrA, AcrB, and TolC was performed on the most potent derivative 7 (3-O-nalidixoyl UA). Results Derivative 7 showed MIC of 62.5 µg/mL and a strong synergistic effect with AZT reducing the MIC of AZT from 100 to 0.19 µg/mL (512-fold reduction) against E. coli at a concentration of 12.5 µg/mL. Other derivatives neither showed antibacterial activity of their own (MIC > 1000 µg/mL) nor any significant synergistic interaction in combination with AZT. The in silico studies on 7 revealed improved druggability parameters over the parent UA and NAL. Conclusion The findings highlight derivative 7 as strong synergistic agent in combination with AZT which may be further investigated to render its efficient use for the treatment of MDR bacterial infections.
Send Orders for Reprints to reprints@benthamscience.net
Letters in Drug Design & Discovery, XXXX, XX, 1-10 1
RESEARCH ARTICLE
1570-1808/XX $65.00+.00 © XXXX Bentham Science Publishers
Synergy Potential of Ursolic Acid-Based Hybrid Molecules
Krishna N. Mishra1, Sonam Singh1, Harish C. Upadhyay1,#,*, Swaroop Sanket2, Manoj Kumar3,
Umme Yashmeen4, Rajni Kant3 and Gaurav R. Dwivedi3,#
1Laboratory of Chemistry, Department of Applied Sciences, Rajkiya Engineering College (Affiliated with Dr. A.P.J. Ab-
dul Kalam Technical University, Lucknow), Churk, Sonbhadra-231206, India; 2Department of Microbiology, Utkal Uni-
versity, Bhubaneswar, Odisha- 751004, India; 3ICMR-Regional Medical Research Center, Gorakhpur-273013, India;
4Department of Biotechnology, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur-273009, India
Abstract: Background: Ursolic acid (UA, 3β-hydroxy-urs-12-en-28-oic acid), a pentacyclic triterpenoid
from various medicinal plants, has been blessed with proven biological effects, including anti-
inflammatory, anticancer, antidiabetic, antioxidant and antibacterial, but its bioavailability and solubility
limit its clinical application.
Objective: Synthesis of UA-based hybrid molecules to explore their antibacterial and synergy potential in
combination with azithromycin (AZT) for the treatment of multidrug-resistant (MDR) bacterial infec-
tions.
Methods: Hybrid molecules of UA with menthol, eugenol, and nalidixic acid (NAL) along with some
other ester derivatives were synthesized, and evaluated for their antibacterial and synergy potential in
combination with AZT against the clinical isolate of Escherichia coli in terms of their minimum
inhibitory concentration (MIC), fold reduction in MIC, fractional inhibitory concentration index (FICI)
and type of interaction. In silico screening of pharmacokinetic parameters, docking affinity against efflux
pump proteins AcrA, AcrB, and TolC was performed on the most potent derivative 7 (3-O-nalidixoyl
UA).
Results: Derivative 7 showed MIC of 62.5 µg/mL and a strong synergistic effect with AZT reducing the
MIC of AZT from 100 to 0.19 µg/mL (512-fold reduction) against E. coli at a concentration of 12.5
µg/mL. Other derivatives neither showed antibacterial activity of their own (MIC > 1000 µg/mL) nor any
significant synergistic interaction in combination with AZT. The in silico studies on 7 revealed improved
druggability parameters over the parent UA and NAL.
Conclusion: The findings highlight derivative 7 as strong synergistic agent in combination with AZT
which may be further investigated to render its efficient use for the treatment of MDR bacterial infections.
A R T I C L E H I S T O R Y
Received: October 28, 2021
Revised: April 01, 2022
Accepted: July 18, 2022
DOI:
10.2174/1570180819666220929143234
Keywords: Hybrid molecules, nalidixic acid, azithromycin, ursolic acid, antibacterial, multidrug-resistant, drug resistance re-
versal.
1. INTRODUCTION
The rapid spread of antimicrobial-resistant bacterial
pathogens marks the onset of a possible severe global health
crisis [1-3]. Antibiotics are becoming increasingly ineffec-
tive in treating infections because of the development of new
resistance mechanisms acquired by pathogenic bacteria [4,
5]. The situation of antimicrobial resistance (AMR) in de-
veloping countries is at an alarming rate due to gross abuse
*Address correspondence to this author at the Laboratory of Chemistry,
Department of Applied Sciences, Rajkiya Engineering College (Affiliated
with Dr. A.P.J. Abdul Kalam Technical University, Lucknow), Churk,
Sonbhadra-231206, India; Tel/Fax: +91-9415164409;
E-mail: harishcu@gmail.com
#These authors contributed equally to this work.
in the use of antimicrobials without any control and effective
monitoring [6, 7]. WHO has declared AMR as one of the top
ten global public health threats facing humanity which re-
quires urgent multisectoral action to achieve sustainable de-
velopment goals [8]. In view of drug resistance, the WHO
listed 12 human pathogenic bacteria (dirty dozen), including
Acinetobacter baumannii, Pseudomonas aeruginosa and
Enterobacteriaceae on which the existing antibiotics are
becoming critically ineffective [9, 10]. The current rate of
emergence and spread of AMR expects to reach 10 million
deaths with an estimated economic cost of $100 trillion
annually by the year 2050 [10, 11]. It is noticeable that the
current antibiotic pipeline is almost dry, also the molecules
under trial are not robust enough to address the current and
projected clinical needs [12, 13]. The study indicates that in
8 Letters in Drug Design & Discovery, XXXX, Vol. XX, No. XX Mishra et al.
Careful analysis of the results of in silico studies indi-
cates that 7 follows the Lipinski Rule of five with violations
for molecular weight and logP values. These violations do
not limit this molecule to being a potential drug candidate as
being a natural product-derived molecule, it may be the sub-
strate for biological transporters allowing it to bypass the
limits of the rule of five for intestinal wall permeability [52].
Further, the binding affinity of 7 with all three receptors is
higher than the parent compounds NAL and UA. The results
of in silico docking and ADMET studies are in good agree-
ment with the assumption that the hybrid molecules may
interact with the target receptors with higher affinity than an
individual bioactive molecule and hence may be a better
alternative to overcome the problem of MDR.
CONCLUSION
Ester derivatives of UA were semi-synthesized, affording
hybrid molecules of UA with menthol, eugenol and NAL in
mild yield. The derivatives were evaluated for their
antibacterial and drug resistance reversal potential in
combination with conventional antibiotic AZT against the
clinical isolates of Escherichia coli. The derivative 7, a
hybrid molecule of UA with NAL was found to be most
potent in reducing MIC of AZT by 512-fold. In further in
silico studies, the 7 showed the highest binding affinity with
receptors AcrA, AcrB and TolC involved in the efflux pump
mechanism responsible for MDR. Our study suggests that
after detailed investigations, derivative 7 may find its poten-
tial use as a co-drug in the treatment of multidrug-resistant
bacterial infections.
LIST OF ABBREVIATIONS
ADME = Absorption, Distribution, Metabolism and
Excretion
DCC = Dicyclohexylcarbodiimide
DMAP = 4-dimethylaminopyridine
FICI = Fractional inhibitory concentration index
IC50 = Half Maximal Inhibitory Concentration
MDR = Multidrug Resistant
MIC = Minimum Inhibitory concentration
MRSA = Methicillin-resistant Staphylococcus
aureus
NAL = Nalidixic acid
SAR = Structure-Activity Relationship
UA = Ursolic acis
USFDA = United States Food and Drug
Administration
WHO = World Health Organization
ETHICS APPROVAL AND CONSENT TO
PARTICIPATE
Not applicable.
HUMAN AND ANIMAL RIGHTS
No animals/humans were used for studies that are the
basis of this research.
CONSENT FOR PUBLICATION
Not applicable.
AVAILABILITY OF DATA AND MATERIALS!
The authors confirm that the data supporting the findings
of this study are available within the article and it’s
supplementary material.
FUNDING!
None.!
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
HCU thanks Dr. S.K. Srivastava, Scientist CSIR-Central
Institute of medicinal and Aromatic Plants, Lucknow (India),
for providing ursolic acid and helping in spectroscopic anal-
ysis of the samples and Director, Rajkiya Engineering
College, Sonbhadra for his constant encouragement and
providing all types of facilities for this study.
SUPPLEMENTARY MATERIAL
A detailed description of the receptor proteins AcrA,
AcrB and TolC and ligand-protein interaction profiles have
been provided as supporting information.
REFERENCES
[1] Yang, Y.H.; Buttery, J. Antimicrobial resistance: A global one-
health problem for all ages. World J. Pediatr., 2018, 14(6), 521-
522.
http://dx.doi.org/10.1007/s12519-018-0194-y PMID: 30298235
[2] Maddocks, S. Antimicrobial resistance: Global problems need
global solutions. Med. J. Aust., 2013, 198(5), 241.
http://dx.doi.org/10.5694/mja13.c0318 PMID: 23496384
[3] Hu, X.Y.; Logue, M.; Robinson, N. Antimicrobial resistance is a
global problem-A UK perspective. Eur. J. Integr. Med., 2020, 36,
101136.
http://dx.doi.org/10.1016/j.eujim.2020.101136 PMID: 32399092
[4] Christaki, E.; Marcou, M.; Tofarides, A. antimicrobial resistance in
bacteria: Mechanisms, evolution, and persistence. J. Mol. Evol.,
2020, 88(1), 26-40.
http://dx.doi.org/10.1007/s00239-019-09914-3 PMID: 31659373
[5] Morrison, L.; Zembower, T.R. Antimicrobial resistance. Gastroin-
test. Endosc. Clin. N. Am., 2020, 30(4), 619-635.
http://dx.doi.org/10.1016/j.giec.2020.06.004 PMID: 32891221
[6] WHO. Global antimicrobial resistance and use surveillance system
(GLASS) report. 2020. Available from: https://www.who.int/initiatives/
glass (Accessed on: August 01, 2021).
[7] Sonam Singh, ; Sanket, A.S.; Dwivedi, G.R.; Upadhyay, H.C. In-
silico Druggability Studies of 4-hydroxy-α-tetralone and its deriva-
tives with RND Efflux pump of E. coli. Pharma. Biosci. J., 2020,
8(2), 21-26.
http://dx.doi.org/10.20510/ukjpb/8/i2/1586224632
Synergy Potential of Ursolic Acid-Based Hybrid Molecules Letters in Drug Design & Discovery, XXXX, Vol. XX, No. XX 9
[8] WHO. Antimicrobial resistance. Available from: https://www.who.
int/health-topics/antimicrobial-resistance (Accessed on: August 01,
2021).
[9] Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson,
M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Car-
meli, Y.; Ouellette, M.; Outterson, K.; Patel, J.; Cavaleri, M.; Cox,
E.M.; Houchens, C.R.; Grayson, M.L.; Hansen, P.; Singh, N.;
Theuretzbacher, U.; Magrini, N.; Aboderin, A.O.; Al-Abri, S.S.;
Awang Jalil, N.; Benzonana, N.; Bhattacharya, S.; Brink, A.J.;
Burkert, F.R.; Cars, O.; Cornaglia, G.; Dyar, O.J.; Friedrich, A.W.;
Gales, A.C.; Gandra, S.; Giske, C.G.; Goff, D.A.; Goossens, H.;
Gottlieb, T.; Guzman Blanco, M.; Hryniewicz, W.; Kattula, D.;
Jinks, T.; Kanj, S.S.; Kerr, L.; Kieny, M-P.; Kim, Y.S.; Kozlov,
R.S.; Labarca, J.; Laxminarayan, R.; Leder, K.; Leibovici, L.;
Levy-Hara, G.; Littman, J.; Malhotra-Kumar, S.; Manchanda, V.;
Moja, L.; Ndoye, B.; Pan, A.; Paterson, D.L.; Paul, M.; Qiu, H.;
Ramon-Pardo, P.; Rodríguez-Baño, J.; Sanguinetti, M.; Sengupta,
S.; Sharland, M.; Si-Mehand, M.; Silver, L.L.; Song, W.; Stein-
bakk, M.; Thomsen, J.; Thwaites, G.E.; van der Meer, J.W.M.; Van
Kinh, N.; Vega, S.; Villegas, M.V.; Wechsler-Fördös, A.;
Wertheim, H.F.L.; Wesangula, E.; Woodford, N.; Yilmaz, F.O.;
Zorzet, A. Discovery, research, and development of new antibiot-
ics: The WHO priority list of antibiotic-resistant bacteria and tu-
berculosis. Lancet Infect. Dis., 2018, 18(3), 318-327.
http://dx.doi.org/10.1016/S1473-3099(17)30753-3 PMID:
29276051
[10] Upadhyay, H.C. Coumarin-1,2,3-triazole hybrid molecules: An
emerging scaffold for combating drug resistance. Curr. Top. Med.
Chem., 2021, 21(8), 737-752.
http://dx.doi.org/10.2174/1568026621666210303145759 PMID:
33655863
[11] de Kraker, M.E.A.; Stewardson, A.J.; Harbarth, S. Will 10 million
people die a year due to antimicrobial resistance by 2050? PLoS
Med., 2016, 13(11), e1002184.
http://dx.doi.org/10.1371/journal.pmed.1002184 PMID: 27898664
[12] Li, X.Z.; Plésiat, P.; Nikaido, H. The challenge of efflux-mediated
antibiotic resistance in Gram-negative bacteria. Clin. Microbiol.
Rev., 2015, 28(2), 337-418.
http://dx.doi.org/10.1128/CMR.00117-14 PMID: 25788514
[13] Simpkin, V.L.; Renwick, M.J.; Kelly, R.; Mossialos, E. Incentivis-
ing innovation in antibiotic drug discovery and development: Pro-
gress, challenges and next steps. J. Antibiot. (Tokyo), 2017, 70(12),
1087-1096.
http://dx.doi.org/10.1038/ja.2017.124 PMID: 29089600
[14] Fair, R.J.; Tor, Y. Antibiotics and bacterial resistance in the 21st
century. Perspect. Medicin. Chem., 2014, 6, PMC.S14459.
http://dx.doi.org/10.4137/PMC.S14459 PMID: 25232278
[15] Upadhyay, H.C. Medicinal chemistry of alternative therapeutics:
Novelty and hopes with genus Ammannia. Curr. Top. Med. Chem.,
2019, 19(10), 784-794.
http://dx.doi.org/10.2174/1568026619666190412101047 PMID:
30977452
[16] Upadhyay, H.C.; Verma, R.K.; Srivastava, S.K. Quantitative de-
termination of bioactive 4-hydroxy-α-tetralone, tetralone-4-O-β-D-
glucopyranoside and ellagic acid in Ammannia baccifera (Linn.) by
reversed-phase high-performance liquid chromatography. J. Chro-
matogr. Sci., 2013, 51(1), 21-25.
http://dx.doi.org/10.1093/chromsci/bms099 PMID: 22700790
[17] Upadhyay, H.C.; Thakur, J.P.; Saikia, D.; Srivastava, S.K. Anti-
tubercular agents from Ammannia baccifera (Linn.). Med. Chem.
Res., 2013, 22(1), 16-21.
http://dx.doi.org/10.1007/s00044-012-9998-9
[18] Upadhyay, H.C.; Sisodia, B.S.; Verma, R.K.; Darokar, M.P.; Sri-
vastava, S.K. Antiplasmodial potential of extracts from two species
of genus Blumea. Pharm. Biol., 2013, 51(10), 1326-1330.
http://dx.doi.org/10.3109/13880209.2013.790453 PMID: 23767769
[19] Katiyar, C.; Kanjilal, S.; Gupta, A.; Katiyar, S. Drug discovery
from plant sources: An integrated approach. Ayu, 2012, 33(1), 10-
19.
http://dx.doi.org/10.4103/0974-8520.100295 PMID: 23049178
[20] Newman, D.J.; Cragg, G.M. Natural products as sources of new
drugs over the nearly four decades from 01/1981 to 09/2019. J.
Nat. Prod., 2020, 83(3), 770-803.
http://dx.doi.org/10.1021/acs.jnatprod.9b01285 PMID: 32162523
[21] Upadhyay, H.C.; Mishra, A.; Pandey, J.; Sharma, P.; Tamrakar,
A.K.; Srivastava, A.K.; Khan, F.; Srivastava, S.K. In vitro, in vivo
and in silico antihyperglycemic activity of some semi-synthetic
phytol derivatives. Med. Chem., 2021, 18(1), 115-121.
http://dx.doi.org/10.2174/1573406417666201216124018 PMID:
33327922
[22] Saxena, A.; Upadhyay, H.C.; Cheema, H.S.; Srivastava, S.K.;
Darokar, M.P.; Bawankule, D.U. Antimalarial activity of phytol de-
rivatives: In vitro and in vivo study. Med. Chem. Res., 2018, 27(5),
1345-1354.
http://dx.doi.org/10.1007/s00044-017-2132-2
[23] Sultana, N. Clinically useful anticancer, antitumor, and antiwrinkle
agent, ursolic acid and related derivatives as medicinally important
natural product. J. Enzyme Inhib. Med. Chem., 2011, 26(5), 616-
642.
http://dx.doi.org/10.3109/14756366.2010.546793 PMID: 21417964
[24] Mlala, S.; Oyedeji, A.O.; Gondwe, M.; Oyedeji, O.O. Ursolic acid
and its derivatives as bioactive agents. Molecules, 2019, 24(15),
2751.
http://dx.doi.org/10.3390/molecules24152751 PMID: 31362424
[25] Maurya, A.; Srivastava, S.K. Preparative-scale separation of anti-
cancer triterpenes from Eucalyptus hybrid by centrifugal partition
chromatography. Sep. Sci. Technol., 2011, 46(7), 1189-1194.
http://dx.doi.org/10.1080/01496395.2010.545793
[26] Pironi, A.M.; de Araújo, P.R.; Fernandes, M.A.; Salgado, H.R.N.;
Chorilli, M. Characteristics, biological properties and analytical
methods of ursolic acid: A review. Crit. Rev. Anal. Chem., 2018,
48(1), 86-93.
http://dx.doi.org/10.1080/10408347.2017.1390425 PMID:
29039968
[27] Feng, X.M.; Su, X.L. Anticancer effect of ursolic acid via mito-
chondria-dependent pathways. Oncol. Lett., 2019, 17(6), 4761-
4767.
http://dx.doi.org/10.3892/ol.2019.10171 PMID: 31186681
[28] Chen, H.; Gao, Y.; Wang, A.; Zhou, X.; Zheng, Y.; Zhou, J. Evolu-
tion in medicinal chemistry of ursolic acid derivatives as anticancer
agents. Eur. J. Med. Chem., 2015, 92, 648-655.
http://dx.doi.org/10.1016/j.ejmech.2015.01.031 PMID: 25617694
[29] Yin, R.; Li, T.; Tian, J.X.; Xi, P.; Liu, R.H. Ursolic acid, a potential
anticancer compound for breast cancer therapy. Crit. Rev. Food
Sci. Nutr., 2018, 58(4), 568-574.
http://dx.doi.org/10.1080/10408398.2016.1203755 PMID:
27469428
[30] Stiti, N.; Hartmann, M.A. Nonsterol triterpenoids as major constit-
uents of Olea Europaea. J. Lipids, 2012, 2012, 1-13.
http://dx.doi.org/10.1155/2012/476595 PMID: 22523691
[31] Batra, A.; Sastry, V.G. Extraction of ursolic acid from Ocimum
sanctum and synthesis of its novel derivatives: Effects on extracel-
lular homocysteine, dihydrofolate reductase activity and prolifera-
tion of HepG2 human hepatoma cells. Pteridines, 2013, 24(3-4),
191-199.
http://dx.doi.org/10.1515/pterid-2013-0023
[32] Sharma, S.B.; Gupta, R. Drug development from natural resource:
A systematic approach. Mini Rev. Med. Chem., 2015, 15(1), 52-57.
http://dx.doi.org/10.2174/138955751501150224160518 PMID:
25986040
[33] Balunas, M.J.; Kinghorn, A.D. Drug discovery from medicinal
plants. Life Sci., 2005, 78(5), 431-441.
http://dx.doi.org/10.1016/j.lfs.2005.09.012 PMID: 16198377
[34] Hussain, H.; Green, I.R.; Ali, I.; Khan, I.A.; Ali, Z.; Al-Sadi, A.M.;
Ahmed, I. Ursolic acid derivatives for pharmaceutical use: A patent
review (2012-2016). Expert Opin. Ther. Pat., 2017, 27(9), 1061-
1072.
http://dx.doi.org/10.1080/13543776.2017.1344219 PMID:
28637397
[35] Shaveta,; Mishra, S.; Singh, P. Hybrid molecules: The privileged
scaffolds for various pharmaceuticals. Eur. J. Med. Chem., 2016,
124, 500-536.
http://dx.doi.org/10.1016/j.ejmech.2016.08.039 PMID: 27598238
[36] Wayne, P. Clinical and Laboratory Standards Institute. Perfor-
mance standards for antimicrobial susceptibility testing, 29th Ed.;
CLSI supplement M100. Clinical and Laboratory Standards Insti-
tute 2019, 39.
[37] Dwivedi, G.R.; Maurya, A.; Yadav, D.K.; Singh, V.; Khan, F.;
Gupta, M.K.; Singh, M.; Darokar, M.P.; Srivastava, S.K. Synergy
10 Letters in Drug Design & Discovery, XXXX, Vol. XX, No. XX Mishra et al.
of clavine alkaloid ‘chanoclavine’ with tetracycline against multi-
drug-resistant E. coli. J. Biomol. Struct. Dyn., 2019, 37(5), 1307-
1325.
http://dx.doi.org/10.1080/07391102.2018.1458654 PMID:
29595093
[38] Dwivedi, G.R.; Maurya, A.; Yadav, D.K.; Khan, F.; Gupta, M.K.;
Gupta, P.; Darokar, M.P.; Srivastava, S.K. Comparative drug re-
sistance reversal potential of natural glycosides: Potential of syner-
gy Niaziridin & Niazirin. Curr. Top. Med. Chem., 2019, 19(10),
847-860.
http://dx.doi.org/10.2174/1568026619666190412120008 PMID:
30977451
[39] Odds, F.C. Synergy, antagonism, and what the chequerboard puts
between them. J. Antimicrob. Chemother., 2003, 52(1), 1.
http://dx.doi.org/10.1093/jac/dkg301 PMID: 12805255
[40] Tian, W.; Chen, C.; Lei, X.; Zhao, J.; Liang, J. CASTp 3.0: Com-
puted atlas of surface topography of proteins. Nucleic Acids Res.,
2018, 46(W1), W363-W367.
http://dx.doi.org/10.1093/nar/gky473 PMID: 29860391
[41] Yu, E.W.; McDermott, G.; Zgurskaya, H.I.; Nikaido, H.; Koshland,
D.E., Jr Structural basis of multiple drug-binding capacity of the
AcrB multidrug efflux pump. Science, 2003, 300(5621), 976-980.
http://dx.doi.org/10.1126/science.1083137 PMID: 12738864
[42] Wouatsa, V.N.A.; Misra, L.; Kumar, S.; Prakash, O.; Khan, F.;
Tchoumbougnang, F.; Venkatesh, R.K. Aromatase and glycosyl
transferase inhibiting acridone alkaloids from fruits of Came-
roonian zanthoxylumspecies. Chem. Cent. J., 2013, 7(1), 125.
http://dx.doi.org/10.1186/1752-153X-7-125 PMID: 23866063
[43] Upadhyay, H.C.; Singh, M.; Prakash, O.; Khan, F.; Srivastava,
S.K.; Bawankule, D.U. QSAR, ADME and docking guided semi-
synthesis and in vitro evaluation of 4-hydroxy-α-tetralone analogs
for anti-inflammatory activity. SN Appl. Sci., 2020, 2(12), 2069.
http://dx.doi.org/10.1007/s42452-020-03798-5
[44] Upadhyay, H.C.; Dwivedi, G.R.; Roy, S.; Sharma, A.; Darokar,
M.P.; Srivastava, S.K. Phytol derivatives as drug resistance rever-
sal agents. ChemMedChem, 2014, 9(8), 1860-8.
http://dx.doi.org/10.1002/cmdc.201402027 PMID: 24891085
[45] Neises, B.; Steglich, W. Simple method for the esterification of
carboxylic acids. Angew. Chem. Int. Ed. Engl., 1978, 17(7), 522-
524.
http://dx.doi.org/10.1002/anie.197805221
[46] Papi Reddy, K.; Singh, A.B.; Puri, A.; Srivastava, A.K.; Narender,
T. Synthesis of novel triterpenoid (lupeol) derivatives and their in
vivo antihyperglycemic and antidyslipidemic activity. Bioorg. Med.
Chem. Lett., 2009, 19(15), 4463-4466.
http://dx.doi.org/10.1016/j.bmcl.2009.05.034 PMID: 19515563
[47] Cos, P.; Vlietinck, A.J.; Berghe, D.V.; Maes, L. Anti-infective
potential of natural products: How to develop a stronger in vitro
‘proof-of-concept’. J. Ethnopharmacol., 2006, 106(3), 290-302.
http://dx.doi.org/10.1016/j.jep.2006.04.003 PMID: 16698208
[48] Dwivedi, G.R.; Rai, R.; Pratap, R.; Singh, K.; Pati, S.; Sahu, S.N.;
Kant, R.; Darokar, M.P.; Yadav, D.K. Drug resistance reversal po-
tential of multifunctional thieno[3,2-c]pyran via potentiation of an-
tibiotics in MDR P. aeruginosa. Biomed. Pharmacother., 2021,
142, 112084.
http://dx.doi.org/10.1016/j.biopha.2021.112084 PMID: 34449308
[49] Mulani, M.S.; Kamble, E.E.; Kumkar, S.N.; Tawre, M.S.; Pardesi,
K.R. Emerging strategies to combat eskape pathogens in the era of
antimicrobial resistance: A review. Front. Microbiol., 2019, 10,
539.
http://dx.doi.org/10.3389/fmicb.2019.00539 PMID: 30988669
[50] Upadhyay, H.; Dwivedi, G.; Darokar, M.; Chaturvedi, V.; Srivasta-
va, S. Bioenhancing and antimycobacterial agents from Ammannia
multiflora. Planta Med., 2012, 78(1), 79-81.
http://dx.doi.org/10.1055/s-0031-1280256 PMID: 21969115
[51] Nikaido, H.; Takatsuka, Y. Mechanisms of RND multidrug efflux
pumps. Biochim. Biophys. Acta. Proteins Proteomics, 2009,
1794(5), 769-781.
http://dx.doi.org/10.1016/j.bbapap.2008.10.004 PMID: 19026770
[52] Pollastri, M.P. Overview on the rule of five. Curr. Protoc. Phar-
macol., 2010.
http://dx.doi.org/10.1002/0471141755.ph0912s49
DISCLAIMER: The above article has been published, as is, ahead-of-print, to provide early visibility but is not the final ver-
sion. Major publication processes like copyediting, proofing, typesetting and further review are still to be done and may lead to
changes in the final published version, if it is eventually published. All legal disclaimers that apply to the final published article
also apply to this ahead-of-print version.
... Scientists explore the structure-activity relationships (SAR) between existing functional groups and a specific disease or debility in an effort to increase the selectivity of such molecules [9,10]. This strategy has produced many exciting leads with improved efficiency over the parent phytomolecules [11][12][13]. The fact that over half of the drugs licensed by the United States Food and Drug Administration (USFDA) between 1981 and 2019 contain molecules that were initially found from plants or their derivatives is a testimony to the sector's mastery of natural products in drug discovery [14]. ...
Article
Full-text available
The growing prevalence of pathogens resistant to several drugs poses an urgent health concern, which can only be addressed by increasing support for the research and development of newer antibiotics. Drug discovery through the exploration of nature's treasures is a successful and ongoing endeavor. However, the conventional method of discovering new drugs from natural sources has become an adjunct choice of the pharmaceutical corporations since it is time-consuming, complicated, expensive, and fraught with uncertainty. The application of several computational techniques, including combinatorial chemistry, metagenomics, and high throughput screening, has accelerated the drug development process simultaneously reducing costs and time. It is a well-established fact that synthetic drugs generated through computer-aided research are frequently linked to a restricted spectrum of pharmacological activity and adverse side effects as compared to treatments that are either directly or indirectly derived from natural sources. Hence, an innovative drug discovery program may be centered on the virtual design of molecules using a scaffold with a natural origin in order to develop effective and affordable therapies with a wide variety of functionality.
Article
Full-text available
A series of maslinic acid (MA) and oleanolic acid (OA)-based analogs with potent antibacterial and antibiofilm activities were designed and semi-synthesized starting from isolated pentacyclic triterpene acids as natural products...
Article
Full-text available
Green synthesis of silver nanoparticles (AgNPs) using the Alysicarpus vaginalis alcoholic extract as a reducing agent and a capping ligand was carried out. Notable peaks of λmax 442–602 nm indicated the reduction of silver ions to elemental AgNPs. Particle size (Zavg) was in the range of 32.3±0.76 to 252.3±0.342 nm). The negative Zeta potential (ZP, −2.9±0.31 to −21.7±0.13 mV) and low polydispersity index (PDI, 0.181±0.02 to 0.635±0.02) supported the homogeneous nature of AgNPs. Further, crystalline spherical nature of AgNPs with a smooth surface was confirmed by high‐resolution transmission electron microscopy (HRTEM) studies. The AgNPs showed 1.2 to 1.4‐fold enhanced antibacterial activity than extract (6–10 mm zone of inhibition) against Staphylococcus aureus, Pseudomonas aeruginosa, Listeria monocytogenes, and Salmonella enterica ser. Typhi by disc diffusion assay. The AgNP formulation F9 showed equal (against Gram‐positive L. monocytogenes) or even better (against Gram‐negative S. enterica Typhi) antimicrobial activity than the positive control, ciprofloxacin. Further investigations on F9 are suggested in order to develop a broad‐spectrum, cost‐effective antibacterial formulation of natural origin. This study represents a significant step forward in the field of bio‐fabrication and presents a promising alternative to traditional chemical synthesis methods for the production of antimicrobial AgNPs.
Article
Full-text available
We explored the antibacterial potential (alone and combination) against multidrug resistant (MDR) Pseudomonas aeruginosa isolates KG-P2 using synthesized thieno[3,2-c]pyran-2-ones in combination with different antibiotics. Out of 14 compounds, two compounds (3g and 3l) abridged the MIC of tetracycline (TET) by 16 folds. Compounds was killing the KG-P2 cells, in time dependent manner, lengthened post-antibiotic effect (PAE) of TET and found decreased the mutant prevention concentration (MPC) of TET. In ethidium bromide efflux experiment, two compounds repressed the drug transporter (efflux pumps) which is further supported by molecular docking of these compounds with efflux complex MexAB-OprM. In another study, these compounds inhibited the synthesis of biofilm.
Article
Full-text available
No doubt antibiotics have saved billions of lives, but lack of novel antibiotics, development of resistance mechanisms in almost all clinical isolates of bacteria, and recurrent infections caused by persistent bacteria hamper the successful treatment of infections. Due to widespread emergence of resistance, even the new families of antimicrobial agents have a short life expectancy. Drugs acting on single target often lead to drug resistance and are associated with various side effects. To overcome this problem either multidrug therapy or single drug acting on multiple targets may be used. The later are called 'hybrid molecules' which are formed by clubbing two biologically active pharmacophores together with or without an appropriate linker. In this rapidly evolving era, the development of natural product-based hybrid molecules may be a super-alternative to multidrug therapy to combat drug resistance caused by various bacterial and fungal strains. Coumarins (benzopyran-2-one) are one of the earliest reported plant secondary metabolites having clinically proven diverse range of pharmacological properties. On the other hand, 1,2,3-triazole is a common pharmacophore in many drugs responsible for polar interactions improving the solubility and binding affinity to biomolecular targets. In this review we discuss recent advances in Coumarin-1,2,3-triazole hybrids as potential antibacterial agents aiming to provide a useful platform for the exploration of new leads with broader spectrum, more effectiveness, less toxicity with multiple modes of action for the development of cost-effective and safer drugs in the future.
Article
Full-text available
Background Due to the prevalence of type-2 diabetes across the globe, there is unmet need to explore new molecular targets for the development of cost-effective and safer antihyperglycemic agents. Objective Structural modification of phytol and evaluation of in vitro, in vivo and in silico antihyperglycemic activity of derivatives establishing the preliminary structure activity relationship (SAR). Methods The semi-synthetic derivatives of phytol were prepared following previously described methods. The antihyperglycemic potential was measured in vitro in terms of increase in 2-deoxyglucose (2-DG) uptake by L-6 rat skeletal muscle cells as well as in vivo in sucrose-loaded (SLM) and streptozotocin (STZ)-induced diabetic rat models. The blood glucose profile was measured at 30, 60, 90, 120, 180, 240, 300 and 1440 min post administration of sucrose in rats. The in silico docking was performed on peroxisome proliferator-activated receptor gamma (PPARγ) as anti-diabetic target along with absorption, distribution, metabolism, excretion and toxicity (ADMET) studies. Results Nine semi-synthetic ester derivatives: acetyl (1), lauroyl (2), palmitoyl (3), pivaloyl (4), trans-crotonyl (5), benzoyl (6), m-anisoyl (7), 3,4,5-trimethoxy benzoyl (8) cinnamoyl (9) along with bromo derivative (10) of phytol were prepared. The derivatives 9, 8 and 2 caused 4.5, 3.2 and 2.7 times more in vitro uptake of 2-DG respectively than rosiglitazone (ROSI). The derivatives showed significant improvement on oral glucose tolerance both in SLM (29.6-21%) as well as STZ-induced diabetic (30.8-19.0%) rats. The in silico ADMET, docking studies showed non-toxicity and high binding affinity with PPARγ. Conclusion The potent antihyperglycemic activity with favorable pharmacokinetics supports phytol derivatives as suitable antidiabetic lead.
Article
Full-text available
The compound 4-hydroxy-α-tetralone (1) is major bioactive secondary metabolite of genus Ammannia (Family- Lythraceae). The compound 1 and its various derivatives are reported to possess anti-tubercular, anti-diabetic, anti-leishmanial and bioenhancing activities. A quantitative structure activity relationship (QSAR) model for predicting anti-inflammatory activity of 4-hydroxy-α-tetralone derivatives against the tumour necrosis factor (TNF)-α, a pro-inflammatory cytokine was developed by non-linear model using artificial neural network (ANN). The regression coefficient (r²) and the leave-one-out cross-validation regression coefficient (LOO rCV²) of the QSAR model were 0.6976 and 0.4016, respectively, while the regression coefficient (r²) for the external set of experimental compounds was 0.835. The 4-hydroxy-α-tetralone virtual derivatives, which showed significant inhibition of TNF-α were subjected to docking and in-silico absorption, distribution, metabolism and excretion (ADME) studies and the results showed similar binding affinity and bioavailability in compliance with the standard drug, Diclofenac. Finally, in order to validate the developed QSAR model, the most and least active virtual derivatives were semi-synthesized, characterized on the basis of their ¹H and ¹³C NMR spectroscopic data and in vitro tested for concentration dependent inhibition of TNF-α. The experimental results obtained, agreed well with the predicted values.
Article
Full-text available
The compound 4-hydroxy-α-tetralone (1) has been reported to possess potent anti-tubercular, anti-diabetic and anti-leishmanial activities. In our earlier studies the compound 1 and its various semi-synthetic derivatives showed potent bioenhancing activity in combination with nalidixic acid (NAL) and tetracycline (TET) reducing the minimum inhibitory concentration (MIC) of antibiotics up to 8 folds by inhibition of ABC efflux pump. However, in gram negative bacteria, resistance nodulation division (RND) family are considered as major efflux pump responsible for multidrug resistance (MDR). Hence, the current study was carried out to access the in-silico docking potential of compound 1 and its cinnamoyl (1a), 3, 4, 5-trimethoxybenzoyl (1b) derivatives against RND efflux pump target proteins AcrA, AcrB, TolC of E. coli. The docking study showed that the test compounds have good binding affinity with target proteins. The derivative 1a showed highest interaction with AcrA followed by AcrB showing binding energies-8.7 and-8.2 kcal/mol respectively. The low molecular weight ≤500, high hydrogen bonding, high log p value (>1) with hydroxy, methoxy and aromatic group of ligands make these compounds as effective efflux pump inhibitor. In drug likeliness studies, these compounds pass the safety criteria with enhanced bioavailability and absorption, less acute oral toxicity, less hepatotoxicity. This study promises that the compound 1 and its derivatives (1a & 1b) might be RND efflux pump inhibitors providing the initial platform for development of safer and cost-effective antibacterial drug to manage MDR infections.
Article
Full-text available
This review is an updated and expanded version of the five prior reviews that were published in this journal in 1997, 2003, 2007, 2012, and 2016. For all approved therapeutic agents, the time frame has been extended to cover the almost 39 years from the first of January 1981 to the 30th of September 2019 for all diseases worldwide and from ∼1946 (earliest so far identified) to the 30th of September 2019 for all approved antitumor drugs worldwide. As in earlier reviews, only the first approval of any drug is counted, irrespective of how many “biosimilars” or added approvals were subsequently identified. As in the 2012 and 2016 reviews, we have continued to utilize our secondary subdivision of a “natural product mimic”, or “NM”, to join the original primary divisions, and the designation “natural product botanical”, or “NB”, to cover those botanical “defined mixtures” now recognized as drug entities by the FDA (and similar organizations). From the data presented in this review, the utilization of natural products and/or synthetic variations using their novel structures, in order to discover and develop the final drug entity, is still alive and well. For example, in the area of cancer, over the time frame from 1946 to 1980, of the 75 small molecules, 40, or 53.3%, are N or ND. In the 1981 to date time frame the equivalent figures for the N* compounds of the 185 small molecules are 62, or 33.5%, though to these can be added the 58 S* and S*/NMs, bringing the figure to 64.9%. In other areas, the influence of natural product structures is quite marked with, as expected from prior information, the anti-infective area being dependent on natural products and their structures, though as can be seen in the review there are still disease areas (shown in Table 2) for which there are no drugs derived from natural products. Although combinatorial chemistry techniques have succeeded as methods of optimizing structures and have been used very successfully in the optimization of many recently approved agents, we are still able to identify only two de novo combinatorial compounds (one of which is a little speculative) approved as drugs in this 39-year time frame, though there is also one drug that was developed using the “fragment-binding methodology” and approved in 2012. We have also added a discussion of candidate drug entities currently in clinical trials as “warheads” and some very interesting preliminary reports on sources of novel antibiotics from Nature due to the absolute requirement for new agents to combat plasmid-borne resistance genes now in the general populace. We continue to draw the attention of readers to the recognition that a significant number of natural product drugs/leads are actually produced by microbes and/or microbial interactions with the “host from whence it was isolated”; thus we consider that this area of natural product research should be expanded significantly.
Article
Full-text available
In recent years, we have seen antimicrobial resistance rapidly emerge at a global scale and spread from one country to the other faster than previously thought. Superbugs and multidrug-resistant bacteria are endemic in many parts of the world. There is no question that the widespread use, overuse, and misuse of antimicrobials during the last 80 years have been associated with the explosion of antimicrobial resistance. On the other hand, the molecular pathways behind the emergence of antimicrobial resistance in bacteria were present since ancient times. Some of these mechanisms are the ancestors of current resistance determinants. Evidently, there are plenty of putative resistance genes in the environment, however, we cannot yet predict which ones would be able to be expressed as phenotypes in pathogenic bacteria and cause clinical disease. In addition, in the presence of inhibitory and sub-inhibitory concentrations of antibiotics in natural habitats, one could assume that novel resistance mechanisms will arise against antimicrobial compounds. This review presents an overview of antimicrobial resistance mechanisms, and describes how these have evolved and how they continue to emerge. As antimicrobial strategies able to bypass the development of resistance are urgently needed, a better understanding of the critical factors that contribute to the persistence and spread of antimicrobial resistance may yield innovative perspectives on the design of such new therapeutic targets.
Article
Full-text available
Non-communicable diseases (NCDs) such as cancer, diabetes, and chronic respiratory and cardiovascular diseases continue to be threatening and deadly to human kind. Resistance to and side effects of known drugs for treatment further increase the threat, while at the same time leaving scientists to search for alternative sources from nature, especially from plants. Pentacyclic triterpenoids (PT) from medicinal plants have been identified as one class of secondary metabolites that could play a critical role in the treatment and management of several NCDs. One of such PT is ursolic acid (UA, 3 β-hydroxy-urs-12-en-28-oic acid), which possesses important biological effects, including anti-inflammatory, anticancer, antidiabetic, antioxidant and antibacterial effects, but its bioavailability and solubility limits its clinical application. Mimusops caffra, Ilex paraguarieni, and Glechoma hederacea, have been reported as major sources of UA. The chemistry of UA has been studied extensively based on the literature, with modifications mostly having been made at positions C-3 (hydroxyl), C12-C13 (double bonds) and C-28 (carboxylic acid), leading to several UA derivatives (esters, amides, oxadiazole quinolone, etc.) with enhanced potency, bioavailability and water solubility. This article comprehensively reviews the information that has become available over the last decade with respect to the sources, chemistry, biological potency and clinical trials of UA and its derivatives as potential therapeutic agents, with a focus on addressing NCDs.
Article
Antimicrobial resistance is developing rapidly and threatens to outstrip the rate at which new antimicrobials are introduced. Genetic recombination allows bacteria to rapidly disseminate genes encoding for antimicrobial resistance within and across species. Antimicrobial use creates a selective evolutionary pressure, which leads to further resistance. Antimicrobial stewardship, best use, and infection prevention are the most effective ways to slow the spread and development of antimicrobial resistance.
Article
Increasingly we are reaching a situation where current antimicrobial medicines are no longer effective for common infections, and antimicrobial resistance (AMR) is becoming a global public health crisis. The reliance on antimicrobials such as antibiotics has become a major issue for both medicine and agriculture, particularly given the slow development of new medicines and pharmaceutical industry investment. The UK government has been working with other international bodies in the search for solutions to the many challenges AMR poses. Herbal medicines may provide a useful modality in the fight against AMR and can work solely or in tandem with current antimicrobial approaches. Recommendations for herbal medicine use during the COVID-19 outbreak have featured in Chinese national guidelines and policies, but UK strategies have no such guidance. More research is urgently needed to explore the biological plausibility and safety of herbal medicines to manage AMR. AMR is universal, affecting anyone and everyone, at any age and in any country. Investigating how such approaches can be integrated into western medicine will be important to elucidate.