Full-text resources of PSJD and other databases are now available in the new Library of Science.
Visit https://bibliotekanauki.pl

PL EN


Preferences help
enabled [disable] Abstract
Number of results

Journal

2010 | 5 | 1 | 12-29

Article title

Surmounting antimicrobial resistance in the Millennium Superbug: Staphylococcus aureus

Content

Title variants

Languages of publication

EN

Abstracts

EN
Staphylococcus aureus is the third most dreaded pathogen posing a severe threat due to its refractory behavior against the current armamentarium of antimicrobial drugs. This is attributed to the evolution of an array of resistance mechanisms responsible for morbidity and mortality globally. Local and international travel has resulted in the movement of drug resistant S. aureus clones from hospitals into communities and further into different geographical areas where they have been responsible for epidemic outbreaks. Thus, there is a dire necessity to refrain further cross movement of these multidrug resistant clones across the globe. The plausible alternative to prevent this situation is by thorough implementation of regulatory aspects of sanitation, formulary usage and development of new therapeutic interventions. Various strategies like exploring novel antibacterial targets, high throughput screening of microbes, combinatorial and synthetic chemistry, combinatorial biosynthesis and vaccine development are being extensively sought to overcome multidrug resistant chronic Staphylococcal infections. The majority of the antibacterial drugs are of microbial origin and are prone to being resisted. Anti-staphylococcal plant natural products that may provide a new alternative to overcome the refractory S.aureus under clinical settings have grossly been unnoticed. The present communication highlights the new chemical entities and therapeutic modalities that are entering the pharmaceutical market or are in the late stages of clinical evaluation to overcome multidrug resistant Staphylococcal infections. The review also explores the possibility of immunity and enzyme-based interventions as new therapeutic modalities and highlights the regulatory concerns on the prescription, usage and formulary development in the developed and developing world to keep the new chemical entities and therapeutic modalities viable to overcome antimicrobial resistance in S. aureus.

Publisher

Journal

Year

Volume

5

Issue

1

Pages

12-29

Physical description

Dates

published
1 - 2 - 2010
online
29 - 1 - 2010

Contributors

author
  • Natural Products & Drug Discovery, Department of Biotechnology and Environmental Sciences, Thapar University, Patiala, Punjab, 147004, India
author
  • Natural Products & Drug Discovery, Department of Biotechnology and Environmental Sciences, Thapar University, Patiala, Punjab, 147004, India

References

  • [1] Peacock S.J., de Silva I., Lowy F.D., What determines the nasal carriage of Staphylococcus aureus?, Trends Microbiol., 2001, 9, 605–610 http://dx.doi.org/10.1016/S0966-842X(01)02254-5[Crossref]
  • [2] Pettit C.A., Fowler V.G., Staphylococcus aureus bacteremia and endocarditis, Cardiology Clinics, 2003, 21(2), 219–233 http://dx.doi.org/10.1016/S0733-8651(03)00030-4[Crossref]
  • [3] Fang G., Keys T.F., Gentry L.O., Harris A.A., Rivera N., Getz K. et al., Prosthetic valve endocarditis resulting from nosocomial bacteremia. A prospective, multicenter study, Ann. Intern. Med.,1993, 119(7), 560–567 [Crossref]
  • [4] Jevons, M.P., Celbenin-resistant staphylococci., Br. Med. J.,1961,1,124–125 http://dx.doi.org/10.1136/bmj.1.5219.124-a[Crossref]
  • [5] Parker M.T., Hewitt J.H., Methicillin resistance in Staphylococcus aureus, Lancet,1970,1, 800–804 http://dx.doi.org/10.1016/S0140-6736(70)92408-6[Crossref]
  • [6] Ogston A., Micrococcus poisoning., J. Anal. Physiol.,1883,17 24–58
  • [7] Skinner D., Keefer C.S., Significance of bacteremia caused by Staphylococcus aureus., Arch. Intern. Med.,1941,68, 851–875
  • [8] Archer G.L., Scott G., Conjugative transfer genes in Staphylococcal isolates from the United States, Antimicrob. Agents Chemother.,1991, 33, 2500–2504
  • [9] Trucksis M., Hooper D.C., Wolfson J.S., Emerging resistance to Fluoroquinolones in Staphylococci, Ann. Intern. Med., 1991, 114, 424–426 [Crossref]
  • [10] Lacey R.W., Mitchell A.A.B., Gentamicin-resistant Staphylococcus aureus, Lancet, 1969, II, 1425–1426 http://dx.doi.org/10.1016/S0140-6736(69)90967-2[Crossref]
  • [11] Lowy F.D., Staphylococcus aureus infections, N. Engl.J. Med.,1998, 339, 520–532 http://dx.doi.org/10.1056/NEJM199808203390806[Crossref]
  • [12] Moellering R.C. Jr., Problems with antimicrobial resistance in gram-positive cocci, Clin. Infect. Dis., 1998, 26,1177–1178 http://dx.doi.org/10.1086/520288[Crossref]
  • [13] Lelievre H., Lina G., Jones M.E., Olive C., Forey F., Roussel-Delvallez M., et al., Emergence and Spread in French Hospitals of Methicillin-Resistant Staphylococcus aureus with Increasing Susceptibility to Gentamicin and Other Antibiotics. J. Clin. Microbiol.,1999, 11, 3452–3457
  • [14] Mulligan M.E., Ruane P.J., Johnston L., Wong P., Wheelock J.P., MacDonald K.,et al. Ciprofloxacin for eradication of methicillin-resistant Staphylococcus aureus colonization. Am. J. Med.,1987,82(4A), 215–219
  • [15] Harnett N., Brown S., Krishnan C., Emergence of Quinolone Resistance among Clinical Isolates of Methicillin-Resistant Staphylococcus aureus in Ontario, Canada, Antimicrob. Agents Chemother.,1991,35(9),1911–1913
  • [16] Kuehnert M.J., Hill H.A., Kupronis B.A., Tokars J.I., Solomon S.L., Jernigan D.B., Methicillin-resistant Staphylococcus aureus-related hospitalizations, United States. Emerg.Infect. Dis.,2005,11:868–872 [Crossref]
  • [17] Hiramatsu K., Vancomycin-resistant Staphylococcus aureus: a new model of antibiotic resistance, Lancet Infect. Dis.,2001,1(3),147–155 http://dx.doi.org/10.1016/S1473-3099(01)00091-3[Crossref]
  • [18] Hiramatsu, K., Reduced susceptibility of Staphylococcus aureus to vancomycin - Japan 1996, Morb. Mortal. Wkly. Rep.,1997,27,624–626
  • [19] Centers for Disease Control and Prevention S. aureus resistant to Vancomycin in the US, Morb. Mortal. Wkly. Rep., 2002, 51, 565–567
  • [20] Centers for Disease Control and Prevention, Vancomycin resistant S. aureus Pennysylvania 2002, Morb. Mortal. Wkly. Rep.,2002, 51, 902
  • [21] Riley T.V., Pearman J.W., Rouse I.L., Changing epidemiology of methicillin-resistant Staphylococcus aureus in Western Australia, Med. J. Aust.,1995, 163,412–414
  • [22] Cookson B.D., Methicillin-resistant Staphylococcus aureus in the community: new battlefronts, or are the battles lost?, Infect. Control Hosp. Epidemiol., 2000, 21, 398–403 http://dx.doi.org/10.1086/501781[Crossref]
  • [23] Chambers H.F., Community associated Methicillin resistant Staphylococcus aureus- resistance and virulence coverage. N. Engl. J. Med., 2005, 325, 1485–1487 http://dx.doi.org/10.1056/NEJMe058023[Crossref]
  • [24] Moran G.J., Krishnadasan A., Gorwitz R.J. Fosheim G.E., McDougal L.K., Carey R.B. et al., Methicillin resistant S. aureus infections among patients in the emergency department, N. Engl. J Med., 2006, 355, 666–674 http://dx.doi.org/10.1056/NEJMoa055356[Crossref]
  • [25] Moreno F., Crisp F., Jorgenson J.H., Patterson and Patterson J.E., Methicillin-resistant Staphylococcus aureus as a community organism,Clin. Infect. Dis., 1995, 21, 1308–1312. [Crossref]
  • [26] Bukharie H.A., Abdelhadi M.S., Saeed I.A., Rubaish A.M., Larbi E.B., Emergence of methicillin- resistant Staphylococcus aureus as a community pathogen. Diagn.Microbiol.Infect.Dis., 2001, 40,1–4 http://dx.doi.org/10.1016/S0732-8893(01)00242-5[Crossref]
  • [27] Salgado C.D., Farr B.M., Calfee D.P., Community acquired methicillin-resistant Staphylococcus aureus: A meta-analysis of prevalence and risk factors, Clin.Infect. Dis., 2003, 36, 131–139 http://dx.doi.org/10.1086/345436[Crossref]
  • [28] Adcock P.M., Pator P., Medly F., Patterson J.E., Murphy T.V., Methicillin-resistant Staphylococcus aureus in two child care centers, J. Infect. Dis., 1998, 178, 577–580 [Crossref]
  • [29] Lindenmayer J.M., Schoenfeld S., O’Grady R., Carney J.K., Methicillin-resistant Staphylococcus aureus in a high school wrestling team and the surrounding community, Arch.Intern. Med.,1998,158, 895–899 http://dx.doi.org/10.1001/archinte.158.8.895[Crossref]
  • [30] Centers for Disease Control and Prevention. Outbreaks of community-associated methicillin-resistant Staphylococcus aureus skin infections-Los Angeles County, California, 2002–2003, Morb. Mortal. Wkly. Rep., 2003, 52, 88
  • [31] Cohen P.R., Kurzrock R., Community-acquired methicillin-resistant Staphylococcus aureus skin infection: an emerging clinical problem, J. Am. Acad. Dermatol., 2004, 50,277–280 http://dx.doi.org/10.1016/j.jaad.2003.06.005[Crossref]
  • [32] Olayinka B.O., Olnitola O.S., Olayinka A.T., Raji B., Antibiotic susceptibility pattern and multiple antibiotic resistance wider of S. aureus isolates in Zaria, Nigeria, J. Trop. Biosci., 2004, 451–454
  • [33] Kowalski T.J., Berbari E.F., Osmon D.R., Epidemiology, treatment and prevention of community acquired methicillin resistant Staphylococcus aureus infections, Mayo Clin. Proc., 2005, 80(9), 1201–1208 http://dx.doi.org/10.4065/80.9.1201[Crossref]
  • [34] Francis J.S., Doherty M.C., Lopatin U., Johnston C.P., Sinha G., Ross T., Severe community onset pneumonia in healthy adults caused by methicillin resistant Staphylococcus aureus carrying panton valentine leukocidin genes, Clin. Infect. Dis.,2005, 40(1),100–107 http://dx.doi.org/10.1086/427148[Crossref]
  • [35] O’Brien F.G., Pearman J.W., Gracey M., Riley T.V., Grubb W.B., Community strains of MRSA involved in hospital outbreak, J. Clin. Microbiol. 1999, 37(9), 2858–2862
  • [36] Pelag A.Y. and Munckhof W.J., Fatal necrotizing pneumonias due to community acquired Methicillin resistant Staphylococcus aureus, Med. J. Aus., 2004,181(4), 228–229
  • [37] Klevens M.R., Morrison A., Nadle J., Petit S., Gershman K., Ray S. et al., Invasive Methicillin resistant Staphylococcus aureus infections in the United States, JAMA, 2007, 298,1763–1771 http://dx.doi.org/10.1001/jama.298.15.1763[Crossref]
  • [38] Fuda C., Suvorov M., Vakulenko S.B., Mobashrey S., The Basis for Resistance to β-Lactam antibiotics by Penicillin-binding Protein2a of Methicillin-resistant Staphylococcus aureus, J. Biol. Chem., 2004, 279(39): 40802–40806 http://dx.doi.org/10.1074/jbc.M403589200[Crossref]
  • [39] Brown D.F.J., Reynolds P.E., Intrinsic resistance to beta-lactam antibiotics in Staphylococcus aureus. FEBS Lett.,1980,122,275–278 http://dx.doi.org/10.1016/0014-5793(80)80455-8[Crossref]
  • [40] Hartman B.J., Tomasz, A., Low-affinity penicillin binding protein associated with β-lactam resistance in Staphylococcus aureus. J. Bacteriol.,1984,58, 513–516
  • [41] Georgopapadakou N.H., Dix B.A., Mauriz, Y.R., Possible physiological functions of penicillin-binding proteins in Staphylococcus aureus, Antimicrob. Agents Chemother., 1986; 29, 333–336
  • [42] Utsui Y., Yokota T., Role of altered penicillin binding protein in methicillin and cepham resistant Staphylococcus aureus. Antimicrob. Agents Chemother., 1985, 28, 397–403
  • [43] Song M.D., Wachi M., Doi M., Ishino F., Matsuhashi M., Evolution of inducible penicillin resistant target protein in methicillin resistant S. aureus by gene fusion, FEBS Lett.,1987,221, 167–171 http://dx.doi.org/10.1016/0014-5793(87)80373-3[Crossref]
  • [44] Walsh T.R., Howe R.A., The prevalence and mechanisms of vancomycin resistance in Staphylococcus aureus, Ann. Rev. Microbiol., 2002, 56, 657–75 http://dx.doi.org/10.1146/annurev.micro.56.012302.160806[Crossref]
  • [45] Chopra I., Antibiotic resistance in Staphylococcus aureus: Causes, concerns and cures?, Exp. Rev. Ant. Infect. Ther., 2003, 1(1), 45–55 http://dx.doi.org/10.1586/14787210.1.1.45[Crossref]
  • [46] Weisblum B., Erythromycin resistance by ribosome modification, Antimicrob. Agents Chemother., 1995,39,577–585
  • [47] Ross J.I., Eady E.A., Cove J.H., Canliffe W.J., Cunliffe W.J., Baumberg S., Wootton J.C., Inducible erythromycin resistance in Staphylococci is encoded by a member of ATP binding transport supergene family, Mol. Microbiol.,1990, 4, 1207–1214 http://dx.doi.org/10.1111/j.1365-2958.1990.tb00696.x[Crossref]
  • [48] Saxena, S., Combating multidrug resistance microbes: A burgeoning problem In Microbes & Human Health, Vol.4, 2007; Edited by Dr. A.K. Chauhan, Dr.Harsha Kharkwal & Dr. Ajit Varma,pp.589–607,I.K.International Publishing House, New Delhi,India ISBN81-89866-05-02
  • [49] Bismuth R., Zilhao R., Sakamoto H. Guesdon J.L., Courvalin P., Gene heterogeneity for tetracycline resistance in Staphylococcus spp. Antimicrob. Agents Chemother., 1990,34,1611–1614
  • [50] Warsa U.C., Nonoyama M., Ida T., Okamoto R., Okubo T., and Shimauchi C., et.al., Detection of tet (K) and tet(M) in Staphylococcus aureus of Asian countries by Polymerase chain reaction, J. Antibiot., 1996, 49(11),1127–1132 [Crossref]
  • [51] Trzcinski K., Cooper B.S., Hryniewicz W., Dawson C.G., Expression of resistance to tetracyclines in strains of methicillin- resistant Staphylococcus aureus, J. Antimicrob. Chemother., 2000, 45, 763–770 http://dx.doi.org/10.1093/jac/45.6.763[Crossref]
  • [52] Schimtz, F.J., Angela K., Sadurski, R., Milatovic D, Fluit A.C., et al. Resistance to tetracycline and distribution of tetracycline resistance genes in European Staphylococcus aureus isolates. J. Antimicrob. Chemother., 2001, 47, 239–240 http://dx.doi.org/10.1093/jac/47.2.239[Crossref]
  • [53] Shinabarger D.L., Marotti K.R., Murray R.W., Lin A.H., Melchior E.P., Swaney S.M. et al. Mechanisms of actions of oxazlidinones: effects of linezolid and eperezolid on translation reactions. Antimicrob. Agents Chemother., 1997, 41, 2132–2136
  • [54] Tsiodras S., Gold H.S., Sakouloulas G., Eliopoulos P.M., Wennersten C., Venkataraman L., et al., Linezolid resistance in a clinical isolate of Staphylococcus aureus, Lancet, 2001, 358, 207–208 http://dx.doi.org/10.1016/S0140-6736(01)05410-1[Crossref]
  • [55] Kloss P., Xiong L., Shinabarger D.L., Mankin, A.S. Resistance mutations in 23 S rRNA identify the site of action of the protein synthesis inhibitor linezolid in the ribosomal peptidyl transferase center, J. Mol. Biol.,1999, 294(1), 93–101 http://dx.doi.org/10.1006/jmbi.1999.3247[Crossref]
  • [56] Vanuffel P., Giambattista Di M., Cocito, C., Chemical probing of virginiamycin M -promoted conformational change of the peptidyltransferase domain, Nuc. Acids Res.,1994, 22, 4449–4453 http://dx.doi.org/10.1093/nar/22.21.4449[Crossref]
  • [57] Mitchell B.A., Brown M.H., Skurray, R.A., Qac A efflux pumps from Staphylococcus aureus: Comparative analysis of resistance to diamidines, biguanidines and guanylhydrazones, Antimicrob. Agents Chemother., 1998, 42, 475–471 http://dx.doi.org/10.1093/jac/42.4.475[Crossref]
  • [58] Walmsley M.B., Mckeegan K.S., Walmsley, A.R., Structure and function of efflux pumps that confer resistance to drugs, Biochem. J.,2003, 376,313–338 http://dx.doi.org/10.1042/BJ20020957[Crossref]
  • [59] Allignet J., Solh N.El., Characterization of a new staphylococcal gene, vga B, encoding a putative ABC transfer conferring resistance to streptogramin A and related compounds,Gene,1997, 202, 133–138 http://dx.doi.org/10.1016/S0378-1119(97)00464-2[Crossref]
  • [60] Prunier A.L., Malbruny B., Laurans M., Brouard J., Duhamel J.F. and Leclercq R., High rate of macrolide resistance in Staphylococcus aureus strains from patients with cystic fibrosis reveals high proportions of hypermutable strains, J. Infect. Dis., 2003, 187, 1709–1716 http://dx.doi.org/10.1086/374937[Crossref]
  • [61] Trucksis M., Wolfson J.S., Hooper D.C., A novel locus conferring fluoroquinolone resistance in Staphylococcus aureus, J Bact., 1991, 173(18), 5854–5860
  • [62] Ng, E.Y., Trucksis, M., Hooper, D.C., Quinolone resistance mutations in topoisomerase IV: relationship to the flqA locus and genetic evidence that topoisomerase IV is the primary target and DNA gyrase is the secondary target of fluoroquinolones in Staphylococcus aureus, Antimicrob. Agents Chemother.,1996, 40, 1881–1888
  • [63] Allignet J., Loncle V., Simenel C., Delepierre M., El Solh N., Sequence of a staphylococcal gene, vat, encoding an acetyltransferase inactivating the A-type compounds of virginiamycin-like antibiotics, Gene,1993,130, 91–98 http://dx.doi.org/10.1016/0378-1119(93)90350-C[Crossref]
  • [64] McGowan, J.E. Jr, Gerding D.N., Does antibiotic restriction prevent resistance?, New Horiz.,1996,4: 370–376
  • [65] Niederman M.S., Is “Crop rotation” of antibiotic the solution to a “resistant” problem to ICU?, Am. J. Resp. Crit. Care Med., 1997; 156: 1029–1031 [Crossref]
  • [66] Jarvis, W.R., Handwashing- the Semmelweis lesson forgotten?, Lancet, 1994, 344(8933): 1311–1312 http://dx.doi.org/10.1016/S0140-6736(94)90687-4[Crossref]
  • [67] Casewell, M.W., Hill R.L., Minimal dose requirements for nasal mupirocin and its role in the control of epidemic MRSA., J. Hosp. Infect., 1991;19: 35–40 http://dx.doi.org/10.1016/0195-6701(91)90201-I[Crossref]
  • [68] Jones M.E., In vitro profile of a new β-lactam, Ceftobiprole with activity against MRSA., Clin. Microbiol. Infect., 2007, 13(2):17–24 http://dx.doi.org/10.1111/j.1469-0691.2007.01722.x[Crossref]
  • [69] Koga T., Abe T., Harumi I., Takenouchi T., Kitayama A., Yoshida T., et al. In vitro and in vivo antibacterial activities of CS-023 (RO4908463), a novel parenteral carbapenem. Antimicrob. Agents Chemother., 2005, 49(8), 3239–3250 http://dx.doi.org/10.1128/AAC.49.8.3239-3250.2005[Crossref]
  • [70] Sum P.E., Lee V.J., Testa R.T., Hlavka J.J., Ellestad G.A., et al., Glycylcyclines - A new generation of potent antibacterial agents through modification of 9-aminotetracyclines, J. Med. Chem., 1994, 37, 184–188 http://dx.doi.org/10.1021/jm00027a023[Crossref]
  • [71] Goldstein F.W., Kitzis M.D., Acar, J.F.N., N-Dimethylglycylamido derivatives of minocycline and 6-demethyl-6-desoxytetracycline. Two new glycylcyclines highly effective against tetracyclineresistant gram-positive cocci., Antimicrob. Agents Chemother., 1994, 38, 2218–2220
  • [72] Livermore D.M., Tigecycline: what is it, and where should it be used?, J.Antimicrob. Chemother., 2005,56(4), 611–14 http://dx.doi.org/10.1093/jac/dki291[Crossref]
  • [73] Bergeron J., Ammirati M., Danley D., Glycylcyclines bind to the high-affinity tetracycline ribosomal binding site and evade Tet (M),-and Tet(O)-mediated ribosomal protection, Antimicrob. Agents Chemother., 1996, 40, 2226–2228
  • [74] Harris, R. and Cruz, M., Tigecycline (Tygacil): A Novel First-in-Class, Broad-Spectrum Intravenous Antibiotic For the Treatment of Serious Bacterial Infections, Pharmacy & Therapeutics, 31(1), 18–27 and 57
  • [75] McKenney D., Quinn J.M., Jackson C.L., Guilmet J.L., Landry J.A., Tanaka S.K., et al. Evaluation of PTK 0796 in experimental model of infections caused by gram positive and gram-negative pathogen. Abstr. InterSc. Conf. Antimicrob. Agents Chemother., 2003, Sep 14–17, abstract no. F-757
  • [76] Shah, P.M., The need for new therapeutic agents: what is in the pipeline?,Clinical Microbiol.Infect., 11, 36–42
  • [77] Zeckel M.L., Preston D.A., Allen B.S., In vitro activities of AntiLY333328 and comparative agents against nosocomial gram positive pathogens collected in a 1997 global surveillance study, Antimicrob. Agents Chemother., 2000, 44(5), 1370–1368 http://dx.doi.org/10.1128/AAC.44.5.1370-1374.2000[Crossref]
  • [78] Barrett J.F., Oritavanacin: Eli Lilly & Co., Curr. Opinion Investig. Drugs, 2001, 2(8), 1039–1044
  • [79] Jabés D., Candiani G., Romanó G., Brunati C., Riva S., and Cavaleri, M., Efficacy of Dalbavancin against Methicillin-Resistant Staphylococcus aureus in the Rat Granuloma Pouch Infection Model. Antimicrob. Agents Chemother., 48(4), 1118–1123
  • [80] Lin G., Credito K., Ednie L.M. and Appelbaum, P.C., Antistaphylococcal activity of Dalbvancin, an experimental glycopeptide, Antimicrob. Agents Chemother., 2005,49(2), 770–772 http://dx.doi.org/10.1128/AAC.49.2.770-772.2005[Crossref]
  • [81] O’Hare M.D., Ghosh G., Felmingham D. and Grüeberg R.N., In vitro studies with ramoplanin (MDL 62,198): a novel lipoglycopeptide antimicrobial, J. Antimicrob.Chemother.,1990,25,217–220 http://dx.doi.org/10.1093/jac/25.2.217[Crossref]
  • [82] Montecalvo M.A., Ramoplanin: a novel antimicrobial agent with the potential to prevent vancomycinresistant enterococcal infection in high-risk patients, J. Antimicrob.Chemother., 2003, 51(Suppl. S3), iii31–iii35
  • [83] Takahata M., Mitsuyama J., Yamashiro Y., Yonezawa M., Araki H., Todo Y., et al., In vitro and in vivo antimicrobial activities of T-3811ME, a novel des-F(6)-quinolone, Antimicrob. Agents Chemother., 1999, 43:1077–84
  • [84] Noviello S., Ianniello F., Leone S. and Esposito S., Comparative activity of garenoxacin and other agents by susceptibility and time-kill testing against Staphylococcus aureus, Streptococcus pyogenes and respiratory pathogens, J. Antimicrob. Chemother., 2003, 52,869–872 http://dx.doi.org/10.1093/jac/dkg429[Crossref]
  • [85] Schmitz FJ, Fluit AC, Milatovic D, Verhoef J., Heinz H.P. and Brisse S., In vitro potency of moxifloxacin, clinafloxacin and sitafloxacin against 248 genetically defined clinical isolates of S.aureus, J. Antimicrob. Chemother., 2006, 46, 109–113 http://dx.doi.org/10.1093/jac/46.1.109[Crossref]
  • [86] Bhagwat SS, Mundkar LA, Gupte SV, Patel M.V., and Khorakiwala H.F., The anti- MRSA quinolone WCK771 has potent activity against sequentially labeled mutants and has a narrow mutant selection windows against quinolone resistant S. aureus and preferentially targets DNA gyrase. Antimicrob. Agents Chemother., 2006, 50(11), 3568–3579 http://dx.doi.org/10.1128/AAC.00641-06
  • [87] Das B., Rudra S., Yadav A., Ray A., Raja Rao A.V.S., Srinivas A.S.S.V., et al., Synthesis and SAR of novel oxazolidinones: discovery of ranbezolid, Bioorg.Med.Chem.Lett., 2005, 15(19), 4261–4267 http://dx.doi.org/10.1016/j.bmcl.2005.06.063[Crossref]
  • [88] Mathur T., Bhateja P., Pandya M., Fatma T., Rattan A., In vitro activity of RBx 7644(ranbezolid) on biofilm producing bacteria, Int.J. Antimicrob. Agents, 2004, 24(4),369–373 http://dx.doi.org/10.1016/j.ijantimicag.2004.04.012[Crossref]
  • [89] Rattan A., RBx-7644: Oxazolidinone antibacterial, Drugs of the future, 2003, 28(11),1070–1077 http://dx.doi.org/10.1358/dof.2003.028.11.769931[Crossref]
  • [90] Bush K., Macielag M. and Weidner-Wells, M., Taking inventory: antibacterial agents currently at or beyond Phase 1, Curr. Opin. Microbiol., 2004, 7(5), 466–476 http://dx.doi.org/10.1016/j.mib.2004.08.013[Crossref]
  • [91] Gill C.J., Abruzzo G.K., Flattery A.M., Misura A.S., Bartizal K., Hickey E.J., In Vivo Efficacy of a Novel Oxazolidinone Compound in Two Mouse Models of Infection, Antimicrob. Agents Chemother., 2007, 51(9), 3434–3436 http://dx.doi.org/10.1128/AAC.01567-06[Crossref]
  • [92] Yuan Z., Trias J., White R.J., Deformylase as a novel antibacterial target, Drug Discovery Today, 2001,6(18), 954–961 http://dx.doi.org/10.1016/S1359-6446(01)01925-0[Crossref]
  • [93] Credito K, Lin G, Ednie LM and Appelbaum P.C., Antistaphylococcal activity of LBM415, a new peptide deformylase inhibitor, compared with those of other agents, Antimicrob. Agents Chemother. 2004, 48, 4033–4036 http://dx.doi.org/10.1128/AAC.48.10.4033-4036.2004[Crossref]
  • [94] Hoang T.T., Schweizer H.P., Fatty acid biosynthesis in Pseudomonas aeruginosa: cloning and characterization of the fabAB operon encoding β-hydroxydecanoyl-acyl carrier protein dehydratase (FabA) and β-ketoacyl-acyl carrier protein synthase I (FabB). J Bacteriol., 1997,179, 5326–5332
  • [95] Heath R.J., Rubin J.R., Holland D.R., Zhang E., Snow M.E., and Rock C.O., Mechanism of Triclosan Inhibition of Bacterial Fatty Acid Synthesis, J. Biol. Chem. 1999, 274(16), 11110–11114 http://dx.doi.org/10.1074/jbc.274.16.11110[Crossref]
  • [96] Payne D.J., Warren P.V., Holmes D.J., Ji Y., Lonsdale J.T., Bacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discovery, Drug Discovery Today, 2001, 6(10), 537–4444 http://dx.doi.org/10.1016/S1359-6446(01)01774-3[Crossref]
  • [97] Oh K.B., Oh M.N., Kim J.G., Shin D.S., Shin J, Inhibition of sortase-mediated Staphylococcus aureus adhesion to fibronectin via fibronectin-binding protein by sortase inhibitors, Appl. Microbiol. Biotechnol., 2005, 70(1),102–106 http://dx.doi.org/10.1007/s00253-005-0040-8[Crossref]
  • [98] Kim S.H., Shin D.S., Oh M.N., Chung S.C., Lee J.S., Chang I.M., Oh K.B., Inhibition of Sortase, a Bacterial Surface Protein Anchoring Transpeptidase, by β-Sitosterol-3-O-glucopyranoside from Fritillaria verticillata, BioSci. Biotech. Biochem., 2003, 67(11): 2477–2479 http://dx.doi.org/10.1271/bbb.67.2477[Crossref]
  • [99] Riedlinger J., Reicke A., Zähner H., Krismer B., Bull A.T., Maldonado L.A., Ward A.C., Goodfellow M., Bister B., Bischoff D., Süssmuth R.D., Fiedler H.P., “Abyssomicins, inhibitors of the paraaminobenzoic acid pathway produced by the marine Verrucosispora strain AB-18-032” J. Antibiot., 2004, 57, 271–279 [Crossref]
  • [100] Allsop A.E., New Antibiotic discovery, novel screens, novel targets and impact of genomics, Curr. Opin. Microbiol.,1998, 1(5),530–534 http://dx.doi.org/10.1016/S1369-5274(98)80085-4[Crossref]
  • [101] Dunman PM, Murphy E, Haney S, Kellogg G.T., Wu S., et al., Transcription Profiling-Based Identification of Staphylococcus aureus Genes Regulated by the agr and/or sarA Loci, J. Bacteriol. 2001, 183, 7341–7353 http://dx.doi.org/10.1128/JB.183.24.7341-7353.2001[Crossref]
  • [102] Liu J., Dehbi M, Moeck G, Arhin F., Bauda P., Bergeron D., et al., Antimicrobial drug discovery through bacteriophage genomics, Nature Biotech., 2004, 22(2), 185–191 http://dx.doi.org/10.1038/nbt932[Crossref]
  • [103] Moreillon P.,The efficacy of amoxicillin /clavulanate (Augmentin) in treatment of severe staphylococcal infections, J Chemother.1994, 6(2). 51–57
  • [104] Prieto J, Aguilar L, Gimenez MJ, Toro D., Gómez-Lus M. L., Dal-Ré R., et al. In vitro Activities of co-amoxiclav at concentrations achieved in human serum against the resistant subpopulation of heteroresistant Staphylococcus aureus: a Controlled Study with vancomycin, Antimicrob. Agents Chemother.,1998,42(7),1574–1577
  • [105] Edouard R.S., Pestel-Caron M., Lemeland J.F., Caron F., In vitro synergistic effects of double and triple combinations of β-Lactams, vancomycin, and netilmicin against methicillin-resistant Staphylococcus aureus strains, Antimicrob. Agents Chemother., 2004, 44(11), 3055–3060 http://dx.doi.org/10.1128/AAC.44.11.3055-3060.2000[Crossref]
  • [106] Shelburne S.A., Musher D.M., Hulten K., Ceasar H., Lu M.Y., Bhaila I., et al. In vitro killing of community-associated methicillin-resistant Staphylococcus aureus with drug combinations, Antimicrob. Agents Chemother., 2004, 48, 4016–4019 http://dx.doi.org/10.1128/AAC.48.10.4016-4019.2004[Crossref]
  • [107] Rand K.H. and Houck H., Synergy of daptomycin with oxacillin and other β-Lactams against methicillin resistant Staphylococcus aureus, Antimicrob. Agents Chemother., 2004,48(8),2871–2875 http://dx.doi.org/10.1128/AAC.48.8.2871-2875.2004[Crossref]
  • [108] Kono K., Tatara I., Takeda S., Arakawa K., Shirotani T., Okada M. et al. Antibacterial activity of epigallocatechin gallate methicillin resistant Staphylococcus aureus. Journal of Japan Association of Infectious Diseases,1994,68,1518–1522
  • [109] Shiota S., Shimizu M., Mizushima T., Ito H., Hatano T., Yoshida T., Tsuchiya T., Marked reduction in the minimum inhibitory concentration (MIC) of betalactams in methicillin-resistant Staphylococcus aureus produced by epicatechin gallate, an ingredient of green tea (Camellia sinensis)., Biol. Pharm. Bull.,1999, 22(12), 1388–1390 [Crossref]
  • [110] Takahashi O., Cai Z., Toda M., Hara Y., Shimamura T. et al., Appearance of antibacterial activity of oxacillin againt methicillin resistant Staphylococcus aureus (MRSA) in the presence of catechin. Journal of Japan Association of Infectious Diseases, 1995,69: 1126–1134
  • [111] Hamilton-Miller J.M, Shah S., Activity of tea component epicatechin gallate and analogues against methicillin resistant Staphylococcus aureus, J Antimicrob. Chemother., 2000, 46, 852–853 http://dx.doi.org/10.1093/jac/46.5.852[Crossref]
  • [112] Zhao W.H., Hu Z., Okubo S., Hara Y. and Shimamura T., Mechanism of synergy between epicatechin gallate and β-Lactams against methicillin resistant Staphylococcus aureus, Antimicrob. Agents Chemother., 2001, 45(6), 1737–1742 http://dx.doi.org/10.1128/AAC.45.6.1737-1742.2001[Crossref]
  • [113] Nicolson K., Evan G. and O’Toole P.W., Potentiation of methicillin activity against methicillin-resistant Staphylococcus aureus by diterpenes, FEMS Microbiol. Lett.,1999, 179(2), 233–239 http://dx.doi.org/10.1111/j.1574-6968.1999.tb08733.x[Crossref]
  • [114] Smith E.C.J., Kaatz G.W., Seo S.M., Wareham N., Williamson E.M. et al., The phenolic diterpene totarol inhibits multidrug efflux pump activity in S. aureus. Antimicrob. Agents Chemother.,2007, 51(12): 4480–4483 http://dx.doi.org/10.1128/AAC.00216-07[Crossref]
  • [115] Schmitz F.J., Fluit A.C., Luckefahr M., Engler B., Hofmann B., Verhoef J. et al. The effect of reserpine, an inhibitor of multidrug efflux pumps, on the in vitro activities of ciprofloxacin, sparfloxacin and moxifloxacin against clinical isolates of Staphylococcus aureus, J. Antimicrob. Chemother.,1998, 42, 807–810 http://dx.doi.org/10.1093/jac/42.6.807[Crossref]
  • [116] Gibbons S. and Udo E.E., The effect of reserpine, a modulator of multidrug efflux pumps on the in vitro activity of tetracycline against clinical isolates of methicillin resistant Staphylococcus aureus possessing tet(k) determinant, Phytother. Res., 2000, 74,139–140 http://dx.doi.org/10.1002/(SICI)1099-1573(200003)14:2<139::AID-PTR608>3.0.CO;2-8[Crossref]
  • [117] Stermitz F.R., Lorenz P., Tawara J.N., Zenewicz L.A., Lewis K., Synergy in a medicinal plant: antimicrobial action of berberine potentiated by 5-methoxyhydnocarpin, a multidrug pump inhibitor, Proc. Natl. Acad. Sci. USA, 2000,97,1433–1437 http://dx.doi.org/10.1073/pnas.030540597[Crossref]
  • [118] Stermitz F.R., Matsuda J.T., Lorenz P., Mueller P., Zenewicz L., Lewis K., et al.,5-Methoxy-hydnocarpin and pheophorbide A: Berberis species components which potentiate berberine growth inhibition of resistant Staphylococcus aureus. J. Nat. Prod., 2000, 63, 1146–1149 http://dx.doi.org/10.1021/np990639k[Crossref]
  • [119] Oluwatuyi M., Kaatz G.W. and Gibbons S., Antibacterial and resistance modifying activity of Rosmarinus officinalis, Phytochem., 2004, 65(2) 3249–3254 http://dx.doi.org/10.1016/j.phytochem.2004.10.009[Crossref]
  • [120] Fujita M., Shiota S., Kuroda T., Tsutomu H., Takashi Y., Tohru M., et al., Remarkable synergies between baicalein and tetracycline and baicalein and β-Lactams against methicillin resistant Staphylococcus aureus, Microbiol. Immunol., 2005, 49, 391–396 [Crossref]
  • [121] Khan I.A., Mirza Z.M., Kumar A., Verma V., Qazi G.N., Piperine, a phytochemical potentiator of ciprofloxacin against Staphylococcus aureus. Antimicrob. Agents Chemother., 2006, 50(2), 810–812 http://dx.doi.org/10.1128/AAC.50.2.810-812.2006[Crossref]
  • [122] Smith P., Stewart J., Fyfe L., Influence of subinhibitory concentrations of plant essential oils on the production of enterotoxins A and B and α-toxin by Staphylococcus aureus, J. Med. Microbiol., 2004, 53, 1023–1027 http://dx.doi.org/10.1099/jmm.0.45567-0[Crossref]
  • [123] Dickson R.A., Houghton P.J., Hylands P.J., Gibbons S.,; Antimicrobial, resistance-modifying effects, antioxidant and free radical scavenging activities of Mezoneuron benthamianum Baill., Securinega virosa Roxb. & Wlld. and Microglossa pyrifolia Lam., Phytother. Res., 2006, 20, 41–45 http://dx.doi.org/10.1002/ptr.1799[Crossref]
  • [124] Braga C., Leite A.A.M., Xavier K.G.S., Takahashi, J A., Bemquerer, M P., Chartone-Souza E., et al. Synergic interaction between pomegranate extract and antibiotics against Staphylococcus aureus, Can. J Microbiol., 2005, 51(7), 541–547 http://dx.doi.org/10.1139/w05-022[Crossref]
  • [125] Okusa P.N., Penge O., Devleeschouwer M., Duez P., Direct and indirect antimicrobial effects and antioxidant activity of Cordia gilletii De Wild (Boraginaceae), J. Ethnopharmacol., 2007, 112(3), 476–481 http://dx.doi.org/10.1016/j.jep.2007.04.003[Crossref]
  • [126] Wright G.D., Resisting resistance: New chemical strategies for battling superbugs,Chem. Biol., 2000,7, R127–32 http://dx.doi.org/10.1016/S1074-5521(00)00126-5[Crossref]
  • [127] Kristiansen J.E., The antimicrobial activity of psychotherapeutic drugs and stereo-isomeric analogues, Dan. Med. Bull.,1990,37,165–182
  • [128] Kristiansen J.E., Chlorpromazine: non-antibiotics with antimicrobial activity-new insights in managing resistance?, Curr.Opin.Investig. Drugs,1993, 2, 587–591
  • [129] Kristiansen J.E., Amaral L., The potential management of resistant infections with nonantibiotics, J. Antimicrob. Chemother., 1997, 40, 319–327 http://dx.doi.org/10.1093/jac/40.3.319[Crossref]
  • [130] Kaatz G.W., Moudgal V.V., Seo S.M, Kristiansen J.E., Phenothiazines and thioxanthenes inhibit multidrug efflux pump activity in Staphylococcus aureus, Antimicrob. Agents Chemother., 2003, 47, 719–726 http://dx.doi.org/10.1128/AAC.47.2.719-726.2003[Crossref]
  • [131] LAM K.S.,Discovery of novel metabolites from marine actinomycetes, Curr. Opin. Microbiol. 2006, 9, 245–251 [Crossref]
  • [132] Gomber C., Saxena S.. Anti-staphylococcal potential of Callistemon rigidus, Central European Journal of Medicine, 2007, 2(1),79–88 http://dx.doi.org/10.2478/s11536-007-0004-8[Crossref]
  • [133] Katarere D.R., Eloff J.N., Antibacterial and Antioxidant activity of Sutherlandia frutescens (Fabaceae) a reputed anti HIV/AIDS phytomedicine, Phytother. Res., 2005, 19(9), 779–781 http://dx.doi.org/10.1002/ptr.1719[Crossref]
  • [134] Akinyemi K.O., Oladapo O., Okwara C.E., Ibe C.C. and Fasure K.A., Screening of crude extracts of six medicinal plants used in South-West Nigerian unorthodox medicine for anti-methicillin resistant Staphylococcus aureus activity, BMC Complementary and Alternative Medicine,2005, 5, 6 (doi:10.1186/1472-6882-5-6) http://dx.doi.org/10.1186/1472-6882-5-6[Crossref]
  • [135] Nitta T., Arai T., Takamatsu H., Inatomi Y., Murata H., Iinuma M., Tanaka T., Ito T., Asai F., Ibrahim I., Nakanishi T. and Watabe K., Antibacterial Activity of Extracts Prepared from Tropical and Subtropical Plants on Methicillin-Resistant Staphylococcus aureus, Jour. Health Sciences,. 2002, 4, 273–276 http://dx.doi.org/10.1248/jhs.48.273[Crossref]
  • [136] Gibbons S., Anti-Staphylococcal plant natural products, Nat. Prod. Rep., 2004, 21, 263–277 http://dx.doi.org/10.1039/b212695h[Crossref]
  • [137] Schempp C.M., Pelz K., Wittmer A., Schöpf E., Simon J.C., Antibacterial activity of hyperforin from St John’s wort, against multiresistant Staphylococcus aureus and gram-positive bacteria, Lancet, 1999, 353(9170), 2129 http://dx.doi.org/10.1016/S0140-6736(99)00214-7[Crossref]
  • [138] Iinuma M., Tosa H., Tanaka T., Asai F., Kobayashi Y., Shimano R., Miyauchi K., Antibacterial activity of Xanthones from guttiferous plants against Methicillin resistant Staphylococcus aureus, J. Pharm. Pharmacol.,1996,48(8), 861–65 [Crossref]
  • [139] Keller M. and Zengler K. Tapping into microbial diversity, Nature Rev. Microb. 2004, 2(2),141–150. http://dx.doi.org/10.1038/nrmicro819[Crossref]
  • [140] Demain A.L., Microbial natural products: Alive and well in 1998, Nature Biotechnol., 1998, 16, 3–4 http://dx.doi.org/10.1038/nbt0198-3[Crossref]
  • [141] Demain A.L., Pharmacologically active secondary metabolites of microorganisms, Appl. Microbiol. Biotechnol., 1999, 52, 455–463 http://dx.doi.org/10.1007/s002530051546[Crossref]
  • [142] Tulp M. and Bohlin L. Functional versus chemical diversity: is biodiversity important for drug discovery?, Trends Pharmacol. Sci., 2002, 23, 225–231 http://dx.doi.org/10.1016/S0165-6147(02)02007-2[Crossref]
  • [143] Wang J., Soisson S.M., Young K., Shoop W., Kodali S., Galgoci A., Painter R.,et al., Platensimycin is a selective Fab F inhibitor with potent antibiotic properties, Nature, 2006, 441: 358–361 http://dx.doi.org/10.1038/nature04784[Crossref]
  • [144] Bull A.T., Stach J.E., Ward A.C., Goodfellow M., Marine actinobacteria: Perspectives, challenges, future directions, Antonie Van Leeuwenhoek, 2005, 87, 65–79 http://dx.doi.org/10.1007/s10482-004-6562-8[Crossref]
  • [145] Strobel G. and Daisy B., Bioprospecting for microbial endophytes and their natural products, Microb. Mol.Biol. Rev. 2003, 67(4), 491–502 http://dx.doi.org/10.1128/MMBR.67.4.491-502.2003[Crossref]
  • [146] Castillo U., Strobel G.A., Ford E.J., Hess W.M., Porter H., Jensen J.B., et.al., Munumbicins, widespectrum antibiotics produced by Streptomyces NRRL 30562, endophytic on Kennedia nigricans, Microbiol., 2002, 148, 2675–2685
  • [147] Lee L.Y, Miyamoto Y.J., McIntyre B.W., Hook M., McCrea K.W., McDevitt D., Brown E.L., The Staphylococcus Map Protein is an immunomodulator that interferes with T-cell mediated responses, J. Clin. Invest., 2002, 110, 1461–1471 [Crossref]
  • [148] Shinefield H., Black S., Fattom A., Horwith G., Rasgon S., Ordonez J.,et al., Use of a Staphylococcus aureus conjugate vaccine in patients receiving hemodialysis, N. Engl. J. Med., 2002, 346, 491–496 http://dx.doi.org/10.1056/NEJMoa011297[Crossref]
  • [149] Burnie J.P., Matthews R.C., Carter T., Beaulieu E., Donohoe M., Chapman C., Williamson P. and Hodgetts S.J., Identification of an Immunodominant ABC Transporter in Methicillin-Resistant Staphylococcus aureus Infections, Infect. Immun., 2000, 68(6), 3200–3209 http://dx.doi.org/10.1128/IAI.68.6.3200-3209.2000[Crossref]
  • [150] Schuhardt V.T., Schindler C.A., Lysostaphin therapy in mice infected with Staphylococcus aureus., J Bacteriol., 1964,88, 815–816
  • [151] Kusuma C.M., Kokai-Kun J.F., Comparison of Four Methods for Determining Lysostaphin Susceptibility of Various Strains of Staphylococcus aureus, Antimicrob. Agents Chemother., 2005, 49, 3256–3263 http://dx.doi.org/10.1128/AAC.49.8.3256-3263.2005[Crossref]
  • [152] Kokai-Kun J.F., Walsh S.M., Chanturiya T., Mond J.J., Lysostaphin cream eradicates Staphylococcus aureus nasal colonization in a cotton rat model. Antimicrob. Agents Chemother., 2003, 47(5),1589–1597 http://dx.doi.org/10.1128/AAC.47.5.1589-1597.2003[Crossref]
  • [153] Yang X., Cong-Ran L., Ren-Hui L., Wang Y.M., Zhang W.X., Chen H.Z., et al., In vitro activity of recombinant lysostaphin against Staphylococcus aureus isolates from hospitals in Beijing, China Journal of Medical Microbiology, 2007, 56, 71–76 http://dx.doi.org/10.1099/jmm.0.46788-0[Crossref]
  • [154] Kiri N., Gordon A., Climo M.W., Combinations of Lysostaphin with β-Lactams are synergistic against oxacillin-resistant Staphylococcus epidermidis,Antimicrob.Agents Chemother., 2002, 6(6), 2017–2020 http://dx.doi.org/10.1128/AAC.46.6.2017-2020.2002[Crossref]
  • [155] Vavra S.B., Roberta B.C., Robert S.D., Development of vancomycin and lysostaphin resistance in a methicillin-resistant Staphylococcus aureus isolate, J. Antimicrob. Chemother., 2001,48, 617–625 http://dx.doi.org/10.1093/jac/48.5.617[Crossref]
  • [156] Stranden A., Ehlert K., Labischinski H., Berger-Bachi B., Cell wall monoglycine cross-bridges and methicillin hypersusceptibility in a fem AB null mutant of methicillin-resistant Staphylococcus aureus,J. Bacteriol.,1997,179, 9–16
  • [157] Ling B. and Berger-Bachi, B., Increased overall antibiotic susceptibility in Staphylococcus aureus femAB null mutants. Antimicrob. Agents Chemother., 1998, 42,936–938
  • [158] Schneider M.M.S., Berger-Bächi B., Tossi A., Sahl A.G., Wiedemann I., In vitro assembly of a complete, pentaglycine interpeptide bridge containing cell wall precursor (lipid II-Gly5) of Staphylococcus aureus, Mol. Microbiol., 2004, 53(2), 675–685 http://dx.doi.org/10.1111/j.1365-2958.2004.04149.x
  • [159] Hutchinson C.R., Combinatorial biosynthesis for new drug discovery, Curr.Opin Microb.,1998, 1, 319–329 http://dx.doi.org/10.1016/S1369-5274(98)80036-2[Crossref]
  • [160] Borchardt J.K., Genetic engineering may keep one of the richest drug gold mines from being played out, Modern Drug Discovery,1999, 2(4), 22–29
  • [161] Jacobsen J.R., Khosla C.,New directions in metabolic engineering.,Curr.Opin. Chem. Biol.,1998, 2,133–137 http://dx.doi.org/10.1016/S1367-5931(98)80045-8[Crossref]
  • [162] Saxena, S. and Kumar D., Human pathogenic bacteria-plant interaction: Potential as novel antimicrobials, International Journal of Biomedical and Pharmaceutical Sciences, 2007,1(2), 120–123
  • [163] Baltz R.H., Vivian M. Stephen K.W., Natural products to drugs: daptomycin and related lipopeptide antibiotics, Nat. Prod. Rep., 2005, 22, 717–717 http://dx.doi.org/10.1039/b416648p[Crossref]
  • [164] Yin X, Zabriskie TM., The enduracidin biosynthetic gene cluster from Streptomyces fungicidicus, Microbiol., 2006, 152, 2969–2983 http://dx.doi.org/10.1099/mic.0.29043-0[Crossref]

Document Type

Publication order reference

Identifiers

YADDA identifier

bwmeta1.element.-psjd-doi-10_2478_s11536-009-0079-5
JavaScript is turned off in your web browser. Turn it on to take full advantage of this site, then refresh the page.