메뉴 건너뛰기




Volumn 31, Issue 2, 2018, Pages

Antibiotic hybrids: The next generation of agents and adjuvants against gram-negative pathogens?

Author keywords

Antibacterial; Antibiotic; Antimicrobial; Efflux; Hybrid; Permeability

Indexed keywords

ADJUVANT; ANTIBIOTIC AGENT; ANTIINFECTIVE AGENT; ASPERGILLOMARASMINE A; AVIBACTAM PLUS CEFTAZIDIME; BETA LACTAMASE; BETA LACTAMASE INHIBITOR; CEFIDEROCOL; CEFTAROLINE FOSAMIL; CEFTOLOZANE PLUS TAZOBACTAM; CILASTATIN; CIPROFLOXACIN; DALBAVANCIN; DELAFLOXACIN; FIDAXOMICIN; FINAFLOXACIN; FRAMYCETIN; IMIPENEM; MEROPENEM PLUS VABORBACTAM; MINOCYCLINE; MOXIFLOXACIN; ORITAVANCIN; POLYAMINOACID; RELEBACTAM; RIFAMPICIN; SECNIDAZOLE; TEDIZOLID; TOBRAMYCIN; UNCLASSIFIED DRUG;

EID: 85044227303     PISSN: 08938512     EISSN: 10986618     Source Type: Journal    
DOI: 10.1128/CMR.00077-17     Document Type: Review
Times cited : (218)

References (330)
  • 1
    • 0038665502 scopus 로고    scopus 로고
    • Antimicrobial resistance: the example of Staphylococcus aureus
    • Lowy, F. 2003. Antimicrobial resistance: the example of Staphylococcus aureus. J Clin Invest 111:1265–1273. https://doi.org/10.1172/JCI18535.
    • (2003) J Clin Invest , vol.111 , pp. 1265-1273
    • Lowy, F.1
  • 2
    • 33646818699 scopus 로고    scopus 로고
    • Antibiotic resistance in bacteria and its future for novel antibiotic development
    • Yoneyama, H, Katsumata R. 2006. Antibiotic resistance in bacteria and its future for novel antibiotic development. Biosci Biotechnol Biochem 70:1060–1075. https://doi.org/10.1271/bbb.70.1060.
    • (2006) Biosci Biotechnol Biochem , vol.70 , pp. 1060-1075
    • Yoneyama, H.1    Katsumata, R.2
  • 4
    • 42549171131 scopus 로고    scopus 로고
    • Federal funding for the study of antimicrobial resistance in nosocomial pathogens: No ESKAPE
    • Rice, LB. 2008. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J Infect Dis 197:1079–1081. https://doi.org/10.1086/533452.
    • (2008) J Infect Dis , vol.197 , pp. 1079-1081
    • Rice, L.B.1
  • 5
    • 78449256152 scopus 로고    scopus 로고
    • Progress and challenges in implementing the research on ESKAPE pathogens
    • Rice, LB. 2010. Progress and challenges in implementing the research on ESKAPE pathogens. Infect Control Hosp Epidemiol 31:S7–S10. https://doi.org/10.1086/655995.
    • (2010) Infect Control Hosp Epidemiol , vol.31 , pp. SS7-S10
    • Rice, L.B.1
  • 6
    • 85018309136 scopus 로고    scopus 로고
    • Resistance among Gram-negative ESKAPE pathogens isolated from hospitalized patients with intra-abdominal and urinary tract infections in Latin American countries: SMART 2013-2015. Braz
    • Karlowsky, JA, Hoban DJ, Hackel MA, Lob SH, Sahm DF. 2017. Resistance among Gram-negative ESKAPE pathogens isolated from hospitalized patients with intra-abdominal and urinary tract infections in Latin American countries: SMART 2013-2015. Braz, J Infect Dis 21:343–348. https://doi.org/10.1016/j.bjid.2017.03.006.
    • (2017) J Infect Dis , vol.21 , pp. 343-348
    • Karlowsky, J.A.1    Hoban, D.J.2    Hackel, M.A.3    Lob, S.H.4    Sahm, D.F.5
  • 7
    • 85014390378 scopus 로고    scopus 로고
    • Antimicrobial susceptibility of Gram-negative ESKAPE pathogens isolated from hospitalized patients with intra-abdominal and urinary tract infections in Asia-Pacific countries: SMART 2013-2015
    • Karlowsky, JA, Hoban DJ, Hackel MA, Lob SH, Sahm DF. 2017. Antimicrobial susceptibility of Gram-negative ESKAPE pathogens isolated from hospitalized patients with intra-abdominal and urinary tract infections in Asia-Pacific countries: SMART 2013-2015. J Med Microbiol 66:61–69. https://doi.org/10.1099/jmm.0.000421.
    • (2017) J Med Microbiol , vol.66 , pp. 61-69
    • Karlowsky, J.A.1    Hoban, D.J.2    Hackel, M.A.3    Lob, S.H.4    Sahm, D.F.5
  • 8
    • 84871528845 scopus 로고    scopus 로고
    • Additional risk factors for infection by multidrug-resistant pathogens in healthcare-associated infection: A large cohort study
    • Cardoso, T, Ribeiro O, Aragao IC, Costa-Pereira A, Sarmento AE. 2012. Additional risk factors for infection by multidrug-resistant pathogens in healthcare-associated infection: a large cohort study. BMC Infect Dis 12:375. https://doi.org/10.1186/1471-2334-12-375.
    • (2012) BMC Infect Dis , vol.12 , pp. 375
    • Cardoso, T.1    Ribeiro, O.2    Aragao, I.C.3    Costa-Pereira, A.4    Sarmento, A.E.5
  • 10
    • 78349311509 scopus 로고    scopus 로고
    • Multiresistant, Gram-negative infections: A global perspective
    • Ho, J, Tambyah PA, Paterson DL. 2010. Multiresistant, Gram-negative infections: a global perspective. Curr Opin Infect Dis 23:546–553. https://doi.org/10.1097/QCO.0b013e32833f0d3e.
    • (2010) Curr Opin Infect Dis , vol.23 , pp. 546-553
    • Ho, J.1    Tambyah, P.A.2    Paterson, D.L.3
  • 11
    • 84886803095 scopus 로고    scopus 로고
    • Antibiotics in the clinical pipeline in 2013
    • Butler, MS, Blaskovich MA, Cooper MA. 2013. Antibiotics in the clinical pipeline in 2013. J Antibiot (Tokyo) 66:571–591. https://doi.org/10.1038/ja.2013.86.
    • (2013) J Antibiot (Tokyo) , vol.66 , pp. 571-591
    • Butler, M.S.1    Blaskovich, M.A.2    Cooper, M.A.3
  • 13
    • 85002808374 scopus 로고    scopus 로고
    • Antimicrobial resistance (AMR) and the United Nations (UN)
    • Houghton, F. 2017. Antimicrobial resistance (AMR) and the United Nations (UN). J Infect Public Health 10:139–140. https://doi.org/10.1016/j.jiph.2016.10.002.
    • (2017) J Infect Public Health , vol.10 , pp. 139-140
    • Houghton, F.1
  • 14
    • 85002931239 scopus 로고    scopus 로고
    • The, United Nations and the urgent need for coordinated global action in the fight against antimicrobial resistance
    • Boucher, HW, Bakken JS, Murray BE. 2016. The, United Nations and the urgent need for coordinated global action in the fight against antimicrobial resistance. Ann Intern Med 165:812–813. https://doi.org/10.7326/M16-2079.
    • (2016) Ann Intern Med , vol.165 , pp. 812-813
    • Boucher, H.W.1    Bakken, J.S.2    Murray, B.E.3
  • 15
    • 84983348040 scopus 로고    scopus 로고
    • Achieving global targets for antimicrobial resistance
    • Laxminarayan, R, Sridhar D, Blaser M, Wang M, Woolhouse M. 2016. Achieving global targets for antimicrobial resistance. Science 353: 874–875. https://doi.org/10.1126/science.aaf9286.
    • (2016) Science , vol.353 , pp. 874-875
    • Laxminarayan, R.1    Sridhar, D.2    Blaser, M.3    Wang, M.4    Woolhouse, M.5
  • 17
    • 84878278251 scopus 로고    scopus 로고
    • 10 × ‘20 progress— development of new drugs active against Gram-negative bacilli: An update from the Infectious Diseases Society of America
    • Boucher, HW, Talbot GH, Benjamin DK, Jr, Bradley J, Guidos RJ, Jones RN, Murray BE, Bonomo RA, Gilbert D. 2013. 10 × ‘20 progress— development of new drugs active against Gram-negative bacilli: an update from the Infectious Diseases Society of America. Clin Infect Dis 56:1685–1694. https://doi.org/10.1093/cid/cit152.
    • (2013) Clin Infect Dis , vol.56 , pp. 1685-1694
    • Boucher, H.W.1    Talbot, G.H.2    Benjamin, D.K.3    Bradley, J.4    Guidos, R.J.5    Jones, R.N.6    Murray, B.E.7    Bonomo, R.A.8    Gilbert, D.9
  • 18
    • 85002213837 scopus 로고    scopus 로고
    • A Gestalt approach to Gram-negative entry
    • Silver, LL. 2016. A Gestalt approach to Gram-negative entry. Bioorg Med Chem 24:6379–6389. https://doi.org/10.1016/j.bmc.2016.06.044.
    • (2016) Bioorg Med Chem , vol.24 , pp. 6379-6389
    • Silver, L.L.1
  • 19
    • 73649107562 scopus 로고    scopus 로고
    • Impact of the glycostructure of amphiphilic membrane components on the function of the outer membrane of Gram-negative bacteria as a matrix for incorporated channels and a target for antimicrobial peptides or proteins
    • Gutsmann, T, Seydel U. 2010. Impact of the glycostructure of amphiphilic membrane components on the function of the outer membrane of Gram-negative bacteria as a matrix for incorporated channels and a target for antimicrobial peptides or proteins. Eur J Cell Biol 89:11–23. https://doi.org/10.1016/j.ejcb.2009.10.011.
    • (2010) Eur J Cell Biol , vol.89 , pp. 11-23
    • Gutsmann, T.1    Seydel, U.2
  • 20
    • 84884303247 scopus 로고    scopus 로고
    • Molecular dynamics and NMR spectroscopy studies of E. Coli lipopolysaccharide structure and dynamics
    • Wu, EL, Engstrom O, Jo S, Stuhlsatz D, Yeom MS, Klauda JB, Widmalm G, Im W. 2013. Molecular dynamics and NMR spectroscopy studies of E. coli lipopolysaccharide structure and dynamics. Biophys J 105: 1444–1455. https://doi.org/10.1016/j.bpj.2013.08.002.
    • (2013) Biophys J , vol.105 , pp. 1444-1455
    • Wu, E.L.1    Engstrom, O.2    Jo, S.3    Stuhlsatz, D.4    Yeom, M.S.5    Klauda, J.B.6    Widmalm, G.7    Im, W.8
  • 21
    • 84934928427 scopus 로고    scopus 로고
    • The membranes of Gram-negative bacteria: Progress in molecular modelling and simulation
    • Khalid, S, Berglund NA, Holdbrook DA, Leung YM, Parkin J. 2015. The membranes of Gram-negative bacteria: progress in molecular modelling and simulation. Biochem Soc Trans 43:162–167. https://doi.org/10.1042/BST20140262.
    • (2015) Biochem Soc Trans , vol.43 , pp. 162-167
    • Khalid, S.1    Berglund, N.A.2    Holdbrook, D.A.3    Leung, Y.M.4    Parkin, J.5
  • 22
    • 80855139866 scopus 로고    scopus 로고
    • Electroporation of the E. Coli and S. Aureus membranes: Molecular dynamics simulations of complex bacterial membranes
    • Piggot, TJ, Holdbrook DA, Khalid S. 2011. Electroporation of the E. coli and S. Aureus membranes: molecular dynamics simulations of complex bacterial membranes. J Phys Chem B 115:13381–13388. https://doi.org/10.1021/jp207013v.
    • (2011) J Phys Chem B , vol.115 , pp. 13381-13388
    • Piggot, T.J.1    Holdbrook, D.A.2    Khalid, S.3
  • 23
    • 23444456920 scopus 로고
    • Prevention of drug access to bacterial targets: Permeability barriers and active efflux
    • Nikaido, H. 1994. Prevention of drug access to bacterial targets: permeability barriers and active efflux. Science 264:382–388. https://doi.org/10.1126/science.8153625.
    • (1994) Science , vol.264 , pp. 382-388
    • Nikaido, H.1
  • 24
    • 84987664888 scopus 로고    scopus 로고
    • Molecular interactions of lipopolysaccharide with an outer membrane protein from Pseudomonas aeruginosa probed by solution NMR
    • Kucharska, I, Liang B, Ursini N, Tamm LK. 2016. Molecular interactions of lipopolysaccharide with an outer membrane protein from Pseudomonas aeruginosa probed by solution NMR. Biochemistry 55:5061–5072. https://doi.org/10.1021/acs.biochem.6b00630.
    • (2016) Biochemistry , vol.55 , pp. 5061-5072
    • Kucharska, I.1    Liang, B.2    Ursini, N.3    Tamm, L.K.4
  • 25
    • 85006736282 scopus 로고    scopus 로고
    • Quantitative measurement of the outer membrane permeability in Escherichia coli lpp and tol-pal mutants defines the significance of Tol-Pal function for maintaining drug resistance
    • Kowata, H, Tochigi S, Kusano T, Kojima S. 2016. Quantitative measurement of the outer membrane permeability in Escherichia coli lpp and tol-pal mutants defines the significance of Tol-Pal function for maintaining drug resistance. J Antibiot (Tokyo) 69:863–870. https://doi.org/10.1038/ja.2016.50.
    • (2016) J Antibiot (Tokyo) , vol.69 , pp. 863-870
    • Kowata, H.1    Tochigi, S.2    Kusano, T.3    Kojima, S.4
  • 26
    • 0025218933 scopus 로고
    • Partitioning of hydrophobic probes into lipopolysaccharide bilayers
    • Vaara, M, Plachy WZ, Nikaido H. 1990. Partitioning of hydrophobic probes into lipopolysaccharide bilayers. Biochim Biophys Acta 1024: 152–158. https://doi.org/10.1016/0005-2736(90)90218-D.
    • (1990) Biochim Biophys Acta , vol.1024 , pp. 152-158
    • Vaara, M.1    Plachy, W.Z.2    Nikaido, H.3
  • 27
    • 84879418353 scopus 로고    scopus 로고
    • Fortifying the barrier: The impact of lipid A remodelling on bacterial pathogenesis
    • Needham, BD, Trent MS. 2013. Fortifying the barrier: the impact of lipid A remodelling on bacterial pathogenesis. Nat Rev Microbiol 11: 467–481. https://doi.org/10.1038/nrmicro3047.
    • (2013) Nat Rev Microbiol , vol.11 , pp. 467-481
    • Needham, B.D.1    Trent, M.S.2
  • 28
    • 79960923273 scopus 로고    scopus 로고
    • Pseudomonas aeruginosa: All roads lead to resistance
    • Breidenstein, EBM, de la Fuente-Nunez C, Hancock REW. 2011. Pseudomonas aeruginosa: all roads lead to resistance. Trends Microbiol 19:419–426. https://doi.org/10.1016/j.tim.2011.04.005.
    • (2011) Trends Microbiol , vol.19 , pp. 419-426
    • Breidenstein, E.1    de La Fuente-Nunez, C.2    Hancock, R.3
  • 30
    • 0034885143 scopus 로고    scopus 로고
    • Vancomycin, teicoplanin, and ramoplanin: Synthetic and mechanistic studies
    • Boger, D. 2001. Vancomycin, teicoplanin, and ramoplanin: synthetic and mechanistic studies. Med Res Rev 21:356–381. https://doi.org/10.1002/med.1014.
    • (2001) Med Res Rev , vol.21 , pp. 356-381
    • Boger, D.1
  • 31
    • 44649092678 scopus 로고    scopus 로고
    • Are natural products still the best source for antibacterial discovery? The bacterial entry factor
    • Silver, LL. 2008. Are natural products still the best source for antibacterial discovery? The bacterial entry factor. Expert Opin Drug Discov 3:487–500. https://doi.org/10.1517/17460441.3.5.487.
    • (2008) Expert Opin Drug Discov , vol.3 , pp. 487-500
    • Silver, L.L.1
  • 32
    • 84887294881 scopus 로고    scopus 로고
    • Structural basis for substrate specificity in the Escherichia coli maltose transport system
    • Oldham, ML, Chen S, Chen J. 2013. Structural basis for substrate specificity in the Escherichia coli maltose transport system. Proc Natl Acad Sci U S A 110:18132–18137. https://doi.org/10.1073/pnas.1311407110.
    • (2013) Proc Natl Acad Sci U S A , vol.110 , pp. 18132-18137
    • Oldham, M.L.1    Chen, S.2    Chen, J.3
  • 33
    • 79960619992 scopus 로고    scopus 로고
    • Maltodextrin-based imaging probes detect bacteria in vivo with high sensitivity and specificity
    • Ning, X, Lee S, Wang Z, Kim D, Stubblefield B, Gilbert E, Murthy N. 2011. Maltodextrin-based imaging probes detect bacteria in vivo with high sensitivity and specificity. Nat Mater 10:602–607. https://doi.org/10.1038/nmat3074.
    • (2011) Nat Mater , vol.10 , pp. 602-607
    • Ning, X.1    Lee, S.2    Wang, Z.3    Kim, D.4    Stubblefield, B.5    Gilbert, E.6    Murthy, N.7
  • 34
    • 80052080449 scopus 로고    scopus 로고
    • The structural biology of beta-barrel membrane proteins: A summary of recent reports
    • Fairman, JW, Noinaj N, Buchanan SK. 2011. The structural biology of beta-barrel membrane proteins: a summary of recent reports. Curr Opin Struct Biol 21:523–531. https://doi.org/10.1016/j.sbi.2011.05.005.
    • (2011) Curr Opin Struct Biol , vol.21 , pp. 523-531
    • Fairman, J.W.1    Noinaj, N.2    Buchanan, S.K.3
  • 35
    • 77956634838 scopus 로고    scopus 로고
    • Porins in prokaryotes and eukaryotes: Common themes and variations
    • Zeth, K, Thein M. 2010. Porins in prokaryotes and eukaryotes: common themes and variations. Biochem J 431:13–22. https://doi.org/10.1042/BJ20100371.
    • (2010) Biochem J , vol.431 , pp. 13-22
    • Zeth, K.1    Thein, M.2
  • 37
    • 64649088018 scopus 로고    scopus 로고
    • Outer membrane permeability and antibiotic resistance
    • Delcour, AH. 2009. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta 1794:808–816. https://doi.org/10.1016/j.bbapap.2008.11.005.
    • (2009) Biochim Biophys Acta , pp. 808-816
    • Delcour, A.H.1
  • 38
    • 0016713447 scopus 로고
    • Outer membrane of Salmonella typhimurium: Reconstitution of sucrose-permeable membrane vesicles
    • Nakae, T. 1975. Outer membrane of Salmonella typhimurium: reconstitution of sucrose-permeable membrane vesicles. Biochem Biophys Res Commun 64:1224–1230. https://doi.org/10.1016/0006-291X(75)90823-2.
    • (1975) Biochem Biophys Res Commun , vol.64 , pp. 1224-1230
    • Nakae, T.1
  • 39
    • 0347479229 scopus 로고    scopus 로고
    • Molecular basis of bacterial outer membrane permeability revisited
    • Nikaido, H. 2003. Molecular basis of bacterial outer membrane permeability revisited. Microbiol Mol Biol Rev 67:593–656. https://doi.org/10.1128/MMBR.67.4.593-656.2003.
    • (2003) Microbiol Mol Biol Rev , vol.67 , pp. 593-656
    • Nikaido, H.1
  • 40
    • 0018357034 scopus 로고
    • Identification of the protein producing transmembrane diffusion pores in the outer membrane of Pseudomonas aeruginosa PA01
    • Hancock, RE, Decad GM, Nikaido H. 1979. Identification of the protein producing transmembrane diffusion pores in the outer membrane of Pseudomonas aeruginosa PA01. Biochim, Biophys Acta 554:323–331. https://doi.org/10.1016/0005-2736(79)90373-0.
    • (1979) Biochim. Biophys Acta , vol.554 , pp. 323-331
    • Hancock, R.E.1    Decad, G.M.2    Nikaido, H.3
  • 42
    • 0025968741 scopus 로고
    • Identification and characterization of porins in Pseudomonas aeruginosa
    • Nikaido, H, Nikaido K, Harayama S. 1991. Identification and characterization of porins in Pseudomonas aeruginosa. J Biol Chem 266: 770–779.
    • (1991) J Biol Chem , vol.266 , pp. 770-779
    • Nikaido, H.1    Nikaido, K.2    Harayama, S.3
  • 43
    • 0031879743 scopus 로고    scopus 로고
    • Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria
    • Hancock, RE. 1998. Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin Infect Dis 27(Suppl 1):S93–S99. https://doi.org/10.1086/514909.
    • (1998) Clin Infect Dis , vol.27 , pp. S93-S99
    • Hancock, R.E.1
  • 44
    • 0020574169 scopus 로고
    • Purification and properties of Pseudomonas aeruginosa porin
    • Yoshimura, F, Zalman LS, Nikaido H. 1983. Purification and properties of Pseudomonas aeruginosa porin. J Biol Chem 258:2308–2314.
    • (1983) J Biol Chem , vol.258 , pp. 2308-2314
    • Yoshimura, F.1    Zalman, L.S.2    Nikaido, H.3
  • 45
    • 85011940598 scopus 로고    scopus 로고
    • The intrinsic resistome of Pseudomonas aeruginosa to betalactams
    • Alvarez-Ortega C, Wiegand I, Olivares J, Hancock REW, Martinez JL. 2011. The intrinsic resistome of Pseudomonas aeruginosa to betalactams. Virulence 2:144–146. https://doi.org/10.4161/viru.2.2.15014.
    • (2011) Virulence , vol.2 , pp. 144-146
    • Alvarez-Ortega, C.1    Wiegand, I.2    Olivares, J.3    Hancock, R.4    Martinez, J.L.5
  • 46
    • 0025862413 scopus 로고
    • Interaction of aminoglycosides with the outer membranes and purified lipopolysaccharide and OmpF porin of Escherichia coli
    • Hancock, RE, Farmer SW, Li ZS, Poole K. 1991. Interaction of aminoglycosides with the outer membranes and purified lipopolysaccharide and OmpF porin of Escherichia coli. Antimicrob Agents Chemother 35: 1309–1314. https://doi.org/10.1128/AAC.35.7.1309.
    • (1991) Antimicrob Agents Chemother , vol.35 , pp. 1309-1314
    • Hancock, R.E.1    Farmer, S.W.2    Li, Z.S.3    Poole, K.4
  • 47
    • 0021185399 scopus 로고
    • Alterations in outer membrane permeability
    • Hancock, RE. 1984. Alterations in outer membrane permeability. Annu Rev Microbiol 38:237–264. https://doi.org/10.1146/annurev.mi.38.100184.001321.
    • (1984) Annu Rev Microbiol , vol.38 , pp. 237-264
    • Hancock, R.E.1
  • 49
    • 84921389409 scopus 로고    scopus 로고
    • Effect of divalent cation removal on the structure of gram-negative bacterial outer membrane models
    • Clifton, LA, Skoda MWA, Le Brun AP, Ciesielski F, Kuzmenko I, Holt SA, Lakey JH. 2015. Effect of divalent cation removal on the structure of gram-negative bacterial outer membrane models. Langmuir 31: 404–412. https://doi.org/10.1021/la504407v.
    • (2015) Langmuir , vol.31 , pp. 404-412
    • Clifton, L.A.1    Skoda, M.2    Le Brun, A.P.3    Ciesielski, F.4    Kuzmenko, I.5    Holt, S.A.6    Lakey, J.H.7
  • 50
    • 84937509566 scopus 로고    scopus 로고
    • Bacterial lipopolysaccharides form physically cross-linked, twodimensional gels in the presence of divalent cations
    • Herrmann, M, Schneck E, Gutsmann T, Brandenburg K, Tanaka M. 2015. Bacterial lipopolysaccharides form physically cross-linked, twodimensional gels in the presence of divalent cations. Soft Matter 11: 6037–6044. https://doi.org/10.1039/C5SM01002K.
    • (2015) Soft Matter , vol.11 , pp. 6037-6044
    • Herrmann, M.1    Schneck, E.2    Gutsmann, T.3    Brandenburg, K.4    Tanaka, M.5
  • 51
    • 67449167509 scopus 로고    scopus 로고
    • Uptake pathways of anionic and cationic photosensitizers into bacteria
    • George, S, Hamblin MR, Kishen A. 2009. Uptake pathways of anionic and cationic photosensitizers into bacteria. Photochem Photobiol Sci 8:788–795. https://doi.org/10.1039/b809624d.
    • (2009) Photochem Photobiol Sci , vol.8 , pp. 788-795
    • George, S.1    Hamblin, M.R.2    Kishen, A.3
  • 52
    • 0024239039 scopus 로고
    • Antibiotic uptake into gram-negative bacteria
    • Hancock, RE, Bell A. 1988. Antibiotic uptake into gram-negative bacteria. Eur J Clin Microbiol Infect Dis 7:713–720. https://doi.org/10.1007/BF01975036.
    • (1988) Eur J Clin Microbiol Infect Dis , vol.7 , pp. 713-720
    • Hancock, R.E.1    Bell, A.2
  • 53
    • 0019312991 scopus 로고
    • Outer membrane protein H1 of Pseudomonas aeruginosa: Involvement in adaptive and mutational resistance to ethylenediaminetetraacetate, polymyxin B, and gentamicin
    • Nicas, TI, Hancock RE. 1980. Outer membrane protein H1 of Pseudomonas aeruginosa: involvement in adaptive and mutational resistance to ethylenediaminetetraacetate, polymyxin B, and gentamicin. J Bacteriol 143:872–878.
    • (1980) J Bacteriol , vol.143 , pp. 872-878
    • Nicas, T.I.1    Hancock, R.E.2
  • 54
    • 0015792721 scopus 로고
    • Activity of aminoglycoside antibiotics against Pseudomonas aeruginosa: Specificity and site of calcium and magnesium antagonism
    • Zimelis, VM, Jackson GG. 1973. Activity of aminoglycoside antibiotics against Pseudomonas aeruginosa: specificity and site of calcium and magnesium antagonism. J Infect Dis 127:663–669. https://doi.org/10.1093/infdis/127.6.663.
    • (1973) J Infect Dis , vol.127 , pp. 663-669
    • Zimelis, V.M.1    Jackson, G.G.2
  • 55
    • 0015178295 scopus 로고
    • Activity of colistin against Pseudomonas aeruginosa: Inhibition by calcium
    • Davis, SD, Iannetta A, Wedgwood RJ. 1971. Activity of colistin against Pseudomonas aeruginosa: inhibition by calcium. J Infect Dis 124: 610–612. https://doi.org/10.1093/infdis/124.6.610.
    • (1971) J Infect Dis , vol.124 , pp. 610-612
    • Davis, S.D.1    Iannetta, A.2    Wedgwood, R.J.3
  • 56
    • 84965184470 scopus 로고
    • Site of action of polymyxin on Pseudomonas aeruginosa: Antagonism by cations
    • Newton, BA. 1954. Site of action of polymyxin on Pseudomonas aeruginosa: antagonism by cations. J Gen Microbiol 10:491–499. https://doi.org/10.1099/00221287-10-3-491.
    • (1954) J Gen Microbiol , vol.10 , pp. 491-499
    • Newton, B.A.1
  • 57
    • 0021282341 scopus 로고
    • Compounds which increase the permeability of the Pseudomonas aeruginosa outer membrane
    • Hancock, RE, Wong PG. 1984. Compounds which increase the permeability of the Pseudomonas aeruginosa outer membrane. Antimicrob Agents Chemother 26:48–52. https://doi.org/10.1128/AAC.26.1.48.
    • (1984) Antimicrob Agents Chemother , vol.26 , pp. 48-52
    • Hancock, R.E.1    Wong, P.G.2
  • 58
    • 1542673165 scopus 로고    scopus 로고
    • On the mechanism of solute uptake in Pseudomonas
    • Tamber, S, Hancock REW. 2003. On the mechanism of solute uptake in Pseudomonas. Front Biosci 8:s472–s483. https://doi.org/10.2741/1075.
    • (2003) Front Biosci , vol.8 , pp. s472-s483
    • Tamber, S.1    Hancock, R.E.W.2
  • 59
    • 84884512993 scopus 로고    scopus 로고
    • Collapsing the proton motive force to identify synergistic combinations against Staphylococcus aureus
    • Farha, MA, Verschoor CP, Bowdish D, Brown ED. 2013. Collapsing the proton motive force to identify synergistic combinations against Staphylococcus aureus. Chem Biol 20:1168–1178. https://doi.org/10.1016/j.chembiol.2013.07.006.
    • (2013) Chem Biol , vol.20 , pp. 1168-1178
    • Farha, M.A.1    Verschoor, C.P.2    Bowdish, D.3    Brown, E.D.4
  • 60
    • 0023876359 scopus 로고
    • Molecular mechanisms involved in the transport of antibiotics into bacteria
    • Chopra, I. 1988. Molecular mechanisms involved in the transport of antibiotics into bacteria. Parasitology 96(Suppl):S25–S44. https://doi.org/10.1017/S0031182000085966.
    • (1988) Parasitology , vol.96 , pp. S25-S44
    • Chopra, I.1
  • 61
    • 33744780736 scopus 로고    scopus 로고
    • Outer membrane active transport: Structure of the BtuB:TonB complex
    • Shultis, DD, Purdy MD, Banchs CN, Wiener MC. 2006. Outer membrane active transport: structure of the BtuB:TonB complex. Science 312: 1396–1399. https://doi.org/10.1126/science.1127694.
    • (2006) Science , vol.312 , pp. 1396-1399
    • Shultis, D.D.1    Purdy, M.D.2    Banchs, C.N.3    Wiener, M.C.4
  • 62
    • 0021783513 scopus 로고
    • The proton motive force in bacteria: A critical assessment of methods
    • Kashket, ER. 1985. The proton motive force in bacteria: a critical assessment of methods. Annu Rev Microbiol 39:219–242. https://doi.org/10.1146/annurev.mi.39.100185.001251.
    • (1985) Annu Rev Microbiol , vol.39 , pp. 219-242
    • Kashket, E.R.1
  • 63
    • 36349000205 scopus 로고    scopus 로고
    • A mechanistical model for the uptake of sulfonamides by bacteria
    • Zarfl, C, Matthies M, Klasmeier J. 2008. A mechanistical model for the uptake of sulfonamides by bacteria. Chemosphere 70:753–760. https://doi.org/10.1016/j.chemosphere.2007.07.045.
    • (2008) Chemosphere , vol.70 , pp. 753-760
    • Zarfl, C.1    Matthies, M.2    Klasmeier, J.3
  • 66
    • 84963979272 scopus 로고    scopus 로고
    • Permeability barrier of Gram-negative cell envelopes and approaches to bypass it
    • Zgurskaya, HI, Lopez CA, Gnanakaran S. 2015. Permeability barrier of Gram-negative cell envelopes and approaches to bypass it. ACS Infect Dis 1:512–522. https://doi.org/10.1021/acsinfecdis.5b00097.
    • (2015) ACS Infect Dis , vol.1 , pp. 512-522
    • Zgurskaya, H.I.1    Lopez, C.A.2    Gnanakaran, S.3
  • 67
    • 84926035587 scopus 로고    scopus 로고
    • The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria
    • Li, X-Z, Plesiat P, Nikaido H. 2015. The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev 28:337–418. https://doi.org/10.1128/CMR.00117-14.
    • (2015) Clin Microbiol Rev , vol.28 , pp. 337-418
    • Li, X.-Z.1    Plesiat, P.2    Nikaido, H.3
  • 68
    • 84930202295 scopus 로고    scopus 로고
    • Molecular architecture of the bacterial tripartite multidrug efflux pump focusing on the adaptor bridging model
    • Song, S, Kim J-S, Lee K, Ha N-C. 2015. Molecular architecture of the bacterial tripartite multidrug efflux pump focusing on the adaptor bridging model. J Microbiol 53:355–364. https://doi.org/10.1007/s12275-015-5248-4.
    • (2015) J Microbiol , vol.53 , pp. 355-364
    • Song, S.1    Kim, J.-S.2    Lee, K.3    Ha, N.-C.4
  • 69
    • 84901992360 scopus 로고    scopus 로고
    • Bacterial multidrug efflux transporters
    • Delmar, JA, Su C-C, Yu EW. 2014. Bacterial multidrug efflux transporters. Annu Rev Biophys 43:93–117. https://doi.org/10.1146/annurev-biophys-051013-022855.
    • (2014) Annu Rev Biophys , vol.43 , pp. 93-117
    • Delmar, J.A.1    Su, C.-C.2    Yu, E.W.3
  • 70
    • 84860163257 scopus 로고    scopus 로고
    • Biochemistry of bacterial multidrug efflux pumps
    • Kumar, S, Varela MF. 2012. Biochemistry of bacterial multidrug efflux pumps. Int J Mol Sci 13:4484–4495. https://doi.org/10.3390/ijms13044484.
    • (2012) Int J Mol Sci , vol.13 , pp. 4484-4495
    • Kumar, S.1    Varela, M.F.2
  • 71
    • 65249146929 scopus 로고    scopus 로고
    • Multidrug resistance in bacteria
    • Nikaido, H. 2009. Multidrug resistance in bacteria. Annu Rev Biochem 78:119–146. https://doi.org/10.1146/annurev.biochem.78.082907.145923.
    • (2009) Annu Rev Biochem , vol.78 , pp. 119-146
    • Nikaido, H.1
  • 72
    • 33646251815 scopus 로고    scopus 로고
    • Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria
    • Piddock, LJV. 2006. Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria. Clin Microbiol Rev 19: 382–402. https://doi.org/10.1128/CMR.19.2.382-402.2006.
    • (2006) Clin Microbiol Rev , vol.19 , pp. 382-402
    • Piddock, L.J.V.1
  • 73
    • 85009812112 scopus 로고    scopus 로고
    • What is an “ideal” antibiotic? Discovery challenges and path forward
    • Singh, SB, Young K, Silver LL. 2017. What is an “ideal” antibiotic? Discovery challenges and path forward. Biochem Pharmacol 133:63–73. https://doi.org/10.1016/j.bcp.2017.01.003.
    • (2017) Biochem Pharmacol , vol.133 , pp. 63-73
    • Singh, S.B.1    Young, K.2    Silver, L.L.3
  • 74
    • 0035289779 scopus 로고    scopus 로고
    • Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings
    • Lipinski, CA, Lombardo F, Dominy BW, Feeney PJ. 2001. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46:3–26. https://doi.org/10.1016/S0169-409X(00)00129-0.
    • (2001) Adv Drug Deliv Rev , vol.46 , pp. 3-26
    • Lipinski, C.A.1    Lombardo, F.2    Dominy, B.W.3    Feeney, P.J.4
  • 75
    • 84966707674 scopus 로고    scopus 로고
    • Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions
    • Lipinski, CA. 2016. Rule of five in 2015 and beyond: target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions. Adv Drug Deliv Rev 101:34–41. https://doi.org/10.1016/j.addr.2016.04.029.
    • (2016) Adv Drug Deliv Rev , vol.101 , pp. 34-41
    • Lipinski, C.A.1
  • 76
    • 84975796082 scopus 로고    scopus 로고
    • Are the physicochemical properties of antibacterial compounds really different from other drugs?
    • Ebejer, J-P, Charlton MH, Finn PW. 2016. Are the physicochemical properties of antibacterial compounds really different from other drugs? J Cheminform 8:30. https://doi.org/10.1186/s13321-016-0143-5.
    • (2016) J Cheminform , vol.8 , pp. 30
    • Ebejer, J.-P.1    Charlton, M.H.2    Finn, P.W.3
  • 77
    • 43949129098 scopus 로고    scopus 로고
    • Physicochemical properties of antibacterial compounds: Implications for drug discovery
    • O’Shea R, Moser HE. 2008. Physicochemical properties of antibacterial compounds: implications for drug discovery. J Med Chem 51: 2871–2878. https://doi.org/10.1021/jm700967e.
    • (2008) J Med Chem , vol.51 , pp. 2871-2878
    • O’Shea, R.1    Moser, H.E.2
  • 78
    • 85006186498 scopus 로고    scopus 로고
    • Progress against Escherichia coli with the oxazolidinone class of antibacterials: Test case for a general approach to improving whole-cell Gram-negative activity
    • Takrouri, K, Cooper HD, Spaulding A, Zucchi P, Koleva B, Cleary DC, Tear W, Beuning PJ, Hirsch EB, Aggen JB. 2016. Progress against Escherichia coli with the oxazolidinone class of antibacterials: test case for a general approach to improving whole-cell Gram-negative activity. ACS Infect Dis 2:405–426. https://doi.org/10.1021/acsinfecdis.6b00003.
    • (2016) ACS Infect Dis , vol.2 , pp. 405-426
    • Takrouri, K.1    Cooper, H.D.2    Spaulding, A.3    Zucchi, P.4    Koleva, B.5    Cleary, D.C.6    Tear, W.7    Beuning, P.J.8    Hirsch, E.B.9    Aggen, J.B.10
  • 79
    • 34447553357 scopus 로고    scopus 로고
    • Intracellular accumulation of linezolid in Escherichia coli, Citrobacter freundii and Enterobacter aerogenes: Role of enhanced efflux pump activity and inactivation
    • Schumacher, A, Trittler R, Bohnert JA, Kummerer K, Pages J-M, Kern WV. 2007. Intracellular accumulation of linezolid in Escherichia coli, Citrobacter freundii and Enterobacter aerogenes: role of enhanced efflux pump activity and inactivation. J Antimicrob Chemother 59:1261–1264. https://doi.org/10.1093/jac/dkl380.
    • (2007) J Antimicrob Chemother , vol.59 , pp. 1261-1264
    • Schumacher, A.1    Trittler, R.2    Bohnert, J.A.3    Kummerer, K.4    Pages, J.-M.5    Kern, W.V.6
  • 80
    • 84926648408 scopus 로고    scopus 로고
    • Thinking outside the “bug”: A unique assay to measure intracellular drug penetration in gram-negative bacteria
    • Zhou, Y, Joubran C, Miller-Vedam L, Isabella V, Nayar A, Tentarelli S, Miller A. 2015. Thinking outside the “bug”: a unique assay to measure intracellular drug penetration in gram-negative bacteria. Anal Chem 87:3579–3584. https://doi.org/10.1021/ac504880r.
    • (2015) Anal Chem , vol.87 , pp. 3579-3584
    • Zhou, Y.1    Joubran, C.2    Miller-Vedam, L.3    Isabella, V.4    Nayar, A.5    Tentarelli, S.6    Miller, A.7
  • 81
    • 84880434138 scopus 로고    scopus 로고
    • LC-MS based assay to measure intracellular compound levels in Mycobacterium smegmatis: Linking compound levels to cellular potency
    • Bhat, J, Narayan A, Venkatraman J, Chatterji M. 2013. LC-MS based assay to measure intracellular compound levels in Mycobacterium smegmatis: linking compound levels to cellular potency. J Microbiol Methods 94:152–158. https://doi.org/10.1016/j.mimet.2013.05.010.
    • (2013) J Microbiol Methods , vol.94 , pp. 152-158
    • Bhat, J.1    Narayan, A.2    Venkatraman, J.3    Chatterji, M.4
  • 82
    • 84928974565 scopus 로고    scopus 로고
    • Measuring propargyl-linked drug populations inside bacterial cells, and their interaction with a dihydrofolate reductase target, by Raman microscopy
    • Heidari-Torkabadi H, Che T, Lombardo MN, Wright DL, Anderson AC, Carey PR. 2015. Measuring propargyl-linked drug populations inside bacterial cells, and their interaction with a dihydrofolate reductase target, by Raman microscopy. Biochemistry 54:2719–2726. https://doi.org/10.1021/acs.biochem.5b00202.
    • (2015) Biochemistry , vol.54 , pp. 2719-2726
    • Heidari-Torkabadi, H.1    Che, T.2    Lombardo, M.N.3    Wright, D.L.4    Erson, A.C.5    Carey, P.R.6
  • 84
    • 84914142159 scopus 로고    scopus 로고
    • General platform for systematic quantitative evaluation of small-molecule permeability in bacteria
    • Davis, TD, Gerry CJ, Tan DS. 2014. General platform for systematic quantitative evaluation of small-molecule permeability in bacteria. ACS Chem Biol 9:2535–2544. https://doi.org/10.1021/cb5003015.
    • (2014) ACS Chem Biol , vol.9 , pp. 2535-2544
    • Davis, T.D.1    Gerry, C.J.2    Tan, D.S.3
  • 86
    • 78650293305 scopus 로고    scopus 로고
    • Targeting bacterial membrane function: An underexploited mechanism for treating persistent infections
    • Hurdle, JG, O’Neill AJ, Chopra I, Lee RE. 2011. Targeting bacterial membrane function: an underexploited mechanism for treating persistent infections. Nat Rev Microbiol 9:62–75. https://doi.org/10.1038/nrmicro2474.
    • (2011) Nat Rev Microbiol , vol.9 , pp. 62-75
    • Hurdle, J.G.1    O’Neill, A.J.2    Chopra, I.3    Lee, R.E.4
  • 87
    • 84942550117 scopus 로고    scopus 로고
    • Membrane-active small molecules: Designs inspired by antimicrobial peptides
    • Ghosh, C, Haldar J. 2015. Membrane-active small molecules: designs inspired by antimicrobial peptides. ChemMedChem 10:1606–1624. https://doi.org/10.1002/cmdc.201500299.
    • (2015) Chemmedchem , vol.10 , pp. 1606-1624
    • Ghosh, C.1    Haldar, J.2
  • 88
    • 84904824145 scopus 로고    scopus 로고
    • Design and synthesis of membranetargeting antibiotics: From peptides- to aminosugar-based antimicrobial cationic amphiphiles
    • Herzog, IM, Fridman M. 2014. Design and synthesis of membranetargeting antibiotics: from peptides- to aminosugar-based antimicrobial cationic amphiphiles. Medchemcomm 5:1014–1026. https://doi.org/10.1039/C4MD00012A.
    • (2014) Medchemcomm , vol.5 , pp. 1014-1026
    • Herzog, I.M.1    Fridman, M.2
  • 89
    • 84961721905 scopus 로고    scopus 로고
    • Short antimicrobial peptides and peptide scaffolds as promising antibacterial agents
    • Domalaon, R, Zhanel GG, Schweizer F. 2016. Short antimicrobial peptides and peptide scaffolds as promising antibacterial agents. Curr Top Med Chem 16:1217–1230. https://doi.org/10.2174/1568026615666150915112459.
    • (2016) Curr Top Med Chem , vol.16 , pp. 1217-1230
    • Domalaon, R.1    Zhanel, G.G.2    Schweizer, F.3
  • 90
    • 84946140850 scopus 로고    scopus 로고
    • Di-N-methylation of anti-Gram-positive aminoglycoside-derived membrane disruptors improves antimicrobial potency and broadens spectrum to Gramnegative bacteria
    • Benhamou, RI, Shaul P, Herzog IM, Fridman M. 2015. Di-N-methylation of anti-Gram-positive aminoglycoside-derived membrane disruptors improves antimicrobial potency and broadens spectrum to Gramnegative bacteria. Angew Chem Int Ed Engl 54:13617–13621. https://doi.org/10.1002/anie.201506814.
    • (2015) Angew Chem Int Ed Engl , vol.54 , pp. 13617-13621
    • Benhamou, R.I.1    Shaul, P.2    Herzog, I.M.3    Fridman, M.4
  • 93
    • 84879072389 scopus 로고    scopus 로고
    • Microbial persistence and the road to drug resistance
    • Cohen, NR, Lobritz MA, Collins JJ. 2013. Microbial persistence and the road to drug resistance. Cell Host Microbe 13:632–642. https://doi.org/10.1016/j.chom.2013.05.009.
    • (2013) Cell Host Microbe , vol.13 , pp. 632-642
    • Cohen, N.R.1    Lobritz, M.A.2    Collins, J.J.3
  • 94
    • 85028502175 scopus 로고    scopus 로고
    • Using engineered bacteria to characterize infection dynamics and antibiotic effects in vivo
    • Certain, LK, Way JC, Pezone MJ, Collins JJ. 2017. Using engineered bacteria to characterize infection dynamics and antibiotic effects in vivo. Cell Host Microbe 22:263.e4–268.e4. https://doi.org/10.1016/j.chom.2017.08.001.
    • (2017) Cell Host Microbe , vol.22 , pp. 263e4-268e4
    • Certain, L.K.1    Way, J.C.2    Pezone, M.J.3    Collins, J.J.4
  • 96
    • 84937628033 scopus 로고    scopus 로고
    • Collateral sensitivity of antibiotic-resistant microbes
    • Pál, C, Papp B, Lázár V. 2015. Collateral sensitivity of antibiotic-resistant microbes. Trends Microbiol 23:401–407. https://doi.org/10.1016/j.tim.2015.02.009.
    • (2015) Trends Microbiol , vol.23 , pp. 401-407
    • Pál, C.1    Papp, B.2    Lázár, V.3
  • 97
    • 65549165914 scopus 로고    scopus 로고
    • What antimicrobial resistance has taught us about horizontal gene transfer
    • Barlow, M. 2009. What antimicrobial resistance has taught us about horizontal gene transfer. Methods Mol Biol 532:397–411. https://doi.org/10.1007/978-1-60327-853-9_23.
    • (2009) Methods Mol Biol , vol.532 , pp. 397-411
    • Barlow, M.1
  • 98
    • 84905667072 scopus 로고    scopus 로고
    • News feature: Next-generation antibiotics
    • Williams, SCP. 2014. News feature: next-generation antibiotics. Proc Natl Acad Sci U S A 111:11227–11229. https://doi.org/10.1073/pnas.1413117111.
    • (2014) Proc Natl Acad Sci U S A , vol.111 , pp. 11227-11229
    • Williams, S.C.P.1
  • 99
    • 84971336976 scopus 로고    scopus 로고
    • The arsenal of pathogens and antivirulence therapeutic strategies for disarming them
    • Brannon, JR, Hadjifrangiskou M. 2016. The arsenal of pathogens and antivirulence therapeutic strategies for disarming them. Drug Des Devel Ther 10:1795–1806. https://doi.org/10.2147/DDDT.S98939.
    • (2016) Drug Des Devel Ther , vol.10 , pp. 1795-1806
    • Brannon, J.R.1    Hadjifrangiskou, M.2
  • 100
    • 85015935510 scopus 로고    scopus 로고
    • Different drugs for bad bugs: Antivirulence strategies in the age of antibiotic resistance
    • Dickey, SW, Cheung GYC, Otto M. 2017. Different drugs for bad bugs: antivirulence strategies in the age of antibiotic resistance. Nat Rev Drug Discov 16:457–471. https://doi.org/10.1038/nrd.2017.23.
    • (2017) Nat Rev Drug Discov , vol.16 , pp. 457-471
    • Dickey, S.W.1    Cheung, G.2    Otto, M.3
  • 101
    • 84905917315 scopus 로고    scopus 로고
    • The art of antibacterial warfare: Deception through interference with quorum sensing-mediated communication
    • Rampioni, G, Leoni L, Williams P. 2014. The art of antibacterial warfare: deception through interference with quorum sensing-mediated communication. Bioorg Chem 55:60–68. https://doi.org/10.1016/j.bioorg.2014.04.005.
    • (2014) Bioorg Chem , vol.55 , pp. 60-68
    • Rampioni, G.1    Leoni, L.2    Williams, P.3
  • 102
    • 85014007232 scopus 로고    scopus 로고
    • Social interactions in bacterial cell-cell signaling
    • Asfahl, KL, Schuster M. 2017. Social interactions in bacterial cell-cell signaling. FEMS Microbiol Rev 41:92–107. https://doi.org/10.1093/femsre/fuw038.
    • (2017) FEMS Microbiol Rev , vol.41 , pp. 92-107
    • Asfahl, K.L.1    Schuster, M.2
  • 104
    • 84922071627 scopus 로고    scopus 로고
    • The hierarchy quorum sensing network in Pseudomonas aeruginosa
    • Lee, J, Zhang L. 2015. The hierarchy quorum sensing network in Pseudomonas aeruginosa. Protein Cell 6:26–41. https://doi.org/10.1007/s13238-014-0100-x.
    • (2015) Protein Cell , vol.6 , pp. 26-41
    • Lee, J.1    Zhang, L.2
  • 105
    • 84982121502 scopus 로고    scopus 로고
    • Quorum sensing signal-response systems in Gram-negative bacteria
    • Papenfort, K, Bassler BL. 2016. Quorum sensing signal-response systems in Gram-negative bacteria. Nat Rev Microbiol 14:576–588. https://doi.org/10.1038/nrmicro.2016.89.
    • (2016) Nat Rev Microbiol , vol.14 , pp. 576-588
    • Papenfort, K.1    Bassler, B.L.2
  • 106
    • 84991237869 scopus 로고    scopus 로고
    • Considerations and caveats in anti-virulence drug development
    • Maura, D, Ballok AE, Rahme LG. 2016. Considerations and caveats in anti-virulence drug development. Curr Opin Microbiol 33:41–46. https://doi.org/10.1016/j.mib.2016.06.001.
    • (2016) Curr Opin Microbiol , vol.33 , pp. 41-46
    • Maura, D.1    Ballok, A.E.2    Rahme, L.G.3
  • 107
    • 84990223696 scopus 로고    scopus 로고
    • Quorum quenching strategy targeting Gram-positive pathogenic bacteria
    • Singh, RP, Desouky SE, Nakayama J. 2016. Quorum quenching strategy targeting Gram-positive pathogenic bacteria. Adv Exp Med Biol 901: 109–130. https://doi.org/10.1007/5584_2016_1.
    • (2016) Adv Exp Med Biol , vol.901 , pp. 109-130
    • Singh, R.P.1    Desouky, S.E.2    Nakayama, J.3
  • 108
    • 84918814054 scopus 로고    scopus 로고
    • Development of antivirulence compounds: A biochemical review
    • Zambelloni, R, Marquez R, Roe AJ. 2015. Development of antivirulence compounds: a biochemical review. Chem Biol Drug Des 85:43–55. https://doi.org/10.1111/cbdd.12430.
    • (2015) Chem Biol Drug Des , vol.85 , pp. 43-55
    • Zambelloni, R.1    Marquez, R.2    Roe, A.J.3
  • 112
    • 84990063713 scopus 로고    scopus 로고
    • Inhibition of quorum sensing-controlled virulence factors and biofilm formation in Pseudomonas aeruginosa by culture extract from novel bacterial species of Paenibacillus using a rat model of chronic lung infection
    • Alasil, SM, Omar R, Ismail S, Yusof MY. 2015. Inhibition of quorum sensing-controlled virulence factors and biofilm formation in Pseudomonas aeruginosa by culture extract from novel bacterial species of Paenibacillus using a rat model of chronic lung infection. Int J Bacteriol 2015:671562. https://doi.org/10.1155/2015/671562.
    • (2015) Int J Bacteriol , vol.2015
    • Alasil, S.M.1    Omar, R.2    Ismail, S.3    Yusof, M.Y.4
  • 113
    • 77952741849 scopus 로고    scopus 로고
    • Therapeutic effect of (Z)-3-(2,5-dimethoxyphenyl)-2-(4-methoxyphenyl) acrylonitrile (DMMA) against Staphylococcus aureus infection in a murine model
    • Oh, K-B, Nam K-W, Ahn H, Shin J, Kim S, Mar W. 2010. Therapeutic effect of (Z)-3-(2,5-dimethoxyphenyl)-2-(4-methoxyphenyl) acrylonitrile (DMMA) against Staphylococcus aureus infection in a murine model. Biochem Biophys Res Commun 396:440–444. https://doi.org/10.1016/j.bbrc.2010.04.113.
    • (2010) Biochem Biophys Res Commun , vol.396 , pp. 440-444
    • Oh, K.-B.1    Nam, K.-W.2    Ahn, H.3    Shin, J.4    Kim, S.5    Mar, W.6
  • 114
    • 84947976626 scopus 로고    scopus 로고
    • Mechanistic analysis of a synthetic inhibitor of the Pseudomonas aeruginosa LasI quorumsensing signal synthase
    • Lidor, O, Al-Quntar A, Pesci EC, Steinberg D. 2015. Mechanistic analysis of a synthetic inhibitor of the Pseudomonas aeruginosa LasI quorumsensing signal synthase. Sci Rep 5:16569. https://doi.org/10.1038/srep16569.
    • (2015) Sci Rep , vol.5 , pp. 16569
    • Lidor, O.1    Al-Quntar, A.2    Pesci, E.C.3    Steinberg, D.4
  • 115
    • 84897011636 scopus 로고    scopus 로고
    • Evolution of resistance to quorumsensing inhibitors
    • Kalia, VC, Wood TK, Kumar P. 2014. Evolution of resistance to quorumsensing inhibitors. Microb Ecol 68:13–23. https://doi.org/10.1007/s00248-013-0316-y.
    • (2014) Microb Ecol , vol.68 , pp. 13-23
    • Kalia, V.C.1    Wood, T.K.2    Kumar, P.3
  • 116
    • 84896908713 scopus 로고    scopus 로고
    • Novel approaches for the design and discovery of quorum-sensing inhibitors
    • Scutera, S, Zucca M, Savoia D. 2014. Novel approaches for the design and discovery of quorum-sensing inhibitors. Expert Opin Drug Discov 9:353–366. https://doi.org/10.1517/17460441.2014.894974.
    • (2014) Expert Opin Drug Discov , vol.9 , pp. 353-366
    • Scutera, S.1    Zucca, M.2    Savoia, D.3
  • 118
    • 0024387119 scopus 로고
    • Antibiotic therapy for Pseudomonas aeruginosa bacteremia: Outcome correlations in a prospective study of 200 patients
    • Hilf, M, Yu VL, Sharp J, Zuravleff JJ, Korvick JA, Muder RR. 1989. Antibiotic therapy for Pseudomonas aeruginosa bacteremia: outcome correlations in a prospective study of 200 patients. Am J Med 87:540–546. https://doi.org/10.1016/S0002-9343(89)80611-4.
    • (1989) Am J Med , vol.87 , pp. 540-546
    • Hilf, M.1    Yu, V.L.2    Sharp, J.3    Zuravleff, J.J.4    Korvick, J.A.5    Muder, R.R.6
  • 119
    • 84994518759 scopus 로고    scopus 로고
    • Antibiotic adjuvants: Rescuing antibiotics from resistance
    • Wright, GD. 2016. Antibiotic adjuvants: rescuing antibiotics from resistance. Trends Microbiol 24:862–871. https://doi.org/10.1016/j.tim.2016.06.009.
    • (2016) Trends Microbiol , vol.24 , pp. 862-871
    • Wright, G.D.1
  • 120
    • 1242352586 scopus 로고    scopus 로고
    • Augmentin (Amoxicillin/clavulanate) in the treatment of communityacquired respiratory tract infection: A review of the continuing development of an innovative antimicrobial agent
    • White, AR, Kaye C, Poupard J, Pypstra R, Woodnutt G, Wynne B. 2004. Augmentin (amoxicillin/clavulanate) in the treatment of communityacquired respiratory tract infection: a review of the continuing development of an innovative antimicrobial agent. J Antimicrob Chemother 53:i3–i20. https://doi.org/10.1093/jac/dkh050.
    • (2004) J Antimicrob Chemother , vol.53 , pp. ii3-i20
    • White, A.R.1    Kaye, C.2    Poupard, J.3    Pypstra, R.4    Woodnutt, G.5    Wynne, B.6
  • 121
    • 74249108028 scopus 로고    scopus 로고
    • Three decades of beta-lactamase inhibitors
    • Drawz, SM, Bonomo RA. 2010. Three decades of beta-lactamase inhibitors. Clin Microbiol Rev 23:160–201. https://doi.org/10.1128/CMR.00037-09.
    • (2010) Clin Microbiol Rev , vol.23 , pp. 160-201
    • Drawz, S.M.1    Bonomo, R.A.2
  • 122
    • 0035522323 scopus 로고    scopus 로고
    • Evolution and epidemiology of extendedspectrum beta-lactamases (ESBLs) and ESBL-producing microorganisms
    • Gniadkowski, M. 2001. Evolution and epidemiology of extendedspectrum beta-lactamases (ESBLs) and ESBL-producing microorganisms. Clin Microbiol Infect 7:597–608. https://doi.org/10.1046/j.1198-743x.2001.00330.x.
    • (2001) Clin Microbiol Infect , vol.7 , pp. 597-608
    • Gniadkowski, M.1
  • 123
    • 27144490073 scopus 로고    scopus 로고
    • Extended-spectrum beta-lactamases: A clinical update
    • Paterson, DL, Bonomo RA. 2005. Extended-spectrum beta-lactamases: a clinical update. Clin Microbiol Rev 18:657–686. https://doi.org/10.1128/CMR.18.4.657-686.2005.
    • (2005) Clin Microbiol Rev , vol.18 , pp. 657-686
    • Paterson, D.L.1    Bonomo, R.A.2
  • 124
    • 36048938508 scopus 로고    scopus 로고
    • Conclusions: The future of antimicrobial therapy— Augmentin and beyond
    • Ball, P. 2007. Conclusions: the future of antimicrobial therapy— Augmentin and beyond. Int J Antimicrob Agents 30(Suppl 2): S139–S141. https://doi.org/10.1016/j.ijantimicag.2007.08.016.
    • (2007) Int J Antimicrob Agents , vol.30 , pp. S139-S141
    • Ball, P.1
  • 125
    • 36148995090 scopus 로고    scopus 로고
    • Introduction: Historical perspective and development of amoxicillin/clavulanate
    • Geddes, AM, Klugman KP, Rolinson GN. 2007. Introduction: historical perspective and development of amoxicillin/clavulanate. Int J Antimicrob Agents 30(Suppl 2):S109–S112. https://doi.org/10.1016/j.ijantimicag.2007.07.015.
    • (2007) Int J Antimicrob Agents , vol.30 , pp. S109-S112
    • Geddes, A.M.1    Klugman, K.P.2    Rolinson, G.N.3
  • 126
    • 84976565309 scopus 로고    scopus 로고
    • Insights into newer antimicrobial agents against Gram-negative bacteria
    • Taneja, N, Kaur H. 2016. Insights into newer antimicrobial agents against Gram-negative bacteria. Microbiol Insights 9:9–19. https://doi.org/10.4137/MBI.S29459.
    • (2016) Microbiol Insights , vol.9 , pp. 9-19
    • Taneja, N.1    Kaur, H.2
  • 127
    • 85008400067 scopus 로고    scopus 로고
    • Antibiotics in the clinical pipeline at the end of 2015
    • Butler, MS, Blaskovich MA, Cooper MA. 2017. Antibiotics in the clinical pipeline at the end of 2015. J Antibiot (Tokyo) 70:3–24. https://doi.org/10.1038/ja.2016.72.
    • (2017) J Antibiot (Tokyo) , vol.70 , pp. 3-24
    • Butler, M.S.1    Blaskovich, M.A.2    Cooper, M.A.3
  • 128
    • 84918777580 scopus 로고    scopus 로고
    • Antibiotic adjuvants: Diverse strategies for controlling drug-resistant pathogens
    • Gill, EE, Franco OL, Hancock REW. 2015. Antibiotic adjuvants: diverse strategies for controlling drug-resistant pathogens. Chem Biol Drug Des 85:56–78. https://doi.org/10.1111/cbdd.12478.
    • (2015) Chem Biol Drug Des , vol.85 , pp. 56-78
    • Gill, E.E.1    Franco, O.L.2    Hancock, R.E.W.3
  • 129
    • 84867332729 scopus 로고    scopus 로고
    • Adjuvant strategies for potentiation of antibiotics to overcome antimicrobial resistance
    • Pieren, M, Tigges M. 2012. Adjuvant strategies for potentiation of antibiotics to overcome antimicrobial resistance. Curr Opin Pharmacol 12:551–555. https://doi.org/10.1016/j.coph.2012.07.005.
    • (2012) Curr Opin Pharmacol , vol.12 , pp. 551-555
    • Pieren, M.1    Tigges, M.2
  • 130
    • 79956079536 scopus 로고    scopus 로고
    • Antibiotic adjuvants: Multicomponent antiinfective strategies
    • Kalan, L, Wright GD. 2011. Antibiotic adjuvants: multicomponent antiinfective strategies. Expert Rev Mol Med 13:e5. https://doi.org/10.1017/S1462399410001766.
    • (2011) Expert Rev Mol Med , vol.13 , pp. 5
    • Kalan, L.1    Wright, G.D.2
  • 131
    • 0022623610 scopus 로고
    • Imipenem-cilastatin: The first thienamycin antibiotic
    • Jacobs, RF. 1986. Imipenem-cilastatin: the first thienamycin antibiotic. Pediatr Infect Dis 5:444–448. https://doi.org/10.1097/00006454-198607000-00015.
    • (1986) Pediatr Infect Dis , vol.5 , pp. 444-448
    • Jacobs, R.F.1
  • 132
    • 0026732827 scopus 로고
    • Inactivation of new carbapenem antibiotics by dehydropeptidase-I from porcine and human renal cortex
    • Hikida, M, Kawashima K, Yoshida M, Mitsuhashi S. 1992. Inactivation of new carbapenem antibiotics by dehydropeptidase-I from porcine and human renal cortex. J Antimicrob Chemother 30:129–134. https://doi.org/10.1093/jac/30.2.129.
    • (1992) J Antimicrob Chemother , vol.30 , pp. 129-134
    • Hikida, M.1    Kawashima, K.2    Yoshida, M.3    Mitsuhashi, S.4
  • 134
    • 84978763327 scopus 로고    scopus 로고
    • The antibiotic pipeline for multi-drug resistant gram negative bacteria: What can we expect?
    • Falagas, ME, Mavroudis AD, Vardakas KZ. 2016. The antibiotic pipeline for multi-drug resistant gram negative bacteria: what can we expect? Expert Rev Anti Infect Ther 14:747–763. https://doi.org/10.1080/14787210.2016.1204911.
    • (2016) Expert Rev Anti Infect Ther , vol.14 , pp. 747-763
    • Falagas, M.E.1    Mavroudis, A.D.2    Vardakas, K.Z.3
  • 139
    • 85007825474 scopus 로고
    • Aspergillomarasmine, A and B, potent microbial inhibitors of endothelin-converting enzyme
    • Arai, K, Ashikawa N, Nakakita Y, Matsuura A, Ashizawa N, Munekata M. 1993. Aspergillomarasmine, A and B, potent microbial inhibitors of endothelin-converting enzyme. Biosci Biotechnol Biochem 57: 1944–1945. https://doi.org/10.1271/bbb.57.1944.
    • (1993) Biosci Biotechnol Biochem , vol.57 , pp. 1944-1945
    • Arai, K.1    Ashikawa, N.2    Nakakita, Y.3    Matsuura, A.4    Ashizawa, N.5    Munekata, M.6
  • 141
    • 84903642660 scopus 로고    scopus 로고
    • A variety of roles for versatile zinc in metallo-beta-lactamases
    • Karsisiotis, AI, Damblon CF, Roberts GCK. 2014. A variety of roles for versatile zinc in metallo-beta-lactamases. Metallomics 6:1181–1197. https://doi.org/10.1039/C4MT00066H.
    • (2014) Metallomics , vol.6 , pp. 1181-1197
    • Karsisiotis, A.I.1    Damblon, C.F.2    Roberts, G.C.K.3
  • 142
    • 84946491160 scopus 로고    scopus 로고
    • Overcoming differences: The catalytic mechanism of metallo-beta-lactamases
    • Meini, M-R, Llarrull LI, Vila AJ. 2015. Overcoming differences: the catalytic mechanism of metallo-beta-lactamases. FEBS Lett 589:3419–3432. https://doi.org/10.1016/j.febslet.2015.08.015.
    • (2015) FEBS Lett , vol.589 , pp. 3419-3432
    • Meini, M.-R.1    Llarrull, L.I.2    Vila, A.J.3
  • 143
    • 84957845289 scopus 로고    scopus 로고
    • Total synthesis and activity of the metallo-beta-lactamase inhibitor aspergillomarasmine A
    • Koteva, K, King AM, Capretta A, Wright GD. 2016. Total synthesis and activity of the metallo-beta-lactamase inhibitor aspergillomarasmine A. Angew Chem Int Ed Engl 55:2210–2212. https://doi.org/10.1002/anie.201510057.
    • (2016) Angew Chem Int Ed Engl , vol.55 , pp. 2210-2212
    • Koteva, K.1    King, A.M.2    Capretta, A.3    Wright, G.D.4
  • 144
    • 84992093037 scopus 로고    scopus 로고
    • Total synthesis of aspergillomarasmine A and related compounds: A sulfamidate approach enables exploration of structure-activity relationships
    • Albu, SA, Koteva K, King AM, Al-Karmi S, Wright GD, Capretta A. 2016. Total synthesis of aspergillomarasmine A and related compounds: a sulfamidate approach enables exploration of structure-activity relationships. Angew Chem Int Ed Engl 55:13259–13262. https://doi.org/10.1002/anie.201606657.
    • (2016) Angew Chem Int Ed Engl , vol.55 , pp. 13259-13262
    • Albu, S.A.1    Koteva, K.2    King, A.M.3    Al-Karmi, S.4    Wright, G.D.5    Capretta, A.6
  • 145
    • 84947976362 scopus 로고    scopus 로고
    • Total synthesis and structural reassignment of aspergillomarasmine A
    • Liao, D, Yang S, Wang J, Zhang J, Hong B, Wu F, Lei X. 2016. Total synthesis and structural reassignment of aspergillomarasmine A. Angew Chem Int Ed Engl 55:4291–4295. https://doi.org/10.1002/anie.201509960.
    • (2016) Angew Chem Int Ed Engl , vol.55 , pp. 4291-4295
    • Liao, D.1    Yang, S.2    Wang, J.3    Zhang, J.4    Hong, B.5    Wu, F.6    Lei, X.7
  • 146
    • 84969597463 scopus 로고    scopus 로고
    • Treatment of Gramnegative bacterial infections by potentiation of antibiotics
    • Zabawa, TP, Pucci MJ, Parr TR, Jr, Lister T. 2016. Treatment of Gramnegative bacterial infections by potentiation of antibiotics. Curr Opin Microbiol 33:7–12. https://doi.org/10.1016/j.mib.2016.05.005.
    • (2016) Curr Opin Microbiol , vol.33 , pp. 7-12
    • Zabawa, T.P.1    Pucci, M.J.2    Parr, T.R.3    Lister, T.4
  • 150
    • 77955345521 scopus 로고    scopus 로고
    • A novel polymyxin derivative that lacks the fatty acid tail and carries only three positive charges has strong synergism with agents excluded by the intact outer membrane
    • Vaara, M, Siikanen O, Apajalahti J, Fox J, Frimodt-Moller N, He H, Poudyal A, Li J, Nation RL, Vaara T. 2010. A novel polymyxin derivative that lacks the fatty acid tail and carries only three positive charges has strong synergism with agents excluded by the intact outer membrane. Antimicrob Agents Chemother 54:3341–3346. https://doi.org/10.1128/AAC.01439-09.
    • (2010) Antimicrob Agents Chemother , vol.54 , pp. 3341-3346
    • Vaara, M.1    Siikanen, O.2    Apajalahti, J.3    Fox, J.4    Frimodt-Moller, N.5    He, H.6    Poudyal, A.7    Li, J.8    Nation, R.L.9    Vaara, T.10
  • 153
    • 85014283729 scopus 로고    scopus 로고
    • Nephrotoxicity of polymyxins: Is there any difference between colistimethate and polymyxin B?
    • Zavascki, AP, Nation RL. 2017. Nephrotoxicity of polymyxins: is there any difference between colistimethate and polymyxin B? Antimicrob Agents Chemother 61:e02319-16. https://doi.org/10.1128/AAC.02319-16.
    • (2017) Antimicrob Agents Chemother , vol.61 , pp. e02316-e02319
    • Zavascki, A.P.1    Nation, R.L.2
  • 154
    • 85000997421 scopus 로고    scopus 로고
    • Are there any ways around the exposure-limiting nephrotoxicity of the polymyxins?
    • Pogue, JM, Ortwine JK, Kaye KS. 2016. Are there any ways around the exposure-limiting nephrotoxicity of the polymyxins? Int J Antimicrob Agents 48:622–626. https://doi.org/10.1016/j.ijantimicag.2016.11.001.
    • (2016) Int J Antimicrob Agents , vol.48 , pp. 622-626
    • Pogue, J.M.1    Ortwine, J.K.2    Kaye, K.S.3
  • 156
    • 0035370341 scopus 로고    scopus 로고
    • Resistance to trimethoprim-sulfamethoxazole
    • Huovinen, P. 2001. Resistance to trimethoprim-sulfamethoxazole. Clin Infect Dis 32:1608–1614. https://doi.org/10.1086/320532.
    • (2001) Clin Infect Dis , vol.32 , pp. 1608-1614
    • Huovinen, P.1
  • 158
    • 57149110892 scopus 로고    scopus 로고
    • The management of acute uncomplicated cystitis in adult women by family physicians in Canada
    • McIsaac WJ, Prakash P, Ross S. 2008. The management of acute uncomplicated cystitis in adult women by family physicians in Canada. Can J Infect Dis Med Microbiol 19:287-293.
    • (2008) Can J Infect Dis Med Microbiol , vol.19 , pp. 287-293
    • McIsaac, W.J.1    Prakash, P.2    Ross, S.3
  • 159
    • 7044264386 scopus 로고    scopus 로고
    • Trimethoprim/sulfamethoxazole: Clinical update
    • Libecco, JA, Powell KR. 2004. Trimethoprim/sulfamethoxazole: clinical update. Pediatr Rev 25:375–380.
    • (2004) Pediatr Rev , vol.25 , pp. 375-380
    • Libecco, J.A.1    Powell, K.R.2
  • 160
    • 84896442957 scopus 로고    scopus 로고
    • Comparative in vitro activity of sulfametrole/trimethoprim and sulfamethoxazole/ trimethoprim and other agents against multiresistant Gram-negative bacteria
    • Livermore, DM, Mushtaq S, Warner M, Woodford N. 2014. Comparative in vitro activity of sulfametrole/trimethoprim and sulfamethoxazole/ trimethoprim and other agents against multiresistant Gram-negative bacteria. J Antimicrob Chemother 69:1050–1056. https://doi.org/10.1093/jac/dkt455.
    • (2014) J Antimicrob Chemother , vol.69 , pp. 1050-1056
    • Livermore, D.M.1    Mushtaq, S.2    Warner, M.3    Woodford, N.4
  • 161
    • 84893435097 scopus 로고    scopus 로고
    • Antibiotic treatment of infections due to carbapenem-resistant Enterobacteriaceae: Systematic evaluation of the available evidence
    • Falagas, ME, Lourida P, Poulikakos P, Rafailidis PI, Tansarli GS. 2014. Antibiotic treatment of infections due to carbapenem-resistant Enterobacteriaceae: systematic evaluation of the available evidence. Antimicrob Agents Chemother 58:654–663. https://doi.org/10.1128/AAC.01222-13.
    • (2014) Antimicrob Agents Chemother , vol.58 , pp. 654-663
    • Falagas, M.E.1    Lourida, P.2    Poulikakos, P.3    Rafailidis, P.I.4    Tansarli, G.S.5
  • 162
    • 84888075146 scopus 로고    scopus 로고
    • Combination therapy for Gram-negative bacteria: What is the evidence?
    • Kmeid, JG, Youssef MM, Kanafani ZA, Kanj SS. 2013. Combination therapy for Gram-negative bacteria: what is the evidence? Expert Rev Anti Infect Ther 11:1355–1362. https://doi.org/10.1586/14787210.2013.846215.
    • (2013) Expert Rev Anti Infect Ther , vol.11 , pp. 1355-1362
    • Kmeid, J.G.1    Youssef, M.M.2    Kanafani, Z.A.3    Kanj, S.S.4
  • 163
    • 79955484275 scopus 로고    scopus 로고
    • Clinical implications of beta-lactam-aminoglycoside synergism: Systematic review of randomised trials
    • Marcus, R, Paul M, Elphick H, Leibovici L. 2011. Clinical implications of beta-lactam-aminoglycoside synergism: systematic review of randomised trials. Int J Antimicrob Agents 37:491–503. https://doi.org/10.1016/j.ijantimicag.2010.11.029.
    • (2011) Int J Antimicrob Agents , vol.37 , pp. 491-503
    • Marcus, R.1    Paul, M.2    Elphick, H.3    Leibovici, L.4
  • 164
    • 84875371294 scopus 로고    scopus 로고
    • β-Lactam plus aminoglycoside or fluoroquinolone combination versus β-lactam monotherapy for Pseudomonas aeruginosa infections: A meta-analysis
    • Vardakas, KZ, Tansarli GS, Bliziotis IA, Falagas ME. 2013. β-Lactam plus aminoglycoside or fluoroquinolone combination versus β-lactam monotherapy for Pseudomonas aeruginosa infections: a meta-analysis. Int J Antimicrob Agents 41:301–310. https://doi.org/10.1016/j.ijantimicag.2012.12.006.
    • (2013) Int J Antimicrob Agents , vol.41 , pp. 301-310
    • Vardakas, K.Z.1    Tansarli, G.S.2    Bliziotis, I.A.3    Falagas, M.E.4
  • 165
    • 0042424618 scopus 로고    scopus 로고
    • Effectiveness of combination antimicrobial therapy for Pseudomonas aeruginosa bacteremia
    • Chamot, E, Boffi El Amari E, Rohner P, Van Delden C. 2003. Effectiveness of combination antimicrobial therapy for Pseudomonas aeruginosa bacteremia. Antimicrob Agents Chemother 47:2756–2764. https://doi.org/10.1128/AAC.47.9.2756-2764.2003.
    • (2003) Antimicrob Agents Chemother , vol.47 , pp. 2756-2764
    • Chamot, E.1    Boffi El Amari, E.2    Rohner, P.3    van Delden, C.4
  • 166
    • 33645155050 scopus 로고    scopus 로고
    • The role of aminoglycosides in combination with a beta-lactam for the treatment of bacterial endocarditis: A meta-analysis of comparative trials
    • Falagas, ME, Matthaiou DK, Bliziotis IA. 2006. The role of aminoglycosides in combination with a beta-lactam for the treatment of bacterial endocarditis: a meta-analysis of comparative trials. J Antimicrob Chemother 57:639–647. https://doi.org/10.1093/jac/dkl044.
    • (2006) J Antimicrob Chemother , vol.57 , pp. 639-647
    • Falagas, M.E.1    Matthaiou, D.K.2    Bliziotis, I.A.3
  • 167
    • 84911003547 scopus 로고    scopus 로고
    • Combination antibiotic treatment versus monotherapy for multidrug-resistant, extensively drug-resistant, and pandrug-resistant Acinetobacter infections: A systematic review
    • Poulikakos, P, Tansarli GS, Falagas ME. 2014. Combination antibiotic treatment versus monotherapy for multidrug-resistant, extensively drug-resistant, and pandrug-resistant Acinetobacter infections: a systematic review. Eur J Clin Microbiol Infect Dis 33:1675–1685. https://doi.org/10.1007/s10096-014-2124-9.
    • (2014) Eur J Clin Microbiol Infect Dis , vol.33 , pp. 1675-1685
    • Poulikakos, P.1    Tansarli, G.S.2    Falagas, M.E.3
  • 168
    • 84863654489 scopus 로고    scopus 로고
    • Combination therapy for treatment of infections with Gram-negative bacteria
    • Tamma, PD, Cosgrove SE, Maragakis LL. 2012. Combination therapy for treatment of infections with Gram-negative bacteria. Clin Microbiol Rev 25:450–470. https://doi.org/10.1128/CMR.05041-11.
    • (2012) Clin Microbiol Rev , vol.25 , pp. 450-470
    • Tamma, P.D.1    Cosgrove, S.E.2    Maragakis, L.L.3
  • 169
    • 0028216801 scopus 로고
    • Dual-action antibiotic hybrids
    • Hamilton-Miller JM. 1994. Dual-action antibiotic hybrids. J Antimicrob Chemother 33:197–200. https://doi.org/10.1093/jac/33.2.197.
    • (1994) J Antimicrob Chemother , vol.33 , pp. 197-200
    • Hamilton-Miller, J.M.1
  • 170
    • 34250160226 scopus 로고    scopus 로고
    • Dual action-based approaches to antibacterial agents
    • Bremner, JB, Ambrus JI, Samosorn S. 2007. Dual action-based approaches to antibacterial agents. Curr Med Chem 14:1459–1477. https://doi.org/10.2174/092986707780831168.
    • (2007) Curr Med Chem , vol.14 , pp. 1459-1477
    • Bremner, J.B.1    Ambrus, J.I.2    Samosorn, S.3
  • 172
    • 16244398678 scopus 로고    scopus 로고
    • Synthesis of beta-lactam nucleoside chimera via Kinugasa reaction and evaluation of their antibacterial activity
    • Basak, A, Pal R. 2005. Synthesis of beta-lactam nucleoside chimera via Kinugasa reaction and evaluation of their antibacterial activity. Bioorg Med Chem Lett 15:2015–2018. https://doi.org/10.1016/j.bmcl.2005.02.064.
    • (2005) Bioorg Med Chem Lett , vol.15 , pp. 2015-2018
    • Basak, A.1    Pal, R.2
  • 173
    • 84857175339 scopus 로고    scopus 로고
    • Antistaphylococcal activity of TD-1792, a multivalent glycopeptide-cephalosporin antibiotic
    • Blais, J, Lewis SR, Krause KM, Benton BM. 2012. Antistaphylococcal activity of TD-1792, a multivalent glycopeptide-cephalosporin antibiotic. Antimicrob Agents Chemother 56:1584–1587. https://doi.org/10.1128/AAC.05532-11.
    • (2012) Antimicrob Agents Chemother , vol.56 , pp. 1584-1587
    • Blais, J.1    Lewis, S.R.2    Krause, K.M.3    Benton, B.M.4
  • 177
    • 84872533264 scopus 로고    scopus 로고
    • Aminoglycoside antibiotics in the 21st century
    • Becker, B, Cooper MA. 2013. Aminoglycoside antibiotics in the 21st century. ACS Chem Biol 8:105–115. https://doi.org/10.1021/cb3005116.
    • (2013) ACS Chem Biol , vol.8 , pp. 105-115
    • Becker, B.1    Cooper, M.A.2
  • 178
    • 54849413943 scopus 로고    scopus 로고
    • How many modes of action should an antibiotic have?
    • Brötz-Oesterhelt H, Brunner NA. 2008. How many modes of action should an antibiotic have? Curr Opin Pharmacol 8:564–573. https://doi.org/10.1016/j.coph.2008.06.008.
    • (2008) Curr Opin Pharmacol , vol.8 , pp. 564-573
    • Brötz-Oesterhelt, H.1    Brunner, N.A.2
  • 179
    • 85026053788 scopus 로고    scopus 로고
    • Glossary of terms used in medicinal chemistry (IUPAC recommendations 1998)
    • Wermuth, CG, Ganellin CR, Lindberg P, Mitscher LA. 1998. Glossary of terms used in medicinal chemistry (IUPAC recommendations 1998). Pure Appl Chem 70:1129–1143. https://doi.org/10.1351/pac199870051129.
    • (1998) Pure Appl Chem , vol.70 , pp. 1129-1143
    • Wermuth, C.G.1    Ganellin, C.R.2    Lindberg, P.3    Mitscher, L.A.4
  • 180
    • 84922572705 scopus 로고    scopus 로고
    • Design of dual action antibiotics as an approach to search for new promising drugs
    • Preobrazhenskaya, AN, Olsufyeva EN, Preyobrazhenskaya MN. 2015. Design of dual action antibiotics as an approach to search for new promising drugs. Russ Chem Rev 84:61. https://doi.org/10.1070/RCR4448.
    • (2015) Russ Chem Rev , vol.84 , pp. 61
    • Preobrazhenskaya, A.N.1    Olsufyeva, E.N.2    Preyobrazhenskaya, M.N.3
  • 181
    • 84969617464 scopus 로고    scopus 로고
    • Analysis of past and present synthetic methodologies on medicinal chemistry: Where have all the new reactions gone?
    • Brown, DG, Bostrom J. 2016. Analysis of past and present synthetic methodologies on medicinal chemistry: where have all the new reactions gone? J Med Chem 59:4443–4458. https://doi.org/10.1021/acs.jmedchem.5b01409.
    • (2016) J Med Chem , vol.59 , pp. 4443-4458
    • Brown, D.G.1    Bostrom, J.2
  • 182
    • 0013797654 scopus 로고
    • Cephalosporinase and penicillinase activities of a beta-lactamase from Pseudomonas pyocyanea
    • Sabath, LD, Jago M, Abraham EP. 1965. Cephalosporinase and penicillinase activities of a beta-lactamase from Pseudomonas pyocyanea. Biochem J 96:739–752. https://doi.org/10.1042/bj0960739.
    • (1965) Biochem J , vol.96 , pp. 739-752
    • Sabath, L.D.1    Jago, M.2    Abraham, E.P.3
  • 183
    • 0030879493 scopus 로고    scopus 로고
    • Review dual β-lactam-fluoroquinolone compounds: A novel approach to antibacterial treatment
    • Bryskier, A. 1997. Review dual β-lactam-fluoroquinolone compounds: a novel approach to antibacterial treatment. Expert Opin Invest Drugs 6:1479–1499. https://doi.org/10.1517/13543784.6.10.1479.
    • (1997) Expert Opin Invest Drugs , vol.6 , pp. 1479-1499
    • Bryskier, A.1
  • 184
    • 0032582734 scopus 로고    scopus 로고
    • S-Aminosulfeniminopenicillins: Multimode β-lactamase inhibitors and template structures for penicillin-based β-lactamase substrates as prodrugs
    • Smyth, TP, O’Donnell ME, O’Connor MJ, St Ledger JO. 1998. S-Aminosulfeniminopenicillins: multimode β-lactamase inhibitors and template structures for penicillin-based β-lactamase substrates as prodrugs. J Org Chem 63:7600–7618. https://doi.org/10.1021/jo970737f.
    • (1998) J Org Chem , vol.63 , pp. 7600-7618
    • Smyth, T.P.1    O’Donnell, M.E.2    O’Connor, M.J.3    St Ledger, J.O.4
  • 185
    • 0034725844 scopus 로고    scopus 로고
    • β-Lactamase-dependent prodrugs—recent developments
    • Smyth, TP, O’Donnell ME, O’Connor MJ, St Ledger JO. 2000. β-Lactamase-dependent prodrugs—recent developments. Tetrahedron 56:5699–5707. https://doi.org/10.1016/S0040-4020(00)00419-1.
    • (2000) Tetrahedron , vol.56 , pp. 5699-5707
    • Smyth, T.P.1    O’Donnell, M.E.2    O’Connor, M.J.3    St Ledger, J.O.4
  • 186
    • 8644249388 scopus 로고    scopus 로고
    • Penicillins as beta-lactamase-dependent prodrugs: Enabling role of a vinyl ester exocyclic to the lactam ring
    • Ruddle, CC, Smyth TP. 2004. Penicillins as beta-lactamase-dependent prodrugs: enabling role of a vinyl ester exocyclic to the lactam ring. Chem Commun (Camb) 2004:2332–2333. https://doi.org/10.1039/B409517K.
    • (2004) Chem Commun (Camb) , vol.2004 , pp. 2332-2333
    • Ruddle, C.C.1    Smyth, T.P.2
  • 187
    • 62949166438 scopus 로고    scopus 로고
    • Metallo-beta-lactamases in Gram-negative bacteria: Introducing the era of pan-resistance?
    • Maltezou, HC. 2009. Metallo-beta-lactamases in Gram-negative bacteria: introducing the era of pan-resistance? Int J Antimicrob Agents 33: 405.e1–405.e7. https://doi.org/10.1016/j.ijantimicag.2008.09.003.
    • (2009) Int J Antimicrob Agents , vol.33
    • Maltezou, H.C.1
  • 190
    • 0023648350 scopus 로고
    • Inactivation of alanine racemase by beta-chloro-L-alanine released enzymatically from amino acid and peptide C10-esters of deacetylcephalothin
    • Mobashery, S, Johnston M. 1987. Inactivation of alanine racemase by beta-chloro-L-alanine released enzymatically from amino acid and peptide C10-esters of deacetylcephalothin. Biochemistry 26:5878–5884. https://doi.org/10.1021/bi00392a045.
    • (1987) Biochemistry , vol.26 , pp. 5878-5884
    • Mobashery, S.1    Johnston, M.2
  • 191
    • 0022618178 scopus 로고
    • Conscripting betalactamase for use in drug delivery Synthesis and biological activity of a cephalosporin C10-ester of an antibiotic dipeptide
    • Mobashery, S, Lerner SA, Johnston M. 1986. Conscripting betalactamase for use in drug delivery Synthesis and biological activity of a cephalosporin C10-ester of an antibiotic dipeptide. J Am Chem Soc 108:1685–1686. https://doi.org/10.1021/ja00267a045.
    • (1986) J am Chem Soc , vol.108 , pp. 1685-1686
    • Mobashery, S.1    Lerner, S.A.2    Johnston, M.3
  • 194
    • 0024389916 scopus 로고
    • Antimicrobial activity of Ro 23-9424, a novel ester-linked codrug of fleroxacin and desacetylcefotaxime
    • Jones, RN, Barry AL, Thornsberry C. 1989. Antimicrobial activity of Ro 23-9424, a novel ester-linked codrug of fleroxacin and desacetylcefotaxime. Antimicrob Agents Chemother 33:944–950. https://doi.org/10.1128/AAC.33.6.944.
    • (1989) Antimicrob Agents Chemother , vol.33 , pp. 944-950
    • Jones, R.N.1    Barry, A.L.2    Thornsberry, C.3
  • 196
    • 0025754851 scopus 로고
    • Escherichia coli resistant to cephalosporins and quinolones is still susceptible to the cephalosporin-quinolone ester Ro 23-9424. Antimicrob
    • Pace, J, Bertasso A, Georgopapadakou NH. 1991. Escherichia coli resistant to cephalosporins and quinolones is still susceptible to the cephalosporin-quinolone ester Ro 23-9424. Antimicrob, Agents Chemother 35:910–915. https://doi.org/10.1128/AAC.35.5.910.
    • (1991) Agents Chemother , vol.35 , pp. 910-915
    • Pace, J.1    Bertasso, A.2    Georgopapadakou, N.H.3
  • 197
    • 0027415754 scopus 로고
    • Mechanisms of action of cephalosporin 3=-quinolone esters, carbamates, and tertiary amines in Escherichia coli
    • Georgopapadakou, NH, Bertasso A. 1993. Mechanisms of action of cephalosporin 3=-quinolone esters, carbamates, and tertiary amines in Escherichia coli. Antimicrob Agents Chemother 37:559–565. https://doi.org/10.1128/AAC.37.3.559.
    • (1993) Antimicrob Agents Chemother , vol.37 , pp. 559-565
    • Georgopapadakou, N.H.1    Bertasso, A.2
  • 199
    • 0029803606 scopus 로고    scopus 로고
    • Effect of RO 23-9424, cefotaxime and fleroxacin on functions of human polymorphonuclear cells and cytokine production by human monocytes
    • Matera, G, Berlinghieri MC, Foti F, Barreca GS, Foca A. 1996. Effect of RO 23-9424, cefotaxime and fleroxacin on functions of human polymorphonuclear cells and cytokine production by human monocytes. J Antimicrob Chemother 38:799–807. https://doi.org/10.1093/jac/38.5.799.
    • (1996) J Antimicrob Chemother , vol.38 , pp. 799-807
    • Matera, G.1    Berlinghieri, M.C.2    Foti, F.3    Barreca, G.S.4    Foca, A.5
  • 201
    • 0025062589 scopus 로고
    • In vivo evaluation of a dual-action antibacterial, Ro 23-9424, compared to cefotaxime and fleroxacin
    • Beskid, G, Siebelist J, McGarry CM, Cleeland R, Chan K, Keith DD. 1990. In vivo evaluation of a dual-action antibacterial, Ro 23-9424, compared to cefotaxime and fleroxacin. Chemotherapy 36:109–116. https://doi.org/10.1159/000238756.
    • (1990) Chemotherapy , vol.36 , pp. 109-116
    • Beskid, G.1    Siebelist, J.2    McGarry, C.M.3    Cleeland, R.4    Chan, K.5    Keith, D.D.6
  • 202
    • 33845894155 scopus 로고    scopus 로고
    • Multi-targeting by monotherapeutic antibacterials
    • Silver, LL. 2007. Multi-targeting by monotherapeutic antibacterials. Nat Rev Drug Discov 6:41–55. https://doi.org/10.1038/nrd2202.
    • (2007) Nat Rev Drug Discov , vol.6 , pp. 41-55
    • Silver, L.L.1
  • 203
    • 0025159235 scopus 로고
    • In vitro activity of Ro 23-9424, a dual-action cephalosporin, compared with activities of other antibiotics
    • Gu, JW, Neu HC. 1990. In vitro activity of Ro 23-9424, a dual-action cephalosporin, compared with activities of other antibiotics. Antimicrob Agents Chemother 34:189–195. https://doi.org/10.1128/AAC.34.2.189.
    • (1990) Antimicrob Agents Chemother , vol.34 , pp. 189-195
    • Gu, J.W.1    Neu, H.C.2
  • 204
    • 0028852737 scopus 로고
    • Low-level resistance to the cephalosporin 3=-quinolone ester Ro 23- 9424 in Escherichia coli
    • Chapman, JS, Bertasso A, Cummings LM, Georgopapadakou NH. 1995. Low-level resistance to the cephalosporin 3=-quinolone ester Ro 23- 9424 in Escherichia coli. Antimicrob Agents Chemother 39:564–566. https://doi.org/10.1128/AAC.39.2.564.
    • (1995) Antimicrob Agents Chemother , vol.39 , pp. 564-566
    • Chapman, J.S.1    Bertasso, A.2    Cummings, L.M.3    Georgopapadakou, N.H.4
  • 205
    • 0028217509 scopus 로고
    • Quinolone resistance mediated by norA: Physiologic characterization and relationship to flqB, a quinolone resistance locus on the Staphylococcus aureus chromosome
    • Ng, EY, Trucksis M, Hooper DC. 1994. Quinolone resistance mediated by norA: physiologic characterization and relationship to flqB, a quinolone resistance locus on the Staphylococcus aureus chromosome. Antimicrob Agents Chemother 38:1345–1355. https://doi.org/10.1128/AAC.38.6.1345.
    • (1994) Antimicrob Agents Chemother , vol.38 , pp. 1345-1355
    • Ng, E.Y.1    Trucksis, M.2    Hooper, D.C.3
  • 206
    • 84880996459 scopus 로고    scopus 로고
    • Speculative strategies for new antibacterials: All roads should not lead to Rome
    • Shapiro, S. 2013. Speculative strategies for new antibacterials: all roads should not lead to Rome. J Antibiot (Tokyo) 66:371–386. https://doi.org/10.1038/ja.2013.27.
    • (2013) J Antibiot (Tokyo) , vol.66 , pp. 371-386
    • Shapiro, S.1
  • 207
    • 77955967899 scopus 로고    scopus 로고
    • Dual-acting hybrid antibiotics: A promising strategy to combat bacterial resistance
    • Pokrovskaya, V, Baasov T. 2010. Dual-acting hybrid antibiotics: a promising strategy to combat bacterial resistance. Expert Opin Drug Discov 5:883–902. https://doi.org/10.1517/17460441.2010.508069.
    • (2010) Expert Opin Drug Discov , vol.5 , pp. 883-902
    • Pokrovskaya, V.1    Baasov, T.2
  • 208
    • 84904717224 scopus 로고    scopus 로고
    • Third and fourth generation fluoroquinolone antibacterials: A systematic review of safety and toxicity profiles
    • Sousa, J, Alves G, Fortuna A, Falcao A. 2014. Third and fourth generation fluoroquinolone antibacterials: a systematic review of safety and toxicity profiles. Curr Drug Saf 9:89–105. https://doi.org/10.2174/1574886308666140106154754.
    • (2014) Curr Drug Saf , vol.9 , pp. 89-105
    • Sousa, J.1    Alves, G.2    Fortuna, A.3    Falcao, A.4
  • 209
    • 74949119366 scopus 로고    scopus 로고
    • Fluoroquinolone antibacterials: A review on chemistry, microbiology and therapeutic prospects
    • Sharma, PC, Jain A, Jain S. 2009. Fluoroquinolone antibacterials: a review on chemistry, microbiology and therapeutic prospects. Acta Pol Pharm 66:587–604.
    • (2009) Acta Pol Pharm , vol.66 , pp. 587-604
    • Sharma, P.C.1    Jain, A.2    Jain, S.3
  • 213
    • 80053179826 scopus 로고    scopus 로고
    • Tyrosyl-tRNA synthetase inhibitors as antibacterial agents: Synthesis, molecular docking and structure-activity relationship analysis of 3-aryl-4-arylaminofuran-2(5H)-ones
    • Xiao, Z-P, Ma T-W, Liao M-L, Feng Y-T, Peng X-C, Li J-L, Li Z-P, Wu Y, Luo Q, Deng Y, Liang X, Zhu H-L. 2011. Tyrosyl-tRNA synthetase inhibitors as antibacterial agents: synthesis, molecular docking and structure-activity relationship analysis of 3-aryl-4-arylaminofuran-2(5H)-ones. Eur J Med Chem 46:4904–4914. https://doi.org/10.1016/j.ejmech.2011.07.047.
    • (2011) Eur J Med Chem , vol.46 , pp. 4904-4914
    • Xiao, Z.-P.1    Ma, T.-W.2    Liao, M.-L.3    Feng, Y.-T.4    Peng, X.-C.5    Li, J.-L.6    Li, Z.-P.7    Wu, Y.8    Luo, Q.9    Deng, Y.10    Liang, X.11    Zhu, H.-L.12
  • 215
    • 26944434828 scopus 로고    scopus 로고
    • Antimicrobial activity of flavonoids
    • Cushnie, TPT, Lamb AJ. 2005. Antimicrobial activity of flavonoids. Int J Antimicrob Agents 26:343–356. https://doi.org/10.1016/j.ijantimicag.2005.09.002.
    • (2005) Int J Antimicrob Agents , vol.26 , pp. 343-356
    • Cushnie, T.1    Lamb, A.J.2
  • 217
    • 80051693256 scopus 로고    scopus 로고
    • Antibacterial activity of naringin derivatives against pathogenic strains
    • Céliz, G, Daz M, Audisio MC. 2011. Antibacterial activity of naringin derivatives against pathogenic strains. J Appl Microbiol 111:731–738. https://doi.org/10.1111/j.1365-2672.2011.05070.x.
    • (2011) J Appl Microbiol , vol.111 , pp. 731-738
    • Céliz, G.1    Daz, M.2    Audisio, M.C.3
  • 218
    • 61449258247 scopus 로고    scopus 로고
    • Naringenin enhances the anti-tumor effect of doxorubicin through selectively inhibiting the activity of multidrug resistance-associated proteins but not P-glycoprotein
    • Zhang, FY, Du GJ, Zhang L, Zhang CL, Lu WL, Liang W. 2009. Naringenin enhances the anti-tumor effect of doxorubicin through selectively inhibiting the activity of multidrug resistance-associated proteins but not P-glycoprotein. Pharm Res 26:914–925. https://doi.org/10.1007/s11095-008-9793-y.
    • (2009) Pharm Res , vol.26 , pp. 914-925
    • Zhang, F.Y.1    Du, G.J.2    Zhang, L.3    Zhang, C.L.4    Lu, W.L.5    Liang, W.6
  • 219
    • 65249160092 scopus 로고    scopus 로고
    • Design, synthesis, and evaluation of novel fluoroquinoloneaminoglycoside hybrid antibiotics
    • Pokrovskaya, V, Belakhov V, Hainrichson M, Yaron S, Baasov T. 2009. Design, synthesis, and evaluation of novel fluoroquinoloneaminoglycoside hybrid antibiotics. J Med Chem 52:2243–2254. https://doi.org/10.1021/jm900028n.
    • (2009) J Med Chem , vol.52 , pp. 2243-2254
    • Pokrovskaya, V.1    Belakhov, V.2    Hainrichson, M.3    Yaron, S.4    Baasov, T.5
  • 221
    • 84964642773 scopus 로고    scopus 로고
    • A hybrid drug limits resistance by evading the action of the multiple antibiotic resistance pathway
    • Wang, KK, Stone LK, Lieberman TD, Shavit M, Baasov T, Kishony R. 2015. A hybrid drug limits resistance by evading the action of the multiple antibiotic resistance pathway. Mol Biol Evol 33:492–500. https://doi.org/10.1093/molbev/msv243.
    • (2015) Mol Biol Evol , vol.33 , pp. 492-500
    • Wang, K.K.1    Stone, L.K.2    Lieberman, T.D.3    Shavit, M.4    Baasov, T.5    Kishony, R.6
  • 222
    • 84879030355 scopus 로고    scopus 로고
    • Synthesis and bioactive evaluation of novel hybrids of metronidazole and berberine as new type of antimicrobial agents and their transportation behavior by human serum albumin
    • Zhang, L, Chang J-J, Zhang S-L, Damu GLV, Geng R-X, Zhou C-H. 2013. Synthesis and bioactive evaluation of novel hybrids of metronidazole and berberine as new type of antimicrobial agents and their transportation behavior by human serum albumin. Bioorg Med Chem 21: 4158–4169. https://doi.org/10.1016/j.bmc.2013.05.007.
    • (2013) Bioorg Med Chem , vol.21 , pp. 4158-4169
    • Zhang, L.1    Chang, J.-J.2    Zhang, S.-L.3    Damu, G.4    Geng, R.-X.5    Zhou, C.-H.6
  • 223
    • 84976496761 scopus 로고    scopus 로고
    • Design, synthesis and biological evaluation of berberine-benzimidazole hybrids as new type of potentially DNA-targeting antimicrobial agents
    • Jeyakkumar, P, Zhang L, Avula SR, Zhou C-H. 2016. Design, synthesis and biological evaluation of berberine-benzimidazole hybrids as new type of potentially DNA-targeting antimicrobial agents. Eur J Med Chem 122:205–215. https://doi.org/10.1016/j.ejmech.2016.06.031.
    • (2016) Eur J Med Chem , vol.122 , pp. 205-215
    • Jeyakkumar, P.1    Zhang, L.2    Avula, S.R.3    Zhou, C.-H.4
  • 224
  • 225
    • 84953338655 scopus 로고    scopus 로고
    • Synthesis of tridecaptin-antibiotic conjugates with in vivo activity against Gramnegative bacteria
    • Cochrane, SA, Li X, He S, Yu M, Wu M, Vederas JC. 2015. Synthesis of tridecaptin-antibiotic conjugates with in vivo activity against Gramnegative bacteria. J Med Chem 58:9779–9785. https://doi.org/10.1021/acs.jmedchem.5b01578.
    • (2015) J Med Chem , vol.58 , pp. 9779-9785
    • Cochrane, S.A.1    Li, X.2    He, S.3    Yu, M.4    Wu, M.5    Vederas, J.C.6
  • 226
    • 84912077873 scopus 로고    scopus 로고
    • Unacylated tridecaptin A(1) acts as an effective sensitiser of Gram-negative bacteria to other antibiotics
    • Cochrane, SA, Vederas JC. 2014. Unacylated tridecaptin A(1) acts as an effective sensitiser of Gram-negative bacteria to other antibiotics. Int J Antimicrob Agents 44:493–499. https://doi.org/10.1016/j.ijantimicag.2014.08.008.
    • (2014) Int J Antimicrob Agents , vol.44 , pp. 493-499
    • Cochrane, S.A.1    Vederas, J.C.2
  • 227
    • 84894051365 scopus 로고    scopus 로고
    • Synthesis and structure-activity relationship studies of N-terminal analogues of the antimicrobial peptide tridecaptin A(1)
    • Cochrane, SA, Lohans CT, Brandelli JR, Mulvey G, Armstrong GD, Vederas JC. 2014. Synthesis and structure-activity relationship studies of N-terminal analogues of the antimicrobial peptide tridecaptin A(1). J Med Chem 57:1127–1131. https://doi.org/10.1021/jm401779d.
    • (2014) J Med Chem , vol.57 , pp. 1127-1131
    • Cochrane, S.A.1    Lohans, C.T.2    Brandelli, J.R.3    Mulvey, G.4    Armstrong, G.D.5    Vederas, J.C.6
  • 228
    • 84867155796 scopus 로고    scopus 로고
    • Structure, function and binding selectivity and stereoselectivity of siderophore-iron outer membrane transporters
    • Schalk, IJ, Mislin GLA, Brillet K. 2012. Structure, function and binding selectivity and stereoselectivity of siderophore-iron outer membrane transporters. Curr Top Membr 69:37–66. https://doi.org/10.1016/B978-0-12-394390-3.00002-1.
    • (2012) Curr Top Membr , vol.69 , pp. 37-66
    • Schalk, I.J.1    Mislin, G.2    Brillet, K.3
  • 230
    • 34548739613 scopus 로고    scopus 로고
    • Siderophore-based iron acquisition and pathogen control
    • Miethke, M, Marahiel MA. 2007. Siderophore-based iron acquisition and pathogen control. Microbiol Mol Biol Rev 71:413–451. https://doi.org/10.1128/MMBR.00012-07.
    • (2007) Microbiol Mol Biol Rev , vol.71 , pp. 413-451
    • Miethke, M.1    Marahiel, M.A.2
  • 231
    • 9244234392 scopus 로고    scopus 로고
    • Bacterial iron sources: From siderophores to hemophores
    • Wandersman, C, Delepelaire P. 2004. Bacterial iron sources: from siderophores to hemophores. Annu Rev Microbiol 58:611–647. https://doi.org/10.1146/annurev.micro.58.030603.123811.
    • (2004) Annu Rev Microbiol , vol.58 , pp. 611-647
    • Wandersman, C.1    Delepelaire, P.2
  • 232
    • 84973641976 scopus 로고    scopus 로고
    • Urinary concentrations and antibacterial activity of BAL30072, a novel siderophore monosulfactam, against uropathogens after intravenous administration in healthy subjects
    • Straubinger, M, Blenk H, Naber KG, Wagenlehner FME. 2016. Urinary concentrations and antibacterial activity of BAL30072, a novel siderophore monosulfactam, against uropathogens after intravenous administration in healthy subjects. Antimicrob Agents Chemother 60: 3309–3315. https://doi.org/10.1128/AAC.02425-15.
    • (2016) Antimicrob Agents Chemother , vol.60 , pp. 3309-3315
    • Straubinger, M.1    Blenk, H.2    Naber, K.G.3    Wagenlehner, F.M.E.4
  • 234
    • 84928895631 scopus 로고    scopus 로고
    • Potentiation of antibacterial activity of the MB-1 siderophore-monobactam conjugate using an efflux pump inhibitor
    • Tomaras, AP, Crandon JL, McPherson CJ, Nicolau DP. 2015. Potentiation of antibacterial activity of the MB-1 siderophore-monobactam conjugate using an efflux pump inhibitor. Antimicrob Agents Chemother 59:2439–2442. https://doi.org/10.1128/AAC.04172-14.
    • (2015) Antimicrob Agents Chemother , vol.59 , pp. 2439-2442
    • Tomaras, A.P.1    Crandon, J.L.2    McPherson, C.J.3    Nicolau, D.P.4
  • 236
    • 0026068667 scopus 로고
    • In vitro antibacterial activity of KP-736, a new cephem antibiotic
    • Maejima, T, Inoue M, Mitsuhashi S. 1991. In vitro antibacterial activity of KP-736, a new cephem antibiotic. Antimicrob Agents Chemother 35: 104–110. https://doi.org/10.1128/AAC.35.1.104.
    • (1991) Antimicrob Agents Chemother , vol.35 , pp. 104-110
    • Maejima, T.1    Inoue, M.2    Mitsuhashi, S.3
  • 237
    • 84957895337 scopus 로고    scopus 로고
    • In vitro antimicrobial activity of a siderophore cephalosporin, S-649266, against Enterobacteriaceae clinical isolates, including carbapenem-resistant strains
    • Kohira, N, West J, Ito A, Ito-Horiyama T, Nakamura R, Sato T, Rittenhouse S, Tsuji M, Yamano Y. 2016. In vitro antimicrobial activity of a siderophore cephalosporin, S-649266, against Enterobacteriaceae clinical isolates, including carbapenem-resistant strains. Antimicrob Agents Chemother 60:729–734. https://doi.org/10.1128/AAC.01695-15.
    • (2016) Antimicrob Agents Chemother , vol.60 , pp. 729-734
    • Kohira, N.1    West, J.2    Ito, A.3    Ito-Horiyama, T.4    Nakamura, R.5    Sato, T.6    Rittenhouse, S.7    Tsuji, M.8    Yamano, Y.9
  • 239
    • 85032468440 scopus 로고    scopus 로고
    • Efficacy of humanized exposures of cefiderocol (S-649266) against a diverse population of Gram-negative bacteria in a murine thigh infection model
    • Monogue, ML, Tsuji M, Yamano Y, Echols R, Nicolau DP. 2017. Efficacy of humanized exposures of cefiderocol (S-649266) against a diverse population of Gram-negative bacteria in a murine thigh infection model. Antimicrob Agents Chemother 61:e01022-17. https://doi.org/10.1128/AAC.01022-17.
    • (2017) Antimicrob Agents Chemother , vol.61 , pp. e01017-e01022
    • Monogue, M.L.1    Tsuji, M.2    Yamano, Y.3    Echols, R.4    Nicolau, D.P.5
  • 241
    • 84996478385 scopus 로고    scopus 로고
    • Siderophore cephalosporin cefiderocol utilizes ferric iron transporter systems for antibacterial activity against Pseudomonas aeruginosa
    • Ito, A, Nishikawa T, Matsumoto S, Yoshizawa H, Sato T, Nakamura R, Tsuji M, Yamano Y. 2016. Siderophore cephalosporin cefiderocol utilizes ferric iron transporter systems for antibacterial activity against Pseudomonas aeruginosa. Antimicrob Agents Chemother 60:7396–7401.
    • (2016) Antimicrob Agents Chemother , vol.60 , pp. 7396-7401
    • Ito, A.1    Nishikawa, T.2    Matsumoto, S.3    Yoshizawa, H.4    Sato, T.5    Nakamura, R.6    Tsuji, M.7    Yamano, Y.8
  • 242
    • 85028348466 scopus 로고    scopus 로고
    • S-649266, a novel siderophore cephalosporin: In vivo efficacy in murine infection model caused by multidrug-resistant Gram-negative bacteria, abstr P0253
    • Tsuji, M, Horiyama T, Toba S, Nakamura R, Yamano Y. 2015. S-649266, a novel siderophore cephalosporin: in vivo efficacy in murine infection model caused by multidrug-resistant Gram-negative bacteria, abstr P0253. Abstr 25th Eur Congr Clin Microbiol Infect Dis.
    • (2015) Abstr 25Th Eur Congr Clin Microbiol Infect Dis
    • Tsuji, M.1    Horiyama, T.2    Toba, S.3    Nakamura, R.4    Yamano, Y.5
  • 243
    • 85005784390 scopus 로고    scopus 로고
    • Cefiderocol, a siderophore cephalosporin for Gram-negative bacterial infections: Pharmacokinetics and safety in subjects with renal impairment
    • Katsube, T, Echols R, Arjona Ferreira JC, Krenz HK, Berg JK, Galloway C. 2017. Cefiderocol, a siderophore cephalosporin for Gram-negative bacterial infections: pharmacokinetics and safety in subjects with renal impairment. J Clin Pharmacol 57:584–591. https://doi.org/10.1002/jcph.841.
    • (2017) J Clin Pharmacol , vol.57 , pp. 584-591
    • Katsube, T.1    Echols, R.2    Arjona Ferreira, J.C.3    Krenz, H.K.4    Berg, J.K.5    Galloway, C.6
  • 244
    • 85009274824 scopus 로고    scopus 로고
    • Pharmacokinetic/pharmacodynamic modeling and simulation of cefiderocol, a parenteral siderophore cephalosporin, for dose adjustment based on renal function
    • Katsube, T, Wajima T, Ishibashi T, Arjona Ferreira JC, Echols R. 2017. Pharmacokinetic/pharmacodynamic modeling and simulation of cefiderocol, a parenteral siderophore cephalosporin, for dose adjustment based on renal function. Antimicrob Agents Chemother 61: e01381-16. https://doi.org/10.1128/AAC.01381-16.
    • (2017) Antimicrob Agents Chemother , vol.61 , pp. e01316-e01381
    • Katsube, T.1    Wajima, T.2    Ishibashi, T.3    Arjona Ferreira, J.C.4    Echols, R.5
  • 245
    • 85022194756 scopus 로고    scopus 로고
    • Bifunctional antimicrobial conjugates and hybrid antimicrobials
    • Klahn, P, Bronstrup M. 2017. Bifunctional antimicrobial conjugates and hybrid antimicrobials. Nat Prod Rep 34:832–885. https://doi.org/10.1039/C7NP00006E.
    • (2017) Nat Prod Rep , vol.34 , pp. 832-885
    • Klahn, P.1    Bronstrup, M.2
  • 246
    • 84975217786 scopus 로고    scopus 로고
    • Hybrid antibiotics— clinical progress and novel designs
    • Parkes, AL, Yule IA. 2016. Hybrid antibiotics— clinical progress and novel designs. Expert Opin Drug Discov 11:665–680. https://doi.org/10.1080/17460441.2016.1187597.
    • (2016) Expert Opin Drug Discov , vol.11 , pp. 665-680
    • Parkes, A.L.1    Yule, I.A.2
  • 247
    • 84958760300 scopus 로고    scopus 로고
    • Adjuvants based on hybrid antibiotics overcome resistance in Pseudomonas aeruginosa and enhance fluoroquinolone efficacy
    • Gorityala, BK, Guchhait G, Fernando DM, Deo S, McKenna SA, Zhanel GG, Kumar A, Schweizer F. 2016. Adjuvants based on hybrid antibiotics overcome resistance in Pseudomonas aeruginosa and enhance fluoroquinolone efficacy. Angew Chem Int Ed Engl 55:555–559. https://doi.org/10.1002/anie.201508330.
    • (2016) Angew Chem Int Ed Engl , vol.55 , pp. 555-559
    • Gorityala, B.K.1    Guchhait, G.2    Fernando, D.M.3    Deo, S.4    McKenna, S.A.5    Zhanel, G.G.6    Kumar, A.7    Schweizer, F.8
  • 248
    • 84988701271 scopus 로고    scopus 로고
    • Hybrid antibiotic overcomes resistance in P. Aeruginosa by enhancing outer membrane penetration and reducing efflux
    • Gorityala, BK, Guchhait G, Goswami S, Fernando DM, Kumar A, Zhanel GG, Schweizer F. 2016. Hybrid antibiotic overcomes resistance in P. aeruginosa by enhancing outer membrane penetration and reducing efflux. J Med Chem 59:8441–8455. https://doi.org/10.1021/acs.jmedchem.6b00867.
    • (2016) J Med Chem , vol.59 , pp. 8441-8455
    • Gorityala, B.K.1    Guchhait, G.2    Goswami, S.3    Fernando, D.M.4    Kumar, A.5    Zhanel, G.G.6    Schweizer, F.7
  • 249
    • 85019215287 scopus 로고    scopus 로고
    • A tobramycin vector enhances synergy and efficacy of efflux pump inhibitors against multidrug-resistant Gramnegative bacteria
    • Yang, X, Goswami S, Gorityala BK, Domalaon R, Lyu Y, Kumar A, Zhanel GG, Schweizer F. 2017. A tobramycin vector enhances synergy and efficacy of efflux pump inhibitors against multidrug-resistant Gramnegative bacteria. J Med Chem 60:3913–3932. https://doi.org/10.1021/acs.jmedchem.7b00156.
    • (2017) J Med Chem , vol.60 , pp. 3913-3932
    • Yang, X.1    Goswami, S.2    Gorityala, B.K.3    Domalaon, R.4    Lyu, Y.5    Kumar, A.6    Zhanel, G.G.7    Schweizer, F.8
  • 250
    • 85019177780 scopus 로고    scopus 로고
    • Amphiphilic tobramycin-lysine conjugates sensitize multidrug resistant Gram-negative bacteria to rifampicin and minocycline
    • Lyu, Y, Yang X, Goswami S, Gorityala BK, Idowu T, Domalaon R, Zhanel GG, Shan A, Schweizer F. 2017. Amphiphilic tobramycin-lysine conjugates sensitize multidrug resistant Gram-negative bacteria to rifampicin and minocycline. J Med Chem 60:3684–3702. https://doi.org/10.1021/acs.jmedchem.6b01742.
    • (2017) J Med Chem , vol.60 , pp. 3684-3702
    • Lyu, Y.1    Yang, X.2    Goswami, S.3    Gorityala, B.K.4    Idowu, T.5    Domalaon, R.6    Zhanel, G.G.7    Shan, A.8    Schweizer, F.9
  • 251
    • 85108124509 scopus 로고    scopus 로고
    • Amphiphilic lysine conjugated to tobramycin synergizes legacy antibiotics against wild-type and multidrug-resistant Pseudomonas aeruginosa
    • December
    • Lyu, Y, Domalaon R, Yang X, Schweizer F. 4 December 2017. Amphiphilic lysine conjugated to tobramycin synergizes legacy antibiotics against wild-type and multidrug-resistant Pseudomonas aeruginosa. Biopolymers https://doi.org/10.1002/bip.23091.
    • (2017) Biopolymers , vol.4
    • Lyu, Y.1    Domalaon, R.2    Yang, X.3    Schweizer, F.4
  • 252
    • 85033796462 scopus 로고    scopus 로고
    • Polymyxin, B3-tobramycin hybrids with Pseudomonas aeruginosa-selective antibacterial activity and strong potentiation of rifampicin, minocycline, and vancomycin
    • Domalaon, R, Yang X, Lyu Y, Zhanel GG, Schweizer F. 2017. Polymyxin, B3-tobramycin hybrids with Pseudomonas aeruginosa-selective antibacterial activity and strong potentiation of rifampicin, minocycline, and vancomycin. ACS Infect Dis 3:941–954. https://doi.org/10.1021/acsinfecdis.7b00145.
    • (2017) ACS Infect Dis , vol.3 , pp. 941-954
    • Domalaon, R.1    Yang, X.2    Lyu, Y.3    Zhanel, G.G.4    Schweizer, F.5
  • 253
    • 0038601510 scopus 로고    scopus 로고
    • Synergy, antagonism, and what the chequerboard puts between them
    • Odds, FC. 2003. Synergy, antagonism, and what the chequerboard puts between them. J Antimicrob Chemother 52:1. https://doi.org/10.1093/jac/dkg301.
    • (2003) J Antimicrob Chemother , vol.52 , pp. 1
    • Odds, F.C.1
  • 254
    • 84962061598 scopus 로고    scopus 로고
    • Breaking the spell: Combating multidrug resistant “superbugs
    • Khan, SN, Khan AU. 2016. Breaking the spell: combating multidrug resistant “superbugs.” Front Microbiol 7:174. https://doi.org/10.3389/fmicb.2016.00174.
    • (2016) Front Microbiol , vol.7 , pp. 174
    • Khan, S.N.1    Khan, A.U.2
  • 255
    • 84995446644 scopus 로고    scopus 로고
    • Multidrug-resistant Gramnegative bacilli: Infection control implications
    • Adler, A, Friedman ND, Marchaim D. 2016. Multidrug-resistant Gramnegative bacilli: infection control implications. Infect Dis Clin North Am 30:967–997. https://doi.org/10.1016/j.idc.2016.08.001.
    • (2016) Infect Dis Clin North Am , vol.30 , pp. 967-997
    • Adler, A.1    Friedman, N.D.2    Marchaim, D.3
  • 256
    • 77950391683 scopus 로고    scopus 로고
    • Aminoglycoside activity observed on single pre-translocation ribosome complexes
    • Feldman, MB, Terry DS, Altman RB, Blanchard SC. 2010. Aminoglycoside activity observed on single pre-translocation ribosome complexes. Nat Chem Biol 6:54–62. https://doi.org/10.1038/nchembio.274.
    • (2010) Nat Chem Biol , vol.6 , pp. 54-62
    • Feldman, M.B.1    Terry, D.S.2    Altman, R.B.3    Blanchard, S.C.4
  • 257
    • 0242609342 scopus 로고    scopus 로고
    • Inhaled tobramycin (TOBI): A review of its use in the management of Pseudomonas aeruginosa infections in patients with cystic fibrosis
    • Cheer, SM, Waugh J, Noble S. 2003. Inhaled tobramycin (TOBI): a review of its use in the management of Pseudomonas aeruginosa infections in patients with cystic fibrosis. Drugs 63:2501–2520. https://doi.org/10.2165/00003495-200363220-00015.
    • (2003) Drugs , vol.63 , pp. 2501-2520
    • Cheer, S.M.1    Waugh, J.2    Noble, S.3
  • 258
    • 84964526748 scopus 로고    scopus 로고
    • Galleria mellonella infection models for the study of bacterial diseases and for antimicrobial drug testing
    • Tsai, CJ-Y, Loh JMS, Proft T. 2016. Galleria mellonella infection models for the study of bacterial diseases and for antimicrobial drug testing. Virulence 7:214–229. https://doi.org/10.1080/21505594.2015.1135289.
    • (2016) Virulence , vol.7 , pp. 214-229
    • Tsai, C.-Y.1    Loh, J.2    Proft, T.3
  • 259
    • 84933517638 scopus 로고    scopus 로고
    • Structure-activity relationships in ultrashort cationic lipopeptides: The effects of amino acid ring constraint on antibacterial activity
    • Domalaon, R, Yang X, O’Neil J, Zhanel GG, Mookherjee N, Schweizer F. 2014. Structure-activity relationships in ultrashort cationic lipopeptides: the effects of amino acid ring constraint on antibacterial activity. Amino Acids 46:2517–2530. https://doi.org/10.1007/s00726-014-1806-z.
    • (2014) Amino Acids , vol.46 , pp. 2517-2530
    • Domalaon, R.1    Yang, X.2    O’Neil, J.3    Zhanel, G.G.4    Mookherjee, N.5    Schweizer, F.6
  • 260
    • 84948709890 scopus 로고    scopus 로고
    • Antibiotic resistance breakers: Can repurposed drugs fill the antibiotic discovery void?
    • Brown, D. 2015. Antibiotic resistance breakers: can repurposed drugs fill the antibiotic discovery void? Nat Rev Drug Discov 14:821–832. https://doi.org/10.1038/nrd4675.
    • (2015) Nat Rev Drug Discov , vol.14 , pp. 821-832
    • Brown, D.1
  • 261
    • 85033439702 scopus 로고    scopus 로고
    • Amphiphilic modulation of glycosylated antitumor ether lipids results in a potent triamino scaffold against epithelial cancer cell lines and BT474 cancer stem cells
    • Idowu, T, Samadder P, Arthur G, Schweizer F. 2017. Amphiphilic modulation of glycosylated antitumor ether lipids results in a potent triamino scaffold against epithelial cancer cell lines and BT474 cancer stem cells. J Med Chem 60:9724–9738. https://doi.org/10.1021/acs.jmedchem.7b01198.
    • (2017) J Med Chem , vol.60 , pp. 9724-9738
    • Idowu, T.1    Samadder, P.2    Arthur, G.3    Schweizer, F.4
  • 262
    • 85033460657 scopus 로고    scopus 로고
    • Ubiquitous nature of fluoroquinolones: The oscillation between antibacterial and anticancer activities
    • Idowu, T, Schweizer F. 2017. Ubiquitous nature of fluoroquinolones: the oscillation between antibacterial and anticancer activities. Antibiotics (Basel) 6:E26. https://doi.org/10.3390/antibiotics6040026.
    • (2017) Antibiotics (Basel) , vol.6 , pp. E26
    • Idowu, T.1    Schweizer, F.2
  • 264
    • 84905383217 scopus 로고    scopus 로고
    • Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease
    • Turner, MD, Nedjai B, Hurst T, Pennington DJ. 2014. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 1843:2563–2582. https://doi.org/10.1016/j.bbamcr.2014.05.014.
    • (2014) Biochim Biophys Acta , vol.1843 , pp. 2563-2582
    • Turner, M.D.1    Nedjai, B.2    Hurst, T.3    Pennington, D.J.4
  • 265
    • 0036141647 scopus 로고    scopus 로고
    • Modulation of release of proinflammatory bacterial compounds by antibacterials: Potential impact on course of inflammation and outcome in sepsis and meningitis
    • Nau, R, Eiffert H. 2002. Modulation of release of proinflammatory bacterial compounds by antibacterials: potential impact on course of inflammation and outcome in sepsis and meningitis. Clin Microbiol Rev 15:95–110. https://doi.org/10.1128/CMR.15.1.95-110.2002.
    • (2002) Clin Microbiol Rev , vol.15 , pp. 95-110
    • Nau, R.1    Eiffert, H.2
  • 266
    • 0033919034 scopus 로고    scopus 로고
    • Anti-inflammatory therapies in sepsis and septic shock
    • Freeman, BD, Natanson C. 2000. Anti-inflammatory therapies in sepsis and septic shock. Expert Opin Invest Drugs 9:1651–1663. https://doi.org/10.1517/13543784.9.7.1651.
    • (2000) Expert Opin Invest Drugs , vol.9 , pp. 1651-1663
    • Freeman, B.D.1    Natanson, C.2
  • 267
    • 84874263897 scopus 로고    scopus 로고
    • Anti-inflammatory effects of tobramycin and a copper-tobramycin complex with superoxide dismutase-like activity
    • Gziut, M, MacGregor HJ, Nevell TG, Mason T, Laight D, Shute JK. 2013. Anti-inflammatory effects of tobramycin and a copper-tobramycin complex with superoxide dismutase-like activity. Br J Pharmacol 168: 1165–1181. https://doi.org/10.1111/bph.12018.
    • (2013) Br J Pharmacol , vol.168 , pp. 1165-1181
    • Gziut, M.1    Macgregor, H.J.2    Nevell, T.G.3    Mason, T.4    Laight, D.5    Shute, J.K.6
  • 268
    • 84904860097 scopus 로고    scopus 로고
    • Human cathelicidin LL-37 and its derivative IG-19 regulate interleukin-32-induced inflammation
    • Choi, K-YG, Napper S, Mookherjee N. 2014. Human cathelicidin LL-37 and its derivative IG-19 regulate interleukin-32-induced inflammation. Immunology 143:68–80. https://doi.org/10.1111/imm.12291.
    • (2014) Immunology , vol.143 , pp. 68-80
    • Choi, K.-Y.1    Napper, S.2    Mookherjee, N.3
  • 269
    • 18244366046 scopus 로고    scopus 로고
    • Immunomodulatory activities of small host defense peptides
    • Bowdish, DME, Davidson DJ, Scott MG, Hancock REW. 2005. Immunomodulatory activities of small host defense peptides. Antimicrob Agents Chemother 49:1727–1732. https://doi.org/10.1128/AAC.49.5.1727-1732.2005.
    • (2005) Antimicrob Agents Chemother , vol.49 , pp. 1727-1732
    • Bowdish, D.1    Davidson, D.J.2    Scott, M.G.3    Hancock, R.E.W.4
  • 270
    • 84863331797 scopus 로고    scopus 로고
    • Cationic host defence peptides: Multifaceted role in immune modulation and inflammation
    • Choi, K-Y, Chow LNY, Mookherjee N. 2012. Cationic host defence peptides: multifaceted role in immune modulation and inflammation. J Innate Immun 4:361–370. https://doi.org/10.1159/000336630.
    • (2012) J Innate Immun , vol.4 , pp. 361-370
    • Choi, K.-Y.1    Chow, L.2    Mookherjee, N.3
  • 271
    • 0036785559 scopus 로고    scopus 로고
    • The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses
    • Scott, MG, Davidson DJ, Gold MR, Bowdish D, Hancock REW. 2002. The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses. J Immunol 169:3883–3891. https://doi.org/10.4049/jimmunol.169.7.3883.
    • (2002) J Immunol , vol.169 , pp. 3883-3891
    • Scott, M.G.1    Davidson, D.J.2    Gold, M.R.3    Bowdish, D.4    Hancock, R.E.W.5
  • 272
    • 0035884910 scopus 로고    scopus 로고
    • Quinolone molecular structure-activity relationships: What we have learned about improving antimicrobial activity
    • Peterson, LR. 2001. Quinolone molecular structure-activity relationships: what we have learned about improving antimicrobial activity. Clin Infect Dis 33(Suppl 3):S180–S186. https://doi.org/10.1086/321846.
    • (2001) Clin Infect Dis , vol.33 , pp. S180-S186
    • Peterson, L.R.1
  • 273
    • 38549088345 scopus 로고    scopus 로고
    • Energy source of flagellar type III secretion
    • Paul, K, Erhardt M, Hirano T, Blair DF, Hughes KT. 2008. Energy source of flagellar type III secretion. Nature 451:489–492. https://doi.org/10.1038/nature06497.
    • (2008) Nature , vol.451 , pp. 489-492
    • Paul, K.1    Erhardt, M.2    Hirano, T.3    Blair, D.F.4    Hughes, K.T.5
  • 275
    • 23144431968 scopus 로고    scopus 로고
    • Inhibitors of efflux pumps in Gramnegative bacteria
    • Pages, J-M, Masi M, Barbe J. 2005. Inhibitors of efflux pumps in Gramnegative bacteria. Trends Mol Med 11:382–389. https://doi.org/10.1016/j.molmed.2005.06.006.
    • (2005) Trends Mol Med , vol.11 , pp. 382-389
    • Pages, J.-M.1    Masi, M.2    Barbe, J.3
  • 278
    • 0012481773 scopus 로고    scopus 로고
    • 26th ed. Clinical and Laboratory Standards Institute, Wayne, PA
    • Clinical and Laboratory Standards Institute. 2016. Performance standards for antimicrobial susceptibility testing. CLSI supplement M100S, 26th ed. Clinical and Laboratory Standards Institute, Wayne, PA.
    • (2016) Performance Standards for Antimicrobial Susceptibility Testing
  • 279
    • 22144471145 scopus 로고    scopus 로고
    • Bacterial resistance to antibiotics: Active efflux and reduced uptake
    • Kumar, A, Schweizer HP. 2005. Bacterial resistance to antibiotics: active efflux and reduced uptake. Adv Drug Deliv Rev 57:1486–1513. https://doi.org/10.1016/j.addr.2005.04.004.
    • (2005) Adv Drug Deliv Rev , vol.57 , pp. 1486-1513
    • Kumar, A.1    Schweizer, H.P.2
  • 280
    • 84899803467 scopus 로고    scopus 로고
    • MexY-promoted aminoglycoside resistance in Pseudomonas aeruginosa: Involvement of a putative proximal binding pocket in aminoglycoside recognition
    • Lau, CH-F, Hughes D, Poole K. 2014. MexY-promoted aminoglycoside resistance in Pseudomonas aeruginosa: involvement of a putative proximal binding pocket in aminoglycoside recognition. mBio 5:e01068-14. https://doi.org/10.1128/mBio.01068-14.
    • (2014) Mbio , vol.5 , pp. e01014-e01068
    • Lau, C.-F.1    Hughes, D.2    Poole, K.3
  • 281
    • 0141959305 scopus 로고    scopus 로고
    • Contribution of the MexXY multidrug transporter to aminoglycoside resistance in Pseudomonas aeruginosa clinical isolates
    • Sobel, ML, McKay GA, Poole K. 2003. Contribution of the MexXY multidrug transporter to aminoglycoside resistance in Pseudomonas aeruginosa clinical isolates. Antimicrob Agents Chemother 47: 3202–3207. https://doi.org/10.1128/AAC.47.10.3202-3207.2003.
    • (2003) Antimicrob Agents Chemother , vol.47 , pp. 3202-3207
    • Sobel, M.L.1    McKay, G.A.2    Poole, K.3
  • 282
    • 84918539871 scopus 로고    scopus 로고
    • Trends and exceptions of physical properties on antibacterial activity for Grampositive and Gram-negative pathogens
    • Brown, DG, May-Dracka TL, Gagnon MM, Tommasi R. 2014. Trends and exceptions of physical properties on antibacterial activity for Grampositive and Gram-negative pathogens. J Med Chem 57:10144–10161. https://doi.org/10.1021/jm501552x.
    • (2014) J Med Chem , vol.57 , pp. 10144-10161
    • Brown, D.G.1    May-Dracka, T.L.2    Gagnon, M.M.3    Tommasi, R.4
  • 283
    • 33749511442 scopus 로고    scopus 로고
    • Method for regulated expression of single-copy efflux pump genes in a surrogate Pseudomonas aeruginosa strain: Identification of the BpeEF-OprC chloramphenicol and trimethoprim efflux pump of Burkholderia pseudomallei 1026b
    • Kumar, A, Chua K-L, Schweizer HP. 2006. Method for regulated expression of single-copy efflux pump genes in a surrogate Pseudomonas aeruginosa strain: identification of the BpeEF-OprC chloramphenicol and trimethoprim efflux pump of Burkholderia pseudomallei 1026b. Antimicrob Agents Chemother 50:3460–3463. https://doi.org/10.1128/AAC.00440-06.
    • (2006) Antimicrob Agents Chemother , vol.50 , pp. 3460-3463
    • Kumar, A.1    Chua, K.-L.2    Schweizer, H.P.3
  • 285
    • 21144453890 scopus 로고    scopus 로고
    • Acetobacter aceti possesses a proton motive force-dependent efflux system for acetic acid
    • Matsushita, K, Inoue T, Adachi O, Toyama H. 2005. Acetobacter aceti possesses a proton motive force-dependent efflux system for acetic acid. J Bacteriol 187:4346–4352. https://doi.org/10.1128/JB.187.13.4346-4352.2005.
    • (2005) J Bacteriol , vol.187 , pp. 4346-4352
    • Matsushita, K.1    Inoue, T.2    Adachi, O.3    Toyama, H.4
  • 286
    • 31544464121 scopus 로고    scopus 로고
    • Effect of 1-(1-naphthylmethyl)-piperazine, a novel putative efflux pump inhibitor, on antimicrobial drug susceptibility in clinical isolates of Enterobacteriaceae other than Escherichia coli
    • Schumacher, A, Steinke P, Bohnert JA, Akova M, Jonas D, Kern WV. 2006. Effect of 1-(1-naphthylmethyl)-piperazine, a novel putative efflux pump inhibitor, on antimicrobial drug susceptibility in clinical isolates of Enterobacteriaceae other than Escherichia coli. J Antimicrob Chemother 57:344–348. https://doi.org/10.1093/jac/dki446.
    • (2006) J Antimicrob Chemother , vol.57 , pp. 344-348
    • Schumacher, A.1    Steinke, P.2    Bohnert, J.A.3    Akova, M.4    Jonas, D.5    Kern, W.V.6
  • 287
    • 33645828405 scopus 로고    scopus 로고
    • Multidrug efflux inhibition in Acinetobacter baumannii: Comparison between 1-(1-naphthylmethyl)-piperazine and phenyl-arginine-beta-naphthylamide
    • Pannek, S, Higgins PG, Steinke P, Jonas D, Akova M, Bohnert JA, Seifert H, Kern WV. 2006. Multidrug efflux inhibition in Acinetobacter baumannii: comparison between 1-(1-naphthylmethyl)-piperazine and phenyl-arginine-beta-naphthylamide. J Antimicrob Chemother 57: 970–974. https://doi.org/10.1093/jac/dkl081.
    • (2006) J Antimicrob Chemother , vol.57 , pp. 970-974
    • Pannek, S.1    Higgins, P.G.2    Steinke, P.3    Jonas, D.4    Akova, M.5    Bohnert, J.A.6    Seifert, H.7    Kern, W.V.8
  • 288
    • 31544441414 scopus 로고    scopus 로고
    • Effect of 1-(1-naphthylmethyl)-piperazine, a novel putative efflux pump inhibitor, on antimicrobial drug susceptibility in clinical isolates of Escherichia coli
    • Kern, WV, Steinke P, Schumacher A, Schuster S, von Baum H, Bohnert JA. 2006. Effect of 1-(1-naphthylmethyl)-piperazine, a novel putative efflux pump inhibitor, on antimicrobial drug susceptibility in clinical isolates of Escherichia coli. J Antimicrob Chemother 57:339–343. https://doi.org/10.1093/jac/dki445.
    • (2006) J Antimicrob Chemother , vol.57 , pp. 339-343
    • Kern, W.V.1    Steinke, P.2    Schumacher, A.3    Schuster, S.4    Von Baum, H.5    Bohnert, J.A.6
  • 289
    • 12944316448 scopus 로고    scopus 로고
    • Selected arylpiperazines are capable of reversing multidrug resistance in Escherichia coli overexpressing RND efflux pumps
    • Bohnert, JA, Kern WV. 2005. Selected arylpiperazines are capable of reversing multidrug resistance in Escherichia coli overexpressing RND efflux pumps. Antimicrob Agents Chemother 49:849–852. https://doi.org/10.1128/AAC.49.2.849-852.2005.
    • (2005) Antimicrob Agents Chemother , vol.49 , pp. 849-852
    • Bohnert, J.A.1    Kern, W.V.2
  • 291
    • 34248400414 scopus 로고    scopus 로고
    • Inhibitors of bacterial efflux pumps as adjuvants in antibiotic treatments and diagnostic tools for detection of resistance by efflux
    • Van Bambeke F, Pages J-M, Lee VJ. 2006. Inhibitors of bacterial efflux pumps as adjuvants in antibiotic treatments and diagnostic tools for detection of resistance by efflux. Recent Pat Antiinfect Drug Discov 1:157–175. https://doi.org/10.2174/157489106777452692.
    • (2006) Recent Pat Antiinfect Drug Discov , vol.1 , pp. 157-175
    • van Bambeke, F.1    Pages, J.-M.2    Lee, V.J.3
  • 292
    • 12944249381 scopus 로고    scopus 로고
    • Use of an efflux pump inhibitor to determine the prevalence of efflux pump-mediated fluoroquinolone resistance and multidrug resistance in Pseudomonas aeruginosa
    • Kriengkauykiat, J, Porter E, Lomovskaya O, Wong-Beringer A. 2005. Use of an efflux pump inhibitor to determine the prevalence of efflux pump-mediated fluoroquinolone resistance and multidrug resistance in Pseudomonas aeruginosa. Antimicrob Agents Chemother 49: 565–570. https://doi.org/10.1128/AAC.49.2.565-570.2005.
    • (2005) Antimicrob Agents Chemother , vol.49 , pp. 565-570
    • Kriengkauykiat, J.1    Porter, E.2    Lomovskaya, O.3    Wong-Beringer, A.4
  • 293
    • 52449130017 scopus 로고    scopus 로고
    • Clinical impact of the over-expression of efflux pump in nonfermentative Gram-negative bacilli, development of efflux pump inhibitors
    • Vila, J, Martinez JL. 2008. Clinical impact of the over-expression of efflux pump in nonfermentative Gram-negative bacilli, development of efflux pump inhibitors. Curr Drug Targets 9:797–807. https://doi.org/10.2174/138945008785747806.
    • (2008) Curr Drug Targets , vol.9 , pp. 797-807
    • Vila, J.1    Martinez, J.L.2
  • 294
    • 84954386847 scopus 로고    scopus 로고
    • Design, synthesis and antitumor properties of glycosylated antitumor ether lipid (GAEL)- chlorambucil-hybrids
    • Idowu, T, Samadder P, Arthur G, Schweizer F. 2016. Design, synthesis and antitumor properties of glycosylated antitumor ether lipid (GAEL)- chlorambucil-hybrids. Chem Phys Lipids 194:139–148. https://doi.org/10.1016/j.chemphyslip.2015.07.003.
    • (2016) Chem Phys Lipids , vol.194 , pp. 139-148
    • Idowu, T.1    Samadder, P.2    Arthur, G.3    Schweizer, F.4
  • 295
    • 85015081962 scopus 로고    scopus 로고
    • Replacing, D-glucosamine with its L-enantiomer in glycosylated antitumor ether lipids (GAELs) retains cytotoxic effects against epithelial cancer cells and cancer stem cells
    • Ogunsina, M, Samadder P, Idowu T, Arthur G, Schweizer F. 2017. Replacing, D-glucosamine with its L-enantiomer in glycosylated antitumor ether lipids (GAELs) retains cytotoxic effects against epithelial cancer cells and cancer stem cells. J Med Chem 60:2142–2147. https://doi.org/10.1021/acs.jmedchem.6b01773.
    • (2017) J Med Chem , vol.60 , pp. 2142-2147
    • Ogunsina, M.1    Samadder, P.2    Idowu, T.3    Arthur, G.4    Schweizer, F.5
  • 297
    • 81155124280 scopus 로고    scopus 로고
    • Chemiosmotic coupling in oxidative and photosynthetic phosphorylation. 1966
    • Mitchell, P. 2011. Chemiosmotic coupling in oxidative and photosynthetic phosphorylation. 1966. Biochim, Biophys Acta 1807:1507–1538. https://doi.org/10.1016/j.bbabio.2011.09.018.
    • (2011) Biochim, Biophys Acta , pp. 1507-1538
    • Mitchell, P.1
  • 298
    • 84960877623 scopus 로고    scopus 로고
    • Controlling bacterial infections by inhibiting proton-dependent processes
    • Kaneti, G, Meir O, Mor A. 2016. Controlling bacterial infections by inhibiting proton-dependent processes. Biochim Biophys Acta 1858: 995–1003. https://doi.org/10.1016/j.bbamem.2015.10.022.
    • (2016) Biochim Biophys Acta , vol.1858 , pp. 995-1003
    • Kaneti, G.1    Meir, O.2    Mor, A.3
  • 300
    • 0032717048 scopus 로고    scopus 로고
    • Diversity of antimicrobial peptides and their mechanisms of action
    • Epand, RM, Vogel HJ. 1999. Diversity of antimicrobial peptides and their mechanisms of action. Biochim Biophys Acta 1462:11–28. https://doi.org/10.1016/S0005-2736(99)00198-4.
    • (1999) Biochim Biophys Acta , vol.1462 , pp. 11-28
    • Epand, R.M.1    Vogel, H.J.2
  • 301
    • 77952395366 scopus 로고    scopus 로고
    • Probing the “charge cluster mechanism” in amphipathic helical cationic antimicrobial peptides
    • Epand, RF, Maloy WL, Ramamoorthy A, Epand RM. 2010. Probing the “charge cluster mechanism” in amphipathic helical cationic antimicrobial peptides. Biochemistry 49:4076–4084. https://doi.org/10.1021/bi100378m.
    • (2010) Biochemistry , vol.49 , pp. 4076-4084
    • Epand, R.F.1    Maloy, W.L.2    Ramamoorthy, A.3    Epand, R.M.4
  • 302
    • 0025833449 scopus 로고
    • Hemolytic and antimicrobial activities of the twenty-four individual omission analogues of melittin
    • Blondelle, SE, Houghten RA. 1991. Hemolytic and antimicrobial activities of the twenty-four individual omission analogues of melittin. Biochemistry 30:4671–4678. https://doi.org/10.1021/bi00233a006.
    • (1991) Biochemistry , vol.30 , pp. 4671-4678
    • Blondelle, S.E.1    Houghten, R.A.2
  • 303
    • 19544382893 scopus 로고    scopus 로고
    • Effects of acyl versus aminoacyl conjugation on the properties of antimicrobial peptides
    • Radzishevsky, IS, Rotem S, Zaknoon F, Gaidukov L, Dagan A, Mor A. 2005. Effects of acyl versus aminoacyl conjugation on the properties of antimicrobial peptides. Antimicrob Agents Chemother 49:2412–2420. https://doi.org/10.1128/AAC.49.6.2412-2420.2005.
    • (2005) Antimicrob Agents Chemother , vol.49 , pp. 2412-2420
    • Radzishevsky, I.S.1    Rotem, S.2    Zaknoon, F.3    Gaidukov, L.4    Dagan, A.5    Mor, A.6
  • 304
    • 33746867147 scopus 로고    scopus 로고
    • Physicochemical properties that enhance discriminative antibacterial activity of short dermaseptin de- rivatives
    • Rotem, S, Radzishevsky I, Mor A. 2006. Physicochemical properties that enhance discriminative antibacterial activity of short dermaseptin de- rivatives. Antimicrob Agents Chemother 50:2666–2672. https://doi.org/10.1128/AAC.00030-06.
    • (2006) Antimicrob Agents Chemother , vol.50 , pp. 2666-2672
    • Rotem, S.1    Radzishevsky, I.2    Mor, A.3
  • 305
    • 33846397753 scopus 로고    scopus 로고
    • In vitro discriminative antipseudomonal properties resulting from acyl substitution of N-terminal sequence of dermaseptin s4 derivatives
    • Marynka, K, Rotem S, Portnaya I, Cogan U, Mor A. 2007. In vitro discriminative antipseudomonal properties resulting from acyl substitution of N-terminal sequence of dermaseptin s4 derivatives. Chem Biol 14:75–85. https://doi.org/10.1016/j.chembiol.2006.11.009.
    • (2007) Chem Biol , vol.14 , pp. 75-85
    • Marynka, K.1    Rotem, S.2    Portnaya, I.3    Cogan, U.4    Mor, A.5
  • 307
    • 75149123575 scopus 로고    scopus 로고
    • A comparative review of the lipoglycopeptides: Oritavancin, dalbavancin, and telavancin
    • Guskey, MT, Tsuji BT. 2010. A comparative review of the lipoglycopeptides: oritavancin, dalbavancin, and telavancin. Pharmacotherapy 30:80–94. https://doi.org/10.1592/phco.30.1.80.
    • (2010) Pharmacotherapy , vol.30 , pp. 80-94
    • Guskey, M.T.1    Tsuji, B.T.2
  • 309
    • 33644509227 scopus 로고    scopus 로고
    • Targeting mechanisms of Pseudomonas aeruginosa pathogenesis
    • Kipnis, E, Sawa T, Wiener-Kronish J. 2006. Targeting mechanisms of Pseudomonas aeruginosa pathogenesis. Med Mal Infect 36:78–91. https://doi.org/10.1016/j.medmal.2005.10.007.
    • (2006) Med Mal Infect , vol.36 , pp. 78-91
    • Kipnis, E.1    Sawa, T.2    Wiener-Kronish, J.3
  • 310
    • 79960934532 scopus 로고    scopus 로고
    • Short native antimicrobial peptides and engineered ultrashort lipopeptides: Similarities and differences in cell specificities and modes of action
    • Mangoni, ML, Shai Y. 2011. Short native antimicrobial peptides and engineered ultrashort lipopeptides: similarities and differences in cell specificities and modes of action. Cell Mol Life Sci 68:2267–2280. https://doi.org/10.1007/s00018-011-0718-2.
    • (2011) Cell Mol Life Sci , vol.68 , pp. 2267-2280
    • Mangoni, M.L.1    Shai, Y.2
  • 311
    • 71749100398 scopus 로고    scopus 로고
    • Cationic amphiphilic peptides with cancer-selective toxicity
    • Schweizer, F. 2009. Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol 625:190–194. https://doi.org/10.1016/j.ejphar.2009.08.043.
    • (2009) Eur J Pharmacol , vol.625 , pp. 190-194
    • Schweizer, F.1
  • 312
    • 0026802197 scopus 로고
    • Agents that increase the permeability of the outer membrane
    • Vaara, M. 1992. Agents that increase the permeability of the outer membrane. Microbiol Rev 56:395–411.
    • (1992) Microbiol Rev , vol.56 , pp. 395-411
    • Vaara, M.1
  • 314
    • 0023615466 scopus 로고
    • Mechanism of bactericidal action of aminoglycosides
    • Davis, BD. 1987. Mechanism of bactericidal action of aminoglycosides. Microbiol Rev 51:341–350.
    • (1987) Microbiol Rev , vol.51 , pp. 341-350
    • Davis, B.D.1
  • 315
    • 84976421894 scopus 로고    scopus 로고
    • Negatively charged lipids as a potential target for new amphiphilic aminoglycoside antibiotics: A biophysical study
    • Sautrey, G, El Khoury M, Dos Santos AG, Zimmermann L, Deleu M, Lins L, Decout J-L, Mingeot-Leclercq M-P. 2016. Negatively charged lipids as a potential target for new amphiphilic aminoglycoside antibiotics: a biophysical study. J Biol Chem 291:13864–13874. https://doi.org/10.1074/jbc.M115.665364.
    • (2016) J Biol Chem , vol.291 , pp. 13864-13874
    • Sautrey, G.1    El Khoury, M.2    Dos Santos, A.G.3    Zimmermann, L.4    Deleu, M.5    Lins, L.6    Decout, J.-L.7    Mingeot-Leclercq, M.-P.8
  • 316
    • 84878243608 scopus 로고    scopus 로고
    • Di-alkylated paromomycin derivatives: Targeting the membranes of gram positive pathogens that cause skin infections
    • Berkov-Zrihen Y, Herzog IM, Feldman M, Sonn-Segev A, Roichman Y, Fridman M. 2013. Di-alkylated paromomycin derivatives: targeting the membranes of gram positive pathogens that cause skin infections. Bioorg Med Chem 21:3624–3631. https://doi.org/10.1016/j.bmc.2013.03.046.
    • (2013) Bioorg Med Chem , vol.21 , pp. 3624-3631
    • Berkov-Zrihen, Y.1    Herzog, I.M.2    Feldman, M.3    Sonn-Segev, A.4    Roichman, Y.5    Fridman, M.6
  • 317
    • 84923646618 scopus 로고    scopus 로고
    • Tobramycin and nebramine as pseudo-oligosaccharide scaffolds for the development of antimicrobial cationic amphiphiles
    • Berkov-Zrihen Y, Herzog IM, Benhamou RI, Feldman M, Steinbuch KB, Shaul P, Lerer S, Eldar A, Fridman M. 2015. Tobramycin and nebramine as pseudo-oligosaccharide scaffolds for the development of antimicrobial cationic amphiphiles. Chemistry 21:4340–4349. https://doi.org/10.1002/chem.201406404.
    • (2015) Chemistry , vol.21 , pp. 4340-4349
    • Berkov-Zrihen, Y.1    Herzog, I.M.2    Benhamou, R.I.3    Feldman, M.4    Steinbuch, K.B.5    Shaul, P.6    Lerer, S.7    Eldar, A.8    Fridman, M.9
  • 318
    • 84994030706 scopus 로고    scopus 로고
    • New broad-spectrum antibacterial amphiphilic aminoglycosides active against resistant bacteria: From neamine derivatives to smaller neosamine analogues
    • Zimmermann, L, Das I, Desire J, Sautrey G, Barros RSV, El Khoury M, Mingeot-Leclercq M-P, Decout J-L. 2016. New broad-spectrum antibacterial amphiphilic aminoglycosides active against resistant bacteria: from neamine derivatives to smaller neosamine analogues. J Med Chem 59:9350–9369. https://doi.org/10.1021/acs.jmedchem.6b00818.
    • (2016) J Med Chem , vol.59 , pp. 9350-9369
    • Zimmermann, L.1    Das, I.2    Desire, J.3    Sautrey, G.4    Barros, R.5    El Khoury, M.6    Mingeot-Leclercq, M.-P.7    Decout, J.-L.8
  • 321
    • 77953507637 scopus 로고    scopus 로고
    • Antibacterial activity of guanidinylated neomycin B- and kanamycin A-derived amphiphilic lipid conjugates
    • Bera, S, Zhanel GG, Schweizer F. 2010. Antibacterial activity of guanidinylated neomycin B- and kanamycin A-derived amphiphilic lipid conjugates. J Antimicrob Chemother 65:1224–1227. https://doi.org/10.1093/jac/dkq083.
    • (2010) J Antimicrob Chemother , vol.65 , pp. 1224-1227
    • Bera, S.1    Zhanel, G.G.2    Schweizer, F.3
  • 322
    • 53549117489 scopus 로고    scopus 로고
    • Design, synthesis, and antibacterial activities of neomycin-lipid conjugates: Polycationic lipids with potent Gram-positive activity
    • Bera, S, Zhanel GG, Schweizer F. 2008. Design, synthesis, and antibacterial activities of neomycin-lipid conjugates: polycationic lipids with potent Gram-positive activity. J Med Chem 51:6160–6164. https://doi.org/10.1021/jm800345u.
    • (2008) J Med Chem , vol.51 , pp. 6160-6164
    • Bera, S.1    Zhanel, G.G.2    Schweizer, F.3
  • 323
    • 85016146767 scopus 로고    scopus 로고
    • Cadazolid for the treatment of Clostridium difficile
    • Endres, BT, Basseres E, Alam MJ, Garey KW. 2017. Cadazolid for the treatment of Clostridium difficile. Expert Opin Invest Drugs 26:509–514. https://doi.org/10.1080/13543784.2017.1304538.
    • (2017) Expert Opin Invest Drugs , vol.26 , pp. 509-514
    • Endres, B.T.1    Basseres, E.2    Alam, M.J.3    Garey, K.W.4
  • 325
    • 84893515123 scopus 로고    scopus 로고
    • Investigations of the mode of action and resistance development of cadazolid, a new antibiotic for treatment of Clostridium difficile infections
    • Locher, HH, Caspers P, Bruyere T, Schroeder S, Pfaff P, Knezevic A, Keck W, Ritz D. 2014. Investigations of the mode of action and resistance development of cadazolid, a new antibiotic for treatment of Clostridium difficile infections. Antimicrob Agents Chemother 58:901–908. https://doi.org/10.1128/AAC.01831-13.
    • (2014) Antimicrob Agents Chemother , vol.58 , pp. 901-908
    • Locher, H.H.1    Caspers, P.2    Bruyere, T.3    Schroeder, S.4    Pfaff, P.5    Knezevic, A.6    Keck, W.7    Ritz, D.8
  • 326
    • 84942850284 scopus 로고    scopus 로고
    • Multicenter, double-blind, randomized, phase 2 study evaluating the novel antibiotic cadazolid in patients with Clostridium difficile infection
    • Louie, T, Nord CE, Talbot GH, Wilcox M, Gerding DN, Buitrago M, Kracker H, Charef P, Cornely OA. 2015. Multicenter, double-blind, randomized, phase 2 study evaluating the novel antibiotic cadazolid in patients with Clostridium difficile infection. Antimicrob Agents Chemother 59: 6266–6273. https://doi.org/10.1128/AAC.00504-15.
    • (2015) Antimicrob Agents Chemother , vol.59 , pp. 6266-6273
    • Louie, T.1    Nord, C.E.2    Talbot, G.H.3    Wilcox, M.4    Gerding, D.N.5    Buitrago, M.6    Kracker, H.7    Charef, P.8    Cornely, O.A.9
  • 327
    • 84894045611 scopus 로고    scopus 로고
    • Cadazolid, a novel antibiotic with potent activity against Clostridium difficile: Safety, tolerability and pharmacokinetics in healthy subjects following single and multiple oral doses
    • Baldoni, D, Gutierrez M, Timmer W, Dingemanse J. 2014. Cadazolid, a novel antibiotic with potent activity against Clostridium difficile: safety, tolerability and pharmacokinetics in healthy subjects following single and multiple oral doses. J Antimicrob Chemother 69:706–714. https://doi.org/10.1093/jac/dkt401.
    • (2014) J Antimicrob Chemother , vol.69 , pp. 706-714
    • Baldoni, D.1    Gutierrez, M.2    Timmer, W.3    Dingemanse, J.4
  • 328
    • 46249084681 scopus 로고    scopus 로고
    • In vitro evaluation of CBR-2092, a novel rifamycin-quinolone hybrid antibiotic: Studies of the mode of action in Staphylococcus aureus
    • Robertson, GT, Bonventre EJ, Doyle TB, Du Q, Duncan L, Morris TW, Roche ED, Yan D, Lynch AS. 2008. In vitro evaluation of CBR-2092, a novel rifamycin-quinolone hybrid antibiotic: studies of the mode of action in Staphylococcus aureus. Antimicrob Agents Chemother 52: 2313–2323. https://doi.org/10.1128/AAC.01649-07.
    • (2008) Antimicrob Agents Chemother , vol.52 , pp. 2313-2323
    • Robertson, G.T.1    Bonventre, E.J.2    Doyle, T.B.3    Du, Q.4    Duncan, L.5    Morris, T.W.6    Roche, E.D.7    Yan, D.8    Lynch, A.S.9
  • 329
    • 46249112774 scopus 로고    scopus 로고
    • In vitro evaluation of CBR-2092, a novel rifamycin-quinolone hybrid antibiotic: Microbiology profiling studies with staphylococci and streptococci
    • Robertson, GT, Bonventre EJ, Doyle TB, Du Q, Duncan L, Morris TW, Roche ED, Yan D, Lynch AS. 2008. In vitro evaluation of CBR-2092, a novel rifamycin-quinolone hybrid antibiotic: microbiology profiling studies with staphylococci and streptococci. Antimicrob Agents Chemother 52:2324–2334. https://doi.org/10.1128/AAC.01651-07.
    • (2008) Antimicrob Agents Chemother , vol.52 , pp. 2324-2334
    • Robertson, G.T.1    Bonventre, E.J.2    Doyle, T.B.3    Du, Q.4    Duncan, L.5    Morris, T.W.6    Roche, E.D.7    Yan, D.8    Lynch, A.S.9
  • 330
    • 79959740064 scopus 로고    scopus 로고
    • Antibiotics in the clinical pipeline in 2011
    • Butler, MS, Cooper MA. 2011. Antibiotics in the clinical pipeline in 2011. J Antibiot (Tokyo) 64:413–425. https://doi.org/10.1038/ja.2011.44.
    • (2011) J Antibiot (Tokyo) , vol.64 , pp. 413-425
    • Butler, M.S.1    Cooper, M.A.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.