Vancomyxins: Vancomycin-Polymyxin Nonapeptide Conjugates That Retain Anti-Gram-Positive Activity with Enhanced Potency against Gram-Negative StrainsClick to copy article linkArticle link copied!
- Emma van GroesenEmma van GroesenBiological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The NetherlandsMore by Emma van Groesen
- Cornelis J. SlingerlandCornelis J. SlingerlandBiological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The NetherlandsMore by Cornelis J. Slingerland
- Paolo InnocentiPaolo InnocentiBiological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The NetherlandsMore by Paolo Innocenti
- Milos MihajlovicMilos MihajlovicDivision of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The NetherlandsMore by Milos Mihajlovic
- Rosalinde MasereeuwRosalinde MasereeuwDivision of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The NetherlandsMore by Rosalinde Masereeuw
- Nathaniel I. Martin*Nathaniel I. Martin*Email: [email protected]Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The NetherlandsMore by Nathaniel I. Martin
Abstract
Vancomycin functions by binding to lipid II, the penultimate bacterial cell wall building block used by both Gram-positive and Gram-negative species. However, vancomycin is generally only able to exert its antimicrobial effect against Gram-positive strains as it cannot pass the outer membrane (OM) of Gram-negative bacteria. To address this challenge, we here describe efforts to conjugate vancomycin to the OM disrupting polymyxin E nonapeptide (PMEN) to yield the hybrid “vancomyxins”. In designing these hybrid antibiotics, different spacers and conjugation sites were explored for connecting vancomycin and PMEN. The vancomyxins show improved activity against Gram-negative strains compared with the activity of vancomycin or vancomycin supplemented with PMEN separately. In addition, the vancomyxins maintain the antimicrobial effect of vancomycin against Gram-positive strains and, in some cases, show enhanced activity against vancomycin-resistant strains. The hybrid antibiotics described here have reduced nephrotoxicity when compared with clinically used polymyxin antibiotics. This study demonstrates that covalent conjugation to an OM disruptor contributes to sensitizing Gram-negative strains to vancomycin while retaining anti-Gram-positive activity.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results and Discussion
MIC (μg/mL)a | |||||||
---|---|---|---|---|---|---|---|
Strain ID | Vancomycin | 8 | 9 | 10 | 11 | 12 | 13 |
Gram-negative bacteria | |||||||
E. coli ATCC25922 | >128 | 16 | 16 | 32 | 16 | 16 | 32 |
K. pneumoniae ATCC27736 | >128 | 32 | 16 | 64 | 8 | 16 | 32 |
Gram-positive bacteria | |||||||
B. subtilis 168 | 0.25 | 0.5 | 0.25 | 0.5 | 0.25 | 0.25 | 1 |
S. aureus ATCC29213 | 0.125 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.5 |
MIC = Minimum inhibitory concentration.
MIC (μg/mL) | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|
Strain ID | Vancomycin | PMEN | Vancomycin +8 μg/mL PMEN | 8 | 9 | 11 | 12 | 14 | 15 | |
Gram-negative bacteria | ||||||||||
E. coli | ATCC25922 | >128 | >128 | 32 | 16 | 16 | 16 | 16 | 16 | 16 |
ATCC35218 | 128 | >128 | 32 | 16 | 16 | 16 | 16 | 8 | 8 | |
K. pneumoniae | ATCC13883 | >128 | >128 | 128 | 32 | 32 | 8 | 8 | 16 | 32 |
ATCC27736 | >128 | >128 | 128 | 32 | 16 | 8 | 16 | 16 | 16 | |
A. baumannii | ATCC17978 | >128 | >128 | 128 | 128 | 64 | 32 | 32 | 128 | 128 |
BAA-747 | >128 | >128 | 128 | 32 | 64 | 32 | 32 | 16 | 32 | |
P. aeruginosa | ATCC10145 | >128 | >128 | 16 | >128 | >128 | 64 | 64 | 32 | 32 |
ATCC27853 | >128 | >128 | 4 | 16 | 32 | 16 | 16 | 16 | 16 | |
Gram-positive bacteria | ||||||||||
B. subtilis | 168 | 0.25 | >128 | ND | 0.5 | 0.25 | 0.25 | 0.25 | ≤0.008 | ≤0.008 |
S. simulans | 22 | 0.125 | >128 | ND | ≤0.008 | 0.031 | 0.031 | 0.016 | ≤0.008 | ≤0.008 |
S. aureus | MSSA ATCC29213 | 0.125 | >128 | ND | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 1 |
MRSA USA300 | 0.25 | >128 | ND | 0.5 | 0.25 | 0.25 | 0.25 | 0.25 | 1 | |
VISA LIM-2 | 4 | >128 | 4 | 2 | 2 | 2 | 4 | 2 | 8 | |
VISA NRS402 | 8 | >128 | 8 | 16 | 8 | 8 | 2 | 8 | 4 | |
VRSA 2 (vanA) | 128 | >128 | 128 | 32 | 64 | 64 | 128 | >128 | >128 | |
VRSA 3b (vanA) | >128 | >128 | >128 | 32 | >128 | 32 | >128 | >128 | >128 | |
E. faecalis | VRE E1246 (vanA) | >128 | >128 | >128 | >128 | >128 | >128 | >128 | >128 | >128 |
VRE E7406(vanB) | 32 | >128 | 32 | 8 | 64 | 64 | 128 | 128 | 128 | |
E. faecium | VSE E980 | 0.5 | >128 | ND | 0.25 | 0.25 | 0.25 | 0.25 | 0.125 | 0.125 |
VRE E155 (vanA) | >128 | >128 | >128 | 64 | 64 | 64 | 64 | 64 | 8 | |
VRE E7314 (vanB) | 128 | >128 | 128 | 2 | 8 | 2 | 8 | 0.5 | 0.031 |
MIC = Minimum inhibitory concentration, ND = not determined, PMEN = Polymyxin E nonapeptide, MSSA = Methicillin-sensitive S. aureus, MRSA = Methicillin-resistant S. aureus, VISA = Vancomycin-intermediate S. aureus, VRSA = Vancomycin-resistant S. aureus, VSE = Vancomycin-sensitive Enterococci, VRE = Vancomycin-resistant Enterococci.
Activity against Gram-Negative Bacteria
Activity against Gram-Positive Bacteria
Cell-based toxicity studies
Conclusion
Methods
Synthetic Procedures
Broth Microdilution Assays
Hemolysis Assays
PTECs Assay
Cell culture
Cell Viability Assay
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.1c00318.
Synthetic schemes and synthesis of all building blocks; analytical RP-HPLC traces, HRMS analysis and yields for all new compounds synthesized; additional MIC analysis of building blocks and control compounds; MIC analysis in μM; hemolytic and PTEC toxicity data (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Financial support provided by the European Research Council (ERC consolidator grant to NIM, grant agreement no. 725523).
References
This article references 49 other publications.
- 1Pootoolal, J., Neu, J., and Wright, G. D. (2002) Glycopeptide Antibiotic Resistance. Annu. Rev. Pharmacol. Toxicol. 42 (1), 381– 408, DOI: 10.1146/annurev.pharmtox.42.091601.142813Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhvFKntr4%253D&md5=629fa4246d555e5f836debb1de8c9fc0Glycopeptide antibiotic resistancePootoolal, Jeff; Neu, John; Wright, Gerard D.Annual Review of Pharmacology and Toxicology (2002), 42 (), 381-408CODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews Inc.)A review. Glycopeptide antibiotics are integral components of the current antibiotic arsenal that is under strong pressures as a result of the emergence of a variety of resistance mechanisms over the past 15 yr. Resistance has manifested itself largely through the expression of genes that encode proteins that reprogram cell wall biosynthesis and thus evade the action of the antibiotic in the enterococci, though recently new mechanisms have appeared that afford resistance and tolerance in the more virulent staphylococci and streptococci. Overcoming glycopeptide resistance will require innovative approaches to generate new antibiotics or otherwise to inhibit the action of resistance elements in various bacteria. The chem. complexity of the glycopeptides, the challenges of discovering and successfully exploiting new targets, and the growing no. of distinct resistance types all increase the difficulty of the current problem we face as a result of the emergence of glycopeptide resistance.
- 2Walsh, C. T., Fisher, S. L., Park, I. S., Prahalad, M., and Wu, Z. (1996) Bacterial Resistance to Vancomycin: Five Genes and One Missing Hydrogen Bond Tell the Story. Chem. Biol. 3 (1), 21– 28, DOI: 10.1016/S1074-5521(96)90079-4Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XpvVyntQ%253D%253D&md5=2be5db81641667e7a34e911621c316acBacterial resistance to vancomycin: five genes and one missing hydrogen bond tell the storyWalsh, C. T.; Fisher, S. L.; Park, I.-S.; Prahalad, M.; Wu, Z.Chemistry & Biology (1996), 3 (1), 21-8CODEN: CBOLE2; ISSN:1074-5521. (Current Biology)A review with 32 refs. A plasmid-borne transposon encodes enzymes and regulator proteins that confer resistance of enterococcal bacteria to the antibiotic vancomycin. Purifn. and characterization of individual proteins encoded by this operon has helped to elucidate the mol. basis of vancomycin resistance. Addnl., the loss of the H bond from the peptidoglycan N-acyl-Ala-D-lactate moiety to the vancomycin backbone carbonyl can quant. account for the resistance phenotype. This new understanding provides opportunities for intervention to reverse resistance.
- 3Beauregard, D. A., Williams, D. H., Gwynn, M. N., and Knowles, D. J. (1995) Dimerization and Membrane Anchors in Extracellular Targeting of Vancomycin Group Antibiotics. Antimicrob. Agents Chemother. 39 (3), 781– 785, DOI: 10.1128/AAC.39.3.781Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXktFWlu74%253D&md5=3285719a930c38ea61299a9e93425ec8Dimerization and membrane anchors in extracellular targeting of vancomycin group antibioticsBeauregard, Daniel A.; Williams, Dudley H.; Gwynn, Michael N.; Knowles, David J. C.Antimicrobial Agents and Chemotherapy (1995), 39 (3), 781-5CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Antibiotics of the vancomycin group are shown to enhance their affinities for the bacterial cell wall by the devices of either dimerization (vancomycin and other glycopeptides which dimerize even more strongly) or use of a membrane anchor (teicoplanin); a chelate mechanism is suggested in both cases, as supported by antagonism expts. with the cell wall analog di-N-acetyl-L-Lys-D-Ala-D-Ala. These results may have implications for other binding processes which occur near membrane surfaces.
- 4Blaskovich, M. A. T., Hansford, K. A., Butler, M. S., Jia, Z., Mark, A. E., and Cooper, M. A. (2018) Developments in Glycopeptide Antibiotics. ACS Infect. Dis. 4, 715– 735, DOI: 10.1021/acsinfecdis.7b00258Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVGhtLs%253D&md5=c384f5eb681053896c19c0d33e268b34Developments in Glycopeptide AntibioticsBlaskovich, Mark A. T.; Hansford, Karl A.; Butler, Mark S.; Jia, ZhiGuang; Mark, Alan E.; Cooper, Matthew A.ACS Infectious Diseases (2018), 4 (5), 715-735CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. Glycopeptide antibiotics (GPA) are a key weapon in the fight against drug resistant bacteria, with vancomycin still a mainstream therapy against serious Gram-pos. infections more than 50 years after it was first introduced. New, more potent semisynthetic derivs. that have entered the clinic, such as dalbavancin and oritavancin, have superior pharmacokinetic and target engagement profiles that enable successful treatment of vancomycin-resistant infections. In the face of resistance development, with multi-drug resistant (MDR) S. pneumonia and MRSA together causing 20-fold more infections than all MDR Gram-neg. infections combined, further improvements are desirable to ensure the Gram-pos. armamentarium is adequately maintained for future generations. A range of modified glycopeptides has been generated in the last decade via total syntheses, semisynthetic modifications of a natural product or biol. engineering. Several of these have undergone extensive characterization with demonstrated in vivo efficacy, good PK/PD profiles and no reported pre-clin. toxicity; some may be suitable for formal preclin. development. The natural product monobactam, cephalosporin and beta-lactam antibiotics all spawned multiple generations of com. and clin. successful semi-synthetic derivs. Similarly, next-generation glycopeptides are now tech. well positioned to advance to the clinic, if sufficient funding and market support returns to antibiotic development.
- 5Mackay, J. P., Gerhard, U., Beauregard, D. A., Williams, D. H., Westwell, M. S., and Searle, M. S. (1994) Glycopeptide Antibiotic Activity and the Possible Role of Dimerization: A Model for Biological Signaling. J. Am. Chem. Soc. 116 (11), 4581– 4590, DOI: 10.1021/ja00090a006Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXjtF2hsbo%253D&md5=cc44b7b483f949cac3268973fcab5134Glycopeptide Antibiotic Activity and the Possible Role of Dimerization: A Model for Biological SignalingMackay, Joel P.; Gerhard, Ute; Beauregard, Daniel A.; Williams, Dudley H.; Westwell, Martin S.; Searle, Mark S.Journal of the American Chemical Society (1994), 116 (11), 4581-90CODEN: JACSAT; ISSN:0002-7863.It is demonstrated that the presence of bacterial cell wall analogs may either enhance or, in the case of ristocetin A, oppose dimerization of glycopeptide antibiotics. These observations may imply that dimerization plays a role in the mode of action of these antibiotics, and a mechanism is proposed to take account of this possibility. The glycopeptide dimers are also found to be formed more exothermically in the presence of cell wall analogs, and the nature of biol. signaling events is discussed in this context. It is pointed out that binding enthalpy (rather than simply binding free energy, ΔG) may be an important quantity in signaling events. If this is so, then oligomers may be abundant in signaling processes partly because the extended aggregates they form are able to cooperatively amplify the conformational changes which are incurred on ligand binding, which occur through relatively small changes in free energy but larger opposing changes in enthalpy and entropy.
- 6Shlaes, D. M., Shlaes, J. H., Davies, J., and Williamson, R. (1989) Escherichia Coli Susceptible to Glycopeptide Antibiotics. Antimicrob. Agents Chemother. 33 (2), 192– 197, DOI: 10.1128/AAC.33.2.192Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXhtlajtro%253D&md5=b1128c013af843352471b14c14a8f08dEscherichia coli susceptible to glycopeptide antibioticsShlaes, David M.; Shlaes, Janet H.; Davies, Julian; Williamson, RussellAntimicrobial Agents and Chemotherapy (1989), 33 (2), 192-7CODEN: AMACCQ; ISSN:0066-4804.Mutants of E. coli susceptible to vancomycin were isolated after mutagenesis with nitrosoguanidine. One such mutant was studied extensively. Multiple regression anal. of the relationship between phys. properties of 20 glycopeptides and their in vitro activities against the vancomycin-susceptible mutant revealed a significant correlation with mol. mass. PI, hydrophobicity, and affinity of the glycopeptide for the pentapeptide target were not as important for activity. This suggested that a block of access of the antibiotic to its target could be the major factor detg. activity. Outer membrane proteins of the vancomycin-susceptible mutant, resistant parent, and revertant strains appeared normal. The mutant exhibited increased susceptibility to both erythromycin and fusidic acid which was lost in single-step revertants to vancomycin resistance. Polymyxin B nonapeptide was synergistic with erythromycin and fusidic acid against the parent and revertant but not against the susceptible mutant. Anal. of the susceptibilities of control strains of E. coli and Salmonella thyphimurium with known defects in lipopolysaccharide (LPS) synthesis revealed that core LPS mutants (Re chemotype) were phenotypically similar to the E. coli mutant under study. However, the LPS core of the mutant migrated slightly less rapidly on SDS-PAGE than wild-type or revertant core LPS and did not resemble Re chemotype LPS core obtained from Salmonella rfaC and rfaD mutants. These data suggest that defects in LPS core structure other than loss of heptose moieties may also be important in loss of resistance to large, hydrophilic mols. such as glycopeptides.
- 7Henderson, J. C., Zimmerman, S. M., Crofts, A. A., Boll, J. M., Kuhns, L. G., Herrera, C. M., and Trent, M. S. (2016) The Power of Asymmetry: Architecture and Assembly of the Gram-Negative Outer Membrane Lipid Bilayer. Annu. Rev. Microbiol. 70 (1), 255– 278, DOI: 10.1146/annurev-micro-102215-095308Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFSqtbzJ&md5=ceccc1176d3a50d3aff8594a8e049a63The Power of Asymmetry: Architecture and Assembly of the Gram-Negative Outer Membrane Lipid BilayerHenderson, Jeremy C.; Zimmerman, Shawn M.; Crofts, Alexander A.; Boll, Joseph M.; Kuhns, Lisa G.; Herrera, Carmen M.; Trent, M. StephenAnnual Review of Microbiology (2016), 70 (), 255-278CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews)Detg. the chem. compn. of biol. materials is paramount to the study of natural phenomena. Here, we describe the compn. of model gram-neg. outer membranes, focusing on the predominant assembly, an asym. bilayer of lipid mols. We also give an overview of lipid biosynthetic pathways and mol. mechanisms that organize this material into the outer membrane bilayer. An emphasis is placed on the potential of these pathways as targets for antibiotic development. We discuss deviations in compn., through bacterial cell surface remodeling, and alternative modalities to the asym. lipid bilayer. Outer membrane lipid alterations of current microbiol. interest, such as lipid structures found in commensal bacteria, are emphasized. Addnl., outer membrane components could potentially be engineered to develop vaccine platforms. Observations related to compn. and assembly of gram-neg. outer membranes will continue to generate novel discoveries, broaden biotechnologies, and reveal profound mysteries to compel future research.
- 8Heesterbeek, D. A. C., Martin, N. I., Velthuizen, A., Duijst, M., Ruyken, M., Wubbolts, R., Rooijakkers, S. H. M., and Bardoel, B. W. (2019) Complement-Dependent Outer Membrane Perturbation Sensitizes Gram-Negative Bacteria to Gram-Positive Specific Antibiotics. Sci. Rep. 9 (1), 3074Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cfos1Ghtg%253D%253D&md5=31cb8135c1978b8a306b4f34fb3dc779Complement-dependent outer membrane perturbation sensitizes Gram-negative bacteria to Gram-positive specific antibioticsHeesterbeek D A C; Velthuizen A; Duijst M; Ruyken M; Rooijakkers S H M; Bardoel B W; Martin N I; Wubbolts RScientific reports (2019), 9 (1), 3074 ISSN:.Gram-negative bacteria are refractory to the action of many antibiotics due to their impermeable outer membrane. An important player of the immune system is the complement system, a protein network in serum that directly kills Gram-negative bacteria through pore-formation by the Membrane Attack Complexes (MAC). We here show that the MAC rapidly perforates the outer membrane but that inner membrane damage, which is essential for killing, is relatively slow. Importantly, we demonstrate that MAC-induced outer membrane damage sensitizes Gram-negative bacteria to otherwise ineffective, Gram-positive-specific, antimicrobials. Synergy between serum and nisin was observed for 22 out of 53 tested Gram-negative clinical isolates and for multi-drug resistant (MDR) blood isolates. The in vivo relevance of this process is further highlighted by the fact that blood sensitizes a MDR K. pneumoniae strain to vancomycin. Altogether, these data imply that antibiotics that are considered ineffective to treat infections with Gram-negatives may have different functional outcomes in patients, due to the presence of the complement system.
- 9Stokes, J. M., MacNair, C. R., Ilyas, B., French, S., Côté, J.-P., Bouwman, C., Farha, M. A., Sieron, A. O., Whitfield, C., Coombes, B. K., and Brown, E. D. (2017) Pentamidine Sensitizes Gram-Negative Pathogens to Antibiotics and Overcomes Acquired Colistin Resistance. Nat. Microbiol. 2, 17028, DOI: 10.1038/nmicrobiol.2017.28Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czjtFyltQ%253D%253D&md5=0e886f7f98b65634e1354466157b744fPentamidine sensitizes Gram-negative pathogens to antibiotics and overcomes acquired colistin resistanceStokes Jonathan M; MacNair Craig R; Ilyas Bushra; French Shawn; Cote Jean-Philippe; Farha Maya A; Sieron Arthur O; Coombes Brian K; Brown Eric D; Bouwman Catrien; Whitfield ChrisNature microbiology (2017), 2 (), 17028 ISSN:.The increasing use of polymyxins(1) in addition to the dissemination of plasmid-borne colistin resistance threatens to cause a serious breach in our last line of defence against multidrug-resistant Gram-negative pathogens, and heralds the emergence of truly pan-resistant infections. Colistin resistance often arises through covalent modification of lipid A with cationic residues such as phosphoethanolamine-as is mediated by Mcr-1 (ref. 2)-which reduce the affinity of polymyxins for lipopolysaccharide(3). Thus, new strategies are needed to address the rapidly diminishing number of treatment options for Gram-negative infections(4). The difficulty in eradicating Gram-negative bacteria is largely due to their highly impermeable outer membrane, which serves as a barrier to many otherwise effective antibiotics(5). Here, we describe an unconventional screening platform designed to enrich for non-lethal, outer-membrane-active compounds with potential as adjuvants for conventional antibiotics. This approach identified the antiprotozoal drug pentamidine(6) as an effective perturbant of the Gram-negative outer membrane through its interaction with lipopolysaccharide. Pentamidine displayed synergy with antibiotics typically restricted to Gram-positive bacteria, yielding effective drug combinations with activity against a wide range of Gram-negative pathogens in vitro, and against systemic Acinetobacter baumannii infections in mice. Notably, the adjuvant activity of pentamidine persisted in polymyxin-resistant bacteria in vitro and in vivo. Overall, pentamidine and its structural analogues represent unexploited molecules for the treatment of Gram-negative infections, particularly those having acquired polymyxin resistance determinants.
- 10Li, Q., Cebrián, R., Montalbán-López, M., Ren, H., Wu, W., and Kuipers, O. P. (2021) Outer-Membrane-Acting Peptides and Lipid II-Targeting Antibiotics Cooperatively Kill Gram-Negative Pathogens. Commun. Biol. 4 (1), 31, DOI: 10.1038/s42003-020-01511-1Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsVGmu7c%253D&md5=2eee7e204b41c868067fb12ec037fa69Outer-membrane-acting peptides and lipid II-targeting antibiotics cooperatively kill Gram-negative pathogensLi, Qian; Cebrian, Ruben; Montalban-Lopez, Manuel; Ren, Huan; Wu, Weihui; Kuipers, Oscar P.Communications Biology (2021), 4 (1), 31CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)The development and dissemination of antibiotic-resistant bacterial pathogens is a growing global threat to public health. Novel compds. and/or therapeutic strategies are required to face the challenge posed, in particular, by Gram-neg. bacteria. Here we assess the combined effect of potent cell-wall synthesis inhibitors with either natural or synthetic peptides that can act on the outer-membrane. Thus, several linear peptides, either alone or combined with vancomycin or nisin, were tested against selected Gram-neg. pathogens, and the best one was improved by further engineering. Finally, peptide D-11 and vancomycin displayed a potent antimicrobial activity at low μM concns. against a panel of relevant Gram-neg. pathogens. This combination was highly active in biol. fluids like blood, but was non-hemolytic and non-toxic against cell lines. We conclude that vancomycin and D-11 are safe at >50-fold their MICs. Based on the results obtained, and as a proof of concept for the newly obsd. synergy, a Pseudomonas aeruginosa mouse infection model expt. was also performed, showing a 4 log10 redn. of the pathogen after treatment with the combination. This approach offers a potent alternative strategy to fight (drug-resistant) Gram-neg. pathogens in humans and mammals.
- 11Ghosh, M., Lin, Y.-M., Miller, P. A., Möllmann, U., Boggess, W. C., and Miller, M. J. (2018) Siderophore Conjugates of Daptomycin Are Potent Inhibitors of Carbapenem Resistant Strains of Acinetobacter Baumannii. ACS Infect. Dis. 4 (10), 1529– 1535, DOI: 10.1021/acsinfecdis.8b00150Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOktLvP&md5=dfa51d335aad7089d5575b148b8b44ebSiderophore Conjugates of Daptomycin are Potent Inhibitors of Carbapenem Resistant Strains of Acinetobacter baumanniiGhosh, Manuka; Lin, Yun-Ming; Miller, Patricia A.; Mollmann, Ute; Boggess, William C.; Miller, Marvin J.ACS Infectious Diseases (2018), 4 (10), 1529-1535CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Development of resistance to antibiotics is a major medical problem. One approach to extending the utility of our limited antibiotic arsenal is to repurpose antibiotics by altering their bacterial selectivity. Many antibiotics that are used to treat infections caused by Gram-pos. bacteria might be made effective against Gram-neg. bacterial infections, if they could circumvent permeability barriers and antibiotic deactivation processes assocd. with Gram-neg. bacteria. Herein, we report that covalent attachment of the normally Gram-pos.-only antibiotic, daptomycin, with iron sequestering siderophore mimetics that are recognized by Gram-neg. bacteria, provides conjugates that are active against virulent strains of Acinetobacter baumannii, including carbapenemase and cephalosporinase producers. The result is the generation of a new set of antibiotics designed to target bacterial infections that have been designated as being of dire concern.
- 12Li, X.-Z., Plésiat, P., and Nikaido, H. (2015) The Challenge of Efflux-Mediated Antibiotic Resistance in Gram-Negative Bacteria. Clin. Microbiol. Rev. 28 (2), 337– 418, DOI: 10.1128/CMR.00117-14Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXltlyhs7o%253D&md5=f481bdc9d3813f008c4c69502e9c9639The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteriaLi, Xian-Zhi; Plesiat, Patrick; Nikaido, HiroshiClinical Microbiology Reviews (2015), 28 (2), 337-418CODEN: CMIREX; ISSN:1098-6618. (American Society for Microbiology)The global emergence of multidrug-resistant Gram-neg. bacteria is a growing threat to antibiotic therapy. The chromosomally encoded drug efflux mechanisms that are ubiquitous in these bacteria greatly contribute to antibiotic resistance and present a major challenge for antibiotic development. Multidrug pumps, particularly those represented by the clin. relevant AcrAB-TolC and Mex pumps of the resistance-nodulation-division (RND) superfamily, not only mediate intrinsic and acquired multidrug resistance (MDR) but also are involved in other functions, including the bacterial stress response and pathogenicity. Addnl., efflux pumps interact synergistically with other resistance mechanisms (e.g., with the outer membrane permeability barrier) to increase resistance levels. Since the discovery of RND pumps in the early 1990s, remarkable scientific and technol. advances have allowed for an in-depth understanding of the structural and biochem. basis, substrate profiles, mol. regulation, and inhibition of MDR pumps. However, the development of clin. useful efflux pump inhibitors and/or new antibiotics that can bypass pump effects continues to be a challenge. Plasmid-borne efflux pump genes (including those for RND pumps) have increasingly been identified. This article highlights the recent progress obtained for organisms of clin. significance, together with methodol. considerations for the characterization of MDR pumps.
- 13Zabawa, T. P., Pucci, M. J., Parr, T. R., and Lister, T. (2016) Treatment of Gram-Negative Bacterial Infections by Potentiation of Antibiotics. Curr. Opin. Microbiol. 33, 7– 12, DOI: 10.1016/j.mib.2016.05.005Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XotFOrtbY%253D&md5=ef37d51a73f4b36352f113243e55f189Treatment of Gram-negative bacterial infections by potentiation of antibioticsZabawa, Thomas P.; Pucci, Michael J.; Parr, Thomas R.; Lister, TroyCurrent Opinion in Microbiology (2016), 33 (), 7-12CODEN: COMIF7; ISSN:1369-5274. (Elsevier Ltd.)Infections caused by antibiotic-resistant pathogens, particularly Gram-neg. bacteria, represent significant treatment challenges for physicians resulting in high rates of morbidity and mortality. The outer membrane of Gram-neg. bacteria acts as a permeability barrier to many compds. that would otherwise be effective antibacterial agents, including those effective against Gram-pos. pathogens. Potentiator mols. disrupt this barrier allowing entry of otherwise impermeant mols., thus providing a strategy to render multi-drug resistant pathogens susceptible to a broader range of antibiotics. Potentiator mols. are cationic and the mechanism of disruption involves interaction with the neg. charged outer membrane. This phys. attribute, along with an often high degree of lipophilicity typically endears these mols. with unacceptable toxicity. Presented herein are examples of advanced potentiator mols. being evaluated for use in combination therapy for the treatment of resistant Gram-neg. infections.
- 14Ghosh, M. and Miller, M. J. (1996) Synthesis and in Vitro Antibacterial Activity of Spermidine-Based Mixed Catechol- and Hydroxamate-Containing Siderophore--Vancomycin Conjugates. Bioorg. Med. Chem. 4 (1), 43– 48, DOI: 10.1016/0968-0896(95)00161-1Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xhs1agtbs%253D&md5=353be1938c9db511358c42da8654d606Synthesis and in vitro antibacterial activity of spermidine-based mixed catechol- and hydroxamate-containing siderophore-vancomycin conjugatesGhosh, Manuka; Miller, Marvin J.Bioorganic & Medicinal Chemistry (1996), 4 (1), 43-8CODEN: BMECEP; ISSN:0968-0896. (Elsevier)The first antibiotic conjugates of vancomycin with siderophore analogs contg. spermidine-based catechol ligands or mixed catechol and hydroxamate ligands are described. The design of the conjugates was based on the earlier observation that conjugation of siderophore components to β-lactam antibiotics induced active iron transport-mediated drug delivery. The novel conjugates were synthesized by selective acylation of the primary amino group of vancomycin. Preliminary biol. studies indicted that siderophore modified vancomycins lost some activity (4- to 16-fold) against Gram-pos. bacteria relative to vancomycin itself, and were generally similar to vancomycin in activity against Gram-neg. bacteria under iron-sufficient conditions. However, under iron-depleted conditions which mimic human serum, the conjugate contg. the spermidine-based catechol ligand displayed enhanced antibacterial activity against an antibiotic hypersensitive strain of Pseudomonas aeruginosa.
- 15Sarkar, P., Samaddar, S., Ammanathan, V., Yarlagadda, V., Ghosh, C., Shukla, M., Kaul, G., Manjithaya, R., Chopra, S., and Haldar, J. (2020) Vancomycin Derivative Inactivates Carbapenem-Resistant Acinetobacter Baumannii and Induces Autophagy. ACS Chem. Biol. 15 (4), 884– 889, DOI: 10.1021/acschembio.0c00091Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltlentbY%253D&md5=930db0ad9b8f9a67238d9ab7d5f5dc89Vancomycin Derivative Inactivates Carbapenem-Resistant Acinetobacter baumannii and Induces AutophagySarkar, Paramita; Samaddar, Sandip; Ammanathan, Veena; Yarlagadda, Venkateswarlu; Ghosh, Chandradhish; Shukla, Manjulika; Kaul, Grace; Manjithaya, Ravi; Chopra, Sidharth; Haldar, JayantaACS Chemical Biology (2020), 15 (4), 884-889CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Vancomycin is a std. drug for the treatment of multidrug-resistant Gram-pos. bacterial infections. Albeit, development of resistance (VRE, VRSA) and its inefficacy against persistent infections is a demerit. It is also intrinsically inactive against Gram-neg. bacteria. Herein, we report a vancomycin deriv., VanQAmC10, that addresses these challenges. VanQAmC10 was rapidly bactericidal against carbapenem-resistant A. baumannii (6 log10 CFU/mL redn. in 6 h), disrupted A. baumannii biofilms, and eradicated their stationary phase cells. In MRSA infected macrophages, the compd. reduced the bacterial burden by 1.3 log10 CFU/mL while vancomycin exhibited a static effect. Further investigation indicated that the compd., unlike vancomycin, promoted the intracellular degradative mechanism, autophagy, in mammalian cells, which may have contributed to its intracellular activity. The findings of the work provide new perspectives on the field of glycopeptide antibiotics.
- 16Neville, L. F., Shalit, I., Warn, P. A., Scheetz, M. H., Sun, J., Chosy, M. B., Wender, P. A., Cegelski, L., and Rendell, J. T. (2021) In Vivo Targeting of Escherichia Coli with Vancomycin-Arginine. Antimicrob. Agents Chemother. 65 (4), e02416– 20, DOI: 10.1128/AAC.02416-20Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvVKru7Y%253D&md5=9a27ab1a8c2b613b8ede21e749d06facIn vivo targeting of Escherichia coli with vancomycin-arginineNeville, Lewis f.; Shalit, Itamar; Warn, Peter a.; Scheetz, Marc h.; Sun, Jiuzhi; Chosy, Madeline b.; Wender, Paul a.; Cegelski, Lynette; Rendell, Jacob t.Antimicrobial Agents and Chemotherapy (2021), 65 (4), e02416CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)The ability of vancomycin-arginine (V-r) to extend the spectrum of activity of glycopeptides to Gram-neg. bacteria was investigated. Its MIC towards Escherichia coli, including β-lactamase expressing Ambler classes A, B, and D, was 8 to 16 μg/mL. Addn. of 8 times the MIC of V-r to E. coli was acutely bactericidal and assocd. with a low frequency of resistance (<2.32 x 10-10). In vivo, V-r markedly reduced E. coli burden by >7 log10 CFU/g in a thigh muscle model. These data warrant further development of V-r in combating E. coli, including resistant forms.
- 17Antonoplis, A., Zang, X., Wegner, T., Wender, P. A., and Cegelski, L. (2019) Vancomycin-Arginine Conjugate Inhibits Growth of Carbapenem-Resistant E. Coli and Targets Cell-Wall Synthesis. ACS Chem. Biol. 14 (9), 2065– 2070, DOI: 10.1021/acschembio.9b00565Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12jtrnP&md5=b1153f445707292b4d98469462d5b286Vancomycin-Arginine Conjugate Inhibits Growth of Carbapenem-Resistant E. coli and Targets Cell-Wall SynthesisAntonoplis, Alexandra; Zang, Xiaoyu; Wegner, Tristan; Wender, Paul A.; Cegelski, LynetteACS Chemical Biology (2019), 14 (9), 2065-2070CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The emergence of multi-drug-resistant Gram-neg. bacteria, including carbapenem-resistant Enterobacteriaceae, is a major health problem that necessitates the development of new antibiotics. Vancomycin inhibits cell-wall synthesis in Gram-pos. bacteria but is generally ineffective against Gram-neg. bacteria and is unable to penetrate the outer membrane barrier. To det. whether vancomycin and other antibiotics effective against Gram-pos. bacteria could, through modification, be rendered effective against Gram-neg. bacteria, we discovered that the covalent attachment of a single arginine to vancomycin yielded conjugates with order-of-magnitude improvements in activity against Gram-neg. bacteria, including pathogenic E. coli. The vancomycin-arginine conjugate (V-R) exhibited efficacy against actively growing bacteria, induced the loss of rod cellular morphol., and resulted in the intracellular accumulation of peptidoglycan precursors, all consistent with cell-wall synthesis disruption as its mechanism of action. Membrane permeabilization studies demonstrated an enhanced outer membrane permeability of V-R as compared with vancomycin. The conjugate exhibited no mammalian cell toxicity or hemolytic activity in MTT and hemolysis assays. Our study introduces a new vancomycin deriv. effective against Gram-neg. bacteria and underscores the broader potential of generating new antibiotics through combined mode-of-action and synthesis-informed design studies.
- 18Vaara, M. (1992) Agents That Increase the Permeability of the Outer Membrane. Microbiol. Rev. 56 (3), 395– 411, DOI: 10.1128/mr.56.3.395-411.1992Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXjsFCktQ%253D%253D&md5=2416c67d643c9be119bfb1c0d2323b62Agents that increase the permeability of the outer membraneVaara, MarttiMicrobiological Reviews (1992), 56 (3), 395-411CODEN: MBRED3; ISSN:0146-0749.A review with 241 refs. on cationic agents and chelators as outer membrane permeabilizers, the mol. mechanism of outer membrane permeability increase, and bacterial strains resistant to the permeabilizers.
- 19Ofek, I., Cohen, S., Rahmani, R., Kabha, K., Tamarkin, D., Herzig, Y., and Rubinstein, E. (1994) Antibacterial Synergism of Polymyxin B Nonapeptide and Hydrophobic Antibiotics in Experimental Gram-Negative Infections in Mice. Antimicrob. Agents Chemother. 38 (2), 374– 377, DOI: 10.1128/AAC.38.2.374Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXhvVagurw%253D&md5=ef5c957b98f952f4563426839261b79fAntibacterial synergism of polymyxin B nonapeptide and hydrophobic antibiotics in experimental gram-negative infections in miceOfek, Izhak; Cohen, Sofie; Rahmani, Rita; Kabha, Kisra; Tamarkin, Dove; Herzig, Yaacov; Rubinstein, EthanAntimicrobial Agents and Chemotherapy (1994), 38 (2), 374-7CODEN: AMACCQ; ISSN:0066-4804.Polymyxin B nonapeptide, derived by cleavage of the fatty acyl diaminobutryic acid from polymyxin B, is considerably less toxic, less bactericidal activity, and retains its ability to render gram-neg. bacteria susceptible to several antibiotics by permeabilizing their outer membranes. The peptide rendered all 53 polymyxin-susceptible strains tested more susceptible to novobiocin, lowering the MIC of novobiocin 8-fold or more. The combination of polymyxin B nonapeptide with novobiocin or with erythromycin administered i.p. in multiple doses synergistically protected mice infected with gram-neg. bacteria. This combination may be clin. useful because of the apparent rarity of the acquisition of resistance.
- 20Velkov, T., Thompson, P. E., Nation, R. L., and Li, J. (2010) Structure--Activity Relationships of Polymyxin Antibiotics. J. Med. Chem. 53 (5), 1898– 1916, DOI: 10.1021/jm900999hGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtleisLrP&md5=8156cb2be8057a3bd6403011ea47ad3cStructure-Activity Relationships of Polymyxin AntibioticsVelkov, Tony; Thompson, Philip E.; Nation, Roger L.; Li, JianJournal of Medicinal Chemistry (2010), 53 (5), 1898-1916CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review discussing the mechanisms of antibacterial activity of polymyxins, the mechanisms of resistance to polymyxins, the polymyxin-LPS complex, the synthesis of polymyxins, the structure-activity relationships of polymyxins, the polymyxin pharmacophore, and toxicol. properties of polymyxins.
- 21Chiharha, S., Ito, A., Yahata, M., Tobita, T., and Koyama, Y. (1974) Chemical Synthesis, Isolation and Characterization of α-N-Fattyacyl Colistin Nonapeptide with Special Reference to the Correlation between Antimicrobial Activity and Carbon Number of Fattyacyl Moiety. Agric. Biol. Chem. 38 (3), 521– 529, DOI: 10.1080/00021369.1974.10861184Google ScholarThere is no corresponding record for this reference.
- 22Keirstead, N. D., Wagoner, M. P., Bentley, P., Blais, M., Brown, C., Cheatham, L., Ciaccio, P., Dragan, Y., Ferguson, D., Fikes, J., Galvin, M., Gupta, A., Hale, M., Johnson, N., Luo, W., McGrath, F., Pietras, M., Price, S., Sathe, A. G., Sasaki, J. C., Snow, D., Walsky, R. L., and Kern, G. (2014) Early Prediction of Polymyxin-Induced Nephrotoxicity with next-Generation Urinary Kidney Injury Biomarkers. Toxicol. Sci. 137 (2), 278– 291, DOI: 10.1093/toxsci/kft247Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1elu7s%253D&md5=4d054fe2a41556455af798738444037dEarly Prediction of Polymyxin-Induced Nephrotoxicity With Next-Generation Urinary Kidney Injury BiomarkersKeirstead, Natalie D.; Wagoner, Matthew P.; Bentley, Patricia; Blais, Marie; Brown, Crystal; Cheatham, Letitia; Ciaccio, Paul; Dragan, Yvonne; Ferguson, Douglas; Fikes, Jim; Galvin, Melanie; Gupta, Anshul; Hale, Michael; Johnson, Nakpangi; Luo, Wenli; McGrath, Frank; Pietras, Mark; Price, Sally; Sathe, Abhishek G.; Sasaki, Jennifer C.; Snow, Debra; Walsky, Robert L.; Kern, GuntherToxicological Sciences (2014), 137 (2), 278-291CODEN: TOSCF2; ISSN:1096-0929. (Oxford University Press)Despite six decades of clin. experience with the polymyxin class of antibiotics, their dose-limiting nephrotoxicity remains difficult to predict due to a paucity of sensitive biomarkers. Here, we evaluate the performance of std. of care and next-generation biomarkers of renal injury in the detection and monitoring of polymyxin-induced acute kidney injury in male Han Wistar rats using colistin (polymyxin E) and a polymyxin B (PMB) deriv. with reduced nephrotoxicity, PMB nonapeptide (PMBN). This study provides the first histopathol. and biomarker anal. of PMBN, an important test of the hypothesis that fatty acid modifications and charge redns. in polymyxins can reduce their nephrotoxicity. The results indicate that alterations in a panel of urinary kidney injury biomarkers can be used to monitor histopathol. injury, with Kim-1 and α-GST emerging as the most sensitive biomarkers outperforming clin. stds. of care, serum or plasma creatinine and blood urea nitrogen. To enable the prediction of polymyxin-induced nephrotoxicity, an in vitro cytotoxicity assay was employed using human proximal tubule epithelial cells (HK-2). Cytotoxicity data in these HK-2 cells correlated with the renal toxicity detected via safety biomarker data and histopathol. evaluation, suggesting that in vitro and in vivo methods can be incorporated within a screening cascade to prioritize polymyxin class analogs with more favorable renal toxicity profiles.
- 23Vaara, M. (1983) Polymyxin B Nonapeptide Complexes with Lipopolysaccharide. FEMS Microbiol. Lett. 18 (1–2), 117– 121, DOI: 10.1111/j.1574-6968.1983.tb00461.xGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXktlGru7s%253D&md5=89942f52c643635faa861f8e938d7f8fPolymyxin B nonapeptide complexes with lipopolysaccharideVaara, M.FEMS Microbiology Letters (1983), 18 (1-2), 117-21CODEN: FMLED7; ISSN:0378-1097.Polymyxin B nonapeptide (I) did not sensitize a polymyxin-resistant pmrA mutant strain of Salmonella typhimurium to the hydrophobic agents novobiocin and fusidic acid, in contrast to the parent strain, and I, like polymyxin, inhibited the biol. activities of isolated lipopolysaccharide (gelation of Limulus hemocyte lysate, consumption of complement). Apparently, I causes a disorganization of the outer membrane by binding to the acidic groups of the lipopolysaccharide, as does polymyxin.
- 24Brandenburg, K., Moriyon, I., Arraiza, M. D., Lewark-Yvetot, G., Koch, M. H. J., and Seydel, U. (2002) Biophysical Investigations into the Interaction of Lipopolysaccharide with Polymyxins. Thermochim. Acta 382 (1), 189– 198, DOI: 10.1016/S0040-6031(01)00731-6Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsV2qtg%253D%253D&md5=1834b8332ada1c60f3c89e42377c242aBiophysical investigations into the interaction of lipopolysaccharide with polymyxinsBrandenburg, K.; Moriyon, I.; Arraiza, M. D.; Lewark-Yvetot, G.; Koch, M. H. J.; Seydel, U.Thermochimica Acta (2002), 382 (1-2), 189-198CODEN: THACAS; ISSN:0040-6031. (Elsevier Science B.V.)It is reported on the interaction of bacterial lipopolysaccharide (LPS, endotoxin), having different sugar chain length covalently bound to its hydrophobic moiety lipid A, with the polycationic antibiotics polymyxin B (PMB) and PMB-nonapeptide (PMBN). The binding enthalpies and the lipid:peptide binding stoichiometries were detd. by isothermal titrn. calorimetry (ITC). For LPS with a long sugar chain (S: smooth form LPS from Yersinia enterocolitica), the titrn. curves exhibit a strong exo-therm which can be interpreted to result from the electrostatic interaction of the neg. charges of the LPS with the pos. charges of the peptides. In contrast, the titrn. curves of LPS with a short sugar chain (LPS Re from Escherichia coli) and of free lipid A yield complex patterns of endo- and exo-therms, which result from the superposition of electrostatic binding, fluidization of the acyl chains and a transition between different three-dimensional aggregate structures of the endo-toxins due to peptide binding. IR spectroscopy indicates that the fluidizing effect of the polymyxins is similar for both types of LPS and for lipid A. Small-angle X-ray diffraction reveals, however, that the Re-type LPS and lipid A are converted from a cubic into a multilamellar structure, whereas, the S-form LPS transforms from a unilamellar into a multilamellar structure with a small no. of lamellae. The presented data allow a better understanding of the interaction of peptides with endotoxin mols.
- 25Domalaon, R., Idowu, T., Zhanel, G. G., and Schweizer, F. (2018) Antibiotic Hybrids: The Next Generation of Agents and Adjuvants against Gram-Negative Pathogens?. Clin. Microbiol. Rev. 31 (2), e00077– 17, DOI: 10.1128/CMR.00077-17Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1yjurvL&md5=cbaac686173643862c15d1a3b8f8e4ceAntibiotic hybrids: the next generation of agents and adjuvants against gram-negative pathogens?Domalaon, Ronald; Idowu, Temilolu; Zhanel, George G.; Schweizer, FrankClinical Microbiology Reviews (2018), 31 (2), e00077-17/1-e00077-17/45CODEN: CMIREX; ISSN:1098-6618. (American Society for Microbiology)The global incidence of drug-resistant Gram-neg. bacillary infections has been increasing, and there is a dire need to develop novel strategies to overcome this problem. Intrinsic resistance in Gram-neg. bacteria, such as their protective outer membrane and constitutively overexpressed efflux pumps, is a major survival weapon that renders them refractory to current antibiotics. Several potential avenues to overcome this problem have been at the heart of antibiotic drug discovery in the past few decades. We review some of these strategies, with emphasis on antibiotic hybrids either as stand-alone antibacterial agents or as adjuvants that potentiate a primary antibiotic in Gram-neg. bacteria. Antibiotic hybrid is defined in this review as a synthetic construct of two or more pharmacophores belonging to an established agent known to elicit a desired antimicrobial effect. The concepts, advances, and challenges of antibiotic hybrids are elaborated in this article. Moreover, we discuss several antibiotic hybrids that were or are in clin. evaluation. Mechanistic insights into how tobramycin-based antibiotic hybrids are able to potentiate legacy antibiotics in multidrug-resistant Gram-neg. bacilli are also highlighted. Antibiotic hybrids indeed have a promising future as a therapeutic strategy to overcome drug resistance in Gram-neg. pathogens and/or expand the usefulness of our current antibiotic arsenal.
- 26Duwe, A. K., Rupar, C. A., Horsman, G. B., and Vas, S. I. (1986) In Vitro Cytotoxicity and Antibiotic Activity of Polymyxin B Nonapeptide. Antimicrob. Agents Chemother. 30 (2), 340– 341, DOI: 10.1128/AAC.30.2.340Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XltFWqs78%253D&md5=8fe41e60b13039bda45e999d0e6a8656In vitro cytotoxicity and antibiotic activity of polymyxin B nonapeptideDuwe, Axel K.; Rupar, C. Anthony; Horsman, Greg B.; Vas, Stephen I.Antimicrobial Agents and Chemotherapy (1986), 30 (2), 340-1CODEN: AMACCQ; ISSN:0066-4804.Polymyxin B nonapeptide, prepd. by enzymic removal of the fatty acyl diaminobutyric acid side chain from polymyxin B, was ∼100-fold less toxic to K562 cells than polymyxin B. Min. inhibitory concns. of polymyxin B nonapeptide against a test panel of bacteria were 2-64-fold lower than those of polymyxin B.
- 27Domalaon, R., Yang, X., Lyu, Y., Zhanel, G. G., and Schweizer, F. (2017) Polymyxin B(3)-Tobramycin Hybrids with Pseudomonas Aeruginosa-Selective Antibacterial Activity and Strong Potentiation of Rifampicin, Minocycline, and Vancomycin. ACS Infect. Dis. 3 (12), 941– 954, DOI: 10.1021/acsinfecdis.7b00145Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1KqsrjJ&md5=2f17be1865925aa4d8a33204e13d2d95Polymyxin B3-Tobramycin Hybrids with Pseudomonas aeruginosa-Selective Antibacterial Activity and Strong Potentiation of Rifampicin, Minocycline, and VancomycinDomalaon, Ronald; Yang, Xuan; Lyu, Yinfeng; Zhanel, George G.; Schweizer, FrankACS Infectious Diseases (2017), 3 (12), 941-954CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)There is an urgent need to develop novel antibacterial agents able to eradicate drug-resistant Gram-neg. pathogens such as Pseudomonas aeruginosa. Antimicrobial hybrids have emerged as a promising strategy to combat bacterial resistance, as a stand-alone drug but also as an adjuvant in combination with existing antibiotics. Herein, we report for the first time the synthesis and biol. evaluation of polymyxin-aminoglycoside heterodimers composed of polymyxin B3 covalently linked to tobramycin via an aliph. hydrocarbon linker. The polymyxin B3-tobramycin hybrids demonstrate potent activity against carbapenem-resistant as well as multidrug- or extensively drug-resistant (MDR/XDR) P. aeruginosa clin. isolates. Furthermore, the most potent hybrid was synergistic with currently used antibiotics against wild-type and MDR/XDR P. aeruginosa but also against Acinetobacter baumannii as well. The promising biol. activity described herein warrants addnl. studies into design and development of new antimicrobial hybrids able to surmount the problem of antimicrobial resistance.
- 28Luther, A., Urfer, M., Zahn, M., Müller, M., Wang, S.-Y., Mondal, M., Vitale, A., Hartmann, J.-B., Sharpe, T., Monte, F. Lo, Kocherla, H., Cline, E., Pessi, G., Rath, P., Modaresi, S. M., Chiquet, P., Stiegeler, S., Verbree, C., Remus, T., Schmitt, M., Kolopp, C., Westwood, M.-A., Desjonquères, N., Brabet, E., Hell, S., LePoupon, K., Vermeulen, A., Jaisson, R., Rithié, V., Upert, G., Lederer, A., Zbinden, P., Wach, A., Moehle, K., Zerbe, K., Locher, H. H., Bernardini, F., Dale, G. E., Eberl, L., Wollscheid, B., Hiller, S., Robinson, J. A., and Obrecht, D. (2019) Chimeric Peptidomimetic Antibiotics against Gram-Negative Bacteria. Nature 576 (7787), 452– 458, DOI: 10.1038/s41586-019-1665-6Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVagsLjJ&md5=209ed92936450f62dd2b0d7a96e90ba1Chimeric peptidomimetic antibiotics against Gram-negative bacteriaLuther, Anatol; Urfer, Matthias; Zahn, Michael; Muller, Maik; Wang, Shuang-Yan; Mondal, Milon; Vitale, Alessandra; Hartmann, Jean-Baptiste; Sharpe, Timothy; Monte, Fabio Lo; Kocherla, Harsha; Cline, Elizabeth; Pessi, Gabriella; Rath, Parthasarathi; Modaresi, Seyed Majed; Chiquet, Petra; Stiegeler, Sarah; Verbree, Carolin; Remus, Tobias; Schmitt, Michel; Kolopp, Caroline; Westwood, Marie-Anne; Desjonqueres, Nicolas; Brabet, Emile; Hell, Sophie; LePoupon, Karen; Vermeulen, Annie; Jaisson, Regis; Rithie, Virginie; Upert, Gregory; Lederer, Alexander; Zbinden, Peter; Wach, Achim; Moehle, Kerstin; Zerbe, Katja; Locher, Hans H.; Bernardini, Francesca; Dale, Glenn E.; Eberl, Leo; Wollscheid, Bernd; Hiller, Sebastian; Robinson, John A.; Obrecht, DanielNature (London, United Kingdom) (2019), 576 (7787), 452-458CODEN: NATUAS; ISSN:0028-0836. (Nature Research)There is an urgent need for new antibiotics against Gram-neg. pathogens that are resistant to carbapenem and 3rd-generation cephalosporins, against which antibiotics of last resort have lost most of their efficacy. We describe a class of synthetic antibiotics inspired by scaffolds derived from natural products. These chimeric antibiotics contain a β-hairpin peptide macrocycle linked to the macrocycle found in the polymyxin and colistin family of natural products. They are bactericidal and have a mechanism of action that involves binding to both lipopolysaccharide and the main component (BamA) of the β-barrel folding complex (BAM) that is required for the folding and insertion of β-barrel proteins into the outer membrane of Gram-neg. bacteria. Extensively optimized derivs. show potent activity against multidrug-resistant pathogens, including all of the Gram-neg. members of the ESKAPE pathogens. These derivs. also show favorable drug properties and overcome colistin resistance, both in vitro and in vivo. The lead candidate is currently in preclin. toxicol. studies that, if successful, will allow progress into clin. studies that have the potential to address life-threatening infections by the Gram-neg. pathogens, and thus to resolve a considerable unmet medical need.
- 29Guan, D., Chen, F., Qiu, Y., Jiang, B., Gong, L., Lan, L., and Huang, W. (2019) Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug-Resistant Bacteria. Angew. Chem., Int. Ed. 58 (20), 6678– 6682, DOI: 10.1002/anie.201902210Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmvFWht7c%253D&md5=6dd58bcc9afae58d6bc9080f14a5cc45Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug-Resistant BacteriaGuan, Dongliang; Chen, Feifei; Qiu, Yunguang; Jiang, Bofeng; Gong, Likun; Lan, Lefu; Huang, WeiAngewandte Chemie, International Edition (2019), 58 (20), 6678-6682CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)In the antibiotics arsenal, vancomycin is a last resort for the treatment of intractable infections. However, this situation is under threat because of the increasing appearance of vancomycin-resistant bacteria (VRB). Herein, we report a series of novel vancomycin derivs. carrying a sulfonium moiety. The sulfonium-vancomycin derivs. exhibited enhanced antibacterial activity against VRB both in vitro and in vivo. These derivs. also exhibited activity against some Gram-neg. bacteria. The sulfonium modification enhanced the interaction of vancomycin with the bacterial cell membrane and disrupts membrane integrity. Furthermore, the in vivo pharmacokinetic profile, stability, and toxicity of these derivs. demonstrated good druggability of the sulfonium-vancomycin analogs. This work provides a promising strategy for combating drug-resistant bacterial infection, and advances the knowledge on sulfonium derivs. for structural optimization and drug development.
- 30Shi, W., Chen, F., Zou, X., Jiao, S., Wang, S., Hu, Y., Lan, L., Tang, F., and Huang, W. (2021) Design, Synthesis, and Antibacterial Evaluation of Vancomycin-LPS Binding Peptide Conjugates. Bioorg. Med. Chem. Lett. 45, 128122, DOI: 10.1016/j.bmcl.2021.128122Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1yrsL%252FE&md5=922f291986a2bed64742b69bbb5cbf7aDesign, synthesis, and antibacterial evaluation of vancomycin-LPS binding peptide conjugatesShi, Weiwei; Chen, Feifei; Zou, Xiangman; Jiao, Shang; Wang, Siqi; Hu, Yu; Lan, Lefu; Tang, Feng; Huang, WeiBioorganic & Medicinal Chemistry Letters (2021), 45 (), 128122CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Developing novel antibiotics is urgently needed with emergency of drug resistance. Vancomycin, the last resort for intractable Gram-pos. bacterial infections, is ineffective against Gram-neg. bacteria and vancomycin resistant bacteria. Herein, we report a series of novel vancomycin derivs. carrying LPS binding peptides, vancomycin-LPS binding peptide conjugates (VPCs). The LPS binding peptides were conjugated onto 4 sites of vancomycin via CuAAC or maleimide- sulfydryl addn., and the formed VPCs were screened against VISA/VRE and Gram-neg. strains. VPCs exhibited enhanced activity against vancomycin resistant bacteria and obtained the activity against Gram-neg. bacteria in vitro, providing a novel strategy for vancomycin modification and glycopeptide antibiotics synthesis.
- 31Yarlagadda, V., Akkapeddi, P., Manjunath, G. B., and Haldar, J. (2014) Membrane Active Vancomycin Analogues: A Strategy to Combat Bacterial Resistance. J. Med. Chem. 57 (11), 4558– 4568, DOI: 10.1021/jm500270wGoogle Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntlKgtrk%253D&md5=30e565c1d5f25b1e8d69b83ea5cab397Membrane active vancomycin analogs: a strategy to combat bacterial resistanceYarlagadda, Venkateswarlu; Akkapeddi, Padma; Manjunath, Goutham B.; Haldar, JayantaJournal of Medicinal Chemistry (2014), 57 (11), 4558-4568CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The alarming growth of antibiotic-resistant superbugs such as vancomycin-resistant enterococci and staphylococci has become a major global health hazard. To address this issue, the authors report the development of lipophilic cationic vancomycin analogs possessing excellent antibacterial activity against several drug-resistant strains. Compared to vancomycin, efficacy greater than 1000-fold was demonstrated against vancomycin-resistant Enterococci (VRE). Significantly, unlike vancomycin, these compds. were shown to be bactericidal at low concns. and did not induce bacterial resistance. An optimized compd. in the series, compared to vancomycin, showed higher activity in methicillin-resistant Staphylococcus aureus (MRSA) infected mouse model and exhibited superior antibacterial activity in whole blood with no obsd. toxicity. The remarkable activity of these compds. is attributed to the incorporation of a new membrane disruption mechanism into vancomycin and opens up a great opportunity for the development of novel antibiotics.
- 32Okano, A., Isley, N. A., and Boger, D. L. (2017) Peripheral Modifications of [Ψ[CH2NH]Tpg4]Vancomycin with Added Synergistic Mechanisms of Action Provide Durable and Potent Antibiotics. Proc. Natl. Acad. Sci. U. S. A. 114, 5052– 5061, DOI: 10.1073/pnas.1704125114Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXovVSitr8%253D&md5=c98e49ee55fa7f42ab921a43e6d66ed2Peripheral modifications of [Ψ[CH2NH]Tpg4]vancomycin with added synergistic mechanisms of action provide durable and potent antibioticsOkano, Akinori; Isley, Nicholas A.; Boger, Dale L.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (26), E5052-E5061CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Subsequent to binding pocket modifications designed to provide dual d-Ala-d-Ala/d-Ala-d-Lac binding that directly overcome the mol. basis of vancomycin resistance, peripheral structural changes have been explored to improve antimicrobial potency and provide addnl. synergistic mechanisms of action. A C-terminal peripheral modification, introducing a quaternary ammonium salt, is reported and was found to provide a binding pocket-modified vancomycin analog with a second mechanism of action that is independent of d-Ala-d-Ala/d-Ala-d-Lac binding. This modification, which induces cell wall permeability and is complementary to the glycopeptide inhibition of cell wall synthesis, was found to provide improvements in antimicrobial potency (200-fold) against vancomycin-resistant Enterococci (VRE). Furthermore, it is shown that this type of C-terminal modification may be combined with a second peripheral (4-chlorobiphenyl)methyl (CBP) addn. to the vancomycin disaccharide to provide even more potent antimicrobial agents [VRE min. inhibitory concn. (MIC) = 0.01-0.005 μg/mL] with activity that can be attributed to three independent and synergistic mechanisms of action, only one of which requires d-Ala-d-Ala/d-Ala-d-Lac binding. Finally, it is shown that such peripherally and binding pocket-modified vancomycin analogs display little propensity for acquired resistance by VRE and that their durability against such challenges as well as their antimicrobial potency follow now predictable trends (three > two > one mechanisms of action). Such antibiotics are expected to display durable antimicrobial activity not prone to rapidly acquired clin. resistance.
- 33Blaskovich, M. A. T., Hansford, K. A., Gong, Y., Butler, M. S., Muldoon, C., Huang, J. X., Ramu, S., Silva, A. B., Cheng, M., Kavanagh, A. M., Ziora, Z., Premraj, R., Lindahl, F., Bradford, T. A., Lee, J. C., Karoli, T., Pelingon, R., Edwards, D. J., Amado, M., Elliott, A. G., Phetsang, W., Daud, N. H., Deecke, J. E., Sidjabat, H. E., Ramaologa, S., Zuegg, J., Betley, J. R., Beevers, A. P. G., Smith, R. A. G., Roberts, J. A., Paterson, D. L., and Cooper, M. A. (2018) Protein-Inspired Antibiotics Active against Vancomycin- and Daptomycin-Resistant Bacteria. Nat. Commun. 9 (1), 1– 17, DOI: 10.1038/s41467-017-02123-wGoogle Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpsVWhtLs%253D&md5=ee332a92ea1fedfa78f83f3b3fe834beProtein-inspired antibiotics active against vancomycin- and daptomycin-resistant bacteriaBlaskovich, Mark A. T.; Hansford, Karl A.; Gong, Yujing; Butler, Mark S.; Muldoon, Craig; Huang, Johnny X.; Ramu, Soumya; Silva, Alberto B.; Cheng, Mu; Kavanagh, Angela M.; Ziora, Zyta; Premraj, Rajaratnam; Lindahl, Fredrik; Bradford, Tanya A.; Lee, June C.; Karoli, Tomislav; Pelingon, Ruby; Edwards, David J.; Amado, Maite; Elliott, Alysha G.; Phetsang, Wanida; Daud, Noor Huda; Deecke, Johan E.; Sidjabat, Hanna E.; Ramaologa, Sefetogi; Zuegg, Johannes; Betley, Jason R.; Beevers, Andrew P. G.; Smith, Richard A. G.; Roberts, Jason A.; Paterson, David L.; Cooper, Matthew A.Nature Communications (2018), 9 (1), 1-17CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The public health threat posed by a looming 'post-antibiotic' era necessitates new approaches to antibiotic discovery. Drug development has typically avoided exploitation of membrane-binding properties, in contrast to nature's control of biol. pathways via modulation of membrane-assocd. proteins and membrane lipid compn. Here, we describe the rejuvenation of the glycopeptide antibiotic vancomycin via selective targeting of bacterial membranes. Peptide libraries based on pos. charged electrostatic effector sequences are ligated to N-terminal lipophilic membrane-insertive elements and then conjugated to vancomycin. These modified lipoglycopeptides, the 'vancapticins', possess enhanced membrane affinity and activity against methicillin-resistant Staphylococcus aureus (MRSA) and other Gram-pos. bacteria, and retain activity against glycopeptide-resistant strains. Optimized antibiotics show in vivo efficacy in multiple models of bacterial infection. This membrane-targeting strategy has potential to 'revitalise' antibiotics that have lost effectiveness against recalcitrant bacteria, or enhance the activity of other i.v.-administered drugs that target membrane-assocd. receptors.
- 34Wu, Z.-C., Cameron, M. D., and Boger, D. L. (2020) Vancomycin C-Terminus Guanidine Modifications and Further Insights into an Added Mechanism of Action Imparted by a Peripheral Structural Modification. ACS Infect. Dis. 6 (8), 2169– 2180, DOI: 10.1021/acsinfecdis.0c00258Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1yntbbM&md5=3a506f57a34ebd0921392db6e6ed9248Vancomycin C-Terminus Guanidine Modifications and Further Insights into an Added Mechanism of Action Imparted by a Peripheral Structural ModificationWu, Zhi-Chen; Cameron, Michael D.; Boger, Dale L.ACS Infectious Diseases (2020), 6 (8), 2169-2180CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A series of vancomycin C-terminus guanidine modifications is disclosed that improves antimicrobial activity, enhances the durability of antimicrobial action against selection or induction of resistance, and introduces a synergistic mechanism of action independent of D-Ala-D-Ala binding and inhibition of cell wall biosynthesis. The added mechanism of action results in induced bacterial cell permeability, which we show may involve interaction with cell envelope teichoic acid. Significantly, the compds. examd. that contain two combined peripheral modifications, a (4-chlorobiphenyl)methyl (CBP) and C-terminus guanidinium modification, offer opportunities for new treatments against not only vancomycin-sensitive but esp. vancomycin-resistant bacteria where they act by two synergistic and now durable mechanisms of action independent of D-Ala-D-Ala/D-Lac binding and display superb antimicrobial potencies (MIC 0.6-0.15 μg/mL, VanA VRE). For the first time, we demonstrate that the synergistic behavior of the peripheral modifications examd. requires the presence of both the CBP and guanidine modifications in a single mol. vs. their combined use as an equimolar mixt. of singly modified compds. Finally, we show that a prototypical member of the series, G3-CBP-vancomycin (15), exhibits no hemolytic activity, displays no mammalian cell growth inhibition, possesses improved and esp. attractive in vivo pharmacokinetic (PK) properties, and displays excellent in vivo efficacy and potency against an esp. challenging multidrug-resistant (MRSA) and VanA vancomycin-resistant (VRSA) Staphylococcus aureus bacterial strain.
- 35Sarkar, P., Basak, D., Mukherjee, R., Bandow, J. E., and Haldar, J. Alkyl-Aryl-Vancomycins: Multimodal Glycopeptides with Weak Dependence on the Bacterial Metabolic State. J. Med. Chem. 2021 64 (14), 10185− 10202 DOI: 10.1021/acs.jmedchem.1c00449Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVygsrnL&md5=d7cc9f643c569d95486b76337de078fdAlkyl-Aryl-Vancomycins: Multimodal Glycopeptides with Weak Dependence on the Bacterial Metabolic StateSarkar, Paramita; Basak, Debajyoti; Mukherjee, Riya; Bandow, Julia E.; Haldar, JayantaJournal of Medicinal Chemistry (2021), 64 (14), 10185-10202CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Resistance to last-resort antibiotics such as vancomycin for Gram-pos. bacterial infections necessitates the development of new therapeutics. Furthermore, the ability of bacteria to survive antibiotic therapy through formation of biofilms and persister cells complicates treatment. Toward this, we report alkyl-aryl-vancomycins (AAVs), with high potency against vancomycin-resistant enterococci and staphylococci. Unlike vancomycin, the lead compd. AAV-qC10 was bactericidal and weakly dependent on bacterial metab. This resulted in complete eradication of non-growing cells of MRSA and disruption of its biofilms. In addn. to inhibiting cell wall biosynthesis like vancomycin, AAV-qC10 also depolarizes and permeabilizes the membrane. More importantly, the compd. delocalized the cell division protein MinD, thereby impairing bacterial growth through multiple pathways. The potential of AAV-qC10 is exemplified by its superior efficacy against MRSA in a murine thigh infection model as compared to vancomycin. This work paves the way for structural optimization and drug development for combating drug-resistant bacterial infections.
- 36Rostovtsev, V. V., Green, L. G., Fokin, V. V., and Sharpless, K. B. (2002) A Stepwise Huisgen Cycloaddition Process: Copper(I)-Catalyzed Regioselective “Ligation” of Azides and Terminal Alkynes. Angew. Chem., Int. Ed. 41 (14), 2596– 2599, DOI: 10.1002/1521-3773(20020715)41:14<2596::AID-ANIE2596>3.0.CO;2-4Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xls1Ohsr4%253D&md5=4603664be6639353b5e70f19b9f8d59fA stepwise Huisgen cycloaddition process: copper(I)-catalyzed regioselective "ligation" of azides and terminal alkynesRostovtsev, Vsevolod V.; Green, Luke G.; Fokin, Valery V.; Sharpless, K. BarryAngewandte Chemie, International Edition (2002), 41 (14), 2596-2599CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH)1,4-Disubstituted 1,2,3-triazoles I (R1 = PhCH2, PhCH2OCH2, 1-adamantyl, etc.; R2 = HO2C, Ph, PhOCH2, Et2NCH2, etc.) were readily and cleanly prepd. via highly efficient and regioselective copper(I)-catalyzed cycloaddn. of azides R1N3 with terminal alkynes R2C≡CH in 82-93% yields.
- 37Tornøe, C. W., Christensen, C., and Meldal, M. (2002) Peptidotriazoles on Solid Phase: [1,2,3]-Triazoles by Regiospecific Copper(I)-Catalyzed 1,3-Dipolar Cycloadditions of Terminal Alkynes to Azides. J. Org. Chem. 67 (9), 3057– 3064, DOI: 10.1021/jo011148jGoogle Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisVeks7w%253D&md5=6b3b805572783873952871f1b69f46ddPeptidotriazoles on Solid Phase: [1,2,3]-Triazoles by Regiospecific Copper(I)-Catalyzed 1,3-Dipolar Cycloadditions of Terminal Alkynes to AzidesTornoe, Christian W.; Christensen, Caspar; Meldal, MortenJournal of Organic Chemistry (2002), 67 (9), 3057-3064CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)The cycloaddn. of azides to alkynes is one of the most important synthetic routes to 1H-[1,2,3]-triazoles. This work reports a novel regiospecific copper(I)-catalyzed 1,3-dipolar cycloaddn. of terminal alkynes to azides on solid-phase. Primary, secondary, and tertiary alkyl azides, aryl azides, and an azido sugar were used successfully in the copper(I)-catalyzed cycloaddn. producing diversely 1,4-substituted [1,2,3]-triazoles in peptide backbones or side chains. The reaction conditions were fully compatible with solid-phase peptide synthesis on polar supports. The copper(I) catalysis is mild and efficient (>95% conversion and purity in most cases) and furthermore, the x-ray structure of 2-azido-2-methylpropanoic acid has been solved, to yield structural information on the 1,3-dipoles entering the reaction. Novel Fmoc-protected amino azides were prepd. from Fmoc-amino alcs. by Mitsunobu reaction.
- 38Silverman, S. M., Moses, J. E., and Sharpless, K. B. (2017) Reengineering Antibiotics to Combat Bacterial Resistance: Click Chemistry [1,2,3]-Triazole Vancomycin Dimers with Potent Activity against MRSA and VRE. Chem. - Eur. J. 23 (1), 79– 83, DOI: 10.1002/chem.201604765Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVGjtbvP&md5=614570f145edfc6e6579a44706b32f7cReengineering Antibiotics to Combat Bacterial Resistance: Click Chemistry [1,2,3]-Triazole Vancomycin Dimers with Potent Activity against MRSA and VRESilverman, Steven M.; Moses, John E.; Sharpless, K. BarryChemistry - A European Journal (2017), 23 (1), 79-83CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Vancomycin has long been considered a drug of last resort. Its efficiency in treating multiple drug-resistant bacterial infections, particularly methicillin-resistant Staphylococcus aureus (MRSA), has had a profound effect on the treatment of life-threatening infections. However, the emergence of resistance to vancomycin is a cause for significant worldwide concern, prompting the urgent development of new effective treatments for antibiotic resistant bacterial infections. Harnessing the benefits of multivalency and cooperativity against vancomycin-resistant strains, we report a Click Chem. approach towards reengineered vancomycin derivs. and the synthesis of a no. of dimers with increased potency against MRSA and vancomycin resistant Enterococci (VRE; VanB). These semi-synthetic dimeric ligands were linked together with great efficiency using the powerful CuAAC reaction, demonstrating high levels of selectivity and purity.
- 39O’Dowd, H., Kim, B., Margolis, P., Wang, W., Wu, C., Lopez, S. L., and Blais, J. (2007) Preparation of Tetra-Boc-Protected Polymyxin B Nonapeptide. Tetrahedron Lett. 48 (11), 2003– 2005, DOI: 10.1016/j.tetlet.2007.01.071Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvVOrtrk%253D&md5=ce84a22fff3bb014682e102ada9a3de4Preparation of tetra-Boc-protected polymyxin B nonapeptideO'Dowd, Hardwin; Kim, Bum; Margolis, Peter; Wang, Wen; Wu, Charlotte; Lopez, Sara L.; Blais, JohanneTetrahedron Letters (2007), 48 (11), 2003-2005CODEN: TELEAY; ISSN:0040-4039. (Elsevier Ltd.)A method for the selective tetra-Boc-protection of polymyxin B nonapeptide (PMBN) has been developed. Boc-ON selectively protects the amino side chains of the four diaminobutyric acid (Dab) residues in the presence of the N-terminal free amine.
- 40Chevalier, S., Bouffartigues, E., Bodilis, J., Maillot, O., Lesouhaitier, O., Feuilloley, M. G. J., Orange, N., Dufour, A., and Cornelis, P. (2017) Structure, Function and Regulation of Pseudomonas Aeruginosa Porins. FEMS Microbiol. Rev. 41 (5), 698– 722, DOI: 10.1093/femsre/fux020Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGjsLnM&md5=9264cfacba2796f9e5f5fd367f4a423eStructure, function and regulation of Pseudomonas aeruginosa porinsChevalier, Sylvie; Bouffartigues, Emeline; Bodilis, Josselin; Maillot, Olivier; Lesouhaitier, Olivier; Feuilloley, Marc G. J.; Orange, Nicole; Dufour, Alain; Cornelis, PierreFEMS Microbiology Reviews (2017), 41 (5), 698-722CODEN: FMREE4; ISSN:1574-6976. (Oxford University Press)A review. Pseudomonas aeruginosa is a Gram-neg. bacterium belonging to the γ -proteobacteria. Like other members of the Pseudomonas genus, it is known for its metabolic versatility and its ability to colonize a wide range of ecol. niches, such as rhizosphere, water environments and animal hosts, including humans where it can cause severe infections. Another particularity of P. aeruginosa is its high intrinsic resistance to antiseptics and antibiotics, which is partly due to its low outer membrane permeability. In contrast to Enterobacteria, pseudomonads do not possess general diffusion porins in their outer membrane, but rather express specific channel proteins for the uptake of different nutrients. The major outer membrane 'porin', OprF, has been extensively investigated, and displays structural, adhesion and signaling functions while its role in the diffusion of nutrients is still under discussion. Other porins include OprB and OprB2 for the diffusion of glucose, the two small outer membrane proteins OprG and OprH, and the two porins involved in phosphate/pyrophosphate uptake, OprP and OprO. The remaining nineteen porins belong to the so-called OprD (Occ) family, which is further split into two subfamilies termed OccD (8 members) and OccK (11 members). In the past years, a large amt. of information concerning the structure, function and regulation of these porins has been published, justifying why an updated review is timely.
- 41Moore, R. A., Bates, N. C., and Hancock, R. E. (1986) Interaction of Polycationic Antibiotics with Pseudomonas Aeruginosa Lipopolysaccharide and Lipid A Studied by Using Dansyl-Polymyxin. Antimicrob. Agents Chemother. 29 (3), 496– 500, DOI: 10.1128/AAC.29.3.496Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28Xhs1Ghs78%253D&md5=96445b43f1deaf3b03cf778f7be35648Interaction of polycationic antibiotics with Pseudomonas aeruginosa lipopolysaccharide and lipid A studied by using dansyl-polymyxinMoore, Richard A.; Bates, Nancy C.; Hancock, Robert E. W.Antimicrobial Agents and Chemotherapy (1986), 29 (3), 496-500CODEN: AMACCQ; ISSN:0066-4804.A fluorescent deriv. of polymyxin B (dansyl-polymyxin) was used to study the interaction of polycations with lipopolysaccharide (LPS) and lipid A from P. aeruginosa. Dansyl-polymyxin became bound to LPS and lipid A sites, including Mg2+-binding sites, resulting in a 20-fold enhancement of fluorescence. A Hill plot of the binding data showed that the binding of dansyl-polymyxin to LPS was cooperative (n = 1.98) and of high affinity (S0.5 = 0.38 μM). The maximal binding capacity of LPS was ∼4 mol of dansyl-polymyxin/mol of LPS. The dansyl-polymyxin interaction with lipid A displayed similar kinetics (n = 2.26; S0.5 = 0.38 μM), and the maximal binding capacity was ∼2 mol of dansyl-polymyxin/mol lipid A. A variety of polycationic compds., including gentamicin, streptomycin, and polymyxin B, as well as Mg2+, were able to displace dansyl-polymyxin bound to LPS or to lipid A. Marked differences both in terms of the degree of displacement and in terms of the amt. of competing polycation required to displace a given amt. of dansyl-polymyxin were obsd. Addn. of excess polymyxin B resulted in displacement of all of the dansyl-polymyxin, demonstrating that only polymyxin-binding sites were being probed. Apparently, polymyxin B binds to multiple sites on LPS, including sites which bind aminoglycoside antibiotics and other polycationic compds.
- 42Viljanen, P. and Vaara, M. (1984) Susceptibility of Gram-Negative Bacteria to Polymyxin B Nonapeptide. Antimicrob. Agents Chemother. 25 (6), 701– 705, DOI: 10.1128/AAC.25.6.701Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXks12ht7g%253D&md5=f612e1b3713f440e844845f548399319Susceptibility of gram-negative bacteria to polymyxin B nonapeptideViljanen, P.; Vaara, M.Antimicrobial Agents and Chemotherapy (1984), 25 (6), 701-5CODEN: AMACCQ; ISSN:0066-4804.Subinhibitory concns. of polymyxin B nonapeptide sensitized all 21 polymyxin-susceptible gram-neg. bacterial strains studied to hydrophobic antibiotics such as fusidic acid, novobiocin, and erythromycin. The susceptibility increases were usually 30- and 300-fold. The strains included representatives of Escherichia coli with different O- and K-antigens, Klebsiella pneumoniae, K. oxytoca, Enterobacter cloacae, Enterobacter agglomerans, Salmonella typhimurium, Acinetobacter calcoaceticus, Pseudomonas aeruginosa, and P. maltophilia. In contrast, polymyxin-resistant strains (Proteus mirabilis, Proteus vulgaris, Morganella morganii, Providencia stuartii, and Serratia marcescens) were resistant to the action of polyyxin B nonapeptide.
- 43Healy, V. L., Lessard, I. A., Roper, D. I., Knox, J. R., and Walsh, C. T. (2000) Vancomycin Resistance in Enterococci: Reprogramming of the D-Ala-D-Ala Ligases in Bacterial Peptidoglycan Biosynthesis. Chem. Biol. 7, 109– 119, DOI: 10.1016/S1074-5521(00)00116-2Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXjvFenu78%253D&md5=843f504cb857b523e57ebb0fea4a0036Vancomycin resistance in enterococci: reprogramming of the D-Ala-D-Ala ligases in bacterial peptidoglycan biosynthesisHealy, Vicki L.; Lessard, Ivan A. D.; Roper, David I.; Knox, James R.; Walsh, Christopher T.Chemistry & Biology (2000), 7 (5), R109-R119CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Science Ltd.)A review with 53 refs. Vancomycin binds to bacterial cell wall intermediates to achieve its antibiotic effect. Infections of vancomycin-resistant enterococci are, however, becoming an increasing problem; the bacteria are resistant because they synthesize different cell wall intermediates. The enzymes involved in cell wall biosynthesis, therefore, are potential targets for combating this resistance. Recent biochem. and crystallog. results are providing mechanistic and structural details about some of these targets.
- 44Arnusch, C. J., Bonvin, A. M. J. J., Verel, A. M., Jansen, W. T. M., Liskamp, R. M. J., de Kruijff, B., Pieters, R. J., and Breukink, E. (2008) The Vancomycin–Nisin(1–12) Hybrid Restores Activity against Vancomycin Resistant Enterococci. Biochemistry 47 (48), 12661– 12663, DOI: 10.1021/bi801597bGoogle Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCns7fE&md5=cfdcca0f596cbe5272f7968bfd53f9f4The vancomycin-nisin(1-12) hybrid restores activity against vancomycin resistant enterococciArnusch, Christopher J.; Bonvin, Alexandre M. J. J.; Verel, Anne Marie; Jansen, Wouter T. M.; Liskamp, Rob M. J.; de Kruijff, Ben; Pieters, Roland J.; Breukink, EefjanBiochemistry (2008), 47 (48), 12661-12663CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Lipid II is a crucial component in bacterial cell wall synthesis. It is the target of a no. of important antibiotics, which include vancomycin and nisin. Here we show that a hybrid antibiotic that consists of vancomycin and nisin fragments is significantly more active than the sep. fragments against vancomycin-resistant enterococci. Three different hybrids were synthesized using click chem. and compared. Optimal spacer lengths and connection points were predicted using computer modeling.
- 45van Hal, S. J., Paterson, D. L., and Lodise, T. P. (2013) Systematic Review and Meta-Analysis of Vancomycin-Induced Nephrotoxicity Associated with Dosing Schedules That Maintain Troughs between 15 and 20 mg per Liter. Antimicrob. Agents Chemother. 57 (2), 734– 744, DOI: 10.1128/AAC.01568-12Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntVCrtL4%253D&md5=6c8b0b3b29b0f93ce978b2dda8571ef6Systematic review and meta-analysis of vancomycin-induced nephrotoxicity associated with dosing schedules that maintain troughs between 15 and 20 milligrams per litervan Hal, S. J.; Paterson, D. L.; Lodise, T. P.Antimicrobial Agents and Chemotherapy (2013), 57 (2), 734-744CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)In an effort to maximize outcomes, recent expert guidelines recommend more-intensive vancomycin dosing schedules to maintain vancomycin troughs between 15 and 20 mg/L. The widespread use of these more-intensive regimens has been assocd. with an increase in vancomycin-induced nephrotoxicity reports. The purpose of this systematic literature review is to det. the nephrotoxicity potential of maintaining higher troughs in clin. practice. All studies pertaining to vancomycin-induced nephrotoxicity between 1996 and Apr. 2012 were identified from PubMed, Embase, Cochrane Controlled Trial Registry, and Medline databases and analyzed according to Cochrane guidelines. Of the initial 240 studies identified, 38 were reviewed, and 15 studies met the inclusion criteria. Overall, higher troughs (≥15 mg/L) were assocd. with increased odds of nephrotoxicity (odds ratio [OR], 2.67; 95% confidence interval [CI], 1.95 to 3.65) relative to lower troughs of <15 mg/L. The relationship between a trough of ≥15 mg/L and nephrotoxicity persisted when the anal. was restricted to studies that examd. only initial trough concns. (OR, 3.12; 95% CI, 1.81 to 5.37). The relationship between troughs of ≥15 mg/L and nephrotoxicity persisted after adjustment for covariates known to independently increase the risk of a nephrotoxicity event. An incremental increase in nephrotoxicity was also obsd. with longer durations of vancomycin administration. Vancomycin-induced nephrotoxicity was reversible in the majority of cases, with short-term dialysis required only in 3% of nephrotoxic episodes. The collective literature indicates that an exposure-nephrotoxicity relationship for vancomycin exists. The probability of a nephrotoxic event increased as a function of the trough concn. and duration of therapy.
- 46Azad, M. A. K., Roberts, K. D., Yu, H. H., Liu, B., Schofield, A. V., James, S. A., Howard, D. L., Nation, R. L., Rogers, K., de Jonge, M. D., Thompson, P. E., Fu, J., Velkov, T., and Li, J. (2015) Significant Accumulation of Polymyxin in Single Renal Tubular Cells: A Medicinal Chemistry and Triple Correlative Microscopy Approach. Anal. Chem. 87 (3), 1590– 1595, DOI: 10.1021/ac504516kGoogle Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtlej&md5=e5079c031fe529836d002da09d6bfddeSignificant Accumulation of Polymyxin in Single Renal Tubular Cells: A Medicinal Chemistry and Triple Correlative Microscopy ApproachAzad, Mohammad A. K.; Roberts, Kade D.; Yu, Heidi H.; Liu, Boyin; Schofield, Alice V.; James, Simon A.; Howard, Daryl L.; Nation, Roger L.; Rogers, Kelly; de Jonge, Martin D.; Thompson, Philip E.; Fu, Jing; Velkov, Tony; Li, JianAnalytical Chemistry (Washington, DC, United States) (2015), 87 (3), 1590-1595CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Polymyxin is the last-line therapy against Gram-neg. 'superbugs'; however, dose-limiting nephrotoxicity can occur in up to 60% of patients after i.v. administration. Understanding the accumulation and concn. of polymyxin within renal tubular cells is essential for the development of novel strategies to ameliorate its nephrotoxicity and to develop safer, new polymyxins. We designed and synthesized a novel dual-modality iodine-labeled fluorescent probe for quant. mapping of polymyxin in kidney proximal tubular cells. Measured by synchrotron X-ray fluorescence microscopy, polymyxin concns. in single rat (NRK-52E) and human (HK-2) kidney tubular cells were approx. 1930- to 4760-fold higher than extracellular concns. Our study is the first to quant. measure the significant uptake of polymyxin in renal tubular cells and provides crucial information for the understanding of polymyxin-induced nephrotoxicity. Importantly, our approach represents a significant methodol. advancement in detn. of drug uptake for single-cell pharmacol.
- 47Sakamoto, Y., Yano, T., Hanada, Y., Takeshita, A., Inagaki, F., Masuda, S., Matsunaga, N., Koyanagi, S., and Ohdo, S. (2017) Vancomycin Induces Reactive Oxygen Species-Dependent Apoptosis via Mitochondrial Cardiolipin Peroxidation in Renal Tubular Epithelial Cells. Eur. J. Pharmacol. 800, 48– 56, DOI: 10.1016/j.ejphar.2017.02.025Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1Klsb4%253D&md5=f2027e646c3dacbc9d6e92558755d224Vancomycin induces reactive oxygen species-dependent apoptosis via mitochondrial cardiolipin peroxidation in renal tubular epithelial cellsSakamoto, Yuya; Yano, Takahisa; Hanada, Yuki; Takeshita, Aki; Inagaki, Fumika; Masuda, Satohiro; Matsunaga, Naoya; Koyanagi, Satoru; Ohdo, ShigehiroEuropean Journal of Pharmacology (2017), 800 (), 48-56CODEN: EJPHAZ; ISSN:0014-2999. (Elsevier B.V.)Vancomycin (VCM) is a first-line antibiotic for serious infections caused by methicillin-resistant Staphylococcus aureus. However, nephrotoxicity is one of the most complaint in VCM therapy. We previously reported that VCM induced apoptosis in a porcine proximal tubular epithelial cell line (LLC-PK1), in which mitochondrial complex I may generate superoxide, leading to cell death. In the present study, VCM caused prodn. of mitochondrial reactive oxygen species and peroxidn. of the mitochondrial phospholipid cardiolipin that was reversed by administration of the mitochondrial uncoupler carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP). FCCP also significantly suppressed VCM-induced depolarization of the mitochondrial membrane and apoptosis. Moreover, the lipophilic antioxidant vitamin E and a mitochondria-targeted antioxidant, mitoTEMPO, also significantly suppressed VCM-induced depolarization of mitochondrial membrane and apoptosis, whereas vitamin C, n-acetyl cysteine, or glutathione did not provide significant protection. These findings suggest that peroxidn. of the mitochondrial membrane cardiolipin mediated the VCM-induced prodn. of intracellular reactive oxygen species and initiation of apoptosis in LLC-PK1 cells. Furthermore, regulation of mitochondrial function using a mitochondria-targeted antioxidant, such as mitoTEMPO, may constitute a potential strategy for mitigation of VCM-induced proximal tubular epithelial cell injury.
- 48Jansen, J., Schophuizen, C. M. S., Wilmer, M. J., Lahham, S. H. M., Mutsaers, H. A. M., Wetzels, J. F. M., Bank, R. A., van den Heuvel, L. P., Hoenderop, J. G., and Masereeuw, R. (2014) A Morphological and Functional Comparison of Proximal Tubule Cell Lines Established from Human Urine and Kidney Tissue. Exp. Cell Res. 323 (1), 87– 99, DOI: 10.1016/j.yexcr.2014.02.011Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjslOhuro%253D&md5=751059bcefbd159e429f53d11a3970d6A morphological and functional comparison of proximal tubule cell lines established from human urine and kidney tissueJansen, J.; Schophuizen, C. M. S.; Wilmer, M. J.; Lahham, S. H. M.; Mutsaers, H. A. M.; Wetzels, J. F. M.; Bank, R. A.; van den Heuvel, L. P.; Hoenderop, J. G.; Masereeuw, R.Experimental Cell Research (2014), 323 (1), 87-99CODEN: ECREAL; ISSN:0014-4827. (Elsevier B.V.)Promising renal replacement therapies include the development of a bioartificial kidney using functional human kidney cell models. In this study, human conditionally immortalized proximal tubular epithelial cell (ciPTEC) lines originating from kidney tissue (ciPTEC-T1 and ciPTEC-T2) were compared to ciPTEC previously isolated from urine (ciPTEC-U).Subclones of all ciPTEC isolates formed tight cell layers on Transwell inserts as detd. by transepithelial resistance, inulin diffusion, E-cadherin expression and immunocytochemisty. Extracellular matrix genes collagen I and -IV α1 were highly present in both kidney tissue derived matured cell lines (p<0.001) compared to matured ciPTEC-U, whereas matured ciPTEC-U showed a more pronounced fibronectin I and laminin 5 gene expression (p<0.01 and p<0.05, resp.). Expression of the influx carrier Org. Cation Transporter 2 (OCT-2), and the efflux pumps P-glycoprotein (P-gp), Multidrug Resistance Protein 4 (MRP4) and Breast Cancer Resistance Protein (BCRP) were confirmed in the three cell lines using real-time PCR and Western blotting. The activities of OCT-2 and P-gp were sensitive to specific inhibition in all models (p<0.001). The highest activity of MRP4 and BCRP was demonstrated in ciPTEC-U (p<0.05). Finally, active albumin reabsorption was highest in ciPTEC-T2 (p<0.001), while Na+-dependent phosphate reabsorption was most abundant in ciPTEC-U (p<0.01).In conclusion, ciPTEC established from human urine or kidney tissue display comparable functional PTEC specific transporters and physiol. characteristics, providing ideal human tools for bioartificial kidney development.
- 49Nieskens, T. T. G., Peters, J. G. P., Schreurs, M. J., Smits, N., Woestenenk, R., Jansen, K., van der Made, T. K., Röring, M., Hilgendorf, C., Wilmer, M. J., and Masereeuw, R. (2016) A Human Renal Proximal Tubule Cell Line with Stable Organic Anion Transporter 1 and 3 Expression Predictive for Antiviral-Induced Toxicity. AAPS J. 18 (2), 465– 475, DOI: 10.1208/s12248-016-9871-8Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFSiurg%253D&md5=303c4ad4c9977e39981b967b3f4466d6A Human Renal Proximal Tubule Cell Line with Stable Organic Anion Transporter 1 and 3 Expression Predictive for Antiviral-Induced ToxicityNieskens, Tom T. G.; Peters, Janny G. P.; Schreurs, Marieke J.; Smits, Niels; Woestenenk, Rob; Jansen, Katja; van der Made, Thom K.; Roering, Melanie; Hilgendorf, Constanze; Wilmer, Martijn J.; Masereeuw, RosalindeAAPS Journal (2016), 18 (2), 465-475CODEN: AJAOB6; ISSN:1550-7416. (Springer)Drug-induced nephrotoxicity still hampers drug development, because current translation from in vitro or animal studies to human lacks high predictivity. Often, renal adverse effects are recognized only during clin. stages of drug development. The current study aimed to establish a robust and a more complete human cell model suitable for screening of drug-related interactions and nephrotoxicity. In addn. to endogenously expressed renal org. cation transporters and efflux transporters, conditionally immortalized proximal tubule epithelial cells (ciPTEC) were completed by transduction of cells with the org. anion transporter (OAT) 1 or OAT3. Fluorescence-activated cell sorting upon exposure to the OAT substrate fluorescein successfully enriched transduced cells. A panel of org. anions was screened for drug-interactions in ciPTEC-OAT1 and ciPTEC-OAT3. The cytotoxic response to the drug-interactions with antivirals was further examd. by cell viability assays. Upon subcloning, concn.-dependent fluorescein uptake was found with a higher affinity for ciPTEC-OAT1 (Km = 0.8 ± 0.1 μM) than ciPTEC-OAT3 (Km = 3.7 ± 0.5 μM). Co-exposure to known OAT1 and/or OAT3 substrates (viz. para-aminohippurate, estrone sulfate, probenecid, furosemide, diclofenac, and cimetidine) in cultures spanning 29 passage nos. revealed relevant inhibitory potencies, confirming the robustness of our model for drug-drug interactions studies. Functional OAT1 was directly responsible for cytotoxicity of adefovir, cidofovir, and tenofovir, while a drug interaction with zidovudine was not assocd. with decreased cell viability. Our data demonstrate that human-derived ciPTEC-OAT1 and ciPTEC-OAT3 are promising platforms for highly predictive drug screening during early phases of drug development.
Cited By
This article is cited by 13 publications.
- Trevor T. Dean, Juliet Jelú-Reyes, A’Lester C. Allen, Terry W. Moore. Peptide–Drug Conjugates: An Emerging Direction for the Next Generation of Peptide Therapeutics. Journal of Medicinal Chemistry 2024, 67
(3)
, 1641-1661. https://doi.org/10.1021/acs.jmedchem.3c01835
- Yuya Komatani, Kyoka Momosaki, Akira Katsuyama, Kazuki Yamamoto, Rintaro Kaguchi, Satoshi Ichikawa. Solid-Phase Total Synthesis of Sandramycin and Its Analogues. Organic Letters 2023, 25
(3)
, 543-548. https://doi.org/10.1021/acs.orglett.2c04327
- Charlotte M. J. Wesseling, Nathaniel I. Martin. Synergy by Perturbing the Gram-Negative Outer Membrane: Opening the Door for Gram-Positive Specific Antibiotics. ACS Infectious Diseases 2022, 8
(9)
, 1731-1757. https://doi.org/10.1021/acsinfecdis.2c00193
- Emma van Groesen, Paolo Innocenti, Nathaniel I. Martin. Recent Advances in the Development of Semisynthetic Glycopeptide Antibiotics: 2014–2022. ACS Infectious Diseases 2022, 8
(8)
, 1381-1407. https://doi.org/10.1021/acsinfecdis.2c00253
- Paramita Sarkar, Kathakali De, Malvika Modi, Geetika Dhanda, Richa Priyadarshini, Julia E. Bandow, Jayanta Haldar. Next-generation membrane-active glycopeptide antibiotics that also inhibit bacterial cell division. Chemical Science 2023, 14
(9)
, 2386-2398. https://doi.org/10.1039/D2SC05600C
- Mengzhen Zhao, Fei Cao, Jiaheng Chen, Junbo Hong, Dan Deng, Qingqing Wang, Yue Sun, Qun Li, Hongbo Xin, Xiaolei Wang. Rapid, direct, visualized and antibody-free bacterial detection with extra species identification and susceptibility evaluation capabilities. Biosensors and Bioelectronics 2023, 221 , 114902. https://doi.org/10.1016/j.bios.2022.114902
- Anna Dvoretckaia, Tatiana Egorova, Apollinariia Dzhuzha, Mariia Levit, Eugene Sivtsov, Elena Demyanova, Evgenia Korzhikova-Vlakh. Polymyxin B Conjugates with Bio-Inspired Synthetic Polymers of Different Nature. International Journal of Molecular Sciences 2023, 24
(3)
, 1832. https://doi.org/10.3390/ijms24031832
- Yuanyuan Qian, Shahriar Mobashery, Jed F. Fisher. Macrolide, lincosamide, glycopeptide, and other antibacterial antibiotics. 2023, 157-213. https://doi.org/10.1016/B978-0-323-90575-6.00008-9
- Rajesh Kuppusamy, Katrina Browne, Dittu Suresh, Romano Maximo Do Rosario, Sudip Chakraborty, Sandy Yang, Mark Willcox, David Black, Renxun Chen, Naresh Kumar. Transition Towards Antibiotic Hybrid Vehicles: The Next Generation
Antibacterials. Current Medicinal Chemistry 2023, 30
(1)
, 104-125. https://doi.org/10.2174/0929867329666220613105424
- Zsolt Szűcs, Ilona Bereczki, Ferenc Fenyvesi, Pál Herczegh, Eszter Ostorházi, Anikó Borbás. Synthesis of an amphiphilic vancomycin aglycone derivative inspired by polymyxins: overcoming glycopeptide resistance in Gram-positive and Gram-negative bacteria in synergy with teicoplanin in vitro. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-24807-0
- Michael I. Treshchalin, Vasilisa A. Polozkova, Elena I. Moiseenko, Helen M. Treshalina, Andrey E. Shchekotikhin, Eleonora R. Pereverzeva. Evaluation of Toxic Properties of New Glycopeptide Flavancin on Rats. Pharmaceuticals 2022, 15
(6)
, 661. https://doi.org/10.3390/ph15060661
- Kristina Klobucar, Eric D. Brown. New potentiators of ineffective antibiotics: Targeting the Gram-negative outer membrane to overcome intrinsic resistance. Current Opinion in Chemical Biology 2022, 66 , 102099. https://doi.org/10.1016/j.cbpa.2021.102099
- Macarena Funes Chabán, Martina Hrast, Rok Frlan, Dafni G. Graikioti, Constantinos M. Athanassopoulos, María Cecilia Carpinella. Inhibition of MurA Enzyme from Escherichia coli and Staphylococcus aureus by Diterpenes from Lepechinia meyenii and Their Synthetic Analogs. Antibiotics 2021, 10
(12)
, 1535. https://doi.org/10.3390/antibiotics10121535
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 49 other publications.
- 1Pootoolal, J., Neu, J., and Wright, G. D. (2002) Glycopeptide Antibiotic Resistance. Annu. Rev. Pharmacol. Toxicol. 42 (1), 381– 408, DOI: 10.1146/annurev.pharmtox.42.091601.1428131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhvFKntr4%253D&md5=629fa4246d555e5f836debb1de8c9fc0Glycopeptide antibiotic resistancePootoolal, Jeff; Neu, John; Wright, Gerard D.Annual Review of Pharmacology and Toxicology (2002), 42 (), 381-408CODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews Inc.)A review. Glycopeptide antibiotics are integral components of the current antibiotic arsenal that is under strong pressures as a result of the emergence of a variety of resistance mechanisms over the past 15 yr. Resistance has manifested itself largely through the expression of genes that encode proteins that reprogram cell wall biosynthesis and thus evade the action of the antibiotic in the enterococci, though recently new mechanisms have appeared that afford resistance and tolerance in the more virulent staphylococci and streptococci. Overcoming glycopeptide resistance will require innovative approaches to generate new antibiotics or otherwise to inhibit the action of resistance elements in various bacteria. The chem. complexity of the glycopeptides, the challenges of discovering and successfully exploiting new targets, and the growing no. of distinct resistance types all increase the difficulty of the current problem we face as a result of the emergence of glycopeptide resistance.
- 2Walsh, C. T., Fisher, S. L., Park, I. S., Prahalad, M., and Wu, Z. (1996) Bacterial Resistance to Vancomycin: Five Genes and One Missing Hydrogen Bond Tell the Story. Chem. Biol. 3 (1), 21– 28, DOI: 10.1016/S1074-5521(96)90079-42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XpvVyntQ%253D%253D&md5=2be5db81641667e7a34e911621c316acBacterial resistance to vancomycin: five genes and one missing hydrogen bond tell the storyWalsh, C. T.; Fisher, S. L.; Park, I.-S.; Prahalad, M.; Wu, Z.Chemistry & Biology (1996), 3 (1), 21-8CODEN: CBOLE2; ISSN:1074-5521. (Current Biology)A review with 32 refs. A plasmid-borne transposon encodes enzymes and regulator proteins that confer resistance of enterococcal bacteria to the antibiotic vancomycin. Purifn. and characterization of individual proteins encoded by this operon has helped to elucidate the mol. basis of vancomycin resistance. Addnl., the loss of the H bond from the peptidoglycan N-acyl-Ala-D-lactate moiety to the vancomycin backbone carbonyl can quant. account for the resistance phenotype. This new understanding provides opportunities for intervention to reverse resistance.
- 3Beauregard, D. A., Williams, D. H., Gwynn, M. N., and Knowles, D. J. (1995) Dimerization and Membrane Anchors in Extracellular Targeting of Vancomycin Group Antibiotics. Antimicrob. Agents Chemother. 39 (3), 781– 785, DOI: 10.1128/AAC.39.3.7813https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXktFWlu74%253D&md5=3285719a930c38ea61299a9e93425ec8Dimerization and membrane anchors in extracellular targeting of vancomycin group antibioticsBeauregard, Daniel A.; Williams, Dudley H.; Gwynn, Michael N.; Knowles, David J. C.Antimicrobial Agents and Chemotherapy (1995), 39 (3), 781-5CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Antibiotics of the vancomycin group are shown to enhance their affinities for the bacterial cell wall by the devices of either dimerization (vancomycin and other glycopeptides which dimerize even more strongly) or use of a membrane anchor (teicoplanin); a chelate mechanism is suggested in both cases, as supported by antagonism expts. with the cell wall analog di-N-acetyl-L-Lys-D-Ala-D-Ala. These results may have implications for other binding processes which occur near membrane surfaces.
- 4Blaskovich, M. A. T., Hansford, K. A., Butler, M. S., Jia, Z., Mark, A. E., and Cooper, M. A. (2018) Developments in Glycopeptide Antibiotics. ACS Infect. Dis. 4, 715– 735, DOI: 10.1021/acsinfecdis.7b002584https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsVGhtLs%253D&md5=c384f5eb681053896c19c0d33e268b34Developments in Glycopeptide AntibioticsBlaskovich, Mark A. T.; Hansford, Karl A.; Butler, Mark S.; Jia, ZhiGuang; Mark, Alan E.; Cooper, Matthew A.ACS Infectious Diseases (2018), 4 (5), 715-735CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. Glycopeptide antibiotics (GPA) are a key weapon in the fight against drug resistant bacteria, with vancomycin still a mainstream therapy against serious Gram-pos. infections more than 50 years after it was first introduced. New, more potent semisynthetic derivs. that have entered the clinic, such as dalbavancin and oritavancin, have superior pharmacokinetic and target engagement profiles that enable successful treatment of vancomycin-resistant infections. In the face of resistance development, with multi-drug resistant (MDR) S. pneumonia and MRSA together causing 20-fold more infections than all MDR Gram-neg. infections combined, further improvements are desirable to ensure the Gram-pos. armamentarium is adequately maintained for future generations. A range of modified glycopeptides has been generated in the last decade via total syntheses, semisynthetic modifications of a natural product or biol. engineering. Several of these have undergone extensive characterization with demonstrated in vivo efficacy, good PK/PD profiles and no reported pre-clin. toxicity; some may be suitable for formal preclin. development. The natural product monobactam, cephalosporin and beta-lactam antibiotics all spawned multiple generations of com. and clin. successful semi-synthetic derivs. Similarly, next-generation glycopeptides are now tech. well positioned to advance to the clinic, if sufficient funding and market support returns to antibiotic development.
- 5Mackay, J. P., Gerhard, U., Beauregard, D. A., Williams, D. H., Westwell, M. S., and Searle, M. S. (1994) Glycopeptide Antibiotic Activity and the Possible Role of Dimerization: A Model for Biological Signaling. J. Am. Chem. Soc. 116 (11), 4581– 4590, DOI: 10.1021/ja00090a0065https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXjtF2hsbo%253D&md5=cc44b7b483f949cac3268973fcab5134Glycopeptide Antibiotic Activity and the Possible Role of Dimerization: A Model for Biological SignalingMackay, Joel P.; Gerhard, Ute; Beauregard, Daniel A.; Williams, Dudley H.; Westwell, Martin S.; Searle, Mark S.Journal of the American Chemical Society (1994), 116 (11), 4581-90CODEN: JACSAT; ISSN:0002-7863.It is demonstrated that the presence of bacterial cell wall analogs may either enhance or, in the case of ristocetin A, oppose dimerization of glycopeptide antibiotics. These observations may imply that dimerization plays a role in the mode of action of these antibiotics, and a mechanism is proposed to take account of this possibility. The glycopeptide dimers are also found to be formed more exothermically in the presence of cell wall analogs, and the nature of biol. signaling events is discussed in this context. It is pointed out that binding enthalpy (rather than simply binding free energy, ΔG) may be an important quantity in signaling events. If this is so, then oligomers may be abundant in signaling processes partly because the extended aggregates they form are able to cooperatively amplify the conformational changes which are incurred on ligand binding, which occur through relatively small changes in free energy but larger opposing changes in enthalpy and entropy.
- 6Shlaes, D. M., Shlaes, J. H., Davies, J., and Williamson, R. (1989) Escherichia Coli Susceptible to Glycopeptide Antibiotics. Antimicrob. Agents Chemother. 33 (2), 192– 197, DOI: 10.1128/AAC.33.2.1926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXhtlajtro%253D&md5=b1128c013af843352471b14c14a8f08dEscherichia coli susceptible to glycopeptide antibioticsShlaes, David M.; Shlaes, Janet H.; Davies, Julian; Williamson, RussellAntimicrobial Agents and Chemotherapy (1989), 33 (2), 192-7CODEN: AMACCQ; ISSN:0066-4804.Mutants of E. coli susceptible to vancomycin were isolated after mutagenesis with nitrosoguanidine. One such mutant was studied extensively. Multiple regression anal. of the relationship between phys. properties of 20 glycopeptides and their in vitro activities against the vancomycin-susceptible mutant revealed a significant correlation with mol. mass. PI, hydrophobicity, and affinity of the glycopeptide for the pentapeptide target were not as important for activity. This suggested that a block of access of the antibiotic to its target could be the major factor detg. activity. Outer membrane proteins of the vancomycin-susceptible mutant, resistant parent, and revertant strains appeared normal. The mutant exhibited increased susceptibility to both erythromycin and fusidic acid which was lost in single-step revertants to vancomycin resistance. Polymyxin B nonapeptide was synergistic with erythromycin and fusidic acid against the parent and revertant but not against the susceptible mutant. Anal. of the susceptibilities of control strains of E. coli and Salmonella thyphimurium with known defects in lipopolysaccharide (LPS) synthesis revealed that core LPS mutants (Re chemotype) were phenotypically similar to the E. coli mutant under study. However, the LPS core of the mutant migrated slightly less rapidly on SDS-PAGE than wild-type or revertant core LPS and did not resemble Re chemotype LPS core obtained from Salmonella rfaC and rfaD mutants. These data suggest that defects in LPS core structure other than loss of heptose moieties may also be important in loss of resistance to large, hydrophilic mols. such as glycopeptides.
- 7Henderson, J. C., Zimmerman, S. M., Crofts, A. A., Boll, J. M., Kuhns, L. G., Herrera, C. M., and Trent, M. S. (2016) The Power of Asymmetry: Architecture and Assembly of the Gram-Negative Outer Membrane Lipid Bilayer. Annu. Rev. Microbiol. 70 (1), 255– 278, DOI: 10.1146/annurev-micro-102215-0953087https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFSqtbzJ&md5=ceccc1176d3a50d3aff8594a8e049a63The Power of Asymmetry: Architecture and Assembly of the Gram-Negative Outer Membrane Lipid BilayerHenderson, Jeremy C.; Zimmerman, Shawn M.; Crofts, Alexander A.; Boll, Joseph M.; Kuhns, Lisa G.; Herrera, Carmen M.; Trent, M. StephenAnnual Review of Microbiology (2016), 70 (), 255-278CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews)Detg. the chem. compn. of biol. materials is paramount to the study of natural phenomena. Here, we describe the compn. of model gram-neg. outer membranes, focusing on the predominant assembly, an asym. bilayer of lipid mols. We also give an overview of lipid biosynthetic pathways and mol. mechanisms that organize this material into the outer membrane bilayer. An emphasis is placed on the potential of these pathways as targets for antibiotic development. We discuss deviations in compn., through bacterial cell surface remodeling, and alternative modalities to the asym. lipid bilayer. Outer membrane lipid alterations of current microbiol. interest, such as lipid structures found in commensal bacteria, are emphasized. Addnl., outer membrane components could potentially be engineered to develop vaccine platforms. Observations related to compn. and assembly of gram-neg. outer membranes will continue to generate novel discoveries, broaden biotechnologies, and reveal profound mysteries to compel future research.
- 8Heesterbeek, D. A. C., Martin, N. I., Velthuizen, A., Duijst, M., Ruyken, M., Wubbolts, R., Rooijakkers, S. H. M., and Bardoel, B. W. (2019) Complement-Dependent Outer Membrane Perturbation Sensitizes Gram-Negative Bacteria to Gram-Positive Specific Antibiotics. Sci. Rep. 9 (1), 30748https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cfos1Ghtg%253D%253D&md5=31cb8135c1978b8a306b4f34fb3dc779Complement-dependent outer membrane perturbation sensitizes Gram-negative bacteria to Gram-positive specific antibioticsHeesterbeek D A C; Velthuizen A; Duijst M; Ruyken M; Rooijakkers S H M; Bardoel B W; Martin N I; Wubbolts RScientific reports (2019), 9 (1), 3074 ISSN:.Gram-negative bacteria are refractory to the action of many antibiotics due to their impermeable outer membrane. An important player of the immune system is the complement system, a protein network in serum that directly kills Gram-negative bacteria through pore-formation by the Membrane Attack Complexes (MAC). We here show that the MAC rapidly perforates the outer membrane but that inner membrane damage, which is essential for killing, is relatively slow. Importantly, we demonstrate that MAC-induced outer membrane damage sensitizes Gram-negative bacteria to otherwise ineffective, Gram-positive-specific, antimicrobials. Synergy between serum and nisin was observed for 22 out of 53 tested Gram-negative clinical isolates and for multi-drug resistant (MDR) blood isolates. The in vivo relevance of this process is further highlighted by the fact that blood sensitizes a MDR K. pneumoniae strain to vancomycin. Altogether, these data imply that antibiotics that are considered ineffective to treat infections with Gram-negatives may have different functional outcomes in patients, due to the presence of the complement system.
- 9Stokes, J. M., MacNair, C. R., Ilyas, B., French, S., Côté, J.-P., Bouwman, C., Farha, M. A., Sieron, A. O., Whitfield, C., Coombes, B. K., and Brown, E. D. (2017) Pentamidine Sensitizes Gram-Negative Pathogens to Antibiotics and Overcomes Acquired Colistin Resistance. Nat. Microbiol. 2, 17028, DOI: 10.1038/nmicrobiol.2017.289https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czjtFyltQ%253D%253D&md5=0e886f7f98b65634e1354466157b744fPentamidine sensitizes Gram-negative pathogens to antibiotics and overcomes acquired colistin resistanceStokes Jonathan M; MacNair Craig R; Ilyas Bushra; French Shawn; Cote Jean-Philippe; Farha Maya A; Sieron Arthur O; Coombes Brian K; Brown Eric D; Bouwman Catrien; Whitfield ChrisNature microbiology (2017), 2 (), 17028 ISSN:.The increasing use of polymyxins(1) in addition to the dissemination of plasmid-borne colistin resistance threatens to cause a serious breach in our last line of defence against multidrug-resistant Gram-negative pathogens, and heralds the emergence of truly pan-resistant infections. Colistin resistance often arises through covalent modification of lipid A with cationic residues such as phosphoethanolamine-as is mediated by Mcr-1 (ref. 2)-which reduce the affinity of polymyxins for lipopolysaccharide(3). Thus, new strategies are needed to address the rapidly diminishing number of treatment options for Gram-negative infections(4). The difficulty in eradicating Gram-negative bacteria is largely due to their highly impermeable outer membrane, which serves as a barrier to many otherwise effective antibiotics(5). Here, we describe an unconventional screening platform designed to enrich for non-lethal, outer-membrane-active compounds with potential as adjuvants for conventional antibiotics. This approach identified the antiprotozoal drug pentamidine(6) as an effective perturbant of the Gram-negative outer membrane through its interaction with lipopolysaccharide. Pentamidine displayed synergy with antibiotics typically restricted to Gram-positive bacteria, yielding effective drug combinations with activity against a wide range of Gram-negative pathogens in vitro, and against systemic Acinetobacter baumannii infections in mice. Notably, the adjuvant activity of pentamidine persisted in polymyxin-resistant bacteria in vitro and in vivo. Overall, pentamidine and its structural analogues represent unexploited molecules for the treatment of Gram-negative infections, particularly those having acquired polymyxin resistance determinants.
- 10Li, Q., Cebrián, R., Montalbán-López, M., Ren, H., Wu, W., and Kuipers, O. P. (2021) Outer-Membrane-Acting Peptides and Lipid II-Targeting Antibiotics Cooperatively Kill Gram-Negative Pathogens. Commun. Biol. 4 (1), 31, DOI: 10.1038/s42003-020-01511-110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsVGmu7c%253D&md5=2eee7e204b41c868067fb12ec037fa69Outer-membrane-acting peptides and lipid II-targeting antibiotics cooperatively kill Gram-negative pathogensLi, Qian; Cebrian, Ruben; Montalban-Lopez, Manuel; Ren, Huan; Wu, Weihui; Kuipers, Oscar P.Communications Biology (2021), 4 (1), 31CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)The development and dissemination of antibiotic-resistant bacterial pathogens is a growing global threat to public health. Novel compds. and/or therapeutic strategies are required to face the challenge posed, in particular, by Gram-neg. bacteria. Here we assess the combined effect of potent cell-wall synthesis inhibitors with either natural or synthetic peptides that can act on the outer-membrane. Thus, several linear peptides, either alone or combined with vancomycin or nisin, were tested against selected Gram-neg. pathogens, and the best one was improved by further engineering. Finally, peptide D-11 and vancomycin displayed a potent antimicrobial activity at low μM concns. against a panel of relevant Gram-neg. pathogens. This combination was highly active in biol. fluids like blood, but was non-hemolytic and non-toxic against cell lines. We conclude that vancomycin and D-11 are safe at >50-fold their MICs. Based on the results obtained, and as a proof of concept for the newly obsd. synergy, a Pseudomonas aeruginosa mouse infection model expt. was also performed, showing a 4 log10 redn. of the pathogen after treatment with the combination. This approach offers a potent alternative strategy to fight (drug-resistant) Gram-neg. pathogens in humans and mammals.
- 11Ghosh, M., Lin, Y.-M., Miller, P. A., Möllmann, U., Boggess, W. C., and Miller, M. J. (2018) Siderophore Conjugates of Daptomycin Are Potent Inhibitors of Carbapenem Resistant Strains of Acinetobacter Baumannii. ACS Infect. Dis. 4 (10), 1529– 1535, DOI: 10.1021/acsinfecdis.8b0015011https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlOktLvP&md5=dfa51d335aad7089d5575b148b8b44ebSiderophore Conjugates of Daptomycin are Potent Inhibitors of Carbapenem Resistant Strains of Acinetobacter baumanniiGhosh, Manuka; Lin, Yun-Ming; Miller, Patricia A.; Mollmann, Ute; Boggess, William C.; Miller, Marvin J.ACS Infectious Diseases (2018), 4 (10), 1529-1535CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Development of resistance to antibiotics is a major medical problem. One approach to extending the utility of our limited antibiotic arsenal is to repurpose antibiotics by altering their bacterial selectivity. Many antibiotics that are used to treat infections caused by Gram-pos. bacteria might be made effective against Gram-neg. bacterial infections, if they could circumvent permeability barriers and antibiotic deactivation processes assocd. with Gram-neg. bacteria. Herein, we report that covalent attachment of the normally Gram-pos.-only antibiotic, daptomycin, with iron sequestering siderophore mimetics that are recognized by Gram-neg. bacteria, provides conjugates that are active against virulent strains of Acinetobacter baumannii, including carbapenemase and cephalosporinase producers. The result is the generation of a new set of antibiotics designed to target bacterial infections that have been designated as being of dire concern.
- 12Li, X.-Z., Plésiat, P., and Nikaido, H. (2015) The Challenge of Efflux-Mediated Antibiotic Resistance in Gram-Negative Bacteria. Clin. Microbiol. Rev. 28 (2), 337– 418, DOI: 10.1128/CMR.00117-1412https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXltlyhs7o%253D&md5=f481bdc9d3813f008c4c69502e9c9639The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteriaLi, Xian-Zhi; Plesiat, Patrick; Nikaido, HiroshiClinical Microbiology Reviews (2015), 28 (2), 337-418CODEN: CMIREX; ISSN:1098-6618. (American Society for Microbiology)The global emergence of multidrug-resistant Gram-neg. bacteria is a growing threat to antibiotic therapy. The chromosomally encoded drug efflux mechanisms that are ubiquitous in these bacteria greatly contribute to antibiotic resistance and present a major challenge for antibiotic development. Multidrug pumps, particularly those represented by the clin. relevant AcrAB-TolC and Mex pumps of the resistance-nodulation-division (RND) superfamily, not only mediate intrinsic and acquired multidrug resistance (MDR) but also are involved in other functions, including the bacterial stress response and pathogenicity. Addnl., efflux pumps interact synergistically with other resistance mechanisms (e.g., with the outer membrane permeability barrier) to increase resistance levels. Since the discovery of RND pumps in the early 1990s, remarkable scientific and technol. advances have allowed for an in-depth understanding of the structural and biochem. basis, substrate profiles, mol. regulation, and inhibition of MDR pumps. However, the development of clin. useful efflux pump inhibitors and/or new antibiotics that can bypass pump effects continues to be a challenge. Plasmid-borne efflux pump genes (including those for RND pumps) have increasingly been identified. This article highlights the recent progress obtained for organisms of clin. significance, together with methodol. considerations for the characterization of MDR pumps.
- 13Zabawa, T. P., Pucci, M. J., Parr, T. R., and Lister, T. (2016) Treatment of Gram-Negative Bacterial Infections by Potentiation of Antibiotics. Curr. Opin. Microbiol. 33, 7– 12, DOI: 10.1016/j.mib.2016.05.00513https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XotFOrtbY%253D&md5=ef37d51a73f4b36352f113243e55f189Treatment of Gram-negative bacterial infections by potentiation of antibioticsZabawa, Thomas P.; Pucci, Michael J.; Parr, Thomas R.; Lister, TroyCurrent Opinion in Microbiology (2016), 33 (), 7-12CODEN: COMIF7; ISSN:1369-5274. (Elsevier Ltd.)Infections caused by antibiotic-resistant pathogens, particularly Gram-neg. bacteria, represent significant treatment challenges for physicians resulting in high rates of morbidity and mortality. The outer membrane of Gram-neg. bacteria acts as a permeability barrier to many compds. that would otherwise be effective antibacterial agents, including those effective against Gram-pos. pathogens. Potentiator mols. disrupt this barrier allowing entry of otherwise impermeant mols., thus providing a strategy to render multi-drug resistant pathogens susceptible to a broader range of antibiotics. Potentiator mols. are cationic and the mechanism of disruption involves interaction with the neg. charged outer membrane. This phys. attribute, along with an often high degree of lipophilicity typically endears these mols. with unacceptable toxicity. Presented herein are examples of advanced potentiator mols. being evaluated for use in combination therapy for the treatment of resistant Gram-neg. infections.
- 14Ghosh, M. and Miller, M. J. (1996) Synthesis and in Vitro Antibacterial Activity of Spermidine-Based Mixed Catechol- and Hydroxamate-Containing Siderophore--Vancomycin Conjugates. Bioorg. Med. Chem. 4 (1), 43– 48, DOI: 10.1016/0968-0896(95)00161-114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xhs1agtbs%253D&md5=353be1938c9db511358c42da8654d606Synthesis and in vitro antibacterial activity of spermidine-based mixed catechol- and hydroxamate-containing siderophore-vancomycin conjugatesGhosh, Manuka; Miller, Marvin J.Bioorganic & Medicinal Chemistry (1996), 4 (1), 43-8CODEN: BMECEP; ISSN:0968-0896. (Elsevier)The first antibiotic conjugates of vancomycin with siderophore analogs contg. spermidine-based catechol ligands or mixed catechol and hydroxamate ligands are described. The design of the conjugates was based on the earlier observation that conjugation of siderophore components to β-lactam antibiotics induced active iron transport-mediated drug delivery. The novel conjugates were synthesized by selective acylation of the primary amino group of vancomycin. Preliminary biol. studies indicted that siderophore modified vancomycins lost some activity (4- to 16-fold) against Gram-pos. bacteria relative to vancomycin itself, and were generally similar to vancomycin in activity against Gram-neg. bacteria under iron-sufficient conditions. However, under iron-depleted conditions which mimic human serum, the conjugate contg. the spermidine-based catechol ligand displayed enhanced antibacterial activity against an antibiotic hypersensitive strain of Pseudomonas aeruginosa.
- 15Sarkar, P., Samaddar, S., Ammanathan, V., Yarlagadda, V., Ghosh, C., Shukla, M., Kaul, G., Manjithaya, R., Chopra, S., and Haldar, J. (2020) Vancomycin Derivative Inactivates Carbapenem-Resistant Acinetobacter Baumannii and Induces Autophagy. ACS Chem. Biol. 15 (4), 884– 889, DOI: 10.1021/acschembio.0c0009115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltlentbY%253D&md5=930db0ad9b8f9a67238d9ab7d5f5dc89Vancomycin Derivative Inactivates Carbapenem-Resistant Acinetobacter baumannii and Induces AutophagySarkar, Paramita; Samaddar, Sandip; Ammanathan, Veena; Yarlagadda, Venkateswarlu; Ghosh, Chandradhish; Shukla, Manjulika; Kaul, Grace; Manjithaya, Ravi; Chopra, Sidharth; Haldar, JayantaACS Chemical Biology (2020), 15 (4), 884-889CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Vancomycin is a std. drug for the treatment of multidrug-resistant Gram-pos. bacterial infections. Albeit, development of resistance (VRE, VRSA) and its inefficacy against persistent infections is a demerit. It is also intrinsically inactive against Gram-neg. bacteria. Herein, we report a vancomycin deriv., VanQAmC10, that addresses these challenges. VanQAmC10 was rapidly bactericidal against carbapenem-resistant A. baumannii (6 log10 CFU/mL redn. in 6 h), disrupted A. baumannii biofilms, and eradicated their stationary phase cells. In MRSA infected macrophages, the compd. reduced the bacterial burden by 1.3 log10 CFU/mL while vancomycin exhibited a static effect. Further investigation indicated that the compd., unlike vancomycin, promoted the intracellular degradative mechanism, autophagy, in mammalian cells, which may have contributed to its intracellular activity. The findings of the work provide new perspectives on the field of glycopeptide antibiotics.
- 16Neville, L. F., Shalit, I., Warn, P. A., Scheetz, M. H., Sun, J., Chosy, M. B., Wender, P. A., Cegelski, L., and Rendell, J. T. (2021) In Vivo Targeting of Escherichia Coli with Vancomycin-Arginine. Antimicrob. Agents Chemother. 65 (4), e02416– 20, DOI: 10.1128/AAC.02416-2016https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvVKru7Y%253D&md5=9a27ab1a8c2b613b8ede21e749d06facIn vivo targeting of Escherichia coli with vancomycin-arginineNeville, Lewis f.; Shalit, Itamar; Warn, Peter a.; Scheetz, Marc h.; Sun, Jiuzhi; Chosy, Madeline b.; Wender, Paul a.; Cegelski, Lynette; Rendell, Jacob t.Antimicrobial Agents and Chemotherapy (2021), 65 (4), e02416CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)The ability of vancomycin-arginine (V-r) to extend the spectrum of activity of glycopeptides to Gram-neg. bacteria was investigated. Its MIC towards Escherichia coli, including β-lactamase expressing Ambler classes A, B, and D, was 8 to 16 μg/mL. Addn. of 8 times the MIC of V-r to E. coli was acutely bactericidal and assocd. with a low frequency of resistance (<2.32 x 10-10). In vivo, V-r markedly reduced E. coli burden by >7 log10 CFU/g in a thigh muscle model. These data warrant further development of V-r in combating E. coli, including resistant forms.
- 17Antonoplis, A., Zang, X., Wegner, T., Wender, P. A., and Cegelski, L. (2019) Vancomycin-Arginine Conjugate Inhibits Growth of Carbapenem-Resistant E. Coli and Targets Cell-Wall Synthesis. ACS Chem. Biol. 14 (9), 2065– 2070, DOI: 10.1021/acschembio.9b0056517https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12jtrnP&md5=b1153f445707292b4d98469462d5b286Vancomycin-Arginine Conjugate Inhibits Growth of Carbapenem-Resistant E. coli and Targets Cell-Wall SynthesisAntonoplis, Alexandra; Zang, Xiaoyu; Wegner, Tristan; Wender, Paul A.; Cegelski, LynetteACS Chemical Biology (2019), 14 (9), 2065-2070CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The emergence of multi-drug-resistant Gram-neg. bacteria, including carbapenem-resistant Enterobacteriaceae, is a major health problem that necessitates the development of new antibiotics. Vancomycin inhibits cell-wall synthesis in Gram-pos. bacteria but is generally ineffective against Gram-neg. bacteria and is unable to penetrate the outer membrane barrier. To det. whether vancomycin and other antibiotics effective against Gram-pos. bacteria could, through modification, be rendered effective against Gram-neg. bacteria, we discovered that the covalent attachment of a single arginine to vancomycin yielded conjugates with order-of-magnitude improvements in activity against Gram-neg. bacteria, including pathogenic E. coli. The vancomycin-arginine conjugate (V-R) exhibited efficacy against actively growing bacteria, induced the loss of rod cellular morphol., and resulted in the intracellular accumulation of peptidoglycan precursors, all consistent with cell-wall synthesis disruption as its mechanism of action. Membrane permeabilization studies demonstrated an enhanced outer membrane permeability of V-R as compared with vancomycin. The conjugate exhibited no mammalian cell toxicity or hemolytic activity in MTT and hemolysis assays. Our study introduces a new vancomycin deriv. effective against Gram-neg. bacteria and underscores the broader potential of generating new antibiotics through combined mode-of-action and synthesis-informed design studies.
- 18Vaara, M. (1992) Agents That Increase the Permeability of the Outer Membrane. Microbiol. Rev. 56 (3), 395– 411, DOI: 10.1128/mr.56.3.395-411.199218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXjsFCktQ%253D%253D&md5=2416c67d643c9be119bfb1c0d2323b62Agents that increase the permeability of the outer membraneVaara, MarttiMicrobiological Reviews (1992), 56 (3), 395-411CODEN: MBRED3; ISSN:0146-0749.A review with 241 refs. on cationic agents and chelators as outer membrane permeabilizers, the mol. mechanism of outer membrane permeability increase, and bacterial strains resistant to the permeabilizers.
- 19Ofek, I., Cohen, S., Rahmani, R., Kabha, K., Tamarkin, D., Herzig, Y., and Rubinstein, E. (1994) Antibacterial Synergism of Polymyxin B Nonapeptide and Hydrophobic Antibiotics in Experimental Gram-Negative Infections in Mice. Antimicrob. Agents Chemother. 38 (2), 374– 377, DOI: 10.1128/AAC.38.2.37419https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXhvVagurw%253D&md5=ef5c957b98f952f4563426839261b79fAntibacterial synergism of polymyxin B nonapeptide and hydrophobic antibiotics in experimental gram-negative infections in miceOfek, Izhak; Cohen, Sofie; Rahmani, Rita; Kabha, Kisra; Tamarkin, Dove; Herzig, Yaacov; Rubinstein, EthanAntimicrobial Agents and Chemotherapy (1994), 38 (2), 374-7CODEN: AMACCQ; ISSN:0066-4804.Polymyxin B nonapeptide, derived by cleavage of the fatty acyl diaminobutryic acid from polymyxin B, is considerably less toxic, less bactericidal activity, and retains its ability to render gram-neg. bacteria susceptible to several antibiotics by permeabilizing their outer membranes. The peptide rendered all 53 polymyxin-susceptible strains tested more susceptible to novobiocin, lowering the MIC of novobiocin 8-fold or more. The combination of polymyxin B nonapeptide with novobiocin or with erythromycin administered i.p. in multiple doses synergistically protected mice infected with gram-neg. bacteria. This combination may be clin. useful because of the apparent rarity of the acquisition of resistance.
- 20Velkov, T., Thompson, P. E., Nation, R. L., and Li, J. (2010) Structure--Activity Relationships of Polymyxin Antibiotics. J. Med. Chem. 53 (5), 1898– 1916, DOI: 10.1021/jm900999h20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtleisLrP&md5=8156cb2be8057a3bd6403011ea47ad3cStructure-Activity Relationships of Polymyxin AntibioticsVelkov, Tony; Thompson, Philip E.; Nation, Roger L.; Li, JianJournal of Medicinal Chemistry (2010), 53 (5), 1898-1916CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review discussing the mechanisms of antibacterial activity of polymyxins, the mechanisms of resistance to polymyxins, the polymyxin-LPS complex, the synthesis of polymyxins, the structure-activity relationships of polymyxins, the polymyxin pharmacophore, and toxicol. properties of polymyxins.
- 21Chiharha, S., Ito, A., Yahata, M., Tobita, T., and Koyama, Y. (1974) Chemical Synthesis, Isolation and Characterization of α-N-Fattyacyl Colistin Nonapeptide with Special Reference to the Correlation between Antimicrobial Activity and Carbon Number of Fattyacyl Moiety. Agric. Biol. Chem. 38 (3), 521– 529, DOI: 10.1080/00021369.1974.10861184There is no corresponding record for this reference.
- 22Keirstead, N. D., Wagoner, M. P., Bentley, P., Blais, M., Brown, C., Cheatham, L., Ciaccio, P., Dragan, Y., Ferguson, D., Fikes, J., Galvin, M., Gupta, A., Hale, M., Johnson, N., Luo, W., McGrath, F., Pietras, M., Price, S., Sathe, A. G., Sasaki, J. C., Snow, D., Walsky, R. L., and Kern, G. (2014) Early Prediction of Polymyxin-Induced Nephrotoxicity with next-Generation Urinary Kidney Injury Biomarkers. Toxicol. Sci. 137 (2), 278– 291, DOI: 10.1093/toxsci/kft24722https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1elu7s%253D&md5=4d054fe2a41556455af798738444037dEarly Prediction of Polymyxin-Induced Nephrotoxicity With Next-Generation Urinary Kidney Injury BiomarkersKeirstead, Natalie D.; Wagoner, Matthew P.; Bentley, Patricia; Blais, Marie; Brown, Crystal; Cheatham, Letitia; Ciaccio, Paul; Dragan, Yvonne; Ferguson, Douglas; Fikes, Jim; Galvin, Melanie; Gupta, Anshul; Hale, Michael; Johnson, Nakpangi; Luo, Wenli; McGrath, Frank; Pietras, Mark; Price, Sally; Sathe, Abhishek G.; Sasaki, Jennifer C.; Snow, Debra; Walsky, Robert L.; Kern, GuntherToxicological Sciences (2014), 137 (2), 278-291CODEN: TOSCF2; ISSN:1096-0929. (Oxford University Press)Despite six decades of clin. experience with the polymyxin class of antibiotics, their dose-limiting nephrotoxicity remains difficult to predict due to a paucity of sensitive biomarkers. Here, we evaluate the performance of std. of care and next-generation biomarkers of renal injury in the detection and monitoring of polymyxin-induced acute kidney injury in male Han Wistar rats using colistin (polymyxin E) and a polymyxin B (PMB) deriv. with reduced nephrotoxicity, PMB nonapeptide (PMBN). This study provides the first histopathol. and biomarker anal. of PMBN, an important test of the hypothesis that fatty acid modifications and charge redns. in polymyxins can reduce their nephrotoxicity. The results indicate that alterations in a panel of urinary kidney injury biomarkers can be used to monitor histopathol. injury, with Kim-1 and α-GST emerging as the most sensitive biomarkers outperforming clin. stds. of care, serum or plasma creatinine and blood urea nitrogen. To enable the prediction of polymyxin-induced nephrotoxicity, an in vitro cytotoxicity assay was employed using human proximal tubule epithelial cells (HK-2). Cytotoxicity data in these HK-2 cells correlated with the renal toxicity detected via safety biomarker data and histopathol. evaluation, suggesting that in vitro and in vivo methods can be incorporated within a screening cascade to prioritize polymyxin class analogs with more favorable renal toxicity profiles.
- 23Vaara, M. (1983) Polymyxin B Nonapeptide Complexes with Lipopolysaccharide. FEMS Microbiol. Lett. 18 (1–2), 117– 121, DOI: 10.1111/j.1574-6968.1983.tb00461.x23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL3sXktlGru7s%253D&md5=89942f52c643635faa861f8e938d7f8fPolymyxin B nonapeptide complexes with lipopolysaccharideVaara, M.FEMS Microbiology Letters (1983), 18 (1-2), 117-21CODEN: FMLED7; ISSN:0378-1097.Polymyxin B nonapeptide (I) did not sensitize a polymyxin-resistant pmrA mutant strain of Salmonella typhimurium to the hydrophobic agents novobiocin and fusidic acid, in contrast to the parent strain, and I, like polymyxin, inhibited the biol. activities of isolated lipopolysaccharide (gelation of Limulus hemocyte lysate, consumption of complement). Apparently, I causes a disorganization of the outer membrane by binding to the acidic groups of the lipopolysaccharide, as does polymyxin.
- 24Brandenburg, K., Moriyon, I., Arraiza, M. D., Lewark-Yvetot, G., Koch, M. H. J., and Seydel, U. (2002) Biophysical Investigations into the Interaction of Lipopolysaccharide with Polymyxins. Thermochim. Acta 382 (1), 189– 198, DOI: 10.1016/S0040-6031(01)00731-624https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsV2qtg%253D%253D&md5=1834b8332ada1c60f3c89e42377c242aBiophysical investigations into the interaction of lipopolysaccharide with polymyxinsBrandenburg, K.; Moriyon, I.; Arraiza, M. D.; Lewark-Yvetot, G.; Koch, M. H. J.; Seydel, U.Thermochimica Acta (2002), 382 (1-2), 189-198CODEN: THACAS; ISSN:0040-6031. (Elsevier Science B.V.)It is reported on the interaction of bacterial lipopolysaccharide (LPS, endotoxin), having different sugar chain length covalently bound to its hydrophobic moiety lipid A, with the polycationic antibiotics polymyxin B (PMB) and PMB-nonapeptide (PMBN). The binding enthalpies and the lipid:peptide binding stoichiometries were detd. by isothermal titrn. calorimetry (ITC). For LPS with a long sugar chain (S: smooth form LPS from Yersinia enterocolitica), the titrn. curves exhibit a strong exo-therm which can be interpreted to result from the electrostatic interaction of the neg. charges of the LPS with the pos. charges of the peptides. In contrast, the titrn. curves of LPS with a short sugar chain (LPS Re from Escherichia coli) and of free lipid A yield complex patterns of endo- and exo-therms, which result from the superposition of electrostatic binding, fluidization of the acyl chains and a transition between different three-dimensional aggregate structures of the endo-toxins due to peptide binding. IR spectroscopy indicates that the fluidizing effect of the polymyxins is similar for both types of LPS and for lipid A. Small-angle X-ray diffraction reveals, however, that the Re-type LPS and lipid A are converted from a cubic into a multilamellar structure, whereas, the S-form LPS transforms from a unilamellar into a multilamellar structure with a small no. of lamellae. The presented data allow a better understanding of the interaction of peptides with endotoxin mols.
- 25Domalaon, R., Idowu, T., Zhanel, G. G., and Schweizer, F. (2018) Antibiotic Hybrids: The Next Generation of Agents and Adjuvants against Gram-Negative Pathogens?. Clin. Microbiol. Rev. 31 (2), e00077– 17, DOI: 10.1128/CMR.00077-1725https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1yjurvL&md5=cbaac686173643862c15d1a3b8f8e4ceAntibiotic hybrids: the next generation of agents and adjuvants against gram-negative pathogens?Domalaon, Ronald; Idowu, Temilolu; Zhanel, George G.; Schweizer, FrankClinical Microbiology Reviews (2018), 31 (2), e00077-17/1-e00077-17/45CODEN: CMIREX; ISSN:1098-6618. (American Society for Microbiology)The global incidence of drug-resistant Gram-neg. bacillary infections has been increasing, and there is a dire need to develop novel strategies to overcome this problem. Intrinsic resistance in Gram-neg. bacteria, such as their protective outer membrane and constitutively overexpressed efflux pumps, is a major survival weapon that renders them refractory to current antibiotics. Several potential avenues to overcome this problem have been at the heart of antibiotic drug discovery in the past few decades. We review some of these strategies, with emphasis on antibiotic hybrids either as stand-alone antibacterial agents or as adjuvants that potentiate a primary antibiotic in Gram-neg. bacteria. Antibiotic hybrid is defined in this review as a synthetic construct of two or more pharmacophores belonging to an established agent known to elicit a desired antimicrobial effect. The concepts, advances, and challenges of antibiotic hybrids are elaborated in this article. Moreover, we discuss several antibiotic hybrids that were or are in clin. evaluation. Mechanistic insights into how tobramycin-based antibiotic hybrids are able to potentiate legacy antibiotics in multidrug-resistant Gram-neg. bacilli are also highlighted. Antibiotic hybrids indeed have a promising future as a therapeutic strategy to overcome drug resistance in Gram-neg. pathogens and/or expand the usefulness of our current antibiotic arsenal.
- 26Duwe, A. K., Rupar, C. A., Horsman, G. B., and Vas, S. I. (1986) In Vitro Cytotoxicity and Antibiotic Activity of Polymyxin B Nonapeptide. Antimicrob. Agents Chemother. 30 (2), 340– 341, DOI: 10.1128/AAC.30.2.34026https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28XltFWqs78%253D&md5=8fe41e60b13039bda45e999d0e6a8656In vitro cytotoxicity and antibiotic activity of polymyxin B nonapeptideDuwe, Axel K.; Rupar, C. Anthony; Horsman, Greg B.; Vas, Stephen I.Antimicrobial Agents and Chemotherapy (1986), 30 (2), 340-1CODEN: AMACCQ; ISSN:0066-4804.Polymyxin B nonapeptide, prepd. by enzymic removal of the fatty acyl diaminobutyric acid side chain from polymyxin B, was ∼100-fold less toxic to K562 cells than polymyxin B. Min. inhibitory concns. of polymyxin B nonapeptide against a test panel of bacteria were 2-64-fold lower than those of polymyxin B.
- 27Domalaon, R., Yang, X., Lyu, Y., Zhanel, G. G., and Schweizer, F. (2017) Polymyxin B(3)-Tobramycin Hybrids with Pseudomonas Aeruginosa-Selective Antibacterial Activity and Strong Potentiation of Rifampicin, Minocycline, and Vancomycin. ACS Infect. Dis. 3 (12), 941– 954, DOI: 10.1021/acsinfecdis.7b0014527https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1KqsrjJ&md5=2f17be1865925aa4d8a33204e13d2d95Polymyxin B3-Tobramycin Hybrids with Pseudomonas aeruginosa-Selective Antibacterial Activity and Strong Potentiation of Rifampicin, Minocycline, and VancomycinDomalaon, Ronald; Yang, Xuan; Lyu, Yinfeng; Zhanel, George G.; Schweizer, FrankACS Infectious Diseases (2017), 3 (12), 941-954CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)There is an urgent need to develop novel antibacterial agents able to eradicate drug-resistant Gram-neg. pathogens such as Pseudomonas aeruginosa. Antimicrobial hybrids have emerged as a promising strategy to combat bacterial resistance, as a stand-alone drug but also as an adjuvant in combination with existing antibiotics. Herein, we report for the first time the synthesis and biol. evaluation of polymyxin-aminoglycoside heterodimers composed of polymyxin B3 covalently linked to tobramycin via an aliph. hydrocarbon linker. The polymyxin B3-tobramycin hybrids demonstrate potent activity against carbapenem-resistant as well as multidrug- or extensively drug-resistant (MDR/XDR) P. aeruginosa clin. isolates. Furthermore, the most potent hybrid was synergistic with currently used antibiotics against wild-type and MDR/XDR P. aeruginosa but also against Acinetobacter baumannii as well. The promising biol. activity described herein warrants addnl. studies into design and development of new antimicrobial hybrids able to surmount the problem of antimicrobial resistance.
- 28Luther, A., Urfer, M., Zahn, M., Müller, M., Wang, S.-Y., Mondal, M., Vitale, A., Hartmann, J.-B., Sharpe, T., Monte, F. Lo, Kocherla, H., Cline, E., Pessi, G., Rath, P., Modaresi, S. M., Chiquet, P., Stiegeler, S., Verbree, C., Remus, T., Schmitt, M., Kolopp, C., Westwood, M.-A., Desjonquères, N., Brabet, E., Hell, S., LePoupon, K., Vermeulen, A., Jaisson, R., Rithié, V., Upert, G., Lederer, A., Zbinden, P., Wach, A., Moehle, K., Zerbe, K., Locher, H. H., Bernardini, F., Dale, G. E., Eberl, L., Wollscheid, B., Hiller, S., Robinson, J. A., and Obrecht, D. (2019) Chimeric Peptidomimetic Antibiotics against Gram-Negative Bacteria. Nature 576 (7787), 452– 458, DOI: 10.1038/s41586-019-1665-628https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVagsLjJ&md5=209ed92936450f62dd2b0d7a96e90ba1Chimeric peptidomimetic antibiotics against Gram-negative bacteriaLuther, Anatol; Urfer, Matthias; Zahn, Michael; Muller, Maik; Wang, Shuang-Yan; Mondal, Milon; Vitale, Alessandra; Hartmann, Jean-Baptiste; Sharpe, Timothy; Monte, Fabio Lo; Kocherla, Harsha; Cline, Elizabeth; Pessi, Gabriella; Rath, Parthasarathi; Modaresi, Seyed Majed; Chiquet, Petra; Stiegeler, Sarah; Verbree, Carolin; Remus, Tobias; Schmitt, Michel; Kolopp, Caroline; Westwood, Marie-Anne; Desjonqueres, Nicolas; Brabet, Emile; Hell, Sophie; LePoupon, Karen; Vermeulen, Annie; Jaisson, Regis; Rithie, Virginie; Upert, Gregory; Lederer, Alexander; Zbinden, Peter; Wach, Achim; Moehle, Kerstin; Zerbe, Katja; Locher, Hans H.; Bernardini, Francesca; Dale, Glenn E.; Eberl, Leo; Wollscheid, Bernd; Hiller, Sebastian; Robinson, John A.; Obrecht, DanielNature (London, United Kingdom) (2019), 576 (7787), 452-458CODEN: NATUAS; ISSN:0028-0836. (Nature Research)There is an urgent need for new antibiotics against Gram-neg. pathogens that are resistant to carbapenem and 3rd-generation cephalosporins, against which antibiotics of last resort have lost most of their efficacy. We describe a class of synthetic antibiotics inspired by scaffolds derived from natural products. These chimeric antibiotics contain a β-hairpin peptide macrocycle linked to the macrocycle found in the polymyxin and colistin family of natural products. They are bactericidal and have a mechanism of action that involves binding to both lipopolysaccharide and the main component (BamA) of the β-barrel folding complex (BAM) that is required for the folding and insertion of β-barrel proteins into the outer membrane of Gram-neg. bacteria. Extensively optimized derivs. show potent activity against multidrug-resistant pathogens, including all of the Gram-neg. members of the ESKAPE pathogens. These derivs. also show favorable drug properties and overcome colistin resistance, both in vitro and in vivo. The lead candidate is currently in preclin. toxicol. studies that, if successful, will allow progress into clin. studies that have the potential to address life-threatening infections by the Gram-neg. pathogens, and thus to resolve a considerable unmet medical need.
- 29Guan, D., Chen, F., Qiu, Y., Jiang, B., Gong, L., Lan, L., and Huang, W. (2019) Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug-Resistant Bacteria. Angew. Chem., Int. Ed. 58 (20), 6678– 6682, DOI: 10.1002/anie.20190221029https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmvFWht7c%253D&md5=6dd58bcc9afae58d6bc9080f14a5cc45Sulfonium, an Underestimated Moiety for Structural Modification, Alters the Antibacterial Profile of Vancomycin Against Multidrug-Resistant BacteriaGuan, Dongliang; Chen, Feifei; Qiu, Yunguang; Jiang, Bofeng; Gong, Likun; Lan, Lefu; Huang, WeiAngewandte Chemie, International Edition (2019), 58 (20), 6678-6682CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)In the antibiotics arsenal, vancomycin is a last resort for the treatment of intractable infections. However, this situation is under threat because of the increasing appearance of vancomycin-resistant bacteria (VRB). Herein, we report a series of novel vancomycin derivs. carrying a sulfonium moiety. The sulfonium-vancomycin derivs. exhibited enhanced antibacterial activity against VRB both in vitro and in vivo. These derivs. also exhibited activity against some Gram-neg. bacteria. The sulfonium modification enhanced the interaction of vancomycin with the bacterial cell membrane and disrupts membrane integrity. Furthermore, the in vivo pharmacokinetic profile, stability, and toxicity of these derivs. demonstrated good druggability of the sulfonium-vancomycin analogs. This work provides a promising strategy for combating drug-resistant bacterial infection, and advances the knowledge on sulfonium derivs. for structural optimization and drug development.
- 30Shi, W., Chen, F., Zou, X., Jiao, S., Wang, S., Hu, Y., Lan, L., Tang, F., and Huang, W. (2021) Design, Synthesis, and Antibacterial Evaluation of Vancomycin-LPS Binding Peptide Conjugates. Bioorg. Med. Chem. Lett. 45, 128122, DOI: 10.1016/j.bmcl.2021.12812230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1yrsL%252FE&md5=922f291986a2bed64742b69bbb5cbf7aDesign, synthesis, and antibacterial evaluation of vancomycin-LPS binding peptide conjugatesShi, Weiwei; Chen, Feifei; Zou, Xiangman; Jiao, Shang; Wang, Siqi; Hu, Yu; Lan, Lefu; Tang, Feng; Huang, WeiBioorganic & Medicinal Chemistry Letters (2021), 45 (), 128122CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Developing novel antibiotics is urgently needed with emergency of drug resistance. Vancomycin, the last resort for intractable Gram-pos. bacterial infections, is ineffective against Gram-neg. bacteria and vancomycin resistant bacteria. Herein, we report a series of novel vancomycin derivs. carrying LPS binding peptides, vancomycin-LPS binding peptide conjugates (VPCs). The LPS binding peptides were conjugated onto 4 sites of vancomycin via CuAAC or maleimide- sulfydryl addn., and the formed VPCs were screened against VISA/VRE and Gram-neg. strains. VPCs exhibited enhanced activity against vancomycin resistant bacteria and obtained the activity against Gram-neg. bacteria in vitro, providing a novel strategy for vancomycin modification and glycopeptide antibiotics synthesis.
- 31Yarlagadda, V., Akkapeddi, P., Manjunath, G. B., and Haldar, J. (2014) Membrane Active Vancomycin Analogues: A Strategy to Combat Bacterial Resistance. J. Med. Chem. 57 (11), 4558– 4568, DOI: 10.1021/jm500270w31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntlKgtrk%253D&md5=30e565c1d5f25b1e8d69b83ea5cab397Membrane active vancomycin analogs: a strategy to combat bacterial resistanceYarlagadda, Venkateswarlu; Akkapeddi, Padma; Manjunath, Goutham B.; Haldar, JayantaJournal of Medicinal Chemistry (2014), 57 (11), 4558-4568CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The alarming growth of antibiotic-resistant superbugs such as vancomycin-resistant enterococci and staphylococci has become a major global health hazard. To address this issue, the authors report the development of lipophilic cationic vancomycin analogs possessing excellent antibacterial activity against several drug-resistant strains. Compared to vancomycin, efficacy greater than 1000-fold was demonstrated against vancomycin-resistant Enterococci (VRE). Significantly, unlike vancomycin, these compds. were shown to be bactericidal at low concns. and did not induce bacterial resistance. An optimized compd. in the series, compared to vancomycin, showed higher activity in methicillin-resistant Staphylococcus aureus (MRSA) infected mouse model and exhibited superior antibacterial activity in whole blood with no obsd. toxicity. The remarkable activity of these compds. is attributed to the incorporation of a new membrane disruption mechanism into vancomycin and opens up a great opportunity for the development of novel antibiotics.
- 32Okano, A., Isley, N. A., and Boger, D. L. (2017) Peripheral Modifications of [Ψ[CH2NH]Tpg4]Vancomycin with Added Synergistic Mechanisms of Action Provide Durable and Potent Antibiotics. Proc. Natl. Acad. Sci. U. S. A. 114, 5052– 5061, DOI: 10.1073/pnas.170412511432https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXovVSitr8%253D&md5=c98e49ee55fa7f42ab921a43e6d66ed2Peripheral modifications of [Ψ[CH2NH]Tpg4]vancomycin with added synergistic mechanisms of action provide durable and potent antibioticsOkano, Akinori; Isley, Nicholas A.; Boger, Dale L.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (26), E5052-E5061CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Subsequent to binding pocket modifications designed to provide dual d-Ala-d-Ala/d-Ala-d-Lac binding that directly overcome the mol. basis of vancomycin resistance, peripheral structural changes have been explored to improve antimicrobial potency and provide addnl. synergistic mechanisms of action. A C-terminal peripheral modification, introducing a quaternary ammonium salt, is reported and was found to provide a binding pocket-modified vancomycin analog with a second mechanism of action that is independent of d-Ala-d-Ala/d-Ala-d-Lac binding. This modification, which induces cell wall permeability and is complementary to the glycopeptide inhibition of cell wall synthesis, was found to provide improvements in antimicrobial potency (200-fold) against vancomycin-resistant Enterococci (VRE). Furthermore, it is shown that this type of C-terminal modification may be combined with a second peripheral (4-chlorobiphenyl)methyl (CBP) addn. to the vancomycin disaccharide to provide even more potent antimicrobial agents [VRE min. inhibitory concn. (MIC) = 0.01-0.005 μg/mL] with activity that can be attributed to three independent and synergistic mechanisms of action, only one of which requires d-Ala-d-Ala/d-Ala-d-Lac binding. Finally, it is shown that such peripherally and binding pocket-modified vancomycin analogs display little propensity for acquired resistance by VRE and that their durability against such challenges as well as their antimicrobial potency follow now predictable trends (three > two > one mechanisms of action). Such antibiotics are expected to display durable antimicrobial activity not prone to rapidly acquired clin. resistance.
- 33Blaskovich, M. A. T., Hansford, K. A., Gong, Y., Butler, M. S., Muldoon, C., Huang, J. X., Ramu, S., Silva, A. B., Cheng, M., Kavanagh, A. M., Ziora, Z., Premraj, R., Lindahl, F., Bradford, T. A., Lee, J. C., Karoli, T., Pelingon, R., Edwards, D. J., Amado, M., Elliott, A. G., Phetsang, W., Daud, N. H., Deecke, J. E., Sidjabat, H. E., Ramaologa, S., Zuegg, J., Betley, J. R., Beevers, A. P. G., Smith, R. A. G., Roberts, J. A., Paterson, D. L., and Cooper, M. A. (2018) Protein-Inspired Antibiotics Active against Vancomycin- and Daptomycin-Resistant Bacteria. Nat. Commun. 9 (1), 1– 17, DOI: 10.1038/s41467-017-02123-w33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpsVWhtLs%253D&md5=ee332a92ea1fedfa78f83f3b3fe834beProtein-inspired antibiotics active against vancomycin- and daptomycin-resistant bacteriaBlaskovich, Mark A. T.; Hansford, Karl A.; Gong, Yujing; Butler, Mark S.; Muldoon, Craig; Huang, Johnny X.; Ramu, Soumya; Silva, Alberto B.; Cheng, Mu; Kavanagh, Angela M.; Ziora, Zyta; Premraj, Rajaratnam; Lindahl, Fredrik; Bradford, Tanya A.; Lee, June C.; Karoli, Tomislav; Pelingon, Ruby; Edwards, David J.; Amado, Maite; Elliott, Alysha G.; Phetsang, Wanida; Daud, Noor Huda; Deecke, Johan E.; Sidjabat, Hanna E.; Ramaologa, Sefetogi; Zuegg, Johannes; Betley, Jason R.; Beevers, Andrew P. G.; Smith, Richard A. G.; Roberts, Jason A.; Paterson, David L.; Cooper, Matthew A.Nature Communications (2018), 9 (1), 1-17CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The public health threat posed by a looming 'post-antibiotic' era necessitates new approaches to antibiotic discovery. Drug development has typically avoided exploitation of membrane-binding properties, in contrast to nature's control of biol. pathways via modulation of membrane-assocd. proteins and membrane lipid compn. Here, we describe the rejuvenation of the glycopeptide antibiotic vancomycin via selective targeting of bacterial membranes. Peptide libraries based on pos. charged electrostatic effector sequences are ligated to N-terminal lipophilic membrane-insertive elements and then conjugated to vancomycin. These modified lipoglycopeptides, the 'vancapticins', possess enhanced membrane affinity and activity against methicillin-resistant Staphylococcus aureus (MRSA) and other Gram-pos. bacteria, and retain activity against glycopeptide-resistant strains. Optimized antibiotics show in vivo efficacy in multiple models of bacterial infection. This membrane-targeting strategy has potential to 'revitalise' antibiotics that have lost effectiveness against recalcitrant bacteria, or enhance the activity of other i.v.-administered drugs that target membrane-assocd. receptors.
- 34Wu, Z.-C., Cameron, M. D., and Boger, D. L. (2020) Vancomycin C-Terminus Guanidine Modifications and Further Insights into an Added Mechanism of Action Imparted by a Peripheral Structural Modification. ACS Infect. Dis. 6 (8), 2169– 2180, DOI: 10.1021/acsinfecdis.0c0025834https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1yntbbM&md5=3a506f57a34ebd0921392db6e6ed9248Vancomycin C-Terminus Guanidine Modifications and Further Insights into an Added Mechanism of Action Imparted by a Peripheral Structural ModificationWu, Zhi-Chen; Cameron, Michael D.; Boger, Dale L.ACS Infectious Diseases (2020), 6 (8), 2169-2180CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A series of vancomycin C-terminus guanidine modifications is disclosed that improves antimicrobial activity, enhances the durability of antimicrobial action against selection or induction of resistance, and introduces a synergistic mechanism of action independent of D-Ala-D-Ala binding and inhibition of cell wall biosynthesis. The added mechanism of action results in induced bacterial cell permeability, which we show may involve interaction with cell envelope teichoic acid. Significantly, the compds. examd. that contain two combined peripheral modifications, a (4-chlorobiphenyl)methyl (CBP) and C-terminus guanidinium modification, offer opportunities for new treatments against not only vancomycin-sensitive but esp. vancomycin-resistant bacteria where they act by two synergistic and now durable mechanisms of action independent of D-Ala-D-Ala/D-Lac binding and display superb antimicrobial potencies (MIC 0.6-0.15 μg/mL, VanA VRE). For the first time, we demonstrate that the synergistic behavior of the peripheral modifications examd. requires the presence of both the CBP and guanidine modifications in a single mol. vs. their combined use as an equimolar mixt. of singly modified compds. Finally, we show that a prototypical member of the series, G3-CBP-vancomycin (15), exhibits no hemolytic activity, displays no mammalian cell growth inhibition, possesses improved and esp. attractive in vivo pharmacokinetic (PK) properties, and displays excellent in vivo efficacy and potency against an esp. challenging multidrug-resistant (MRSA) and VanA vancomycin-resistant (VRSA) Staphylococcus aureus bacterial strain.
- 35Sarkar, P., Basak, D., Mukherjee, R., Bandow, J. E., and Haldar, J. Alkyl-Aryl-Vancomycins: Multimodal Glycopeptides with Weak Dependence on the Bacterial Metabolic State. J. Med. Chem. 2021 64 (14), 10185− 10202 DOI: 10.1021/acs.jmedchem.1c0044935https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVygsrnL&md5=d7cc9f643c569d95486b76337de078fdAlkyl-Aryl-Vancomycins: Multimodal Glycopeptides with Weak Dependence on the Bacterial Metabolic StateSarkar, Paramita; Basak, Debajyoti; Mukherjee, Riya; Bandow, Julia E.; Haldar, JayantaJournal of Medicinal Chemistry (2021), 64 (14), 10185-10202CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Resistance to last-resort antibiotics such as vancomycin for Gram-pos. bacterial infections necessitates the development of new therapeutics. Furthermore, the ability of bacteria to survive antibiotic therapy through formation of biofilms and persister cells complicates treatment. Toward this, we report alkyl-aryl-vancomycins (AAVs), with high potency against vancomycin-resistant enterococci and staphylococci. Unlike vancomycin, the lead compd. AAV-qC10 was bactericidal and weakly dependent on bacterial metab. This resulted in complete eradication of non-growing cells of MRSA and disruption of its biofilms. In addn. to inhibiting cell wall biosynthesis like vancomycin, AAV-qC10 also depolarizes and permeabilizes the membrane. More importantly, the compd. delocalized the cell division protein MinD, thereby impairing bacterial growth through multiple pathways. The potential of AAV-qC10 is exemplified by its superior efficacy against MRSA in a murine thigh infection model as compared to vancomycin. This work paves the way for structural optimization and drug development for combating drug-resistant bacterial infections.
- 36Rostovtsev, V. V., Green, L. G., Fokin, V. V., and Sharpless, K. B. (2002) A Stepwise Huisgen Cycloaddition Process: Copper(I)-Catalyzed Regioselective “Ligation” of Azides and Terminal Alkynes. Angew. Chem., Int. Ed. 41 (14), 2596– 2599, DOI: 10.1002/1521-3773(20020715)41:14<2596::AID-ANIE2596>3.0.CO;2-436https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xls1Ohsr4%253D&md5=4603664be6639353b5e70f19b9f8d59fA stepwise Huisgen cycloaddition process: copper(I)-catalyzed regioselective "ligation" of azides and terminal alkynesRostovtsev, Vsevolod V.; Green, Luke G.; Fokin, Valery V.; Sharpless, K. BarryAngewandte Chemie, International Edition (2002), 41 (14), 2596-2599CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH)1,4-Disubstituted 1,2,3-triazoles I (R1 = PhCH2, PhCH2OCH2, 1-adamantyl, etc.; R2 = HO2C, Ph, PhOCH2, Et2NCH2, etc.) were readily and cleanly prepd. via highly efficient and regioselective copper(I)-catalyzed cycloaddn. of azides R1N3 with terminal alkynes R2C≡CH in 82-93% yields.
- 37Tornøe, C. W., Christensen, C., and Meldal, M. (2002) Peptidotriazoles on Solid Phase: [1,2,3]-Triazoles by Regiospecific Copper(I)-Catalyzed 1,3-Dipolar Cycloadditions of Terminal Alkynes to Azides. J. Org. Chem. 67 (9), 3057– 3064, DOI: 10.1021/jo011148j37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisVeks7w%253D&md5=6b3b805572783873952871f1b69f46ddPeptidotriazoles on Solid Phase: [1,2,3]-Triazoles by Regiospecific Copper(I)-Catalyzed 1,3-Dipolar Cycloadditions of Terminal Alkynes to AzidesTornoe, Christian W.; Christensen, Caspar; Meldal, MortenJournal of Organic Chemistry (2002), 67 (9), 3057-3064CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)The cycloaddn. of azides to alkynes is one of the most important synthetic routes to 1H-[1,2,3]-triazoles. This work reports a novel regiospecific copper(I)-catalyzed 1,3-dipolar cycloaddn. of terminal alkynes to azides on solid-phase. Primary, secondary, and tertiary alkyl azides, aryl azides, and an azido sugar were used successfully in the copper(I)-catalyzed cycloaddn. producing diversely 1,4-substituted [1,2,3]-triazoles in peptide backbones or side chains. The reaction conditions were fully compatible with solid-phase peptide synthesis on polar supports. The copper(I) catalysis is mild and efficient (>95% conversion and purity in most cases) and furthermore, the x-ray structure of 2-azido-2-methylpropanoic acid has been solved, to yield structural information on the 1,3-dipoles entering the reaction. Novel Fmoc-protected amino azides were prepd. from Fmoc-amino alcs. by Mitsunobu reaction.
- 38Silverman, S. M., Moses, J. E., and Sharpless, K. B. (2017) Reengineering Antibiotics to Combat Bacterial Resistance: Click Chemistry [1,2,3]-Triazole Vancomycin Dimers with Potent Activity against MRSA and VRE. Chem. - Eur. J. 23 (1), 79– 83, DOI: 10.1002/chem.20160476538https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVGjtbvP&md5=614570f145edfc6e6579a44706b32f7cReengineering Antibiotics to Combat Bacterial Resistance: Click Chemistry [1,2,3]-Triazole Vancomycin Dimers with Potent Activity against MRSA and VRESilverman, Steven M.; Moses, John E.; Sharpless, K. BarryChemistry - A European Journal (2017), 23 (1), 79-83CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)Vancomycin has long been considered a drug of last resort. Its efficiency in treating multiple drug-resistant bacterial infections, particularly methicillin-resistant Staphylococcus aureus (MRSA), has had a profound effect on the treatment of life-threatening infections. However, the emergence of resistance to vancomycin is a cause for significant worldwide concern, prompting the urgent development of new effective treatments for antibiotic resistant bacterial infections. Harnessing the benefits of multivalency and cooperativity against vancomycin-resistant strains, we report a Click Chem. approach towards reengineered vancomycin derivs. and the synthesis of a no. of dimers with increased potency against MRSA and vancomycin resistant Enterococci (VRE; VanB). These semi-synthetic dimeric ligands were linked together with great efficiency using the powerful CuAAC reaction, demonstrating high levels of selectivity and purity.
- 39O’Dowd, H., Kim, B., Margolis, P., Wang, W., Wu, C., Lopez, S. L., and Blais, J. (2007) Preparation of Tetra-Boc-Protected Polymyxin B Nonapeptide. Tetrahedron Lett. 48 (11), 2003– 2005, DOI: 10.1016/j.tetlet.2007.01.07139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvVOrtrk%253D&md5=ce84a22fff3bb014682e102ada9a3de4Preparation of tetra-Boc-protected polymyxin B nonapeptideO'Dowd, Hardwin; Kim, Bum; Margolis, Peter; Wang, Wen; Wu, Charlotte; Lopez, Sara L.; Blais, JohanneTetrahedron Letters (2007), 48 (11), 2003-2005CODEN: TELEAY; ISSN:0040-4039. (Elsevier Ltd.)A method for the selective tetra-Boc-protection of polymyxin B nonapeptide (PMBN) has been developed. Boc-ON selectively protects the amino side chains of the four diaminobutyric acid (Dab) residues in the presence of the N-terminal free amine.
- 40Chevalier, S., Bouffartigues, E., Bodilis, J., Maillot, O., Lesouhaitier, O., Feuilloley, M. G. J., Orange, N., Dufour, A., and Cornelis, P. (2017) Structure, Function and Regulation of Pseudomonas Aeruginosa Porins. FEMS Microbiol. Rev. 41 (5), 698– 722, DOI: 10.1093/femsre/fux02040https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFGjsLnM&md5=9264cfacba2796f9e5f5fd367f4a423eStructure, function and regulation of Pseudomonas aeruginosa porinsChevalier, Sylvie; Bouffartigues, Emeline; Bodilis, Josselin; Maillot, Olivier; Lesouhaitier, Olivier; Feuilloley, Marc G. J.; Orange, Nicole; Dufour, Alain; Cornelis, PierreFEMS Microbiology Reviews (2017), 41 (5), 698-722CODEN: FMREE4; ISSN:1574-6976. (Oxford University Press)A review. Pseudomonas aeruginosa is a Gram-neg. bacterium belonging to the γ -proteobacteria. Like other members of the Pseudomonas genus, it is known for its metabolic versatility and its ability to colonize a wide range of ecol. niches, such as rhizosphere, water environments and animal hosts, including humans where it can cause severe infections. Another particularity of P. aeruginosa is its high intrinsic resistance to antiseptics and antibiotics, which is partly due to its low outer membrane permeability. In contrast to Enterobacteria, pseudomonads do not possess general diffusion porins in their outer membrane, but rather express specific channel proteins for the uptake of different nutrients. The major outer membrane 'porin', OprF, has been extensively investigated, and displays structural, adhesion and signaling functions while its role in the diffusion of nutrients is still under discussion. Other porins include OprB and OprB2 for the diffusion of glucose, the two small outer membrane proteins OprG and OprH, and the two porins involved in phosphate/pyrophosphate uptake, OprP and OprO. The remaining nineteen porins belong to the so-called OprD (Occ) family, which is further split into two subfamilies termed OccD (8 members) and OccK (11 members). In the past years, a large amt. of information concerning the structure, function and regulation of these porins has been published, justifying why an updated review is timely.
- 41Moore, R. A., Bates, N. C., and Hancock, R. E. (1986) Interaction of Polycationic Antibiotics with Pseudomonas Aeruginosa Lipopolysaccharide and Lipid A Studied by Using Dansyl-Polymyxin. Antimicrob. Agents Chemother. 29 (3), 496– 500, DOI: 10.1128/AAC.29.3.49641https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL28Xhs1Ghs78%253D&md5=96445b43f1deaf3b03cf778f7be35648Interaction of polycationic antibiotics with Pseudomonas aeruginosa lipopolysaccharide and lipid A studied by using dansyl-polymyxinMoore, Richard A.; Bates, Nancy C.; Hancock, Robert E. W.Antimicrobial Agents and Chemotherapy (1986), 29 (3), 496-500CODEN: AMACCQ; ISSN:0066-4804.A fluorescent deriv. of polymyxin B (dansyl-polymyxin) was used to study the interaction of polycations with lipopolysaccharide (LPS) and lipid A from P. aeruginosa. Dansyl-polymyxin became bound to LPS and lipid A sites, including Mg2+-binding sites, resulting in a 20-fold enhancement of fluorescence. A Hill plot of the binding data showed that the binding of dansyl-polymyxin to LPS was cooperative (n = 1.98) and of high affinity (S0.5 = 0.38 μM). The maximal binding capacity of LPS was ∼4 mol of dansyl-polymyxin/mol of LPS. The dansyl-polymyxin interaction with lipid A displayed similar kinetics (n = 2.26; S0.5 = 0.38 μM), and the maximal binding capacity was ∼2 mol of dansyl-polymyxin/mol lipid A. A variety of polycationic compds., including gentamicin, streptomycin, and polymyxin B, as well as Mg2+, were able to displace dansyl-polymyxin bound to LPS or to lipid A. Marked differences both in terms of the degree of displacement and in terms of the amt. of competing polycation required to displace a given amt. of dansyl-polymyxin were obsd. Addn. of excess polymyxin B resulted in displacement of all of the dansyl-polymyxin, demonstrating that only polymyxin-binding sites were being probed. Apparently, polymyxin B binds to multiple sites on LPS, including sites which bind aminoglycoside antibiotics and other polycationic compds.
- 42Viljanen, P. and Vaara, M. (1984) Susceptibility of Gram-Negative Bacteria to Polymyxin B Nonapeptide. Antimicrob. Agents Chemother. 25 (6), 701– 705, DOI: 10.1128/AAC.25.6.70142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXks12ht7g%253D&md5=f612e1b3713f440e844845f548399319Susceptibility of gram-negative bacteria to polymyxin B nonapeptideViljanen, P.; Vaara, M.Antimicrobial Agents and Chemotherapy (1984), 25 (6), 701-5CODEN: AMACCQ; ISSN:0066-4804.Subinhibitory concns. of polymyxin B nonapeptide sensitized all 21 polymyxin-susceptible gram-neg. bacterial strains studied to hydrophobic antibiotics such as fusidic acid, novobiocin, and erythromycin. The susceptibility increases were usually 30- and 300-fold. The strains included representatives of Escherichia coli with different O- and K-antigens, Klebsiella pneumoniae, K. oxytoca, Enterobacter cloacae, Enterobacter agglomerans, Salmonella typhimurium, Acinetobacter calcoaceticus, Pseudomonas aeruginosa, and P. maltophilia. In contrast, polymyxin-resistant strains (Proteus mirabilis, Proteus vulgaris, Morganella morganii, Providencia stuartii, and Serratia marcescens) were resistant to the action of polyyxin B nonapeptide.
- 43Healy, V. L., Lessard, I. A., Roper, D. I., Knox, J. R., and Walsh, C. T. (2000) Vancomycin Resistance in Enterococci: Reprogramming of the D-Ala-D-Ala Ligases in Bacterial Peptidoglycan Biosynthesis. Chem. Biol. 7, 109– 119, DOI: 10.1016/S1074-5521(00)00116-243https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXjvFenu78%253D&md5=843f504cb857b523e57ebb0fea4a0036Vancomycin resistance in enterococci: reprogramming of the D-Ala-D-Ala ligases in bacterial peptidoglycan biosynthesisHealy, Vicki L.; Lessard, Ivan A. D.; Roper, David I.; Knox, James R.; Walsh, Christopher T.Chemistry & Biology (2000), 7 (5), R109-R119CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Science Ltd.)A review with 53 refs. Vancomycin binds to bacterial cell wall intermediates to achieve its antibiotic effect. Infections of vancomycin-resistant enterococci are, however, becoming an increasing problem; the bacteria are resistant because they synthesize different cell wall intermediates. The enzymes involved in cell wall biosynthesis, therefore, are potential targets for combating this resistance. Recent biochem. and crystallog. results are providing mechanistic and structural details about some of these targets.
- 44Arnusch, C. J., Bonvin, A. M. J. J., Verel, A. M., Jansen, W. T. M., Liskamp, R. M. J., de Kruijff, B., Pieters, R. J., and Breukink, E. (2008) The Vancomycin–Nisin(1–12) Hybrid Restores Activity against Vancomycin Resistant Enterococci. Biochemistry 47 (48), 12661– 12663, DOI: 10.1021/bi801597b44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCns7fE&md5=cfdcca0f596cbe5272f7968bfd53f9f4The vancomycin-nisin(1-12) hybrid restores activity against vancomycin resistant enterococciArnusch, Christopher J.; Bonvin, Alexandre M. J. J.; Verel, Anne Marie; Jansen, Wouter T. M.; Liskamp, Rob M. J.; de Kruijff, Ben; Pieters, Roland J.; Breukink, EefjanBiochemistry (2008), 47 (48), 12661-12663CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Lipid II is a crucial component in bacterial cell wall synthesis. It is the target of a no. of important antibiotics, which include vancomycin and nisin. Here we show that a hybrid antibiotic that consists of vancomycin and nisin fragments is significantly more active than the sep. fragments against vancomycin-resistant enterococci. Three different hybrids were synthesized using click chem. and compared. Optimal spacer lengths and connection points were predicted using computer modeling.
- 45van Hal, S. J., Paterson, D. L., and Lodise, T. P. (2013) Systematic Review and Meta-Analysis of Vancomycin-Induced Nephrotoxicity Associated with Dosing Schedules That Maintain Troughs between 15 and 20 mg per Liter. Antimicrob. Agents Chemother. 57 (2), 734– 744, DOI: 10.1128/AAC.01568-1245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntVCrtL4%253D&md5=6c8b0b3b29b0f93ce978b2dda8571ef6Systematic review and meta-analysis of vancomycin-induced nephrotoxicity associated with dosing schedules that maintain troughs between 15 and 20 milligrams per litervan Hal, S. J.; Paterson, D. L.; Lodise, T. P.Antimicrobial Agents and Chemotherapy (2013), 57 (2), 734-744CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)In an effort to maximize outcomes, recent expert guidelines recommend more-intensive vancomycin dosing schedules to maintain vancomycin troughs between 15 and 20 mg/L. The widespread use of these more-intensive regimens has been assocd. with an increase in vancomycin-induced nephrotoxicity reports. The purpose of this systematic literature review is to det. the nephrotoxicity potential of maintaining higher troughs in clin. practice. All studies pertaining to vancomycin-induced nephrotoxicity between 1996 and Apr. 2012 were identified from PubMed, Embase, Cochrane Controlled Trial Registry, and Medline databases and analyzed according to Cochrane guidelines. Of the initial 240 studies identified, 38 were reviewed, and 15 studies met the inclusion criteria. Overall, higher troughs (≥15 mg/L) were assocd. with increased odds of nephrotoxicity (odds ratio [OR], 2.67; 95% confidence interval [CI], 1.95 to 3.65) relative to lower troughs of <15 mg/L. The relationship between a trough of ≥15 mg/L and nephrotoxicity persisted when the anal. was restricted to studies that examd. only initial trough concns. (OR, 3.12; 95% CI, 1.81 to 5.37). The relationship between troughs of ≥15 mg/L and nephrotoxicity persisted after adjustment for covariates known to independently increase the risk of a nephrotoxicity event. An incremental increase in nephrotoxicity was also obsd. with longer durations of vancomycin administration. Vancomycin-induced nephrotoxicity was reversible in the majority of cases, with short-term dialysis required only in 3% of nephrotoxic episodes. The collective literature indicates that an exposure-nephrotoxicity relationship for vancomycin exists. The probability of a nephrotoxic event increased as a function of the trough concn. and duration of therapy.
- 46Azad, M. A. K., Roberts, K. D., Yu, H. H., Liu, B., Schofield, A. V., James, S. A., Howard, D. L., Nation, R. L., Rogers, K., de Jonge, M. D., Thompson, P. E., Fu, J., Velkov, T., and Li, J. (2015) Significant Accumulation of Polymyxin in Single Renal Tubular Cells: A Medicinal Chemistry and Triple Correlative Microscopy Approach. Anal. Chem. 87 (3), 1590– 1595, DOI: 10.1021/ac504516k46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtlej&md5=e5079c031fe529836d002da09d6bfddeSignificant Accumulation of Polymyxin in Single Renal Tubular Cells: A Medicinal Chemistry and Triple Correlative Microscopy ApproachAzad, Mohammad A. K.; Roberts, Kade D.; Yu, Heidi H.; Liu, Boyin; Schofield, Alice V.; James, Simon A.; Howard, Daryl L.; Nation, Roger L.; Rogers, Kelly; de Jonge, Martin D.; Thompson, Philip E.; Fu, Jing; Velkov, Tony; Li, JianAnalytical Chemistry (Washington, DC, United States) (2015), 87 (3), 1590-1595CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Polymyxin is the last-line therapy against Gram-neg. 'superbugs'; however, dose-limiting nephrotoxicity can occur in up to 60% of patients after i.v. administration. Understanding the accumulation and concn. of polymyxin within renal tubular cells is essential for the development of novel strategies to ameliorate its nephrotoxicity and to develop safer, new polymyxins. We designed and synthesized a novel dual-modality iodine-labeled fluorescent probe for quant. mapping of polymyxin in kidney proximal tubular cells. Measured by synchrotron X-ray fluorescence microscopy, polymyxin concns. in single rat (NRK-52E) and human (HK-2) kidney tubular cells were approx. 1930- to 4760-fold higher than extracellular concns. Our study is the first to quant. measure the significant uptake of polymyxin in renal tubular cells and provides crucial information for the understanding of polymyxin-induced nephrotoxicity. Importantly, our approach represents a significant methodol. advancement in detn. of drug uptake for single-cell pharmacol.
- 47Sakamoto, Y., Yano, T., Hanada, Y., Takeshita, A., Inagaki, F., Masuda, S., Matsunaga, N., Koyanagi, S., and Ohdo, S. (2017) Vancomycin Induces Reactive Oxygen Species-Dependent Apoptosis via Mitochondrial Cardiolipin Peroxidation in Renal Tubular Epithelial Cells. Eur. J. Pharmacol. 800, 48– 56, DOI: 10.1016/j.ejphar.2017.02.02547https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1Klsb4%253D&md5=f2027e646c3dacbc9d6e92558755d224Vancomycin induces reactive oxygen species-dependent apoptosis via mitochondrial cardiolipin peroxidation in renal tubular epithelial cellsSakamoto, Yuya; Yano, Takahisa; Hanada, Yuki; Takeshita, Aki; Inagaki, Fumika; Masuda, Satohiro; Matsunaga, Naoya; Koyanagi, Satoru; Ohdo, ShigehiroEuropean Journal of Pharmacology (2017), 800 (), 48-56CODEN: EJPHAZ; ISSN:0014-2999. (Elsevier B.V.)Vancomycin (VCM) is a first-line antibiotic for serious infections caused by methicillin-resistant Staphylococcus aureus. However, nephrotoxicity is one of the most complaint in VCM therapy. We previously reported that VCM induced apoptosis in a porcine proximal tubular epithelial cell line (LLC-PK1), in which mitochondrial complex I may generate superoxide, leading to cell death. In the present study, VCM caused prodn. of mitochondrial reactive oxygen species and peroxidn. of the mitochondrial phospholipid cardiolipin that was reversed by administration of the mitochondrial uncoupler carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP). FCCP also significantly suppressed VCM-induced depolarization of the mitochondrial membrane and apoptosis. Moreover, the lipophilic antioxidant vitamin E and a mitochondria-targeted antioxidant, mitoTEMPO, also significantly suppressed VCM-induced depolarization of mitochondrial membrane and apoptosis, whereas vitamin C, n-acetyl cysteine, or glutathione did not provide significant protection. These findings suggest that peroxidn. of the mitochondrial membrane cardiolipin mediated the VCM-induced prodn. of intracellular reactive oxygen species and initiation of apoptosis in LLC-PK1 cells. Furthermore, regulation of mitochondrial function using a mitochondria-targeted antioxidant, such as mitoTEMPO, may constitute a potential strategy for mitigation of VCM-induced proximal tubular epithelial cell injury.
- 48Jansen, J., Schophuizen, C. M. S., Wilmer, M. J., Lahham, S. H. M., Mutsaers, H. A. M., Wetzels, J. F. M., Bank, R. A., van den Heuvel, L. P., Hoenderop, J. G., and Masereeuw, R. (2014) A Morphological and Functional Comparison of Proximal Tubule Cell Lines Established from Human Urine and Kidney Tissue. Exp. Cell Res. 323 (1), 87– 99, DOI: 10.1016/j.yexcr.2014.02.01148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjslOhuro%253D&md5=751059bcefbd159e429f53d11a3970d6A morphological and functional comparison of proximal tubule cell lines established from human urine and kidney tissueJansen, J.; Schophuizen, C. M. S.; Wilmer, M. J.; Lahham, S. H. M.; Mutsaers, H. A. M.; Wetzels, J. F. M.; Bank, R. A.; van den Heuvel, L. P.; Hoenderop, J. G.; Masereeuw, R.Experimental Cell Research (2014), 323 (1), 87-99CODEN: ECREAL; ISSN:0014-4827. (Elsevier B.V.)Promising renal replacement therapies include the development of a bioartificial kidney using functional human kidney cell models. In this study, human conditionally immortalized proximal tubular epithelial cell (ciPTEC) lines originating from kidney tissue (ciPTEC-T1 and ciPTEC-T2) were compared to ciPTEC previously isolated from urine (ciPTEC-U).Subclones of all ciPTEC isolates formed tight cell layers on Transwell inserts as detd. by transepithelial resistance, inulin diffusion, E-cadherin expression and immunocytochemisty. Extracellular matrix genes collagen I and -IV α1 were highly present in both kidney tissue derived matured cell lines (p<0.001) compared to matured ciPTEC-U, whereas matured ciPTEC-U showed a more pronounced fibronectin I and laminin 5 gene expression (p<0.01 and p<0.05, resp.). Expression of the influx carrier Org. Cation Transporter 2 (OCT-2), and the efflux pumps P-glycoprotein (P-gp), Multidrug Resistance Protein 4 (MRP4) and Breast Cancer Resistance Protein (BCRP) were confirmed in the three cell lines using real-time PCR and Western blotting. The activities of OCT-2 and P-gp were sensitive to specific inhibition in all models (p<0.001). The highest activity of MRP4 and BCRP was demonstrated in ciPTEC-U (p<0.05). Finally, active albumin reabsorption was highest in ciPTEC-T2 (p<0.001), while Na+-dependent phosphate reabsorption was most abundant in ciPTEC-U (p<0.01).In conclusion, ciPTEC established from human urine or kidney tissue display comparable functional PTEC specific transporters and physiol. characteristics, providing ideal human tools for bioartificial kidney development.
- 49Nieskens, T. T. G., Peters, J. G. P., Schreurs, M. J., Smits, N., Woestenenk, R., Jansen, K., van der Made, T. K., Röring, M., Hilgendorf, C., Wilmer, M. J., and Masereeuw, R. (2016) A Human Renal Proximal Tubule Cell Line with Stable Organic Anion Transporter 1 and 3 Expression Predictive for Antiviral-Induced Toxicity. AAPS J. 18 (2), 465– 475, DOI: 10.1208/s12248-016-9871-849https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvFSiurg%253D&md5=303c4ad4c9977e39981b967b3f4466d6A Human Renal Proximal Tubule Cell Line with Stable Organic Anion Transporter 1 and 3 Expression Predictive for Antiviral-Induced ToxicityNieskens, Tom T. G.; Peters, Janny G. P.; Schreurs, Marieke J.; Smits, Niels; Woestenenk, Rob; Jansen, Katja; van der Made, Thom K.; Roering, Melanie; Hilgendorf, Constanze; Wilmer, Martijn J.; Masereeuw, RosalindeAAPS Journal (2016), 18 (2), 465-475CODEN: AJAOB6; ISSN:1550-7416. (Springer)Drug-induced nephrotoxicity still hampers drug development, because current translation from in vitro or animal studies to human lacks high predictivity. Often, renal adverse effects are recognized only during clin. stages of drug development. The current study aimed to establish a robust and a more complete human cell model suitable for screening of drug-related interactions and nephrotoxicity. In addn. to endogenously expressed renal org. cation transporters and efflux transporters, conditionally immortalized proximal tubule epithelial cells (ciPTEC) were completed by transduction of cells with the org. anion transporter (OAT) 1 or OAT3. Fluorescence-activated cell sorting upon exposure to the OAT substrate fluorescein successfully enriched transduced cells. A panel of org. anions was screened for drug-interactions in ciPTEC-OAT1 and ciPTEC-OAT3. The cytotoxic response to the drug-interactions with antivirals was further examd. by cell viability assays. Upon subcloning, concn.-dependent fluorescein uptake was found with a higher affinity for ciPTEC-OAT1 (Km = 0.8 ± 0.1 μM) than ciPTEC-OAT3 (Km = 3.7 ± 0.5 μM). Co-exposure to known OAT1 and/or OAT3 substrates (viz. para-aminohippurate, estrone sulfate, probenecid, furosemide, diclofenac, and cimetidine) in cultures spanning 29 passage nos. revealed relevant inhibitory potencies, confirming the robustness of our model for drug-drug interactions studies. Functional OAT1 was directly responsible for cytotoxicity of adefovir, cidofovir, and tenofovir, while a drug interaction with zidovudine was not assocd. with decreased cell viability. Our data demonstrate that human-derived ciPTEC-OAT1 and ciPTEC-OAT3 are promising platforms for highly predictive drug screening during early phases of drug development.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.1c00318.
Synthetic schemes and synthesis of all building blocks; analytical RP-HPLC traces, HRMS analysis and yields for all new compounds synthesized; additional MIC analysis of building blocks and control compounds; MIC analysis in μM; hemolytic and PTEC toxicity data (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.