1
|
He X, Karlsson PA, Xiong R, Moodie LWK, Wang H, Bergström CAS, Hubert M. Liquid crystal nanoparticles for oral combination antibiotic therapies: A strategy towards protecting commensal gut bacteria during treatment. J Colloid Interface Sci 2025; 678:287-300. [PMID: 39245019 DOI: 10.1016/j.jcis.2024.08.230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Antibiotics are essential for treating infections and reducing risks during medical interventions. However, many commonly used antibiotics lack the physiochemical properties for an efficient oral administration when treating systemic infection. Instead, we are reliant on intravenous delivery, which presents complications outside of clinical settings. Developing novel formulations for oral administration is a potential solution to this problem. We engineered hexosome and cubosome liquid crystal nanoparticles (LCNPs) characterized by small-angle X-ray scattering and cryogenic transmission electron microscopy, and could encapsulate the antibiotics vancomycin (VAN) and clarithromycin (CLA) with high loading efficiencies. By rationally choosing stable lipid building blocks, the loaded LCNPs demonstrated excellent resilience against enzymatic degradation in an in vitro gut model LCNP stability is crucial as premature antibiotic leakage can negatively impact the gut microbiota. In screens against the representative gut bacteria Enterococcus faecalis and Escherichia coli, our LCNPs provided a protective effect. Furthermore, we explored co-administration and dual loading strategies of VAN and CLA, and demonstrated effective loading, stability and protection for E. faecalis and E. coli. This work represents a proof of concept for the early-stage development of antibiotic-loaded LCNPs to treat systemic infection via oral administration, opening opportunities for combination antibiotic therapies.
Collapse
Affiliation(s)
- Xiguo He
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Philip A Karlsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ruisheng Xiong
- Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - Lindon W K Moodie
- Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helen Wang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Madlen Hubert
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
Bian X, Chen Z, Li F, Xie Y, Li Y, Luo Y, Zou X, Wang H, Zhang J, Wang X, Zhang J, Guan D. Single Amine or Guanidine Modification on Norvancomycin and Vancomycin to Overcome Multidrug-Resistance through Augmented Lipid II Binding and Increased Membrane Activity. J Med Chem 2024; 67:20639-20663. [PMID: 39504470 DOI: 10.1021/acs.jmedchem.4c02196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Vancomycin and norvancomycin have diminished antibacterial efficacy due to acquired or intrinsic resistance from mutations in the terminal dipeptide of lipid II in Gram-positive bacteria or failure to penetrate into the periplasm in Gram-negative bacteria. Herein, we rationally designed and synthesized a series of vancomycin analogues bearing single amine or guanidine functionality, altering various linkers and modification sites, to combat the resistance. Extensive antibacterial screening was performed to delineate a comprehensive SAR. Many derivatives revitalized the activity in vitro, exhibiting a 4-128-fold or 2-16-fold enhancement against the acquired or intrinsic resistance with lower toxicity. Significantly, the optimal compound 4g demonstrated greater pharmacokinetic and pharmacodynamic profiles. Further studies uncovered additional independent and synergistic mechanisms for 4g, including the enhanced membrane activity and augmented inhibition of peptidoglycan biosynthesis via increased lipid II binding, highlighting its potential as a future lead candidate to replenish the glycopeptide antibiotic arsenal.
Collapse
Affiliation(s)
- Xiaolei Bian
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Fang Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Yuanyuan Xie
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Yi Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Youhong Luo
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang 261031, China
| | - Xiangman Zou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Hui Wang
- Nanjing Cantech Microbial Technology Co. Ltd., No. 18, Zhilan Rd, Jiangning, Nanjing 211100, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang 261031, China
| | - Xiaowen Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Dongliang Guan
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
3
|
Rahn HP, Liu X, Chosy MB, Sun J, Cegelski L, Wender PA. Biguanide-Vancomycin Conjugates are Effective Broad-Spectrum Antibiotics against Actively Growing and Biofilm-Associated Gram-Positive and Gram-Negative ESKAPE Pathogens and Mycobacteria. J Am Chem Soc 2024; 146:22541-22552. [PMID: 39088791 PMCID: PMC11624893 DOI: 10.1021/jacs.4c06520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Strategies to increase the efficacy and/or expand the spectrum of activity of existing antibiotics provide a potentially fast path to clinically address the growing crisis of antibiotic-resistant infections. Here, we report the synthesis, antibacterial efficacy, and mechanistic activity of an unprecedented class of biguanide-antibiotic conjugates. Our lead biguanide-vancomycin conjugate, V-C6-Bg-PhCl (5e), induces highly effective cell killing with up to a 2 orders-of-magnitude improvement over its parent compound, vancomycin (V), against vancomycin-resistant enterococcus. V-C6-Bg-PhCl (5e) also exhibits improved activity against mycobacteria and each of the ESKAPE pathogens, including the Gram-negative organisms. Furthermore, we uncover broad-spectrum killing activity against biofilm-associated Gram-positive and Gram-negative bacteria as well as mycobacteria not observed for clinically used antibiotics such as oritavancin. Mode-of-action studies reveal that vancomycin-like cell wall synthesis inhibition with improved efficacy attributed to enhanced engagement at vancomycin binding sites through biguanide association with relevant cell-surface anions for Gram-positive and Gram-negative bacteria. Due to its potency, remarkably broad activity, and lack of acute mammalian cell toxicity, V-C6-Bg-PhCl (5e) is a promising candidate for treating antibiotic-resistant infections and notoriously difficult-to-treat slowly growing and antibiotic-tolerant bacteria associated with chronic and often incurable infections. More generally, this study offers a new strategy (biguanidinylation) to enhance antibiotic activity and facilitate clinical entry.
Collapse
Affiliation(s)
- Harrison P. Rahn
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Xinyu Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Madeline B. Chosy
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
van Groesen E, Mons E, Kotsogianni I, Arts M, Tehrani KHME, Wade N, Lysenko V, Stel FM, Zwerus JT, De Benedetti S, Bakker A, Chakraborty P, van der Stelt M, Scheffers DJ, Gooskens J, Smits WK, Holden K, Gilmour PS, Willemse J, Hitchcock CA, van Hasselt JGC, Schneider T, Martin NI. Semisynthetic guanidino lipoglycopeptides with potent in vitro and in vivo antibacterial activity. Sci Transl Med 2024; 16:eabo4736. [PMID: 39110780 DOI: 10.1126/scitranslmed.abo4736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/23/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
Gram-positive bacterial infections present a major clinical challenge, with methicillin- and vancomycin-resistant strains continuing to be a cause for concern. In recent years, semisynthetic vancomycin derivatives have been developed to overcome this problem as exemplified by the clinically used telavancin, which exhibits increased antibacterial potency but has also raised toxicity concerns. Thus, glycopeptide antibiotics with enhanced antibacterial activities and improved safety profiles are still necessary. We describe the development of a class of highly potent semisynthetic glycopeptide antibiotics, the guanidino lipoglycopeptides, which contain a positively charged guanidino moiety bearing a variable lipid group. These glycopeptides exhibited enhanced in vitro activity against a panel of Gram-positive bacteria including clinically relevant methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant strains, showed minimal toxicity toward eukaryotic cells, and had a low propensity for resistance selection. Mechanistically, guanidino lipoglycopeptides engaged with bacterial cell wall precursor lipid II with a higher binding affinity than vancomycin. Binding to both wild-type d-Ala-d-Ala lipid II and the vancomycin-resistant d-Ala-d-Lac variant was confirmed, providing insight into the enhanced activity of guanidino lipoglycopeptides against vancomycin-resistant isolates. The in vivo efficacy of guanidino lipoglycopeptide EVG7 was evaluated in a S. aureus murine thigh infection model and a 7-day sepsis survival study, both of which demonstrated superiority to vancomycin. Moreover, the minimal to mild kidney effects at supratherapeutic doses of EVG7 indicate an improved therapeutic safety profile compared with vancomycin. These findings position guanidino lipoglycopeptides as candidates for further development as antibacterial agents for the treatment of clinically relevant multidrug-resistant Gram-positive infections.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Elma Mons
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Kamaleddin H M E Tehrani
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Nicola Wade
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Vladyslav Lysenko
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Florence M Stel
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Jordy T Zwerus
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | - Stefania De Benedetti
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Alexander Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Parichita Chakraborty
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, 2300 RA Leiden, Netherlands
| | - Dirk-Jan Scheffers
- Department of Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9700 AB Groningen, Netherlands
| | - Jairo Gooskens
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Wiep Klaas Smits
- Experimental Bacteriology, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Kirsty Holden
- Evotec (U.K.) Ltd., Alderley Park, Macclesfield, Cheshire, SK10 4TG UK
| | | | - Joost Willemse
- Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| | | | - J G Coen van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, Netherlands
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, 53113 Bonn, Germany
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, 2300 RA Leiden, Netherlands
| |
Collapse
|
5
|
McGlumphy S, Damai A, Salameh L, Corbin GB, Wang Q, Markiewicz J, Mosher JJ, Spitzer N, Quiñones R. Biocompatible antibiotic-coupled nickel-titanium nanoparticles as a potential coating material for biomedical devices. Heliyon 2024; 10:e31434. [PMID: 38831845 PMCID: PMC11145499 DOI: 10.1016/j.heliyon.2024.e31434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
The challenges facing metallic implants for reconstructive surgery include the leaching of toxic metal ions, a mismatch in elastic modulus between the implant and the treated tissue, and the risk of infection. These problems can be addressed by passivating the metal surface with an organic substrate and incorporating antibiotic molecules. Nitinol (NiTi), a nickel-titanium alloy, is used in devices for biomedical applications due to its shape memory and superelasticity. However, unmodified NiTi carries a risk of localized nickel toxicity and inadequately supports angiogenesis or neuroregeneration due to limited cell adhesion, poor biomineralization, and little antibacterial activity. To address these challenges, NiTi nanoparticles were modified using self-assembled phosphonic acid monolayers and functionalized with the antibiotics ceftriaxone and vancomycin via the formation of an amide. Surface modifications were monitored to confirm that phosphonic acid modifications were present on NiTi nanoparticles and 100% of the samples formed ordered films. Modifications were stable for more than a year. Elemental composition showed the presence of nickel, titanium, and phosphorus (1.9% for each sample) after surface modifications. Dynamic light scattering analysis suggested some agglomeration in solution. However, scanning electron microscopy coupled with energy-dispersive X-ray spectroscopy confirmed a particle size distribution of <100 nm, the even distribution of nanoparticles on coverslips, and elemental composition before and after cell culture. B35 neuroblastoma cells exhibited no inhibition of survival and extended neurites of approximately 100 μm in total length when cultured on coverslips coated with only poly-l-lysine or with phosphonic acid-modified NiTi, indicating high biocompatibility. The ability to support neural cell growth and differentiation makes modified NiTi nanoparticles a promising coating for surfaces in metallic bone and nerve implants. NiTi nanoparticles functionalized with ceftriaxone inhibited Escherichia coli and Serratia marcescens (SM6) at doses of 375 and 750 μg whereas the growth of Bacillus subtilis was inhibited by a dose of only 37.5 μg. NiTi-vancomycin was effective against B. subtilis at all doses even after mammalian cell culture. These are common bacteria associated with infected implants, further supporting the potential use of functionalized NiTi in coating reconstructive implants.
Collapse
Affiliation(s)
- Sarah McGlumphy
- Department of Chemistry, Marshall University, Huntington, WV, 25755, USA
- Department of Biological Sciences, Marshall University, Huntington, WV, 25755, USA
| | - Aakriti Damai
- Department of Chemistry, Marshall University, Huntington, WV, 25755, USA
- Department of Biological Sciences, Marshall University, Huntington, WV, 25755, USA
| | - Lena Salameh
- Department of Chemistry, Marshall University, Huntington, WV, 25755, USA
| | - Gabriell B. Corbin
- Department of Biological Sciences, Marshall University, Huntington, WV, 25755, USA
| | - Qiang Wang
- Shared Research Facilities, West Virginia University, Morgantown, WV, 25606, USA
| | - John Markiewicz
- Department of Chemistry, Marshall University, Huntington, WV, 25755, USA
| | - Jennifer J. Mosher
- Department of Biological Sciences, Marshall University, Huntington, WV, 25755, USA
| | - Nadja Spitzer
- Department of Biological Sciences, Marshall University, Huntington, WV, 25755, USA
| | - Rosalynn Quiñones
- Department of Chemistry, Marshall University, Huntington, WV, 25755, USA
| |
Collapse
|
6
|
Nithiya P, Alagarsamy G, Sathish PB, Rajarathnam D, Li X, Jeyaraj S, Satheesh M, Selvakumar R. Impact of effluent parameters and vancomycin concentration on vancomycin resistant Escherichia coli and its host specific bacteriophage lytic activity in hospital effluent. ENVIRONMENTAL RESEARCH 2024; 247:118334. [PMID: 38316381 DOI: 10.1016/j.envres.2024.118334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/07/2024]
Abstract
Vancomycin resistance in bacteria has been classified under high priority category by World Health Organization (WHO) and its presence in hospital effluent is reported to be increasing owing to excess antibiotics use. Among various strategies, bacteriophage has been recently considered as a promising biological agent for combating such antimicrobial resistant bacteria (ARB). However, the influence of effluent's properties on phage-ARB interaction in actual hospital effluent is not completely understood. The present works intends to study this influence of hospital effluent and its parameters on the interaction between vancomycin resistant E. coli (VRE) and its host specific bacteriophage. The isolated VRE was identified by 16S rRNA sequencing, matrix-assisted laser desorption/ionization-time of flight (MALDI - TOF) and whole genome sequencing. The infectivity of phage onto host bacteria was investigated using electron microscopic techniques, dynamic light scattering (DLS), spectrofluorophotometer and confirmed using double agar overlay method. The monovalency and polyvalency of isolated phage against various bacterial species were determined. The phage morphology was identical to T7 phage belonging to Podoviridae. The phage lysis was maximum at pH 7 (90.2%), 37 °C (91.6%) and vancomycin concentration of 50 μg/mL in both synthetic media (89.13%) and effluent (100%). At a maximum vancomycin concentration of 100 μg/mL, decrease in Ca, K, Mg and P (up to 19.70, 14.18, 28, and 15.82% respectively) concentration in effluent was observed due to phage infectivity when compared to control. The whole genome sequencing was performed and the bioinformatics analysis presented the role of mdfA gene encoding the efflux pump in causing vancomycin resistance in E. coli. It also depicted the presence of multiple genes responsible for mercury, cobalt, zinc and cadmium resistance in VRE. These results clearly indicate that bacteriophage mediated combating of VRE is possible in actual hospital effluent and can be used as one of the treatment methods.
Collapse
Affiliation(s)
- P Nithiya
- Department of Nanobiotechnology, PSG Institute of Advanced Studies, Coimbatore, 641004, India
| | - G Alagarsamy
- Department of Nanobiotechnology, PSG Institute of Advanced Studies, Coimbatore, 641004, India
| | - P B Sathish
- Department of Nanobiotechnology, PSG Institute of Advanced Studies, Coimbatore, 641004, India
| | - D Rajarathnam
- Australian Centre for Water and Environmental Biotechnology, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Xu Li
- Department of Civil and Environmental Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sankarganesh Jeyaraj
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, 641004, India; PSG Center for Genetics and Molecular Biology, Off Avinashi Road, Coimbatore, 641004, India
| | - Manjima Satheesh
- PSG Center for Molecular Medicine and Therapeutics, PSG Institute of Medical Sciences and Research, Coimbatore, 641004, India; PSG Center for Genetics and Molecular Biology, Off Avinashi Road, Coimbatore, 641004, India
| | - R Selvakumar
- Department of Nanobiotechnology, PSG Institute of Advanced Studies, Coimbatore, 641004, India.
| |
Collapse
|
7
|
Chosy MB, Sun J, Rahn HP, Liu X, Brčić J, Wender PA, Cegelski L. Vancomycin-Polyguanidino Dendrimer Conjugates Inhibit Growth of Antibiotic-Resistant Gram-Positive and Gram-Negative Bacteria and Eradicate Biofilm-Associated S. aureus. ACS Infect Dis 2024; 10:384-397. [PMID: 38252999 PMCID: PMC11646489 DOI: 10.1021/acsinfecdis.3c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The global challenge of antibiotic resistance necessitates the introduction of more effective antibiotics. Here we report a potentially general design strategy, exemplified with vancomycin, that improves and expands antibiotic performance. Vancomycin is one of the most important antibiotics in use today for the treatment of Gram-positive infections. However, it fails to eradicate difficult-to-treat biofilm populations. Vancomycin is also ineffective in killing Gram-negative bacteria due to its inability to breach the outer membrane. Inspired by our seminal studies on cell penetrating guanidinium-rich transporters (e.g., octaarginine), we recently introduced vancomycin conjugates that effectively eradicate Gram-positive biofilm bacteria, persister cells and vancomycin-resistant enterococci (with V-r8, vancomycin-octaarginine), and Gram-negative pathogens (with V-R, vancomycin-arginine). Having shown previously that the spatial array (linear versus dendrimeric) of multiple guanidinium groups affects cell permeation, we report here for the first time vancomycin conjugates with dendrimerically displayed guanidinium groups that exhibit superior efficacy and breadth, presenting the best activity of V-r8 and V-R in single broad-spectrum compounds active against ESKAPE pathogens. Mode-of-action studies reveal cell-surface activity and enhanced vancomycin-like killing. The vancomycin-polyguanidino dendrimer conjugates exhibit no acute mammalian cell toxicity or hemolytic activity. Our study introduces a new class of broad-spectrum vancomycin derivatives and a general strategy to improve or expand antibiotic performance through combined mode-of-action and function-oriented design studies.
Collapse
Affiliation(s)
- Madeline B. Chosy
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Harrison P. Rahn
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Xinyu Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jasna Brčić
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Brčić J, Tong A, Wender PA, Cegelski L. Conjugation of Vancomycin with a Single Arginine Improves Efficacy against Mycobacteria by More Effective Peptidoglycan Targeting. J Med Chem 2023; 66:10226-10237. [PMID: 37477249 PMCID: PMC10783851 DOI: 10.1021/acs.jmedchem.3c00565] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Drug resistant bacterial infections have emerged as one of the greatest threats to public health. The discovery and development of new antimicrobials and anti-infective strategies are urgently needed to address this challenge. Vancomycin is one of the most important antibiotics for the treatment of Gram-positive infections. Here, we introduce the vancomycin-arginine conjugate (V-R) as a highly effective antimicrobial against actively growing mycobacteria and difficult-to-treat mycobacterial biofilm populations. Further improvement in efficacy through combination treatment of V-R to inhibit peptidoglycan synthesis and ethambutol to inhibit arabinogalactan synthesis underscores the ability to identify compound synergies to more effectively target the Achilles heel of the cell-wall assembly. Moreover, we introduce mechanistic activity data and a molecular model derived from a d-Ala-d-Ala-bound vancomycin structure that we hypothesize underlies the molecular basis for the antibacterial improvement attributed to the arginine modification that is specific to peptidoglycan chemistry employed by mycobacteria and distinct from Gram-positive pathogens.
Collapse
Affiliation(s)
- Jasna Brčić
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Alan Tong
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
9
|
Werby SH, Brčić J, Chosy MB, Sun J, Rendell JT, Neville LF, Wender PA, Cegelski L. Detection of intact vancomycin-arginine as the active antibacterial conjugate in E. coli by whole-cell solid-state NMR. RSC Med Chem 2023; 14:1192-1198. [PMID: 37360389 PMCID: PMC10285746 DOI: 10.1039/d3md00173c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/13/2023] [Indexed: 06/28/2023] Open
Abstract
The introduction of new and improved antibacterial agents based on facile synthetic modifications of existing antibiotics represents a promising strategy to deliver urgently needed antibacterial candidates to treat multi-drug resistant bacterial infections. Using this strategy, vancomycin was transformed into a highly active agent against antibiotic-resistant Gram-negative organisms in vitro and in vivo through the addition of a single arginine to yield vancomycin-arginine (V-R). Here, we report detection of the accumulation of V-R in E. coli by whole-cell solid-state NMR using 15N-labeled V-R. 15N CPMAS NMR revealed that the conjugate remained fully amidated without loss of arginine, demonstrating that intact V-R represents the active antibacterial agent. Furthermore, C{N}REDOR NMR in whole cells with all carbons at natural abundance 13C levels exhibited the sensitivity and selectivity to detect the directly bonded 13C-15N pairs of V-R within E. coli cells. Thus, we also present an effective methodology to directly detect and evaluate active drug agents and their accumulation within bacteria without the need for potentially perturbative cell lysis and analysis protocols.
Collapse
Affiliation(s)
- Sabrina H Werby
- Department of Chemistry, Stanford University Stanford CA 94305 USA
| | - Jasna Brčić
- Department of Chemistry, Stanford University Stanford CA 94305 USA
| | - Madeline B Chosy
- Department of Chemistry, Stanford University Stanford CA 94305 USA
| | - Jiuzhi Sun
- Department of Chemistry, Stanford University Stanford CA 94305 USA
| | | | | | - Paul A Wender
- Department of Chemistry, Stanford University Stanford CA 94305 USA
- Department of Chemical and Systems Biology, Stanford University Stanford CA 94305 USA
| | - Lynette Cegelski
- Department of Chemistry, Stanford University Stanford CA 94305 USA
| |
Collapse
|
10
|
Almeida MC, da Costa PM, Sousa E, Resende DISP. Emerging Target-Directed Approaches for the Treatment and Diagnosis of Microbial Infections. J Med Chem 2023; 66:32-70. [PMID: 36586133 DOI: 10.1021/acs.jmedchem.2c01212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With the rising levels of drug resistance, developing efficient antimicrobial therapies has become a priority. A promising strategy is the conjugation of antibiotics with relevant moieties that can potentiate their activity by target-directing. The conjugation of siderophores with antibiotics allows them to act as Trojan horses by hijacking the microorganisms' highly developed iron transport systems and using them to carry the antibiotic into the cell. Through the analysis of relevant examples of the past decade, this Perspective aims to reveal the potential of siderophore-antibiotic Trojan horses for the treatment of infections and the role of siderophores in diagnostic techniques. Other conjugated molecules will be the subject of discussion, namely those involving vitamin B12, carbohydrates, and amino acids, as well as conjugated compounds targeting protein degradation and β-lactamase activated prodrugs.
Collapse
Affiliation(s)
- Mariana C Almeida
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, FFUP - Faculdade de Farmácia, Universidade do Porto, Rua de Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.,CIIMAR- Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Paulo M da Costa
- CIIMAR- Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, FFUP - Faculdade de Farmácia, Universidade do Porto, Rua de Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.,CIIMAR- Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Diana I S P Resende
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, FFUP - Faculdade de Farmácia, Universidade do Porto, Rua de Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.,CIIMAR- Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| |
Collapse
|
11
|
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. Sci Rep 2022; 12:20921. [PMID: 36463278 PMCID: PMC9719540 DOI: 10.1038/s41598-022-24807-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Gram-negative bacteria possess intrinsic resistance to glycopeptide antibiotics so these important antibacterial medications are only suitable for the treatment of Gram-positive bacterial infections. At the same time, polymyxins are peptide antibiotics, structurally related to glycopeptides, with remarkable activity against Gram-negative bacteria. With the aim of breaking the intrinsic resistance of Gram-negative bacteria against glycopeptides, a polycationic vancomycin aglycone derivative carrying an n-decanoyl side chain and five aminoethyl groups, which resembles the structure of polymyxins, was prepared. Although the compound by itself was not active against the Gram-negative bacteria tested, it synergized with teicoplanin against Escherichia coli, Pseudomonas aeruginosa and Acinetobacter baumannii, and it was able to potentiate vancomycin against these Gram-negative strains. Moreover, it proved to be active against vancomycin- and teicoplanin-resistant Gram-positive bacteria.
Collapse
|
12
|
van Groesen E, Innocenti P, Martin NI. Recent Advances in the Development of Semisynthetic Glycopeptide Antibiotics: 2014-2022. ACS Infect Dis 2022; 8:1381-1407. [PMID: 35895325 PMCID: PMC9379927 DOI: 10.1021/acsinfecdis.2c00253] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The accelerated appearance of drug-resistant bacteria poses an ever-growing threat to modern medicine's capacity to fight infectious diseases. Gram-positive species such as methicillin-resistant Staphylococcus aureus (MRSA) and Streptococcus pneumoniae continue to contribute significantly to the global burden of antimicrobial resistance. For decades, the treatment of serious Gram-positive infections relied upon the glycopeptide family of antibiotics, typified by vancomycin, as a last line of defense. With the emergence of vancomycin resistance, the semisynthetic glycopeptides telavancin, dalbavancin, and oritavancin were developed. The clinical use of these compounds is somewhat limited due to toxicity concerns and their unusual pharmacokinetics, highlighting the importance of developing next-generation semisynthetic glycopeptides with enhanced antibacterial activities and improved safety profiles. This Review provides an updated overview of recent advancements made in the development of novel semisynthetic glycopeptides, spanning the period from 2014 to today. A wide range of approaches are covered, encompassing innovative strategies that have delivered semisynthetic glycopeptides with potent activities against Gram-positive bacteria, including drug-resistant strains. We also address recent efforts aimed at developing targeted therapies and advances made in extending the activity of the glycopeptides toward Gram-negative organisms.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University 2333 BE Leiden, The Netherlands
| | - Paolo Innocenti
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University 2333 BE Leiden, The Netherlands
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University 2333 BE Leiden, The Netherlands
| |
Collapse
|
13
|
Acharya Y, Bhattacharyya S, Dhanda G, Haldar J. Emerging Roles of Glycopeptide Antibiotics: Moving beyond Gram-Positive Bacteria. ACS Infect Dis 2022; 8:1-28. [PMID: 34878254 DOI: 10.1021/acsinfecdis.1c00367] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glycopeptides, a class of cell wall biosynthesis inhibitors, have been the antibiotics of choice against drug-resistant Gram-positive bacterial infections. Their unique mechanism of action involving binding to the substrate of cell wall biosynthesis and substantial longevity in clinics makes this class of antibiotics an attractive choice for drug repurposing and reprofiling. However, resistance to glycopeptides has been observed due to alterations in the substrate, cell wall thickening, or both. The emergence of glycopeptide resistance has resulted in the development of synthetic and semisynthetic glycopeptide analogues to target acquired resistance. Recent findings demonstrate that these derivatives, along with some of the FDA approved glycopeptides have been shown to have antimicrobial activity against Gram-negative bacteria, Mycobacteria, and viruses thus expanding their spectrum of activity across the microbial kingdom. Additional mechanisms of action and identification of novel targets have proven to be critical in broadening the spectrum of activity of glycopeptides. This review focuses on the applications of glycopeptides beyond their traditional target group of Gram-positive bacteria. This will aid in making the scientific community aware about the nontraditional activity profiles of glycopeptides, identify the existing loopholes, and further explore this antibiotic class as a potential broad-spectrum antimicrobial agent.
Collapse
Affiliation(s)
- Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Shaown Bhattacharyya
- Biochemistry and Molecular Biology Program, Departments of Chemistry and Biology, College of Arts and Science, Boston University, Boston, Massachusetts 02215, United States
| | - Geetika Dhanda
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
14
|
Neville LF, Shalit I, Warn PA, Rendell JT. OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1706-1709. [PMID: 35229156 PMCID: PMC9155633 DOI: 10.1093/jac/dkac063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives STM-001, a retargeted glycopeptide, is active against MDR E. coli expressing ESBLs including carbapenemases. Herein, we assessed its capability to combat E. coli complicated urinary tract infections (cUTI) in mice driven by clinically important serine (CTX-M-15) and metallo-β-lactamases (NDM-1). Methods Plasma and urine pharmacokinetics following IV administration of STM-001 (1–50 mg/kg) were determined in mice via LC-MS/MS. The effects on bacterial burden (kidney, bladder and urine) were determined in a 7 day mouse cUTI model whereby STM-001 was administered q12h or q24h at 2–100 mg/kg/day from Day 4. Efficacy was assessed by the change in log10 cfu/g or log10 cfu/mL from vehicle-treated infected mice. Results MICs of STM-001 for CTX-M-15 and NDM-1 E. coli were 8 and 16 mg/L, respectively. Blood pharmacokinetic profile was linear and dose-dependent with low clearance of 9.49 ± 0.31 mL/min/kg, V = 0.63 ± 0.02 L/kg and t½ = 1.16 ± 0.03 h. High STM-001 concentrations were recovered in urine 0–8 h post-administration, reaching up to 120-fold above its MIC. In cUTI efficacy studies, STM-001 (1–50 mg/kg, q12h) reduced CTX-M-15 burden by log10 4.31 (kidney), 3.95 (bladder) and 4.82 (urine) compared with vehicle-treated animals (P < 0.0001). STM-001 also reduced NDM-1 burden by log10 3.89 (kidney), 3.76 (bladder) and 3.08 (urine) (P < 0.0001), with similar inhibitory effects following q24h dosing. Conclusions STM-001 was highly effective in reducing E. coli burden in kidney, bladder and urine in mouse cUTI models. The observed efficacy with either dosing regimen indicates potential low humanized doses of 1–5 mg/kg. These data support further development of STM-001 as an innovative, carbapenem-sparing antibiotic to combat human cUTIs.
Collapse
Affiliation(s)
- Lewis F. Neville
- SuperTrans Medical Ltd, Tel Aviv-Jaffa, Israel
- Corresponding author. E-mail:
| | | | | | | |
Collapse
|
15
|
Poulhazan A, Dickwella Widanage MC, Muszyński A, Arnold AA, Warschawski DE, Azadi P, Marcotte I, Wang T. Identification and Quantification of Glycans in Whole Cells: Architecture of Microalgal Polysaccharides Described by Solid-State Nuclear Magnetic Resonance. J Am Chem Soc 2021; 143:19374-19388. [PMID: 34735142 PMCID: PMC8630702 DOI: 10.1021/jacs.1c07429] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 12/15/2022]
Abstract
Microalgae are photosynthetic organisms widely distributed in nature and serve as a sustainable source of bioproducts. Their carbohydrate components are also promising candidates for bioenergy production and bioremediation, but the structural characterization of these heterogeneous polymers in cells remains a formidable problem. Here we present a widely applicable protocol for identifying and quantifying the glycan content using magic-angle-spinning (MAS) solid-state NMR (ssNMR) spectroscopy, with validation from glycosyl linkage and composition analysis deduced from mass-spectrometry (MS). Two-dimensional 13C-13C correlation ssNMR spectra of a uniformly 13C-labeled green microalga Parachlorella beijerinckii reveal that starch is the most abundant polysaccharide in a naturally cellulose-deficient strain, and this polymer adopts a well-organized and highly rigid structure in the cell. Some xyloses are present in both the mobile and rigid domains of the cell wall, with their chemical shifts partially aligned with the flat-ribbon 2-fold xylan identified in plants. Surprisingly, most other carbohydrates are largely mobile, regardless of their distribution in glycolipids or cell walls. These structural insights correlate with the high digestibility of this cellulose-deficient strain, and the in-cell ssNMR methods will facilitate the investigations of other economically important algae species.
Collapse
Affiliation(s)
- Alexandre Poulhazan
- Department
of Chemistry, University of Quebec at Montreal, Montreal H2X 2J6, Canada
| | | | - Artur Muszyński
- Complex
Carbohydrate Research Center, University
of Georgia, Athens, Georgia 30602, United States
| | - Alexandre A. Arnold
- Department
of Chemistry, University of Quebec at Montreal, Montreal H2X 2J6, Canada
| | - Dror E. Warschawski
- Laboratoire
des Biomolécules, LBM, CNRS UMR 7203,
Sorbonne Université, École Normale Supérieure,
PSL University, 75005 Paris, France
| | - Parastoo Azadi
- Complex
Carbohydrate Research Center, University
of Georgia, Athens, Georgia 30602, United States
| | - Isabelle Marcotte
- Department
of Chemistry, University of Quebec at Montreal, Montreal H2X 2J6, Canada
| | - Tuo Wang
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| |
Collapse
|
16
|
New potentiators of ineffective antibiotics: Targeting the Gram-negative outer membrane to overcome intrinsic resistance. Curr Opin Chem Biol 2021; 66:102099. [PMID: 34808425 DOI: 10.1016/j.cbpa.2021.102099] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022]
Abstract
Because of the rise in antibiotic resistance and the dwindling pipeline of effective antibiotics, it is imperative to explore avenues that breathe new life into existing drugs. This is particularly important for intrinsically resistant Gram-negative bacteria, which are exceedingly difficult to treat. The Gram-negative outer membrane (OM) prevents the entry of a plethora of antibiotics that are effective against Gram-positive bacteria, despite the presence of the targets of these drugs. Uncovering molecules that increase the permeability of the OM to sensitize Gram-negative bacteria to otherwise ineffective antibiotics is an approach that has recently garnered increased attention in the field. In this review, we survey chemical matter which has been shown to potentiate antibiotics against Gram-negative bacteria by perturbing the OM. These include peptides, nanoparticles, macromolecules, antibiotic conjugates, and small molecules.
Collapse
|
17
|
van Groesen E, Slingerland CJ, Innocenti P, Mihajlovic M, Masereeuw R, Martin NI. Vancomyxins: Vancomycin-Polymyxin Nonapeptide Conjugates That Retain Anti-Gram-Positive Activity with Enhanced Potency against Gram-Negative Strains. ACS Infect Dis 2021; 7:2746-2754. [PMID: 34387988 PMCID: PMC8438664 DOI: 10.1021/acsinfecdis.1c00318] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
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.
Collapse
Affiliation(s)
- Emma van Groesen
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Cornelis J. Slingerland
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Paolo Innocenti
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| |
Collapse
|
18
|
Zeiders SM, Chmielewski J. Antibiotic-cell-penetrating peptide conjugates targeting challenging drug-resistant and intracellular pathogenic bacteria. Chem Biol Drug Des 2021; 98:762-778. [PMID: 34315189 DOI: 10.1111/cbdd.13930] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/09/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022]
Abstract
The failure to treat everyday bacterial infections is a current threat as pathogens are finding new ways to thwart antibiotics through mechanisms of resistance and intracellular refuge, thus rendering current antibiotic strategies ineffective. Cell-penetrating peptides (CPPs) are providing a means to improve antibiotics that are already approved for use. Through coadministration and conjugation of antibiotics with CPPs, improved accumulation and selectivity with alternative and/or additional modes of action against infections have been observed. Herein, we review the recent progress of this antibiotic-cell-penetrating peptide strategy in combatting sensitive and drug-resistant pathogens. We take a closer look into the specific antibiotics that have been enhanced, and in some cases repurposed as broad-spectrum drugs. Through the addition and conjugation of cell-penetrating peptides to antibiotics, increased permeation across mammalian and/or bacterial membranes and a broader range in bacterial selectivity have been achieved.
Collapse
Affiliation(s)
| | - Jean Chmielewski
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|