1
|
Keck JM, Viteri A, Schultz J, Fong R, Whitman C, Poush M, Martin M. New Agents Are Coming, and So Is the Resistance. Antibiotics (Basel) 2024; 13:648. [PMID: 39061330 PMCID: PMC11273847 DOI: 10.3390/antibiotics13070648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance is a global threat that requires urgent attention to slow the spread of resistant pathogens. The United States Centers for Disease Control and Prevention (CDC) has emphasized clinician-driven antimicrobial stewardship approaches including the reporting and proper documentation of antimicrobial usage and resistance. Additional efforts have targeted the development of new antimicrobial agents, but narrow profit margins have hindered manufacturers from investing in novel antimicrobials for clinical use and therefore the production of new antibiotics has decreased. In order to combat this, both antimicrobial drug discovery processes and healthcare reimbursement programs must be improved. Without action, this poses a high probability to culminate in a deadly post-antibiotic era. This review will highlight some of the global health challenges faced both today and in the future. Furthermore, the new Infectious Diseases Society of America (IDSA) guidelines for resistant Gram-negative pathogens will be discussed. This includes new antimicrobial agents which have gained or are likely to gain FDA approval. Emphasis will be placed on which human pathogens each of these agents cover, as well as how these new agents could be utilized in clinical practice.
Collapse
Affiliation(s)
- J. Myles Keck
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alina Viteri
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | - Rebecca Fong
- Department of Pharmacy, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Charles Whitman
- Department of Pharmacy, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Madeline Poush
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marlee Martin
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
2
|
Hammerle F, Fiala J, Höck A, Huymann L, Vrabl P, Husiev Y, Bonnet S, Peintner U, Siewert B. Fungal Anthraquinone Photoantimicrobials Challenge the Dogma of Cationic Photosensitizers. JOURNAL OF NATURAL PRODUCTS 2023; 86:2247-2257. [PMID: 37708055 PMCID: PMC10616806 DOI: 10.1021/acs.jnatprod.2c01157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 09/16/2023]
Abstract
The photoantimicrobial potential of four mushroom species (i.e., Cortinarius cinnabarinus, C. holoxanthus, C. malicorius, and C. sanguineus) was explored by studying the minimal inhibitory concentrations (MIC) via a light-modified broth microdilution assay based on the recommended protocols of the European Committee on Antimicrobial Susceptibility Testing (EUCAST). The extracts were tested against Candida albicans, Escherichia coli, and Staphylococcus aureus under blue (λ = 428 and 478 nm, H = 30 J/cm2) and green light (λ = 528 nm, H = 30 J/cm2) irradiation. Three extracts showed significant photoantimicrobial effects at concentrations below 25 μg/mL. Targeted isolation of the major pigments from C. sanguineus led to the identification of two new potent photoantimicrobials, one of them (i.e., dermocybin) being active against S. aureus and C. albicans under green light irradiation [PhotoMIC530 = 39.5 μM (12.5 μg/mL) and 2.4 μM (0.75 μg/mL), respectively] and the other one (i.e., emodin) being in addition active against E. coli in a low micromolar range [PhotoMIC428 = 11.1 μM (3 μg/mL)]. Intriguingly, dermocybin was not (photo)cytotoxic against the three tested cell lines, adding an additional level of selectivity. Since both photoantimicrobials are not charged, this discovery shifts the paradigm of cationic photosensitizers.
Collapse
Affiliation(s)
- Fabian Hammerle
- Department
of Department of Pharmacognosy, Institute of Pharmacy, CCB −
Centrum of Chemistry and Biomedicine, CMBI − Center for Molecular
Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Johannes Fiala
- Department
of Department of Pharmacognosy, Institute of Pharmacy, CCB −
Centrum of Chemistry and Biomedicine, CMBI − Center for Molecular
Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Anja Höck
- Department
of Department of Pharmacognosy, Institute of Pharmacy, CCB −
Centrum of Chemistry and Biomedicine, CMBI − Center for Molecular
Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
- Department
of Biotechnology & Food Engineering, MCI-The Entrepreneurial School, 6020 Innsbruck, Austria
| | - Lesley Huymann
- Department
of Department of Pharmacognosy, Institute of Pharmacy, CCB −
Centrum of Chemistry and Biomedicine, CMBI − Center for Molecular
Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
- Institute
of Microbiology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Pamela Vrabl
- Institute
of Microbiology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Yurii Husiev
- Leiden
Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands
| | - Sylvestre Bonnet
- Leiden
Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands
| | - Ursula Peintner
- Institute
of Microbiology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Bianka Siewert
- Department
of Department of Pharmacognosy, Institute of Pharmacy, CCB −
Centrum of Chemistry and Biomedicine, CMBI − Center for Molecular
Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Wang H, Xie Y, Chen Y, Zhao H, Lv X, Zhang Z, Li G, Pan J, Wang J, Liu Z. Transdermal Delivery of Photosensitizer-Catalase Conjugate by Fluorinated Polyethylenimine for Enhanced Topical Photodynamic Therapy of Bacterial Infections. Adv Healthc Mater 2023; 12:e2300848. [PMID: 37178381 DOI: 10.1002/adhm.202300848] [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: 03/16/2023] [Revised: 04/25/2023] [Indexed: 05/15/2023]
Abstract
Pathogenic bacteria induce subcutaneous infections pose serious threats to global public health. Recently, photodynamic therapy (PDT) has been proposed as a non-invasive approach for anti-microbial treatment without the risk to induce drug resistance. However, due to the hypoxic environment of most anaerobiont-infected sites, the therapeutic efficacy of oxygen consuming PDT has been limited. Herein, a transdermal delivery system is reported to allow effective delivery of photosensitizers into infected skin for PDT treatment of skin infections by bacteria. Considering the overproduction of hydrogen peroxide (H2 O2 ) in the abscess area, catalase (CAT), an enzyme that triggers H2 O2 decomposition to generate O2 , is conjugated with chlorine e6 (Ce6) to form a photosensitizer conjugate (Ce6-CAT) as an enhanced PDT agent against Staphylococcus Aureus. After screening a series of fluorinated low molecular weight polyethylenimine (F-PEI) with different fluorination degrees, the optimized F-PEI formulation is identified with the best transdermal delivery ability system. Upon mixing, the formed Ce6-CAT@F-PEI nanocomplex shows effective transdermal penetration after being applied to the skin surface. With light exposure of the infected skin, highly effective in vivo anti-bacterial PDT therapeutic effect with Ce6-CAT@F-PEI is observed. This work proposes a transdermal PDT therapeutic nanomedicine particularly promising for the anti-bacterial treatment of skin infections.
Collapse
Affiliation(s)
- Hairong Wang
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yi Xie
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yanling Chen
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - He Zhao
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xinjing Lv
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zimu Zhang
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Gen Li
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jian Pan
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jian Wang
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
4
|
Abstract
TonB-dependent transporters (TBDTs) are present in all gram-negative bacteria and mediate energy-dependent uptake of molecules that are too scarce or large to be taken up efficiently by outer membrane (OM) diffusion channels. This process requires energy that is derived from the proton motive force and delivered to TBDTs by the TonB-ExbBD motor complex in the inner membrane. Together with the need to preserve the OM permeability barrier, this has led to an extremely complex and fascinating transport mechanism for which the fundamentals, despite decades of research, are still unclear. In this review, we describe our current understanding of the transport mechanism of TBDTs, their potential role in the delivery of novel antibiotics, and the important contributions made by TBDT-associated (lipo)proteins.
Collapse
Affiliation(s)
- Augustinas Silale
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom; ,
| | - Bert van den Berg
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom; ,
| |
Collapse
|
5
|
Al-Mijalli SH, Mrabti NN, Ouassou H, Sheikh RA, Abdallah EM, Assaggaf H, Bakrim S, Alshahrani MM, Awadh AAA, Qasem A, Attar A, Lee LH, Bouyahya A, Goh KW, Ming LC, Mrabti HN. Phytochemical Variability, In Vitro and In Vivo Biological Investigations, and In Silico Antibacterial Mechanisms of Mentha piperita Essential Oils Collected from Two Different Regions in Morocco. Foods 2022; 11:3466. [PMID: 36360079 PMCID: PMC9658668 DOI: 10.3390/foods11213466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 08/13/2023] Open
Abstract
The objective of this work is to explore the phytochemical profile of Mentha piperita essential oils (MPEO) collected from two different Moroccan regions using gas chromatography-mass spectrophotometer (GC-MS) and to investigate their antioxidant, anti-inflammatory, antidiabetic and, antimicrobial effects using in vivo and in vitro assays. The chemical constituent of MPEO from the Azrou zone is dominated by carvone (70.25%), while MPEO from the Ouazzane zone is rich in Menthol (43.32%) and Menthone (29.4%). MPEO from Ouezzane showed higher antioxidant activity than EO from Azrou. Nevertheless, EO from Ouezzane considerably inhibited 5-Lipoxygenase (IC50 = 11.64 ± 0.02 µg/mL) compared to EO from Azro (IC50 = 23.84 ± 0.03 µg/mL). Both EOs from Azrou and Ouazzane inhibited the α-amylase activity in vitro, with IC50 values of 131.62 ± 0.01 µg/mL and 91.64 ± 0.03 µg/mL, respectively. The EOs were also tested for minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). The discdiffusion test revealed that MPEOs from both regions have significant antibacterial efficacy, and MPEOs from the north region showed the highest effect. The gram-positive bacteria were the most susceptible organisms. The MIC concentrations were in the range of 0.05 to 6.25 mg/mL, and the MBC concentrations were within 0.05-25.0 mg/mL. The MBC/MIC index indicated that MPEO has strong bactericidal effects.
Collapse
Affiliation(s)
- Samiah Hamad Al-Mijalli
- Department of Biology, College of Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Nidal Naceiri Mrabti
- Computer Chemistry and Modeling Team, Laboratory of Materials, Modeling and Environmental Engineering (LIMME), Faculty of Sciences Dhar El Mehraz, Sidi Mohamed Ben Abdellah University (USMBA), BP 1796, Atlas, Fez 30000, Morocco
| | - Hayat Ouassou
- Faculty of Sciences, University Mohammed First, Boulevard Mohamed VI BP 717, Oujda 60000, Morocco
| | - Ryan A. Sheikh
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Emad M. Abdallah
- Department of Science Laboratories, College of Science and Arts, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Hamza Assaggaf
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, 1988, Najran 61441, Saudi Arabia
| | - Ahmed Abdullah Al Awadh
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, 1988, Najran 61441, Saudi Arabia
| | - Ahmed Qasem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ammar Attar
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat BP 6203, Morocco
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Long Chiau Ming
- Pengiran Anak Puteri Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Hanae Naceiri Mrabti
- Laboratoires TBC, Faculty of Pharmaceutical and Biological Sciences, B.P. 8359006 Lille, France
- Laboratory of Pharmacology and Toxicology, Bio Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Rabat BP 6203, Morocco
| |
Collapse
|
6
|
Fluoroquinolones Hybrid Molecules as Promising Antibacterial Agents in the Fight against Antibacterial Resistance. Pharmaceutics 2022; 14:pharmaceutics14081749. [PMID: 36015376 PMCID: PMC9414178 DOI: 10.3390/pharmaceutics14081749] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
The emergence of bacterial resistance has motivated researchers to discover new antibacterial agents. Nowadays, fluoroquinolones keep their status as one of the essential classes of antibacterial agents. The new generations of fluoroquinolones are valuable therapeutic tools with a spectrum of activity, including Gram-positive, Gram-negative, and atypical bacteria. This review article surveys the design of fluoroquinolone hybrids with other antibacterial agents or active compounds and underlines the new hybrids' antibacterial properties. Antibiotic fluoroquinolone hybrids have several advantages over combined antibiotic therapy. Thus, some challenges related to joining two different molecules are under study. Structurally, the obtained hybrids may contain a cleavable or non-cleavable linker, an essential element for their pharmacokinetic properties and mechanism of action. The design of hybrids seems to provide promising antibacterial agents helpful in the fight against more virulent and resistant strains. These hybrid structures have proven superior antibacterial activity and less susceptibility to bacterial resistance than the component molecules. In addition, fluoroquinolone hybrids have demonstrated other biological effects such as anti-HIV, antifungal, antiplasmodic/antimalarial, and antitumor activity. Many fluoroquinolone hybrids are in various phases of clinical trials, raising hopes that new antibacterial agents will be approved shortly.
Collapse
|
7
|
Cochrane W, Fitzgerald PR, Paegel BM. Antibacterial Discovery via Phenotypic DNA-Encoded Library Screening. ACS Chem Biol 2021; 16:2752-2756. [PMID: 34806373 PMCID: PMC8688339 DOI: 10.1021/acschembio.1c00714] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The global rise of multidrug resistant infections poses an imminent, existential threat. Numerous pipelines have failed to convert biochemically active molecules into bona fide antibacterials, owing to a lack of chemical material with antibacterial-like physical properties in high-throughput screening compound libraries. Here, we demonstrate scalable design and synthesis of an antibacterial-like solid-phase DNA-encoded library (DEL, 7488 members) and facile hit deconvolution from whole-cell Escherichia coli and Bacillus subtilis cytotoxicity screens. The screen output identified two low-micromolar inhibitors of B. subtilis growth and recapitulated known structure-activity relationships of the fluoroquinolone antibacterial class. This phenotypic DEL screening strategy is also potentially applicable to adherent cells and will broadly enable the discovery and optimization of cell-active molecules.
Collapse
Affiliation(s)
- Wesley
G. Cochrane
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
| | - Patrick R. Fitzgerald
- Skaggs
Doctoral Program in the Chemical and Biological Sciences, Scripps Research, La Jolla, California 92037, United States
| | - Brian M. Paegel
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
- Departments
of Chemistry & Biomedical Engineering, University of California, Irvine, California 92697, United States
| |
Collapse
|
8
|
Miethke M, Pieroni M, Weber T, Brönstrup M, Hammann P, Halby L, Arimondo PB, Glaser P, Aigle B, Bode HB, Moreira R, Li Y, Luzhetskyy A, Medema MH, Pernodet JL, Stadler M, Tormo JR, Genilloud O, Truman AW, Weissman KJ, Takano E, Sabatini S, Stegmann E, Brötz-Oesterhelt H, Wohlleben W, Seemann M, Empting M, Hirsch AKH, Loretz B, Lehr CM, Titz A, Herrmann J, Jaeger T, Alt S, Hesterkamp T, Winterhalter M, Schiefer A, Pfarr K, Hoerauf A, Graz H, Graz M, Lindvall M, Ramurthy S, Karlén A, van Dongen M, Petkovic H, Keller A, Peyrane F, Donadio S, Fraisse L, Piddock LJV, Gilbert IH, Moser HE, Müller R. Towards the sustainable discovery and development of new antibiotics. Nat Rev Chem 2021; 5:726-749. [PMID: 34426795 PMCID: PMC8374425 DOI: 10.1038/s41570-021-00313-1] [Citation(s) in RCA: 564] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/08/2023]
Abstract
An ever-increasing demand for novel antimicrobials to treat life-threatening infections caused by the global spread of multidrug-resistant bacterial pathogens stands in stark contrast to the current level of investment in their development, particularly in the fields of natural-product-derived and synthetic small molecules. New agents displaying innovative chemistry and modes of action are desperately needed worldwide to tackle the public health menace posed by antimicrobial resistance. Here, our consortium presents a strategic blueprint to substantially improve our ability to discover and develop new antibiotics. We propose both short-term and long-term solutions to overcome the most urgent limitations in the various sectors of research and funding, aiming to bridge the gap between academic, industrial and political stakeholders, and to unite interdisciplinary expertise in order to efficiently fuel the translational pipeline for the benefit of future generations.
Collapse
Affiliation(s)
- Marcus Miethke
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Marco Pieroni
- Food and Drug Department, University of Parma, Parma, Italy
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Mark Brönstrup
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Chemical Biology (CBIO), Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Peter Hammann
- Infectious Diseases & Natural Product Research at EVOTEC, and Justus Liebig University Giessen, Giessen, Germany
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, Paris, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, Paris, France
| | - Philippe Glaser
- Ecology and Evolution of Antibiotic Resistance Unit, Microbiology Department, Institut Pasteur, CNRS UMR3525, Paris, France
| | | | - Helge B. Bode
- Department of Biosciences, Goethe University Frankfurt, Frankfurt, Germany
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, Marburg, Germany
| | - Rui Moreira
- Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Yanyan Li
- Unit MCAM, CNRS, National Museum of Natural History (MNHN), Paris, France
| | - Andriy Luzhetskyy
- Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Marnix H. Medema
- Bioinformatics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Jean-Luc Pernodet
- Institute for Integrative Biology of the Cell (I2BC) & Microbiology Department, University of Paris-Saclay, Gif-sur-Yvette, France
| | - Marc Stadler
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Microbial Drugs (MWIS), Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | | | | | - Andrew W. Truman
- Department of Molecular Microbiology, John Innes Centre, Norwich, United Kingdom
| | - Kira J. Weissman
- Molecular and Structural Enzymology Group, Université de Lorraine, CNRS, IMoPA, Nancy, France
| | - Eriko Takano
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, University of Manchester, Manchester, United Kingdom
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Evi Stegmann
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Wolfgang Wohlleben
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Department of Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Myriam Seemann
- Institute for Chemistry UMR 7177, University of Strasbourg/CNRS, ITI InnoVec, Strasbourg, France
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
| | - Alexander Titz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Timo Jaeger
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Silke Alt
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | | | | | - Andrea Schiefer
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Kenneth Pfarr
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- German Center for Infection Research (DZIF), Braunschweig, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Heather Graz
- Biophys Ltd., Usk, Monmouthshire, United Kingdom
| | - Michael Graz
- School of Law, University of Bristol, Bristol, United Kingdom
| | | | | | - Anders Karlén
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Hrvoje Petkovic
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany
| | | | | | - Laurent Fraisse
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Laura J. V. Piddock
- The Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Ian H. Gilbert
- Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Heinz E. Moser
- Novartis Institutes for BioMedical Research (NIBR), Emeryville, CA USA
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University Campus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| |
Collapse
|
9
|
Boyd NK, Teng C, Frei CR. Brief Overview of Approaches and Challenges in New Antibiotic Development: A Focus On Drug Repurposing. Front Cell Infect Microbiol 2021; 11:684515. [PMID: 34079770 PMCID: PMC8165386 DOI: 10.3389/fcimb.2021.684515] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 12/19/2022] Open
Abstract
Drug repurposing, or identifying new uses for existing drugs, has emerged as an alternative to traditional drug discovery processes involving de novo synthesis. Drugs that are currently approved or under development for non-antibiotic indications may possess antibiotic properties, and therefore may have repurposing potential, either alone or in combination with an antibiotic. They might also serve as "antibiotic adjuvants" to enhance the activity of certain antibiotics.
Collapse
Affiliation(s)
- Natalie K Boyd
- College of Pharmacy, The University of Texas at Austin, San Antonio, TX, United States.,Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Chengwen Teng
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, The University of South Carolina, Columbia, SC, United States
| | - Christopher R Frei
- College of Pharmacy, The University of Texas at Austin, San Antonio, TX, United States.,Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States.,Research Department, South Texas Veterans Health Care System, San Antonio, TX, United States.,Pharmacy Department, University Health System, San Antonio, TX, United States
| |
Collapse
|
10
|
Yang H, Liu K, Jin S, Huigens Iii RW. Design, synthesis and biological evaluation of a halogenated phenazine-erythromycin conjugate prodrug for antibacterial applications. Org Biomol Chem 2021; 19:1483-1487. [PMID: 33521803 PMCID: PMC7939049 DOI: 10.1039/d0ob02428g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There is a significant need for new antibacterial agents as pathogenic bacteria continue to threaten human health through the acquisition of resistance and tolerance towards existing antibiotics. Over the last several years, our group has been developing a novel series of halogenated phenazines that demonstrate potent antibacterial and biofilm eradication activities against critical Gram-positive pathogens, including: Staphylococcus aureus, Staphylococcus epidermidis and Enterococcus faecium. Here, we report the design, chemical synthesis and initial biological assessment of a halogenated phenazine-erythromycin conjugate prodrug 5 aimed at enhancing the translational potential for halogenated phenazines as a treatment of bacterial infections.
Collapse
Affiliation(s)
- Hongfen Yang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Robert W Huigens Iii
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, USA.
| |
Collapse
|
11
|
Phoon BL, Ong CC, Mohamed Saheed MS, Show PL, Chang JS, Ling TC, Lam SS, Juan JC. Conventional and emerging technologies for removal of antibiotics from wastewater. JOURNAL OF HAZARDOUS MATERIALS 2020; 400:122961. [PMID: 32947727 DOI: 10.1016/j.jhazmat.2020.122961] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 05/27/2023]
Abstract
Antibiotics and pharmaceuticals related products are used to enhance public health and quality of life. The wastewater that is produced from pharmaceutical industries still contains noticeable amount of antibiotics, and this has remained one of the major environmental problems facing public health. The conventional wastewater remediation approach employed by the pharmaceutical industries for the antibiotics wastewater removal is unable to remove the antibiotics completely. Besides, municipal and livestock wastewater also contain unmetabolized antibiotics released by human and animal, respectively. The antibiotic found in wastewater leads to antibiotic resistance challenges, also emergence of superbugs. Currently, numerous technological approaches have been developed to remove antibiotics from the wastewater. Therefore, it was imperative to critically review the weakness and strength of these current advanced technological approaches in use. Besides, the conventional methods for removal of antibiotics such as Klavaroti et al., Homem and Santos also discussed. Although, membrane treatment is discovered as the ultimate choice of approach, to completely remove the antibiotics, while the filtered antibiotics are still retained on the membrane. This study found, hybrid processes to be the best solution antibiotics removal from wastewater. Nevertheless, real-time monitoring system is also recommended to ascertain that, wastewater is cleared of antibiotics.
Collapse
Affiliation(s)
- Bao Lee Phoon
- Nanotechnology & Catalysis Research Centre (NANOCAT), Level 3 Block A, Institute for Advanced Studies, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chong Cheen Ong
- Department of Fundamental & Applied Sciences, Universiti Teknologi PETRONAS, 32610 Seri Iskandar, Perak Darul Ridzuan, Malaysia; Centre of Innovative Nanostructures & Nanodevices (COINN), Universiti Teknologi PETRONAS, 32610 Seri Iskandar, Perak Darul Ridzuan, Malaysia
| | - Mohamed Shuaib Mohamed Saheed
- Department of Fundamental & Applied Sciences, Universiti Teknologi PETRONAS, 32610 Seri Iskandar, Perak Darul Ridzuan, Malaysia; Centre of Innovative Nanostructures & Nanodevices (COINN), Universiti Teknologi PETRONAS, 32610 Seri Iskandar, Perak Darul Ridzuan, Malaysia
| | - Pau-Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor Darul Ehsan, Malaysia
| | - Jo-Shu Chang
- Department of Chemical and Materials Engineering, College of Engineering, Tunghai University, Taichung 407, Taiwan; Center for Nanotechnology, Tunghai University, Taichung 407, Taiwan; Department of Chemical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Tau Chuan Ling
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Su Shiung Lam
- Pyrolysis Technology Research Group, Institute of Tropical Aquaculture and Fisheries (AKUATROP) & Institute of Tropical Biodiversity and Sustainable Development (Bio-D Tropika), Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Joon Ching Juan
- Nanotechnology & Catalysis Research Centre (NANOCAT), Level 3 Block A, Institute for Advanced Studies, University of Malaya, 50603 Kuala Lumpur, Malaysia; School of Science, Monash University, Sunway Campus, Jalan Lagoon Selatan, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
12
|
Cruz CD, Wrigstedt P, Moslova K, Iashin V, Mäkkylä H, Ghemtio L, Heikkinen S, Tammela P, Perea-Buceta JE. Installation of an aryl boronic acid function into the external section of N-aryl-oxazolidinones: Synthesis and antimicrobial evaluation. Eur J Med Chem 2020; 211:113002. [PMID: 33223262 DOI: 10.1016/j.ejmech.2020.113002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 10/23/2022]
Abstract
N-aryl-oxazolidinones is a prominent family of antimicrobials used for treating infections caused by clinically prevalent Gram-positive bacteria. Recently, boron-containing compounds have displayed intriguing potential in the antibiotic discovery setting. Herein, we report the unprecedented introduction of a boron-containing moiety such as an aryl boronic acid in the external region of the oxazolidinone structure via a chemoselective acyl coupling reaction. As a result, we accessed a series of analogues with a distal aryl boronic pharmacophore on the oxazolidinone scaffold. We identified that a peripheric linear conformation coupled with freedom of rotation and no further substitution on the external aryl boronic ring, an amido linkage with hydrogen bonding character, in addition to a para-relative disposition between boronic group and linker, are the optimal combination of structural features in this series for antimicrobial activity. In comparison to linezolid, the analogue comprising all those features, compound 20b, displayed levels of antimicrobial activity augmented by an eight-fold to a thirty-two-fold against a panel of Gram-positive strains, and a near one hundred-fold against Escherichia coli JW5503, a Gram-negative mutant strain with a defective efflux capability.
Collapse
Affiliation(s)
- Cristina D Cruz
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Finland
| | - Pauli Wrigstedt
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014, Finland
| | - Karina Moslova
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014, Finland
| | - Vladimir Iashin
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014, Finland
| | - Heidi Mäkkylä
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Finland
| | - Léo Ghemtio
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Finland
| | - Sami Heikkinen
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014, Finland
| | - Päivi Tammela
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Finland
| | - Jesus E Perea-Buceta
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014, Finland.
| |
Collapse
|
13
|
Xiao T, Liu K, Huigens RW. Progress towards a stable cephalosporin-halogenated phenazine conjugate for antibacterial prodrug applications. Bioorg Med Chem Lett 2020; 30:127515. [PMID: 32860978 DOI: 10.1016/j.bmcl.2020.127515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023]
Abstract
Resistant bacteria successfully evade the action of conventional antibiotic therapies during infection, often leading to significant illness and death. Our lab has discovered halogenated phenazine (HP) analogues which demonstrate potent antibacterial activities through a unique iron-starving mechanism. Herein, we describe synthetic efforts towards a stable cephalosporin-HP conjugate prodrug with the aim of translating HPs into useful clinical agents. Cephalosporin-antibiotic conjugates offer multiple advantages for antibacterial design, including the release of active agents through the targeting of intracellular cephalosporinase following selective ring-opening of the beta-lactam warhead. During these studies, carbonate-linked cephalosporin-HP conjugate 16 was synthesized; however, we were unable to successfully remove the ester group required for cephalosporinase processing. Cephalosporin-HP 16 was then utilized as a probe to investigate the stability of the carbonate linker in antibacterial assays and, as predicted, this compound proved to be inactive against Staphylococcus aureus (MIC > 100 µM). The lack of 16's antibacterial activity can be attributed to the carbonate linker remaining intact throughout the MIC assay, thus not liberating the active HP moiety. These efforts have led to a more stable cephalosporin-HP conjugate joined through a carbonate linker compared to a highly unstable ether linked analogue we previously reported.
Collapse
Affiliation(s)
- Tao Xiao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, FL 32610, United States
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, FL 32610, United States
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, FL 32610, United States
| |
Collapse
|
14
|
Fisher JF, Mobashery S. Constructing and deconstructing the bacterial cell wall. Protein Sci 2020; 29:629-646. [PMID: 31747090 PMCID: PMC7021008 DOI: 10.1002/pro.3737] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
The history of modern medicine cannot be written apart from the history of the antibiotics. Antibiotics are cytotoxic secondary metabolites that are isolated from Nature. The antibacterial antibiotics disproportionately target bacterial protein structure that is distinct from eukaryotic protein structure, notably within the ribosome and within the pathways for bacterial cell-wall biosynthesis (for which there is not a eukaryotic counterpart). This review focuses on a pre-eminent class of antibiotics-the β-lactams, exemplified by the penicillins and cephalosporins-from the perspective of the evolving mechanisms for bacterial resistance. The mechanism of action of the β-lactams is bacterial cell-wall destruction. In the monoderm (single membrane, Gram-positive staining) pathogen Staphylococcus aureus the dominant resistance mechanism is expression of a β-lactam-unreactive transpeptidase enzyme that functions in cell-wall construction. In the diderm (dual membrane, Gram-negative staining) pathogen Pseudomonas aeruginosa a dominant resistance mechanism (among several) is expression of a hydrolytic enzyme that destroys the critical β-lactam ring of the antibiotic. The key sensing mechanism used by P. aeruginosa is monitoring the molecular difference between cell-wall construction and cell-wall deconstruction. In both bacteria, the resistance pathways are manifested only when the bacteria detect the presence of β-lactams. This review summarizes how the β-lactams are sensed and how the resistance mechanisms are manifested, with the expectation that preventing these processes will be critical to future chemotherapeutic control of multidrug resistant bacteria.
Collapse
Affiliation(s)
- Jed F. Fisher
- Department of Chemistry and BiochemistryUniversity of Notre DameSouth BendIndiana
| | - Shahriar Mobashery
- Department of Chemistry and BiochemistryUniversity of Notre DameSouth BendIndiana
| |
Collapse
|
15
|
Synthesis and Antibacterial Activity of Difluoromethyl Cinnamoyl Amides. Molecules 2020; 25:molecules25040789. [PMID: 32059479 PMCID: PMC7070587 DOI: 10.3390/molecules25040789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 01/18/2023] Open
Abstract
Series of novel amides of isoferulic acid, where the phenolic hydroxyl was replaced by a difluoromethyl group, were synthesized and their in vitro antibacterial activities assayed against fourteen bacterial strains (six Gram-positive and eight Gram-negative). A one-pot methodology was developed to obtain the 3′-(difluoromethyl)-4′-methoxycinnamoyl amides using Deoxofluor® as a fluorinating agent. The N-isopropyl, N-isopentyl, and N-(2-phenylethyl) amides 11b, 11d and 11g were the most active and selective against Mycobacterium smegmatis (MIC = 8 µg/mL) with 11b and 11g displaying negligible or no cytotoxicity against HepG2 and A549 cells. Thirteen analogs of N-isopropylamide 11b were also synthesized and their antibacterial activity assayed. Results show that the difluoromethyl moiety enhanced antibacterial activity and selectivity towards M. smegmatis, changing the microorganism inhibition profile of the parent compound. The selectivity exhibited by some of the compounds towards M. smegmatis makes them potential leads in the search for new narrow spectrum antibiotics against M. tuberculosis.
Collapse
|
16
|
Pyta K, Janas A, Skrzypczak N, Schilf W, Wicher B, Gdaniec M, Bartl F, Przybylski P. Specific Interactions between Rifamycin Antibiotics and Water Influencing Ability To Overcome Natural Cell Barriers and the Range of Antibacterial Potency. ACS Infect Dis 2019; 5:1754-1763. [PMID: 31461259 DOI: 10.1021/acsinfecdis.9b00176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Rifamycins are a group of macrocyclic antibiotics mainly used for the treatment of various bacterial infections including tuberculosis. Spectroscopic studies of rifamycins evidence the formation of temperature- and solvent-dependent equilibria between A-, B-, and C-type conformers in solutions. The B- and C-type conformers of rifamycin antibiotics are exclusively formed in the presence of water molecules. A- and B-type conformers exhibit a hydrophilic and "open" ansa-bridge nature whereas the C-type conformer is more lipophilic due to the presence of a "closed" ansa-bridge structure. The involvement of the lactam moiety of the ansa-bridge in intramolecular H-bonds within rifapentine and rifampicin implicates the formation of a more hydrophilic A-type conformer. In contrast to rifampicin and rifapentine, for rifabutin and rifaximin, the "free" lactam group enhances conformational flexibility of the ansa-bridge, thereby enabling interconversion between A- and C-type conformers. In turn, an equilibrium between A- and C-type conformers for rifamycins improves their adaptation to the changing nature of bacteria cell membranes, especially those of Gram-negative strains and/or to efflux pump systems.
Collapse
Affiliation(s)
- Krystian Pyta
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Anna Janas
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Natalia Skrzypczak
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Wojciech Schilf
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Barbara Wicher
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland
| | - Maria Gdaniec
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Franz Bartl
- Humboldt-Universität zu Berlin, Lebenswissenschaftliche Fakultät, Institut für Biologie, Biophysikalische Chemie, Invalidenstr. 42, 10099 Berlin, Germany
| | - Piotr Przybylski
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| |
Collapse
|
17
|
Sati GC, Sarpe VA, Furukawa T, Mondal S, Mantovani M, Hobbie SN, Vasella A, Böttger EC, Crich D. Modification at the 2'-Position of the 4,5-Series of 2-Deoxystreptamine Aminoglycoside Antibiotics To Resist Aminoglycoside Modifying Enzymes and Increase Ribosomal Target Selectivity. ACS Infect Dis 2019; 5:1718-1730. [PMID: 31436080 PMCID: PMC6788953 DOI: 10.1021/acsinfecdis.9b00128] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
![]()
A series
of derivatives of the 4,5-disubstituted class of 2-deoxystreptamine
aminoglycoside antibiotics neomycin, paromomycin, and ribostamycin
was prepared and assayed for (i) their ability to inhibit protein
synthesis by bacterial ribosomes and by engineered bacterial ribosomes
carrying eukaryotic decoding A sites, (ii) antibacterial activity
against wild type Gram negative and positive pathogens, and (iii)
overcoming resistance due to the presence of aminoacyl transferases
acting at the 2′-position. The presence of five suitably positioned
residual basic amino groups was found to be necessary for activity
to be retained upon removal or alkylation of the 2′-position
amine. As alkylation of the 2′-amino group overcomes the action
of resistance determinants acting at that position and in addition
results in increased selectivity for the prokaryotic over eukaryotic
ribosomes, it constitutes an attractive modification for introduction
into next generation aminoglycosides. In the neomycin series, the
installation of small (formamide) or basic (glycinamide) amido groups
on the 2′-amino group is tolerated.
Collapse
Affiliation(s)
- Girish C. Sati
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Vikram A. Sarpe
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Takayuki Furukawa
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Sujit Mondal
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Matilde Mantovani
- Institute of Medical Microbiology, University of Zurich, 28 Gloriastrasse, 8006 Zürich, Switzerland
| | - Sven N. Hobbie
- Institute of Medical Microbiology, University of Zurich, 28 Gloriastrasse, 8006 Zürich, Switzerland
| | - Andrea Vasella
- Organic Chemistry Laboratory, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Erik C. Böttger
- Institute of Medical Microbiology, University of Zurich, 28 Gloriastrasse, 8006 Zürich, Switzerland
| | - David Crich
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| |
Collapse
|
18
|
Jadhav RW, Kobaisi MA, Jones LA, Vinu A, Bhosale SV. The Supramolecular Self-Assembly of Aminoglycoside Antibiotics and their Applications. ChemistryOpen 2019; 8:1154-1166. [PMID: 31497469 PMCID: PMC6718072 DOI: 10.1002/open.201900193] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Aminoglycosides, a class of antibiotics that includes gentamicin, kanamycin, neomycin, streptomycin, tobramycin and apramycin, are derived from various streptomyces species. Despite the significant increase in the antibacterial resistant pathogens, aminoglycosides remain an important class of antimicrobial drugs due to their unique chemical structure which offers a broad spectrum of activity. The modification of antibiotics and their subsequent use in supramolecular chemistry is rarely reported. Given the importance of aminoglycosides, here we give a brief overview on the modification of 4,5- and 4,6-disubstituted deoxystreptamine classes of aminoglycosides through supramolecular chemistry and their potential for real world applications. We also make the case that the work in this area is gaining momentum, and there are significant opportunities to meet the challenges of modern antibiotics through the modification of aminoglycosides by harnessing the advantages of supramolecular chemistry.
Collapse
Affiliation(s)
- Ratan W. Jadhav
- School of Chemical SciencesGoa University Taleigao PlateauGoa403 206INDIA
| | - Mohammad Al Kobaisi
- School of Science, Faculty of Science, Engineering and TechnologySwinburne University of TechnologyHawthornAustralia
| | - Lathe A. Jones
- CAMIC, School of ScienceRMIT University, GPO Box2476Melbourne, VIC-3001Australia
| | - Ajayan Vinu
- Global Innovative Center for Advanced Nanomaterials (GICAN)The University of Newcastle (UON), University Drive, CallaghanNSW 2308Australia
| | | |
Collapse
|
19
|
Bacterial metabolic state more accurately predicts antibiotic lethality than growth rate. Nat Microbiol 2019; 4:2109-2117. [PMID: 31451773 DOI: 10.1038/s41564-019-0536-0] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/08/2019] [Indexed: 01/19/2023]
Abstract
Growth rate and metabolic state of bacteria have been separately shown to affect antibiotic efficacy1-3. However, the two are interrelated as bacterial growth inherently imposes a metabolic burden4; thus, determining individual contributions from each is challenging5,6. Indeed, faster growth is often correlated with increased antibiotic efficacy7,8; however, the concurrent role of metabolism in that relationship has not been well characterized. As a result, a clear understanding of the interdependence between growth and metabolism, and their implications for antibiotic efficacy, are lacking9. Here, we measured growth and metabolism in parallel across a broad range of coupled and uncoupled conditions to determine their relative contribution to antibiotic lethality. We show that when growth and metabolism are uncoupled, antibiotic lethality uniformly depends on the bacterial metabolic state at the time of treatment, rather than growth rate. We further reveal a critical metabolic threshold below which antibiotic lethality is negligible. These findings were general for a wide range of conditions, including nine representative bactericidal drugs and a diverse range of Gram-positive and Gram-negative species (Escherichia coli, Acinetobacter baumannii and Staphylococcus aureus). This study provides a cohesive metabolic-dependent basis for antibiotic-mediated cell death, with implications for current treatment strategies and future drug development.
Collapse
|
20
|
Meng Z, Tang ML, Yu L, Liang Y, Han J, Zhang C, Hu F, Yu JM, Sun X. Novel Mercapto Propionamide Derivatives with Potent New Delhi Metallo-β-Lactamase-1 Inhibitory Activity and Low Toxicity. ACS Infect Dis 2019; 5:903-916. [PMID: 30838850 DOI: 10.1021/acsinfecdis.8b00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The emergence and worldwide prevalence of New Delhi metallo-β-lactamase 1 (NDM-1) expressing Gram-negative bacteria with resistance against most β-lactam antibiotics pose a serious threat to human health. However, no NDM-1 inhibitors are clinically approved at present. Herein, based on the lead compound captopril, a series of compounds were designed, synthesized, and evaluated for NDM-1 inhibitory activities. All designed compounds showed single digit micromolar or submicromolar NDM-1 inhibitory activities, which were much more potent than that of captopril. Among them, compounds 14a and 14m exhibited excellent NDM-1 inhibitory activities, with IC50 values of 0.10 and 0.12 μM, respectively. Further studies demonstrated that compound 14m displayed low cytotoxicity, good water solubility, high metabolic stability, and low acute toxicity in mice. Importantly, compound 14m exhibited potent synergistic antimicrobial activities with Meropenem (MEM) for the treatment of clinically isolated NDM-1-expressing strains.
Collapse
Affiliation(s)
- Zhi Meng
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Mei-Lin Tang
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Molecular Engineering and Institutes of Biomedical Sciences, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Liting Yu
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yongxi Liang
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jilai Han
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - ChenChen Zhang
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Fupin Hu
- Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jian-Ming Yu
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xun Sun
- Department of Natural Products Chemistry, School of PharmacyFudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
21
|
Synthetic sideromycins (skepticism and optimism): selective generation of either broad or narrow spectrum Gram-negative antibiotics. Biometals 2019; 32:425-451. [PMID: 30919118 DOI: 10.1007/s10534-019-00192-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 02/07/2023]
Abstract
New or repurposed antibiotics are desperately needed since bacterial resistance has risen to essentially all of our current antibiotics, and few new antibiotics have been developed over the last several decades. A primary cause of drug resistance is the overuse of antibiotics that can result in alteration of microbial permeability, alteration of drug target binding sites, induction of enzymes that destroy antibiotics (i.e., β-lactamases) and even induction of efflux mechanisms. Research efforts are described that are designed to determine if the known critical dependence of iron assimilation by microbes for growth and virulence can be exploited for the development of new approaches to antibiotic therapy. Iron recognition and active transport relies on the biosyntheses and use of microbe-selective iron chelating compounds called siderophores. Several natural siderophore-antibiotic conjugates (sideromycins) have been discovered and studied. The natural sideromycins consist of an iron binding siderophore linked to a warhead that exerts antibiotic activity once assimilated by targeted bacteria. Inspired these natural conjugates, a combination of chemical syntheses, microbiological and biochemical studies have been used to generate semi-synthetic and totally synthetic sideromycin analogs. The results demonstrate that siderophores and analogs can be used for iron transport-mediated drug delivery ("Trojan Horse" antibiotics or sideromycins) and induction of iron limitation/starvation (development of new agents to block iron assimilation). While several examples illustrate that this approach can generate microbe selective antibiotics that are active in vitro, the scope and limitations of this approach, especially related to development of resistance, siderophore based molecular recognition requirements, appropriate linker and drug choices, will be described.
Collapse
|
22
|
Kuppusamy R, Yasir M, Yee E, Willcox M, Black DS, Kumar N. Guanidine functionalized anthranilamides as effective antibacterials with biofilm disruption activity. Org Biomol Chem 2019; 16:5871-5888. [PMID: 30070287 DOI: 10.1039/c8ob01699b] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We describe a library of amphiphilic anthranilamide compounds as antimicrobial peptide (AMP) mimics. These contain a hydrophobic naphthoyl side chain and different hydrophilic cationic groups such as amino, quaternary ammonium and guanidino groups. These are prepared via the ring-opening of different isatoic anhydrides. The antibacterial activity against S. aureus and E. coli of compounds containing guanidino cationic groups was greater than that for amino and quaternary ammonium cationic groups. The fluoro-substituted guanidinium compound 9b showed a minimum inhibitory concentration (MIC) of 2.0 μM against S. aureus, and reduced established biofilms of S. aureus by 92% at 64 μM concentration. The bromo-substituted guanidinium compound 9d exhibited good MIC against S. aureus (3.9 μM) and E. coli (15.6 μM) and disrupted established biofilms of S. aureus by 83% at 62.4 μM concentration. Cytoplasmic membrane permeability studies suggested that depolarization and disruption of the bacterial cell membrane could be a possible mechanism for antibacterial activity and the in vitro toxicity studies against MRC-5 human lung fibroblast cells showed that the potent compounds are non-toxic against mammalian cells.
Collapse
Affiliation(s)
- Rajesh Kuppusamy
- School of Chemistry, UNSW Australia, Sydney, NSW 2052, Australia.
| | | | | | | | | | | |
Collapse
|
23
|
Hickey SM, Ashton TD, Boer G, Bader CA, Thomas M, Elliott AG, Schmuck C, Yu HY, Li J, Nation RL, Cooper MA, Plush SE, Brooks DA, Pfeffer FM. Norbornane-based cationic antimicrobial peptidomimetics targeting the bacterial membrane. Eur J Med Chem 2018; 160:9-22. [PMID: 30316060 DOI: 10.1016/j.ejmech.2018.09.072] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/14/2018] [Accepted: 09/29/2018] [Indexed: 11/27/2022]
Abstract
The design, synthesis and evaluation of a small series of potent amphiphilic norbornane antibacterial agents has been performed (compound 10 MIC = 0.25 μg/mL against MRSA). Molecular modelling indicates rapid aggregation of this class of antibacterial agent prior to membrane association and insertion. Two fluorescent analogues (compound 29 with 4-amino-naphthalimide and 34 with 4-nitrobenz-2-oxa-1,3-diazole fluorophores) with good activity (MIC = 0.5 μg/mL against MRSA) were also constructed and confocal microscopy studies indicate that the primary site of interaction for this family of compounds is the bacterial membrane.
Collapse
Affiliation(s)
- Shane M Hickey
- Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| | - Trent D Ashton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Gareth Boer
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Christie A Bader
- Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Michael Thomas
- Research School of Chemistry, The Australian National University, Acton, ACT, 2601, Australia
| | - Alysha G Elliott
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Carsten Schmuck
- Institute for Organic Chemistry, University of Duisburg-Essen, 45117, Essen, Germany
| | - Heidi Y Yu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Science, Royal Parade, Parkville, Victoria, 3052, Australia
| | - Jian Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Science, Royal Parade, Parkville, Victoria, 3052, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Science, Royal Parade, Parkville, Victoria, 3052, Australia
| | - Matthew A Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Sally E Plush
- Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Douglas A Brooks
- Cancer Research Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Frederick M Pfeffer
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, 3216, Australia.
| |
Collapse
|
24
|
Schnaars C, Kildahl-Andersen G, Prandina A, Popal R, Radix S, Le Borgne M, Gjøen T, Andresen AMS, Heikal A, Økstad OA, Fröhlich C, Samuelsen Ø, Lauksund S, Jordheim LP, Rongved P, Åstrand OAH. Synthesis and Preclinical Evaluation of TPA-Based Zinc Chelators as Metallo-β-lactamase Inhibitors. ACS Infect Dis 2018; 4:1407-1422. [PMID: 30022668 DOI: 10.1021/acsinfecdis.8b00137] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The rise of antimicrobial resistance (AMR) worldwide and the increasing spread of multi-drug-resistant organisms expressing metallo-β-lactamases (MBL) require the development of efficient and clinically available MBL inhibitors. At present, no such inhibitor is available, and research is urgently needed to advance this field. We report herein the development, synthesis, and biological evaluation of chemical compounds based on the selective zinc chelator tris-picolylamine (TPA) that can restore the bactericidal activity of Meropenem (MEM) against Pseudomonas aeruginosa and Klebsiella pneumoniae expressing carbapenemases Verona integron-encoded metallo-β-lactamase (VIM-2) and New Delhi metallo-β-lactamase 1 (NDM-1), respectively. These adjuvants were prepared via standard chemical methods and evaluated in biological assays for potentiation of MEM against bacteria and toxicity (IC50) against HepG2 human liver carcinoma cells. One of the best compounds, 15, lowered the minimum inhibitory concentration (MIC) of MEM by a factor of 32-256 at 50 μM within all tested MBL-expressing clinical isolates and showed no activity toward serine carbapenemase expressing isolates. Biochemical assays with purified VIM-2 and NDM-1 and 15 resulted in inhibition kinetics with kinact/ KI of 12.5 min-1 mM-1 and 0.500 min-1 mM-1, respectively. The resistance frequency of 15 at 50 μM was in the range of 10-7 to 10-9. 15 showed good tolerance in HepG2 cells with an IC50 well above 100 μM, and an in vivo study in mice showed no acute toxic effects even at a dose of 128 mg/kg.
Collapse
Affiliation(s)
| | | | - Anthony Prandina
- Université de Lyon, Université Lyon 1, Faculté de
Pharmacie - ISPB, EA 4446 Bioactive Molecules and Medicinal Chemistry,
SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, 69373 Lyon Cedex 8, France
| | | | - Sylvie Radix
- Université de Lyon, Université Lyon 1, Faculté de
Pharmacie - ISPB, EA 4446 Bioactive Molecules and Medicinal Chemistry,
SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, 69373 Lyon Cedex 8, France
| | - Marc Le Borgne
- Université de Lyon, Université Lyon 1, Faculté de
Pharmacie - ISPB, EA 4446 Bioactive Molecules and Medicinal Chemistry,
SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, 69373 Lyon Cedex 8, France
| | | | | | - Adam Heikal
- Centre for Integrative
Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, Blindern, Oslo, Norway
| | - Ole Andreas Økstad
- Centre for Integrative
Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, Blindern, Oslo, Norway
| | - Christopher Fröhlich
- Norwegian National
Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, 9038 Tromsø, Norway
- NorStruct, Department of Chemistry, Faculty of Science and Technology,
SIVA Innovation Centre, UiT The Arctic University of Norway, 9037 Tromsø, Norway
| | - Ørjan Samuelsen
- Norwegian National
Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Pharmacy, UiT − The Arctic University of Norway, 9037 Tromsø, Norway
| | - Silje Lauksund
- Norwegian National
Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Lars Petter Jordheim
- Université Lyon, Université Claude Bernard Lyon 1, INSERM
1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche
en Cancérologie de Lyon, Lyon 69008, France
| | | | | |
Collapse
|
25
|
Sonousi A, Sarpe VA, Brilkova M, Schacht J, Vasella A, Böttger EC, Crich D. Effects of the 1- N-(4-Amino-2 S-hydroxybutyryl) and 6'- N-(2-Hydroxyethyl) Substituents on Ribosomal Selectivity, Cochleotoxicity, and Antibacterial Activity in the Sisomicin Class of Aminoglycoside Antibiotics. ACS Infect Dis 2018; 4:1114-1120. [PMID: 29708331 DOI: 10.1021/acsinfecdis.8b00052] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Syntheses of the 6'- N-(2-hydroxyethyl) and 1- N-(4-amino-2 S-hydroxybutyryl) derivatives of the 4,6-aminoglycoside sisomicin and that of the doubly modified 1- N-(4-amino-2 S-hydroxybutyryl)-6'- N-(2-hydroxyethyl) derivative known as plazomicin are reported together with their antibacterial and antiribosomal activities and selectivities. The 6'- N-(2-hydroxyethyl) modification results in a moderate increase in prokaryotic/eukaryotic ribosomal selectivity, whereas the 1- N-(4-amino-2 S-hydroxybutyryl) modification has the opposite effect. When combined in plazomicin, the effects of the two groups on ribosomal selectivity cancel each other out, leading to the prediction that plazomicin will exhibit ototoxicity comparable to those of the parent and the current clinical aminoglycoside antibiotics gentamicin and tobramycin, as borne out by ex vivo studies with mouse cochlear explants. The 6'- N-(2-hydroxyethyl) modification restores antibacterial activity in the presence of the AAC(6') aminoglycoside-modifying enzymes, while the 1- N-(4-amino-2 S-hydroxybutyryl) modification overcomes resistance to the AAC(2') class but is still affected to some extent by the AAC(3) class. Neither modification is able to circumvent the ArmA ribosomal methyltransferase-induced aminoglycoside resistance. The use of phenyltriazenyl protection for the secondary amino group of sisomicin facilitates the synthesis of each derivative and their characterization through the provision of sharp NMR spectra for all intermediates.
Collapse
Affiliation(s)
- Amr Sonousi
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Vikram A. Sarpe
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Margarita Brilkova
- Institut für Medizinische Mikrobiologie, Universität Zürich, Gloriastrasse 28/30, 8006 Zürich, Switzerland
| | - Jochen Schacht
- Kresge Hearing Research Institute, Department of Otolaryngology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Andrea Vasella
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Erik C. Böttger
- Institut für Medizinische Mikrobiologie, Universität Zürich, Gloriastrasse 28/30, 8006 Zürich, Switzerland
| | - David Crich
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| |
Collapse
|
26
|
Tomoshige S, Dik DA, Akabane-Nakata M, Madukoma CS, Fisher JF, Shrout JD, Mobashery S. Total Syntheses of Bulgecins A, B, and C and Their Bactericidal Potentiation of the β-Lactam Antibiotics. ACS Infect Dis 2018; 4:860-867. [PMID: 29716193 PMCID: PMC5996343 DOI: 10.1021/acsinfecdis.8b00105] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Indexed: 12/15/2022]
Abstract
The bulgecins are iminosaccharide secondary metabolites of the Gram-negative bacterium Paraburkholderia acidophila and inhibitors of lytic transglycosylases of bacterial cell-wall biosynthesis and remodeling. The activities of the bulgecins are intimately intertwined with the mechanism of a cobiosynthesized β-lactam antibiotic. β-Lactams inhibit the penicillin-binding proteins, enzymes also critical to cell-wall biosynthesis. The simultaneous loss of the lytic transglycosylase (by bulgecin) and penicillin-binding protein (by β-lactams) activities results in deformation of the septal cell wall, observed microscopically as a bulge preceding bacterial cell lysis. We describe a practical synthesis of the three naturally occurring bulgecin iminosaccharides and their mechanistic evaluation in a series of microbiological studies. These studies identify potentiation by the bulgecin at subminimum inhibitory concentrations of the β-lactam against three pathogenic Gram-negative bacteria and establish for the first time that this potentiation results in a significant increase in the bactericidal efficacy of a clinical β-lactam.
Collapse
Affiliation(s)
- Shusuke Tomoshige
- Department of Chemistry
& Biochemistry, University of Notre
Dame, 352 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| | - David A. Dik
- Department of Chemistry
& Biochemistry, University of Notre
Dame, 352 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| | - Masaaki Akabane-Nakata
- Department of Chemistry
& Biochemistry, University of Notre
Dame, 352 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| | - Chinedu S. Madukoma
- Department of Civil & Environmental Engineering & Earth Sciences, University of Notre Dame, 156 Fitzpatrick Hall, Notre
Dame, Indiana 46556, United States
| | - Jed F. Fisher
- Department of Chemistry
& Biochemistry, University of Notre
Dame, 352 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| | - Joshua D. Shrout
- Department of Civil & Environmental Engineering & Earth Sciences, University of Notre Dame, 156 Fitzpatrick Hall, Notre
Dame, Indiana 46556, United States
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry
& Biochemistry, University of Notre
Dame, 352 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
27
|
Bouquet J, King DT, Vadlamani G, Benzie GR, Iorga B, Ide D, Adachi I, Kato A, Vocadlo DJ, Mark BL, Blériot Y, Désiré J. Selective trihydroxylated azepane inhibitors of NagZ, a glycosidase involved in Pseudomonas aeruginosa resistance to β-lactam antibiotics. Org Biomol Chem 2018; 15:4609-4619. [PMID: 28513749 DOI: 10.1039/c7ob00838d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of a series of d-gluco-like configured 4,5,6-trihydroxyazepanes bearing a triazole, a sulfonamide or a fluorinated acetamide moiety at C-3 is described. These synthetic derivatives have been tested for their ability to selectively inhibit the muropeptide recycling glucosaminidase NagZ and to thereby increase sensitivity of Pseudomonas aeruginosa to β-lactams, a pathway with substantial therapeutic potential. While introduction of triazole and sulfamide groups failed to lead to glucosaminidase inhibitors, the NHCOCF3 analog proved to be a selective inhibitor of NagZ over other glucosaminidases including human O-GlcNAcase and lysosomal hexosaminidases HexA and B.
Collapse
Affiliation(s)
- J Bouquet
- Equipe Synthèse Organique, Groupe Glycochimie, IC2MP, UMR CNRS 7285, Université de Poitiers, 4 rue Michel Brunet, 86073 Poitiers cedex 09, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Liu R, Miller PA, Vakulenko SB, Stewart NK, Boggess WC, Miller MJ. A Synthetic Dual Drug Sideromycin Induces Gram-Negative Bacteria To Commit Suicide with a Gram-Positive Antibiotic. J Med Chem 2018; 61:3845-3854. [PMID: 29554424 DOI: 10.1021/acs.jmedchem.8b00218] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Many antibiotics lack activity against Gram-negative bacteria because they cannot permeate the outer membrane or suffer from efflux and, in the case of β-lactams, are degraded by β-lactamases. Herein, we describe the synthesis and studies of a dual drug conjugate (1) of a siderophore linked to a cephalosporin with an attached oxazolidinone. The cephalosporin component of 1 is rapidly hydrolyzed by purified ADC-1 β-lactamase to release the oxazolidinone. Conjugate 1 is active against clinical isolates of Acinetobacter baumannii as well as strains producing large amounts of ADC-1 β-lactamase. Overall, the results are consistent with siderophore-mediated active uptake, inherent activity of the delivered dual drug, and in the presence of β-lactamases, intracellular release of the oxazolidinone upon cleavage of the cephalosporin to allow the freed oxazolidinone to inactivate its target. The ultimate result demonstrates that Gram-positive oxazolidinone antibiotics can be made to be effective against Gram-negative bacteria by β-lactamase triggered release.
Collapse
Affiliation(s)
- Rui Liu
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| | - Patricia A Miller
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| | - Sergei B Vakulenko
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| | - Nichole K Stewart
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| | - William C Boggess
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| | - Marvin J Miller
- Department of Chemistry and Biochemistry , University of Notre Dame , Notre Dame , Indiana 46556 , United States
| |
Collapse
|
29
|
van Geelen L, Meier D, Rehberg N, Kalscheuer R. (Some) current concepts in antibacterial drug discovery. Appl Microbiol Biotechnol 2018; 102:2949-2963. [PMID: 29455386 DOI: 10.1007/s00253-018-8843-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/30/2022]
Abstract
The rise of multidrug resistance in bacteria rendering pathogens unresponsive to many clinical drugs is widely acknowledged and considered a critical global healthcare issue. There is broad consensus that novel antibacterial chemotherapeutic options are extremely urgently needed. However, the development pipeline of new antibacterial drug lead structures is poorly filled and not commensurate with the scale of the problem since the pharmaceutical industry has shown reduced interest in antibiotic development in the past decades due to high economic risks and low profit expectations. Therefore, academic research institutions have a special responsibility in finding novel treatment options for the future. In this mini review, we want to provide a broad overview of the different approaches and concepts that are currently pursued in this research field.
Collapse
Affiliation(s)
- Lasse van Geelen
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University Düsseldorf, 40225, Dusseldorf, Germany
| | - Dieter Meier
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University Düsseldorf, 40225, Dusseldorf, Germany
| | - Nidja Rehberg
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University Düsseldorf, 40225, Dusseldorf, Germany
| | - Rainer Kalscheuer
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University Düsseldorf, 40225, Dusseldorf, Germany.
| |
Collapse
|
30
|
Deng F, Yu H, Pan X, Hu G, Wang Q, Peng R, Tan L, Yang Z. Ultra-high performance liquid chromatography tandem mass spectrometry for the determination of five glycopeptide antibiotics in food and biological samples using solid-phase extraction. J Chromatogr A 2018; 1538:54-59. [DOI: 10.1016/j.chroma.2018.01.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/14/2017] [Accepted: 01/17/2018] [Indexed: 10/18/2022]
|
31
|
The Road from Host-Defense Peptides to a New Generation of Antimicrobial Drugs. Molecules 2018; 23:molecules23020311. [PMID: 29389911 PMCID: PMC6017364 DOI: 10.3390/molecules23020311] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/23/2018] [Accepted: 01/30/2018] [Indexed: 01/28/2023] Open
Abstract
Host-defense peptides, also called antimicrobial peptides (AMPs), whose protective action has been used by animals for millions of years, fulfill many requirements of the pharmaceutical industry, such as: (1) broad spectrum of activity; (2) unlike classic antibiotics, they induce very little resistance; (3) they act synergically with conventional antibiotics; (4) they neutralize endotoxins and are active in animal models. However, it is considered that many natural peptides are not suitable for drug development due to stability and biodisponibility problems, or high production costs. This review describes the efforts to overcome these problems and develop new antimicrobial drugs from these peptides or inspired by them. The discovery process of natural AMPs is discussed, as well as the development of synthetic analogs with improved pharmacological properties. The production of these compounds at acceptable costs, using different chemical and biotechnological methods, is also commented. Once these challenges are overcome, a new generation of versatile, potent and long-lasting antimicrobial drugs is expected.
Collapse
|
32
|
Understanding drug resistance will improve the treatment of bacterial infections. Nat Rev Microbiol 2017; 15:639-640. [DOI: 10.1038/nrmicro.2017.121] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Carosso S, Liu R, Miller PA, Hecker SJ, Glinka T, Miller MJ. Methodology for Monobactam Diversification: Syntheses and Studies of 4-Thiomethyl Substituted β-Lactams with Activity against Gram-Negative Bacteria, Including Carbapenemase Producing Acinetobacter baumannii. J Med Chem 2017; 60:8933-8944. [PMID: 28994597 DOI: 10.1021/acs.jmedchem.7b01164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Bromine induced lactamization of vinyl acetohydroxamates facilitated syntheses of monocyclic β-lactams suitable for incorporation of a thiomethyl and extended functionality at the C(4) position. Elaboration of the resulting substituted N-hydroxy-2-azetidinones allowed incorporation of functionalized α-amino substituents appropriate for enhancement of antibiotic activity. Evaluation of antibacterial activity against a panel of Gram-positive and Gram-negative bacteria revealed structure-activity relationships (SAR) and identification of potent new monobactam antibiotics. The corresponding bis-catechol conjugate, 42, has excellent activity against Gram-negative bacteria including carbapenemase and carbacephalosporinase producing strains of Acinetobacter baumannii, which have been listed by the WHO as being of critical concern worldwide.
Collapse
Affiliation(s)
- Serena Carosso
- Department of Chemistry and Biochemistry, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Rui Liu
- Department of Chemistry and Biochemistry, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Patricia A Miller
- Department of Chemistry and Biochemistry, University of Notre Dame , Notre Dame, Indiana 46556, United States
| | - Scott J Hecker
- Rempex Pharmaceuticals, The Medicines Company , 3013 Science Park Road, First Floor, San Diego, California 92121, United States
| | - Tomasz Glinka
- Rempex Pharmaceuticals, The Medicines Company , 3013 Science Park Road, First Floor, San Diego, California 92121, United States
| | - Marvin J Miller
- Department of Chemistry and Biochemistry, University of Notre Dame , Notre Dame, Indiana 46556, United States
| |
Collapse
|
34
|
Galappathie S, Edwards DJ, Elliott AG, Cooper MA, Palombo EA, Butler MS, Mahon PJ. Antibacterial Nerol Cinnamates from the Australian Plant Eremophila longifolia. JOURNAL OF NATURAL PRODUCTS 2017; 80:1178-1181. [PMID: 28257200 DOI: 10.1021/acs.jnatprod.6b00888] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Two new antimicrobial agents, neryl ferulate (1) and neryl p-coumarate (2), were identified using bioassay-guided isolation from the leaves of Eremophila longifolia, which is a medicinal plant used by some Australian Aboriginal communities. Although gradual autoxidation of the nerol subunit hindered the initial attempts to purify and characterize 1 and 2, it was found that the autoxidation could be stopped through storage under argon at -20 °C. Biological evaluation showed that neryl ferulate (1) had moderate activity against various Gram-positive bacteria, while neryl p-coumarate (2) was active only against Enterococcus faecium.
Collapse
Affiliation(s)
- Savithri Galappathie
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology , Hawthorn 3122, Victoria, Australia
| | - David J Edwards
- Institute for Molecular Bioscience, University of Queensland , St Lucia 4072, Queensland, Australia
| | - Alysha G Elliott
- Institute for Molecular Bioscience, University of Queensland , St Lucia 4072, Queensland, Australia
| | - Matthew A Cooper
- Institute for Molecular Bioscience, University of Queensland , St Lucia 4072, Queensland, Australia
| | - Enzo A Palombo
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology , Hawthorn 3122, Victoria, Australia
| | - Mark S Butler
- Institute for Molecular Bioscience, University of Queensland , St Lucia 4072, Queensland, Australia
| | - Peter J Mahon
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology , Hawthorn 3122, Victoria, Australia
| |
Collapse
|
35
|
Ghosh M, Miller PA, Möllmann U, Claypool WD, Schroeder VA, Wolter WR, Suckow M, Yu H, Li S, Huang W, Zajicek J, Miller MJ. Targeted Antibiotic Delivery: Selective Siderophore Conjugation with Daptomycin Confers Potent Activity against Multidrug Resistant Acinetobacter baumannii Both in Vitro and in Vivo. J Med Chem 2017; 60:4577-4583. [DOI: 10.1021/acs.jmedchem.7b00102] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Manuka Ghosh
- Hsiri Therapeutics, Innovation Park, 1400 East Angela
Boulevard, South Bend, Indiana 46617, United States
| | - Patricia A. Miller
- Hsiri Therapeutics, Innovation Park, 1400 East Angela
Boulevard, South Bend, Indiana 46617, United States
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Ute Möllmann
- Hsiri Therapeutics, Innovation Park, 1400 East Angela
Boulevard, South Bend, Indiana 46617, United States
| | - William D. Claypool
- Hsiri Therapeutics, LLC, Rosetree Corporate Center, 1400 N. Providence Road,
Building 1, Suite 115S, Media, Pennsylvania 19063, United States
| | - Valerie A. Schroeder
- Frieman Life Sciences Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - William R. Wolter
- Frieman Life Sciences Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mark Suckow
- Frieman Life Sciences Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Honglin Yu
- PracticaChem, 5 Lanyuan Road, Room D-603, Huayuan
Industrial Park, Tianjin, 300384, China
| | - Shuang Li
- PracticaChem, 5 Lanyuan Road, Room D-603, Huayuan
Industrial Park, Tianjin, 300384, China
| | - Weiqiang Huang
- PracticaChem, 5 Lanyuan Road, Room D-603, Huayuan
Industrial Park, Tianjin, 300384, China
| | - Jaroslav Zajicek
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Marvin J. Miller
- Hsiri Therapeutics, Innovation Park, 1400 East Angela
Boulevard, South Bend, Indiana 46617, United States
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre
Dame, Indiana 46556, United States
| |
Collapse
|
36
|
New antibiotics from Nature’s chemical inventory. Bioorg Med Chem 2016; 24:6227-6252. [DOI: 10.1016/j.bmc.2016.09.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/07/2016] [Indexed: 01/07/2023]
|