1
|
Stappert M, Kohnhäuser D, Seedorf T, Coetzee J, Rox K, Fuchs HLS, Cirnski K, Leitner C, Herrmann J, Kirschning A, Müller R, Brönstrup M. Synthetic studies on the tetrasubstituted D-ring of cystobactamids lead to potent terephthalic acid antibiotics. Commun Chem 2024; 7:252. [PMID: 39501041 PMCID: PMC11538350 DOI: 10.1038/s42004-024-01337-6] [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: 08/08/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
Novel scaffolds for broad-spectrum antibiotics are rare and in strong demand because of the increase in antimicrobial resistance. The cystobactamids, discovered from myxobacterial sources, have a unique hexapeptidic scaffold with five arylamides and possess potent, resistance-breaking properties. This study investigates the role of the central D-ring pharmacophore in cystobactamids, a para-aminobenzoic acid (PABA) moiety that is additionally substituted by hydroxy and isopropoxy functions. We varied the two oxygenated substituents and replaced both amide connectors with bioisosteres. Synthetic routes were developed that included metal-mediated aromatic functionalization or heterocycle formations, leading to 19 novel analogues. The antibiotic efficacy of all analogues was determined against bacteria from the ESKAPE pathogen panel. While the replacement and the repositioning of hydroxy and isopropoxy substituents was not advantageous, exchanging PABA by terephthalic acid amides led to the highly potent analogue 42 with broad-spectrum activity, insensitivity towards AlbD-mediated degradation and promising pharmacokinetic properties in mice. The study highlights the steep structure-activity relationships in the tetrasubstituted D-ring and a surprisingly favorable reversion of the amide connecting C and D.
Collapse
Affiliation(s)
- Moritz Stappert
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover Schneiderberg 1B, Hanover, Germany
| | - Daniel Kohnhäuser
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tim Seedorf
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover Schneiderberg 1B, Hanover, Germany
| | - Janetta Coetzee
- Helmholtz Institute for Pharmaceutical Research Saarland, Universitätscampus E8.1, Saarbrücken, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), site Hannover Braunschweig, Braunschweig, Germany
| | - Hazel L S Fuchs
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katarina Cirnski
- Helmholtz Institute for Pharmaceutical Research Saarland, Universitätscampus E8.1, Saarbrücken, Germany
| | - Christian Leitner
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover Schneiderberg 1B, Hanover, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Universitätscampus E8.1, Saarbrücken, Germany
| | - Andreas Kirschning
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover Schneiderberg 1B, Hanover, Germany
- Uppsala Biomedical Center (BMC), University Uppsala, Uppsala, Sweden
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Universitätscampus E8.1, Saarbrücken, Germany
- German Center for Infection Research (DZIF), site Hannover Braunschweig, Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover Schneiderberg 1B, Hanover, Germany.
- German Center for Infection Research (DZIF), site Hannover Braunschweig, Braunschweig, Germany.
| |
Collapse
|
2
|
Solga D, Wieske LHE, Wilcox S, Zeilinger C, Jansen-Olliges L, Cirnski K, Herrmann J, Müller R, Erdelyi M, Kirschning A. Is Simultaneous Binding to DNA and Gyrase Important for the Antibacterial Activity of Cystobactamids? Chemistry 2024; 30:e202303796. [PMID: 38217886 DOI: 10.1002/chem.202303796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Cystobactamids are aromatic oligoamides that exert their natural antibacterial properties by inhibition of bacterial gyrases. Such aromatic oligoamides were proposed to inhibit α-helix-mediated protein-protein interactions and may serve for specific recognition of DNA. Based on this suggestion, we designed new derivatives that have duplicated cystobactamid triarene units as model systems to decipher the specific binding mode of cystobactamids to double stranded DNA. Solution NMR analyses revealed that natural cystobactamids as well as their elongated analogues show an overall bent shape at their central aliphatic unit, with an average CX-CY-CZ angle of ~110 degrees. Our finding is corroborated by the target-bound structure of close analogues, as established by cryo-EM very recently. Cystobactamid CN-861-2 binds directly to the bacterial gyrase with an affinity of 9 μM, and also exhibits DNA-binding properties with specificity for AT-rich DNA. Elongation/dimerization of the triarene subunit of native cystobactamids is demonstrated to lead to an increase in DNA binding affinity. This implies that cystobactamids' gyrase inhibitory activity necessitates not just interaction with the gyrase itself, but also with DNA via their triarene unit.
Collapse
Affiliation(s)
- Danny Solga
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Lianne H E Wieske
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, SE-752 37, Uppsala, Sweden
| | - Scott Wilcox
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, SE-752 37, Uppsala, Sweden
| | - Carsten Zeilinger
- Institute of Biophysics and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 38, 30167, Hannover, Germany
| | - Linda Jansen-Olliges
- Institute of Biophysics and Center of Biomolecular Drug Research (BMWZ), Leibniz University Hannover, Schneiderberg 38, 30167, Hannover, Germany
| | - Katarina Cirnski
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Mate Erdelyi
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, SE-752 37, Uppsala, Sweden
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| |
Collapse
|
3
|
Zborovsky L, Kleebauer L, Seidel M, Kostenko A, von Eckardstein L, Gombert FO, Weston J, Süssmuth RD. Improvement of the antimicrobial potency, pharmacokinetic and pharmacodynamic properties of albicidin by incorporation of nitrogen atoms. Chem Sci 2021; 12:14606-14617. [PMID: 34881013 PMCID: PMC8580050 DOI: 10.1039/d1sc04019g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/17/2021] [Indexed: 01/22/2023] Open
Abstract
The worrisome development and spread of multidrug-resistant bacteria demands new antibacterial agents with strong bioactivities particularly against Gram-negative bacteria. Albicidins were recently structurally characterized as highly active antibacterial natural products from the bacterium Xanthomonas albilineans. Albicidin, which effectively targets the bacterial DNA-gyrase, is a lipophilic hexapeptide mostly consisting of para amino benzoic acid units and only one α-amino acid. In this study, we report on the design and synthesis of new albicidins, containing N-atoms on each of the 5 different phenyl rings. We systematically introduced N-atoms into the aromatic backbone to monitor intramolecular H-bonds and for one derivative correlated them with a significant enhancement of the antibacterial activity and activity spectrum, particularly also towards Gram-positive bacteria. In parallel we conducted DFT calculations to find the most stable conformation of each derivative. A drastic angle-change was observed for the lead compound and shows a preferred planarity through H-bonding with the introduced N-atom at the D-fragment of albicidin. Finally, we went to the next level and conducted the first in vivo experiments with an albicidin analogue. Our lead compound was evaluated in two different mouse experiments: In the first we show a promising PK profile and the absence of toxicity and in the second very good efficiency and reduction of the bacterial titre in an E. coli infection model with FQ-resistant clinically relevant strains. These results qualify albicidins as active antibacterial substances with the potential to be developed as a drug for treatment of infections caused by Gram-negative and Gram-positive bacteria.
Collapse
Affiliation(s)
- Lieby Zborovsky
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Leonardo Kleebauer
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Maria Seidel
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Arseni Kostenko
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Leonard von Eckardstein
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Frank Otto Gombert
- Gombert Pharma Research Solutions (GPRS) Dornacherstrasse 120 CH 4053 Basel Switzerland
| | - John Weston
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| | - Roderich D Süssmuth
- Institut für Organische Chemie, Technische Universität Berlin Straße des 17. Juni 124 10623 Berlin Germany
| |
Collapse
|
4
|
Wang Z, Kasper A, Mehmood R, Ternei M, Li S, Freundlich JS, Brady SF. Metagenome-Guided Analogue Synthesis Yields Improved Gram-Negative-Active Albicidin- and Cystobactamid-Type Antibiotics. Angew Chem Int Ed Engl 2021; 60:22172-22177. [PMID: 34355488 DOI: 10.1002/anie.202104874] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/05/2021] [Indexed: 11/11/2022]
Abstract
Natural products are a major source of new antibiotics. Here we utilize biosynthetic instructions contained within metagenome-derived congener biosynthetic gene clusters (BGCs) to guide the synthesis of improved antibiotic analogues. Albicidin and cystobactamid are the first members of a new class of broad-spectrum ρ-aminobenzoic acid (PABA)-based antibiotics. Our search for PABA-specific adenylation domain sequences in soil metagenomes revealed that BGCs in this family are common in nature. Twelve BGCs that were bio-informatically predicted to encode six new congeners were recovered from soil metagenomic libraries. Synthesis of these six predicted structures led to the identification of potent antibiotics with changes in their spectrum of activity and the ability to circumvent resistance conferred by endopeptidase cleavage enzymes.
Collapse
Affiliation(s)
- Zongqiang Wang
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Amanda Kasper
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Rabia Mehmood
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Melinda Ternei
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Shaogang Li
- Department of Medicine, Center for Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Joel S Freundlich
- Department of Medicine, Center for Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
5
|
Wang Z, Kasper A, Mehmood R, Ternei M, Li S, Freundlich JS, Brady SF. Metagenome‐Guided Analogue Synthesis Yields Improved Gram‐Negative‐Active Albicidin‐ and Cystobactamid‐Type Antibiotics. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202104874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Zongqiang Wang
- Laboratory of Genetically Encoded Small Molecules The Rockefeller University 1230 York Avenue New York NY 10065 USA
| | - Amanda Kasper
- Laboratory of Genetically Encoded Small Molecules The Rockefeller University 1230 York Avenue New York NY 10065 USA
| | - Rabia Mehmood
- Laboratory of Genetically Encoded Small Molecules The Rockefeller University 1230 York Avenue New York NY 10065 USA
| | - Melinda Ternei
- Laboratory of Genetically Encoded Small Molecules The Rockefeller University 1230 York Avenue New York NY 10065 USA
| | - Shaogang Li
- Department of Medicine, Center for Emerging and Re-emerging Pathogens Rutgers University—New Jersey Medical School Newark NJ 07103 USA
| | - Joel S. Freundlich
- Department of Medicine, Center for Emerging and Re-emerging Pathogens Rutgers University—New Jersey Medical School Newark NJ 07103 USA
| | - Sean F. Brady
- Laboratory of Genetically Encoded Small Molecules The Rockefeller University 1230 York Avenue New York NY 10065 USA
| |
Collapse
|
6
|
Hawkins PME, Liu DY, Linington RG, Payne RJ. Solid-phase synthesis of coralmycin A/ epi-coralmycin A and desmethoxycoralmycin A. Org Biomol Chem 2021; 19:6291-6300. [PMID: 34212970 DOI: 10.1039/d1ob01062j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The total synthesis of the natural product coralmycin A/epi-coralmycin A, as well as a desmethoxy analogue is described. Synthesis was achieved via a divergent, bidirectional solid-phase strategy, including a key on-resin O-acylation, O to N acyl shift, and O-alkylation protocol to incorporate the unusual 4-amino-2-hydroxy-3-isopropoxybenzoic acid motifs. The synthetic natural product was generated as a 1 : 1 mixture of epimers at the central β-methoxyasparagine residue and exhibited potent antibacterial activity against a panel of ten Gram-negative and seven Gram-positive organisms. The desmethoxy analogue possessed significantly more potent antimicrobial activity against this panel with minimal inhibitory concentrations (MICs) as low as 50 nM.
Collapse
Affiliation(s)
- Paige M E Hawkins
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia. and Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, NSW 2006, Australia
| | - Dennis Y Liu
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia. and Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Behroz I, Kleebauer L, Hommernick K, Seidel M, Grätz S, Mainz A, Weston JB, Süssmuth RD. Acetylenic Replacement of Albicidin's Methacrylamide Residue Circumvents Detrimental E/Z Photoisomerization and Preserves Antibacterial Activity. Chemistry 2021; 27:9077-9086. [PMID: 33769627 PMCID: PMC8362182 DOI: 10.1002/chem.202100523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 01/07/2023]
Abstract
The natural product albicidin is a highly potent inhibitor of bacterial DNA gyrase. Its outstanding activity, particularly against Gram-negative pathogens, qualifies it as a promising lead structure in the search for new antibacterial drugs. However, as we show here, the N-terminal cinnamoyl moiety of albicidin is susceptible to photochemical E/Z isomerization. Moreover, the newly formed Z isomer exhibits significantly reduced antibacterial activity, which hampers the development and biological evaluation of albicidin and potent derivatives thereof. Hence, we synthesized 13 different variants of albicidin in which the vulnerable para-coumaric acid moiety was replaced; this yielded photostable analogues. Biological activity assays revealed that diaryl alkyne analogues exhibited virtually undiminished antibacterial efficacy. This promising scaffold will therefore serve as a blueprint for the design of a potent albicidin-based drug.
Collapse
Affiliation(s)
- Iraj Behroz
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Leonardo Kleebauer
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Kay Hommernick
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Maria Seidel
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Stefan Grätz
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Andi Mainz
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - John B. Weston
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| | - Roderich D. Süssmuth
- Institut für Organische ChemieTechnische Universität BerlinStraße des 17. Juni 12410623BerlinGermany
| |
Collapse
|
8
|
Seedorf T, Kirschning A, Solga D. Natural and Synthetic Oligoarylamides: Privileged Structures for Medical Applications. Chemistry 2021; 27:7321-7339. [PMID: 33481284 PMCID: PMC8251530 DOI: 10.1002/chem.202005086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 12/13/2022]
Abstract
The term "privileged structure" refers to a single molecular substructure or scaffold that can serve as a starting point for high-affinity ligands for more than one receptor type. In this report, a hitherto overlooked group of privileged substructures is addressed, namely aromatic oligoamides, for which there are natural models in the form of cystobactamids, albicidin, distamycin A, netropsin, and others. The aromatic and heteroaromatic core, together with a flexible selection of substituents, form conformationally well-defined scaffolds capable of specifically binding to conformationally well-defined regions of biomacromolecules such as helices in proteins or DNA often by acting as helices mimics themselves. As such, these aromatic oligoamides have already been employed to inhibit protein-protein and nucleic acid-protein interactions. This article is the first to bring together the scattered knowledge about aromatic oligoamides in connection with biomedical applications.
Collapse
Affiliation(s)
- Tim Seedorf
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum, (BMWZ)Leibniz Universität HannoverSchneiderberg 1B30167HannoverGermany
| | - Andreas Kirschning
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum, (BMWZ)Leibniz Universität HannoverSchneiderberg 1B30167HannoverGermany
| | - Danny Solga
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum, (BMWZ)Leibniz Universität HannoverSchneiderberg 1B30167HannoverGermany
| |
Collapse
|
9
|
Groß S, Schnell B, Haack PA, Auerbach D, Müller R. In vivo and in vitro reconstitution of unique key steps in cystobactamid antibiotic biosynthesis. Nat Commun 2021; 12:1696. [PMID: 33727542 PMCID: PMC7966384 DOI: 10.1038/s41467-021-21848-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 02/13/2021] [Indexed: 01/31/2023] Open
Abstract
Cystobactamids are myxobacteria-derived topoisomerase inhibitors with potent anti-Gram-negative activity. They are formed by a non-ribosomal peptide synthetase (NRPS) and consist of tailored para-aminobenzoic acids, connected by a unique α-methoxy-L-isoasparagine or a β-methoxy-L-asparagine linker moiety. We describe the heterologous expression of the cystobactamid biosynthetic gene cluster (BGC) in Myxococcus xanthus. Targeted gene deletions produce several unnatural cystobactamids. Using in vitro experiments, we reconstitute the key biosynthetic steps of linker formation and shuttling via CysB to the NRPS. The biosynthetic logic involves a previously uncharacterized bifunctional domain found in the stand-alone NRPS module CysH, albicidin biosynthesis and numerous BGCs of unknown natural products. This domain performs either an aminomutase (AM) or an amide dehydratase (DH) type of reaction, depending on the activity of CysJ which hydroxylates CysH-bound L-asparagine. Furthermore, CysQ O-methylates hydroxyl-L-(iso)asparagine only in the presence of the AMDH domain. Taken together, these findings provide direct evidence for unique steps in cystobactamid biosynthesis.
Collapse
Affiliation(s)
- Sebastian Groß
- grid.11749.3a0000 0001 2167 7588Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany ,grid.452463.2DZIF - German Centre for Infection Research, Partnersite Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Bastien Schnell
- grid.11749.3a0000 0001 2167 7588Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany ,grid.452463.2DZIF - German Centre for Infection Research, Partnersite Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Patrick A. Haack
- grid.11749.3a0000 0001 2167 7588Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany ,grid.452463.2DZIF - German Centre for Infection Research, Partnersite Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - David Auerbach
- grid.11749.3a0000 0001 2167 7588Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany ,grid.452463.2DZIF - German Centre for Infection Research, Partnersite Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Rolf Müller
- grid.11749.3a0000 0001 2167 7588Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany ,grid.11749.3a0000 0001 2167 7588Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany ,grid.452463.2DZIF - German Centre for Infection Research, Partnersite Hannover-Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
10
|
Moreira R, Noden M, Taylor SD. Synthesis of Azido Acids and Their Application in the Preparation of Complex Peptides. SYNTHESIS-STUTTGART 2020. [DOI: 10.1055/s-0040-1707314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractAzido acids are important synthons for the synthesis of complex peptides. As a protecting group, the azide moiety is atom-efficient, easy to install and can be reduced in the presence of many other protecting groups, making it ideal for the synthesis of branched and/or cyclic peptides. α-Azido acids are less bulky than urethane-protected counterparts and react more effectively in coupling reactions of difficult-to-form peptide and ester bonds. Azido acids can also be used to form azoles on complex intermediates. This review covers the synthesis of azido acids and their application to the total synthesis of complex peptide natural products.1 Introduction2 Synthesis of α-Azido Acids2.1 From α-Amino Acids or Esters2.2 Via α-Substitution2.3 Via Electrophilic Azidation2.4 Via Condensation of N-2-Azidoacetyl-4-Phenylthiazolidin- 2-Thi one Enolates with Aldehydes and Acetals2.5 Synthesis of α,β-Unsaturated α-Azido Acids and Esters3 Synthesis of β-Azido Acids3.1 Preparation of Azidoalanine and 3-Azido-2-aminobutanoic Acids3.2 General Approaches to Preparing β-Azido Acids Other Than Azi doalanine and AABA4 Azido Acids in Total Synthesis4.1 α-Azido Acids4.2 β-Azido Acids and Azido Acids Containing an Azide on the Side
Chain5 Conclusions
Collapse
|
11
|
Elgaher WAM, Hamed MM, Baumann S, Herrmann J, Siebenbürger L, Krull J, Cirnski K, Kirschning A, Brönstrup M, Müller R, Hartmann RW. Cystobactamid 507: Concise Synthesis, Mode of Action, and Optimization toward More Potent Antibiotics. Chemistry 2020; 26:7219-7225. [PMID: 31984562 PMCID: PMC7317206 DOI: 10.1002/chem.202000117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Indexed: 12/23/2022]
Abstract
Lack of new antibiotics and increasing antimicrobial resistance are among the main concerns of healthcare communities nowadays, and these concerns necessitate the search for novel antibacterial agents. Recently, we discovered the cystobactamids—a novel natural class of antibiotics with broad‐spectrum antibacterial activity. In this work, we describe 1) a concise total synthesis of cystobactamid 507, 2) the identification of the bioactive conformation using noncovalently bonded rigid analogues, and 3) the first structure–activity relationship (SAR) study for cystobactamid 507 leading to new analogues with high metabolic stability, superior topoisomerase IIA inhibition, antibacterial activity and, importantly, stability toward the resistant factor AlbD. Deeper insight into the mode of action revealed that the cystobactamids employ DNA minor‐groove binding as part of the drug–target interaction without showing significant intercalation. By designing a new analogue of cystobactamid 919‐2, we finally demonstrated that these findings could be further exploited to obtain more potent hexapeptides against Gram‐negative bacteria.
Collapse
Affiliation(s)
- Walid A M Elgaher
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Mostafa M Hamed
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Sascha Baumann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | | | - Jana Krull
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Katarina Cirnski
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University of Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| | - Rolf W Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
| |
Collapse
|
12
|
Planke T, Cirnski K, Herrmann J, Müller R, Kirschning A. Synthetic and Biological Studies on New Urea and Triazole Containing Cystobactamid Derivatives. Chemistry 2020; 26:4289-4296. [PMID: 31834653 PMCID: PMC7186842 DOI: 10.1002/chem.201904073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Indexed: 12/02/2022]
Abstract
Cystobactamids belong to the group of arene-based oligoamides that effectively inhibit bacterial type IIa topoisomerases. Cystobactamid 861-2 is the most active member of these antibiotics. Most amide bonds present in the cystobactamids link benzoic acids with anilines and it was found that some of these amide bonds undergo chemical and enzymatic hydrolysis, especially the one linking ring C with ring D. This work reports on the chemical synthesis and biological evaluation of thirteen new cystobactamids that still contain the methoxyaspartate hinge. However, we exchanged selected amide bonds either by the urea or the triazole groups and modified ring A in the latter case. While hydrolytic stability could be improved with these structural substitutes, the high antibacterial potency of cystobactamid 861-2 could only be preserved in selected cases. This includes derivatives, in which the urea group is positioned between rings A and B and where the triazole is found between rings C and D.
Collapse
Affiliation(s)
- Therese Planke
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Katarina Cirnski
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Jennifer Herrmann
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Rolf Müller
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Andreas Kirschning
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ), Leibniz Universität Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| |
Collapse
|
13
|
Goes A, Lapuhs P, Kuhn T, Schulz E, Richter R, Panter F, Dahlem C, Koch M, Garcia R, Kiemer AK, Müller R, Fuhrmann G. Myxobacteria-Derived Outer Membrane Vesicles: Potential Applicability Against Intracellular Infections. Cells 2020; 9:cells9010194. [PMID: 31940898 PMCID: PMC7017139 DOI: 10.3390/cells9010194] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
In 2019, it was estimated that 2.5 million people die from lower tract respiratory infections annually. One of the main causes of these infections is Staphylococcus aureus, a bacterium that can invade and survive within mammalian cells. S. aureus intracellular infections are difficult to treat because several classes of antibiotics are unable to permeate through the cell wall and reach the pathogen. This condition increases the need for new therapeutic avenues, able to deliver antibiotics efficiently. In this work, we obtained outer membrane vesicles (OMVs) derived from the myxobacteria Cystobacter velatus strain Cbv34 and Cystobacter ferrugineus strain Cbfe23, that are naturally antimicrobial, to target intracellular infections, and investigated how they can affect the viability of epithelial and macrophage cell lines. We evaluated by cytometric bead array whether they induce the expression of proinflammatory cytokines in blood immune cells. Using confocal laser scanning microscopy and flow cytometry, we also investigated their interaction and uptake into mammalian cells. Finally, we studied the effect of OMVs on planktonic and intracellular S. aureus. We found that while Cbv34 OMVs were not cytotoxic to cells at any concentration tested, Cbfe23 OMVs affected the viability of macrophages, leading to a 50% decrease at a concentration of 125,000 OMVs/cell. We observed only little to moderate stimulation of release of TNF-alpha, IL-8, IL-6 and IL-1beta by both OMVs. Cbfe23 OMVs have better interaction with the cells than Cbv34 OMVs, being taken up faster by them, but both seem to remain mostly on the cell surface after 24 h of incubation. This, however, did not impair their bacteriostatic activity against intracellular S. aureus. In this study, we provide an important basis for implementing OMVs in the treatment of intracellular infections.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Philipp Lapuhs
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Thomas Kuhn
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Eilien Schulz
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Robert Richter
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Drug Delivery (DDEL), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Fabian Panter
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Marcus Koch
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany;
| | - Ronald Garcia
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Rolf Müller
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
| | - Gregor Fuhrmann
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Correspondence: ; Tel.: +49-68-198-806 (ext. 1500)
| |
Collapse
|
14
|
Testolin G, Cirnski K, Rox K, Prochnow H, Fetz V, Grandclaudon C, Mollner T, Baiyoumy A, Ritter A, Leitner C, Krull J, van den Heuvel J, Vassort A, Sordello S, Hamed MM, Elgaher WAM, Herrmann J, Hartmann RW, Müller R, Brönstrup M. Synthetic studies of cystobactamids as antibiotics and bacterial imaging carriers lead to compounds with high in vivo efficacy. Chem Sci 2019; 11:1316-1334. [PMID: 34123255 PMCID: PMC8148378 DOI: 10.1039/c9sc04769g] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is an alarming scarcity of novel chemical matter with bioactivity against multidrug-resistant Gram-negative bacterial pathogens. Cystobactamids, recently discovered natural products from myxobacteria, are an exception to this trend. Their unusual chemical structure, composed of oligomeric para-aminobenzoic acid moieties, is associated with a high antibiotic activity through the inhibition of gyrase. In this study, structural determinants of cystobactamid's antibacterial potency were defined at five positions, which were varied using three different synthetic routes to the cystobactamid scaffold. The potency against Acinetobacter baumannii could be increased ten-fold to an MIC (minimum inhibitory concentration) of 0.06 μg mL-1, and the previously identified spectrum gap of Klebsiella pneumoniae could be closed compared to the natural products (MIC of 0.5 μg mL-1). Proteolytic degradation of cystobactamids by the resistance factor AlbD was prevented by an amide-triazole replacement. Conjugation of cystobactamid's N-terminal tetrapeptide to a Bodipy moiety induced the selective localization of the fluorophore for bacterial imaging purposes. Finally, a first in vivo proof of concept was obtained in an E. coli infection mouse model, where derivative 22 led to the reduction of bacterial loads (cfu, colony-forming units) in muscle, lung and kidneys by five orders of magnitude compared to vehicle-treated mice. These findings qualify cystobactamids as highly promising lead structures against infections caused by Gram-positive and Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Giambattista Testolin
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Katarina Cirnski
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany.,Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany .,German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Hans Prochnow
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Verena Fetz
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Charlotte Grandclaudon
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany .,German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Tim Mollner
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Alain Baiyoumy
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Antje Ritter
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Christian Leitner
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany .,German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Jana Krull
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Joop van den Heuvel
- Group Recombinant Protein Expression, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Aurelie Vassort
- Evotec ID 1541 Avenue Marcel Merieux 69289 Marcy l'Etoile France
| | | | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Walid A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Jennifer Herrmann
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany.,Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Rolf W Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Rolf Müller
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany.,Helmholtz Institute for Pharmaceutical Research Saarland Universitätscampus E8.1 66123 Saarbrücken Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research Inhoffenstrasse 7 38124 Braunschweig Germany .,German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany.,Center of Biomolecular Drug Research (BMWZ), Leibniz Universität 30167 Hannover Germany
| |
Collapse
|
15
|
Moeller M, Norris MD, Planke T, Cirnski K, Herrmann J, Müller R, Kirschning A. Scalable Syntheses of Methoxyaspartate and Preparation of the Antibiotic Cystobactamid 861-2 and Highly Potent Derivatives. Org Lett 2019; 21:8369-8372. [DOI: 10.1021/acs.orglett.9b03143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Malte Moeller
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ) der Leibniz Universität Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Matthew D. Norris
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ) der Leibniz Universität Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Therese Planke
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ) der Leibniz Universität Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Katarina Cirnski
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123 Saarbrücken, Germany
| | - Jennifer Herrmann
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Abteilung Mikrobielle Naturstoffe, Helmholtz Institut für Pharmazeutische Forschung Saarland, Helmholtz Zentrum für Infektionsforschung und Universität des Saarlandes, Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreas Kirschning
- Institut für Organische Chemie und Biomolekulares Wirkstoffzentrum (BMWZ) der Leibniz Universität Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| |
Collapse
|