1
|
Kircheva N, Angelova S, García-Iriepa C, Marazzi M, Dudev T. Thermodynamics of the Ga 3+/ Fe 3+ Competition in a Model of the Heme B-Containing Bacterial Catalase Active Center. Inorg Chem 2025. [PMID: 40329694 DOI: 10.1021/acs.inorgchem.4c05527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Antibiotic resistance presents an enormous threat to human well-being due to the overconsumption and misuse of these essential drugs in recent years. A novel and intriguing path to overcoming the ever-pressing problem appears in the "Trojan horse" strategy exploiting bacteria's internalization systems and their exceptional capability to scavenge metal ions, iron in particular, from the surrounding media when evading the host organism. A promising candidate in this field is the abiogenic cation gallium─a ferric mimetic species, known to exert diverse effects, with its well-pronounced antibacterial activity attracting the attention of scientists in the past decade. In the study presented herewith, the computational chemistry methods, based on Density Functional Theory (DFT), are utilized in order to differentiate those outer factors contributing to gallium's ability to substitute the native ferric ion in the active site of the enzyme catalase. The characteristics of the surrounding media such as pH and solvent exposure, the composition of the protein shell, the nature of the metal, and different substrate molecules have been taken into account. The obtained results are interpreted in light of the experimentally reported observations and aim to contribute to deciphering this aspect of gallium's mechanism of antibacterial activity.
Collapse
Affiliation(s)
- Nikoleta Kircheva
- Institute of Optical Materials and Technologies "Acad. J. Malinowski", Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
| | - Silvia Angelova
- Institute of Optical Materials and Technologies "Acad. J. Malinowski", Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
- University of Chemical Technology and Metallurgy, 8 St. Kliment Ohridski Blvd, Sofia 1756, Bulgaria
| | - Cristina García-Iriepa
- Universidad de Alcalá, Departamento de Química Analítica, Química Física e Ingeniería Química, Functional Molecular Systems (FuMSys) group, Ctra, Madrid-Barcelona km. 33, 600, Alcalá de Henares, Madrid 28801, Spain
- Universidad de Alcalá, Instituto de Investigación Química "Andrés M. del Río" (IQAR), Ctra, Madrid-Barcelona km. 33, 600, Alcalá de Henares, Madrid 28801, Spain
| | - Marco Marazzi
- Universidad de Alcalá, Departamento de Química Analítica, Química Física e Ingeniería Química, Functional Molecular Systems (FuMSys) group, Ctra, Madrid-Barcelona km. 33, 600, Alcalá de Henares, Madrid 28801, Spain
- Universidad de Alcalá, Instituto de Investigación Química "Andrés M. del Río" (IQAR), Ctra, Madrid-Barcelona km. 33, 600, Alcalá de Henares, Madrid 28801, Spain
| | - Todor Dudev
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kl. Ohridski", Sofia 1164, Bulgaria
| |
Collapse
|
2
|
Bredael K, Vanhecke F, Vandenheede E, Stevens CV, Gobec S, D'hooghe M. Synthesis and Antibacterial Properties of 3-Amino-β-Lactams Bearing a Heteroatom-Containing C4 Substituent. ChemMedChem 2025; 20:e202400994. [PMID: 39906996 DOI: 10.1002/cmdc.202400994] [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: 12/12/2024] [Revised: 01/21/2025] [Accepted: 02/04/2025] [Indexed: 02/06/2025]
Abstract
The rise of antimicrobial resistance has spurred the search for innovative antibiotics, with monocyclic 3-amino-β-lactams - aztreonam standing out as key example - showing significant potential. In particular, C4-functionalized 3-amino-β-lactams have emerged as a promising subclass that can potentially improve the activity, stability and cellular permeability of the compounds. This review outlines various synthetic methodologies available for the construction of 3-amino-β-lactams bearing a heteroatom-containing substituent at C4, with the heteroatom connected to the ring system either directly or via a methylene bridge. Special attention is devoted to 3-amino-4-hydroxymethyl-β-lactams and 3-amino-4-acetoxy-β-lactams as versatile synthetic intermediates. Moreover, the effect of these C4 substituents on the biological activity of the corresponding 3-amino-β-lactams is discussed in detail. A better understanding of synthetic protocols and antibacterial properties related to this underexplored class of monocyclic 3-amino-β-lactams might contribute to address the current antibiotics problems we are facing more efficiently.
Collapse
Affiliation(s)
- Kato Bredael
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Fien Vanhecke
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Emma Vandenheede
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Christian V Stevens
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Bresson J, Burke K, Ethève‐Quelquejeu M, Iannazzo L, Cariou K. Metal-Catalyzed Cross-Coupling for the Synthesis of β-Lactam Drugs and Related Chemical Probes. ChemMedChem 2025; 20:e202400960. [PMID: 39963822 PMCID: PMC12058247 DOI: 10.1002/cmdc.202400960] [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: 11/27/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Antimicrobial resistance is a major public health threat, due to the emergence of new bacterial strains not responding to classical antibiotics. This review focuses on the use of transition metal cross-coupling strategies used to access new β-lactam derivatives, the most well-known and commonly used antibiotics. This manuscript covers the seminal studies for the synthesis of antibiotics up to the current need of accessing specific probes (by functionalizing existing drugs), crucial for the detection of resistances. These strategies also allow the linkage of a cargo to a β-lactam antibiotic for selective release for either therapeutic effect or for diagnostic purposes (in the case of probes), which will be explained in this article.
Collapse
Affiliation(s)
- Jules Bresson
- Institute of Chemistry for Life and Health SciencesLaboratory for Inorganic Chemical BiologyChimie ParisTechPSL UniversityCNRS11 rue Pierre et Marie Curie75005ParisFrance
| | - Katie Burke
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesUniversité Paris CitéCNRSF-75006ParisFrance
| | - Mélanie Ethève‐Quelquejeu
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesUniversité Paris CitéCNRSF-75006ParisFrance
- Institut Universitaire de FranceParis75006France
| | - Laura Iannazzo
- Laboratoire de Chimie et Biochimie Pharmacologiques et ToxicologiquesUniversité Paris CitéCNRSF-75006ParisFrance
| | - Kevin Cariou
- Institute of Chemistry for Life and Health SciencesLaboratory for Inorganic Chemical BiologyChimie ParisTechPSL UniversityCNRS11 rue Pierre et Marie Curie75005ParisFrance
| |
Collapse
|
4
|
Li X, Dong S, Pan Q, Liu N, Zhang Y. Antibiotic conjugates: Using molecular Trojan Horses to overcome drug resistance. Biomed Pharmacother 2025; 186:118007. [PMID: 40268370 DOI: 10.1016/j.biopha.2025.118007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 04/25/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a global health crisis due to the rapid emergence of multi-drug-resistant bacteria. The paucity of novel antibiotics in the clinical pipeline has exacerbated this issue, thereby warranting the development of new antibacterial therapies. The 'Trojan Horse' strategy entails conjugating antibiotics with bioactive components that not only facilitate the entry of antibiotic molecules into bacterial cells by circumventing the membrane barriers, but also augment the effects of conventional antibiotics against recalcitrant pathogens. These Trojan Horse elements can also serve as a promising tool for repurposing drugs with hitherto unexamined antimicrobial activity, or drugs with limited clinical utility due to considerable toxic side effects. In this review, we have discussed the current state of research on antibiotic conjugates with monoclonal antibodies (mAbs), antimicrobial peptides (AMPs) and the iron-chelating siderophores. The rationale and mechanisms of different antibiotic conjugates have been summarized, and the preclinical and clinical evidence pertaining to the activity of these conjugates against drug-resistant pathogens have been reviewed. Furthermore, the challenges associated with the clinical translation of these novel antimicrobials, and the future research directions have also been discussed. While antibiotic conjugates offer an attractive alternative to conventional antimicrobials, there are several obstacles to their clinical translation. A greater understanding of the mechanisms underlying AMR, and continuing advances in genetic engineering, synthetic biology, and bioinformatics will be crucial in designing more selective, potent, and safe antibiotic conjugates for tackling multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Rehabilitation, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Miao ZY, Lin J, Chen WM. Natural sideromycins and siderophore-conjugated natural products as inspiration for novel antimicrobial agents. Eur J Med Chem 2025; 287:117333. [PMID: 39892091 DOI: 10.1016/j.ejmech.2025.117333] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
The widespread emergence of multidrug-resistant (MDR) Gram-negative pathogens has posed a major challenge to clinical anti-infective therapy, and new effective treatments are urgently needed. A promising "Trojan horse" strategy involves conjugating antibiotics to siderophore molecules; the resulting siderophore-antibiotic conjugates (SACs) deliver antibiotics directly into cells by hijacking the sophisticated iron transport systems of Gram-negative bacteria, bypassing the outer membrane permeability barrier to enhance uptake and antibacterial efficacy. The clinical release of the first siderophore-antibiotic conjugate, cefiderocol, has aroused tremendous interest in the field among researchers and pharmaceutical companies. To date, most of the reported SACs have focused on the conjugation of siderophores to traditional antibacterial drugs. However, these antibacterial agents designed on the basis of the traditional antibiotic skeleton theoretically bear the risk of cross-resistance caused by shared molecular scaffolds. In this case, exploring novel natural product antibacterial conjugate scaffolds to circumvent the risk of early cross-resistance represents a presumably more sustainable approach for the development of SACs. In this review, we systematically summarize the research progress on siderophore-natural product conjugates as novel antimicrobial agents reported since 2010. Additionally, we propose challenges to be overcome and prospects for future development in this field.
Collapse
Affiliation(s)
- Zhi-Ying Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China
| | - Jing Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| | - Wei-Min Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 511400, China.
| |
Collapse
|
6
|
Mayegowda SB, Gadilingappa MN. Microbial Siderophores: A New Insight on Healthcare Applications. BME FRONTIERS 2025; 6:0112. [PMID: 40124737 PMCID: PMC11927942 DOI: 10.34133/bmef.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Globally, increased illness and disorders have gained importance in improvising therapeutics to help extend the lifespan of an individual. In this scenario, understanding the mechanism of bacterial pathogenicity linked to the interaction between the host and the pathogen focusing on essential metal ions is necessary. Numerous studies indicate that the severity of a disease might be due to the reduced availability of iron, linked to abnormal production or lack of acquisition systems. However, several microbes produce siderophores as virulence factors, low-molecular-weight organic compounds for acquisition of iron by iron-chelating systems. In medical applications, siderophores are employed in novel strategies in order to design effective new drugs and vaccines, targeting and delivering antibiotics to target sites in multidrug-resistant pathogens. Meanwhile, some types of siderophores are used as drug delivery modalities and antimalarial, anticancer, and antibacterial agents, for example, by employing conjugation techniques such as Trojan horse delivery. Hence, the current review integrates several applications of siderophores with an overview covering taxonomy, organisms producing iron affinity carriers, and their acquisition mechanism. This understanding may delineate newer opportunities to adapt possible therapies and/or treatments against several multidrug-resistant pathogens, representing a crucial solution for public health problems worldwide.
Collapse
|
7
|
Davidov Y, Tejman-Yarden N, Robinson A, Rahav G, Nissan I. Enterobactin and salmochelin S4 inhibit the growth of Staphylococcus aureus. Front Cell Infect Microbiol 2025; 15:1456046. [PMID: 40110026 PMCID: PMC11919883 DOI: 10.3389/fcimb.2025.1456046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
There is increasing demand for novel antimicrobial agents to tackle the antimicrobial resistance crisis. Here we report that two Enterobacteriaceae-produced siderophores, enterobactin and salmochelin S4, inhibit the growth of Staphylococcus aureus isolates, including methicillin-resistance S. aureus (MRSA) clinical isolates. The IC50 for different S. aureus isolates were 2-5 µM for salmochelin S4 and 5-10 µM for enterobactin. This inhibitory activity was partially repressed by adding Fe+3. These siderophores also inhibited the growth of Enterococcus strains, including vancomycin-resistant enterococci (VRE) clinical isolates, though less effectively than for S. aureus. The growth of various Gram-negative bacteria was barely affected by these siderophores. These results shed new light on the role of enterobactin and salmochelin in bacterial physiology and ecology and have potential for the development of novel strategies to combat the rapid rise of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yaacov Davidov
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Noa Tejman-Yarden
- Department of Laboratories, Public Health Directorate, Ministry of Health, Jerusalem, Israel
| | - Ari Robinson
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Israel Nissan
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Department of Avian Diseases, Kimron Veterinary Institute, Beit Dagan, Israel
| |
Collapse
|
8
|
Kharga K, Jha S, Vishwakarma T, Kumar L. Current developments and prospects of the antibiotic delivery systems. Crit Rev Microbiol 2025; 51:44-83. [PMID: 38425122 DOI: 10.1080/1040841x.2024.2321480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Antibiotics have remained the cornerstone for the treatment of bacterial infections ever since their discovery in the twentieth century. The uproar over antibiotic resistance among bacteria arising from genome plasticity and biofilm development has rendered current antibiotic therapies ineffective, urging the development of innovative therapeutic approaches. The development of antibiotic resistance among bacteria has further heightened the clinical failure of antibiotic therapy, which is often linked to its low bioavailability, side effects, and poor penetration and accumulation at the site of infection. In this review, we highlight the potential use of siderophores, antibodies, cell-penetrating peptides, antimicrobial peptides, bacteriophages, and nanoparticles to smuggle antibiotics across impermeable biological membranes to achieve therapeutically relevant concentrations of antibiotics and combat antimicrobial resistance (AMR). We will discuss the general mechanisms via which each delivery system functions and how it can be tailored to deliver antibiotics against the paradigm of mechanisms underlying antibiotic resistance.
Collapse
Affiliation(s)
- Kusum Kharga
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Shubhang Jha
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Tanvi Vishwakarma
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| |
Collapse
|
9
|
Romanenko MN, Shikov AE, Savina IA, Nizhnikov AA, Antonets KS. Whole-Genome Sequencing of Peribacillus frigoritolerans Strain d21.2 Isolated in the Republic of Dagestan, Russia. Microorganisms 2024; 12:2410. [PMID: 39770615 PMCID: PMC11678259 DOI: 10.3390/microorganisms12122410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Pesticide-free agriculture is a fundamental pillar of environmentally friendly agriculture. To this end, there is an active search for new bacterial strains capable of synthesizing secondary metabolites and toxins that protect crops from pathogens and pests. In this study, we isolated a novel strain d21.2 of Peribacillus frigoritolerans from a soil sample collected in the Republic of Dagestan, Russia. Leveraging several bioinformatic approaches on Illumina-based whole-genome assembly, we revealed that the strain harbors certain insecticidal loci (coding for putative homologs of Bmp and Vpa) and also contains multiple BGCs (biosynthetic gene clusters), including paeninodin, koranimine, schizokinen, and fengycin. In total, 21 BGCs were predicted as synthesizing metabolites with bactericidal and/or fungicidal effects. Importantly, by applying a re-scaffolding pipeline, we managed to robustly predict MGEs (mobile genetic elements) associated with BGCs, implying high genetic plasticity. In addition, the d21.2's genome was free from genes encoding for enteric toxins, implying its safety in use. A comparison with available genomes of the Peribacillus frigoritolerans strain revealed that the strain described here contains more functionally important loci than other members of the species. Therefore, strain d21.2 holds potential for use in agriculture due to the probable manifestation of bactericidal, fungicidal, growth-stimulating, and other useful properties. The assembled genome is available in the NCBI GeneBank under ASM4106054v1.
Collapse
Affiliation(s)
- Maria N. Romanenko
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.N.R.); (A.E.S.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anton E. Shikov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.N.R.); (A.E.S.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Iuliia A. Savina
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.N.R.); (A.E.S.)
| | - Anton A. Nizhnikov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.N.R.); (A.E.S.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Kirill S. Antonets
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.N.R.); (A.E.S.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
10
|
Rodríguez-Robles E, Müller D, Künzl T, Nemat SJ, Edelmann MP, Srivastava P, Louis D, Groaz E, Tiefenbacher K, Roberts TM, Herdewijn P, Marlière P, Panke S. Rational design of a bacterial import system for new-to-nature molecules. Metab Eng 2024; 85:26-34. [PMID: 38802041 DOI: 10.1016/j.ymben.2024.05.005] [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: 01/14/2024] [Revised: 04/27/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Integration of novel compounds into biological processes holds significant potential for modifying or expanding existing cellular functions. However, the cellular uptake of these compounds is often hindered by selectively permeable membranes. We present a novel bacterial transport system that has been rationally designed to address this challenge. Our approach utilizes a highly promiscuous sulfonate membrane transporter, which allows the passage of cargo molecules attached as amides to a sulfobutanoate transport vector molecule into the cytoplasm of the cell. These cargoes can then be unloaded from the sulfobutanoyl amides using an engineered variant of the enzyme γ-glutamyl transferase, which hydrolyzes the amide bond and releases the cargo molecule within the cell. Here, we provide evidence for the broad substrate specificity of both components of the system by evaluating a panel of structurally diverse sulfobutanoyl amides. Furthermore, we successfully implement the synthetic uptake system in vivo and showcase its functionality by importing an impermeant non-canonical amino acid.
Collapse
Affiliation(s)
- Emilio Rodríguez-Robles
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - David Müller
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Tilmann Künzl
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Suren J Nemat
- Department of Chemistry, University of Basel, Basel, Switzerland
| | - Martin Peter Edelmann
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Puneet Srivastava
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Elisabetta Groaz
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Tania Michelle Roberts
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Piet Herdewijn
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Sven Panke
- Bioprocess Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
11
|
Luo VC, Peczuh MW. Location, Location, Location: Establishing Design Principles for New Antibacterials from Ferric Siderophore Transport Systems. Molecules 2024; 29:3889. [PMID: 39202968 PMCID: PMC11357680 DOI: 10.3390/molecules29163889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
This review strives to assemble a set of molecular design principles that enables the delivery of antibiotic warheads to Gram-negative bacterial targets (ESKAPE pathogens) using iron-chelating siderophores, known as the Trojan Horse strategy for antibiotic development. Principles are derived along two main lines. First, archetypical siderophores and their conjugates are used as case studies for native iron transport. They enable the consideration of the correspondence of iron transport and antibacterial target location. The second line of study charts the rationale behind the clinical antibiotic cefiderocol. It illustrates the potential versatility for the design of new Trojan Horse-based antibiotics. Themes such as matching the warhead to a location where the siderophore delivers its cargo (i.e., periplasm vs. cytoplasm), whether or not a cleavable linker is required, and the relevance of cheaters to the effectiveness and selectivity of new conjugates will be explored. The effort to articulate rules has identified gaps in the current understanding of iron transport pathways and suggests directions for new investigations.
Collapse
Affiliation(s)
| | - Mark W. Peczuh
- Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, U3060, Storrs, CT 06269, USA;
| |
Collapse
|
12
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: a review. Front Pharmacol 2024; 15:1444781. [PMID: 39221153 PMCID: PMC11362070 DOI: 10.3389/fphar.2024.1444781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Antibacterial drug resistance poses a significant challenge to modern healthcare systems, threatening our ability to effectively treat bacterial infections. This review aims to provide a comprehensive overview of the types and mechanisms of antibacterial drug resistance. To achieve this aim, a thorough literature search was conducted to identify key studies and reviews on antibacterial resistance mechanisms, strategies and next-generation antimicrobials to contain antimicrobial resistance. In this review, types of resistance and major mechanisms of antibacterial resistance with examples including target site modifications, decreased influx, increased efflux pumps, and enzymatic inactivation of antibacterials has been discussed. Moreover, biofilm formation, and horizontal gene transfer methods has also been included. Furthermore, measures (interventions) taken to control antimicrobial resistance and next-generation antimicrobials have been discussed in detail. Overall, this review provides valuable insights into the diverse mechanisms employed by bacteria to resist the effects of antibacterial drugs, with the aim of informing future research and guiding antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of clinical pharmacy, College of medicine and health sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of environmental health science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
13
|
Motz RN, Kamyabi G, Nolan EM. Experimental methods for evaluating siderophore-antibiotic conjugates. Methods Enzymol 2024; 702:21-50. [PMID: 39155112 DOI: 10.1016/bs.mie.2024.06.004] [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] [Indexed: 08/20/2024]
Abstract
Siderophore-antibiotic conjugates (SACs) are of past and current interest for delivering antibacterials into Gram-negative bacterial pathogens that express siderophore receptors. Studies of SACs are often multifaceted and involve chemical and biological approaches. Major goals are to evaluate the antimicrobial activity and uptake of novel SACs and use the resulting data to inform further mode-of-action studies and molecular design strategies. In this chapter, we describe four key methods that we apply when investigating the antimicrobial activity and uptake of novel SACs based on the siderophore enterobactin (Ent). These methods are based on approaches from the siderophore literature as well as established protocols for antimicrobial activity testing, and include assays for evaluating SAC antimicrobial activity, time-kill kinetics, siderophore competition, and bacterial cell uptake using 57Fe. These assays have served us well in characterizing our Ent-based conjugates and can be applied to study SACs that use other siderophores as targeting vectors.
Collapse
Affiliation(s)
- Rachel N Motz
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ghazal Kamyabi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
14
|
Moreno-Latorre M, de la Torre MC, Cabeza JA, García-Álvarez P, Sierra MA. Attaching Metal-Containing Moieties to β-Lactam Antibiotics: The Case of Penicillin and Cephalosporin. Inorg Chem 2024; 63:12593-12603. [PMID: 38923955 PMCID: PMC11234371 DOI: 10.1021/acs.inorgchem.4c01548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Procedures for the preparation of transition metal complexes having intact bicyclic cepham or penam systems as ligands have been developed. Starting from readily available 4-azido-2-azetidinones, a synthetic approach has been tuned using a copper-catalyzed azide-alkyne cycloaddition between 3-azido-2-azetinones and alkynes, followed by methylation and transmetalation to Au(I) and Ir(III) complexes from the mesoionic carbene Ag(I) complexes. This methodology was applied to 6-azido penam and 7-azido cepham derivatives to build 6-(1,2,3-triazolyl)penam and 7-(1,2,3-triazolyl)cepham proligands, which upon methylation and metalation with Au(I) and Ir(III) complexes yielded products derived from the coordination of the metal to the penam C6 and cepham C7 positions, preserving intact the bicyclic structure of the penicillin and cephalosporin scaffolds. The crystal structure of complex 28b, which has an Ir atom directly bonded to the intact penicillin bicycle, was determined by X-ray diffraction. This is the first structural report of a penicillin-transition-metal complex having the bicyclic system of these antibiotics intact. The selectivity of the coordination processes was interpreted using DFT calculations.
Collapse
Affiliation(s)
- María Moreno-Latorre
- Instituto
de Química Orgánica General, Consejo Superior de Investigaciones Científicas (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
- Centro
de Innovación en Química Avanzada ORFEO-CINQA, https://orfeocinqa.es/
| | - María C. de la Torre
- Instituto
de Química Orgánica General, Consejo Superior de Investigaciones Científicas (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
- Centro
de Innovación en Química Avanzada ORFEO-CINQA, https://orfeocinqa.es/
| | - Javier A. Cabeza
- Departamento
de Química Orgánica e Inorgánica, Facultad de
Química, Universidad de Oviedo, 33071 Oviedo, Spain
- Centro
de Innovación en Química Avanzada ORFEO-CINQA, https://orfeocinqa.es/
| | - Pablo García-Álvarez
- Departamento
de Química Orgánica e Inorgánica, Facultad de
Química, Universidad de Oviedo, 33071 Oviedo, Spain
- Centro
de Innovación en Química Avanzada ORFEO-CINQA, https://orfeocinqa.es/
| | - Miguel A. Sierra
- Departamento
de Química Orgánica I, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain
- Centro
de Innovación en Química Avanzada ORFEO-CINQA, https://orfeocinqa.es/
| |
Collapse
|
15
|
Huang YJ, Yang MH, Lin LY, Liu J, Zang YP, Lin J, Chen WM. Exploring the Localization of Siderophore-Mediated Cargo Delivery in Gram-Negative Bacteria Using 3-Hydroxypyridin-4(1 H)-one-Fluorescein Probes. ACS Infect Dis 2024; 10:2303-2317. [PMID: 38725130 DOI: 10.1021/acsinfecdis.4c00287] [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] [Indexed: 06/15/2024]
Abstract
The design of siderophore-antibiotic conjugates is a promising strategy to overcome drug resistance in negative bacteria. However, accumulating studies have shown that only those antibiotics acting on the cell wall or cell membrane multiply their antibacterial effects when coupled with siderophores, while antibiotics acting on targets in the cytoplasm of bacteria do not show an obvious enhancement of their antibacterial effects when coupled with siderophores. To explore the causes of this phenomenon, we synthesized several conjugate probes using 3-hydroxypyridin-4(1H)-ones as siderophores and replacing the antibiotic cargo with 5-carboxyfluorescein (5-FAM) or malachite green (MG) cargo. By monitoring changes in the fluorescence intensity of FAM conjugate 20 in bacteria, the translocation of the conjugate across the outer membranes of Gram-negative pathogens was confirmed. Further, the use of the fluorogen activating protein(FAP)/MG system revealed that 3-hydroxypyridin-4(1H)-one-MG conjugate 26 was ultimately distributed mainly in the periplasm rather than being translocated into the cytosol of Escherichia coli and Pseudomonas aeruginosa PAO1. Additional mechanistic studies suggested that the uptake of the conjugate involved the siderophore-dependent iron transport pathway and the 3-hydroxypyridin-4(1H)-ones siderophore receptor-dependent mechanism. Meanwhile, we demonstrated that the conjugation of 3-hydroxypyridin-4(1H)-ones to the fluorescein 5-FAM can reduce the possibility of the conjugates crossing the membrane layers of mammalian Vero cells by passive diffusion, and the advantages of the mono-3-hydroxypyridin-4(1H)-ones as a delivery vehicle in the design of conjugates compared to the tri-3-hydroxypyridin-4(1H)-ones. Overall, this work reveals the localization rules of 3-hydroxypyridin-4(1H)-ones as siderophores to deliver the cargo into Gram-negative bacteria. It provides a theoretical basis for the subsequent design of siderophore-antibiotic conjugates, especially based on 3-hydroxypyridin-4(1H)-ones as siderophores.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Ling-Yin Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jun Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 511400, China
| |
Collapse
|
16
|
Liu F, Kou Q, Li H, Cao Y, Chen M, Meng X, Zhang Y, Wang T, Wang H, Zhang D, Yang Y. Discovery of YFJ-36: Design, Synthesis, and Antibacterial Activities of Catechol-Conjugated β-Lactams against Gram-Negative Bacteria. J Med Chem 2024; 67:6705-6725. [PMID: 38596897 DOI: 10.1021/acs.jmedchem.4c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cefiderocol is the first approved catechol-conjugated cephalosporin against multidrug-resistant Gram-negative bacteria, while its application was limited by poor chemical stability associated with the pyrrolidinium linker, moderate potency against Klebsiella pneumoniae and Acinetobacter baumannii, intricate procedures for salt preparation, and potential hypersensitivity. To address these issues, a series of novel catechol-conjugated derivatives were designed, synthesized, and evaluated. Extensive structure-activity relationships and structure-metabolism relationships (SMR) were conducted, leading to the discovery of a promising compound 86b (Code no. YFJ-36) with a new thioether linker. 86b exhibited superior and broad-spectrum in vitro antibacterial activity, especially against A. baumannii and K. pneumoniae, compared with cefiderocol. Potent in vivo efficacy was observed in a murine systemic infection model. Furthermore, the physicochemical stability of 86b in fluid medium at pH 6-8 was enhanced. 86b also reduced potential the risk of allergy owing to the quaternary ammonium linker. The improved properties of 86b supported its further research and development.
Collapse
Affiliation(s)
- Fangjun Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Qunhuan Kou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Hongyuan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Yangzhi Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Meng Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Xin Meng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yinyong Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Ting Wang
- Department of Microbiology, Sichuan Primed Bio-Tech Group Co., Ltd., Chengdu, Sichuan Province 610041, P. R. China
| | - Hui Wang
- China Pharmaceutical University, Jiangsu 211198, China
| | - Dan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
17
|
Kircheva N, Dobrev S, Petkova V, Yocheva L, Angelova S, Dudev T. In Silico Analysis of the Ga 3+/Fe 3+ Competition for Binding the Iron-Scavenging Siderophores of P. aeruginosa-Implementation of Three Gallium-Based Complexes in the "Trojan Horse" Antibacterial Strategy. Biomolecules 2024; 14:487. [PMID: 38672503 PMCID: PMC11048449 DOI: 10.3390/biom14040487] [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: 03/21/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of multidrug-resistant (MDR) microorganisms combined with the ever-draining antibiotic pipeline poses a disturbing and immensely growing public health challenge that requires a multidisciplinary approach and the application of novel therapies aimed at unconventional targets and/or applying innovative drug formulations. Hence, bacterial iron acquisition systems and bacterial Fe2+/3+-containing enzymes have been identified as a plausible target of great potential. The intriguing "Trojan horse" approach deprives microorganisms from the essential iron. Recently, gallium's potential in medicine as an iron mimicry species has attracted vast attention. Different Ga3+ formulations exhibit diverse effects upon entering the cell and thus supposedly have multiple targets. The aim of the current study is to specifically distinguish characteristics of great significance in regard to the initial gallium-based complex, allowing the alien cation to effectively compete with the native ferric ion for binding the siderophores pyochelin and pyoverdine secreted by the bacterium P. aeruginosa. Therefore, three gallium-based formulations were taken into consideration: the first-generation gallium nitrate, Ga(NO3)3, metabolized to Ga3+-hydrated forms, the second-generation gallium maltolate (tris(3-hydroxy-2-methyl-4-pyronato)gallium), and the experimentally proven Ga carrier in the bloodstream-the protein transferrin. We employed a reliable in silico approach based on DFT computations in order to understand the underlying biochemical processes that govern the Ga3+/Fe3+ rivalry for binding the two bacterial siderophores.
Collapse
Affiliation(s)
- Nikoleta Kircheva
- Institute of Optical Materials and Technologies “Acad. J. Malinowski”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.K.); (S.D.); (V.P.); (S.A.)
| | - Stefan Dobrev
- Institute of Optical Materials and Technologies “Acad. J. Malinowski”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.K.); (S.D.); (V.P.); (S.A.)
| | - Vladislava Petkova
- Institute of Optical Materials and Technologies “Acad. J. Malinowski”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.K.); (S.D.); (V.P.); (S.A.)
| | - Lyubima Yocheva
- Faculty of Chemistry and Pharmacy, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| | - Silvia Angelova
- Institute of Optical Materials and Technologies “Acad. J. Malinowski”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.K.); (S.D.); (V.P.); (S.A.)
- University of Chemical Technology and Metallurgy, 1756 Sofia, Bulgaria
| | - Todor Dudev
- Faculty of Chemistry and Pharmacy, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| |
Collapse
|
18
|
Huang YJ, Zang YP, Peng LJ, Yang MH, Lin J, Chen WM. Cajaninstilbene acid derivatives conjugated with siderophores of 3-hydroxypyridin-4(1H)-ones as novel antibacterial agents against Gram-negative bacteria based on the Trojan horse strategy. Eur J Med Chem 2024; 269:116339. [PMID: 38537513 DOI: 10.1016/j.ejmech.2024.116339] [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: 02/03/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/07/2024]
Abstract
The low permeability of the outer membrane of Gram-negative bacteria is a serious obstacle to the development of new antibiotics against them. Conjugation of antibiotic with siderophore based on the "Trojan horse strategy" is a promising strategy to overcome the outer membrane obstacle. In this study, series of antibacterial agents were designed and synthesized by conjugating the 3-hydroxypyridin-4(1H)-one based siderophores with cajaninstilbene acid (CSA) derivative 4 which shows good activity against Gram-positive bacteria by targeting their cell membranes but is ineffective against Gram-negative bacteria. Compared to the inactive parent compound 4, the conjugates 45c or 45d exhibits significant improvement in activity against Gram-negative bacteria, including Escherichia coli, Klebsiella pneumoniae and especially P. aeruginosa (minimum inhibitory concentrations, MICs = 7.8-31.25 μM). The antibacterial activity of the conjugates is attributed to the CSA derivative moiety, and the action mechanism is by disruption of bacterial cell membranes. Further studies on the uptake mechanisms showed that the bacterial siderophore-dependent iron transport system was involved in the uptake of the conjugates. In addition, the conjugates 45c and 45d showed a lower cytotoxic effects in vivo and in vitro and a positive therapeutic effect in the treatment of C. elegans infected by P. aeruginosa. Overall, our work describes a new class and a promising 3-hydroxypyridin-4(1H)-one-CSA derivative conjugates for further development as antibacterial agents against Gram-negative bacteria.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Li-Jun Peng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| |
Collapse
|
19
|
Motz RN, Guo C, Sargun A, Walker GT, Sassone-Corsi M, Raffatellu M, Nolan EM. Conjugation to Native and Nonnative Triscatecholate Siderophores Enhances Delivery and Antibacterial Activity of a β-Lactam to Gram-Negative Bacterial Pathogens. J Am Chem Soc 2024; 146:7708-7722. [PMID: 38457782 PMCID: PMC11037102 DOI: 10.1021/jacs.3c14490] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Developing new antibiotics and delivery strategies is of critical importance for treating infections caused by Gram-negative bacterial pathogens. Hijacking bacterial iron uptake machinery, such as that of the siderophore enterobactin (Ent), represents one promising approach toward these goals. Here, we report a novel Ent-inspired siderophore-antibiotic conjugate (SAC) employing an alternative siderophore moiety as the delivery vector and demonstrate the potency of our SACs harboring the β-lactam antibiotic ampicillin (Amp) against multiple pathogenic Gram-negative bacterial strains. We establish the ability of N,N',N''-(nitrilotris(ethane-2,1-diyl))tris(2,3-dihydroxybenzamide) (TRENCAM, hereafter TC), a synthetic mimic of Ent, to facilitate drug delivery across the outer membrane (OM) of Gram-negative pathogens. Conjugation of Amp to a new monofunctionalized TC scaffold affords TC-Amp, which displays markedly enhanced antibacterial activity against the gastrointestinal pathogen Salmonella enterica serovar Typhimurium (STm) compared with unmodified Amp. Bacterial uptake, antibiotic susceptibility, and microscopy studies with STm show that the TC moiety facilitates TC-Amp uptake by the OM receptors FepA and IroN and that the Amp warhead inhibits penicillin-binding proteins. Moreover, TC-Amp achieves targeted activity, selectively killing STm in the presence of a commensal lactobacillus. Remarkably, we uncover that TC-Amp and its Ent-based predecessor Ent-Amp achieve enhanced antibacterial activity against diverse Gram-negative ESKAPE pathogens that express Ent uptake machinery, including strains that possess intrinsic β-lactam resistance. TC-Amp and Ent-Amp exhibit potency comparable to that of the FDA-approved SAC cefiderocol against Gram-negative pathogens. These results demonstrate the effective application of native and appropriately designed nonnative siderophores as vectors for drug delivery across the OM of multiple Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Rachel N. Motz
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Artur Sargun
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gregory T. Walker
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Martina Sassone-Corsi
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, CA 92697, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, CA 92697, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines, La Jolla, CA 92093, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Bo L, Sun H, Li YD, Zhu J, Wurpel JND, Lin H, Chen ZS. Combating antimicrobial resistance: the silent war. Front Pharmacol 2024; 15:1347750. [PMID: 38420197 PMCID: PMC10899355 DOI: 10.3389/fphar.2024.1347750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Once hailed as miraculous solutions, antibiotics no longer hold that status. The excessive use of antibiotics across human healthcare, agriculture, and animal husbandry has given rise to a broad array of multidrug-resistant (MDR) pathogens, posing formidable treatment challenges. Antimicrobial resistance (AMR) has evolved into a pressing global health crisis, linked to elevated mortality rates in the modern medical era. Additionally, the absence of effective antibiotics introduces substantial risks to medical and surgical procedures. The dwindling interest of pharmaceutical industries in developing new antibiotics against MDR pathogens has aggravated the scarcity issue, resulting in an exceedingly limited pipeline of new antibiotics. Given these circumstances, the imperative to devise novel strategies to combat perilous MDR pathogens has become paramount. Contemporary research has unveiled several promising avenues for addressing this challenge. The article provides a comprehensive overview of these innovative therapeutic approaches, highlighting their mechanisms of action, benefits, and drawbacks.
Collapse
Affiliation(s)
- Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Haidong Sun
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jonathan Zhu
- Carle Place Middle and High School, Carle Place, NY, United States
| | - John N. D. Wurpel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Hanli Lin
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
| |
Collapse
|
21
|
Batey SFD, Davie MJ, Hems ES, Liston JD, Scott TA, Alt S, Francklyn CS, Wilkinson B. The catechol moiety of obafluorin is essential for antibacterial activity. RSC Chem Biol 2023; 4:926-941. [PMID: 37920400 PMCID: PMC10619133 DOI: 10.1039/d3cb00127j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/11/2023] [Indexed: 11/04/2023] Open
Abstract
Obafluorin is a Pseudomonas fluorescens antibacterial natural product that inhibits threonyl-tRNA synthetase (ThrRS). It acts as a broad-spectrum antibiotic against a range of clinically relevant pathogens and comprises a strained β-lactone ring decorated with catechol and 4-nitro-benzyl moieties. The catechol moiety is widespread in nature and its role in the coordination of ferric iron has been well-characterised in siderophores and Trojan horse antibiotics. Here we use a combination of mutasynthesis, bioassays, enzyme assays and metal binding studies to delineate the role of the catechol moiety in the bioactivity of obafluorin. We use P. fluorescens biosynthetic mutants to generate obafluorin analogues with modified catechol moieties. We demonstrate that an intact catechol is required for both antibacterial activity and inhibition of the ThrRS molecular target. Although recent work showed that the obafluorin catechol coordinates Zn2+ in the ThrRS active site, we find that obafluorin is a weak Zn2+ binder in vitro, contrasting with a strong, specific 1 : 1 interaction with Fe3+. We use bioassays with siderophore transporter mutants to probe the role of the obafluorin catechol in Fe3+-mediated uptake. Surprisingly, obafluorin does not behave as a Trojan horse antibiotic but instead exhibits increased antibacterial activity in the presence of Fe3+. We further demonstrate that Fe3+ binding prevents the hydrolytic breakdown of the β-lactone ring, revealing a hitherto unreported function for the catechol moiety in natural product bioactivity.
Collapse
Affiliation(s)
- Sibyl F D Batey
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Melissa J Davie
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Edward S Hems
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Jonathon D Liston
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Thomas A Scott
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Silke Alt
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| | - Christopher S Francklyn
- Department of Biochemistry, College of Medicine, University of Vermont, Burlington Vermont 05405 USA
| | - Barrie Wilkinson
- Department of Molecular Microbiology, John Innes Centre Norwich Research Park Norwich NR4 7UH UK
| |
Collapse
|
22
|
Krajnc A, Gobec S. Conjugates of monocyclic β-lactams and siderophore mimetics: a patent evaluation (WO2023023393). Expert Opin Ther Pat 2023; 33:471-476. [PMID: 37902072 DOI: 10.1080/13543776.2023.2262135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023]
Abstract
INTRODUCTION β-Lactams, which include monobactams, remain the most important class of antibiotics worldwide. Aztreonam, the only monobactam in clinical use, has remarkable activity against many Gram-negative bacteria, but limited activity against some of the most problematic multidrug-resistant (MDR) pathogens, such as MDR Pseudomonas aeruginosa and Acinetobacter baumannii co-expressing extended-spectrum- and metallo-β-lactamases, which can inactivate aztreonam by hydrolysis. AREAS COVERED Structurally novel siderophore-conjugated aztreonam derivatives with improved antibacterial properties against several high-priority pathogens are claimed. This invention reports that sidechain extension of aztreonam is tolerated; the coupling of its aminothiazoloxime carboxylic acid part with a siderophore mimetic significantly improved the antibacterial activity against several problematic strains, including MDR A. baumannii isolates with carbapenemase/cephalosporinase activity. EXPERT OPINION Finding new strategies to tackle bacterial resistance to β-lactam antibiotics is critical. Considering that β lactams are validated and safe drugs, this research may stimulate the field to develop new ideas in the arena of antimicrobial drug discovery, particularly with respect to siderophore mimetics.
Collapse
Affiliation(s)
- Alen Krajnc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
23
|
Domingues S, Lima T, Saavedra MJ, Da Silva GJ. An Overview of Cefiderocol's Therapeutic Potential and Underlying Resistance Mechanisms. Life (Basel) 2023; 13:1427. [PMID: 37511802 PMCID: PMC10382032 DOI: 10.3390/life13071427] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Antimicrobial resistance continues to increase globally and treatment of difficult-to-treat (DTT) infections, mostly associated with carbapenem-resistant (CR) Pseudomonas aeruginosa, CR Acinetobacter baumannii, and CR- and third-generation-cephalosporins-resistant Enterobacterales remains a challenge for the clinician. The recent approval of cefiderocol has broaden the armamentarium for the treatment of patients with DTT infections. Cefiderocol is a siderophore cephalosporin that has shown excellent antibacterial activity, in part due to its innovative way of cell permeation. It is relatively stable compared to most commonly found carbapenamases. However, some resistant mechanisms to cefiderocol have already been identified and reduced susceptibility has developed during patient treatment, highlighting that the clinical use of cefiderocol must be rational. In this review, we summarize the current available treatments against the former resistant bacteria, and we revise and discuss the mechanism of action of cefiderocol, underlying the biological function of siderophores, the therapeutic potential of cefiderocol, and the mechanisms of resistance reported so far.
Collapse
Affiliation(s)
- Sara Domingues
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Tiago Lima
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maria José Saavedra
- CITAB-Inov4Agro, Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- CECAV-AL4AnimalS, Animal and Veterinary Research Center, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Gabriela Jorge Da Silva
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
24
|
Rayner B, Verderosa AD, Ferro V, Blaskovich MAT. Siderophore conjugates to combat antibiotic-resistant bacteria. RSC Med Chem 2023; 14:800-822. [PMID: 37252105 PMCID: PMC10211321 DOI: 10.1039/d2md00465h] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/21/2023] [Indexed: 10/31/2023] Open
Abstract
Antimicrobial resistance (AMR) is a global threat to society due to the increasing emergence of multi-drug resistant bacteria that are not susceptible to our last line of defence antibiotics. Exacerbating this issue is a severe gap in antibiotic development, with no new clinically relevant classes of antibiotics developed in the last two decades. The combination of the rapidly increasing emergence of resistance and scarcity of new antibiotics in the clinical pipeline means there is an urgent need for new efficacious treatment strategies. One promising solution, known as the 'Trojan horse' approach, hijacks the iron transport system of bacteria to deliver antibiotics directly into cells - effectively tricking bacteria into killing themselves. This transport system uses natively produced siderophores, which are small molecules with a high affinity for iron. By linking antibiotics to siderophores, to make siderophore antibiotic conjugates, the activity of existing antibiotics can potentially be reinvigorated. The success of this strategy was recently exemplified with the clinical release of cefiderocol, a cephalosporin-siderophore conjugate with potent antibacterial activity against carbapenem-resistant and multi-drug resistant Gram-negative bacilli. This review discusses the recent advancements in siderophore antibiotic conjugates and the challenges associated with the design of these compounds that need to be overcome to deliver more efficacious therapeutics. Potential strategies have also been suggested for new generations of siderophore-antibiotics with enhanced activity.
Collapse
Affiliation(s)
- Beth Rayner
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
| | - Anthony D Verderosa
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
| | - Vito Ferro
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, University of Queensland Brisbane Queensland Australia
- Australian Infectious Disease Research Centre, The University of Queensland Brisbane Queensland Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland Australia
| |
Collapse
|
25
|
Fernandes GFS, Scarim CB, Kim SH, Wu J, Castagnolo D. Oxazolidinones as versatile scaffolds in medicinal chemistry. RSC Med Chem 2023; 14:823-847. [PMID: 37252095 PMCID: PMC10211318 DOI: 10.1039/d2md00415a] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/06/2023] [Indexed: 11/19/2023] Open
Abstract
Oxazolidinone is a five-member heterocyclic ring with several biological applications in medicinal chemistry. Among the three possible isomers, 2-oxazolidinone is the most investigated in drug discovery. Linezolid was pioneered as the first approved drug containing an oxazolidinone ring as the pharmacophore group. Numerous analogues have been developed since its arrival on the market in 2000. Some have succeeded in reaching the advanced stages of clinical studies. However, most oxazolidinone derivatives reported in recent decades have not reached the initial stages of drug development, despite their promising pharmacological applications in a variety of therapeutic areas, including antibacterial, antituberculosis, anticancer, anti-inflammatory, neurologic, and metabolic diseases, among other areas. Therefore, this review article aims to compile the efforts of medicinal chemists who have explored this scaffold over the past decades and highlight the potential of the class for medicinal chemistry.
Collapse
Affiliation(s)
| | - Cauê Benito Scarim
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University Araraquara 14800903 Brazil
| | - Seong-Heun Kim
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
- School of Cancer and Pharmaceutical Sciences, King's College London 150 Stamford Street SE1 9NH London UK
| | - Jingyue Wu
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Daniele Castagnolo
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| |
Collapse
|
26
|
Huang YJ, Zhong XL, Zang YP, Yang MH, Lin J, Chen WM. 3-Hydroxy-pyridin-4(1H)-ones as siderophores mediated delivery of isobavachalcone enhances antibacterial activity against pathogenic Pseudomonas aeruginosa. Eur J Med Chem 2023; 257:115454. [PMID: 37210837 DOI: 10.1016/j.ejmech.2023.115454] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
The natural prenylated chalcone isobavachalcone (IBC) shows good antibacterial activity against Gram-positive bacteria but is ineffective against Gram-negative bacteria, most likely due to the outer membrane barrier of Gram-negative bacteria. The Trojan horse strategy has been shown to be an effective strategy to overcome the reduction in the permeability of the outer membrane of Gram-negative bacteria. In this study, eight different 3-hydroxy-pyridin-4(1H)-one-isobavachalcone conjugates were designed and synthesized based on the siderophore Trojan horse strategy. The conjugates exhibited 8- to 32-fold lower minimum inhibitory concentrations (MICs) and 32- to 177-fold lower half-inhibitory concentrations (IC50s) against Pseudomonas aeruginosa PAO1 as well as clinical multidrug-resistant (MDR) strains compared to the parent IBC under iron limitation. Further studies showed that the antibacterial activity of the conjugates was regulated by the bacterial iron uptake pathway under different iron concentration conditions. Studies on the antibacterial mechanism of conjugate 1b showed that it exerts antibacterial activity by disrupting cytoplasmic membrane integrity and inhibiting cell metabolism. Finally, conjugate 1b showed a lower cytotoxic effects on Vero cells than IBC and a positive therapeutic effect in the treatment of bacterial infections caused by Gram-negative bacteria PAO1. Overall, this work demonstrates that IBC can be delivered to Gram-negative bacteria when combined with 3-hydroxy-pyridin-4(1H)-ones as siderophores and provides a scientific basis for the development of effective antibacterial agents against Gram-negative bacteria.
Collapse
Affiliation(s)
- Yong-Jun Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Xiao-Lin Zhong
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Yi-Peng Zang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Ming-Han Yang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China
| | - Jing Lin
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| | - Wei-Min Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 511400, China.
| |
Collapse
|
27
|
Evenson GE, Powell WC, Hinds AB, Walczak MA. Catalytic Amide Activation with Thermally Stable Molybdenum(VI) Dioxide Complexes. J Org Chem 2023; 88:6192-6202. [PMID: 37027833 PMCID: PMC10422866 DOI: 10.1021/acs.joc.3c00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Oxazolines and thiazolines are important constituents of bioactive natural products and pharmaceuticals. Here, we report the development of an effective and practical method of oxazoline and thiazoline formation, which can facilitate the synthesis of natural products, chiral ligands, and pharmaceutical intermediates. This method capitalized on a Mo(VI) dioxide catalyst stabilized by substituted picolinic acid ligands, which is tolerant to many functional groups that would otherwise be sensitive to highly electrophilic alternative reagents.
Collapse
Affiliation(s)
- Garrett E Evenson
- University of Colorado, Department of Chemistry, Boulder, Colorado 80309, United States
| | - Wyatt C Powell
- University of Colorado, Department of Chemistry, Boulder, Colorado 80309, United States
| | - Aaron B Hinds
- University of Colorado, Department of Chemistry, Boulder, Colorado 80309, United States
| | - Maciej A Walczak
- University of Colorado, Department of Chemistry, Boulder, Colorado 80309, United States
| |
Collapse
|
28
|
Cui F, Fan R, Wang D, Li J, Li T. Research progress on iron uptake pathways and mechanisms of foodborne microorganisms and their application in the food sector. Crit Rev Food Sci Nutr 2023; 64:8892-8910. [PMID: 37099732 DOI: 10.1080/10408398.2023.2204491] [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] [Indexed: 04/28/2023]
Abstract
Iron is one of the essential nutrients for almost all microorganisms. Under iron-limited conditions, bacteria can secrete siderophores to the outside world to absorb iron for survival. This process requires the coordinated action of energy-transducing proteins, transporters, and receptors. The spoilage factors of some spoilage bacteria and the pathogenic mechanism of pathogenic bacteria are also closely related to siderophores. Meanwhile, some siderophores have also gradually evolved toward beneficial aspects. First, a variety of siderophores are classified into three aspects. In addition, representative iron uptake systems of Gram-negative and Gram-positive bacteria are described in detail to understand the common and specific pathways of iron uptake by various bacteria. In particular, the causes of siderophore-induced bacterial pathogenicity and the methods and mechanisms of inhibiting bacterial iron absorption under the involvement of siderophores are presented. Then, the application of siderophores in the food sector is mainly discussed, such as improving the food quality of dairy products and meat, inhibiting the attack of pathogenic bacteria on food, improving the plant growth environment, and promoting plant growth. Finally, this review highlights the unresolved fate of siderophores in the iron uptake system and emphasizes further development of siderophore-based substitutes for traditional drugs, new antibiotic-resistance drugs, and vaccines in the food and health sectors.
Collapse
Affiliation(s)
- Fangchao Cui
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Rongsen Fan
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Dangfeng Wang
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
- College of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianrong Li
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, Jinzhou, China
| | - Tingting Li
- Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, Dalian, China
| |
Collapse
|
29
|
Southwell JW, Herman R, Raines DJ, Clarke JE, Böswald I, Dreher T, Gutenthaler SM, Schubert N, Seefeldt J, Metzler‐Nolte N, Thomas GH, Wilson KS, Duhme‐Klair A. Siderophore-Linked Ruthenium Catalysts for Targeted Allyl Ester Prodrug Activation within Bacterial Cells. Chemistry 2023; 29:e202202536. [PMID: 36355416 PMCID: PMC10108276 DOI: 10.1002/chem.202202536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/12/2022]
Abstract
Due to rising resistance, new antibacterial strategies are needed, including methods for targeted antibiotic release. As targeting vectors, chelating molecules called siderophores that are released by bacteria to acquire iron have been investigated for conjugation to antibacterials, leading to the clinically approved drug cefiderocol. The use of small-molecule catalysts for prodrug activation within cells has shown promise in recent years, and here we investigate siderophore-linked ruthenium catalysts for the activation of antibacterial prodrugs within cells. Moxifloxacin-based prodrugs were synthesised, and their catalyst-mediated activation was demonstrated under anaerobic, biologically relevant conditions. In the absence of catalyst, decreased antibacterial activities were observed compared to moxifloxacin versus Escherichia coli K12 (BW25113). A series of siderophore-linked ruthenium catalysts were investigated for prodrug activation, all of which displayed a combinative antibacterial effect with the prodrug, whereas a representative example displayed little toxicity against mammalian cell lines. By employing complementary bacterial growth assays, conjugates containing siderophore units based on catechol and azotochelin were found to be most promising for intracellular prodrug activation.
Collapse
Affiliation(s)
| | - Reyme Herman
- University of YorkDepartment of BiologyHeslingtonWentworth WayYO10 5DDUK
| | - Daniel J. Raines
- University of YorkDepartment of ChemistryHeslingtonYorkYO10 5DDUK
| | - Justin E. Clarke
- University of YorkYork Structural Biology LaboratoryHeslingtonYO10 5DDUK
| | - Isabelle Böswald
- University of YorkDepartment of ChemistryHeslingtonYorkYO10 5DDUK
| | - Thorsten Dreher
- University of YorkDepartment of ChemistryHeslingtonYorkYO10 5DDUK
| | | | - Nicole Schubert
- Anorganische ChemieRuhr-Universität BochumUniversitätsstraße 15044801BochumGermany
| | - Jana Seefeldt
- Anorganische ChemieRuhr-Universität BochumUniversitätsstraße 15044801BochumGermany
| | - Nils Metzler‐Nolte
- Anorganische ChemieRuhr-Universität BochumUniversitätsstraße 15044801BochumGermany
| | - Gavin H. Thomas
- University of YorkDepartment of BiologyHeslingtonWentworth WayYO10 5DDUK
| | - Keith S. Wilson
- University of YorkYork Structural Biology LaboratoryHeslingtonYO10 5DDUK
| | | |
Collapse
|
30
|
Tabcheh J, Vergalli J, Davin-Régli A, Ghanem N, Pages JM, Al-Bayssari C, Brunel JM. Rejuvenating the Activity of Usual Antibiotics on Resistant Gram-Negative Bacteria: Recent Issues and Perspectives. Int J Mol Sci 2023; 24:1515. [PMID: 36675027 PMCID: PMC9864949 DOI: 10.3390/ijms24021515] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Antibiotic resistance continues to evolve and spread beyond all boundaries, resulting in an increase in morbidity and mortality for non-curable infectious diseases. Due to the failure of conventional antimicrobial therapy and the lack of introduction of a novel class of antibiotics, novel strategies have recently emerged to combat these multidrug-resistant infectious microorganisms. In this review, we highlight the development of effective antibiotic combinations and of antibiotics with non-antibiotic activity-enhancing compounds to address the widespread emergence of antibiotic-resistant strains.
Collapse
Affiliation(s)
- Jinane Tabcheh
- Aix Marseille University, INSERM, SSA, MCT, 13385 Marseille, France
- Faculty of Science 3, Lebanese University, Michel Slayman Tripoli Campus, Tripoli 1352, Lebanon
| | - Julia Vergalli
- Aix Marseille University, INSERM, SSA, MCT, 13385 Marseille, France
| | - Anne Davin-Régli
- Aix Marseille University, INSERM, SSA, MCT, 13385 Marseille, France
| | - Noha Ghanem
- Faculty of Science 3, Lebanese University, Michel Slayman Tripoli Campus, Tripoli 1352, Lebanon
| | - Jean-Marie Pages
- Aix Marseille University, INSERM, SSA, MCT, 13385 Marseille, France
| | - Charbel Al-Bayssari
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut P.O. Box 55251, Lebanon
| | | |
Collapse
|
31
|
Synthesis and study of new siderophore analog-ciprofloxacin conjugates with antibiotic activities against Pseudomonas aeruginosa and Burkholderia spp. Eur J Med Chem 2022; 245:114921. [DOI: 10.1016/j.ejmech.2022.114921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
|
32
|
Cole MS, Hegde PV, Aldrich CC. β-Lactamase-Mediated Fragmentation: Historical Perspectives and Recent Advances in Diagnostics, Imaging, and Antibacterial Design. ACS Infect Dis 2022; 8:1992-2018. [PMID: 36048623 DOI: 10.1021/acsinfecdis.2c00315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The discovery of β-lactam (BL) antibiotics in the early 20th century represented a remarkable advancement in human medicine, allowing for the widespread treatment of infectious diseases that had plagued humanity throughout history. Yet, this triumph was followed closely by the emergence of β-lactamase (BLase), a bacterial weapon to destroy BLs. BLase production is a primary mechanism of resistance to BL antibiotics, and the spread of new homologues with expanded hydrolytic activity represents a pressing threat to global health. Nonetheless, researchers have developed strategies that take advantage of this defense mechanism, exploiting BLase activity in the creation of probes, diagnostic tools, and even novel antibiotics selective for resistant organisms. Early discoveries in the 1960s and 1970s demonstrating that certain BLs expel a leaving group upon BLase cleavage have spawned an entire field dedicated to employing this selective release mechanism, termed BLase-mediated fragmentation. Chemical probes have been developed for imaging and studying BLase-expressing organisms in the laboratory and diagnosing BL-resistant infections in the clinic. Perhaps most promising, new antibiotics have been developed that use BLase-mediated fragmentation to selectively release cytotoxic chemical "warheads" at the site of infection, reducing off-target effects and allowing for the repurposing of putative antibiotics against resistant organisms. This Review will provide some historical background to the emergence of this field and highlight some exciting recent reports that demonstrate the promise of this unique release mechanism.
Collapse
Affiliation(s)
- Malcolm S Cole
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard St SE, Minneapolis, Minnesota 55455, United States
| | - Pooja V Hegde
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard St SE, Minneapolis, Minnesota 55455, United States
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard St SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
33
|
Moynié L, Hoegy F, Milenkovic S, Munier M, Paulen A, Gasser V, Faucon AL, Zill N, Naismith JH, Ceccarelli M, Schalk IJ, Mislin GLA. Hijacking of the Enterobactin Pathway by a Synthetic Catechol Vector Designed for Oxazolidinone Antibiotic Delivery in Pseudomonas aeruginosa. ACS Infect Dis 2022; 8:1894-1904. [PMID: 35881068 DOI: 10.1021/acsinfecdis.2c00202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Enterobactin (ENT) is a tris-catechol siderophore used to acquire iron by multiple bacterial species. These ENT-dependent iron uptake systems have often been considered as potential gates in the bacterial envelope through which one can shuttle antibiotics (Trojan horse strategy). In practice, siderophore analogues containing catechol moieties have shown promise as vectors to which antibiotics may be attached. Bis- and tris-catechol vectors (BCVs and TCVs, respectively) were shown using structural biology and molecular modeling to mimic ENT binding to the outer membrane transporter PfeA in Pseudomonas aeruginosa. TCV but not BCV appears to cross the outer membrane via PfeA when linked to an antibiotic (linezolid). TCV is therefore a promising vector for Trojan horse strategies against P. aeruginosa, confirming the ENT-dependent iron uptake system as a gate to transport antibiotics into P. aeruginosa cells.
Collapse
Affiliation(s)
- Lucile Moynié
- The Rosalind Franklin Institute, Harwell Campus, Oxfordshire OX11 0QS, U.K
| | - Françoise Hoegy
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Stefan Milenkovic
- Department of Physics, University of Cagliari, 09042 Monserrato, Italy
| | - Mathilde Munier
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Aurélie Paulen
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Véronique Gasser
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Aline L Faucon
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Nicolas Zill
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - James H Naismith
- The Rosalind Franklin Institute, Harwell Campus, Oxfordshire OX11 0QS, U.K.,Division of Structural Biology, Wellcome Trust Centre of Human Genomics, 7 Roosevelt Drive, Oxford OX3 7BN, U.K
| | - Matteo Ceccarelli
- Department of Physics, University of Cagliari, 09042 Monserrato, Italy.,IOM/CNR, Sezione di Cagliari, University of Cagliari, 09042 Monserrato, Italy
| | - Isabelle J Schalk
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| | - Gaëtan L A Mislin
- CNRS, UMR7242 Biotechnologie et Signalisation Cellulaire, 300 Boulevard Sébastien Brant, F-67412 Illkirch, France.,Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg (IREBS), 300 Boulevard Sébastien Brant, F-67412 Illkirch, France
| |
Collapse
|
34
|
Wesseling CJ, Martin NI. Synergy by Perturbing the Gram-Negative Outer Membrane: Opening the Door for Gram-Positive Specific Antibiotics. ACS Infect Dis 2022; 8:1731-1757. [PMID: 35946799 PMCID: PMC9469101 DOI: 10.1021/acsinfecdis.2c00193] [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] [Indexed: 01/29/2023]
Abstract
New approaches to target antibacterial agents toward Gram-negative bacteria are key, given the rise of antibiotic resistance. Since the discovery of polymyxin B nonapeptide as a potent Gram-negative outer membrane (OM)-permeabilizing synergist in the early 1980s, a vast amount of literature on such synergists has been published. This Review addresses a range of peptide-based and small organic compounds that disrupt the OM to elicit a synergistic effect with antibiotics that are otherwise inactive toward Gram-negative bacteria, with synergy defined as a fractional inhibitory concentration index (FICI) of <0.5. Another requirement for the inclusion of the synergists here covered is their potentiation of a specific set of clinically used antibiotics: erythromycin, rifampicin, novobiocin, or vancomycin. In addition, we have focused on those synergists with reported activity against Gram-negative members of the ESKAPE family of pathogens namely, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and/or Acinetobacter baumannii. In cases where the FICI values were not directly reported in the primary literature but could be calculated from the published data, we have done so, allowing for more direct comparison of potency with other synergists. We also address the hemolytic activity of the various OM-disrupting synergists reported in the literature, an effect that is often downplayed but is of key importance in assessing the selectivity of such compounds for Gram-negative bacteria.
Collapse
|
35
|
Hausmann S, Geiser J, Valentini M. Mechanism of inhibition of bacterial RNA helicases by diazo dyes and implications for antimicrobial drug development. Biochem Pharmacol 2022; 204:115194. [DOI: 10.1016/j.bcp.2022.115194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
|
36
|
Mangal S, Dua T, Chauhan M, Dhingra N, Chhibber S, Singh V, Harjai K. Design, Synthesis, and Quorum Quenching Potential of Novel Catechol–Zingerone Conjugate to Find an Elixir to Tackle Pseudomonas aeruginosa Through the Trojan Horse Strategy. Front Chem 2022; 10:902719. [PMID: 35783213 PMCID: PMC9240400 DOI: 10.3389/fchem.2022.902719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 11/27/2022] Open
Abstract
To address the issue of multidrug resistance in Pseudomonas aeruginosa, a novel catechol–zingerone conjugate (1) linked via a non-hydrolyzable 1,2,3-triazole linker was synthesized and subjected to biological evaluation based on the Trojan horse strategy. To enhance the efficacy, catechol, a xenosiderophore, utilized by P. aeruginosa for iron assimilation, and the dietary phytochemical zingerone, known for its anti-virulent activity against Pseudomonas aeruginosa, were exploited in the present study. Theoretical validation of conjugate (1) was conducted by in silico molecular docking analysis to determine the interaction with outer membrane transport receptor PirA and quorum sensing signal receptors. In addition, nine-fold binding affinity of Conjugate (1) toward PirA (5FP2) in comparison to its natural ligand catechol with D-score −1.13 Å authenticated the designed Trojan horse drug. Conjugate (1) showed stronger anti-virulent activity than zingerone; hence, it exhibited a promising anti-biofilm efficacy as assessed by crystal violet assay and visualized by FESEM toward P. aeruginosa. Encouraging results against P. aeruginosa in terms of quorum sensing regulated virulence factors, motility phenotypes, and biofilm formation with no cell cytotoxicity and could help open hitherto unexplored possibilities of establishing Trojan horse drugs as a successful approach against multidrug resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Surabhi Mangal
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Tamanna Dua
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| | - Monika Chauhan
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Vasundhara Singh
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
- *Correspondence: Vasundhara Singh, ; Kusum Harjai,
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
- *Correspondence: Vasundhara Singh, ; Kusum Harjai,
| |
Collapse
|
37
|
Prasad NK, Seiple IB, Cirz RT, Rosenberg OS. Leaks in the Pipeline: a Failure Analysis of Gram-Negative Antibiotic Development from 2010 to 2020. Antimicrob Agents Chemother 2022; 66:e0005422. [PMID: 35471042 PMCID: PMC9112940 DOI: 10.1128/aac.00054-22] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The World Health Organization (WHO) has warned that our current arsenal of antibiotics is not innovative enough to face impending infectious diseases, especially those caused by multidrug-resistant Gram-negative pathogens. Although the current preclinical pipeline is well stocked with novel candidates, the last U.S. Food and Drug Administration (FDA)-approved antibiotic with a novel mechanism of action against Gram-negative bacteria was discovered nearly 60 years ago. Of all the antibiotic candidates that initiated investigational new drug (IND) applications in the 2000s, 17% earned FDA approval within 12 years, while an overwhelming 62% were discontinued in that time frame. These "leaks" in the clinical pipeline, where compounds with clinical potential are abandoned during clinical development, indicate that scientific innovations are not reaching the clinic and providing benefits to patients. This is true for not only novel candidates but also candidates from existing antibiotic classes with clinically validated targets. By identifying the sources of the leaks in the clinical pipeline, future developmental efforts can be directed toward strategies that are more likely to flow into clinical use. In this review, we conduct a detailed failure analysis of clinical candidates with Gram-negative activity that have fallen out of the clinical pipeline over the past decade. Although limited by incomplete data disclosure from companies engaging in antibiotic development, we attempt to distill the developmental challenges faced by each discontinued candidate. It is our hope that this insight can help de-risk antibiotic development and bring new, effective antibiotics to the clinic.
Collapse
Affiliation(s)
- Neha K. Prasad
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Ian B. Seiple
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | | | - Oren S. Rosenberg
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Biochemistry, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
38
|
Klahn P, Zscherp R, Jimidar CC. Advances in the Synthesis of Enterobactin, Artificial Analogues, and Enterobactin-Derived Antimicrobial Drug Conjugates and Imaging Tools for Infection Diagnosis. SYNTHESIS-STUTTGART 2022. [DOI: 10.1055/a-1783-0751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AbstractIron is an essential growth factor for bacteria, but although highly abundant in nature, its bioavailability during infection in the human host or the environment is limited. Therefore, bacteria produce and secrete siderophores to ensure their supply of iron. The triscatecholate siderophore enterobactin and its glycosylated derivatives, the salmochelins, play a crucial role for iron acquisition in several bacteria. As these compounds can serve as carrier molecules for the design of antimicrobial siderophore drug conjugates as well as siderophore-derived tool compounds for the detection of infections with bacteria, their synthesis and the design of artificial analogues is of interest. In this review, we give an overview on the synthesis of enterobactin, biomimetic as well as totally artificial analogues, and related drug-conjugates covering up to 12/2021.1 Introduction2 Antibiotic Crisis and Sideromycins as Natural Templates for New Antimicrobial Drugs3 Biosynthesis of Enterobactin, Salmochelins, and Microcins4 Total Synthesis of Enterobactin and Salmochelins5 Chemoenzymatic Semi-synthesis of Salmochelins and Microcin E492m Derivatives6 Synthesis of Biomimetic Enterobactin Derivatives with Natural Tris-lactone Backbone7 Synthesis of Artificial Enterobactin Derivatives without Tris-lactone Backbone8 Conclusions
Collapse
Affiliation(s)
- Philipp Klahn
- Institute of Organic Chemistry, Technische Universität Braunschweig
- Department for Chemistry and Molecular Biology, University of Gothenburg
| | - Robert Zscherp
- Institute of Organic Chemistry, Technische Universität Braunschweig
| | | |
Collapse
|
39
|
Murugaiyan J, Kumar PA, Rao GS, Iskandar K, Hawser S, Hays JP, Mohsen Y, Adukkadukkam S, Awuah WA, Jose RAM, Sylvia N, Nansubuga EP, Tilocca B, Roncada P, Roson-Calero N, Moreno-Morales J, Amin R, Kumar BK, Kumar A, Toufik AR, Zaw TN, Akinwotu OO, Satyaseela MP, van Dongen MBM. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics (Basel) 2022; 11:200. [PMID: 35203804 PMCID: PMC8868457 DOI: 10.3390/antibiotics11020200] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic resistance, and, in a broader perspective, antimicrobial resistance (AMR), continues to evolve and spread beyond all boundaries. As a result, infectious diseases have become more challenging or even impossible to treat, leading to an increase in morbidity and mortality. Despite the failure of conventional, traditional antimicrobial therapy, in the past two decades, no novel class of antibiotics has been introduced. Consequently, several novel alternative strategies to combat these (multi-) drug-resistant infectious microorganisms have been identified. The purpose of this review is to gather and consider the strategies that are being applied or proposed as potential alternatives to traditional antibiotics. These strategies include combination therapy, techniques that target the enzymes or proteins responsible for antimicrobial resistance, resistant bacteria, drug delivery systems, physicochemical methods, and unconventional techniques, including the CRISPR-Cas system. These alternative strategies may have the potential to change the treatment of multi-drug-resistant pathogens in human clinical settings.
Collapse
Affiliation(s)
- Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - P. Anand Kumar
- Department of Veterinary Microbiology, NTR College of Veterinary Science, Sri Venkateswara Veterinary University, Gannavaram 521102, India;
| | - G. Srinivasa Rao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517502, India;
| | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France;
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | | | - John P. Hays
- Department of Medical Microbiology, Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Yara Mohsen
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria 21544, Egypt;
- Infectious Disease Clinical Pharmacist, Antimicrobial Stewardship Department, International Medical Center Hospital, Cairo 11511, Egypt
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - Wireko Andrew Awuah
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Ruiz Alvarez Maria Jose
- Research Coordination and Support Service, National Institute of Health (ISS) Viale Regina -Elena, 299, 00161 Rome, Italy;
| | - Nanono Sylvia
- Infectious Diseases Institute (IDI), College of Health Sciences, Makerere University, Kampala 7072, Uganda;
| | | | - Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Natalia Roson-Calero
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Javier Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Rohul Amin
- James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh;
| | - Ballamoole Krishna Kumar
- Nitte (Deemed to be University), Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangalore 575018, India;
| | - Abishek Kumar
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Abdul-Rahman Toufik
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Thaint Nadi Zaw
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Oluwatosin O. Akinwotu
- Department of Microbiology and Biotechnology Centre, Maharaja Sayajirao University of Baroda, Vadodara 390002, India;
- Environmental and Biotechnology Unit, Department of Microbiology, University of Ibadan, 200132 Ibadan, Nigeria
| | | | | |
Collapse
|
40
|
Price TK, Davar K, Contreras D, Ward KW, Garner OB, Simner PJ, Yang S, Chandrasekaran S. Case Report and Genomic Analysis of Cefiderocol-Resistant Escherichia coli Clinical Isolates. Am J Clin Pathol 2022; 157:257-265. [PMID: 34542575 DOI: 10.1093/ajcp/aqab115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/01/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Cefiderocol is a novel siderophore cephalosporin with in vitro activity against multidrug-resistant (MDR), gram-negative bacteria and intrinsic structural stability to all classes of carbapenemases. We sought to identify gene variants that could affect the mechanism of action (MOA) of cefiderocol. METHODS We report a case of bacteremia in a liver transplant candidate with a strain of carbapenem-resistant Escherichia coli that was found to be resistant to cefiderocol despite no prior treatment with this antimicrobial agent. Using whole-genome sequencing, we characterized the genomic content of this E coli isolate and assessed for genetic variants between related strains that were found to be cefiderocol susceptible. RESULTS We identified several variants in genes with the potential to affect the mechanism of action of cefiderocol. CONCLUSIONS The cefiderocol resistance in the E coli isolate identified in this study is likely due to mutations in the cirA gene, an iron transporter gene.
Collapse
Affiliation(s)
- Travis K Price
- Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | - Kusha Davar
- Division of Infectious Disease, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Deisy Contreras
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kevin W Ward
- Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | - Omai B Garner
- Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | - Patricia J Simner
- Division of Medical Microbiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shangxin Yang
- Department of Pathology and Laboratory Medicine, Los Angeles, CA, USA
| | | |
Collapse
|
41
|
Kamińska K, Mular A, Olshvang E, Nolte NM, Kozłowski H, Wojaczyńska E, Gumienna-Kontecka E. The diversity and utility of arylthiazoline and aryloxazoline siderophores: challenges of total synthesis. RSC Adv 2022; 12:25284-25322. [PMID: 36199325 PMCID: PMC9450019 DOI: 10.1039/d2ra03841b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Siderophores are unique ferric ion chelators produced and secreted by some organisms like bacteria, fungi and plants under iron deficiency conditions. These molecules possess immense affinity and specificity for Fe3+ and other metal ions, which attracts great interest due to the numerous possibilities of application, including antibiotics delivery to resistant bacteria strains. Total synthesis of siderophores is a must since the compounds are present in natural sources at extremely small concentrations. These molecules are extremely diverse in terms of molecular structure and physical and chemical properties. This review is focused on achievements and developments in the total synthesis strategies of naturally occurring siderophores bearing arylthiazoline and aryloxazoline units. A review presents advances in total synthesis of thiazoline and oxazoline-bearing siderophores, unique ferric ion chelators found in some bacteria, fungi and plants.![]()
Collapse
Affiliation(s)
- Karolina Kamińska
- Faculty of Chemistry, University of Wrocław, Fryderyka Joliot-Curie 14, 50-383 Wrocław, Poland
- Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Andrzej Mular
- Faculty of Chemistry, University of Wrocław, Fryderyka Joliot-Curie 14, 50-383 Wrocław, Poland
| | - Evgenia Olshvang
- Inorganic Chemistry I-Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitaetsstrasse, 44801 Bochum, Germany
| | - Nils Metzler Nolte
- Inorganic Chemistry I-Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Universitaetsstrasse, 44801 Bochum, Germany
| | - Henryk Kozłowski
- Faculty of Chemistry, University of Wrocław, Fryderyka Joliot-Curie 14, 50-383 Wrocław, Poland
- Department of Health Sciences, University of Opole, Katowicka 68, 45-060 Opole, Poland
| | - Elżbieta Wojaczyńska
- Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | | |
Collapse
|
42
|
β-lactam Resistance in Pseudomonas aeruginosa: Current Status, Future Prospects. Pathogens 2021; 10:pathogens10121638. [PMID: 34959593 PMCID: PMC8706265 DOI: 10.3390/pathogens10121638] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/06/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa is a major opportunistic pathogen, causing a wide range of acute and chronic infections. β-lactam antibiotics including penicillins, carbapenems, monobactams, and cephalosporins play a key role in the treatment of P. aeruginosa infections. However, a significant number of isolates of these bacteria are resistant to β-lactams, complicating treatment of infections and leading to worse outcomes for patients. In this review, we summarize studies demonstrating the health and economic impacts associated with β-lactam-resistant P. aeruginosa. We then describe how β-lactams bind to and inhibit P. aeruginosa penicillin-binding proteins that are required for synthesis and remodelling of peptidoglycan. Resistance to β-lactams is multifactorial and can involve changes to a key target protein, penicillin-binding protein 3, that is essential for cell division; reduced uptake or increased efflux of β-lactams; degradation of β-lactam antibiotics by increased expression or altered substrate specificity of an AmpC β-lactamase, or by the acquisition of β-lactamases through horizontal gene transfer; and changes to biofilm formation and metabolism. The current understanding of these mechanisms is discussed. Lastly, important knowledge gaps are identified, and possible strategies for enhancing the effectiveness of β-lactam antibiotics in treating P. aeruginosa infections are considered.
Collapse
|
43
|
Mular A, Shanzer A, Kozłowski H, Hubmann I, Misslinger M, Krzywik J, Decristoforo C, Gumienna-Kontecka E. Cyclic Analogs of Desferrioxamine E Siderophore for 68Ga Nuclear Imaging: Coordination Chemistry and Biological Activity in Staphylococcus aureus. Inorg Chem 2021; 60:17846-17857. [PMID: 34783539 PMCID: PMC8653149 DOI: 10.1021/acs.inorgchem.1c02453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
As multidrug-resistant
bacteria are an emerging problem and threat
to humanity, novel strategies for treatment and diagnostics are actively
sought. We aim to utilize siderophores, iron-specific strong chelating
agents produced by microbes, as gallium ion carriers for diagnosis,
applying that Fe(III) can be successfully replaced by Ga(III) without
losing biological properties of the investigated complex, which allows
molecular imaging by positron emission tomography (PET). Here, we
report synthesis, full solution chemistry, thermodynamic characterization,
and the preliminary biological evaluation of biomimetic derivatives
(FOX) of desferrioxamine E (FOXE) siderophore, radiolabeled with 68Ga for possible applications in PET imaging of S.
aureus. From a series of six biomimetic analogs, which differ
from FOXE with cycle length and position of hydroxamic and amide groups,
the highest Fe(III) and Ga(III) stability was determined for the most
FOXE alike compounds–FOX 2-4 and FOX 2-5; we have also established
the stability constant of the Ga-FOXE complex. For this purpose, spectroscopic
and potentiometric titrations, together with the Fe(III)–Ga(III)
competition method, were used. [68Ga]Ga-FOXE derivatives
uptake and microbial growth promotion studies conducted on S. aureus were efficient for compounds with a larger cavity,
i.e., FOX 2-5, 2-6, and 3-5. Even though showing low uptake values,
Fe-FOX 2-4 seems to be also a good Fe-source to support the growth
of S. aureus. Overall, proposed derivatives may hold
potential as inert and stable carrier agents for radioactive Ga(III)
ions for diagnostic medical applications or interesting starting compounds
for further modifications. In this work,
the authors have investigated a set of novel
ferrioxamine E analogs as potential Ga-68 chelators and tools for
infection imaging.
Collapse
Affiliation(s)
- Andrzej Mular
- Faculty of Chemistry, University of Wrocław, 50-383 Wrocław, Poland
| | - Abraham Shanzer
- Department of Organic Chemistry, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Henryk Kozłowski
- Faculty of Chemistry, University of Wrocław, 50-383 Wrocław, Poland.,Department of Health Sciences, University of Opole, 45-060 Opole, Poland
| | - Isabella Hubmann
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Matthias Misslinger
- Institute of Molecular Biology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Julia Krzywik
- TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | | |
Collapse
|
44
|
Xiao G, Alphey MS, Tran F, Pirrie L, Milbeo P, Zhou Y, Bickel JK, Kempf O, Kempf K, Naismith JH, Westwood NJ. Next generation Glucose-1-phosphate thymidylyltransferase (RmlA) inhibitors: An extended SAR study to direct future design. Bioorg Med Chem 2021; 50:116477. [PMID: 34757294 PMCID: PMC8613358 DOI: 10.1016/j.bmc.2021.116477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 11/29/2022]
Abstract
The monosaccharide l-Rhamnose is an important component of bacterial cell walls. The first step in the l-rhamnose biosynthetic pathway is catalysed by glucose-1-phosphate thymidylyltransferase (RmlA), which condenses glucose-1-phosphate (Glu-1-P) with deoxythymidine triphosphate (dTTP) to yield dTDP-d-glucose. In addition to the active site where catalysis of this reaction occurs, RmlA has an allosteric site that is important for its function. Building on previous reports, SAR studies have explored further the allosteric site, leading to the identification of very potent P. aeruginosa RmlA inhibitors. Modification at the C6-NH2 of the inhibitor's pyrimidinedione core structure was tolerated. X-ray crystallographic analysis of the complexes of P. aeruginosa RmlA with the novel analogues revealed that C6-aminoalkyl substituents can be used to position a modifiable amine just outside the allosteric pocket. This opens up the possibility of linking a siderophore to this class of inhibitor with the goal of enhancing bacterial cell wall permeability.
Collapse
Affiliation(s)
- Ganyuan Xiao
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Magnus S Alphey
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Fanny Tran
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Lisa Pirrie
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Pierre Milbeo
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Yi Zhou
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Jasmine K Bickel
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Oxana Kempf
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - Karl Kempf
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK
| | - James H Naismith
- Division of Structural Biology, University of Oxford, and The Rosalind Franklin Institute, Harwell Campus, OX11 0FA, UK.
| | - Nicholas J Westwood
- School of Chemistry and Biomedical Sciences Research Complex, University of St Andrews and EaStCHEM, St Andrews Fife KY16 9ST, UK.
| |
Collapse
|
45
|
Liu R, Miller PA, Miller MJ. Conjugation of Aztreonam, a Synthetic Monocyclic β-Lactam Antibiotic, to a Siderophore Mimetic Significantly Expands Activity Against Gram-Negative Bacteria. ACS Infect Dis 2021; 7:2979-2986. [PMID: 34668698 DOI: 10.1021/acsinfecdis.1c00458] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Monocyclic β-lactams with antibiotic activity were first synthesized more than 40 years ago. Extensive early structure-activity relationship (SAR) studies, especially in the 1980s, emphasized the need for heteroatom activation of monocyclic β-lactams and led to studies of oxamazins, monobactams, monosulfactams, and monocarbams with various side chains and peripheral substitution that revealed potent activity against select strains of Gram-negative bacteria. Aztreonam, still the only clinically used monobactam, has notable activity against many Gram-negative bacteria but limited activity against some of the most problematic multidrug resistant (MDR) strains of Pseudomonas aeruginosa and Acinetobacter baumannii. Herein, we report that extension of the side chain of aztreonam is tolerated and especially that coupling of the side chain free acid with a bis-catechol siderophore mimetic significantly improves activity against the MDR strains of Gram-negative bacteria that are of most significant concern.
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
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
46
|
Ribeiro M, Sousa CA, Simões M. Harnessing microbial iron chelators to develop innovative therapeutic agents. J Adv Res 2021; 39:89-101. [PMID: 35777919 PMCID: PMC9263657 DOI: 10.1016/j.jare.2021.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/24/2021] [Accepted: 10/27/2021] [Indexed: 01/19/2023] Open
Abstract
Microbial iron chelators as a new route to develop inspiring antimicrobials. Siderophore-mimicking antibiotics as a pathogen-targeted strategy. Effectiveness of iron chelators on antibiotic-resistant Gram-negative bacteria. Iron chelators and the treatment of iron overload diseases. Iron chelators as powerful tools for cancer therapy.
Background Aim of Review Key Scientific Concepts of Review
Collapse
|
47
|
Oluwabusola ET, Adebisi OO, Reyes F, Acquah KS, De La Cruz M, Mweetwa LL, Rajakulendran JE, Warner DF, Hai D, Ebel R, Jaspars M. Isolation and characterization of new phenolic siderophores with antimicrobial properties from Pseudomonas sp. UIAU-6B. Beilstein J Org Chem 2021; 17:2390-2398. [PMID: 34621401 PMCID: PMC8450953 DOI: 10.3762/bjoc.17.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 08/30/2021] [Indexed: 01/21/2023] Open
Abstract
Five new phenolic siderophores 1–5 were isolated from the organic extract of a culture broth in a modified SGG medium of Pseudomonas sp. UIAU-6B, obtained from sediments collected from the Oyun river in North Central Nigeria. The structure of the new compounds, pseudomonin A–C (1–3) and pseudomobactin A and B (4 and 5) isolated alongside two known compounds, pseudomonine (6) and salicylic acid (7), were elucidated based on high-resolution mass spectrometry, 1D and 2D NMR analyses. The absolute configuration of the threonine residue in compounds 1–5 was determined by Marfey analysis. The antimicrobial evaluation of compound 4 exhibited the most potent activity against vancomycin-sensitive Enterococcus faecium VS144754, followed by 3 and 5, with MIC values ranging from 8 to 32 µg/mL. Compounds 2 and 3 exhibited moderate activity against Mycobacterium tuberculosis H37Rv, with MIC values of 7.8 and 15.6 µg/mL, respectively. Plausible biosynthetic hypotheses toward the new compounds 1–5 were proposed.
Collapse
Affiliation(s)
| | - Olusoji O Adebisi
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, Kwara State, Ilorin, Nigeria
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avenida del Conocimiento 34, Parque Tecnoloógico de Ciencias de la Salud, E-18016 Granada, Spain
| | - Kojo S Acquah
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - Mercedes De La Cruz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avenida del Conocimiento 34, Parque Tecnoloógico de Ciencias de la Salud, E-18016 Granada, Spain
| | - Larry L Mweetwa
- Marine Biodiscovery Centre, Department of Chemistry, University of Aberdeen, Scotland, UK
| | - Joy E Rajakulendran
- Marine Biodiscovery Centre, Department of Chemistry, University of Aberdeen, Scotland, UK
| | - Digby F Warner
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - Deng Hai
- Marine Biodiscovery Centre, Department of Chemistry, University of Aberdeen, Scotland, UK
| | - Rainer Ebel
- Marine Biodiscovery Centre, Department of Chemistry, University of Aberdeen, Scotland, UK
| | - Marcel Jaspars
- Marine Biodiscovery Centre, Department of Chemistry, University of Aberdeen, Scotland, UK
| |
Collapse
|
48
|
Prabhakar PK. Bacterial Siderophores and Their Potential Applications: A Review. Curr Mol Pharmacol 2021; 13:295-305. [PMID: 32418535 DOI: 10.2174/1874467213666200518094445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/15/2020] [Accepted: 04/23/2020] [Indexed: 12/16/2022]
Abstract
The bacterial infection is one of the major health issues throughout the world. To protect humans from the infection and infectious agents, it is important to understand the mechanism of interaction of pathogens along with their susceptible hosts. This will help us to develop a novel strategy for designing effective new drugs or vaccines. As iron is an essential metal ion required for all the living systems for their growth, as well, it is needed by pathogenic bacterial cells for their growth and development inside host tissues. To get iron from the host tissues, microbes developed an iron-chelating system called siderophore and also corresponding receptors. Siderophores are low molecular weight organic complex produced by different strains of bacteria for the procurement of iron from the environment or host body under the iron deficient-conditions. Mostly in the environment at physiological pH, the iron is present in the ferric ionic form (Fe3+), which is water- insoluble and thus inaccessible for them. Such a condition promotes the generation of siderophores. These siderophores have been used in different areas such as agriculture, treatment of diseases, culture the unculturable strains of bacteria, promotion of plant growth, controlling phytopathogens, detoxification of heavy metal contamination, etc. In the medical field, siderophores can be used as "Trojan Horse Strategy", which forms a complex with antibiotics and also delivers these antibiotics to the desired locations, especially in antibiotic-resistant bacteria. The promising application of siderophore-based use of antibiotics for the management of bacterial resistance can be strategies to be used.
Collapse
Affiliation(s)
- Pranav Kumar Prabhakar
- Department of Transdisciplinary Research, Lovely Professional University, Phagwara, Punjab-144411, India
| |
Collapse
|
49
|
Zscherp R, Coetzee J, Vornweg J, Grunenberg J, Herrmann J, Müller R, Klahn P. Biomimetic enterobactin analogue mediates iron-uptake and cargo transport into E. coli and P. aeruginosa. Chem Sci 2021; 12:10179-10190. [PMID: 34377407 PMCID: PMC8336463 DOI: 10.1039/d1sc02084f] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
The design, synthesis and biological evaluation of the artificial enterobactin analogue EntKL and several fluorophore-conjugates thereof are described. EntKL provides an attachment point for cargos such as fluorophores or antimicrobial payloads. Corresponding conjugates are recognized by outer membrane siderophore receptors of Gram-negative pathogens and retain the natural hydrolyzability of the tris-lactone backbone. Initial density-functional theory (DFT) calculations of the free energies of solvation (ΔG(sol)) and relaxed Fe-O force constants of the corresponding [Fe-EntKL]3- complexes indicated a similar iron binding constant compared to natural enterobactin (Ent). The synthesis of EntKL was achieved via an iterative assembly based on a 3-hydroxylysine building block over 14 steps with an overall yield of 3%. A series of growth recovery assays under iron-limiting conditions with Escherichia coli and Pseudomonas aeruginosa mutant strains that are defective in natural siderophore synthesis revealed a potent concentration-dependent growth promoting effect of EntKL similar to natural Ent. Additionally, four cargo-conjugates differing in molecular size were able to restore growth of E. coli indicating an uptake into the cytosol. P. aeruginosa displayed a stronger uptake promiscuity as six different cargo-conjugates were found to restore growth under iron-limiting conditions. Imaging studies utilizing BODIPYFL-conjugates, demonstrated the ability of EntKL to overcome the Gram-negative outer membrane permeability barrier and thus deliver molecular cargos via the bacterial iron transport machinery of E. coli and P. aeruginosa.
Collapse
Affiliation(s)
- Robert Zscherp
- Institute of Organic Chemistry, Technische Universität Braunschweig Hagenring 30 D-38106 Braunschweig Germany
| | - Janetta Coetzee
- Department for Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research and Department of Pharmacy at Universität des Saarlandes Campus Building E 8.1 D-66123 Saarbrücken Germany
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Johannes Vornweg
- Institute of Organic Chemistry, Technische Universität Braunschweig Hagenring 30 D-38106 Braunschweig Germany
| | - Jörg Grunenberg
- Institute of Organic Chemistry, Technische Universität Braunschweig Hagenring 30 D-38106 Braunschweig Germany
| | - Jennifer Herrmann
- Department for Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research and Department of Pharmacy at Universität des Saarlandes Campus Building E 8.1 D-66123 Saarbrücken Germany
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Rolf Müller
- Department for Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research and Department of Pharmacy at Universität des Saarlandes Campus Building E 8.1 D-66123 Saarbrücken Germany
- German Center for Infection Research (DZIF) Site Hannover-Braunschweig Germany
| | - Philipp Klahn
- Institute of Organic Chemistry, Technische Universität Braunschweig Hagenring 30 D-38106 Braunschweig Germany
| |
Collapse
|
50
|
Colorimetric Sensing of Amoxicillin Facilitated by Molecularly Imprinted Polymers. Polymers (Basel) 2021; 13:polym13132221. [PMID: 34279364 PMCID: PMC8271505 DOI: 10.3390/polym13132221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/30/2021] [Accepted: 07/04/2021] [Indexed: 12/02/2022] Open
Abstract
The scope of the presented research orientates itself towards the development of a Molecularly Imprinted Polymer (MIP)-based dye displacement assay for the colorimetric detection of the antibiotic amoxicillin in aqueous medium. With this in mind, the initial development of an MIP capable of such a task sets focus on monolithic bulk polymerization to assess monomer/crosslinker combinations that have potential towards the binding of amoxicillin. The best performing composition (based on specificity and binding capacity) is utilized in the synthesis of MIP particles by emulsion polymerization, yielding particles that prove to be more homogenous in size and morphology compared to that of the crushed monolithic MIP, which is an essential trait when it comes to the accuracy of the resulting assay. The specificity and selectivity of the emulsion MIP proceeds to be highlighted, demonstrating a higher affinity towards amoxicillin compared to other compounds of the aminopenicillin class (ampicillin and cloxacillin). Conversion of the polymeric receptor is then undertaken, identifying a suitable dye for the displacement assay by means of binding experiments with malachite green, crystal violet, and mordant orange. Once identified, the optimal dye is then loaded onto the synthetic receptor, and the displaceability of the dye deduced by means of a dose response experiment. Alongside the sensitivity, the selectivity of the assay is scrutinized against cloxacillin and ampicillin. Yielding a dye displacement assay that can be used (semi-)quantitatively in a rapid manner.
Collapse
|