1
|
Heckler EP, Ali L, Bhattarai S, Cagle-White B, Smith NC, Moore ED, Coover RA, Abdel Aziz MH, Sarkar A. A benzoxazolyl urea inhibits VraS and enhances antimicrobials against vancomycin intermediate-resistant Staphylococcus aureus. Bioorg Med Chem Lett 2025; 120:130113. [PMID: 39880176 DOI: 10.1016/j.bmcl.2025.130113] [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: 08/08/2024] [Revised: 01/04/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Vancomycin intermediate-resistant Staphylococcus aureus (VISA) is a pathogen of concern. VraS, a histidine kinase, facilitates the VISA phenotype. Here, we reveal a benzoxazolyl urea (chemical 1) that directly inhibits VraS and enhances vancomycin to below the clinical breakpoint against an archetypal VISA strain, Mu50. 50 μM of 1 enhances vancomycin 16-fold to 0.25 μg/mL. The MIC of oxacillin is enhanced 32-fold to 8 μg/mL, only slightly above its clinical breakpoint. The chemical also showed promising enhancement of oxacillin against several MRSA strains. 1 shows ∼30 % inhibition of ATPase activity in VraS and reduces vra gene auto-upregulation typical upon vancomycin exposure. Therefore, 1 inhibits VraS to block normal vra operon function, leading to potent enhancement of cell wall-directed antibiotics. Interestingly, a molecular modeling approach suggests 1 does not displace ATP from the active site, but acts elsewhere. While VraS inhibitors have previously been reported to function against MRSA, to the best of our knowledge, this is the first direct VraS inhibitor ever reported that shows significant enhancement of vancomycin against VISA.
Collapse
Affiliation(s)
- Emerson P Heckler
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Liaqat Ali
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, TX, USA
| | - Shrijan Bhattarai
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, TX, USA
| | - Brittnee Cagle-White
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, TX, USA
| | - Nickalus C Smith
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Erik D Moore
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| | - Robert A Coover
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - May H Abdel Aziz
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, TX, USA.
| | - Aurijit Sarkar
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, NC, USA; Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA.
| |
Collapse
|
2
|
Sibai RE, Farahat ZEM, Qasem HH, Hassan H. The power of DNA-encoded chemical libraries in the battle against drug-resistant bacteria. RSC Adv 2025; 15:14001-14029. [PMID: 40309121 PMCID: PMC12042081 DOI: 10.1039/d5ra00016e] [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: 01/01/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Drug-resistant bacteria are increasingly posing an imminent existential threat, as many bacteria have developed resistance mechanisms that render most antibiotics ineffective. In the meantime, the number of newly approved antibiotics or new clinical antibacterial drug candidates is sharply declining. A key challenge is finding effective pharmacophores that can penetrate and accumulate inside bacterial cells. DNA-encoded chemical libraries (DECLs) play vital roles in accelerating hit identification and screening against various bacterial protein targets. In this review, we highlight the pivotal role of DECLs in accelerating the identification of new pharmacophores and hit compounds against drug-resistant bacteria. This review focuses on the protein targets, where DECLs have directly contributed to the rapid identification of new inhibitors. In addition, this review explores the methods used to screen DECLs against various bacterial targets and discusses the current outlook and perspectives on the role of DECLs in tackling antimicrobial resistance.
Collapse
Affiliation(s)
- Riyad E Sibai
- Department of Microbiology and Biochemistry, Faculty of Science, Zagazig University Zagazig 44519 Egypt
| | - Zainab E M Farahat
- Department of Biochemistry, Faculty of Science, Cairo University Giza 12613 Egypt
| | - Hasnaa H Qasem
- Department of Zoology, Faculty of Science, Ain Shams University Abbassia Cairo 11566 Egypt
| | - Haitham Hassan
- Chemistry Department, School of Life Sciences, University of Sussex Falmer, Brighton East Sussex BN1 9QJ UK
| |
Collapse
|
3
|
Liu X, Song M, Liu Y, Yang S, Chen S, Kang J, Shen J, Zhu K. Rational Design of Natural Xanthones Against Gram-negative Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411923. [PMID: 39888292 PMCID: PMC11984908 DOI: 10.1002/advs.202411923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/21/2024] [Indexed: 02/01/2025]
Abstract
Most antibiotics are ineffective against Gram-negative bacteria owing to the existence of the outer membrane (OM) barrier. The rational design of compounds to expand their antibacterial spectra of antibiotics solely targeting Gram-positive pathogens remains challenging. Here, the design of skeletons from natural products to penetrate the OM are deciphered. Structure-activity relationship analysis shows the optimization of the model of natural xanthones α-mangostin endows the broad-spectrum antibacterial activity. Mechanistic studies demonstrate the lead compound A20 penetrates the OM in a self-promoted pathway through electronic and hydrophobic interactions with lipopolysaccharides and phospholipids in OM. A20 displays rapid bactericidal activity by targeting the cofactor heme in the respiratory complex. The therapeutic efficacy of A20 is demonstrated in two animal models infected with multidrug-resistant Gram-negative bacterial pathogens. The findings elucidate the structural property and self-promoted transportation of a class of antibacterial compounds, to facilitate the design and discovery of antibacterial agents against increasingly prevalent Gram-negative pathogens associated with infections.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Meirong Song
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Ying Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shuyu Yang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shang Chen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jijun Kang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| |
Collapse
|
4
|
Kench T, Sultana Chowdhury N, Rahman KM, Vilar R. Discovery of Phototoxic Metal Complexes with Antibacterial Properties via a Combinatorial Approach. Inorg Chem 2025; 64:5113-5121. [PMID: 40021484 PMCID: PMC11920948 DOI: 10.1021/acs.inorgchem.4c05414] [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: 03/03/2025]
Abstract
Antimicrobial resistance is one of the biggest global healthcare challenges. Therefore, there is an urgent need to develop new molecules that display distinct antibacterial properties to overcome resistance. With this aim, we have developed a combinatorial and semiautomated platform to synthesize and screen a library of 78 compounds against Gram-positive and Gram-negative bacteria. This library is based on octahedral iridium(III) complexes with general formula [Ir(CN)2(NN)]Cl (where CN are cyclometallating polyaromatic ligands and NN are phenanthroline-imidazole or dipyridophenazine derivatives) which are known to generate reactive oxygen species (ROS) upon light irradiation. From the initial screen of the entire library (in the dark and under light irradiation) against Escherichia coli and Staphylococcus aureus, we show that this scaffold is highly effective at inhibiting growth of Gram-positive bacteria at an intermediate dose (16 μg/mL), displaying a hit rate of >30% in the dark and rising to 56% under light irradiation. Six complexes were selected for further studies against a panel of five Gram-positive strains, allowing us to identify two lead complexes with MICs as low as 2 μg/mL. These complexes were studied in more detail to establish their mode of action using a time-kill study against the S. aureus USA300 strain.
Collapse
Affiliation(s)
- Timothy Kench
- Department of Chemistry, Imperial College London, White City Campus, 82 Wood Lane, London W12 OBZ, U.K
| | - Nasima Sultana Chowdhury
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Ramon Vilar
- Department of Chemistry, Imperial College London, White City Campus, 82 Wood Lane, London W12 OBZ, U.K
| |
Collapse
|
5
|
Cardona ST, Rahman ASMZ, Novomisky Nechcoff J. Innovative perspectives on the discovery of small molecule antibiotics. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:19. [PMID: 40082593 PMCID: PMC11906701 DOI: 10.1038/s44259-025-00089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
Antibiotics are essential to modern medicine, but multidrug-resistant (MDR) bacterial infections threaten their efficacy. Resistance evolution shortens antibiotic lifespans, limiting investment returns and slowing new approvals. Consequently, the WHO defines four innovation criteria: new chemical class, target, mode of action (MoA), and lack of cross-resistance. This review explores innovative discovery approaches, including AI-driven screening, metagenomics, and target-based strategies, to develop novel antibiotics that meet these criteria and combat MDR infections.
Collapse
Affiliation(s)
- Silvia T Cardona
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada.
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.
| | | | | |
Collapse
|
6
|
Süssmuth RD, Kulike‐Koczula M, Gao P, Kosol S. Fighting Antimicrobial Resistance: Innovative Drugs in Antibacterial Research. Angew Chem Int Ed Engl 2025; 64:e202414325. [PMID: 39611429 PMCID: PMC11878372 DOI: 10.1002/anie.202414325] [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: 07/29/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
In the fight against bacterial infections, particularly those caused by multi-resistant pathogens known as "superbugs", the need for new antibacterials is undoubted in scientific communities and is by now also widely perceived by the general population. However, the antibacterial research landscape has changed considerably over the past years. With few exceptions, the majority of big pharma companies has left the field and thus, the decline in R&D on antibacterials severely impacts the drug pipeline. In recent years, antibacterial research has increasingly relied on smaller companies or academic research institutions, which mostly have only limited financial resources, to carry a drug discovery and development process from the beginning and through to the beginning of clinical phases. This review formulates the requirements for an antibacterial in regard of targeted pathogens, resistance mechanisms and drug discovery. Strategies are shown for the discovery of new antibacterial structures originating from natural sources, by chemical synthesis and more recently from artificial intelligence approaches. This is complemented by principles for the computer-aided design of antibacterials and the refinement of a lead structure. The second part of the article comprises a compilation of antibacterial molecules classified according to bacterial target structures, e.g. cell wall synthesis, protein synthesis, as well as more recently emerging target classes, e.g. fatty acid synthesis, proteases and membrane proteins. Aspects of the origin, the antibacterial spectrum, resistance and the current development status of the presented drug molecules are highlighted.
Collapse
Affiliation(s)
- Roderich D. Süssmuth
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Marcel Kulike‐Koczula
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Peng Gao
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Simone Kosol
- Medical School BerlinDepartment Human MedicineRüdesheimer Strasse 5014195BerlinGermany
| |
Collapse
|
7
|
He Y, Wang X, He S, Han X, Wang A, Zhang F, Deng J, Long X, Lin J, Feng Y, He X, Li Z, Li J, Luo F, Tan H. Enhancing Antibiotic-Resistant Bacterial Infection Therapy: Self-Assembling Gemini Quaternary Ammonium-Functionalized Peptide Nanoassemblies with Multiple Antibacterial Mechanisms. ACS NANO 2025; 19:6977-6992. [PMID: 39936387 DOI: 10.1021/acsnano.4c14689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
The rising threat of antimicrobial-resistant (AMR) infections highlights the urgent need for effective antimicrobial agents and therapies. Peptide-based antimicrobial nanomaterials are well-placed to meet this need. Here, we explore the conjugation of antimicrobial gemini quaternary ammonium compounds (GQAs) with designed short hexapeptides to create cationic antimicrobial nanomaterials with low cytotoxicity and minimal resistance tendency. (WA)3GQA8C self-assembles into nanoparticles and exhibits potent antimicrobial activity against drug-resistant pathogens and enhanced stability. (WA)3GQA8C protects against subcutaneous abscess infection and rescues mice from acute peritonitis infection by reducing the systemic bacterial burden and alleviating organ damage, with superior effects to vancomycin. Notably, (WA)3GQA8C thoroughly disrupts bacterial membrane integrity akin to peeling fruit to induce bacterial membrane disintegration, a feat inaccessible to conventional antibiotics. Mechanistic studies suggest that (WA)3GQA8C targets the bacterial membrane phospholipids phosphatidylglycerol (PG), inducing PG deformation to form fibrous or lamellar structures, which leads to the disruption of the bacterial membrane. Furthermore, the interference in lipoprotein trafficking exacerbates damage to bacterial membrane integrity. (WA)3GQA8C also synergizes antimicrobial activity by impairing the protein synthesis function of the ribosome. These quaternized peptide nanoassemblies provide a rational strategy for designing peptide-based antimicrobial nanomaterials to combat the growing threats of resistant bacteria.
Collapse
Affiliation(s)
- Yuanyuan He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Shanshan He
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jielun Deng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xirui Long
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jingjing Lin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yuan Feng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xueling He
- Laboratory animal center, Sichuan University, Chengdu, Sichuan 610207, China
| | - Zhen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Feng Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X center of materials, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
8
|
Kumari R, Saraogi I. Navigating Antibiotic Resistance in Gram-Negative Bacteria: Current Challenges and Emerging Therapeutic Strategies. Chemphyschem 2025:e202401057. [PMID: 39970066 DOI: 10.1002/cphc.202401057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The rapid rise of antibiotic resistance poses a severe global health crisis, necessitating new approaches to counter this growing threat. The problem is exacerbated in Gram-negative bacterial pathogens as many antibiotics are unable to enter these cells owing to their unique additional outer membrane barrier. In this review, we discuss the challenges of targeting Gram-negative bacteria, including the complexity of the outer membrane, as well as the presence of efflux pumps and β-lactamases that contribute to resistance. We also review solutions proposed to facilitate the entry and accumulation of antibiotics in Gram-negative bacteria. These involve using existing antibiotics in combination with other inhibitors to attack the bacterial cell synergistically. We also highlight approaches to target Gram-negative pathogens via novel modes of action, providing new strategies to tackle antibiotic resistance.
Collapse
Affiliation(s)
- Reshma Kumari
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| |
Collapse
|
9
|
Jung E, Griesser T, Costafrolaz J, Duverger O, Mattenberger Y, Dittmann S, Dorst A, Major A, Dailler D, Schäfle D, Sievers S, Brodolin K, Viollier PH, Sander P, Gademann K. Switching Residues: A Platform for the Synthesis of Fidaxomicin Antibiotics. Angew Chem Int Ed Engl 2025; 64:e202419095. [PMID: 39584779 PMCID: PMC11811690 DOI: 10.1002/anie.202419095] [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: 10/03/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Peripheral modification is often the main approach to optimize natural products for improved biological activity or desired physicochemical properties. This procedure inevitably increases molecular weight, often accompanied by undesired increased lipophilicity. Removing structural elements from natural products is not always tolerated. This is also the case for the antibiotic fidaxomicin (Fdx), where every structural component has been shown to be crucial for antibiotic activity. In this work, we demonstrate how the residue switching approach can maintain biological activity of Fdx derivatives by replacing the rhamnoside-dichlorohomoorsellinate moiety of Fdx with smaller, more polar building blocks. We used palladium-catalysed allylic substitution to selectively install N-nucleophiles on the core of Fdx. The new derivatives were designed to mimic the binding of Fdx to the bacterial RNA polymerase. Evaluation against Mycobacterium tuberculosis, Clostridioides difficile, and the Gram-negative model organism Caulobacter crescentus demonstrated that the newly introduced residues can restore antibiotic activity, which was further supported by on-target RNA polymerase assays. We combined the allylic substitution with an organocatalysed novioside acylation protocol to enable the functionalisation of two vectors on Fdx in one pot. This platform greatly expands the accessible chemical space for Fdx derivatives and enables the future development of systemic Fdx antibiotics.
Collapse
Affiliation(s)
- Erik Jung
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Tizian Griesser
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerland
| | - Jordan Costafrolaz
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Ondine Duverger
- Institut de Recherche en Infectiologie de MontpellierUniv. MontpellierCNRSMontpellier34293France
| | - Yves Mattenberger
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Silvia Dittmann
- Department of Microbial Physiology and Molecular BiologyInstitute of MicrobiologyCenter for Functional Genomics of MicrobesUniversity of GreifswaldGreifswaldGermany
| | - Andrea Dorst
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Alexander Major
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - David Dailler
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| | - Daniel Schäfle
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerland
| | - Susanne Sievers
- Department of Microbial Physiology and Molecular BiologyInstitute of MicrobiologyCenter for Functional Genomics of MicrobesUniversity of GreifswaldGreifswaldGermany
| | - Konstantin Brodolin
- Institut de Recherche en Infectiologie de MontpellierUniv. MontpellierCNRSMontpellier34293France
| | - Patrick H. Viollier
- Department of Microbiology and Molecular MedicineFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Peter Sander
- Institute of Medical MicrobiologyUniversity of ZurichZurichSwitzerlandNational Reference Laboratory for MycobacteriaUniversity of ZurichZurichSwitzerland
| | - Karl Gademann
- Department of ChemistryUniversity of Zurich8057ZürichSwitzerland
| |
Collapse
|
10
|
George M, Wright GD. Revisiting the potential of natural products in antimycobacterial therapy: advances in drug discovery and semisynthetic solutions. Curr Opin Microbiol 2025; 83:102576. [PMID: 39742555 DOI: 10.1016/j.mib.2024.102576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025]
Abstract
Natural products have been pivotal in treating mycobacterial infections with early antibiotics such as streptomycin, forming the foundation of tuberculosis therapy. However, the emergence of multidrug-resistant and extensively drug-resistant Mycobacterium species has intensified the need for novel antimycobacterial agents. In this review, we revisit the historical contributions of natural products to antimycobacterial drug discovery and highlight recent advances in the field. We assess the application of molecular networking and the exploration of unculturable bacteria in identifying new antimycobacterial compounds such as amycobactin and levesquamides. We also highlight the role of semisynthesis in optimizing natural products, exemplified by sequanamycins and spectinomycin analogs that evade M. tuberculosis' intrinsic resistance. Finally, we discuss emerging technologies that are promising to accelerate the discovery and development of next-generation antimycobacterial therapies. Despite ongoing challenges, these innovative approaches offer renewed hope in addressing the growing crisis of drug-resistant mycobacterial infections.
Collapse
Affiliation(s)
- Maya George
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
11
|
Farha MA, Tu MM, Brown ED. Important challenges to finding new leads for new antibiotics. Curr Opin Microbiol 2025; 83:102562. [PMID: 39603107 DOI: 10.1016/j.mib.2024.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Identification of new antibiotics remains a huge challenge. The last antibiotic of new chemical class and mechanism was discovered more than 30 years ago. Advances since have been largely incremental modifications to a limited number of chemical scaffolds. Discovering and developing truly new antibiotics is challenging: the science is complex, and the development process is time consuming and expensive. Herein, we focus on the discovery phase of modern antibacterial research and development. We argue that antibacterial discovery has been challenged by a poor understanding of bacterial permeability, by generic in vitro conventions that ignore the host, and by the inherent complexity of bacterial systems. Together, these factors have colluded to challenge modern, industrial, and reductionist approaches to antibiotic discovery. Nevertheless, advances in our understanding of many of these obstacles, including a new appreciation for the complexity of both host and pathogen biology, bode well for future efforts.
Collapse
Affiliation(s)
- Maya A Farha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Megan M Tu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada; Michael G. DeGroote Institute of Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
12
|
Carnevale Neto F, Vessecchi R. Structural elucidation of 14-membered ring macrolide antibiotics using electrospray ionization tandem mass spectrometry and density functional theory calculations. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9913. [PMID: 39448384 DOI: 10.1002/rcm.9913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/07/2024] [Accepted: 09/02/2024] [Indexed: 10/26/2024]
Abstract
RATIONALE Macrolides are critical antibiotics featuring a macrocyclic lactone core with deoxy sugars. Understanding their gas-phase fragmentation is challenging but essential for improving structural elucidation in mass spectrometry, which has implications for drug discovery and development. METHODS We used electrospray ionization collision-induced dissociation tandem mass spectrometry (ESI-CID-MS) combined with quantum chemical calculations to investigate the fragmentation pathways of erythromycin A and roxithromycin. This approach helps elucidate the preferred fragmentation routes influenced by protonation sites. RESULTS Macrolides showed similar fragmentation patterns, including sequential losses of saccharide or amino sugar units and dehydration from the macrocycle core. Multiple competitive pathways were observed, influenced by protonation sites. Computational studies confirmed the most favorable protonation sites and their impact on fragmentation, providing insights into key diagnostic product ions. Subsequent fragments involved rearrangement pathways such as alkene formation and cleavages via remote hydrogen transfers and pericyclic reactions. CONCLUSIONS Our integrated approach offers a comprehensive understanding of macrolide fragmentation, enhancing structural elucidation and potential applications in drug development. This study advances mass spectrometry analysis of macrolides, contributing to pharmaceutical research by integrating orthogonal annotation methods and fragmentation studies.
Collapse
Affiliation(s)
- Fausto Carnevale Neto
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Ricardo Vessecchi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
13
|
Bogyo M, Upadhyay T, Woods E, Ahator S, Julin K, Faucher F, Hollander M, Pedowitz N, Abegg D, Hammond I, Eke I, Wang S, Chen S, Bennett J, Jo J, Lentz C, Adibekian A, Fellner M. Covalent-fragment screening identifies selective inhibitors of multiple Staphylococcus aureus serine hydrolases important for growth and biofilm formation. RESEARCH SQUARE 2024:rs.3.rs-5494070. [PMID: 39711551 PMCID: PMC11661381 DOI: 10.21203/rs.3.rs-5494070/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Staphylococcus aureus is a leading cause of bacteria-associated mortality worldwide. This is largely because infection sites are often difficult to localize and the bacteria forms biofilms which are not effectively cleared using classical antibiotics. Therefore, there is a need for new tools to both image and treat S. aureus infections. We previously identified a group of S. aureus serine hydrolases known as fluorophosphonate-binding hydrolases (Fphs), which regulate aspects of virulence and lipid metabolism. However, because their structures are similar and their functions overlap, it remains challenging to distinguish the specific roles of individual members of this family. In this study, we applied a high-throughput screening approach using a library of covalent electrophiles to identify inhibitors for FphB, FphE, and FphH. We identified inhibitors that irreversibly bind to the active-site serine residue of each enzyme with high potency and selectivity without requiring extensive medicinal chemistry optimization. Structural and biochemical analysis identified novel binding modes for several of the inhibitors. Selective inhibitors of FphH impaired both bacterial growth and biofilm formation while Inhibitors of FphB and FphE had no impact on cell growth and only limited impact on biofilm formation. These results suggest that all three hydrolases likely play functional, but non-equivalent roles in biofilm formation and FphH is a potential target for development of therapeutics that have both antibiotic and anti-biofilm activity. Overall, we demonstrate that focused covalent fragment screening can be used to rapidly identify highly potent and selective electrophiles targeting bacterial serine hydrolases. This approach could be applied to other classes of lipid hydrolases in diverse pathogens or higher eukaryotes.
Collapse
|
14
|
Lee S, Lee T, Kim MK, Ahn JH, Jeong S, Park KH, Chong Y. 7- O-Carboxylic Acid-Substituted 3- O-Alkyl Difluoroquercetin; An Aztreonam-Potentiating Agent Against Carbapenemase-Producing Pseudomonas aeruginosa Through Simultaneous Inhibition of Metallo-β-Lactamase and Efflux Pump. Antibiotics (Basel) 2024; 13:1202. [PMID: 39766592 PMCID: PMC11672637 DOI: 10.3390/antibiotics13121202] [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: 10/31/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Previously, we reported that 3-O-alkyl difluoroquercetins (di-F-Q) potentiates the antimicrobial activity of aztreonam (ATM) against metallo-β-lactamase (MBL)-producing P. aeruginosa through simultaneous inhibition of MBLs and efflux pumps. However, the ATM-potentiating activity of the 3-O-alkyl di-F-Q was observed only at high and potentially toxic concentrations (32 mg/L). Methods: As both MBLs and efflux pumps reside in the periplasm of Gram-negative bacteria, their inhibitors should accumulate in the periplasmic space. However, the outer membrane porins, the major entry pathway in Gram-negative bacteria, allow the passive diffusion of hydrophilic polar molecules across the outer membrane. Thus, we reasoned that the introduction of a polar substituent at 7-OH position of 3-O-alkyl di-F-Q would enhance its periplasmic concentration to result in potentiation of ATM at lower concentrations. Results: The title compound 5 exhibited inhibitory activity against NDM-1 as well as the efflux pump of P. aeruginosa, which resulted in synergistical potentiation of ATM. A combination of ATM (8 mg/L) and 5 (8 mg/L) inhibited 80% of the ATM-resistant CPPA, while ATM alone did not show any inhibition. In addition, only 4 mg/L of 5 was needed to reduce the MIC90 of ATM four-fold in ATM-resistant CPPA (n = 15). The time-kill data further supported the effectiveness of the combined treatment of ATM with 5, and the combination of ATM (1xMIC) with 8 mg/L of 5 showed bactericidal effects in every bacterial strain tested (PA-002, blaIMP, PA-003, blaVIM, PA-014, blaGES, and PA-017, blaNDM) by reducing the bacterial loads by 5.1 log10~8.9 log10. Conclusions: The title compound 5 exhibited inhibitory activity against NDM-1 as well as the efflux pump of P. aeruginosa, and the combined inhibitory activity resulted in synergistical potentiation of ATM. It should be noted that most CPPA isolates tested were sensitized to 8 mg/L of ATM upon combination with 4~8 mg/L of 5.
Collapse
Affiliation(s)
- Seongyeon Lee
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (S.L.); (T.L.); (M.K.K.); (J.H.A.)
| | - Taegum Lee
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (S.L.); (T.L.); (M.K.K.); (J.H.A.)
| | - Mi Kyoung Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (S.L.); (T.L.); (M.K.K.); (J.H.A.)
| | - Joong Hoon Ahn
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (S.L.); (T.L.); (M.K.K.); (J.H.A.)
| | - Seri Jeong
- Department of Laboratory Medicine, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
| | - Ki-Ho Park
- Department of Infectious Disease, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Youhoon Chong
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (S.L.); (T.L.); (M.K.K.); (J.H.A.)
| |
Collapse
|
15
|
Burillo A, Bouza E. The evolution of knowledge for treating Gram-negative bacterial infections. Curr Opin Infect Dis 2024; 37:573-581. [PMID: 39259682 DOI: 10.1097/qco.0000000000001055] [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: 09/13/2024]
Abstract
PURPOSE OF REVIEW Infections caused by nonprimarily pathogenic Gram-negative bacilli (GNB) have been increasingly reported from the second half of the 20th century to the present. This phenomenon has expanded during the antibiotic era and in the presence of immunodeficiency.Before the discovery of sulphonamides and penicillin G, infections caused by GNB were rare compared to Gram-positive infections. The advent of anticancer therapy, the expansion of surgical procedures, the use of corticosteroids, and the implantation of prosthetic materials, along with better control of Gram-positive infections, have promoted the current increase in GNB infections.GNB have similar antimicrobial targets to Gram-positive bacteria. However, only antibiotics that can penetrate the double membrane of GNB and remain in them for a sufficient duration have antibacterial activity against them. RECENT FINDINGS Sulphonamides and early penicillins had limited activity against GNB. Ampicillin and subsequent beta-lactams expanded their spectrum to treat GNB. Aminoglycosides may re-surge with less toxic drugs, as highly resistant to beta-lactams GNB rise. Polymyxins, tetracyclines, and fluoroquinolones are also used for GNB. Combinations with other agents may be needed in specific cases, such as in the central nervous system and prostate, where beta-lactams may have difficulty reaching the infection site.Alternatives to current treatments must be sought in the discovery of new drug families and therapies such as phage therapy combined with antibiotics. SUMMARY Narrower-spectrum immunosuppressive therapies and antibiotics, antimicrobials that minimally intervene with the human microbiota, and instant diagnostic methods are necessary to imagine a future where currently dominant bacteria in infectious pathology lose their preeminence.
Collapse
Affiliation(s)
- Almudena Burillo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón
- Medicine Department, School of Medicine, Universidad Complutense de Madrid
- CIBER of Respiratory Diseases (CIBERES CB06/06/0058)
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón
- Medicine Department, School of Medicine, Universidad Complutense de Madrid
- CIBER of Respiratory Diseases (CIBERES CB06/06/0058)
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| |
Collapse
|
16
|
Bennett F, Huang Y, Dong S, Jiang J, Hunter D, Zhao Z, Gu X, Scott JD, Tang H, Yang D, Xiao L, Scapin G, Fischmann T, Mirza A, Dayananth P, Painter RE, Villafania A, Garlisi CG, Zhang R, Mayhood TW, Si Q, Li N, Amin RP, Chen F, Bhatt B, Regan CP, Regan H, Lin X, Wu J, Leithead A, Young K, Pasternak A. Discovery of sulfone containing metallo-β-lactamase inhibitors with reduced bacterial cell efflux and histamine release issues. Bioorg Med Chem Lett 2024; 114:129989. [PMID: 39396683 DOI: 10.1016/j.bmcl.2024.129989] [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: 07/30/2024] [Revised: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
The design, syntheses and antibacterial evaluation of sulfone analogues of previously disclosed metallo-β-lactamase inhibitors (MBLis) are described. The novel derivatives were overall more effective in gram-negative bacterial cell-based assays when combined with imipenem and relebactam. The major contributors to the improved anti-bacterial activity are enhanced enzyme-inhibitor interactions and reduced bacterial cell efflux monitored via an efflux assay involving isogenic Pseudomonas aeruginosa efflux + and efflux - tool strains.
Collapse
Affiliation(s)
- Frank Bennett
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA.
| | - Yuhua Huang
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Shuzhi Dong
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Jinlong Jiang
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - David Hunter
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Zhiqiang Zhao
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Xin Gu
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Jack D Scott
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Haiqun Tang
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Dexi Yang
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| | - Li Xiao
- Merck & Co., Inc., Department of Computational and Structural Chemistry, 126 E. Lincoln Avenue., Rahway, NJ 07065, USA
| | - Giovanna Scapin
- Merck & Co., Inc., Department of Computational and Structural Chemistry, 126 E. Lincoln Avenue., Rahway, NJ 07065, USA
| | - Thierry Fischmann
- Merck & Co., Inc., Department of Computational and Structural Chemistry, 126 E. Lincoln Avenue., Rahway, NJ 07065, USA
| | - Asra Mirza
- Merck & Co., Inc., Department of Infectious Diseases, 770 Sumneytown Pike., West Point, PA 19486, USA
| | - Priya Dayananth
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Ronald E Painter
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Artjohn Villafania
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Charles G Garlisi
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Rumin Zhang
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Todd W Mayhood
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Qian Si
- Merck & Co., Inc., Department of Pharmacology, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Nianyu Li
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Rupesh P Amin
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Feifei Chen
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Bhavana Bhatt
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Christopher P Regan
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Hilary Regan
- Merck & Co., Inc., Department of Nonclinical Drug Safety, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Xinjie Lin
- Merck & Co., Inc., Department of Pharmacokinetics and Pharmacodynamics, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Jin Wu
- Merck & Co., Inc., Department of Pharmacokinetics and Pharmacodynamics, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Andrew Leithead
- Merck & Co., Inc., Department of Pharmaceutical Sciences, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Katherine Young
- Merck & Co., Inc., Department of Infectious Diseases, 770 Sumneytown Pike., West Point, PA 19486, USA
| | - Alexander Pasternak
- Merck & Co., Inc., Department of Medicinal Chemistry, 126 E. Lincoln Avenue, Rahway, NJ 07065, USA
| |
Collapse
|
17
|
Vergalli J, Réfrégiers M, Ruggerone P, Winterhalter M, Pagès JM. Advances in methods and concepts provide new insight into antibiotic fluxes across the bacterial membrane. Commun Biol 2024; 7:1508. [PMID: 39543341 PMCID: PMC11564671 DOI: 10.1038/s42003-024-07168-4] [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: 05/16/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
The sophisticated envelope of Gram-negative bacteria modulates the uptake of small molecules in a side-chain-sensitive manner. Despite intensive theoretical and experimental investigations, a general set of pathways underpinning antibiotic uptake has not been identified. This manuscript discusses the passive influx versus active efflux of antibiotics, considering the responsible membrane proteins and the transported molecules. Recent methods have analyzed drug transport across the bacterial membrane in order to understand their activity. The combination of in vitro, in cellulo and in silico methods shed light on the key, mainly electrostatic, interactions between the molecule surface, porins and transporters during permeation. A key factor is the relationship between the dose of an active compound near its target and its antibacterial activity during the critical early window. Today, methodology breakthroughs provide fruitful tools to precisely dissect drug transport, identify key steps in drug resistance associated with membrane impermeability and efflux, and highlight key parameters to generate more effective drugs.
Collapse
Affiliation(s)
| | | | - Paolo Ruggerone
- Department of Physics, University of Cagliari, 09042, Monserrato, CA, Italy
| | - Mathias Winterhalter
- Department of Life Sciences and Chemistry, Constructor University, 28719, Bremen, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
| | | |
Collapse
|
18
|
Andrade Meirelles M, Almeida VM, Sullivan JR, de Toledo I, Dos Reis CV, Cunha MR, Zigweid R, Shim A, Sankaran B, Woodward EL, Seibold S, Liu L, Mian MR, Battaile KP, Riley J, Duncan C, Simeons FRC, Ferguson L, Joji H, Read KD, Lovell S, Staker BL, Behr MA, Pilli RA, Couñago RM. Rational Exploration of 2,4-Diaminopyrimidines as DHFR Inhibitors Active against Mycobacterium abscessus and Mycobacterium avium, Two Emerging Human Pathogens. J Med Chem 2024; 67:19143-19164. [PMID: 39468773 DOI: 10.1021/acs.jmedchem.4c01594] [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: 10/30/2024]
Abstract
Nontuberculous mycobacteria (NTM) are emerging human pathogens linked to severe pulmonary diseases. Current treatments involve the prolonged use of multiple drugs and are often ineffective. Bacterial dihydrofolate reductase (DHFR) is a key enzyme targeted by antibiotics in Gram-negative bacterial infections. However, existing DHFR inhibitors designed for Gram-negative bacteria often fail against mycobacterial DHFRs. Here, we detail the rational design of NTM DHFR inhibitors based on P218, a malarial DHFR inhibitor. We identified compound 8, a 2,4-diaminopyrimidine exhibiting improved pharmacological properties and activity against purified DHFR, and whole cell cultures of two predominant NTM species: Mycobacterium avium and Mycobacterium abscessus. This study underscores the potential of compound 8 as a promising candidate for the in vivo validation of DHFR as an effective treatment against NTM infections.
Collapse
Affiliation(s)
- Matheus Andrade Meirelles
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Vitor M Almeida
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Jaryd R Sullivan
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
| | - Ian de Toledo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Caio Vinicius Dos Reis
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Micael Rodrigues Cunha
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Rachel Zigweid
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Abraham Shim
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Elijah L Woodward
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Steve Seibold
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Lijun Liu
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mohammad Rasel Mian
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Kevin P Battaile
- New York Structural Biology Center, Upton, New York 11973, United States
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Christina Duncan
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Frederick R C Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Liam Ferguson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Halimatu Joji
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Scott Lovell
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Bart L Staker
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Marcel A Behr
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
- Department of Medicine, McGill University Health Centre, Montréal H4A 3J1, Canada
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Rafael M Couñago
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
19
|
Deylami J, Chng SS, Yong EH. Elucidating Antibiotic Permeation through the Escherichia coli Outer Membrane: Insights from Molecular Dynamics. J Chem Inf Model 2024; 64:8310-8321. [PMID: 39480067 PMCID: PMC11558678 DOI: 10.1021/acs.jcim.4c01249] [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: 07/16/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024]
Abstract
Antibiotic resistance represents a critical public health threat, with an increasing number of Gram-negative pathogens demonstrating resistance to a broad range of clinical drugs. A primary challenge in enhancing antibiotic efficacy is overcoming the robust barrier presented by the bacterial outer membrane. Our research addresses a longstanding question: What is the rate of antibiotic permeation across the outer membrane (OM) of Gram-negative bacteria? Utilizing molecular dynamics (MD) simulations, we assess the passive permeability profiles of four commercially available antibiotics─gentamicin, novobiocin, rifampicin, and tetracycline across an asymmetric atomistic model of the Escherichia coli (E. coli) OM, employing the inhomogeneous solubility-diffusion model. Our examination of the interactions between these drugs and their environmental context during OM permeation reveals that extended hydrogen bond formation and drug-cation interactions significantly hinder the energetics of passive permeation, notably affecting novobiocin. Our MD simulations corroborate well with experimental data and reveal new implications of solvation on drug permeability, overall advancing the possible use of computational prediction of membrane permeability in future antibiotic discovery.
Collapse
Affiliation(s)
- Javad Deylami
- School
of Physical and Mathematical Sciences, Nanyang
Technological University, Singapore 637371, Singapore
| | - Shu Sin Chng
- Department
of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Ee Hou Yong
- School
of Physical and Mathematical Sciences, Nanyang
Technological University, Singapore 637371, Singapore
| |
Collapse
|
20
|
Kumar P, Saini S, Gangwar A, Sharma R, Anal JMH. Antibacterial activity of structurally diverse natural prenylated isobavachalcone derivatives. RSC Adv 2024; 14:32771-32785. [PMID: 39429936 PMCID: PMC11484510 DOI: 10.1039/d4ra05370b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Isobavachalcone (IBC) is a natural prenylated flavonoid containing chalcone and prenyl chain moieties with a wide range of biological and pharmacological properties. In this work, we synthesized structurally diversified derivatives (IBC-2 to IBC-10) from the natural prenylated chalcone IBC isolated from Psoralea corylifolia and assessed their antibacterial potency against the Gram-positive and Gram-negative bacterial strains S. aureus ATCC 29213, MRSA ATCC 15187, E. coli ATCC25922 and P. aeruginosa ATCC 27853. IBC and IBC-2 exhibited a minimum inhibition concentration (MIC) of 5.0 μM against S. aureus ATCC 29213, whereas IBC-3 exhibited a broad-spectrum activity against Gram-positive and Gram-negative pathogens. Cytotoxicity assessments on the murine RAW 264.7 macrophage cell line revealed minimal to moderate cytotoxicity for IBC-2 and IBC-3 with a favorable selectivity index (>10). Time- and concentration-dependent studies further supported the bactericidal nature of the compounds, as IBC, IBC-2, and IBC-3 exhibited concentration-dependent killing of S. aureus in a time-dependent manner. Furthermore, combination studies, SEM analysis, and PI staining suggest that IBC-3's mechanism of action targets the bacteria's cytoplasmic membrane or cell wall. The bioactive compounds displayed promising drug-like characteristics and a favorable pharmacokinetic profile (ADME-Tox), indicating a projected high oral bioavailability. Structure-activity relationships (SARs) drawn from this study reveal that a prenyl chain at the A-ring and hydroxy functional groups attached to the aromatic rings of chalcone scaffolds are responsible for this antibacterial potential, which will be helpful in the future discovery and development of antibiotics from natural products to overcome the antibiotic resistance crisis.
Collapse
Affiliation(s)
- Puneet Kumar
- Natural Products and Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sapna Saini
- Infectious Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Anjali Gangwar
- Infectious Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Rashmi Sharma
- Infectious Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Jasha Momo H Anal
- Natural Products and Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| |
Collapse
|
21
|
Kohnhäuser D, Seedorf T, Cirnski K, Heimann D, Coetzee J, Sordello S, Richter J, Stappert M, Sabuco JF, Corbett D, Bacqué E, Rox K, Herrmann J, Vassort A, Müller R, Kirschning A, Brönstrup M. Optimization of the Central α-Amino Acid in Cystobactamids to the Broad-Spectrum, Resistance-Breaking Antibiotic CN-CC-861. J Med Chem 2024; 67:17162-17190. [PMID: 39303218 PMCID: PMC11472329 DOI: 10.1021/acs.jmedchem.4c00927] [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: 04/19/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Cystobactamids have a unique oligoarylamide structure and exhibit broad-spectrum activity against Gram-negative and Gram-positive bacteria. In this study, the central α-amino acid of the cystobactamid scaffold was modified to address the relevance of stereochemistry, hydrogen bonding and polarity by 33 derivatives. As demonstrated by three matched molecular pairs, l-amino acids were preferred over d-amino acids. A rigidification to a six-membered system stabilized the bioactive conformation for the on-target Escherichia coli gyrase, but did not improve antimicrobial activity. Compound CN-CC-861, carrying a propargyl side chain, had more than 16-fold lower minimal inhibitory concentration (MIC) values against Enterococcus faecalis, Staphylococci and Acinetobacter strains, compared to known analogues. Moreover, CN-CC-861 retained activity against multidrug-resistant enterococci, displayed strong bactericidal activity, moderate-low frequencies of resistance and in vivo efficacy in a neutropenic thigh infection model with E. coli. Overall, the findings will guide the design of new promising structures with higher activities and broader spectrum.
Collapse
Affiliation(s)
- Daniel Kohnhäuser
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Tim Seedorf
- Institute
of Organic Chemistry and Biomolecular Drug Research Centre (BMWZ), Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Katarina Cirnski
- Microbial
Natural Products, Helmholtz Institute for Pharmaceutical Research
Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dominik Heimann
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Janetta Coetzee
- Microbial
Natural Products, Helmholtz Institute for Pharmaceutical Research
Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Sylvie Sordello
- Evotec
ID, 1541 Avenue Marcel
Merieux, 69289 Marcy
l’Etoile, France
| | - Jana Richter
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Moritz Stappert
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | | | - David Corbett
- Evotec
ID, 1541 Avenue Marcel
Merieux, 69289 Marcy
l’Etoile, France
| | - Eric Bacqué
- Evotec
ID, 1541 Avenue Marcel
Merieux, 69289 Marcy
l’Etoile, France
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Microbial
Natural Products, Helmholtz Institute for Pharmaceutical Research
Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Aurelie Vassort
- Evotec
ID, 1541 Avenue Marcel
Merieux, 69289 Marcy
l’Etoile, France
| | - Rolf Müller
- Microbial
Natural Products, Helmholtz Institute for Pharmaceutical Research
Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Pharmacy at Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Andreas Kirschning
- Institute
of Organic Chemistry and Biomolecular Drug Research Centre (BMWZ), Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
- Uppsala Biomedical
Center (BMC), Uppsala University, Husargatan 3, 752 37 Uppsala, Sweden
| | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute
of Organic Chemistry and Biomolecular Drug Research Centre (BMWZ), Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
- German
Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
22
|
Mielniczuk S, Hoff K, Baselious F, Li Y, Haupenthal J, Kany AM, Riedner M, Rohde H, Rox K, Hirsch AKH, Krimm I, Sippl W, Holl R. Development of Fragment-Based Inhibitors of the Bacterial Deacetylase LpxC with Low Nanomolar Activity. J Med Chem 2024; 67:17363-17391. [PMID: 39303295 PMCID: PMC11472313 DOI: 10.1021/acs.jmedchem.4c01262] [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: 06/03/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
In a fragment-based approach using NMR spectroscopy, benzyloxyacetohydroxamic acid-derived inhibitors of the bacterial deacetylase LpxC bearing a substituent to target the uridine diphosphate-binding site of the enzyme were developed. By appending privileged fragments via a suitable linker, potent LpxC inhibitors with promising antibacterial activities could be obtained, like the one-digit nanomolar LpxC inhibitor (S)-13j [Ki (EcLpxC C63A) = 9.5 nM; Ki (PaLpxC): 5.6 nM]. To rationalize the observed structure-activity relationships, molecular docking and molecular dynamics studies were performed. Initial in vitro absorption-distribution-metabolism-excretion-toxicity (ADMET) studies of the most potent compounds have paved the way for multiparameter optimization of our newly developed isoserine-based amides.
Collapse
Affiliation(s)
- Sebastian Mielniczuk
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Katharina Hoff
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Fady Baselious
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Yunqi Li
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
- Shanghai
Key Laboratory of Regulatory Biology, The Institute of Biomedical
Sciences & School of Life Sciences, East China Normal University, 200241 Shanghai, China
| | - Jörg Haupenthal
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Maria Riedner
- Technology
Platform Mass Spectrometry, Universität
Hamburg, Mittelweg 177, 20148 Hamburg, Germany
| | - Holger Rohde
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
- Institute
of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Helmholtz International Lab for Anti-infectives, Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabelle Krimm
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Ralph Holl
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| |
Collapse
|
23
|
Li W, Tao Z, Zhou M, Jiang H, Wang L, Ji B, Zhao Y. Antibiotic adjuvants against multidrug-resistant Gram-negative bacteria: important component of future antimicrobial therapy. Microbiol Res 2024; 287:127842. [PMID: 39032266 DOI: 10.1016/j.micres.2024.127842] [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: 04/24/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
The swift emergence and propagation of multidrug-resistant (MDR) bacterial pathogens constitute a tremendous global health crisis. Among these pathogens, the challenge of antibiotic resistance in Gram-negative bacteria is particularly pressing due to their distinctive structure, such as highly impermeable outer membrane, overexpressed efflux pumps, and mutations. Several strategies have been documented to combat MDR Gram-negative bacteria, including the structural modification of existing antibiotics, the development of antimicrobial adjuvants, and research on novel targets that MDR bacteria are sensitive to. Drugs functioning as adjuvants to mitigate resistance to existing antibiotics may play a pivotal role in future antibacterial therapy strategies. In this review, we provide a brief overview of potential antibacterial adjuvants against Gram-negative bacteria and their mechanisms of action, and discuss the application prospects and potential for bacterial resistance to these adjuvants, along with strategies to reduce this risk.
Collapse
Affiliation(s)
- Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Zhen Tao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Motan Zhou
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Huilin Jiang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Liudi Wang
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Bingjie Ji
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
24
|
Yildirim O, Barman D, Chung M, Stone S, Geißen R, Boby ML, Sherborne BS, Tan DS. Design and synthesis of a library of C8-substituted sulfamidoadenosines to probe bacterial permeability. Bioorg Med Chem Lett 2024; 110:129844. [PMID: 38851357 PMCID: PMC11361631 DOI: 10.1016/j.bmcl.2024.129844] [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: 04/24/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Gram-negative bacteria pose a major challenge in antibiotic drug discovery because their cell envelope presents a permeability barrier that affords high intrinsic resistance to small-molecule drugs. The identification of correlations between chemical structure and Gram-negative permeability would thus enable development of predictive tools to facilitate antibiotic discovery. Toward this end, have advanced a library design paradigm in which various chemical scaffolds are functionalized at different regioisomeric positions using a uniform reagent set. This design enables decoupling of scaffold, regiochemistry, and substituent effects upon Gram-negative permeability of these molecules. Building upon our recent synthesis of a library of C2-substituted sulfamidoadenosines, we have now developed an efficient synthetic route to an analogous library of regioisomeric C8-substituted congeners. The C8 library samples a region of antibiotic-relevant chemical space that is similar to that addressed by the C2 library, but distinct from that sampled by a library of analogously substituted oxazolidinones. Selected molecules were tested for accumulation in Escherichia coli in a pilot analysis, setting the stage for full comparative evaluation of these libraries in the future.
Collapse
Affiliation(s)
- Okan Yildirim
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Dipti Barman
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Mia Chung
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Samantha Stone
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Raphael Geißen
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Doctoral Program, Faculty of Biology, Albert-Ludwigs-Universität Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Melissa L Boby
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Pharmacology Graduate Program, Weill Cornell Graduate School, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | - Derek S Tan
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Pharmacology Graduate Program, Weill Cornell Graduate School, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Tri-Institutional Research Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
25
|
Skudlarek JW, Cooke AJ, Mitchell HJ, Babaoglu K, Shaw AW, Tong L, Nomland AB, Labroli M, Sha D, Mulhearn JJ, Wu C, Li SW, Beshore DC, Hughes JME, Jouffroy M, Wang H, Balibar CJ, Painter RE, Shen P, Lange HS, Ishchenko A, Chen YT, Klein DJ, Tracy RW, Miller RR, Cabalu TD, Wu Z, Leithead A, Scapin G, Hruza AW, Dzhekieva L, Bukhtiyarova M, Homsher MF, Xu M, Bahnck-Teets C, McKenney D, Buevich AV, Liu J, Zhang LK, Meng T, Kelly T, DiNunzio E, Soisson S, Cheng RKY, Hennig M, Raheem I, Walker SS. Cerastecin Inhibition of the Lipooligosaccharide Transporter MsbA to Combat Acinetobacter baumannii: From Screening Impurity to In Vivo Efficacy. J Med Chem 2024; 67:15620-15675. [PMID: 39172133 DOI: 10.1021/acs.jmedchem.4c01277] [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: 08/23/2024]
Abstract
Acinetobacter baumannii, a commonly multidrug-resistant Gram-negative bacterium responsible for large numbers of bloodstream and lung infections worldwide, is increasingly difficult to treat and constitutes a growing threat to human health. Structurally novel antibacterial chemical matter that can evade existing resistance mechanisms is essential for addressing this critical medical need. Herein, we describe our efforts to inhibit the essential A. baumannii lipooligosaccharide (LOS) ATP-binding cassette (ABC) transporter MsbA. An unexpected impurity from a phenotypic screening was optimized as a series of dimeric compounds, culminating with 1 (cerastecin D), which exhibited antibacterial activity in the presence of human serum and a pharmacokinetic profile sufficient to achieve efficacy against A. baumannii in murine septicemia and lung infection models.
Collapse
Affiliation(s)
| | - Andrew J Cooke
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Kerim Babaoglu
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Anthony W Shaw
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ling Tong
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Marc Labroli
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Deyou Sha
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Chengwei Wu
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Sarah W Li
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | | | | | - Hao Wang
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Carl J Balibar
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Pamela Shen
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Henry S Lange
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Yun-Ting Chen
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel J Klein
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Rodger W Tracy
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Randy R Miller
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Zhe Wu
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | | | - Alan W Hruza
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | | | | | - Min Xu
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - David McKenney
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Jian Liu
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Li-Kang Zhang
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Tao Meng
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Terri Kelly
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | | | | | | | - Izzat Raheem
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Scott S Walker
- Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
26
|
Schum D, Elsen FAV, Ruddell S, Schorpp K, Junca H, Müsken M, Chen SY, Fiedler MK, Pickl T, Pieper DH, Hadian K, Zacharias M, Sieber SA. Screening Privileged Alkyl Guanidinium Motifs under Host-Mimicking Conditions Reveals a Novel Antibiotic with an Unconventional Mode of Action. JACS AU 2024; 4:3125-3134. [PMID: 39211621 PMCID: PMC11350587 DOI: 10.1021/jacsau.4c00449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 09/04/2024]
Abstract
Screening large molecule libraries against pathogenic bacteria is often challenged by a low hit rate due to limited uptake, underrepresentation of antibiotic structural motifs, and assays that do not resemble the infection conditions. To address these limitations, we present a screen of a focused library of alkyl guanidinium compounds, a structural motif associated with antibiotic activity and enhanced uptake, under host-mimicking infection conditions against a panel of disease-associated bacteria. Several hit molecules were identified with activities against Gram-positive and Gram-negative bacteria, highlighting the fidelity of the general concept. We selected one compound (L15) for in-depth mode of action studies that exhibited bactericidal activity against methicillin-resistant Staphylococcus aureus USA300 with a minimum inhibitory concentration of 1.5 μM. Structure-activity relationship studies confirmed the necessity of the guanidinium motif for antibiotic activity. The mode of action was investigated using affinity-based protein profiling with an L15 probe and identified the signal peptidase IB (SpsB) as the most promising hit. Validation by activity assays, binding site identification, docking, and molecular dynamics simulations demonstrated SpsB activation by L15, a recently described mechanism leading to the dysregulation of protein secretion and cell death. Overall, this study highlights the need for unconventional screening strategies to identify novel antibiotics.
Collapse
Affiliation(s)
- Dominik Schum
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Franziska A. V. Elsen
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Stuart Ruddell
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Kenji Schorpp
- Helmholtz
Zentrum München, Research Unit Signaling and Translation, Ingolstädter Landstraße
1, Neuherberg, Munich 85764, Germany
| | - Howard Junca
- Helmholtz
Centre for Infection Research, Microbial Interactions and Processes, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz
Centre for Infection Research, Central Facility for Microscopy, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Shu-Yu Chen
- TUM
School of Natural Sciences, Department of Bioscience, Theoretical Biophysics (T38), Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Michaela K. Fiedler
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Thomas Pickl
- TUM School
of Natural Sciences, Department of Chemistry, Catalysis Research Center (CRC), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 1, Garching 85748, Germany
| | - Dietmar H. Pieper
- Helmholtz
Centre for Infection Research, Microbial Interactions and Processes, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Kamyar Hadian
- Helmholtz
Zentrum München, Research Unit Signaling and Translation, Ingolstädter Landstraße
1, Neuherberg, Munich 85764, Germany
| | - Martin Zacharias
- TUM
School of Natural Sciences, Department of Bioscience, Theoretical Biophysics (T38), Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| | - Stephan A. Sieber
- TUM
School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II, Center for Functional Protein Assemblies
(CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer Str. 8, Garching 85748, Germany
| |
Collapse
|
27
|
Bellucci MC, Romani C, Sani M, Volonterio A. Dual Antibiotic Approach: Synthesis and Antibacterial Activity of Antibiotic-Antimicrobial Peptide Conjugates. Antibiotics (Basel) 2024; 13:783. [PMID: 39200083 PMCID: PMC11352213 DOI: 10.3390/antibiotics13080783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
In recent years, bacterial resistance to conventional antibiotics has become a major concern in the medical field. The global misuse of antibiotics in clinics, personal use, and agriculture has accelerated this resistance, making infections increasingly difficult to treat and rendering new antibiotics ineffective more quickly. Finding new antibiotics is challenging due to the complexity of bacterial mechanisms, high costs and low financial incentives for the development of new molecular scaffolds, and stringent regulatory requirements. Additionally, innovation has slowed, with many new antibiotics being modifications of existing drugs rather than entirely new classes. Antimicrobial peptides (AMPs) are a valid alternative to small-molecule antibiotics offering several advantages, including broad-spectrum activity and a lower likelihood of inducing resistance due to their multifaceted mechanisms of action. However, AMPs face challenges such as stability issues in physiological conditions, potential toxicity to human cells, high production costs, and difficulties in large-scale manufacturing. A reliable strategy to overcome the drawbacks associated with the use of small-molecule antibiotics and AMPs is combination therapy, namely the simultaneous co-administration of two or more antibiotics or the synthesis of covalently linked conjugates. This review aims to provide a comprehensive overview of the literature on the development of antibiotic-AMP conjugates, with a particular emphasis on critically analyzing the design and synthetic strategies employed in their creation. In addition to the synthesis, the review will also explore the reported antibacterial activity of these conjugates and, where available, examine any data concerning their cytotoxicity.
Collapse
Affiliation(s)
- Maria Cristina Bellucci
- Department of Food, Environmental, and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20131 Milano, Italy;
| | - Carola Romani
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| | - Monica Sani
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimica “G. Natta” (SCITEC), Via Mario Bianco 9, 20131 Milano, Italy;
| | - Alessandro Volonterio
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| |
Collapse
|
28
|
Li H, Zhu X, Zhang X, Dong C. Caspofungin enhances the potency of rifampin against Gram-negative bacteria. Front Microbiol 2024; 15:1447485. [PMID: 39211315 PMCID: PMC11358092 DOI: 10.3389/fmicb.2024.1447485] [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: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Developing antibiotic adjuvants is an effective strategy to combat antimicrobial resistance (AMR). The envelope of Gram-negative bacteria (GNB) is a barrier to prevent the entry of antibiotics, making it an attractive target for novel antibiotic and adjuvant development. Methods and Results In this study, we identified Caspofungin acetate (CAS) as an antibiotic adjuvant against GNB in the repurposing screen of 3,158 FDA-approved drugs. Checkerboard assays suggested that CAS could enhance the antimicrobial activity of rifampin or colistin against various GNB strains in vitro, Moreover, Galleria mellonella larvae infection model also indicated that CAS significantly potentiated the efficacy of rifampin against multidrug-resistant Escherichia coli 72 strain in vivo. Most importantly, resistance development assay showed that CAS was less susceptible to accelerating the resistance development of drug-sensitive strain E. coli MG1655. Functional studies and RNA-seq analysis confirmed that the mechanisms by which CAS enhanced the antimicrobial activities of antibiotics were involved in permeabilizing the bacterial cell envelope, disrupting proton motive force and inhibiting bacterial biofilm formation. Additionally, it has been found that PgaC is the CAS target and enzymatic assay has confirmed the inhibition activity. Discussion Our results illustrate the feasibility of CAS as an antibiotic adjuvant against GNB, which is an alternative strategy of anti-infection.
Collapse
Affiliation(s)
- Haotian Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiaojing Zhu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xing Zhang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Changjiang Dong
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Lewis K, Lee RE, Brötz-Oesterhelt H, Hiller S, Rodnina MV, Schneider T, Weingarth M, Wohlgemuth I. Sophisticated natural products as antibiotics. Nature 2024; 632:39-49. [PMID: 39085542 PMCID: PMC11573432 DOI: 10.1038/s41586-024-07530-w] [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: 12/28/2022] [Accepted: 05/07/2024] [Indexed: 08/02/2024]
Abstract
In this Review, we explore natural product antibiotics that do more than simply inhibit an active site of an essential enzyme. We review these compounds to provide inspiration for the design of much-needed new antibacterial agents, and examine the complex mechanisms that have evolved to effectively target bacteria, including covalent binders, inhibitors of resistance, compounds that utilize self-promoted entry, those that evade resistance, prodrugs, target corrupters, inhibitors of 'undruggable' targets, compounds that form supramolecular complexes, and selective membrane-acting agents. These are exemplified by β-lactams that bind covalently to inhibit transpeptidases and β-lactamases, siderophore chimeras that hijack import mechanisms to smuggle antibiotics into the cell, compounds that are activated by bacterial enzymes to produce reactive molecules, and antibiotics such as aminoglycosides that corrupt, rather than merely inhibit, their targets. Some of these mechanisms are highly sophisticated, such as the preformed β-strands of darobactins that target the undruggable β-barrel chaperone BamA, or teixobactin, which binds to a precursor of peptidoglycan and then forms a supramolecular structure that damages the membrane, impeding the emergence of resistance. Many of the compounds exhibit more than one notable feature, such as resistance evasion and target corruption. Understanding the surprising complexity of the best antimicrobial compounds provides a roadmap for developing novel compounds to address the antimicrobial resistance crisis by mining for new natural products and inspiring us to design similarly sophisticated antibiotics.
Collapse
Affiliation(s)
- Kim Lewis
- Antimicrobial Discovery Center, Northeastern University, Boston, MA, USA.
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Tubingen, Germany
- Controlling Microbes to Fight Infection-Cluster of Excellence, Tubingen, Germany
| | | | - Marina V Rodnina
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Cologne-Bonn, Bonn, Germany
| | - Markus Weingarth
- Chemistry Department, Utrecht University, Utrecht, the Netherlands
| | - Ingo Wohlgemuth
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| |
Collapse
|
30
|
Parkhill SL, Johnson EO. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochem J 2024; 481:839-864. [PMID: 38958473 PMCID: PMC11346456 DOI: 10.1042/bcj20220062] [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: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The application of dyes to understanding the aetiology of infection inspired antimicrobial chemotherapy and the first wave of antibacterial drugs. The second wave of antibacterial drug discovery was driven by rapid discovery of natural products, now making up 69% of current antibacterial drugs. But now with the most prevalent natural products already discovered, ∼107 new soil-dwelling bacterial species must be screened to discover one new class of natural product. Therefore, instead of a third wave of antibacterial drug discovery, there is now a discovery bottleneck. Unlike natural products which are curated by billions of years of microbial antagonism, the vast synthetic chemical space still requires artificial curation through the therapeutics science of antibacterial drugs - a systematic understanding of how small molecules interact with bacterial physiology, effect desired phenotypes, and benefit the host. Bacterial molecular genetics can elucidate pathogen biology relevant to therapeutics development, but it can also be applied directly to understanding mechanisms and liabilities of new chemical agents with new mechanisms of action. Therefore, the next phase of antibacterial drug discovery could be enabled by integrating chemical expertise with systematic dissection of bacterial infection biology. Facing the ambitious endeavour to find new molecules from nature or new-to-nature which cure bacterial infections, the capabilities furnished by modern chemical biology and molecular genetics can be applied to prospecting for chemical modulators of new targets which circumvent prevalent resistance mechanisms.
Collapse
Affiliation(s)
- Susannah L. Parkhill
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
| | - Eachan O. Johnson
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
- Department of Chemistry, Imperial College, London, U.K
- Department of Chemistry, King's College London, London, U.K
| |
Collapse
|
31
|
MacNair CR, Rutherford ST, Tan MW. Alternative therapeutic strategies to treat antibiotic-resistant pathogens. Nat Rev Microbiol 2024; 22:262-275. [PMID: 38082064 DOI: 10.1038/s41579-023-00993-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 04/19/2024]
Abstract
Resistance threatens to render antibiotics - which are essential for modern medicine - ineffective, thus posing a threat to human health. The discovery of novel classes of antibiotics able to overcome resistance has been stalled for decades, with the developmental pipeline relying almost entirely on variations of existing chemical scaffolds. Unfortunately, this approach has been unable to keep pace with resistance evolution, necessitating new therapeutic strategies. In this Review, we highlight recent efforts to discover non-traditional antimicrobials, specifically describing the advantages and limitations of antimicrobial peptides and macrocycles, antibodies, bacteriophages and antisense oligonucleotides. These approaches have the potential to stem the tide of resistance by expanding the physicochemical property space and target spectrum occupied by currently approved antibiotics.
Collapse
Affiliation(s)
- Craig R MacNair
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Steven T Rutherford
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
32
|
Blasco B, Jang S, Terauchi H, Kobayashi N, Suzuki S, Akao Y, Ochida A, Morishita N, Takagi T, Nagamiya H, Suzuki Y, Watanabe T, Lee H, Lee S, Shum D, Cho A, Koh D, Park S, Lee H, Kim K, Ropponen HK, Augusto da Costa RM, Dunn S, Ghosh S, Sjö P, Piddock LJV. High-throughput screening of small-molecules libraries identified antibacterials against clinically relevant multidrug-resistant A. baumannii and K. pneumoniae. EBioMedicine 2024; 102:105073. [PMID: 38520916 PMCID: PMC10963893 DOI: 10.1016/j.ebiom.2024.105073] [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: 12/11/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The current pipeline for new antibiotics fails to fully address the significant threat posed by drug-resistant Gram-negative bacteria that have been identified by the World Health Organization (WHO) as a global health priority. New antibacterials acting through novel mechanisms of action are urgently needed. We aimed to identify new chemical entities (NCEs) with activity against Klebsiella pneumoniae and Acinetobacter baumannii that could be developed into a new treatment for drug-resistant infections. METHODS We developed a high-throughput phenotypic screen and selection cascade for generation of hit compounds active against multidrug-resistant (MDR) strains of K. pneumoniae and A. baumannii. We screened compound libraries selected from the proprietary collections of three pharmaceutical companies that had exited antibacterial drug discovery but continued to accumulate new compounds to their collection. Compounds from two out of three libraries were selected using "eNTRy rules" criteria associated with increased likelihood of intracellular accumulation in Escherichia coli. FINDINGS We identified 72 compounds with confirmed activity against K. pneumoniae and/or drug-resistant A. baumannii. Two new chemical series with activity against XDR A. baumannii were identified meeting our criteria of potency (EC50 ≤50 μM) and absence of cytotoxicity (HepG2 CC50 ≥100 μM and red blood cell lysis HC50 ≥100 μM). The activity of close analogues of the two chemical series was also determined against A. baumannii clinical isolates. INTERPRETATION This work provides proof of principle for the screening strategy developed to identify NCEs with antibacterial activity against multidrug-resistant critical priority pathogens such as K. pneumoniae and A. baumannii. The screening and hit selection cascade established here provide an excellent foundation for further screening of new compound libraries to identify high quality starting points for new antibacterial lead generation projects. FUNDING BMBF and GARDP.
Collapse
Affiliation(s)
- Benjamin Blasco
- Global Antibiotic Research and Development Partnership (GARDP), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | - Soojin Jang
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Hiroki Terauchi
- Eisai Co., Ltd., Tsukuba Research Laboratories, 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Naoki Kobayashi
- Eisai Co., Ltd., Tsukuba Research Laboratories, 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Shuichi Suzuki
- Eisai Co., Ltd., Tsukuba Research Laboratories, 5-1-3 Tokodai, Tsukuba, Ibaraki, 300-2635, Japan
| | - Yuichiro Akao
- Takeda Pharmaceutical Company Ltd, 261, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Atsuko Ochida
- Takeda Pharmaceutical Company Ltd, 261, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Nao Morishita
- Takeda Pharmaceutical Company Ltd, 261, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Terufumi Takagi
- Takeda Pharmaceutical Company Ltd, 261, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Hiroyuki Nagamiya
- Takeda Pharmaceutical Company Ltd, 261, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yamato Suzuki
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Toshiaki Watanabe
- Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Hyunjung Lee
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Sol Lee
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - David Shum
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Ahreum Cho
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Dahae Koh
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Soonju Park
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Honggun Lee
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Kideok Kim
- Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Henni-Karoliina Ropponen
- Global Antibiotic Research and Development Partnership (GARDP), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | | | | | - Sunil Ghosh
- TCG Lifesciences Private Limited, Block BN, Plot 7, Salt Lake Electronics Complex, Sector V, Kolkata, 700091, West Bengal, India
| | - Peter Sjö
- Drugs for Neglected Diseases Initiative, 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | - Laura J V Piddock
- Global Antibiotic Research and Development Partnership (GARDP), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland.
| |
Collapse
|
33
|
Gurvic D, Zachariae U. Multidrug efflux in Gram-negative bacteria: structural modifications in active compounds leading to efflux pump avoidance. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:6. [PMID: 39843816 PMCID: PMC11721645 DOI: 10.1038/s44259-024-00023-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2025]
Abstract
Gram-negative bacteria cause the majority of critically drug-resistant infections, necessitating the rapid development of new drugs with Gram-negative activity. However, drug design is hampered by the low permeability of the Gram-negative cell envelope and the function of drug efflux pumps, which extrude foreign molecules from the cell. A better understanding of the molecular determinants of compound recognition by efflux pumps is, therefore, essential. Here, we quantitatively analysed the activity of 73,737 compounds, recorded in the publicly accessible database CO-ADD, across three strains of E. coli - the wild-type, the efflux-deficient tolC variant, and the hyper-permeable lpxC variant, to elucidate the molecular principles of evading efflux pumps. We computationally investigated molecular features within this dataset that promote, or reduce, the propensity of being recognised by the TolC-dependent efflux systems in E. coli. Our results show that, alongside a range of physicochemical features, the presence or absence of specific chemical groups in the compounds substantially increases the probability of avoiding efflux. A comparison of our findings with inward permeability data further underscores the primary role of efflux in determining drug bioactivity in Gram-negative bacteria.
Collapse
Affiliation(s)
- Dominik Gurvic
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
- Biochemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
34
|
Dong S, Zhao Z, Tang H, Li G, Pan J, Gu X, Jiang J, Xiao L, Scapin G, Hunter DN, Yang D, Huang Y, Bennett F, Yang SW, Mandal M, Tang H, Su J, Tudge C, deJesus RK, Ding FX, Lombardo M, Hicks JD, Fischmann T, Mirza A, Dayananth P, Painter RE, Villafania A, Garlisi CG, Zhang R, Mayhood TW, Si Q, Li N, Amin RP, Bhatt B, Chen F, Regan CP, Regan H, Lin X, Wu J, Leithead A, Pollack SR, Scott JD, Nargund RP, Therien AG, Black T, Young K, Pasternak A. Structure Guided Discovery of Novel Pan Metallo-β-Lactamase Inhibitors with Improved Gram-Negative Bacterial Cell Penetration. J Med Chem 2024; 67:3400-3418. [PMID: 38387069 DOI: 10.1021/acs.jmedchem.3c01614] [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: 02/24/2024]
Abstract
The use of β-lactam (BL) and β-lactamase inhibitor combination to overcome BL antibiotic resistance has been validated through clinically approved drug products. However, unmet medical needs still exist for the treatment of infections caused by Gram-negative (GN) bacteria expressing metallo-β-lactamases. Previously, we reported our effort to discover pan inhibitors of three main families in this class: IMP, VIM, and NDM. Herein, we describe our work to improve the GN coverage spectrum in combination with imipenem and relebactam. This was achieved through structure- and property-based optimization to tackle the GN cell penetration and efflux challenges. A significant discovery was made that inhibition of both VIM alleles, VIM-1 and VIM-2, is essential for broad GN coverage, especially against VIM-producing P. aeruginosa. In addition, pharmacokinetics and nonclinical safety profiles were investigated for select compounds. Key findings from this drug discovery campaign laid the foundation for further lead optimization toward identification of preclinical candidates.
Collapse
Affiliation(s)
- Shuzhi Dong
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Zhiqiang Zhao
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Haiqun Tang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Guoqing Li
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jianping Pan
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Xin Gu
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jinlong Jiang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Li Xiao
- Computational and Structural Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Giovanna Scapin
- Computational and Structural Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - David N Hunter
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Dexi Yang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Yuhua Huang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Frank Bennett
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Shu-Wei Yang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Mihirbaran Mandal
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Haifeng Tang
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jing Su
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Clare Tudge
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Fa-Xiang Ding
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Matthew Lombardo
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jacqueline D Hicks
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Thierry Fischmann
- Computational and Structural Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Asra Mirza
- Antibacterial/Antifungal, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Priya Dayananth
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Ronald E Painter
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Artjohn Villafania
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Charles G Garlisi
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rumin Zhang
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Todd W Mayhood
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Qian Si
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Nianyu Li
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Rupesh P Amin
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Bhavana Bhatt
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Feifei Chen
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Christopher P Regan
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Hillary Regan
- Nonclinical Drug Safety, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xinjie Lin
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jin Wu
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Andrew Leithead
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Scott R Pollack
- Discovery Process Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jack D Scott
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ravi P Nargund
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Alex G Therien
- Exploratory Science Center, Merck & Co., Inc., Cambridge, Massachusetts 02139, United States
| | - Todd Black
- Antibacterial/Antifungal, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Katherine Young
- Antibacterial/Antifungal, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Alexander Pasternak
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
35
|
Kuralt V, Frlan R. Navigating the Chemical Space of ENR Inhibitors: A Comprehensive Analysis. Antibiotics (Basel) 2024; 13:252. [PMID: 38534687 DOI: 10.3390/antibiotics13030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
Antimicrobial resistance is a global health threat that requires innovative strategies against drug-resistant bacteria. Our study focuses on enoyl-acyl carrier protein reductases (ENRs), in particular FabI, FabK, FabV, and InhA, as potential antimicrobial agents. Despite their promising potential, the lack of clinical approvals for inhibitors such as triclosan and isoniazid underscores the challenges in achieving preclinical success. In our study, we curated and analyzed a dataset of 1412 small molecules recognized as ENR inhibitors, investigating different structural variants. Using advanced cheminformatic tools, we mapped the physicochemical landscape and identified specific structural features as key determinants of bioactivity. Furthermore, we investigated whether the compounds conform to Lipinski rules, PAINS, and Brenk filters, which are crucial for the advancement of compounds in development pipelines. Furthermore, we investigated structural diversity using four different representations: Chemotype diversity, molecular similarity, t-SNE visualization, molecular complexity, and cluster analysis. By using advanced bioinformatics tools such as matched molecular pairs (MMP) analysis, machine learning, and SHAP analysis, we were able to improve our understanding of the activity cliques and the precise effects of the functional groups. In summary, this chemoinformatic investigation has unraveled the FAB inhibitors and provided insights into rational antimicrobial design, seamlessly integrating computation into the discovery of new antimicrobial agents.
Collapse
Affiliation(s)
- Vid Kuralt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Rok Frlan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
36
|
Bonacorsi A, Trespidi G, Scoffone VC, Irudal S, Barbieri G, Riabova O, Monakhova N, Makarov V, Buroni S. Characterization of the dispirotripiperazine derivative PDSTP as antibiotic adjuvant and antivirulence compound against Pseudomonas aeruginosa. Front Microbiol 2024; 15:1357708. [PMID: 38435690 PMCID: PMC10904629 DOI: 10.3389/fmicb.2024.1357708] [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: 12/18/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Abstract
Pseudomonas aeruginosa is a major human pathogen, able to establish difficult-to-treat infections in immunocompromised and people with cystic fibrosis (CF). The high rate of antibiotic treatment failure is due to its notorious drug resistance, often mediated by the formation of persistent biofilms. Alternative strategies, capable of overcoming P. aeruginosa resistance, include antivirulence compounds which impair bacterial pathogenesis without exerting a strong selective pressure, and the use of antimicrobial adjuvants that can resensitize drug-resistant bacteria to specific antibiotics. In this work, the dispirotripiperazine derivative PDSTP, already studied as antiviral, was characterized for its activity against P. aeruginosa adhesion to epithelial cells, its antibiotic adjuvant ability and its biofilm inhibitory potential. PDSTP was effective in impairing the adhesion of P. aeruginosa to various immortalized cell lines. Moreover, the combination of clinically relevant antibiotics with the compound led to a remarkable enhancement of the antibiotic efficacy towards multidrug-resistant CF clinical strains. PDSTP-ceftazidime combination maintained its efficacy in vivo in a Galleria mellonella infection model. Finally, the compound showed a promising biofilm inhibitory activity at low concentrations when tested both in vitro and using an ex vivo pig lung model. Altogether, these results validate PDSTP as a promising compound, combining the ability to decrease P. aeruginosa virulence by impairing its adhesion and biofilm formation, with the capability to increase antibiotic efficacy against antibiotic resistant strains.
Collapse
Affiliation(s)
- Andrea Bonacorsi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Gabriele Trespidi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Viola C. Scoffone
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Samuele Irudal
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Olga Riabova
- Research Center of Biotechnology RAS, Moscow, Russia
| | | | - Vadim Makarov
- Research Center of Biotechnology RAS, Moscow, Russia
| | - Silvia Buroni
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
37
|
Choi Y, Choe HW, Kook M, Choo S, Park TW, Bae S, Kim H, Yang J, Jeong WS, Yu J, Lee KR, Kim YS, Yu J. Proline-Hinged α-Helical Peptides Sensitize Gram-Positive Antibiotics, Expanding Their Physicochemical Properties to Be Used as Gram-Negative Antibiotics. J Med Chem 2024; 67:1825-1842. [PMID: 38124427 PMCID: PMC10860147 DOI: 10.1021/acs.jmedchem.3c01473] [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: 08/09/2023] [Revised: 11/13/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
The outer membrane (OM) of Gram-negative bacteria is the most difficult obstacle for small-molecule antibiotics to reach their targets in the cytosol. The molecular features of Gram-negative antibiotics required for passing through the OM are that they should be positively charged rather than neutral, flat rather than globular, less flexible, or more increased amphiphilic moment. Because of these specific molecular characteristics, developing Gram-negative antibiotics is difficult. We focused on sensitizer peptides to facilitate the passage of hydrophobic Gram-positive antibiotics through the OM. We explored ways of improving the sensitizing ability of proline-hinged α-helical peptides by adjusting their length, hydrophobicity, and N-terminal groups. A novel peptide, 1403, improves the potentiation of rifampicin in vitro and in vivo and potentiates most Gram-positive antibiotics. The "sensitizer" approach is more plausible than those that rely on conventional drug discovery methods concerning drug development costs and the development of drug resistance.
Collapse
Affiliation(s)
- Yoonhwa Choi
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
- CAMP
Therapeutics, Seoul 08826, Republic of Korea
| | - Hyeong Woon Choe
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Minsoo Kook
- Department
of Infectious Disease, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Seolah Choo
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Tae Woo Park
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Soeun Bae
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Heeseung Kim
- Department
of Infectious Disease, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jihye Yang
- Department
of Infectious Disease, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Woo-Seong Jeong
- Laboratory
Animal Resource Center, Korea Research Institute
of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jiyoung Yu
- Asan
Medical Center, Seoul 05505, Republic
of Korea
| | - Kyeong-Ryoon Lee
- Laboratory
Animal Resource Center, Korea Research Institute
of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Yang Soo Kim
- Department
of Infectious Disease, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jaehoon Yu
- Department
of Chemistry & Education, Seoul National
University, Seoul 08826, Republic
of Korea
- CAMP
Therapeutics, Seoul 08826, Republic of Korea
| |
Collapse
|
38
|
Shin E, Zhang Y, Zhou J, Lang Y, Sayed ARM, Werkman C, Jiao Y, Kumaraswamy M, Bulman ZP, Luna BM, Bulitta JB. Improved characterization of aminoglycoside penetration into human lung epithelial lining fluid via population pharmacokinetics. Antimicrob Agents Chemother 2024; 68:e0139323. [PMID: 38169309 PMCID: PMC10848756 DOI: 10.1128/aac.01393-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Aminoglycosides are important treatment options for serious lung infections, but modeling analyses to quantify their human lung epithelial lining fluid (ELF) penetration are lacking. We estimated the extent and rate of penetration for five aminoglycosides via population pharmacokinetics from eight published studies. The area under the curve in ELF vs plasma ranged from 50% to 100% and equilibration half-lives from 0.61 to 5.80 h, indicating extensive system hysteresis. Aminoglycoside ELF peak concentrations were blunted, but overall exposures were moderately high.
Collapse
Affiliation(s)
- Eunjeong Shin
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Yongzhen Zhang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jieqiang Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Yinzhi Lang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Alaa R. M. Sayed
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Carolin Werkman
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Monika Kumaraswamy
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, California, USA
- Infectious Diseases Section, VA San Diego Healthcare System, San Diego, California, USA
| | - Zackery P. Bulman
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois, USA
| | - Brian M. Luna
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jürgen B. Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| |
Collapse
|
39
|
Wong F, Zheng EJ, Valeri JA, Donghia NM, Anahtar MN, Omori S, Li A, Cubillos-Ruiz A, Krishnan A, Jin W, Manson AL, Friedrichs J, Helbig R, Hajian B, Fiejtek DK, Wagner FF, Soutter HH, Earl AM, Stokes JM, Renner LD, Collins JJ. Discovery of a structural class of antibiotics with explainable deep learning. Nature 2024; 626:177-185. [PMID: 38123686 PMCID: PMC10866013 DOI: 10.1038/s41586-023-06887-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
The discovery of novel structural classes of antibiotics is urgently needed to address the ongoing antibiotic resistance crisis1-9. Deep learning approaches have aided in exploring chemical spaces1,10-15; these typically use black box models and do not provide chemical insights. Here we reasoned that the chemical substructures associated with antibiotic activity learned by neural network models can be identified and used to predict structural classes of antibiotics. We tested this hypothesis by developing an explainable, substructure-based approach for the efficient, deep learning-guided exploration of chemical spaces. We determined the antibiotic activities and human cell cytotoxicity profiles of 39,312 compounds and applied ensembles of graph neural networks to predict antibiotic activity and cytotoxicity for 12,076,365 compounds. Using explainable graph algorithms, we identified substructure-based rationales for compounds with high predicted antibiotic activity and low predicted cytotoxicity. We empirically tested 283 compounds and found that compounds exhibiting antibiotic activity against Staphylococcus aureus were enriched in putative structural classes arising from rationales. Of these structural classes of compounds, one is selective against methicillin-resistant S. aureus (MRSA) and vancomycin-resistant enterococci, evades substantial resistance, and reduces bacterial titres in mouse models of MRSA skin and systemic thigh infection. Our approach enables the deep learning-guided discovery of structural classes of antibiotics and demonstrates that machine learning models in drug discovery can be explainable, providing insights into the chemical substructures that underlie selective antibiotic activity.
Collapse
Affiliation(s)
- Felix Wong
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Integrated Biosciences, San Carlos, CA, USA
| | - Erica J Zheng
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Chemical Biology, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jacqueline A Valeri
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina M Donghia
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Melis N Anahtar
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Satotaka Omori
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Integrated Biosciences, San Carlos, CA, USA
| | - Alicia Li
- Integrated Biosciences, San Carlos, CA, USA
| | - Andres Cubillos-Ruiz
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Aarti Krishnan
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wengong Jin
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Abigail L Manson
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jens Friedrichs
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, Dresden, Germany
| | - Ralf Helbig
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, Dresden, Germany
| | - Behnoush Hajian
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dawid K Fiejtek
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Florence F Wagner
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Holly H Soutter
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan M Stokes
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research and David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Lars D Renner
- Leibniz Institute of Polymer Research and the Max Bergmann Center of Biomaterials, Dresden, Germany
| | - James J Collins
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
40
|
Stoorza AM, Duerfeldt AS. Guiding the Way: Traditional Medicinal Chemistry Inspiration for Rational Gram-Negative Drug Design. J Med Chem 2024; 67:65-80. [PMID: 38134355 PMCID: PMC11342810 DOI: 10.1021/acs.jmedchem.3c01831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
The discovery and development of small-molecule therapeutics effective against Gram-negative pathogens are highly challenging tasks. Most compounds that are active in biochemical settings fail to exhibit whole-cell activity. The major reason for this lack of activity is the effectiveness of bacterial cell envelopes as permeability barriers. These barriers originate from the nutrient-selective outer membranes, which act synergistically with polyspecific efflux pumps. Guiding principles to enable rational optimization of small molecules for efficient penetration and intracellular accumulation in Gram-negative bacteria would have a transformative impact on the discovery and design of chemical probes and therapeutics. In this Perspective, we draw on inspiration from traditional medicinal chemistry approaches for eukaryotic drug design to present a broader call for action in developing comparable approaches for Gram-negative bacteria.
Collapse
Affiliation(s)
- Alexis M Stoorza
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55414, United States
| | - Adam S Duerfeldt
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55414, United States
| |
Collapse
|
41
|
Ferrando N, Pino-Otín MR, Ballestero D, Lorca G, Terrado EM, Langa E. Enhancing Commercial Antibiotics with Trans-Cinnamaldehyde in Gram-Positive and Gram-Negative Bacteria: An In Vitro Approach. PLANTS (BASEL, SWITZERLAND) 2024; 13:192. [PMID: 38256746 PMCID: PMC10820649 DOI: 10.3390/plants13020192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024]
Abstract
One strategy to mitigate the emergence of bacterial resistance involves reducing antibiotic doses by combining them with natural products, such as trans-cinnamaldehyde (CIN). The objective of this research was to identify in vitro combinations (CIN + commercial antibiotic (ABX)) that decrease the minimum inhibitory concentration (MIC) of seven antibiotics against 14 different Gram-positive and Gram-negative pathogenic bacteria, most of them classified as ESKAPE. MIC values were measured for all compounds using the broth microdilution method. The effect of the combinations on these microorganisms was analyzed through the checkboard assay to determine the type of activity (synergy, antagonism, or addition). This analysis was complemented with a kinetic study of the synergistic combinations. Fifteen synergistic combinations were characterized for nine of the tested bacteria. CIN demonstrated effectiveness in reducing the MIC of chloramphenicol, streptomycin, amoxicillin, and erythromycin (94-98%) when tested on Serratia marcescens, Staphylococcus aureus, Pasteurella aerogenes, and Salmonella enterica, respectively. The kinetic study revealed that when the substances were tested alone at the MIC concentration observed in the synergistic combination, bacterial growth was not inhibited. However, when CIN and the ABX, for which synergy was observed, were tested simultaneously in combination at these same concentrations, the bacterial growth inhibition was complete. This demonstrates the highly potent in vitro synergistic activity of CIN when combined with commercial ABXs. This finding could be particularly beneficial in livestock farming, as this sector witnesses the highest quantities of antimicrobial usage, contributing significantly to antimicrobial resistance issues. Further research focused on this natural compound is thus warranted for this reason.
Collapse
Affiliation(s)
- Natalia Ferrando
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Autovía A-23 Zaragoza-Huesca, km. 510, 50830 Villanueva de Gállego, Spain; (N.F.); (M.R.P.-O.); (D.B.); (G.L.)
| | - María Rosa Pino-Otín
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Autovía A-23 Zaragoza-Huesca, km. 510, 50830 Villanueva de Gállego, Spain; (N.F.); (M.R.P.-O.); (D.B.); (G.L.)
| | - Diego Ballestero
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Autovía A-23 Zaragoza-Huesca, km. 510, 50830 Villanueva de Gállego, Spain; (N.F.); (M.R.P.-O.); (D.B.); (G.L.)
| | - Guillermo Lorca
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Autovía A-23 Zaragoza-Huesca, km. 510, 50830 Villanueva de Gállego, Spain; (N.F.); (M.R.P.-O.); (D.B.); (G.L.)
| | - Eva María Terrado
- Departamento de Didácticas Específicas, Facultad de Educación, Universisad de Zaragoza, Calle Pedro Cerbuna 12, 50009 Zaragoza, Spain;
| | - Elisa Langa
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Autovía A-23 Zaragoza-Huesca, km. 510, 50830 Villanueva de Gállego, Spain; (N.F.); (M.R.P.-O.); (D.B.); (G.L.)
| |
Collapse
|
42
|
Dos Santos Bezerra WA, Tavares CP, Lima VAS, da Rocha CQ, da Silva Vaz Junior I, Michels PAM, Costa Junior LM, Dos Santos Soares AM. In silico and In vitro Assessment of Dimeric Flavonoids (Brachydins) on Rhipicephalus microplus Glutathione S-transferase. Med Chem 2024; 20:912-919. [PMID: 38847259 DOI: 10.2174/0115734064298481240517072216] [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: 12/14/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Rhipicephalus microplus, an important cattle ectoparasite, is responsible for a substantial negative impact on the economy due to productivity loss. The emergence of resistance to widely used commercial acaricides has sparked efforts to explore alternative products for tick control. METHODS To address this challenge, innovative solutions targeting essential tick enzymes, like glutathione S-transferase (GST), have gained attention. Dimeric flavonoids, particularly brachydins (BRAs), have demonstrated various biological activities, including antiparasitic effects. The objectives of this study were to isolate four dimeric flavonoids from Fridericia platyphylla roots and to evaluate their potential as inhibitors of R. microplus GST. RESULTS In vitro assays confirmed the inhibition of R. microplus GST by BRA-G, BRA-I, BRA-J, and BRA-K with IC50 values of 0.075, 0.079, 0.075, and 0.058 mg/mL, respectively, with minimal hemolytic effects. Molecular docking of BRA-G, BRA-I, BRA-J, and BRA-K in a threedimensional model of R. microplus GST revealed predicted interactions with MolDock Scores of - 142.537, -126.831, -108.571, and -123.041, respectively. Both in silico and in vitro analyses show that brachydins are potential inhibitors of R. microplus GST. CONCLUSION The findings of this study deepen our understanding of GST inhibition in ticks, affirming its viability as a drug target. This knowledge contributes to the advancement of treatment modalities and strategies for improved tick control.
Collapse
Affiliation(s)
- Wallyson André Dos Santos Bezerra
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia - Bionorte, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Caio Pavão Tavares
- Laboratório de Controle de Parasitos, Departamento de Patologia, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Victor Antônio Silva Lima
- Laboratório de Química de Produtos Naturais, Departamento de Química, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Cláudia Quintino da Rocha
- Laboratório de Química de Produtos Naturais, Departamento de Química, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Itabajara da Silva Vaz Junior
- Faculdade de Veterinária, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Paul A M Michels
- School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Livio Martins Costa Junior
- Laboratório de Química de Produtos Naturais, Departamento de Química, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | - Alexandra Martins Dos Santos Soares
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia da Amazônia - Bionorte, Universidade Federal do Maranhão, São Luís, MA, Brazil
- Laboratório de Bioquímica Vegetal, Departamento de Engenharia Química, Universidade Federal do Maranhão, São Luís, MA, Brazil
| |
Collapse
|
43
|
Li C, Ye G, Jiang Y, Wang Z, Yu H, Yang M. Artificial Intelligence in battling infectious diseases: A transformative role. J Med Virol 2024; 96:e29355. [PMID: 38179882 DOI: 10.1002/jmv.29355] [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: 10/16/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024]
Abstract
It is widely acknowledged that infectious diseases have wrought immense havoc on human society, being regarded as adversaries from which humanity cannot elude. In recent years, the advancement of Artificial Intelligence (AI) technology has ushered in a revolutionary era in the realm of infectious disease prevention and control. This evolution encompasses early warning of outbreaks, contact tracing, infection diagnosis, drug discovery, and the facilitation of drug design, alongside other facets of epidemic management. This article presents an overview of the utilization of AI systems in the field of infectious diseases, with a specific focus on their role during the COVID-19 pandemic. The article also highlights the contemporary challenges that AI confronts within this domain and posits strategies for their mitigation. There exists an imperative to further harness the potential applications of AI across multiple domains to augment its capacity in effectively addressing future disease outbreaks.
Collapse
Affiliation(s)
- Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Guoguo Ye
- Shenzhen Key Laboratory of Pathogen and Immunity, National Clinical Research Center for Infectious Disease, The Third People's Hospital of Shenzhen, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yinghan Jiang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Zhiming Wang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Haiyang Yu
- Hangzhou Yalla Information Technology Service Co., Ltd., Hangzhou, People's Republic of China
| | - Minghui Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People's Republic of China
| |
Collapse
|
44
|
Renard S, Versluys S, Taillier T, Dubarry N, Leroi-Geissler C, Rey A, Cornaire E, Sordello S, Carry JCB, Angouillant-Boniface O, Gouyon T, Thompson F, Lebourg G, Certal V, Balazs L, Arranz E, Doerflinger G, Bretin F, Gervat V, Brohan E, Kraft V, Boulenc X, Ducelier C, Bacqué E, Couturier C. Optimization of the Antibacterial Spectrum and the Developability Profile of the Novel-Class Natural Product Corramycin. J Med Chem 2023; 66:16869-16887. [PMID: 38088830 DOI: 10.1021/acs.jmedchem.3c01564] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Corramycin 1 is a novel zwitterionic antibacterial peptide isolated from a culture of the myxobacterium Corallococcus coralloides. Though Corramycin displayed a narrow spectrum and modest MICs against sensitive bacteria, its ADMET and physchem profile as well as its high tolerability in mice along with an outstanding in vivo efficacy in an Escherichia coli septicemia mouse model were promising and prompted us to embark on an optimization program aiming at enlarging the spectrum and at increasing the antibacterial activities by modulating membrane permeability. Scanning the peptidic moiety by the Ala-scan strategy followed by key stabilization and introduction of groups such as a primary amine or siderophore allowed us to enlarge the spectrum and increase the overall developability profile. The optimized Corramycin 28 showed an improved mouse IV PK and a broader spectrum with high potency against key Gram-negative bacteria that translated into excellent efficacy in several in vivo mouse infection models.
Collapse
Affiliation(s)
| | | | - Thomas Taillier
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | | | | | - Astrid Rey
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | - Emilie Cornaire
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | | | | | | | - Thierry Gouyon
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | | | - Gilles Lebourg
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Victor Certal
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Laszlo Balazs
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Esther Arranz
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | | | | | - Vincent Gervat
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Eric Brohan
- Sanofi, 13 Quai Jules Guesde, Vitry-sur-Seine 94403, France
| | - Volker Kraft
- Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt am Main 65926, Germany
| | | | - Cécile Ducelier
- Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin 91385, France
| | - Eric Bacqué
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| | - Cédric Couturier
- Evotec, 1541, Avenue Marcel Mérieux, Marcy L'Etoile 69280, France
| |
Collapse
|
45
|
Saxena D, Maitra R, Bormon R, Czekanska M, Meiers J, Titz A, Verma S, Chopra S. Tackling the outer membrane: facilitating compound entry into Gram-negative bacterial pathogens. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:17. [PMID: 39843585 PMCID: PMC11721184 DOI: 10.1038/s44259-023-00016-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/23/2023] [Indexed: 01/17/2025]
Abstract
Emerging resistance to all available antibiotics highlights the need to develop new antibiotics with novel mechanisms of action. Most of the currently used antibiotics target Gram-positive bacteria while Gram-negative bacteria easily bypass the action of most drug molecules because of their unique outer membrane. This additional layer acts as a potent barrier restricting the entry of compounds into the cell. In this scenario, several approaches have been elucidated to increase the accumulation of compounds into Gram-negative bacteria. This review includes a brief description of the physicochemical properties that can aid compounds to enter and accumulate in Gram-negative bacteria and covers different strategies to target or bypass the outer membrane-mediated barrier in Gram-negative bacterial pathogens.
Collapse
Affiliation(s)
- Deepanshi Saxena
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India
| | - Rahul Maitra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India
| | - Rakhi Bormon
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, UP, India
| | - Marta Czekanska
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Joscha Meiers
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123, Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123, Saarbrücken, Germany.
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany.
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Kanpur, 208016, UP, India.
- Center for Nanoscience, IIT Kanpur, Kanpur, 208016, UP, India.
| | - Sidharth Chopra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, 226031, UP, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
46
|
Koehler MFT, Chen YC, Chen Y, Chen Y, Crawford JJ, Durk MR, Garland K, Hanan EJ, Higuchi RI, Hu H, Ly CQ, Paraselli PG, Roberts TC, Schwarz JB, Smith PA, Yu Z, Heise CE. Lipid Tales: Optimizing Arylomycin Membrane Anchors. ACS Med Chem Lett 2023; 14:1524-1530. [PMID: 37974942 PMCID: PMC10641904 DOI: 10.1021/acsmedchemlett.3c00327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/22/2023] [Indexed: 11/19/2023] Open
Abstract
Multidrug-resistant bacteria are spreading at alarming rates, and despite extensive efforts, no new antibiotic class with activity against Gram-negative bacteria has been approved in over 50 years. LepB inhibitors (LepBi) based on the arylomycin class of natural products are a novel class of antibiotics and function by inhibiting the bacterial type I signal peptidase (SPase) in Gram-negative bacteria. One critical aspect of LepBi development involves optimization of the membrane-anchored lipophilic portion of the molecule. We therefore developed an approach that assesses the effect of this portion on the complicated equilibria of plasma protein binding, crossing the outer membrane of Gram-negative bacteria and anchoring in the bacterial inner membrane to facilitate SPase binding. Our findings provide important insights into the development of antibacterial agents where the target is associated with the inner membrane of Gram-negative bacteria.
Collapse
Affiliation(s)
- Michael F. T. Koehler
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | - Yi-Chen Chen
- Department
of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Yongsheng Chen
- Department
of Chemistry, WuXi AppTec, Shanghai 200131, China
| | - Yuan Chen
- Department
of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - James J. Crawford
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | - Matthew R. Durk
- Department
of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Keira Garland
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | - Emily J. Hanan
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | | | - Huiyong Hu
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | - Cuong Q. Ly
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | | | | | - Jacob B. Schwarz
- Department
of Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080, United States
| | - Peter A. Smith
- Department
of Infectious Disease, Genentech, Inc., South San Francisco, California 94080, United States
| | - Zhiyong Yu
- Department
of Chemistry, WuXi AppTec, Shanghai 200131, China
| | - Christopher E. Heise
- Department
of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| |
Collapse
|
47
|
Belousov MV, Kosolapova AO, Fayoud H, Sulatsky MI, Sulatskaya AI, Romanenko MN, Bobylev AG, Antonets KS, Nizhnikov AA. OmpC and OmpF Outer Membrane Proteins of Escherichia coli and Salmonella enterica Form Bona Fide Amyloids. Int J Mol Sci 2023; 24:15522. [PMID: 37958507 PMCID: PMC10649029 DOI: 10.3390/ijms242115522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Outer membrane proteins (Omps) of Gram-negative bacteria represent porins involved in a wide range of virulence- and pathogenesis-related cellular processes, including transport, adhesion, penetration, and the colonization of host tissues. Most outer membrane porins share a specific spatial structure called the β-barrel that provides their structural integrity within the membrane lipid bilayer. Recent data suggest that outer membrane proteins from several bacterial species are able to adopt the amyloid state alternative to their β-barrel structure. Amyloids are protein fibrils with a specific spatial structure called the cross-β that gives them an unusual resistance to different physicochemical influences. Various bacterial amyloids are known to be involved in host-pathogen and host-symbiont interactions and contribute to colonization of host tissues. Such an ability of outer membrane porins to adopt amyloid state might represent an important mechanism of bacterial virulence. In this work, we investigated the amyloid properties of the OmpC and OmpF porins from two species belonging to Enterobacteriaceae family, Escherichia coli, and Salmonella enterica. We demonstrated that OmpC and OmpF of E. coli and S. enterica form toxic fibrillar aggregates in vitro. These aggregates exhibit birefringence upon binding Congo Red dye and show characteristic reflections under X-ray diffraction. Thus, we confirmed amyloid properties for OmpC of E. coli and demonstrated bona fide amyloid properties for three novel proteins: OmpC of S. enterica and OmpF of E. coli and S. enterica in vitro. All four studied porins were shown to form amyloid fibrils at the surface of E. coli cells in the curli-dependent amyloid generator system. Moreover, we found that overexpression of recombinant OmpC and OmpF in the E. coli BL21 strain leads to the formation of detergent- and protease-resistant amyloid-like aggregates and enhances the birefringence of bacterial cultures stained with Congo Red. We also detected detergent- and protease-resistant aggregates comprising OmpC and OmpF in S. enterica culture. These data are important in the context of understanding the structural dualism of Omps and its relation to pathogenesis.
Collapse
Affiliation(s)
- Mikhail V. Belousov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anastasiia O. Kosolapova
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Haidar Fayoud
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Maksim I. Sulatsky
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (A.I.S.)
| | - Anna I. Sulatskaya
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (M.I.S.); (A.I.S.)
| | - Maria N. Romanenko
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alexander G. Bobylev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Kirill S. Antonets
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anton A. Nizhnikov
- All-Russia Research Institute for Agricultural Microbiology, 196608 St. Petersburg, Russia; (M.V.B.); (A.O.K.); (H.F.); (M.N.R.); (K.S.A.)
- Faculty of Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
48
|
Skrzyniarz K, Kuc-Ciepluch D, Lasak M, Arabski M, Sanchez-Nieves J, Ciepluch K. Dendritic systems for bacterial outer membrane disruption as a method of overcoming bacterial multidrug resistance. Biomater Sci 2023; 11:6421-6435. [PMID: 37605901 DOI: 10.1039/d3bm01255g] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
The alarming rise of multi-drug resistant microorganisms has increased the need for new approaches through the development of innovative agents that are capable of attaching to the outer layers of bacteria and causing permanent damage by penetrating the bacterial outer membrane. The permeability (disruption) of the outer membrane of Gram-negative bacteria is now considered to be one of the main ways to overcome multidrug resistance in bacteria. Natural and synthetic permeabilizers such as AMPs and dendritic systems seem promising. However, due to their advantages in terms of biocompatibility, antimicrobial capacity, and wide possibilities for modification and synthesis, highly branched polymers and dendritic systems have gained much more interest in recent years. Various forms of arrangement, and structure of the skeleton, give dendritic systems versatile applications, especially the possibility of attaching other ligands to their surface. This review will focus on the mechanisms used by different types of dendritic polymers, and their complexes with macromolecules to enhance their antimicrobial effect, and to permeabilize the bacterial outer membrane. In addition, future challenges and potential prospects are illustrated in the hope of accelerating the advancement of nanomedicine in the fight against resistant pathogens.
Collapse
Affiliation(s)
- Kinga Skrzyniarz
- Division of Medical Biology, Jan Kochanowski University, Kielce, Poland.
| | | | - Magdalena Lasak
- Division of Medical Biology, Jan Kochanowski University, Kielce, Poland.
| | - Michał Arabski
- Division of Medical Biology, Jan Kochanowski University, Kielce, Poland.
| | - Javier Sanchez-Nieves
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares, Madrid, Spain
- Instituto de Investigación Química "Andrés M. del Río" (IQAR, UAH), Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
- Institute for Health Research Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Karol Ciepluch
- Division of Medical Biology, Jan Kochanowski University, Kielce, Poland.
| |
Collapse
|
49
|
Ordaz G, Dagà U, Budia A, Pérez-Lanzac A, Fernández JM, Jordán C. Urinary pH and antibiotics, choose carefully. A systematic review. Actas Urol Esp 2023; 47:408-415. [PMID: 36754205 DOI: 10.1016/j.acuroe.2023.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 02/08/2023]
Abstract
Uncomplicated urinary tract infection (UTI) is the most common bacterial infection in women. Since 1948, the relationship between urinary pH and antibiotics (ABs) has been established. We aimed to search for the best urinary pH for each family of antibiotics and to assess whether pH changes bacterial susceptibility to them. We included in vitro research and in vivo studies including one or more bacterial species and tested the effect of one or more ABs at different pH values. We also included randomized controlled clinical trials (RCTs) in uncomplicated UTI (EAU guidelines 2019 definition), choosing the ABs based on urinary pH or using an antibiotic plus urinary pH modifiers (L-methionine, vitamin C…) vs. an antibiotic and a placebo. Quadas-2 tool was used as a quality assessment of the studies and PRISMA set of items for systematic reviews. Two authors independently screened and evaluated the papers, while two additional authors individually repeated the search. A fifth researcher acted as an arbiter, and another author collaborated as a hospital pharmaceutical consultant. Alkaline-friendly antibiotics are most fluoroquinolones, aminoglycosides, trimethoprim. Acidic-friendly antibiotics are fosfomycin, tetracycline, nitrofurantoin and some β-lactams. We suggest performing urine cultures with antibiogram tests, in both acidic and alkaline media, to define the bacterial susceptibility profile. There is insufficient in vivo evidence to support whether choosing an antibiotic based on a patient's urinary pH or adding urinary pH modifiers will lead to a higher cure rate.
Collapse
Affiliation(s)
- G Ordaz
- GEMA: Spanish Group of Urology of Meta-Analysis and Systematic Reviews, Spain; HUP: University and Polytechnic Hospital, La Fe, Valencia, Spain.
| | - U Dagà
- HUP: University and Polytechnic Hospital, La Fe, Valencia, Spain
| | - A Budia
- GEMA: Spanish Group of Urology of Meta-Analysis and Systematic Reviews, Spain; HUP: University and Polytechnic Hospital, La Fe, Valencia, Spain
| | - A Pérez-Lanzac
- GEMA: Spanish Group of Urology of Meta-Analysis and Systematic Reviews, Spain
| | - J M Fernández
- GEMA: Spanish Group of Urology of Meta-Analysis and Systematic Reviews, Spain
| | - C Jordán
- ICO: Catalan Institute of Oncology, Barcelona, Spain
| |
Collapse
|
50
|
Phan NKN, Huynh TKC, Nguyen HP, Le QT, Nguyen TCT, Ngo KKH, Nguyen THA, Ton KA, Thai KM, Hoang TKD. Exploration of Remarkably Potential Multitarget-Directed N-Alkylated-2-(substituted phenyl)-1 H-benzimidazole Derivatives as Antiproliferative, Antifungal, and Antibacterial Agents. ACS OMEGA 2023; 8:28733-28748. [PMID: 37576624 PMCID: PMC10413844 DOI: 10.1021/acsomega.3c03530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
Improving lipophilicity for drugs to penetrate the lipid membrane and decreasing bacterial and fungal coinfections for patients with cancer pose challenges in the drug development process. Here, a series of new N-alkylated-2-(substituted phenyl)-1H-benzimidazole derivatives were synthesized and characterized by 1H and 13C NMR, FTIR, and HRMS spectrum analyses to address these difficulties. All the compounds were evaluated for their antiproliferative, antibacterial, and antifungal activities. Results indicated that compound 2g exhibited the best antiproliferative activity against the MDA-MB-231 cell line and also displayed significant inhibition at minimal inhibitory concentration (MIC) values of 8, 4, and 4 μg mL-1 against Streptococcus faecalis, Staphylococcus aureus, and methicillin-resistant Staphylococcus aureus compared with amikacin. The antifungal data of compounds 1b, 1c, 2e, and 2g revealed their moderate activities toward Candida albicans and Aspergillus niger, with MIC values of 64 μg mL-1 for both strains. Finally, the molecular docking study found that 2g interacted with crucial amino acids in the binding site of complex dihydrofolate reductase with nicotinamide adenine dinucleotide phosphate.
Collapse
Affiliation(s)
- Ngoc-Kim-Ngan Phan
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Thi-Kim-Chi Huynh
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology, No.18, Hoang Quoc Viet Str., Cau Giay Dist., Hanoi City 100000, Vietnam
| | - Hoang-Phuc Nguyen
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Quoc-Tuan Le
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Thi-Cam-Thu Nguyen
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Kim-Khanh-Huy Ngo
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Thi-Hong-An Nguyen
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Khoa Anh Ton
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
| | - Khac-Minh Thai
- Department
of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, No.41-43, Dinh Tien Hoang Str.,
Dist. 1, Ho Chi Minh City 70000, Vietnam
| | - Thi-Kim-Dung Hoang
- Institute
of Chemical Technology, Vietnam Academy of Science and Technology, No.1A, TL29 Str., Thanh Loc Ward,
Dist. 12, Ho Chi Minh City 70000, Vietnam
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology, No.18, Hoang Quoc Viet Str., Cau Giay Dist., Hanoi City 100000, Vietnam
| |
Collapse
|