1
|
Qiu J, Qin R, Zhi S, Liu L. Recent advance in macrolactams: Structure, bioactivity, and biosynthesis. Bioorg Chem 2025; 159:108406. [PMID: 40184666 DOI: 10.1016/j.bioorg.2025.108406] [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: 10/01/2024] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
Macrolactams have garnered significant attention in recent years due to their diverse structures and remarkable biological activities. Despite the increasing number of new members being reported, a systematic discussion of recent advancements in this family is still lacking, particularly in areas such as structure-activity relationship and newly identified biosynthetic pathways that deviate from the traditional collinear rule. To address this gap, we compiled 105 macrolactams reported between 2004 and 2023, produced by microbial strains isolated from diverse environments, including marine sediments, soil, plants, and animals. This review not only highlights the sources, structures, and biological activities of these macrolactams but also delves into 17 known biosynthetic pathways. We provide an in-depth analysis of the associated biosynthetic gene clusters, the mechanisms of key enzymes, and their roles in the biosynthesis process. By offering these insights, this review serves as a valuable reference for the discovery of novel macrolactams and their sustainable production using synthetic biology approaches in the future.
Collapse
Affiliation(s)
- Jiawei Qiu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; College of Food Science and Engineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Ruochang Qin
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Shuai Zhi
- School of Public Health, Ningbo University, Ningbo, Zhejiang 315000, China
| | - Liwei Liu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
2
|
Prinster T, Harrison A, Dick C, Horvath DJ, Li B, Sievers G, Madamsetty R, Zhang J, Mason KM, Khoo C, Justice SS. Cranberry constituents prevent SOS-mediated filamentation of uropathogenic Escherichia coli. Infect Immun 2025:e0060024. [PMID: 40208062 DOI: 10.1128/iai.00600-24] [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: 02/20/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
The diameter, length, and shape of bacteria are maintained with such high fidelity that these parameters are classically used as metrics in the distinction of bacterial species. Increasing evidence indicates that bacteria transiently shift their shapes into distinctive morphologies in response to environmental changes. Elongation of bacterial length into a filamentous shape provides unique survival advantages for many bacterial species. Analysis of 42 clinical isolates of uropathogenic Escherichia coli (UPEC) revealed that filamentation to host-derived antimicrobials is a conserved phenotype. Therefore, we hypothesize that filamentation represents a conserved mechanism of pathogenic bacterial persistence that can be targeted for narrow-spectrum, anti-virulence therapies. We demonstrate that cranberries prevent SulA-mediated filamentation of UPEC. Furthermore, we identify multiple fractions of cranberries that retain anti-filamentation properties. These studies provide mechanistic insight into the clinical efficacy of cranberry for patients with recurrent urinary tract infections. Inhibition of filamentation represents a novel approach to promote bacterial pathogen susceptibility to immune and antibiotic-mediated clearance to attenuate disease.
Collapse
Affiliation(s)
- Tracy Prinster
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Alistair Harrison
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Christopher Dick
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Dennis J Horvath
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Birong Li
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Grace Sievers
- The College of Nursing, The Ohio State University, Columbus, Ohio, USA
| | | | - Jingwen Zhang
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Kevin M Mason
- The Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Christina Khoo
- Ocean Spray Cranberries, Inc, Lakeville, Massachusetts, USA
| | - Sheryl S Justice
- The College of Nursing, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
3
|
Goh MH, Rabiner RA, Connolly JJ, Lozano-Calderon SA, Chen AF. A Novel Isotropic Optical Fiber: Antimicrobial Effect of Blue Light on Drug Resistant Organisms. J Orthop Res 2025; 43:881-888. [PMID: 39775972 DOI: 10.1002/jor.26042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025]
Abstract
Drug-resistant organisms (DROs) necessitate the development of new therapies. Antimicrobial blue light (ABL) is a promising option, utilizing photoexcitation of endogenous bacterial components to generate reactive oxygen species, leading to bacterial death. The aim of this study is to investigate the effects of a novel isotropic optical fiber under in-vitro conditions on multidrug-resistant gram-negative Pseudomonas aeruginosa (MDR-Pa) and methicillin-resistant Staphylococcus aureus (MRSA). Time-to-kill assays were conducted in tubes containing 10 mL of 0.9% NaCl solution with an inoculum of 1 × 10⁵ CFU/mL for MDR-Pa or MRSA. The experiments were repeated at least three times per strain. Experimental tubes had either one (low power, LP) or two (high power, HP) optical fibers delivering five ABL wavelengths (405, 415, 435, 450, and 475 nm) over 60 min. Control tubes lacked optical fibers. Samples were taken at 0, 10, 20, 30, and 60 min, streaked on agar, and incubated to determine CFU/mL. Bactericidal reduction was defined as a ≥ 99.9% (≥ 3 log10) reduction in CFU/mL. One-way ANOVA were conducted. The novel isotropic optical fiber was able to exhibit bactericidal effects for MDR-Pa only under HP-ABL with a log10CFU/mL ± SD difference of -3.71 ± 0.01 at 60 min (p = 0.03). Conversely, the optical fiber exhibited bactericidal effects on MRSA under both LP-ABL and HP-ABL with a log10CFU/mL±SD difference of -3.73 ± 0.08 at 60 min (p = 0.03) and -3.07 ± 0.28 at 20 min (p = 0.02), respectively. The isotropic optical fiber demonstrated bactericidal effects on MRSA and MDR-Pa in in-vitro studies and shows potential as a therapeutic option for DROs.
Collapse
Affiliation(s)
- Megan H Goh
- Department of Orthopaedic Surgery, Division of Orthopaedic Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Joseph J Connolly
- Department of Orthopaedic Surgery, Division of Orthopaedic Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Santiago A Lozano-Calderon
- Department of Orthopaedic Surgery, Division of Orthopaedic Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Antonia F Chen
- Department of Orthopaedic Surgery, Division of Arthroplasty and Joint Reconstruction, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Muir BW, Payne JAE, Martin JH, O' Shea R, Rajesh S, Blackman LD, Shen HH, Heazlewood C, Bansal V, Morgan B. An Australian perspective on clinical, economic and regulatory considerations in emerging nanoparticle therapies for infections. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:9. [PMID: 39966608 PMCID: PMC11836273 DOI: 10.1038/s44259-024-00070-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/28/2024] [Indexed: 02/20/2025]
Abstract
Antimicrobial resistance (AMR) poses a growing global health threat. Nanomedicine, combined with drug repurposing, may help extend the effective lifespan of current and new antimicrobials. This review, presents an Australian perspective on nanotechnology-based therapies, highlighting scientific and clinical challenges. Early consideration of the potential barriers to market access may help to accelerate research translation, regulatory approval and patient access to nano-antimicrobial (NAM) drugs for resistant pathogens, not only in Australia, but globally.
Collapse
Affiliation(s)
- Benjamin W Muir
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia.
| | - Jennifer A E Payne
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia.
| | - Jennifer H Martin
- University of Newcastle School of Medicine and Public Health, Callaghan New South Wales, Australia
| | - Riley O' Shea
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Sarigama Rajesh
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Lewis D Blackman
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | | | - Chad Heazlewood
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Vipul Bansal
- Sir Ian Potter NanoBioSensing Facility, RMIT University, Melbourne, VIC, Australia
| | - Branwen Morgan
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| |
Collapse
|
5
|
Shin E, Dousa KM, Taracila MA, Bethel CR, Nantongo M, Nguyen DC, Akusobi C, Kurz SG, Plummer MS, Daley CL, Holland SM, Rubin EJ, Bulitta JB, Boom WH, Kreiswirth BN, Bonomo RA. Durlobactam in combination with β-lactams to combat Mycobacterium abscessus. Antimicrob Agents Chemother 2025; 69:e0117424. [PMID: 39714147 PMCID: PMC11823594 DOI: 10.1128/aac.01174-24] [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: 08/05/2024] [Accepted: 11/11/2024] [Indexed: 12/24/2024] Open
Abstract
Mycobacterium abscessus (Mab) presents significant clinical challenges. This study evaluated the synergistic effects of a β-lactam and β-lactamase inhibitor combination against Mab and explored the underlying mechanisms. Synergy was assessed through MIC tests and time-kill studies, and binding affinities of nine β-lactams and BLIs to eight target receptors (L,D-transpeptidases [LDT] 1-5, D,D-carboxypeptidase, penicillin-binding protein [PBP] B, and PBP-lipo) were assessed using mass spectrometry and kinetic studies. Thermal stability and morphological changes were determined. Imipenem demonstrated high binding affinity to LDTs and PBPs, with extremely low inhibition constants (Ki,app; ≤0.002 mg/L for LDT1-2, ≤0.6 mg/L for PBPs), while cephalosporins, sulopenem, tebipenem, and amoxicillin exhibited moderate to low binding affinity. Durlobactam inactivated BlaMab and LDT/PBPs more potently than avibactam. The Ki,apps of durlobactam for PBP B, PBP-lipo, and LDT2 were below clinically achievable unbound concentrations, while avibactam's Ki,app for LDT/PBPs exceeded the clinical concentrations. Single β-lactam treatments resulted in minimal killing (~1 log10 reduction). Although avibactam yielded no effect, combinations with avibactam showed a significant reduction (~4 log10 CFU/mL). Durlobactam alone showed ~2 log10 reduction, and when combined with imipenem or two β-lactams, durlobactam achieved near-eradication of Mab, surpassing the current therapy (amikacin + clarithromycin + imipenem/cefoxitin). Inactivation of PBP-lipo by sulopenem, imipenem, durlobactam, and amoxicillin (with avibactam) led to morphological changes, showing filaments. This study demonstrates the mechanistic basis of combinations therapy, particularly imipenem + durlobactam, in overcoming β-lactam resistance in Mab.
Collapse
Affiliation(s)
- Eunjeong Shin
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Khalid M. Dousa
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Magdalena A. Taracila
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Mary Nantongo
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - David C. Nguyen
- Division of Infectious Diseases, Department of Pediatrics and Division of Infectious Diseases, and Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Chidiebere Akusobi
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | - Charles L. Daley
- Division of Mycobacterial and Respiratory Infections, National Jewish Health, Denver, Colorado, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Eric J. Rubin
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jürgen B. Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - W. Henry Boom
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Barry N. Kreiswirth
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Robert A. Bonomo
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- CWRU-Cleveland VAMC Center for Antibiotic Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
- Departments of Biochemistry, Pharmacology, and Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Cleveland Geriatric Research Education and Clinical Center (GRECC), VANEOHS, Cleveland, Ohio, USA
| |
Collapse
|
6
|
El Dougdoug NK, Attia MS, Malash MN, Abdel-Maksoud MA, Malik A, Kiani BH, Fesal AA, Rizk SH, El-Sayyad GS, Harb N. Aspergillus fumigatus-induced biogenic silver nanoparticles' efficacy as antimicrobial and antibiofilm agents with potential anticancer activity: An in vitro investigation. Microb Pathog 2025; 199:106950. [PMID: 39303958 DOI: 10.1016/j.micpath.2024.106950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/18/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
A worldwide hazard to human health is posed by the growth of pathogenic bacteria that have contaminated fresh, processed, cereal, and seed products in storage facilities. As the number of multidrug-resistant (MDR) pathogenic microorganisms rises, we must find safe, and effective antimicrobials. The use of green synthesis of nanoparticles to combat microbial pathogens has gained a rising interest. The current study showed that Aspergillus fumigatus was applied as a promising biomass for the green synthesis of biogenic silver nanoparticles (Ag NPs). The UV-visible spectra of biosynthesized Ag NPs appeared characteristic surface plasmon absorption at 475 nm, round-shaped with sizes ranging from 17.11 to 75.54 nm and an average size of 50.37 ± 2.3 nm. In vitro tests were conducted to evaluate the antibacterial, antioxidant, and anticancer effects of various treatment procedures for Ag NP applications. The synthesized Ag NPs was revealed antimicrobial activity against Aspergillus flauvas, A. niger, Bacillus cereus, Candida albicans, Esherichia coli, Pseudomonas aerugonosa, and Staphylococcus aureus under optimum conditions. The tested bacteria were sensitive to low Ag NPs concentrations (5, 10, 11, 8, 7, 10, and 7 mg/mL) which was observed for the mentioned-before tested microorganisms, respectively. The tested bacterial pathogens experienced their biofilm formation effectively suppressed by Ag NPs at sub-inhibitory doses. Antibacterial reaction mechanism of Ag NPs were tested using scanning electron microscopy (SEM) to verify their antibacterial efficacy towards S. aureus and P. aeruginosa. These findings clearly show how harmful Ag NPs are to pathogenic bacteria. The synthesized Ag NPs showed antitumor activity with IC50 at 5 μg/mL against human HepG-2 and MCF-7 cellular carcinoma cells, while 50 mg/mL was required to induce 70 % of normal Vero cell mortality. These findings imply that green synthetic Ag NPs can be used on cancer cell lines in vitro for anticancer effect beside their potential as a lethal factor against some tested pathogenic microbes.
Collapse
Affiliation(s)
- Noha K El Dougdoug
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Mohamed S Attia
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
| | - Mohamed N Malash
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Mostafa A Abdel-Maksoud
- Botany and Microbiology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Bushra H Kiani
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachuesetts, 01609, USA
| | - Abeer A Fesal
- Higher Institute for Agriculture, Shoubra El-Kheima, Cairo, Egypt
| | - Samar H Rizk
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Gharieb S El-Sayyad
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Galala City, Suez, Egypt; Drug Microbiology Lab., Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt; Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Cairo, Egypt.
| | - Nashwa Harb
- Department of Biology and Microbiology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
7
|
Liu Z, Zhou Q, Xue J, Cui M, Xu L, Fang T, Wen Z, Li D, Wang J, Deng X, Zhou Y. Recent discoveries of propyl gallate restore the antibacterial effect of tigecycline against tet(X4)-positive Escherichia coli. Biochem Pharmacol 2025; 231:116638. [PMID: 39571915 DOI: 10.1016/j.bcp.2024.116638] [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: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Propyl gallate (PG), an approved food additive, can be added to different foods and drugs to provide health benefits with minimal danger. However, no clinical application of PG as an antibacterial agent for the treatment of antimicrobial resistance (AMR) has been documented. The aim of this study was to elucidate the effects and mechanisms by which PG inhibits the activity of Tet(X4). Enzyme activity inhibition assay, antimicrobial tests, scanning electron microscopy (SEM) assay, molecular docking and dynamics simulation assays, and animal infection models were used to confirm the synergistic efficacy and mechanism. Here, we found that PG efficiently inhibited Tet(X4) enzyme activity (IC50 = 34.83 μg/mL) while affecting the expression of tet(X4). PG has a synergistic effect with tigecycline (fractional inhibitory concentration index (FICI) < 0.5) against tet(X4)-positive Escherichia coli (E. coli) isolates of animal origin. The survival rates of G. mellonella larvae and the mouse systemic infection model increased by 60 % and 39 %, respectively. The combination of PG and tigecycline showed remarkable treatment benefits in terms of the bacterial load and inflammatory factors in mice. Our results indicate that PG is a valuable adjuvant with tetracyclines and can be considered to address the inevitable infection caused by tet(X4)-positive bacteria, which is a feasible way to extend the lifespan of existing antibiotics.
Collapse
Affiliation(s)
- Zhiying Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qianyu Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jinjing Xue
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China
| | - Minhe Cui
- Jilin Mushuo Breeding Co., Ltd, Changchun 130052, Jilin, China
| | - Lei Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tianqi Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; College of Food Science and Engineering, Jilin University, Changchun, China
| | - Zhongmei Wen
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Yonglin Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China.
| |
Collapse
|
8
|
Niu H, Gu J, Zhang Y. Bacterial persisters: molecular mechanisms and therapeutic development. Signal Transduct Target Ther 2024; 9:174. [PMID: 39013893 PMCID: PMC11252167 DOI: 10.1038/s41392-024-01866-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 07/18/2024] Open
Abstract
Persisters refer to genetically drug susceptible quiescent (non-growing or slow growing) bacteria that survive in stress environments such as antibiotic exposure, acidic and starvation conditions. These cells can regrow after stress removal and remain susceptible to the same stress. Persisters are underlying the problems of treating chronic and persistent infections and relapse infections after treatment, drug resistance development, and biofilm infections, and pose significant challenges for effective treatments. Understanding the characteristics and the exact mechanisms of persister formation, especially the key molecules that affect the formation and survival of the persisters is critical to more effective treatment of chronic and persistent infections. Currently, genes related to persister formation and survival are being discovered and confirmed, but the mechanisms by which bacteria form persisters are very complex, and there are still many unanswered questions. This article comprehensively summarizes the historical background of bacterial persisters, details their complex characteristics and their relationship with antibiotic tolerant and resistant bacteria, systematically elucidates the interplay between various bacterial biological processes and the formation of persister cells, as well as consolidates the diverse anti-persister compounds and treatments. We hope to provide theoretical background for in-depth research on mechanisms of persisters and suggest new ideas for choosing strategies for more effective treatment of persistent infections.
Collapse
Affiliation(s)
- Hongxia Niu
- School of Basic Medical Science and Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiaying Gu
- School of Basic Medical Science and Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Ying Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250022, Shandong, China.
| |
Collapse
|
9
|
Laxminarayan R, Impalli I, Rangarajan R, Cohn J, Ramjeet K, Trainor BW, Strathdee S, Sumpradit N, Berman D, Wertheim H, Outterson K, Srikantiah P, Theuretzbacher U. Expanding antibiotic, vaccine, and diagnostics development and access to tackle antimicrobial resistance. Lancet 2024; 403:2534-2550. [PMID: 38797178 DOI: 10.1016/s0140-6736(24)00878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024]
Abstract
The increasing number of bacterial infections globally that do not respond to any available antibiotics indicates a need to invest in-and ensure access to-new antibiotics, vaccines, and diagnostics. The traditional model of drug development, which depends on substantial revenues to motivate investment, is no longer economically viable without push and pull incentives. Moreover, drugs developed through these mechanisms are unlikely to be affordable for all patients in need, particularly in low-income and middle-income countries. New, publicly funded models based on public-private partnerships could support investment in antibiotics and novel alternatives, and lower patients' out-of-pocket costs, making drugs more accessible. Cost reductions can be achieved with public goods, such as clinical trial networks and platform-based quality assurance, manufacturing, and product development support. Preserving antibiotic effectiveness relies on accurate and timely diagnosis; however scaling up diagnostics faces technological, economic, and behavioural challenges. New technologies appeared during the COVID-19 pandemic, but there is a need for a deeper understanding of market, physician, and consumer behaviour to improve the use of diagnostics in patient management. Ensuring sustainable access to antibiotics also requires infection prevention. Vaccines offer the potential to prevent infections from drug-resistant pathogens, but funding for vaccine development has been scarce in this context. The High-Level Meeting of the UN General Assembly in 2024 offers an opportunity to rethink how research and development can be reoriented to serve disease management, prevention, patient access, and antibiotic stewardship.
Collapse
Affiliation(s)
- Ramanan Laxminarayan
- One Health Trust, Bengaluru, India; High Meadows Environmental Institute, Princeton University, Princeton, NJ, USA.
| | | | | | - Jennifer Cohn
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | | | | | - Steffanie Strathdee
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Nithima Sumpradit
- Food and Drug Administration, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Heiman Wertheim
- Department of Medical Microbiology and Radboudumc Center for Infectious Diseases, Radboudumc, Netherlands
| | | | | | | |
Collapse
|
10
|
Yun IJ, Park HJ, Chae J, Heo SJ, Kim YC, Kim B, Choi JY. Nationwide Analysis of Antimicrobial Prescription in Korean Hospitals between 2018 and 2021: The 2023 KONAS Report. Infect Chemother 2024; 56:256-265. [PMID: 38960739 PMCID: PMC11224044 DOI: 10.3947/ic.2024.0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/15/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Data on antimicrobial use at the national level are crucial for establishing domestic antimicrobial stewardship policies and enabling medical institutions to benchmark each other. This study aimed to analyze antimicrobial use in Korean hospitals. MATERIALS AND METHODS We investigated antimicrobials prescribed in Korean hospitals between 2018 and 2021 using data from the Health Insurance Review and Assessment. Primary care hospitals (PCHs), secondary care hospitals (SCHs), and tertiary care hospitals (TCHs) were included in this analysis. Antimicrobials were categorized according to the Korea National Antimicrobial Use Analysis System (KONAS) classification, which is suitable for measuring antimicrobial use in Korean hospitals. RESULTS Among over 1,900 hospitals, PCHs constituted the highest proportion, whereas TCHs had the lowest representation. The most frequently prescribed antimicrobials in 2021 were piperacillin/β-lactamase inhibitor (9.3%) in TCHs, ceftriaxone (11.0%) in SCHs, and cefazedone (18.9%) in PCHs. Between 2018 and 2021, the most used antimicrobial classes according to the KONAS classification were 'broad-spectrum antibacterial agents predominantly used for community-acquired infections' in SCHs and TCHs and 'narrow spectrum beta-lactam agents' in PCHs. Total consumption of antimicrobials decreased from 951.7 to 929.9 days of therapy (DOT)/1,000 patient-days in TCHs and 817.8 to 752.2 DOT/1,000 patient-days in SCHs during study period; however, no reduction was noted in PCHs (from 504.3 to 527.2 DOT/1,000 patient-days). Moreover, in 2021, the use of reserve antimicrobials decreased from 13.6 to 10.7 DOT/1,000 patient-days in TCHs and from 4.6 to 3.3 DOT/1,000 patient-days in SCHs. However, in PCHs, the use increased from 0.7 to 0.8 DOT/1,000 patient-days. CONCLUSION This study confirmed that antimicrobial use differed according to hospital type in Korea. Recent increases in the use of total and reserve antimicrobials in PCHs reflect the challenges that must be addressed.
Collapse
Affiliation(s)
- I Ji Yun
- Department of Pharmacy, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hyo Jung Park
- Department of Pharmaceutical Services, Samsung Medical Center, Seoul, Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Jungmi Chae
- Health Insurance Review and Assessment (HIRA) Assessment Policy Institute, Wonju, Korea
| | - Seok-Jae Heo
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Chan Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Bongyoung Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea.
| | - Jun Yong Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Batuman O, Britt-Ugartemendia K, Kunwar S, Yilmaz S, Fessler L, Redondo A, Chumachenko K, Chakravarty S, Wade T. The Use and Impact of Antibiotics in Plant Agriculture: A Review. PHYTOPATHOLOGY 2024; 114:885-909. [PMID: 38478738 DOI: 10.1094/phyto-10-23-0357-ia] [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: 05/22/2024]
Abstract
Growers have depended on the specificity and efficacy of streptomycin and oxytetracycline as a part of their plant disease arsenal since the middle of the 20th century. With climate change intensifying plant bacterial epidemics, the established success of these antibiotics remains threatened. Our strong reliance on certain antibiotics for devastating diseases eventually gave way to resistance development. Although antibiotics in plant agriculture equal to less than 0.5% of overall antibiotic use in the United States, it is still imperative for humans to continue to monitor usage, environmental residues, and resistance in bacterial populations. This review provides an overview of the history and use, resistance and mitigation, regulation, environmental impact, and economics of antibiotics in plant agriculture. Bacterial issues, such as the ongoing Huanglongbing (citrus greening) epidemic in Florida citrus production, may need antibiotics for adequate control. Therefore, preserving the efficacy of our current antibiotics by utilizing more targeted application methods, such as trunk injection, should be a major focus. [Formula: see text] Copyright © 2024 The Author(s). This is an open access article distributed under the CC BY 4.0 International license.
Collapse
Affiliation(s)
- Ozgur Batuman
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Kellee Britt-Ugartemendia
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Sanju Kunwar
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Salih Yilmaz
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Lauren Fessler
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Ana Redondo
- Department of Plant Pathology, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Kseniya Chumachenko
- Department of Wildlife Ecology and Conservation, University of Florida, Gainesville, FL
| | - Shourish Chakravarty
- Department of Food and Resource Economics, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| | - Tara Wade
- Department of Food and Resource Economics, Southwest Florida Research and Education Center, University of Florida, Immokalee, FL
| |
Collapse
|
12
|
Kinch MS, Kraft Z, Schwartz T. Antibiotic Development: Lessons from the Past and Future Opportunities. Pharm Res 2024; 41:839-848. [PMID: 38561581 DOI: 10.1007/s11095-024-03694-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
The challenge of antimicrobial resistance is broadly appreciated by the clinical and scientific communities. To assess progress in the development of medical countermeasures to combat bacterial infections, we deployed information gleaned from clinical trials conducted from 2000 to 2021. Whereas private sector interest in cancer grew dramatically over this period, activity to combat bacterial infections remained stagnant. The comparative ambivalence to antimicrobial resistance is reflected in the number of investigative drugs under clinical investigation, their stage of development and most troublingly, a declining number of organizations that are actively involved in the development of new products to treat bacterial infections. This drop reflects the exits of many companies that had previously developed antibacterial agents.
Collapse
Affiliation(s)
- Michael S Kinch
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY, 11548, USA.
| | - Zachary Kraft
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY, 11548, USA
| | - Tyler Schwartz
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY, 11548, USA
| |
Collapse
|
13
|
Sapko MT, Manyak M, Panicucci R, Javitt JC. NRX-101 (D-Cycloserine + Lurasidone) Is Active against Drug-Resistant Urinary Pathogens In Vitro. Antibiotics (Basel) 2024; 13:308. [PMID: 38666984 PMCID: PMC11047644 DOI: 10.3390/antibiotics13040308] [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: 02/18/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/29/2024] Open
Abstract
D-Cycloserine (DCS) is a broad-spectrum antibiotic that is currently FDA-approved to treat tuberculosis (TB) disease and urinary tract infection (UTI). Despite numerous reports showing good clinical efficacy, DCS fell out of favor as a UTI treatment because of its propensity to cause side effects. NRX-101, a fixed-dose combination of DCS and lurasidone, has been awarded Qualified Infectious Disease Product and Fast Track Designation by the FDA. In this study, we tested NRX-101 against the urinary tract pathogens Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii in cation-adjusted Mueller-Hinton broth (caMHB) and artificial urine media (AUM). Several strains were multidrug resistant. Test compounds were serially diluted in broth/media. Minimum inhibitory concentration (MIC) was defined as the lowest concentration of the test compound at which no bacterial growth was observed. DCS exhibited antibacterial efficacy against all strains tested while lurasidone did not appreciably affect the antibacterial action of DCS in vitro. In AUM, the MICs ranged from 128 to 512 mcg/mL for both DCS and NRX-101. In caMHB, MICs ranged from 8 to 1024 mcg/mL for NRX-101 and 32 to 512 mcg/mL for DCS alone. Our data confirm that DCS has antibacterial activity against reference and drug-resistant urinary pathogens. Furthermore, lurasidone does not interfere with DCS's antimicrobial action in vitro. These results support the clinical development of NRX-101 as a treatment for complicated urinary tract infections.
Collapse
Affiliation(s)
- Michael T. Sapko
- NRx Pharmaceuticals, 1201 N Market St, Suite 111, Wilmington, DE 19801, USA
| | - Michael Manyak
- Department of Urology, George Washington University, 900 23rd Street NW, Washington, DC 20037, USA
| | - Riccardo Panicucci
- NRx Pharmaceuticals, 1201 N Market St, Suite 111, Wilmington, DE 19801, USA
| | - Jonathan C. Javitt
- NRx Pharmaceuticals, 1201 N Market St, Suite 111, Wilmington, DE 19801, USA
- Department of Ophthalmology, Johns Hopkins University, 1800 Orleans St, Baltimore, MD 21287, USA
| |
Collapse
|
14
|
Pérez-Palacios P, Rodríguez-Ochoa JL, Velázquez-Escudero A, Rodríguez-Baño J, Rodríguez-Martínez JM, Pascual Á, Docobo-Pérez F. Implications of two-component systems EnvZ/OmpR and BaeS/BaeR in in vitro temocillin resistance in Escherichia coli. J Antimicrob Chemother 2024; 79:641-647. [PMID: 38305703 PMCID: PMC10904727 DOI: 10.1093/jac/dkae021] [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: 10/31/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND BaeS/BaeR is a two-component system of Escherichia coli that controls the expression of porins and efflux pumps. Its role in beta-lactam resistance is limited. OBJECTIVES To study the role of baeS/baeR two-component system in temocillin resistance in E. coli. METHODS E. coli strain BW25113 and single-gene deletion mutants related to two-component systems were collected from the KEIO collection. Double-gen deletion mutants were generated. Temocillin-resistant mutant frequencies were determined at 32 mg/L. E. coli BW25113 mutants were selected by selective pressure from serial passages. Biological costs were analysed by growth curves. Genomes of the generated mutants were sequenced. The expression level of the mdtA, mdtB, mdtC, acrD and tolC in the ΔbaeS mutant was determined by RT-PCR (with/without temocillin exposure). RESULTS The frequency of temocillin mutants ranged from 2.12 × 10-8 to 4.51 × 10-8 in single-porin mutants. No mutants were recovered from E. coli BW25113 (>10-9). Selection of temocillin-resistant variants by serial passage yielded mutants up to 128 mg/L. Mutations were found in the baeS gene. Temocillin MICs ranged from 4 to 32 mg/L (highest MICs for ΔbaeS and ΔompR). The efflux pumps mdtA, mdtB, mdtC and acrD pumps were overexpressed 3-10-fold in the presence of temocillin in ΔbaeS compared to control. CONCLUSIONS Mutations in the sensor histidine kinase, baeS, may be involved in temocillin resistance through the expression of the efflux pumps mdtABC and acrD. In addition, the low mutation rate may be a good predictor of temocillin activity.
Collapse
Affiliation(s)
- Patricia Pérez-Palacios
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - José Luis Rodríguez-Ochoa
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ana Velázquez-Escudero
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Jesús Rodríguez-Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José Manuel Rodríguez-Martínez
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Álvaro Pascual
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Seville, Spain
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Fernando Docobo-Pérez
- Instituto de Biomedicina de Sevilla IBIS, Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Microbiología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
15
|
Franck E, Crofts TS. History of the streptothricin antibiotics and evidence for the neglect of the streptothricin resistome. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:3. [PMID: 39843956 PMCID: PMC11702664 DOI: 10.1038/s44259-023-00020-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2025]
Abstract
The streptothricin antibiotics were among the first antibiotics to be discovered from the environment and remain some of the most recovered antimicrobials in natural product screens. Increasing rates of antibiotic resistance and recognition that streptothricin antibiotics may play a role in countering so-called super-bugs has led to the re-evaluation of their clinical potential. Here we will review the current state of knowledge of streptothricins and their resistance in bacteria, with a focus on the potential for new resistance mechanisms and determinants to emerge in the context of potential widespread clinical adoption of this antibiotic class.
Collapse
Affiliation(s)
- Ezabelle Franck
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Terence S Crofts
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
16
|
da Silva ET, de Andrade GF, Lourenço MCS, De Souza MVN. Synthesis and Anti-bacterial Activity of New Substituted 2-trifluoromethyl-4-quinolinylhydrazone Analogs against Mycobacterium tuberculosis Strains. Curr Med Chem 2024; 31:6713-6721. [PMID: 39420718 DOI: 10.2174/0109298673267136231003113803] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/26/2023] [Accepted: 09/01/2023] [Indexed: 10/19/2024]
Abstract
BACKGROUND Tuberculosis (TB) is a serious disease that still affects humanity, despite being old, caused by the bacterium Mycobacterium tuberculosis (Mtb). The emergence of drug-resistant strains has alarmed governments and international organizations, such as the World Health Organization (WHO). The need for research on new drugs that are effective in a shorter treatment time and active against resistant strains still persists. OBJECTIVE The objective of this study is to synthesize and evaluate forty-four substituted 2-trifluoromethyl-4-quinolinylhydrazone analogs, as probable inhibitors of Mycobacterium tuberculosis growth. METHODS The anti-mycobacterial activities of all tested compounds against Mycobacterium tuberculosis strains, as well as the cytotoxicity test, were evaluated using the in vitro microplate procedure with broth microdilution assay. RESULTS Thirteen compounds exhibited some activity against sensitive strain ATCC 27294, six of which were the most active: 4a, 4c, 6a, 6b, 6c, and 6g; with MIC around 7 - 8 μM, close to that presented by ethambutol (15.9 μM), a drug used in the treatment of tuberculosis. These same compounds also were active against a resistant strain of Mtb (T113), with MIC around 7 - 8 μM. Three of these compounds 4a, 6a, and 6c were not cytotoxic against Vero cells at concentrations near the MIC. CONCLUSION This study indicates the importance of the hydrazone function to obtain promising anti-TB compounds and open new perspectives for drug development.
Collapse
Affiliation(s)
- Emerson Teixeira da Silva
- Instituto de Tecnologia em Fármacos, Farmanguinhos, Fiocruz - Rua Sizenando Nabuco, 100, 21041-250, Manguinhos, Rio de Janeiro, Brazil
| | - Gabriel Fernandes de Andrade
- Instituto de Tecnologia em Fármacos, Farmanguinhos, Fiocruz - Rua Sizenando Nabuco, 100, 21041-250, Manguinhos, Rio de Janeiro, Brazil
| | | | - Marcus Vinicius Nora De Souza
- Instituto de Tecnologia em Fármacos, Farmanguinhos, Fiocruz - Rua Sizenando Nabuco, 100, 21041-250, Manguinhos, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Delbrouck JA, Murza A, Diachenko I, Ben Jamaa A, Devi R, Larose A, Chamberland S, Malouin F, Boudreault PL. From garden to lab: C-3 chemical modifications of tomatidine unveil broad-spectrum ATP synthase inhibitors to combat bacterial resistance. Eur J Med Chem 2023; 262:115886. [PMID: 37924710 DOI: 10.1016/j.ejmech.2023.115886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 11/06/2023]
Abstract
Antibiotic resistance is escalating alarmingly worldwide. Bacterial resistance mechanisms are surfacing and proliferating across the globe, jeopardizing our capacity to manage prevalent infectious illnesses. Without drastic measures, we risk entering a post-antibiotic era, where even trivial infections and injuries can cause death again. In this context, we have developed a new class of antibiotics based on tomatidine (TO), a natural product derived from tomato plants, with a novel mode of action by targeting bacterial ATP synthases. The first generation of compounds proved highly specific for small-colony variants (SCVs) of Staphylococcus aureus. However, optimization of this scaffold through extensive structure-activity relationship studies has enabled us to broaden its effectiveness to include both Gram-positive and Gram-negative bacteria. Notably, the results showed that specific C3-modification of TO could improve ATP synthase inhibition and also bypass the outer membrane barrier of Gram-negative bacteria to gain substantial growth inhibition including against multi-resistant strains.
Collapse
Affiliation(s)
- Julien A Delbrouck
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Alexandre Murza
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Iryna Diachenko
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Abdelkhalek Ben Jamaa
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Runjun Devi
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada
| | - Audrey Larose
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, J1K 2R1, QC, Canada
| | - Suzanne Chamberland
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, J1K 2R1, QC, Canada
| | - François Malouin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, J1K 2R1, QC, Canada.
| | - Pierre-Luc Boudreault
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4, QC, Canada; Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, J1H 5N4, Québec, Canada.
| |
Collapse
|
18
|
Campaniço A, Harjivan SG, Freitas E, Serafini M, Gaspar MM, Capela R, Gomes P, Jordaan A, Madureira AM, André V, Silva AB, Duarte MT, Portugal I, Perdigão J, Moreira R, Warner DF, Lopes F. Structural Optimization of Antimycobacterial Azaaurones Towards Improved Solubility and Metabolic Stability. ChemMedChem 2023; 18:e202300410. [PMID: 37845182 DOI: 10.1002/cmdc.202300410] [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: 08/02/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/18/2023]
Abstract
While N-acetyl azaaurones have already been disclosed for their potential against tuberculosis (TB), their low metabolic stability remains an unaddressed liability. We now report a study designed to improve the metabolic stability and solubility of the azaaurone scaffold and to identify the structural requirements for antimycobacterial activity. Replacing the N-acetyl moiety for a N-carbamoyl group led to analogues with sub- and nanomolar potencies against M. tuberculosis H37Rv, as well as equipotent against drug-susceptible and drug-resistant M. tuberculosis isolates. The new N-carbamoyl azaaurones exhibited improved microsomal stability, compared to their N-acetylated counterparts, with several compounds displaying moderate to high kinetic solubility. The frequency of spontaneous resistance to azaaurones was observed to be in the range of 10-8 , a value that is comparable to current TB drugs in the market. Overall, these results reveal that azaaurones are amenable to structural modifications to improve metabolic and solubility liabilities, and highlight their potential as antimycobacterial agents.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Shrika G Harjivan
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Elisabete Freitas
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Marco Serafini
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - M Manuela Gaspar
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Rita Capela
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Pedro Gomes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Audrey Jordaan
- Molecular Mycobacteriology Research Unit, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, Rondebosch, 7701, South Africa
| | - Ana M Madureira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Vânia André
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
- Associação do Instituto Superior Técnico para a Investigação e Desenvolvimento (IST-ID), Avenida António José de Almeida, n.° 12, 1000-043, Lisboa, Portugal
| | - Andreia B Silva
- Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - M Teresa Duarte
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| | - Isabel Portugal
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - João Perdigão
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Digby F Warner
- Molecular Mycobacteriology Research Unit, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, Rondebosch, 7701, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, Rondebosch, 7701, South Africa
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
19
|
Mills B, Kiang A, Mohanan SMPC, Bradley M, Klausen M. Riboflavin-Vancomycin Conjugate Enables Simultaneous Antibiotic Photo-Release and Photodynamic Killing against Resistant Gram-Positive Pathogens. JACS AU 2023; 3:3014-3023. [PMID: 38034955 PMCID: PMC10685426 DOI: 10.1021/jacsau.3c00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023]
Abstract
Decades of antibiotic misuse have led to alarming levels of antimicrobial resistance, and the development of alternative diagnostic and therapeutic strategies to delineate and treat infections is a global priority. In particular, the nosocomial, multidrug-resistant "ESKAPE" pathogens such as Gram-positive methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus spp (VRE) urgently require alternative treatments. Here, we developed light-activated molecules based on the conjugation of the FDA-approved photosensitizer riboflavin to the Gram-positive specific ligand vancomycin to enable targeted antimicrobial photodynamic therapy. The riboflavin-vancomycin conjugate proved to be a potent and versatile antibacterial agent, enabling the rapid, light-mediated, killing of MRSA and VRE with no significant off-target effects. The attachment of riboflavin on vancomycin also led to an increase in antibiotic activity against S. aureus and VRE. Simultaneously, we evidenced for the first time that the flavin subunit undergoes an efficient photoinduced bond cleavage reaction to release vancomycin, thereby acting as a photoremovable protecting group with potential applications in drug delivery.
Collapse
Affiliation(s)
- Bethany Mills
- Translational Healthcare Technologies group, Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Alex Kiang
- Translational Healthcare Technologies group, Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Syam Mohan P C Mohanan
- Translational Healthcare Technologies group, Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, U.K
| | - Mark Bradley
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K
| | - Maxime Klausen
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K
| |
Collapse
|
20
|
Tarín-Pelló A, Suay-García B, Forés-Martos J, Falcó A, Pérez-Gracia MT. Computer-aided drug repurposing to tackle antibiotic resistance based on topological data analysis. Comput Biol Med 2023; 166:107496. [PMID: 37793206 DOI: 10.1016/j.compbiomed.2023.107496] [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: 04/23/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
The progressive emergence of antimicrobial resistance has become a global health problem in need of rapid solution. Research into new antimicrobial drugs is imperative. Drug repositioning, together with computational mathematical prediction models, could be a fast and efficient method of searching for new antibiotics. The aim of this study was to identify compounds with potential antimicrobial capacity against Escherichia coli from US Food and Drug Administration-approved drugs, and the similarity between known drug targets and E. coli proteins using a topological structure-activity data analysis model. This model has been shown to identify molecules with known antibiotic capacity, such as carbapenems and cephalosporins, as well as new molecules that could act as antimicrobials. Topological similarities were also found between E. coli proteins and proteins from different bacterial species such as Mycobacterium tuberculosis, Pseudomonas aeruginosa and Salmonella Typhimurium, which could imply that the selected molecules have a broader spectrum than expected. These molecules include antitumor drugs, antihistamines, lipid-lowering agents, hypoglycemic agents, antidepressants, nucleotides, and nucleosides, among others. The results presented in this study prove the ability of computational mathematical prediction models to predict molecules with potential antimicrobial capacity and/or possible new pharmacological targets of interest in the design of new antibiotics and in the better understanding of antimicrobial resistance.
Collapse
Affiliation(s)
- Antonio Tarín-Pelló
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud Universidad Cardenal Herrera-CEU, CEU Universities, C/ Santiago Ramón y Cajal, 46115, Alfara del Patriarca, Valencia, Spain
| | - Beatriz Suay-García
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - Jaume Forés-Martos
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - Antonio Falcó
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ San Bartolomé 55, 46115, Alfara del Patriarca, Valencia, Spain
| | - María-Teresa Pérez-Gracia
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud Universidad Cardenal Herrera-CEU, CEU Universities, C/ Santiago Ramón y Cajal, 46115, Alfara del Patriarca, Valencia, Spain.
| |
Collapse
|
21
|
Torres-Boncompte J, Catalá-Gregori P, Garcia-Llorens J, Soriano JM, Sevilla-Navarro S. Simultaneous Salmonella and bacteriophage isolation on Modified Semisolid Rappaport Vassiliadis media. Poult Sci 2023; 102:102960. [PMID: 37579648 PMCID: PMC10429287 DOI: 10.1016/j.psj.2023.102960] [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: 04/30/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/16/2023] Open
Abstract
Salmonella represents a food safety concern worldwide. Despite the application of National Control Programs (NCP) against Salmonella, regulated by the European Union, every year the European Food Safety Authority reports new cases. On the look for new alternatives to antibiotics, bacteriophages, or phages, rise as a promising alternative to treat multidrug resistance infections. Although they are known to be ubiquitous in the environment, their high specificity to host cells hinders their isolation and usage for phage therapy. The ISO 6579-1:2017 is performed as a reference method in the NCP and uses an unspecific media to enrich the sample the same way most phage isolation protocols do. Later, the protocol uses a more selective media to isolate the Salmonella, Modified Semisolid Rappaport Vassiliadis (MSRV). This paper aims to find out whether, due to the similarity between phage isolation protocols and the ISO 6579-1:2017, this last one could be used as a protocol to also isolate phages against the same bacterium that is being simultaneously isolated. To do so, 2 experiments were performed to assess phage isolation from MSRV media in in-vivo conditions. The results from experiments 1 and 2 proved that the MSRV media was usable for simultaneous phage and pathogen isolation through a single procedure. Additionally, there is a correlation between the antigenic formulae from the bacteria and the phage's host range, seeming to be effective against bacteria with similar antigenic formulae.
Collapse
Affiliation(s)
- Jan Torres-Boncompte
- Food & Health Lab, Institute of Materials Science, University of Valencia, Valencia, 46980, Spain; Centro de Calidad Avícola y Alimentación Animal de la Comunidad Valenciana (CECAV), Castellón, 12539, Spain
| | - Pablo Catalá-Gregori
- Centro de Calidad Avícola y Alimentación Animal de la Comunidad Valenciana (CECAV), Castellón, 12539, Spain; Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Instituto de Ciencias Biomédicas, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113, Spain
| | - Josep Garcia-Llorens
- Food & Health Lab, Institute of Materials Science, University of Valencia, Valencia, 46980, Spain; Centro de Calidad Avícola y Alimentación Animal de la Comunidad Valenciana (CECAV), Castellón, 12539, Spain
| | - José M Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, Valencia, 46980, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute La Fe, Valencia, 46026, Spain
| | - Sandra Sevilla-Navarro
- Centro de Calidad Avícola y Alimentación Animal de la Comunidad Valenciana (CECAV), Castellón, 12539, Spain; Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Instituto de Ciencias Biomédicas, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113, Spain.
| |
Collapse
|
22
|
Soares JM, Yakovlev VV, Blanco KC, Bagnato VS. Recovering the susceptibility of antibiotic-resistant bacteria using photooxidative damage. Proc Natl Acad Sci U S A 2023; 120:e2311667120. [PMID: 37729197 PMCID: PMC10523486 DOI: 10.1073/pnas.2311667120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Multidrug-resistant bacteria are one of the most serious threats to infection control. Few new antibiotics have been developed; however, the lack of an effective new mechanism of their action has worsened the situation. Photodynamic inactivation (PDI) can break antimicrobial resistance, since it potentiates the effect of antibiotics, and induces oxidative stress in microorganisms through the interaction of light with a photosensitizer. This paper addresses the application of PDI for increasing bacterial susceptibility to antibiotics and helping in bacterial persistence and virulence. The effect of photodynamic action on resistant bacteria collected from patients and bacteria cells with induced resistance in the laboratory was investigated. Staphylococcus aureus resistance breakdown levels for each antibiotic (amoxicillin, erythromycin, and gentamicin) from the photodynamic effect (10 µM curcumin, 10 J/cm2) and its maintenance in descendant microorganisms were demonstrated within five cycles after PDI application. PDI showed an innovative feature for modifying the degree of bacterial sensitivity to antibiotics according to dosages, thus reducing resistance and persistence of microorganisms from standard and clinical strains. We hypothesize a reduction in the degree of antimicrobial resistance through photooxidative action combats antibiotic failures.
Collapse
Affiliation(s)
- Jennifer M. Soares
- Institute of Physics of São Carlos, University of São Paulo, São Carlos13566-590, Brazil
- Biomedical Engineering, Texas A&M University, College Station, TX77840
| | | | - Kate C. Blanco
- Institute of Physics of São Carlos, University of São Paulo, São Carlos13566-590, Brazil
| | - Vanderlei S. Bagnato
- Institute of Physics of São Carlos, University of São Paulo, São Carlos13566-590, Brazil
- Biomedical Engineering, Texas A&M University, College Station, TX77840
| |
Collapse
|
23
|
Alnezary FS, Almutairi MS, Gonzales-Luna AJ, Thabit AK. The Significance of Bayesian Pharmacokinetics in Dosing for Critically Ill Patients: A Primer for Clinicians Using Vancomycin as an Example. Antibiotics (Basel) 2023; 12:1441. [PMID: 37760737 PMCID: PMC10525617 DOI: 10.3390/antibiotics12091441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Antibiotic use is becoming increasingly challenging with the emergence of multidrug-resistant organisms. Pharmacokinetic (PK) alterations result from complex pathophysiologic changes in some patient populations, particularly those with critical illness. Therefore, antibiotic dose individualization in such populations is warranted. Recently, there have been advances in dose optimization strategies to improve the utilization of existing antibiotics. Bayesian-based dosing is one of the novel approaches that could help clinicians achieve target concentrations in a greater percentage of their patients earlier during therapy. This review summarizes the advantages and disadvantages of current approaches to antibiotic dosing, with a focus on critically ill patients, and discusses the use of Bayesian methods to optimize vancomycin dosing. The Bayesian method of antibiotic dosing was developed to provide more precise predictions of drug concentrations and target achievement early in therapy. It has benefits such as the incorporation of personalized PK/PD parameters, improved predictive abilities, and improved patient outcomes. Recent vancomycin dosing guidelines emphasize the importance of using the Bayesian method. The Bayesian method is able to achieve appropriate antibiotic dosing prior to the patient reaching the steady state, allowing the patient to receive the right drug at the right dose earlier in therapy.
Collapse
Affiliation(s)
- Faris S. Alnezary
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah 41477, Saudi Arabia;
| | - Masaad Saeed Almutairi
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Anne J. Gonzales-Luna
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX 77204, USA;
| | - Abrar K. Thabit
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, 7027 Abdullah Al-Sulaiman Rd, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
24
|
Szepe CK, Kafle A, Bhattarai S, Handy ST, Farone MB. Evaluation of the Antibacterial Effect of Aurone-Derived Triazoles on Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1370. [PMID: 37760667 PMCID: PMC10525585 DOI: 10.3390/antibiotics12091370] [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: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Infections caused by antibiotic-resistant bacteria continue to pose a significant public health threat despite their overall decreasing numbers in the last two decades. One group of compounds fundamental to the search for new agents is low-cost natural products. In this study, we explored a group of newly synthesized novel aurone-derived triazole compounds to identify those with pharmaceutical potential as inhibitors of antibiotic-resistant Staphylococcus aureus. Using the broth microdilution method, antibacterial activities against methicillin-resistant S. aureus ATCC 43300 (MRSA) and methicillin-sensitive S. aureus ATCC 29213 (MSSA) were identified for four aurone-derived triazole compounds, AT106, AT116, AT125, and AT137, using the half-maximal inhibitory concentrations for the bacteria (IC50) and mammalian cell lines (CC50). Compounds AT125 and AT137 were identified to have pharmaceutical potential as the IC50 values against MRSA were 5.412 µM and 3.870 µM, whereas the CC50 values measured on HepG2 cells were 50.57 µM and 39.81 µM, respectively, resulting in selectivity indexes (SI) > 10. Compounds AT106 and AT116 were also selected for further study. IC50 values for these compounds were 5.439 µM and 3.178 µM, and the CC50 values were 60.33 µM and 50.87 µM, respectively; however, SI values > 10 were for MSSA only. Furthermore, none of the selected compounds showed significant hemolytic activity for human erythrocytes. We also tested the four compounds against S. aureus biofilms. Although AT116 and AT125 successfully disrupted MSSA biofilms, there was no measurable potency against MRSA biofilms. Checkerboard antibiotic assays to identify inhibitory mechanisms for these compounds indicated activity against bacterial cell membranes and cell walls, supporting the pharmaceutical potential for aurone-derived triazoles against antibiotic-resistant bacteria. Examining structure-activity relationships between the four compounds in this study and other aurone-derived triazoles in our library suggest that substitution with a halogen on either the salicyl ring or triazole aryl group along with triazoles having nitrile groups improves anti-Staphylococcal activity with the location of the functionality being very important.
Collapse
Affiliation(s)
- Csilla Klara Szepe
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Arjun Kafle
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Shrijana Bhattarai
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Scott T. Handy
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA (S.T.H.)
| | - Mary B. Farone
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| |
Collapse
|
25
|
Martínez-Fructuoso L, Arends SJR, Freire VF, Evans JR, DeVries S, Peyser BD, Akee RK, Thornburg CC, Kumar R, Ensel S, Morgan GM, McConachie GD, Veeder N, Duncan LR, Grkovic T, O’Keefe BR. Screen for New Antimicrobial Natural Products from the NCI Program for Natural Product Discovery Prefractionated Extract Library. ACS Infect Dis 2023; 9:1245-1256. [PMID: 37163243 PMCID: PMC10262198 DOI: 10.1021/acsinfecdis.3c00067] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Indexed: 05/11/2023]
Abstract
The continuing emergence of antibiotic-resistant microbes highlights the need for the identification of new chemotypes with antimicrobial activity. One of the most prolific sources of antimicrobial molecules has been the systematic screening of natural product samples. The National Institute of Allergy and Infectious Diseases and the National Cancer Institute here report a large screen of 326,656 partially purified natural product fractions against a panel of four microbial pathogens, resulting in the identification of >3000 fractions with antifungal and/or antibacterial activity. A small sample of these active fractions was further purified and the chemical structures responsible for the antimicrobial activity were elucidated. The proof-of-concept study identified many different chemotypes, several of which have not previously been reported to have antimicrobial activity. The results show that there remain many unidentified antibiotic compounds from nature.
Collapse
Affiliation(s)
- Lucero Martínez-Fructuoso
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
| | | | - Vitor F. Freire
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
| | - Jason R. Evans
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
| | - Sean DeVries
- JMI
Laboratories, North Liberty, Iowa 52317, United States
| | - Brian D. Peyser
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
| | - Rhone K. Akee
- Natural
Products Support Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United
States
| | - Christopher C. Thornburg
- Natural
Products Support Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United
States
| | - Rohitesh Kumar
- Natural
Products Support Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United
States
| | - Susan Ensel
- Natural
Products Support Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United
States
- Department
of Chemistry and Physics, Hood College, Frederick, Maryland 21701-8599, United
States
| | - Gina M. Morgan
- JMI
Laboratories, North Liberty, Iowa 52317, United States
| | - Grant D. McConachie
- Natural
Products Support Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United
States
| | - Nathan Veeder
- JMI
Laboratories, North Liberty, Iowa 52317, United States
| | | | - Tanja Grkovic
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
- Molecular
Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Barry R. O’Keefe
- Natural
Products Branch, Developmental Therapeutic Program, Division of Cancer
Treatment and Diagnosis, National Cancer
Institute, Frederick, Maryland 21702-1201, United States
- Molecular
Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| |
Collapse
|
26
|
Xu Q, Guo M, Yu F. β-Barrel Assembly Machinery (BAM) Complex as Novel Antibacterial Drug Target. Molecules 2023; 28:molecules28093758. [PMID: 37175168 PMCID: PMC10180388 DOI: 10.3390/molecules28093758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 05/15/2023] Open
Abstract
The outer membrane of Gram-negative bacteria is closely related to the pathogenicity and drug resistance of bacteria. Outer membrane proteins (OMPs) are a class of proteins with important biological functions on the outer membrane. The β-barrel assembly machinery (BAM) complex plays a key role in OMP biogenesis, which ensures that the OMP is inserted into the outer membrane in a correct folding manner and performs nutrient uptake, antibiotic resistance, cell adhesion, cell signaling, and maintenance of membrane stability and other functions. The BAM complex is highly conserved among Gram-negative bacteria. The abnormality of the BAM complex will lead to the obstruction of OMP folding, affect the function of the outer membrane, and eventually lead to bacterial death. In view of the important role of the BAM complex in OMP biogenesis, the BAM complex has become an attractive target for the development of new antibacterial drugs against Gram-negative bacteria. Here, we summarize the structure and function of the BAM complex and review the latest research progress of antibacterial drugs targeting BAM in order to provide a new perspective for the development of antibiotics.
Collapse
Affiliation(s)
- Qian Xu
- Laboratory of Molecular Pathology, Department of Pathology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Min Guo
- Allergy Clinic, Zibo Central Hospital, Zibo 255000, China
| | - Feiyuan Yu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
27
|
Fisher JF, Mobashery S. β-Lactams from the Ocean. Mar Drugs 2023; 21:86. [PMID: 36827127 PMCID: PMC9963991 DOI: 10.3390/md21020086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The title of this essay is as much a question as it is a statement. The discovery of the β-lactam antibiotics-including penicillins, cephalosporins, and carbapenems-as largely (if not exclusively) secondary metabolites of terrestrial fungi and bacteria, transformed modern medicine. The antibiotic β-lactams inactivate essential enzymes of bacterial cell-wall biosynthesis. Moreover, the ability of the β-lactams to function as enzyme inhibitors is of such great medical value, that inhibitors of the enzymes which degrade hydrolytically the β-lactams, the β-lactamases, have equal value. Given this privileged status for the β-lactam ring, it is therefore a disappointment that the exemplification of this ring in marine secondary metabolites is sparse. It may be that biologically active marine β-lactams are there, and simply have yet to be encountered. In this report, we posit a second explanation: that the value of the β-lactam to secure an ecological advantage in the marine environment might be compromised by its close structural similarity to the β-lactones of quorum sensing. The steric and reactivity similarities between the β-lactams and the β-lactones represent an outside-of-the-box opportunity for correlating new structures and new enzyme targets for the discovery of compelling biological activities.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry & Biochemistry, 354 McCourtney Hall, University of Note Dame, Notre Dame, IN 46656-5670, USA
| | - Shahriar Mobashery
- Department of Chemistry & Biochemistry, 354 McCourtney Hall, University of Note Dame, Notre Dame, IN 46656-5670, USA
| |
Collapse
|
28
|
Blasey N, Rehrmann D, Riebisch AK, Mühlen S. Targeting bacterial pathogenesis by inhibiting virulence-associated Type III and Type IV secretion systems. Front Cell Infect Microbiol 2023; 12:1065561. [PMID: 36704108 PMCID: PMC9872159 DOI: 10.3389/fcimb.2022.1065561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Infections caused by Gram-negative pathogens pose a major health burden. Both respiratory and gastrointestinal infections are commonly associated with these pathogens. With the increase in antimicrobial resistance (AMR) over the last decades, bacterial infections may soon become the threat they have been before the discovery of antibiotics. Many Gram-negative pathogens encode virulence-associated Type III and Type IV secretion systems, which they use to inject bacterial effector proteins across bacterial and host cell membranes into the host cell cytosol, where they subvert host cell functions in favor of bacterial replication and survival. These secretion systems are essential for the pathogens to cause disease, and secretion system mutants are commonly avirulent in infection models. Hence, these structures present attractive targets for anti-virulence therapies. Here, we review previously and recently identified inhibitors of virulence-associated bacterial secretions systems and discuss their potential as therapeutics.
Collapse
|
29
|
Hamed S, Emara M. Antibacterial and Antivirulence Activities of Acetate, Zinc Oxide Nanoparticles, and Vitamin C Against E. coli O157:H7 and P. aeruginosa. Curr Microbiol 2023; 80:57. [PMID: 36588146 PMCID: PMC9805986 DOI: 10.1007/s00284-022-03151-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023]
Abstract
Infectious diseases remain one of the major health challenges worldwide due to the problem of antimicrobial resistance. Conventional antimicrobials have the disadvantage that bacteria rapidly acquire resistance to them, so alternatives must be developed to combat antibiotic resistance. Nanotechnology and the repurposing of existing drugs with known biological profiles are new approaches to replacing conventional antimicrobials. In this paper, we have tested the antibacterial activity of sodium acetate (NaA), vitamin C (VC), and zinc oxide nanoparticles (ZnO NPs) against Escherichia coli O157:H7 ATCC 51659 and Pseudomonas aeruginosa ATCC 27853. MIC values for tested compounds ranged from 0.08 to 6.5 mg ml-1, and the effect of combinations and safety profiles against HepG2 cell line of these compounds were also evaluated. At sub-MIC values, tested compounds had a potential antivirulence effect by inhibiting motility and reducing biofilm formation and maturation. Collectively, ZnO NPs and VC are considered safe alternatives to traditional antibiotics that are capable of reducing the development of antibiotic resistance in microbes. Graphical abstract representing the main aim and the final findings of our work. Spread of multidrug-resistant (MDR) bacterial strains created an urge for alternative safe antimicrobial agents. In this work, we found that ZnO NPs and vitamin C are potential candidates that could be used against MDR E.coli and P. aeruginosa.
Collapse
Affiliation(s)
- Selwan Hamed
- grid.412093.d0000 0000 9853 2750Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University - Ain Helwan, Helwan, 11795 Egypt
| | - Mohamed Emara
- grid.412093.d0000 0000 9853 2750Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University - Ain Helwan, Helwan, 11795 Egypt
| |
Collapse
|
30
|
Pipiya SO, Mirzoeva NZ, Baranova MN, Eliseev IE, Mokrushina YA, Shamova OV, Gabibov AG, Smirnov IV, Terekhov SS. Creation of Recombinant Biocontrol Agents by Genetic Programming of Yeast. Acta Naturae 2023; 15:74-80. [PMID: 37153506 PMCID: PMC10154779 DOI: 10.32607/actanaturae.11878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/21/2023] [Indexed: 05/09/2023] Open
Abstract
Bacterial infections caused by antibiotic-resistant pathogens pose an extremely serious and elusive problem in healthcare. The discovery and targeted creation of new antibiotics are today among the most important public health issues. Antibiotics based on antimicrobial peptides (AMPs) are of particular interest due to their genetically encoded nature. A distinct advantage of most AMPs is their direct mechanism of action that is mediated by their membranolytic properties. The low rate of emergence of antibiotic resistance associated with the killing mechanism of action of AMPs attracts heightened attention to this field. Recombinant technologies enable the creation of genetically programmable AMP producers for large-scale generation of recombinant AMPs (rAMPs) or the creation of rAMP-producing biocontrol agents. The methylotrophic yeast Pichia pastoris was genetically modified for the secreted production of rAMP. Constitutive expression of the sequence encoding the mature AMP protegrin-1 provided the yeast strain that effectively inhibits the growth of target gram-positive and gram-negative bacteria. An antimicrobial effect was also observed in the microculture when a yeast rAMP producer and a reporter bacterium were co-encapsulated in droplets of microfluidic double emulsion. The heterologous production of rAMPs opens up new avenues for creating effective biocontrol agents and screening antimicrobial activity using ultrahigh-throughput technologies.
Collapse
Affiliation(s)
- S. O. Pipiya
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - N. Z. Mirzoeva
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - M. N. Baranova
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - I. E. Eliseev
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - Yu. A. Mokrushina
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
- Lomonosov Moscow State University M.V. Lomonosov, Moscow, 119234 Russian Federation
| | - O. V. Shamova
- Institute of Experimental Medicine, St. Petersburg, 197022 Russian Federation
| | - A. G. Gabibov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
- Lomonosov Moscow State University M.V. Lomonosov, Moscow, 119234 Russian Federation
| | - I. V. Smirnov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
- Federal State Budgetary Institution “National Medical Research Center of Endocrinology” of the Ministry of Health of the Russian Federation, Moscow, 115478 Russian Federation
| | - S. S. Terekhov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, academicians M.M. Shemyakin and Yu.A. Ovchinnikov, Russian Academy of Sciences, Moscow, 117997 Russian Federation
- Lomonosov Moscow State University M.V. Lomonosov, Moscow, 119234 Russian Federation
| |
Collapse
|
31
|
Rosalia M, Chiesa E, Tottoli EM, Dorati R, Genta I, Conti B, Pisani S. Tobramycin Nanoantibiotics and Their Advantages: A Minireview. Int J Mol Sci 2022; 23:ijms232214080. [PMID: 36430555 PMCID: PMC9692674 DOI: 10.3390/ijms232214080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Nowadays, antimicrobial resistance (AMR) represents a challenge for antibiotic therapy, mostly involving Gram-negative bacteria. Among the strategies activated to overcome AMR, the repurposing of already available antimicrobial molecules by encapsulating them in drug delivery systems, such as nanoparticles (NPs) and also engineered NPs, seems to be promising. Tobramycin is a powerful and effective aminoglycoside, approved for complicated infections and reinfections and indicated mainly against Gram-negative bacteria, such as Pseudomonas aeruginosa, Escherichia coli, Proteus, Klebsiella, Enterobacter, Serratia, Providencia, and Citrobacter species. However, the drug presents several side effects, mostly due to dose frequency, and for this reason, it is a good candidate for nanomedicine formulation. This review paper is focused on what has been conducted in the last 20 years for the development of Tobramycin nanosized delivery systems (nanoantibiotics), with critical discussion and comparison. Tobramycin was selected as the antimicrobial drug because it is a wide-spectrum antibiotic that is effective against both Gram-positive and Gram-negative aerobic bacteria, and it is characterized by a fast bactericidal effect, even against multidrug-resistant microorganisms (MDR).
Collapse
Affiliation(s)
- Mariella Rosalia
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Enrica Chiesa
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Erika Maria Tottoli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-03-8298-7378
| | - Silvia Pisani
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
32
|
Elfadil D, Elkhatib WF, El-Sayyad GS. Promising advances in nanobiotic-based formulations for drug specific targeting against multidrug-resistant microbes and biofilm-associated infections. Microb Pathog 2022; 170:105721. [PMID: 35970290 DOI: 10.1016/j.micpath.2022.105721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Antimicrobial agents and alternative strategies to combat bacterial infections have become urgent due to the rapid development of multidrug-resistant bacteria caused by the misuse and overuse of antibiotics, as well as the ineffectiveness of antibiotics against difficult-to-treat infectious diseases. Nanobiotics is one of the strategies being explored to counter the increase in antibiotic-resistant bacteria. Nanobiotics are antibiotic molecules encapsulated in nanoparticles or artificially engineered pure antibiotics that are ≤ 100 nm in size in at least one dimension. Formulation scientists recognize nanobiotic delivery systems as an effective strategy to overcome the limitations associated with conventional antibiotic therapy. This review highlights the general mechanisms by which nanobiotics can be used to target resistant microbes and biofilm-associated infections. We focus on the design elements, properties, characterization, and toxicity assessment of organic nanoparticles, inorganic nanoparticle and molecularly imprinted polymer-based nano-formulations that can be designed to improve the efficacy of nanobiotic formulation.
Collapse
Affiliation(s)
- Dounia Elfadil
- Biology and Chemistry Department, Hassan II University of Casablanca, Morocco
| | - Walid F Elkhatib
- Microbiology and Immunology Department, Ain Shams University, African Union Organization St., Abbassia, Cairo, 11566, Egypt; Department of Microbiology and Immunology, Galala University, New Galala City, Suez, Egypt.
| | - Gharieb S El-Sayyad
- Department of Microbiology and Immunology, Galala University, New Galala City, Suez, Egypt; Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| |
Collapse
|
33
|
Gregory E, Martin C. The Intersection of Antimicrobial Stewardship, the Pharmaceutical Industry, and the Federal Legislature. Open Forum Infect Dis 2022; 9:ofac404. [PMID: 36046701 PMCID: PMC9423378 DOI: 10.1093/ofid/ofac404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
To mitigate the dangers of inappropriate antimicrobial use leading to increased multidrug-resistant organisms and mortality, antimicrobial stewardship programs have become a mainstay in many health systems. Unfortunately, some pharmaceutical manufacturers simultaneously have ended antimicrobial research and development efforts altogether due to suboptimal return on investments. An optimal and sustainable antimicrobial armamentarium requires a broad alliance between antimicrobial stewardship programs, the pharmaceutical industry, the legislature, and federal and state agencies. Public-private relationships such as the Combating Antibiotic-Resistant Bacteria Biopharmaceutical Accelerator (CARB-X) and legislative policies creating push and pull incentives, including the Generating Antibiotic Incentives Now (GAIN), Developing an Innovative Strategy for Antimicrobial-Resistant Microorganisms (DISARM), and Pioneering Antimicrobial Subscriptions to End Upsurging Resistance (PASTEUR) Acts, are each a step in the right direction, but more work remains. Understanding these legislative actions is imperative for all clinicians, as is teamwork from those involved in the antimicrobial field to develop and maintain the life cycle of each drug that harbors societal value.
Collapse
Affiliation(s)
- Eric Gregory
- Department of Pharmacy, University of Kansas Health System, Kansas City, Kansas, USA
| | - Craig Martin
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| |
Collapse
|