1
|
Gu L, Wang Y, Wang H, Xu D. Analysis of clinical and microbiological characteristics of invasive Klebsiella pneumoniae liver abscess syndrome. BMC Infect Dis 2025; 25:626. [PMID: 40301787 PMCID: PMC12039297 DOI: 10.1186/s12879-025-10981-9] [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: 10/12/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Invasive Klebsiella pneumoniae liver abscess syndrome (IKPLAS) is emerging as a new disease worldwide, threatening human health. This study aimed to investigate the clinical and microbiological features of IKPLAS in order to detect this syndrome early and select antibiotics appropriately. METHODS Medical data from patients in Tongji Hospital, China, diagnosed with Klebsiella pneumoniae liver abscess (KPLA) between 2015 and 2023 was collected and analyzed retrospectively. RESULTS The study included 208 patients with KPLA, 41 with IKPLAS, and 167 with non-IKPLAS (NIKPLAS). Multivariate logistic regression analysis demonstrated that symptoms in other organ systems (including ocular, pulmonary, and neurological symptoms) (p = 0.001) and a sequential organ failure assessment (SOFA) score ≥ 4 within 48 h of admission (P = 0.002) were significant risk factors for IKPLAS. Patients with IKPLAS had a higher risk of developing multiple organ dysfunction (MODS), and a PCT ≥ 10 ng/mL was identified as an independent risk factor for MODS (p = 0.01). IKPLAS was associated with significantly prolonged hospital stays and unfavorable outcomes (all p < 0.05). There were no significant differences in microbiological characteristics between IKPLAS and NIKPLAS, including the antimicrobial susceptibility pattern and resistance profile of Klebsiella pneumoniae (KP) (all p > 0.05). In this study, KP isolates were susceptible to most antibiotics, with low rates of drug resistance. Specifically, a total of five carbapenem-resistant strains (2.6%) and seven multidrug-resistant strains (3.6%) were detected, all of which were derived from the NIKPLAS group. CONCLUSIONS Symptoms in other organ systems and the SOFA score ≥ 4 within 48 h of admission were significant predictors for IKPLAS. This study elucidated the antimicrobial susceptibility profile of liver abscess-associated KP strains, providing a reference for the early initiation of rational and effective antimicrobial therapy in patients with KPLA.
Collapse
Affiliation(s)
- Li Gu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong Xu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Chamizo-López FJ, Gutiérrez-Fernández J, Rojo-Martín MD, Borrego-Alcaide AB, González-Hevilla A, Lara-Oya A, Palop-Borrás B, Navarro-Marí JM, Pérez-Ruiz M. Development and Validation of a Multiplex Real-Time PCR Assay for Rapid Screening of Main Carbapenemase Genes in Clinical Isolates and Surveillance Samples. Antibiotics (Basel) 2025; 14:363. [PMID: 40298527 PMCID: PMC12024442 DOI: 10.3390/antibiotics14040363] [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: 12/11/2024] [Revised: 02/26/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND/OBJECTIVES Carbapenem-resistant Enterobacterales, largely due to carbapenemase production, are significant public health threats, which compromise treatment with key β-lactam antibiotics. Early detection is essential for guiding therapy and controlling spread. This study describes the design, optimisation and validation of a multiplex real-time PCR for the screening of the most frequent carbapenemases in our area. METHODS Primers and probes targeted at genes encoding carbapenemases blaKPC, blaIMP, blaVIM, blaNDM and blaOXA-48-group were designed and adapted for the development, and in silico and experimental validation of a single-tube real-time PCR. RESULTS A good linear correlation between the fluorescence values in the real-time PCR and the log10 of bacterial concentration of each carbapenemase-containing bacterial suspension was observed (R2 > 0.98). The limit of detection was 2-15, 16-256, 42-184, 4-42, 42-226 CFU/reaction of VIM-, IMP-, NDM-, KPC- and OXA-48-carbapenemase-containing bacteria, respectively. Intra-assay coefficient of variation for the mean Ct values ranged from 0.99% for OXA-48 to 3.34% for KPC. Inter-assay variability remained below 7%. Real-time PCR tested on bacterial isolates yielded 100% sensitivity and specificity. Analysis of rectal swabs using extracted DNA and a DNA extraction-free protocol showed good concordance with culture-based phenotypic methods. Additionally, the molecular method could detect all targets, except for one sample where only the DNA extraction-free protocol detected NDM. CONCLUSIONS The assay offers a rapid, sensitive and specific method for the screening of major carbapenemase genes, providing an effective tool for surveillance and infection control in clinical settings. The DNA extraction-free protocol converts this method into a good alternative for screening in 24/7 clinical laboratories. Further multiplexing to target other resistance genes, on demand, could add potential benefits to this laboratory-developed method.
Collapse
Affiliation(s)
- Francisco Javier Chamizo-López
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
- Department de Microbiología, Universidad de Granada, Fuerzas Armadas, s/n. 18014 Granada, Spain;
| | - José Gutiérrez-Fernández
- Department de Microbiología, Universidad de Granada, Fuerzas Armadas, s/n. 18014 Granada, Spain;
| | - María Dolores Rojo-Martín
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
| | - Ana Belén Borrego-Alcaide
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
| | - Alba González-Hevilla
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
| | - Ana Lara-Oya
- Servicio de Microbiología, Complejo Hospitalario de Jaén, Av. del Ejército Español, 10, 23007 Jaén, Spain;
| | - Begoña Palop-Borrás
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
| | - José María Navarro-Marí
- Instituto Biosanitario de Granada, Avda, Servicio de Microbiología, Hospital Universitario Virgen de las Nieves, Fuerzas Armadas, s/n. 18014 Granada, Spain;
| | - Mercedes Pérez-Ruiz
- Servicio de Microbiología, Hospital Regional Universitario de Málaga, Carlos Haya, s/n. 29010 Málaga, Spain; (F.J.C.-L.); (M.D.R.-M.); (A.B.B.-A.); (A.G.-H.); (B.P.-B.)
- Department de Microbiología, Universidad de Granada, Fuerzas Armadas, s/n. 18014 Granada, Spain;
| |
Collapse
|
3
|
Carmeli Y, Cisneros JM, Paul M, Daikos GL, Wang M, Torre-Cisneros J, Singer G, Titov I, Gumenchuk I, Zhao Y, Jiménez-Rodríguez RM, Liang L, Chen G, Pyptiuk O, Aksoy F, Rogers H, Wible M, Arhin FF, Luckey A, Leaney JL, Pypstra R, Chow JW. Aztreonam-avibactam versus meropenem for the treatment of serious infections caused by Gram-negative bacteria (REVISIT): a descriptive, multinational, open-label, phase 3, randomised trial. THE LANCET. INFECTIOUS DISEASES 2025; 25:218-230. [PMID: 39389071 DOI: 10.1016/s1473-3099(24)00499-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND There is a need for additional therapeutic options for serious infections caused by Gram-negative pathogens. In the phase 3, descriptive REVISIT study, we investigated the safety and efficacy of aztreonam-avibactam in the treatment of complicated intra-abdominal infections or hospital-acquired pneumonia or ventilator-associated pneumonia (HAP-VAP) caused, or suspected to be caused, by Gram-negative bacteria. METHODS This prospective, multinational, open-label, central assessor-masked study enrolled adults who were hospitalised with a complicated intra-abdominal infection or HAP-VAP. Patients were randomly allocated via block randomisation using interactive response technology stratified by infection type in a 2:1 ratio to aztreonam-avibactam (with metronidazole for complicated intra-abdominal infection) or meropenem with or without colistin for 5-14 days for complicated intra-abdominal infection or 7-14 days for HAP-VAP. The primary endpoint was clinical cure at the test-of-cure visit (within 3 days before or after day 28) in the intention-to-treat (ITT) population. Secondary endpoints included 28-day mortality in the ITT population and safety in patients in the ITT population who received study drug (safety analysis set). No formal hypothesis testing was planned. The study was registered with ClinicalTrials.gov (NCT03329092) and EudraCT (2017-002742-68) and is complete. FINDINGS Between April 5, 2018, and Feb 23, 2023, we screened 461 patients. 422 patients were enrolled and randomly allocated (282 in the aztreonam-avibactam group and 140 in the meropenem group, forming the ITT analysis set), of whom ten patients (seven in the aztreonam-avibactam group and three in the meropenem group) were randomly allocated but did not receive study treatment. 271 (64%) of 422 patients had at least one Gram-negative pathogen from an adequate specimen identified at baseline. The most frequent baseline pathogens were Enterobacterales (252 [93%] of 271). Overall, 19 (24%) of 80 isolates tested for carbapenemases were carbapenemase-positive (serine, metallo-β-lactamase, or both). 193 (68·4%) of 282 patients in the aztreonam-avibactam group and 92 (65·7%) of 140 in the meropenem group had clinical cure at the test-of-cure visit (treatment difference 2·7% [95% CI -6·6 to 12·4]). For patients with complicated intra-abdominal infection, the adjudicated clinical cure rate was 76·4% (159 of 208) for the aztreonam-avibactam group and 74·0% (77 of 104) for the meropenem group. Cure rates in patients with HAP-VAP were 45·9% (34 of 74) for aztreonam-avibactam and 41·7% (15 of 36) for meropenem. 28-day all-cause mortality rates were 4% (12 of 282) for aztreonam-avibactam and 7% (ten of 140) for meropenem; in patients with complicated intra-abdominal infection, mortality was 2% (four of 208) and 3% (three of 104) for aztreonam-avibactam and meropenem, respectively, and in patients with HAP-VAP, mortality was 11% (eight of 74) and 19% (seven of 36), respectively. Aztreonam-avibactam was generally well tolerated, and safety findings were consistent with the known safety profile of aztreonam monotherapy. There were no treatment-related serious adverse events in the aztreonam-avibactam group. INTERPRETATION These phase 3 efficacy and safety data provide support for aztreonam-avibactam as a potential therapeutic option for complicated intra-abdominal infection or HAP-VAP caused by Gram-negative bacteria. FUNDING Pfizer.
Collapse
Affiliation(s)
- Yehuda Carmeli
- The National Center for Antibiotic Resistance and Infection Control, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - José Miguel Cisneros
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Hospital Universitario Virgen del Rocío, IBiS/CSIC, Universidad de Sevilla. CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Mical Paul
- Infectious Diseases Institute, Rambam Health Care Campus, Technion - Israel Institute of Technology, Haifa, Israel; Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - George L Daikos
- National and Kapodistrian University of Athens, Athens, Greece
| | - Minggui Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Heath Commission of People's Republic of China, Shanghai, China
| | | | - George Singer
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ivan Titov
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Illia Gumenchuk
- Communal Non-Profit Enterprise Vinnytsia Regional Clinical Hospital Named after M I Pyrogov Vinnytsia Regional Council, Vinnytsia, Ukraine
| | | | - Rosa-María Jiménez-Rodríguez
- Unidad Clínica de Cirugía General, Hospital Universitario Virgen del Rocío, IBiS/CSIC, Universidad de Sevilla, Spain
| | - Lu Liang
- Baotou Central Hospital, Baotou, Inner Mongolia, China
| | - Gang Chen
- First People's Hospital of Kunming, Kunming, China
| | - Oleksandr Pyptiuk
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Firdevs Aksoy
- Karadeniz Technical University, School of Medicine, Trabzon, Turkey
| | | | | | | | - Alison Luckey
- Global Antibiotic R&D Partnership, Geneva, Switzerland
| | | | | | | |
Collapse
|
4
|
Sharma R, Rodriguez-Rios M, Crossland J, Septiyana M, Megia-Fernandez A, Klausen M, Bradley M. A multi-valent polymyxin-based fluorescent probe for the detection of Gram-negative infections. J Mater Chem B 2025; 13:882-887. [PMID: 39717883 DOI: 10.1039/d4tb01786b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
A multi-branched fluorogenic probe for the rapid and specific detection of Gram-negative bacteria is reported. Three Gram-negative-targeting azido-modified polymyxins were clicked onto a trivalent scaffold functionalised with the environmental green-emitting fluorophore 7-nitrobenz-2-oxa-1,3-diazole. The probe allowed wash-free detection of target bacteria with increased sensitivity and lower limits of detection compared to monovalent probes.
Collapse
Affiliation(s)
- Richa Sharma
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - Maria Rodriguez-Rios
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - James Crossland
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - Maulida Septiyana
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Mataram, Mataram, Indonesia
| | - Alicia Megia-Fernandez
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
| | - Maxime Klausen
- School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, UK
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005 Paris, France.
| | - Mark Bradley
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House, London, E1 1HH, UK.
| |
Collapse
|
5
|
Abdullah SJ, Guan JS, Mu Y, Bhattacharjya S. Single Disulfide Bond in Host Defense Thanatin Analog Peptides: Antimicrobial Activity, Atomic-Resolution Structures and Target Interactions. Int J Mol Sci 2024; 26:51. [PMID: 39795909 PMCID: PMC11720011 DOI: 10.3390/ijms26010051] [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: 11/25/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Host defense antimicrobial peptides (AMPs) are promising lead molecules with which to develop antibiotics against drug-resistant bacterial pathogens. Thanatin, an inducible antimicrobial peptide involved in the host defense of Podisus maculiventris insects, is gaining considerable attention in the generation of novel classes of antibiotics. Thanatin or thanatin-based analog peptides are extremely potent in killing bacterial pathogens in the Enterobacteriaceae family, including drug-resistant strains of Escherichia coli and Klebsiella pneumoniae. A single disulfide bond that covalently links two anti-parallel β-strands in thanatin could be pivotal to its selective antibacterial activity and mode of action. However, potential correlations of the disulfide covalent bond with structure, activity and target binding in thanatin peptides are currently unclear to. Here, we examined a 16-residue designed thanatin peptide, namely disulfide-bonded VF16QK, and its Cys to Ser substituted variant, VF16QKSer, to delineate their structure-activity relationships. Bacterial growth inhibitory activity was only detected for the disulfide-bonded VF16QK peptide. Mechanistically, both peptides vastly differ in their bacterial cell permeabilizations, atomic-resolution structures, interactions with the LPS-outer membrane and target periplasmic protein LptAm binding. In particular, analysis of the 3-D structures of the two peptides revealed an altered folded conformation for the VF16QKSer peptide that was correlated with diminished LPS-outer membrane permeabilization and target interactions. Analysis of docked complexes of LPS-thanatin peptides indicated potential structural requirements and conformational adaptation for antimicrobial activity. Collectively, these observations contrast with those for the disulfide-bonded β-hairpin antimicrobial protegrin and tachyplesin peptides, where disulfide bonds are dispensable for activity. We surmise that the atomistic structures and associated molecular interactions presented in this work can be utilized to design novel thanatin-based antibiotics.
Collapse
Affiliation(s)
| | | | | | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
6
|
Huzum B, Aprotosoaie AC, Alexa O, Sîrbu PD, Puha B, Veliceasa B, Huzum RM. Antimicrobials in Orthopedic Infections: Overview of Clinical Perspective and Microbial Resistance. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1988. [PMID: 39768868 PMCID: PMC11728363 DOI: 10.3390/medicina60121988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Orthopedic infections are challenging pathologies that impose a heavy burden on patients and the healthcare system. Antimicrobial therapy is a critical component of the successful management of orthopedic infections, but its effectiveness depends on patient-, surgery-, drug-, and hospital-related factors. The dramatic increase in the emergence of multidrug-resistant microbial strains necessitates new clinical approaches in order to prevent or limit this phenomenon and to ensure a favorable therapeutic outcome. The present paper reviews the currently available antimicrobial strategies in the management of orthopedic infections, highlighting their clinical use related to the occurrence of microbial resistance. Some approaches for reducing antibiotic resistance emergence in orthopedics are also presented. The use of antibiotics tailored to the microorganism's sensitivity profile, patient factors, and pharmacokinetic profile in terms of monotherapy or combinations, the understanding of microbial pathogenicity and resistance patterns, strict control measures in healthcare facilities, the development of new antimicrobial therapies (drugs, devices, technologies), and patient education for improving compliance and tolerance are some of the most important tools for overcoming microbial resistance.
Collapse
Affiliation(s)
- Bogdan Huzum
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Ana Clara Aprotosoaie
- Faculty of Pharmacy “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania
| | - Ovidiu Alexa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Paul Dan Sîrbu
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Puha
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Bogdan Veliceasa
- Department of Orthopaedic and Traumatology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania; (B.H.); (O.A.); (P.D.S.); (B.P.); (B.V.)
| | - Riana Maria Huzum
- Department of Radiology, Faculty of Medicine “Grigore T. Popa”, University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania;
| |
Collapse
|
7
|
Darwiche FI, Hussein HM, Harb SB, Nahhal S, Kurdi A, Sleiman A, Hamadeh L, Barada S, Gerges JR, Araj GF, Zahreddine NK, Ibrahim A, Kanafani Z, Mahfouz R, Kanj SS, Matar GM, Fayad AGA. Investigating an outbreak of extensively drug-resistant Acinetobacter baumannii in a tertiary healthcare centre in lebanon using next-generation sequencing. Int J Antimicrob Agents 2024; 64:107353. [PMID: 39362611 DOI: 10.1016/j.ijantimicag.2024.107353] [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: 10/31/2023] [Revised: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The frequent occurrence of Acinetobacter baumannii in hospital settings and the elevated rate of antimicrobial resistance in this pathogen represent a serious clinical and public health threat worldwide, and particularly in Lebanon where outbreak surveillance and control are still insufficient. Whole-genome sequencing (WGS) is a fast and reliable tool to study outbreaks at the molecular level and obtain actionable knowledge, leading to better control measures. A total of 59 A. baumannii isolates were collected from intensive care unit (ICU) patients (57 isolates) and from the hospital environment (2 isolates) between August 2022 and May 2023, antimicrobial susceptibility testing (AST) was performed and gDNA was subjected to WGS. Analysis was performed to reveal the sequence types (ST), the relatedness to strains that caused other outbreaks and the arsenal of resistance genes harboured by these bacteria. Of 59 isolates, 85% were categorised as extensively drug-resistant (XDR), 13.6% as multidrug-resistant (MDR) and 1.7% as pan-drug-resistant. All isolates belonged to international clone (IC)2, of which the majority were of ST2 (91.5%). The isolates clustered well with those of a previous outbreak in the same hospital. In addition, isolates from hospitals in Lebanon clustered well together and some clustered with those originating from other countries. The observed genetic relatedness between the current isolates and those from the previous outbreaks underscores the importance of strict surveillance to limit the threat of outbreaks. Moreover, the clustering of isolates from Lebanon with others from distant countries proves the necessity to further investigate the international spread of drug-resistant pathogens and the implementation of control strategies.
Collapse
Affiliation(s)
- Fatima I Darwiche
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - Hadi M Hussein
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - Souad Bou Harb
- Department of Internal medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sarah Nahhal
- Department of Internal medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Abdallah Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmad Sleiman
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - Lama Hamadeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Pillar Genomics Laboratory, American University of Beirut, Beirut, Lebanon
| | - Sara Barada
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - Jose-Rita Gerges
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - George F Araj
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nada Kara Zahreddine
- Infection control and prevention program, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ahmad Ibrahim
- Infection control and prevention program, American University of Beirut Medical Center, Beirut, Lebanon
| | - Zeina Kanafani
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rami Mahfouz
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon; Pillar Genomics Laboratory, American University of Beirut, Beirut, Lebanon
| | - Souha S Kanj
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| | - Antoine G Abou Fayad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; World Health Organization (WHO) Collaborating Center for Reference and Research on Bacterial Pathogens, Beirut, Lebanon.
| |
Collapse
|
8
|
Liu Z, Liang X, Zhang Y, Deng W, Wang Y, Lu Z, Liu Q, Wei L. Drug Repurposing: Research Progress of Niclosamide and Its Derivatives on Antibacterial Activity. Infect Drug Resist 2024; 17:4539-4556. [PMID: 39464831 PMCID: PMC11505561 DOI: 10.2147/idr.s490998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
The development of antibiotic resistance complicates the treatment of infectious diseases and is a global public health threat. However, drug repurposing can address this resistance issue and reduce research and development costs. Niclosamide is a salicylanilide compound approved by the Food and Drug Administration (FDA), and it has been used clinically for treating parasitic infections for many years. Recent studies have shown that niclosamide can inhibit bacterial and fungus activity by affecting the quorum sensing system, biofilm formation, cell membrane potential, and other mechanisms. Here, we discuss recent advances in the antimicrobial applications of niclosamide and its derivatives to provide new perspectives in treating infectious diseases.
Collapse
Affiliation(s)
- Zhihong Liu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Xiaofang Liang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yu Zhang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Wenbo Deng
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yulin Wang
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Zhangping Lu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Qianqian Liu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Lianhua Wei
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
9
|
Nasr J, Abdessamad H, Mina J, Haykal T, Jamil Y, Abboud E, Mahdi A, Asmar R, Abi Assaad R, Alameddine D, Bourji A, Mahdi M, Abdulaal R, Tomassian S, El Ahmadieh H, Azzam W, Mokhbat JE, Moghnieh R, Rodriguez-Morales AJ, Husni R. The epidemiology of gram-negative bacteremia in Lebanon: a study in four hospitals. Ann Clin Microbiol Antimicrob 2024; 23:90. [PMID: 39385237 PMCID: PMC11465513 DOI: 10.1186/s12941-024-00740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Gram-negative bacteremia is a life-threatening infection with high morbidity and mortality. Its incidence is rising worldwide, and treatment has become more challenging due to emerging bacterial resistance. Little data is available on the burden and outcome of such infections in Lebanon. METHODS We conducted this retrospective study in four Lebanese hospitals. Data on medical conditions and demographics of 2400 patients diagnosed with a bloodstream infection based on a positive blood culture were collected between January 2014 and December 2020. RESULTS Most bacteremias were caused by Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii, with the more resistant organisms being hospital-acquired. Third-generation cephalosporin and quinolone resistance was steady throughout the study, but carbapenem resistance increased. Mortality with such infections is high, but carbapenem resistance or infection with Pseudomonas or Acinetobacter species were significant risk factors for poor outcomes. CONCLUSION This is the first multi-center study from Lebanon on gram-negative bacteremia, resistance patterns, and factors associated with a poor outcome. More surveillance is needed to provide data to guide empirical treatment for bacteremia in Lebanon.
Collapse
Affiliation(s)
- Janane Nasr
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Hilal Abdessamad
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Johnathan Mina
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Tony Haykal
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Yasser Jamil
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Emma Abboud
- Laboratory Director, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Ahmad Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rana Asmar
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rawad Abi Assaad
- Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Dana Alameddine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alaa Bourji
- Department of Surgery, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Mahmoud Mahdi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Razan Abdulaal
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Serge Tomassian
- Department of Internal Medicine, University of Balamand, Balamand, Lebanon
| | - Hanane El Ahmadieh
- Infection Control Coordination, Mount Lebanon Hospital University Medical Center, Beirut, 1102, Lebanon
| | - Wael Azzam
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Jacques E Mokhbat
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Rima Moghnieh
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
| | - Alfonso J Rodriguez-Morales
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon
- Master of Clinical Epidemiology and Biostatistics, Universidad Cientifica del Sur, Lima, 15067, Peru
| | - Rola Husni
- Division of Infectious Diseases, Department of Internal Medicine, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 1102, Lebanon.
| |
Collapse
|
10
|
Kuo LK, Chang HT, Hsueh SC, Liu IM, Hsieh PC, Jean SS. Bacterial profile, and independent predictors for healthcare-associated pneumonia persistently caused by multidrug-resistant Gram-negative bacteria for patients with the preceding multidrug-resistant Gram-negative pneumonia in Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:801-811. [PMID: 39147627 DOI: 10.1016/j.jmii.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/18/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES To understand the microbial profile and investigate the independent predictors for healthcare-associated pneumonia (HCAP) pertinaciously caused by isolates of multidrug-resistant (MDR) Gram-negative bacteria (GNB). METHODS Multicenter ICU patients who received appropriate antibiotic treatments for preceding pneumonia due to MDR GNB isolates and subsequently developed HCAP caused by either MDR GNB (n = 126) or non-MDR GNB (n = 40) isolates in Taiwan between 2018 and 2023 were enrolled. Between the groups of patients with HCAP due to MDR GNB and non-MDR GNB, the proportions of the following variables, including demographic characteristics, important co-morbidities, nursing home residence, physiological severity, intervals between two hospitalizations, steroid use, the tracheostomy tube use alone, ventilator support, and the predominant GNB species involving HCAP, were analyzed using the chi-square test. Logistic regression was employed to explore the independent predictors for HCAP persistently caused by MDR GNB in the aforementioned variables with a P-value of <0.15 in the univariate analysis. RESULTS MDR-Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii complex were the three predominant species causing HCAP. Chronic structural lung disorders, diabetes mellitus, intervals of ≤30 days between two hospitalizations, use of the tracheostomy tube alone, and prior pneumonia caused by MDR A. baumannii complex were shown to independently predict the HCAP tenaciously caused by MDR GNB. Conversely, the preceding pneumonia caused by MDR P. aeruginosa was a negative predictor. CONCLUSION Identifying predictors for HCAP persistently caused by MDR GNB is crucial for prescribing appropriate antibiotics.
Collapse
Affiliation(s)
- Li-Kuo Kuo
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Critical Care Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Hou-Tai Chang
- Department of Critical Care Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Industrial Engineering and Management, Yuan Ze University, Taoyuan, Taiwan
| | - Shun-Chung Hsueh
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - I-Min Liu
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Po-Chuen Hsieh
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Shio-Shin Jean
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan; Departments of Internal Medicine and Critical Care Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan.
| |
Collapse
|
11
|
Berry L, Ramirez D, Domalaon R, Schweizer F. Sulfonamide Bioisosteres of Niclosamide Enhance Antibacterial Activity of Colistin and Bacitracin. ChemMedChem 2024; 19:e202400175. [PMID: 38679656 DOI: 10.1002/cmdc.202400175] [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: 03/05/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Multicomponent therapy combining antibiotics with enhancer molecules known as adjuvants is an emerging strategy to combat antimicrobial resistance. Niclosamide is a clinically relevant anthelmintic drug with potential to be repurposed for its inherent antibacterial activity against Gram-positive bacteria and its ability to potentiate the antibacterial activity of colistin against susceptible and resistant Gram-negative bacteria. Herein, sulfonamide analogs of niclosamide were prepared and found to enhance colistin activity against Gram-negative bacteria. The ability of niclosamide and the new sulfonamide analogs to synergize with bacitracin against vancomycin-resistant Enterococcus faecium was also discovered.
Collapse
Affiliation(s)
- Liam Berry
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Danyel Ramirez
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Ronald Domalaon
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Keck JM, Viteri A, Schultz J, Fong R, Whitman C, Poush M, Martin M. New Agents Are Coming, and So Is the Resistance. Antibiotics (Basel) 2024; 13:648. [PMID: 39061330 PMCID: PMC11273847 DOI: 10.3390/antibiotics13070648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance is a global threat that requires urgent attention to slow the spread of resistant pathogens. The United States Centers for Disease Control and Prevention (CDC) has emphasized clinician-driven antimicrobial stewardship approaches including the reporting and proper documentation of antimicrobial usage and resistance. Additional efforts have targeted the development of new antimicrobial agents, but narrow profit margins have hindered manufacturers from investing in novel antimicrobials for clinical use and therefore the production of new antibiotics has decreased. In order to combat this, both antimicrobial drug discovery processes and healthcare reimbursement programs must be improved. Without action, this poses a high probability to culminate in a deadly post-antibiotic era. This review will highlight some of the global health challenges faced both today and in the future. Furthermore, the new Infectious Diseases Society of America (IDSA) guidelines for resistant Gram-negative pathogens will be discussed. This includes new antimicrobial agents which have gained or are likely to gain FDA approval. Emphasis will be placed on which human pathogens each of these agents cover, as well as how these new agents could be utilized in clinical practice.
Collapse
Affiliation(s)
- J. Myles Keck
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alina Viteri
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | - Rebecca Fong
- Department of Pharmacy, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Charles Whitman
- Department of Pharmacy, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Madeline Poush
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Marlee Martin
- Department of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
13
|
Caffrey AR, Appaneal HJ, Lopes VV, Riccobene TA, LaPlante KL. Real-world utilization of ceftazidime/avibactam among inpatients in the national Veterans Affairs Healthcare System. Am J Health Syst Pharm 2024; 81:509-520. [PMID: 38365226 DOI: 10.1093/ajhp/zxae038] [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] [Indexed: 02/18/2024] Open
Abstract
PURPOSE Multidrug-resistant (MDR) infections are challenging to treat due to underlying patient conditions, pathogen characteristics, and high antibiotic resistance rates. As newer antibiotic therapies come to market, limited data exist about their real-world utilization. METHODS This was a national retrospective cohort study of ceftazidime/avibactam (approved in 2015) utilization among inpatients from the Veterans Affairs (VA) Healthcare System, from 2015 through 2021. Joinpoint regression was used to estimate time trends in utilization. RESULTS Ceftazidime/avibactam use increased by 52.3% each year (days of therapy per 1,000 bed days; 95% confidence interval, 12.4%-106.4%). We identified 1,048 unique predominantly male (98.3%) and white (66.2%; Black, 27.7%) patients treated with ceftazidime/avibactam, with a mean (SD) age of 71.5 (11.9) years. The most commonly isolated organisms were Pseudomonas aeruginosa (36.3%; carbapenem resistant, 80.6%; MDR, 65.0%) and Klebsiella species (34.1%; carbapenem resistant, 78.4%; extended-spectrum cephalosporin resistant, 90.7%). Common comorbid conditions included hypertension (74.8%), nervous system disorders (60.2%), diabetes mellitus (48.7%), and cancer (45.1%). Median time to ceftazidime/avibactam initiation from admission was 6 days, with a median of 3 changes in therapy before ceftazidime/avibactam initiation and a subsequent median length of inpatient stay of 14 days (median of 8 days of ceftazidime/avibactam therapy). Treatment heterogeneity was high, both before ceftazidime/avibactam initiation (89.6%) and during ceftazidime/avibactam treatment (85.6%), and common concomitant antibiotics included vancomycin (41.4%), meropenem (24.1%), cefepime (15.2%), and piperacillin/tazobactam (15.2%). The inpatient mortality rate was 23.6%, and 20.8% of patients had a subsequent admission with ceftazidime/avibactam treatment. CONCLUSION Utilization of ceftazidime/avibactam increased from 2015 to 2021 in the national VA Healthcare System. Ceftazidime/avibactam was utilized in complex, difficult-to-treat patients, with substantial treatment heterogeneity and variation in the causative organism and culture sites.
Collapse
Affiliation(s)
- Aisling R Caffrey
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Haley J Appaneal
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Vrishali V Lopes
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | | | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
14
|
Nag P, Sadani K, Pisharody L, Thian XY, Ratnakar TS, Ansari A, Mukherji S, Mukherji S. Essential oil mediated synthesis and application of highly stable copper nanoparticles as coatings on textiles and surfaces for rapid and sustained disinfection of microorganisms. NANOTECHNOLOGY 2024; 35:345602. [PMID: 38788697 DOI: 10.1088/1361-6528/ad501b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 05/24/2024] [Indexed: 05/26/2024]
Abstract
Rampant pathogenesis induced by communicable microbes has necessitated development of technologies for rapid and sustained disinfection of surfaces. Copper nanoparticles (CuNPs) have been widely reported for their antimicrobial properties. However, nanostructured copper is prone to oxidative dissolution in the oil phase limiting its sustained use on surfaces and coatings. The current study reports a systematic investigation of a simple synthesis protocol using fatty acid stabilizers (particularly essential oils) for synthesis of copper nanoparticles in the oil phase. Of the various formulations synthesized, rosemary oil stabilized copper nanoparticles (RMO CuNPs) were noted to have the best inactivation kinetics and were also most stable. Upon morphological characterization by TEM and EELS, these were found to be monodispersed (φ5-8 nm) with copper coexisting in all three oxidation states on the surface of the nanoparticles. The nanoparticles were drop cast on woven fabric of around 500 threads per inch and exposed to gram positive bacteria (Staphylococcus aureus), gram negative bacteria (Escherichia coliandPseudomonas aeruginosa), enveloped RNA virus (phi6), non-enveloped RNA virus (MS2) and non-enveloped DNA virus (T4) to encompass the commonly encountered groups of pathogens. It was possible to completely disinfect 107copies of all microorganisms within 40 min of exposure. Further, this formulation was incorporated with polyurethane as thinners and used to coat non-woven fabrics. These also exhibited antimicrobial properties. Sustained disinfection with less than 9% cumulative copper loss for upto 14 washes with soap water was observed while the antioxidant activity was also preserved. Based on the studies conducted, RMO CuNP in oil phase was found to have excellent potential of integration on surface coatings, paints and polymers for rapid and sustained disinfection of microbes on surfaces.
Collapse
Affiliation(s)
- Pooja Nag
- Department of Mechatronics, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | - Kapil Sadani
- Department of Instrumentation and Control, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | - Lakshmi Pisharody
- Environmental Science and Engineering Department, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Xiao Yun Thian
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Tadi Sai Ratnakar
- Department of Mechatronics, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | - Arhama Ansari
- Environmental Science and Engineering Department, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Suparna Mukherji
- Environmental Science and Engineering Department, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Soumyo Mukherji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
15
|
Manrique PD, Leus IV, López CA, Mehla J, Malloci G, Gervasoni S, Vargiu AV, Kinthada RK, Herndon L, Hengartner NW, Walker JK, Rybenkov VV, Ruggerone P, Zgurskaya HI, Gnanakaran S. Predicting permeation of compounds across the outer membrane of P. aeruginosa using molecular descriptors. Commun Chem 2024; 7:84. [PMID: 38609430 PMCID: PMC11015012 DOI: 10.1038/s42004-024-01161-y] [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: 10/04/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The ability Gram-negative pathogens have at adapting and protecting themselves against antibiotics has increasingly become a public health threat. Data-driven models identifying molecular properties that correlate with outer membrane (OM) permeation and growth inhibition while avoiding efflux could guide the discovery of novel classes of antibiotics. Here we evaluate 174 molecular descriptors in 1260 antimicrobial compounds and study their correlations with antibacterial activity in Gram-negative Pseudomonas aeruginosa. The descriptors are derived from traditional approaches quantifying the compounds' intrinsic physicochemical properties, together with, bacterium-specific from ensemble docking of compounds targeting specific MexB binding pockets, and all-atom molecular dynamics simulations in different subregions of the OM model. Using these descriptors and the measured inhibitory concentrations, we design a statistical protocol to identify predictors of OM permeation/inhibition. We find consistent rules across most of our data highlighting the role of the interaction between the compounds and the OM. An implementation of the rules uncovered in our study is shown, and it demonstrates the accuracy of our approach in a set of previously unseen compounds. Our analysis sheds new light on the key properties drug candidates need to effectively permeate/inhibit P. aeruginosa, and opens the gate to similar data-driven studies in other Gram-negative pathogens.
Collapse
Affiliation(s)
- Pedro D Manrique
- Physics Department, George Washington University, Washington, 20052, DC, USA.
| | - Inga V Leus
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - César A López
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - Jitender Mehla
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Silvia Gervasoni
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Attilio V Vargiu
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Rama K Kinthada
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, 63103, MO, USA
| | - Liam Herndon
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - Nicolas W Hengartner
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA
| | - John K Walker
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, 63103, MO, USA
| | - Valentin V Rybenkov
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Monserrato, 20052, CA, Italy
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, 73019, OK, USA
| | - S Gnanakaran
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, 87545, NM, USA.
| |
Collapse
|
16
|
Dombach JL, Christensen GL, Allgood SC, Quintana JLJ, Detweiler CS. Inhibition of multiple staphylococcal growth states by a small molecule that disrupts membrane fluidity and voltage. mSphere 2024; 9:e0077223. [PMID: 38445864 PMCID: PMC10964410 DOI: 10.1128/msphere.00772-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 03/07/2024] Open
Abstract
New molecular approaches to disrupting bacterial infections are needed. The bacterial cell membrane is an essential structure with diverse potential lipid and protein targets for antimicrobials. While rapid lysis of the bacterial cell membrane kills bacteria, lytic compounds are generally toxic to whole animals. In contrast, compounds that subtly damage the bacterial cell membrane could disable a microbe, facilitating pathogen clearance by the immune system with limited compound toxicity. A previously described small molecule, D66, terminates Salmonella enterica serotype Typhimurium (S. Typhimurium) infection of macrophages and reduces tissue colonization in mice. The compound dissipates bacterial inner membrane voltage without rapid cell lysis under broth conditions that permeabilize the outer membrane or disable efflux pumps. In standard media, the cell envelope protects Gram-negative bacteria from D66. We evaluated the activity of D66 in Gram-positive bacteria because their distinct envelope structure, specifically the absence of an outer membrane, could facilitate mechanism of action studies. We observed that D66 inhibited Gram-positive bacterial cell growth, rapidly increased Staphylococcus aureus membrane fluidity, and disrupted membrane voltage while barrier function remained intact. The compound also prevented planktonic staphylococcus from forming biofilms and a disturbed three-dimensional structure in 1-day-old biofilms. D66 furthermore reduced the survival of staphylococcal persister cells and of intracellular S. aureus. These data indicate that staphylococcal cells in multiple growth states germane to infection are susceptible to changes in lipid packing and membrane conductivity. Thus, agents that subtly damage bacterial cell membranes could have utility in preventing or treating disease.IMPORTANCEAn underutilized potential antibacterial target is the cell membrane, which supports or associates with approximately half of bacterial proteins and has a phospholipid makeup distinct from mammalian cell membranes. Previously, an experimental small molecule, D66, was shown to subtly damage Gram-negative bacterial cell membranes and to disrupt infection of mammalian cells. Here, we show that D66 increases the fluidity of Gram-positive bacterial cell membranes, dissipates membrane voltage, and inhibits the human pathogen Staphylococcus aureus in several infection-relevant growth states. Thus, compounds that cause membrane damage without lysing cells could be useful for mitigating infections caused by S. aureus.
Collapse
Affiliation(s)
- Jamie L. Dombach
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Grace L. Christensen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samual C. Allgood
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Joaquin L. J. Quintana
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Corrella S. Detweiler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
17
|
Bhattacharjya S, Zhang Z, Ramamoorthy A. LL-37: Structures, Antimicrobial Activity, and Influence on Amyloid-Related Diseases. Biomolecules 2024; 14:320. [PMID: 38540740 PMCID: PMC10968335 DOI: 10.3390/biom14030320] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 11/11/2024] Open
Abstract
Antimicrobial peptides (AMPs), as well as host defense peptides (HDPs), constitute the first line of defense as part of the innate immune system. Humans are known to express antimicrobial precursor proteins, which are further processed to generate AMPs, including several types of α/β defensins, histatins, and cathelicidin-derived AMPs like LL37. The broad-spectrum activity of AMPs is crucial to defend against infections caused by pathogenic bacteria, viruses, fungi, and parasites. The emergence of multi-drug resistant pathogenic bacteria is of global concern for public health. The prospects of targeting antibiotic-resistant strains of bacteria with AMPs are of high significance for developing new generations of antimicrobial agents. The 37-residue long LL37, the only cathelicidin family of AMP in humans, has been the major focus for the past few decades of research. The host defense activity of LL37 is likely underscored by its expression throughout the body, spanning from the epithelial cells of various organs-testis, skin, respiratory tract, and gastrointestinal tract-to immune cells. Remarkably, apart from canonical direct killing of pathogenic organisms, LL37 exerts several other host defense activities, including inflammatory response modulation, chemo-attraction, and wound healing and closure at the infected sites. In addition, LL37 and its derived peptides are bestowed with anti-cancer and anti-amyloidogenic properties. In this review article, we aim to develop integrative, mechanistic insight into LL37 and its derived peptides, based on the known biophysical, structural, and functional studies in recent years. We believe that this review will pave the way for future research on the structures, biochemical and biophysical properties, and design of novel LL37-based molecules.
Collapse
Affiliation(s)
- Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Zhizhuo Zhang
- Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, The University of Michigan, Ann Arbor, MI 48109, USA;
| | - Ayyalusamy Ramamoorthy
- Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, Michigan Neuroscience Institute, The University of Michigan, Ann Arbor, MI 48109, USA;
- National High Magnetic Field Laboratory, Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
18
|
Berry L, Neale Q, Arora R, Ramirez D, Brizuela M, Domalaon R, Arthur G, Schweizer F. Exploring Structure-Activity Relationships of Niclosamide-Based Colistin Potentiators in Colistin-Resistant Gram-Negative Bacteria. Antibiotics (Basel) 2024; 13:43. [PMID: 38247602 PMCID: PMC10812775 DOI: 10.3390/antibiotics13010043] [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: 12/12/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Colistin is primarily used as a last resort antibiotic against highly resistant Gram-negative bacteria (GNB). Rising rates of colistin resistance, however, may limit future use of this agent. The anthelmintic drug niclosamide has been shown to enhance colistin activity in combination therapy, but a detailed structure-activity relationship (SAR) for niclosamide against GNB has yet to be studied. A series of niclosamide analogs were synthesized to perform an SAR, leading to the discovery of a lead compound that displayed comparable colistin-potentiating activity to niclosamide with reduced cytotoxicity. Overall, this work provides important insights into synthetic strategies for the future development of new niclosamide derivatives and demonstrates that toxicity to mammalian cells can be reduced while maintaining colistin potentiation.
Collapse
Affiliation(s)
- Liam Berry
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Quinn Neale
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Rajat Arora
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Danyel Ramirez
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Marc Brizuela
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Ronald Domalaon
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 3N4, Canada;
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (L.B.); (Q.N.); (R.A.); (D.R.); (M.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
19
|
Allgood SC, Su CC, Crooks AL, Meyer CT, Zhou B, Betterton MD, Barbachyn MR, Yu EW, Detweiler CS. Bacterial efflux pump modulators prevent bacterial growth in macrophages and under broth conditions that mimic the host environment. mBio 2023; 14:e0249223. [PMID: 37921493 PMCID: PMC10746280 DOI: 10.1128/mbio.02492-23] [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: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
IMPORTANCE Bacterial efflux pumps are critical for resistance to antibiotics and for virulence. We previously identified small molecules that inhibit efflux pumps (efflux pump modulators, EPMs) and prevent pathogen replication in host cells. Here, we used medicinal chemistry to increase the activity of the EPMs against pathogens in cells into the nanomolar range. We show by cryo-electron microscopy that these EPMs bind an efflux pump subunit. In broth culture, the EPMs increase the potency (activity), but not the efficacy (maximum effect), of antibiotics. We also found that bacterial exposure to the EPMs appear to enable the accumulation of a toxic metabolite that would otherwise be exported by efflux pumps. Thus, inhibitors of bacterial efflux pumps could interfere with infection not only by potentiating antibiotics, but also by allowing toxic waste products to accumulate within bacteria, providing an explanation for why efflux pumps are needed for virulence in the absence of antibiotics.
Collapse
Affiliation(s)
- Samual C. Allgood
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Chih-Chia Su
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Amy L. Crooks
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Christian T. Meyer
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado, USA
- Duet Biosystems, Nashville, Tennessee, USA
- Antimicrobial Research Consortium (ARC) Labs, Boulder, Colorado, USA
| | - Bojun Zhou
- Department of Physics, University of Colorado, Boulder, Colorado, USA
| | - Meredith D. Betterton
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Physics, University of Colorado, Boulder, Colorado, USA
- Center for Computational Biology, Flatiron Institute, New York, New York, USA
| | | | - Edward W. Yu
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Corrella S. Detweiler
- Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
20
|
Dang J, Shu J, Wang R, Yu H, Chen Z, Yan W, Zhao B, Ding L, Wang Y, Hu H, Li Z. The glycopatterns of Pseudomonas aeruginosa as a potential biomarker for its carbapenem resistance. Microbiol Spectr 2023; 11:e0200123. [PMID: 37861315 PMCID: PMC10714932 DOI: 10.1128/spectrum.02001-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/08/2023] [Indexed: 10/21/2023] Open
Abstract
Bacterial surface glycans are an attractive therapeutic target in response to antibiotics; however, current knowledge of the corresponding mechanisms is rather limited. Antimicrobial susceptibility testing, genome sequencing, and MALDI-TOF MS, commonly used in recent years to analyze bacterial resistance, are unable to rapidly and efficiently establish associations between glycans and resistance. The discovery of new antimicrobial strategies still requires the introduction of promising analytical methods. In this study, we applied lectin microarray technology and a machine-learning model to screen for important glycan structures associated with carbapenem-resistant P. aeruginosa. This work highlights that specific glycopatterns can be important biomarkers associated with bacterial antibiotic resistance, which promises to provide a rapid entry point for exploring new resistance mechanisms in pathogens.
Collapse
Affiliation(s)
- Jing Dang
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jian Shu
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Ruiying Wang
- Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, China
| | - Hanjie Yu
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zhuo Chen
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Wenbo Yan
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Bingxiang Zhao
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Li Ding
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yuzi Wang
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Huizheng Hu
- Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, China
| | - Zheng Li
- Laboratory of Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
21
|
Chang DH, Lee MR, Wang N, Lynn DM, Palecek SP. Establishing Quantifiable Guidelines for Antimicrobial α/β-Peptide Design: A Partial Least-Squares Approach to Improve Antimicrobial Activity and Reduce Mammalian Cell Toxicity. ACS Infect Dis 2023; 9:2632-2651. [PMID: 38014670 PMCID: PMC10807133 DOI: 10.1021/acsinfecdis.3c00468] [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] [Indexed: 11/29/2023]
Abstract
Antimicrobial peptides (AMPs) are promising candidates to combat pathogens that are resistant to conventional antimicrobial drugs because they operate through mechanisms that involve membrane disruption. However, the use of AMPs in clinical settings has been limited, at least in part, by their susceptibility to proteolytic degradation and their lack of selectivity toward pathogenic microbes vs mammalian cells. We recently reported on the design of α- and β-peptide oligomers structurally templated upon the naturally occurring α-helical AMP aurein 1.2. These α/β-peptide oligomers are more proteolytically stable than aurein 1.2 and have several other attributes that render them attractive as alternatives to conventional AMPs. This study describes the influence of peptide physicochemical properties on the broad-spectrum activity of aurein 1.2-based α/β-peptide mimics against nine bacterial, fungal, and mammalian cell lines. We used a partial least-squares regression (PLSR)-supervised machine learning model to quantify and visualize relationships between experimentally determined physicochemical properties (e.g., hydrophobicity, charge, and helicity) and experimentally measured cell-type-specific activities of 21 peptides in a 149-member α/β-peptide library. Using this approach, we identified several peptides that were predicted to exhibit enhanced broad-spectrum selectivity, a measure that evaluates antimicrobial activity relative to mammalian cell toxicity compared to aurein 1.2. Experimental validation demonstrated high model predictive performance, and characterization of compounds with the highest broad-spectrum selectivity revealed peptide hydrophobicity, helicity, and helical rigidity to be strong predictors of broad-spectrum selectivity. The most selective peptide identified from the model prediction has more than a 13-fold improvement in broad-spectrum selectivity than that of aurein 1.2, demonstrating the ability of using PLSR models to identify quantitative structure-function relationships for nonstandard amino acid-containing peptides. Overall, this work establishes quantifiable guidelines for the rational design of helical antimicrobial α/β-peptides and identifies promising new α/β-peptides with significantly reduced mammalian toxicities and improved antifungal and antibacterial activities relative to aurein 1.2.
Collapse
Affiliation(s)
- Douglas H. Chang
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - Myung-Ryul Lee
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - Nathan Wang
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - David M. Lynn
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Sean P. Palecek
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| |
Collapse
|
22
|
Bradley JS, Makieieva N, Tøndel C, Roilides E, Kelly MS, Patel M, Vaddady P, Maniar A, Zhang Y, Paschke A, Chen LF. Pharmacokinetics, Safety, and Tolerability of Imipenem/Cilastatin/Relebactam in Children with Confirmed or Suspected Gram-Negative Bacterial Infections: A Phase 1b, Open-Label, Single-Dose Clinical Trial. J Clin Pharmacol 2023; 63:1387-1397. [PMID: 37562063 DOI: 10.1002/jcph.2334] [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: 04/18/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Imipenem/cilastatin/relebactam is approved for the treatment of serious gram-negative bacterial infections in adults. This study assessed the pharmacokinetics (PK), safety, and tolerability of a single dose of imipenem/cilastatin/relebactam (with a fixed 2:1 ratio of imipenem/cilastatin to relebactam, and with a maximum dose of 15 mg/kg imipenem and 15 mg/kg cilastatin [≤500 mg imipenem and ≤500 mg cilastatin] and 7.5 mg/kg relebactam [≤250 mg relebactam]) in children with confirmed/suspected gram-negative bacterial infections receiving standard-of-care antibacterial therapy. In this phase 1, noncomparative study (ClinicalTrials.gov identifier, NCT03230916), PK parameters from 46 children were analyzed using both population modeling and noncompartmental analysis. The PK/pharmacodynamic (PD) target for imipenem was percent time of the dosing interval that unbound plasma concentration exceeded the minimum inhibitory concentration (%fT>MIC) of ≥30% (MIC = 2 mcg/mL). For relebactam, the PK/PD target was a free drug area under the plasma concentration-time curve (AUC) normalized to MIC (at 2 mcg/mL) of ≥8.0 (equivalent to an AUC from time zero extrapolated to infinity of ≥20.52 mcg·h/mL). Safety was assessed up to 14 days after drug infusion. For imipenem, the ranges for the geometric mean %fT>MIC and maximum concentration (Cmax ) across age cohorts were 56.5%-93.7% and 32.2-38.2 mcg/mL, respectively. For relebactam, the ranges of the geometric mean Cmax and AUC from 0 to 6 hours across age cohorts were 16.9-21.3 mcg/mL and 26.1-55.3 mcg·h/mL, respectively. In total, 8/46 (17%) children experienced ≥1 adverse events (AEs) and 2/46 (4%) children experienced nonserious AEs that were deemed drug related by the investigator. Imipenem and relebactam exceeded plasma PK/PD targets; single doses of imipenem/cilastatin/relebactam were well tolerated with no significant safety concerns identified. These results informed imipenem/cilastatin/relebactam dose selection for further pediatric clinical evaluation.
Collapse
Affiliation(s)
- John S Bradley
- Department of Pediatrics, University of California San Diego School of Medicine and Rady Children's Hospital of San Diego, San Diego, CA, USA
| | - Nataliia Makieieva
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, and Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Emmanuel Roilides
- Third Department of Pediatrics, Infectious Diseases Unit, School of Medicine, Aristotle University and Hippokration General Hospital, Thessaloniki, Greece
| | - Matthew S Kelly
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | - Pavan Vaddady
- Merck & Co. Inc, Rahway, NJ, USA
- Daiichi Sankyo, Inc., Basking Ridge, NJ, USA
| | | | | | | | | |
Collapse
|
23
|
Duda-Madej A, Viscardi S, Topola E. Meropenem/Vaborbactam: β-Lactam/β-Lactamase Inhibitor Combination, the Future in Eradicating Multidrug Resistance. Antibiotics (Basel) 2023; 12:1612. [PMID: 37998814 PMCID: PMC10668789 DOI: 10.3390/antibiotics12111612] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/04/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Due to the fact that there is a steadily increasing trend in the area of antimicrobial resistance in microorganisms, there is a need to look for new treatment alternatives. One of them is the search for new β-lactamase inhibitors and combining them with β-lactam antibiotics, with the aim of increasing the low-dose efficacy, as well as lowering the resistance potential of bacterial strains. This review presents the positive effect of meropenem in combination with a vaborbactam (MER-VAB). This latest antibiotic-inhibitor combination has found particular use in the treatment of infections with the etiology of carbapenem-resistant Enterobacterales (CRE), Gram-negative bacteria, with a high degree of resistance to available antimicrobial drugs.
Collapse
Affiliation(s)
- Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-368 Wrocław, Poland
| | - Szymon Viscardi
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (E.T.)
| | - Ewa Topola
- Faculty of Medicine, Wroclaw Medical University, Ludwika Pasteura 1, 50-367 Wrocław, Poland; (S.V.); (E.T.)
| |
Collapse
|
24
|
Nguyen HT, Venter H, Woolford L, Young KA, McCluskey A, Garg S, Sapula SS, Page SW, Ogunniyi AD, Trott DJ. Oral administration of a 2-aminopyrimidine robenidine analogue (NCL195) significantly reduces Staphylococcus aureus infection and reduces Escherichia coli infection in combination with sub-inhibitory colistin concentrations in a bioluminescent mouse model. Antimicrob Agents Chemother 2023; 67:e0042423. [PMID: 37695304 PMCID: PMC10583667 DOI: 10.1128/aac.00424-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/06/2023] [Indexed: 09/12/2023] Open
Abstract
We have previously reported promising in vivo activity of the first-generation 2-aminopyramidine robenidine analogue NCL195 against Gram-positive bacteria (GPB) when administered via the systemic route. In this study, we examined the efficacy of oral treatment with NCL195 (± low-dose colistin) in comparison to oral moxifloxacin in bioluminescent Staphylococcus aureus and Escherichia coli peritonitis-sepsis models. Four oral doses of 50 mg/kg NCL195, commencing immediately post-infection, were administered at 4 h intervals in the S. aureus peritonitis-sepsis model. We used a combination of four oral doses of 50 mg/kg NCL195 and four intraperitoneal doses of colistin at 0.125 mg/kg, 0.25 mg/kg, or 0.5 mg/kg in the E. coli peritonitis-sepsis model. Subsequently, the dose rates of four intraperitoneal doses of colistin were increased to 0.5 mg/kg, 1 mg/kg, or 2 mg/kg at 4 h intervals to treat a colistin-resistant E. coli infection. In the S. aureus infection model, oral treatment of mice with NCL195 resulted in significantly reduced S. aureus infection loads (P < 0.01) and longer survival times (P < 0.001) than vehicle-only treated mice. In the E. coli infection model, co-administration of NCL195 and graded doses of colistin resulted in a dose-dependent significant reduction in colistin-susceptible (P < 0.01) or colistin-resistant (P < 0.05) E. coli loads compared to treatment with colistin alone at similar concentrations. Our results confirm that NCL195 is a potential candidate for further preclinical development as a specific treatment for multidrug-resistant infections, either as a stand-alone antibiotic for GPB or in combination with sub-inhibitory concentrations of colistin for Gram-negative bacteria.
Collapse
Affiliation(s)
- Hang Thi Nguyen
- Australian Center for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Pharmacology, Toxicology, Internal Medicine and Diagnostics, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Lucy Woolford
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kelly A. Young
- Chemistry, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
| | - Adam McCluskey
- Chemistry, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
| | - Sanjay Garg
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sylvia S. Sapula
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | | | - Abiodun David Ogunniyi
- Australian Center for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Darren J. Trott
- Australian Center for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
25
|
López-Rodríguez JC, Hancock SJ, Li K, Crotta S, Barrington C, Suárez-Bonnet A, Priestnall SL, Aubé J, Wack A, Klenerman P, Bengoechea JA, Barral P. Type I interferons drive MAIT cell functions against bacterial pneumonia. J Exp Med 2023; 220:e20230037. [PMID: 37516912 PMCID: PMC10373297 DOI: 10.1084/jem.20230037] [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: 01/06/2023] [Revised: 05/31/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are abundant in the lung and contribute to host defense against infections. During bacterial infections, MAIT cell activation has been proposed to require T cell receptor (TCR)-mediated recognition of antigens derived from the riboflavin synthesis pathway presented by the antigen-presenting molecule MR1. MAIT cells can also be activated by cytokines in an MR1-independent manner, yet the contribution of MR1-dependent vs. -independent signals to MAIT cell functions in vivo remains unclear. Here, we use Klebsiella pneumoniae as a model of bacterial pneumonia and demonstrate that MAIT cell activation is independent of MR1 and primarily driven by type I interferons (IFNs). During Klebsiella infection, type I IFNs stimulate activation of murine and human MAIT cells, induce a Th1/cytotoxic transcriptional program, and modulate MAIT cell location within the lungs. Consequently, adoptive transfer or boosting of pulmonary MAIT cells protect mice from Klebsiella infection, with protection being dependent on direct type I IFN signaling on MAIT cells. These findings reveal type I IFNs as new molecular targets to manipulate MAIT cell functions during bacterial infections.
Collapse
Affiliation(s)
- Juan Carlos López-Rodríguez
- The Peter Gorer Department of Immunobiology, King’s College London, London, UK
- The Francis Crick Institute, London, UK
| | - Steven J. Hancock
- Wellcome-Wolfson Institute for Experimental Medicine. School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Kelin Li
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Alejandro Suárez-Bonnet
- The Francis Crick Institute, London, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
| | - Simon L. Priestnall
- The Francis Crick Institute, London, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Oxford, UK
| | - Jose A. Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine. School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King’s College London, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
26
|
Cang HQ, Quan XH, Chu XH, Liang Y, Yang X, Li J. Carbapenems versus β-lactam and β-lactamase inhibitors for treatment of nosocomial pneumonia: A systematic review and meta-analysis. Heliyon 2023; 9:e20108. [PMID: 37767465 PMCID: PMC10520732 DOI: 10.1016/j.heliyon.2023.e20108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/24/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Background Carbapenems and β-lactam and β-lactamase inhibitors (BLBLIs) have been used empirically in nosocomial pneumonia, but their efficacy and safety are controversial. Objective We carried out a systematic review with meta-analysis to evaluate the efficacy and safety of carbapenems versus BLBLIs against nosocomial pneumonia. Methods PubMed, Embase, Cochrane Central Register of Controlled Trials, CNKI, Wangfang, VIP and Sinomed were searched systematically through April 29, 2023 for clinical trials comparing carbapenems with BLBLIs for treatment of nosocomial pneumonia. Random-effects models were used to evaluate the impact of treatment on the risk ratio (RR) of all-cause mortality, clinical response, microbiologic response, resistance by Pseudomonas aeruginosa, adverse effects (AEs), and serious adverse effects. The quality of the evidence was assessed with the Cochrane risk of bias tool. The review was registerted in the INPLASY (INPLASY202340113). Results Seven randomized controlled trials containing 3306 patients met our inclusion criteria Our meta-analysis showed no significant difference in all-cause mortality (RR = 0.88, 95% confidence interval [CI] = 0.75-1.03, I2 = 0%) or clinical cure (1.02, 0.96-1.09, 30%) or clinical failure (1.19, 0.97-1.47, 0%) or microbiologic clinical cure (0.98, 0.89-1.06, 40%) or Pseudomonas aeruginosa resistance (RR 2.43, CI 0.86-6.81, 49%, P = 0.09) or adverse events (0.98, 0.93-1.02, 0%) between carbapenems groups versus BLBLIs groups, but a significant difference was found for severe adverse events (RR 0.83, CI 0.73-0.94, 0%). Conclusion Differences in the prevalence of mortality, clinical cure, or clinical failure were not observed between carbapenems groups versus BLBLIs groups in terms of nosocomial pneumonia. The use of carbapenems was linked to a tendency towards the emergence of P. aeruginosa resistance, however, no statistically significant difference was observed.
Collapse
Affiliation(s)
- Huai Qin Cang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| | - Xiang Hua Quan
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| | - Xiang Hua Chu
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| | - Yu Liang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| | - Xue Yang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| | - Jing Li
- Department of Pharmacy, Affiliated Hospital of Qingdao University, China
| |
Collapse
|
27
|
Allgood SC, Su CC, Crooks AL, Meyer CT, Zhou B, Betterton MD, Barbachyn MR, Yu EW, Detweiler CS. Bacterial Efflux Pump Modulators Prevent Bacterial Growth in Macrophages and Under Broth Conditions that Mimic the Host Environment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558466. [PMID: 37786697 PMCID: PMC10541609 DOI: 10.1101/2023.09.20.558466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
New approaches for combatting microbial infections are needed. One strategy for disrupting pathogenesis involves developing compounds that interfere with bacterial virulence. A critical molecular determinant of virulence for Gram-negative bacteria are efflux pumps of the resistance-nodulation-division (RND) family, which includes AcrAB-TolC. We previously identified small molecules that bind AcrB, inhibit AcrAB-TolC, and do not appear to damage membranes. These efflux pump modulators (EPMs) were discovered in an in-cell screening platform called SAFIRE (Screen for Anti-infectives using Fluorescence microscopy of IntracellulaR Enterobacteriaceae). SAFIRE identifies compounds that disrupt the growth of a Gram-negative human pathogen, Salmonella enterica serotype Typhimurium (S. Typhimurium) in macrophages. We used medicinal chemistry to iteratively design ~200 EPM35 analogs and test them for activity in SAFIRE, generating compounds with nanomolar potency. Analogs were demonstrated to bind AcrB in a substrate binding pocket by cryo-electron microscopy (cryo-EM). Despite having amphipathic structures, the EPM analogs do not disrupt membrane voltage, as monitored by FtsZ localization to the cell septum. The EPM analogs had little effect on bacterial growth in standard Mueller Hinton Broth. However, under broth conditions that mimic the micro-environment of the macrophage phagosome, acrAB is required for growth, the EPM analogs are bacteriostatic, and increase the potency of antibiotics. These data suggest that under macrophage-like conditions the EPM analogs prevent the export of a toxic bacterial metabolite(s) through AcrAB-TolC. Thus, compounds that bind AcrB could disrupt infection by specifically interfering with the export of bacterial toxic metabolites, host defense factors, and/or antibiotics.
Collapse
Affiliation(s)
- Samual C Allgood
- Molecular, Cellular Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Chih-Chia Su
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Amy L Crooks
- Molecular, Cellular Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Christian T Meyer
- Molecular, Cellular Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
- Duet Biosystems, Nashville, TN, USA
- Antimicrobial Research Consortium (ARC) Labs, Boulder, CO, USA
| | - Bojun Zhou
- Department of Physics, University of Colorado, Boulder, CO, USA
| | - Meredith D Betterton
- Molecular, Cellular Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Physics, University of Colorado, Boulder, CO, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | | | - Edward W Yu
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Corrella S Detweiler
- Molecular, Cellular Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
28
|
Mohseni P, Ghorbani A, Fariborzi N. Exploring the potential of cold plasma therapy in treating bacterial infections in veterinary medicine: opportunities and challenges. Front Vet Sci 2023; 10:1240596. [PMID: 37720476 PMCID: PMC10502341 DOI: 10.3389/fvets.2023.1240596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Cold plasma therapy is a novel approach that has shown significant promise in treating bacterial infections in veterinary medicine. Cold plasma possesses the potential to eliminate various bacteria, including those that are resistant to antibiotics, which renders it a desirable substitute for traditional antibiotics. Furthermore, it can enhance the immune system and facilitate the process of wound healing. However, there are some challenges associated with the use of cold plasma in veterinary medicine, such as achieving consistent and uniform exposure to the affected area, determining optimal treatment conditions, and evaluating the long-term impact on animal health. This paper explores the potential of cold plasma therapy in veterinary medicine for managing bacterial diseases, including respiratory infections, skin infections, and wound infections such as Clostridium botulinum, Clostridium perfringens, Bacillus cereus, and Bacillus subtilis. It also shows the opportunities and challenges associated with its use. In conclusion, the paper highlights the promising potential of utilizing cold plasma in veterinary medicine. However, to gain a comprehensive understanding of its benefits and limitations, further research is required. Future studies should concentrate on refining treatment protocols and assessing the long-term effects of cold plasma therapy on bacterial infections and the overall health of animals.
Collapse
Affiliation(s)
- Parvin Mohseni
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Abozar Ghorbani
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute (NSTRI), Karaj, Iran
| | - Niloofar Fariborzi
- Department of Biology and Control of Diseases Vector, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Ilham D, Souad L, Asmae LH, Kawtar N, Mohammed T, Nabila S. Prevalence, antibiotic resistance profile, MBLs encoding genes, and biofilm formation among clinical carbapenem-resistant Enterobacterales isolated from patients in Mohammed VI University Hospital Centre, Morocco. Lett Appl Microbiol 2023; 76:ovad107. [PMID: 37699792 DOI: 10.1093/lambio/ovad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023]
Abstract
Enterobacterales are frequently a major cause of human infections. The emergence of carbapenem resistance as well as the biofilm formation complicate their management. In this regard, this study aimed to investigate the prevalence, antibiogram, carbapenemase genes, and biofilm production among Enterobacterales. For this purpose, 18 172 clinical specimens from hospitalized patients at Mohammed VI University Hospital were collected over two years (2018-2019). The bacteriological investigation was performed to isolate Enterobacterales. Subsequently, BD-Phoenix and MALDI-TOF-MS were used for bacterial identification. The production of ESBLs and carbapenemases was assessed using phenotypic tests and PCR. The biofilm formation was eventually carried out. Out of 195 carbapenem-resistant Enterobacterales strains, 190 were carbapenemase producers, and 74 Enterobacterales produced metallo-beta-lactamases (MBLs). The PCR results revealed that blaNDM was the most common carbapenemase gene, present in 62 cases, followed by the co-existence of blaNDM and blaOXA-48 in 12 cases. Klebsiella pneumoniae was the most frequently identified species among the 74 New Delhi metallo-β-lactamase (NDM) isolates and the XDR resistance phenotype was the most prevalent with 58.10%. Additionally, all 74 NDM-positive Enterobacterales were able to form biofilms, with 82.4% being strong producers. This study highlights the need for rapid detection of carbapenemase and biofilm production in our hospital to manage this health concern.
Collapse
Affiliation(s)
- Dilagui Ilham
- Laboratoire de Lutte Contre les Maladies Infectieuses, Faculty of Medicine and Pharmacy, University Cadi Ayyad, Marrakesh 40000, Morocco
- Laboratory of Microbiology-Virology of Ar-Razi Hospital, Mohammed VI University Hospital Centre, BP 2360, Marrakesh, Morocco
| | - Loqman Souad
- Laboratoire de Lutte Contre les Maladies Infectieuses, Faculty of Medicine and Pharmacy, University Cadi Ayyad, Marrakesh 40000, Morocco
- Laboratory of Microbiology-Virology of Ar-Razi Hospital, Mohammed VI University Hospital Centre, BP 2360, Marrakesh, Morocco
| | - Lamrani Hanchi Asmae
- Laboratoire de Lutte Contre les Maladies Infectieuses, Faculty of Medicine and Pharmacy, University Cadi Ayyad, Marrakesh 40000, Morocco
- Laboratory of Microbiology-Virology of Ar-Razi Hospital, Mohammed VI University Hospital Centre, BP 2360, Marrakesh, Morocco
| | - Nayme Kawtar
- Laboratory of Molecular Bacteriology, Pasteur Institute, Casablanca 20250, Morocco
| | - Timinouni Mohammed
- Laboratory of Molecular Bacteriology, Pasteur Institute, Casablanca 20250, Morocco
| | - Soraa Nabila
- Laboratoire de Lutte Contre les Maladies Infectieuses, Faculty of Medicine and Pharmacy, University Cadi Ayyad, Marrakesh 40000, Morocco
- Laboratory of Microbiology-Virology of Ar-Razi Hospital, Mohammed VI University Hospital Centre, BP 2360, Marrakesh, Morocco
| |
Collapse
|
30
|
Karim MR, Zakaria Z, Hassan L, Faiz NM, Ahmad NI. The occurrence and molecular detection of mcr-1 and mcr-5 genes in Enterobacteriaceae isolated from poultry and poultry meats in Malaysia. Front Microbiol 2023; 14:1208314. [PMID: 37601372 PMCID: PMC10435970 DOI: 10.3389/fmicb.2023.1208314] [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: 04/18/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
The advent of antimicrobials-resistant (AMR), including colistin-resistant bacteria, poses a significant challenge to animal and human health, food safety, socio-economic growth, and the global environment. This study aimed to ascertain the colistin resistance prevalence and molecular mechanisms of colistin resistance in Enterobacteriaceae. The colistin resistance was determined using broth microdilution assay, PCR; and Sanger sequencing of mcr genes responsible for colistin resistance in Enterobacteriaceae (n = 627), including Escherichia coli (436), Salmonella spp. (n = 140), and Klebsiella pneumoniae (n = 51), obtained from chicken and chicken meats. Out of 627 Enterobacteriaceae, 8.6% of isolates exhibited colistin resistance phenotypically. Among these colistin resistant isolates, 9.3% (n = 37) were isolated from chicken meat, 7.2% (n = 11) from the cloacal swab of chicken and 7.9% (n = 6) from the litter samples. Overall, 12.96% of colistin-resistant isolates were positive with mcr genes, in which mcr-1 and mcr-5 genes were determined in 11.11% and 1.85% of colistin-resistant isolates, respectively. The E. coli isolates obtained from chicken meats, cloacal swabs and litter samples were found positive for mcr-1, and Salmonella spp. originated from the chicken meat sample was observed with mcr-5, whereas no mcr genes were observed in K. pneumoniae strains isolated from any of the collected samples. The other colistin resistance genes, including mcr-2, mcr-3, mcr-4, mcr-6, mcr-7, mcr-8, mcr-9, and mcr-10 were not detected in the studied samples. The mcr-1 and mcr-5 genes were sequenced and found to be 100% identical to the mcr-1 and mcr-5 gene sequences available in the NCBI database. This is the first report of colistin resistance mcr-5 gene in Malaysia which could portend the emergence of mcr-5 harboring bacterial strains for infection. Further studies are needed to characterize the mr-5 harbouring bacteria for the determination of plasmid associated with mcr-5 gene.
Collapse
Affiliation(s)
- Md Rezaul Karim
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
- Bangladesh Livestock Research Institute, Dhaka, Bangladesh
| | - Zunita Zakaria
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
- Institute of Bioscience, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
| | - Latiffah Hassan
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
| | - Nik Mohd Faiz
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
| | - Nur Indah Ahmad
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
31
|
Elek CKA, Brown TL, Le Viet T, Evans R, Baker DJ, Telatin A, Tiwari SK, Al-Khanaq H, Thilliez G, Kingsley RA, Hall LJ, Webber MA, Adriaenssens EM. A hybrid and poly-polish workflow for the complete and accurate assembly of phage genomes: a case study of ten przondoviruses. Microb Genom 2023; 9:mgen001065. [PMID: 37463032 PMCID: PMC10438801 DOI: 10.1099/mgen.0.001065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/17/2023] [Indexed: 07/21/2023] Open
Abstract
Bacteriophages (phages) within the genus Przondovirus are T7-like podoviruses belonging to the subfamily Studiervirinae, within the family Autographiviridae, and have a highly conserved genome organisation. The genomes of these phages range from 37 to 42 kb in size, encode 50-60 genes and are characterised by the presence of direct terminal repeats (DTRs) flanking the linear chromosome. These DTRs are often deleted during short-read-only and hybrid assemblies. Moreover, long-read-only assemblies are often littered with sequencing and/or assembly errors and require additional curation. Here, we present the isolation and characterisation of ten novel przondoviruses targeting Klebsiella spp. We describe HYPPA, a HYbrid and Poly-polish Phage Assembly workflow, which utilises long-read assemblies in combination with short-read sequencing to resolve phage DTRs and correcting errors, negating the need for laborious primer walking and Sanger sequencing validation. Our assembly workflow utilised Oxford Nanopore Technologies for long-read sequencing for its accessibility, making it the more relevant long-read sequencing technology at this time, and Illumina DNA Prep for short-read sequencing, representing the most commonly used technologies globally. Our data demonstrate the importance of careful curation of phage assemblies before publication, and prior to using them for comparative genomics.
Collapse
Affiliation(s)
- Claire K. A. Elek
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich Research Park, Norwich, UK
| | - Teagan L. Brown
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Thanh Le Viet
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Rhiannon Evans
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - David J. Baker
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Andrea Telatin
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Sumeet K. Tiwari
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Haider Al-Khanaq
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Gaëtan Thilliez
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
| | - Robert A. Kingsley
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lindsay J. Hall
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich Research Park, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL—Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mark A. Webber
- Quadram Institute Bioscience, Rosalind Franklin Road, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich Research Park, Norwich, UK
| | | |
Collapse
|
32
|
Rashid MH, Bukhari SNY, Mousa A, Aziz AA, Hakobyan K. Cefiderocol as a Treatment Option for Stenotrophomonas maltophilia Causing Hospital-Acquired/Ventilator-Associated Pneumonia. Cureus 2023; 15:e38613. [PMID: 37284369 PMCID: PMC10239792 DOI: 10.7759/cureus.38613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/08/2023] Open
Abstract
Multidrug-resistant (MDR) gram-negative bacteria have been causing havoc for the healthcare system because of the rarity of the treatment options available. Stenotrophomonas maltophilia is a non-fermenting gram-negative bacterium that causes different infections, particularly respiratory tract infections. It displays resistance to many antibiotics (e.g., carbapenems, fluoroquinolones, and trimethoprim-sulfamethoxazole). Cefiderocol is a novel antibiotic which still in the preclinical stages of Food and Drug Administration (FDA) approval for S. maltophilia. We present the case of a 76-year-old male with end-stage renal disease (ESRD), intubated for acute hypoxemic respiratory failure due to volume overload and worsening oxygenation, who subsequently developed ventilator-associated pneumonia, found to be due to MDR Stenotrophomonas maltophilia. The patient ultimately showed clinical improvement with a 7-day course with a renally adjusted dose of cefiderocol. This shows that cefiderocol can prove to be a potential treatment option against serious infections caused by difficult-to-treat S. maltophilia.
Collapse
Affiliation(s)
| | | | - Aliaa Mousa
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| | - Ahmed Ali Aziz
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| | - Knkush Hakobyan
- Internal Medicine, Capital Health Regional Medical Center, Trenton, USA
| |
Collapse
|
33
|
You Y, Liu H, Zhu Y, Zheng H. Rational design of stapled antimicrobial peptides. Amino Acids 2023; 55:421-442. [PMID: 36781451 DOI: 10.1007/s00726-023-03245-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023]
Abstract
The global increase in antimicrobial drug resistance has dramatically reduced the effectiveness of traditional antibiotics. Structurally diverse antibiotics are urgently needed to combat multiple-resistant bacterial infections. As part of innate immunity, antimicrobial peptides have been recognized as the most promising candidates because they comprise diverse sequences and mechanisms of action and have a relatively low induction rate of resistance. However, because of their low chemical stability, susceptibility to proteases, and high hemolytic effect, their usage is subject to many restrictions. Chemical modifications such as D-amino acid substitution, cyclization, and unnatural amino acid modification have been used to improve the stability of antimicrobial peptides for decades. Among them, a side-chain covalent bridge modification, the so-called stapled peptide, has attracted much attention. The stapled side-chain bridge stabilizes the secondary structure, induces protease resistance, and increases cell penetration and biological activity. Recent progress in computer-aided drug design and artificial intelligence methods has also been used in the design of stapled antimicrobial peptides and has led to the successful discovery of many prospective peptides. This article reviews the possible structure-activity relationships of stapled antimicrobial peptides, the physicochemical properties that influence their activity (such as net charge, hydrophobicity, helicity, and dipole moment), and computer-aided methods of stapled peptide design. Antimicrobial peptides under clinical trial: Pexiganan (NCT01594762, 2012-05-07). Omiganan (NCT02576847, 2015-10-13).
Collapse
Affiliation(s)
- YuHao You
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - HongYu Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - YouZhuo Zhu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
34
|
Too RJ, Gitao GC, Bebora LC, Mollenkopf DF, Kariuki SM, Wittum TE. Frequency and diversity of carbapenemase-producing Enterobacterales recovered from untreated wastewater impacted by selective media containing cefotaxime and meropenem in Ohio, USA. PLoS One 2023; 18:e0281909. [PMID: 36812188 PMCID: PMC9946209 DOI: 10.1371/journal.pone.0281909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/03/2023] [Indexed: 02/24/2023] Open
Abstract
As safe agents of last resort, carbapenems are reserved for the treatment of infections caused by multidrug-resistant organisms. The impact of β-lactam antibiotics, cefotaxime, and meropenem on the frequency and diversity of carbapenemase-producing organisms recovered from environmental samples has not been fully established. Therefore, this methodological study aimed at determining β-lactam drugs used in selective enrichment and their impact on the recovery of carbapenemase-producing Enterobacterales (CPE) from untreated wastewater. We used a longitudinal study design where 1L wastewater samples were collected weekly from wastewater treatment plant (WWTP) influent and quarterly from contributing sanitary sewers in Columbus, Ohio USA with 52 total samples collected. Aliquots of 500 mL were passed through membrane filters of decreasing pore sizes to enable all the water to pass through and capture bacteria. For each sample, the resulting filters were placed into two modified MacConkey (MAC) broths, one supplemented with 0.5 μg/mL of meropenem and 70 μg/mL of ZnSO4 and the other supplemented with 2 μg/mL cefotaxime. The inoculated broth was then incubated at 37° C overnight, after which they were streaked onto two types of correspondingly-modified MAC agar plates supplemented with 0.5 μg/mL and 1.0 μg/mL of meropenem and 70 μg/mL of ZnSO4 and incubated at 37°C overnight. The isolates were identified based on morphological and biochemical characteristics. Then, up to four distinct colonies of each isolate's pure culture per sample were tested for carbapenemase production using the Carba-NP test. Matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry (MS) MALDI-TOF MS was used to identify carbapenemase-producing organisms. In total 391 Carba-NP positive isolates were recovered from the 52 wastewater samples: 305 (78%) isolates had blaKPC, 73 (19%) carried blaNDM, and 14 (4%) harbored both blaKPC and blaNDM resistance genes. CPE genes of both blaKPC and blaNDM were recovered in both types of modified MAC broths, with 84 (21%) having a blaKPC gene, 22 (6%) carrying blaNDM and 9 (2%) harbored both a blaKPC and blaNDM of isolates recovered from MAC medium incorporated with 0.5ug/mL meropenem and 70ug/mL ZnSO4. The most prevalent isolates were Klebsiella pneumoniae, Escherichia coli, and Citrobacter spp.
Collapse
Affiliation(s)
- Rael J. Too
- Kenya Medical Research Institute (KEMRI-Kenya), Nairobi, Kenya
- The University of Nairobi (UoN-Kenya), Nairobi, Kenya
- The Ohio State University (OSU-OH, USA), Columbus, OH, United States of America
- * E-mail:
| | | | | | - Dixie F. Mollenkopf
- The Ohio State University (OSU-OH, USA), Columbus, OH, United States of America
| | | | - Thomas E. Wittum
- The Ohio State University (OSU-OH, USA), Columbus, OH, United States of America
| |
Collapse
|
35
|
Muacevic A, Adler JR, Almefleh AA, Alamri AH, Alobud AS, Bawazeer RA, Alswaji AA, Alalwan B, Aldriwesh MG, Al Johani SM, Alghoribi MF. Prevalence of Carbapenem Non-susceptible Gram-Negative Bacteria at Tertiary Care Hospitals in Saudi Arabia. Cureus 2023; 15:e33767. [PMID: 36655153 PMCID: PMC9840728 DOI: 10.7759/cureus.33767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2023] [Indexed: 01/16/2023] Open
Abstract
Background Antibiotics significantly increased life expectancy and decreased mortality rates due to infections. However, this trend is starting to fade with the rise of multidrug-resistant organisms (MDR); these strains are becoming a global burden on healthcare and the economy. The dramatic increase and spread of carbapenem-resistant gram-negative bacteria (CRGNB) has become a serious global public health concern. In this retrospective cross-sectional study, we aimed to estimate the rates of gram-negative bacteremia in five tertiary care hospitals in different geographical locations in Saudi Arabia for five years. Methods A retrospective cross-sectional study was conducted in five tertiary care hospitals in Saudi Arabia among patients with bacteremia due to CRGNB. Electronic medical records were used to retrieve data regarding patient demographics and antimicrobial susceptibility testing (AST) over five years between January 2016 and December 2020. Patients with positive blood cultures for carbapenem-resistant Escherichia (E.) coli, Klebsiella (K.) pneumonia, Pseudomonas (P.) aeruginosa, and Acinetobacter (A.) baumannii comprise the final study population. Results This retrospective multicentric study was conducted between 2016 and 2020 in five tertiary care hospitals across five cities in Saudi Arabia. E. coli (n=2190, 38.03%), K. pneumoniae (n=2154, 37.41%), P. aeruginosa (n = 918, 15.94%), and A. baumannii (n=496, 8.61%) constitute the 5758 gram-negative bacteria isolates. E. coli was the most frequently identified species in Riyadh, AlAhsa, Dammam, and Madinah (40%, 46.50%, 61.67%, and 43.66%, respectively), with a p-value of (p<0.001), except in Jeddah, where K. pneumoniae was the most prevalent (42%). The mean age of patients across Riyadh, AlAhsa, Dammam, and Madinah was 62.2 years (± 4.24). In contrast to Jeddah, where the majority of isolates (702; 41.8%) belonged to the adult age group. Most isolates were from male patients (3045; 52.9%), compared to 2713 (47.1%) from female patients. K. pneumoniae 1226 (40.3%) was the most prevalent isolate among male patients while E. coli (1135; 41.8%) was the most prevalent isolate among female patients. Conclusion Our study showed that the prevalence of carbapenem non-susceptible Gram-negative bacteria is relatively high, which therefore makes them very challenging to treat. The results show an urgent need for improved antibiotic stewardship strategies, including better surveillance and more effective infection control measures to reduce this issue. Further research into the molecular epidemiology and risk factors associated with these infections is necessary to guide public health policymakers in developing interventions to help control the spread of carbapenem-resistant Gram-negative bacteria.
Collapse
|
36
|
Sinha S, Dhanabal VB, Manivannen VL, Cappiello F, Tan SM, Bhattacharjya S. Ultra-Short Cyclized β-Boomerang Peptides: Structures, Interactions with Lipopolysaccharide, Antibiotic Potentiator and Wound Healing. Int J Mol Sci 2022; 24:ijms24010263. [PMID: 36613707 PMCID: PMC9820106 DOI: 10.3390/ijms24010263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Many antibiotics are ineffective in killing Gram-negative bacteria due to the permeability barrier of the outer-membrane LPS. Infections caused by multi-drug-resistant Gram-negative pathogens require new antibiotics, which are often difficult to develop. Antibiotic potentiators disrupt outer-membrane LPS and can assist the entry of large-scaffold antibiotics to the bacterial targets. In this work, we designed a backbone-cyclized ultra-short, six-amino-acid-long (WKRKRY) peptide, termed cWY6 from LPS binding motif of β-boomerang bactericidal peptides. The cWY6 peptide does not exhibit any antimicrobial activity; however, it is able to permeabilize the LPS outer membrane. Our results demonstrate the antibiotic potentiator activity in the designed cWY6 peptide for several conventional antibiotics (vancomycin, rifampicin, erythromycin, novobiocin and azithromycin). Remarkably, the short cWY6 peptide exhibits wound-healing activity in in vitro assays. NMR, computational docking and biophysical studies describe the atomic-resolution structure of the peptide in complex with LPS and mode of action in disrupting the outer membrane. The dual activities of cWY6 peptide hold high promise for further translation to therapeutics.
Collapse
Affiliation(s)
- Sheetal Sinha
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Advanced Environmental Biotechnology Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, 1 Cleantech Loop, Singapore 637141, Singapore
- Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Vidhya Bharathi Dhanabal
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Veronica Lavanya Manivannen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Floriana Cappiello
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185 Rome, Italy
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Correspondence:
| |
Collapse
|
37
|
Krishna A, Zere T, Mistry S, Ismaiel O, Stone H, Sacks LV, Weaver JL. Evaluation of a Sequential Antibiotic Treatment Regimen of Ampicillin, Ciprofloxacin and Fosfomycin against Escherichia coli CFT073 in the Hollow Fiber Infection Model Compared with Simultaneous Combination Treatment. Antibiotics (Basel) 2022; 11:antibiotics11121705. [PMID: 36551362 PMCID: PMC9774593 DOI: 10.3390/antibiotics11121705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Employ the hollow fiber infection model (HFIM) to study sequential antibiotic administration (ampicillin, ciprofloxacin and fosfomycin) using human pharmacokinetic profiles to measure changes in the rate of antibiotic resistance development and compare this to simultaneous combination therapy with the same antibiotic combinations. METHODS Escherichia coli CFT073, a clinical uropathogenic strain, was exposed individually to clinically relevant pharmacokinetic concentrations of ampicillin on day 1, ciprofloxacin on day 2 and fosfomycin on day 3. This sequence was continued for 10 days in the HFIM. Bacterial samples were collected at different time points to enumerate total and resistant bacterial populations. The results were compared with the simultaneous combination therapy previously studied. RESULTS Sequential antibiotic treatment (ampicillin-ciprofloxacin-fosfomycin sequence) resulted in the early emergence of single and multi-antibiotic-resistant subpopulations, while the simultaneous treatment regimen significantly delayed or prevented the emergence of resistant subpopulations. CONCLUSION Sequential administration of these antibiotic monotherapies did not significantly delay the emergence of resistant subpopulations compared to simultaneous treatment with combinations of the same antibiotics. Further studies are warranted to evaluate different sequences of the same antibiotics in delaying emergent resistance.
Collapse
Affiliation(s)
- Ashok Krishna
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
- Correspondence:
| | - Tesfalem Zere
- Office of Product Evaluation and Quality, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Sabyasachy Mistry
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Omnia Ismaiel
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Heather Stone
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Leonard V. Sacks
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - James L. Weaver
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
38
|
Mongalo NI, Raletsena MV. An Inventory of South African Medicinal Plants Used in the Management of Sexually Transmitted and Related Opportunistic Infections: An Appraisal and Some Scientific Evidence (1990-2020). PLANTS (BASEL, SWITZERLAND) 2022; 11:3241. [PMID: 36501281 PMCID: PMC9738887 DOI: 10.3390/plants11233241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 06/17/2023]
Abstract
The current work is aimed at generating the first inventory of South African medicinal plants used in the treatment of sexually transmitted and related opportunistic infections associated with HIV-AIDS. This is important in assisting researchers to access a list of plant species to evaluate for potential phytocompounds, as this area of research is greatly lagging in South Africa. A total of 335 medicinal plants from 103 families have been documented in the current work. The most represented families are Fabaceae (11.64%) and Asteraceae (6.27%). Herbs constitute 36.53%, trees 32.34%, shrubs 29.04%, climbers 1.80% and parasites 0.30%. It is worrying that on the plant parts used, the roots constitute 47.18%, while leaves and stem bark yield 16.62 and 15.01%, respectively. Catharanthus roseus exhibited the highest number of citations (19), while Peltophorum africanum had 14 and both Carica papaya and Vachelia karoo had 12. In the mode of administration of the reported medicinal plant species, most of the plants are boiled and taken orally (48.22%), while other plant species are used as mouth washes (3.25%). Although there is reasonable in vitro activity of some of the plant species, validating the relevance of use, there is still a need to explore the mode of action of such plant species; isolated compounds and possible derivatives thereof are of paramount importance and need to be explored as well. Furthermore, toxicological aspects of such plant species need to be explored.
Collapse
Affiliation(s)
- Nkoana I. Mongalo
- The College of Agriculture and Environmental Sciences (CAES), University of South Africa, Johannesburg 2092, South Africa
| | | |
Collapse
|
39
|
In Vitro Activity of Robenidine Analogues NCL259 and NCL265 against Gram-Negative Pathogens. Antibiotics (Basel) 2022; 11:antibiotics11101301. [PMID: 36289959 PMCID: PMC9598656 DOI: 10.3390/antibiotics11101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Multidrug-resistant (MDR) Gram-negative pathogens, especially Acinetobacter baumannii, Pseudomonas aeruginosa, Escherichia coli and Enterobacter spp., are recognized by the World Health Organization as the most critical priority pathogens in urgent need of drug development. In this study, the in vitro antimicrobial activity of robenidine analogues NCL259 and NCL265 was tested against key human and animal Gram-negative clinical isolates and reference strains. NCL259 and NCL265 demonstrated moderate antimicrobial activity against these Gram-negative priority pathogens with NCL265 consistently more active, achieving lower minimum inhibitory concentrations (MICs) in the range of 2−16 µg/mL. When used in combination with sub-inhibitory concentrations of polymyxin B to permeabilize the outer membrane, NCL259 and NCL265 elicited a synergistic or additive activity against the reference strains tested, reducing the MIC of NCL259 by 8- to 256- fold and the MIC of NCL265 by 4- to 256- fold. A small minority of Klebsiella spp. isolates (three) were resistant to both NCL259 and NCL265 with MICs > 256 µg/mL. This resistance was completely reversed in the presence of the efflux pump inhibitor phenylalanine-arginine-beta-naphthylamide (PAβN) to yield MIC values of 8−16 µg/mL and 2−4 µg/mL for NCL259 and NCL256, respectively. When NCL259 and NCL265 were tested against wild-type E. coli isolate BW 25113 and its isogenic multidrug efflux pump subunit AcrB deletion mutant (∆AcrB), the MIC of both compounds against the mutant ∆AcrB isolate was reduced 16-fold compared to the wild-type parent, indicating a significant role for the AcrAB-TolC efflux pump from Enterobacterales in imparting resistance to these robenidine analogues. In vitro cytotoxicity testing revealed that NCL259 and NCL265 had much higher levels of toxicity to a range of human cell lines compared to the parent robenidine, thus precluding their further development as novel antibiotics against Gram-negative pathogens.
Collapse
|
40
|
Curcumin Stimulates the Overexpression of Virulence Factors in Salmonella enterica Serovar Typhimurium: In Vitro and Animal Model Studies. Antibiotics (Basel) 2022; 11:antibiotics11091230. [PMID: 36140009 PMCID: PMC9494991 DOI: 10.3390/antibiotics11091230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Salmonella spp. is one of the most common food poisoning pathogens and the main cause of diarrheal diseases in humans in developing countries. The increased Salmonella resistance to antimicrobials has led to the search for new alternatives, including natural compounds such as curcumin, which has already demonstrated a bactericidal effect; however, in Gram-negatives, there is much controversy about this effect, as it is highly variable. In this study, we aimed to verify the antibacterial activity of curcumin against the Salmonella enterica serovar Typhimurium growth rate, virulence, and pathogenicity. The strain was exposed to 110, 220 or 330 µg/mL curcumin, and by complementary methods (spectrophotometric, pour plate and MTT assays), we determined its antibacterial activity. To elucidate whether curcumin regulates the expression of virulence genes, Salmonella invA, fliC and siiE genes were investigated by quantitative real-time reverse transcription (qRT-PCR). Furthermore, to explore the effect of curcumin on the pathogenesis process in vivo, a Caenorhabditis elegans infection model was employed. No antibacterial activity was observed, even at higher concentrations of curcumin. All concentrations of curcumin caused overgrowth (35−69%) and increased the pathogenicity of the bacterial strain through the overexpression of virulence factors. The latter coincided with a significant reduction in both the lifespan and survival time of C. elegans when fed with curcumin-treated bacteria. Our data provide relevant information that may support the selective antibacterial effects of curcumin to reconsider the indiscriminate use of this phytochemical, especially in outbreaks of pathogenic Gram-negative bacteria.
Collapse
|
41
|
Rizk NA, Zahreddine N, Haddad N, Ahmadieh R, Hannun A, Bou Harb S, Haddad SF, Zeenny RM, Kanj SS. The Impact of Antimicrobial Stewardship and Infection Control Interventions on Acinetobacter baumannii Resistance Rates in the ICU of a Tertiary Care Center in Lebanon. Antibiotics (Basel) 2022; 11:911. [PMID: 35884165 PMCID: PMC9311570 DOI: 10.3390/antibiotics11070911] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance is a serious threat to global health, causing increased mortality and morbidity especially among critically ill patients. This toll is expected to rise following the COVID-19 pandemic. Carbapenem-resistant Acinetobacter baumannii (CRAb) is among the Gram-negative pathogens leading antimicrobial resistance globally; it is listed as a critical priority pathogen by the WHO and is implicated in hospital-acquired infections and outbreaks, particularly in critically ill patients. Recent reports from Lebanon describe increasing rates of infection with CRAb, hence the need to develop concerted interventions to control its spread. We set to describe the impact of combining antimicrobial stewardship and infection control measures on resistance rates and colonization pressure of CRAb in the intensive care units of a tertiary care center in Lebanon before the COVID-19 pandemic. The antimicrobial stewardship program introduced a carbapenem-sparing initiative in April 2019. During the same period, infection control interventions involved focused screening, monitoring, and tracking of CRAb, as well as compliance with specific measures. From January 2018 to January 2020, we report a statistically significant decrease in carbapenem consumption and a decrease in resistance rates of isolated A. baumannii. The colonization pressure of CRAb also decreased significantly, reaching record low levels at the end of the intervention period. The results indicate that a multidisciplinary approach and combined interventions between the stewardship and infection control teams can lead to a sustained reduction in resistance rates and CRAb spread in ICUs.
Collapse
Affiliation(s)
- Nesrine A. Rizk
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.A.R.); (S.B.H.); (S.F.H.)
| | - Nada Zahreddine
- Infection Control and Prevention Program, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.Z.); (R.A.)
| | - Nisrine Haddad
- Department of Pharmacy, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.H.); (A.H.); (R.M.Z.)
| | - Rihab Ahmadieh
- Infection Control and Prevention Program, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.Z.); (R.A.)
| | - Audra Hannun
- Department of Pharmacy, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.H.); (A.H.); (R.M.Z.)
| | - Souad Bou Harb
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.A.R.); (S.B.H.); (S.F.H.)
| | - Sara F. Haddad
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.A.R.); (S.B.H.); (S.F.H.)
| | - Rony M. Zeenny
- Department of Pharmacy, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.H.); (A.H.); (R.M.Z.)
| | - Souha S. Kanj
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon; (N.A.R.); (S.B.H.); (S.F.H.)
| |
Collapse
|
42
|
Gaibani P, Giani T, Bovo F, Lombardo D, Amadesi S, Lazzarotto T, Coppi M, Rossolini GM, Ambretti S. Resistance to Ceftazidime/Avibactam, Meropenem/Vaborbactam and Imipenem/Relebactam in Gram-Negative MDR Bacilli: Molecular Mechanisms and Susceptibility Testing. Antibiotics (Basel) 2022; 11:antibiotics11050628. [PMID: 35625273 PMCID: PMC9137602 DOI: 10.3390/antibiotics11050628] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 01/25/2023] Open
Abstract
Multidrug resistance (MDR) represents a serious global threat due to the rapid global spread and limited antimicrobial options for treatment of difficult-to-treat (DTR) infections sustained by MDR pathogens. Recently, novel β-lactams/β-lactamase inhibitor combinations (βL-βLICs) have been developed for the treatment of DTR infections due to MDR Gram-negative pathogens. Although novel βL-βLICs exhibited promising in vitro and in vivo activities against MDR pathogens, emerging resistances to these novel molecules have recently been reported. Resistance to novel βL-βLICs is due to several mechanisms including porin deficiencies, increasing carbapenemase expression and/or enzyme mutations. In this review, we summarized the main mechanisms related to the resistance to ceftazidime/avibactam, meropenem/vaborbactam and imipenem/relebactam in MDR Gram-negative micro-organisms. We focused on antimicrobial activities and resistance traits with particular regard to molecular mechanisms related to resistance to novel βL-βLICs. Lastly, we described and discussed the main detection methods for antimicrobial susceptibility testing of such molecules. With increasing reports of resistance to novel βL-βLICs, continuous attention should be maintained on the monitoring of the phenotypic traits of MDR pathogens, into the characterization of related mechanisms, and on the emergence of cross-resistance to these novel antimicrobials.
Collapse
Affiliation(s)
- Paolo Gaibani
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
- Correspondence:
| | - Tommaso Giani
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Federica Bovo
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Donatella Lombardo
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Stefano Amadesi
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Tiziana Lazzarotto
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40100 Bologna, Italy
| | - Marco Coppi
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Gian Maria Rossolini
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Simone Ambretti
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| |
Collapse
|
43
|
In Vitro Potency and Spectrum of the Novel Polymyxin MRX-8 Tested against Clinical Isolates of Gram-Negative Bacteria. Antimicrob Agents Chemother 2022; 66:e0013922. [PMID: 35475635 DOI: 10.1128/aac.00139-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The polymyxins display excellent in vitro antimicrobial activity against most Enterobacterales, Pseudomonas aeruginosa, and Acinetobacter baumannii isolates, but their clinical utility has been limited because of class-specific toxicity problems. Therefore, new polymyxin analogs with improved safety properties are needed to combat serious infections caused by resistant Gram-negative pathogens. MRX-8 is a novel polymyxin B analog that displays reduced toxicity in in vitro and animal assays and is currently being evaluated in a phase 1 clinical trial. In this nonclinical study, the in vitro potency and spectrum of MRX-8 and comparators were evaluated against a large set of Gram-negative clinical isolates collected in the United States in 2017 to 2020. MRX-8, colistin, and polymyxin B exhibited nearly identical antimicrobial activities against the Enterobacterales, Pseudomonas aeruginosa, and Acinetobacter baumannii isolate sets. MRX-8 MIC50 and MIC90 values were 0.12 and 0.25 mg/L, respectively, for the set of Enterobacterales isolates not intrinsically resistant to colistin and 0.5 and 1 mg/L, respectively, against both the A. baumannii and P. aeruginosa isolate sets. All three polymyxin-class compounds retained activity against meropenem-resistant and multidrug-resistant isolate subsets but were inactive against isolates displaying acquired or intrinsic resistance to polymyxins. These results support the continued development of MRX-8 to treat serious Gram-negative infections.
Collapse
|
44
|
Önal U, Akyol D, Mert M, Başkol D, Memetali SC, Şanlıdağ G, Kenanoğlu B, Uyan-Önal A, Quliyeva G, Avşar CB, Akdağ D, Demir M, Erdem HA, Kahraman Ü, Bozbıyık O, Özgiray E, Bozkurt D, Akarca FK, Demirağ K, Çankayalı İ, Uyar M, Çilli F, Arda B, Yamazhan T, Pullukçu H, Taşbakan MI, Sipahi H, Ulusoy S, Sipahi OR. Carbapenem-resistant Gram-negative pathogens associated with septic shock: a review of 120 cases. J Chemother 2022; 34:436-445. [PMID: 35446235 DOI: 10.1080/1120009x.2022.2064703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study aimed to evaluate the influencing variables for outcomes in patients with septic shock having culture-proven carbapenem-resistant Gram-negative pathogens. It included 120 patients (mean age 64.29 ± 1.35 years and 58.3% female). The mean Sequential Organ Failure Assessment score during septic shock diagnosis was found to be 11.22 ± 0.43 and 9 ± 0.79 among the patients with mortality and among the survivors, respectively (P = 0.017). The logistic regression analysis showed that empirical treatment as mono Gram-negative bacteria-oriented antibiotic therapy (P = 0.016, odds ratio (OR) = 17.730, 95% confidence interval (CI): 1.728-182.691), Charlson Comorbidity Index >2 (P = 0.032, OR = 7.312, 95% CI: 5.7-18.3), and systemic inflammatory response syndrome score 3 or 4 during septic shock diagnosis (P = 0.014, OR = 5.675, 95% CI: 1.424-22.619) were found as independent risk factors for day 30 mortality. Despite early diagnosis and effective management of patients with septic shock, the mortality rates are quite high in CRGNP-infected patients.
Collapse
Affiliation(s)
- Uğur Önal
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey.,Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Uludag University, Bursa, Turkey
| | - Deniz Akyol
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Merve Mert
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Dilşah Başkol
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Seichan Chousein Memetali
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Gamze Şanlıdağ
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Buse Kenanoğlu
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Ayşe Uyan-Önal
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey.,Yüksek İhtisas Research and Teaching Hospital, Department of Infectious Diseases and Clinical Microbiology, Bursa, Turkey
| | - Günel Quliyeva
- Bona Dea International Hospital, Infectious Diseases Clinic, Baku, Azerbaijan
| | - Cansu Bulut Avşar
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Damla Akdağ
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Melike Demir
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Hüseyin Aytaç Erdem
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Ümit Kahraman
- Faculty of Medicine, Department of Cardiovascular Surgery, Ege University, Bornova, Izmir, Turkey
| | - Osman Bozbıyık
- Faculty of Medicine, Department of General Surgery, Ege University, Bornova, Izmir, Turkey
| | - Erkin Özgiray
- Faculty of Medicine, Department of Neurosurgery, Ege University, Bornova, Izmir, Turkey
| | - Devrim Bozkurt
- Faculty of Medicine, Department of Internal Medicine, Ege University, Bornova, Izmir, Turkey
| | - Funda Karbek Akarca
- Faculty of Medicine, Department of Emergency Medicine, Ege University, Bornova, Izmir, Turkey
| | - Kubilay Demirağ
- Faculty of Medicine, Department of Anaesthesiology and Reanimation, Ege University, Bornova, Izmir, Turkey
| | - İlkin Çankayalı
- Faculty of Medicine, Department of Anaesthesiology and Reanimation, Ege University, Bornova, Izmir, Turkey
| | - Mehmet Uyar
- Faculty of Medicine, Department of Anaesthesiology and Reanimation, Ege University, Bornova, Izmir, Turkey
| | - Feriha Çilli
- Faculty of Medicine, Department of Medical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Bilgin Arda
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Tansu Yamazhan
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Hüsnü Pullukçu
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Meltem Işıkgöz Taşbakan
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Hilal Sipahi
- Department of Medical Microbiology, Bornova Directorate of Health, Bornova, Izmir, Turkey
| | - Sercan Ulusoy
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| | - Oguz Resat Sipahi
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
45
|
Atomic-Resolution Structures and Mode of Action of Clinically Relevant Antimicrobial Peptides. Int J Mol Sci 2022; 23:ijms23094558. [PMID: 35562950 PMCID: PMC9100274 DOI: 10.3390/ijms23094558] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Global rise of infections and deaths caused by drug-resistant bacterial pathogens are among the unmet medical needs. In an age of drying pipeline of novel antibiotics to treat bacterial infections, antimicrobial peptides (AMPs) are proven to be valid therapeutics modalities. Direct in vivo applications of many AMPs could be challenging; however, works are demonstrating encouraging results for some of them. In this review article, we discussed 3-D structures of potent AMPs e.g., polymyxin, thanatin, MSI, protegrin, OMPTA in complex with bacterial targets and their mode of actions. Studies on human peptide LL37 and de novo-designed peptides are also discussed. We have focused on AMPs which are effective against drug-resistant Gram-negative bacteria. Since treatment options for the infections caused by super bugs of Gram-negative bacteria are now extremely limited. We also summarize some of the pertinent challenges in the field of clinical trials of AMPs.
Collapse
|
46
|
In vitro Activity of Imipenem-Relebactam Alone and in Combination with Fosfomycin against Carbapenem-Resistant Gram-negative Pathogens. Diagn Microbiol Infect Dis 2022; 103:115712. [DOI: 10.1016/j.diagmicrobio.2022.115712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/13/2021] [Accepted: 04/17/2022] [Indexed: 11/19/2022]
|
47
|
Fernández Álvaro E, Voong Vinh P, de Cozar C, Willé DR, Urones B, Cortés A, Price A, Tran Do Hoang N, Ha Thanh T, McCloskey M, Shaheen S, Dayao D, Martinot A, de Mercado J, Castañeda P, García-Perez A, Singa B, Pavlinac P, Walson J, Martínez-Martínez MS, Arnold SLM, Tzipori S, Ballell Pages L, Baker S. The repurposing of Tebipenem pivoxil as alternative therapy for severe gastrointestinal infections caused by extensively drug-resistant Shigella spp. eLife 2022; 11:e69798. [PMID: 35289746 PMCID: PMC8959600 DOI: 10.7554/elife.69798] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
Background Diarrhoea remains one of the leading causes of childhood mortality globally. Recent epidemiological studies conducted in low-middle income countries (LMICs) identified Shigella spp. as the first and second most predominant agent of dysentery and moderate diarrhoea, respectively. Antimicrobial therapy is often necessary for Shigella infections; however, we are reaching a crisis point with efficacious antimicrobials. The rapid emergence of resistance against existing antimicrobials in Shigella spp. poses a serious global health problem. Methods Aiming to identify alternative antimicrobial chemicals with activity against antimicrobial resistant Shigella, we initiated a collaborative academia-industry drug discovery project, applying high-throughput phenotypic screening across broad chemical diversity and followed a lead compound through in vitro and in vivo characterisation. Results We identified several known antimicrobial compound classes with antibacterial activity against Shigella. These compounds included the oral carbapenem Tebipenem, which was found to be highly potent against broadly susceptible Shigella and contemporary MDR variants for which we perform detailed pre-clinical testing. Additional in vitro screening demonstrated that Tebipenem had activity against a wide range of other non-Shigella enteric bacteria. Cognisant of the risk for the development of resistance against monotherapy, we identified synergistic behaviour of two different drug combinations incorporating Tebipenem. We found the orally bioavailable prodrug (Tebipenem pivoxil) had ideal pharmacokinetic properties for treating enteric pathogens and was effective in clearing the gut of infecting organisms when administered to Shigella-infected mice and gnotobiotic piglets. Conclusions Our data highlight the emerging antimicrobial resistance crisis and shows that Tebipenem pivoxil (licenced for paediatric respiratory tract infections in Japan) should be accelerated into human trials and could be repurposed as an effective treatment for severe diarrhoea caused by MDR Shigella and other enteric pathogens in LMICs. Funding Tres Cantos Open Lab Foundation (projects TC239 and TC246), the Bill and Melinda Gates Foundation (grant OPP1172483) and Wellcome (215515/Z/19/Z).
Collapse
Affiliation(s)
| | - Phat Voong Vinh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | | | | | | | | | - Alan Price
- GSK Global Health, Tres CantosMadridSpain
| | - Nhu Tran Do Hoang
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | - Tuyen Ha Thanh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research UnitHo Chi Minh CityViet Nam
| | - Molly McCloskey
- Division of Allergy and Infectious Disease, Center for Emerging and Re-emerging Infectious Diseases University of Washington School of MedicineSeattleUnited States
| | - Shareef Shaheen
- Division of Allergy and Infectious Disease, Center for Emerging and Re-emerging Infectious Diseases University of Washington School of MedicineSeattleUnited States
| | - Denise Dayao
- Department of Infectious Disease and Global Health, Tufts University Cummings School of Veterinary MedicineNorth GraftonUnited States
| | - Amanda Martinot
- Department of Infectious Disease and Global Health, Tufts University Cummings School of Veterinary MedicineNorth GraftonUnited States
| | | | | | | | | | - Patricia Pavlinac
- Department of Global Health, University of WashingtonSeattleUnited States
| | - Judd Walson
- Division of Allergy and Infectious Disease, Center for Emerging and Re-emerging Infectious Diseases University of Washington School of MedicineSeattleUnited States
| | | | - Samuel LM Arnold
- Division of Allergy and Infectious Disease, Center for Emerging and Re-emerging Infectious Diseases University of Washington School of MedicineSeattleUnited States
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Tufts University Cummings School of Veterinary MedicineNorth GraftonUnited States
| | | | - Stephen Baker
- University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| |
Collapse
|
48
|
Sinha S, Dhanabal VB, Sperandeo P, Polissi A, Bhattacharjya S. Linking dual mode of action of host defense antimicrobial peptide thanatin: Structures, lipopolysaccharide and LptA m binding of designed analogs. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183839. [PMID: 34915021 DOI: 10.1016/j.bbamem.2021.183839] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
At present, antibiotics options to cure infections caused by drug resistant Gram-negative pathogens are highly inadequate. LPS outer membrane, proteins involved in LPS transport and biosynthesis pathways are vital targets. Thanatin, an insect derived 21-residue long antimicrobial peptide may be exploited for the development of effective antibiotics against Gram-negative bacteria. As a mode of bacterial cell killing, thanatin disrupts LPS outer membrane and inhibits LPS transport by binding to the periplasmic protein LptAm. Here, we report structure-activity correlation of thanatin and analogs for the purpose of rational design. These analogs of thanatin are investigated, by NMR, ITC and fluorescence, to correlate structure, antibacterial activity and binding with LPS and LptAm, a truncated monomeric variant. Our results demonstrate that an analog thanatin M21F exhibits superior antibacterial activity. In LPS interaction analyses, thanatin M21F demonstrate high affinity binding to outer membrane LPS. The atomic resolution structure of thanatin M21F in LPS micelle reveals four stranded β-sheet structure in a dimeric topology whereby the sidechain of aromatic residues Y10, F21 sustained mutual packing at the interface. Strikingly, LptAm binding affinity of thanatin M21F has been significantly increased with an estimated Kd ~ 0.73 nM vs 13 nM for thanatin. Further, atomic resolution structures and interactions of Ala based thanatin analogs define plausible correlations with antibacterial activity and LPS, LptAm interactions. Taken together, the current work provides a frame-work for the designing of thanatin based potent antimicrobial peptides for the treatment of drug resistance Gram-negative bacteria.
Collapse
Affiliation(s)
- Sheetal Sinha
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; Advanced Environmental Biotechnology Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, 1 Cleantech Loop, Singapore 637141, Singapore
| | - Vidhya Bharathi Dhanabal
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Paola Sperandeo
- Dept. of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Alessandra Polissi
- Dept. of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
49
|
Nguyen HT, Venter H, Woolford L, Young K, McCluskey A, Garg S, Page SW, Trott DJ, Ogunniyi AD. Impact of a Novel Anticoccidial Analogue on Systemic Staphylococcus aureus Infection in a Bioluminescent Mouse Model. Antibiotics (Basel) 2022; 11:antibiotics11010065. [PMID: 35052942 PMCID: PMC8773087 DOI: 10.3390/antibiotics11010065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 02/05/2023] Open
Abstract
In this study, we investigated the potential of an analogue of robenidine (NCL179) to expand its chemical diversity for the treatment of multidrug-resistant (MDR) bacterial infections. We show that NCL179 exhibits potent bactericidal activity, returning minimum inhibitory concentration/minimum bactericidal concentrations (MICs/MBCs) of 1–2 µg/mL against methicillin-resistant Staphylococcus aureus, MICs/MBCs of 1–2 µg/mL against methicillin-resistant S. pseudintermedius and MICs/MBCs of 2–4 µg/mL against vancomycin-resistant enterococci. NCL179 showed synergistic activity against clinical isolates and reference strains of Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa in the presence of sub-inhibitory concentrations of colistin, whereas NCL179 alone had no activity. Mice given oral NCL179 at 10 mg/kg and 50 mg/kg (4 × doses, 4 h apart) showed no adverse clinical effects and no observable histological effects in any of the organs examined. In a bioluminescent S. aureus sepsis challenge model, mice that received four oral doses of NCL179 at 50 mg/kg at 4 h intervals exhibited significantly reduced bacterial loads, longer survival times and higher overall survival rates than the vehicle-only treated mice. These results support NCL179 as a valid candidate for further development to treat MDR bacterial infections as a stand-alone antibiotic or in combination with existing antibiotic classes.
Collapse
Affiliation(s)
- Hang Thi Nguyen
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia;
- Department of Pharmacology, Toxicology, Internal Medicine and Diagnostics, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi 100000, Vietnam
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Lucy Woolford
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus, Roseworthy, SA 5371, Australia;
| | - Kelly Young
- Chemistry, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia; (K.Y.); (A.M.)
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia; (K.Y.); (A.M.)
| | - Sanjay Garg
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | | | - Darren J. Trott
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia;
- Correspondence: (D.J.T.); (A.D.O.); Tel.: +61-8-8313-7989 (D.J.T.); +61-432331914 (A.D.O.); Fax: +61-8-8313-7956 (D.J.T.)
| | - Abiodun David Ogunniyi
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia;
- Correspondence: (D.J.T.); (A.D.O.); Tel.: +61-8-8313-7989 (D.J.T.); +61-432331914 (A.D.O.); Fax: +61-8-8313-7956 (D.J.T.)
| |
Collapse
|
50
|
Rodrigues YC, Lobato ARF, Quaresma AJPG, Guerra LMGD, Brasiliense DM. The Spread of NDM-1 and NDM-7-Producing Klebsiella pneumoniae Is Driven by Multiclonal Expansion of High-Risk Clones in Healthcare Institutions in the State of Pará, Brazilian Amazon Region. Antibiotics (Basel) 2021; 10:1527. [PMID: 34943739 PMCID: PMC8698286 DOI: 10.3390/antibiotics10121527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Carbapenem resistance among Klebsiella pneumoniae isolates is often related to carbapenemase genes, located in genetic transmissible elements, particularly the blaKPC gene, which variants are spread in several countries. Recently, reports of K. pneumoniae isolates harboring the blaNDM gene have increased dramatically along with the dissemination of epidemic high-risk clones (HRCs). In the present study, we report the multiclonal spread of New Delhi metallo-beta-lactamase (NDM)-producing K. pneumoniae in different healthcare institutions in the state of Pará, Northern Brazil. A total of 23 NDM-producing isolates were tested regarding antimicrobial susceptibility testing features, screening of carbapenemase genes, and genotyping by multilocus sequencing typing (MLST). All K. pneumoniae isolates were determined as multidrug-resistant (MDR), being mainly resistant to carbapenems, cephalosporins, and fluoroquinolones. The blaNDM-7 (60.9%-14/23) and blaNDM-1 (34.8%-8/23) variants were detected. MLST genotyping revealed the predomination of HRCs, including ST11/CC258, ST340/CC258, ST15/CC15, ST392/CC147, among others. To conclude, the present study reveals the contribution of HRCs and non-HRCs in the spread of NDM-1 and NDM-7-producing K. pneumoniae isolates in Northern (Amazon region) Brazil, along with the first detection of NDM-7 variant in Latin America and Brazil, highlighting the need for surveillance and control of strains that may negatively impact healthcare and antimicrobial resistance.
Collapse
Affiliation(s)
| | | | | | | | - Danielle Murici Brasiliense
- Bacteriology and Mycology Section, Evandro Chagas Institute (SABMI/IEC), Ananindeua 67030-000, PA, Brazil; (Y.C.R.); (A.R.F.L.); (A.J.P.G.Q.); (L.M.G.D.G.)
| |
Collapse
|