1
|
Butler DA, Patel N, O'Donnell JN, Lodise TP. Combination therapy with IV fosfomycin for adult patients with serious Gram-negative infections: a review of the literature. J Antimicrob Chemother 2024; 79:2421-2459. [PMID: 39215642 DOI: 10.1093/jac/dkae253] [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] [Indexed: 09/04/2024] Open
Abstract
Treatment of patients with serious infections due to resistant Gram-negative bacteria remains highly problematic and has prompted clinicians to use existing antimicrobial agents in innovative ways. One approach gaining increased therapeutic use is combination therapy with IV fosfomycin. This article reviews the preclinical pharmacokinetic/pharmacodynamic (PK/PD) infection model and clinical data surrounding the use of combination therapy with IV fosfomycin for the treatment of serious infections caused by resistant Gram-negative bacteria. Data from dynamic in vitro and animal infection model studies of highly resistant Enterobacterales and non-lactose fermenters are positive and suggest IV fosfomycin in combination with a β-lactam, polymyxin or aminoglycoside produces a synergistic effect that rivals or surpasses that of other aminoglycoside- or polymyxin-containing regimens. Clinical studies performed to date primarily have involved patients with pneumonia and/or bacteraemia due to Klebsiella pneumoniae, Pseudomonas aeruginosa or Acinetobacter baumannii. Overall, the observed success rates with fosfomycin combination regimens were consistent with those reported for other combination regimens commonly used to treat these patients. In studies in which direct treatment comparisons can be derived, the results suggest that patients who received fosfomycin combination therapy had similar or improved outcomes compared with other therapies and combinations, especially when it was used in combination with a β-lactam that (1) targets PBP-3 and (2) has exceptional stability in the presence of β-lactamases. Collectively, the data indicate that combination therapy with IV fosfomycin should be considered as a potential alternative to aminoglycoside or polymyxin combinations for patients with antibiotic-resistant Gram-negative infections when benefits outweigh risks.
Collapse
Affiliation(s)
- David A Butler
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, 106 New Scotland Avenue, Albany, NY 12208, USA
| | - Nimish Patel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9255 Pharmacy Lane, La Jolla, CA, USA
| | - J Nicholas O'Donnell
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, 106 New Scotland Avenue, Albany, NY 12208, USA
| | - Thomas P Lodise
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, 106 New Scotland Avenue, Albany, NY 12208, USA
| |
Collapse
|
2
|
Shein AMS, Hongsing P, Smith OK, Phattharapornjaroen P, Miyanaga K, Cui L, Ishikawa H, Amarasiri M, Monk PN, Kicic A, Chatsuwan T, Pletzer D, Higgins PG, Abe S, Wannigama DL. Current and novel therapies for management of Acinetobacter baumannii-associated pneumonia. Crit Rev Microbiol 2024:1-22. [PMID: 38949254 DOI: 10.1080/1040841x.2024.2369948] [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: 09/25/2023] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Acinetobacter baumannii is a common pathogen associated with hospital-acquired pneumonia showing increased resistance to carbapenem and colistin antibiotics nowadays. Infections with A. baumannii cause high patient fatalities due to their capability to evade current antimicrobial therapies, emphasizing the urgency of developing viable therapeutics to treat A. baumannii-associated pneumonia. In this review, we explore current and novel therapeutic options for overcoming therapeutic failure when dealing with A. baumannii-associated pneumonia. Among them, antibiotic combination therapy administering several drugs simultaneously or alternately, is one promising approach for optimizing therapeutic success. However, it has been associated with inconsistent and inconclusive therapeutic outcomes across different studies. Therefore, it is critical to undertake additional clinical trials to ascertain the clinical effectiveness of different antibiotic combinations. We also discuss the prospective roles of novel antimicrobial therapies including antimicrobial peptides, bacteriophage-based therapy, repurposed drugs, naturally-occurring compounds, nanoparticle-based therapy, anti-virulence strategies, immunotherapy, photodynamic and sonodynamic therapy, for utilizing them as additional alternative therapy while tackling A. baumannii-associated pneumonia. Importantly, these innovative therapies further require pharmacokinetic and pharmacodynamic evaluation for safety, stability, immunogenicity, toxicity, and tolerability before they can be clinically approved as an alternative rescue therapy for A. baumannii-associated pulmonary infections.
Collapse
Affiliation(s)
- Aye Mya Sithu Shein
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - O'Rorke Kevin Smith
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Phatthranit Phattharapornjaroen
- Department of Emergency Medicine, Center of Excellence, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Surgery, Sahlgrenska Academy, Institute of Clinical Sciences, Gothenburg University, Gothenburg, Sweden
| | - Kazuhiko Miyanaga
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Longzhu Cui
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hitoshi Ishikawa
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Japan
| | - Mohan Amarasiri
- Laboratory of Environmental Hygiene, Department of Health Science, School of Allied Health Sciences, Kitasato University, Kitasato, Sagamihara-Minami, Japan
| | - Peter N Monk
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, UK
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia
- School of Population Health, Curtin University, Bentley, Western Australia, Australia
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Paul G Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA receiving countries, The University of Sheffield, Sheffield, UK
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| |
Collapse
|
3
|
Russo A, Gullì SP, D'Avino A, Borrazzo C, Carannante N, Dezza FC, Covino S, Polistina G, Fiorentino G, Trecarichi EM, Mastroianni CM, Torti C, Oliva A. Intravenous fosfomycin for treatment of severe infections caused by carbapenem-resistant Acinetobacter baumannii: A multi-centre clinical experience. Int J Antimicrob Agents 2024; 64:107190. [PMID: 38697579 DOI: 10.1016/j.ijantimicag.2024.107190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Severe infections caused by carbapenem-resistant Acinetobacter baumannii (CRAB) have been reported increasingly over the past few years. Many in-vivo and in-vitro studies have suggested a possible role of intravenous fosfomycin for the treatment of CRAB infections. METHODS This multi-centre, retrospective study included patients treated with intravenous fosfomycin for severe infections caused by CRAB admitted consecutively to four hospitals in Italy from December 2017 to December 2022. The primary goal of the study was to evaluate the risk factors associated with 30-day mortality in the study population. A propensity score matched analysis was added to the model. RESULTS One hundred and two patients with severe infections caused by CRAB treated with an intravenous fosfomycin-containing regimen were enrolled in this study. Ventilator-associated pneumonia (VAP) was diagnosed in 59% of patients, primary bacteraemia in 22% of patients, and central-venous-catheter-related infection in 16% of patients. All patients were treated with a regimen containing intravenous fosfomycin, mainly in combination with cefiderocol (n=54), colistin (n=48) or ampicillin/sulbactam (n=18). Forty-eight (47%) patients died within 30 days. Fifty-eight (57%) patients experienced clinical therapeutic failure. Cox regression analysis showed that diabetes, primary bacteraemia and a colistin-containing regimen were independently associated with 30-day mortality, whereas adequate source control of infection, early 24-h active in-vitro therapy, and a cefiderocol-containing regimen were associated with survival. A colistin-based regimen, A. baumannii colonization and primary bacteraemia were independently associated with clinical failure. Conversely, adequate source control of infection, a cefiderocol-containing regimen, and early 24-h active in-vitro therapy were associated with clinical success. CONCLUSIONS Different antibiotic regimens containing fosfomycin in combination can be used for treatment of severe infections caused by CRAB.
Collapse
Affiliation(s)
- Alessandro Russo
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy.
| | - Sara Palma Gullì
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - Alessandro D'Avino
- Department of Internal Medicine and Risk Management, Cristo Re Hospital, Rome, Italy
| | - Cristian Borrazzo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Novella Carannante
- Emergency Room, Cotugno-Monaldi Hospital, AORN Ospedali dei Colli, Naples, Italy
| | | | - Sara Covino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giorgio Polistina
- Sub-Intensive Care Unit and Respiratory Physiopathology Department, Cotugno-Monaldi Hospital, AORN Ospedali dei Colli, Naples, Italy
| | - Giuseppe Fiorentino
- Sub-Intensive Care Unit and Respiratory Physiopathology Department, Cotugno-Monaldi Hospital, AORN Ospedali dei Colli, Naples, Italy
| | - Enrico Maria Trecarichi
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | | | - Carlo Torti
- UOC Malattie Infettive - Dipartimento Scienze Mediche e Chirurgiche - Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma - Sezione Malattie Infettive - Dipartimento di Sicurezza e Bioetica - Università Cattolica S. Cuore, Roma
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Müderris T, Dursun Manyaslı G, Sezak N, Kaya S, Demirdal T, Gül Yurtsever S. In-vitro evaluation of different antimicrobial combinations with and without colistin against carbapenem-resistant Acinetobacter baumannii clinical isolates. Eur J Med Res 2024; 29:331. [PMID: 38880888 PMCID: PMC11180387 DOI: 10.1186/s40001-024-01885-6] [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: 02/27/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Carbapenem-resistant Acinetobacter baumannii (CRAB) infections are one of the most common causes of nosocomial infections and have high mortality rates due to difficulties in treatment. In this study, the in vitro synergistic interactions of the colistin (CT)-meropenem (MEM) combination and patient clinical outcomes were compared in CRAB-infected patients that receive CT-MEM antimicrobial combination therapy. In addition, in vitro synergistic interactions of MEM-ertapenem (ETP), MEM-fosfomycin (FF) and CT-FF antimicrobial combinations were investigated. Finally, the epsilometer (E) test and checkerboard test results were compared and the compatibility of these two tests was evaluated. METHODS Twenty-one patients were included in the study. Bacterial identification was performed with MALDI-TOF, and antimicrobial susceptibility was assessed with an automated system. Synergy studies were performed using the E test and checkerboard method. RESULTS For the checkerboard method, the synergy rates for CT-MEM, MEM-FF, MEM-ETP and CT-FF were 100%, 52.3%, 23.8% and 28.5%, respectively. In the E test synergy tests, synergistic effects were detected for two isolates each in the CT-MEM and CT-FF combinations. Microbial eradication was achieved in nine (52.9%) of the 17 patients that received CT-MEM combination therapy. The agreement between the E test and the checkerboard test was 6.5%. CONCLUSIONS A synergistic effect was found with the checkerboard method for the CT-MEM combination in all isolates in our study, and approximately 70% of the patients benefited from treatment with this combination. In addition, more than half of the isolates showed a synergistic effect for the MEM-FF combination. Combinations of CT-MEM and MEM-FF may be options for the treatment of CRAB infections. However, a comprehensive understanding of the potential of the microorganism to develop resistant mutants under applied exposures, as well as factors that directly affect antimicrobial activity, such as pharmacokinetics/pharmacodynamics, is essential for providing treatment advice. We found a low rate of agreement between the E test method and the checkerboard test method in our study, in contrast to the literature. Comprehensive studies that compare clinical results with methods are needed to determine the ideal synergy test and interpretation method.
Collapse
Affiliation(s)
- Tuba Müderris
- Faculty of Medicine, Department of Medical Microbiology, İzmir Katip Çelebi University, İzmir, Türkiye.
| | - Gülden Dursun Manyaslı
- Cizre Dr. Selahattin Cizrelioğlu Public Hospital, Department of Medical Microbiology, Şırnak, Türkiye
| | - Nurbanu Sezak
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, İzmir Demokrasi University, İzmir, Türkiye
| | - Selçuk Kaya
- Faculty of Medicine, Department of Medical Microbiology, İzmir Katip Çelebi University, İzmir, Türkiye
| | - Tuna Demirdal
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, İzmir Katip Çelebi University, İzmir, Türkiye
| | - Süreyya Gül Yurtsever
- Faculty of Medicine, Department of Medical Microbiology, İzmir Katip Çelebi University, İzmir, Türkiye
| |
Collapse
|
5
|
Bisaro F, Jackson-Litteken CD, McGuffey JC, Hooppaw AJ, Bodrog S, Jebeli L, Ortiz-Marquez JC, van Opijnen T, Scott NE, Di Venanzio G, Feldman MF. Diclofenac sensitizes multi-drug resistant Acinetobacter baumannii to colistin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594771. [PMID: 38798593 PMCID: PMC11118529 DOI: 10.1101/2024.05.17.594771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Acinetobacter baumannii causes life-threatening infections that are becoming difficult to treat due to increasing rates of multi-drug resistance (MDR) among clinical isolates. This has led the World Health Organization and the CDC to categorize MDR A. baumannii as a top priority for the research and development of new antibiotics. Colistin is the last-resort antibiotic to treat carbapenem-resistant A. baumannii . Not surprisingly, reintroduction of colistin has resulted in the emergence of colistin-resistant strains. Diclofenac is a nonsteroidal anti-inflammatory drug used to treat pain and inflammation associated with arthritis. In this work, we show that diclofenac sensitizes colistin-resistant A. baumannii clinical strains to colistin, in vitro and in a murine model of pneumonia. Diclofenac also reduced the colistin MIC of Klebsiella pneumoniae and Pseudomonas aeruginosa isolates. Transcriptomic and proteomic analyses revealed an upregulation of oxidative stress-related genes and downregulation of type IV pili induced by the combination treatment. Notably, the concentrations of colistin and diclofenac effective in the murine model were substantially lower than those determined in vitro , implying a stronger synergistic effect in vivo compared to in vitro . A pilA mutant strain, lacking the primary component of the type IV pili, became sensitive to colistin in the absence of diclofenac. This suggest that the downregulation of type IV pili is key for the synergistic activity of these drugs in vivo and indicates that colistin and diclofenac exert an anti-virulence effect. Together, these results suggest that the diclofenac can be repurposed with colistin to treat MDR A. baumannii .
Collapse
|
6
|
Basardeh E, Piri-Gavgani S, Moradi HR, Azizi M, Mirzabeigi P, Nazari F, Ghanei M, Mahboudi F, Rahimi-Jamnani F. Anti-Acinetobacter Baumannii single-chain variable fragments provide therapeutic efficacy in an immunocompromised mouse pneumonia model. BMC Microbiol 2024; 24:55. [PMID: 38341536 PMCID: PMC10858608 DOI: 10.1186/s12866-023-03080-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/22/2023] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The emergence of carbapenem-resistant and extensively drug-resistant (XDR) Acinetobacter baumannii as well as inadequate effective antibiotics calls for an urgent effort to find new antibacterial agents. The therapeutic efficacy of two human scFvs, EB211 and EB279, showing growth inhibitory activity against A. baumannii in vitro, was investigated in immunocompromised mice with A. baumannii pneumonia. RESULTS The data revealed that infected mice treated with EB211, EB279, and a combination of the two scFvs showed better survival, reduced bacterial load in the lungs, and no marked pathological abnormalities in the kidneys, liver, and lungs when compared to the control groups receiving normal saline or an irrelevant scFv. CONCLUSIONS The results from this study suggest that the scFvs with direct growth inhibitory activity could offer promising results in the treatment of pneumonia caused by XDR A. baumannii.
Collapse
Affiliation(s)
- Eilnaz Basardeh
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Piri-Gavgani
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Hamid Reza Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Parastoo Mirzabeigi
- Department of Clinical Pharmacy and Pharmacoeconomics, Faculty of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Nazari
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Rahimi-Jamnani
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
İzci F, Ture Z, Dinc G, Yay AH, Eren EE, Bolat D, Gönen ZB, Ünüvar GK, Yıldız O, Aygen B. The efficacy of mesenchymal stem cell treatment and colistin-fosfomycin combination on colistin-resistant Acinetobacter baumannii sepsis model. Eur J Clin Microbiol Infect Dis 2023; 42:1365-1372. [PMID: 37814067 DOI: 10.1007/s10096-023-04674-x] [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: 12/25/2022] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION This study examines the role of mesenchymal stem cells (MSCs) in an experimental sepsis model developed with colistin-resistant Acinetobacter baumannii (CRAB). MATERIALS AND METHODS BALB-c mice were divided into treatment groups (MSC, MSC + colistin (C)-fosfomycin (F), and C-F and control groups (positive and negative)). CRAB was administered to mice through intraperitoneal injection. Three hours later, C, F, and MSC were given intraperitoneally to the treatment groups. Colistin administration was repeated every 12 h, F administration was done every 4 h, and the second dose of MSC was administered after 48 h. Mice were sacrificed at 24 and 72 h. The bacterial load was determined as colony-forming units per gram (cfu/g). Histopathological examination was conducted on the left lung, liver, and both kidneys. IL-6 and C-reactive protein (CRP) levels in mouse sera were determined by enzyme-linked immunosorbent assay. RESULTS Among the treatment groups, the C-F group had the lowest colony count in the lung (1.24 ± 1.66 cfu/g) and liver (1.03 ± 1.08 cfu/g). The highest bacterial clearance was observed at 72 h compared to 24 h in the MSC-treated groups (p = 0.008). The MSC + C-F group showed the lowest histopathological score in the liver and kidney (p = 0.009). In the negative control group, the IL-6 level at the 24th hour was the lowest (p < 0.001). Among the treatment groups, the CRP level was the lowest in the MSC + C-F group at 24 and 72 h. CONCLUSION In a CRAB sepsis model, adding MSCs to a colistin-fosfomycin treatment may be beneficial in terms of reducing bacterial loads and preventing histopathological damage.
Collapse
Affiliation(s)
- Feyza İzci
- Department of Infectious Diseases, Çankırı State Hospital, Çankırı, Turkey
| | - Zeynep Ture
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| | - Gokcen Dinc
- Department of Medical Microbiology, Faculty of Medicine, and Department of Stem Cell GMP Unit of Genome and Stem Cell Centre, Erciyes University, Kayseri, Turkey
| | - Arzu Hanım Yay
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Esma Eryılmaz Eren
- Department of Infectious Diseases and Clinical Microbiology, Kayseri City Education and Research Hospital, Kayseri, Turkey
| | - Demet Bolat
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Zeynep Burcin Gönen
- Oral and Maxillofacial Surgery, Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Gamze Kalın Ünüvar
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Orhan Yıldız
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Bilgehan Aygen
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
8
|
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
|
9
|
Young M, Chojnacki M, Blanchard C, Cao X, Johnson WL, Flaherty D, Dunman PM. Genetic Determinants of Acinetobacter baumannii Serum-Associated Adaptive Efflux-Mediated Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1173. [PMID: 37508269 PMCID: PMC10376123 DOI: 10.3390/antibiotics12071173] [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: 06/09/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen of serious healthcare concern that is becoming increasingly difficult to treat due to antibiotic treatment failure. Recent studies have revealed that clinically defined antibiotic-susceptible strains upregulate the expression of a repertoire of putative drug efflux pumps during their growth under biologically relevant conditions, e.g., in human serum, resulting in efflux-associated resistance to physiologically achievable antibiotic levels within a patient. This phenomenon, termed Adaptive Efflux Mediated Resistance (AEMR), has been hypothesized to account for one mechanism by which antibiotic-susceptible A. baumannii fails to respond to antibiotic treatment. In the current study, we sought to identify genetic determinants that contribute to A. baumannii serum-associated AEMR by screening a transposon mutant library for members that display a loss of the AEMR phenotype. Results revealed that mutation of a putative pirin-like protein, YhaK, results in a loss of AEMR, a phenotype that could be complemented by a wild-type copy of the yhaK gene and was verified in a second strain background. Ethidium bromide efflux assays confirmed that the loss of AEMR phenotype due to pirin-like protein mutation correlated with reduced overarching efflux capacity. Further, flow cytometry and confocal microscopy measures of a fluorophore 7-(dimethylamino)-coumarin-4-acetic acid (DMACA)-tagged levofloxacin isomer, ofloxacin, further verified that YhaK mutation reduces AEMR-mediated antibiotic efflux. RNA-sequencing studies revealed that YhaK may be required for the expression of multiple efflux-associated systems, including MATE and ABC families of efflux pumps. Collectively, the data indicate that the A. baumannii YhaK pirin-like protein plays a role in modulating the organism's adaptive efflux-mediated resistance phenotype.
Collapse
Affiliation(s)
- Mikaeel Young
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (M.Y.); (M.C.); (W.L.J.)
| | - Michaelle Chojnacki
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (M.Y.); (M.C.); (W.L.J.)
| | - Catlyn Blanchard
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (M.Y.); (M.C.); (W.L.J.)
| | - Xufeng Cao
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, Lafayette, IN 47907, USA
| | - William L. Johnson
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (M.Y.); (M.C.); (W.L.J.)
| | - Daniel Flaherty
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, Lafayette, IN 47907, USA
- Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907, USA
| | - Paul M. Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; (M.Y.); (M.C.); (W.L.J.)
| |
Collapse
|
10
|
Russo A, Bruni A, Gullì S, Borrazzo C, Quirino A, Lionello R, Serapide F, Garofalo E, Serraino R, Romeo F, Marascio N, Matera G, Longhini F, Trecarichi EM, Torti C. Efficacy of cefiderocol- vs colistin-containing regimen for treatment of bacteraemic ventilator-associated pneumonia caused by carbapenem-resistant Acinetobacter baumannii in patients with COVID-19. Int J Antimicrob Agents 2023; 62:106825. [PMID: 37088438 PMCID: PMC10121149 DOI: 10.1016/j.ijantimicag.2023.106825] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/25/2023]
Abstract
INTRODUCTION Ventilator-associated pneumonia (VAP) caused by carbapenem-resistant Acinetobacter baumannii (CRAB) in patients hospitalized in intensive care units (ICUs) is an important and challenging complication, including in patients with coronavirus disease 2019 (COVID-19). Considering the poor lung penetration of most antibiotics, including intravenous colistin due to the poor pharmacokinetics/pharmacodynamics at the infection site, the choice of the best antibiotic regimen is still being debated. METHODS This single-centre, observational study was conducted from March 2020 to August 2022, and included all patients hospitalized consecutively with VAP and concomitant bloodstream infection due to CRAB in the COVID-ICU. The main goal of the study was to evaluate risk factors associated with survival or death at 30 days from VAP onset. A propensity score for receiving therapy was added to the model. RESULTS During the study period, 73 patients who developed VAP and concomitant positive blood cultures caused by CRAB were enrolled in the COVID-ICU. Of these patients, 67 (91.7%) developed septic shock, 42 (57.5%) had died at 14 days and 59 (80.8%) had died at 30 days. Overall, 54 (74%) patients were treated with a colistin-containing regimen and 19 (26%) were treated with a cefiderocol-containing regimen. Cox regression analysis showed that chronic obstructive pulmonary disease and age were independently associated with 30-day mortality. Conversely, cefiderocol-containing regimens and cefiderocol + fosfomycin in combination were independently associated with 30-day survival, as confirmed by propensity score analysis. CONCLUSIONS This real-life study in patients with bacteraemic VAP caused by CRAB provides useful suggestions for clinicians, showing a possible benefit of cefiderocol and its association with fosfomycin.
Collapse
Affiliation(s)
- A Russo
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy.
| | - A Bruni
- Anaesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - S Gullì
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - C Borrazzo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - A Quirino
- Clinical Microbiology Unit, Department of Health Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - R Lionello
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - F Serapide
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - E Garofalo
- Anaesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - R Serraino
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - F Romeo
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - N Marascio
- Clinical Microbiology Unit, Department of Health Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - G Matera
- Clinical Microbiology Unit, Department of Health Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - F Longhini
- Anaesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - E M Trecarichi
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| | - C Torti
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, 'Magna Graecia' University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
11
|
Lysitsas M, Chatzipanagiotidou I, Billinis C, Valiakos G. Fosfomycin Resistance in Bacteria Isolated from Companion Animals (Dogs and Cats). Vet Sci 2023; 10:vetsci10050337. [PMID: 37235420 DOI: 10.3390/vetsci10050337] [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: 03/31/2023] [Revised: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Fosfomycin is an old antibacterial agent, which is currently used mainly in human medicine, in uncomplicated Urinary Tract Infections (UTIs). The purpose of this review is to investigate the presence and the characteristics of Fosfomycin resistance in bacteria isolated from canine or feline samples, estimate the possible causes of the dissemination of associated strains in pets, and underline the requirements of prospective relevant studies. Preferred Reporting Items for Systematic Reviews (PRISMA) guidelines were used for the search of current literature in two databases. A total of 33 articles were finally included in the review. Relevant data were tracked down, assembled, and compared. Referring to the geographical distribution, Northeast Asia was the main area of origin of the studies. E. coli was the predominant species detected, followed by other Enterobacteriaceae, Staphylococci, and Pseudomonas spp. FosA and fosA3 were the more frequently encountered Antimicrobial Resistance Genes (ARGs) in the related Gram-negative isolates, while fosB was regularly encountered in Gram-positive ones. The majority of the strains were multidrug-resistant (MDR) and co-carried resistance genes against several classes of antibiotics and especially β-Lactams, such as blaCTX-M and mecA. These results demonstrate the fact that the cause of the spreading of Fosfomycin-resistant bacteria among pets could be the extended use of other antibacterial agents, that promote the prevalence of MDR, epidemic strains among an animal population. Through the circulation of these strains into a community, a public health issue could arise. Further research is essential though, for the comprehensive consideration of the issue, as the current data are limited.
Collapse
Affiliation(s)
- Marios Lysitsas
- Faculty of Veterinary Science, University of Thessaly, 43100 Karditsa, Greece
| | | | | | - George Valiakos
- Faculty of Veterinary Science, University of Thessaly, 43100 Karditsa, Greece
| |
Collapse
|
12
|
Marino A, Stracquadanio S, Campanella E, Munafò A, Gussio M, Ceccarelli M, Bernardini R, Nunnari G, Cacopardo B. Intravenous Fosfomycin: A Potential Good Partner for Cefiderocol. Clinical Experience and Considerations. Antibiotics (Basel) 2022; 12:antibiotics12010049. [PMID: 36671250 PMCID: PMC9854867 DOI: 10.3390/antibiotics12010049] [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: 12/07/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Multidrug resistant Gram-negative bacteremia represents a therapeutic challenge clinicians have to deal with. This concern becomes more difficult when causing germs are represented by carbapenem resistant Acinetobacter baumannii or difficult-to-treat Pseudomonas aeruginosa. Few antibiotics are available against these cumbersome bacteria, although literature data are not conclusive, especially for Acinetobacter. Cefiderocol could represent a valid antibiotic choice, being a molecule with an innovative mechanism of action capable of overcoming common resistance pathways, whereas intravenous fosfomycin may be an appropriate partner either enhancing cefiderocol activity or avoiding resistance development. Here we report two patients with MDR Gram negative bacteremia who were successfully treated with a cefiderocol/fosfomycin combination.
Collapse
Affiliation(s)
- Andrea Marino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS, Garibaldi Hospital, University of Catania, 95123 Catania, Italy
| | - Stefano Stracquadanio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
- Correspondence:
| | - Edoardo Campanella
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS, Garibaldi Hospital, University of Catania, 95123 Catania, Italy
| | - Antonio Munafò
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Maria Gussio
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS, Garibaldi Hospital, University of Catania, 95123 Catania, Italy
| | - Manuela Ceccarelli
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS, Garibaldi Hospital, University of Catania, 95123 Catania, Italy
| | - Renato Bernardini
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuseppe Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98124 Messina, Italy
| | - Bruno Cacopardo
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, ARNAS, Garibaldi Hospital, University of Catania, 95123 Catania, Italy
| |
Collapse
|
13
|
Gao L, Ma X. Transcriptome Analysis of Acinetobacter baumannii in Rapid Response to Subinhibitory Concentration of Minocycline. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16095. [PMID: 36498165 PMCID: PMC9741440 DOI: 10.3390/ijerph192316095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
The increasing emergence of multidrug-resistant Acinetobacter baumannii brings great threats to public health. Minocycline is a kind of semisynthetic derivative of the antibacterial drug tetracycline and is often used to treat infections caused by multidrug-resistant A. baumannii with other antibiotics. However, minocycline-resistant A. baumannii appears constantly. To rapidly explore the response of A. baumannii to minocycline stress, RNA-seq was carried out to compare the difference in the transcriptome of A. baumannii ATCC19606 in the presence or absence of minocycline. The results showed that 25 genes were differentially expressed, including 10 downregulated genes and 15 upregulated genes, and 24 sRNA were upregulated and 24 were downregulated based on the filter criteria (Log2FC > 1 or <−1 and FDR < 0.05). RtcB family protein and ABC transporter ATP-binding protein were upregulated by 2.6- and 11.3-fold, and molecular chaperone GroES, chaperonin GroL, class C beta-lactamase ADC-158, amino acid ABC transporter permease, and APC family permease were downregulated by at least two-fold in the presence of half-MIC minocycline. The differentially expressed genes are mainly involved in the stress response, the GroES/GroEL chaperonin system, and transport metabolic pathways. sRNA 1248 was significantly upregulated, and sRNA 1767, 5182, and 6984 were downregulated in a rapid response to minocycline. These results provide insights into the adaptive mechanism of A. baumannii to minocycline.
Collapse
Affiliation(s)
- Lili Gao
- College of Grassland Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiaochun Ma
- Experimental Animal Center, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
14
|
Al-Madboly LA. A Novel Triple Combination To Combat Serious Infections with Carbapenem-Resistant Acinetobacter baumannii in a Mouse Pneumonia Model. Microbiol Spectr 2022; 10:e0271021. [PMID: 35975993 PMCID: PMC9603289 DOI: 10.1128/spectrum.02710-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/17/2022] [Indexed: 01/04/2023] Open
Abstract
The ongoing crisis of antimicrobial resistance demands novel combinations between antimicrobials and nonantimicrobials to manage infections caused by highly resistant pathogens. This study aimed to evaluate the effect of combining sodium ascorbate and/or apo-transferrin with imipenem, forming double and triple combinations, against 20 multiple-carbapenemase-producing Acinetobacter baumannii strains using the checkerboard test, time-kill assay, and disc diffusion test. The results of the checkerboard assay revealed that all double combinations showed indifference, while only triple combination recorded a synergistic effect (fractional inhibitory concentration index [FICI] < 0.8) in 95% the test isolates. Moreover, the MIC of imipenem (MICimp) was strongly reduced (up to 128-fold reduction) after treatment with the triple combination against highly resistant isolates and reached the susceptible range. The time-kill assay revealed that the triple combination led to a 4-log10 reduction in the CFU at 8 h compared with the initial bacterial count, and no viable count was recorded at 10 h. The mouse pneumonia model showed restoration of lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following administration of the triple rescue therapy. Additionally, normal lungs with normal patent alveoli were detected 72 h following treatment. Accordingly, sodium ascorbate and apo-transferrin are promising adjunct biological agents with the potential to restore the effectiveness of critically essential antibiotics like imipenem, commonly used for the treatment of A. baumannii infections. IMPORTANCE Combination therapy provides a perspective to threat multidrug-resistant (MDR) strains. The present study sheds light on a novel and effective triple combination against carbapenem-resistant A. baumannii. Our in vitro results showed that combining imipenem with apo-transferrin and sodium ascorbate yielded synergism in 95% of test isolates, and this was associated with a marked reduction in imipenem MIC, shifting it below the breakpoint. Furthermore, a bactericidal effect was recorded, with no viable count detected at 10 h. An in vivo murine model of pneumonia was induced to mimic human disease. The triple combination therapy restored lung function and structure, with mild to moderate residual inflammation and moderately congested vessels observed 8 h following the initiation of therapy. Therefore, our findings suggest novel insights about a promising new combination therapy against highly resistant carbapenemase-producing A. baumannii to restore the effectiveness of imipenem.
Collapse
Affiliation(s)
- Lamiaa A. Al-Madboly
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
15
|
Arrazuria R, Kerscher B, Huber KE, Hoover JL, Lundberg CV, Hansen JU, Sordello S, Renard S, Aranzana-Climent V, Hughes D, Gribbon P, Friberg LE, Bekeredjian-Ding I. Variability of murine bacterial pneumonia models used to evaluate antimicrobial agents. Front Microbiol 2022; 13:988728. [PMID: 36160241 PMCID: PMC9493352 DOI: 10.3389/fmicb.2022.988728] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial resistance has become one of the greatest threats to human health, and new antibacterial treatments are urgently needed. As a tool to develop novel therapies, animal models are essential to bridge the gap between preclinical and clinical research. However, despite common usage of in vivo models that mimic clinical infection, translational challenges remain high. Standardization of in vivo models is deemed necessary to improve the robustness and reproducibility of preclinical studies and thus translational research. The European Innovative Medicines Initiative (IMI)-funded “Collaboration for prevention and treatment of MDR bacterial infections” (COMBINE) consortium, aims to develop a standardized, quality-controlled murine pneumonia model for preclinical efficacy testing of novel anti-infective candidates and to improve tools for the translation of preclinical data to the clinic. In this review of murine pneumonia model data published in the last 10 years, we present our findings of considerable variability in the protocols employed for testing the efficacy of antimicrobial compounds using this in vivo model. Based on specific inclusion criteria, fifty-three studies focusing on antimicrobial assessment against Pseudomonas aeruginosa, Klebsiella pneumoniae and Acinetobacter baumannii were reviewed in detail. The data revealed marked differences in the experimental design of the murine pneumonia models employed in the literature. Notably, several differences were observed in variables that are expected to impact the obtained results, such as the immune status of the animals, the age, infection route and sample processing, highlighting the necessity of a standardized model.
Collapse
Affiliation(s)
- Rakel Arrazuria
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Karen E. Huber
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | - Jennifer L. Hoover
- Infectious Diseases Research Unit, GlaxoSmithKline Pharmaceuticals, Collegeville, PA, United States
| | | | - Jon Ulf Hansen
- Department of Bacteria, Parasites & Fungi, Statens Serum Institut, Copenhagen, Denmark
| | | | | | | | - Diarmaid Hughes
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Hamburg, Germany
| | | | - Isabelle Bekeredjian-Ding
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- *Correspondence: Isabelle Bekeredjian-Ding,
| |
Collapse
|
16
|
Unravelling the In Vitro and In Vivo Anti- Helicobacter pylori Effect of Delphinidin-3- O-Glucoside Rich Extract from Pomegranate Exocarp: Enhancing Autophagy and Downregulating TNF-α and COX2. Antioxidants (Basel) 2022; 11:antiox11091752. [PMID: 36139826 PMCID: PMC9495706 DOI: 10.3390/antiox11091752] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
Fruits containing antioxidants, e.g., anthocyanins, exhibit antimicrobial activities. The emergence of drug resistance represents a major challenge in eradicating H. pylori. The current study aims to explore the effect of pomegranate exocarp anthocyanin methanol extract (PEAME) against H. pylori isolates recovered from antral gastric biopsies. The UPLC-PDA-MS/MS and 1H NMR analyses indicated delphinidin-3-O-glucoside as the major anthocyanin in the extract. The PEAME showed activity against all tested resistant isolates in vitro recording minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values of 128 and 256 µg/mL, respectively. In vivo investigation included evaluation of the rat gastric mucosa for malondialdehyde (MDA), catalase activity, COX2, TNF-α, and key autophagy gene expression. The combination of pomegranate with metronidazole markedly reduced the viable count of H. pylori and the level of COX2, with alleviation of H. pylori-induced inflammation and oxidative stress (reduction of MDA, p-value < 0.001; and increase in catalase activity, p-value < 0.001). Autophagy gene expression was significantly upregulated upon treatment, whereas TNF-α was downregulated. In conclusion, we comprehensively assessed the effect of PEAME against H. pylori isolates, suggesting its potential in combination with metronidazole for eradication of this pathogen. The beneficial effect of PEAME may be attributed to its ability to enhance autophagy.
Collapse
|
17
|
Abbey T, Vialichka A, Jurkovic M, Biagi M, Wenzler E. Activity of Omadacycline Alone and in Combination against Carbapenem-Nonsusceptible Acinetobacter baumannii with Varying Minocycline Susceptibility. Microbiol Spectr 2022; 10:e0054222. [PMID: 35647655 PMCID: PMC9241703 DOI: 10.1128/spectrum.00542-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/24/2022] [Indexed: 12/02/2022] Open
Abstract
Tetracycline-based combinations are increasingly used for serious carbapenem-nonsusceptible Acinetobacter baumannii (CNSAb) infections given their potent in vitro activity, synergism with other agents, and acceptable toxicity profile. Omadacycline is a novel aminomethylcycline with activity against minocycline-resistant pathogens, once daily oral dosing, and favorable pharmacokinetic properties. Given these potential advantages, the in vitro potency and antibacterial activity of omadacycline were evaluated alone and in combination against CNSAb with varying minocycline susceptibility. Broth microdilution testing of 41 CNSAb revealed that omadacycline (MIC50/90: 4/8 mg/L) inhibited 68.3% (28/41) of isolates at ≤4 mg/L and its activity was unaffected by minocycline nonsusceptibility (MIC50/90: 4/8 mg/L; 74.2% [23/31] inhibited at ≤4 mg/L). Ten (5 minocycline susceptible and 5 nonsusceptible) of the 41 CNSAb isolates were then evaluated in time-kill analyses against omadacycline and comparator agents alone and in dual- and triple-drug combinations at the free maximum concentration of drug in serum (fCmax). Amikacin, meropenem, and polymyxin B alone were each bactericidal against 4 of 10 (40%) isolates while omadacycline and sulbactam were bactericidal against 0 (0%) and 1 (10%), respectively. In dual-drug combinations with omadacycline, synergy was observed against 80% of isolates with sulbactam followed by 30% with amikacin or polymyxin B and 0% with meropenem or rifampin. The triple-drug combination of omadacycline, sulbactam, and polymyxin B achieved synergy against just one additional strain over the omadacycline-sulbactam dual combination but significantly reduced the time to 99.9% kill by more than 6 h (4.6 ± 2.8 h vs. 11.3 ± 5.9 h, P < 0.01). These results support the continued investigation into tetracycline-based combinations against CNSAb, particularly those including sulbactam, and suggest that omadacycline may have in vitro advantages over existing tetracycline-derivatives. IMPORTANCE Treatment of infections due to Acinetobacter baumannii often involves the use of multiple antibiotics simultaneously as combination therapy, but it is unknown which antibiotics are best used together. Tetracycline agents such as minocycline and tigecycline maintain good activity against A. baumannii and are often used with one or more other agents to achieve better killing of the bacteria. Omadacycline is a new tetracycline that may have a role in the treatment of A. baumannii, but no data are available evaluating its interaction with other commonly used drugs such as polymyxin B and sulbactam. Therefore, the purpose of this study was to investigate the antibacterial activity of omadacycline when combined with one or more other agents against carbapenem-resistant strains of A. baumannii. These findings may then be used to design confirmatory studies that could help decide what drugs work best together and what combination of agents should be used for patients.
Collapse
Affiliation(s)
- Taylor Abbey
- College of Pharmacy, University of Illinois Chicago, Chicago, Illinois, USA
| | - Alesia Vialichka
- College of Pharmacy, University of Illinois Chicago, Chicago, Illinois, USA
| | - Michele Jurkovic
- College of Pharmacy, University of Illinois Chicago, Chicago, Illinois, USA
| | - Mark Biagi
- College of Pharmacy, University of Illinois Chicago, Rockford, Illinois, USA
| | - Eric Wenzler
- College of Pharmacy, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
18
|
Singkham-In U, Chatsuwan T. Synergism of imipenem with fosfomycin associated with the active cell wall recycling and heteroresistance in Acinetobacter calcoaceticus-baumannii complex. Sci Rep 2022; 12:230. [PMID: 34997148 PMCID: PMC8741973 DOI: 10.1038/s41598-021-04303-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022] Open
Abstract
The carbapenem-resistant Acinetobacter calcoaceticus-baumannii (ACB) complex has become an urgent threat worldwide. Here, we determined antibiotic combinations and the feasible synergistic mechanisms against three couples of ACB (A. baumannii (AB250 and A10), A. pittii (AP1 and AP23), and A. nosocomialis (AN4 and AN12)). Imipenem with fosfomycin, the most effective in the time-killing assay, exhibited synergism to all strains except AB250. MurA, a fosfomycin target encoding the first enzyme in the de novo cell wall synthesis, was observed with the wild-type form in all isolates. Fosfomycin did not upregulate murA, indicating the MurA-independent pathway (cell wall recycling) presenting in all strains. Fosfomycin more upregulated the recycling route in synergistic strain (A10) than non-synergistic strain (AB250). Imipenem in the combination dramatically downregulated the recycling route in A10 but not in AB250, demonstrating the additional effect of imipenem on the recycling route, possibly resulting in synergism by the agitation of cell wall metabolism. Moreover, heteroresistance to imipenem was observed in only AB250. Our results indicate that unexpected activity of imipenem on the active cell wall recycling concurrently with the presence of heteroresistance subpopulation to imipenem may lead to the synergism of imipenem and fosfomycin against the ACB isolates.
Collapse
Affiliation(s)
- Uthaibhorn Singkham-In
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Rama VI Road, Bangkok, 10330, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Rama VI Road, Bangkok, 10330, Thailand. .,Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Rama VI Road, Bangkok, 10330, Thailand.
| |
Collapse
|
19
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:2199-2208. [DOI: 10.1093/jac/dkac142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 11/14/2022] Open
|
20
|
He Q, Zhao L, Li G, Shen Y, Hu Y, Wang Y. The antimicrobial cyclic peptide B2 combats multidrug resistant Acinetobacter baumannii infection. NEW J CHEM 2022. [DOI: 10.1039/d1nj05353a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In silico methods were employed for the development of antimicrobial peptides against MDR A. baumannii by binding to BamA.
Collapse
Affiliation(s)
- Qingxiu He
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Linan Zhao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Guangping Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, China, 400054
- Chongqing Key Laboratory of Target Based Drug Screening and Activity Evaluation, Chongqing University of Technology, Chongqing, China, 400054
| | - Yong Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, China, 400054
- Chongqing Key Laboratory of Target Based Drug Screening and Activity Evaluation, Chongqing University of Technology, Chongqing, China, 400054
| | - Yuanqiang Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, China, 400054
- Chongqing Key Laboratory of Target Based Drug Screening and Activity Evaluation, Chongqing University of Technology, Chongqing, China, 400054
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China, 400716
| |
Collapse
|
21
|
Tamma PD, Aitken SL, Bonomo RA, Mathers AJ, van Duin D, Clancy CJ. Infectious Diseases Society of America Guidance on the Treatment of AmpC β-lactamase-Producing Enterobacterales, Carbapenem-Resistant Acinetobacter baumannii, and Stenotrophomonas maltophilia Infections. Clin Infect Dis 2021; 74:2089-2114. [PMID: 34864936 DOI: 10.1093/cid/ciab1013] [Citation(s) in RCA: 270] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The Infectious Diseases Society of America (IDSA) is committed to providing up-to-date guidance on the treatment of antimicrobial-resistant infections. A previous guidance document focused on infections caused by extended-spectrum β-lactamase-producing Enterobacterales (ESBL-E), carbapenem-resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with difficult-to-treat resistance (DTR-P. aeruginosa). Here, guidance is provided for treating AmpC β-lactamase-producing Enterobacterales (AmpC-E), carbapenem-resistant Acinetobacter baumannii (CRAB), and Stenotrophomonas maltophilia infections. METHODS A panel of six infectious diseases specialists with expertise in managing antimicrobial-resistant infections formulated questions about the treatment of AmpC-E, CRAB, and S. maltophilia infections. Answers are presented as suggestions and corresponding rationales. In contrast to guidance in the previous document, published data on optimal treatment of AmpC-E, CRAB, and S. maltophilia infections are limited. As such, guidance in this document is provided as "suggested approaches" based on clinical experience, expert opinion, and a review of the available literature. Because of differences in the epidemiology of resistance and availability of specific anti-infectives internationally, this document focuses on the treatment of infections in the United States. RESULTS Preferred and alternative treatment suggestions are provided, assuming the causative organism has been identified and antibiotic susceptibility results are known. Approaches to empiric treatment, duration of therapy, and other management considerations are also discussed briefly. Suggestions apply for both adult and pediatric populations. CONCLUSIONS The field of antimicrobial resistance is highly dynamic. Consultation with an infectious diseases specialist is recommended for the treatment of antimicrobial-resistant infections. This document is current as of September 17, 2021 and will be updated annually. The most current versions of IDSA documents, including dates of publication, are available at www.idsociety.org/practice-guideline/amr-guidance-2.0/.
Collapse
Affiliation(s)
- Pranita D Tamma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel L Aitken
- Department of Pharmacy, University of Michigan Health, Ann Arbor, Michigan, USA
| | - Robert A Bonomo
- Medical Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, University Hospitals Cleveland Medical Center and Departments of Medicine, Pharmacology, Molecular Biology, and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Amy J Mathers
- Departments of Medicine and Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - David van Duin
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Cornelius J Clancy
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Mohamed Abu El-Wafa W, Abouwarda AM. In vitro assessment of the antibacterial effects of the combinations of fosfomycin, colistin, trimethoprim and nitrofurantoin against multi-drug-resistant Escherichia coli. Lett Appl Microbiol 2021; 74:334-343. [PMID: 34839528 DOI: 10.1111/lam.13619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022]
Abstract
MDR UPEC has become a global health challenge. Our study investigates the pairwise interactions among FOS, COL, NIT and TRI against 29 UPEC strains using the checkerboard method. The synergistic combinations are further evaluated for their bactericidal effects against the most resistant strain (MRS) using the time-kill method. The results showed that 100% of these strains were resistant to TRI and NIT, whereas 75·86% of them were susceptible to FOS and COL. Among all tested strains, only seven strains were highly resistant to all used antibiotics. Remarkably, FOS/COL, COL/NIT and COL/TRI combinations represent the most effective synergistic (fractional inhibitory concentration index <1) combinations against the seven strains at MICs lower than the susceptible breakpoint ranges, followed by FOS/NIT and FOS/TRI, which achieved synergistic interactions against 1/7 and 2/7 of these strains. Importantly, the bactericidal effects (reduction ≥3·0 log10 CFU per ml) were only observed with FOS/COL, COL/NIT and COL/TRI combinations against MRS after 24 h of post-treatment. Our data suggested that FOS/COL, COL/NIT and COL/TRI combinations could be a promising option against MDR UPEC infections. Additionally, FOS/NIT and FOS/TRI probably represent a good option for MDR UPEC with lower MICs.
Collapse
Affiliation(s)
- W Mohamed Abu El-Wafa
- Department of Microbiology, General Division of Basic Medical Sciences, Egyptian Drug Authority (EDA), Formerly National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - A M Abouwarda
- Department of Microbiology, General Division of Basic Medical Sciences, Egyptian Drug Authority (EDA), Formerly National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
23
|
Zhang H, Zhu Y, Yang N, Kong Q, Zheng Y, Lv N, Chen H, Yue C, Liu Y, Li J, Ye Y. In vitro and in vivo Activity of Combinations of Polymyxin B with Other Antimicrobials Against Carbapenem-Resistant Acinetobacter baumannii. Infect Drug Resist 2021; 14:4657-4666. [PMID: 34764660 PMCID: PMC8577563 DOI: 10.2147/idr.s334200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/28/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To study the in vitro and in vivo antibacterial activities of polymyxin B (PB) and other five antimicrobial agents, including imipenem (IMP), meropenem (MEM), tigecycline (TGC), sulbactam (SUL), and rifampicin (RIF), alone or in combination against carbapenem-resistant Acinetobacter baumannii (CRAB). Methods Microbroth dilution method was used to determine the minimum inhibitory concentration (MIC) of ten strains of CRAB against six antibacterial drugs, and the checkerboard method was used to determine the fractional inhibitory concentration index (FICI). A mouse pneumonia model was established by intranasal instillation of Ab5075 to evaluate the antibacterial activity in vivo. Results The resistance rate of ten CRAB strains to IMP, MEM, and SUL was 100%, that to PB and TGC was 0%, and that to RIF was 20%. When PB was used in combination with the other five antibiotics in vitro, it mainly showed synergistic and additive effects on CRAB. The synergistic effect of PB and RIF was maximal, followed by MEM and IMP but was weak with SUL and TGC. In vivo, compared to the model group (untreated with antibiotics), treatment group (six antibiotics alone and PB combined with the other five antibiotics) reduced the bacterial load in the lung tissue and the serum inflammatory factors (IL-1β, IL-6, and TNF-α). The bacterial load and the inflammatory factors of the combined group decreased significantly than that of the single group (P<0.05). The IL-6 and TNF-α values of the PB combined with the RIF group were significantly lower than the two drugs used individually. Conclusion The combination of PB and IMP, MEM, and RIF exerted robust in vitro synergistic effects on CRAB isolates. The combination of PB and the other five antimicrobial agents had a better effect in the mouse pneumonia model than single agent, while the combination of PB and RIF had the best effect.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yunzhu Zhu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ning Yang
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Qinxiang Kong
- Department of Infectious Diseases, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yahong Zheng
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Na Lv
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Haoran Chen
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chengcheng Yue
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yanyan Liu
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Anhui Center for Surveillance of Bacterial Resistance, Hefei, Anhui, People's Republic of China.,Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Department of Infectious Diseases, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Anhui Center for Surveillance of Bacterial Resistance, Hefei, Anhui, People's Republic of China.,Institute of Bacterial Resistance, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ying Ye
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
24
|
Allend SO, Garcia MO, da Cunha KF, de Albernaz DTF, da Silva ME, Ishikame RY, Panagio LA, Nakazaro G, Reis GF, Pereira DB, Hartwig DD. Biogenic silver nanoparticle (Bio-AgNP) has an antibacterial effect against carbapenem-resistant Acinetobacter baumannii with synergism and additivity when combined with polymyxin B. J Appl Microbiol 2021; 132:1036-1047. [PMID: 34496109 DOI: 10.1111/jam.15297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/17/2021] [Accepted: 09/04/2021] [Indexed: 12/23/2022]
Abstract
AIMS Carbapenem-resistant Acinetobacter baumannii represents a public health problem, and the search for new antibacterial drugs has become a priority. Here, we investigate the antibacterial activity of biogenic silver nanoparticles (Bio-AgNPs) synthesized by Fusarium oxysporum, used alone or in combination with polymyxin B against carbapenem-resistant A. baumannii. METHODS AND RESULTS In this study, ATCC® 19606™ strain and four carbapenem-resistant A. baumannii strains were used. The antibacterial activity of Bio-AgNPs and its synergism with polymyxin B were determined using broth microdilution, checkboard methods and time-kill assays. The integrity of the bacterial cell membrane was monitored by protein leakage assay. In addition, the cytotoxicity in the VERO mammalian cell line was also evaluated, and the selectivity index was calculated. Bio-AgNPs have an antibacterial activity with MIC and MBC ranging from 0.460 to 1.870 µg/ml. The combination of polymyxin B and Bio-AgNPs presents synergy against four of the five strains tested and additivity against one strain in the checkerboard assay. Considering the time of cell death, Bio-AgNPs killed all carbapenem-resistant isolates and ATCC® 19606™ within 1 h. When combined, Bio-AgNPs presented 16-fold reduction of the polymyxin B MIC and showed a decrease in terms of viable A. baumannii cells in 4 h of treatment, with synergic and additive effects. Protein leakage was observed with increasing concentrations for Bio-AgNPs treatments. Additionally, Bio-AgNP and polymyxin B showed dose-dependent cytotoxicity against mammalian VERO cells and combined the cytotoxicity which was significantly reduced and presented a greater pharmacological safety. CONCLUSIONS The results presented here indicate that Bio-AgNPs in combination with polymyxin B could represent a good alternative in the treatment of carbapenem-resistant A. baumannii. SIGNIFICANCE AND IMPACT OF STUDY This study demonstrates the synergic effect between Bio-AgNPs and polymyxin B on carbapenem-resistant A. baumannii strains.
Collapse
Affiliation(s)
- Suzane Olachea Allend
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Marcelle Oliveira Garcia
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Kamila Furtado da Cunha
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | | | - Mirian Elert da Silva
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Rodrigo Yudi Ishikame
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | | | - Gerson Nakazaro
- Department of Microbiology, State University of Londrina, Londrina, PR, Brazil
| | | | - Daniela Brayer Pereira
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Daiane Drawanz Hartwig
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, Brazil
| |
Collapse
|
25
|
Lazar V, Holban AM, Curutiu C, Chifiriuc MC. Modulation of Quorum Sensing and Biofilms in Less Investigated Gram-Negative ESKAPE Pathogens. Front Microbiol 2021; 12:676510. [PMID: 34394026 PMCID: PMC8359898 DOI: 10.3389/fmicb.2021.676510] [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: 03/05/2021] [Accepted: 06/30/2021] [Indexed: 01/20/2023] Open
Abstract
Pathogenic bacteria have the ability to sense their versatile environment and adapt by behavioral changes both to the external reservoirs and the infected host, which, in response to microbial colonization, mobilizes equally sophisticated anti-infectious strategies. One of the most important adaptive processes is the ability of pathogenic bacteria to turn from the free, floating, or planktonic state to the adherent one and to develop biofilms on alive and inert substrata; this social lifestyle, based on very complex communication networks, namely, the quorum sensing (QS) and response system, confers them an increased phenotypic or behavioral resistance to different stress factors, including host defense mechanisms and antibiotics. As a consequence, biofilm infections can be difficult to diagnose and treat, requiring complex multidrug therapeutic regimens, which often fail to resolve the infection. One of the most promising avenues for discovering novel and efficient antibiofilm strategies is targeting individual cells and their QS mechanisms. A huge amount of data related to the inhibition of QS and biofilm formation in pathogenic bacteria have been obtained using the well-established gram-positive Staphylococcus aureus and gram-negative Pseudomonas aeruginosa models. The purpose of this paper was to revise the progress on the development of antibiofilm and anti-QS strategies in the less investigated gram-negative ESKAPE pathogens Klebsiella pneumoniae, Acinetobacter baumannii, and Enterobacter sp. and identify promising leads for the therapeutic management of these clinically significant and highly resistant opportunistic pathogens.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Carmen Curutiu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
26
|
Antonello RM, Di Bella S, Maraolo AE, Luzzati R. Fosfomycin in continuous or prolonged infusion for systemic bacterial infections: a systematic review of its dosing regimen proposal from in vitro, in vivo and clinical studies. Eur J Clin Microbiol Infect Dis 2021; 40:1117-1126. [PMID: 33604721 PMCID: PMC8139892 DOI: 10.1007/s10096-021-04181-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Fosfomycin (FOS) administered intravenously has been recently rediscovered for the treatment of systemic infections due to multidrug-resistant bacteria. Its pharmacokinetic properties suggest a time-dependent dosing schedule with more clinical benefits from prolonged (PI) or continuous infusion (CI) than from intermittent infusion. We revised literature concerning PI and CI FOS to identify the best dosing regimen based on current evidence. We performed a MEDLINE/PubMed search. Ninety-one studies and their pertinent references were screened. Seventeen studies were included in the present review. The activity of FOS against Gram-negative and Gram-positive bacteria was evaluated in fourteen and five studies, respectively. Six studies evaluated FOS activity in combination with another antibiotic. Daily dosing of 12, 16, 18 or 24 g, administered with different schedules, were investigated. These regimens resulted active against the tested isolates in most cases. Emergence of resistant isolates has been shown to be preventable through the coadministration of another active antibiotic. FOS is a promising option to treat systemic infections caused by multidrug-resistant bacteria. Coadministration with another active molecule is required to prevent the emergence of resistant bacterial strains. The results of our review suggest that a therapeutic regimen including a loading dose of FOS 8 g followed by a daily dose of 16 g or 24 g CI could be the best therapeutic approach for patients with normal renal function. The dosing regimens in patients with renal insufficiency and CI or PI superiority compared with intermittent infusion in clinical settings should be further investigated.
Collapse
Affiliation(s)
- Roberta Maria Antonello
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy
| | - Alberto Enrico Maraolo
- First Division of Infectious Diseases, Cotugno Hospital, AORN dei Colli, 80131 Naples, Italy
| | - Roberto Luzzati
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy
| |
Collapse
|
27
|
Oh S, Chau R, Nguyen AT, Lenhard JR. Losing the Battle but Winning the War: Can Defeated Antibacterials Form Alliances to Combat Drug-Resistant Pathogens? Antibiotics (Basel) 2021; 10:antibiotics10060646. [PMID: 34071451 PMCID: PMC8227011 DOI: 10.3390/antibiotics10060646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the recent development of antibacterials that are active against multidrug-resistant pathogens, drug combinations are often necessary to optimize the killing of difficult-to-treat organisms. Antimicrobial combinations typically are composed of multiple agents that are active against the target organism; however, many studies have investigated the potential utility of combinations that consist of one or more antibacterials that individually are incapable of killing the relevant pathogen. The current review summarizes in vitro, in vivo, and clinical studies that evaluate combinations that include at least one drug that is not active individually against Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii, or Staphylococcus aureus. Polymyxins were often included in combinations against all three of the Gram-negative pathogens, and carbapenems were commonly incorporated into combinations against K. pneumoniae and A. baumannii. Minocycline, sulbactam, and rifampin were also frequently investigated in combinations against A. baumannii, whereas the addition of ceftaroline or another β-lactam to vancomycin or daptomycin showed promise against S. aureus with reduced susceptibility to vancomycin or daptomycin. Although additional clinical studies are needed to define the optimal combination against specific drug-resistant pathogens, the large amount of in vitro and in vivo studies available in the literature may provide some guidance on the rational design of antibacterial combinations.
Collapse
|
28
|
Russo A, Bassetti M, Bellelli V, Bianchi L, Marincola Cattaneo F, Mazzocchetti S, Paciacconi E, Cottini F, Schiattarella A, Tufaro G, Sabetta F, D'Avino A. Efficacy of a Fosfomycin-Containing Regimen for Treatment of Severe Pneumonia Caused by Multidrug-Resistant Acinetobacter baumannii: A Prospective, Observational Study. Infect Dis Ther 2021; 10:187-200. [PMID: 33068255 PMCID: PMC7568458 DOI: 10.1007/s40121-020-00357-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/08/2020] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Severe pneumonia caused by multidrug-resistant Acinetobacter baumannii (MDR-AB) remains a difficult-to-treat infection. Considering the poor lung penetration of most antibiotics, the choice of the better antibiotic regimen is debated. METHODS We performed a prospective, observational, multicenter study conducted from January 2017 to June 2020. All consecutive hospitalized patients with severe pneumonia due to MDR-AB were included in the study. The primary endpoint of the study was to evaluate risk factors associated with survival or death at 30 days from pneumonia onset. A propensity score for receiving therapy with fosfomycin was added to the model. RESULTS During the study period, 180 cases of hospital-acquired pneumonia, including ventilator-associated pneumonia, caused by MDR-AB strains were observed. Cox regression analysis of factors associated with 30-day mortality, after propensity score, showed that septic shock, and secondary bacteremia were associated with death, while a fosfomycin-containing regimen was associated with 30-day survival. Antibiotic combinations with fosfomycin in definitive therapy for 44 patients were: fosfomycin + colistin in 11 (25%) patients followed by fosfomycin + carbapenem + tigecycline in 8 (18.2%), fosfomycin + colistin + tigecycline in 7 (15.9%), fosfomycin + rifampin in 7 (15.9%), fosfomycin + tigecycline in 6 (13.6%), fosfomycin + carbapenem in 3 (6.8%), and fosfomycin + aminoglycoside in 2 (4.5%). CONCLUSIONS This real-life clinical experience concerning the therapeutic approach to severe pneumonia caused by MDR-AB provides useful suggestions to clinicians, showing the use of different antibiotic regimens with a predominant role for fosfomycin. Further randomized clinical trials are necessary to confirm or exclude these observations.
Collapse
Affiliation(s)
- Alessandro Russo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
- Internal Medicine Unit, Policlinico Casilino, Rome, Italy.
| | - Matteo Bassetti
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | | | - Luigi Bianchi
- Internal Medicine Unit, Policlinico Casilino, Rome, Italy
| | | | | | - Elena Paciacconi
- Department of Intensive Care Unit, Cristo Re Hospital, Rome, Italy
| | - Fabrizio Cottini
- Intensive Care Unit, San Carlo di Nancy Hospital-GVM Care and Research, Rome, Italy
| | | | | | | | - Alessandro D'Avino
- Department of Internal Medicine and Risk Management, Cristo Re Hospital, Rome, Italy
| |
Collapse
|
29
|
Nwabor OF, Terbtothakun P, Voravuthikunchai SP, Chusri S. Evaluation of the Synergistic Antibacterial Effects of Fosfomycin in Combination with Selected Antibiotics against Carbapenem-Resistant Acinetobacter baumannii. Pharmaceuticals (Basel) 2021; 14:185. [PMID: 33668905 PMCID: PMC7996625 DOI: 10.3390/ph14030185] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/22/2022] Open
Abstract
The spread of multi-drug resistant (MDR) pathogens and the lagging pace in the development of novel chemotherapeutic agents warrant the use of combination therapy as a reliable, cost-effective interim option. In this study, the synergistic effects of fosfomycin in combination with other antibiotics were assessed. Of the 193 isolates, 90.6% were non-susceptible to fosfomycin, with minimum inhibitory concentrations (MICs) of ≥128 µg/mL. Antibacterial evaluation of fosfomycin-resistant isolates indicated multi-drug resistance to various antibiotic classes. Combinations of fosfomycin with 12 commonly used antibiotics synergistically inhibited most fosfomycin-resistant isolates. The fractional inhibitory concentration index indicated that combining fosfomycin with either aminoglycosides, glycylcyclines, fluoroquinolones, or colistin resulted in 2- to 16-fold reduction in the MIC of fosfomycin. Time-kill kinetics further confirmed the synergistic bactericidal effects of fosfomycin in combination with either amikacin, gentamicin, tobramycin, minocycline, tigecycline, or colistin, with more than 99.9% reduction in bacterial cells. Fosfomycin-based combination therapy might serve as an alternative option for the treatment of MDR A. baumannii. Further steps including in vivo efficacy and toxicity in experimental models of infection are required prior to clinical applications.
Collapse
Affiliation(s)
- Ozioma F. Nwabor
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; (O.F.N.); (P.T.)
- Division of Biological Science, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand;
| | - Pawarisa Terbtothakun
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; (O.F.N.); (P.T.)
- Division of Biological Science, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand;
| | - Supayang P. Voravuthikunchai
- Division of Biological Science, Faculty of Science and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand;
| | - Sarunyou Chusri
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; (O.F.N.); (P.T.)
| |
Collapse
|
30
|
Rahayuningtyas I, Indrawati A, Wibawan IWT, Palupi MF, Istiyaningsih I. Phylogenetic group determination and plasmid virulence gene profiles of colistin-resistant Escherichia coli originated from the broiler meat supply chain in Bogor, Indonesia. Vet World 2020; 13:1807-1814. [PMID: 33132591 PMCID: PMC7566268 DOI: 10.14202/vetworld.2020.1807-1814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/21/2020] [Indexed: 11/23/2022] Open
Abstract
Background and Aim: Pathogenic Escherichia coli contamination along the broiler meat supply chain is a serious public health concern. This bacterial infection with multidrug-resistant can lead to treatment failure. Several studies have revealed that avian pathogenic E. coli (APEC) and human extraintestinal pathogenic E. coli (ExPEC) showed a close genetic relationship and may share virulence genes. This study aimed to determine the phylogenetic group and virulence gene profiles in colistin-resistant E. coli obtained from the broiler meat supply chain in Bogor, West Java, Indonesia. Materials and Methods: Fifty-eight archive isolates originated from the cloacal swab, litter, drinking water, inside plucker swab, fresh meat at small scale poultry slaughterhouses, and traditional markets were used in this study. All the isolates were characterized by a polymerase chain reaction to determine the phylogenetic group (A, B1, B2, or D) and virulence gene profiles with APEC marker genes (iutA, hlyF, iss, iroN, and ompT). Results: Phylogenetic grouping revealed that the isolates belong to A group (34.48%), D group (34.48%), B1 group (17.24%), and B2 group (13.79%). The virulence gene prevalence was as follows: iutA (36%), hlyF (21%), ompT (21%), iroN (10%), and iss (9%). The B2 group presented with more virulence genes combinations. iroN, hlyF, and ompT genes were positively associated with the B2 group (p≤0.05). Conclusion: Our results highlight the role of colistin-resistant E. coli originated from the broiler meat supply chain as a potential reservoir for human ExPEC virulence genes.
Collapse
Affiliation(s)
- Irma Rahayuningtyas
- Department of Animal Disease and Veterinary Public Health, Faculty of Veterinary Medicine, IPB University-Bogor, Indonesia.,National Veterinary Drug Assay Laboratory, Directorate General of Livestock and Animal Health, Ministry of Agriculture of the Republic of Indonesia, Indonesia
| | - Agustin Indrawati
- Department of Animal Disease and Veterinary Public Health, Faculty of Veterinary Medicine, IPB University-Bogor, Indonesia
| | - I Wayan Teguh Wibawan
- Department of Animal Disease and Veterinary Public Health, Faculty of Veterinary Medicine, IPB University-Bogor, Indonesia
| | - Maria Fatima Palupi
- National Veterinary Drug Assay Laboratory, Directorate General of Livestock and Animal Health, Ministry of Agriculture of the Republic of Indonesia, Indonesia
| | - Istiyaningsih Istiyaningsih
- National Veterinary Drug Assay Laboratory, Directorate General of Livestock and Animal Health, Ministry of Agriculture of the Republic of Indonesia, Indonesia
| |
Collapse
|
31
|
Kareem SM. Emergence of mcr- and fosA3-mediated colistin and fosfomycin resistance among carbapenem-resistant Acinetobacter baumannii in Iraq. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
32
|
Antonello RM, Principe L, Maraolo AE, Viaggi V, Pol R, Fabbiani M, Montagnani F, Lovecchio A, Luzzati R, Di Bella S. Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies. Antibiotics (Basel) 2020; 9:antibiotics9080500. [PMID: 32785114 PMCID: PMC7460049 DOI: 10.3390/antibiotics9080500] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/31/2022] Open
Abstract
Fosfomycin is being increasingly prescribed for multidrug-resistant bacterial infections. In patients with systemic involvement, intravenous fosfomycin is usually administered as a partner drug, as part of an antibiotic regimen. Hence, the knowledge of fosfomycin pharmacodynamic interactions (synergistic, additive, indifferent and antagonistic effect) is fundamental for a proper clinical management of severe bacterial infections. We performed a systematic review to point out fosfomycin’s synergistic properties, when administered with other antibiotics, in order to help clinicians to maximize drug efficacy optimizing its use in clinical practice. Interactions were more frequently additive or indifferent (65.4%). Synergism accounted for 33.7% of total interactions, while antagonism occurred sporadically (0.9%). Clinically significant synergistic interactions were mostly distributed in combination with penicillins (51%), carbapenems (43%), chloramphenicol (39%) and cephalosporins (33%) in Enterobactaerales; with linezolid (74%), tetracyclines (72%) and daptomycin (56%) in Staphylococcus aureus; with chloramphenicol (53%), aminoglycosides (43%) and cephalosporins (36%) against Pseudomonas aeruginosa; with daptomycin (97%) in Enterococcus spp. and with sulbactam (75%) and penicillins (60%) and in Acinetobacter spp. fosfomycin-based antibiotic associations benefit from increase in the bactericidal effect and prevention of antimicrobial resistances. Taken together, the presence of synergistic interactions and the nearly total absence of antagonisms, make fosfomycin a good partner drug in clinical practice.
Collapse
Affiliation(s)
- Roberta Maria Antonello
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | | | - Alberto Enrico Maraolo
- First Division of Infectious Diseases, Cotugno Hospital, AORN dei Colli, 80131 Naples, Italy;
| | | | - Riccardo Pol
- Department of Infectious Diseases, Udine University, 33100 Udine, Italy;
| | - Massimiliano Fabbiani
- Department of Medical Sciences, Tropical and Infectious Diseases Unit, University Hospital of Siena, 53100 Siena, Italy; (M.F.); (F.M.)
| | - Francesca Montagnani
- Department of Medical Sciences, Tropical and Infectious Diseases Unit, University Hospital of Siena, 53100 Siena, Italy; (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Antonio Lovecchio
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | - Roberto Luzzati
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy; (R.M.A.); (A.L.); (R.L.)
- Correspondence:
| |
Collapse
|