1
|
Choi BJ, Choi U, Ryu DB, Lee CR. PhoPQ-mediated lipopolysaccharide modification governs intrinsic resistance to tetracycline and glycylcycline antibiotics in Escherichia coli. mSystems 2024; 9:e0096424. [PMID: 39345149 PMCID: PMC11495068 DOI: 10.1128/msystems.00964-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
Tetracyclines and glycylcycline are among the important antibiotics used to combat infections caused by multidrug-resistant Gram-negative pathogens. Despite the clinical importance of these antibiotics, their mechanisms of resistance remain unclear. In this study, we elucidated a novel mechanism of resistance to tetracycline and glycylcycline antibiotics via lipopolysaccharide (LPS) modification. Disruption of the Escherichia coli PhoPQ two-component system, which regulates the transcription of various genes involved in magnesium transport and LPS modification, leads to increased susceptibility to tetracycline, minocycline, doxycycline, and tigecycline. These phenotypes are caused by enhanced expression of phosphoethanolamine transferase EptB, which catalyzes the modification of the inner core sugar of LPS. PhoPQ-mediated regulation of EptB expression appears to affect the intracellular transportation of doxycycline. Disruption of EptB increases resistance to tetracycline and glycylcycline antibiotics, whereas the other two phosphoethanolamine transferases, EptA and EptC, that participate in the modification of other LPS residues, are not associated with resistance to tetracyclines and glycylcycline. Overall, our results demonstrated that PhoPQ-mediated modification of a specific residue of LPS by phosphoethanolamine transferase EptB governs intrinsic resistance to tetracycline and glycylcycline antibiotics. IMPORTANCE Elucidating the resistance mechanisms of clinically important antibiotics helps in maintaining the clinical efficacy of antibiotics and in the prescription of adequate antibiotic therapy. Although tetracycline and glycylcycline antibiotics are clinically important in combating multidrug-resistant Gram-negative bacterial infections, their mechanisms of resistance are not fully understood. Our research demonstrates that the E. coli PhoPQ two-component system affects resistance to tetracycline and glycylcycline antibiotics by controlling the expression of phosphoethanolamine transferase EptB, which catalyzes the modification of the inner core residue of lipopolysaccharide (LPS). Therefore, our findings highlight a novel resistance mechanism to tetracycline and glycylcycline antibiotics and the physiological significance of LPS core modification in E. coli.
Collapse
Affiliation(s)
- Byoung Jun Choi
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Umji Choi
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Dae-Beom Ryu
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| | - Chang-Ro Lee
- Department of Biological Sciences, Myongji University, Yongin, Gyeonggido, Republic of Korea
| |
Collapse
|
2
|
Bayoudh F, Giot JB, Descy J, Fontaine C, Hayette MP, Baron F, Willems E, Beguin Y, Frippiat F, Servais S. Oral minocycline as systemic therapy for uncomplicated venous access device-related bloodstream infection with coagulase-negative staphylococci after allogeneic hematopoietic cell transplantation. Curr Res Transl Med 2024; 72:103422. [PMID: 38244302 DOI: 10.1016/j.retram.2023.103422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/03/2023] [Accepted: 10/15/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Venous access device-related bloodstream infection (VAD-BSI) with coagulase-negative staphylococci (CoNS) is a common complication after allogeneic hematopoietic cell transplantation (alloHCT). Standard systemic antimicrobial therapy for uncomplicated VAD-BSI with methicillin-resistant CoNS consists of intravenous (IV) vancomycin (vanco). This requires hospitalization, needs new competent venous access, exposes patients to potential toxicity (mainly renal) and increases the risk of commensal flora dysbiosis with selection of vanco-resistant enterococci. Combined with VAD management (removal or antibiotic locks), oral minocycline (mino) has been evaluated as an alternative systemic therapy for the treatment of uncomplicated VAD-BSIs with CoNS at our center, primarily when the reference treatment with IV vanco was not possible (renal failure or allergy) or when hospitalization was refused by patients. Here, we retrospectively report our single center experience with this mino-based approach. PATIENTS AND METHODS From January 2012 to December 2020, 24 uncomplicated VAD-BSIs with CoNS in 23 alloHCT patients were treated with oral mino as systemic antibiotic therapy in combination with VAD management. VAD were implantable ports (n = 17), tunneled catheter (n = 1) or PIC-lines (n = 6). Staphylococci were S. epidermidis (n = 21) or S. haemolyticus (n = 3). Mino was administered with a loading dose of 200 mg followed by 100 mg BID for 7-14 days. For 8 VAD-BSIs, patients were initially treated with IV vanco for the first 1-3 days followed by oral mino, while 16 VAD-BSIs were treated with oral mino as the sole antimicrobial agent for systemic therapy. VAD management consisted of catheter removal (for tunneled catheters and PIC-lines, n = 7) or antibiotic locks with vanco (n = 15) or gentamicin (n = 2) administered at least 3 times a week for 14 days (for ports). RESULTS Overall, clearance of bacteremia (as assessed by negativity for the same CoNS of surveillance peripheral blood cultures drawn between day+ 3 and +30 after initiation of systemic therapy) was achieved in all but 1 patient (with port) who had persistent bacteremia at day +9. No complication such as suppurative thrombophlebitis, endocarditis, distant foci of infection or BSI-related death was observed in any patient during the 3-month period after initiation of treatment. Regarding the 17 port-BSI cases for which VAD conservative strategy was attempted, failure of 3-month VAD preservation was documented in 7/17 cases and 3-month recurrence of VAD-BSI was observed in 3/17 cases (with 1 patient with cellulitis). Treatment with mino was well tolerated except for a mild skin rash in one patient. CONCLUSION Further prospective studies are needed to evaluate efficacy and safety of this approach.
Collapse
Affiliation(s)
- Firas Bayoudh
- Department of Clinical Hematology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Jean-Baptiste Giot
- Department of Infectious Disease, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Julie Descy
- Laboratory of Clinical Microbiology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Corentin Fontaine
- Laboratory of Clinical Microbiology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Marie-Pierre Hayette
- Laboratory of Clinical Microbiology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Clinical Hematology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Evelyne Willems
- Department of Clinical Hematology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Yves Beguin
- Department of Clinical Hematology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Frédéric Frippiat
- Department of Infectious Disease, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium
| | - Sophie Servais
- Department of Clinical Hematology, University Hospital of Liège, CHU Sart-Tilman, 4000 Liège, Belgium.
| |
Collapse
|
3
|
Zheng Q, Lin R, Chen Y, Lv Q, Zhang J, Zhai J, Xu W, Wang W. SARS-CoV-2 induces "cytokine storm" hyperinflammatory responses in RA patients through pyroptosis. Front Immunol 2022; 13:1058884. [PMID: 36532040 PMCID: PMC9751040 DOI: 10.3389/fimmu.2022.1058884] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Background The coronavirus disease (COVID-19) is a pandemic disease that threatens worldwide public health, and rheumatoid arthritis (RA) is the most common autoimmune disease. COVID-19 and RA are each strong risk factors for the other, but their molecular mechanisms are unclear. This study aims to investigate the biomarkers between COVID-19 and RA from the mechanism of pyroptosis and find effective disease-targeting drugs. Methods We obtained the common gene shared by COVID-19, RA (GSE55235), and pyroptosis using bioinformatics analysis and then did the principal component analysis(PCA). The Co-genes were evaluated by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and ClueGO for functional enrichment, the protein-protein interaction (PPI) network was built by STRING, and the k-means machine learning algorithm was employed for cluster analysis. Modular analysis utilizing Cytoscape to identify hub genes, functional enrichment analysis with Metascape and GeneMANIA, and NetworkAnalyst for gene-drug prediction. Network pharmacology analysis was performed to identify target drug-related genes intersecting with COVID-19, RA, and pyroptosis to acquire Co-hub genes and construct transcription factor (TF)-hub genes and miRNA-hub genes networks by NetworkAnalyst. The Co-hub genes were validated using GSE55457 and GSE93272 to acquire the Key gene, and their efficacy was assessed using receiver operating curves (ROC); SPEED2 was then used to determine the upstream pathway. Immune cell infiltration was analyzed using CIBERSORT and validated by the HPA database. Molecular docking, molecular dynamics simulation, and molecular mechanics-generalized born surface area (MM-GBSA) were used to explore and validate drug-gene relationships through computer-aided drug design. Results COVID-19, RA, and pyroptosis-related genes were enriched in pyroptosis and pro-inflammatory pathways(the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome complex, death-inducing signaling complex, regulation of interleukin production), natural immune pathways (Network map of SARS-CoV-2 signaling pathway, activation of NLRP3 inflammasome by SARS-CoV-2) and COVID-19-and RA-related cytokine storm pathways (IL, nuclear factor-kappa B (NF-κB), TNF signaling pathway and regulation of cytokine-mediated signaling). Of these, CASP1 is the most involved pathway and is closely related to minocycline. YY1, hsa-mir-429, and hsa-mir-34a-5p play an important role in the expression of CASP1. Monocytes are high-caspase-1-expressing sentinel cells. Minocycline can generate a highly stable state for biochemical activity by docking closely with the active region of caspase-1. Conclusions Caspase-1 is a common biomarker for COVID-19, RA, and pyroptosis, and it may be an important mediator of the excessive inflammatory response induced by SARS-CoV-2 in RA patients through pyroptosis. Minocycline may counteract cytokine storm inflammation in patients with COVID-19 combined with RA by inhibiting caspase-1 expression.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Rongjie Lin
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Yuchao Chen
- Department of Paediatrics, Fujian Provincial Hospital South Branch, Fuzhou, China
| | - Qi Lv
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Weihong Xu
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China,*Correspondence: Weihong Xu, ; Wanming Wang,
| | - Wanming Wang
- Department of Orthopedics, 900th Hospital of Joint Logistics Support Force, Fuzhou, China,*Correspondence: Weihong Xu, ; Wanming Wang,
| |
Collapse
|
4
|
Paranos P, Vourli S, Pournaras S, Meletiadis J. Assessing Clinical Potential of Old Antibiotics against Severe Infections by Multi-Drug-Resistant Gram-Negative Bacteria Using In Silico Modelling. Pharmaceuticals (Basel) 2022; 15:1501. [PMID: 36558952 PMCID: PMC9781251 DOI: 10.3390/ph15121501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
In the light of increasing antimicrobial resistance among gram-negative bacteria and the lack of new more potent antimicrobial agents, new strategies have been explored. Old antibiotics, such as colistin, temocillin, fosfomycin, mecillinam, nitrofurantoin, minocycline, and chloramphenicol, have attracted the attention since they often exhibit in vitro activity against multi-drug-resistant (MDR) gram-negative bacteria, such as Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The current review provides a summary of the in vitro activity, pharmacokinetics and PK/PD characteristics of old antibiotics. In silico modelling was then performed using Monte Carlo simulation in order to combine all preclinical data with human pharmacokinetics and determine the probability of target (1-log kill in thigh/lung infection animal models) attainment (PTA) of different dosing regimens. The potential of clinical efficacy of a drug against severe infections by MDR gram-negative bacteria was considered when PTA was >95% at the epidemiological cutoff values of corresponding species. In vitro potent activity against MDR gram-negative pathogens has been shown for colistin, polymyxin B, temocillin (against E. coli and K. pneumoniae), fosfomycin (against E. coli), mecillinam (against E. coli), minocycline (against E. coli, K. pneumoniae, A. baumannii), and chloramphenicol (against E. coli) with ECOFF or MIC90 ≤ 16 mg/L. When preclinical PK/PD targets were combined with human pharmacokinetics, Monte Carlo analysis showed that among the old antibiotics analyzed, there is clinical potential for polymyxin B against E. coli, K. pneumoniae, and A. baumannii; for temocillin against K. pneumoniae and E. coli; for fosfomycin against E. coli and K. pneumoniae; and for mecillinam against E. coli. Clinical studies are needed to verify the potential of those antibiotics to effectively treat infections by multi-drug resistant gram-negative bacteria.
Collapse
Affiliation(s)
- Paschalis Paranos
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Sophia Vourli
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Spyros Pournaras
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
5
|
Gao W, Han X, Li Y, Zhou Z, Wang J, Shi R, Jiao J, Qi Y, Zhou Y, Zhao J. Modification strategies for improving antibacterial properties of polyetheretherketone. J Appl Polym Sci 2022. [DOI: 10.1002/app.52847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Weijia Gao
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Xiao Han
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Yongli Li
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Zhe Zhou
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Junyan Wang
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Ruining Shi
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Junjie Jiao
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Yuanzheng Qi
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Yanmin Zhou
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| | - Jinghui Zhao
- Hospital of Stomatology Jilin University Changchun Jilin Province China
| |
Collapse
|
6
|
Qu X, Bian X, Chen Y, Hu J, Huang X, Wang Y, Fan Y, Wu H, Li X, Li Y, Guo B, Liu X, Zhang J. Polymyxin B Combined with Minocycline: A Potentially Effective Combination against blaOXA-23-harboring CRAB in In Vitro PK/PD Model. Molecules 2022; 27:molecules27031085. [PMID: 35164349 PMCID: PMC8840471 DOI: 10.3390/molecules27031085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
Polymyxin-based combination therapy is commonly used to treat carbapenem-resistant Acinetobacter baumannii (CRAB) infections. In the present study, the bactericidal effect of polymyxin B and minocycline combination was tested in three CRAB strains containing blaOXA-23 by the checkerboard assay and in vitro dynamic pharmacokinetics/pharmacodynamics (PK/PD) model. The combination showed synergistic or partial synergistic effect (fractional inhibitory concentration index ≤0.56) on the tested strains in checkboard assays. The antibacterial activity was enhanced in the combination group compared with either monotherapy in in vitro PK/PD model. The combination regimen (simultaneous infusion of 0.75 mg/kg polymyxin B and 100 mg minocycline via 2 h infusion) reduced bacterial colony counts by 0.9–3.5 log10 colony forming units per milliliter (CFU/mL) compared with either drug alone at 24 h. In conclusion, 0.75 mg/kg polymyxin B combined with 100 mg minocycline via 2 h infusion could be a promising treatment option for CRAB bloodstream infections.
Collapse
Affiliation(s)
- Xingyi Qu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xingchen Bian
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuancheng Chen
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiali Hu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Xiaolan Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Yu Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Yaxin Fan
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Hailan Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Xin Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Yi Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Beining Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
| | - Xiaofen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Correspondence: (X.L.); (J.Z.); Tel.: +86-21-52888190 (J.Z.)
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China; (X.Q.); (X.B.); (J.H.); (X.H.); (Y.W.); (Y.F.); (H.W.); (X.L.); (Y.L.); (B.G.)
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai 200040, China
- National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: (X.L.); (J.Z.); Tel.: +86-21-52888190 (J.Z.)
| |
Collapse
|
7
|
Li B, Shi L, Liu R, Li Z, Cao S, Li J. A lingering mouthwash with sustained antibiotic release and biofilm eradication for periodontitis. J Mater Chem B 2021; 9:8694-8707. [PMID: 34622266 DOI: 10.1039/d1tb01742j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dental plaque biofilms are believed to be one of the principal virulence factors in periodontitis resulting in tooth loss. Traditional mouthwashes are limited due to the continuous flow of saliva and poor drug penetration ability in the biofilm. Herein, we fabricated an antibiotic delivery platform based on natural polysaccharides (chitosan and cyclodextrin) as a novel mouthwash for the topical cavity delivery of minocycline. The penetration and residence mechanisms demonstrate that the platform can prolong the residence time up to 12 h on biofilms. Furthermore, sustained release can enhance the penetration of drugs into biofilms. In vitro antibiofilm experimental results indicated that the mouthwash effectively kills bacteria and eradicate biofilms. Effective treatment in vivo was confirmed by the significantly reduced dental plaque and alleviated inflammation observed in a rat periodontitis model. In summary, this novel platform can improve antibiofilm efficiency and prevent drugs from being washed away by saliva, which may provide benefits for many oral infectious diseases.
Collapse
Affiliation(s)
- Bohua Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Liuqi Shi
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China. .,School of Material Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Ruixing Liu
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Zhanrong Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Shaokui Cao
- School of Material Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Jingguo Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China. .,School of Material Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
8
|
Shortridge D, Arends SJR, Streit JM, Castanheira M. Minocycline Activity against Unusual Clinically Significant Gram-Negative Pathogens. Antimicrob Agents Chemother 2021; 65:e0126421. [PMID: 34491809 PMCID: PMC8522744 DOI: 10.1128/aac.01264-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 11/20/2022] Open
Abstract
The minocycline susceptibility of 3,856 isolates including Burkholderia, Achromobacter, Alcaligenes, Aeromonas, and Stenotrophomonas maltophilia from the SENTRY surveillance (2014 to 2019) were analyzed. The susceptibilities of these species (%S) were Achromobacter spp. (n = 411; 92.6%), Burkholderia cepacia species complex (n = 199; 85.9%), Aeromonas spp. (n = 127; 99.2%), Chryseobacterium spp. (n = 59; 94.9%), Alcaligenes faecalis (n = 42; 88.1%), and S. maltophilia (n = 2,287; 99.5%). These data suggest that minocycline is a useful treatment option for infections caused by unusual Gram-negative pathogens.
Collapse
|
9
|
Chang JB, Chen Y, Wang H, Ma X, Zhang X, Wu H, Ma B, Zuo W, Ma W, Wang R, Wei J. Combined Strategy for Post-Operative Patients with Central Nervous System Infections Caused by Extensively Drug-Resistant/Pan-Drug–Resistant Acinetobacter baumannii: A Retrospective Study. Surg Infect (Larchmt) 2020; 21:853-858. [PMID: 32302500 DOI: 10.1089/sur.2019.341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jian-bo Chang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yihao Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - He Wang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojun Ma
- Department of Infectious Disease, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Wu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Baitao Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zuo
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Junji Wei
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Abstract
Omadacycline is a novel aminomethylcycline antibiotic developed as a once-daily, intravenous and oral treatment for acute bacterial skin and skin structure infection (ABSSSI) and community-acquired bacterial pneumonia (CABP). Omadacycline, a derivative of minocycline, has a chemical structure similar to tigecycline with an alkylaminomethyl group replacing the glycylamido group at the C-9 position of the D-ring of the tetracycline core. Similar to other tetracyclines, omadacycline inhibits bacterial protein synthesis by binding to the 30S ribosomal subunit. Omadacycline possesses broad-spectrum antibacterial activity against Gram-positive and Gram-negative aerobic, anaerobic, and atypical bacteria. Omadacycline remains active against bacterial isolates possessing common tetracycline resistance mechanisms such as efflux pumps (e.g., TetK) and ribosomal protection proteins (e.g., TetM) as well as in the presence of resistance mechanisms to other antibiotic classes. The pharmacokinetics of omadacycline are best described by a linear, three-compartment model following a zero-order intravenous infusion or first-order oral administration with transit compartments to account for delayed absorption. Omadacycline has a volume of distribution (Vd) ranging from 190 to 204 L, a terminal elimination half-life (t½) of 13.5-17.1 h, total clearance (CLT) of 8.8-10.6 L/h, and protein binding of 21.3% in healthy subjects. Oral bioavailability of omadacycline is estimated to be 34.5%. A single oral dose of 300 mg (bioequivalent to 100 mg IV) of omadacycline administered to fasted subjects achieved a maximum plasma concentration (Cmax) of 0.5-0.6 mg/L and an area under the plasma concentration-time curve from 0 to infinity (AUC0-∞) of 9.6-11.9 mg h/L. The free plasma area under concentration-time curve divided by the minimum inhibitory concentration (i.e., fAUC24h/MIC), has been established as the pharmacodynamic parameter predictive of omadacycline antibacterial efficacy. Several animal models including neutropenic murine lung infection, thigh infection, and intraperitoneal challenge model have documented the in vivo antibacterial efficacy of omadacycline. A phase II clinical trial on complicated skin and skin structure infection (cSSSI) and three phase III clinical trials on ABSSSI and CABP demonstrated the safety and efficacy of omadacycline. The phase III trials, OASIS-1 (ABSSSI), OASIS-2 (ABSSSI), and OPTIC (CABP), established non-inferiority of omadacycline to linezolid (OASIS-1, OASIS-2) and moxifloxacin (OPTIC), respectively. Omadacycline is currently approved by the FDA for use in treatment of ABSSSI and CABP. Phase II clinical trials involving patients with acute cystitis and acute pyelonephritis are in progress. Mild, transient gastrointestinal events are the predominant adverse effects associated with use of omadacycline. Based on clinical trial data to date, the adverse effect profile of omadacycline is similar to studied comparators, linezolid and moxifloxacin. Unlike tigecycline and eravacycline, omadacycline has an oral formulation that allows for step-down therapy from the intravenous formulation, potentially facilitating earlier hospital discharge, outpatient therapy, and cost savings. Omadacycline has a potential role as part of an antimicrobial stewardship program in the treatment of patients with infections caused by antibiotic-resistant and multidrug-resistant Gram-positive [including methicillin-resistant Staphylococcus aureus (MRSA)] and Gram-negative pathogens.
Collapse
|
11
|
Bart G, Zeller V, Kerroumi Y, Heym B, Meyssonnier V, Desplaces N, Kitzis MD, Ziza JM, Marmor S. Minocycline Combined with Vancomycin for the Treatment of Methicillin-Resistant Coagulase-Negative Staphylococcal Prosthetic Joint Infection Managed with Exchange Arthroplasty. J Bone Jt Infect 2020; 5:110-117. [PMID: 32566448 PMCID: PMC7295649 DOI: 10.7150/jbji.43254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/14/2020] [Indexed: 11/09/2022] Open
Abstract
Introduction: Treatment of methicillin-resistant (MR) staphylococcal prosthetic joint infections (PJIs) remains a matter of discussion, with vancomycin-rifampin combination therapy being the preferred treatment for DAIR and one-stage exchange arthroplasty strategies. This study analyzes the outcomes of patients with chronic methicillin-resistant coagulase-negative staphylococcal PJIs treated with vancomycin-minocycline combination therapy. Methods: This prospective, single center cohort study included all chronic MR coagulase-negative staphylococcal PJIs (01/2004-12/2014) treated with exchange arthroplasty and at least 4 weeks of minocycline-vancomycin. The following endpoints were considered: reinfection including relapse (same microorganism) and a new infection (different microorganism) and PJI-related deaths. Their outcomes were compared with PJIs treated with rifampin-vancomycin during the same period. Results: Thirty-four patients (median age, 69 years) with 22 hip and 12 knee arthroplasty infections were included. Sixteen (47%) had previously been managed in another center. Median vancomycin MIC of strains was 3 mg/L. Nineteen underwent one-stage, 15 two-stage exchange arthroplasty. After a median [IQR] follow-up of 43 [26-68] months, 2 patients relapsed and 6 developed a new PJI. Compared to 36 rifampin-vancomycin treated PJIs, relapse- or reinfection-free survival rates didn't differ, but more new infections developed in the minocycline group (6 vs 3; P 0.3). Conclusions: Minocycline-vancomycin combination therapy for chronic MR coagulase-negative staphylococcal PJIs seems to be an interesting therapeutic alternative.
Collapse
Affiliation(s)
- Géraldine Bart
- Service de Médecine Interne et Rhumatologie, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Valérie Zeller
- Service de Médecine Interne et Rhumatologie, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France.,Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Younes Kerroumi
- Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Beate Heym
- Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France.,Laboratoire des Centres de Santé et Hôpitaux d'Ile de France, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Vanina Meyssonnier
- Service de Médecine Interne et Rhumatologie, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France.,Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Nicole Desplaces
- Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | | | - Jean Marc Ziza
- Service de Médecine Interne et Rhumatologie, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France.,Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| | - Simon Marmor
- Centre de Référence des Infections Ostéo-Articulaires Complexes, Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France.,Service de Chirurgie Osseuse et Traumatologique; Groupe Hospitalier Diaconesses-Croix Saint-Simon, Paris, France
| |
Collapse
|
12
|
Tsakris A, Koumaki V, Dokoumetzidis A. Minocycline susceptibility breakpoints for Acinetobacter baumannii: do we need to re-evaluate them? J Antimicrob Chemother 2020; 74:295-297. [PMID: 30412249 DOI: 10.1093/jac/dky448] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Minocycline is an old broad-spectrum tetracycline indicated for the treatment of various infections, including those due to minocycline-susceptible Acinetobacter spp. Susceptibility data worldwide are showing increasing rates of resistance of Acinetobacter baumannii to almost all antimicrobial classes, whereas minocycline seems to remain relatively potent against this significant pathogen. Since no new effective drugs have been released against MDR A. baumannii, minocycline is an attractive choice. Tracing back minocycline CLSI susceptibility breakpoints, it is evident that they have been based on old pharmacokinetic approaches. In an attempt to integrate the scarce new pharmacodynamic data, a Monte Carlo simulation was performed. It seems that the currently used breakpoints are, 8-fold elevated according to the approved dosage regimen, giving erroneously higher rates of minocycline susceptibility of A. baumannii. Therefore, current minocycline breakpoints merit re-evaluation in order to deliver reliable susceptibility profiles for selecting the appropriate therapy.
Collapse
Affiliation(s)
- Athanasios Tsakris
- Department of Microbiology, Medical School, University of Athens, Athens, Greece
| | - Vasiliki Koumaki
- Department of Microbiology, Medical School, University of Athens, Athens, Greece
| | | |
Collapse
|
13
|
Zuckerman ST, Rivera-Delgado E, Haley RM, Korley JN, von Recum HA. Elucidating the Structure-Function Relationship of Solvent and Cross-Linker on Affinity-Based Release from Cyclodextrin Hydrogels. Gels 2020; 6:gels6010009. [PMID: 32235748 PMCID: PMC7151216 DOI: 10.3390/gels6010009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
Minocycline (MNC) is a tetracycline antibiotic capable of associating with cyclodextrin (CD), and it is a frontline drug for many instances of implant infection. Due to its broad-spectrum activity and long half-life, MNC represents an ideal drug for localized delivery; however, classic polymer formulations, particularly hydrogels, result in biphasic release less suitable for sustained anti-microbial action. A polymer delivery system capable of sustained, steady drug delivery rates poses an attractive target to maximize the antimicrobial activity of MNC. Here, we formed insoluble hydrogels of polymerized CD (pCD) with a range of crosslinking densities, and then assessed loading, release, and antimicrobial activity of MNC. MNC loads between 5-12 wt % and releases from pCD hydrogels for >14 days. pCD loaded with MNC shows extended antimicrobial activity against S. aureus for >40 days and E. coli for >70 days. We evaluated a range of water/ethanol blends to test our hypothesis that solvent polarity will impact drug-CD association as a function of hydrogel swelling and crosslinking. Increased polymer crosslinking and decreased solvent polarity both reduced MNC loading, but solvent polarity showed a dramatic reduction independent of hydrogel swelling. Due to its high solubility and excellent delivery profile, MNC represents a unique drug to probe the structure-function relationship between drug, affinity group, and polymer crosslinking ratio.
Collapse
Affiliation(s)
- Sean T. Zuckerman
- Affinity Therapeutics, LLC, 11000 Cedar Avenue, Suite 285, Cleveland, OH 44106, USA; (S.T.Z.); (J.N.K.)
| | - Edgardo Rivera-Delgado
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue Cleveland, OH 44106, USA; (E.R.-D.); (R.M.H.)
| | - Rebecca M. Haley
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue Cleveland, OH 44106, USA; (E.R.-D.); (R.M.H.)
| | - Julius N. Korley
- Affinity Therapeutics, LLC, 11000 Cedar Avenue, Suite 285, Cleveland, OH 44106, USA; (S.T.Z.); (J.N.K.)
| | - Horst A. von Recum
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue Cleveland, OH 44106, USA; (E.R.-D.); (R.M.H.)
- Correspondence:
| |
Collapse
|
14
|
Asadi A, Abdi M, Kouhsari E, Panahi P, Sholeh M, Sadeghifard N, Amiriani T, Ahmadi A, Maleki A, Gholami M. Minocycline, focus on mechanisms of resistance, antibacterial activity, and clinical effectiveness: Back to the future. J Glob Antimicrob Resist 2020; 22:161-174. [PMID: 32061815 DOI: 10.1016/j.jgar.2020.01.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/17/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES The increasing crisis regarding multidrug-resistant (MDR) and extensively drug-resistant microorganisms leads to appealing therapeutic options. METHODS During the last 30 years, minocycline, a wide-spectrum antimicrobial agent, has been effective against MDR Gram-positive and Gram-negative bacterial infections. As with other tetracyclines, the mechanism of action of minocycline involves attaching to the bacterial 30S ribosomal subunit and preventing protein synthesis. RESULTS This antimicrobial agent has been approved for the treatment of acne vulgaris, some sexually transmitted diseases and rheumatoid arthritis. Although many reports have been published, there remains limited information regarding the prevalence, mechanism of resistance and clinical effectiveness of minocycline. CONCLUSION Thus, we summarize here the currently available data concerning pharmacokinetics and pharmacodynamics, mechanism of action and resistance, antibacterial activity and clinical effectiveness of minocycline.
Collapse
Affiliation(s)
- Arezoo Asadi
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Abdi
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Kouhsari
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran; Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Pegah Panahi
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Sholeh
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nourkhoda Sadeghifard
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Ahmadi
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abbas Maleki
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mehrdad Gholami
- Department of Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
15
|
Konreddy AK, Rani GU, Lee K, Choi Y. Recent Drug-Repurposing-Driven Advances in the Discovery of Novel Antibiotics. Curr Med Chem 2019; 26:5363-5388. [PMID: 29984648 DOI: 10.2174/0929867325666180706101404] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
Drug repurposing is a safe and successful pathway to speed up the novel drug discovery and development processes compared with de novo drug discovery approaches. Drug repurposing uses FDA-approved drugs and drugs that failed in clinical trials, which have detailed information on potential toxicity, formulation, and pharmacology. Technical advancements in the informatics, genomics, and biological sciences account for the major success of drug repurposing in identifying secondary indications of existing drugs. Drug repurposing is playing a vital role in filling the gap in the discovery of potential antibiotics. Bacterial infections emerged as an ever-increasing global public health threat by dint of multidrug resistance to existing drugs. This raises the urgent need of development of new antibiotics that can effectively fight multidrug-resistant bacterial infections (MDRBIs). The present review describes the key role of drug repurposing in the development of antibiotics during 2016-2017 and of the details of recently FDA-approved antibiotics, pipeline antibiotics, and antibacterial properties of various FDA-approved drugs of anti-cancer, anti-fungal, anti-hyperlipidemia, antiinflammatory, anti-malarial, anti-parasitic, anti-viral, genetic disorder, immune modulator, etc. Further, in view of combination therapies with the existing antibiotics, their potential for new implications for MDRBIs is discussed. The current review may provide essential data for the development of quick, safe, effective, and novel antibiotics for current needs and suggest acuity in its effective implications for inhibiting MDRBIs by repurposing existing drugs.
Collapse
Affiliation(s)
- Ananda Kumar Konreddy
- College of Life Sciences and Biotechnology, Korea University, Seoul 136- 713, South Korea
| | - Grandhe Usha Rani
- College of Pharmacy, Dongguk University-Seoul, Goyang 410-820, South Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 410-820, South Korea
| | - Yongseok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul 136- 713, South Korea
| |
Collapse
|
16
|
Carruthers NJ, Stemmer PM, Media J, Swartz K, Wang X, Aube N, Hamann MT, Valeriote F, Shaw J. The anti-MRSA compound 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) inhibits protein synthesis in Staphylococcus aureus. J Proteomics 2019; 210:103539. [PMID: 31629958 DOI: 10.1016/j.jprot.2019.103539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Methicillin-resistant S aureus (MRSA) contributes to patient mortality and extended hospital stays. 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) is a natural product antibiotic that is effective against MRSA but has no known mechanism of action (MOA). We used proteomics to identify the MOA for KCR. Methicillin sensitive S aureus and a mixture of four KCR stereoisomers were tested. A time-kill assay was used to choose a 4 h treatment using KCR at 5× its MIC for proteomic analysis. S aureus was treated in triplicate with KCR, oxacillin or vehicle and quantitative proteomic analysis was carried out using isobaric tags and mass spectrometry. 1190 proteins were identified and 552 were affected by KCR (q < 0.01). Ontology analysis identified 6 distinct translation-related categories that were affected by KCR (PIANO, 10% false-discovery rate) including structural constituent of ribosome, translation, rRNA binding, tRNA binding, tRNA processing and aminoacyl-tRNA ligase activity. Median fold changes (KCR vs Control) for small and large ribosomal components were 1.46 and 1.43 respectively. KCR inhibited the production of luciferase protein in an in vitro assay (IC50 39.6 μg/ml). Upregulation of translation-related proteins in response to KCR indicates that KCR acts to disrupt S aureus protein synthesis. This was confirmed with an in vitro transcription/translation assay. SIGNIFICANCE: Methicillin-resistant S aureus (MRSA) contributes to patient mortality and extended hospital stays. 3-O-alpha-L-(2″,3″-di-p-coumaroyl)rhamnoside (KCR) is a natural product antibiotic that is effective against MRSA but has no known mechanism of action (MOA). Using proteomic analysis we determined that KCR acts by inhibiting protein synthesis. KCR is an exciting novel antibiotic and this work represents an important step in its development towards clinical use.
Collapse
Affiliation(s)
- Nicholas J Carruthers
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201, USA; Wayne State University, Institute of Environmental Health Sciences, 2309 Scott Hall, 540 E Canfield Ave, Detroit, MI 48202, United States of America.
| | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Joe Media
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Ken Swartz
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Xiaojuan Wang
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nicholas Aube
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Frederick Valeriote
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48201, USA.
| | - Jiajiu Shaw
- Henry Ford Health System, Detroit, MI, USA; 21st Century Therapeutics, Detroit, MI 48201, USA
| |
Collapse
|
17
|
Bayliss MA, Rigdova K, Kyriakides M, Grier S, Lovering AM, Williams H, Griffith DC, MacGowan A. Development, validation and application of a novel HPLC-MS/MS method for the measurement of minocycline in human plasma and urine. J Pharm Biomed Anal 2019; 169:90-98. [DOI: 10.1016/j.jpba.2019.02.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/01/2022]
|
18
|
Pharmacodynamics of Minocycline against Acinetobacter baumannii in a Rat Pneumonia Model. Antimicrob Agents Chemother 2019; 63:AAC.01671-18. [PMID: 30397059 DOI: 10.1128/aac.01671-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/29/2018] [Indexed: 11/20/2022] Open
Abstract
Minocycline is currently approved in the United States for the treatment of infections caused by susceptible isolates of Acinetobacter spp. The objective of these studies was to determine the minocycline exposures associated with an antibacterial effect against Acinetobacter baumannii in a rat pneumonia model. Rats received minocycline doses as 30-min intravenous infusions. In the rat pneumonia model, six clinical isolates of A. baumannii with MICs ranging from 0.03 to 4 mg/liter were studied. In this model, minocycline produced a bacteriostatic effect with a free 24-h area under the concentration-time curve (AUC)/MIC ratio of 10 to 16 and produced 1 log of bacterial killing with a free 24-h AUC/MIC of 13 to 24. These exposures can be achieved with the current FDA-approved dosage regimens of intravenous minocycline.
Collapse
|
19
|
Shaw J, Swartz K, Valeriote F, Media J, Chen B, Hamann MT, Wang X. Identification of the Metabolites of a Novel Anti-MRSA Compound, Kaempferol-3- O-Alpha-L-(2″,3″-di- p-coumaroyl)rhamnoside (KCR), Extracted from American Sycamore. Nat Prod Commun 2018. [DOI: 10.1177/1934578x1801300321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fractionation of an extract from American sycamore leaves produced the small molecule, kaempferol-3- O-alpha-L-(2″,3″-di- p-coumaroyl)rhamnoside (KCR), which exists in four stereoisomeric forms ( EE, EZ, ZE, and ZZ) at the olefin in the p-coumaroyl; all four isomers exhibit potent anti-MRSA activity in vitro. As part of the preclinical development of KCR, we set out to investigate the metabolites of KCR in mouse plasma as a prelude of ADME studies and therapeutic assessment. When KCR was added to mouse plasma at 37 °C, two new HPLC peaks appeared with increasing intensity as the incubation time increased; their retention times were shorter than that of KCR indicating that KCR was metabolized to produce two compounds that were more polar. HPLC results indicated that the two metabolites mainly came from the ZE and EE isomers and that the ZZ isomer was the most stable. Based on their respective HPLC retention times and UV spectra, these two metabolites were tentatively identified as p-coumaric acid and afzelin; both of which are more polar than KCR. The molecular weights of both metabolites were then confirmed by a Waters Acquity UPLC system with a QDa mass detector. UPLC chromatograms and molecular ions of metabolites 1 and 2 match well with those of reference materials, p-coumaric acid and afzelin, thus confirming the identities of the two major metabolites of KCR. In summary, KCR was metabolized in mouse plasma and two major metabolites ( p-coumaric acid and afzelin) were identified; the metabolites were mainly converted from the ZE and EE isomers.
Collapse
Affiliation(s)
- Jiajiu Shaw
- 21st Century Therapeutics, 440 Burroughs St., Suite 447, Detroit, Michigan 48202, USA
- Henry Ford Health System, Department of Internal Medicine, 440 Burroughs St., Rm 415, Detroit, Michigan 48202, USA
| | - Kenneth Swartz
- 21st Century Therapeutics, 440 Burroughs St., Suite 447, Detroit, Michigan 48202, USA
| | - Frederick Valeriote
- Henry Ford Health System, Department of Internal Medicine, 440 Burroughs St., Rm 415, Detroit, Michigan 48202, USA
| | - Joseph Media
- Henry Ford Health System, Department of Internal Medicine, 440 Burroughs St., Rm 415, Detroit, Michigan 48202, USA
| | - Ben Chen
- 21st Century Therapeutics, 440 Burroughs St., Suite 447, Detroit, Michigan 48202, USA
| | - Mark T. Hamann
- Oxford Laboratories, 213 Timber Lane, Oxford, MS 38655, USA
| | - Xiaojuan Wang
- Medical University of South Carolina, Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA
| |
Collapse
|
20
|
Intravenous minocycline in multidrug-resistant infections: a profile of its use in the USA with a focus on Acinetobacter infections. DRUGS & THERAPY PERSPECTIVES 2017. [DOI: 10.1007/s40267-017-0453-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Echeverria KO, Lascola KM, Giguère S, Foreman JH, Austin SA. Pulmonary disposition and pharmacokinetics of minocycline in adult horses. Am J Vet Res 2017; 78:1319-1328. [DOI: 10.2460/ajvr.78.11.1319] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Echeverria KO, Lascola KM, Giguère S, Foreman JH. Effect of feeding on the pharmacokinetics of oral minocycline in healthy adult horses. J Vet Pharmacol Ther 2017; 41:e53-e56. [DOI: 10.1111/jvp.12456] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/16/2017] [Indexed: 11/26/2022]
Affiliation(s)
- K. O. Echeverria
- Department of Veterinary Clinical Medicine; College of Veterinary Medicine; University of Illinois; Urbana IL USA
| | - K. M. Lascola
- Department of Veterinary Clinical Medicine; College of Veterinary Medicine; University of Illinois; Urbana IL USA
| | - S. Giguère
- Department of Large Animal Medicine; College of Veterinary Medicine; University of Georgia; Athens GA USA
| | - J. H. Foreman
- Department of Veterinary Clinical Medicine; College of Veterinary Medicine; University of Illinois; Urbana IL USA
| |
Collapse
|
23
|
In Vitro Assessment of Combined Polymyxin B and Minocycline Therapy against Klebsiella pneumoniae Carbapenemase (KPC)-Producing K. pneumoniae. Antimicrob Agents Chemother 2017; 61:AAC.00073-17. [PMID: 28438930 DOI: 10.1128/aac.00073-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/15/2017] [Indexed: 01/03/2023] Open
Abstract
The multidrug resistance profiles of Klebsiella pneumoniae carbapenemase (KPC) producers have led to increased clinical polymyxin use. Combination therapy with polymyxins may improve treatment outcomes, but it is uncertain which combinations are most effective. Clinical successes with intravenous minocycline-based combination treatments have been reported for infections caused by carbapenemase-producing bacteria. The objective of this study was to evaluate the in vitro activity of polymyxin B and minocycline combination therapy against six KPC-2-producing K. pneumoniae isolates (minocycline MIC range, 2 to 32 mg/liter). Polymyxin B monotherapy (0.5, 1, 2, 4, and 16 mg/liter) resulted in a rapid reduction of up to 6 log in bactericidal activity followed by regrowth by 24 h. Minocycline monotherapy (1, 2, 4, 8, and 16 mg/liter) showed no reduction of activity of >1.34 log against all isolates, although concentrations of 8 and 16 mg/liter prolonged the time to regrowth. When the therapies were used in combination, rapid bactericidal activity was followed by slower regrowth, with synergy (60 of 120 combinations at 24 h, 19 of 120 combinations at 48 h) and additivity (43 of 120 combinations at 24 h, 44 of 120 combinations at 48 h) against all isolates. The extent of killing was greatest against the more susceptible polymyxin B isolates (MICs of ≤0.5 mg/liter) regardless of the minocycline MIC. The pharmacodynamic activity of combined polymyxin B-minocycline therapy against KPC-producing K. pneumoniae is dependent on polymyxin B susceptibility. Further in vitro and animal studies must be performed to fully evaluate the efficacy of this drug combination.
Collapse
|
24
|
Monk CS, Jeong SY, Gibson DJ, Plummer CE. The presence of minocycline in the tear film of normal horses following oral administration and its anticollagenase activity. Vet Ophthalmol 2017; 21:58-65. [DOI: 10.1111/vop.12479] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Caroline S. Monk
- Department of Small Animal Clinical Sciences; University of Florida College of Veterinary Medicine; Gainesville FL USA
| | - Sun Young Jeong
- Department of Obstetrics; Shands School of Medicine; University of Florida; Gainesville FL USA
| | - Daniel James Gibson
- Department of Obstetrics; Shands School of Medicine; University of Florida; Gainesville FL USA
| | - Caryn E. Plummer
- Department of Small Animal Clinical Sciences; University of Florida College of Veterinary Medicine; Gainesville FL USA
- Department of Large Animal Clinical Sciences; University of Florida College of Veterinary Medicine; Gainesville FL USA
| |
Collapse
|
25
|
Lashinsky JN, Henig O, Pogue JM, Kaye KS. Minocycline for the Treatment of Multidrug and Extensively Drug-Resistant A. baumannii: A Review. Infect Dis Ther 2017; 6:199-211. [PMID: 28357705 PMCID: PMC5446366 DOI: 10.1007/s40121-017-0153-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Indexed: 11/28/2022] Open
Abstract
Acinetobacter baumannii can cause life-threatening nosocomial infections associated with high rates of morbidity and mortality. In recent years, the increasing number of infections due to extensively drug-resistant Acinetobacter with limited treatment options has resulted in a need for additional therapeutic agents, and a renaissance of older, neglected antimicrobials. This has led to an increased interest in the use of minocycline to treat these infections. Minocycline has been shown to overcome many resistance mechanisms affecting other tetracyclines in A. baumannii, including tigecycline. Additionally, it has favorable pharmacokinetic and pharmacodynamic properties, as well as excellent in vitro activity against drug-resistant A. baumannii. Available data support therapeutic success with minocycline, while ease of dosing with no need for renal or hepatic dose adjustments and improved safety have made it an appealing therapy. This review will focus on the mechanisms of action and resistance to tetracyclines in A. baumannii, the in vitro activity, pharmacokinetic and pharmacodynamic properties of minocycline against A. baumannii, and finally the clinical experience with minocycline for the treatment of invasive infections due to this pathogen.
Collapse
Affiliation(s)
| | - Oryan Henig
- Department of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jason M Pogue
- Department of Pharmacy Services, Sinai-Grace Hospital, Detroit Medical Center, Detroit, MI, USA
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Keith S Kaye
- Department of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Matos AC, Pinto RV, Bettencourt AF. Easy-Assessment of Levofloxacin and Minocycline in Relevant Biomimetic Media by HPLC–UV Analysis. J Chromatogr Sci 2017; 55:757-765. [DOI: 10.1093/chromsci/bmx033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/04/2017] [Indexed: 11/14/2022]
|
27
|
Abstract
Intravenous minocycline (Minocin®) is approved in the USA for use in patients with infections due to susceptible strains of Gram-positive and Gram-negative pathogens, including infections due to Acinetobacter spp. Minocycline is a synthetic tetracycline derivative that was originally introduced in the 1960s. A new intravenous formulation of minocycline was recently approved and introduced to address the increasing prevalence of multidrug-resistant (MDR) pathogens. Minocycline shows antibacterial activity against A. baumannii clinical isolates worldwide, and exhibits synergistic bactericidal activity against MDR and extensively drug-resistant (XDR) A. baumannii isolates when combined with other antibacterial agents. In retrospective studies, intravenous minocycline provided high rates of clinical success or improvement and was generally well tolerated among patients with MDR or carbapenem-resistant A. baumannii infections. While randomized clinical trial data would be useful to fully establish the place of minocycline in the management of these infections for which there are currently very few available options, clinical trials in patients with infections due to Acinetobacter spp. are difficult to perform. Nevertheless, current data indicate a potential role for intravenous minocycline in the treatment of patients MDR A. baumannii infections, particularly when combined with a second antibacterial agent (e.g. colistin).
Collapse
Affiliation(s)
- Sarah L Greig
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| | - Lesley J Scott
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand
| |
Collapse
|
28
|
Vasconcellos FMD, Casas MRT, Tavares LCB, Garcia DDO, Camargo CH. In vitro activity of antimicrobial agents against multidrug- and extensively drug-resistant Acinetobacter baumannii. J Med Microbiol 2017; 66:98-102. [PMID: 28056222 DOI: 10.1099/jmm.0.000422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | | | - Laís Calissi Brisolla Tavares
- Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil.,Bacteriology Division, Adolfo Lutz Institute, São Paulo, SP, Brazil
| | | | | |
Collapse
|
29
|
Abstract
Staphylococcus aureus, although generally identified as a commensal, is also a common cause of human bacterial infections, including of the skin and other soft tissues, bones, bloodstream, and respiratory tract. The history of S. aureus treatment is marked by the development of resistance to each new class of antistaphylococcal antimicrobial drugs, including the penicillins, sulfonamides, tetracyclines, glycopeptides, and others, complicating therapy. S. aureus isolates identified in the 1960s were sometimes resistant to methicillin, a ß-lactam antimicrobial active initially against a majority S. aureus strains. These MRSA isolates, resistant to nearly all ß-lactam antimicrobials, were first largely confined to the health care environment and the patients who attended it. However, in the mid-1990s, new strains, known as community-associated (CA-) MRSA strains, emerged. CA-MRSA organisms, compared with health care-associated (HA-) MRSA strain types, are more often susceptible to multiple classes of non ß-lactam antimicrobials. While infections caused by methicillin-susceptible S. aureus (MSSA) strains are usually treated with drugs in the ß-lactam class, such as cephalosporins, oxacillin or nafcillin, MRSA infections are treated with drugs in other antimicrobial classes. The glycopeptide drug vancomycin, and in some countries teicoplanin, is the most common drug used to treat severe MRSA infections. There are now other classes of antimicrobials available to treat staphylococcal infections, including several that have been approved after 2009. The antimicrobial management of invasive and noninvasive S. aureus infections in the ambulatory and in-patient settings is the topic of this review. Also discussed are common adverse effects of antistaphylococcal antimicrobial agents, advantages of one agent over another for specific clinical syndromes, and the use of adjunctive therapies such as surgery and intravenous immunoglobulin. We have detailed considerations in the therapy of noninvasive and invasive S. aureus infections. This is followed by sections on specific clinical infectious syndromes including skin and soft tissue infections, bacteremia, endocarditis and intravascular infections, pneumonia, osteomyelitis and vertebral discitis, epidural abscess, septic arthritis, pyomyositis, mastitis, necrotizing fasciitis, orbital infections, endophthalmitis, parotitis, staphylococcal toxinoses, urogenital infections, and central nervous system infections.
Collapse
|
30
|
Reynolds LJ, Roberts AP, Anjum MF. Efflux in the Oral Metagenome: The Discovery of a Novel Tetracycline and Tigecycline ABC Transporter. Front Microbiol 2016; 7:1923. [PMID: 27999567 PMCID: PMC5138185 DOI: 10.3389/fmicb.2016.01923] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 11/16/2016] [Indexed: 01/07/2023] Open
Abstract
Antibiotic resistance in human bacterial pathogens and commensals is threatening our ability to treat infections and conduct common medical procedures. As novel antibiotics are discovered and marketed it is important that we understand how resistance to them may arise and know what environments may act as reservoirs for such resistance genes. In this study a tetracycline and tigecycline resistant clone was identified by screening a human saliva metagenomic library in Escherichia coli EPI300 on agar containing 5 μg/ml tetracycline. Sequencing of the DNA insert present within the tetracycline resistant clone revealed it to contain a 7,765 bp fragment harboring novel ABC half transporter genes, tetAB(60). Mutagenesis studies performed on these genes confirmed that they were responsible for the tetracycline and tigecycline resistance phenotypes. Growth studies performed using E. coli EPI300 clones that harbored either the wild type, the mutated, or none of these genes indicated that there was a fitness cost associated with presence of these genes, with the isolate harboring both genes exhibiting a significantly slower growth than control strains. Given the emergence of E. coli strains that are sensitive only to tigecycline and doxycycline it is concerning that such a resistance mechanism has been identified in the human oral cavity.
Collapse
Affiliation(s)
- Liam J Reynolds
- Department of Microbial Diseases, UCL Eastman Dental Institute, Faculty of Medical Sciences, University College LondonLondon, UK; Department of Bacteriology, Animal and Plant Health AgencyAddlestone, UK
| | - Adam P Roberts
- Department of Microbial Diseases, UCL Eastman Dental Institute, Faculty of Medical Sciences, University College London London, UK
| | - Muna F Anjum
- Department of Microbial Diseases, UCL Eastman Dental Institute, Faculty of Medical Sciences, University College LondonLondon, UK; Department of Bacteriology, Animal and Plant Health AgencyAddlestone, UK
| |
Collapse
|
31
|
Shankar C, Nabarro LEB, Anandan S, Veeraraghavan B. Minocycline and Tigecycline: What Is Their Role in the Treatment of Carbapenem-Resistant Gram-Negative Organisms? Microb Drug Resist 2016; 23:437-446. [PMID: 27564414 DOI: 10.1089/mdr.2016.0043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Carbapenem-resistant organisms are increasingly common worldwide, particularly in India and are associated with high mortality rates especially in patients with severe infection such as bacteremia. Existing drugs such as carbapenems and polymyxins have a number of disadvantages, but remain the mainstay of treatment. The tetracycline class of antibiotics was first produced in the 1940s. Minocycline, tetracycline derivative, although licensed for treatment of wide range of infections, has not been considered for treatment of multidrug-resistant organisms until recently and needs further in vivo studies. Tigecycline, a derivative of minocycline, although with certain disadvantages, has been frequently used in the treatment of carbapenem-resistant organisms. In this article, we review the properties of minocycline and tigecycline, the common mechanisms of resistance, and assess their role in the management of carbapenem-resistant organisms.
Collapse
Affiliation(s)
- Chaitra Shankar
- Department of Clinical Microbiology, Christian Medical College and Hospital , Vellore, India
| | - Laura E B Nabarro
- Department of Clinical Microbiology, Christian Medical College and Hospital , Vellore, India
| | - Shalini Anandan
- Department of Clinical Microbiology, Christian Medical College and Hospital , Vellore, India
| | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College and Hospital , Vellore, India
| |
Collapse
|
32
|
Dimitriadis P, Protonotariou E, Varlamis S, Poulou A, Vasilaki O, Metallidis S, Tsakris A, Malisiovas N, Skoura L, Pournaras S. Comparative evaluation of minocycline susceptibility testing methods in carbapenem-resistant Acinetobacter baumannii. Int J Antimicrob Agents 2016; 48:321-3. [PMID: 27451087 DOI: 10.1016/j.ijantimicag.2016.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/26/2016] [Accepted: 05/28/2016] [Indexed: 10/21/2022]
Abstract
In this study, the performance of two commonly used routine antimicrobial susceptibility testing methods, the automated VITEK(®)2 system and Etest (bioMérieux, Marcy-l'Étoile, France), was compared with the standard broth microdilution (BMD) method on 87 multidrug- and carbapenem-resistant Acinetobacter baumannii clinical isolates. Clinical and Laboratory Standards Institute (CLSI) 2015 breakpoints (susceptible, ≤4 mg/L; intermediate, 8 mg/L; and resistant, ≥16 mg/L) were used. Minocycline showed excellent activity, with 94.3% of isolates susceptible by BMD (VITEK(®)2, 73.6%; Etest, 63.2%). The MIC50/90 values (minimum inhibitory concentrations required to inhibit 50% and 90% of the isolates, respectively) were as follows: BMD, 1/4 mg/L; VITEK(®)2, ≤1/8 mg/L; and Etest, 4/16 mg/L. Etest produced 14.9% major/20.7% minor errors and VITEK(®)2 produced 3.4% major/17.2% minor errors. These data indicate that VITEK(®)2 may be more reliable than Etest for routine susceptibility testing of minocycline for A. baumannii isolates. As both VITEK(®)2 and Etest produced higher minocycline MICs compared with the reference method, BMD may be needed to validate the categorisation of carbapenem-resistant A. baumannii by these assays as minocycline non-susceptible.
Collapse
Affiliation(s)
- Pavlos Dimitriadis
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece
| | - Efthymia Protonotariou
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece.
| | - Sotiris Varlamis
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece
| | - Aggeliki Poulou
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece
| | - Olga Vasilaki
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece
| | - Simeon Metallidis
- First Internal Medicine Department, Infectious Diseases Unit, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, University of Athens, Athens, Greece
| | - Nikos Malisiovas
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lemonia Skoura
- Department of Microbiology, AHEPA University Hospital, Aristotle University of Thessaloniki, S. Kiriakidi 1, 546 36 Thessaloniki, Greece
| | - Spyros Pournaras
- Department of Microbiology, Medical School, University of Athens, Athens, Greece
| |
Collapse
|
33
|
Diversity of mechanisms conferring resistance to β-lactams among OXA-23–producing Acinetobacter baumannii clones. Diagn Microbiol Infect Dis 2016; 85:90-7. [DOI: 10.1016/j.diagmicrobio.2016.01.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/18/2016] [Accepted: 01/25/2016] [Indexed: 11/16/2022]
|
34
|
Abstract
OBJECTIVES Recently, the increased cost and decreased availability of doxycycline has sparked an interest in using minocycline as an alternative. The purpose of this study was to determine the pharmacokinetics of minocycline in domestic cats in order to facilitate dosage decisions. METHODS Purpose-bred, young adult cats were administered a single dose of either intravenous (IV; n = 4; 5 mg/kg) or oral (n = 6; 50 mg/cat) minocycline. Blood was collected from each at intervals up to 24 h afterwards. Minocycline was measured using high performance liquid chromatography with ultraviolet detection. A one-compartment pharmacokinetic model was fit to the oral data and a two-compartment model to the IV data via a computer program. Plasma protein binding was measured by fortifying blank plasma from untreated healthy cats with minocycline at two concentrations and applying an ultracentrifugation method. RESULTS Two cats became transiently lethargic and tachypneic during IV drug infusion. One cat vomited 6.0 h after infusion, and two cats vomited either 1.5 h or ~5.0 h after oral drug administration. The mean oral dose administered was 13.9 ± 0.47 mg/kg. Oral bioavailability was approximately 62%. Plasma protein binding was 60% at 5 µg/ml and 46% at 1 μg/ml. After IV administration, elimination half-life (t(½)), apparent volume of distribution at steady-state, and systemic clearance were 6.7 h (coefficient of variation [CV] 14.4%), 1.5 l/kg (CV 34.5%) and 2.9 ml/kg/min (CV 40.8%), respectively. After oral administration the terminal t(½) and peak concentration (Cmax) were 6.3 h (CV 9%) and 4.77 µg/ml (CV 36%), respectively. CONCLUSIONS AND RELEVANCE Because most bacteria will have a minimum inhibitory concentration of ⩽0.5 μg/ml, an oral dose of 8.8 mg/kg q24h would be adequate to meet pharmacokinetic-pharmacodynamic targets after adjusting for protein binding. Although some gastrointestinal upset may occur, one 50 mg capsule orally q24h would provide appropriate dosing for most cats.
Collapse
Affiliation(s)
- Beth E Tynan
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Mark G Papich
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Marie E Kerl
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Leah A Cohn
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
35
|
Leite GC, Oliveira MS, Perdigão-Neto LV, Rocha CKD, Guimarães T, Rizek C, Levin AS, Costa SF. Antimicrobial Combinations against Pan-Resistant Acinetobacter baumannii Isolates with Different Resistance Mechanisms. PLoS One 2016; 11:e0151270. [PMID: 26998609 PMCID: PMC4801211 DOI: 10.1371/journal.pone.0151270] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 02/25/2016] [Indexed: 01/12/2023] Open
Abstract
The study investigated the effect of antibiotic combinations against 20 clinical isolates of A. baumannii (seven colistin-resistant and 13 colistin-susceptible) with different resistance mechanisms. Clinical data, treatment, and patient mortality were evaluated. The following methods were used: MIC, PCRs, and outer membrane protein (OMP) analysis. Synergy was investigated using the checkerboard and time-kill methods. Clonality was evaluated by PFGE. Based on clonality, the whole genome sequence of six A. baumannii isolates was analyzed. All isolates were resistant to meropenem, rifampicin, and fosfomycin. OXA-23 and OXA-143 were the most frequent carbapenemases found. Four isolates showed loss of a 43kDa OMP. The colistin-susceptible isolates belonged to different clones and showed the highest synergistic effect with fosfomycin-amikacin. Among colistin-resistant isolates, the highest synergistic effect was observed with the combinations of colistin-rifampicin followed by colistin-vancomycin. All colistin-resistant isolates harbored blaOXA-23-like and belonged to CC113. Clinical and demographic data were available for 18 of 20 patients. Fourteen received treatment and eight patients died during treatment. The most frequent site of infection was the blood in 13 of 14 patients. Seven patients received vancomycin plus an active drug against A. baumannii; however, mortality did not differ in this group. The synergistic effect was similar for colistin-susceptible isolates of distinct clonal origin presenting with the same resistance mechanism. Overall mortality and death during treatment was high, and despite the high synergism in vitro with vancomycin, death did not differ comparing the use or not of vancomycin plus an active drug against A. baumannii.
Collapse
Affiliation(s)
- Gleice Cristina Leite
- Department of Infectious Diseases, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Investigation 54 (LIM-54), São Paulo, Brazil
| | - Maura Salaroli Oliveira
- Department of Infectious Diseases, University of São Paulo, São Paulo, Brazil
- Hospital Das Clínicas FMUSP, São Paulo, Brazil
| | - Lauro Vieira Perdigão-Neto
- Department of Infectious Diseases, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Investigation 54 (LIM-54), São Paulo, Brazil
- Hospital Das Clínicas FMUSP, São Paulo, Brazil
| | | | - Thais Guimarães
- Department of Infection Control, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Camila Rizek
- Laboratory of Medical Investigation 54 (LIM-54), São Paulo, Brazil
| | - Anna Sara Levin
- Department of Infectious Diseases, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Investigation 54 (LIM-54), São Paulo, Brazil
- Hospital Das Clínicas FMUSP, São Paulo, Brazil
| | - Silvia Figueiredo Costa
- Department of Infectious Diseases, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Investigation 54 (LIM-54), São Paulo, Brazil
- Hospital Das Clínicas FMUSP, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
36
|
Colton B, McConeghy KW, Schreckenberger PC, Danziger LH. I.V. minocycline revisited for infections caused by multidrug-resistant organisms. Am J Health Syst Pharm 2016; 73:279-85. [DOI: 10.2146/ajhp150290] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
37
|
Hnot ML, Cole LK, Lorch G, Rajala-Schultz PJ, Papich MG. Effect of feeding on the pharmacokinetics of oral minocycline in healthy research dogs. Vet Dermatol 2015; 26:399-405, e92-3. [DOI: 10.1111/vde.12246] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 12/01/2022]
Affiliation(s)
- Melanie L. Hnot
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Lynette K. Cole
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Gwendolen Lorch
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Paivi J. Rajala-Schultz
- Department of Veterinary Preventive Medicine; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Mark G. Papich
- Department of Molecular Biomedical Sciences; College of Veterinary Medicine; North Carolina State University; 1060 William Moore Drive Raleigh NC 27607 USA
| |
Collapse
|
38
|
Zhang Y, Valeriote F, Swartz K, Chen B, Hamann MT, Rodenburg DL, McChesney JD, Shaw J. HPLC Plasma Assay of a Novel Anti-MRSA Compound, Kaempferol-3- O-Alpha-L-(2″,3″-di- p-coumaroyl)rhamnoside, from Sycamore Leaves. Nat Prod Commun 2015. [DOI: 10.1177/1934578x1501000818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a serious pathogen that is resistant to current antibiotic therapy. Thus, there is an urgent need for novel antimicrobial agents that can effectively combat these new strains of drug-resistant “superbugs”. Recently, fractionation of an extract from Platanus occidentalis (American sycamore) leaves produced an active kaempferol molecule, 3- O-alpha-L-(2″,3″-di- p-coumaroyl)rhamnoside (KCR), in four isomeric forms; all four isomers exhibit potent anti-MRSA activity. In order to further the preclinical development of KCR as a new antibiotic class, we developed and validated a simple analytical method for assaying KCR plasma concentration. Because KCR will be developed as a new drug, although comprising four stereoisomers, the analytical method was devised to assay the total amount of all four isomers. In the present work, both a plasma processing procedure and an HPLC method have been developed and validated. Mouse plasma containing KCR was first treated with ethanol and then centrifuged. The supernatant was dried, suspended in ethanol, centrifuged, and the supernatant was injected into an HPLC system comprising a Waters C18, a mobile phase composing methanol, acetonitrile, and trifluoroacetic acid and monitored at 313 nm. The method was validated by parameters including a good linear correlation, a limit of quantification of 0.27 μg/mL, and high accuracy. In summary, this method allows a rapid analysis of KCR in the plasma samples for pharmacokinetics studies.
Collapse
Affiliation(s)
- Yiguan Zhang
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
| | | | - Kenneth Swartz
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
| | - Ben Chen
- 21st Century Therapeutics, 1366 Hilton Rd, Ferndale, MI 48220, USA
| | - Mark T. Hamann
- Oxford Pharmaceutical Development, Oxford, MS 38655, USA
| | | | | | - Jiajiu Shaw
- Henry Ford Health System, 440 Burroughs St, Detroit, MI 48202, USA
- 21st Century Therapeutics, 1366 Hilton Rd, Ferndale, MI 48220, USA
| |
Collapse
|
39
|
Hnot ML, Cole LK, Lorch G, Papich MG, Rajala-Schultz PJ, Daniels JB. Evaluation of canine-specific minocycline and doxycycline susceptibility breakpoints for meticillin-resistantStaphylococcus pseudintermediusisolates from dogs. Vet Dermatol 2015. [DOI: 10.1111/vde.12227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Melanie L. Hnot
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Lynette K. Cole
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Gwendolen Lorch
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Mark G. Papich
- Department of Molecular Biomedical Sciences; College of Veterinary Medicine; North Carolina State University; 1060 William Moore Drive Raleigh NC 27607 USA
| | - Paivi J. Rajala-Schultz
- Department of Veterinary Preventive Medicine; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| | - Joshua B. Daniels
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; The Ohio State University; 601 Vernon L. Tharp St. Columbus OH 43210 USA
| |
Collapse
|
40
|
Adibhesami H, Douraghi M, Rahbar M, Abdollahi A. Minocycline activity against clinical isolates of multidrug-resistant Acinetobacter baumannii. Clin Microbiol Infect 2015. [PMID: 26197214 DOI: 10.1016/j.cmi.2015.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- H Adibhesami
- Division of Microbiology, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - M Douraghi
- Division of Microbiology, Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Food Microbiology Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| | - M Rahbar
- Department of Microbiology, Reference Health Laboratories, Ministry of Health, Tehran, Iran
| | - A Abdollahi
- Department of Pathology, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Ritchie DJ, Garavaglia-Wilson A. A review of intravenous minocycline for treatment of multidrug-resistant Acinetobacter infections. Clin Infect Dis 2015; 59 Suppl 6:S374-80. [PMID: 25371513 DOI: 10.1093/cid/ciu613] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Options for treatment of multidrug-resistant (MDR) Acinetobacter baumannii infections are extremely limited. Minocycline intravenous is active against many MDR strains of Acinetobacter, and Clinical and Laboratory Standards Institute breakpoints exist to guide interpretation of minocycline susceptibility results with Acinetobacter. In addition, minocycline intravenous holds a US Food and Drug Administration indication for treatment of infections caused by Acinetobacter. There is an accumulating amount of literature reporting successful use of minocycline intravenous for treatment of serious MDR Acinetobacter infections, particularly for nosocomial pneumonia. These results, coupled with the generally favorable tolerability of minocycline intravenous, support its use as a viable therapeutic option for treatment of MDR Acinetobacter infections.
Collapse
Affiliation(s)
- David J Ritchie
- Department of Pharmacy, Barnes-Jewish Hospital Pharmacy Practice Department, St Louis College of Pharmacy
| | - Alexandria Garavaglia-Wilson
- Pharmacy Practice Department, St Louis College of Pharmacy Infectious Diseases Clinic, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
42
|
Castanheira M, Mendes RE, Jones RN. Update on Acinetobacter species: mechanisms of antimicrobial resistance and contemporary in vitro activity of minocycline and other treatment options. Clin Infect Dis 2015; 59 Suppl 6:S367-73. [PMID: 25371512 DOI: 10.1093/cid/ciu706] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Among Acinetobacter species, A. baumannii and other closely related species are commonly implicated in nosocomial infections. These organisms are usually multidrug resistant (MDR), and therapeutic options to treat A. baumannii infections are very limited. Clinicians have been resorting to older antimicrobial agents to treat infections caused by MDR A. baumannii, and some of these agents have documented toxicity and/or are not optimized for the infection type to be treated. Recent clinical experience supported by antimicrobial susceptibility data suggests that minocycline has greater activity than other tetracyclines and glycylcyclines against various MDR pathogens that have limited therapeutic options available, including Acinetobacter species. An intravenous formulation of minocycline has recently become available for clinical use, and in contrast to most older tetracyclines, minocycline has high activity against Acinetobacter species. In this report, we summarized some of the characteristics of the tetracycline class, and quantified the minocycline activity against contemporary (2007-2011) isolates and its potential therapeutic role against a collection of 5477 A. baumannii and other relevant gram-negative organisms when compared directly with tetracycline, doxycycline, and other broad-spectrum antimicrobial agents. Acinetobacter baumannii strains were highly resistant to all agents tested, with the exception of minocycline (79.1% susceptible) and colistin (98.8% susceptible). Minocycline (minimum inhibitory concentration that inhibits 50% and 90% of the isolates [MIC(50/90)]: 1/8 µg/mL) displayed greater activity than doxycycline (MIC(50/90): 2/>8 µg/mL) and tetracycline hydrochloride (HCL) (only 30.2% susceptible) against A. baumannii isolates, and was significantly more active than other tetracyclines against Burkholderia cepacia, Escherichia coli, Serratia marcescens, and Stenotrophomonas maltophilia isolates. In vitro susceptibility testing using tetracycline HCL as a surrogate for the susceptibility other tetracyclines fails to detect minocycline-susceptible isolates and the potential utility of minocycline for the treatment of many MDR A. baumannii infections and other difficult-to-treat species, where there are often limited choices of antimicrobials.
Collapse
|
43
|
Falagas ME, Vardakas KZ, Kapaskelis A, Triarides NA, Roussos NS. Tetracyclines for multidrug-resistant Acinetobacter baumannii infections. Int J Antimicrob Agents 2015; 45:455-60. [PMID: 25801348 DOI: 10.1016/j.ijantimicag.2014.12.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
Multidrug-resistant (MDR) Acinetobacter baumannii infections have emerged as a serious threat worldwide. As novel agents have yet to be developed, understanding the effectiveness and safety of older antibiotics has become a priority. The purpose of this systematic review was to summarise the available clinical evidence on the use of tetracyclines for the treatment of A. baumannii infections. Ten retrospective studies regarding doxycycline and minocycline for the treatment of 185 A. baumannii infections (of which 65.4% were respiratory infections and 13% were bloodstream infections) in 156 patients were available. In most cases (86.4%), tetracyclines were administered in combination with another agent. The usual dosage of doxycycline or minocycline was 100mg intravenous or per os twice daily (usually with a 200mg loading dose for minocycline). Clinical success was achieved in 120 (76.9%) of 156 patients; in 87 (71.9%) of 121 respiratory infections and in 21 (87.5%) of 24 bloodstream infections. Twenty-two deaths occurred in 100 recorded cases. Microbiological eradication was attained in 72 (71.3%) of 101 available cases and documented microbiological eradication was reached in 59 (66.3%) of 89 available cases. Adverse events were noted in only 1 of 88 cases. Overall, although tetracycline-containing regimens showed encouraging results, more data from larger comparative trials are required to establish a role for these antibiotics in the treatment of MDR A. baumannii infections.
Collapse
Affiliation(s)
- Matthew E Falagas
- Alfa Institute of Biomedical Sciences (AIBS), Athens, Greece; Department of Internal Medicine-Infectious Diseases, Iaso General Hospital, Athens, Greece; Department of Medicine, Tufts University School of Medicine, Boston, MA, USA.
| | - Konstantinos Z Vardakas
- Alfa Institute of Biomedical Sciences (AIBS), Athens, Greece; Department of Internal Medicine-Infectious Diseases, Iaso General Hospital, Athens, Greece
| | - Anastasios Kapaskelis
- Alfa Institute of Biomedical Sciences (AIBS), Athens, Greece; Department of Internal Medicine-Infectious Diseases, Iaso General Hospital, Athens, Greece
| | - Nikolaos A Triarides
- Alfa Institute of Biomedical Sciences (AIBS), Athens, Greece; Department of Internal Medicine-Infectious Diseases, Iaso General Hospital, Athens, Greece
| | | |
Collapse
|
44
|
New Thermoresistant Polymorph from CO2 Recrystallization of Minocycline Hydrochloride. Pharm Res 2014; 31:3136-49. [DOI: 10.1007/s11095-014-1406-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/28/2014] [Indexed: 11/25/2022]
|
45
|
Spiliopoulou A, Jelastopulu E, Vamvakopoulou S, Bartzavali C, Kolonitsiou F, Anastassiou ED, Christofidou M. In vitroactivity of tigecycline and colistin againstA. baumanniiclinical bloodstream isolates during an 8-year period. J Chemother 2014; 27:266-70. [DOI: 10.1179/1973947814y.0000000193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
46
|
Matos AC, Gonçalves LM, Rijo P, Vaz MA, Almeida AJ, Bettencourt AF. A novel modified acrylic bone cement matrix. A step forward on antibiotic delivery against multiresistant bacteria responsible for prosthetic joint infections. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 38:218-26. [DOI: 10.1016/j.msec.2014.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 01/20/2014] [Accepted: 02/03/2014] [Indexed: 01/30/2023]
|
47
|
Neonakis IK, Spandidos DA, Petinaki E. Is minocycline a solution for multidrug-resistant Acinetobacter baumannii? Future Microbiol 2014; 9:299-305. [DOI: 10.2217/fmb.13.167] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT: Minocycline is an old, safe, second-line antimicrobial agent that has drawn attention over the last few years as a possible therapeutic option against multidrug-resistant Acinetobacter baumannii (MDRAB) clinical isolates. Recent in vitro and in vivo results indicate that minocycline is a valid, alternative treatment option for minocycline-susceptible MDRAB. Although effective alone, its administration as monotherapy should be avoided. Combinations with other antimicrobials can reduce the MIC of each component, present synergism and minimize the risk for drug resistance. Owing to its limited solubility in urine, it should be avoided for urinary pathogens. The present article reports all available information regarding its use as a therapeutic option against MDRAB.
Collapse
Affiliation(s)
- Ioannis K Neonakis
- Department of Microbiology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Demetrios A Spandidos
- Department of Laboratory Medicine, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Efthimia Petinaki
- Department of Microbiology, Medical School, University of Thessaly, Larissa, Greece
| |
Collapse
|
48
|
Rajamuthiah R, Fuchs BB, Jayamani E, Kim Y, Larkins-Ford J, Conery A, Ausubel FM, Mylonakis E. Whole animal automated platform for drug discovery against multi-drug resistant Staphylococcus aureus. PLoS One 2014; 9:e89189. [PMID: 24586584 PMCID: PMC3929655 DOI: 10.1371/journal.pone.0089189] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/15/2014] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus, the leading cause of hospital-acquired infections in the United States, is also pathogenic to the model nematode Caenorhabditis elegans. The C. elegans-S. aureus infection model was previously carried out on solid agar plates where the bacteriovorous C. elegans feeds on a lawn of S. aureus. However, agar-based assays are not amenable to large scale screens for antibacterial compounds. We have developed a high throughput liquid screening assay that uses robotic instrumentation to dispense a precise amount of methicillin resistant S. aureus (MRSA) and worms in 384-well assay plates, followed by automated microscopy and image analysis. In validation of the liquid assay, an MRSA cell wall defective mutant, MW2ΔtarO, which is attenuated for killing in the agar-based assay, was found to be less virulent in the liquid assay. This robust assay with a Z'-factor consistently greater than 0.5 was utilized to screen the Biomol 4 compound library consisting of 640 small molecules with well characterized bioactivities. As proof of principle, 27 of the 30 clinically used antibiotics present in the library conferred increased C. elegans survival and were identified as hits in the screen. Surprisingly, the antihelminthic drug closantel was also identified as a hit in the screen. In further studies, we confirmed the anti-staphylococcal activity of closantel against vancomycin-resistant S. aureus isolates and other Gram-positive bacteria. The liquid C. elegans-S. aureus assay described here allows screening for anti-staphylococcal compounds that are not toxic to the host.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Younghoon Kim
- Department of Animal Science, Chonbuk National University, Jeonju, Republic of Korea
| | - Jonah Larkins-Ford
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annie Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Frederick M. Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
49
|
Curcio D. Resistant pathogen-associated skin and skin-structure infections: antibiotic options. Expert Rev Anti Infect Ther 2014; 8:1019-36. [DOI: 10.1586/eri.10.87] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
50
|
Use of Intravenous Minocycline for the Treatment of Methicillin-Resistant Staphylococcus aureus (MRSA) and Resistant Gram-Negative Organisms. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2014. [DOI: 10.1097/ipc.0b013e31828bbb82] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|