1
|
Bonifaz A, García-Sotelo RS, Lumbán-Ramirez F, Vázquez-González D, Inclán-Reyes JI, Sierra-Garduño ME, Araiza J, Chandler D. Update on actinomycetoma treatment: linezolid in the treatment of actinomycetomas due to Nocardia spp and Actinomadura madurae resistant to conventional treatments. Expert Rev Anti Infect Ther 2025; 23:79-89. [PMID: 39760435 DOI: 10.1080/14787210.2024.2448723] [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: 09/14/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
INTRODUCTION Mycetoma is a chronic granulomatous infection, common throughout tropical regions, and is considered a neglected disease that mostly affects impoverished populations. Mycetoma is divided into eumycetoma, caused by fungi, and actinomycetoma, caused by filamentous bacteria. Clinical presentation is distinctive, and making the diagnosis is usually not difficult; however, access to safe and effective treatments is a major challenge. There is not a single best treatment, with the choice of treatment depending on etiology, severity and extent of disease, and patient comorbidities. AREAS COVERED The following topics regarding actinomycetoma are discussed. I) Background information on actinomycetoma, and etiology. II) Differences between actinomycetoma caused by Nocardia, and cutaneous nocardiosis. III) Review of the treatment options for actinomycetoma, caused by three species - Nocardia, Actinomadura, and Streptomyces. IV) Experience using linezolid in the treatment of mycetoma caused by Nocardia spp. and Actinomadura madurae. EXPERT OPINION Multiple treatment regimens for actinomycetoma were discussed according to the causative agent. Experience in using linezolid in combined therapy for actinomycetoma caused by Nocardia in which conventional treatment options failed was also presented. The first case report of treatment with linezolid for mycetoma caused by Actinomadura madurae is presented in this article.
Collapse
Affiliation(s)
- Alexandro Bonifaz
- Dermatology & Mycology Service, Hospital General de México "Dr. Eduardo Liceaga", CDMX, Mexico
| | - Roxana S García-Sotelo
- Dermatology & Mycology Service, Hospital General de México "Dr. Eduardo Liceaga", CDMX, Mexico
| | - Fabiola Lumbán-Ramirez
- Dermatology & Mycology Service, Hospital General de México "Dr. Eduardo Liceaga", CDMX, Mexico
| | | | | | | | - Javier Araiza
- Infectology Service, Hospital General de México "Dr. Eduardo Liceaga", CDMX, Mexico
| | - David Chandler
- Dermatology Department, Brighton General Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
- Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
2
|
Nosek K, Samiec M, Ziółkowski H, Markowska-Buńka P, Czuczwar M, Borys M, Onichimowski D. Linezolid Adsorption on Filters during Continuous Renal Replacement Therapy: An In Vitro Study. Pharmaceuticals (Basel) 2024; 17:1317. [PMID: 39458958 PMCID: PMC11510572 DOI: 10.3390/ph17101317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/30/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Renal replacement therapy (RRT), widely used in the treatment of renal injury during sepsis, aims to eliminate the toxins and proinflammatory cytokines involved in the pathomechanism underlying septic shock. Dialysis filters are characterized by a high adsorption potential for cytokines in RRT in the case of septic renal injury. For the treatment of sepsis with antibiotics, it is of key importance to achieve the desired values of PK/PD indices. Continuous renal replacement therapy (CRRT) may affect antimicrobial clearance, increasing their elimination in some cases. Methods: The aim of this study was to determine the degree of adsorption for linezolid on three different types of filters used in CRRT. In our in vitro study, a continuous veno-venous hemofiltration (CVVH) was conducted using three types of filters: polysulfone (PS), polyethyleneimine-treated polyacrylonitrile (PAN PEI), and non-PEI-treated polyacrylonitrile (PAN). Each type of filter was used in three CVVH cycles, involving the use of 600 mg of linezolid dissolved in 700 mL of bovine blood or in 700 mL of 0.9% NaCl. In each case, the total volume of the obtained solution was 1000 mL. Blood samples were collected at particular time points to measure their drug concentration. The differences in mean drug/NaCl adsorption and drug/blood adsorption were determined using a one-way ANOVA with multiple comparisons via Tukey's post hoc test; a p-value of <0.05 was considered significant. Results: A significant adsorption of linezolid was found for PAN PEI filters, both in samples obtained from bovine blood and 0.9% NaCl solutions, at the endpoint. In PAN PEI samples, the concentration of linezolid in 0.9% NaCl solutions decreased from 594.74 μg/mL to 310.66 μg/mL after 120 min (the difference was established at 52%). In blood samples, the initial concentration was 495.18 μg/mL, which then decreased to 359.84 μg/mL (73% of the beginning value). No significant adsorption was demonstrated on PAN or PS filters. Conclusion: There is a need for in vivo research to confirm the effect of filter type on linezolid concentration in patients undergoing CRRT.
Collapse
Affiliation(s)
- Krzysztof Nosek
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury, Al. Warszawska 30, 10-082 Olsztyn, Poland
| | - Milena Samiec
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury, Al. Warszawska 30, 10-082 Olsztyn, Poland
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, University of Warmia and Mazury, Al. Warszawska 30, 11-082 Olsztyn, Poland
| | - Hubert Ziółkowski
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland
| | - Paulina Markowska-Buńka
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland
| | - Mirosław Czuczwar
- 2nd Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Borys
- 2nd Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Dariusz Onichimowski
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, University of Warmia and Mazury, Al. Warszawska 30, 11-082 Olsztyn, Poland
| |
Collapse
|
3
|
Otri I, Medaglia S, Martínez-Máñez R, Aznar E, Sancenón F. Exploring the Synergy between Nano-Formulated Linezolid and Polymyxin B as a Gram-Negative Effective Antibiotic Delivery System Based on Mesoporous Silica Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:228. [PMID: 38276746 PMCID: PMC10818268 DOI: 10.3390/nano14020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Antimicrobial resistance is a current silent pandemic that needs new types of antimicrobial agents different from the classic antibiotics that are known to lose efficiency over time. Encapsulation of antibiotics inside nano-delivery systems could be a promising, effective strategy that is able to delay the capability of pathogens to develop resistance mechanisms against antimicrobials. These systems can be adapted to deliver already discovered antibiotics to specific infection sites in a more successful way. Herein, mesoporous silica nanomaterials are used for an efficient delivery of a linezolid gram-positive antibiotic that acts synergistically with gram-negative antimicrobial polymyxin B. For this purpose, linezolid is encapsulated in the pores of the mesoporous silica, whose outer surface is coated with a polymyxin B membrane disruptor. The nanomaterial showed a good controlled-release performance in the presence of lipopolysaccharide, found in bacteria cell membranes, and the complete bacteria E. coli DH5α. The performed studies demonstrate that when the novel formulation is near bacteria, polymyxin B interacts with the cell membrane, thereby promoting its permeation. After this step, linezolid can easily penetrate the bacteria and act with efficacy to kill the microorganism. The nano-delivery system presents a highly increased antimicrobial efficacy against gram-negative bacteria, where the use of free linezolid is not effective, with a fractional inhibitory concentration index of 0.0063 for E. coli. Moreover, enhanced toxicity against gram-positive bacteria was confirmed thanks to the combination of both antibiotics in the same nanoparticles. Although this new nanomaterial should be further studied to reach clinical practice, the obtained results pave the way to the development of new nanoformulations which could help in the fight against bacterial infections.
Collapse
Affiliation(s)
- Ismael Otri
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (I.O.); (S.M.); (R.M.-M.)
| | - Serena Medaglia
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (I.O.); (S.M.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 46022 Valencia, Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (I.O.); (S.M.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe, Universitat Politècnica de València, 46026 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Centro de Investigación Príncipe Felipe, Universitat Politècnica de València, 46012 Valencia, Spain
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Elena Aznar
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (I.O.); (S.M.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe, Universitat Politècnica de València, 46026 Valencia, Spain
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022 Valencia, Spain; (I.O.); (S.M.); (R.M.-M.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 46022 Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Instituto de Investigación Sanitaria La Fe, Universitat Politècnica de València, 46026 Valencia, Spain
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Centro de Investigación Príncipe Felipe, Universitat Politècnica de València, 46012 Valencia, Spain
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
4
|
Miller LG, Flores EA, Launer B, Lee P, Kalkat P, Derrah K, Agrawal S, Schwartz M, Steele G, Kim T, Kuvhenguhwa MS. Safety and tolerability of tedizolid as oral treatment for bone and joint infections. Microbiol Spectr 2023; 11:e0128223. [PMID: 37750695 PMCID: PMC10581034 DOI: 10.1128/spectrum.01282-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/07/2023] [Indexed: 09/27/2023] Open
Abstract
Bone and joint infections (BJIs) are common infections increasingly managed with oral therapy. However, there are limited safe oral options for many Gram-positive pathogens. In animal studies and short-term human use, tedizolid lacks the hematologic and neurologic toxicity of the other available oxazolidinone, linezolid. However, there are limited prospective safety data. We conducted an open-label, non-comparative trial of oral tedizolid for BJI treatment. Primary outcomes were safety and cure rate. Eligible patients had a BJI caused by documented or suspected Gram-positive pathogen, required 4-12 weeks of therapy, and did not have myelosuppression or peripheral/optic neuropathy. Subjects underwent weekly evaluation for cytopenias and neuropathy. We enrolled 44 subjects; five were lost to follow-up. Two subjects did not complete planned treatment because of rash (n = 1) and urgent surgery (n = 1). Of 37 patients with evaluable outcomes, 17 (46%) had hardware-associated infection, 13 (35%) had osteomyelitis, 5 (14%) had prosthetic joint infection, and 2 (5%) had other BJIs. Median (mean, range) treatment duration was 12 (10.1, 4-12) weeks. There were no cases of cytopenias or peripheral or optic neuropathy. Treatment cure occurred in 13 (35%); 19 (51%) required antibiotic continuation after 12 weeks of tedizolid related to retained hardware at the BJI site, and failure occurred in four (11%), two unlikely, one possibly, and one probably due to tedizolid. We found that oral tedizolid was well tolerated for prolonged BJI treatment without significant toxicity. Clinical failure rate was similar to that of other published BJI investigations. (This study has been registered at Clinicaltrials.gov under identifier NCT03009045.) IMPORTANCE Bone and joint infections are common infections with limited effective and safe oral options for Gram-positive infections. The largest prospective clinical trial of tedizolid therapy for bone and joint infections enrolled 44 patients and tested each in person weekly with detailed safety monitoring including tests for leukopenia, anemia, thrombocytopenia, peripheral neuropathy, and optic neuropathy for up to 12 weeks. Findings demonstrated tedizolid was generally well tolerated and there were no incident cases of cytopenias or neuropathy. Cure rates were similar to that in other bone and joint infection studies. In summary, oral tedizolid appears to be a well-tolerated oral option for Gram-positive bone and joint infections.
Collapse
Affiliation(s)
- Loren G. Miller
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
- Lundquist David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Evelyn A. Flores
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Bryn Launer
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Pamela Lee
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Praneet Kalkat
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Kelli Derrah
- University of California Davis Medical Center, Sacramento, California, USA
| | - Shalini Agrawal
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Matthew Schwartz
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Grant Steele
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tae Kim
- Lundquist Institute for Biomedical Research at Harbor-UCLA Medical Center, Torrance, California, USA
| | | |
Collapse
|
5
|
Bi H, Deng R, Liu Y. Linezolid decreases Staphylococcus aureus biofilm formation by affecting the IcaA and IcaB proteins. Acta Microbiol Immunol Hung 2022. [PMID: 35579972 DOI: 10.1556/030.2022.01689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/21/2022] [Indexed: 02/05/2023]
Abstract
Background The ica gene of Staphylococcus aureus (S. aureus) plays a vital role in its growth and biofilm formation. Among them, IcaA and IcaB are critical proteins for synthesizing extracellular polysaccharides and biofilms in S. aureus. To investigate whether the formation of S. aureus biofilms can be inhibited through the IcaA and IcaB proteins by the presence of linezolid. Methods The icaA and icaB genes of S. aureus ATCC 25923 were silenced by homologous recombination. The critical roles of icaA and icaB in S. aureus were analysed by observing the growth curve and biofilm formation after linezolid treatment. Then, the effect of linezolid on the morphology of S. aureus was observed by scanning electron microscopy. Finally, the potential binding ability of linezolid to Ica proteins was predicted by molecular docking. Results The icaA- and icaB-silenced strains were successfully constructed, and the sensitivity of S. aureus to linezolid was decreased after icaA and icaB silencing. Scanning electron microscopy showed that linezolid caused invagination of the S. aureus surface and reduced the production of biofilms. Molecular docking results showed that linezolid could bind to IcaA and IcaB proteins. Conclusion IcaA and IcaB are potential targets of linezolid in inhibiting the biofilm formation of S. aureus (ATCC 25923).
Collapse
Affiliation(s)
- Hongxia Bi
- Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Deng
- Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yanbin Liu
- Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Inhibition of penicillin-binding protein 2a (PBP2a) in methicillin resistant Staphylococcus aureus (MRSA) by combination of oxacillin and a bioactive compound from Ramalinaroesleri. Microb Pathog 2020; 150:104676. [PMID: 33278518 DOI: 10.1016/j.micpath.2020.104676] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/29/2020] [Accepted: 11/25/2020] [Indexed: 11/22/2022]
Abstract
Lichens are known to be useful and important in ethanopharmacology since ages and still possess substantial interest in alternative medical practices around the world. The intent of this investigation was to evaluate and to understand the antibacterial potential of usnic acid which was isolated from Himalyan fruticose lichen Ramalina roesleri. Usnic acid is predicted for its pharmaceutical properties through in -silico studies. Binding efficiency of usnic acid with Penicillin binding protein-PBP2a, a protein which is responsible for conferring resistance in Staphylococcus aureus was accessed using in-silico interaction assays comparing with oxacillin and ceftaroline. Further, the validation of in-silico modelling was checked by determining the antibacterial potential of usnic acid against methicillin-resistant Staphylococcus aureus (MRSA) clinical isolates. In total, 28 clinical isolates collected from hospitals/medical students were included in the study and the anti-Staphylococcal activity was determined using agar plate dilution method followed by time-kill kinetics and synergistic studies. The scanning electron microscopic (SEM) pictures were obtained to show the cell wall disruption of MRSA by usnic acid. Docking results clearly indicated the enhanced binding potential of usnic acid (Glide XP G Score: 10.968; Glide energy -64.869) with PBP2a which is better than the energy range of reference compound, oxacillin (Glide XP G Score: 6.596; Glide energy -53.285) and roughly comparable to the co-crystallized ligand ceftaroline (Glide XP G Score: 12.20; Glide energy -70.322). Cefteroline is known to be more active against MRSA compared to oxacillin. The minimum inhibitory concentrations (MICs) of usnic acid against the clinical isolates of MRSA and reference strain (NCTC-6571) were in the range of 32-128 μg/ml. The high affinity of usnic acid to bind with PBP2a which is demonstrated via in-silico studies is further confirmed by the impressive inhibitory activity of usnic acid on MRSA clinical isolates.
Collapse
|
7
|
Choi Y, Lee SW, Kim A, Jang K, Nam H, Cho YL, Yu KS, Jang IJ, Chung JY. Safety, tolerability and pharmacokinetics of 21 day multiple oral administration of a new oxazolidinone antibiotic, LCB01-0371, in healthy male subjects. J Antimicrob Chemother 2018; 73:183-190. [PMID: 29069400 DOI: 10.1093/jac/dkx367] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/08/2017] [Indexed: 11/12/2022] Open
Abstract
Background LCB01-0371 is a new oxazolidinone antibiotic, which targets most Gram-positive organisms. High rates of adverse reactions including myelosuppression have been reported for existing oxazolidinones, limiting their long-term use. Objectives The safety, tolerability and pharmacokinetics (PK) of 21 day multiple oral administrations of LCB01-0371 in healthy male subjects (clinicaltrials.gov: NCT02540460) were investigated. Methods In this randomized, double-blind, placebo-controlled study, subjects received 800 mg of LCB01-0371 once or twice daily or 1200 mg of LCB01-0371 twice-daily for 21 days in a fasting state. Safety and tolerability profiles including laboratory tests were evaluated during the study and on a post-study visit and the results were analysed using repeated-measures analysis of variance (RM-ANOVA). Serial blood samples for PK analysis were collected up to 12 h after dosing on day 21. Results A total of 40 subjects were enrolled and 34 subjects completed the study. Two subjects dropped out according to stopping rules. In the 1200 mg twice-daily dose group, the absolute value of red blood cell count, haematocrit and haemoglobin decreased by 500 × 106/L (6.5%), 4.5% (6.8%) and 1.6 g/dL (6.9%), respectively, after 21 day administrations of LCB01-0371. However, mean relative changes from baseline of all haematology values were not significantly different among doses, including placebo (all, P < 0.05). PK profiles of LCB01-0371 in the dose range of 800 mg once daily to 1200 mg twice daily were consistent with previous studies. Conclusions LCB01-0371 is well tolerated in healthy male subjects with comparable haematology profiles to placebo, after multiple doses of up to 1200 mg twice daily for 21 days.
Collapse
Affiliation(s)
- Yewon Choi
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Sang Won Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Anhye Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Clinical Trial Center, Ajou University Medical Center, Suwon, Korea
| | - Kyungho Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | | | | | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Korea
| |
Collapse
|
8
|
|
9
|
Hedaya MA, Thomas V, Abdel-Hamid ME, Kehinde EO, Phillips OA. Comparative Pharmacokinetic Study for Linezolid and Two Novel Antibacterial Oxazolidinone Derivatives in Rabbits: Can Differences in the Pharmacokinetic Properties Explain the Discrepancies between Their In Vivo and In Vitro Antibacterial Activities? Pharmaceutics 2017; 9:pharmaceutics9030034. [PMID: 28880210 PMCID: PMC5620575 DOI: 10.3390/pharmaceutics9030034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/04/2022] Open
Abstract
This is a comparative pharmacokinetics study of linezolid (Lzd), and two novel oxazolidinone antibacterial agents—PH027 and PH051—in rabbits to determine if the discrepancy between the in vitro and in vivo activities of the novel compounds is due to pharmacokinetic factors. The pharmacokinetics after IV and oral administration, plasma protein binding and tissue distribution for the three compounds were compared. The elimination half-lives were 52.4 ± 6.3, 68.7 ± 12.1 and 175 ± 46.1 min for Lzd, PH027 and PH051, respectively. The oral bioavailability for Lzd, PH027 and PH051 administered as suspension were 38.7%, 22.1% and 4.73%, which increased significantly when administered as microemulsion to 51.7%, 72.9% and 13.9%. The plasma protein binding were 32–34%, 37–38% and 90–91% for Lzd, PH027 and PH051. The tissue distribution for PH027 and PH051 in all investigated tissues were higher than that for Lzd. It can be concluded that the lower bioavailability of PH027 and PH051 compared to Lzd when administered as suspension is the main cause of their lower in vivo activity, despite their comparable in vitro activity. Differences in the other pharmacokinetic characteristics cannot explain the lower in vivo activity. The in vivo activity of the novel compounds should be re-evaluated using formulations with good oral bioavailability.
Collapse
Affiliation(s)
- Mohsen A Hedaya
- Department of Pharmaceutics, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| | - Vidhya Thomas
- Department of Pharmaceutics, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| | - Mohamed E Abdel-Hamid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| | | | - Oludotun A Phillips
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| |
Collapse
|
10
|
Padayatchi N, Mahomed S, Loveday M, Naidoo K. Antibiotic stewardship for drug resistant tuberculosis. Expert Opin Pharmacother 2016; 17:1981-3. [DOI: 10.1080/14656566.2016.1225724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
11
|
Zhanel GG, Love R, Adam H, Golden A, Zelenitsky S, Schweizer F, Gorityala B, Lagacé-Wiens PRS, Rubinstein E, Walkty A, Gin AS, Gilmour M, Hoban DJ, Lynch JP, Karlowsky JA. Tedizolid: a novel oxazolidinone with potent activity against multidrug-resistant gram-positive pathogens. Drugs 2015; 75:253-70. [PMID: 25673021 DOI: 10.1007/s40265-015-0352-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tedizolid phosphate is a novel oxazolidinone prodrug (converted to the active form tedizolid by phosphatases in vivo) that has been developed and recently approved (June 2014) by the United States FDA for the treatment of acute bacterial skin and skin structure infections (ABSSSIs) caused by susceptible Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). Tedizolid is an oxazolidinone, but differs from other oxazolidinones by possessing a modified side chain at the C-5 position of the oxazolidinone nucleus which confers activity against certain linezolid-resistant pathogens and has an optimized C- and D-ring system that improves potency through additional binding site interactions. The mechanism of action of tedizolid is similar to other oxazolidinones and occurs through inhibition of bacterial protein synthesis by binding to 23S ribosomal RNA (rRNA) of the 50S subunit of the ribosome. As with other oxazolidinones, the spontaneous frequency of resistance development to tedizolid is low. Tedizolid is four- to eightfold more potent in vivo than linezolid against all species of staphylococci, enterococci, and streptococci, including drug-resistant phenotypes such as MRSA and vancomycin-resistant enterococci (VRE) and linezolid-resistant phenotypes. Importantly, tedizolid demonstrates activity against linezolid-resistant bacterial strains harboring the horizontally transmissible cfr gene, in the absence of certain ribosomal mutations conferring reduced oxazolidinone susceptibility. With its half-life of approximately 12 h, tedizolid is dosed once daily. It demonstrates linear pharmacokinetics, has a high oral bioavailability of approximately 90 %, and is primarily excreted by the liver as an inactive, non-circulating sulphate conjugate. Tedizolid does not require dosage adjustment in patients with any degree of renal dysfunction or hepatic dysfunction. Studies in animals have demonstrated that the pharmacodynamic parameter most closely associated with the efficacy of tedizolid is fAUC(0-24h)/MIC. In non-neutropenic animals, a dose-response enhancement was observed with tedizolid and lower exposures were required compared to neutropenic cohorts. Two Phase III clinical trials have demonstrated non-inferiority of a once-daily tedizolid 200 mg dose for 6-10 days versus twice-daily 600 mg linezolid for the treatment of ABSSSIs. Both trials used the primary endpoint of early clinical response at 48-72 h; however, one trial compared oral formulations while the other initiated therapy with the parenteral formulation and allowed oral sequential therapy following initial clinical response. Throughout its development, tedizolid has demonstrated that it is well tolerated and animal studies have shown a lower propensity for neuropathies with long-term use than its predecessor linezolid. Data from the two completed Phase III clinical trials demonstrated that the studied tedizolid regimen (200 mg once daily for 6 days) had significantly less impact on hematologic parameters as well as significantly less gastrointestinal treatment-emergent adverse effects (TEAEs) than its comparator linezolid. As with linezolid, tedizolid is a weak, reversible MAO inhibitor; however, a murine head twitch model validated to assess serotonergic activity reported no increase in the number of head twitches with tedizolid even at doses that exceeded the C max in humans by up to 25-fold. Tyramine and pseudoephedrine challenge studies in humans have also reported no meaningful MAO-related interactions with tedizolid. With its enhanced in vitro activity against a broad-spectrum of Gram-positive aerobic bacteria, convenient once-daily dosing, a short 6-day course of therapy, availability of both oral and intravenous routes of administration, and an adverse effect profile that appears to be more favorable than linezolid, tedizolid is an attractive agent for use in both the hospital and community settings. Tedizolid is currently undergoing additional Phase III clinical trials for the treatment of hospital-acquired bacterial pneumonia (HABP) and ventilated nosocomial pneumonia (VNP).
Collapse
Affiliation(s)
- George G Zhanel
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Lee CR, Lee JH, Park KS, Jeong BC, Lee SH. Quantitative proteomic view associated with resistance to clinically important antibiotics in Gram-positive bacteria: a systematic review. Front Microbiol 2015; 6:828. [PMID: 26322035 PMCID: PMC4531251 DOI: 10.3389/fmicb.2015.00828] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/27/2015] [Indexed: 11/13/2022] Open
Abstract
The increase of methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) poses a worldwide and serious health threat. Although new antibiotics, such as daptomycin and linezolid, have been developed for the treatment of infections of Gram-positive pathogens, the emergence of daptomycin-resistant and linezolid-resistant strains during therapy has now increased clinical treatment failures. In the past few years, studies using quantitative proteomic methods have provided a considerable progress in understanding antibiotic resistance mechanisms. In this review, to understand the resistance mechanisms to four clinically important antibiotics (methicillin, vancomycin, linezolid, and daptomycin) used in the treatment of Gram-positive pathogens, we summarize recent advances in studies on resistance mechanisms using quantitative proteomic methods, and also examine proteins playing an important role in the bacterial mechanisms of resistance to the four antibiotics. Proteomic researches can identify proteins whose expression levels are changed in the resistance mechanism to only one antibiotic, such as LiaH in daptomycin resistance and PrsA in vancomycin resistance, and many proteins simultaneously involved in resistance mechanisms to various antibiotics. Most of resistance-related proteins, which are simultaneously associated with resistance mechanisms to several antibiotics, play important roles in regulating bacterial envelope biogenesis, or compensating for the fitness cost of antibiotic resistance. Therefore, proteomic data confirm that antibiotic resistance requires the fitness cost and the bacterial envelope is an important factor in antibiotic resistance.
Collapse
Affiliation(s)
- Chang-Ro Lee
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University Yongin, South Korea
| | - Jung Hun Lee
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University Yongin, South Korea
| | - Kwang Seung Park
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University Yongin, South Korea
| | - Byeong Chul Jeong
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University Yongin, South Korea
| | - Sang Hee Lee
- National Leading Research Laboratory of Drug Resistance Proteomics, Department of Biological Sciences, Myongji University Yongin, South Korea
| |
Collapse
|
13
|
Rio-Marques L, Hartke A, Bizzini A. The effect of inoculum size on selection of in vitro resistance to vancomycin, daptomycin, and linezolid in methicillin-resistant Staphylococcus aureus. Microb Drug Resist 2015; 20:539-43. [PMID: 25010140 DOI: 10.1089/mdr.2014.0059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The inoculum effect (IE) is an increase in the minimum inhibitory concentration (MIC) at high bacterial densities. The effect of three inoculum sizes on the selection of resistance to vancomycin, daptomycin, and linezolid was investigated in methicillin-resistant Staphylococcus aureus (MRSA). METHODS Low (10(4) CFU/ml), medium (10(6) CFU/ml), and high (10(8) CFU/ml) inocula of MRSA were exposed to twofold increasing concentrations of either drug during 15 days of cycling. MICs for low (MICL), medium (MICM), and high (MICH) inocula were determined daily. Conventional MICs were measured at days 1, 5, 10, and 15. Experiments were performed in triplicate. RESULTS At the beginning of the experiment a small IE was observed for vancomycin (MICL=1 μg/ml, MICM=1-2 μg/ml, and MICH=2 μg/ml) and a significant IE for daptomycin (MICL=0.25 μg/ml, MICM=0.25-0.5 μg/ml, and MICH=2 μg/ml). Linezolid exhibited no IE at low and medium inocula (MICL=1 μg/ml and MICM=1-2 μg/ml), but with the high inoculum, concentrations up to 2,048 μg/ml did not fully inhibit visual growth. During cycling, increase of MIC was observed for all antibiotics. At day 15, MICL, MICM, and MICH of vancomycin were 2-4, 4-8, and 4-16 μg/ml and of daptomycin were 0.5-2, 8-128, and 64-256 μg/ml, respectively. MICL and MICM of linezolid were 1 and 2-4 μg/ml, respectively. Conventional MICs showed vancomycin and daptomycin selection of resistance since day 5 depending on the inocula. No selection of linezolid resistance was observed. CONCLUSIONS Our results showed the importance of the inoculum size in the development of resistance. Measures aimed at lowering the inoculum at the site of infection should be used whenever possible in parallel to antimicrobial therapy.
Collapse
Affiliation(s)
- Laura Rio-Marques
- 1 Service of Infectious Diseases, University of Lausanne, University Hospital Center (CHUV) , Lausanne, Switzerland
| | | | | |
Collapse
|
14
|
El-Assal MI, Helmy SA. Single-dose linezolid pharmacokinetics in critically ill patients with impaired renal function especially chronic hemodialysis patients. Biopharm Drug Dispos 2014; 35:405-16. [DOI: 10.1002/bdd.1910] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/28/2014] [Accepted: 07/07/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Mona I. El-Assal
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries; Future University; Egypt
| | - Sally A. Helmy
- Department of Pharmaceutics, Faculty of Pharmacy; Damanhour University; Damanhour Egypt
- Department of Clinical and Hospital Pharmacy, Faculty of Pharmacy; Taibah University; AL-Madinah AL-Munawarah Kingdom of Saudi Arabia
| |
Collapse
|
15
|
Diep BA, Equils O, Huang DB, Gladue R. Linezolid effects on bacterial toxin production and host immune response: review of the evidence. Curr Ther Res Clin Exp 2014; 73:86-102. [PMID: 24648596 DOI: 10.1016/j.curtheres.2012.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2012] [Indexed: 10/28/2022] Open
Abstract
BACKGROUND Linezolid is active against a broad range of gram-positive pathogens and has the potential to also affect production of bacterial toxins and host immune function. OBJECTIVE To assess the evidence for direct effects of linezolid on bacterial toxin synthesis and modulation of host immune responses. METHODS Literature searches were performed of the PubMed and OVID databases. Reviews and non-English language articles were excluded. Articles with information on the effect of linezolid on bacterial toxin synthesis and immune responses were selected for further review, and data were summarized. RESULTS Substantial in vitro evidence supports effects of linezolid on bacterial toxin production; however, the strength of the evidence and the nature of the effects are mixed. In the case of Staphylococcus aureus, repeated observations support the inhibition of production of certain staphylococcal toxins (Panton-Valentine leukocidin, protein A, and α- and β-hemolysin) by linezolid, whereas only solitary reports indicate inhibition (toxic shock syndrome toxin-1, coagulase, autolysins, and enterotoxins A and B) or stimulation (phenol-soluble modulins) of toxin production by linezolid. In the case of Streptococcus pyogenes, there are solitary reports of linezolid inhibition (protein M, deoxyribonuclease, and streptococcal pyrogenic exotoxins A, B, and F) or stimulation (immunogenic secreted protein 2 and streptococcal inhibitor of complement-mediated lysis) of toxin production, whereas published evidence for effects on streptolysin O production is conflicting. In vitro data are limited, but suggest that linezolid might also have indirect effects on host cytokine expression through inhibition of bacterial production of toxins. In vivo data from preclinical animal studies and a single clinical study in humans are limited and equivocal insofar as a potential role for linezolid in modulating the host inflammatory response; this is due in part to the difficulty in isolating antimicrobial effects and toxin synthesis inhibitory effects of linezolid from any secondary effects on host inflammatory response. CONCLUSIONS Available evidence supports the possibility that linezolid can inhibit, and in some cases stimulate, toxin production in clinically relevant pathogens. However, more research will be needed to determine the potential clinical relevance of those findings for linezolid.
Collapse
Affiliation(s)
- Binh An Diep
- Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Ozlem Equils
- Medical Division, Pfizer Inc, Collegeville, Pennsylvania
| | - David B Huang
- Medical Division, Pfizer Inc, Collegeville, Pennsylvania
| | - Ron Gladue
- Pfizer Global Biotherapeutic Technologies, Cambridge, Massachusetts
| |
Collapse
|
16
|
Kaur S, Harjai K, Chhibber S. Bacteriophage mediated killing of Staphylococcus aureus in vitro on orthopaedic K wires in presence of linezolid prevents implant colonization. PLoS One 2014; 9:e90411. [PMID: 24594764 PMCID: PMC3940871 DOI: 10.1371/journal.pone.0090411] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 01/27/2014] [Indexed: 01/21/2023] Open
Abstract
Background Infections of bone and joint tissues following arthroplasty surgeries remain a major challenge in orthopaedic settings. Methicillin resistant Staphylococcus aureus (MRSA) is recognised as an established pathogen in such infections. Combination therapy using linezolid and bacteriophage impregnated in biopolymer was investigated in the present study as an alternative strategy to prevent MRSA colonisation on the orthopaedic implant surface. Methodology Coating of stainless steel orthopaedic grade K-wires was achieved using hydroxypropylmethlycellulose (HPMC) mixed with phage alone, linezolid alone and phage and linezolid together. The potential of these agents to inhibit adhesion of S.aureus (MRSA) 43300 on K-wires was assessed. Coated and naked wires were analysed by scanning electron microscopy (SEM) and fluorescent staining. Result Significant reduction in bacterial adhesion was achieved on phage/linezolid wires in comparison to naked as well as HPMC coated wires. However, maximum reduction in bacterial adherence (∼4 log cycles) was observed on the wires coated with phage-linezolid combination. The frequency of emergence of resistant mutants was also negligible in presence of both the agents. Conclusion This study provides evidence to confirm that local delivery system employing linezolid (a potent protein synthesis inhibitor) along with a broad spectrum lytic bacteriophage (capable of self-multiplication) is able to attack the adhered as well as surrounding bacteria present near the implant site. Unlike other antibiotic based therapies, this combination has the potential to significantly restrict the emergence of resistant mutants, thus paving the way for effective treatment of MRSA associated infection of medical implants.
Collapse
Affiliation(s)
- Sandeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
- * E-mail:
| |
Collapse
|
17
|
Zaffiri L, Gardner J, Toledo-Pereyra LH. History of antibiotics: from fluoroquinolones to daptomycin (Part 2). J INVEST SURG 2014; 26:167-79. [PMID: 23869821 DOI: 10.3109/08941939.2013.808461] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the Modern Era, physicians attested to the reciprocal influence among a technologically advanced society, rapid scientific progresses in medicine, and the need for new antimicrobials. The results of these changes were not only seen in the prolongation of life expectancy but also by the emergence of new pathogens. We first observed the advent of Gram-negative bacteria as a major source of nosocomial infections. The treatment of these microorganisms was complicated by the appearance and spread of drug resistance. We first focused on the development of two major classes of antimicrobials still currently used for the treatment of Gram-negative bacteria, such as fluoroquinolones and carbapenemes. Subsequently, we directed our attention to the growth of the incidence of infections due to Methicillin-Resistant Staphylococcus aureus (MRSA). Although the first MRSA was already isolated in 1961, the treatment of this new pathogen has been based on the efficacy of vancomycin for more than four decades. Only in the last 15 yr, we assisted in the development of new antimicrobial agents such as linezolid and daptomycin.
Collapse
Affiliation(s)
- Lorenzo Zaffiri
- Western Michigan University School of Medicine, Kalamazoo, Michigan, USA
| | | | | |
Collapse
|
18
|
Mandal K, Roy A, Sen S, Bag T, Kumar N, Moitra S. Disseminated staphylococcal disease in healthy children-experience from two tertiary care hospitals of West Bengal. Indian J Pediatr 2014; 81:133-7. [PMID: 23658063 DOI: 10.1007/s12098-013-1034-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 04/03/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To assess the etiology, precipitating factors, treatment and outcome of disseminated staphylococcal disease (DSD) in healthy immunocompetent children. METHODS This hospital based observational prospective study was conducted in two tertiary care hospitals of West Bengal, India during the period of March, 2011 through February, 2012. Inclusion criteria were 1) children between 1 and 12 y and 2) clinical features DSD characterized by i) involvement of at least two distant organs with presence of gram positive cocci in clusters and/or growth of Staphylococcus aureus from at least one normally sterile body fluid, ii) fever, persistent bacteremia despite antibiotics and focal involvement of two or more separate tissue sites (skin, bone, joint, kidney, lung, liver, heart). RESULTS Thirty six cases fulfilled the criteria of DSD with mean age in years 6.03 ± 3.04 (range 1-12). The age group of 5-12 y was found to be more vulnerable (p < 0.001). Septic arthritis following accidental blunt closed trauma, was the significant precipitating factor for DSD in this age group (p = 0.031). Methicillin resistant Staphylococcus aureus (MRSA) was the causative agent in all the cases. Vancomycin resistant Staphylococcus aureus (VRSA) was detected in 88.9 % of cases. All cases were sensitive to linezolid. No significant side effects were observed with 28 d of linezolid therapy. CONCLUSIONS DSD is more common in 5-12 y age group. Trauma is a significant precipitating factor for DSD in this age group. Linezolid may be considered as the first line drug in DSD with MRSA.
Collapse
Affiliation(s)
- Kalyanbrata Mandal
- Department of Pediatrics, North Bengal Medical College and Hospital, Sushrutanagar, Darjeeling, West Bengal, India
| | | | | | | | | | | |
Collapse
|
19
|
Peppard WJ, Peppard SR, Somberg L. Optimizing drug therapy in the surgical intensive care unit. Surg Clin North Am 2013; 92:1573-620. [PMID: 23153885 DOI: 10.1016/j.suc.2012.08.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This article provides a review of commonly prescribed medications in the surgical ICU, focusing on sedatives, antipsychotics, neuromuscular blocking agents, cardiovascular agents, anticoagulants, and antibiotics. A brief overview of pharmacology is followed by practical considerations to aid prescribers in selecting the best therapy within a given category of drugs to optimize patient outcomes.
Collapse
Affiliation(s)
- William J Peppard
- Department of Pharmacy, Froedtert Hospital, Milwaukee, WI 53226, USA
| | | | | |
Collapse
|
20
|
Traffic jam at the bacterial sec translocase: targeting the SecA nanomotor by small-molecule inhibitors. ACTA ACUST UNITED AC 2011; 18:685-98. [PMID: 21700205 DOI: 10.1016/j.chembiol.2011.04.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 04/27/2011] [Accepted: 04/28/2011] [Indexed: 11/20/2022]
Abstract
The rapid rise of drug-resistant bacteria is one of the most serious unmet medical needs facing the world. Despite this increasing problem of antibiotic resistance, the number of different antibiotics available for the treatment of serious infections is dwindling. Therefore, there is an urgent need for new antibacterial drugs, preferably with novel modes of action to potentially avoid cross-resistance with existing antibacterial agents. In recent years, increasing attention has been paid to bacterial protein secretion as a potential antibacterial target. Among the different protein secretion pathways that are present in bacterial pathogens, the general protein secretory (Sec) pathway is widely considered as an attractive target for antibacterial therapy. One of the key components of the Sec pathway is the peripheral membrane ATPase SecA, which provides the energy for the translocation of preproteins across the bacterial cytoplasmic membrane. In this review, we will provide an overview of research efforts on the discovery and development of small-molecule SecA inhibitors. Furthermore, recent advances on the structure and function of SecA and their potential impact on antibacterial drug discovery will be discussed.
Collapse
|
21
|
Amini S, Tavazoie S. Antibiotics and the post-genome revolution. Curr Opin Microbiol 2011; 14:513-8. [PMID: 21816663 DOI: 10.1016/j.mib.2011.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 12/28/2022]
Abstract
The emergence of pathogenic bacteria resistant to multiple antimicrobial agents is turning into a major crisis in human and veterinary medicine. This necessitates a serious re-evaluation of our approaches toward antibacterial drug discovery and use. Concurrent advances in genomics including whole-genome sequencing, genotyping, and gene expression profiling have the potential to transform our basic understanding of antimicrobial pathways and lead to the discovery of novel targets and therapeutics.
Collapse
Affiliation(s)
- Sasan Amini
- Department of Molecular Biology & Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | | |
Collapse
|
22
|
Hopkins AL, Bickerton GR, Carruthers IM, Boyer SK, Rubin H, Overington JP. Rapid analysis of pharmacology for infectious diseases. Curr Top Med Chem 2011; 11:1292-300. [PMID: 21401504 PMCID: PMC3182413 DOI: 10.2174/156802611795429130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 08/03/2010] [Accepted: 11/15/2010] [Indexed: 11/26/2022]
Abstract
Pandemic, epidemic and endemic infectious diseases are united by a common problem: how do we rapidly and cost-effectively identify potential pharmacological interventions to treat infections? Given the large number of emerging and neglected infectious diseases and the fact that they disproportionately afflict the poorest members of the global society, new ways of thinking are required to developed high productivity discovery systems that can be applied to a larger number of pathogens. The growing availability of parasite genome data provides the basis for developing methods to prioritize, a priori, the potential drug target and pharmacological landscape of an infectious disease. Thus the overall objective of infectious disease informatics is to enable the rapid generation of plausible, novel medical hypotheses of testable pharmacological experiments, by uncovering undiscovered relationships in the wealth of biomedical literature and databases that were collected for other purposes. In particular our goal is to identify potential drug targets present in a pathogen genome and prioritize which pharmacological experiments are most likely to discover drug-like lead compounds rapidly against a pathogen (i.e. which specific compounds and drug targets should be screened, in which assays and where they can be sourced). An integral part of the challenge is the development and integration of methods to predict druggability, essentiality, synthetic lethality and polypharmacology in pathogen genomes, while simultaneously integrating the inevitable issues of chemical tractability and the potential for acquired drug resistance from the start.
Collapse
Affiliation(s)
- Andrew L Hopkins
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, UK.
| | | | | | | | | | | |
Collapse
|
23
|
Davis LT, Kumar N, Nijm LM, Ulanski LJ, Tu EY, Fiscella RG, Peterson RJ, Glickman RD. An adaptable HPLC method for the analysis of frequently used antibiotics in ocular samples. J Chromatogr B Analyt Technol Biomed Life Sci 2010; 878:2421-6. [DOI: 10.1016/j.jchromb.2010.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 07/29/2010] [Accepted: 08/02/2010] [Indexed: 10/19/2022]
|