1
|
Mitsutake K, Shinya N, Seki M, Ohara T, Uemura K, Fukunaga M, Sakai J, Nagao M, Sata M, Hamada Y, Kawasuji H, Yamamoto Y, Nakamatsu M, Koizumi Y, Mikamo H, Ukimura A, Aoyagi T, Sawai T, Tanaka T, Izumikawa K, Takayama Y, Nakamura K, Kanemitsu K, Tokimatsu I, Nakajima K, Akine D. Clinical characteristics and analysis of prognostic factors in methicillin-resistant Staphylococcus aureus endocarditis: A retrospective multicenter study in Japan. J Infect Chemother 2024; 30:1259-1265. [PMID: 38876203 DOI: 10.1016/j.jiac.2024.06.002] [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: 12/14/2023] [Revised: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Infective endocarditis (IE) caused by MRSA (methicillin-resistant Staphylococcus aureus) is associated with a high mortality rate. This study aimed to elucidate the characteristics of patients with MRSA-IE in Japan and identify the factors associated with prognosis. METHODS This retrospective study included patients with a confirmed diagnosis of IE caused by MRSA, between January 2015 and April 2019. RESULTS A total of 65 patients from 19 centers were included, with a mean age of 67 years and 26 % were female. Fifty percent of the patients with IE were had nosocomial infections and 25 % had prosthetic valve involvement. The most common comorbidities were hemodialysis (20 %) and diabetes (20 %). Congestive heart failure was present in 86 % of patients (NYHA class I, II: 48 %; III, IV: 38 %). The 30-day and in-hospital mortality rates were 29 % and 46 %, respectively. Multi-organ failure was the primary cause of death, accounting for 43 % of all causes of death. Prognostic factors for in-hospital mortality were age, disseminated intravascular coagulation, daptomycin and/or linezolid as initial antibiotic therapy, and surgery. Surgical treatment was associated with a lower mortality rate (odds ratio [OR], 0.026; 95 % confidence interval [CI], 0.002-0.382; p = 0.008 for 30-day mortality and OR, 0.130; 95 % CI; 0.029-0.584; p = 0.008 for in-hospital mortality). CONCLUSION Mortality due to MRSA-IE remains high. Surgical treatment is a significant prognostic predictor of MRSA-IE.
Collapse
Affiliation(s)
- Kotaro Mitsutake
- Department of Infectious Diseases and Infection Control, Saitama International Medical Center, Saitama Medical University, 397-1, Hidaka, Saitama, 350-1298, Japan.
| | - Natsuki Shinya
- Department of Infectious Diseases and Infection Control, Saitama International Medical Center, Saitama Medical University, 397-1, Hidaka, Saitama, 350-1298, Japan
| | - Masafumi Seki
- Department of Infectious Diseases and Infection Control, Saitama International Medical Center, Saitama Medical University, 397-1, Hidaka, Saitama, 350-1298, Japan; Department of Infectious Diseases, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai, Miyagi, 983-8536, Japan
| | - Takahiro Ohara
- Division of Geriatric and Community Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai, Miyagi, 983-8536, Japan
| | - Kohei Uemura
- Department of Biostatistics and Bioinformatics, Interfaculty Initiative in Information Studies, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Masato Fukunaga
- Department of Cardiology, Kokura Memorial Hospital, 3-2-1 Asano, Kokurakita, Kitakyushu, Fukuoka, 802-8555, Japan
| | - Jun Sakai
- Department of Infectious Disease and Infection Control, Saitama Medical University Hospital, 1981 Kamoda, Kawagoe, Saitama, 350-8550, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto City, Kyoto, 606-8507, Japan
| | - Makoto Sata
- National Cerebral and Cardiovascular Center, Division of Pulmonology and Infection Control, 6-1, Kishibe Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Yohei Hamada
- Department of Infectious Disease and Hospital Epidemiology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-0937, Japan
| | - Hitoshi Kawasuji
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Masashi Nakamatsu
- Department of Infection Control, University of the Ryukyus Hospital, 207 Aza-Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Yusuke Koizumi
- Department of Clinical Infectious Diseases, Aichi Medical University, 1-1 Iwasaku, Ganmata, Nagakute, Aichi, 480-1195, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, 1-1 Iwasaku, Ganmata, Nagakute, Aichi, 480-1195, Japan
| | - Akira Ukimura
- Infection Control Center, Osaka Medical and Pharmaceutical University Hospital, 2-7 Daigaku-cho, Takatsuki, Osaka, 569-0801, Japan
| | - Tetsuji Aoyagi
- Department of Clinical Microbiology and Infection, Tohoku University Graduate School of Medicine, Department of Comprehensive Infectious Diseases, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Toyomitsu Sawai
- Nagasaki Harbor Medical Center, Department of Respiratory Medicine, 6-39 Shinchi-cho, Nagasaki City, Nagasaki, 850-0842, Japan
| | - Takeshi Tanaka
- Infection Control and Education Center, Nagasaki University Hospital, 1 Chome-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Koichi Izumikawa
- Infection Control and Education Center, Nagasaki University Hospital, 1 Chome-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoko Takayama
- Department of Infection Control and Infectious Diseases, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, 1-15-1, Kitazato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Kiwamu Nakamura
- Department of Infection Control, Fukushima Medical University, 1 Hikarigaoka, Fukushima-shi, Fukushima, 960-1295, Japan
| | - Keiji Kanemitsu
- Department of Infection Control, Fukushima Medical University, 1 Hikarigaoka, Fukushima-shi, Fukushima, 960-1295, Japan
| | - Issei Tokimatsu
- Department of Medicine, Division of Clinical Infectious Diseases, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan
| | - Kazuhiko Nakajima
- Department of Infection Prevention and Control, Hyogo Medical University, 1-1, Mukogawa, Nishinomiya, Hyogo, 663-850, Japan
| | - Dai Akine
- Division of Clinical Infectious Diseases, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
2
|
Jacobs MMJ, Holla M, van Wageningen B, Hermans E, Veerman K. Mismatch Rate of Empirical Antimicrobial Treatment in Fracture-Related Infections. J Orthop Trauma 2024; 38:240-246. [PMID: 38377474 DOI: 10.1097/bot.0000000000002782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
OBJECTIVES To evaluate the current standard of care regarding empirical antimicrobial therapy in fracture-related infections (FRIs). METHODS DESIGN Retrospective cohort study. SETTING Level I Trauma Center. PATIENT SELECTION CRITERIA Adult patients treated for FRI with surgical debridement and empirical antibiotics between September 1, 2014, and August 31, 2022. Patients were excluded if less than 5 tissue samples for culture were taken, culture results were negative, or there was an antibiotic-free window of less than 3 days before debridement. OUTCOME MEASURES AND COMPARISONS FRI microbial etiology, antimicrobial resistance patterns (standardized antimicrobial panels were tested for each pathogen), the mismatch rate between empirical antimicrobial therapy and antibiotic resistance of causative microorganism(s), and mismatching risk factors. RESULTS In total, 75 patients were included [79% (59/75) men, mean age 51 years]. The most prevalent microorganisms were Staphylococcus aureus (52%, 39/75) and Staphylococcus epidermidis (41%, 31/75). The most frequently used empirical antibiotic was clindamycin (59%, 44/75), followed by combinations of gram-positive and gram-negative covering antibiotics (15%, 11/75). The overall mismatch rate was 51% (38/75) [95% confidence interval (CI), 0.39-0.62] and did not differ between extremities [upper: 31% (4/13) (95% CI, 0.09-0.61), lower: 55% (33/60) (95% CI, 0.42-0.68, P = 0.11)]. Mismatching empirical therapy occurred mostly in infections caused by S. epidermidis and gram-negative bacteria. Combination therapy of vancomycin with ceftazidime produced the lowest theoretical mismatch rate (8%, 6/71). Polymicrobial infections were an independent risk factor for mismatching (OR: 8.38, 95% CI, 2.53-27.75, P < 0.001). CONCLUSIONS In patients with FRI, a mismatching of empirical antibiotic therapy occurred in half of patients, mainly due to lack of coverage for S. epidermidis , gram-negative bacteria, and polymicrobial infections. Empirical therapy with vancomycin and ceftazidime produced the lowest theoretical mismatch rates. This study showed the need for the consideration of gram-negative coverage in addition to standard broad gram-positive coverage. Future studies should investigate the effect of the proposed empirical therapy on long-term outcomes. LEVEL OF EVIDENCE Prognostic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Michelle M J Jacobs
- Department of Orthopaedic Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Micha Holla
- Department of Orthopaedic Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Bas van Wageningen
- Department of Trauma Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands ; and
| | - Erik Hermans
- Department of Trauma Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands ; and
| | - Karin Veerman
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
3
|
Bavaro DF, Belati A, Bussini L, Cento V, Diella L, Gatti M, Saracino A, Pea F, Viale P, Bartoletti M. Safety and effectiveness of fifth generation cephalosporins for the treatment of methicillin-resistant staphylococcus aureus bloodstream infections: a narrative review exploring past, present, and future. Expert Opin Drug Saf 2024; 23:9-36. [PMID: 38145925 DOI: 10.1080/14740338.2023.2299377] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infection (BSI) is a major issue in healthcare, since it is often associated with endocarditis or deep site foci. Relevant morbidity and mortality associated with MRSA-BSIs forced the development of new antibiotic strategies; in particular, this review will focus the attention on fifth-generation cephalosporins (ceftaroline/ceftobiprole), that are the only ß-lactams active against MRSA. AREAS COVERED The review discusses the available randomized controlled trials and real-world observational studies conducted on safety and effectiveness of ceftaroline/ceftobiprole for the treatment of MRSA-BSIs. Finally, a proposal of MRSA-BSI treatment flowchart, based on fifth-generation cephalosporins, is described. EXPERT OPINION The use of anti-MRSA cephalosporins is an acceptable choice either in monotherapy or combination therapy for the treatment of MRSA-BSIs due to their relevant effectiveness and safety. Particularly, their use may be advisable in combination therapy in case of severe infections (including endocarditis or persistent bacteriemia) or in monotherapy in subjects at higher risk of drugs-induced toxicity with older regimens. On the contrary, caution should be taken in case of suspected/ascertained central nervous system infections due to inconsistent data regarding penetration of these drugs in cerebrospinal fluid and brain tissues.
Collapse
Affiliation(s)
- Davide Fiore Bavaro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Infectious Disease Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alessandra Belati
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari "Aldo Moro", Bari, Italy
| | - Linda Bussini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Infectious Disease Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valeria Cento
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Microbiology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lucia Diella
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari "Aldo Moro", Bari, Italy
| | - Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Annalisa Saracino
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari "Aldo Moro", Bari, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Viale
- Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Infectious Disease Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michele Bartoletti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Infectious Disease Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
4
|
Kebriaei R, Abdul-Mutakabbir JC, Stamper KC, Lev KL, Rybak MJ. Targeting Dalbavancin Inoculum Effect: Adjunctive Single Dose of Daptomycin. Infect Dis Ther 2023; 12:2485-2494. [PMID: 37798469 PMCID: PMC10600059 DOI: 10.1007/s40121-023-00875-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Daptomycin (DAP) has proven to be a viable alternative amid vancomycin resistance; however, the use of DAP post vancomycin treatment has led to the development of DAP non-susceptible (DNS) strains. Dalbavancin (DAL), a novel single-dosed lipoglycopeptide, has shown enhanced activity against highly resistant Staphylococcus aureus strains. However, on the basis of previous reports and our observations, DAL does not demonstrate similar activity at high versus low inoculum levels. Therefore, we hypothesized that addition of DAP even at minimal concentrations (single dose on day 1) will lower the inoculum to the level that can be cleared by dalbavancin. METHODS Isolates from methicillin-resistant S. aureus (MRSA)-infected patients with varying susceptibility profiles were evaluated using broth microdilution methods. Two DNS-VISA strains (vancomycin intermediate resistant S. aureus) and one MRSA strain were further evaluated in a one-compartment PK/PD model using a high starting initial inoculum of 109 CFU/mL as well as low initial inoculum of 107 CFU/mL over 168 h to assess the activity of DAL and DAP monotherapy and in combination. RESULTS Single therapies were not bactericidal when evaluated in the 168 h in vitro one-compartment model with an initial inoculum of 109; however, the combination of DAL plus single dose of DAP resulted in enhanced killing at the end of the 168-h exposure. DAL single therapy caused reduction in colony counts down to detection limit (2 log10 CFU/ml) at a lower inoculum but did not show enhancement (< 2 log10 CFU/ml) at higher initial inoculums (P < 0.01) for all three strains. Similarly, DAP caused initial bacterial reduction up to 4 log10 CFU/ml with regrowth at about 32 h of exposure, which stayed at initial inoculum levels for the duration of the model for all three strains. CONCLUSIONS Dalbavancin inoculum effect is a major issue in bacterial infections with high bacterial loads and the combination of DAL plus single dose of DAP showed promise in eradicating resistant S. aureus strains at high inoculums.
Collapse
Affiliation(s)
- Razieh Kebriaei
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, Ohio State University, Columbus, USA
| | - Jacinda C Abdul-Mutakabbir
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Division of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Kyle C Stamper
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Prism Labs, LLC, Walled Lake, MI, USA
| | - Katherine L Lev
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Michael J Rybak
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA.
- Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, MI, USA.
- Detroit Receiving Hospital, Detroit, MI, USA.
| |
Collapse
|
5
|
Chang J, Tasellari A, Wagner JL, Scheetz MH. Contemporary pharmacologic treatments of MRSA for hospitalized adults: rationale for vancomycin versus non-vancomycin therapies as first line agents. Expert Rev Anti Infect Ther 2023; 21:1309-1325. [PMID: 37876291 DOI: 10.1080/14787210.2023.2275663] [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: 05/25/2023] [Accepted: 10/23/2023] [Indexed: 10/26/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) remains an important pathogen in the hospital setting and causes significant morbidity and mortality each year. Since the initial discovery over 60 years ago, vancomycin has remained a first-line treatment for many different types of MRSA infections. However, significant concerns related to target attainment and nephrotoxicity have spurred efforts to develop more effective agents in the last two decades. AREAS COVERED Newer anti-MRSA antibiotics that have been approved since 2000 include linezolid, daptomycin, and ceftaroline. As clinical evidence has accumulated, these newer agents have become more frequently used, and some are now recommended as co-first-line options (along with vancomycin) in clinical practice guidelines. For this review, a scoping review of the literature was conducted to support our findings and recommendations. EXPERT OPINION Vancomycin remains an important standard of care for MRSA infections but is limited with respect to nephrotoxicity and rapid target attainment. Newer agents such as linezolid, daptomycin, and ceftaroline have specific indications for treating different types of MRSA infections; however, newer agents also have unique attributes which require consideration during therapy.
Collapse
Affiliation(s)
- Jack Chang
- Department of Pharmacy Practice, Midwestern University College of Pharmacy, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Midwestern University College of Pharmacy, Downers Grove, IL, USA
- Northwestern Memorial Hospital, Department of Pharmacy, Chicago, IL, USA
| | - Ardita Tasellari
- Department of Pharmacy Practice, Midwestern University College of Pharmacy, Downers Grove, IL, USA
| | - Jamie L Wagner
- School of Pharmacy, University of Mississippi, Jackson, MS, USA
| | - Marc H Scheetz
- Department of Pharmacy Practice, Midwestern University College of Pharmacy, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Midwestern University College of Pharmacy, Downers Grove, IL, USA
- Northwestern Memorial Hospital, Department of Pharmacy, Chicago, IL, USA
| |
Collapse
|
6
|
Sidders AE, Kedziora KM, Arts M, Daniel JM, de Benedetti S, Beam JE, Bui DT, Parsons JB, Schneider T, Rowe SE, Conlon BP. Antibiotic-induced accumulation of lipid II synergizes with antimicrobial fatty acids to eradicate bacterial populations. eLife 2023; 12:80246. [PMID: 36876902 PMCID: PMC10030119 DOI: 10.7554/elife.80246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 03/05/2023] [Indexed: 03/07/2023] Open
Abstract
Antibiotic tolerance and antibiotic resistance are the two major obstacles to the efficient and reliable treatment of bacterial infections. Identifying antibiotic adjuvants that sensitize resistant and tolerant bacteria to antibiotic killing may lead to the development of superior treatments with improved outcomes. Vancomycin, a lipid II inhibitor, is a frontline antibiotic for treating methicillin-resistant Staphylococcus aureus and other Gram-positive bacterial infections. However, vancomycin use has led to the increasing prevalence of bacterial strains with reduced susceptibility to vancomycin. Here, we show that unsaturated fatty acids act as potent vancomycin adjuvants to rapidly kill a range of Gram-positive bacteria, including vancomycin-tolerant and resistant populations. The synergistic bactericidal activity relies on the accumulation of membrane-bound cell wall intermediates that generate large fluid patches in the membrane leading to protein delocalization, aberrant septal formation, and loss of membrane integrity. Our findings provide a natural therapeutic option that enhances vancomycin activity against difficult-to-treat pathogens, and the underlying mechanism may be further exploited to develop antimicrobials that target recalcitrant infection.
Collapse
Affiliation(s)
- Ashelyn E Sidders
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Katarzyna M Kedziora
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Bioinformatics and Analytics Research Collaborative, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
| | - Jan-Martin Daniel
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
| | | | - Jenna E Beam
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Duyen T Bui
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Joshua B Parsons
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Division of Infectious Diseases, Duke University, Durham, United States
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
| | - Sarah E Rowe
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Brian P Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
7
|
de Tymowski C, Sahnoun T, Provenchere S, Para M, Derre N, Mutuon P, Duval X, Grall N, Iung B, Kernéis S, Lucet JC, Montravers P. Impact of Antibiotic Prophylaxis on Surgical Site Infections in Cardiac Surgery. Antibiotics (Basel) 2023; 12:85. [PMID: 36671286 PMCID: PMC9854463 DOI: 10.3390/antibiotics12010085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
(1) Background: Cephalosporins (CA) are the first-line antibiotic prophylaxis recommended to prevent surgical site infection (SSI) after cardiac surgery. The combination of vancomycin/gentamicin (VGA) might represent a good alternative, but few studies have evaluated its efficacy in SSI prevention. (2) Methods: A single-centre retrospective study was conducted over a 13-year period in all consecutive adult patients undergoing elective cardiac surgery. Patients were stratified according to the type of antibiotic prophylaxis. CA served as the first-line prophylaxis, and VGA was used as the second-line prophylaxis. The primary endpoint was SSI occurrence at 90 days, which was defined as the need for reoperation due to SSI. (3) Results: In total, 14,960 adult patients treated consecutively from 2006 to 2019 were included in this study, of whom 1774 (12%) received VGA and 540 (3.7%) developed SSI. VGA patients had higher severity with increased 90-day mortality. Nevertheless, the frequency of SSI was similar between CA and VGA patients. However, the microbiological aetiologies were different, with more Gram-negative bacteria noted in the VGA group. (4) Conclusions: VGA seems to be as effective as CA in preventing SSI.
Collapse
Affiliation(s)
- Christian de Tymowski
- Department of Anaesthesiology and Surgical Intensive Care, DMU PARABOL, AP-HP, Hôpital Bichat, 75018 Paris, France
- Université Paris Cité, Centre de Recherche sur l’Inflammation, INSERM UMR 1149, CNRS ERL8252, F-75018 Paris, France
- Laboratory of Excellence, Inflamex, Université Paris Cité, F-75018 Paris, France
- Department of Immunology, DHU Fire, AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Tarek Sahnoun
- Department of Anaesthesiology and Surgical Intensive Care, DMU PARABOL, AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Sophie Provenchere
- Department of Anaesthesiology and Surgical Intensive Care, DMU PARABOL, AP-HP, Hôpital Bichat, 75018 Paris, France
- INSERM Clinical Investigation Center 1425, 75018 Paris, France
| | - Marylou Para
- Department of Cardiac Surgery, AP-HP, Hôpital Bichat, 75018 Paris, France
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
| | - Nicolas Derre
- Department of Anaesthesiology and Surgical Intensive Care, DMU PARABOL, AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Pierre Mutuon
- Service MSI, AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Xavier Duval
- INSERM Clinical Investigation Center 1425, 75018 Paris, France
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
- Université Paris Cité, INSERM, IAME, F-75018 Paris, France
| | - Nathalie Grall
- Université Paris Cité, INSERM, IAME, F-75018 Paris, France
- Service de Bactériologie, AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Bernard Iung
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
- Cardiology Department, AP-HP, Bichat Hospital, Université Paris Cite, INSERM 1148, 46 Rue Henri Huchard, 75018 Paris, France
| | - Solen Kernéis
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
- Université Paris Cité, INSERM, IAME, F-75018 Paris, France
- Equipe de Prévention du Risque Infectieux (EPRI), AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Jean-Christophe Lucet
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
- Université Paris Cité, INSERM, IAME, F-75018 Paris, France
- Equipe de Prévention du Risque Infectieux (EPRI), AP-HP, Hôpital Bichat, 75018 Paris, France
| | - Philippe Montravers
- Department of Anaesthesiology and Surgical Intensive Care, DMU PARABOL, AP-HP, Hôpital Bichat, 75018 Paris, France
- UFR Paris Nord, Université Paris Cité, 75006 Paris, France
- Université Paris Cité, Physiopathologie et Epidémiologie des Maladies Respiratoires, INSERM UMR 1152, F-75018 Paris, France
| |
Collapse
|
8
|
Antonello RM, Canetti D, Riccardi N. Daptomycin synergistic properties from in vitro and in vivo studies: a systematic review. J Antimicrob Chemother 2022; 78:52-77. [PMID: 36227704 DOI: 10.1093/jac/dkac346] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/21/2022] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Daptomycin is a bactericidal lipopeptide antibiotic approved for the treatment of systemic infections (i.e. skin and soft tissue infections, bloodstream infections, infective endocarditis) caused by Gram-positive cocci. It is often prescribed in association with a partner drug to increase its bactericidal effect and to prevent the emergence of resistant strains during treatment; however, its synergistic properties are still under evaluation. METHODS We performed a systematic review to offer clinicians an updated overview of daptomycin synergistic properties from in vitro and in vivo studies. Moreover, we reported all in vitro and in vivo data evaluating daptomycin in combination with other antibiotic agents, subdivided by antibiotic classes, and a summary graph presenting the most favourable combinations at a glance. RESULTS A total of 92 studies and 1087 isolates (723 Staphylococcus aureus, 68 Staphylococcus epidermidis, 179 Enterococcus faecium, 105 Enterococcus faecalis, 12 Enterococcus durans) were included. Synergism accounted for 30.9% of total interactions, while indifferent effect was the most frequently observed interaction (41.9%). Antagonistic effect accounted for 0.7% of total interactions. The highest synergistic rates against S. aureus were observed with daptomycin in combination with fosfomycin (55.6%). For S. epidermidis and Enterococcus spp., the most effective combinations were daptomycin plus ceftobiprole (50%) and daptomycin plus fosfomycin (63.6%) or rifampicin (62.8%), respectively. FUTURE PERSPECTIVES We believe this systematic review could be useful for the future updates of guidelines on systemic infections where daptomycin plays a key role.
Collapse
Affiliation(s)
- Roberta Maria Antonello
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50121, Italy
| | - Diana Canetti
- Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Niccolò Riccardi
- Department of Clinical and Experimental Medicine, Infectious Diseases Unit, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa 56124, Italy
| |
Collapse
|
9
|
Lehman SM, Kongari R, Glass AM, Koert M, Ray MD, Plaut RD, Stibitz S. Phage K gp102 Drives Temperature-Sensitive Antibacterial Activity on USA300 MRSA. Viruses 2022; 15:17. [PMID: 36680060 PMCID: PMC9861931 DOI: 10.3390/v15010017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
There is widespread interest in using obligately lytic bacteriophages ("phages") to treat human bacterial infections. Among Staphylococcus aureus infections, the USA300 lineage is a frequent cause of invasive disease. We observed that phage K, a model S. aureus myophage, exhibits temperature-sensitive growth on USA300 strains, with the wild-type phage providing poorer growth suppression in broth and forming smaller and fainter plaques at 37 °C vs. 30 °C. We isolated 65 mutants of phage K that had improved plaquing characteristics at 37 °C when compared to the parental phage. In all 65 mutants, this phenotype was attributable to loss-of-function (LoF) mutations in gp102, which encodes a protein of unknown function that has homologs only among the Herelleviridae (SPO1-like myophages infecting gram-positive bacteria). Additional experiments with representative mutants consistently showed that the temperature-sensitive plaque phenotype was specific to USA300 MRSA strains and that Gp102 disruption was correlated with improved suppression of bacterial growth in broth and improved antibacterial activity in a mouse model of upper respiratory tract infection. The same genotype and in vitro phenotypes could be replicated in close relatives of phage K. Gp102 disruption did not have a detectable effect on adsorption but did delay cell culture lysis relative to wild-type under permissive infection conditions, suggesting that gp102 conservation might be maintained by selective pressure for more rapid replication. Expression of gp102 on a plasmid was toxic to both an MSSA and a USA300 MRSA strain. Molecular modeling predicts a protein with two helix-turn-helix domains that displays some similarity to DNA-binding proteins such as transcription factors. While its function remains unclear, gp102 is a conserved gene that is important to the infection process of Kayvirus phages, and it appears that the manner in which USA300 strains defend against them at 37 °C can be overcome by gp102 LoF mutations.
Collapse
|
10
|
Pereira JG, Fernandes J, Duarte AR, Fernandes SM. β-Lactam Dosing in Critical Patients: A Narrative Review of Optimal Efficacy and the Prevention of Resistance and Toxicity. Antibiotics (Basel) 2022; 11:antibiotics11121839. [PMID: 36551496 PMCID: PMC9774837 DOI: 10.3390/antibiotics11121839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial prescription in critically ill patients represents a complex challenge due to the difficult balance between infection treatment and toxicity prevention. Underexposure to antibiotics and therapeutic failure or, conversely, drug overexposure and toxicity may both contribute to a worse prognosis. Moreover, changes in organ perfusion and dysfunction often lead to unpredictable pharmacokinetics. In critically ill patients, interindividual and intraindividual real-time β-lactam antibiotic dose adjustments according to the patient's condition are critical. The continuous infusion of β-lactams and the therapeutic monitoring of their concentration have both been proposed to improve their efficacy, but strong data to support their use are still lacking. The knowledge of the pharmacokinetic/pharmacodynamic targets is poor and is mostly based on observational data. In patients with renal or hepatic failure, selecting the right dose is even more tricky due to changes in drug clearance, distribution, and the use of extracorporeal circuits. Intermittent usage may further increase the dosing conundrum. Recent data have emerged linking overexposure to β-lactams to central nervous system toxicity, mitochondrial recovery delay, and microbiome changes. In addition, it is well recognized that β-lactam exposure facilitates resistance selection and that correct dosing can help to overcome it. In this review, we discuss recent data regarding real-time β-lactam antibiotic dose adjustment, options in special populations, and the impacts on mitochondria and the microbiome.
Collapse
Affiliation(s)
- João Gonçalves Pereira
- Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, 4450-681 Matosinhos, Portugal
- Correspondence: ; Tel.: +351-96-244-1546
| | - Joana Fernandes
- Centro Hospitalar de Trás-os-Montes e Alto Douro, 5000-508 Vila Real, Portugal
| | - Ana Rita Duarte
- Nova Medical School, Universidade NOVA de Lisboa, 1099-085 Lisbon, Portugal
| | - Susana Mendes Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, 4450-681 Matosinhos, Portugal
- Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| |
Collapse
|
11
|
Reddy P. Clinical Approach to Nosocomial Bacterial Sepsis. Cureus 2022; 14:e28601. [PMID: 36185840 PMCID: PMC9521889 DOI: 10.7759/cureus.28601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
Bacterial sepsis and septic shock are associated with a high mortality, and when clinically suspected, clinicians must initiate broad-spectrum antimicrobials within the first hour of diagnosis. Thorough review of prior cultures involving multidrug-resistant (MDR) pathogens along with other likely pathogens should be performed to provide an appropriate broad-spectrum empiric antibiotic coverage. The appropriate antibiotic loading dose followed by individualized modification of maintenance dose should be implemented based on the presence of hepatic or renal dysfunction. Use of procalcitonin is no longer recommended to determine need for initial antibacterial therapy and for de-escalation. Daily reevaluation of appropriateness of treatment is necessary based on the culture results and clinical response. All positive cultures should be carefully screened for possible contamination or colonization, which may not represent the true organism causing the sepsis. Culture negative sepsis accounts for one-half of all cases, and de-escalation of initial antibiotic regimen should be done gradually in these patients with close monitoring.
Collapse
|
12
|
La YJ, Kim HR, Oh DH, Ahn JY, Kim YC. Comparison of Clinical Outcomes for Glycopeptides and Beta-Lactams in Methicillin-Susceptible Staphylococcus Aureus Bloodstream Infections. Yonsei Med J 2022; 63:611-618. [PMID: 35748072 PMCID: PMC9226830 DOI: 10.3349/ymj.2022.63.7.611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/20/2022] [Indexed: 11/27/2022] Open
Abstract
PURPOSE This study aimed to provide compelling evidence of anti-staphylococcal beta-lactam use for methicillin-susceptible Staphylococcus aureus bloodstream infection (MSSA BSI). MATERIALS AND METHODS We retrospectively collected data on patients with MSSA BSI who were admitted to two academic tertiary-care hospitals from 2010 to 2018. Only patients who received nafcillin, cefazolin, vancomycin, or teicoplanin as definitive therapy were included. The primary outcome was 28-day mortality. To perform unbiased comparisons between both treatments, we used inverse probability of treatment weighting (IPTW) analysis. RESULTS A total of 359 patients were divided into two groups based on the definitive therapy used: beta-lactams (n=203), including nafcillin or cefazolin; and glycopeptides (n=156), including vancomycin or teicoplanin. In the IPTW analysis, glycopeptides were associated with significantly increased odds of 28-day mortality (adjusted odds ratio, 3.37; 95% confidence interval, 1.71-6.61; p<0.001). The rate of primary outcome in prespecified subgroups was largely consistent with the main analysis. CONCLUSION Definitive therapy with beta-lactams in patients with MSSA BSI was associated with lower 28-day mortality compared to definitive therapy with glycopeptides.
Collapse
Affiliation(s)
- Yeon Ju La
- Department of Internal Medicine, Kangwon National University Hospital, Chuncheon, Korea
| | - Hye Rim Kim
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hyun Oh
- Department of Internal Medicine, Seoul Medical Center, Seoul, Korea
| | - Jin Young Ahn
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Yong Chan Kim
- Department of Internal Medicine, Division of Infectious Disease, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea.
| |
Collapse
|
13
|
Dégi J, Herman V, Igna V, Dégi DM, Hulea A, Muselin F, Cristina RT. Antibacterial Activity of Romanian Propolis against Staphylococcus aureus Isolated from Dogs with Superficial Pyoderma: In Vitro Test. Vet Sci 2022; 9:vetsci9060299. [PMID: 35737351 PMCID: PMC9231063 DOI: 10.3390/vetsci9060299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022] Open
Abstract
Staphylococcal infection treatment in dogs is frequently associated with adverse side effects, high costs, prolonged treatment, and resistant strain selection. Staphylococcus aureus is the most frequently isolated staphylococci in cases of canine superficial pyoderma. The number of Staphylococcus strains to exhibit primary resistance to various drugs in vitro is increasing. Propolis has a diverse chemical composition and well-known therapeutic properties against bacterial infections. The current investigation evaluated in vitro the antimicrobial activity of the commercial allopathic antimicrobials, Romanian propolis ethanolic extracts, against clinical Staphylococcus aureus strains isolated from superficial dermatitis clinical samples in dogs and two reference strains: Staphylococcus aureus ATCC 25923 and Staphylococcus aureus ATCC 43300, as the MSSA and MRSA positive controls, respectively, in western Romania. We used the microdilution broth technique to evaluate the susceptibility profile of the bacteria. The minimum inhibitory concentration (MIC) of the Romanian propolis ethanolic extract ranged from 6 to 10 μg/mL for all isolates, determined by the broth microdilution method. The MICs of ethanolic Romanian propolis extracts had a pronounced antibacterial activity. These results indicate that propolis can potentially be used and recommended for in vivo experiments as a promising therapeutic agent against Staphylococcus aureus infections in superficial dermatitis of dogs.
Collapse
|
14
|
Bian X, Qu X, Zhang J, Nang SC, Bergen PJ, Tony Zhou Q, Chan HK, Feng M, Li J. Pharmacokinetics and pharmacodynamics of peptide antibiotics. Adv Drug Deliv Rev 2022; 183:114171. [PMID: 35189264 PMCID: PMC10019944 DOI: 10.1016/j.addr.2022.114171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 01/05/2023]
Abstract
Antimicrobial resistance is a major global health challenge. As few new efficacious antibiotics will become available in the near future, peptide antibiotics continue to be major therapeutic options for treating infections caused by multidrug-resistant pathogens. Rational use of antibiotics requires optimisation of the pharmacokinetics and pharmacodynamics for the treatment of different types of infections. Toxicodynamics must also be considered to improve the safety of antibiotic use and, where appropriate, to guide therapeutic drug monitoring. This review focuses on the pharmacokinetics/pharmacodynamics/toxicodynamics of peptide antibiotics against multidrug-resistant Gram-negative and Gram-positive pathogens. Optimising antibiotic exposure at the infection site is essential for improving their efficacy and minimising emergence of resistance.
Collapse
Affiliation(s)
- Xingchen Bian
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; School of Pharmacy, Fudan University, Shanghai, China
| | - Xingyi Qu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; School of Pharmacy, Fudan University, Shanghai, China; Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Sue C Nang
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Phillip J Bergen
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Meiqing Feng
- School of Pharmacy, Fudan University, Shanghai, China
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia.
| |
Collapse
|
15
|
Bouchelaghem S, Das S, Naorem RS, Czuni L, Papp G, Kocsis M. Evaluation of Total Phenolic and Flavonoid Contents, Antibacterial and Antibiofilm Activities of Hungarian Propolis Ethanolic Extract against Staphylococcus aureus. Molecules 2022; 27:574. [PMID: 35056886 PMCID: PMC8782033 DOI: 10.3390/molecules27020574] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/24/2022] Open
Abstract
Propolis is a natural bee product that is widely used in folk medicine. This study aimed to evaluate the antimicrobial and antibiofilm activities of ethanolic extract of propolis (EEP) on methicillin-resistant and sensitive Staphylococcus aureus (MRSA and MSSA). Propolis samples were collected from six regions in Hungary. The minimum inhibitory concentrations (MIC) values and the interaction of EEP-antibiotics were evaluated by the broth microdilution and the chequerboard broth microdilution methods, respectively. The effect of EEP on biofilm formation and eradication was estimated by crystal violet assay. Resazurin/propidium iodide dyes were applied for simultaneous quantification of cellular metabolic activities and dead cells in mature biofilms. The EEP1 sample showed the highest phenolic and flavonoid contents. The EEP1 successfully prevented the growth of planktonic cells of S. aureus (MIC value = 50 µg/mL). Synergistic interactions were shown after the co-exposition to EEP1 and vancomycin at 108 CFU/mL. The EEP1 effectively inhibited the biofilm formation and caused significant degradation of mature biofilms (50-200 µg/mL), as a consequence of the considerable decrement of metabolic activity. The EEP acts effectively as an antimicrobial and antibiofilm agent on S. aureus. Moreover, the simultaneous application of EEP and vancomycin could enhance their effect against MRSA infection.
Collapse
Affiliation(s)
- Sarra Bouchelaghem
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Sourav Das
- Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság Str. 13, 7624 Pécs, Hungary;
| | - Romen Singh Naorem
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Lilla Czuni
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Gábor Papp
- Department of General and Environmental Microbiology, Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary; (S.B.); (R.S.N.); (L.C.); (G.P.)
| | - Marianna Kocsis
- Department of Plant Biology, Institute of Biology, University of Pécs, Ifjúság str. 6, 7624 Pécs, Hungary
| |
Collapse
|
16
|
Yang G, Yan Y, Mao J, Liu H, Chen M, Zhang N, Li Y, Gu J, Huang X. Development and Validation of an HPLC-UV Method for Quantitation of Linezolid: Application to Resistance Study Using in vitro PK/PD Model. Infect Drug Resist 2021; 14:5089-5098. [PMID: 34880634 PMCID: PMC8647170 DOI: 10.2147/idr.s343200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/19/2021] [Indexed: 11/23/2022] Open
Abstract
Background Linezolid (LNZ), an oxazolidinone antibiotic, has 100% oral bioavailability and favorable activities against gram-positive pathogens. The in vitro PK/PD model was developed based on concentrations obtained with routine doses in humans can be used to guide dose optimization in the clinic. Methods In this study, we employed an in vitro PK/PD model to simulate the changes in the plasma concentration of linezolid in the human body against a clinical isolate of MRSA in vitro. A high-performance liquid chromatography (HPLC)-UV method was applied to measure the concentration of linezolid. Bacterial samples were collected at different times from the central compartment for count. Results The chromatographic separation was carried out with an AichromBond-AQC18 column(250mm×4.6mm, 5μm), using a mobile phase of water with 0.1% formic acid:acetonitrile 70:30 (v/v), followed by detection at 254 nm, and a single detection run was completed within 10 min. The method was validated by estimating the precision and accuracy for the inter- and intra-day analyses in the concentration range of 0.25-32 mg/L. The method was linear over the investigated range of 0.125-32 mg/L, with all correlation coefficients R2 = 0.9999. The intra-day and inter-day precisions were within 7.598%, and the method recovery ranged from 90.912% to 106.459%. In vitro PK/PD model, both the absorption and elimination of linezolid being simulated can be precisely controlled by computer. In the control group, the bacterial reached 7.9 Log10CFU/mL in the first 48h and maintained until the end, indicating that the colonies grew well in vitro PK/PD model. In the linezolid 600 mg q12h administration group, the colony decreased to 2.39 Log10CFU/mL at 24h, showing a good bactericidal effect; however, the colonies resumed growth to the initial level in 48h, indicating an emergence of resistance. Conclusion We successfully established an in vitro infection PK/PD model and developed an HPLC-UV method to determine linezolid concentration for resistance investigation. The results suggest that the 600 mg q12h dosing regimen may no longer be applicable and requires optimization.
Collapse
Affiliation(s)
- Guang Yang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China.,Department of Pharmacy, The Third People's Hospital of Tongling, Tongling, Anhui, People's Republic of China
| | - Yisong Yan
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China.,Department of Pharmacy, Anhui College of Traditional Chinese Medicine, Wuhu, Anhui, People's Republic of China
| | - Jun Mao
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Huiping Liu
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Mingtao Chen
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Na Zhang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yaowen Li
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jiangjun Gu
- Department of Pharmacy, The Third People's Hospital of Tongling, Tongling, Anhui, People's Republic of China
| | - Xiaohui Huang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
17
|
Golikova MV, Strukova EN, Portnoy YA, Zinner SH, Firsov AA. MPC-Based Prediction of Anti-Mutant Effectiveness of Antibiotic Combinations: In Vitro Model Study with Daptomycin and Gentamicin against Staphylococcus aureus. Antibiotics (Basel) 2021; 10:antibiotics10101148. [PMID: 34680729 PMCID: PMC8532831 DOI: 10.3390/antibiotics10101148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 01/04/2023] Open
Abstract
To explore whether combined treatments with daptomycin and gentamicin can prevent the development of Staphylococcus aureus resistance, and whether the possible restriction is associated with changes in antibiotic mutant prevention concentrations (MPCs), the enrichment of daptomycin- and gentamicin-resistant mutants was studied by simulating 5-day single and combined treatments in an in vitro dynamic model. The MPCs of the antibiotics in the combination were determined at concentration ratios equal to the ratios of 24 h areas, under the concentration–time curve (AUCs) of the antibiotics, as simulated in pharmacodynamic experiments. The MPCs of both daptomycin and gentamicin decreased in the presence of each other; this led to an increase in the time when antibiotic concentrations were above the MPC (T>MPC). The increases in T>MPCs were concurrent with increases of the anti-mutant effects of the combined antibiotics. When anti-mutant effects of the antibiotics in single and combined treatments were plotted against the T>MPCs, significant sigmoid relationships were obtained. These findings suggest that (1) daptomycin–gentamicin combinations prevent the development of S. aureus resistance to each antibiotic; (2) the anti-mutant effects of antibiotic combinations can be predicted using MPCs determined at pharmacokinetic-based antibiotic concentration ratios; (3) T>MPC is a reliable predictor of the anti-mutant efficacy of antibiotic combinations.
Collapse
Affiliation(s)
- Maria V. Golikova
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya Street, 119021 Moscow, Russia; (E.N.S.); (Y.A.P.); (A.A.F.)
- Correspondence: ; Tel.: +7-926-7077198
| | - Elena N. Strukova
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya Street, 119021 Moscow, Russia; (E.N.S.); (Y.A.P.); (A.A.F.)
| | - Yury A. Portnoy
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya Street, 119021 Moscow, Russia; (E.N.S.); (Y.A.P.); (A.A.F.)
| | - Stephen H. Zinner
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mount Auburn St., Cambridge, MA 02138, USA;
| | - Alexander A. Firsov
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, 11 Bolshaya Pirogovskaya Street, 119021 Moscow, Russia; (E.N.S.); (Y.A.P.); (A.A.F.)
| |
Collapse
|
18
|
Van Belkum A, Gros MF, Ferry T, Lustig S, Laurent F, Durand G, Jay C, Rochas O, Ginocchio CC. Novel strategies to diagnose prosthetic or native bone and joint infections. Expert Rev Anti Infect Ther 2021; 20:391-405. [PMID: 34384319 DOI: 10.1080/14787210.2021.1967745] [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] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Bone and Joint Infections (BJI) are medically important, costly and occur in native and prosthetic joints. Arthroplasties will increase significantly in absolute numbers over time as well as the incidence of Prosthetic Joint Infections (PJI). Diagnosis of BJI and PJI is sub-optimal. The available diagnostic tests have variable effectiveness, are often below standard in sensitivity and/or specificity, and carry significant contamination risks during the collection of clinical samples. Improvement of diagnostics is urgently needed. AREAS COVERED We provide a narrative review on current and future diagnostic microbiology technologies. Pathogen identification, antibiotic resistance detection, and assessment of the epidemiology of infections via bacterial typing are considered useful for improved patient management. We confirm the continuing importance of culture methods and successful introduction of molecular, mass spectrometry-mediated and next-generation genome sequencing technologies. The diagnostic algorithms for BJI must be better defined, especially in the context of diversity of both disease phenotypes and clinical specimens rendered available. EXPERT OPINION Whether interventions in BJI or PJI are surgical or chemo-therapeutic (antibiotics and bacteriophages included), prior sensitive and specific pathogen detection remains a therapy-substantiating necessity. Innovative tests for earlier and more sensitive and specific detection of bacterial pathogens in BJI are urgently needed.
Collapse
Affiliation(s)
- Alex Van Belkum
- bioMérieux, Open Innovation and Partnerships, 3 Route De Port Michaud, La Balme Les Grottes, France
| | | | - Tristan Ferry
- Service Des Maladies Infectieuses Et Tropicales, Hospices Civils De Lyon, Hôpital De La Croix-Rousse, Lyon, France.,Maladies Infectieuses, Université Claude Bernard Lyon 1, Villeurbanne, France.,Centre Interrégional De Référence Pour La Prise En Charge Des Infections Ostéo-articulaires Complexes (Crioac Lyon), Hôpital De La Croix-Rousse, Lyon, France.,Ciri - Centre International De Recherche En Infectiologie, Inserm, U1111, Université́ Claude Bernard Lyon 1CNRS, UMR5308, Ecole Normale Supérieure De Lyon, Univ Lyon, Lyon, France
| | - Sebastien Lustig
- Maladies Infectieuses, Université Claude Bernard Lyon 1, Villeurbanne, France.,Service De Chirurgie Orthopédique, Hôpital De La Croix-Rousse, Lyon, France
| | - Frédéric Laurent
- Service Des Maladies Infectieuses Et Tropicales, Hospices Civils De Lyon, Hôpital De La Croix-Rousse, Lyon, France.,Ciri - Centre International De Recherche En Infectiologie, Inserm, U1111, Université́ Claude Bernard Lyon 1CNRS, UMR5308, Ecole Normale Supérieure De Lyon, Univ Lyon, Lyon, France
| | | | - Corinne Jay
- bioMérieux, BioFire Development Emea, Grenoble, France
| | - Olivier Rochas
- Corporate Business Development, bioMérieux, Marcy-l'Étoile, France
| | | |
Collapse
|
19
|
Konstantinovski MM, Veldkamp KE, Lavrijsen APM, Bosch T, Kraakman MEM, Nooij S, Claas ECJ, Gooskens J. Hospital transmission of borderline oxacillin-resistant Staphylococcus aureus evaluated by whole-genome sequencing. J Med Microbiol 2021; 70. [PMID: 34269673 PMCID: PMC8493421 DOI: 10.1099/jmm.0.001384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction Staphylococcus aureus is a major cause of hospital infections worldwide. Awareness towards methicillin-resistant S. aureus (MRSA) infections is high but attention towards borderline oxacillin-resistant S. aureus (BORSA) is limited, possibly due to an underestimated clinical relevance, presumption of low incidence and diagnostic limitations. Gap statement BORSA surveillance has not been routinely implemented, and thus consensus with regard to a definition and infection control measures is lacking. Aim Our goals were to investigate the occurrence, molecular characteristics and clinical manifestations of BORSA infections in the hospital setting. Methodology Following an increased incidence in 2016, BORSA cases in 2014/2016 (in our institution) were more specifically evaluated. Medical records were reviewed to investigate epidemiological links, clinical characteristics and outcomes. Resistance and virulence markers were assessed by whole genome sequencing (WGS). Conventional methods: amplified fragment length polymorphism (AFLP) ; multilocus sequence typing (MLST) and multiple locus variable-number tandem repeat analysis (MLVA) were compared with core genome MLST (cgMLST) and whole-genome single nucleotide polymorphism (wgSNP) analysis to confirm genetic clusters. Results From 2009 to 2013, BORSA comprised 0.1 % of all clinical S. aureus strains. In 2016, the incidence was six-fold higher in comparison to the baseline. Whole-genome SNP and cgMLST confirmed two BORSA clusters among patients with dermatological conditions. Patients with BORSA presented with skin infections, and one case developed a severe invasive infection with a fatal outcome. Infection control measures successfully prevented further transmission in both clusters. WGS findings showed that BORSA strains carried multiple resistance and virulence genes with increased pathogenic potential. Conclusion WGS and cgMLST effectively characterized and confirmed BORSA clusters among at-risk patients with clinical manifestations ranging from mild skin infections to life-threatening bacteraemia. Clinical awareness and active monitoring are therefore warranted for the timely implementation of infection control measures to prevent BORSA transmission in high-risk patients.
Collapse
Affiliation(s)
| | - Karin Ellen Veldkamp
- Medical Microbiology Department, Leiden University Medical Center, Leiden, Netherlands
| | | | - Thijs Bosch
- Infectious Diseases Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment, de Bilt, Netherlands
| | - Margriet E M Kraakman
- Medical Microbiology Department, Leiden University Medical Center, Leiden, Netherlands
| | - Sam Nooij
- Medical Microbiology Department, Leiden University Medical Center, Leiden, Netherlands
| | - Eric C J Claas
- Medical Microbiology Department, Leiden University Medical Center, Leiden, Netherlands
| | - Jairo Gooskens
- Medical Microbiology Department, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
20
|
Bajpai RR, Razdan S, Sanchez-Gonzalez MA, Razdan S. Minimizing transrectal prostate biopsy-related infections; A prospective randomized trial of povidone-iodine intrarectal cleaning versus formalin needle disinfection. Indian J Urol 2021; 37:254-260. [PMID: 34465955 PMCID: PMC8388331 DOI: 10.4103/iju.iju_34_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/31/2021] [Accepted: 06/21/2021] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Transrectal prostate biopsies are associated with post biopsy infection and sepsis. We compared the efficacy of povidone-iodine rectal disinfection versus formalin needle disinfection in preventing post biopsy infection among patients undergoing transrectal ultrasound-guided prostate biopsy. METHODS Patients scheduled to undergo ultrasound-guided transrectal prostate biopsy (n = 621) over 20 months were randomized into 2 groups to receive either povidone-iodine intrarectal disinfection or formalin disinfection of needle after each core. These were compared to assess which methodology better prevented postprocedure infection. Statistical analysis were used to identify independent factors promoting infections. RESULTS Two hundred and ninety-eight patients from povidone-iodine intrarectal disinfection were compared with 300 from formalin needle disinfection group. Formalin needle disinfection was associated with significantly more infections (P = 0.02). Escherichia coli was the dominant pathogen, with >50% of cases being quinolone resistant. Type of disinfection (P = 0.002), BMI (P = 0.001), chronic prostatitis (P = 0.002), and diabetes mellitus (P = 0.01) were independent predictors of infections. BMI at 28.95 kg/m2 provided the best predictive cut-off point for infections, irrespective of method of disinfection. Area under the curve for all these parameters together was 0.91. CONCLUSIONS We conclude that along with oral cephalosporin prophylaxis, povidone-iodine intrarectal disinfection is a superior to formalin needle disinfection alone in preventing post biopsy infection. Patients with BMI >28.95 kg/m2 should be considered at a higher risk for infections.
Collapse
Affiliation(s)
- Rajesh Raj Bajpai
- Department of Urology, Larkin Community Hospital, South Miami, Florida, USA
| | - Shirin Razdan
- Department of Urology, Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, USA
| | | | - Sanjay Razdan
- Department of Urology, International Robotic Prostatectomy Institute, Doral, Florida, USA
| |
Collapse
|
21
|
Lerche CJ, Schwartz F, Theut M, Fosbøl EL, Iversen K, Bundgaard H, Høiby N, Moser C. Anti-biofilm Approach in Infective Endocarditis Exposes New Treatment Strategies for Improved Outcome. Front Cell Dev Biol 2021; 9:643335. [PMID: 34222225 PMCID: PMC8249808 DOI: 10.3389/fcell.2021.643335] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Infective endocarditis (IE) is a life-threatening infective disease with increasing incidence worldwide. From early on, in the antibiotic era, it was recognized that high-dose and long-term antibiotic therapy was correlated to improved outcome. In addition, for several of the common microbial IE etiologies, the use of combination antibiotic therapy further improves outcome. IE vegetations on affected heart valves from patients and experimental animal models resemble biofilm infections. Besides the recalcitrant nature of IE, the microorganisms often present in an aggregated form, and gradients of bacterial activity in the vegetations can be observed. Even after appropriate antibiotic therapy, such microbial formations can often be identified in surgically removed, infected heart valves. Therefore, persistent or recurrent cases of IE, after apparent initial infection control, can be related to biofilm formation in the heart valve vegetations. On this background, the present review will describe potentially novel non-antibiotic, antimicrobial approaches in IE, with special focus on anti-thrombotic strategies and hyperbaric oxygen therapy targeting the biofilm formation of the infected heart valves caused by Staphylococcus aureus. The format is translational from preclinical models to actual clinical treatment strategies.
Collapse
Affiliation(s)
- Christian Johann Lerche
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Franziska Schwartz
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Theut
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil Loldrup Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
22
|
Dua A, Sutphin PD, Siedner MJ, Moran J. Case 16-2021: A 37-Year-Old Woman with Abdominal Pain and Aortic Dilatation. N Engl J Med 2021; 384:2054-2063. [PMID: 34042393 DOI: 10.1056/nejmcpc2100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Anahita Dua
- From the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Massachusetts General Hospital, and the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Harvard Medical School - both in Boston
| | - Patrick D Sutphin
- From the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Massachusetts General Hospital, and the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Harvard Medical School - both in Boston
| | - Mark J Siedner
- From the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Massachusetts General Hospital, and the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Harvard Medical School - both in Boston
| | - Jakob Moran
- From the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Massachusetts General Hospital, and the Departments of Surgery (A.D.), Radiology (P.D.S.), Medicine (M.J.S.), and Pathology (J.M.), Harvard Medical School - both in Boston
| |
Collapse
|
23
|
Minocycline Alone and in Combination with Polymyxin B, Meropenem, and Sulbactam against Carbapenem-Susceptible and -Resistant Acinetobacter baumannii in an In Vitro Pharmacodynamic Model. Antimicrob Agents Chemother 2021; 65:AAC.01680-20. [PMID: 33318006 DOI: 10.1128/aac.01680-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/01/2020] [Indexed: 01/23/2023] Open
Abstract
Acinetobacter baumannii is recognized as an urgent public health threat by the Centers for Disease Control and Prevention (CDC). Current treatment options are scarce, particularly against carbapenem-resistant Acinetobacter baumannii (CRAB). We simulated the impact of minocycline standard (200 mg load + 100 mg Q12h) and high (700 mg load + 350 mg Q12h) doses, polymyxin B (2.5 mg/kg Q12h), sulbactam (1 g Q6h and 9 g/24 h as continuous infusion), and meropenem (intermittent 1 or 2 g Q8h and 6 g/24 h as continuous infusion) alone or in combination against CRAB and non-CRAB isolates by simulating human therapeutic dosing regimens in a 72-h, in vitro pharmacodynamic (IVPD) model. There were no monotherapy regimens that demonstrated bactericidal activity against the tested non-CRAB and CRAB strains. Resistance development was common in monotherapy regimens. Against the CRAB isolate, the triple combination of high-dose minocycline (fAUC/MIC 21.2), polymyxin B (fAUC/MIC 15.6), and continuous-infusion sulbactam (67% T >MIC) was the most consistently active regimen. Against non-CRAB, the triple therapy regimen of high-dose minocycline (fAUC/MIC 84.8) with continuous-infusion meropenem (100% T >MIC) and continuous-infusion sulbactam (83% T >MIC), as well as the double therapy of high-dose minocycline (fAUC/MIC 84.8) with continuous-infusion meropenem (100% T >MIC), resulted in persistently bactericidal activity. In conclusion, triple therapy with high-dose minocycline, continuous-infusion sulbactam, and polymyxin B produced the most significant kill against the carbapenem-resistant Acinetobacter baumannii, with no regrowth and minimal resistance development.
Collapse
|
24
|
Salas JR, Jaberi-Douraki M, Wen X, Volkova VV. Mathematical modeling of the 'inoculum effect': six applicable models and the MIC advancement point concept. FEMS Microbiol Lett 2020; 367:5710933. [PMID: 31960902 PMCID: PMC7317156 DOI: 10.1093/femsle/fnaa012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/17/2020] [Indexed: 01/09/2023] Open
Abstract
Antimicrobial treatment regimens against bacterial pathogens are designed using the drug's minimum inhibitory concentration (MIC) measured at a bacterial density of 5.7 log10(colony-forming units (CFU)/mL) in vitro. However, MIC changes with pathogen density, which varies among infectious diseases and during treatment. Incorporating this into treatment design requires realistic mathematical models of the relationships. We compared the MIC–density relationships for Gram-negative Escherichia coli and non-typhoidal Salmonella enterica subsp. enterica and Gram-positive Staphylococcus aureus and Streptococcus pneumonia (for n = 4 drug-susceptible strains per (sub)species and 1–8 log10(CFU/mL) densities), for antimicrobial classes with bactericidal activity against the (sub)species: β-lactams (ceftriaxone and oxacillin), fluoroquinolones (ciprofloxacin), aminoglycosides (gentamicin), glycopeptides (vancomycin) and oxazolidinones (linezolid). Fitting six candidate mathematical models to the log2(MIC) vs. log10(CFU/mL) curves did not identify one model best capturing the relationships across the pathogen–antimicrobial combinations. Gompertz and logistic models (rather than a previously proposed Michaelis–Menten model) fitted best most often. Importantly, the bacterial density after which the MIC sharply increases (an MIC advancement-point density) and that density's intra-(sub)species range evidently depended on the antimicrobial mechanism of action. Capturing these dependencies for the disease–pathogen–antimicrobial combination could help determine the MICs for which bacterial densities are most informative for treatment regimen design.
Collapse
Affiliation(s)
- Jessica R Salas
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Majid Jaberi-Douraki
- Department of Mathematics, Kansas State University, Manhattan, KS 66506, USA.,Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
| | - Xuesong Wen
- Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA.,Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
| | - Victoriya V Volkova
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA.,Center for Outcomes Research and Epidemiology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
25
|
Verification of a Novel Approach to Predicting Effects of Antibiotic Combinations: In Vitro Dynamic Model Study with Daptomycin and Gentamicin against Staphylococcus aureus. Antibiotics (Basel) 2020; 9:antibiotics9090538. [PMID: 32854240 PMCID: PMC7557373 DOI: 10.3390/antibiotics9090538] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
To explore whether susceptibility testing with antibiotic combinations at pharmacokinetically derived concentration ratios is predictive of the antimicrobial effect, a Staphylococcus aureus strain was exposed to daptomycin and gentamicin alone or in combination in multiple dosing experiments. The susceptibility of the S. aureus strain to daptomycin and gentamicin in combination was tested at concentration ratios equal to the ratios of 24 h areas under the concentration–time curve (AUC24s) of antibiotics simulated in an in vitro dynamic model in five-day treatments. The MICs of daptomycin and gentamicin decreased in the presence of each other; this led to an increase in the antibiotic AUC24/MIC ratios and the antibacterial effects. Effects of single and combined treatments were plotted against the AUC24/MIC ratios of daptomycin or gentamicin, and a significant sigmoid relationship was obtained. Similarly, when the effects of single and combined treatments were related to the total exposure of both drugs (the sum of AUC24/MIC ratios (∑AUC24/MIC)), a significant sigmoid relationship was obtained. These findings suggest that (1) the effects of antibiotic combinations can be predicted by AUC24/MICs using MICs of each antibacterial determined at pharmacokinetically derived concentration ratios; (2) ∑AUC24/MIC is a reliable predictor of the antibacterial effects of antibiotic combinations.
Collapse
|
26
|
Valderrama MJ, Alfaro M, Rodríguez-Avial I, Baos E, Rodríguez-Avial C, Culebras E. Synergy of Linezolid with Several Antimicrobial Agents against Linezolid-Methicillin-Resistant Staphylococcal Strains. Antibiotics (Basel) 2020; 9:E496. [PMID: 32784878 PMCID: PMC7460281 DOI: 10.3390/antibiotics9080496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Linezolid is a synthetic oxazolydinone active against multi-resistant Gram-positive cocci that inhibits proteins synthesis by interacting with the 50S ribosomal subunit. Although linezolid-resistant strains are infrequent, several outbreaks have been recently described, associated with prolonged treatment with the antibiotic. As an alternative to monotherapy, the combination of different antibiotics is a commonly used option to prevent the selection of resistant strains. In this work, we evaluated combinations of linezolid with classic and new aminoglycosides (amikacin, gentamicin and plazomicin), carbapenems (doripenem, imipenem and meropenem) and fosfomycin on several linezolid- and methicillin-resistant strains of Staphylococcus aureus and S. epidermidis, isolated in a hospital intensive care unit in Madrid, Spain. Using checkerboard and time-kill assays, interesting synergistic effects were encountered for the combination of linezolid with imipenem in all the staphylococcal strains, and for linezolid-doripenem in S.epidermidis isolates. The combination of plazomicin seemed to also have a good synergistic or partially synergistic activity against most of the isolates. None of the combinations assayed showed an antagonistic effect.
Collapse
Affiliation(s)
- María-José Valderrama
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Alfaro
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.A.); (I.R.-A.); (E.B.); (E.C.)
| | | | - Elvira Baos
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.A.); (I.R.-A.); (E.B.); (E.C.)
| | | | - Esther Culebras
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (M.A.); (I.R.-A.); (E.B.); (E.C.)
- Departamento de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| |
Collapse
|
27
|
Huang Z, Mao C, Wei Y, Gu X, Cai Q, Shen X, Ding H. Analysis of the mutant selection window and killing of Mycoplasma hyopneumoniae for doxycycline, tylosin, danofloxacin, tiamulin, and valnemulin. PLoS One 2020; 15:e0220350. [PMID: 32544163 PMCID: PMC7297357 DOI: 10.1371/journal.pone.0220350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 05/14/2020] [Indexed: 11/20/2022] Open
Abstract
Mycoplasma hyopneumoniae is the major pathogenic microorganism causing enzootic pneumonia in pigs. With increasing resistance of M. hyopneumoniae to conventional antibiotics, treatment is becoming complicated. Herein, we investigated the mutant selection window (MSW) of doxycycline, tylosin, danofloxacin, tiamulin, and valnemulin for treating the M. hyopneumoniae type strain (ATCC 25934) to determine the likelihood of promoting resistance with continued use of these antibiotics. Minimum inhibitory concentration (MIC) values against M. hyopneumoniae were determined for each antimicrobial agent based on microdilution broth and agar dilution methods (bacterial numbers ranged from 105 colony-forming units (CFU)/mL to 109 CFU/mL). The minimal concentration inhibiting colony formation by 99% (MIC99) and the mutant prevention concentration (MPC) were determined by the agar dilution method with three inoculum sizes. Antimicrobial killing was determined based on MIC99 and MPC values for all five agents. MIC values ranged from 0.001 to 0.25 μg/mL based on the microdilution broth method, and from 0.008 to 1.0 μg/mL based on the agar dilution method. MPC values ranged from 0.0016 to 10.24 μg/mL. MPC/MIC99 values were ordered tylosin > doxycycline > danofloxacin > tiamulin > valnemulin. MPC achieved better bactericidal action than MIC99. Based on pharmacodynamic analyses, danofloxacin, tylosin, and doxycycline are more likely to select resistant mutants than tiamulin and valnemulin.
Collapse
Affiliation(s)
- Zilong Huang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Chunxiao Mao
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yanzhe Wei
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiaoyan Gu
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Qinren Cai
- Technical Center for Inspection and Quarantine, Zhuhai Entry-Exit Inspection and Quarantine Bureau, Zhuhai, China
| | - Xiangguang Shen
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Huanzhong Ding
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
- * E-mail:
| |
Collapse
|
28
|
Rose WE, Bienvenida AM, Xiong YQ, Chambers HF, Bayer AS, Ersoy SC. Ability of Bicarbonate Supplementation To Sensitize Selected Methicillin-Resistant Staphylococcus aureus Strains to β-Lactam Antibiotics in an Ex Vivo Simulated Endocardial Vegetation Model. Antimicrob Agents Chemother 2020; 64:e02072-19. [PMID: 31844004 PMCID: PMC7038310 DOI: 10.1128/aac.02072-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/08/2019] [Indexed: 12/30/2022] Open
Abstract
Supplementation of standard growth media (cation-adjusted Mueller-Hinton Broth [CAMHB]) with bicarbonate (NaHCO3) increases β-lactam susceptibility of selected methicillin-resistant Staphylococcus aureus (MRSA) strains ("NaHCO3 responsive"). This "sensitization" phenomenon translated to enhanced β-lactam efficacy in a rabbit model of endocarditis. The present study evaluated NaHCO3-mediated β-lactam MRSA sensitization using an ex vivo pharmacodynamic model, featuring simulated endocardial vegetations (SEVs), to more closely mimic the host microenvironment. Four previously described MRSA strains were used: two each exhibiting in vitro NaHCO3-responsive or NaHCO3-nonresponsive phenotypes. Cefazolin (CFZ) and oxacillin (OXA) were evaluated in CAMHB with or without NaHCO3 Intra-SEV MRSA killing was determined over 72-h exposures. In both "responsive" strains, supplementation with 25 mM or 44 mM NaHCO3 significantly reduced β-lactam MICs to below the OXA susceptibility breakpoint (≤4 mg/liter) and resulted in bactericidal activity (≥3-log killing) in the model for both OXA and CFZ. In contrast, neither in vitro-defined nonresponsive MRSA strain showed significant sensitization in the SEV model to either β-lactam. At both NaHCO3 concentrations, the fractional time above MIC was >50% for both CFZ and OXA in the responsive MRSA strains. Also, in media containing RPMI plus 10% Luria-Bertani broth (proposed as a more host-mimicking microenvironment and containing 25 mM NaHCO3), both CFZ and OXA exhibited enhanced bactericidal activity against NaHCO3-responsive strains in the SEV model. Neither CFZ nor OXA exposures selected for emergence of high-level β-lactam-resistant mutants within SEVs. Thus, in this ex vivo model of endocarditis, in the presence of NaHCO3 supplementation, both CFZ and OXA are highly active against MRSA strains that demonstrate similar enhanced susceptibility in NaHCO3-supplemented media in vitro.
Collapse
Affiliation(s)
- Warren E Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Yan Q Xiong
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Henry F Chambers
- University of California at San Francisco School of Medicine, San Francisco, California, USA
| | - Arnold S Bayer
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
- David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Selvi C Ersoy
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| |
Collapse
|
29
|
Britt NS, Hazlett DS, Horvat RT, Liesman RM, Steed ME. Activity of pulmonary vancomycin exposures versus planktonic and biofilm isolates of methicillin-resistant Staphylococcus aureus from cystic fibrosis sputum. Int J Antimicrob Agents 2020; 55:105898. [PMID: 31931147 DOI: 10.1016/j.ijantimicag.2020.105898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/30/2019] [Accepted: 01/04/2020] [Indexed: 11/30/2022]
Abstract
Vancomycin is commonly used to treat methicillin-resistant Staphylococcus aureus (MRSA) infections in patients with cystic fibrosis (CF) lung disease. However, there are limited data to support the in vitro activity of this agent against MRSA isolated from CF sputum. The primary objective of this study was to evaluate the activity of vancomycin at pulmonary concentrations (intravenous and inhaled) against four clinical MRSA CF sputum isolates in planktonic and biofilm time-kill (TK) experiments. Vancomycin minimum inhibitory concentrations (MICs) were determined for these isolates at standard inoculum (SI) (~106 CFU/mL) and high inoculum (HI) (~108 CFU/mL) as well as in biofilms cultivated using physiological medium representing the microenvironment of the CF lung. Vancomycin concentrations of 10, 25, 100 and 275 µg/mL were evaluated in TK experiments against planktonic MRSA at varying inocula and versus biofilm MRSA. Vancomycin MICs increased from 0.5 µg/mL when tested at SI to 8-16 µg/mL at HI. Vancomycin MICs were further increased to 16-32 µg/mL in biofilm studies. In TK experiments, vancomycin displayed bactericidal activity (≥3 log10 killing at 24 h) against 1/4 and 0/4 planktonic MRSA isolates at SI and HI, respectively, whereas vancomycin was bactericidal against 0/4 isolates against MRSA biofilms. Based on these findings, vancomycin monotherapy appears unlikely to eradicate MRSA from the respiratory tract of patients with CF, even at high concentrations similar to those observed with inhaled therapy. Novel vancomycin formulations with enhanced biofilm penetration or combination therapy with other potentially synergistic agents should be explored.
Collapse
Affiliation(s)
- Nicholas S Britt
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA; Department of Internal Medicine, Division of Infectious Diseases, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| | - Daniel S Hazlett
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| | - Rebecca T Horvat
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Rachael M Liesman
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Molly E Steed
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| |
Collapse
|
30
|
Pharmacodynamics of ClpP-Activating Antibiotic Combinations against Gram-Positive Pathogens. Antimicrob Agents Chemother 2019; 64:AAC.01554-19. [PMID: 31611348 DOI: 10.1128/aac.01554-19] [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: 08/06/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022] Open
Abstract
It is often difficult to cure endocarditis, osteomyelitis, and device-associated infections caused by Gram-positive pathogens, despite therapy with clinically appropriate antibiotics. This may be due to antibiotic tolerance or resistance development. Acyldepsipeptides (ADEPs) are a class of bactericidal compounds active against a variety of clinically important Gram-positive bacteria, including staphylococci, streptococci, and enterococci. ADEPs activate caseinolytic protease P (ClpP), killing high-density, nondividing cultures of bacteria that are tolerant to approved classes of antibiotics. Acyldepsipeptide analog 4 (ADEP4) was active against a panel of drug-resistant Gram-positive pathogens in MIC assays, with no preexisting resistance detected. Killing of stationary-phase cultures was observed when ADEP4 was combined with multiple classes of approved antibiotics. Additionally, a hollow-fiber infection model was used to assess the effects of ADEP4 antibiotic combinations on bacterial killing and resistance development. These studies were performed on high-density cultures of methicillin-resistant S. aureus (MRSA), methicillin-susceptible S. aureus (MSSA), and vancomycin-resistant Enterococcus faecalis (VRE). None of the approved antibiotics linezolid, ampicillin, and oxacillin tested alone had bactericidal activity under these conditions. ADEP4 initially caused killing, but regrowth of the culture was apparent within 96 h due to resistance. Combinations of ADEP4 with linezolid or oxacillin caused substantially improved killing of MRSA or MSSA cultures, respectively, and no regrowth due to resistance was observed. The combination of ADEP4 and ampicillin eradicated cultures of VRE to the limit of detection within 52 h. These data suggest that combining ClpP activators with traditional antibiotics may be a good strategy to treat complicated Gram-positive infections.
Collapse
|
31
|
Hornak JP, Anjum S, Reynoso D. Adjunctive ceftaroline in combination with daptomycin or vancomycin for complicated methicillin-resistant Staphylococcus aureus bacteremia after monotherapy failure. Ther Adv Infect Dis 2019; 6:2049936119886504. [PMID: 31857898 PMCID: PMC6915839 DOI: 10.1177/2049936119886504] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/14/2019] [Indexed: 11/30/2022] Open
Abstract
Background: Methicillin-resistant Staphylococcus aureus bacteremia (MRSA-B) may fail to improve with standard monotherapy, particularly in patients with multifocal infection, incomplete source control, or persistent bacteremia. Synergy observed in vitro between ceftaroline (CPT) and daptomycin (DAP) or vancomycin (VAN) may translate into clinical benefit. Here, we describe our experience with DAP/CPT and VAN/CPT for complicated MRSA-B after monotherapy failure. Methods: Single-center, retrospective review of consecutive patients treated with DAP/CPT or VAN/CPT for MRSA-B after monotherapy failure from 1 January 2016 to 30 November 2018. Results: We identified 11 instances of combination therapy in 10 patients (DAP/CPT = 6, VAN/CPT = 5) with 1 patient receiving VAN/CPT followed by DAP/CPT. Rates of multifocal infection, incomplete source control, persistent bacteremia, and infective endocarditis were high (100%, 80%, 60%, and 60%, respectively). Combination therapy was initiated most commonly for persistent bacteremia (60%). When patients were persistently bacteremic, median preceding duration was 13 days and median time to clearance was 3 days. Total microbiologic cure rate was 100%. There were zero instances of bacteremia relapse at 30 days (30D) or 60 days (60D). All-cause 30D and 60D mortality rates were 11.1% and 33.3%, respectively. Conclusions: Combination therapy demonstrated success in diverse cases of refractory MRSA-B, including instances of persistent bacteremia paired with incomplete source control. Optimal timing and therapeutic cadence for combination therapy remain unclear. Our findings suggest that DAP/CPT and VAN/CPT can be considered for complicated MRSA bacteremia when other treatment options fail or are unavailable. We propose persistent bacteremia with incomplete source control to be a clinical niche particularly worthy of further investigation.
Collapse
Affiliation(s)
- Joseph Patrik Hornak
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, USA
| | - Seher Anjum
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - David Reynoso
- The University of Texas Medical Branch, Division of Infectious Diseases, 301 University Blvd., Rte. 0435, Marvin Graves Building 4.210H, Galveston, TX, 77555-0435, USA
| |
Collapse
|
32
|
Di Domenico EG, Rimoldi SG, Cavallo I, D’Agosto G, Trento E, Cagnoni G, Palazzin A, Pagani C, Romeri F, De Vecchi E, Schiavini M, Secchi D, Antona C, Rizzardini G, Dichirico RB, Toma L, Kovacs D, Cardinali G, Gallo MT, Gismondo MR, Ensoli F. Microbial biofilm correlates with an increased antibiotic tolerance and poor therapeutic outcome in infective endocarditis. BMC Microbiol 2019; 19:228. [PMID: 31638894 PMCID: PMC6802308 DOI: 10.1186/s12866-019-1596-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Infective endocarditis (IE) is associated with high rates of mortality. Prolonged treatments with high-dose intravenous antibiotics often fail to eradicate the infection, frequently leading to high-risk surgical intervention. By providing a mechanism of antibiotic tolerance, which escapes conventional antibiotic susceptibility profiling, microbial biofilm represents a key diagnostic and therapeutic challenge for clinicians. This study aims at assessing a rapid biofilm identification assay and a targeted antimicrobial susceptibility profile of biofilm-growing bacteria in patients with IE, which were unresponsive to antibiotic therapy. RESULTS Staphylococcus aureus was the most common isolate (50%), followed by Enterococcus faecalis (25%) and Streptococcus gallolyticus (25%). All microbial isolates were found to be capable of producing large, structured biofilms in vitro. As expected, antibiotic treatment either administered on the basis of antibiogram or chosen empirically among those considered first-line antibiotics for IE, including ceftriaxone, daptomycin, tigecycline and vancomycin, was not effective at eradicating biofilm-growing bacteria. Conversely, antimicrobial susceptibility profile of biofilm-growing bacteria indicated that teicoplanin, oxacillin and fusidic acid were most effective against S. aureus biofilm, while ampicillin was the most active against S. gallolyticus and E. faecalis biofilm, respectively. CONCLUSIONS This study indicates that biofilm-producing bacteria, from surgically treated IE, display a high tolerance to antibiotics, which is undetected by conventional antibiograms. The rapid identification and antimicrobial tolerance profiling of biofilm-growing bacteria in IE can provide key information for both antimicrobial therapy and prevention strategies.
Collapse
Affiliation(s)
- Enea Gino Di Domenico
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Sara Giordana Rimoldi
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Ilaria Cavallo
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Giovanna D’Agosto
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Elisabetta Trento
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Giovanni Cagnoni
- UOC Cardiochirurgia, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Alessandro Palazzin
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Cristina Pagani
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Francesca Romeri
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Elena De Vecchi
- Laboratory of Clinical Chemistry and Microbiology, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy
| | - Monica Schiavini
- Dipartimento di Malattie Infettive, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Daniela Secchi
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Carlo Antona
- UOC Cardiochirurgia, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Giuliano Rizzardini
- Dipartimento di Malattie Infettive, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Rita Barbara Dichirico
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Luigi Toma
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Daniela Kovacs
- Cutaneous Physiopathology Lab, San Gallicano Dermatologic Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Giorgia Cardinali
- Cutaneous Physiopathology Lab, San Gallicano Dermatologic Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Maria Teresa Gallo
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| | - Maria Rita Gismondo
- Laboratorio di Microbiologia Clinica, Virologia e Diagnostica delle Bioemergenze, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, Polo Universitario, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology, San Gallicano Dermatology Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), via Elio Chianesi, 53 00144 Rome, Italy
| |
Collapse
|
33
|
Strain-Specific Metabolic Requirements Revealed by a Defined Minimal Medium for Systems Analyses of Staphylococcus aureus. Appl Environ Microbiol 2019; 85:AEM.01773-19. [PMID: 31471305 DOI: 10.1128/aem.01773-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/26/2019] [Indexed: 01/08/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive pathogenic bacterium that colonizes an estimated one-third of the human population and can cause a wide spectrum of disease, ranging from superficial skin infections to life-threatening sepsis. The adaptive mechanisms that contribute to the success of this pathogen remain obscure partially due to a lack of knowledge of its metabolic requirements. Systems biology approaches can be extremely useful in predicting and interpreting metabolic phenotypes; however, such approaches rely on a chemically defined minimal medium as a basis to investigate the requirements of the cell. In this study, a chemically defined minimal medium formulation, termed synthetic minimal medium (SMM), was investigated and validated to support growth of three S. aureus strains: LAC and TCH1516 (USA300 lineage), as well as D592 (USA100 lineage). The formulated SMM was used in an adaptive laboratory evolution experiment to probe the various mutational trajectories of all three strains leading to optimized growth capabilities. The evolved strains were phenotypically characterized for their growth rate and antimicrobial susceptibility. Strains were also resequenced to examine the genetic basis for observed changes in phenotype and to design follow-up metabolite supplementation assays. Our results reveal evolutionary trajectories that arose from strain-specific metabolic requirements. SMM and the evolved strains can also serve as important tools to study antibiotic resistance phenotypes of S. aureus IMPORTANCE As researchers try to understand and combat the development of antibiotic resistance in pathogens, there is a growing need to thoroughly understand the physiology and metabolism of the microbes. Staphylococcus aureus is a threatening pathogen with increased antibiotic resistance and well-studied virulence mechanisms. However, the adaptive mechanisms used by this pathogen to survive environmental stresses remain unclear, mostly due to the lack of information about its metabolic requirements. Defining the minimal metabolic requirements for S. aureus growth is a first step toward unraveling the mechanisms by which it adapts to metabolic stresses. Here, we present the development of a chemically defined minimal medium supporting growth of three S. aureus strains, and we reveal key genetic mutations contributing to improved growth in minimal medium.
Collapse
|
34
|
Brooks Peterson M, Cohen MN, O'Neill BR, Garg S, Child J, Henthorn TK, Galinkin JG. Preoperative Vancomycin Administration for Surgical Site Prophylaxis: Plasma and Soft-Tissue Concentrations in Pediatric Neurosurgical and Orthopedic Patients. Anesth Analg 2019; 130:1435-1444. [PMID: 31397701 DOI: 10.1213/ane.0000000000004340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Vancomycin is used for antibiotic prophylaxis in pediatric surgical patients without a complete understanding of plasma and soft-tissue pharmacokinetics. Guidelines recommend incision within 60 minutes after administration; however, tissue vancomycin concentrations at that early time may not be therapeutic. We conducted a study of plasma and skin concentrations in pediatric neurosurgical and orthopedic patients to characterize intraoperative vancomycin pharmacokinetics. METHODS Patients (0.1-18.8 years of age) undergoing posterior spinal fusion (n = 30) or ventriculoperitoneal shunt placement (n = 30) received intravenous vancomycin 15 mg/kg (maximum 1000 mg) over 1 hour. Skin was biopsied at incision and skin closure. Blood samples were collected at incision, at 2 and 4 hours intraoperatively, and at closure. Population pharmacokinetic analysis was performed to characterize pharmacokinetic parameter estimates and to develop a model of intraoperative plasma and skin vancomycin concentrations versus time. RESULTS Pharmacokinetic analysis included data from 59 subjects, 130 plasma samples, and 107 skin samples. A 2-compartment model, volume of the central (Vc) and volume of the peripheral compartment (V2), proved to have the best fit. Stepwise covariate selection yielded a significant relationship for body surface area on elimination clearance and body weight on V2. Skin vancomycin concentrations rose continuously during surgery. Modeling predicted that equilibration of skin and plasma vancomycin concentrations took ≥5 hours. CONCLUSIONS Skin vancomycin concentrations immediately after a preoperative dose are relatively low compared with concentrations at the end of surgery. It may be advisable to extend the time between dose and incision if higher skin concentrations are desired at the start of surgery.
Collapse
Affiliation(s)
| | | | | | - Sumeet Garg
- Department of Orthopedic Surgery, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Jason Child
- Department of Pharmacy, Children's Hospital Colorado, Aurora, Colorado
| | - Thomas K Henthorn
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
35
|
Tellis M, Joseph J, Khande H, Bhagwat S, Patel M. In vitro bactericidal activity of levonadifloxacin (WCK 771) against methicillin- and quinolone-resistant Staphylococcus aureus biofilms. J Med Microbiol 2019; 68:1129-1136. [DOI: 10.1099/jmm.0.000999] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Melroy Tellis
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Jiji Joseph
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Hemant Khande
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Sachin Bhagwat
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Mahesh Patel
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Skin and soft-tissue infections (SSIs) are among the commonest infections encountered in clinical practice. Spread of methicillin-resistant Staphylococcus aureus SSIs continues to increase in both health care and community settings and presents a challenge for the best treatment choice. Vancomycin has been the mainstay of SSIs treatment, but recently its use has been questioned because of concerns about its efficacy, tolerability, and unfavorable pharmacokinetic/pharmacodynamic profile. The purpose of this review is to establish the current role for vancomycin in light of the literature published from January 2007 to September 2017 on comparison with both old and new alternatives. RECENT FINDINGS Meta-analyses show better clinical and microbiological outcomes for drugs approved for the treatment of SSI, including those sustained by methicillin-resistant S. aureus, in the last 10 years than for vancomycin. The newer glycopeptides and linezolid decrease the total treatment costs compared with vancomycin, by reducing the length of stay or avoiding the hospitalization. SUMMARY Vancomycin is noninferior in efficacy and safety to all comparator drugs, including the newest on the market. However, the SSI treatment evidence base presents several shortcomings limiting the clinical applicability of the results. High-level clinical trials should be performed to obtain results that can be generalized and applied effectively in clinical practice.
Collapse
|
37
|
Luther MK, Mermel LA, LaPlante KL. Comparison of linezolid and vancomycin lock solutions with and without heparin against biofilm-producing bacteria. Am J Health Syst Pharm 2019; 74:e193-e201. [PMID: 28438824 DOI: 10.2146/ajhp150804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The activity of linezolid and vancomycin lock solutions against biofilm-producing strains of Staphylococcus aureus, S. epidermidis, and Enterococcus faecalis was studied. METHODS Two strains each of methicillin-susceptible S. aureus (MSSA), methicillin-resistant S. aureus (MRSA), and S. epidermidis, and 1 strain of vancomycin-susceptible E. faecalis and vancomycin-resistant E. faecalis were tested against vancomycin and linezolid to assess prevention of biofilm formation and eradication of these pathogens within a formed biofilm. Activity was also tested in a 72-hour in vitro central venous catheter (CVC) model. After 24 hours of biofilm growth in a CVC, a lock solution containing vancomycin (2 or 5 mg/mL) or linezolid (1 or 2 mg/mL) alone or in combination with heparin sodium (5,000 units/mL with benzyl alcohol 0.45%) was instilled and incubated at 35 °C for 72 hr. Heparin and 0.9% sodium chloride injection were also tested. RESULTS Linezolid and vancomycin prevented biofilm formation below the minimum inhibitory concentration for 88% and 25% of isolates tested, respectively. The addition of preservative-containing heparin decreased the activity of vancomycin and linezolid lock solutions against all strains. Vancomycin 2- and 5-mg/mL lock solutions had the most activity against MSSA and E. faecalis strains (p < 0.01). Linezolid 2 mg/mL was the most active lock solution against the MRSA strains tested (p < 0.01). There were no significant differences in vancomycin or linezolid lock solution activity against S. epidermidis. CONCLUSION Heparin reduced activity of vancomycin and linezolid lock solutions against S. aureus, S. epidermidis, and E. faecalis biofilms. While linezolid or vancomycin lock solution reduced overall biofilm burden, it did not completely eradicate the bacteria at tested concentrations.
Collapse
Affiliation(s)
- Megan K Luther
- Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.,Department of Pharmacy Practice, University of Rhode Island, Kingston, RI
| | - Leonard A Mermel
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI.,Division of Infectious Diseases, Rhode Island Hospital, Providence, RI
| | - Kerry L LaPlante
- Department of Pharmacy Practice, University of Rhode Island, Kingston, RI .,Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.
| |
Collapse
|
38
|
Daptomycin Dose-Ranging Evaluation with Single-Dose versus Multidose Ceftriaxone Combinations against Streptococcus mitis /oralis in an Ex Vivo Simulated Endocarditis Vegetation Model. Antimicrob Agents Chemother 2019; 63:AAC.00386-19. [PMID: 30962347 DOI: 10.1128/aac.00386-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
The viridans group streptococci (VGS) are a heterogeneous group of organisms which are important components of the normal human oral flora. Among the VGS, the Streptococcus mitis /oralis subgroup is one of the most common causes of infective endocarditis (IE). Daptomycin (DAP) is a potential alternative therapeutic option for invasive S. mitis infections, given high rates of β-lactam resistance and vancomycin tolerance in such strains. However, the ability of these strains to rapidly evolve high-level and durable DAP resistance (DAP-R) is problematic. Recent data suggest that combination DAP-β-lactam therapy circumvents this issue. Human-simulated dose-escalating DAP-alone dose regimens (6, 8, 10, or 12 mg/kg/day times 4 days) versus DAP (6 mg/kg/day) plus ceftriaxone (CRO) (2 g once daily times 4 days or 0.5 g, single dose) were assessed against two prototypical DAP-susceptible (DAP-S) S. mitis /oralis strains (SF100 and 351), as measured by a pharmacokinetic/pharmacodynamic (PK/PD) model of simulated endocardial vegetations (SEVs). No DAP-alone regimen was effective, with regrowth of high-level DAP-R isolates observed for both strains over 96-h exposures. Combinations of DAP-CRO with either single- or multidose regimens yielded significant reductions in log10 CFU/g amounts within SEVs for both strains (∼6 log10 CFU/g) within 24 h. In addition, no DAP-R strains were detected in either DAP-CRO combination regimens over the 96-h exposure. In contrast to prior in vitro studies, no perturbations in two key cardiolipin biosynthetic genes (cdsA and pgsA) were identified in DAP-R SEV isolates emerging from strain 351, despite defective phospholipid production. The combination of DAP-CRO warrants further investigation for treatment of IE due to S. mitis /oralis.
Collapse
|
39
|
Broussou DC, Toutain PL, Woehrlé F, El Garch F, Bousquet-Melou A, Ferran AA. Comparison of in vitro static and dynamic assays to evaluate the efficacy of an antimicrobial drug combination against Staphylococcus aureus. PLoS One 2019; 14:e0211214. [PMID: 30673774 PMCID: PMC6344103 DOI: 10.1371/journal.pone.0211214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022] Open
Abstract
An easily implementable strategy to reduce treatment failures in severe bacterial infections is to combine already available antibiotics. However, most in vitro combination assays are performed by exposing standard bacterial inocula to constant concentrations of antibiotics over less than 24h, which can be poorly representative of clinical situations. The aim of this study was to assess the ability of static and dynamic in vitro Time-Kill Studies (TKS) to identify the potential benefits of an antibiotic combination (here, amikacin and vancomycin) on two different inoculum sizes of two S. aureus strains. In the static TKS (sTKS), performed by exposing both strains over 24h to constant antibiotic concentrations, the activity of the two drugs combined was not significantly different the better drug used alone. However, the dynamic TKS (dTKS) performed over 5 days by exposing one strain to fluctuating concentrations representative of those observed in patients showed that, with the large inoculum, the activities of the drugs, used alone or in combination, significantly differed over time. Vancomycin did not kill bacteria, amikacin led to bacterial regrowth whereas the combination progressively decreased the bacterial load. Thus, dTKS revealed an enhanced effect of the combination on a large inoculum not observed in sTKS. The discrepancy between the sTKS and dTKS results highlights that the assessment of the efficacy of a combination for severe infections associated with a high bacterial load could be demanding. These situations probably require the implementation of dynamic assays over the entire expected treatment duration rather than the sole static assays performed with steady drug concentrations over 24h.
Collapse
Affiliation(s)
- Diane C. Broussou
- UMR 1436 INTHERES, Université de Toulouse, INRA, ENVT, Toulouse, France
- Vetoquinol SA, Lure, France
| | - Pierre-Louis Toutain
- Department of Veterinary Basics Sciences, Royal Veterinary College, London, United Kingdom
| | | | | | | | - Aude A. Ferran
- UMR 1436 INTHERES, Université de Toulouse, INRA, ENVT, Toulouse, France
- * E-mail:
| |
Collapse
|
40
|
Kidd JM, Abdelraouf K, Nicolau DP. Comparative efficacy of human-simulated epithelial lining fluid exposures of tedizolid, linezolid and vancomycin in neutropenic and immunocompetent murine models of staphylococcal pneumonia. J Antimicrob Chemother 2018; 74:970-977. [DOI: 10.1093/jac/dky513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 02/04/2023] Open
Affiliation(s)
- James M Kidd
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Kamilia Abdelraouf
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
41
|
Zinner SH, Golikova MV, Strukova EN, Portnoy YA, Firsov AA. Predicting antibiotic combination effects on the selection of resistant Staphylococcus aureus: In vitro model studies with linezolid and gentamicin. Int J Antimicrob Agents 2018; 52:854-860. [DOI: 10.1016/j.ijantimicag.2018.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/21/2018] [Accepted: 09/09/2018] [Indexed: 12/01/2022]
|
42
|
Use of exogenous volatile organic compounds to detect Salmonella in milk. Anal Chim Acta 2018; 1028:121-130. [DOI: 10.1016/j.aca.2018.03.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/26/2018] [Accepted: 03/30/2018] [Indexed: 01/10/2023]
|
43
|
Maslivetc VA, Turner DN, McNair KN, Frolova L, Rogelj S, Maslivetc AA, Aksenov NA, Rubina M, Rubin M. Desymmetrization of Cyclopropenes via the Potassium-Templated Diastereoselective 7- exo- trig Cycloaddition of Tethered Amino Alcohols toward Enantiopure Cyclopropane-Fused Oxazepanones with Antimycobacterial Activity. J Org Chem 2018; 83:5650-5664. [PMID: 29696970 DOI: 10.1021/acs.joc.8b00640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A strain-release-driven, cation-templated intramolecular nucleophilic addition of tethered alkoxides to prochiral cyclopropenes is described. Employment of chiral β- and γ-amino alkoxides allowed for highly diastereoselective assembly of a small series of enantiopure cyclopropane-fused oxazepanones. It was shown that the chiral center at C-4 plays a crucial role in controlling desymmetrization of the cyclopropenyl moiety, instigated by a profound potassium-templated effect. The preliminary biological activities of the new cyclopropane-fused medium heterocycles against Gram-positive bacteria, Gram-negative bacteria, mycobacteria, cancer cells, and fungus were evaluated.
Collapse
Affiliation(s)
- Vladimir A Maslivetc
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045-7582 , United States
| | - Danielle N Turner
- Departments of Chemistry and Biology , New Mexico Institute of Mining and Technology , Socorro , New Mexico 87801 , United States
| | - Kimberly N McNair
- Departments of Chemistry and Biology , New Mexico Institute of Mining and Technology , Socorro , New Mexico 87801 , United States
| | - Liliya Frolova
- Departments of Chemistry and Biology , New Mexico Institute of Mining and Technology , Socorro , New Mexico 87801 , United States
| | - Snezna Rogelj
- Departments of Chemistry and Biology , New Mexico Institute of Mining and Technology , Socorro , New Mexico 87801 , United States
| | - Anna A Maslivetc
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045-7582 , United States
| | - Nicolai A Aksenov
- Department of Chemistry , North Caucasus Federal University , 1a Pushkin St. , Stavropol 355009 , Russian Federation
| | - Marina Rubina
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045-7582 , United States
| | - Michael Rubin
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045-7582 , United States.,Department of Chemistry , North Caucasus Federal University , 1a Pushkin St. , Stavropol 355009 , Russian Federation
| |
Collapse
|
44
|
Combination of Tedizolid and Daptomycin against Methicillin-Resistant Staphylococcus aureus in an In Vitro Model of Simulated Endocardial Vegetations. Antimicrob Agents Chemother 2018. [PMID: 29530843 DOI: 10.1128/aac.00101-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major pathogen responsible for health care-associated infections, and treatment options are limited. Tedizolid (TZD) is a novel oxazolidinone antibiotic with activity against MRSA. Previously, daptomycin (DAP) has demonstrated synergy with other antibiotics against MRSA. We sought to determine the efficacy of the combination of TZD and DAP against MRSA in an in vitro model of simulated endocardial vegetations (SEVs). TZD simulations of 200 mg once daily and DAP simulations of 6 mg/kg of body weight and 10 mg/kg once daily were tested alone and in the combinations TZD plus DAP at 6 mg/kg or DAP at 10 mg/kg against two clinical strains of MRSA, 494 and 67. These regimens were tested in SEV models over 8 days to determine the antibacterial activity of the regimens and whether synergy or antagonism might be present between the agents. Against both strains 494 and 67 and at both DAP dose regimens, the combination of TZD and DAP was antagonistic at 192 h. In all cases, DAP alone was statistically superior to DAP plus TZD. When the combination was stopped after 96 h, transitioning to DAP at 6 mg/kg or DAP at 10 mg/kg alone resulted in better antibacterial activity than either of the TZD-plus-DAP combinations, further demonstrating antagonistic effects. Against MRSA, we demonstrated that TZD and DAP have antagonistic activity that hinders their overall antimicrobial efficacy. The exact nature of this antagonistic relationship is still undetermined, but its presence warrants further study of the potentially harmful grouping of the two antibiotics in clinical use.
Collapse
|
45
|
Broussou DC, Lacroix MZ, Toutain PL, Woehrlé F, El Garch F, Bousquet-Melou A, Ferran AA. Differential Activity of the Combination of Vancomycin and Amikacin on Planktonic vs. Biofilm-Growing Staphylococcus aureus Bacteria in a Hollow Fiber Infection Model. Front Microbiol 2018; 9:572. [PMID: 29636741 PMCID: PMC5880918 DOI: 10.3389/fmicb.2018.00572] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/13/2018] [Indexed: 11/13/2022] Open
Abstract
Combining currently available antibiotics to optimize their use is a promising strategy to reduce treatment failures against biofilm-associated infections. Nevertheless, most assays of such combinations have been performed in vitro on planktonic bacteria exposed to constant concentrations of antibiotics over only 24 h and the synergistic effects obtained under these conditions do not necessarily predict the behavior of chronic clinical infections associated with biofilms. To improve the predictivity of in vitro combination assays for bacterial biofilms, we first adapted a previously described Hollow-fiber (HF) infection model by allowing a Staphylococcus aureus biofilm to form before drug exposure. We then mimicked different concentration profiles of amikacin and vancomycin, similar to the free plasma concentration profiles that would be observed in patients treated daily over 5 days. We assessed the ability of the two drugs, alone or in combination, to reduce planktonic and biofilm-embedded bacterial populations, and to prevent the selection of resistance within these populations. Although neither amikacin nor vancomycin exhibited any bactericidal activity on S. aureus in monotherapy, the combination had a synergistic effect and significantly reduced the planktonic bacterial population by -3.0 to -6.0 log10 CFU/mL. In parallel, no obvious advantage of the combination, as compared to amikacin alone, was demonstrated on biofilm-embedded bacteria for which the addition of vancomycin to amikacin only conferred a further maximum reduction of 0.3 log10 CFU/mL. No resistance to vancomycin was ever found whereas a few bacteria less-susceptible to amikacin were systematically detected before treatment. These resistant bacteria, which were rapidly amplified by exposure to amikacin alone, could be maintained at a low level in the biofilm population and even suppressed in the planktonic population by adding vancomycin. In conclusion, by adapting the HF model, we were able to demonstrate the different bactericidal activities of the vancomycin and amikacin combination on planktonic and biofilm-embedded bacterial populations, suggesting that, for biofilm-associated infections, the efficacy of this combination would not be much greater than with amikacin monotherapy. However, adding vancomycin could reduce possible resistance to amikacin and provide a relevant strategy to prevent the selection of antibiotic-resistant bacteria during treatments.
Collapse
Affiliation(s)
- Diane C Broussou
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France.,Vétoquinol, Global Drug Development, Lure, France
| | | | - Pierre-Louis Toutain
- Department of Veterinary Basic Sciences, Royal Veterinary College, London, United Kingdom
| | | | | | | | - Aude A Ferran
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France
| |
Collapse
|
46
|
Khan A, Wilson B, Gould IM. Current and future treatment options for community-associated MRSA infection. Expert Opin Pharmacother 2018; 19:457-470. [PMID: 29480032 DOI: 10.1080/14656566.2018.1442826] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Community-associated MRSA (CA-MRSA) represents a global epidemic which beautifully encapsulates the fascinating ability of bacterial organisms to adapt quickly on an evolutionary basis to the extreme selective pressure of antibiotic exposure. In stark contrast to Healthcare-associated MRSA (HA-MRSA), it has become apparent that CA-MRSA is less straight forward of a challenge in terms of controlling its transmission, and has forced clinicians to adjust empiric management of clinical syndromes such as skin and soft tissue infection (SSTI) as well as pneumonia. AREAS COVERED This review details the history and epidemiology of CA-MRSA, while covering both current and future treatment options that are and may be available to clinicians. The authors reviewed both historic and more recent literature on this ever-evolving topic. EXPERT OPINION While development of new anti-MRSA agents should be encouraged, the importance of antimicrobial stewardship in the battle to stay ahead of the curve with regards to the ongoing control of the MRSA epidemic should be emphasised.
Collapse
Affiliation(s)
- A Khan
- a Department of Medical Microbiology , Aberdeen Royal Infirmary (ARI) , Aberdeen , Scotland
| | - B Wilson
- a Department of Medical Microbiology , Aberdeen Royal Infirmary (ARI) , Aberdeen , Scotland
| | - I M Gould
- a Department of Medical Microbiology , Aberdeen Royal Infirmary (ARI) , Aberdeen , Scotland
| |
Collapse
|
47
|
Kato H, Hagihara M, Murakami E, Suematsu H, Nishiyama N, Koizumi Y, Yamagishi Y, Uno B, Mikamo H. Considerations about the Use of a Loading Dose of Daptomycin in a Neutropenic Murine Thigh Infection Model with Methicillin-Resistant Staphylococcus aureus Infection. Chemotherapy 2017; 63:13-19. [DOI: 10.1159/000481158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/31/2017] [Indexed: 11/19/2022]
Abstract
Previous clinical studies have showed the clinical benefits of the initiation of treatment with a daptomycin (DAP) loading dose, but only a few studies have evaluated its antimicrobial benefits. We evaluated the efficacy of a DAP loading dose against methicillin-resistant Staphylococcus aureus (MRSA) infections in a neutropenic murine thigh infection model. Three MRSA isolates (DAP MIC: 0.5, 1, and 2 mg/L) were tested. Four DAP regimens simulating human concentration-time profiles, i.e., (i) day 1: 8 mg/kg and day 2: 6 mg/kg, (ii) days 1 and 2: 6 mg/kg/day, (iii) day 1: 8 mg/kg and day 2: 4 mg/kg, and (iv) days 1 and 2: 4 mg/kg/day, were administered to the mice. Efficacy was calculated as the change in bacterial density. DAP loading-dose regimen iii showed greater antimicrobial activity against MRSA with MIC 1 mg/L than nonloading regimen iv (-3.10 ± 0.63 vs. -0.71 ± 0.34 log10 CFU; p < 0.01). Loading-dose regimen iii achieved greater log10 CFU changes than nonloading regimen ii, while the total DAP dose for 2 days was the same (-3.10 ± 0.63 vs. -1.46 ± 0.48 log10 CFU; p < 0.05). DAP loading-dose regimen iii showed enhanced antimicrobial activity against MRSA with DAP MIC 0.5 mg/L when compared with nonloading regimen iv. However, loading-dose regimens i and iii did not reduce bacterial density for MRSA with DAP MIC 2 mg/L. Our data suggest that a DAP loading-dose regimen would be an advantageous procedure for patients infected with MRSA with DAP MIC ≤1 mg/L.
Collapse
|
48
|
AngioVac System Used for Vegetation Debulking in a Patient with Tricuspid Valve Endocarditis: A Case Report and Review of the Literature. Case Rep Cardiol 2017; 2017:1923505. [PMID: 29238620 PMCID: PMC5697122 DOI: 10.1155/2017/1923505] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022] Open
Abstract
AngioVac is a vacuum-based device approved in 2014 for percutaneous removal of undesirable materials from the intravascular system. Although numerous reports exist with regard to the use of the AngioVac device in aspiration of iliocaval, pulmonary, upper extremity, and right-sided heart chamber thrombi, very few data are present demonstrating its use in treatment of right-sided endocarditis. In this case report, we describe the novel device used in debulking a large right-sided tricuspid valve vegetation reducing the occurrence of septic embolisation and enhancing the efficacy of antibiotics in clearance of bloodstream infection. Further research is needed in larger RSIE patient populations to confirm the benefits and the potential of improved outcomes associated with the AngioVac device as well as identify its potential complications.
Collapse
|
49
|
Firsov AA, Alieva KN, Strukova EN, Golikova MV, Portnoy YA, Dovzhenko SA, Kobrin MB, Romanov AV, Edelstein MV, Zinner SH. Testing the mutant selection window hypothesis with Staphylococcus aureus exposed to linezolid in an in vitro dynamic model. J Antimicrob Chemother 2017; 72:3100-3107. [DOI: 10.1093/jac/dkx249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
|
50
|
|