51
|
Yao Z, Wu Y, Xu H, Lei Y, Long W, Li M, Gu Y, Jiang Z, Cao C. Prevalence and clinical characteristics of methicillin-resistant Staphylococcus aureus infections among dermatology inpatients: A 7-year retrospective study at a tertiary care center in southwest China. Front Public Health 2023; 11:1124930. [PMID: 36998271 PMCID: PMC10043400 DOI: 10.3389/fpubh.2023.1124930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/16/2023] Open
Abstract
BackgroundIncreased rates of methicillin-resistant Staphylococcus aureus (MRSA) antibiotic resistance and the associated morbidity have increased dermatologists' attention to skin and soft tissue MRSA infections. However, the clinical characterization of MRSA skin and soft tissue infections (SSTIs) in Southwest China is lacking, which precludes optimal prevention and treatment of these infections.ObjectivesThis study was conducted to characterize the prevalence, clinical comorbidities and antibiotic susceptibility of MRSA isolates from SSTIs, including community-associated (CA) and healthcare-associated (HA) isolates.MethodsIn the Dermatology Inpatient Department of the First Affiliated Hospital of Guangxi Medical University, a retrospective study was conducted on data, including patient demographics and clinical information, from culture-confirmed S. aureus isolated from skin and soft tissue between January 1, 2015, and December 31, 2021. Isolate susceptibility to 13 antibiotics was determined using the Vitek 2 system.ResultsFrom among 864 S. aureus strains, we identified 283 MRSA (32.75%) isolates comprising 203 CA-MRSA and 80 HA-MRSA isolates. The average rate of CA-MRSA isolation for MRSA SSTIs was 71.73%. The HA-MRSA isolation rate for MRSA SSTIs increased significantly. HA-MRSA-infected patients were older. The most common dermatological presentation of CA-MRSA infection was staphylococcal scalded skin syndrome, while the comorbidity severe drug eruption was significantly associated with HA-MRSA infection. One CA-MRSA strain was resistant to linezolid, and one HA-MRSA strain had an intermediate phenotype for vancomycin; both strains had low sensitivity to clindamycin and erythromycin (3.70%~19.40%). However, HA-MRSA isolates were more susceptible to trimethoprim/sulfamethoxazole.ConclusionsCA-MRSA is a predominant pathogen causing SSTIs, and HA-MRSA infection incidence is increasing gradually. Both strains showed increasing antibiotic resistance. Our data on MRSA susceptibility may guide dermatologist antibiotic treatment decisions. Dermatologists should consider these identified comorbidities of MRSA SSTIs when patients are admitted and initiate early prevention and treatment of MRSA.
Collapse
Affiliation(s)
- Zhijian Yao
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Mycosis Prevention and Treatment, Nanning, Guangxi, China
| | - Yidan Wu
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Mycosis Prevention and Treatment, Nanning, Guangxi, China
| | - Hongming Xu
- Department of Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Ying Lei
- Department of Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Wanyu Long
- Department of Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Meixian Li
- Department of Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Gu
- Department of Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhiwen Jiang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Mycosis Prevention and Treatment, Nanning, Guangxi, China
| | - Cunwei Cao
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Mycosis Prevention and Treatment, Nanning, Guangxi, China
- *Correspondence: Cunwei Cao
| |
Collapse
|
52
|
Population Pharmacokinetic and Pharmacokinetic/Pharmacodynamic Target Attainment Analyses for Dalbavancin in Pediatric Patients. Pediatr Infect Dis J 2023; 42:99-105. [PMID: 36638392 PMCID: PMC9835671 DOI: 10.1097/inf.0000000000003764] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Dalbavancin, approved for the treatment of pediatric and adult patients with acute bacterial skin and skin structure infections, has a terminal half-life of >14 days allowing administration as a single-dose regimen. METHODS We developed a population pharmacokinetic (PK) model using 1124 dalbavancin concentrations from 211 pediatric patients, with allometric scaling of clearance and volume parameter exponents fixed at 0.75 and 1, respectively. Serum albumin was included as a covariate on all PK parameters; creatinine clearance or estimated glomerular filtration rate was a covariate on clearance. The final model, qualified by visual predictive checks and bootstrapping, was used to simulate 1000 PK profiles for a range of pediatric age groups. PK/pharmacodynamic target attainment (PTA) was calculated for targets associated with stasis, 1-log kill, and 2-log kill of Staphylococcus aureus (neutropenic murine thigh infection model). RESULTS Dalbavancin PK was well characterized by a three-compartment model. No additional significant covariates were identified. Simulations showed that single-dose (30-minute intravenous infusion) regimens of 22.5 mg/kg (patients <6 years) and 18 mg/kg (patients 6 years to <18 years) resulted in PTA ≥94% for minimal inhibitory concentrations ≤2 mg/L and ≤0.5 mg/L for the stasis and 2-log kill targets, respectively. PTA for pediatric patients was similar to adults with exposures within the range for adults administered 1500 mg dalbavancin. CONCLUSION Dalbavancin PK in pediatric patients was well characterized by a three-compartment model. Simulations with the final model demonstrated adequate PTA across the entire age range for the approved pediatric dalbavancin doses.
Collapse
|
53
|
Makwela AB, Grootboom WM, Abraham V, Witika B, Godman B, Skosana PP. Antimicrobial Management of Skin and Soft Tissue Infections among Surgical Wards in South Africa: Findings and Implications. Antibiotics (Basel) 2023; 12:antibiotics12020275. [PMID: 36830186 PMCID: PMC9951966 DOI: 10.3390/antibiotics12020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Skin and soft tissue infections (SSTIs) are one of the most common infectious diseases requiring antibiotics. However, complications of SSTIs may lead to the overprescribing of antibiotics and to subsequent antibiotic resistance. Consequently, monitoring the prescribing alignment with the current recommendations from the South African Standard Treatment Guidelines (STG) is necessary in order to improve future care. This study involved reviewing pertinent patients with SSTIs who were prescribed antimicrobials in the surgical ward of a leading South African tertiary public hospital from April to June 2021 using an adapted data collection tool. Sixty-seven patient files were reviewed. Among the patients with SSTIs, hypertension and chronic osteomyelitis were the most frequent co-morbidities at 22.4% and 13.4%, respectively. The most diagnosed SSTIs were surgical site infections (35.1%), wound site infections (23%), and major abscesses (16.2%). Blood cultures were performed on 40.3% of patients, with Staphylococcus aureus (32.7%) and Enterococcus spp. (21.2%) being the most cultured pathogens. Cefazolin was prescribed empirically for 46.3% of patients for their SSTIs. In addition, SSTIs were treated with gentamycin, ciprofloxacin, and rifampicin at 17.5%, 11.3%, and 8.8%, respectively, with treatment fully complying with STG recommendations in 55.2% of cases. Overall, the most common cause of SSTIs was Staphylococcus aureus, and empiric treatment is recommended as the initial management. Subsequently, culture sensitivities should be performed to enhance adherence to STGs and to improve future care.
Collapse
Affiliation(s)
- Atlanta B. Makwela
- Department of Clinical Pharmacy, School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
| | - Wandisile M. Grootboom
- Department of Clinical Pharmacy, School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
- Dr George Mukhari Academic Hospital, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
| | - Veena Abraham
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
| | - Bwalya Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
| | - Brian Godman
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Correspondence: (B.G.); (P.P.S.)
| | - Phumzile P. Skosana
- Department of Clinical Pharmacy, School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Ga-Rankuwa, Pretoria 0208, South Africa
- Correspondence: (B.G.); (P.P.S.)
| |
Collapse
|
54
|
Teymournejad O, Li Z, Beesetty P, Yang C, Montgomery CP. Toxin expression during Staphylococcus aureus infection imprints host immunity to inhibit vaccine efficacy. NPJ Vaccines 2023; 8:3. [PMID: 36693884 PMCID: PMC9873725 DOI: 10.1038/s41541-022-00598-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/05/2022] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus infections are a major public health issue, and a vaccine is urgently needed. Despite a considerable promise in preclinical models, all vaccines tested thus far have failed to protect humans against S. aureus. Unlike laboratory mice, humans are exposed to S. aureus throughout life. In the current study, we hypothesized that prior exposure to S. aureus "imprints" the immune response to inhibit vaccine-mediated protection. We established a mouse model in which S. aureus skin and soft tissue infection (SSTI) is followed by vaccination and secondary SSTI. Unlike naïve mice, S. aureus-sensitized mice were incompletely protected against secondary SSTI by vaccination with the inactivated α-hemolysin (Hla) mutant HlaH35L. Inhibition of protection was specific for the HlaH35L vaccine and required hla expression during primary SSTI. Surprisingly, inhibition occurred at the level of vaccine-elicited effector T cells; hla expression during primary infection limited the expansion of T cells and dendritic cells and impaired vaccine-specific T cell responses. Importantly, the T cell-stimulating adjuvant CAF01 rescued inhibition and restored vaccine-mediated protection. Together, these findings identify a potential mechanism for the failure of translation of promising S. aureus vaccines from mouse models to clinical practice and suggest a path forward to prevent these devastating infections.
Collapse
Affiliation(s)
- Omid Teymournejad
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.185648.60000 0001 2175 0319Present Address: Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL US
| | - Zhaotao Li
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US
| | - Pavani Beesetty
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.231844.80000 0004 0474 0428Present Address: Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario Canada
| | - Ching Yang
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.259180.70000 0001 2298 1899Present Address: Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY US
| | - Christopher P. Montgomery
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.261331.40000 0001 2285 7943Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH US ,grid.240344.50000 0004 0392 3476Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH US
| |
Collapse
|
55
|
Deaconu M, Prelipcean AM, Brezoiu AM, Mitran RA, Isopencu G, Matei C, Berger D. Novel Collagen-Polyphenols-Loaded Silica Composites for Topical Application. Pharmaceutics 2023; 15:pharmaceutics15020312. [PMID: 36839635 PMCID: PMC9962153 DOI: 10.3390/pharmaceutics15020312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Lesions can affect skin functions and cause a simple issue, such as dehydration, or more challenging complications, such as bacterial infections. The purpose of this study was to design composites for topical application that can prevent and/or assist in bacterial infections and support cell regeneration using natural components. A polyphenolic extract obtained from Salvia officinalis was embedded in functionalized mesoporous silica nanoparticles for better stability, followed by their distribution into a collagen porous scaffold. The resulting polyphenols-loaded MSN exhibited enhanced antibacterial activity and good cytocompatibility. Improved thermal stability of the collagen porous scaffold was obtained due to the presence of the functionalized MSN. For the first time, collagen-polyphenols-loaded silica composites were reported in the literature as potential wound dressings. The newly developed composites showed excellent sterility.
Collapse
Affiliation(s)
- Mihaela Deaconu
- CAMPUS Research Institute, University “Politehnica” of Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
- Faculty of Chemical Engineering and Biotechnologies, University “Politehnica” of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
| | - Ana-Maria Prelipcean
- National Institute of R&D for Biological Sciences, 296 Splaiul Independetei, 060031 Bucharest, Romania
- Correspondence: (A.-M.P.); (D.B.)
| | - Ana-Maria Brezoiu
- Faculty of Chemical Engineering and Biotechnologies, University “Politehnica” of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
| | - Raul-Augustin Mitran
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Gabriela Isopencu
- Faculty of Chemical Engineering and Biotechnologies, University “Politehnica” of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
| | - Cristian Matei
- Faculty of Chemical Engineering and Biotechnologies, University “Politehnica” of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
| | - Daniela Berger
- Faculty of Chemical Engineering and Biotechnologies, University “Politehnica” of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
- Correspondence: (A.-M.P.); (D.B.)
| |
Collapse
|
56
|
Bacterial Motility and Its Role in Skin and Wound Infections. Int J Mol Sci 2023; 24:ijms24021707. [PMID: 36675220 PMCID: PMC9864740 DOI: 10.3390/ijms24021707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Skin and wound infections are serious medical problems, and the diversity of bacteria makes such infections difficult to treat. Bacteria possess many virulence factors, among which motility plays a key role in skin infections. This feature allows for movement over the skin surface and relocation into the wound. The aim of this paper is to review the type of bacterial movement and to indicate the underlying mechanisms than can serve as a target for developing or modifying antibacterial therapies applied in wound infection treatment. Five types of bacterial movement are distinguished: appendage-dependent (swimming, swarming, and twitching) and appendage-independent (gliding and sliding). All of them allow bacteria to relocate and aid bacteria during infection. Swimming motility allows bacteria to spread from 'persister cells' in biofilm microcolonies and colonise other tissues. Twitching motility enables bacteria to press through the tissues during infection, whereas sliding motility allows cocci (defined as non-motile) to migrate over surfaces. Bacteria during swarming display greater resistance to antimicrobials. Molecular motors generating the focal adhesion complexes in the bacterial cell leaflet generate a 'wave', which pushes bacterial cells lacking appendages, thereby enabling movement. Here, we present the five main types of bacterial motility, their molecular mechanisms, and examples of bacteria that utilise them. Bacterial migration mechanisms can be considered not only as a virulence factor but also as a target for antibacterial therapy.
Collapse
|
57
|
Jackson BT, Cluck DB, Henao-Martínez AF, Chastain DB. Kimyrsa and Orbactiv - A Tale of Two Formulations. Drug Des Devel Ther 2023; 17:737-742. [PMID: 36923104 PMCID: PMC10010140 DOI: 10.2147/dddt.s324285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Kimyrsa is a new formulation (NF) of the original formulation of oritavancin ([OF] Orbactiv). Comparatively, the obvious benefit with this product is the shortened infusion time and flexibility with solution compatibility, but otherwise maintains a similar pharmacokinetic and microbiologic profile. At present, the NF lacks significant real-world experience relative to other available lipoglycopeptides and thus its place in therapy remains difficult to predict but would not be expected to be significantly different than its OF.
Collapse
Affiliation(s)
- Brittany T Jackson
- Department of Pharmacy, Mount Sinai Morningside, New York, NY, 10025, USA
| | - David B Cluck
- Department of Pharmacy Practice, Bill Gatton College of Pharmacy East Tennessee State University, Johnson City, TN, 37614, USA
| | - Andrés F Henao-Martínez
- Division of Infectious Diseases, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel B Chastain
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Albany, GA, 31701, USA
| |
Collapse
|
58
|
Đorđevski N, Uba AI, Zengin G, Božunović J, Gašić U, Ristanović E, Ćirić A, Nikolić B, Stojković D. Chemical and Biological Investigations of Allium scorodoprasum L. Flower Extracts. Pharmaceuticals (Basel) 2022; 16:ph16010021. [PMID: 36678518 PMCID: PMC9865742 DOI: 10.3390/ph16010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
This study was designed to investigate the impact of different extraction solvent systems on the chemical composition and biological activities of Allium scorodoprasum L. (Amaryllidaceae)-the medicinal plant that was traditionally used as a remedy in the medieval period in the Balkans. Targeted chemical analysis of nine different extracts was performed by UHPLC(-)HESI-QqQ-MS/MS. Antimicrobial and antibiofilm activities of the extracts were investigated on sixteen clinical isolates of bacteria, yeasts and dermatomycetes, all isolated from infected human skin and corneal formations. Cytotoxicity and wound-healing properties were tested on human immortalized keratinocytes (HaCaT cell line). Antioxidant activity was assessed by six different assays, while beneficial potential against certain neurodegenerative diseases and type 2 diabetes was determined in selected enzyme inhibition assays coupled with molecular modeling. The results showed that the obtained extracts were rich in phenolic compounds, especially flavonoid glycosides such as rutin and kaempferol 3-O-glucoside. All of the extracts showed antimicrobial, wound-healing, antioxidant and anti-enzymatic properties. This study is the first of its kind, linking the medieval medicinal use of wild-growing flowers of A. scorodoprasum with contemporary in vitro scientific approaches.
Collapse
Affiliation(s)
- Nikoleta Đorđevski
- Laboratory of Immunology, Institute of Microbiology, Medical Military Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Abdullahi Ibrahim Uba
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Istanbul, Turkey
| | - Gokhan Zengin
- Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, 42130 Konya, Turkey
| | - Jelena Božunović
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - Uroš Gašić
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - Elizabeta Ristanović
- Laboratory of Immunology, Institute of Microbiology, Medical Military Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Ana Ćirić
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
| | - Biljana Nikolić
- University of Belgrade, Faculty of Biology, Studentski trg 16, 11000 Belgrade, Serbia
| | - Dejan Stojković
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
- Correspondence: ; Tel.: +381-112-078-419
| |
Collapse
|
59
|
Zasowski EJ, Trinh TD, Claeys KC, Dryden M, Shlyapnikov S, Bassetti M, Carnelutti A, Khachatryan N, Kurup A, Pulido Cejudo A, Melo L, Cao B, Rybak MJ. International Validation of a Methicillin-Resistant Staphylococcus aureus Risk Assessment Tool for Skin and Soft Tissue Infections. Infect Dis Ther 2022; 11:2253-2263. [PMID: 36319943 PMCID: PMC9669284 DOI: 10.1007/s40121-022-00712-x] [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: 07/14/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION To promote judicious prescribing of methicillin-resistant Staphylococcus aureus (MRSA)-active therapy for skin and soft tissue infections (SSTI), we previously developed an MRSA risk assessment tool. The objective of this study was to validate this risk assessment tool internationally. METHODS A multicenter, prospective cohort study of adults with purulent SSTI was performed at seven international sites from July 2016 to March 2018. Patient MRSA risk scores were computed as follows: MRSA infection/colonization history (2 points); previous hospitalization, previous antibiotics, chronic kidney disease, intravenous drug use, human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), diabetes with obesity (1 point each). Predictive performance of MRSA surveillance percentage, MRSA risk score, and estimated MRSA probability (surveillance percentage adjusted by risk score) were quantified using the area under the receiver operating characteristic curves (aROC) and compared. Performance characteristics of different risk score thresholds across varying baseline MRSA prevalence were examined. RESULTS Two hundred three patients were included. Common SSTI were wounds (28.6%), abscess (25.1%), and cellulitis with abscess (20.7%). Patients with higher risk scores were more likely to have MRSA (P < 0.001). The MRSA risk score aROC (95%CI) [0.748 (0.678-0.819)] was significantly greater than MRSA surveillance percentage [0.646 (0.569-0.722)] (P = 0.016). Estimated MRSA probability aROC [0.781 (0.716-0.845)] was significantly greater than surveillance percentage (P < 0.001) but not the risk score (P = 0.192). The estimated negative predictive value (NPV) of an MRSA score ≥ 1 (i.e., a score of 0) was greater than 90% when MRSA prevalence was 30% or less. CONCLUSION The MRSA risk score and estimated MRSA probability were significantly more predictive of MRSA compared with surveillance percentage. An MRSA risk score of zero had high predictive value and could help avoid unnecessary empiric MRSA coverage in low-acuity patients. Further study, including impact of such risk assessment tools on prescribing patterns and outcomes are required before implementation.
Collapse
Affiliation(s)
- E. J. Zasowski
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201 USA ,Department of Clinical Sciences, Touro University California College of Pharmacy, Vallejo, CA USA ,Geriatrics, Palliative and Extended Care Service Line, San Francisco Veterans Affairs Health Care System, San Francisco, CA USA ,Department of Clinical Pharmacy, University of California, San Francisco School of Pharmacy, San Francisco, CA USA
| | - T. D. Trinh
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201 USA ,Department of Clinical Pharmacy, University of California, San Francisco School of Pharmacy, San Francisco, CA USA
| | - K. C. Claeys
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201 USA ,Department of Pharmacy Practice and Science, University of Maryland School of Pharmacy, Baltimore, MD USA
| | - M. Dryden
- Royal Hampshire County Hospital, Winchester, UK
| | - S. Shlyapnikov
- I.I. Dzhanelidze Institute of Emergency Medicine, Saint Petersburg, Russia
| | - M. Bassetti
- Department of Health Sciences, Infectious Diseases Clinic, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - A. Carnelutti
- Department of Health Sciences, Infectious Diseases Clinic, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - N. Khachatryan
- Department of Surgery, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - A. Kurup
- Infectious Diseases Care, Mount Elizabeth Hospital, Singapore, Singapore
| | | | - L. Melo
- Hospital Dona Helena, Joinville, Brazil
| | - B. Cao
- Department of Pulmonary and Critical Care Medicine, Centre for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201 USA ,Department of Medicine, Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, MI USA ,Department of Pharmacy Services, Detroit Medical Center, Detroit, MI USA
| |
Collapse
|
60
|
Zinzi D, Vlachaki I, Falla E, Mantopoulos T, Nathwani D. Cost-minimisation analysis of oritavancin for the treatment of acute bacterial skin and skin structure infections from a United Kingdom perspective. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2022; 23:1371-1381. [PMID: 35113269 PMCID: PMC9550763 DOI: 10.1007/s10198-022-01432-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Early discharge (ED) from hospital and outpatient parenteral antibiotic therapy (OPAT) are effective approaches for the management of a range of infections, including acute bacterial skin and skin structure infections (ABSSSI). Strategies that facilitate ED, thereby reducing complications such as healthcare-acquired infection whilst enhancing patient quality of life, are being increasingly adopted in line with good antimicrobial stewardship practice. This study presents a cost-minimisation analysis for the use of oritavancin at ED versus relevant comparators from a National Health Service (NHS) and personal and social services United Kingdom perspective. METHODS A cost-minimisation model considering adult patients with ABSSSI with suspected or confirmed methicillin-resistant Staphylococcus aureus (MRSA) infection, was developed based on publicly available NHS costs, practice guidelines for ABSSSI and clinical expert's opinion. Cost of treatment and treatment days were compared for oritavancin at ED to dalbavancin, teicoplanin, daptomycin and linezolid. RESULTS Following the empiric use of either flucloxacillin or vancomycin in the inpatient setting, oritavancin was compared to OPAT with dalbavancin, teicoplanin and daptomycin, and oral linezolid from day 4 of treatment. Oritavancin at ED reduced treatment duration by 0.8 days and led to cost savings of £281 in comparison to dalbavancin. In comparison to teicoplanin, daptomycin and linezolid, oritavancin reduced treatment duration by 5 days, with marginally higher costs (£446, £137, and £1,434, respectively). CONCLUSION Oritavancin, used to support ED, is associated with lower costs compared with dalbavancin and reduced treatment duration relative to all comparators. Its use would support an ED approach in MRSA ABSSSI management.
Collapse
Affiliation(s)
| | | | - Edel Falla
- EMEA Real World Methods and Evidence Generation, IQVIA Ltd, London, UK
| | - Theo Mantopoulos
- EMEA Real World Methods and Evidence Generation, IQVIA Ltd, Athens, Greece.
| | - Dilip Nathwani
- Medical School, University of Dundee, Dundee, DD19SY, UK
| |
Collapse
|
61
|
Kaul S, Sagar P, Gupta R, Garg P, Priyadarshi N, Singhal NK. Mechanobactericidal, Gold Nanostar Hydrogel-Based Bandage for Bacteria-Infected Skin Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44084-44097. [PMID: 36099413 DOI: 10.1021/acsami.2c10844] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The emergence of multidrug resistant (MDR) microorganisms has led to the development of alternative approaches for providing relief from microbial attacks. The mechano-bactericidal action as a substitute for antimicrobials has become the focus of intensive research. In this work, nanostructure-conjugated hydrogel are explored as a flexible dressing against Staphylococcus aureus (S. aureus)-infected skin wounds. Herein gold nanostars (AuNst) with spike lengths reaching 120 nm are probed for antibacterial action. The bacterial killing of >95% is observed for Pseudomonas aeruginosa (P. aeruginosa) and Escherichia coli (E. coli), while up to 60% for Gram-positive S. aureus. AuNst conjugated hydrogel (AuNst120@H) reduced >80% colonies of P. aeruginosa and E. coli. In comparison, around 35.4% reduction of colonies are obtained for S. aureus. The viability assay confirmed the presence of about 85% of living NIH-3T3 cells when grown with hydrogels. An animal wound model is also developed to assess the efficiency of AuNst120@H. A significant reduction in wound size is observed on the 10th day in AuNst120@H treated animals with fully formed epidermal layers, hair follicles, new blood vessels, and arrector muscles. These findings suggest that novel dressing materials can be developed with antimicrobial nanotextured surfaces.
Collapse
Affiliation(s)
- Sunaina Kaul
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
- Department of Biotechnology, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Poonam Sagar
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
- Department of Biotechnology, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Ritika Gupta
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
- Department of Biotechnology, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Priyanka Garg
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
- Department of Biotechnology, Panjab University, Sector 25, Chandigarh, 160014, India
| | - Nitesh Priyadarshi
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute, Sector 81, Sahibzada Ajit Singh Nagar, Mohali, 140306, India
| |
Collapse
|
62
|
Does a Positive Methicillin-Resistant Staphylococcus aureus (MRSA) Nasal Screen Predict the Risk for MRSA Skin and Soft Tissue Infection? Ann Pharmacother 2022; 57:669-676. [PMID: 36189671 DOI: 10.1177/10600280221127389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background: Skin and soft tissue infections (SSTIs) are often caused by gram-positive bacteria that colonize the skin. Given the overuse of antibiotics, SSTIs are increasingly caused by resistant bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). Guidance on the utility of MRSA nasal screening for MRSA SSTI is limited. Objective: To determine whether MRSA nasal screening predicts the risk of MRSA SSTIs. Methods: This was a single-center, retrospective cohort study of adult patients with an SSTI diagnosis that had MRSA nasal screening and wound cultures obtained within 48 hours of starting antibiotics. Sensitivity, specificity, positive and negative predictive value, and positive and negative likelihood ratios were calculated using VassarStats. Pretest and posttest probabilities were estimated with Microsoft Excel. Results: A total of 884 patient encounters were reviewed between December 1, 2018, and October 31, 2021, and 300 patient encounters were included. The prevalence of MRSA SSTI was 18.3%. The MRSA nasal colonization had a sensitivity of 63.6%, specificity of 93.9%, positive predictive value of 70.0% (95% CI = 55.2%-81.7%), negative predictive value of 92.0% (95% CI = 87.7%-94.9%), positive likelihood ratio of 10.39 (95% CI = 6.12-17.65), negative likelihood ratio of 0.39 (95% CI = 0.27-0.55), positive posttest probability of 70.0%, and negative posttest probability of 8.0%. Conclusions: Given the high positive likelihood ratio, a positive MRSA nasal screen was associated with a large increase in the probability of MRSA SSTI at our institution, and a negative MRSA nasal screen was associated with a small but potentially significant decrease in the probability of MRSA SSTI.
Collapse
|
63
|
Fang X, Liu Y, Zhang M, Zhou S, Cui P, Hu H, Jiang P, Wang C, Qiu L, Wang J. Glucose oxidase loaded thermosensitive hydrogel as an antibacterial wound dressing. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
64
|
Rajpoot K, Prajapati SK, Malaiya A, Jain R, Jain A. Meropenem-Loaded Nanostructured Lipid Carriers For Skin and Soft Tissue Infection Caused by Staphylococcus aureus: Formulation, Design, and Evaluation. AAPS PharmSciTech 2022; 23:241. [PMID: 36008695 DOI: 10.1208/s12249-022-02381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Abstract
AIM Meropenem hydrochloride (MpM)-loaded nanostructured lipid carriers were designed for the effective management of skin infection caused by Staphylococcus aureus via topical route. The solvent evaporation tactic was preferred to develop nanostructured lipid carriers (NLCs). Stearic acid was used as a solid fatty acid; oleic acid was used as liquid fatty acid and Tween 80 as a surfactant. The Staphylococcus aureus burden was analyzed by pharmacodynamic studies. The skin retention was analyzed by fluorescence microscopy. Spherical shape of NLCs was confirmed by TEM. The optimum particle size of the MpM-NLCs was ~ 126.5 ± 0.9 nm with 79.1 ± 2.3% entrapment (EE) and 0.967 mV zeta potential. The in vitro release studies revealed 81.5 ± 3.1% release of drug in 48 h, while the pure drug was almost completely released (98.4 ± 1.4%) within 24 h confirming the potential of NLCs for sustained topical drug delivery. Skin permeation study also revealed better permeation of drug from NLCs than of plain drug. The prepared MpM-NLCs when stored at 4 ± 2°C for 90 days were found to be more stable when the formulation was stored at 28 ± 2°C. The S. aureus burden was analyzed by evaluating the zone of inhibition (ZOI). The ZOI of MpM alone and MpM-NLC gel was measured and compared with that of the control group. The MpM was found significantly effective when its gel was prepared with NLCs because of its enhanced adhesion property occlusion and ability to sustain release. In overall, the study's outcomes validated the relevance of NLC's composition as a vehicle for topical MpM administration in skin diseases caused by Staphylococcus aureus.
Collapse
Affiliation(s)
- Kshipra Rajpoot
- Bhagyoday Tirth Pharmacy College, Sagar, M.P., 470002, India
| | - Shiv Kumar Prajapati
- Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, Uttar Pradesh, 201310, India
| | | | - Richa Jain
- Molecular Biotechnology Laboratory, C.S.R.D., People's University, Bhopal, M.P., 462037, India
| | - Aakanchha Jain
- Bhagyoday Tirth Pharmacy College, Sagar, M.P., 470002, India. .,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
65
|
Amin AN, Dellinger EP, Harnett G, Kraft BD, LaPlante KL, LoVecchio F, McKinnell JA, Tillotson G, Valentine S. It's about the patients: Practical antibiotic stewardship in outpatient settings in the United States. Front Med (Lausanne) 2022; 9:901980. [PMID: 35966853 PMCID: PMC9363693 DOI: 10.3389/fmed.2022.901980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 12/01/2022] Open
Abstract
Antibiotic-resistant pathogens cause over 35,000 preventable deaths in the United States every year, and multiple strategies could decrease morbidity and mortality. As antibiotic stewardship requirements are being deployed for the outpatient setting, community providers are facing systematic challenges in implementing stewardship programs. Given that the vast majority of antibiotics are prescribed in the outpatient setting, there are endless opportunities to make a smart and informed choice when prescribing and to move the needle on antibiotic stewardship. Antibiotic stewardship in the community, or "smart prescribing" as we suggest, should factor in antibiotic efficacy, safety, local resistance rates, and overall cost, in addition to patient-specific factors and disease presentation, to arrive at an appropriate therapy. Here, we discuss some of the challenges, such as patient/parent pressure to prescribe, lack of data or resources for implementation, and a disconnect between guidelines and real-world practice, among others. We have assembled an easy-to-use best practice guide for providers in the outpatient setting who lack the time or resources to develop a plan or consult lengthy guidelines. We provide specific suggestions for antibiotic prescribing that align real-world clinical practice with best practices for antibiotic stewardship for two of the most common bacterial infections seen in the outpatient setting: community-acquired pneumonia and skin and soft-tissue infection. In addition, we discuss many ways that community providers, payors, and regulatory bodies can make antibiotic stewardship easier to implement and more streamlined in the outpatient setting.
Collapse
Affiliation(s)
- Alpesh N. Amin
- Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | | | - Glenn Harnett
- No Resistance Consulting, Birmingham, AL, United States
| | - Bryan D. Kraft
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kerry L. LaPlante
- College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Frank LoVecchio
- Department of Emergency Medicine, Valleywise Health, Arizona State University, Phoenix, AZ, United States
| | - James A. McKinnell
- Infectious Disease Clinical Outcomes Research Unit, Division of Infectious Disease, Lundquist Research Institute at Harbor-UCLA, Torrance, CA, United States
| | | | | |
Collapse
|
66
|
Skuhala T, Trkulja V, Rimac M, Dragobratović A, Desnica B. Analysis of Types of Skin Lesions and Diseases in Everyday Infectious Disease Practice-How Experienced Are We? Life (Basel) 2022; 12:978. [PMID: 35888068 PMCID: PMC9319552 DOI: 10.3390/life12070978] [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: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Rashes and skin lesions are a common reason for patient visits to emergency departments and physicians' offices. The differential diagnosis includes a variety of infectious and non-infectious diseases, some of which can be life-threatening. The aim of this retrospective study was to evaluate the quantity and type of skin lesions among outpatients and inpatients at a tertiary care university-affiliated teaching hospital for infectious diseases over a three-year period to assess disease burden and physicians' experience in diagnosing skin lesions. Diagnoses (by ICD-10 codes) were classified into three groups: infectious diseases that include skin lesions, non-infectious skin lesions and undiagnosed skin lesions. During the observed period, out of the total of 142,416 outpatients, 14.8% presented with some form of skin lesion. Among them, 68% had skin lesions inherent to infectious disease, 10.8% suffered from non-infectious skin lesions and 21.2% remained with undiagnosed skin lesions. The most common infectious diagnoses were chickenpox, herpes zoster and unspecified viral infections characterized by skin and mucous membrane lesions. The most common non-infectious diagnoses were urticaria and atopic dermatitis. Overall, the most common individual diagnosis (ICD-10 code) was "nonspecific skin eruption" (n = 4448, 21.1%), which was followed by chickenpox and herpes zoster. Among the 17,401 patients hospitalized over the observed period, 13.1% had skin lesion as the main reason for hospitalization, almost all (97.5%) of which were infectious in etiology. The most common diagnoses were cellulitis, erysipelas and herpes zoster. The presented data suggest that the burden of diseases presenting with skin lesions is significant in everyday infectious disease practice, but the overwhelming number of undiagnosed patients implies the need for further education in this area.
Collapse
Affiliation(s)
- Tomislava Skuhala
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia; (A.D.); (B.D.)
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Marin Rimac
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Anja Dragobratović
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia; (A.D.); (B.D.)
| | - Boško Desnica
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10000 Zagreb, Croatia; (A.D.); (B.D.)
| |
Collapse
|
67
|
Xue Y, Zhou J, Xu BN, Li Y, Bao W, Cheng XL, He Y, Xu CP, Ren J, Zheng YR, Jia CY. Global Burden of Bacterial Skin Diseases: A Systematic Analysis Combined With Sociodemographic Index, 1990-2019. Front Med (Lausanne) 2022; 9:861115. [PMID: 35547219 PMCID: PMC9084187 DOI: 10.3389/fmed.2022.861115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background The latest incidence and disability-adjusted life-years (DALYs) of major bacterial skin diseases (BSD) and their relationship with socioeconomic are not readily available. Objective Describe the global age-standardized incidence and DALYs rates of BSD and analyze their relationship with socioeconomic. Methods All data were obtained from Global Burden of Disease (GBD) 2019 database. The correlation between BSD and socioeconomic development status was analyzed. Results The age-standardized incidence and age-standardized DALYs rate of BSD are: 169.72 million [165.28-175.44] and 0.41 million [0.33-0.48]. Of the two main BSD, pyoderma cause significantly much heavier burden than cellulitis. The change of age-standardized incidence (7.38% [7.06-7.67]) and DALYs (-10.27% [-25.65 to 25.45]) rate of BSD presented an upward or downward trend from 1990 to 2019. The highest burden was in the low-middle sociodemographic index (SDI) area while the area with the lowest burden was recorded in the high-middle SDI area in 2019. Limitations GBD 2019 data of BSD are derived from estimation and mathematical modeling. Conclusion The burden of BSD is related to socioeconomic development status. The results based on GBD2019 data may benefit policymakers in guiding priority-setting decisions for the global burden of BSD.
Collapse
Affiliation(s)
- Yi Xue
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Jie Zhou
- School of Medicine, Xiamen University, Xiamen, China
| | - Bei-Ni Xu
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yue Li
- School of Medicine, Xiamen University, Xiamen, China
| | - Wu Bao
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Xia Lin Cheng
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Yan He
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Chun Peng Xu
- Division of Plastic Surgery, Siming Branch of the First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jun Ren
- Department of Dermatology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Ya Rong Zheng
- Division of Plastic Surgery, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Chi Yu Jia
- Department of Burns and Plastic & Wound Repair Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
68
|
Esposito S, Pagliano P, De Simone G, Pan A, Brambilla P, Gattuso G, Mastroianni C, Kertusha B, Contini C, Massoli L, Francisci D, Priante G, Libanore M, Bicocchi R, Borgia G, Maraolo AE, Brugnaro P, Panese S, Calabresi A, Amendola G, Savalli F, Geraci C, Tedesco A, Fossati S, Carretta A, Santantonio T, Cenderello G, Crisalli MP, Schiaroli E, Rovere P, Masini G, Ferretto R, Cascio A, Colomba C, Gioè C, Tumbarello M, Losito AR, Foti G, Prestileo T, Buscemi C, Iaria C, Iacobello C, Sonia S, Starnini G, Ialungo A, Sapienza M. Epidemiology, aetiology and treatment of skin and soft tissue infections: final report of a prospective multicentre national registry. J Chemother 2022; 34:524-533. [DOI: 10.1080/1120009x.2022.2075170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Silvano Esposito
- Department of Infectious Diseases, University of Salerno, Salerno, Italy
| | - Pasquale Pagliano
- Department of Infectious Diseases, University of Salerno, Salerno, Italy
| | - Giuseppe De Simone
- Department of Infectious Diseases, University of Salerno, Salerno, Italy
| | - Angelo Pan
- Department of Infectious Diseases, Istituti Ospitalieri of Cremona, Cremona, Italy
| | - Paola Brambilla
- Department of Infectious Diseases, Istituti Ospitalieri of Cremona, Cremona, Italy
| | - Gianni Gattuso
- Department of Infectious Diseases, Carlo Poma Hospital, Mantova, Italy
| | - Claudio Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University, Latina, Italy
| | - Blertha Kertusha
- Department of Public Health and Infectious Diseases, Sapienza University, Latina, Italy
| | - Carlo Contini
- Department of Medical Sciences, Section of Infectious Diseases and Dermatology, University of Ferrara, Ferrara, Italy
| | - Lorenzo Massoli
- Department of Medical Sciences, Section of Infectious Diseases and Dermatology, University of Ferrara, Ferrara, Italy
| | - Daniela Francisci
- Infectious Diseases Clinic, University Hospital “S. Maria”, University of Perugia, Perugia, Italy
| | - Giulia Priante
- Infectious Diseases Clinic, University Hospital “S. Maria”, University of Perugia, Perugia, Italy
| | - Marco Libanore
- Infectious Diseases Unit, Department of Infectious Diseases, S. Anna University Hospital, Ferrara, Italy
| | - Roberto Bicocchi
- Infectious Diseases Unit, Department of Infectious Diseases, S. Anna University Hospital, Ferrara, Italy
| | - Guglielmo Borgia
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Albert Enrico Maraolo
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Pierluigi Brugnaro
- Infectious Diseases Department, Ospedale Civile “SS. Giovanni e Paolo”, Venice, Italy
| | - Sandro Panese
- Infectious Diseases Department, Ospedale Civile “SS. Giovanni e Paolo”, Venice, Italy
| | - Alessandra Calabresi
- Emergency Department, Hospital “Santi Antonio e Biagio e C. Arrigo”, Alessandria, Italy
| | - Giovanni Amendola
- Emergency Department, Hospital “Santi Antonio e Biagio e C. Arrigo”, Alessandria, Italy
| | | | - Consuelo Geraci
- Infectious Diseases Unit, Hospital of Trapani, Trapani, Italy
| | - Andrea Tedesco
- Infectious Diseases Unit, Hospital Fracastoro San Bonifacio, Verona, Italy
| | - Sara Fossati
- Infectious Diseases Unit, University Hospital of Trieste, Trieste, Italy
| | - Anna Carretta
- Department of Infectious Diseases, University Hospital “Ospedali Riuniti” of Foggia, Foggia, Italy
| | - Teresa Santantonio
- Department of Infectious Diseases, University Hospital “Ospedali Riuniti” of Foggia, Foggia, Italy
| | | | | | - Elisabetta Schiaroli
- Department of Medicine, Section of Infectious Diseases, University of Perugia, Perugia, Italy
| | | | - Giulia Masini
- Infectious Diseases Unit, Legnago Hospital, Verona, Italy
| | - Roberto Ferretto
- Infectious Diseases Unit, “Alto Vicentino” Santorso Hospital, Vicenza, Italy
| | - Antonio Cascio
- Infectious Diseases Unit, Universita degli Studi di Palermo, Palermo, Italy
| | - Claudia Colomba
- Infectious Diseases Unit, Universita degli Studi di Palermo, Palermo, Italy
| | - Claudia Gioè
- Infectious Diseases Unit, Universita degli Studi di Palermo, Palermo, Italy
| | - Mario Tumbarello
- Institute of Infectious Diseases, Foundation Policlinico Universitario A. Gemelli IRCCS, Cattolica del Sacro Cuore University, Rome, Italy
| | - Angela Raffaella Losito
- Institute of Infectious Diseases, Foundation Policlinico Universitario A. Gemelli IRCCS, Cattolica del Sacro Cuore University, Rome, Italy
| | - Giuseppe Foti
- Infetious Diseases Unit, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | | | | | - Chiara Iaria
- Infectious Diseases Unit, Arnas Civico Hospital, Palermo, Italy
| | | | - Sofia Sonia
- Infectious Diseases Unit, Cannizzaro Hospital, Catania, Italy
| | | | - Anna Ialungo
- Infectious Diseases Unit, Belcolle Hospital, Viterbo, Italy
| | | | | |
Collapse
|
69
|
Close RM, Sutcliffe CG, Galdun P, Reid A, Askew MR, Davidson AM, Kellywood K, Parker D, Patel J, Romancito E, Brown LB, McAuley JB, Hammitt LL. Point-of-care molecular diagnostics for the detection of group A Streptococcus in non-invasive skin and soft tissue infections: a validation study. Diagn Microbiol Infect Dis 2022; 103:115729. [DOI: 10.1016/j.diagmicrobio.2022.115729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/15/2022] [Accepted: 05/20/2022] [Indexed: 01/21/2023]
|
70
|
Dellsperger S, Kramer S, Stoller M, Burger A, Geissbühler E, Amsler I, Hirsig A, Weyer L, Hebeisen U, Aebi P, Burgherr N, Brügger F, Chaix E, Salamoni J, Glauser S, Elisabeth Büchi A, Béguelin C, Waldegg G, Kessler B, Egger M, Sendi P. Early Switch from Intravenous to Oral Antibiotics in Skin- and Soft-tissue Infections: An Algorithm-based Prospective Multicentre Pilot Trial. Open Forum Infect Dis 2022; 9:ofac197. [PMID: 35794940 PMCID: PMC9251671 DOI: 10.1093/ofid/ofac197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background In hospitalized patients with skin and soft tissue infections (SSTIs), intravenous (IV) empiric antibiotic treatment is initiated. The best time point for switching from IV to oral treatment is unknown. We used an algorithm-based decision tree for the switch from IV to oral antibiotics within 48 hours and aimed to investigate the treatment outcome of this concept. Methods In a nonrandomized trial, we prospectively enrolled 128 patients hospitalized with SSTI from July 2019 to May 2021 at 3 institutions. Clinical and biochemical response data during the first week and at follow-up after 30 days were analyzed. Patients fulfilling criteria for the switch from IV to oral antibiotics were assigned to the intervention group. The primary outcome was a composite definition consisting of the proportion of patients with clinical failure or death of any cause. Results Ninety-seven (75.8%) patients were assigned to the intervention group. All of them showed signs of clinical improvement (ie, absence of fever or reduction of pain) within 48 hours of IV treatment, irrespective of erythema finding or biochemical response. The median total antibiotic treatment duration was 11 (interquartile range [IQR], 9–13) days in the invention group and 15 (IQR, 11–24) days in the nonintervention group (P < .001). The median duration of hospitalization was 5 (IQR, 4–6) days in the intervention group and 8 (IQR, 6–12) days in the nonintervention group (P < .001). There were 5 (5.2%) failures in the intervention group and 1 (3.2%) in the nonintervention group after a median follow-up of 37 days. Conclusions In this pilot trial, the proposed decision algorithm for early switch from IV to oral antibiotics for SSTI treatment was successful in 95% of cases. Clinical Trials Registration. ISRCTN15245496
Collapse
Affiliation(s)
| | - Simea Kramer
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Michael Stoller
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Annika Burger
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Elio Geissbühler
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Isabel Amsler
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Anna Hirsig
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Linda Weyer
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Ursula Hebeisen
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Philipp Aebi
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Nicolas Burgherr
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Fabienne Brügger
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Edouard Chaix
- Department of Internal Medicine, Spitalzentrum Biel, Biel/Bienne, Switzerland
| | - Jérôme Salamoni
- Department of Internal Medicine, Spitalzentrum Biel, Biel/Bienne, Switzerland
| | - Sandra Glauser
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Annina Elisabeth Büchi
- Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Emergency Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Charles Béguelin
- Department of Internal Medicine, Spitalzentrum Biel, Biel/Bienne, Switzerland
| | - Gabriel Waldegg
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Bernhard Kessler
- Clinic of Internal Medicine, Emmental Hospital, Burgdorf, Switzerland
| | - Martin Egger
- Clinic of Internal Medicine, Emmental Hospital, Langnau, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
71
|
Rosenblatt LS, King SA, Callahan ME, Wilkerson RG. Which Way Would You Slice It? Evaluation of 3 Educational Models for the Loop Drainage Technique. MEDICAL SCIENCE EDUCATOR 2022; 32:481-494. [PMID: 35528288 PMCID: PMC9054987 DOI: 10.1007/s40670-022-01530-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Emergency department visits for cutaneous abscesses are increasing. It is important for healthcare professionals to be proficient in identifying and treating abscesses. Loop drainage technique (LDT) is a newer technique which has been described in several articles but limited resources for teaching have been studied. The objective of this study was to compare 3 models for learning and teaching the LDT. METHODS This was a prospective survey study of a convenience sample of emergency medicine residents at a large urban academic center. Residents volunteered to participate during a scheduled cadaver and simulation session. After a self-directed review of the LDT, each participant performed ultrasound visualization and then the LDT on 3 simulated abscesses: a cadaveric model, a commercial abscess pad, and a homemade phantom. Participants completed pre- and post-simulation surveys. RESULTS Of 57 residents, 28 participated in the 1-day simulation. The majority (57.1%, p < 0.009) preferred the cadaver model for learning the LDT, and 78.6% reported it to have the most realistic physical examination for an abscess (p = 0.001). Prior to participation, 0% of residents felt proficient performing LDT. After participation, 46.4% of residents felt proficient and 78.6% reported intent to use in clinical practice (p < 0.001). CONCLUSIONS Simulation is an effective educational tool for both learning new skills and improving procedural competency. Residents found cadavers provided the most realistic physical examination, and the majority preferred it for learning the LDT. However, cadavers are not always accessible, an important factor when considering various educational settings.
Collapse
Affiliation(s)
- Lauren S. Rosenblatt
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S. Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201 USA
| | - Samantha A. King
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S. Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201 USA
| | - Michele E. Callahan
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S. Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201 USA
| | - R. Gentry Wilkerson
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 S. Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201 USA
| |
Collapse
|
72
|
Extrapolation as a Default Strategy in Pediatric Drug Development. Ther Innov Regul Sci 2022; 56:883-894. [PMID: 35006587 DOI: 10.1007/s43441-021-00367-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023]
Abstract
Pediatric drug development lags adult development by about 8 years (Mulugeta et al. in Pediatr Clin 64(6):1185-1196, 2017). In such context, many incentives, regulations, and innovative techniques have been proposed to address the disparity for pediatric patients. One such strategy is extrapolation of efficacy from a reference population. Extrapolation is currently justified by providing evidence in support of the effective use of drugs in children when the course of the disease and the expected treatment response would be sufficiently similar in the pediatric and reference population. This paper's position is that, despite uncertainties, pediatric drug development programs should initially assume some degree of extrapolation. The degree to which extrapolation can be used lies along a continuum representing the uncertainties to be addressed through generation of new pediatric evidence. In addressing these uncertainties, the extrapolation strategy should reflect the level of tolerable uncertainty concerning the decision to expose a child to the risks of a new drug. This judgment about the level of tolerable uncertainty should vary with the context (e.g., disease severity, existing therapeutic options) and can be embedded into pediatric drug development archetypes to ascertain the extent of studies needed and whether simultaneous development for adults and adolescents be considered.
Collapse
|
73
|
Eckmann C, Tulkens PM. Current and future options for treating complicated skin and soft tissue infections: focus on fluoroquinolones and long-acting lipoglycopeptide antibiotics. J Antimicrob Chemother 2021; 76:iv9-iv22. [PMID: 34849999 PMCID: PMC8632788 DOI: 10.1093/jac/dkab351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Bacterial skin and soft tissue infections are among the most common bacterial infections and constitute a major burden for patients and healthcare systems. Care is complicated by the variety of potential pathogens, some with resistance to previously effective antimicrobial agents, the wide spectrum of clinical presentations and the risk of progression to life-threatening forms. More-efficient care pathways are needed that can reduce hospital admissions and length of stay, while maintaining a high quality of care and adhering to antimicrobial stewardship principles. Several agents approved recently for treating acute bacterial skin and skin structure infections have characteristics that meet these requirements. We address the clinical and pharmacological characteristics of the fourth-generation fluoroquinolone delafloxacin, and the long-acting lipoglycopeptide agents dalbavancin and oritavancin.
Collapse
Affiliation(s)
- Christian Eckmann
- Department of General, Visceral and Thoracic Surgery, Klinikum Hannoversch-Muenden, Goettingen University, Germany
| | - Paul M Tulkens
- Cellular and Molecular Pharmacology, Louvain Drug Research Institute, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
74
|
Wilcox MH, Dryden M. Update on the epidemiology of healthcare-acquired bacterial infections: focus on complicated skin and skin structure infections. J Antimicrob Chemother 2021; 76:iv2-iv8. [PMID: 34849996 PMCID: PMC8632754 DOI: 10.1093/jac/dkab350] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Healthcare-associated infections (HCAIs) are a threat to patient safety and cause substantial medical and economic burden in acute care and long-term care facilities. Risk factors for HCAIs include patient characteristics, the type of care and the setting. Local surveillance data and microbiological characterization are crucial tools for guiding antimicrobial treatment and informing efforts to reduce the incidence of HCAI. Skin and soft tissue infections, including superficial and deep incisional surgical site infections, are among the most frequent HCAIs. Other skin and soft tissue infections associated with healthcare settings include vascular access site infections, infected burns and traumas, and decubitus ulcer infections.
Collapse
Affiliation(s)
- Mark H Wilcox
- Department of Medical Microbiology, Leeds Teaching Hospitals & University of Leeds, Leeds, UK
| | - Matthew Dryden
- Hampshire Hospitals NHS Foundation Trust, Winchester, UK
| |
Collapse
|
75
|
Scali JT, Son YG, Madison IT, Fink BA, Mueller TJ. Intraperitoneal abscess from perforated diverticulitis with fistualization to extraperitoneal abscess into the scrotum: a case report. AFRICAN JOURNAL OF UROLOGY 2021. [DOI: 10.1186/s12301-021-00251-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Scrotal abscesses are common skin abscesses seen in the general population; however, intraperitoneal abscesses tracking into the scrotum are rare. Intraperitoneal and retroperitoneal abscesses contiguous with the scrotum have been reported in the literature in specific populations. Wound cultures can aid in differentiating the source of the infections. Recurrent abscesses have been observed in high-risk populations, such as those with malignancy or who are immunocompromised.
Case presentation
We present a 71-year-old male with pericolonic abscess following perforated diverticulitis. The abscess was drained with interventional radiology and was complicated by an extraperitoneal abscess that tracked to the scrotum. Incision and drainage of the extraperitoneal abscess and the scrotal abscess were required with intravenous antibiotic therapy. The abscess tracking into the scrotum is reported without evidence of patent processus vaginalis.
Conclusion
We conclude that an intraperitoneal abscess can track to the scrotum through extraperitoneal fascial planes in the absence of a patent processus vaginalis. We show that extraperitoneal abscess spread to the scrotum is possible, with wound cultures helping to differentiate the source of the infection. High-risk patients with recurrent abscesses can also be susceptible to contiguous spread.
Collapse
|
76
|
Oliva A, Stefani S, Venditti M, Di Domenico EG. Biofilm-Related Infections in Gram-Positive Bacteria and the Potential Role of the Long-Acting Agent Dalbavancin. Front Microbiol 2021; 12:749685. [PMID: 34745053 PMCID: PMC8569946 DOI: 10.3389/fmicb.2021.749685] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 01/08/2023] Open
Abstract
Infections caused by Gram-positive bacteria are a major public health problem due to their increasing resistance to antibiotics. Staphylococcus and Enterococcus species' resistance and pathogenicity are enhanced by their ability to form biofilm. The biofilm lifestyle represents a significant obstacle to treatment because bacterial cells become highly tolerant to a wide range of antimicrobial compounds normally effective against their planktonic forms. Thus, novel therapeutic strategies targeting biofilms are urgently needed. The lipoglycopeptide dalbavancin is a long-acting agent for treating acute bacterial skin and skin structure infections caused by a broad range of Gram-positive pathogens. Recent studies have shown promising activity of dalbavancin against Gram-positive biofilms, including methicillin-resistant S. aureus (MRSA), methicillin-resistant S. epidermidis (MRSE), and vancomycin-susceptible enterococci. This review outlines the mechanisms regulating biofilm development in Staphylococcus and Enterococcus species and the clinical impact of biofilm-related infections. In addition, it discusses the clinical implications and potential therapeutic perspectives of the long-acting drug dalbavancin against biofilm-forming Gram-positive pathogens.
Collapse
Affiliation(s)
- Alessandra Oliva
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | - Stefania Stefani
- Laboratory of Molecular Medical Microbiology and Antimicrobial Resistance Research (Mmarl), Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | | |
Collapse
|
77
|
Feng P, Qiu H, Luo Y, Hu J, Cao Y, Pang Q, Mou X, Hou R, Hou W, Zhu Y. Development of Poloxamer Hydrogels Containing Antibacterial Guanidine-Based Polymers for Healing of Full-Thickness Skin Wound. ACS Biomater Sci Eng 2021; 7:4557-4568. [PMID: 34423628 DOI: 10.1021/acsbiomaterials.1c00600] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of hydrogels containing guanidine-based polymers using a poloxamer as the matrix were prepared to provide novel wound dressings with antibacterial and repairing-promotion properties for skin wounds. Herein, we developed a series of antibacterial hydrogels, the cationic guanidine-based polymer polyhexamethylene guanidine hydrochloride (PHMG) with poloxamer aqueous solution (12%, w/w) simplified as PHMGP, chitosan (CS)-cross-linked PHMG (referred to as PHMC) with poloxamer aqueous solution simplified as PHMCP, and hyaluronic acid (HA)-modified PHMG (referred to as PHMH) with poloxamer aqueous solution simplified as PHMHP, for enhancing full-thickness skin wound healing. The characterizations, antimicrobial activity, cytotoxicity, and in vivo full-thickness wound-healing capability of these hydrogels were analyzed and evaluated. The results show that though PHMGP possesses great bactericide properties, its cytotoxicity is too strong to support skin regeneration. However, after modified with CS or HA, PHMCP and PHMHP showed good biocompatibility and antimicrobial properties against Gram-positive and Gram-negative bacteria that are commonly present in injured skin. Both PHMCP and PHMHP hydrogels exhibited upgraded wound-healing efficiency in full-thickness skin defects, characterized by a shorter wound closure time, faster re-regeneration, and the earlier formation of skin appendages, compared with those of control or pure poloxamer treatments. Their biological mechanism was detected. Both PHMCP and PHMHP can regulate the related biofactors during the skin repair process such as interleukin-1β (IL-1β), interleukin-6 (IL-6), transforming growth factor beta-1(TGF-β1), alpha-smooth muscle actin (α-SMA), and vascular endothelial growth factor, to promote wound healing with less serious scarring. In short, hydrogels with excellent capabilities to inhibit microorganism infection and promote wound healing were developed, which will shed light on designing and producing wound dressings with promising applications in future.
Collapse
Affiliation(s)
- Peipei Feng
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Haofeng Qiu
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Yang Luo
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Jiaying Hu
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Yuhao Cao
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Qian Pang
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Xianbo Mou
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Ruixia Hou
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Wenjia Hou
- School of Medicine, Ningbo University, Ningbo 315010, China
| | - Yabin Zhu
- School of Medicine, Ningbo University, Ningbo 315010, China
| |
Collapse
|
78
|
The eternal dilemma of antitoxin antibiotics for skin and soft tissue infection. Curr Opin Infect Dis 2021; 34:80-88. [PMID: 33560018 DOI: 10.1097/qco.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW In standard clinical practice, combined antibiotic treatment is used to treat severe skin and soft tissue infections (SSTIs), whereby one of the drugs is usually a protein synthesis inhibitor antibiotic. However, evidence for this practice is only based on data from 'in vitro' studies, animal models and case reports. There are no randomized controlled trials. In the light of several new drugs marketed for the treatment of these infections, there is a need to revise the state of the art. RECENT FINDINGS New reviews and systematic appraisals of the literature exist on the use of protein synthesis inhibitor antibiotics to treat severe SSTI. Several 'in vitro' studies have assessed the efficacy of some of the new drugs. SUMMARY Combination therapy, including an adjuvant protein synthesis inhibitor antibiotic for toxin suppression, should be used both in patients with severe SSTI and in those with moderate infection and risk factors for methicillin-resistant positive- or Panton-Valentine leukocidin positive-Staphylococcus aureus infection.
Collapse
|
79
|
Rogne T, Liyanarachi KV, Rasheed H, Thomas LF, Flatby HM, Stenvik J, Løset M, Gill D, Burgess S, Willer CJ, Hveem K, Åsvold BO, Brumpton BM, DeWan AT, Solligård E, Damås JK. GWAS Identifies LINC01184/SLC12A2 as a Risk Locus for Skin and Soft Tissue Infections. J Invest Dermatol 2021; 141:2083-2086.e8. [PMID: 33662382 PMCID: PMC7612997 DOI: 10.1016/j.jid.2021.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Tormod Rogne
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Center for Perinatal, Pediatric and Environmental Epidemiology, Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA; Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Kristin V Liyanarachi
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Humaira Rasheed
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Laurent F Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway; BioCore - Bioinformatics Core Facility, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Helene M Flatby
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jørgen Stenvik
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Mari Løset
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Dipender Gill
- Clinical Pharmacology and Therapeutics Section, Institute of Medical and Biomedical Education, St George's University of London, London, United Kingdom; Institute for Infection and Immunity, St George's University of London, London, United Kingdom; Clinical Pharmacology Group, Pharmacy and Medicines Directorate, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom; Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Cristen J Willer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Research, Innovation and Education, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Bjørn O Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Endocrinology, Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Clinic of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Andrew T DeWan
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Center for Perinatal, Pediatric and Environmental Epidemiology, Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Erik Solligård
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jan K Damås
- Gemini Center for Sepsis Research, Department of Circulation and Medical Imaging, NTNU Norwegian University of Science and Technology, Trondheim, Norway; Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
80
|
Alphonse MP, Rubens JH, Ortines RV, Orlando NA, Patel AM, Dikeman D, Wang Y, Vuong I, Joyce DP, Zhang J, Mumtaz M, Liu H, Liu Q, Youn C, Patrick GJ, Ravipati A, Miller RJ, Archer NK, Miller LS. Pan-caspase inhibition as a potential host-directed immunotherapy against MRSA and other bacterial skin infections. Sci Transl Med 2021; 13:13/601/eabe9887. [PMID: 34233954 DOI: 10.1126/scitranslmed.abe9887] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/02/2021] [Accepted: 05/26/2021] [Indexed: 01/01/2023]
Abstract
Staphylococcus aureus causes most skin infections in humans, and the emergence of methicillin-resistant S. aureus (MRSA) strains is a serious public health threat. There is an urgent clinical need for nonantibiotic immunotherapies to treat MRSA infections and prevent the spread of antibiotic resistance. Here, we investigated the pan-caspase inhibitor quinoline-valine-aspartic acid-difluorophenoxymethyl ketone (Q-VD-OPH) for efficacy against MRSA skin infection in mice. A single systemic dose of Q-VD-OPH decreased skin lesion sizes and reduced bacterial burden compared with vehicle-treated or untreated mice. Although Q-VD-OPH inhibited inflammasome-dependent apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC) speck formation and caspase-1-mediated interleukin-1β (IL-1β) production, Q-VD-OPH maintained efficacy in mice deficient in IL-1β, ASC, caspase-1, caspase-11, or gasdermin D. Thus, Q-VD-OPH efficacy was independent of inflammasome-mediated pyroptosis. Rather, Q-VD-OPH reduced apoptosis of monocytes and neutrophils. Moreover, Q-VD-OPH enhanced necroptosis of macrophages with concomitant increases in serum TNF and TNF-producing neutrophils, monocytes/macrophages, and neutrophils in the infected skin. Consistent with this, Q-VD-OPH lacked efficacy in mice deficient in TNF (with associated reduced neutrophil influx and necroptosis), in mice deficient in TNF/IL-1R and anti-TNF antibody-treated WT mice. In vitro studies revealed that combined caspase-3, caspase-8, and caspase-9 inhibition reduced apoptosis, and combined caspase-1, caspase-8, and caspase-11 inhibition increased TNF, suggesting a mechanism for Q-VD-OPH efficacy in vivo. Last, Q-VD-OPH also had a therapeutic effect against Streptococcus pyogenes and Pseudomonas aeruginosa skin infections in mice. Collectively, pan-caspase inhibition represents a potential host-directed immunotherapy against MRSA and other bacterial skin infections.
Collapse
Affiliation(s)
- Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jessica H Rubens
- Divison of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nicholas A Orlando
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aman M Patel
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Dustin Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ivan Vuong
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammed Mumtaz
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Garrett J Patrick
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Advaitaa Ravipati
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
81
|
Felgueiras HP. An Insight into Biomolecules for the Treatment of Skin Infectious Diseases. Pharmaceutics 2021; 13:pharmaceutics13071012. [PMID: 34371704 PMCID: PMC8309093 DOI: 10.3390/pharmaceutics13071012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/31/2022] Open
Abstract
In assigning priorities, skin infectious diseases are frequently classified as minor when compared to infectious diseases of high mortality rates, such as tuberculosis or HIV. However, skin infections are amongst the most common and prevalent diseases worldwide. Elderly individuals present an increased susceptibility to skin infections, which may develop atypical signs and symptoms or even complicate pre-existing chronic disorders. When the skin fails to correct or inhibit the action of certain pathogenic microorganisms, biomolecules endowed with antimicrobial features are frequently administered topically or systemically to assist or treat such conditions. (1) Antibiotics, (2) antimicrobial peptides, or (3) natural extracts display important features that can actively inhibit the propagation of these pathogens and prevent the evolution of infectious diseases. This review highlights the properties and mechanisms of action of these biomolecules, emphasizing their effects on the most prevalent and difficult to treat skin infections caused by pathogenic bacteria, fungi, and viruses. The versatility of biomolecules’ actions, their symbiotic effects with skin cells and other inherent antimicrobial components, and their target-directed signatures are also explored here.
Collapse
Affiliation(s)
- Helena P Felgueiras
- Centre for Textile Science and Technology (2C2T), University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| |
Collapse
|
82
|
Enfrentamiento de las infecciones de piel en el adulto. REVISTA MÉDICA CLÍNICA LAS CONDES 2021. [DOI: 10.1016/j.rmclc.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
83
|
Ziesmer J, Tajpara P, Hempel N, Ehrström M, Melican K, Eidsmo L, Sotiriou GA. Vancomycin-Loaded Microneedle Arrays against Methicillin-Resistant Staphylococcus Aureus Skin Infections. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001307. [PMID: 34307835 PMCID: PMC8281827 DOI: 10.1002/admt.202001307] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/12/2021] [Indexed: 05/24/2023]
Abstract
Skin and soft tissue infections (SSTIs) caused by methicillin-resistant Staphylococcus aureus (MRSA) are a major healthcare burden, often treated with intravenous injection of the glycopeptide antibiotic vancomycin (VAN). However, low local drug concentration in the skin limits its treatment efficiency, while systemic exposure promotes the development of resistant bacterial strains. Topical administration of VAN on skin is ineffective as its high molecular weight prohibits transdermal penetration. In order to implement a local VAN delivery, microneedle (MN) arrays with a water-insoluble support layer for the controlled administration of VAN into the skin are developed. The utilization of such a support layer results in water-insoluble needle shafts surrounded by drug-loaded water-soluble tips with high drug encapsulation. The developed MN arrays can penetrate the dermal barriers of both porcine and fresh human skin. Permeation studies on porcine skin reveal that the majority of the delivered VAN is retained within the skin. It is shown that the VAN-MN array reduces MRSA growth both in vitro and ex vivo on skin. The developed VAN-MN arrays may be extended to several drugs and may facilitate localized treatment of MRSA-caused skin infections while minimizing adverse systemic effects.
Collapse
Affiliation(s)
- Jill Ziesmer
- Department of MicrobiologyTumour and Cell BiologyKarolinska InstitutetStockholmSE‐17177Sweden
| | - Poojabahen Tajpara
- Department of Medicine SolnaUnit of RheumatologyKarolinska InstitutetStockholmSE‐17177Sweden
| | | | - Marcus Ehrström
- Department of Reconstructive Plastic SurgeryKarolinska University Hospital SolnaStockholmSE‐17176Sweden
| | - Keira Melican
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES)Karolinska Institutet and KTH Royal Institute of TechnologyStockholmSE‐171 77Sweden
- Department of NeuroscienceKarolinska InstitutetStockholmSE‐171 77Sweden
| | - Liv Eidsmo
- Department of Medicine SolnaUnit of RheumatologyKarolinska InstitutetStockholmSE‐17177Sweden
- Diagnostiskt Centrum HudStockholmSE‐11137Sweden
- Leo Foundation Skin Immunology CenterUniversity of CopenhagenCopenhagenDK‐2100Denmark
| | - Georgios A. Sotiriou
- Department of MicrobiologyTumour and Cell BiologyKarolinska InstitutetStockholmSE‐17177Sweden
| |
Collapse
|
84
|
Namukobe J, Sekandi P, Byamukama R, Murungi M, Nambooze J, Ekyibetenga Y, Nagawa CB, Asiimwe S. Antibacterial, antioxidant, and sun protection potential of selected ethno medicinal plants used for skin infections in Uganda. Trop Med Health 2021; 49:49. [PMID: 34130746 PMCID: PMC8207782 DOI: 10.1186/s41182-021-00342-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/08/2021] [Indexed: 11/11/2022] Open
Abstract
Background Rural populations in Uganda rely heavily on medicinal plants for the treatment of bacterial skin infections. However, the efficacy of these medicinal plants for their pharmacological action is not known. The study aimed at evaluating the antibacterial, antioxidant, and sun protection potential of Spermacoce princeae, Psorospermum febrifugum, Plectranthus caespitosus, and Erlangea tomentosa extracts. Methods The plant samples were extracted by maceration sequentially using hexane, dichloromethane, ethyl acetate, methanol, and distilled water. Antibacterial activity of each extract was carried out using an agar well diffusion assay against Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, Klebsiella pneumonie, Streptococcus pyogenes, and Salmonella typhi. Acute dermal toxicity of the aqueous extract of S. princeae and P. febrifugum, and E. tomentosa was assessed in young adult healthy Wistar albino rats at a dose of 8000 and 10,000 mg/kg body weight. The antioxidant activity of each extract was carried out using a 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical scavenging assay. The sun protection factor was determined using Shimadzu UltraViolet-Visible double beam spectrophotometer between 290 and 320 nm. Results The plant extracts showed good antibacterial activity against the tested bacterial strains with minimum inhibitory concentration (MIC) ranging between 3.12 and 12.5 mg/ml. There was no significant change in the levels of creatinine, alanine aminotransferase, and aspartate aminotransferase in the rats even at a higher dose of 10,000 mg/kg, which was related to the results of biochemical analysis of the blood samples from the treated and control groups. The aqueous and methanol extracts of S. princeae showed potential antioxidant properties, with half maximal inhibitory concentration (IC50) values of 59.82 and 61.20 μg/ml respectively. The organic and aqueous extracts of P. caespitosus showed high levels of protection against Ultraviolet light with sun protection potential values ranging between 30.67 and 37.84. Conclusions The study demonstrated that the selected medicinal plants possessed good antibacterial, antioxidant, and sun protection properties. Therefore, the plants are alternative sources of antibacterial, antioxidant, and sun protection agents in managing bacterial skin infections.
Collapse
Affiliation(s)
- Jane Namukobe
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Peter Sekandi
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda.
| | - Robert Byamukama
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Moses Murungi
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Jennifer Nambooze
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Yeremiah Ekyibetenga
- Department of Chemistry, College of Natural Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Christine Betty Nagawa
- Department of Forestry, Biodiversity and Tourism, College of Agriculture and Environmental Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Savina Asiimwe
- Department of Plant Sciences, Microbiology & Biotechnology, Makerere University, P.O Box 7062, Kampala, Uganda
| |
Collapse
|
85
|
Sokolowski K, Pham HM, Wenzler E, Gemeinhart RA. Glutathione-Conjugated Hydrogels: Flexible Vehicles for Personalized Treatment of Bacterial Infections. Pharm Res 2021; 38:1247-1261. [PMID: 34117588 DOI: 10.1007/s11095-021-03057-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Skin and soft tissue infections are increasingly prevalent and often complicated by potentially fatal therapeutic hurdles, such as poor drug perfusion and antibiotic resistance. Delivery vehicles capable of versatile loading may improve local bioavailability and minimize systemic toxicities yet such vehicles are not clinically available. Therefore, we aimed to expand upon the use of glutathione-conjugated poly(ethylene glycol) GSH-PEG hydrogels beyond protein delivery and evaluate the ability to deliver traditional therapeutic molecules. METHODS PEG and GSH-PEG hydrogels were prepared using ultraviolet light (UV)-polymerization. Hydrogel loading and release of selected drug candidates was examined using UV-visible spectrometry. Therapeutic molecules and GST-fusion protein loading was examined using UV-visible and fluorescent spectrometry. Efficacy of released meropenem was assessed against meropenem-sensitive and -resistant P. aeruginosa in an agar diffusion bioassay. RESULTS For all tested agents, GSH-PEG hydrogels demonstrated time-dependent loading whereas PEG hydrogels did not. GSH-PEG hydrogels released meropenem over 24 h. Co-loading of biologic and traditional therapeutics into a single vehicle was successfully demonstrated. Meropenem-loaded GSH-PEG hydrogels inhibited the growth of meropenem-sensitive and resistant P. aeruginosa isolates. CONCLUSION GSH ligands within GSH-PEG hydrogels allow loading and effective delivery of charged therapeutic agents, in addition to biologic therapeutics.
Collapse
Affiliation(s)
- Karol Sokolowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Hai M Pham
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Eric Wenzler
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA.
| | - Richard A Gemeinhart
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois, USA. .,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA. .,Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois, USA. .,Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
86
|
Microbial Profile and Antibiogram Pattern Analysis of Skin and Soft Tissue Infections at a Tertiary Care Center in South India. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Skin and soft tissue infections (SSTIs) are the most common type of infection globally and also in India. Periodic evaluation of data on microbial profiling and antibiogram trend analysis is mandatory for creating a timely empirical treatment guideline and a proper antimicrobial cycling plan. In the current study, retrospective analysis of the data from 3,570 samples collected from suspected SSTI over one year was performed. Analysis was done on the clinical condition, causative agent/s identified and their antimicrobial susceptibility according to the standard guidelines. Seventy-three percent of samples yielded positive growth, with majority being unimicrobial infections. Gram-negative bacteria (GNB) were more commonly associated with infections from in-patients while Gram-positive cocci (GPC) were seen among out-patients. More than 70% of isolates among GNB were found susceptible to Carbapenems, piperacillin-tazobactam, amikacin and chloramphenicol. Among GPC, maximum sensitivity was seen to glycopeptides, linezolid followed by chloramphenicol, tetracycline, clindamycin and amikacin. Unusual resistance patterns like Penicillin resistant Ampicillin sensitive Enterococcus faecalis (PRASEF)strains, Multidrug resistance (MDR) and Extended drug resistance (XDR) were higher among Enterococci spp, MRSA, Acinetobacter spp and Klebsiella spp. Resistance to penicillin and cephalosporin drugs were high among GNB and GPC. Carbapenems, aminoglycosides, glycopeptides and broad-spectrum antibiotics are found sensitive and potential choices for empirical therapy. As prevalence of MDR and XDR strains were high and the trend analysis shows likelihood of few of the first and second line drugs becoming sensitive in future, we conclude that continued analysis of bacterial profiling and AMR pattern analysis among SSTI is essential.
Collapse
|
87
|
Thean LJ, Romani L, Engelman D, Jenney A, Wand H, Mani J, Paka J, Devi R, Sahukhan A, Kama M, Tuicakau M, Kado J, Carvalho N, Whitfeld M, Kaldor J, Steer AC. Prospective Surveillance of Primary Healthcare Presentations for Scabies and Bacterial Skin Infections in Fiji, 2018-2019. Am J Trop Med Hyg 2021; 105:230-237. [PMID: 34029210 DOI: 10.4269/ajtmh.20-1459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
Scabies, impetigo, and other skin and soft tissue infections (SSTIs) are highly prevalent in many tropical, low-middle income settings, but information regarding their burden of disease is scarce. We conducted a surveillance of presentations of scabies and SSTIs, including impetigo, abscesses, cellulitis, and severe SSTI, to primary health facilities in Fiji. We established a monthly reporting system over the course of 50 weeks (July 2018-June 2019) for scabies and SSTIs at all 42 public primary health facilities in the Northern Division of Fiji (population, ≈131,914). For each case, information was collected regarding demographics, diagnosis, and treatment. There were 13,736 individual primary healthcare presentations with scabies, SSTI, or both (108.3 presentations per 1000 person-years; 95% confidence interval [CI], 106.6-110 presentations). The incidence was higher for males than for females (incidence rate ratio [IRR], 1.15; 95% CI, 1.11-1.19). Children younger than 5 years had the highest incidence among all age groups (339.1 per 1000 person-years). The incidence was higher among the iTaukei (indigenous) population (159.9 per 1000 person-years) compared with Fijians of Indian descent (30.1 per 1000 person-years; IRR, 5.32; 95% CI, 5.03-5.61). Abscesses had the highest incidence (63.5 per 1,000 person-years), followed by scabies (28.7 per 1,000 person-years) and impetigo (21.6 per 1,000 person-years). Scabies and SSTIs impose a substantial burden in Fiji and represent a high incidence of primary health presentations in this population. The incidence in low-middle income settings is up to 10-times higher than that in high-income settings. New public health strategies and further research are needed to address these conditions.
Collapse
Affiliation(s)
- Li Jun Thean
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,2Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Lucia Romani
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,3Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Daniel Engelman
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,2Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,4Melbourne Children's Global Health, Melbourne Children's Campus, The Royal Children's Hospital, Melbourne, Australia
| | - Adam Jenney
- 5College of Medicine, Nursing and Health Sciences, Fiji National University, Suva, Fiji.,6Department of Infectious Diseases, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Handan Wand
- 3Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Jyotishna Mani
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jessica Paka
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Rachel Devi
- 7Ministry of Health and Medical Services, Suva, Fiji
| | | | - Mike Kama
- 7Ministry of Health and Medical Services, Suva, Fiji
| | | | - Joseph Kado
- 7Ministry of Health and Medical Services, Suva, Fiji.,8Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia.,9Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia
| | - Natalie Carvalho
- 10School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Margot Whitfeld
- 11Department of Dermatology, St. Vincent's Hospital, Sydney, New South Wales, Australia.,12School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - John Kaldor
- 3Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew C Steer
- 1Tropical Diseases Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,2Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,4Melbourne Children's Global Health, Melbourne Children's Campus, The Royal Children's Hospital, Melbourne, Australia
| |
Collapse
|
88
|
Ahmad F, Farooq A, Khan MUG. Deep Learning Model for Pathogen Classification Using Feature Fusion and Data Augmentation. Curr Bioinform 2021. [DOI: 10.2174/1574893615999200707143535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Bacterial pathogens are deadly for animals and humans. The ease of their dissemination, coupled
with their high capacity for ailment and death in infected individuals, makes them a threat to society.
Objective:
Due to high similarity among genera and species of pathogens, it is sometimes difficult for microbiologists to
differentiate between them. Their automatic classification using deep-learning models can help in reliable, and accurate
outcomes.
Method:
Deep-learning models, namely; AlexNet, GoogleNet, ResNet101, and InceptionV3 are used with numerous
variations including training model from scratch, fine-tuning without pre-trained weights, fine-tuning along with freezing
weights of initial layers, fine-tuning along with adjusting weights of all layers and augmenting the dataset by random
translation and reflection. Moreover, as the dataset is small, fine-tuning and data augmentation strategies are applied to
avoid overfitting and produce a generalized model. A merged feature vector is produced using two best-performing models
and accuracy is calculated by xgboost algorithm on the feature vector by applying cross-validation.
Results:
Fine-tuned models where augmentation is applied produces the best results. Out of these, two-best-performing
deep models i.e. (ResNet101, and InceptionV3) selected for feature fusion, produced a similar validation accuracy of 95.83
with a loss of 0.0213 and 0.1066, and a testing accuracy of 97.92 and 93.75, respectively. The proposed model used xgboost
to attained a classification accuracy of 98.17% by using 35-folds cross-validation.
Conclusion:
The automatic classification using these models can help experts in the correct identification of pathogens.
Consequently, they can help in controlling epidemics and thereby minimizing the socio-economic impact on the community.
Collapse
Affiliation(s)
- Fareed Ahmad
- Department of Computer Science and Engineering, Faculty of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| | - Amjad Farooq
- Department of Computer Science and Engineering, Faculty of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| | - Muhammad Usman Ghani Khan
- Department of Computer Science and Engineering, Faculty of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| |
Collapse
|
89
|
Welte T, Scheeren TW, Overcash JS, Saulay M, Engelhardt M, Hamed K. Efficacy and safety of ceftobiprole in patients aged 65 years or older: a post hoc analysis of three Phase III studies. Future Microbiol 2021; 16:543-555. [PMID: 33960817 DOI: 10.2217/fmb-2021-0042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: To evaluate the efficacy and safety of ceftobiprole in patients aged ≥65 years. Materials & methods: We conducted a post hoc analysis of three randomized, double-blind, Phase III studies in patients with acute bacterial skin and skin structure infections, community-acquired pneumonia and hospital-acquired pneumonia. Results: Findings for patients aged ≥65 years (n = 633) were consistent with those for the overall study populations, although a trend toward improved outcomes was reported in some subgroups, for example, patients aged ≥75 years with community-acquired pneumonia were more likely to achieve an early clinical response with ceftobiprole than comparator (treatment difference 16.3% [95% CI:1.8-30.8]). The safety profile was similar between treatment groups in all studies. Conclusion: This analysis further supports the efficacy and safety of ceftobiprole in older patients with acute bacterial skin and skin structure infections or pneumonia. Clinicaltrials.gov trial identifiers: NCT03137173, NCT00326287, NCT00210964, NCT00229008.
Collapse
Affiliation(s)
- Tobias Welte
- Department of Respiratory Medicine & Member of the German Centre for Lung Research, Medizinische Hochschule Hannover, Hannover, Germany
| | - Thomas Wl Scheeren
- Department of Anesthesiology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Mikael Saulay
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Marc Engelhardt
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Kamal Hamed
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| |
Collapse
|
90
|
Håkansson J, Cavanagh JP, Stensen W, Mortensen B, Svendsen JS, Svenson J. In vitro and in vivo antibacterial properties of peptide AMC-109 impregnated wound dressings and gels. J Antibiot (Tokyo) 2021; 74:337-345. [PMID: 33495549 DOI: 10.1038/s41429-021-00406-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/16/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023]
Abstract
Synthetic mimics of antimicrobial peptides (AMPs) is a promising class of molecules for a variety of antimicrobial applications. Several hurdles must be passed before effective systemic infection therapies with AMPs can be achieved, but the path to effective topical treatment of skin, nail, and soft tissue infections appears less challenging to navigate. Skin and soft tissue infection is closely coupled to the emergence of antibiotic resistance and represents a major burden to the healthcare system. The present study evaluates the promising synthetic cationic AMP mimic, AMC-109, for treatment of skin infections in vivo. The compound is evaluated both in impregnated cotton wound dressings and in a gel formulation against skin infections caused by Staphylococcus aureus and methicillin resistant S. aureus. Both the ability to prevent colonization and formation of an infection, as well as eradicate an ongoing infection in vivo with a high bacterial load, were evaluated. The present work demonstrates that AMC-109 displays a significantly higher antibacterial activity with up to a seven-log reduction in bacterial loads compared to current clinical standard therapy; Altargo cream (1% retapamulin) and Fucidin cream (2% fusidic acid) in the in vivo wound models. It is thus concluded that AMC-109 represents a promising entry in the development of new and effective remedies for various skin infections.
Collapse
Affiliation(s)
- Joakim Håkansson
- Department of Chemistry, Biomaterial & Textile, RISE Research Institutes of Sweden, Borås, Sweden.,Department of Laboratory Medicine, Institute of Biomedicine, Gothenburg University, Gothenburg, Sweden
| | - Jorunn Pauline Cavanagh
- Amicoat A/S, Sandvika, Norway.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Wenche Stensen
- Department of Chemistry, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - John-Sigurd Svendsen
- Amicoat A/S, Sandvika, Norway.,Department of Chemistry, UiT The Arctic University of Norway, Tromsø, Norway
| | - Johan Svenson
- Department of Chemistry, Biomaterial & Textile, RISE Research Institutes of Sweden, Borås, Sweden. .,Cawthron Institute, Nelson, New Zealand.
| |
Collapse
|
91
|
Poliseno M, Bavaro DF, Brindicci G, Luzzi G, Carretta DM, Spinarelli A, Messina R, Miolla MP, Achille TI, Dibartolomeo MR, Dell'Aera M, Saracino A, Angarano G, Favale S, D'Agostino C, Moretti B, Signorelli F, Taglietti C, Carbonara S. Dalbavancin Efficacy and Impact on Hospital Length-of-Stay and Treatment Costs in Different Gram-Positive Bacterial Infections. Clin Drug Investig 2021; 41:437-448. [PMID: 33884583 PMCID: PMC8059686 DOI: 10.1007/s40261-021-01028-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2021] [Indexed: 01/07/2023]
Abstract
Background and Objectives The study aimed to evaluate the impact of dalbavancin therapy on both hospital length-of-stay (LOS) and treatment-related costs, as well as to describe the clinical outcome, in a retrospective cohort of patients with diverse Gram-positive bacterial infections, hospitalized in different specialty Units. Methods From July 2017 to July 2019, clinical and sociodemographic data were collected for all hospitalized patients switched to dalbavancin for the treatment of Gram-positive infections. LOS and treatment-related costs were assessed and compared to a hypothetical scenario where the initial standard antimicrobial therapy would have been administered in hospital for the same duration as dalbavancin. Results A total of 50 patients were enrolled. The observed infections were: acute bacterial skin and skin structure infections (ABSSSIs, 12 patients), complicated ABSSSIs (eight patients), osteoarticular infections (18 patients), vascular graft or cardiovascular implantable electronic devices (CIED) infections (12 patients). After a median of 14 [interquartile range (IQR) 7–28] days, the in-hospital antimicrobial therapy was switched to dalbavancin 1500 mg. When appropriate, considering the site and the clinical course of the infection, 1500 mg doses were repeated every 14 days until recovery. Overall, 49/50 (98%) patients reported clinical success at the end of therapy. No relapses were observed in 37 patients for whom a median follow-up of 150 (IQR 30–180) days was available. By switching to dalbavancin, a median of €8,259 (IQR 5644–17,270) and 14 hospital days (IQR 22–47) per patient were saved. Conclusions In this experience, the use of dalbavancin contributed to shorten LOS and treatment-related costs, especially in difficult Gram-positive infections requiring prolonged therapy.
Collapse
Affiliation(s)
- Mariacristina Poliseno
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy. .,Unit of Infectious Diseases, A.O.U. Policlinico Riuniti, Foggia, Italy.
| | - Davide Fiore Bavaro
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy
| | - Gaetano Brindicci
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy
| | - Giovanni Luzzi
- Unit of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | | | - Antonio Spinarelli
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Raffaella Messina
- Division of Neurosurgery, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Maria Paola Miolla
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Teresa Immacolata Achille
- Unit of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | | | - Maria Dell'Aera
- Department of Hospital Pharmacy, Policlinico of Bari, University of Bari, Bari, Italy
| | - Annalisa Saracino
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy
| | - Gioacchino Angarano
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy
| | - Stefano Favale
- Unit of Cardiovascular Diseases, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Carlo D'Agostino
- Cardiology Department, University, Hospital Policlinico Consorziale, Bari, Italy
| | - Biagio Moretti
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Francesco Signorelli
- Division of Neurosurgery, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | | | - Sergio Carbonara
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari, Bari, Italy.,Unit of Infectious Diseases, Hospital V. Emanuele II, Bisceglie, Italy
| |
Collapse
|
92
|
Hoang TPN, Ghori MU, Conway BR. Topical Antiseptic Formulations for Skin and Soft Tissue Infections. Pharmaceutics 2021; 13:558. [PMID: 33921124 PMCID: PMC8071503 DOI: 10.3390/pharmaceutics13040558] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 01/22/2023] Open
Abstract
Skin and soft tissue infections (SSTIs) are usually acute conditions of inflammatory microbial occupation of the skin layers and underlying soft tissues. SSTIs are one of the most frequent types of infection, typically requiring medical intervention and contribute to morbidity and mortality in both primary care and hospitalised patients. Due to the dramatic rise of antibiotic resistance, antiseptic agents can be potential alternatives for the prevention and treatment of SSTIs. Notably, they are commonly recommended in many global practical guidelines for use in per- and post- operative procedures. A range of antiseptics, including chlorhexidine, triclosan, alcohol, and povidone-iodine, are used and are mainly formulated as traditional, simple dosage forms such as solutions and semi-solids. However, in recent years, there have been studies reporting the potential for nanotechnology in the delivery of antiseptics. In this review, we have collated the scientific literature that focuses on topical antiseptic formulations for prevention and treatment of SSTIs, and have divided findings into traditional and advanced formulations. We conclude that although nanotechnological formulations have demonstrated potential advantages for delivering drugs; nevertheless, there is still scope for traditional formulations and further development of optimised topical formulations to address the rise of antimicrobial resistance.
Collapse
Affiliation(s)
- Thi Phuong Nga Hoang
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (T.P.N.H.); (M.U.G.)
| | - Muhammad Usman Ghori
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (T.P.N.H.); (M.U.G.)
| | - Barbara R. Conway
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK; (T.P.N.H.); (M.U.G.)
- Institute of Skin Integrity and Infections Prevention, University of Huddersfield, Huddersfield HD1 3DH, UK
| |
Collapse
|
93
|
Pai MP, Wilcox MH, Chitra S, McGovern PC. Safety and efficacy of omadacycline by BMI categories and diabetes history in two Phase III randomized studies of patients with acute bacterial skin and skin structure infections. J Antimicrob Chemother 2021; 76:1315-1322. [PMID: 33458763 PMCID: PMC8050767 DOI: 10.1093/jac/dkaa558] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES The objectives of this post-hoc analysis were to examine the safety and efficacy of omadacycline by BMI categories and diabetes history in adults with acute bacterial skin and skin structure infections (ABSSSI) from two pivotal Phase III studies. PATIENTS AND METHODS OASIS-1 (ClinicalTrials.gov identifier NCT02378480): patients were randomized 1:1 to IV omadacycline or linezolid for 7-14 days, with optional transition to oral medication. OASIS-2 (ClinicalTrials.gov identifier NCT02877927): patients received once-daily oral omadacycline or twice-daily oral linezolid for 7-14 days. Early clinical response (ECR) was defined as ≥20% reduction in lesion size 48-72 h after the first dose. Clinical success at post-treatment evaluation (PTE; 7-14 days after the last dose) was defined as symptom resolution such that antibacterial therapy was unnecessary. Safety was assessed by treatment-emergent adverse events and laboratory measures. Between-treatment comparisons were made with regard to WHO BMI categories and diabetes history. RESULTS Patients were evenly distributed among healthy weight, overweight and obese groups. Clinical success for omadacycline-treated patients at ECR and PTE was similar across BMI categories. Outcomes by diabetes status were similar in omadacycline- and linezolid-treated patients: at ECR, clinical success rates were lower for those with diabetes; at PTE, clinical success was similar between treatment groups regardless of diabetes history. The safety of omadacycline and linezolid was largely similar across BMI groups and by diabetes history. CONCLUSIONS Omadacycline efficacy in patients with higher BMI and in patients with diabetes was consistent with results from two pivotal Phase III ABSSSI trials. Fixed-dose omadacycline is an appropriate treatment for ABSSSI in adults regardless of BMI.
Collapse
Affiliation(s)
- Manjunath P Pai
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mark H Wilcox
- University of Leeds & Leeds Teaching Hospitals, Leeds LS2 9JT, UK
| | - Surya Chitra
- Paratek Pharmaceuticals, Inc., King of Prussia, PA 19406, USA
| | - Paul C McGovern
- Paratek Pharmaceuticals, Inc., King of Prussia, PA 19406, USA
| |
Collapse
|
94
|
Perni S, Preedy EC, Prokopovich P. Amplify antimicrobial photo dynamic therapy efficacy with poly-beta-amino esters (PBAEs). Sci Rep 2021; 11:7275. [PMID: 33790379 PMCID: PMC8012660 DOI: 10.1038/s41598-021-86773-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/17/2021] [Indexed: 01/03/2023] Open
Abstract
Light-activated antimicrobial agents (photosensitisers) are promising alternatives to antibiotics for the treatment of skin infections and wounds through antimicrobial photo dynamic therapy (aPDT); utilisation of this technique is still restricted by general low efficacy requiring long exposure time (in the order of tens of minutes) that make the treatment very resource intensive. We report for the first time the possibility of harvesting the cell penetrating properties of poly-beta-amino esters (PBAEs) in combination with toluidine blue O (TBO) to shorten aPDT exposure time. Candidates capable of inactivation rates 30 times quicker than pure TBO were discovered and further improvements through PBAE backbone optimisation could be foreseen. Efficacy of the complexes was PBAE-dependent on a combination of TBO uptake and a newly discovered and unexpected role of PBAEs on reactive species production. Chemometric approach of partial least square regression was employed to assess the critical PBAE properties involved in this newly observed phenomenon in order to elicit a possible mechanism. The superior antimicrobial performance of this new approach benefits from the use of well established, low-cost and safe dye (TBO) coupled with inexpensive, widely tested and biodegradable polymers also known to be safe. Moreover, no adverse cytotoxic effects of the PBAEs adjuvated TBO delivery have been observed on a skin cells in vitro model demonstrating the safety profile of this new technology.
Collapse
Affiliation(s)
- Stefano Perni
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Emily C Preedy
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Polina Prokopovich
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
95
|
Improved economic and clinical outcomes with oritavancin versus a comparator group for treatment of acute bacterial skin and skin structure infections in a community hospital. PLoS One 2021; 16:e0248129. [PMID: 33735185 PMCID: PMC7971560 DOI: 10.1371/journal.pone.0248129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/22/2021] [Indexed: 11/19/2022] Open
Abstract
Background Oritavancin is a lipoglycopeptide antibiotic with in vitro bactericidal activity against gram-positive pathogens indicated for use in adults with acute bacterial skin and skin structure infections (ABSSSI). Its concentration-dependent activity and prolonged half-life provide a convenient single-dose alternative to multi-dose daily therapies for ABSSSI. This retrospective cohort study was conducted to quantify the clinical and economic advantages of using oritavancin compared to other antibiotic agents that have been historically effective for ABSSSI. Methods Seventy-nine patients received oritavancin who had failed previous outpatient antibiotic therapy (OPAT) for cellulitis or abscess and were subsequently readmitted to the hospital as an inpatient between 2016 and 2018. These patients were compared to a cohort of 28 patients receiving other antibiotics following OPAT failure and subsequent hospitalization for these two infection types. The primary clinical end point was average length of stay (aLOS) and secondary endpoints included readmission rates for the same indication at 30 and 90 days after discharge and the average hospital cost (aHC). Results A total of 107 patients were hospitalized for treatment of cellulitis or abscess. Demographic characteristics of both the oritavancin and comparator groups were similar except for the presence of diabetes. The primary clinical endpoint showed a non-significant decrease in aLOS between the oritavancin group versus comparator (2.12 days versus 2.59 days; p = 0.097). The secondary endpoints revealed lower readmission rates associated with oritavancin treatment at 30 and 90 days; the average hospital cost was 5.9% lower for patients that received oritavancin. Conclusion The results of this study demonstrate that oritavancin provides not only a single-dose alternative to multi-day therapies for skin and skin structure infections, but also a clinical and economic advantage compared to other antibiotic agents.
Collapse
|
96
|
Karauzum H, Venkatasubramaniam A, Adhikari RP, Kort T, Holtsberg FW, Mukherjee I, Mednikov M, Ortines R, Nguyen NTQ, Doan TMN, Diep BA, Lee JC, Aman MJ. IBT-V02: A Multicomponent Toxoid Vaccine Protects Against Primary and Secondary Skin Infections Caused by Staphylococcus aureus. Front Immunol 2021; 12:624310. [PMID: 33777005 PMCID: PMC7987673 DOI: 10.3389/fimmu.2021.624310] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus causes a wide range of diseases from skin infections to life threatening invasive diseases such as bacteremia, endocarditis, pneumonia, surgical site infections, and osteomyelitis. Skin infections such as furuncles, carbuncles, folliculitis, erysipelas, and cellulitis constitute a large majority of infections caused by S. aureus (SA). These infections cause significant morbidity, healthcare costs, and represent a breeding ground for antimicrobial resistance. Furthermore, skin infection with SA is a major risk factor for invasive disease. Here we describe the pre-clinical efficacy of a multicomponent toxoid vaccine (IBT-V02) for prevention of S. aureus acute skin infections and recurrence. IBT-V02 targets six SA toxins including the pore-forming toxins alpha hemolysin (Hla), Panton-Valentine leukocidin (PVL), leukocidin AB (LukAB), and the superantigens toxic shock syndrome toxin-1 and staphylococcal enterotoxins A and B. Immunization of mice and rabbits with IBT-V02 generated antibodies with strong neutralizing activity against toxins included in the vaccine, as well as cross-neutralizing activity against multiple related toxins, and protected against skin infections by several clinically relevant SA strains of USA100, USA300, and USA1000 clones. Efficacy of the vaccine was also shown in non-naïve mice pre-exposed to S. aureus. Furthermore, vaccination with IBT-V02 not only protected mice from a primary infection but also demonstrated lasting efficacy against a secondary infection, while prior challenge with the bacteria alone was unable to protect against recurrence. Serum transfer studies in a primary infection model showed that antibodies are primarily responsible for the protective response.
Collapse
Affiliation(s)
| | | | | | - Tom Kort
- Integrated BioTherapeutics, Rockville, MD, United States
| | | | | | - Mark Mednikov
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Roger Ortines
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Nhu T. Q. Nguyen
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Thien M. N. Doan
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jean C. Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Rockville, MD, United States
| |
Collapse
|
97
|
Karpuz M, Atlihan-Gundogdu E, Demir ES, Senyigit Z. Radiolabeled Tedizolid Phosphate Liposomes for Topical Application: Design, Characterization, and Evaluation of Cellular Binding Capacity. AAPS PharmSciTech 2021; 22:62. [PMID: 33528714 DOI: 10.1208/s12249-020-01917-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022] Open
Abstract
Nowadays, the incidence of acute bacterial skin and skin structure infection (ABSSSI) is increasing. The increased bioavailability and reduced drug resistance of antibiotics are crucial to obtain a more effective treatment response in these infections. These favorable properties could be achieved by different drug delivery systems such as liposomes. In this study, nanosized, radiolabeled tedizolid phosphate liposomal formulations were prepared and evaluated with their in vitro cellular binding capacity and biocompatible profile for topical treatment of ABSSSI. Liposomes were characterized by evaluation of their visual inspection, particle size (about 190-270 nm), zeta potential value (around 0), and encapsulation efficiency (nearly 10%). The release rate of tedizolid phosphate from liposomes was also studied using dialysis membranes and evaluated kinetically. The stability of formulations was observed at three different temperatures and humidity conditions for 28 days. Afterward, liposomes were labeled with 99mTc, and the optimal amount of reducing agent (stannous chloride) was determined as 500 μg in this direct labeling procedure. All liposome formulations were successfully radiolabeled with high efficiency and exhibited high radiochemical purity (> 80%) during 6 h in different media. Furthermore, the cellular bindings of liposomal formulations were evaluated in human skin fibroblast cells by measuring the radioactivity. Higher radioactivity values were obtained in CCD-1070Sk cells incubated by liposome formulations compared to sodium pertechnetate. This finding suggested that liposomal formulation increased the cellular binding of radioactivity. By the result of our study, nanosized, tedizolid phosphate encapsulated liposome formulation was found to be a favorable carrier system in the treatment of ABSSSI.
Collapse
|
98
|
Gao Y, Zhang W, Cheng YF, Cao Y, Xu Z, Xu LQ, Kang Y, Xue P. Intradermal administration of green synthesized nanosilver (NS) through film-coated PEGDA microneedles for potential antibacterial applications. Biomater Sci 2021; 9:2244-2254. [PMID: 33514957 DOI: 10.1039/d0bm02136a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Skin infections caused by pathogens, including bacteria, fungi and viruses, are difficult to completely eliminate through standard topical administration, owing to the restricted drug permeation into the epidermis layer. Herein, we developed a poly(ethylene glycol) diacrylate (PEGDA) microneedle patch with surface coating of a nanosilver (NS) encapsulated gelatin/sucrose film for antibacterial applications, by virtue of enhanced skin permeation by microneedle penetration and efficient drug delivery through rapid film dissolving. NS was facilely synthesized through a green process based on the bioinspired crystallization of ionic state silver in the presence of a silk fibroin (SF) template. A gelatin/sucrose polymeric film encapsulating NS was dressed on the surface of the mold cavity, and film-coated PEGDA (PEGDA/film-NS) microneedles were subsequently fabricated through standard ultraviolet (UV) light-induced polymerization. To demonstrate their advantages for therapeutic applications, the physicochemical properties of the as-developed microneedles were characterized in terms of their morphology, composition, mechanical strength, etc. Moreover, rapid NS release from PEGDA@film-NS microneedles driven by the aqueous environment was demonstrated under physiological conditions. Additionally, such film-coated microneedles exhibited good mechanical strength for skin penetration, and their antibacterial activity against Gram-positive bacteria (Staphylococcus epidermidis and Staphylococcus aureus) as well as Gram-negative bacteria (Escherichia coli and Pseudomonas aeruginosa) was verified using bacterial suspension in vitro. Altogether, such a minimally invasive strategy exhibited good potential for realizing a broad-spectrum antibacterial effect, which may provide a practical methodology for the management of polymicrobial skin infection during clinical trials.
Collapse
Affiliation(s)
- Ya Gao
- School of Materials and Energy, Southwest University, Chongqing 400715, China.
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Kranjec C, Morales Angeles D, Torrissen Mårli M, Fernández L, García P, Kjos M, Diep DB. Staphylococcal Biofilms: Challenges and Novel Therapeutic Perspectives. Antibiotics (Basel) 2021; 10:131. [PMID: 33573022 PMCID: PMC7911828 DOI: 10.3390/antibiotics10020131] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Staphylococci, like Staphylococcus aureus and S. epidermidis, are common colonizers of the human microbiota. While being harmless in many cases, many virulence factors result in them being opportunistic pathogens and one of the major causes of hospital-acquired infections worldwide. One of these virulence factors is the ability to form biofilms-three-dimensional communities of microorganisms embedded in an extracellular polymeric matrix (EPS). The EPS is composed of polysaccharides, proteins and extracellular DNA, and is finely regulated in response to environmental conditions. This structured environment protects the embedded bacteria from the human immune system and decreases their susceptibility to antimicrobials, making infections caused by staphylococci particularly difficult to treat. With the rise of antibiotic-resistant staphylococci, together with difficulty in removing biofilms, there is a great need for new treatment strategies. The purpose of this review is to provide an overview of our current knowledge of the stages of biofilm development and what difficulties may arise when trying to eradicate staphylococcal biofilms. Furthermore, we look into promising targets and therapeutic methods, including bacteriocins and phage-derived antibiofilm approaches.
Collapse
Affiliation(s)
- Christian Kranjec
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Danae Morales Angeles
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Marita Torrissen Mårli
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Lucía Fernández
- Department of Technology and Biotechnology of Dairy Products, Dairy Research Institute of Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (L.F.); (P.G.)
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Pilar García
- Department of Technology and Biotechnology of Dairy Products, Dairy Research Institute of Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (L.F.); (P.G.)
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Dzung B. Diep
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| |
Collapse
|
100
|
Doostan M, Maleki H, Doostan M, Khoshnevisan K, Faridi-Majidi R, Arkan E. Effective antibacterial electrospun cellulose acetate nanofibrous patches containing chitosan/erythromycin nanoparticles. Int J Biol Macromol 2020; 168:464-473. [PMID: 33316335 DOI: 10.1016/j.ijbiomac.2020.11.174] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/24/2023]
Abstract
Herein, we fabricated the antibacterial nanofibrous mats composed of cellulose acetate (CA) nanofibers loaded with erythromycin-chitosan nanoparticles (Ery-CS NPs) intended for infected wound dressing. The Ery-loaded CS NPs were prepared by ionic gelation process and then incorporated into the CA electrospun nanofibers (NFs). Regarding physiochemical properties, the NPs and obtained mats were characterized using dynamic light scattering (DLS), scanning electron microscopy (SEM), attenuated total reflection fourier transform infrared (ATR-FTIR), and contact angle measurement. The antimicrobial activity and cell viability of fibroblast cells were also evaluated. The results indicated that Ery was loaded into CS NPs with high encapsulation efficiency (95%). The CA NFs (17% w/v) incorporated with the Ery-CS NPs (12 wt%) displayed smooth homogenous morphology with 141.7 ± 91.7 nm average diameter. The relevant analyses confirmed that the NPs incorporated into NFs and provided high water holding capacity with high porosity. Finally, Ery-CS NPs/CA mats were able to inhibit the growth of both Gram-positive and Gram-negative bacteria as well as showed no cytotoxic effect on the human dermal fibroblast cells. Overall, our findings concluded that the proposed system could be potentially applied as the proper antibacterial mats for infected wound dressing applications.
Collapse
Affiliation(s)
- Maryam Doostan
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hassan Maleki
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahtab Doostan
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamyar Khoshnevisan
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Arkan
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|