1
|
Babin P, Delbove A, Gousseff M, Gouin JB, Crabol Y, Akoha M, Cady A, Lemaitre F, Tattevin P, Boulay H. Impact of a kidney-sparing protocol for the treatment of methicillin-susceptible Staphylococcus aureus bloodstream infections (CLOXECO): a quasi-experimental study. J Antimicrob Chemother 2025; 80:1261-1268. [PMID: 40036732 DOI: 10.1093/jac/dkaf062] [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: 10/21/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Antistaphylococcal penicillin (ASP) is the first-line therapy for MSSA bloodstream infection (BSI), with cefazolin as an alternative. However, ASPs are associated with a high risk of acute kidney injury (AKI) and overexposure. We implemented a kidney-sparing protocol based on: (i) systematic use of cefazolin in patients with creatinine clearance of <60 mL/min or any risk factor for AKI; and (ii) reduced ASP dose (75-100 mg/kg/day) with therapeutic drug monitoring. METHODS We compared all episodes of MSSA BSI in adults admitted during the 15 months before (control period) and the 12 months after (optimization period) protocol implementation. Primary outcome was sterile blood cultures by Day 3. Secondary outcomes included AKI, dialysis, MSSA BSI relapses, and mortality. RESULTS We included 100 patients in the control group and 104 in the optimization group. Baseline characteristics were similar in both groups, with a mean ± SD age of 73 ± 20 years, male predominance (73%), and high prevalence of chronic kidney disease (80%) and diabetes (31%). Initial treatment was ASP (cloxacillin) in 80/100 (80%) patients in the control group, versus 24/104 (23%) in the optimization group (P < 0.001). Day 3 sterile blood cultures, and Day 30 mortality remained similar (respectively 95% versus 93%, and 21% versus 24%; not significant). AKI and dialysis requirements were less frequent during the optimization period, respectively 37% versus 56% (P = 0.045), and 1% versus 8% (P = 0.017). CONCLUSIONS A kidney-sparing protocol for MSSA BSI based on systematic use of cefazolin in patients with AKI risk factor, and lower ASP doses, was associated with similar efficacy and lower risk of AKI and dialysis requirements.
Collapse
Affiliation(s)
- Philippine Babin
- Service de néphrologie, Hôpital Pontchaillou, rue le Guilloux, CHU Rennes, France
| | - Agathe Delbove
- Service de réanimation médicale, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Marie Gousseff
- Service de maladies infectieuses et médecine interne, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Jean-Baptiste Gouin
- Service de néphrologie, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Yoann Crabol
- Service de maladies infectieuses et médecine interne, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Mauriac Akoha
- Service de néphrologie, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Anne Cady
- Service de biologie médicale, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| | - Florian Lemaitre
- Pharmacologie Clinique, Hôpital Pontchaillou, rue le Guilloux, CHU Rennes, France
| | - Pierre Tattevin
- Service de néphrologie, Hôpital Pontchaillou, rue le Guilloux, CHU Rennes, France
- Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, rue le Guilloux, CHU Rennes, France
| | - Hugoline Boulay
- Service de néphrologie, Centre Hospitalier Bretagne Atlantique, Boulevard Général Maurice Guillaudot, Vannes, France
| |
Collapse
|
2
|
Villa SDL, Escrihuela-Vidal F, Fernández-Hidalgo N, Escudero-Sánchez R, Cabezón I, Boix-Palop L, Díaz-Pollán B, Goikoetxea AJ, García-País MJ, Pérez-Rodríguez MT, Crespo Á, Buzón-Martín L, Sanz-Peláez O, Ramos-Merino L, Fiorante S, Muñoz P. Ceftaroline for bloodstream infections caused by methicillin-resistant Staphylococcus aureus: a multicentre retrospective cohort study. Clin Microbiol Infect 2025; 31:793-801. [PMID: 39581546 DOI: 10.1016/j.cmi.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES To evaluate the effectiveness of ceftaroline vs. vancomycin or daptomycin in the treatment of methicillin-resistant Staphylococcus aureus bloodstream infections (BSIs) (MRSA-BSIs). METHODS This multicentre retrospective study conducted in 15 Spanish hospitals included data from the first MRSA-BSIs of adult patients between January 2019 and December 2022. The ceftaroline group included patients who received ceftaroline for ≥72 hours within the first week of BSI onset; the standard-of-care (SOC) group included patients who received vancomycin or daptomycin ≥72 hours after BSI onset. Primary outcome was 30-day all-cause mortality; secondary outcomes included 90-day mortality and incidence of adverse events (AEs). Propensity-score matching and Cox proportional hazards analyses were performed. RESULTS A total of 429 MRSA-BSIs were included: 133 in the ceftaroline group and 296 in the SOC group. More patients in the ceftaroline group had a Sequential Organ Failure Assessment score >2 (51.1% vs. 36.5%; p < 0.01), complicated BSI (66.2% vs. 42.2%; p < 0.01), infective endocarditis (18.8% vs. 6.4%; p < 0.01) and prescribed in combination treatment (65.4% vs. 11.5%; p < 0.01), with no statistically significant differences in 30-day mortality: 23.3% ceftaroline (95% CI, 16.1-30.5%) vs. 16.2% SOC (95% CI, 12.0-20.4%), p 0.08. There were no statistically significant differences in 90-day mortality (33.1% ceftaroline vs. 26.7% SOC; p 0.17). After propensity-score matching, 105 patients treated with ceftaroline were matched with 105 controls: the 30-day mortality rates were 21.9% and 16.2% (p 0.38). Cox regression analysis of the entire cohort (n = 429) revealed that age (hazard ratio [HR], 1.05; 95% CI, 1.03-1.07) and Sequential Organ Failure Assessment score >2 (HR, 2.34; 95% CI, 1.50-3.65) were associated with 90-day mortality risk, although ceftaroline treatment did not demonstrate a significant effect (HR, 1.00; 95% CI, 0.97-1.02). Incidence of AEs was 12.0% in ceftaroline vs. 4.4% in the SOC group (p < 0.01). Most AEs occurred when ceftaroline was used in combination vs. monotherapy (17.2% vs. 2.2%; p 0.01). DISCUSSION Ceftaroline was an effective treatment for MRSA-BSIs but was commonly prescribed in combination showing a higher incidence of AEs.
Collapse
Affiliation(s)
- Sofía de la Villa
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | - Francesc Escrihuela-Vidal
- Infectious Diseases Department, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Nuria Fernández-Hidalgo
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Escudero-Sánchez
- CIBER de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, Madrid, Spain; Infectious Disease Department, Ramon y Cajal University Hospital, Madrid, Spain; Instituto de Salud Carlos III (IRYCIS), Madrid, Spain
| | - Itxasne Cabezón
- Infectious Diseases Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Lucía Boix-Palop
- Infectious Diseases Department, Hospital Universitari Mútua de Terrassa, Barcelona, Spain
| | - Beatriz Díaz-Pollán
- CIBER de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, Madrid, Spain; Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain
| | | | - María José García-País
- Internal Medicine Department, Hospital Universitario Lucus Augusti, Lugo, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - María Teresa Pérez-Rodríguez
- Infectious Diseases Unit, Internal Medicine Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain; Galicia Sur Health Research Institute, Vigo, Spain
| | - Ángela Crespo
- Internal Medicine Department, Hospital Universitario de León, León, Spain
| | - Luis Buzón-Martín
- Infectious Diseases Department, Complejo Asistencial Universitario de Burgos, Burgos, Spain
| | - Oscar Sanz-Peláez
- Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | | | - Silvana Fiorante
- Internal Medicine Department, Hospital El Escorial, Madrid, Spain
| | - Patricia Muñoz
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Enfermedades Respiratorias, CIBERES, Instituto de Salud Carlos III, Madrid, Spain; Medicine Department, School of Medicine, Universidad Complutense de Madrid, Spain
| |
Collapse
|
3
|
Strömdahl M, Hagman K, Hedman K, Westman A, Hedenstierna M, Ursing J. Time to Staphylococcus aureus Blood Culture Positivity as a Risk Marker of Infective Endocarditis: A Retrospective Cohort Study. Clin Infect Dis 2025; 80:727-734. [PMID: 39707905 PMCID: PMC12043066 DOI: 10.1093/cid/ciae628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Endocarditis occurs in approximately 10%-15% of patients with Staphylococcus aureus bacteremia (SAB). Short time to positivity (TTP) in blood culture flasks has been linked to endocarditis in smaller studies. This study evaluated the association between TTP and endocarditis in SAB in a large cohort. METHODS Adult patients with ≥1 S. aureus positive blood culture treated at a tertiary-level, 500-bed hospital in Stockholm, Sweden between 2011-2021 were retrospectively identified. The primary outcome was the presence of infective endocarditis. RESULTS A total of 1703 episodes of SAB (23/1703 methicillin-resistant) in 1610 patients were included. Median age was 75 (interquartile range [IQR], 63-84) years and median Charlson comorbidity index score was 2 (IQR, 1-3). Echocardiography was performed in 1102/1703 (65%). Thirty-day mortality was 406/1703 (24%) and endocarditis was found in 154/1703 (9%). Median TTP was shorter in patients with endocarditis (9 [IQR, 7-12] hours) compared to patients without endocarditis (13 [interquartile range, 10-18] hours; P < .001). The risk of endocarditis decreased with 11% per hour (odds ratio [OR], 0.89 [95% confidence interval {CI}, .54-.92]; P < .001) in a univariate analysis using TTP as a continuous variable. In multivariate analysis, TTP <13 hours (the median) was independently associated with endocarditis (OR, 3.59 [95% CI, 2.35-5.3]; P < .001). The negative predictive value of TTP >13 hours for endocarditis was 96% (95% CI, 95%-97%). CONCLUSIONS Short TTP was associated with endocarditis. The negative predictive value of >95% suggests that TTP >13 hours can be used to risk-stratify patients with SAB.
Collapse
Affiliation(s)
- Martin Strömdahl
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm
| | - Karl Hagman
- Department of Infectious Diseases, Sahlgrenska University Hospital
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karolina Hedman
- Department of Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Westman
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
- Department of Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Hedenstierna
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm
| | - Johan Ursing
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm
| |
Collapse
|
4
|
Chua ZH, Tan SH, Mok HT, Teng CB, Vasoo S, Young BE. Antibiotic therapy and clinical outcomes of penicillin-susceptible Staphylococcus aureus (PSSA) bloodstream infection (BSI): a ten-year retrospective cohort study. Sci Rep 2025; 15:12103. [PMID: 40204889 PMCID: PMC11982392 DOI: 10.1038/s41598-025-96383-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
In recent years, the incidence of penicillin-susceptible S. aureus (PSSA) bloodstream infection (BSI) has increased worldwide. However, the preferred antibiotic remains uncertain due to concerns of inducible resistance to benzylpenicillin. We compared outcomes associated with benzylpenicillin versus other antibiotics and investigated risk factors influencing treatment failure. Patients were grouped into benzylpenicillin and non-benzylpenicillin beta-lactam treatment groups (including anti-staphylococcal penicillins and cephalosporins). The primary outcome was overall treatment failure (30-day all-cause mortality and/or 90-day relapse). Of 335 patients, 74 (22.09%) received benzylpenicillin and 261 (77.91%) received a non-benzylpenicillin beta-lactam. While rates of overall treatment failure (13.51% vs. 17.24%; P = 0.45) and occurrence of adverse drug events (6.76% vs. 7.66%; P = 0.79) were comparable to non-benzylpenicillin beta-lactams, benzylpenicillin showed faster microbiological clearance [3.00 days (IQR, 2.00-4.00 days) vs. 4.00 days (IQR, 3.00-5.00 days); P = 0.03] and fewer persistent infections (22.97% vs. 36.02%; P = 0.04), suggesting potential to improve patient outcomes. We also found that unknown source (aOR 4.63, 95% CI 1.47-14.64; P < 0.01) was associated with treatment failure, while review by Infectious Disease (ID) specialists (aOR 0.30, 95% CI 0.12-0.73; P = 0.01) was protective, stressing the importance of early ID referral and thorough source identification. This study highlights benzylpenicillin as an effective treatment for PSSA BSI.
Collapse
Affiliation(s)
- Zheng Hong Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Sock Hoon Tan
- Division of Pharmacy, Tan Tock Seng Hospital, Singapore, Singapore
| | - Hoi Tong Mok
- Division of Pharmacy, Tan Tock Seng Hospital, Singapore, Singapore
| | - Christine B Teng
- Division of Pharmacy, Tan Tock Seng Hospital, Singapore, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore, Singapore
| | - Shawn Vasoo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- National Center for Infectious Diseases, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Barnaby E Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- National Center for Infectious Diseases, Singapore, Singapore
| |
Collapse
|
5
|
Mayoryk S, Song X. Contact Precautions for Preventing Methicillin-Resistant Staphylococcus Aureus in Pediatric Healthcare Settings: Pros, Cons, and Future Actions. J Pediatric Infect Dis Soc 2025; 14:piaf023. [PMID: 40037535 DOI: 10.1093/jpids/piaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Although contact precautions (CP) have proven effective in protecting patients and healthcare providers and preventing the transmission of methicillin-resistant Staphylococcus aureus (MRSA) in healthcare facilities, pediatric patients under CP may experience unintended effects, including psychosocial stress and limited access to developmentally appropriate activities. Modifying or discontinuing the routine use of CP based on risk assessment results may enhance their overall benefits. Facilities that opt to modify or cease the routine use of CP should base their decisions on (1) compliance with the local regulations related to MRSA; (2) institutional compliance with CP for patients with MRSA infection and/or colonization, and (3) assessment of local MRSA data. Irrespective of any changes, all pediatric facilities should conduct ongoing assessments of MRSA-specific risks and monitor compliance with infection control practices. The results of these activities should guide the optimal use of CP to prevent MRSA infections among hospitalized pediatric patients.
Collapse
Affiliation(s)
| | - Xiaoyan Song
- Office of Infection Control and Epidemiology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Science, Washington, DC, USA
| |
Collapse
|
6
|
Tong SYC, Fowler VG, Skalla L, Holland TL. Management of Staphylococcus aureus Bacteremia: A Review. JAMA 2025:2832601. [PMID: 40193249 DOI: 10.1001/jama.2025.4288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Importance Staphylococcus aureus, a gram-positive bacterium, is the leading cause of death from bacteremia worldwide, with a case fatality rate of 15% to 30% and an estimated 300 000 deaths per year. Observations Staphylococcus aureus bacteremia causes metastatic infection in more than one-third of cases, including endocarditis (≈12%), septic arthritis (7%), vertebral osteomyelitis (≈4%), spinal epidural abscess, psoas abscess, splenic abscess, septic pulmonary emboli, and seeding of implantable medical devices. Patients with S aureus bacteremia commonly present with fever or symptoms from metastatic infection, such as pain in the back, joints, abdomen or extremities, and/or change in mental status. Risk factors include intravascular devices such as implantable cardiac devices and dialysis vascular catheters, recent surgical procedures, injection drug use, diabetes, and previous S aureus infection. Staphylococcus aureus bacteremia is detected with blood cultures. Prolonged S aureus bacteremia (≥48 hours) is associated with a 90-day mortality risk of 39%. All patients with S aureus bacteremia should undergo transthoracic echocardiography; transesophageal echocardiography should be performed in patients at high risk for endocarditis, such as those with persistent bacteremia, persistent fever, metastatic infection foci, or implantable cardiac devices. Other imaging modalities, such as computed tomography or magnetic resonance imaging, should be performed based on symptoms and localizing signs of metastatic infection. Staphylococcus aureus is categorized as methicillin-susceptible (MSSA) or methicillin-resistant (MRSA) based on susceptibility to β-lactam antibiotics. Initial treatment for S aureus bacteremia typically includes antibiotics active against MRSA such as vancomycin or daptomycin. Once antibiotic susceptibility results are available, antibiotics should be adjusted. Cefazolin or antistaphylococcal penicillins should be used for MSSA and vancomycin, daptomycin, or ceftobiprole for MRSA. Phase 3 trials for S aureus bacteremia demonstrated noninferiority of daptomycin to standard of care (treatment success, 53/120 [44%] vs 48/115 [42%]) and noninferiority of ceftobiprole to daptomycin (treatment success, 132/189 [70%] vs 136/198 [69%]). Source control is a critical component of treating S aureus bacteremia and may include removal of infected intravascular or implanted devices, drainage of abscesses, and surgical debridement. Conclusions and relevance Staphylococcus aureus bacteremia has a case fatality rate of 15% to 30% and causes 300 000 deaths per year worldwide. Empirical antibiotic treatment should include vancomycin or daptomycin, which are active against MRSA. Once S aureus susceptibilities are known, MSSA should be treated with cefazolin or an antistaphylococcal penicillin. Additional clinical management consists of identifying sites of metastatic infection and pursuing source control for identified foci of infection.
Collapse
Affiliation(s)
- Steven Y C Tong
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University, Durham, North Carolina
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Lesley Skalla
- Duke University Medical Center Library and Archives, Duke University School of Medicine, Durham, North Carolina
| | - Thomas L Holland
- Division of Infectious Diseases, Duke University, Durham, North Carolina
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
7
|
Verkaik D, Westgeest AC, Wu JL, Sigaloff KCE, Lambregts MMC, de Boer MGJ. Clinical revenues of selective use of [18F]-FDG-PET/CT scanning in the management of Staphylococcus aureus bacteremia. Eur J Clin Microbiol Infect Dis 2025; 44:895-904. [PMID: 39921696 PMCID: PMC11946999 DOI: 10.1007/s10096-025-05052-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE Although [18F]-FDG-PET/CT scanning (PET/CT) is recommended for managing Staphylococcus aureus bacteremia (SAB), its added value remains debated. This study investigated the clinical revenues of selective PET/CT use in SAB by considering three consequence-categories: detection of new infection foci, performance of new interventions, and alterations in antimicrobial therapy. METHODS All adult patients with ≥ 1 blood culture (BC) positive with Staphylococcus aureus admitted in a Dutch academic center between 201711 and 202311 were identified. Standard practice was to order PET/CT for patients with community acquired SAB and/or positive BCs after ≥ 48 h of treatment, or if multiple foci, or persistent fever, or endocarditis were present. Clinical- and laboratory data were obtained from electronic health records. Numbers-needed-to-scan (NNT-scan) were calculated for each consequence-category. Regression analyses were performed to identify variables correlated with consequence-bearing PET/CT. RESULTS Of 397 SAB patients, 143 (36%) underwent PET/CT. This led to detection of new foci in 73/143 patients (NNT-scan ≈ 2), new interventions in 33/143 patients (NNT-scan ≈ 4), and a change in antimicrobial therapy in 44/143 patients (NNT-scan ≈ 3). A CRP > 200 mg/L at presentation and positive follow-up BCs at 48 h were independently associated with interventions following PET/CT (adjusted OR and 95%CI 3.2 (1.2-8.3) and 2.6 (1.0-6.7) respectively). PET/CT results instigated changes in antimicrobial therapy predominantly in patients < 65 years and those with a CRP < 100 at presentation. CONCLUSIONS Selective PET/CT ordering in real-life practice resulted in a relatively low NNT-scan across all consequence-categories. Further research is warranted to optimize patient selection for PET/CT using clinical parameters or profiles.
Collapse
Affiliation(s)
- Dewi Verkaik
- Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - Annette C Westgeest
- Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - Jian Ling Wu
- Department of Internal Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Kim C E Sigaloff
- Department of Internal Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Merel M C Lambregts
- Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - Mark G J de Boer
- Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands.
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Room C-5-44, Albinusdreef 2, Leiden, 2333 RZ, The Netherlands.
| |
Collapse
|
8
|
Winkler P, Mao Y. Dual Delivery of Cells and Bioactive Molecules for Wound Healing Applications. Molecules 2025; 30:1577. [PMID: 40286165 PMCID: PMC11990229 DOI: 10.3390/molecules30071577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic wounds not only cause significant patient morbidity but also impose a substantial economic burden on the healthcare system. The primary barriers to wound healing include a deficiency of key modulatory factors needed to progress beyond the stalled inflammatory phase and an increased susceptibility to infections. While antimicrobial agents have traditionally been used to treat infections, stem cells have recently emerged as a promising therapy due to their regenerative properties, including the secretion of cytokines and immunomodulators that support wound healing. This study aims to develop an advanced dual-delivery system integrating stem cells and antibiotics. Stem cells have previously been delivered by encapsulation in gelatin methacrylate (GelMA) hydrogels. To explore a more effective delivery method, GelMA was processed into microparticles (MP). Compared to a bulk GelMA hydrogel (HG) encapsulation, GelMA MP supported greater cell growth and enhanced in vitro wound healing activity of human mesenchymal stem cells (hMSCs), likely due to a larger surface area for cell attachment and improved nutrient exchange. To incorporate antimicrobial properties, the broad-spectrum antibiotics penicillin/streptomycin (PS) were loaded into a bulk GelMA hydrogel, which was then cryo-milled into MPs to serve as carriers for hMSCs. To achieve a more sustained antibiotic release, gelatin nanoparticles (NP) were used as carriers for PS. PS was either incorporated during NP synthesis (NP+PS(S)) or absorbed into NP after synthesis (NP+PS(A)). MPs containing PS, NP+PS(S), or NP+PS(A) were tested for their cell carrier functions and antibacterial activities. The incorporation of PS did not compromise the cell-carrying function of MP configurations. The anti-S. aureus activity was detected in conditioned media from MPs for up to eight days-four days longer than from bulk HG containing PS. Notably, the presence of hMSCs prolonged the antimicrobial activity of MPs, suggesting a synergistic effect between stem cells and antibiotics. PS loaded via synthesis (NP+PS(S)) exhibited a delayed initial release, whereas PS loaded via absorption (NP+PS(A)) provided a more immediate release, with potential for sustained delivery. This study demonstrates the feasibility of a dual-delivery system integrating thera.
Collapse
Affiliation(s)
| | - Yong Mao
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA;
| |
Collapse
|
9
|
Zilberberg MD, Nathanson BH, Wagenaar R, Posthumus J, Shorr AF. Unfavorable Outcomes and Their Risk Factors in Hospitalized Patients with Staphylococcus aureus Bacteremia in the US: A Multicenter Retrospective Cohort Study, 2020-2022. Antibiotics (Basel) 2025; 14:326. [PMID: 40149136 PMCID: PMC11939448 DOI: 10.3390/antibiotics14030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: In the US, 120,000 cases of Staphylococcus aureus bacteremia (SAB) occur annually. Apart from mortality, little is known about other unfavorable outcomes (UOs). We developed a multifaceted definition for UOs in SAB and examined their incidence and predictors. Methods: We conducted a multicenter (~300 hospitals) retrospective cohort study between 2020 and 2022 of adult hospitalized patients with at least one blood culture (BC) positive for S. aureus. UOs were any of the following: hospital mortality, antibiotic escalation, persistently positive BCs, prolonged post-infection length of stay (LOS), 30-day readmission, and disease worsening. We compared the group with UOs to favorable outcomes (FOs). Regression models identified predictors of UOs. Results: Among 4080 patients with SAB, 2427 (59.5%) experienced a UO, most commonly 30-day readmission (42.0%) and antibiotic escalation (37.7%). Those with UOs more frequently had septic shock at admission (5.7% vs. 1.2%), requiring the ICU (18.8% vs. 14.7%) and dialysis (4.4% vs. 1.9%) prior to SAB onset. Community-onset SAB predominated in both groups, with more complicated SAB in the UO group (39.8% vs. 22.3%). Vancomycin use was similar, while daptomycin was more common in the UO group (8.5% vs. 3.0%). Variables with the highest odds ratios predicting a UO were septic shock on admission (3.498, 95% CI 2.145, 5.704), empiric daptomycin (2.723, 95% CI 1.943, 3.821), and complicated SAB (2.476, 95% CI 2.047, 2.994). Conclusions: UOs occur frequently in the setting of SAB. A broader perspective exploring issues other than mortality demonstrates the substantial implications of SAB both for patients and healthcare systems. Select clinical variables are associated with UOs, some of which may not be modifiable.
Collapse
Affiliation(s)
| | | | - Rolf Wagenaar
- Basilea Pharmaceutica International Ltd., 4123 Allschwil, Switzerland; (R.W.); (J.P.)
| | - Jan Posthumus
- Basilea Pharmaceutica International Ltd., 4123 Allschwil, Switzerland; (R.W.); (J.P.)
| | | |
Collapse
|
10
|
Dufresne K, Tuffs SW, Walton NR, Kasper KJ, Mohorovic I, Hasan F, Bentall T, Heinrichs DE, Delport J, Mele TS, McCormick JK. Evidence that staphylococcal superantigens promote within-patient bacterial persistence following post-operative surgical site infection. Infect Immun 2025; 93:e0040724. [PMID: 39878522 PMCID: PMC11895439 DOI: 10.1128/iai.00407-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Staphylococcus aureus is a predominant cause of post-operative surgical site infections and persistent bacteremia. Here, we describe a patient who experienced three episodes of S. aureus infection over a period of 4 months following a total knee arthroplasty. The initial bloodstream isolate (SAB-0429) was a clonal complex 5 (CC5) and methicillin-resistant S. aureus (MRSA), whereas two subsequent isolates (SAB-0485 and SAB-0495) were CC5 isolates but methicillin-sensitive S. aureus. The two latter isolates harbored a plasmid encoding three superantigen genes that were not present in the primary MRSA isolate. SAB-0485 and SAB-0495 both expressed the plasmid-encoded staphylococcal enterotoxin R exotoxin and demonstrated increased superantigen activity compared with SAB-0429. Compared to SAB-0429, the latter isolates also demonstrated an increased bacterial burden in a mouse bacteremia model that was dependent on increased interferon-γ production. Curing of the plasmid from SAB-0485 reduced this virulence phenotype. These findings suggest that the superantigen exotoxins may provide a selective advantage in chronic post-surgical infections.IMPORTANCEIn this study, we investigated bacterial isolates from a patient who experienced three recurrent S. aureus infections over a 4 month period following total knee arthroplasty. Genomic and phenotypic characterization of these isolates revealed that they all belonged to clonal complex 5, yet the latter two strains contained an additional plasmid encoding superantigen exotoxins. Subsequent experimental infection experiments in mice demonstrated that the plasmid-encoded superantigens exacerbated bacteremia by promoting liver abscess formation. These experiments suggest that despite appropriate antibiotic therapy, bacterial superantigens may be able to promote persistent infection following post-surgery.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Stephen W. Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Nicholas R. Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katherine J. Kasper
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Ivor Mohorovic
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Farah Hasan
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tracey Bentall
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Johan Delport
- Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Tina S. Mele
- Division of Critical Care Medicine, Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, London Health Sciences Centre, University Hospital, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
11
|
Zhang XL, Liu J, Fu P, Wang YX, Fan PP, Zhou JL, Xiang XQ, Shen HL, Liu TY, Zhang YY, Zhu T, Zhang CY, Wang CQ, Lu GP, Yan GF. Epidemiological profile and antimicrobial resistance trends of Staphylococcus aureus in Chinese pediatric intensive care units from 2016 to 2022: a multi-center retrospective study. BMC Infect Dis 2025; 25:298. [PMID: 40025450 PMCID: PMC11874852 DOI: 10.1186/s12879-025-10704-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
OBJECTIVE This study aimed to analyze the profiles and evolution of Staphylococcus aureus in the pediatric intensive care units (PICUs) of 17 hospitals in China from 2016 to 2022. METHODS Susceptibility testing was performed to bacterial strains with a uniform monitoring protocol, which was provided by the US Clinical and Laboratory Standards Institute (CLSI) and used by the China Antimicrobial Surveillance Network (CHINET). The results were interpreted in accordance with the performance standards for antimicrobial susceptibility testing issued by the US Clinical and Laboratory Standards Institute. RESULTS Twenty-six thousand six hundred thirteen bacterial strains were isolated from 17 PICUs in China from 2016 to 2022, 3,147 of which were Staphylococcus aureus, ranking second among etiological agents of infections from PICUs. In 2022, Staphylococcus aureus had the highest detection rate, being 36.19%. And in 2021, MRSA had the highest detection rate, being 10.35% in Staphylococcus aureus. There were statistically significant differences in the annual detection rate of gram-positive bacteria, Staphylococcus aureus and MRSA between the years from 2016 to 2022 (P < 0.05). More males were found with Staphylococcus aureus or methicillin-resistant Staphylococcus aureus, but there were no statistical differences in gender distribution between any two years (P < 0.05). The top 3 highest detection rate of Staphylococcus aureus in age groups were infants (1244, 39.7%), toddlers (741, 23.7%), and children at school age and older (731, 23.4%). For MRSA, The top 3 in age groups were infants (91, 38.9%), children at school age and older (87, 29.1%), and toddlers (48, 20.5%). The detection rate of Staphylococcus aureus was statistically different in the distribution of age stratification (P < 0.05). There was no statistically significant difference in these two aspects of MRSA (P > 0.05). The top 3 highest detection rate of Staphylococcus aureus in infected sites were the lower respiratory tract (2,552, 81.7%), bloodstream (217, 6.5%), and skin wounds (110, 3.9%). For MRSA, The top 3 in infected sites were the lower respiratory tract (156, 77.9%), skin wounds (47, 8.8%), and bloodstream (15, 6.6%). The detection rate of Staphylococcus aureus and MRSA was statistically different in the distribution of infected sites (P < 0.05). All the strains of Staphylococcus aureus were sensitive to tigecycline, nitrofurantoin, vancomycin, and linezolid. The resistant rate of Staphylococcus aureus, to penicillin G was as high as 87.5% at least, to erythromycin was as high as 51.8% at least, to benzocillin was as high as 38.0% at least, to cefoxitin was as high as 35.5% at least, and to clindamycin was as high as 32.7% at least. All the strains of MRSA were sensitive to vancomycin, linezolid, quinupristin/dalfopristin, and tigecycline. Of these 234 strains of MRSA, 179 (76.5%) were resistant to erythromycin, 116 (49.6%) to clindamycin, 39 (16.7%) to tetracycline, 29 (12.4%) to levofloxacin, 27 (11.5%) to ciprofloxacin, 27 (11.5%) to moxifloxacin, 14 (6.0%) to TMP-SMX, eight (3.4%) to rifampicin, and six (2.6%) to gentamicin. CONCLUSIONS Staphylococcus aureus is the most common gram-positive bacterium in PICUs. Infants are most likely to be infected by Staphylococcus aureus and methicillin-resistant Staphylococcus aureus. And the lower respiratory tract is the most common infected site of Staphylococcus aureus. Staphylococcus aureus has a high resistant rates to commonly used antimicriobials in pediatrics, but no strains resistant to vancomycin and/or linezolid were found. When considering Staphylococcus aureus infection clinically, it is necessary to select antimicrobials reasonably based on the patient's age, infected site and local epidemiological characteristics.
Collapse
Grants
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- 2021YFC2701800, 2021YFC2701805 the National Key Research and Development Program of China
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- No. 202140442 the Scientific Research Project of Shanghai Municipal Health Commission
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
- 2023ZDFC0103 the Shanghai Municipal Health System major supports discipline projects
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Jing Liu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Pan Fu
- Department of Clinical Laboratory, Lab of Microbiology, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Yi-Xue Wang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Pan-Pan Fan
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Jin-Lan Zhou
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Xian-Qi Xiang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Hui-Li Shen
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Ting-Yan Liu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Ying-Ying Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Ting Zhu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Cai-Yan Zhang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China
| | - Chuan-Qing Wang
- Department of Clinical Laboratory, Lab of Microbiology, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China.
| | - Guo-Ping Lu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China.
| | - Gang-Feng Yan
- Department of Pediatric Intensive Care Unit, Children's Hospital of Fudan University, National Center for Children's Health, 399 Wanyuan Road, Shanghai, 201102, P.R. China.
| |
Collapse
|
12
|
Aung MS, Osada M, Urushibara N, Kawaguchiya M, Ohashi N, Hirose M, Ito M, Yamada K, Tada K, Kobayashi N. Molecular characterization of methicillin-susceptible/resistant Staphylococcus aureus from bloodstream infections in northern Japan: The dominance of CC1-MRSA-IV, the emergence of human-associated ST398 and livestock-associated CC20 and CC97 MSSA. J Glob Antimicrob Resist 2025; 41:77-87. [PMID: 39725318 DOI: 10.1016/j.jgar.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/26/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Staphylococcus aureus (S. aureus) is a major cause of bloodstream infections. The recent epidemiological features and antimicrobial resistance trend were analysed for methicillin-resistant and susceptible S. aureus (MRSA/MSSA) isolates from blood samples in people from northern Japan. METHODS The S. aureus isolates from blood culture were screened by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) and genotyped by the schemes of multilocus sequence typing (MLST), coa, agr, spa, and SCCmec types. Antimicrobial resistance genes and virulence factors were detected by multiplex/uniplex polymerase chain reaction (PCR). Antimicrobial susceptibility was examined using a broth microdilution test. RESULTS A total of 301 isolates (163 MRSA and 138 MSSA) were isolated from bloodstream infections in 2023 (from April to December). The MRSA isolates were classified into three groups, that is, clonal complexes (CC)1-SCCmec-IV (CC1-IV) (52%), CC5-II (36%), and CC8-IV (12%). The prevalence of CC1 was significantly higher than those in our previous studies (2017-2021). Four CC8-IVa isolates with PVL genes on ΦSa2usa were considered to be the USA300 clone (sequence type [ST]8/spa-t008/coa IIIa/agr I) or its variants that were genotyped as those closely related to ST8/t008 or lacking arginine catabolic mobile element (ACME). In contrast, MSSA was genetically highly divergent and classified into 22 STs, with CC1 (ST1 and ST188) being the most common (25%). It was notable that 29 MSSA isolates (21%) were classified into livestock-associated (LA) genotypes, ST20, ST97, and CC398 (ST398 and ST291). Genetic characterization of the CC398 isolates suggested that these belong to human-adapted MSSA clones. CONCLUSIONS The present study revealed the increasing trend of CC1 MRSA surpassing CC5, and the emergence of MSSA representing human-adapted CC398, and LA types ST97 and ST20 from bloodstream infections in people in Japan. © 2024 The Author(s). Published by Elsevier Ltd on behalf of International Society for Antimicrobial Chemotherapy.
Collapse
Affiliation(s)
- Meiji Soe Aung
- Department of Hygiene, Sapporo Medical University School of Medicine, Hokkaido, Sapporo, Japan.
| | - Masako Osada
- Master's Program in Midwifery, Tenshi College Graduate School, Sapporo, Japan
| | - Noriko Urushibara
- Department of Hygiene, Sapporo Medical University School of Medicine, Hokkaido, Sapporo, Japan
| | - Mitsuyo Kawaguchiya
- Department of Hygiene, Sapporo Medical University School of Medicine, Hokkaido, Sapporo, Japan
| | - Nobuhide Ohashi
- Department of Hygiene, Sapporo Medical University School of Medicine, Hokkaido, Sapporo, Japan
| | - Mina Hirose
- Division of Pediatric Dentistry, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Masahiko Ito
- Sapporo Clinical Laboratory, Inc., Sapporo, Japan
| | | | - Kousuke Tada
- Sapporo Clinical Laboratory, Inc., Sapporo, Japan
| | - Nobumichi Kobayashi
- Department of Hygiene, Sapporo Medical University School of Medicine, Hokkaido, Sapporo, Japan
| |
Collapse
|
13
|
Hisano H, Shoji K, Matsui T, Kato H, Fukui K, Kano M, Yamada Y, Gocho Y, Ishiguro A. Subsequent bacteremia associated with intravascular catheter colonization with Staphylococcus aureus, Gram-negative rods, and Candida species in children. J Infect Chemother 2025; 31:102600. [PMID: 39725073 DOI: 10.1016/j.jiac.2024.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Subsequent bacteremia developed in 14 % of patients with positive catheter tip cultures but concurrent negative blood cultures. The occurrence of subsequent bacteremia did not differ significantly by pathogens (Staphylococcus aureus, Gram-negative rods [GNR], and Candida spp.). Mortality was higher in patients with GNR and Candida spp. than with S. aureus.
Collapse
Affiliation(s)
- Hina Hisano
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Kensuke Shoji
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan.
| | - Toshihiro Matsui
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroki Kato
- Division of Critical Care Medicine, Department of Critical Care and Anesthesia, National Center for Child Health and Development, Tokyo, Japan
| | - Kana Fukui
- Division of Neonatology, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Motohiro Kano
- Division of Surgery, Department of Surgical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Yuji Yamada
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Yoshihiro Gocho
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Akira Ishiguro
- Center for Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
14
|
Weikel CA, Pisciotta JM. Recreational Facilities as Reservoirs for Multidrug-Resistant Staphylococcus aureus. Microbiol Immunol 2025; 69:168-173. [PMID: 39716386 DOI: 10.1111/1348-0421.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/14/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Antibiotic-resistant pathogens in public settings present a growing risk to human health. Staphylococcus aureus often asymptomatically colonizes human skin, while virulent strains cause soft tissue infections, osteomyelitis, endocarditis, and are associated with cystic fibrosis. Here we investigated the presence and distribution of multidrug-resistant S. aureus on exercise equipment in university recreation facilities. Equipment sampled included barbells (n = 10), dumbbell handles (n = 15), kettle bell handles (n = 5), ellipticals (n = 5), treadmills (n = 5), cable attachments (n = 5). Mannitol salt agar, CHROMagar-MRSA, Gram staining and latex agglutination testing were useds to isolate and identify S. aureus, including methicillin-resistant S. aureus. Kirby-Bauer disc-diffusion assay was utilized to determine antibiotic susceptibility profiles. Results show 42% of 456 S. aureus isolates from 45 different equipment surfaces were ampicillin resistant. Of 60 representative ampicillin-resistant isolates, 92% were resistant to additional antibiotics with the majority resistant to erythromycin (40%) and sulfisoxazole (75%). Multidrug resistance to three or more drugs was observed in 73% of the ampicillin-resistant subpopulation. These results indicate recreational facilities may serve as reservoirs for multi-drug resistant S. aureus including methicillin-resistant S. aureus (MRSA) and regular disinfection of equipment is warranted for safeguarding public health.
Collapse
Affiliation(s)
- Chase A Weikel
- Department of Biology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| | - John M Pisciotta
- Department of Biology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| |
Collapse
|
15
|
Scheier TC, Schreiber PW, Yao X, Eikelboom JW, Brugger SD, Mertz D. Efficacy of Staphylococcus aureus nasal decolonization at hospital admission on reduction of any infections within 90 days - a systematic review and meta-analysis. J Hosp Infect 2025; 159:1-10. [PMID: 39983921 DOI: 10.1016/j.jhin.2025.01.017] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/23/2025]
Abstract
Infections with Staphylococcus aureus are associated with excess mortality and costs. Simple interventions, such as decolonization protocols, might help to reduce subsequent infections, especially in settings in which S. aureus is of major clinical relevance, such as healthcare-associated infections. The aim of this research was to determine whether S. aureus decolonization protocols, using a nasal ointment and applied at hospital admission, reduce the risk of infection. MEDLINE, EMBASE and clinicaltrials.gov were searched for all randomized controlled trials investigating the use of nasal ointments for decolonization for patients at hospital admission and reporting on infections within 90 days. Data were pooled as risk ratios using a random-effects model. The Cochrane RoB 2 tool and GRADE were used to assess the risk of bias and quality of evidence. Four (0.5%) of 833 screened studies with a total of 4150 patients met eligibility criteria. The overall certainty of evidence ranged from low to moderate across outcomes. Nasal S. aureus decolonization probably reduces infections (6.9% vs 9.5%; rate ratio (RR): 0.73 (95% confidence interval (CI) 0.57; 0.93)) and may increase microbiological eradication (82.7% vs 55.2%; RR: 1.80 (95% CI 0.73; 4.44)) compared with placebo. It may not reduce mortality (2.5 vs 2.4; RR: 1.02 (95% CI 0.67; 1.54)) but also not lead to increased adverse effects (0.7% vs 0.6%; RR: 1.01 (95% CI 0.45; 2.30)). Results were consistent across subgroups and sensitivity analyses. In conclusion, nasal decolonization at acute-care hospital admission probably reduces the risk of infections and may result in a large increase in microbiological eradication. It may not reduce mortality and also not result in an increased number of adverse events.
Collapse
Affiliation(s)
- T C Scheier
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada.
| | - P W Schreiber
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - X Yao
- Department of Health Research Methodology, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - J W Eikelboom
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada; McMaster University, Hamilton, ON, Canada
| | - S D Brugger
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - D Mertz
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada; Department of Health Research Methodology, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada; Division of Infectious Diseases, Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
Bergersen KV, Zheng Y, Rossetti M, Ruffin F, Pickering H, Parmar R, Sunga G, Chan LC, Gjertson D, Fowler VG, Yeaman MR, Reed EF. Early cytokine signatures and clinical phenotypes discriminate persistent from resolving MRSA bacteremia. BMC Infect Dis 2025; 25:231. [PMID: 39966757 PMCID: PMC11834594 DOI: 10.1186/s12879-025-10620-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Staphylococcus aureus bacteremia (SAB) is a prevalent life-threatening infection often caused by methicillin-resistant S. aureus (MRSA). Up to 30% of SAB patients fail to clear infection even with gold-standard anti-MRSA antibiotics. This phenomenon is termed antibiotic-persistent MRSA bacteremia (APMB). The mechanisms driving APMB are complex and involve host phenotypes significantly impacting the immune response. Thus, defining early immune signatures and clinical phenotypes that differentiate APMB from antibiotic resolving (AR)MB could aid therapeutic success. METHODS We assessed 38 circulating cytokines and chemokines using affinity proteomics in 74 matched pairs of vancomycin-treated SAB cases identified as ARMB or APMB after 5 days of blood culture. RESULTS Unsupervised hierarchical clustering segregated APMB from ARMB based on differential levels of IL-10, IL-12p40, IL-13, CCL4, and TGFα. Additionally, CXCL1, CCL22 and IL-17A significantly differed between APMB and ARMB when correlated with diabetes, dialysis, metastatic infection, or cardiac vegetation. Combining immune signatures with these relevant clinical phenotypes sharply increased accuracy of discriminating APMB outcome to 79.1% via logistic regression modeling. Finally, classification-regression tree analysis revealed explicit analyte thresholds associated with APMB outcome at presentation especially in patients with metastatic infection. CONCLUSIONS Collectively, this study identifies previously unrecognized cytokine and chemokine signatures that distinguish APMB and ARMB at presentation and in the context of host clinical characteristics associated with increased disease severity. Validation of a biomarker signature that accurately predicts outcomes could guide early therapeutic strategies and interventions to reduce risks of persistent SAB that are associated with worsened morbidity and mortality.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Ying Zheng
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Maura Rossetti
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gemalene Sunga
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Liana C Chan
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David Gjertson
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA.
- Duke Clinical Research Institute, Duke University, Durham, NC, USA.
| | - Michael R Yeaman
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA.
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Division of Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Divisions of Molecular Medicine and Infectious Diseases, David Geffen School of Medicine and Harbor-UCLA Medical Center, 1124 West Carson Street, Building MRL / 250, Torrance, CA, 90502, USA.
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA.
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Karki P, Ke Y, Zhang CO, Promnares K, Li Y, Williams CH, Hong CC, Birukov KG, Birukova AA. Inhibition of proton sensor GPR68 suppresses endothelial dysfunction and acute lung injury caused by Staphylococcus aureus bacterial particles. FASEB J 2025; 39:e70333. [PMID: 39907683 DOI: 10.1096/fj.202401947r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Lung bacterial infections, including hospital-acquired pneumonia, remain a serious problem for public health. Endothelial cell (EC) exposure to heat-killed Staphylococcus aureus (HKSA) represents a clinical scenario of high titers of killed bacterial particles present in the host after antibiotic therapy, which triggers inflammatory cascades, cytokine storms, and EC dysfunction leading to acute lung injury (ALI). GPR68 is a member of the proton-sensing G protein-coupled receptor family. Acting as a pH sensor, GPR68 becomes activated upon pH reduction and contributes to pathologic cell responses by activating ER stress and unfolded protein response. This study investigated the role of GPR68 in HKSA-induced EC dysfunction and HKSA-induced ALI. HKSA robustly increased GPR68 mRNA levels in human pulmonary EC and directly stimulated GPR68 activity. A selective GPR68 small molecule inhibitor, OGM-8345, attenuated HKSA-induced EC permeability and protected cell junction integrity. OGM-8345 inhibited HKSA-induced activation of inflammatory genes TNF-α, IL-6, IL-8, IL-1β, and CXCL5 and decreased cytokine secretion by HKSA-challenged EC. Co-treatment with the GPR68 activator Ogerin or medium acidification to pH 6.5 augmented HKSA-induced EC dysfunction, which was rescued by OGM-8345. Intratracheal HKSA injection increased vascular leak and lung inflammation in mice which were monitored by lung Evans blue extravasation, increased cell and protein count in bronchoalveolar lavage, and mRNA expression of inflammatory genes. ALI and barrier dysfunction was attenuated by OGM-8345. We show for the first time the role of GPR68 in mediating HKSA-induced lung injury and the strong potential for OGM-8345 as a therapeutic treatment of bacterial pathogen-induced ALI associated with tissue acidification.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chen-Ou Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kamoltip Promnares
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yue Li
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Charles H Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Charles C Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna A Birukova
- Division of Pulmonary and Critical Care, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Wang G, Wen J, Tian Z, Zhou H, Peng X, Zhang P, Li Z. Piceatannol and its analogues alleviate Staphylococcus aureus pathogenesis by targeting β-lactamase biofilms and α-hemolysin. Sci Rep 2025; 15:5551. [PMID: 39952994 PMCID: PMC11828952 DOI: 10.1038/s41598-025-89654-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
β-Lactamases, biofilms and toxins pose challenges for combating S. aureus infection. Thus, identifying inhibitors that can restore bacterial sensitivity to antibiotics, destroy biofilms, and antitoxins is a promising way to develop alternative agents. In this study, we found that piceatannol (pit), along with its analogues resveratrol (ret) and pterostilbene (pts) bind with β-lactamase to inhibit its activity, and 96TYR, 58ILE and 66LYS were identified as the critical binding residues. Pit and pts reduced the ampicillin (Amp) and gentamicin (Gm) MICs against S. aureus and enhanced the bactericidal ability of Amp. Pit and its analogues inhibited the formation of S. aureus USA300. In addition, the pit analogues bound with α-hemolysin and suppressed the hemolysis activity of the bacterial culture supernatant. The mechanism analysis revealed that pit exhibited multiple potential binding modes with α-hemolysin. Pit significantly decreased the cytotoxicity and the adherence effect mediated by S. aureus and increased the survival rate of Galleria mellonella that infected with S. aureus, the pathological tissue damage of Galleria mellonella was alleviated by treatment with pit alone or in combination with Amp. Taken together, our findings identify promising compounds for the development of S. aureus infection inhibitors.
Collapse
Affiliation(s)
- Guizhen Wang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Jingyao Wen
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Zizeng Tian
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Hanbing Zhou
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Xinli Peng
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Peigang Zhang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China
| | - Zhandong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, 130052, China.
- Jilin Engineering Normal University, Changchun, 130052, China.
| |
Collapse
|
19
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
20
|
Al Shaikhli H, Akins RL, Stover KR, Barber KE. Exploring combination treatment options for persistent methicillin-susceptible Staphylococcus aureus bacteremia. Am J Health Syst Pharm 2025; 82:150-163. [PMID: 39230345 DOI: 10.1093/ajhp/zxae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Indexed: 09/05/2024] Open
Abstract
PURPOSE This review explores the management of persistent methicillin-susceptible Staphylococcus aureus bacteremia (SAB), emphasizing the importance of timely intervention due to SAB's association with metastatic dissemination, relapse, and mortality. SUMMARY The literature analysis first delves into risk factors for persistent SAB, highlighting the need for effective treatment strategies. The subsequent focus is on combination strategies for persistent SAB. Daptomycin, ertapenem, ceftaroline, fosfomycin, rifampin, and gentamicin are explored as adjuncts to cefazolin or antistaphylococcal penicillins. Daptomycin combination therapy is assessed through in vivo and clinical studies, indicating potential benefits, especially with higher-risk sources of infection. Ertapenem combination therapy has been demonstrated to have a synergistic effect with cefazolin, presenting a viable salvage option. Rifampin's ability to penetrate biofilm is examined, with discussion of inconclusive evidence on mortality benefits. The review also considers stewardship implications, discussing concerns such as resistance emergence, adverse events, and increased costs associated with combination therapy. Mathematical models suggest combination therapy as an effective approach to prevent resistance. Adverse events vary with each combination, and duration of therapy remains diverse across studies in the absence of well-established dosing guidelines. CONCLUSION The review provides a thorough exploration of the literature on treatment of persistent SAB, underscoring the need for evidence-based guidelines, further studies, and clinical judgment in tailoring treatment strategies. The multifaceted analysis contributes valuable insights for clinicians managing this challenging condition.
Collapse
|
21
|
Hanamoto S, Fujimoto Y, Sugiura K, Haga T. Prevalence of Methicillin-Resistant Staphylococcus aureus in Livestock in Japan: A Systematic Review and Meta-Analysis. EPIDEMIOLOGIA 2025; 6:3. [PMID: 39982255 PMCID: PMC11844002 DOI: 10.3390/epidemiologia6010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 02/22/2025] Open
Abstract
Background: Methicillin-resistant Staphylococcus aureus (MRSA) is an important health issue that is estimated to have caused 130,000 deaths worldwide in 2021. As more instances of cross-species transmission of MRSA have been reported, concerns have been raised regarding the spread of livestock-associated MRSA to humans. The prevalence of MRSA in livestock varies globally. This study systematically reviews the prevalence of MRSA at the farm and animal levels in Japan. Methods: Relevant studies published in English or Japanese between 2000 and 2023 were retrieved from four databases. Pooled prevalences at the farm and animal levels in Japanese farms were calculated using a random-effects model. Subgroup and meta-regression analyses were also performed to explore sources of heterogeneity. Results: The 13 studies included in this meta-analysis yielded an MRSA prevalence of 3.54% (95% confidence interval [CI] 0.65-8.30%) at the individual pig level, 13.07% (95% CI 5.42-23.04%) at the pig farm level, 0.0% (95% CI 0.00-0.04%) at the individual cattle level, and 0% (95% CI 0.00-0.44%) at the individual chicken level. A significant increase in MRSA prevalence over time was evident at the individual pig level by both subgroup analysis (p = 0.020) and meta-regression (p = 0.019). Conclusions: Our results indicated that the proportion of pigs that can be a source of MRSA infection in humans has been steadily increasing in Japan. Despite some limitations, our findings strongly imply a need for more attention to pig-to-human MRSA transmission in Japan.
Collapse
Affiliation(s)
- Sayoko Hanamoto
- Division of Infection Control and Disease Prevention, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
| | - Yuri Fujimoto
- Laboratory of OSG Veterinary Science for Global Disease Management, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Katsuaki Sugiura
- Laboratory of Environmental Science for Sustainable Development, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
- Nippon Institute for Biological Science, Ome, Tokyo 198-0024, Japan
| | - Takeshi Haga
- Division of Infection Control and Disease Prevention, Department of Veterinary Medical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan;
- Laboratory of OSG Veterinary Science for Global Disease Management, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
22
|
Shaaban F, Salem Sokhn E, Khalil C, Saleh FA. Antimicrobial activity of adipose-derived mesenchymal stromal cell secretome against methicillin-resistant Staphylococcus aureus. Stem Cell Res Ther 2025; 16:21. [PMID: 39849590 PMCID: PMC11755800 DOI: 10.1186/s13287-025-04138-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/10/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) is still a growing concern in the field of antimicrobial resistance due to its resistance to conventional antibiotics and its association with high mortality rates. Mesenchymal stromal cells (MSCs) have been shown as a promising and attractive alternative treatment for bacterial infections, due to their antibacterial properties and potential to bypass traditional resistance mechanisms. This study aims to shed light on the antibacterial potential of adipose-derived mesenchymal stromal cell (AD-MSC) secretome against clinical isolates of Staphylococcus spp., including MRSA strains. METHODS Using the Kirby-Bauer disk diffusion method, broth microdilution assays, and colony-forming unit (CFU) counting, the antibacterial activity of AD-MSC secretome was assessed. These tests were first conducted on Staphylococcus (S.) aureus ATCC 25923, then on 73 clinical isolates including MRSA strains. Further molecular analysis was performed to identify resistant genes in MRSA isolates. RESULTS The AD-MSC secretome demonstrated significant antibacterial activity against S. aureus ATCC with a 32 mm inhibition zone. 96% of the collected staphylococcal clinical isolates showed susceptibility to the secretome with 87.5% inhibition observed in MRSA isolates, along with 100% in MSSA, MSSE, and MRSE strains. Molecular analysis revealed that MRSA strains resistant to the secretome harbored mecA, ermA, and ermB genes. Additionally, the mecA-negative MRSA strains remained susceptible to the secretome, suggesting alternative resistance mechanisms. CONCLUSION These findings emphasize the ability of AD-MSCs secretome as a promising alternative for treating antibiotic-resistant infections, with potential applications in combating MRSA. However, further research is required to explore its clinical applications as a complementary or standalone therapy for resistant infections.
Collapse
Affiliation(s)
- Fatimah Shaaban
- Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Elie Salem Sokhn
- Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Charbel Khalil
- Reviva Regenerative Medicine Center, Bsalim, Lebanon
- Bone Marrow Transplant Unit, Burjeel Medical City, Abu Dhabi, UAE
- School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Fatima A Saleh
- Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon.
| |
Collapse
|
23
|
Hicks AS, Dolan MA, Shah MD, Elwood SE, Platts-Mills JA, Madden GR, Elliott ZS, Eby JC. Early initiation of ceftaroline-based combination therapy for methicillin-resistant Staphylococcus aureus bacteremia. Ann Clin Microbiol Antimicrob 2025; 24:3. [PMID: 39806392 PMCID: PMC11730128 DOI: 10.1186/s12941-025-00773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
PURPOSE Monotherapy with vancomycin or daptomycin remains guideline-based care for methicillin-resistant Staphylococcus aureus bacteremia (MRSA-B) despite concerns regarding efficacy. Limited data support potential benefit of combination therapy with ceftaroline as initial therapy. We present an assessment of outcomes of patients initiated on early combination therapy for MRSA-B. METHODS This was a single-center, retrospective study of adult patients admitted with MRSA-B between July 1, 2017 and April 31, 2023. During this period, there was a change in institutional practice from routine administration of monotherapy to initial combination therapy for most patients with MRSA-B. Combination therapy included vancomycin or daptomycin plus ceftaroline within 72 h of index blood culture and monotherapy was vancomycin or daptomycin alone. The primary outcome was a composite of persistent bacteremia, 30-day all-cause mortality, and 30-day bacteremia recurrence. Time to microbiological cure and safety outcomes were assessed. All outcomes were assessed using propensity score-weighted logistic regression. RESULTS Of 213 patients included, 118 received monotherapy (115 vancomycin, 3 daptomycin) and 95 received combination therapy with ceftaroline (76 vancomycin, 19 daptomycin). The mean time from MRSA-positive molecular diagnostic blood culture result to combination therapy was 12.1 h. There was no difference between groups for the primary composite outcome (OR 1.58, 95% CI 0.60, 4.18). Time to microbiological cure was longer with combination therapy (mean difference 1.50 days, 95% CI 0.60, 2.41). Adverse event rates were similar in both groups. CONCLUSIONS Early initiation of ceftaroline-based combination therapy did not improve outcomes for patients with MRSA-B in comparison to monotherapy therapy.
Collapse
Affiliation(s)
- Addison S Hicks
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA.
| | - Mackenzie A Dolan
- Department of Pharmacy, University of Virginia Health, Charlottesville, Virginia, USA
| | - Megan D Shah
- Department of Pharmacy, University of Virginia Health, Charlottesville, Virginia, USA
| | - Sarah E Elwood
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA
| | - Gregory R Madden
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA
| | - Zachary S Elliott
- Department of Pharmacy, University of Virginia Health, Charlottesville, Virginia, USA
| | - Joshua C Eby
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA.
| |
Collapse
|
24
|
Fernandes P, Maia Oliveira J, Rocha AR, Carvalho S, Vaz J. Isolated Pulmonary Valve Infective Endocarditis With Persistent Staphylococcus aureus Bacteremia and Rapid Clearance With Ertapenem Plus Cefazolin. Cureus 2025; 17:e77335. [PMID: 39935929 PMCID: PMC11813253 DOI: 10.7759/cureus.77335] [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] [Accepted: 01/12/2025] [Indexed: 02/13/2025] Open
Abstract
Infective endocarditis is an infection of the heart's native or prosthetic valves, often caused by bacteria such as Staphylococcus aureus. Although infective endocarditis most commonly affects the left heart, cases of right-sided infective endocarditis, involving structures like the tricuspid or pulmonary valves, are also noted. Isolated native pulmonary valve infective endocarditis is exceptionally rare. After suspicion, the diagnosis relies on clinical symptoms and signs, imaging, and microbiological evidence. We report an unusual case of isolated pulmonary valve infective endocarditis in a previously healthy 59-year-old man without typical risk factors. He presented with unspecific symptoms such as fever, chills, dizziness, and left shoulder pain. Radiologically, the patient presented small ground-glass opacities in both lungs that aggravated during the first days after hospital admission with multifocal consolidation areas, and later developing bilateral necrotizing pneumonia. Despite adequate antibiotic treatment, the patient developed septic shock and persistent Staphylococcus aureus bacteremia. Given the persistence of such microorganisms in the bloodstream, despite the initial absence of endocardial involvement on transthoracic echocardiography, transesophageal echocardiography was done and revealed a large vegetation on the pulmonary valve with valvular regurgitation. According to Duke's criteria for infective endocarditis, a definite diagnosis was made, once both major clinical criteria were present, namely, typical microorganisms consistent with infective endocarditis from two separate blood cultures and evidence of endocardial involvement. Given the refractory bacteremia, an unusual combination of antibiotic therapy, including ertapenem and cefazolin, was introduced, leading to rapid clearance of bacteremia. This salvage antibiotic regimen was chosen due to the synergy of carbapenem with cefazolin and their potential improved bactericidal activity within biofilms. The patient subsequently required surgical intervention with bioprosthetic pulmonary valve replacement and ultimately achieved near-full recovery after a prolonged hospital stay. This case illustrates the diagnostic and therapeutic challenges of rare right-sided infective endocarditis since the patient presented with non-specific symptoms, without typical risk factors for right-sided infective endocarditis and the initial transthoracic echocardiography showed no valvular vegetations. Furthermore, the persistence of bacteremia despite adequate antibiotic therapy was a clinical challenge and this case highlights the potential efficacy of ertapenem plus cefazolin in treating persistent Staphylococcus aureus infections. It underscores the importance of individualized management in severe cases and the need for ongoing research to optimize treatment strategies for persistent infections.
Collapse
Affiliation(s)
- Paulo Fernandes
- Intensive Care Unit, Hospital José Joaquim Fernandes, Beja, PRT
| | | | - Ana Rita Rocha
- Internal Medicine Department, Hospital José Joaquim Fernandes, Beja, PRT
| | - Sara Carvalho
- Intensive Care Unit, Hospital José Joaquim Fernandes, Beja, PRT
| | - José Vaz
- Intensive Care Unit, Hospital José Joaquim Fernandes, Beja, PRT
| |
Collapse
|
25
|
Bae S, Kook MS, Chang E, Jung J, Kim MJ, Chong YP, Kim SH, Choi SH, Lee SO, Kim YS. Risk Factors for Infection-Attributable Mortality in Patients With Staphylococcus aureus Bacteremia: A Competing Risk Analysis. Open Forum Infect Dis 2025; 12:ofae734. [PMID: 39776932 PMCID: PMC11704955 DOI: 10.1093/ofid/ofae734] [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: 09/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background Identifying risk factors for mortality in patients with Staphylococcus aureus bacteremia (SAB) is crucial due to its high fatality. However, data on risk factors for infection-attributable deaths considering competing risk events such as non-infection-attributable deaths remain limited. We performed a competing risk analysis to elucidate risk factors associated with 30-day infection-attributable mortality in a large cohort of patients with SAB. Methods This retrospective cohort study included adult patients diagnosed with SAB at a tertiary hospital from August 2008 to December 2019. Competing risk analysis was performed using Fine and Gray models to estimate subdistribution hazard ratios (sHRs) for 30-day infection-attributable death. Results Among 1936 patients, 444 (22.9%) died within 30 days. Of these, 338 (76.1%) were infection-attributable and 106 (23.9%) were non-infection-attributable deaths. The multivariable Fine and Gray model identified significant risk factors for 30-day infection-attributable death (sHRs with 95% confidence intervals): an increase in age by 10 years (1.14 [1.02-1.26]), presence of malignancy (1.54 [1.17-2.02]), liver cirrhosis (2.15 [1.56-2.97]), corticosteroid use (1.61 [1.19-2.17]), septic shock (3.28 [1.98-5.42]), elevated C-reactive protein (1.60 [1.19-2.14]), pneumonia (1.81 [1.21-2.72]), persistent bacteremia (1.73 [1.31-2.30]), and failure to remove the eradicable focus (2.40 [1.38-4.19]) or absence of an eradicable focus (1.49 [1.08-2.04]). Except for age and malignancy, these factors were not significantly associated with non-infection-related death. Conclusions Specific risk factors for infection-attributable death in patients with SAB were identified, distinct from those for nonattributable death. These findings can aid in the early identification of patients at risk for SAB-attributable mortality.
Collapse
Affiliation(s)
- Seongman Bae
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Soo Kook
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Euijin Chang
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiwon Jung
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Jae Kim
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Pil Chong
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Ho Choi
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Oh Lee
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yang Soo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Center for Antimicrobial Resistance and Microbial Genetics, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
26
|
Kim M, Ranganath N, Chesdachai S, Stevens RW, Sohail MR, Abu Saleh OM. Which trial do we need? Combination therapy with daptomycin plus ceftaroline versus standard-of-care monotherapy in the treatment of methicillin-resistant Staphylococcus aureus bacteraemia. Clin Microbiol Infect 2025; 31:18-21. [PMID: 39182576 DOI: 10.1016/j.cmi.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/02/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Affiliation(s)
- Myeongji Kim
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States.
| | - Nischal Ranganath
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Supavit Chesdachai
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryan W Stevens
- Department of Pharmacy, Mayo Clinic, Rochester, MN, United States
| | | | - Omar M Abu Saleh
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
27
|
Recht J, Evans TJ, Chansamouth V, Phommasone K, Mayxay M, Ashley EA. Which trial do we need? A pragmatic randomized trial of trimethoprim-sulfamethoxazole vs. vancomycin for the treatment of methicillin-resistant Staphylococcus aureus bacteraemia in low-resource settings. Clin Microbiol Infect 2025; 31:13-17. [PMID: 39067512 DOI: 10.1016/j.cmi.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Affiliation(s)
- Judith Recht
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Terry John Evans
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao PDR; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vilada Chansamouth
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao PDR; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom; Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Koukeo Phommasone
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao PDR; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom; Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao PDR; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom; Institute of Research and Education Development (IRED), University of Health Sciences, Ministry of Health, Vientiane, Lao PDR
| | - Elizabeth A Ashley
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit (LOMWRU), Mahosot Hospital, Vientiane, Lao PDR; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
28
|
Saito M, McDonald KA, Grier AK, Meghwani H, Rangel-Moreno J, Becerril-Villanueva E, Gamboa-Dominguez A, Bruno J, Beck CA, Proctor RA, Kates SL, Schwarz EM, Muthukrishnan G. Immune Checkpoint Molecules as Biomarkers of Staphylococcus aureus Bone Infection and Clinical Outcome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630837. [PMID: 39803468 PMCID: PMC11722373 DOI: 10.1101/2024.12.30.630837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Staphylococcus aureus prosthetic joint infections (PJIs) are broadly considered incurable, and clinical diagnostics that guide conservative vs. aggressive surgical treatments don't exist. Multi-omics studies in a humanized NSG-SGM3 BLT mouse model demonstrate human T cells: 1) are remarkably heterogenous in gene expression and numbers, and 2) exist as a mixed population of activated, progenitor-exhausted, and terminally-exhausted Th1/Th17 cells with increased expression of immune checkpoint proteins (LAG3, TIM-3). Importantly, these proteins are upregulated in the serum and the bone marrow of S. aureus PJI patients. A multiparametric nomogram combining high serum immune checkpoint protein levels with low proinflammatory cytokine levels (IFN-γ, IL-2, TNF-α, IL-17) revealed that TIM-3 was highly predictive of adverse disease outcomes (AUC=0.89). Hence, T cell impairment in the form of immune checkpoint expression and exhaustion could be a functional biomarker for S. aureus PJI disease outcome, and blockade of checkpoint proteins could potentially improve outcomes following surgery.
Collapse
Affiliation(s)
- Motoo Saito
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Katya A. McDonald
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alex K. Grier
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Himanshu Meghwani
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Enrique Becerril-Villanueva
- Psychoimmunology laboratory, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz.” Mexico City, Mexico
| | - Armando Gamboa-Dominguez
- Deparment of Pathology, Instituto Nacional de Ciencias Médicas Y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jennifer Bruno
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher A. Beck
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward M. Schwarz
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Gowrishankar Muthukrishnan
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Laurier N, Karellis A, Xue X, Afilalo M, Weiss K. Strategies to reduce 28-day mortality in adult patients with bacteremia in the emergency department. BMC Infect Dis 2024; 24:1384. [PMID: 39633299 PMCID: PMC11616233 DOI: 10.1186/s12879-024-10242-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Bacteremia, a common emergency department presentation, has a high burden of mortality, cost and morbidity. We aimed to identify areas for potential improvement in emergency department bacteremia management. METHODS This retrospective cohort study included adults with bacteremia in an emergency department in 2019 and 2022. The primary outcome was 28-day mortality. Descriptive analyses evaluated demographics, comorbidities and clinical characteristics. Univariate and multivariate analyses identified mortality predictors. RESULTS Overall, 433 patients were included [217 males (50.1%), mean ± SD age: 74.1 ± 15.2 years]. The 28-day mortality rate was 15.2% (n = 66). In univariate analysis, age ≥ 70 years, arrival by ambulance, arrhythmia, congestive heart failure, recent steroid use, hypotension (< 90/60 mmHg), mechanical ventilation, cardiac arrest, intensive care unit (ICU) admission, intravenous antibiotics, pneumonia as bacteremia source, non-urinary tract infections, no infectious disease consultation, no antibiotic adjustment and no control blood cultures were significantly associated with 28-day mortality (p < 0.05). Malignancy showed a statistical trend (0.05 < p < 0.15). The above-stated sixteen variables, identified in univariate analysis, were assessed via multivariate analysis. Primarily, clinical relevance and, secondarily, statistical significance were used for multivariate model creation to prioritize pertinent variables. Five risk factors, significantly associated with mortality (p < 0.05), were included in the model: ICU admission [adjusted OR (95% CI): 6.03 (3.08-11.81)], pneumonia as bacteremia source [4.94 (2.62-9.32)], age ≥ 70 [3.16 (1.39-7.17)], hypotension [2.12 (1.02-4.40)], and no infectious disease consultation [2.02 (1.08-3.78)]). Surprisingly, initial antibiotic administration within 6 h, inappropriate initial antibiotic regimen and type of bacteria (Gram-negative, Gram-positive) were non-significant (p > 0.05). CONCLUSIONS We identified significant mortality predictors among emergency department patients presenting with bacteremia. Referral to an infectious disease physician is the only modifiable strategy to decrease 28-day mortality with long-term effect and should be prioritized.
Collapse
Affiliation(s)
- Noémie Laurier
- Division of Infectious Diseases and Microbiology, Jewish General Hospital, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
- McGill University, 845 Sherbrooke St W, Montreal, QC, H3A 0G4, Canada
- Centre of Excellence in Infectious Diseases, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
- Lady Davis Institute, Montreal, QC, H3T 1E2, Canada
| | - Angela Karellis
- Division of Infectious Diseases and Microbiology, Jewish General Hospital, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.
- Centre of Excellence in Infectious Diseases, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.
- Lady Davis Institute, Montreal, QC, H3T 1E2, Canada.
| | - Xiaoqing Xue
- Emergency Department, Jewish General Hospital, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Marc Afilalo
- McGill University, 845 Sherbrooke St W, Montreal, QC, H3A 0G4, Canada
- Emergency Department, Jewish General Hospital, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Karl Weiss
- Division of Infectious Diseases and Microbiology, Jewish General Hospital, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
- McGill University, 845 Sherbrooke St W, Montreal, QC, H3A 0G4, Canada
- Centre of Excellence in Infectious Diseases, 3755 Chemin de La Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
- Lady Davis Institute, Montreal, QC, H3T 1E2, Canada
- Université de Montréal, 2900 Edouard Montpetit Blvd, Montreal, QC, H3T 1J4, Canada
| |
Collapse
|
30
|
Staudacher M, Hotz JF, Kriz R, Schefberger K, Schneider L, Spettel K, Starzengruber P, Hagemann JB, Leutzendorff A, Burgmann H, Lagler H. Differences in oxazolidinone resistance mechanisms and small colony variants emergence of Staphylococcus aureus induced in an in vitro resistance development model. Emerg Microbes Infect 2024; 13:2292077. [PMID: 38055244 PMCID: PMC10849000 DOI: 10.1080/22221751.2023.2292077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/03/2023] [Indexed: 12/07/2023]
Abstract
Invasive Staphylococcus aureus infections are associated with a high burden of disease, case fatality rate and healthcare costs. Oxazolidinones such as linezolid and tedizolid are considered potential treatment choices for conditions involving methicillin resistance or penicillin allergies. Additionally, they are being investigated as potential inhibitors of toxins in toxin-mediated diseases. In this study, linezolid and tedizolid were evaluated in an in vitro resistance development model for induction of resistance in S. aureus. Whole genome sequencing was conducted to elucidate resistance mechanisms through the identification of causal mutations. After inducing resistance to both linezolid and tedizolid, several partially novel single nucleotide variants (SNVs) were detected in the rplC gene, which encodes the 50S ribosome protein L3 in S. aureus. These SNVs were found to decrease the binding affinity, potentially serving as the underlying cause for oxazolidinone resistance. Furthermore, in opposite to linezolid we were able to induce phenotypically small colony variants of S. aureus after induction of resistance with tedizolid for the first time in literature. In summary, even if different antibiotic concentrations were required and SNVs were detected, the principal capacity of S. aureus to develop resistance to oxazolidinones seems to differ between linezolid and tedizolid in-vivo but not in vitro. Stepwise induction of resistance seems to be a time and cost-effective tool for assessing resistance evolution. Inducted-resistant strains should be examined and documented for epidemiological reasons, if MICs start to rise or oxazolidinone-resistant S. aureus outbreaks become more frequent.
Collapse
Affiliation(s)
- Moritz Staudacher
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
- Department of Angiology, Medical University of Vienna, Vienna, Austria
| | - Julian Frederic Hotz
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Evangelic Hospital Vienna, Vienna, Austria
| | - Richard Kriz
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Katharina Schefberger
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Lisa Schneider
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Kathrin Spettel
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Peter Starzengruber
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | | | - Amelie Leutzendorff
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Heimo Lagler
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Grosso R, Nguyen V, Ahmed SK, Wong-Beringer A. Novel Epigallocatechin Gallate (EGCG) Analogs with Improved Biochemical Properties for Targeting Extracellular and Intracellular Staphylococcus aureus. Appl Microbiol 2024; 4:1568-1581. [DOI: 10.3390/applmicrobiol4040107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Staphylococcus aureus is a leading cause of bloodstream infection (SAB), with up to 30% mortality. Despite treatment with standard antibiotics, one in three patients develops a persistent infection, which portends a five-fold increase in the risk of death. Persistent SAB has been attributed in part to the inability of antistaphylococcal antibiotics to eradicate intracellular S. aureus surviving inside macrophages. (-)- Epigallocatechin gallate (EGCG) is a catechin found in green tea that has been widely studied for its broad biological activities, ranging from anticancer to antibacterial activity. However, EGCG is greatly limited by its poor drug-like properties in terms of stability, membrane permeability, and bioavailability. In this study, we established through a series of in vitro experiments that structural modifications of EGCG enhanced drug-like properties while maintaining or improving its antistaphylococcal activity. Our lead EGCG analogs (MCC-1 and MCC-2) showed improved biochemical properties along with increased potency against extracellular S. aureus and restored susceptibility of β-lactam agents to methicillin-resistant S. aureus (MRSA). Importantly, the lead analogs but not EGCG potentiated macrophage- and antibiotic-mediated clearance of intracellular bacteria. Overall, EGCG analogs showed promise for further development as adjunctive therapy candidates for the treatment of SAB.
Collapse
Affiliation(s)
- Riley Grosso
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Vy Nguyen
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Syed Kaleem Ahmed
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Annie Wong-Beringer
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| |
Collapse
|
32
|
Madaline T, Classen DC, Eby JC. Building the Future of Infectious Diseases: A Call to Action for Quality Improvement Research and Measurement. J Infect Dis 2024; 230:1064-1072. [PMID: 38591245 DOI: 10.1093/infdis/jiae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/27/2024] [Accepted: 04/06/2024] [Indexed: 04/10/2024] Open
Abstract
Quality is central to value-based care, and measurement is essential for assessing performance and understanding improvement over time. Both value-based care and methods for quality measurement are evolving. Infectious diseases (ID) has been less engaged than other specialties in quality measure development, and ID providers must seize the opportunity to engage with quality measure development and research. Antimicrobial stewardship programs are an ideal starting point for ID-related quality measure development; antimicrobial stewardship program interventions and best practices are ID specific, measurable, and effective, yet they are grossly undercompensated. Herein, we provide a scheme for prioritizing research focused on development of ID-specific quality measures. Maturation of quality measurement research in ID, beginning with an initial focus on stewardship-related conditions and then expanding to non-stewardship topics, will allow ID to take control of its future in value-based care and promote the growth of ID through greater recognition of its value.
Collapse
Affiliation(s)
- Theresa Madaline
- Division of Infectious Diseases, New York-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - David C Classen
- Division of Epidemiology, University of Utah, Salt Lake City, Utah, USA
| | - Joshua C Eby
- Division of Infectious Diseases and International Health, University of Virginia Health, Charlottesville, Virginia, USA
| |
Collapse
|
33
|
Peng H, Ou Y, Zhang R, Wang R, Wen D, Yang Q, Liu X. Monitoring vancomycin blood concentrations reduces mortality risk in critically ill patients: a retrospective cohort study using the MIMIC-IV database. Front Pharmacol 2024; 15:1458600. [PMID: 39611174 PMCID: PMC11602295 DOI: 10.3389/fphar.2024.1458600] [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: 07/02/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024] Open
Abstract
Background The incidence and mortality of severe Gram-positive cocci infections are particularly high in intensive care units (ICUs). Vancomycin remains the treatment of choice for severe infections caused by Gram-positive cocci, particularly methicillin-resistant Staphylococcus aureus (MRSA). Some guidelines recommend therapeutic drug monitoring (TDM) for critically ill patients treated with vancomycin; however, there is currently a lack of evidence to support that TDM improves the mortality rates of these patients. Therefore, we designed this cohort study to compare the impact of monitoring vancomycin blood concentrations on mortality rates in critically ill patients and to provide evidence to support this routine clinical practice. Methods Data were extracted from the Medical Information Mart for Intensive Care (MIMIC)-IV database for a retrospective cohort analysis of critically ill patients receiving intravenous vancomycin treatment. The primary outcome was the 28 day mortality rate. The propensity score matching (PSM) method was used to match the baseline characteristics between patients in the TDM group and the non-TDM group. The relationship between 28 day mortality and vancomycin TDM in the critically ill cohort was evaluated using Cox proportional hazards regression analysis and Kaplan-Meier survival curves. Validation of the primary outcomes was conducted by comparing the PSM model and the Cox proportional hazards regression model. The robustness of the conclusion was subsequently verified by subgroup and sensitivity analyses. Results Data for 18,056 critically ill patients who met the study criteria were collected from the MIMIC-IV database. Of these, 7,451 patients had at least one record of vancomycin blood concentration monitoring, which we defined as the TDM group. The TDM group exhibited a 28 day mortality rate of 25.7% (1,912/7,451) compared to 16.2% in the non-TDM group (1,723/10,605). After PSM, 4,264 patients were included in each of the TDM and non-TDM groups, with a 28 day mortality rate of 20.0% (1,022/4,264) in the TDM group and 26.4% (1,126/4,264) in the non-TDM group. Multivariate Cox proportional hazards analysis revealed a significantly lower 28 day mortality risk in the TDM group when compared to the non-TDM group (adjusted hazard ratio [HR]: 0.86; 95% confidence interval [CI]: 0.79, 0.93; p < 0.001). Further PSM analyses (adjusted HR: 0.91; 95% CI: 0.84, 0.99; p = 0.033) confirmed the lower risk of mortality in the TDM group. Kaplan-Meier survival analysis revealed a significantly higher survival rate at 28 days for the TDM group (log-rank test, p < 0.001). Subgroup analysis results indicated that patients with sepsis, septic shock, estimated glomerular filtration rate ≤ 60 mL/min/1.73 m2, undergoing renal replacement therapy, using vasoactive drugs, on mechanical ventilation, and those with higher severity scores (Acute Physiology Score III ≥40, Oxford Acute Severity of Illness Score ≥30, Simplified Acute Physiology Score II ≥ 30) significantly benefited from monitoring vancomycin blood concentrations. The results remained unchanged excluding patients staying in ICU for less than 48 h or those infected with MRSA. Conclusion This cohort study showed that monitoring vancomycin blood concentrations is associated with a significantly lower 28 day mortality rate in critically ill patients, highlighting the importance of routinely performing vancomycin TDM in these patients.
Collapse
Affiliation(s)
- Huaidong Peng
- Department of Pharmacy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuantong Ou
- Department of Critical Care, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruichang Zhang
- Department of Critical Care, Guangzhou Twelfth People’ Hospital, Guangzhou, China
| | - Ruolun Wang
- Department of Pharmacy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Deliang Wen
- Department of Critical Care, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qilin Yang
- Department of Critical Care, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaorui Liu
- Department of Pharmacy, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
34
|
Dieperink SS, Nørgaard M, Mehnert F, Oestergaard LB, Benfield T, Torp-Pedersen C, Petersen A, Glintborg B, Hetland ML. Rheumatoid arthritis and risk of osteoarticular infection and death following Staphylococcus aureus bacteraemia: a nationwide cohort study. Rheumatology (Oxford) 2024; 63:2989-2996. [PMID: 38460189 DOI: 10.1093/rheumatology/keae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 03/11/2024] Open
Abstract
OBJECTIVES Osteoarticular infection (OAI) is a feared complication of Staphylococcus aureus bacteraemia (SAB) and is associated with poor outcomes. We aimed to explore the risk of OAI and death following SAB in patients with and without rheumatoid arthritis (RA) and to identify risk factors for OAI in patients with RA. METHODS Danish nationwide cohort study of all patients with microbiologically verified first-time SAB between 2006-18. We identified RA, SAB, comorbidities, and RA-related characteristics (e.g. orthopaedic implants and antirheumatic treatment) in national registries including the rheumatology registry DANBIO. We estimated the cumulative incidence of OAI and death and adjusted hazard ratios (HRs, multivariate Cox regression). RESULTS We identified 18 274 patients with SAB (n = 367 with RA). The 90-day cumulative incidence of OAI was 23.1% (95% CI 18.8; 27.6) for patients with RA and 12.5% (12.1; 13.0) for patients without RA (non-RA) [HR 1.93 (1.54; 2.41)]. For RA patients with orthopaedic implants cumulative incidence was 29.4% (22.9; 36.2) [HR 1.75 (1.08; 2.85)], and for current users of tumor necrosis factor inhibitors (TNFi) it was 41.9% (27.0; 56.1) [HR 2.27 (1.29; 3.98) compared with non-users]. All-cause 90-day mortality following SAB was similar in RA [35.4% (30.6; 40.3)] and non-RA [33.9% (33.2; 34.5), HR 1.04 (0.87; 1.24)]. CONCLUSION Following SAB, almost one in four patients with RA contracted OAI corresponding to a doubled risk compared with non-RA. In RA, orthopaedic implants and current TNFi use were associated with approximately doubled OAI risk. One in three died within 90 days in both RA and non-RA. These findings encourage vigilance in RA patients with SAB to avoid treatment delay of OAI.
Collapse
Affiliation(s)
- Sabine S Dieperink
- Copenhagen Center for Arthritis Research (COPECARE), Centre for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Copenhagen University Hospital-Rigshospitalet, Glostrup, Denmark
- Department of Internal Medicine and Emergency Department, Odense University Hospital, Svendborg, Denmark
| | - Mette Nørgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Frank Mehnert
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Louise B Oestergaard
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Hellerup, Denmark
| | - Thomas Benfield
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital-Amager and Hvidovre, Copenhagen, Denmark
| | - Christian Torp-Pedersen
- Department of Cardiology, Copenhagen University Hospital-North Zealand, Hillerød, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Petersen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Bente Glintborg
- Copenhagen Center for Arthritis Research (COPECARE), Centre for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Copenhagen University Hospital-Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- DANBIO Registry, Centre for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Copenhagen University Hospital-Rigshospitalet, Glostrup, Denmark
| | - Merete L Hetland
- Copenhagen Center for Arthritis Research (COPECARE), Centre for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Copenhagen University Hospital-Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- DANBIO Registry, Centre for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Copenhagen University Hospital-Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
35
|
Gerrity RC, Parkinson M, Strength R, Animalu CN, Davidson N, Fuchs CJ, Jackson CD, Summers NA. Effect of HIV Status and Charlson Comorbidity Index on COVID-19 Clinical Outcomes in a Case-Control Study. South Med J 2024; 117:651-656. [PMID: 39486450 DOI: 10.14423/smj.0000000000001753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
OBJECTIVES During the course of the coronavirus disease 2019 (COVID-19) pandemic, numerous comorbidities were identified as risk factors for increased morbidity and mortality. Few studies have examined human immunodeficiency virus (HIV) and COVID-19 co-infection and the impact of HIV on COVID-19 outcomes. In this study, we compared outcomes of people living with HIV with COVID-19 with a control group to examine outcomes. METHODS We identified 45 people living with HIV admitted with COVID-19 to one of three large healthcare systems in Memphis, Tennessee, between March 1 and October 31, 2020. We matched the people living with HIV in a 1:1 fashion to a control group of COVID-19-positive patients without a recorded history of HIV and compared clinical outcomes. Nine pairs were not able to be optimally matched, so a sensitivity analysis was completed by repeating the same analyses in the primary analysis while excluding the nine mismatched pairs. RESULTS Patients did not differ significantly in demographic variables due to the matching algorithm, and there was no significant difference in measured outcomes between people living with HIV and controls. A CD4 count of <200 cells per microliter was not significantly associated with increased morbidity or mortality. Controlling for HIV status, an elevated Charlson Comorbidity Index score of >3 was associated with increased intubation (P = 0.02), vasopressor use (odds ratio [OR] 4.81, P = 0.04), intensive care unit level of care (OR 4.37, P = 0.007), mortality (OR 7.14, P = 0.02), and length of overall hospital stay in days (P = 0.004). CONCLUSIONS We found no difference in outcomes of people living with HIV in comparison to matched controls based on HIV status but found that an increased Charlson Comorbidity Index score led to increased morbidity and mortality regardless of HIV status.
Collapse
Affiliation(s)
| | | | | | - Chinelo N Animalu
- the Department of Medicine, Division of Infectious Diseases, University of Tennessee Health Science Center College of Medicine, Memphis
| | | | | | - Christopher D Jackson
- the Department of Medicine, Division of Infectious Diseases, University of Tennessee Health Science Center College of Medicine, Memphis
| | | |
Collapse
|
36
|
Thornley EM, Alexander R, Popelka J, Montgomery CP. Risk Factors for Persistent Staphylococcus aureus Bacteremia in Children. Pediatr Infect Dis J 2024; 43:1040-1045. [PMID: 38905222 PMCID: PMC11662081 DOI: 10.1097/inf.0000000000004439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of pediatric bacteremia. Persistent S. aureus bacteremia (SAB) is associated with increased morbidity and mortality in adults and children. Risk factors for S. aureus bacteremia have been well established, but there is a limited understanding of the factors that contribute to the development of persistent SAB in children. METHODS This is a single-center retrospective secondary analysis of a prospective observational study of pediatric patients hospitalized with S. aureus infection over a 3.5-year period at a large, quaternary, children's hospital. RESULTS Two hundred fifty-nine children with confirmed S. aureus infection were enrolled in the study. Sixty-five of these were found to have bacteremia, with 28 (43%) developing persistent bacteremia. Patients with persistent SAB were culture-positive for a median of 3.5 days compared with 1 day for those without ( P ≤ 0.001). Children with persistent SAB were more likely to have an identified osteoarticular source of infection (93%, n = 26 vs. 62%, n = 23; P = 0.008) and had a shorter median duration to culture positivity than those without persistent SAB (16 hours vs. 20 hours; P ≤ 0.001). In addition, children with persistent SAB had higher median values of presenting erythrocyte sedimentation rate, peak erythrocyte sedimentation rate, presenting C-reactive protein and peak C-reactive protein. Not surprisingly, hospital length of stay was longer in children with persistent SAB compared with those without. CONCLUSIONS These findings suggest that a shorter time to culture positivity, osteoarticular infection, and higher presenting and peak values for select inflammatory markers are potential risk factors for persistent SAB in children.
Collapse
Affiliation(s)
- Erin M. Thornley
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Robin Alexander
- Biostatistics Resource at Nationwide Children’s Hospital (BRANCH), Columbus, OH
| | - Jill Popelka
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Christopher P. Montgomery
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
37
|
Li Y, Farhan MHR, Yang X, Guo Y, Sui Y, Chu J, Huang L, Cheng G. A review on the development of bacterial multi-epitope recombinant protein vaccines via reverse vaccinology. Int J Biol Macromol 2024; 282:136827. [PMID: 39476887 DOI: 10.1016/j.ijbiomac.2024.136827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/21/2024] [Indexed: 11/10/2024]
Abstract
Bacterial vaccines play a crucial role in combating bacterial infectious diseases. Apart from the prevention of disease, bacterial vaccines also help to reduce the mortality rates in infected populations. Advancements in vaccine development technologies have addressed the constraints of traditional vaccine design, providing novel approaches for the development of next-generation vaccines. Advancements in reverse vaccinology, bioinformatics, and comparative proteomics have opened horizons in vaccine development. Specifically, the use of protein structural data in crafting multi-epitope vaccines (MEVs) to target pathogens has become an important research focus in vaccinology. In this review, we focused on describing the methodologies and tools for epitope vaccine development, along with recent progress in this field. Moreover, this article also discusses the challenges in epitope vaccine development, providing insights for the future development of bacterial multi-epitope genetically engineered vaccines.
Collapse
Affiliation(s)
- Yuxin Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Muhammad Haris Raza Farhan
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Xiaohan Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Ying Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; MOA Laboratory of Risk Assessment for Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
38
|
Waked R, Coats L, Rosato A, Yen CF, Wood E, Diekema DJ, Rokas KE, Mercuro NJ. Clinical outcome of combination of vancomycin and ceftaroline versus vancomycin monotherapy for treatment of methicillin resistant Staphylococcus aureus bloodstream infection. BMC Infect Dis 2024; 24:1212. [PMID: 39468491 PMCID: PMC11514869 DOI: 10.1186/s12879-024-10107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND The role of combination therapies for serious methicillin-resistant Staphylococcus aureus (MRSA) infections is widely debated. METHODS This retrospective cohort study included adults with MRSA bacteraemia treated between January 1, 2013, to December 31, 2022. Patients receiving combination therapy with vancomycin and ceftaroline were matched in a 2:1 ratio with those on vancomycin monotherapy based on bacteraemia source and illness severity. The primary outcome was frequency of bacteraemia recurrence. Secondary outcomes were all cause 30/90-day mortality, recurrence or mortality at 30/90 days and in hospital length of stay. RESULTS Of 57 patients included, 37 (65%) were in the combination group. The overall intensive care unit admission rate was 63.2% (36/57) and the Pitt Bacteraemia Score was 1 [0-4] at the time of diagnosis. The most common source of infection was endovascular/endocarditis (n = 36, 63.2%). Demographic and clinical characteristics were similar between the monotherapy and combination group of patients, except for higher body mass index (32.5 [25.5-36.4] vs. 24.4 [20.9-29], p = 0.004) and a greater immunosuppression prevalence (3 (15%) vs. 0 (0%), p = 0.039) in monotherapy group. There was no significant difference in bacteraemia recurrence (3 (15%) vs. 4 (10.8%), p = 0.7) or all-cause 30-day mortality (3 (15%) vs. 4 (10.8%), p = 0.7) between the two groups. CONCLUSION The results of this study are limited by a retrospective observational design; however, combination of vancomycin and ceftaroline for MRSA bacteraemia was not associated with lower bacteraemia recurrence or mortality compared to vancomycin monotherapy.
Collapse
Affiliation(s)
- Rami Waked
- Department of Infectious Diseases, MaineHealth Maine Medical Centre, 22 Bramhall St, Portland, ME, 04102, USA.
- Department of Internal Medicine, MaineHealth Maine Medical Centre, Portland, ME, USA.
| | - Leslie Coats
- Pharmacy, MaineHealth Maine Medical Centre, Portland, ME, USA
| | - Adriana Rosato
- Maine Health Institute for Research, Scarborough, ME, USA
| | - Christina F Yen
- Department of Infectious Diseases, MaineHealth Maine Medical Centre, 22 Bramhall St, Portland, ME, 04102, USA
| | - Emily Wood
- Department of Infectious Diseases, MaineHealth Maine Medical Centre, 22 Bramhall St, Portland, ME, 04102, USA
| | - Daniel J Diekema
- Department of Infectious Diseases, MaineHealth Maine Medical Centre, 22 Bramhall St, Portland, ME, 04102, USA
| | | | | |
Collapse
|
39
|
Matthews EL, Dilworth TJ. Determining the Prognostic Value of Complete Blood Count Subgroup Parameters in Staphylococcus aureus Bacteremia. J Patient Cent Res Rev 2024; 11:197-203. [PMID: 39439536 PMCID: PMC11493308 DOI: 10.17294/2330-0698.2073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Purpose Serum cytokine alterations are associated with increased Staphylococcus aureus bacteremia (SAB) mortality. Unfortunately, clinical use of these cytokines is uncommon due to limited availability and high cost. Complete blood count (CBC) with differential reflects the host immune response, and CBC subgroup parameters may have prognostic value in SAB. We sought to determine the association between CBC subgroup parameters on the day of index blood culture and 30-day all-cause mortality in SAB patients. Methods We conducted a retrospective study of adult SAB patients with infectious diseases consultation to evaluate the discriminatory capacity of CBC subgroup parameters in predicting SAB mortality. Clinical and microbiological data were collected, including severity of illness and CBC subgroup parameters, on the day of index blood culture. The primary outcome was 30-day all-cause mortality. A multivariable logistic regression model was used to determine the association between patient-level variables and mortality. Results A total of 119 patients were included. The overall 30-day all-cause mortality rate was 10.1%. The median neutrophil-to-lymphocyte count ratio (NLCR) among survivors was 13.6 vs 23.2 among non-survivors (p = .007). Median lymphocyte count among survivors was 0.9 x 103 cells/μL vs 0.6 x 103 cells/μL among non-survivors (p = .031). Median platelet count was higher among survivors than non-survivors (239 x 103 cells/μL vs 171 x 103 cells/μL, respectively; p = .018). All other CBC subgroup parameters were similar between the two groups. Known SAB mortality predictors, including age, were also associated with increased mortality. Lower lymphocyte count was independently associated with increased mortality (adjusted odds ratio [aOR] 0.236, 95% confidence interval [CI] 0.064-0.872), as was higher PITT bacteremia score (aOR 2.439, 95% CI 1.565-3.803). Conclusions CBC subgroup parameters may have prognostic value in SAB. Additional study is warranted to further ascertain the prognostic value of these readily available laboratory values.
Collapse
Affiliation(s)
- Emily L. Matthews
- Department of Pharmacy Services, Aurora St. Luke’s Medical Center, Advocate Health, Milwaukee, WI
| | - Thomas J. Dilworth
- Department of Pharmacy Services, Advocate Health Midwest, Advocate Health, Milwaukee, WI
| |
Collapse
|
40
|
Ferreira M, Pinto M, Aires-da-Silva F, Bettencourt A, Gaspar MM, Aguiar SI. Rifabutin: a repurposed antibiotic with high potential against planktonic and biofilm staphylococcal clinical isolates. Front Microbiol 2024; 15:1475124. [PMID: 39450290 PMCID: PMC11499150 DOI: 10.3389/fmicb.2024.1475124] [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: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 10/26/2024] Open
Abstract
Staphylococcus aureus poses a significant threat as an opportunistic pathogen in humans, and animal medicine, particularly in the context of hospital-acquired infections (HAIs). Effective treatment is a significant challenge, contributing substantially to the global health burden. While antibiotic therapy remains the primary approach for staphylococcal infections, its efficacy is often compromised by the emergence of resistant strains and biofilm formation. The anticipated solution is the discovery and development of new antibacterial agents. However, this is a time consuming and expensive process with limited success rates. One potential alternative for addressing this challenge is the repurposing of existing antibiotics. This study investigated the potential of rifabutin (RFB) as a repurposed antibiotic for treating S. aureus infections. The minimum inhibitory concentration (MIC) of rifabutin was assessed by the broth microdilution method, in parallel to vancomycin, against 114 clinical isolates in planktonic form. The minimum biofilm inhibitory concentration (MBIC50) was determined by an adaptation of the broth microdilution method, followed by MTT assay, against a subset of selected 40 clinical isolates organized in biofilms. The study demonstrated that RFB MIC ranged from 0.002 to 6.250 μg/mL with a MIC50 of 0.013 μg/mL. RFB also demonstrated high anti-biofilm activity in the subset of 40 clinical isolates, with confirmed biofilm formation, with no significant MBIC50 differences observed between the MSSA and MRSA strains, in contrast to that observed for the VAN. These results highlight the promising efficacy of RFB against staphylococcal clinical isolates with different resistance patterns, whether in planktonic and biofilm forms.
Collapse
Affiliation(s)
- Magda Ferreira
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Margarida Pinto
- Laboratório de Microbiologia do Serviço de Patologia Clínica do Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Frederico Aires-da-Silva
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Ana Bettencourt
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Maria Manuela Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Faculty of Sciences, Institute of Biophysics and Biomedical Engineering (IBEB), Universidade de Lisboa, Lisbon, Portugal
| | - Sandra Isabel Aguiar
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
41
|
Gallego Rodríguez A, Duch Llorach P, Grillo S, Piriz Marabajan M, Pomar Solchaga V, Hornero López A, Jimenez Martínez E, Oriol Bermudez I, Rivera A, Pujol Rojo M, López-Contreras González J. Risk factors for mortality and complications in peripheral venous catheter-associated Staphylococcus aureus bacteraemia: a large multicentre cohort study. J Hosp Infect 2024; 152:13-20. [PMID: 39032565 DOI: 10.1016/j.jhin.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Peripheral venous catheter-associated Staphylococcus aureus bacteraemia (PVC-SAB) is a potentially life-threatening nosocomial infection. AIM This cohort study aims to identify the risk factors associated with its mortality and complications. METHODS Retrospective analysis of a prospective cohort study conducted at two tertiary-care hospitals in Spain. Adult patients admitted between January 2011 and July 2019 which developed PVC-SAB during their hospital stay were included. Primary outcome was all-cause 30- and 90-day mortality. Secondary outcomes were sepsis or septic shock at the onset of bacteraemia, metastatic infection and length of hospital stay. Univariate and multivariate analyses were performed. FINDINGS A total of 256 PVC-SAB were diagnosed in 243 patients between 2011 and 2019. Thirty-day and 90-day all-cause mortality were 18.3% and 24.2%, respectively. Lack of susceptible antibiotic administration the day after blood culture collection (odds ratio: 4.14; 95% confidence interval: 1.55-11.03; P = 0.005), sepsis and complicated bacteraemia were identified as independent risk factors for 30- and 90-day mortality; meticillin-resistant S. aureus bacteraemia was identified as an independent risk factor only for 30-day mortality and functional dependence only for 90-day mortality. Persistent bacteraemia and sepsis were associated with septic metastases, which significantly increased hospital stay, and endocarditis. A greater proportion of patients experiencing septic shock were subsequently institutionalized compared to those without. CONCLUSION PVC-SAB remains linked to high mortality rates. Prompt administration of appropriate antibiotics is crucial for lowering mortality. A comprehensive diagnostic approach is essential, especially in patients with persistent bacteraemia and implanted cardiovascular devices, to rule out metastatic complications and endocarditis.
Collapse
Affiliation(s)
- A Gallego Rodríguez
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - P Duch Llorach
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S Grillo
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - M Piriz Marabajan
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - V Pomar Solchaga
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - A Hornero López
- Infectious Diseases Department, Hospital Universitari de Bellvitge - IDIBELL, Bellvitge Institute of Biomedical Research, Nursing Research Group, Barcelona, Spain
| | - E Jimenez Martínez
- Infectious Diseases Department, Hospital Universitari de Bellvitge - IDIBELL, Bellvitge Institute of Biomedical Research, Nursing Research Group, Barcelona, Spain
| | - I Oriol Bermudez
- Infectious Diseases Department, Hospital Universitari de Bellvitge - IDIBELL, Bellvitge Institute of Biomedical Research, Nursing Research Group, Barcelona, Spain; CIBERINFEC, ISCIII CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - A Rivera
- Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain; Microbiology Department, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - M Pujol Rojo
- Infectious Diseases Department, Hospital Universitari de Bellvitge - IDIBELL, Bellvitge Institute of Biomedical Research, Nursing Research Group, Barcelona, Spain; CIBERINFEC, ISCIII CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - J López-Contreras González
- Infectious Diseases Unit, Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau-Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Boubour A, Kim CY, Torres S, Jia DT, Hess E, Liu S, Sun Y, Fong K, Epstein S, Yan H, Luche N, Gao K, Glassberg B, Harmon M, Hoang H, Navis A, Schorr E, Gofshteyn JS, Yeshokumar AK, Thakur KT. Sociodemographic and Clinical Factors Associated With Clinical Outcome in Neuroinfectious Diseases: A Multicenter Retrospective Cohort Study. Neurohospitalist 2024; 14:396-405. [PMID: 39308466 PMCID: PMC11412469 DOI: 10.1177/19418744241263138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Objective To evaluate sociodemographic and clinical factors associated with clinical outcomes in patients hospitalized with neuroinfectious diseases at three tertiary care centers in New York City. Methods This retrospective cohort study was conducted at three large urban tertiary care centers between January 1, 2010 and December 31, 2017. Poor clinical outcome was defined as length of hospital stay (LOS) ≥2 weeks and/or discharge to a location other than home. Sociodemographic and clinical factors were obtained from electronic medical records and descriptively analyzed. Multivariate logistic regression analysis investigated relationships between sociodemographic and clinical factors, and outcomes. Results Among 205 patients with definitive neuroinfectious diagnoses, older patients were more likely to have a LOS ≥2 weeks (odds ratio [OR]: 1.03; 95% confidence interval [CI]: 1.01-1.05) and less likely to be discharged home (OR: 0.96; 95% CI: 0.94-0.98) than younger patients. Patients with an immunocompromised state were more likely to have a LOS ≥2 weeks (OR: 2.80; 95% CI: 1.17-6.69). Additionally, patients admitted to the intensive care unit (ICU) were more likely to have a LOS ≥2 weeks (OR: 4.65; 95% CI: 2.13-10.16) and less likely to be discharged home (OR: 0.14; 95% CI: 0.06-0.34). There were no statistically significant associations between sex, race, ethnicity, English proficiency, substance use, or poverty index, and clinical outcome. Conclusions In this multicenter cohort of hospitalized neuroinfectious diseases, older age, history of immunocompromised state, and admission to the ICU were significantly associated with poor clinical outcome.
Collapse
Affiliation(s)
- Alexandra Boubour
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Carla Y. Kim
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah Torres
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dan T. Jia
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Evan Hess
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sibei Liu
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Yifei Sun
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Kathryn Fong
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Multiple Sclerosis Center, Columbia University Irving Medical Center, New York, NY USA
| | - Samantha Epstein
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Multiple Sclerosis Center, Columbia University Irving Medical Center, New York, NY USA
| | - Helena Yan
- Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA
| | - Nicole Luche
- Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA
| | - Kerry Gao
- Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA
| | - Brittany Glassberg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Harmon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hai Hoang
- Department of Pediatrics, Weill Cornell Medical Center, New York, NY, USA
| | - Allison Navis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Schorr
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anusha K. Yeshokumar
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran T. Thakur
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Briol S, Gheysens O, Jamar F, Yildiz H, De Greef J, Cyr Yombi J, Verroken A, Belkhir L. Impact of [ 18F] FDG PET/CT on outcomes in patients with Staphylococcus aureus bacteremia: A retrospective single-center experience. Infect Dis Now 2024; 54:104977. [PMID: 39276874 DOI: 10.1016/j.idnow.2024.104977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVE Staphylococcus aureus bacteremia (SAB) is a leading cause of community and hospital-acquired bacteremia with significant morbidity and mortality. Effective management depends on accurate diagnosis, source control and assessment of metastatic infections. [18F] FDG PET/CT has been shown to reduce mortality in high-risk SAB patients. This study aims to evaluate the impact of [18F] FDG PET/CT on outcomes in patients with SAB. METHODS Single-center, retrospective, real-life setting study including all consecutive SAB cases from 2017 to 2019. Medical records were analyzed to collect information. RESULTS Out of the 315 included patients, 132 underwent [18F] FDG PET/CT. In those patients, a clear focus of infection was more frequently identified, leading to better adapted treatments and extended hospital stays. Overall mortality rates at 30 days, 90 days and one year were 25.1 %, 36.8 % and 44.8 % respectively. Mortality was significantly lower in the [18F] FDG PET/CT group (p < 0.0001) and persisted (p < 0.05) after adjusting for imbalances between groups regarding oncologic patients and deaths within 7 days. The difference in mortality remained significant irrespective of prolonged bacteremia but was not significant with regard to hospital-acquired SAB. Supplementary analysis using the Cox proportional hazards model confirmed that [18F] FDG PET/CT was significantly associated with reduced mortality (p < 0.05). CONCLUSION In this real-life cohort, patients with SAB having undergone [18F] FDG PET/CT experienced lower mortality rates, highlighting the additional value of [18F] FDG PET/CT in SAB management. Further research is needed to identify the subpopulations that would benefit most from the integration of [18F] FDG PET/CT in their work-up.
Collapse
Affiliation(s)
- Sébastien Briol
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium.
| | - Olivier Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium
| | - François Jamar
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium
| | - Halil Yildiz
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium
| | - Julien De Greef
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Jean Cyr Yombi
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Alexia Verroken
- Department of Microbiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Leïla Belkhir
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 av Hippocrate, 1200 Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
44
|
Campbell AJ, Anpalagan K, Best EJ, Britton PN, Gwee A, Hatcher J, Manley BJ, Marsh J, Webb RH, Davis JS, Mahar RK, McGlothlin A, McMullan B, Meyer M, Mora J, Murthy S, Nourse C, Papenburg J, Schwartz KL, Scheuerman O, Snelling T, Strunk T, Stark M, Voss L, Tong SYC, Bowen AC. Whole-of-Life Inclusion in Bayesian Adaptive Platform Clinical Trials. JAMA Pediatr 2024; 178:1066-1071. [PMID: 39158898 DOI: 10.1001/jamapediatrics.2024.2697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Importance There is a recognized unmet need for clinical trials to provide evidence-informed care for infants, children and adolescents. This Special Communication outlines the capacity of 3 distinct trial design strategies, sequential, parallel, and a unified adult-pediatric bayesian adaptive design, to incorporate children into clinical trials and transform this current state of evidence inequity. A unified adult-pediatric whole-of-life clinical trial is demonstrated through the Staphylococcus aureus Network Adaptive Platform (SNAP) trial. Observations Bayesian methods provide a framework for synthesizing data in the form of a probability model that can be used in the design and analysis of a clinical trial. Three trial design strategies are compared: (1) a sequential adult-pediatric bayesian approach that involves a separate, deferred pediatric trial that incorporates existing adult trial data into the analysis model to potentially reduce the pediatric trial sample size; (2) a parallel adult-pediatric bayesian trial whereby separate pediatric enrollment occurs in a parallel trial, running alongside an adult randomized clinical trial; and (3) a unified adult-pediatric bayesian adaptive design that supports the enrollment of both children and adults simultaneously in a whole-of-life bayesian adaptive randomized clinical trial. The SNAP trial whole-of-life design uses a bayesian hierarchical model that allows information sharing (also known as borrowing) between trial age groups by linking intervention effects of children and adults, thereby improving inference in both groups. Conclusion and Relevance Bayesian hierarchical models may provide more precision for estimates of safety and efficacy of treatments in trials with heterogenous populations compared to traditional methods of analysis. They facilitate the inclusion of children in clinical trials and a shift from children deemed therapeutic orphans to the vision of no child left behind in clinical trials to ensure evidence for clinical practice exists across the life course. The SNAP trial provides an example of a bayesian adaptive whole-of-life inclusion design that enhances trial population inclusivity and diversity overall, as well as generalizability and translation of findings into clinical practice.
Collapse
Affiliation(s)
- Anita J Campbell
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Keerthi Anpalagan
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Emma J Best
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
- The National Immunisation Advisory Centre, The University of Auckland, Auckland, New Zealand
- Department of Infectious Diseases, Starship Children's Hospital, Auckland, New Zealand
| | - Philip N Britton
- Sydney Medical School and Sydney Infectious Diseases, University of Sydney, Sydney, New South Wales, Australia
- Department of Infectious Diseases and Microbiology, the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Amanda Gwee
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Antimicrobials Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - James Hatcher
- Department of Microbiology, Great Ormond Street Hospital for Children, London, United Kingdom
- Infection, Immunity, and Inflammation Research Department, University College London, London, United Kingdom
| | - Brett J Manley
- The Royal Women's Hospital, Melbourne, Victoria, Australia
- The Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Julie Marsh
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
- Centre for Child Health research, School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Rachel H Webb
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
- Department of Infectious Diseases, Starship Children's Hospital, Auckland, New Zealand
- Department of Paediatrics, Kidz First Children's 'Hospital, Auckland, New Zealand
| | - Joshua S Davis
- Menzies School of Health Research, Charles Darwin Hospital, Darwin, Northern Territory, Australia
- John Hunter Hospital, University of Newcastle, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Robert K Mahar
- Clinical Epidemiology and Biostatistics, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Centre for Epidemiology and Biostatistics Unit, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Centre for Data Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Brendan McMullan
- Department of Infectious Diseases, Sydney Children's Hospital, Randwick, Sydney, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Michael Meyer
- Neonatal Unit, Kidz First Middlemore Hospital Auckland, Auckland, New Zealand
- Department of Paediatrics: Child and Youth Health University of Auckland, Auckland, Auckland, New Zealand
| | - Jocelyn Mora
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Srinivas Murthy
- Division of Critical Care, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Clare Nourse
- Queensland Children's Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Jesse Papenburg
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Division of Microbiology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Kevin L Schwartz
- Division of Infectious Diseases, St Joseph's Health Centre - Unity Health Toronto, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Oded Scheuerman
- Pediatrics B and Pediatric Infectious Diseases Unit, Schneider Children Medical Center Israel, Petach Tikva, Israel
- Tel Aviv University, Tel Aviv, Israel
| | - Thomas Snelling
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
- School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Tobias Strunk
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Neonatal Directorate Child and Adolescent Health Service, King Edward Memorial Hospital for Women, Subiaco, Western Australia, Australia
- Telethon Kids Institute, Perth, Western Australia, Australia
| | - Michael Stark
- The Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- The Department of Neonatal Medicine, The Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Lesley Voss
- Department of Infectious Diseases, Starship Children's Hospital, Auckland, New Zealand
| | - Steven Y C Tong
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Asha C Bowen
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
45
|
Russell CD, Berry K, Cooper G, Sim W, Lee RS, Gan TY, Donlon W, Besu A, Heppenstall E, Tysall L, Robb A, Dewar S, Smith A, Fowler VG. Distinct Clinical Endpoints of Staphylococcus aureus Bacteraemia Complicate Assessment of Outcome. Clin Infect Dis 2024; 79:604-611. [PMID: 38767234 PMCID: PMC11426269 DOI: 10.1093/cid/ciae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND We aimed to test the hypothesis that development of metastatic infection represents a distinct clinical endpoint from death due to Staphylococcus aureus bacteremia (SAB). METHODS We conducted a retrospective observational study of adults with SAB between 20 December 2019 and 23 August st2022 (n = 464). Simple logistic regression, odds ratios, and z-scores were used to compare host, clinical, and microbiologic features. RESULTS Co-occurrence of attributable mortality and metastatic infection was infrequent. Charlson Comorbidity Index and age were strongly associated with attributable mortality, but not metastatic infection. We compared patients with fatal SAB (without clinically-apparent metastatic complications, 14.4% of cohort), metastatic SAB (without attributable mortality, 22.2%), neither complication (56.7%), and overlapping fatal/metastatic SAB (6.7%). Compared to SAB without complications, fatal SAB was specifically associated with older age and multi-morbidity. Metastatic SAB was specifically associated with community acquisition, persistent fever, persistent bacteremia, and recurrence. Endocarditis was over-represented in the fatal/metastatic SAB overlap group, which shared patient characteristics with fatal SAB. In contrast to other (predominantly musculoskeletal) metastatic complications, endocarditis was associated with increased mortality, with death occurring in older multi-morbid patients later after SAB onset. CONCLUSIONS Patients with SAB experience distinct clinical endpoints: (i) early death, associated with multi-morbidity and age; (ii) metastatic (predominantly musculoskeletal) SAB; (iii) endocarditis, associated with late death occurring in older people with multi-morbidity, and (iv) bacteraemia without complications. These distinctions could be important for selecting appropriate outcomes in clinical trials: different interventions might be required to reduce mortality versus improve clinical response in patients with metastatic SAB.
Collapse
Affiliation(s)
- Clark D Russell
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
- Medical Microbiology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Karla Berry
- Clinical Infection Research Group, Western General Hospital, Edinburgh, United Kingdom
| | - George Cooper
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Wynne Sim
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rui Shian Lee
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tze Yi Gan
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - William Donlon
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Antonia Besu
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emily Heppenstall
- Medical Microbiology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Luke Tysall
- Medical Microbiology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Robb
- Scottish Microbiology Reference Laboratory, New Lister Building, Glasgow, United Kingdom
| | - Simon Dewar
- Medical Microbiology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
- Clinical Infection Research Group, Western General Hospital, Edinburgh, United Kingdom
| | - Andrew Smith
- Scottish Microbiology Reference Laboratory, New Lister Building, Glasgow, United Kingdom
- College of Medical, Veterinary & Life Sciences, Glasgow Dental Hospital & School, University of Glasgow, Glasgow, United Kingdom
| | - Vance G Fowler
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
46
|
Hendriks A, Kerkman PF, Varkila MRJ, Haitsma Mulier JLG, Ali S, Ten Doesschate T, van der Vaart TW, de Haas CJC, Aerts PC, Cremer OL, Bonten MJM, Nizet V, Liu GY, Codée JDC, Rooijakkers SHM, van Strijp JAG, van Sorge NM. Glycan-specific IgM is critical for human immunity to Staphylococcus aureus. Cell Rep Med 2024; 5:101734. [PMID: 39293400 PMCID: PMC11525025 DOI: 10.1016/j.xcrm.2024.101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/18/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
Staphylococcus aureus is a major human pathogen, yet the immune factors that protect against infection remain elusive. High titers of opsonic IgG antibodies, achieved in preclinical animal immunization studies, have consistently failed to provide protection in humans. Here, we investigate antibody responses to the conserved S. aureus surface glycan wall teichoic acid (WTA) and detect the presence of WTA-specific IgM and IgG antibodies in the plasma of healthy individuals. Functionally, WTA-specific IgM outperforms IgG in opsonophagocytic killing of S. aureus and protects against disseminated S. aureus bacteremia through passive immunization. In a clinical setting, patients with S. aureus bacteremia have significantly lower WTA-specific IgM but similar IgG levels compared to healthy controls. Importantly, low WTA-IgM levels correlate with disease mortality and impaired bacterial opsonization. Our findings may guide risk stratification of hospitalized patients and inform future design of antibody-based therapies and vaccines against serious S. aureus infection.
Collapse
Affiliation(s)
- Astrid Hendriks
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Priscilla F Kerkman
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Meri R J Varkila
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jelle L G Haitsma Mulier
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sara Ali
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Thijs Ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Thomas W van der Vaart
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Carla J C de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Piet C Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marc J M Bonten
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - George Y Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands; Netherlands Reference Center for Bacterial Meningitis, Amsterdam UMC, location AMC, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
47
|
Solís N, Pérez C, Ramírez M, Castro J, Rodríguez C. Clinical presentation and microbiological characteristics of community-acquired Staphylococcus aureus bacteraemia at a tertiary hospital in Costa Rica. J Med Microbiol 2024; 73. [PMID: 39234813 DOI: 10.1099/jmm.0.001883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Introduction. Staphylococcus aureus is a leading agent in community-acquired bacteraemia (CAB) and has been linked to elevated mortality rates and methicillin resistance in Costa Rica.Gap statement and aim. To update and enhance previous data obtained in this country, we analysed the clinical manifestations of 54 S. aureus CAB cases in a tertiary hospital and delineated the sequence types (STs), virulome, and resistome of the implicated isolates.Methodology. Clinical information was retrieved from patient files. Antibiotic susceptibility profiles were obtained with disc diffusion and automated phenotypic tests. Genomic data were exploited to type the isolates and for detection of resistance and virulence genes.Results. Primary infections predominantly manifested as bone and joint infections, followed by skin and soft tissue infections. Alarmingly, 70% of patients continued to exhibit positive haemocultures beyond 48 h of treatment modification, with nearly a quarter requiring mechanical ventilation or developing septic shock. The 30-day mortality rate reached an alarming 40%. More than 60% of the patients were found to have received suboptimal or inappropriate antibiotic treatment, and there was an alarming tendency towards the overuse of third-generation cephalosporins as empirical treatment. Laboratory tests indicated elevated creatinine levels, leukocytosis, and bandaemia within the first 24 h of hospitalization. However, most showed improvement after 48 h. The isolates were categorized into 13 STs, with a predominance of representatives from the clonal complexes CC72 (ST72), CC8 (ST8), CC5 (ST5, ST6), and CC1 (ST188). Twenty-four isolates tested positive for mecA, with ST72 strains accounting for 20. In addition, we detected genes conferring acquired resistance to aminoglycosides, MLSB antibiotics, trimethoprim/sulfamethoxazole, and mutations for fluoroquinolone resistance in the isolate collection. Genes associated with biofilm formation, capsule synthesis, and exotoxin production were prevalent, in contrast to the infrequent detection of enterotoxins or exfoliative toxin genes.Conclusions. Our findings broaden our understanding of S. aureus infections in a largely understudied region and can enhance patient management and treatment strategies.
Collapse
Affiliation(s)
- Natalia Solís
- Department of Haematology, Hospital San Juan de Dios, San José, Costa Rica
| | - Cristian Pérez
- Clinical Laboratory, Hospital Nacional de Niños, San José, Costa Rica
| | - Manuel Ramírez
- Department of Infectious Diseases, Hospital San Juan de Dios, San José, Costa Rica
| | - José Castro
- Department of Biostatistics, Caja Costarricense del Seguro Social, San José, Costa Rica
| | - César Rodríguez
- Faculty of Microbiology and Research Center for Tropical Diseases (CIET), Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
48
|
Desposito L, Bascara C. Review: sepsis guidelines and core measure bundles. Postgrad Med 2024; 136:702-711. [PMID: 39092891 DOI: 10.1080/00325481.2024.2388021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Sepsis is a major cause of mortality worldwide and is the third-leading cause of death in the United States. Sepsis is resource-intensive and requires prompt recognition and treatment to reduce mortality. The impact of sepsis is not only on in-hospital survival but extends into post-discharge quality of life and risk of re-admission. As the understanding of sepsis physiology evolved, so have the recommended screening tools and treatment protocol which challenge prior standards of care. There have been noteworthy efforts by the Surviving Sepsis Campaign, the Third International Consensus Definitions for Sepsis and the Centers for Medicare and Medicaid Services to establish core measure bundles. This review highlights both the 2021 SSC International Guidelines and the 2015 CMS Severe Sepsis/Septic Shock Core Measure Bundle, or SEP-1. Notably, the SEP-1 bundle was implemented as a value-based purchasing program, linking care of sepsis patients to financial incentives. The objective is to explore the most current evidence-based data to inform clinical practice while utilizing the available guidelines as a roadmap.
Collapse
Affiliation(s)
- Lia Desposito
- Internal Medicine, Division of Hospital Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| | - Christina Bascara
- Internal Medicine, Division of Hospital Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| |
Collapse
|
49
|
Ohta R, Sano C. Factors Affecting Recurrent Staphylococcus aureus Bacteremia Among Older Patients in Rural Community Hospitals: A Retrospective Cohort Study. Cureus 2024; 16:e70120. [PMID: 39449886 PMCID: PMC11502118 DOI: 10.7759/cureus.70120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Staphylococcus aureus bacteremia (SAB) poses a significant health risk, particularly among adults over 65 years old, due to age-related vulnerabilities and comorbidities. Recurrent SAB is associated with increased morbidity, prolonged hospitalizations, and higher healthcare costs, necessitating the identification of risk factors that contribute to these recurrent infections. Methods A retrospective cohort study was conducted at a rural community hospital to identify factors associated with recurrent SAB in older patients. Data were extracted from electronic medical records of patients diagnosed with SAB between April 2016 and December 2023. Multivariate logistic regression was employed to analyze the relationship between recurrent SAB and potential risk factors, including age, sex, BMI, dependency on Japanese long-term health insurance, and comorbidities. Results Among 99 patients with SAB, 36 (36.4%) experienced recurrence. Higher BMI was significantly associated with recurrent SAB (OR: 1.15, 95% CI: 1.01-1.31, p = 0.036), while dependency on long-term care was associated with a lower risk of recurrence (OR: 0.20, 95% CI: 0.06-0.64, p = 0.007). Age and sex did not show significant associations with recurrence. Conclusion This study identified higher BMI as a risk factor for recurrent SAB in older patients, while dependency on long-term care was protective. These findings highlight the need for targeted management strategies for patients with higher BMI to prevent recurrent SAB. Further research is needed to explore these associations and confirm their relevance in other clinical settings.
Collapse
Affiliation(s)
| | - Chiaki Sano
- Community Medicine Management, Shimane University Faculty of Medicine, Izumo, JPN
| |
Collapse
|
50
|
Omar A, El-Banna TE, Sonbol FI, El-Bouseary MM. Potential antivirulence and antibiofilm activities of sub-MIC of oxacillin against MDR S. aureus isolates: an in-vitro and in-vivo study. BMC Microbiol 2024; 24:295. [PMID: 39123138 PMCID: PMC11312681 DOI: 10.1186/s12866-024-03429-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Multi-drug resistant Staphylococcus aureus is one of the most common causes of nosocomial and community-acquired infections, with high morbidity and mortality. Treatment of such infections is particularly problematic; hence, it is complicated by antibiotic resistance, and there is currently no reliable vaccine. Furthermore, it is well known that S. aureus produces an exceptionally large number of virulence factors that worsen infection. Consequently, the urgent need for anti-virulent agents that inhibit biofilm formation and virulence factors has gained momentum. Therefore, we focused our attention on an already-approved antibiotic and explored whether changing the dosage would still result in the intended anti-virulence effect. METHODS In the present study, we determined the antibiotic resistance patterns and the MICs of oxacillin against 70 MDR S. aureus isolates. We also investigated the effect of sub-MICs of oxacillin (at 1/4 and 1/8 MICs) on biofilm formation using the crystal violet assay, the phenol-sulphuric acid method, and confocal laser scanning microscopy (CLSM). We examined the effect of sub-MICs on virulence factors and bacterial morphology using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and electron microscopy, respectively. Moreover, we studied the effect of sub-MICs of oxacillin (OX) in-vivo using a wound infection model. RESULTS Oxacillin at 1/2 MIC showed a significant decrease in bacterial viability, while 1/4 and 1/8 MICs had negligible effects on treated bacterial isolates. Treatment of MDR isolates with 1/4 or 1/8 MICs of oxacillin significantly reduced biofilm formation (64% and 40%, respectively). The treated MDR S. aureus with sub-MICs of OX exhibited a dramatic reduction in several virulence factors, including protease, hemolysin, coagulase, and toxic shock syndrome toxin-1 (TSST-1) production. The sub-MICs of OX significantly decreased (P < 0.05) the gene expression of biofilm and virulence-associated genes such as agrA, icaA, coa, and tst. Furthermore, oxacillin at sub-MICs dramatically accelerated wound healing, according to the recorded scoring of histological parameters. CONCLUSION The treatment of MDR S. aureus with sub-MICs of oxacillin can help in combating the bacterial resistance and may be considered a promising approach to attenuating the severity of S. aureus infections due to the unique anti-biofilm and anti-virulence activities.
Collapse
Affiliation(s)
- Amira Omar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Tarek E El-Banna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Fatma I Sonbol
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Maisra M El-Bouseary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| |
Collapse
|