1
|
Maraolo AE, Gatti M, Principe L, Marino A, Pipitone G, De Pascale G, Ceccarelli G. Management of methicillin-resistant Staphylococcus aureus bloodstream infections: a comprehensive narrative review of available evidence focusing on current controversies and the challenges ahead. Expert Rev Anti Infect Ther 2025:1-26. [PMID: 40165471 DOI: 10.1080/14787210.2025.2487163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Bloodstream infections (BSIs) caused by Staphylococcus aureus are common worldwide, representing one of the most relevant issues in clinical infectious diseases practice. In particular, BSIs by methicillin-resistant S. aureus (MRSA-BSI) are still today a challenge since mortality burden remains elevated although decades of research. AREAS COVERED The following topics regarding MRSA-BSI were reviewed and discussed by resorting to best available evidence retrieved from PubMed/MEDLINE up to October 2024: i) epidemiology; ii) microbiology; iii) classification, with a focus on complicated and not complicated forms; iv) the structured approach to the patient; v) pharmacokinetics and pharmacodynamics of the main antimicrobial options; vi) controversies regarding the best therapeutic approach. EXPERT OPINION Despite ongoing efforts to better stratify and manage MRSA-BSI, there is no universally accepted classification system accurately distinguishing between uncomplicated/low risk and complicated/high risk forms. Biomarkers such as interleukin(IL)-10 hold promise in order to enable a more precise stratification, premise for an appropriate treatment plan. There is a theoretical rationale for implementing a combination therapy including a beta-lactam agent upfront, especially for patients considered at higher risk of unfavorable outcomes, but further data are necessary, and the same applies to newer adjuvants. Novel microbiological techniques may help in guiding antimicrobial duration.
Collapse
Affiliation(s)
- Alberto Enrico Maraolo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luigi Principe
- Microbiology and Virology Unit, Great Metropolitan Hospital "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Andrea Marino
- Department of Clinical and Experimental Medicine, Infectious Diseases Unit, ARNAS Garibaldi Hospital, University of Catania, Catania, Italy
| | | | - Gennaro De Pascale
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze dell 'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University Hospital Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
2
|
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
|
3
|
Clegg J, Mnich ME, Carignano A, Cova G, Tavarini S, Sammicheli C, Clemente B, Smith M, Siena E, Bardelli M, Brazzoli M, Bagnoli F, McLoughlin RM, Soldaini E. Staphylococcus aureus-specific TIGIT + Treg are present in the blood of healthy subjects - a hurdle for vaccination? Front Immunol 2025; 15:1500696. [PMID: 39981298 PMCID: PMC11840346 DOI: 10.3389/fimmu.2024.1500696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/12/2024] [Indexed: 02/22/2025] Open
Abstract
Staphylococcus aureus poses an enormous burden of morbidity and mortality worldwide. Making an efficacious vaccine has however proven extremely challenging. Due to colonizing interactions, pre-existing S. aureus-specific CD4+ T cells are often found in the human population and yet a detailed characterization of their phenotypes and how they might in turn impact vaccine efficacy are thus far unknown. Using an activation induced marker assay to sort for S. aureus-specific CD4+ T cells in an effector function-independent manner, single cell transcriptomic analysis was conducted. Remarkably, S. aureus-specific CD4+ T cells consisted not only of a broader spectrum of conventional T cells (Tcon) than previously described but also of regulatory T cells (Treg). As compared to polyclonally-activated CD4+ T cells, S. aureus-specific Tcon were enriched for the expression of the Th17-type cytokine genes IL17A, IL22 and IL26, while higher percentages of S. aureus-specific Treg expressed the T Cell Immunoreceptor with Ig and ITIM domains (TIGIT), a pleiotropic immune checkpoint. Notably, the antagonistic anti-TIGIT mAb Tiragolumab increased IL-1β production in response to S. aureus in vitro. Therefore, these results uncover the presence of S. aureus-specific TIGIT+ Treg in the blood of healthy subjects that could blunt responses to vaccination and indicate TIGIT as a potential targetable biomarker to overcome pre-exposure-induced immunosuppression.
Collapse
Affiliation(s)
- Jonah Clegg
- GSK, Research Center, Siena, Italy
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Malgorzata E. Mnich
- GSK, Research Center, Siena, Italy
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | | | | | | | - Megan Smith
- GSK, Research Center, Siena, Italy
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Rachel M. McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
4
|
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
|
5
|
Tsai CM, Hajam IA, Caldera JR, Chiang AW, Gonzalez C, Du X, Choudhruy B, Li H, Suzuki E, Askarian F, Clark T, Lin B, Wierzbicki IH, Riestra AM, Conrad DJ, Gonzalez DJ, Nizet V, Lewis NE, Liu GY. Pathobiont-driven antibody sialylation through IL-10 undermines vaccination. J Clin Invest 2024; 134:e179563. [PMID: 39680460 DOI: 10.1172/jci179563] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024] Open
Abstract
The pathobiont Staphylococcus aureus (Sa) induces nonprotective antibody imprints that underlie ineffective staphylococcal vaccination. However, the mechanism by which Sa modifies antibody activity is not clear. Herein, we demonstrate that IL-10 is the decisive factor that abrogates antibody protection in mice. Sa-induced B10 cells drive antigen-specific vaccine suppression that affects both recalled and de novo developed B cells. Released IL-10 promotes STAT3 binding upstream of the gene encoding sialyltransferase ST3gal4 and increases its expression by B cells, leading to hyper-α2,3sialylation of antibodies and loss of protective activity. IL-10 enhances α2,3sialylation on cell-wall-associated IsdB, IsdA, and MntC antibodies along with suppression of the respective Sa vaccines. Consistent with mouse findings, human anti-Sa antibodies as well as anti-pseudomonal antibodies from cystic fibrosis subjects (high IL-10) are hypersialylated, compared with anti-Streptococcus pyogenes and pseudomonal antibodies from normal individuals. Overall, we demonstrate a pathobiont-centric mechanism that modulates antibody glycosylation through IL-10, leading to loss of staphylococcal vaccine efficacy.
Collapse
Affiliation(s)
- Chih-Ming Tsai
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - Irshad A Hajam
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - J R Caldera
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - Austin Wt Chiang
- Immunology Center of Georgia and Department of Medicine, Augusta University, Augusta, Georgia, USA
| | - Cesia Gonzalez
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - Xin Du
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - Biswa Choudhruy
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | - Haining Li
- Department of Bioengineering, University of California, La Jolla, California, USA
| | - Emi Suzuki
- Division of Gastroenterology, Department of Pediatrics, UCSD, La Jolla, California, USA
- Division of Gastroenterology, Rady Children's Hospital, San Diego, California, USA
| | - Fatemeh Askarian
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
| | - Ty'Tianna Clark
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Brian Lin
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
| | - Igor H Wierzbicki
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Angelica M Riestra
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Douglas J Conrad
- Division of Pulmonary, Critical Care and Sleep Medicine, UCSD, La Jolla, California, USA
| | - David J Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Nathan E Lewis
- Department of Bioengineering, University of California, La Jolla, California, USA
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, California, USA
| | - George Y Liu
- Division of Infectious Diseases, Department of Pediatrics, University of California, La Jolla, California, USA
- Division of Infectious Diseases, Rady Children's Hospital, San Diego, California, USA
| |
Collapse
|
6
|
Torres VJ. Interleukin 10 drives Staphylococcus aureus imprinting and vaccine failure in murine models via antibody glycosylation. J Clin Invest 2024; 134:e187055. [PMID: 39680461 DOI: 10.1172/jci187055] [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: 12/18/2024] Open
Abstract
Despite many attempts, there is currently no approved vaccine to prevent Staphylococcus aureus infections. Preclinical vaccination models have failed to predict vaccine efficacy in humans as S. aureus exposure in humans imprints an immune response that is lacking in naive animals. In this issue of the JCI, Tsai and colleagues identify the cytokine IL-10 as the driver of humoral imprinting by S. aureus. Upon vaccination, S. aureus-experienced animals produced copious levels of IL-10, resulting in the hyper-α2,3 sialylation of antibodies, which interfered with the phagocytic-promoting properties of the vaccine-elicited anti-S. aureus antibodies. These findings correlate with the observation that hyperproduction of IL-10 in humans also induces hyper-α2,3 sialylation of antibodies and provide a possible mechanism for previous vaccine failures.
Collapse
|
7
|
Bakthavatchalam YD, Karthik R, Ashok A, Walia K, Ragothaman H, Krishnamoorthy S, Manokaran Y, Veeraraghavan B. Synergistic approach of ertapenem and flucloxacillin or cefazolin for addressing persistent methicillin susceptible Staphylococcus aureus bacteremia. Indian J Med Microbiol 2024; 52:100755. [PMID: 39550069 DOI: 10.1016/j.ijmmb.2024.100755] [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: 06/29/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
In the present study, we determine the synergistic activity of ertapenem with cefazolin and flucloxacillin against type A beta-lactamase producing Methicillin susceptible Staphylococcus aureus isolate. In the time kill assay, at standard inoculum, ertapenem with cefazolin showed >1 log kill and ertapenem with flucloxacillin demonstrated >2 log kill. When tested at high inoculum, both combinations achieved >1 log kill at 1x MIC.
Collapse
Affiliation(s)
| | - Rajiv Karthik
- Department of Infectious Disease, Christian Medical College, Vellore, India.
| | - Anand Ashok
- Department of Orthopaedics, Christian Medical College, Vellore, India.
| | - Kamini Walia
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India.
| | - Harathi Ragothaman
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
| | | | - Yuvasri Manokaran
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
| | | |
Collapse
|
8
|
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
|
9
|
Volk CF, Proctor RA, Rose WE. The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia. Int J Mol Sci 2024; 25:6486. [PMID: 38928191 PMCID: PMC11203666 DOI: 10.3390/ijms25126486] [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: 05/20/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.
Collapse
Affiliation(s)
- Cecilia F. Volk
- Pharmacy Practice and Translational Research Division, School of Pharmacy, Pharmacy University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Richard A. Proctor
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Warren E. Rose
- Pharmacy Practice and Translational Research Division, School of Pharmacy, Pharmacy University of Wisconsin-Madison, Madison, WI 53705, USA;
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
10
|
Parmar R, Pickering H, Ahn R, Rossetti M, Gjertson DW, Ruffin F, Chan LC, Fowler VG, Yeaman MR, Reed EF. Integrated transcriptomic analysis reveals immune signatures distinguishing persistent versus resolving outcomes in MRSA bacteremia. Front Immunol 2024; 15:1373553. [PMID: 38846955 PMCID: PMC11153731 DOI: 10.3389/fimmu.2024.1373553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Staphylococcus aureus bacteremia (SAB) is a life-threatening infection particularly involving methicillin-resistant S. aureus (MRSA). In contrast to resolving MRSA bacteremia (RB), persistent MRSA bacteremia (PB) blood cultures remain positive despite appropriate antibiotic treatment. Host immune responses distinguishing PB vs. RB outcomes are poorly understood. Here, integrated transcriptomic, IL-10 cytokine levels, and genomic analyses sought to identify signatures differentiating PB vs. RB outcomes. Methods Whole-blood transcriptomes of propensity-matched PB (n=28) versus RB (n=30) patients treated with vancomycin were compared in one independent training patient cohort. Gene expression (GE) modules were analyzed and prioritized relative to host IL-10 cytokine levels and DNA methyltransferase-3A (DNMT3A) genotype. Results Differential expression of T and B lymphocyte gene expression early in MRSA bacteremia discriminated RB from PB outcomes. Significant increases in effector T and B cell signaling pathways correlated with RB, lower IL-10 cytokine levels and DNMT3A heterozygous A/C genotype. Importantly, a second PB and RB patient cohort analyzed in a masked manner demonstrated high predictive accuracy of differential signatures. Discussion Collectively, the present findings indicate that human PB involves dysregulated immunity characterized by impaired T and B cell responses associated with excessive IL-10 expression in context of the DNMT3A A/A genotype. These findings reveal distinct immunologic programs in PB vs. RB outcomes, enable future studies to define mechanisms by which host and/or pathogen drive differential signatures and may accelerate prediction of PB outcomes. Such prognostic assessment of host risk could significantly enhance early anti-infective interventions to avert PB and improve patient outcomes.
Collapse
Affiliation(s)
- Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Richard Ahn
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Liana C. Chan
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Vance G. Fowler
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Michael R. Yeaman
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
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. RESEARCH SQUARE 2024:rs.3.rs-4095478. [PMID: 38559201 PMCID: PMC10980158 DOI: 10.21203/rs.3.rs-4095478/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
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 hours 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 hours. 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
|
12
|
Sakoulas G, Nizet V. Measuring beta-lactam minimum inhibitory concentrations in Staphylococcus aureus in the clinical microbiology laboratory: pinning the tail on the donkey. J Clin Microbiol 2024; 62:e0036623. [PMID: 37966224 PMCID: PMC10793257 DOI: 10.1128/jcm.00366-23] [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] [Indexed: 11/16/2023] Open
Abstract
Significant shortcomings have been identified in standard methods of susceptibility testing in bacteriological media, not only because the media fails to recapitulate the in vivo environment, but susceptibility testing itself fails to capture sub-MIC effects that significantly attenuate bacterial virulence properties. Until susceptibility testing conditions better recapitulate the in vivo environment, attempts to establish the quantitative relevance of beta-lactam MIC using current clinical microbiology standards in Staphylococcus aureus infections will likely prove unsuccessful.
Collapse
Affiliation(s)
- George Sakoulas
- Sharp Rees-Stealy Medical Group, San Diego, California, USA
- UCSD School of Medicine, La Jolla, California, USA
| | - Victor Nizet
- UCSD School of Medicine, La Jolla, California, USA
- Skaggs School of Pharmacy, UCSD School of Medicine, La Jolla, California, USA
| |
Collapse
|
13
|
Alfano DN, Miller MJ, Bubeck Wardenburg J. Endothelial ADAM10 utilization defines a molecular pathway of vascular injury in mice with bacterial sepsis. J Clin Invest 2023; 133:e168450. [PMID: 37788087 PMCID: PMC10688991 DOI: 10.1172/jci168450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
The endothelium plays a critical role in the host response to infection and has been a focus of investigation in sepsis. While it is appreciated that intravascular thrombus formation, severe inflammation, and loss of endothelial integrity impair tissue oxygenation during sepsis, the precise molecular mechanisms that lead to endothelial injury remain poorly understood. We demonstrate here that endothelial ADAM10 was essential for the pathogenesis of Staphylococcus aureus sepsis, contributing to α-toxin-mediated (Hla-mediated) microvascular thrombus formation and lethality. As ADAM10 is essential for endothelial development and homeostasis, we examined whether other major human sepsis pathogens also rely on ADAM10-dependent pathways in pathogenesis. Mice harboring an endothelium-specific knockout of ADAM10 were protected against lethal Pseudomonas aeruginosa and Streptococcus pneumoniae sepsis, yet remained fully susceptible to group B streptococci and Candida albicans sepsis. These studies illustrate a previously unknown role for ADAM10 in sepsis-associated endothelial injury and suggest that understanding pathogen-specific divergent host pathways in sepsis may enable more precise targeting of disease.
Collapse
Affiliation(s)
| | - Mark J. Miller
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
14
|
Wang Y, Ahmadi MZ, Dikeman DA, Youn C, Archer NK. γδ T cell-intrinsic IL-1R promotes survival during Staphylococcus aureus bacteremia. Front Immunol 2023; 14:1171934. [PMID: 37483624 PMCID: PMC10361057 DOI: 10.3389/fimmu.2023.1171934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Staphylococcus aureus is a leading cause of bacteremia, further complicated by the emergence of antibiotic-resistant strains such as methicillin-resistant S. aureus (MRSA). A better understanding of host defense mechanisms is needed for the development of host-directed therapies as an alternative approach to antibiotics. The levels of IL-1, IL-17, and TNF-α cytokines in circulation have been associated with predictive outcomes in patients with S. aureus bacteremia. However, their causative role in survival and the cell types involved in these responses during bacteremia is not entirely clear. Using a mouse model of S. aureus bacteremia, we demonstrated that IL-17A/F and TNF-α had no significant impact on survival, whereas IL-1R signaling was critical for survival during S. aureus bacteremia. Furthermore, we identified that T cells, but not neutrophils, monocytes/macrophages, or endothelial cells were the crucial cell type for IL-1R-mediated survival against S. aureus bacteremia. Finally, we determined that the expression of IL-1R on γδ T cell, but not CD4+ or CD8+ T cells was responsible for survival against the S. aureus bacteremia. Taken together, we uncovered a role for IL-1R, but not IL-17A/F and TNF-α in protection against S. aureus bacteremia. Importantly, γδ T cell-intrinsic expression of IL-1R was crucial for survival, but not on other immune cells or endothelial cells. These findings reveal potential cellular and immunological targets for host-directed therapies for improved outcomes against S. aureus bacteremia.
Collapse
Affiliation(s)
| | | | | | | | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
Heithoff DM, Barnes V L, Mahan SP, Fried JC, Fitzgibbons LN, House JK, Mahan MJ. Re-evaluation of FDA-approved antibiotics with increased diagnostic accuracy for assessment of antimicrobial resistance. Cell Rep Med 2023; 4:101023. [PMID: 37116500 PMCID: PMC10213814 DOI: 10.1016/j.xcrm.2023.101023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/16/2023] [Accepted: 04/05/2023] [Indexed: 04/30/2023]
Abstract
Accurate assessment of antibiotic susceptibility is critical for treatment of antimicrobial resistant (AMR) infections. Here, we examine whether antimicrobial susceptibility testing in media more physiologically representative of in vivo conditions improves prediction of clinical outcome relative to standard bacteriologic medium. This analysis reveals that ∼15% of minimum inhibitory concentration (MIC) values obtained in physiologic media predicted a change in susceptibility that crossed a clinical breakpoint used to categorize patient isolates as susceptible or resistant. The activities of antibiotics having discrepant results in different media were evaluated in murine sepsis models. Testing in cell culture medium improves the accuracy by which MIC assays predict in vivo efficacy. This analysis identifies several antibiotics for treatment of AMR infections that standard testing failed to identify and those that are ineffective despite indicated use by standard testing. Methods with increased diagnostic accuracy mitigate the AMR crisis via utilizing existing agents and optimizing drug discovery.
Collapse
Affiliation(s)
- Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Lucien Barnes V
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Scott P Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, Santa Barbara, CA 93106, USA; Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Jeffrey C Fried
- Department of Medical Education, Santa Barbara Cottage Hospital, Santa Barbara, CA 93105, USA; Department of Pulmonary and Critical Care Medicine, Santa Barbara Cottage Hospital, Santa Barbara, CA 93105, USA
| | - Lynn N Fitzgibbons
- Department of Medical Education, Santa Barbara Cottage Hospital, Santa Barbara, CA 93105, USA; Division of Infectious Diseases, Santa Barbara Cottage Hospital, Santa Barbara, CA 93105, USA
| | - John K House
- Faculty of Science, School of Veterinary Science, The University of Sydney, Camden, NSW 2570, Australia.
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
16
|
Cuervo G. PET/CT to diagnose and manage patients with infectious diseases: "¡vamos a brillar, mi amor! (Let's shine, my love!)". ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2023; 41:1-2. [PMID: 36621242 DOI: 10.1016/j.eimce.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/09/2023]
Affiliation(s)
- Guillermo Cuervo
- Department of Infectious Diseases, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
17
|
Xiao L, Ran X, Zhong Y, Le Y, Li S. A combined ratio change of inflammatory biomarkers at 72 h could predict the severity and prognosis of sepsis from pulmonary infections. Immunobiology 2022; 227:152290. [DOI: 10.1016/j.imbio.2022.152290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/24/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022]
|
18
|
Cuervo G. PET/CT to diagnose and manage patients with infectious diseases: “¡vamos a brillar, mi amor! (Let's shine, my love!)”. Enferm Infecc Microbiol Clin 2022. [DOI: 10.1016/j.eimc.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
19
|
Weiss-Tessbach M, Ratzinger F, Obermueller M, Burgmann H, Staudinger T, Robak O, Schmid M, Roessler B, Jilma B, Kussmann M, Traby L. Biomarkers for differentiation of coronavirus disease 2019 or extracorporeal membrane oxygenation related inflammation and bacterial/fungal infections in critically ill patients: A prospective observational study. Front Med (Lausanne) 2022; 9:917606. [PMID: 36275812 PMCID: PMC9582266 DOI: 10.3389/fmed.2022.917606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Secondary infections in coronavirus disease 2019 (COVID-19) patients are difficult to distinguish from inflammation associated with COVID-19 and/or extracorporeal membrane oxygenation (ECMO). Therefore, highly specific and sensitive biomarkers are needed to identify patients in whom antimicrobial therapy can be safely withheld. In this prospective monocentric study, 66 COVID-19 patients admitted to the intensive care unit (ICU) for ECMO evaluation were included. A total of 46 (70%) patients with secondary infections were identified by using broad microbiological and virological panels and standardized diagnostic criteria. Various laboratory parameters including C-reactive protein (CRP), interleukin (IL)-6, procalcitonin (PCT), and IL-10 were determined at time of study inclusion. The best test performance for differentiating bacterial/fungal secondary infections and COVID-19 and/or ECMO associated inflammation was achieved by IL-10 (ROC-AUC 0.84) and a multivariant step-wise regression model including CRP, IL-6, PCT, and IL-10 (ROC-AUC 0.93). Data obtained in the present study highlights the use of IL-10 to differentiate secondary bacterial/fungal infections from COVID-19 and/or ECMO associated inflammation in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Matthias Weiss-Tessbach
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Franz Ratzinger
- Ihr Labor, Medical Diagnostics Laboratories, Vienna, Austria
| | - Markus Obermueller
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas Staudinger
- Department of Medicine I, Intensive Care Unit 13i2, Medical University of Vienna, Vienna, Austria
| | - Oliver Robak
- Department of Medicine I, Intensive Care Unit 13i2, Medical University of Vienna, Vienna, Austria
| | - Monika Schmid
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bernhard Roessler
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical Simulation and Emergency Management Research Group, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuel Kussmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria,*Correspondence: Manuel Kussmann,
| | - Ludwig Traby
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria,Ludwig Traby,
| |
Collapse
|
20
|
TLR2, TLR4, and NLRP3 mediated the balance between host immune-driven resistance and tolerance in Staphylococcus aureus-infected mice. Microb Pathog 2022; 169:105671. [PMID: 35811022 DOI: 10.1016/j.micpath.2022.105671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 01/02/2023]
Abstract
Staphylococcus aureus (S. aureus) is a gram-positive pathogen that can cause infectious diseases in mammals. S. aureus-induced host innate immune responses have a relationship with Toll-like receptor 2 (TLR2), TLR4, and Nod-like receptor pyrin domain-containing protein 3 (NLRP3). However, the detailed roles of TLR2, TLR4, and NLRP3 in regulating the host inflammatory response to S. aureus infection remain unclear. Our data indicated that the S. aureus-induced mortality was aggravated by deficiency of TLR2, TLR4, and NLRP3 in mice. In the subsequent experiment, we found that during S. aureus infection, the roles of TLR2, TLR4, and NLRP3 seemed to be different at multiple timepoints. The deficiency of TLR2, TLR4, or NLRP3 attenuated the expression of High-mobility group box protein 1 (HMGB1) and Hyaluronic acid-binding protein 2 (HABP2), which is accompanied by decreased proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum at 3 h and 6 h post-infection. However, with S. aureus infection prolonged (24 h post-infection), the trend was diametrically opposite. The results showed that deficiency of TLR2, TLR4, or NLRP3 aggravated HABP2 and HMGB1 expression, which is accompanied by enhanced proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum. These results were consistent with the data observed in S. aureus-infected bone marrow-derived macrophages (BMDMs). All these results suggested that during S. aureus infection, TLR2, TLR4, and NLRP3 has time-dependent effect in regulating the balance between immune-driven resistance and tolerance.
Collapse
|
21
|
Tsai CM, Hajam IA, Caldera JR, Liu GY. Integrating complex host-pathogen immune environments into S. aureus vaccine studies. Cell Chem Biol 2022; 29:730-740. [PMID: 35594849 DOI: 10.1016/j.chembiol.2022.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/16/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022]
Abstract
Staphylococcus aureus (SA) is a leading cause of bacterial infection and antibiotic resistance globally. Therefore, development of an effective vaccine has been a major goal of the SA field for the past decades. With the wealth of understanding of pathogenesis, the failure of all SA vaccine trials has been a surprise. We argue that experimental SA vaccines have not worked because vaccines have been studied in naive laboratory animals, whereas clinical vaccine efficacy is tested in immune environments reprogrammed by SA. Here, we review the failed SA vaccines that have seemingly defied all principles of vaccinology. We describe major SA evasion strategies and suggest that they reshape the immune environment in a way that makes vaccines prone to failures. We propose that appropriate integration of concepts of host-pathogen interaction into vaccine study designs could lead to insight critical for the development of an effective SA vaccine.
Collapse
Affiliation(s)
- Chih-Ming Tsai
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Irshad A Hajam
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - J R Caldera
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - George Y Liu
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Division of Infectious Diseases, Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
22
|
Gilbertie J, Ulloa ER, Daiker JC, Nguyen K, Smelter D, Rose W, Geriak M, Schnabel LV, Nizet V, Sakoulas G. Potent Activity of Ertapenem Plus Cefazolin Within Staphylococcal Biofilms: A Contributing Factor in the Treatment of Methicillin-Susceptible Staphylococcus aureus Endocarditis. Open Forum Infect Dis 2022; 9:ofac159. [PMID: 35493130 PMCID: PMC9045957 DOI: 10.1093/ofid/ofac159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022] Open
Abstract
Background Besides antistaphylococcal beta-lactams and source control, there are limited validated antimicrobial salvage options in patients with prolonged methicillin-susceptible Staphylococcus aureus (MSSA) bacteremia, including infective endocarditis (IE). Methods MSSA IE cases treated with ertapenem (ETP) plus cefazolin (CZ) were compared with matched IE cases treated with standard beta-lactam monotherapy. The bactericidal activity of ETP plus CZ was also compared with nafcillin (NAF), CZ, and ETP alone using an in vitro MSSA biofilm model. Results The median duration of bacteremia experienced by patients (n = 12) while on CZ or NAF was 4 days (range 1-16 days) compared with 1 day (range 1-3 days) for patients (n = 5) treated with ETP + CZ (P = .01, Mann-Whitney U test). Cefazolin and NAF alone or in combination did not achieve biofilm eradication at clinically relevant concentrations. However, the addition of ETP to CZ led to bactericidal eradication within biofilms at standard dosing. Conclusions Ertapenem reduces CZ concentrations required to eradicate MSSA biofilms to those achievable in vivo by standard dosing, translating into shorter bacteremia duration in patients with MSSA endocarditis. Larger studies are needed to investigate ETP plus CZ therapy in the treatment of biofilm-related MSSA infections such as endocarditis.
Collapse
Affiliation(s)
- Jessica Gilbertie
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Erlinda R Ulloa
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, California, USA.,Division of Infectious Disease, Children's Hospital of Orange County, Orange, California, USA
| | - Jennifer C Daiker
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Khanh Nguyen
- Sharp Memorial Hospital, San Diego, California, USA
| | - Dan Smelter
- Department and School of Pharmacy, University of Wisconsin Health, Madison, Wisconsin, USA
| | - Warren Rose
- Department and School of Pharmacy, University of Wisconsin Health, Madison, Wisconsin, USA
| | | | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Victor Nizet
- Collaborative to Halt Antibiotic-Resistant Microbes (CHARM), Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA.,Skaggs School of Pharmacy, University of California San Diego, La Jolla, California, USA
| | - George Sakoulas
- Sharp Memorial Hospital, San Diego, California, USA.,Collaborative to Halt Antibiotic-Resistant Microbes (CHARM), Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
23
|
Rose W, Volk C, Dilworth TJ, Sakoulas G. Approaching 65 Years: Is it Time to Consider Retirement of Vancomycin for Treating Methicillin-Resistant Staphylococcus aureus Endovascular Infections? Open Forum Infect Dis 2022; 9:ofac137. [PMID: 35493116 PMCID: PMC9043000 DOI: 10.1093/ofid/ofac137] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022] Open
Abstract
Vancomycin was introduced nearly 65 years ago and remains the standard antibiotic for serious methicillin-resistant Staphylococcus aureus (MRSA) infections. Staphylococcus aureus remains highly susceptibility to vancomycin (>97%). Despite this, MRSA treatment failure with vancomycin is high in complicated bacteremia. Additionally, vancomycin can cause nephrotoxicity, leading to new therapeutic drug monitoring guidance. This demonstrates how difficult it is to dose vancomycin in a way that is both efficacious and safe, especially during long courses of therapy. Often underappreciated are the cost, resources, and complexity of vancomycin care at a time when alternative antibiotics are becoming cost comparable. This perspective highlights a bigger picture of how the treatment repertoires of many other diseases have changed and advanced since vancomycin’s introduction in the 1950s, yet the vancomycin MRSA treatment standard remains. While vancomycin can still have a role, 65 years may be a practical retirement age for vancomycin in highly complex endovascular infections.
Collapse
Affiliation(s)
- Warren Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Cecilia Volk
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas J Dilworth
- Department of Pharmacy Services, Advocate Aurora Health, Milwaukee, WI, USA
| | - George Sakoulas
- Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
24
|
Infection Biomarkers Based on Metabolomics. Metabolites 2022; 12:metabo12020092. [PMID: 35208167 PMCID: PMC8877834 DOI: 10.3390/metabo12020092] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/18/2022] Open
Abstract
Current infection biomarkers are highly limited since they have low capability to predict infection in the presence of confounding processes such as in non-infectious inflammatory processes, low capability to predict disease outcomes and have limited applications to guide and evaluate therapeutic regimes. Therefore, it is critical to discover and develop new and effective clinical infection biomarkers, especially applicable in patients at risk of developing severe illness and critically ill patients. Ideal biomarkers would effectively help physicians with better patient management, leading to a decrease of severe outcomes, personalize therapies, minimize antibiotics overuse and hospitalization time, and significantly improve patient survival. Metabolomics, by providing a direct insight into the functional metabolic outcome of an organism, presents a highly appealing strategy to discover these biomarkers. The present work reviews the desired main characteristics of infection biomarkers, the main metabolomics strategies to discover these biomarkers and the next steps for developing the area towards effective clinical biomarkers.
Collapse
|
25
|
Is the Success of Cefazolin plus Ertapenem in Methicillin-Susceptible
Staphylococcus aureus
Bacteremia Based on Release of Interleukin 1-beta? Antimicrob Agents Chemother 2022; 66:e0216621. [DOI: 10.1128/aac.02166-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cefazolin and ertapenem has been shown to be an effective salvage regimen for refractory methicillin-susceptible
Staphylococcus aureus
bacteremia. Our findings suggest cefazolin plus ertapenem
in vitro
stimulates interleukin-1β release from peripheral blood monocytes both with and without
S. aureus
presence. This IL-1β augmentation was primarily driven by ertapenem. These findings support further exploration of cefazolin plus ertapenem in MSSA bacteremia and may partially explain its marked potency
in vivo
despite modest synergy
in vitro
.
Collapse
|
26
|
Duan L, Zhang J, Chen Z, Gou Q, Xiong Q, Yuan Y, Jing H, Zhu J, Ni L, Zheng Y, Liu Z, Zhang X, Zeng H, Zou Q, Zhao Z. Antibiotic Combined with Epitope-Specific Monoclonal Antibody Cocktail Protects Mice Against Bacteremia and Acute Pneumonia from Methicillin-Resistant Staphylococcus aureus Infection. J Inflamm Res 2021; 14:4267-4282. [PMID: 34511967 PMCID: PMC8415768 DOI: 10.2147/jir.s325286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose We previously reported that monoclonal antibody (mAb) cocktail improves survival in Staphylococcus aureus infection. In this study, we used acute pneumonia model and lethal sepsis model to investigate the efficacy of antibiotic combined with epitope-specific mAb cocktail in treating MRSA252 infection. Methods MRSA252 was challenged by tail vein injection or tracheal intubation to establish sepsis model or pneumonia model. One hour after infection, the mice received a single intravenous injection of normal saline, vancomycin, and vancomycin combined monoclonal antibody, linezolid alone or linezolid combined monoclonal antibody. Daily record survival rate (total 7 days), bacterial load, histology, cytokine analysis of serum and alveolar lavage fluid, and in vitro determination of the neutralizing ability of antibodies to SEB toxin and Hla toxin explained the mechanism of antibody action. Results The mAb cocktail combined with low doses of vancomycin or linezolid improved survival rates in acute pneumonia model (70%, 80%) and lethal sepsis model (80%, 80%). Epitope-specific monoclonal antibodies reduced bacterial colonization in the kidneys and lungs of mice and inhibited the biological functions of the toxins Hla and SEB in vitro. Compared to the antibiotic alone or PBS groups, the combination group had higher levels of IL-1α, IL-1β and IFN-γ and lower levels of IL-6, IL-10, TNF-α. Further, the combination of antibiotic and mAb cocktail improved infection survival against the clinical MRSA isolates in a lethal sepsis model. Conclusion This study demonstrates a novel method to treat people with low immunity against drug-resistant S. aureus infections.
Collapse
Affiliation(s)
- LianLi Duan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Qingshan Xiong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Jiang Zhu
- Department of Pathology, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Li Ni
- Obstetrics and Gynecology, The First People's Hospital of Jiulongpo District, Chongqing, 400050, People's Republic of China
| | - Yuling Zheng
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, People's Republic of China
| | - Zhiyong Liu
- Department of Laboratory Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Xiaokai Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, People's Republic of China
| |
Collapse
|
27
|
Sun J, Uchiyama S, Olson J, Morodomi Y, Cornax I, Ando N, Kohno Y, Kyaw MMT, Aguilar B, Haste NM, Kanaji S, Kanaji T, Rose WE, Sakoulas G, Marth JD, Nizet V. Repurposed drugs block toxin-driven platelet clearance by the hepatic Ashwell-Morell receptor to clear Staphylococcus aureus bacteremia. Sci Transl Med 2021; 13:13/586/eabd6737. [PMID: 33762439 DOI: 10.1126/scitranslmed.abd6737] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus (SA) bloodstream infections cause high morbidity and mortality (20 to 30%) despite modern supportive care. In a human bacteremia cohort, we found that development of thrombocytopenia was correlated to increased mortality and increased α-toxin expression by the pathogen. Platelet-derived antibacterial peptides are important in bloodstream defense against SA, but α-toxin decreased platelet viability, induced platelet sialidase to cause desialylation of platelet glycoproteins, and accelerated platelet clearance by the hepatic Ashwell-Morell receptor (AMR). Ticagrelor (Brilinta), a commonly prescribed P2Y12 receptor inhibitor used after myocardial infarction, blocked α-toxin-mediated platelet injury and resulting thrombocytopenia, thereby providing protection from lethal SA infection in a murine intravenous challenge model. Genetic deletion or pharmacological inhibition of AMR stabilized platelet counts and enhanced resistance to SA infection, and the anti-influenza sialidase inhibitor oseltamivir (Tamiflu) provided similar therapeutic benefit. Thus, a "toxin-platelet-AMR" regulatory pathway plays a critical role in the pathogenesis of SA bloodstream infection, and its elucidation provides proof of concept for repurposing two commonly prescribed drugs as adjunctive therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Josh Sun
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Satoshi Uchiyama
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yosuke Morodomi
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Ingrid Cornax
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nao Ando
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yohei Kohno
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - May M T Kyaw
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Bernice Aguilar
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nina M Haste
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Sachiko Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Taisuke Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Warren E Rose
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - George Sakoulas
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA
| | - Jamey D Marth
- Center for Nanomedicine, UC Santa Barbara, Santa Barbara, CA 93106, USA.,Sanford Burnham Prebys Medical Discovery Institute, UC Santa Barbara, Santa Barbara, CA 93106, USA
| | - Victor Nizet
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA. .,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
28
|
Eichenberger EM, Ruffin F, Dagher M, Lerebours R, Jung SH, Sharma-Kuinkel B, Macintyre AN, Thaden JT, Sinclair M, Hale L, Kohler C, Palmer SM, Alexander BD, Fowler VG, Maskarinec SA. Bacteremia in solid organ transplant recipients as compared to immunocompetent patients: Acute phase cytokines and outcomes in a prospective, matched cohort study. Am J Transplant 2021; 21:2113-2122. [PMID: 33131212 PMCID: PMC8085168 DOI: 10.1111/ajt.16388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/08/2020] [Accepted: 10/25/2020] [Indexed: 01/25/2023]
Abstract
We undertook a prospective, matched cohort study of patients with Staphylococcus aureus bacteremia (SAB) and gram-negative bacteremia (GNB) to compare the characteristics, outcomes, and chemokine and cytokine response in transplant recipients to immunocompetent, nontransplant recipients. Fifty-five transplant recipients (GNB n = 29; SAB n = 26) and 225 nontransplant recipients (GNB n = 114; SAB n = 111) were included for clinical analysis. Transplant GNB had a significantly lower incidence of septic shock than nontransplant GNB (10.3% vs 30.7%, p = .03). Thirty-day mortality did not differ significantly between transplant and nontransplant recipients with GNB (10.3% vs 15.8%, p = .57) or SAB (0.0% vs 11.7%, p = .13). Next, transplant patients were matched 1:1 with nontransplant patients for the chemokine and cytokine analysis. Five cytokines and chemokines were significantly lower in transplant GNB vs nontransplant GNB: IL-2 (median [IQR]: 7.1 pg/ml [7.1, 7.1] vs 32.6 pg/ml [7.1, 88.0]; p = .001), MIP-1β (30.7 pg/ml [30.7, 30.7] vs 243.3 pg/ml [30.7, 344.4]; p = .001), IL-8 (32.0 pg/ml [5.6, 53.1] vs 59.1 pg/ml [39.2, 119.4]; p = .003), IL-15 (12.0 pg/ml [12.0, 12.0] vs 12.0 pg/ml [12.0, 126.7]; p = .03), and IFN-α (5.1 pg/mL [5.1, 5.1] vs 5.1 pg/ml [5.1, 26.3]; p = .04). Regulated upon Activation, Normal T Cell Expressed and Secreted (RANTES) was higher in transplant SAB vs nontransplant SAB (mean [SD]: 750.2 pg/ml [194.6] vs 656.5 pg/ml [147.6]; p = .046).
Collapse
Affiliation(s)
- Emily M Eichenberger
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Felicia Ruffin
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Michael Dagher
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Reginald Lerebours
- Department of Biostatistics & Bioinformatics, Duke University, Durham, North Carolina, United States of America
| | - Sin-Ho Jung
- Department of Biostatistics & Bioinformatics, Duke University, Durham, North Carolina, United States of America
| | - Batu Sharma-Kuinkel
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Andrew N Macintyre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | - Joshua T Thaden
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Matthew Sinclair
- United States of America, Department of Nephrology, Duke University, Durham, North Carolina, United States of America,,Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Lauren Hale
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Celia Kohler
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Scott M Palmer
- Department of Transplant Pulmonology, Duke University, Durham, North Carolina, United States,,Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Barbara D Alexander
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Vance G Fowler
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America,,Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America,Corresponding author: Vance G Fowler Jr., MD, MHS, Duke University Medical Center, Division of Infectious Diseases, 315 Trent Drive Hanes House, Durham, NC 27710, , (P): 919 668-6053, (F): 919 684-8902
| | - Stacey A Maskarinec
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
29
|
Alosaimy S, Lagnf AM, Morrisette T, Jorgensen SCJ, Trinh TD, Zasowski EJ, Scipione MR, Zhao JJ, Mynatt R, Herbin S, Dhar S, Chopra T, Janisse J, Rebold N, Pogue JM, Rybak MJ. Standardized Treatment and Assessment Pathway Improves Mortality in Adults With Methicillin-resistant Staphylococcus aureus Bacteremia: STAPH Study. Open Forum Infect Dis 2021; 8:ofab261. [PMID: 34258313 PMCID: PMC8271135 DOI: 10.1093/ofid/ofab261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infection (BSI) management remains challenging for clinicians. Numerous in vitro studies report synergy when vancomycin (VAN) and daptomycin (DAP) are combined with beta-lactams (BLs), which has led to clinical implementation of these combinations. While shorter durations of bacteremia have often been reported, there has been no significant impact on mortality. Methods The Detroit Medical Center (DMC) developed and implemented a clinical pathway algorithm for MRSA BSI treatment in 2016 that included the early use of BL combination therapy with standard of care (VAN or DAP) and a mandatory Infectious Diseases consultation. This was a retrospective, quasi-experimental study at the DMC between 2013 and 2020. Multivariable logistic regression was used to assess the independent association between pathway implementation and 30-day mortality while adjusting for confounding variables. Results Overall, 813 adult patients treated for MRSA BSI were evaluated. Compared with prepathway (PRE) patients (n = 379), those treated postpathway (POST; n = 434) had a significant reduction in 30-day and 90-day mortality: 9.7% in POST vs 15.6% in PRE (P = .011) and 12.2% in POST vs 19.0% in PRE (P = .007), respectively. The incidence of acute kidney injury (AKI) was higher in the PRE compared with the POST group: 9.6% vs 7.2% (P = .282), respectively. After adjusting for confounding variables including Infectious Diseases consult, POST was independently associated with a reduction in 30-day mortality (adjusted odds ratio [aOR], 0.608; 95% CI, 0.375–0.986). Conclusions Implementation of an MRSA BSI treatment pathway with early use of BL reduced mortality with no increased rate of AKI. Further prospective evaluation of this pathway approach is warranted.
Collapse
Affiliation(s)
- Sara Alosaimy
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Abdalhamid M Lagnf
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Taylor Morrisette
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Sarah C J Jorgensen
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Trang D Trinh
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Evan J Zasowski
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Marco R Scipione
- Department of Pharmacy, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
| | - Jing J Zhao
- Harper University Hospital, Detroit, Michigan, USA
| | - Ryan Mynatt
- University of Kentucky, Lexington, Kentucky, USA
| | - Shelbye Herbin
- Department of Pharmacy, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
| | - Sorabh Dhar
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Wayne State University, Detroit, Michigan, USA.,Division of Infectious Diseases, John D. Dingell, Veterans Administration Medical Center, Detroit, Michigan, USA
| | - Teena Chopra
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Wayne State University, Detroit, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA
| | - James Janisse
- Department of Family Medicine and Public Health Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Nicholas Rebold
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Jason M Pogue
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Michael J Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA.,Department of Pharmacy, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA.,Department of Family Medicine and Public Health Sciences, School of Medicine, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
30
|
Rose W, Fantl M, Geriak M, Nizet V, Sakoulas G. Current Paradigms of Combination therapy in Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia: Does it Work, Which Combination and For Which Patients? Clin Infect Dis 2021; 73:2353-2360. [PMID: 33993226 DOI: 10.1093/cid/ciab452] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
The last several years have seen an emergence of literature documenting the utility of combination antimicrobial therapy, particularly in the salvage of refractory MRSA bacteremia. Recent clinical data are shaping conundrums of which regimens may be more beneficial, which can be potentially harmful, and which subset of patients stand to benefit from more aggressive treatment regimens than called for by current standards. In addition, the incorporation of combination therapy for MRSA bacteremia should be accompanied by the reminder that antimicrobial therapy does not need to be uniform for the entire duration, with an early intensive phase in high inoculum infections (e.g. with combination therapy), followed by a consolidation phase (i.e. monotherapy). This review and perspective consolidates the recent data on this subject and directs future goals in filling the knowledge gaps to methodically move forward towards improving patient outcomes.
Collapse
Affiliation(s)
- Warren Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Fantl
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew Geriak
- Pharmacy Department, Sharp Memorial Hospital, San Diego, CA, USA
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - George Sakoulas
- Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
31
|
Tan K, Minejima E, Lou M, Mack WJ, Nieberg P, Wong-Beringer A. Cytokine measurements add value to clinical variables in predicting outcomes for Staphylococcus aureus bacteremia. BMC Infect Dis 2021; 21:317. [PMID: 33820537 PMCID: PMC8022397 DOI: 10.1186/s12879-021-06010-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/22/2021] [Indexed: 11/12/2022] Open
Abstract
Background We demonstrated that an early dysregulated cytokine response [high interleukin-10 to tissue necrosis factor (IL-10/TNF) ratio] predicted poor outcomes in patients with Staphylococcus aureus bacteremia (SAB). However, high interpatient variability in cytokine levels were observed. We grouped cytokine measurements in quartiles and assessed their additive value to clinical variables for predicting bacterial persistence and 30-day mortality in patients with SAB. Methods A multicenter observational study was conducted in hospitalized patients with SAB. Medical charts were reviewed for relevant information. Blood samples were obtained for cytokine measurements by ELISA: interferon-gamma (IFNγ), interleukin (IL-1β, IL-6, IL-8, IL-10, IL-17) and tissue necrosis factor (TNF). Cytokine measurements were grouped into quartiles. Significant predictors for bacterial persistence and 30-day mortality were determined by multivariable logistic regression analysis. Area under the ROC curve (AUC) analysis was performed and predictive performance was compared between models with and without cytokine quartiles. Results Among 606 patients with SAB, a subset of patients (n = 239) had Day 1 cytokine measurements and clinical data collected; of those, 53 (22%) had persistent bacteremia. Accounting for septic shock, the addition of either IL-10 (AUC 0.708) or TNF (AUC 0.714) quartiles measured on Day 1 improved model performance for predicting bacterial persistence. All patients had Day 4 cytokine measurements; 52 patients (8.5%) died within 30-days of SAB onset. Inclusion of either IL-10 (AUC 0.873) or TNF (AUC 0.879) quartiles, but not both, measured on Day 4 to the significant clinical predictors (coronary artery disease, Pitt bacteremia score ≥ 4, and septic shock) improved model performance for mortality. Conclusions IL-10 or TNF levels falling within the range in the upper quartiles, when combined with clinical variables, improved model performance for predicting outcomes, and may potentially be used to support aggressive management and biomarker-guided studies to evaluate the benefit of adjunctive immunotherapy for SAB in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06010-0.
Collapse
Affiliation(s)
- Karen Tan
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Emi Minejima
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Mimi Lou
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Paul Nieberg
- Department of Medicine - Infectious Diseases, Huntington Hospital, Pasadena, USA
| | - Annie Wong-Beringer
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA. .,Department of Pharmacy, Huntington Hospital, Pasadena, USA.
| |
Collapse
|
32
|
Li Y, Gou H, Chu P, Zhang K, Jiang Z, Cai R, Song S, Bian Z, Li C. Comparison of Host Cytokine Response in Piglets Infected With Toxigenic and Non-toxigenic Staphylococcus hyicus. Front Vet Sci 2021; 8:639141. [PMID: 33665221 PMCID: PMC7920954 DOI: 10.3389/fvets.2021.639141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/26/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus hyicus is the most common causative agent of exudative epidermitis (EE) in piglets. Staphylococcus hyicus can be grouped into toxigenic and non-toxigenic strains based on its ability to cause EE in pigs. However, the inflammatory response of piglets infected with toxigenic and non-toxigenic S. hyicus has not been elucidated. In this study, we evaluated the serum cytokine profile in piglets inoculated with toxigenic and non-toxigenic S. hyicus strains and recorded the clinical signs in piglets. Fifteen piglets were divided into three groups (n = 5) and inoculated with a toxigenic strain (ZC-4), a non-toxigenic strain (CF-1), and PBS (control), respectively. The changes in serum levels of cytokines (interleukin [IL]-1β, IL-4, IL-6, IL-8, IL-10, IL-12, granulocyte-macrophage colony-stimulating factor, interferon-γ, transforming growth factor-β1, and tumor necrosis factor-α) were evaluated using a cytokine array at 6, 24, 48, and 72 h post inoculation. The results showed that piglets infected with the toxigenic strain exhibited more severe clinical signs and higher mortality than those infected with the non-toxigenic strain. The serum levels of pro-inflammatory cytokine IL-1β were significantly increased in toxigenic-and non-toxigenic-strain-infected piglets compared to those in the control group (p < 0.05), while the anti-inflammatory cytokine IL-10 was significantly up-regulated only in toxigenic group than in control group (p < 0.05). These results indicated that piglets infected with toxigenic and non-toxigenic S. hyicus showed differential infection status and inflammatory responses. Both toxigenic- and non-toxigenic- S. hyicus infection could induce a pro-inflammatory reaction in piglets. In addition, the toxigenic strain induced a strong anti-inflammatory response in piglets as indicated by the increased serum level of IL-10, which may be associated with the severe clinical signs and increased mortality and may be the key cytokine response responsible for pathogenic mechanisms of S. hyicus.
Collapse
Affiliation(s)
- Yan Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Hongchao Gou
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Pinpin Chu
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Kunli Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Zhiyong Jiang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Rujian Cai
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Shuai Song
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Zhibiao Bian
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Chunling Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China.,Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| |
Collapse
|
33
|
Algorri M, Jorth P, Wong-Beringer A. Variable Release of Lipoteichoic Acid From Staphylococcus aureus Bloodstream Isolates Relates to Distinct Clinical Phenotypes, Strain Background, and Antibiotic Exposure. Front Microbiol 2021; 11:609280. [PMID: 33519759 PMCID: PMC7840697 DOI: 10.3389/fmicb.2020.609280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022] Open
Abstract
Background Staphylococcus aureus is a leading cause of bacterial bloodstream infections. The heterogeneity in patient outcomes in S. aureus bacteremia (SAB) can be attributed in part to strain characteristics, which may influence host response to infection. We specifically examined the relationship between lipoteichoic acid (LTA) release from S. aureus and disease phenotype, strain background, and antibiotic exposure. Methods Seven strains of S. aureus causing different clinical phenotypes of bacteremia and two reference strains (LAC USA 300 and Mu3) were analyzed for LTA release at baseline and following exposure to antibiotics from different pharmacologic classes (vancomycin, ceftaroline, and tedizolid). LTA release was quantified by LTA-specific ELISA. Whole genome sequencing was performed on the clinical strains and analyzed using open-source bioinformatics tools. Results Lipoteichoic acid release varied by 4-fold amongst the clinical strains and appeared to be related to duration of bacteremia, independent of MLST type. Low LTA releasing strains were isolated from patients who had prolonged duration of bacteremia and died. Antibiotic-mediated differences in LTA release appeared to be associated with MLST type, as ST8 strains released maximal LTA in response to tedizolid while other non-ST8 strains demonstrated high LTA release with vancomycin. Genetic variations related to the LTA biosynthesis pathway were detected in all non-ST8 strains, though ST8 strains showed no variations despite demonstrating differential LTA release. Conclusion Our findings provide the basis for future studies to evaluate the relationship between LTA release-mediated host immune response and clinical outcomes as well as the potential for antibiotic modulation of LTA release as a therapeutic strategy and deserve confirmation with larger number of strains with known clinical phenotypes.
Collapse
Affiliation(s)
- Marquerita Algorri
- School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Peter Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Annie Wong-Beringer
- School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
34
|
Volk CF, Burgdorf S, Edwardson G, Nizet V, Sakoulas G, Rose WE. Interleukin (IL)-1β and IL-10 Host Responses in Patients With Staphylococcus aureus Bacteremia Determined by Antimicrobial Therapy. Clin Infect Dis 2021; 70:2634-2640. [PMID: 31365924 DOI: 10.1093/cid/ciz686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/18/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Patient interleukin (IL)-1β and IL-10 responses early in Staphylococcus aureus bacteremia (SaB) are associated with bacteremia duration and mortality. We hypothesized that these responses vary depending on antimicrobial therapy, with particular interest in whether the superiority of β-lactams links to key cytokine pathways. METHODS Three medical centers included 59 patients with SaB (47 methicillin-resistant S. aureus [MRSA], 12 methicillin-sensitive S. aureus [MSSA]) from 2015-2017. In the first 48 hours, patients were treated with either a β-lactam (n = 24), including oxacillin, cefazolin, or ceftaroline, or a glyco-/lipopeptide (n = 35), that is, vancomycin or daptomycin. Patient sera from days 1, 3, and 7 were assayed for IL-1β and IL-10 by enzyme-linked immunosorbent assay and compared using the Mann-Whitney U test. RESULTS On presentation, IL-10 was elevated in mortality (P = .008) and persistent bacteremia (P = .034), while no difference occurred in IL-1β. Regarding treatment groups, IL-1β and IL-10 were similar prior to receiving antibiotic. Patients treated with β-lactam had higher IL-1β on days 3 (median +5.6 pg/mL; P = .007) and 7 (+10.9 pg/mL; P = .016). Ex vivo, addition of the IL-1 receptor antagonist anakinra to whole blood reduced staphylococcal killing, supporting an IL-1β functional significance in SaB clearance. β-lactam-treated patients had sharper declines in IL-10 than vancomycin or daptomycin -treated patients over 7 days. CONCLUSIONS These data underscore the importance of β-lactams for SaB, including consideration that the adjunctive role of β-lactams for MRSA in select patients helps elicit favorable host cytokine responses.
Collapse
Affiliation(s)
- Cecilia F Volk
- School of Pharmacy, University of Wisconsin-Madison, La Jolla
| | - Sarah Burgdorf
- Department of Pediatrics, University of California-San Diego School of Medicine, La Jolla
| | | | - Victor Nizet
- Department of Pediatrics, University of California-San Diego School of Medicine, La Jolla
| | - George Sakoulas
- Department of Pediatrics, University of California-San Diego School of Medicine, La Jolla
| | - Warren E Rose
- School of Pharmacy, University of Wisconsin-Madison, La Jolla
| |
Collapse
|
35
|
Minejima E, Mai N, Bui N, Mert M, Mack WJ, She RC, Nieberg P, Spellberg B, Wong-Beringer A. Defining the Breakpoint Duration of Staphylococcus aureus Bacteremia Predictive of Poor Outcomes. Clin Infect Dis 2021; 70:566-573. [PMID: 30949675 DOI: 10.1093/cid/ciz257] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Persistent Staphylococcus aureus bacteremia (SAB) is defined based on varying duration in literature. The primary objective was to determine the risk of poor outcomes in relation to bacteremia duration. METHODS Multicenter, prospective, observational study of adult hospitalized patients with SAB. Medical records were reviewed for pertinent data. Patients were grouped by bacteremia duration: short (1-2 days), intermediate (3-6 days), and prolonged (≥7 days) and compared for risk factors and outcomes. RESULTS Of 884 patients, 63% had short, 28% intermediate, and 9% prolonged bacteremia. Overall mean age was 57 years, and 70% were male. The prolonged group had the highest proportion of methicillin-resistant SAB (P < .0001). Choice of antibiotic therapy did not significantly affect bacteremia duration; however, time to source-control procedure was delayed in the prolonged and intermediate groups compared with the short group (3.5 vs 3 vs 1 day, P < .0001). Metastatic complications, length of stay, and 30-day mortality were progressively worse as bacteremia duration increased (P < .0001). Every continued day of bacteremia was associated with a relative risk of death of 1.16 (95% confidence interval, 1.10-1.22; P < .0001), with a significant increase in risk starting at 3 days as determined by receiver operating characteristic analysis. CONCLUSIONS Optimal management of SAB should target bacterial clearance as soon as possible to minimize incremental risk of mortality with each day of positive blood culture. Delay in source control but not type of antistaphylococcal therapy was significantly associated with prolonged bacteremia and worse outcomes.
Collapse
Affiliation(s)
- Emi Minejima
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, Los Angeles.,Los Angeles County and USC Medical Center, Los Angeles
| | - Nikki Mai
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, Los Angeles
| | - Nancy Bui
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, Los Angeles
| | - Melissa Mert
- Departments of Preventive Medicine and Clinical and Translational Science Institute, Los Angeles
| | - Wendy J Mack
- Departments of Preventive Medicine of Keck School of Medicine, Los Angeles
| | - Rosemary C She
- Departments of Pathology, Keck School of Medicine at USC, Los Angeles
| | - Paul Nieberg
- Department of Medicine-Infectious Diseases, Huntington Hospital, Pasadena, California
| | - Brad Spellberg
- Los Angeles County and USC Medical Center, Los Angeles.,Department of Medicine, Keck School of Medicine at USC, Los Angeles
| | - Annie Wong-Beringer
- Department of Clinical Pharmacy, University of Southern California (USC) School of Pharmacy, Los Angeles.,Department of Pharmacy, Huntington Hospital, Pasadena, California
| |
Collapse
|
36
|
Kim NH, Sung JY, Choi YJ, Choi SJ, Ahn S, Ji E, Kim M, Kim CJ, Song KH, Choe PG, Park WB, Kim ES, Park KU, Kim NJ, Oh MD, Kim HB. Toll-like receptor 2 downregulation and cytokine dysregulation predict mortality in patients with Staphylococcus aureus bacteremia. BMC Infect Dis 2020; 20:901. [PMID: 33256638 PMCID: PMC7706030 DOI: 10.1186/s12879-020-05641-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Staphylococcus aureus bacteremia (SAB) presents heterogeneously, owing to the differences in underlying host conditions and immune responses. Although Toll-like receptor 2 (TLR2) is important in recognizing S. aureus, its function during S. aureus infection remains controversial. We aimed to examine the association of TLR2 expression and associated cytokine responses with clinical SAB outcomes. METHODS Patients from a prospective SAB cohort at two tertiary-care medical centers were enrolled. Blood was sampled at several timepoints (≤5 d, 6-9 d, 10-13 d, 14-19 d, and ≥ 20 d) after SAB onset. TLR2 mRNA levels were determined via real-time PCR and serum tumor necrosis factor [TNF]-α, interleukin [IL]-6, and IL-10 levels were analyzed with multiplex-high-sensitivity electrochemiluminescent ELISA. RESULTS TLR2 levels varied among 59 SAB patients. On days 2-5, TLR2 levels were significantly higher in SAB survivors than in healthy controls (p = 0.040) and slightly but not significantly higher than non-survivors (p = 0.120), and SAB patients dying within 7 d had lower TLR2 levels than survivors (P = 0.077) although statistically insignificant. IL-6 and IL-10 levels were significantly higher in non-survivors than in survivors on days 2-5 post-bacteremia (P = 0.010 and P = 0.021, respectively), and those dying within 7 d of SAB (n = 3) displayed significantly higher IL-10/TNF-α ratios than the survivors did (P = 0.007). CONCLUSION TLR2 downregulation and IL-6 and IL-10 concentrations suggestive of immune dysregulation during early bacteremia may be associated with mortality from SAB. TLR2 expression levels and associated cytokine reactions during early-phase SAB may be potential prognostic factors in SAB, although larger studies are warranted.
Collapse
Affiliation(s)
- Nak-Hyun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea
| | - Ji Yeon Sung
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea.,Present Address: Roche Korea, Seoul, Republic of Korea
| | - Yoon Jung Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea
| | - Su-Jin Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soyeon Ahn
- Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Eunjeong Ji
- Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Moonsuk Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea
| | - Chung Jong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea.,Present Address: Department of Internal Medicine, Ewha Womans University, Seoul Hospital, Seoul, Republic of Korea
| | - Kyoung-Ho Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea
| | - Pyoeng Gyun Choe
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wan Beom Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eu Suk Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea
| | - Kyoung Un Park
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Nam-Joong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myoung-Don Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hong Bin Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam, 463-707, Republic of Korea.
| |
Collapse
|
37
|
Algorri M, Wong-Beringer A. Differential effects of antibiotics on neutrophils exposed to lipoteichoic acid derived from Staphylococcus aureus. Ann Clin Microbiol Antimicrob 2020; 19:50. [PMID: 33143710 PMCID: PMC7641855 DOI: 10.1186/s12941-020-00392-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Persistent bacteremia occurs in at least 30% of patients with Staphylococcus aureus bloodstream infection (SAB) and may be attributable to a dysregulated host immune response. Neutrophils interact with a variety of S. aureus microbial factors, including lipoteichoic acid (LTA), to activate phagocytic function in a concentration-dependent manner. Antibiotics have been shown to exert both direct antimicrobial action as well as immunomodulatory effects. In this study, we compared the effects of different anti-staphylococcal antibiotics on LTA-mediated immune activation of neutrophils. METHODS Neutrophils obtained from healthy volunteers were exposed to two levels of LTA (1 and 10 μg/ml) with or without addition of antibiotics from different pharmacologic classes (vancomycin, daptomycin, ceftaroline). Neutrophil function was assessed by examining phagocytic response, activation (CD11b, CD62L expression), Toll-like receptor-2 expression, cell survival and apoptosis, and CXCL8 release. RESULTS Differential LTA-mediated antibiotic effects on neutrophil function were observed primarily at the high LTA exposure level. Ceftaroline in the presence of 10 μg/ml LTA had the most prominent effects on phagocytosis and CD11b and CD62L expression, with trends towards increased neutrophil survival and preservation of CXCL8 release when compared to daptomycin and vancomycin with the latter significantly dampening PMN CXCL8 release. CONCLUSIONS Select antimicrobial agents, such as ceftaroline, exert immunostimulatory effects on neutrophils exposed to S. aureus LTA, which when confirmed in vivo, could be leveraged for its dual immunomodulatory and antibacterial actions for the treatment of persistent SAB mediated by a dysregulated host response.
Collapse
Affiliation(s)
- Marquerita Algorri
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Annie Wong-Beringer
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA.
| |
Collapse
|
38
|
Miller LS, Fowler VG, Shukla SK, Rose WE, Proctor RA. Development of a vaccine against Staphylococcus aureus invasive infections: Evidence based on human immunity, genetics and bacterial evasion mechanisms. FEMS Microbiol Rev 2020; 44:123-153. [PMID: 31841134 PMCID: PMC7053580 DOI: 10.1093/femsre/fuz030] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Invasive Staphylococcus aureus infections are a leading cause of morbidity and mortality in both hospital and community settings, especially with the widespread emergence of virulent and multi-drug resistant methicillin-resistant S. aureus strains. There is an urgent and unmet clinical need for non-antibiotic immune-based approaches to treat these infections as the increasing antibiotic resistance is creating a serious threat to public health. However, all vaccination attempts aimed at preventing S. aureus invasive infections have failed in human trials, especially all vaccines aimed at generating high titers of opsonic antibodies against S. aureus surface antigens to facilitate antibody-mediated bacterial clearance. In this review, we summarize the data from humans regarding the immune responses that protect against invasive S. aureus infections as well as host genetic factors and bacterial evasion mechanisms, which are important to consider for the future development of effective and successful vaccines and immunotherapies against invasive S. aureus infections in humans. The evidence presented form the basis for a hypothesis that staphylococcal toxins (including superantigens and pore-forming toxins) are important virulence factors, and targeting the neutralization of these toxins are more likely to provide a therapeutic benefit in contrast to prior vaccine attempts to generate antibodies to facilitate opsonophagocytosis.
Collapse
Affiliation(s)
- Lloyd S Miller
- Immunology, Janssen Research and Development, 1400 McKean Road, Spring House, PA, 19477, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Cancer Research Building 2, Suite 209, Baltimore, MD, 21231, USA.,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, 1830 East Monument Street, Baltimore, MD, 21287, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, 601 North Caroline Street, Baltimore, MD, 21287, USA.,Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA
| | - Vance G Fowler
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, 315 Trent Drive, Hanes House, Durham, NC, 27710, USA.,Duke Clinical Research Institute, Duke University Medical Center, 40 Duke Medicine Circle, Durham, NC, 27710, USA
| | - Sanjay K Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, 1000 North Oak Avenue, Marshfield, WI, 54449, USA.,Computation and Informatics in Biology and Medicine, University of Wisconsin, 425 Henry Mall, Room 3445, Madison, WI, 53706, USA
| | - Warren E Rose
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Pharmacy Practice Division, University of Wisconsin-Madison, 777 Highland Avenue, 4123 Rennebohm Hall, Madison, WI, 53705 USA
| | - Richard A Proctor
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA.,Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, 1550 Linden Drive, Microbial Sciences Building, Room 1334, Madison, WI, 53705, USA
| |
Collapse
|
39
|
Hendrickson JA, Spitznogle SL, Gonzales-Luna AJ, Babic JT, Britt RS, Knight JM, Pham SN, McDaneld PM. Significant Publications on Infectious Diseases Pharmacotherapy in 2019. J Pharm Pract 2020; 34:800-813. [PMID: 32935641 DOI: 10.1177/0897190020951348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To provide a summary of the most prominent peer-reviewed infectious diseases (ID) pharmacotherapy and Human Immunodeficiency Virus (HIV)-related articles published in 2019. SUMMARY Houston Infectious Diseases Network (HIDN) members were asked to nominate articles that they believed were most influential within the ID and HIV pharmacotherapy science communities. A total of 48 general ID and 6 HIV-related articles were nominated. Following nominations, an online survey was distributed via e-mail to Society of Infectious Diseases Pharmacists (SIDP) members, with a total of 156 and 54 members voting for general ID and HIV-related articles, respectively. The results of this survey were ranked to determine the top 10 general ID and top HIV articles. The top articles were then summarized by HIDN members, including residents, fellows, and clinical pharmacists. CONCLUSION This review covers many of the most influential ID articles published in 2019, including 3 practice guideline updates. Due to the high rate of ID literature published each year, this review continues to help summarize these articles for the ID community, allowing clinicians to remain up-to-date on practice-changing publications in ID and HIV pharmacotherapy.
Collapse
Affiliation(s)
- Joshua A Hendrickson
- Department of Pharmacy Practice and Translational Research, 15507University of Houston College of Pharmacy, Houston, TX, USA
| | - Sarah L Spitznogle
- Division of Pharmacy, 4002The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne J Gonzales-Luna
- Department of Pharmacy Practice and Translational Research, 15507University of Houston College of Pharmacy, Houston, TX, USA
| | - Jessica T Babic
- Department of Pharmacy, 23464Memorial Hermann-Texas Medical Center, Houston, TX, USA
| | - Rachel S Britt
- Department of Pharmacy, 12338The University of Texas Medical Branch, Galveston, TX, USA
| | - Joshua M Knight
- Department of Pharmacy, 23534Houston Methodist Hospital, Houston, TX, USA
| | - Selena N Pham
- Department of Pharmacy, 20116Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Patrick M McDaneld
- Division of Pharmacy, 4002The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Wozniak JM, Mills RH, Olson J, Caldera JR, Sepich-Poore GD, Carrillo-Terrazas M, Tsai CM, Vargas F, Knight R, Dorrestein PC, Liu GY, Nizet V, Sakoulas G, Rose W, Gonzalez DJ. Mortality Risk Profiling of Staphylococcus aureus Bacteremia by Multi-omic Serum Analysis Reveals Early Predictive and Pathogenic Signatures. Cell 2020; 182:1311-1327.e14. [PMID: 32888495 PMCID: PMC7494005 DOI: 10.1016/j.cell.2020.07.040] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/11/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus bacteremia (SaB) causes significant disease in humans, carrying mortality rates of ∼25%. The ability to rapidly predict SaB patient responses and guide personalized treatment regimens could reduce mortality. Here, we present a resource of SaB prognostic biomarkers. Integrating proteomic and metabolomic techniques enabled the identification of >10,000 features from >200 serum samples collected upon clinical presentation. We interrogated the complexity of serum using multiple computational strategies, which provided a comprehensive view of the early host response to infection. Our biomarkers exceed the predictive capabilities of those previously reported, particularly when used in combination. Last, we validated the biological contribution of mortality-associated pathways using a murine model of SaB. Our findings represent a starting point for the development of a prognostic test for identifying high-risk patients at a time early enough to trigger intensive monitoring and interventions.
Collapse
Affiliation(s)
- Jacob M Wozniak
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert H Mills
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - J R Caldera
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gregory D Sepich-Poore
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marvic Carrillo-Terrazas
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chih-Ming Tsai
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fernando Vargas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rob Knight
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Y Liu
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Sakoulas
- Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Warren Rose
- School of Pharmacy, School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medicine, School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Collaborative to Halt Antibiotic-Resistant Microbes, University of California, San Diego, La Jolla, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
41
|
Kuehl R, Morata L, Boeing C, Subirana I, Seifert H, Rieg S, Kern WV, Kim HB, Kim ES, Liao CH, Tilley R, Lopez-Cortés LE, Llewelyn MJ, Fowler VG, Thwaites G, Cisneros JM, Scarborough M, Nsutebu E, Gurgui Ferrer M, Pérez JL, Barlow G, Hopkins S, Ternavasio-de la Vega HG, Török ME, Wilson P, Kaasch AJ, Soriano A. Defining persistent Staphylococcus aureus bacteraemia: secondary analysis of a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2020; 20:1409-1417. [PMID: 32763194 DOI: 10.1016/s1473-3099(20)30447-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Staphylococcus aureus persistent bacteraemia is only vaguely defined and the effect of different durations of bacteraemia on mortality is not well established. Our primary aim was to analyse mortality according to duration of bacteraemia and to derive a clinically relevant definition for persistent bacteraemia. METHODS We did a secondary analysis of a prospective observational cohort study at 17 European centres (nine in the UK, six in Spain, and two in Germany), with recruitment between Jan 1, 2013, and April 30, 2015. Adult patients who were consecutively hospitalised with monomicrobial S aureus bacteraemia were included. Patients were excluded if no follow-up blood culture was taken, if the first follow-up blood-culture was after 7 days, or if active antibiotic therapy was started more than 3 days after first blood culture. The primary outcome was 90-day mortality. Univariable and time-dependent multivariable Cox regression analysis were used to assess predictors of mortality. Duration of bacteraemia was defined as bacteraemic days under active antibiotic therapy counting the first day as day 1. FINDINGS Of 1588 individuals assessed for eligibility, 987 were included (median age 65 years [IQR 51-75]; 625 [63%] male). Death within 90 days occurred in 273 (28%) patients. Patients with more than 1 day of bacteraemia (315 [32%]) had higher Charlson comorbidity index and sequential organ failure assessment scores and a longer interval from first symptom to first blood culture. Crude 90-day mortality increased from 22% (148 of 672) with 1 day of bacteraemia, to 39% (85 of 218) with 2-4 days, 43% (30 of 69) with 5-7 days, and 36% (10 of 28) with more than 7 days of bacteraemia. Metastatic infections developed in 39 (6%) of 672 patients with 1 day of bacteraemia versus 40 (13%) of 315 patients if bacteraemia lasted for at least 2 days. The second day of bacteraemia had the highest HR and earliest cutoff significantly associated with mortality (adjusted hazard ratio 1·93, 95% CI 1·51-2·46; p<0·0001). INTERPRETATION We suggest redefining the cutoff duration for persistent bacteraemia as 2 days or more despite active antibiotic therapy. Our results favour follow-up blood cultures after 24 h for early identification of all patients with increased risk of death and metastatic infection. FUNDING None.
Collapse
Affiliation(s)
- Richard Kuehl
- Service of Infectious Diseases, Hospital Clínic of Barcelona, Barcelona, Spain; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Laura Morata
- Service of Infectious Diseases, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Christian Boeing
- Institute of Medical Microbiology and Hospital Hygiene, Faculty of Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Isaac Subirana
- CIBER en Epidemiología y Salud Pública, Barcelona, Spain
| | - Harald Seifert
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University of Cologne, Cologne, Germany; German Center for Infection Research, Partner site Bonn-Cologne, Cologne, Germany
| | - Siegbert Rieg
- Division of Infectious Diseases, Department of Medicine II, Medical Center-University of Freiburg, Freiburg, Germany
| | - Winfried V Kern
- Division of Infectious Diseases, Department of Medicine II, Medical Center-University of Freiburg, Freiburg, Germany
| | - Hong Bin Kim
- Division of Infectious Diseases, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Eu Suk Kim
- Division of Infectious Diseases, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Chun-Hsing Liao
- Infectious Diseases, Department of Internal Medicine, Far Eastern Memorial Hospital, Taipei City, Taiwan
| | - Robert Tilley
- Department of Microbiology, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Luis Eduardo Lopez-Cortés
- Infectious Diseases and Clinical Microbiology Unit, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Martin J Llewelyn
- Department of Infectious Diseases and Microbiology, Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Vance G Fowler
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Guy Thwaites
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - José Miguel Cisneros
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Instituto de Biomedicina de Sevilla, Seville, Spain
| | - Matt Scarborough
- Nuffield Department of Medicine, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Emmanuel Nsutebu
- Tropical and Infectious Disease Unit, Royal Liverpool University Hospital, Liverpool, UK
| | | | - José L Pérez
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
| | - Gavin Barlow
- Department of Infection, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - Susan Hopkins
- Infectious Diseases Unit, Royal Free London NHS Foundation Trust, London, UK
| | | | - M Estée Török
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Peter Wilson
- Department of Microbiology and Virology, University College London Hospital NHS Foundation Trust, London, UK
| | - Achim J Kaasch
- Institute of Medical Microbiology and Hospital Hygiene, Faculty of Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Alex Soriano
- Service of Infectious Diseases, Hospital Clínic of Barcelona, Barcelona, Spain.
| | | |
Collapse
|
42
|
Berti A, Rose W, Nizet V, Sakoulas G. Antibiotics and Innate Immunity: A Cooperative Effort Toward the Successful Treatment of Infections. Open Forum Infect Dis 2020; 7:ofaa302. [PMID: 32818143 PMCID: PMC7423293 DOI: 10.1093/ofid/ofaa302] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the common ancestry of antimicrobial and immunological science, a divergence driven by artificially construed paradigms in microbiology has placed limits on how we understand the mechanisms of antibiotics in vivo. We summarize recent updates on data that shed light on how antibiotics interact with components of innate immunity.
Collapse
Affiliation(s)
- Andrew Berti
- Department of Pharmacy Practice, Wayne State University College of Pharmacy and Health Sciences, Detroit, Michigan, USA.,Department of Biochemistry, Microbiology and Immunology, Wayne State University College of Medicine, Detroit, Michigan, USA
| | - Warren Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Victor Nizet
- Collaborative to Halt Antimicrobial Resistant Microbes, University of California San Diego School of Medicine, La Jolla, California, USA.,Skaggs School of Pharmacy, University of California San Diego, La Jolla, California, USA
| | - George Sakoulas
- Collaborative to Halt Antimicrobial Resistant Microbes, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
43
|
Eichenberger EM, Dagher M, Ruffin F, Park L, Hersh L, Sivapalasingam S, Fowler VG, Prasad BC. Complement levels in patients with bloodstream infection due to Staphylococcus aureus or Gram-negative bacteria. Eur J Clin Microbiol Infect Dis 2020; 39:2121-2131. [PMID: 32621149 PMCID: PMC7334117 DOI: 10.1007/s10096-020-03955-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
The complement system is a vital component of the innate immune system, though its role in bacteremia is poorly understood. We present complement levels in Staphylococcus aureus bacteremia (SAB) and Gram-negative bacteremia (GNB) and describe observed associations of complement levels with clinical outcomes. Complement and cytokine levels were measured in serum samples from 20 hospitalized patients with SAB, 20 hospitalized patients with GNB, 10 non-infected hospitalized patients, and 10 community controls. C5a levels were significantly higher in patients with SAB as compared to patients with GNB. Low C4 and C3 levels were associated with septic shock and 30-day mortality in patients with GNB, and elevated C3 was associated with a desirable outcome defined as absence of (1) septic shock, (2) acute renal failure, and (3) death within 30 days of bacteremia. Low levels of C9 were associated with septic shock in patients with GNB but not SAB. Elevated IL-10 was associated with increased 30-day mortality in patients with SAB. Complement profiles differ in patients with SAB and those with GNB. Measurement of IL-10 in patients with SAB and of C4, C3, and C9 in patients with GNB may help to identify those at higher risk for poor outcomes.
Collapse
Affiliation(s)
- Emily M Eichenberger
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, 27710, USA.
| | - Michael Dagher
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Lawrence Park
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, 27710, USA
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Lisa Hersh
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA
| | | | - Vance G Fowler
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, 2301 Erwin Road, Durham, NC, 27710, USA
| | | |
Collapse
|
44
|
Guimaraes AO, Cao Y, Hong K, Mayba O, Peck MC, Gutierrez J, Ruffin F, Carrasco-Triguero M, Dinoso JB, Clemenzi-Allen A, Koss CA, Maskarinec SA, Chambers HF, Fowler VG, Baruch A, Rosenberger CM. A Prognostic Model of Persistent Bacteremia and Mortality in Complicated Staphylococcus aureus Bloodstream Infection. Clin Infect Dis 2020; 68:1502-1511. [PMID: 30165412 DOI: 10.1093/cid/ciy739] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of bacteremia, yet there remains a significant knowledge gap in the identification of relevant biomarkers that predict clinical outcomes. Heterogeneity in the host response to invasive S. aureus infection suggests that specific biomarker signatures could be utilized to differentiate patients prone to severe disease, thereby facilitating earlier implementation of more aggressive therapies. METHODS To further elucidate the inflammatory correlates of poor clinical outcomes in patients with S. aureus bacteremia, we evaluated the association between a panel of blood proteins at initial presentation of bacteremia and disease severity outcomes using 2 cohorts of patients with S. aureus bacteremia (n = 32 and n = 124). RESULTS We identified 13 candidate proteins that were correlated with mortality and persistent bacteremia. Prognostic modeling identified interleukin (IL)-8 and CCL2 as the strongest individual predictors of mortality, with the combination of these biomarkers classifying fatal outcome with 89% sensitivity and 77% specificity (P < .0001). Baseline IL-17A levels were elevated in patients with persistent bacteremia (P < .0001), endovascular (P = .026) and metastatic tissue infections (P = .012). CONCLUSIONS These results demonstrate the potential utility of selected biomarkers to distinguish patients with the highest risk for treatment failure and bacteremia-related complications, providing a valuable tool for clinicians in the management of S. aureus bacteremia. Additionally, these biomarkers could identify patients with the greatest potential to benefit from novel therapies in clinical trials.
Collapse
Affiliation(s)
| | - Yi Cao
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Kyu Hong
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Oleg Mayba
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Melicent C Peck
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Johnny Gutierrez
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | | | - Jason B Dinoso
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Angelo Clemenzi-Allen
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Catherine A Koss
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | | | - Henry F Chambers
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | - Amos Baruch
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | | |
Collapse
|
45
|
Association of variants in selected genes mediating host immune response with duration of Staphylococcus aureus bacteremia. Genes Immun 2020; 21:240-248. [PMID: 32507857 DOI: 10.1038/s41435-020-0101-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 12/26/2022]
Abstract
Host genetic variation may be a contributing factor to variability in Staphylococcus aureus bacteremia duration. We assessed whether 28 single nucleotide polymorphisms (SNPs) in seven genes (TLR2, TLR4, TIRAP, IRAK4, TRAF6, NOD2, and CISH) that mediate host immune response were associated with S. aureus bacteremia duration. Subjects included 158 patients with short-term (≤4 days) and 44 with persistent (>4 days) S. aureus bacteremia from an academic medical center. In single SNP analyses, the minor allele frequencies of three TIRAP SNPs (rs655540, rs563011, and rs8177376) were higher in persistent bacteremia (P < 0.05). A haplotype with all three minor alleles was also associated with persistent bacteremia (P = 0.037). The minor allele frequencies of four other TIRAP SNPs (rs8177342, rs4937114, rs3802813, and rs4937115) were higher in short-term bacteremia (P < 0.05), and a haplotype containing the four minor alleles was associated with short-term bacteremia (P = 0.045). All seven SNPs are located in binding sites for proteins or noncoding RNAs that regulate transcription. None of the associations remained statistically significant after adjustment for multiple comparisons. Further investigation is needed to understand how genetic variation in TIRAP and other host immune genes may influence the duration of S. aureus bacteremia.
Collapse
|
46
|
Genetic Determinants Enabling Medium-Dependent Adaptation to Nafcillin in Methicillin-Resistant Staphylococcus aureus. mSystems 2020; 5:5/2/e00828-19. [PMID: 32234776 PMCID: PMC7112963 DOI: 10.1128/msystems.00828-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial susceptibility testing standards driving clinical decision-making have centered around the use of cation-adjusted Mueller-Hinton broth (CA-MHB) as the medium with the notion of supporting bacterial growth, without consideration of recapitulating the in vivo environment. However, it is increasingly recognized that various medium conditions have tremendous influence on antimicrobial activity, which in turn may have major implications on the ability of in vitro susceptibility assays to predict antibiotic activity in vivo. To elucidate differential growth optimization and antibiotic resistance mechanisms, adaptive laboratory evolution was performed in the presence or absence of the antibiotic nafcillin with methicillin-resistant Staphylococcus aureus (MRSA) TCH1516 in either (i) CA-MHB, a traditional bacteriological nutritionally rich medium, or (ii) Roswell Park Memorial Institute (RPMI), a medium more reflective of the in vivo host environment. Medium adaptation analysis showed an increase in growth rate in RPMI, but not CA-MHB, with mutations in apt, adenine phosphoribosyltransferase, and the manganese transporter subunit, mntA, occurring reproducibly in parallel replicate evolutions. The medium-adapted strains showed no virulence attenuation. Continuous exposure of medium-adapted strains to increasing concentrations of nafcillin led to medium-specific evolutionary strategies. Key reproducibly occurring mutations were specific for nafcillin adaptation in each medium type and did not confer resistance in the other medium environment. Only the vraRST operon, a regulator of membrane- and cell wall-related genes, showed mutations in both CA-MHB- and RPMI-evolved strains. Collectively, these results demonstrate the medium-specific genetic adaptive responses of MRSA and establish adaptive laboratory evolution as a platform to study clinically relevant resistance mechanisms.IMPORTANCE The ability of pathogens such as Staphylococcus aureus to evolve resistance to antibiotics used in the treatment of infections has been an important concern in the last decades. Resistant acquisition usually translates into treatment failure and puts patients at risk of unfavorable outcomes. Furthermore, the laboratory testing of antibiotic resistance does not account for the different environment the bacteria experiences within the human body, leading to results that do not translate into the clinic. In this study, we forced methicillin-resistant S. aureus to develop nafcillin resistance in two different environments, a laboratory environment and a physiologically more relevant environment. This allowed us to identify genetic changes that led to nafcillin resistance under both conditions. We concluded that not only does the environment dictate the evolutionary strategy of S. aureus to nafcillin but also that the evolutionary strategy is specific to that given environment.
Collapse
|
47
|
Cao Y, Guimaraes AO, Peck MC, Mayba O, Ruffin F, Hong K, Carrasco-Triguero M, Fowler VG, Maskarinec SA, Rosenberger CM. Risk stratification biomarkers for Staphylococcus aureus bacteraemia. Clin Transl Immunology 2020; 9:e1110. [PMID: 32082571 PMCID: PMC7018520 DOI: 10.1002/cti2.1110] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/18/2019] [Accepted: 01/23/2020] [Indexed: 11/28/2022] Open
Abstract
Objectives To identify risk stratification biomarkers to enrich for the subset of Staphylococcus aureus bacteraemia patients who develop deep‐seated tissue infections with high morbidity and mortality to guide clinical trial enrolment and clinical management. Methods We evaluated the prognostic value of eight biomarkers for persistent bacteraemia, mortality and endovascular infection foci in a validation cohort of 160 patients with S. aureus bacteraemia enrolled consecutively over 3 years. Results High levels of IL‐17A, IL‐10 or soluble E‐selectin at bacteraemia diagnosis correlated with the duration of positive blood cultures. When thresholds defined in an independent cohort were applied, these biomarkers were robust predictors of persistent bacteraemia or endovascular infection. High serum levels of IL‐17A and IL‐10 often preceded the radiographic diagnosis of infective endocarditis, suggesting potential utility for prioritising diagnostic radiographic imaging. High IL‐8 was prognostic for all‐cause mortality, while IL‐17A and IL‐10 were superior to clinical metrics in discriminating between attributable mortality and non‐attributable mortality. High IL‐17A and IL‐10 identified more patients who developed microbiological failure or mortality than were identified by infective endocarditis diagnosis. Conclusion These biomarkers offer potential utility to identify patients at risk of persistent bacteraemia to guide diagnostic imaging and clinical management. Low biomarker levels could be used to rule out the need for more invasive TEE imaging in patients at lower risk of infective endocarditis. These biomarkers could enable clinical trials by enriching for patients with the greatest need for novel therapies.
Collapse
Affiliation(s)
- Yi Cao
- Bioinformatics and Computational Biology Genentech, Inc. South San Francisco CA USA
| | | | - Melicent C Peck
- Clinical Sciences Genentech, Inc. South San Francisco CA USA
| | - Oleg Mayba
- Bioinformatics and Computational Biology Genentech, Inc. South San Francisco CA USA
| | - Felicia Ruffin
- Division of Infectious Diseases Duke University Durham NC USA
| | - Kyu Hong
- BioAnalytical Sciences Genentech, Inc. South San Francisco CA USA.,BioAnalysis, Immune-Onc Therapeutics Palo Alto CA USA
| | | | - Vance G Fowler
- Division of Infectious Diseases Duke University Durham NC USA
| | | | | |
Collapse
|
48
|
Sakoulas G, Geriak M, Nizet V. Is a Reported Penicillin Allergy Sufficient Grounds to Forgo the Multidimensional Antimicrobial Benefits of β-Lactam Antibiotics? Clin Infect Dis 2020; 68:157-164. [PMID: 29986019 DOI: 10.1093/cid/ciy557] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/07/2018] [Indexed: 12/18/2022] Open
Abstract
The majority of patients with reported penicillin allergy are not allergic when tested or challenged. Penicillin allergy testing has been shown to significantly reduce annual healthcare expenditures. Data have emerged showing β-lactams have multidimensional antibacterial effects in vivo, far beyond what is appreciated in standard bacteriological susceptibility testing media. These include enhancing bacterial killing by the innate immune system. Supporting the clinical relevance of these secondary underappreciated effects are recent clinical and pharmacoeconomic analyses that show worse outcomes in patients with reported penicillin allergies who receive non-β-lactam antibiotics when compared to their non-penicillin-allergic counterparts. This is particularly relevant in the treatment of Staphylococcus aureus bacteremia. This article reviews the tremendous advantages offered by β-lactam therapy and makes a strong case that the debunking of false penicillin allergies through a detailed allergy history and penicillin allergy testing should be a vital component of antimicrobial stewardship practices.
Collapse
Affiliation(s)
- George Sakoulas
- Sharp Memorial Hospital, San Diego, La Jolla.,School of Medicine, University of California, San Diego, La Jolla
| | | | - Victor Nizet
- School of Medicine, University of California, San Diego, La Jolla.,Skaggs School of Pharmacy, University of California, San Diego, La Jolla
| |
Collapse
|
49
|
McCreary EK, Kullar R, Geriak M, Zasowski EJ, Rizvi K, Schulz LT, Ouellette K, Vasina L, Haddad F, Rybak MJ, Zervos MJ, Sakoulas G, Rose WE. Multicenter Cohort of Patients With Methicillin-Resistant Staphylococcus aureus Bacteremia Receiving Daptomycin Plus Ceftaroline Compared With Other MRSA Treatments. Open Forum Infect Dis 2019; 7:ofz538. [PMID: 31938716 PMCID: PMC6951465 DOI: 10.1093/ofid/ofz538] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/17/2019] [Indexed: 01/11/2023] Open
Abstract
Background Daptomycin and ceftaroline (DAP-CPT) have been used for persistent methicillin-resistant Staphylococcus aureus bacteremia (MRSAB), but have rarely been compared with other therapies. This study provides an exploratory analysis of patients placed on DAP-CPT vs standard of care (SOC) for MRSAB. Methods This is a retrospective, matched cohort study MRSAB patients at 4 hospitals in the United States. Patients receiving DAP-CPT for ≥72 hours at any point in therapy were matched 2:1 when possible, 1:1 otherwise, to SOC, first by infection source, then age and renal function. SOC was empiric treatment with vancomycin or daptomycin and any subsequent combination antibiotic(s), except for DAP-CPT. Results Fifty-eight patients received DAP-CPT with 113 matched SOC. Ninety-six percent of SOC received vancomycin, and 56% (63/113) escalated therapy at least once in the treatment course. Twenty-four patients received DAP-CPT within 72 hours of index culture; 2 (8.3%) died within 30 days vs 14.2% (16/113) with SOC (P > .05). Subgroup analysis identified numerically lower mortality in DAP-CPT patients with a Charlson comorbidity index ≥3, endovascular source, and receipt of DAP-CPT within 72 hours of index culture. The median MRSAB duration was 9.3 vs 4.8 days for DAP-CPT and SOC, respectively. DAP-CPT was initiated on day 6 on average; after receipt of DAP-CPT, MRSAB duration was 3.3 days. Conclusions DAP-CPT treatment is often delayed in MRSAB. Combination therapy may be more beneficial if initiated earlier, particularly in patients at higher risk for mortality. Blinded, randomized, prospective studies are needed to eliminate selection bias inherent in retrospective analyses when examining DAP-CPT vs SOC.
Collapse
Affiliation(s)
- Erin K McCreary
- Department of Pharmacy, University of Wisconsin Health, Madison, Wisconsin, USA
| | | | - Matthew Geriak
- Pharmacy Department, Sharp Memorial Hospital, San Diego, California, USA
| | - Evan J Zasowski
- Infectious Disease, Sharp Healthcare, San Diego, California, USA
| | - Khulood Rizvi
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lucas T Schulz
- Department of Pharmacy, University of Wisconsin Health, Madison, Wisconsin, USA
| | - Krista Ouellette
- Pharmacy Department, Sharp Memorial Hospital, San Diego, California, USA
| | - Logan Vasina
- Pharmacy Department, Sharp Memorial Hospital, San Diego, California, USA
| | - Fadi Haddad
- Infectious Disease, Sharp Healthcare, San Diego, California, USA
| | - Michael J Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA.,Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Marcus J Zervos
- Wayne State University School of Medicine, Detroit, Michigan, USA.,Division of Infectious Diseases, Henry Ford Health System, Detroit, Michigan, USA
| | - George Sakoulas
- Infectious Disease, Sharp Healthcare, San Diego, California, USA.,Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, California, USA
| | - Warren E Rose
- Department of Pharmacy, University of Wisconsin Health, Madison, Wisconsin, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
50
|
Ferraro A, Buonocore SM, Auquier P, Nicolas I, Wallemacq H, Boutriau D, van der Most RG. Role and plasticity of Th1 and Th17 responses in immunity to Staphylococcus aureus. Hum Vaccin Immunother 2019; 15:2980-2992. [PMID: 31149870 PMCID: PMC6930085 DOI: 10.1080/21645515.2019.1613126] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The human commensal Staphylococcus aureus (SA) is a leading cause of skin/soft tissue and surgical-site infections, and bacteremia. Functional antibodies and T-cell-mediated immunity, particularly Th1/Th17 responses, are thought to mediate protection. Vaccine development may be hindered by modulation of vaccine-induced T cells by pathogen-activated immunoregulatory responses, e.g., via IL-10.We screened SA proteins for CD4+ T-cell-activating and IL-10/IL-17-inducing capacities using healthy donor-derived PBMCs. Responses were characterized (Th1/Th17/Th22/immunosuppressive IL-10-producing cells) using intracellular cytokine staining and flow cytometry. Phenotypic plasticity of Th1/Th17 cells was evaluated under pro- or anti-inflammatory conditions using modulatory cytokines. The impact of vaccination on SA-specific memory responses was assessed using samples from a clinical trial evaluating AS03-adjuvanted and non-adjuvanted multicomponent (CPS5/CPS8/α-toxin/ClfA) vaccines (NCT01160172).The donors exhibited SA-specific memory T-cell responses, indicative of pre-existing immunity to SA. We identified effective activators of Th1 responses (EbhA/IsaA/SdrE/MntC/Aaa/α-toxin), and Th17 and Th1/Th17 responses (EbhA/IsaA/SdrE and, to a lesser extent, α-toxin), but not of Th22 responses or IL-10 production. MRPII, IsdA, and ClfA were inefficient CD4+ T-cell activators in our assays. IL-10, likely produced by innate immune cells, influenced mainly Th1 cells by suppressing IFN-γ production. The memory CD4+ T-cells observed after long-term stimulation with α-toxin and ClfA indicated that vaccination with these proteins had induced expansion of pre-existing Th1 but not Th17 responses, without apparent adjuvant effect, confirming the trial data. The Th1/Th17-driving proteins (EbhA/IsaA/SdrE) shared low IL-10-promoting abilities and restricted phenotypic plasticity under pro- and anti-inflammatory conditions.Given the complex immunopathology and multiple virulence factors, identification of Th1/Th17-driving antigens, adjuvants and administration routes, and delineation of the role of memory responses, may advance vaccine development.
Collapse
|