1
|
Sanz-Codina M, van Os W, Pham AD, Jorda A, Wölf-Duchek M, Bergmann F, Lackner E, Lier C, van Hasselt JGC, Minichmayr IK, Dorn C, Zeitlinger M, al Jalali V. Target-site cefiderocol pharmacokinetics in soft tissues of healthy volunteers. J Antimicrob Chemother 2024; 79:3281-3288. [PMID: 39373642 PMCID: PMC11638555 DOI: 10.1093/jac/dkae359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/21/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Cefiderocol may potentially be used to treat skin and soft tissue infections (SSTIs). However, the pharmacokinetics of cefiderocol in human soft tissues have not yet been determined. The objective of the present PK study was to investigate whether target-site concentrations of cefiderocol are sufficiently high for the treatment of SSTIs. METHODS In this pharmacokinetic study, a single intravenous dose of 2 g cefiderocol was administered to eight healthy male volunteers. Drug concentrations were determined in plasma, muscle and subcutis over 8 h. Free plasma concentrations were calculated using the plasma protein binding determined with ultrafiltration. Free tissue concentrations were obtained using microdialysis. Penetration ratios were calculated as AUC0-8h_free_tissue/AUC0-8h_free_plasma. A population pharmacokinetic model was developed, and the probability of target attainment (PTA) was determined using Monte Carlo simulations. RESULTS Cefiderocol showed good tissue penetration, with mean penetration ratios ± standard deviation of 0.99 ± 0.33 and 0.92 ± 0.30 for subcutis and muscle, respectively. Cefiderocol pharmacokinetics in plasma were best described with a two-compartment model, and tissue concentrations were described by scaling the tissue concentrations to concentrations in the peripheral compartment of the plasma model. For a thrice-daily regimen with 2 g doses intravenously infused over 3 h, PTA was ≥90% for MIC values up to 4 mg/L, both based on free plasma and soft tissue pharmacokinetics. CONCLUSIONS This study indicates that a dose of 2 g cefiderocol achieves concentrations in plasma considered sufficient for treating relevant bacterial species. Assuming a comparable PK/PD target for soft tissues, sufficiently high concentrations would also be achieved in soft tissues.
Collapse
Affiliation(s)
- Maria Sanz-Codina
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Wisse van Os
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anh Duc Pham
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Wölf-Duchek
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Felix Bergmann
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Edith Lackner
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Constantin Lier
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - J G Coen van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Iris K Minichmayr
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christoph Dorn
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Valentin al Jalali
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Tian T, Yu Q, Yang D, Zhang X, Zhang C, Li J, Luo T, Zhang K, Lv X, Wang Y, Wang H, Li H. Endothelial α 1-adrenergic receptor activation improves cardiac function in septic mice via PKC-ERK/p38MAPK signaling pathway. Int Immunopharmacol 2024; 141:112937. [PMID: 39182270 DOI: 10.1016/j.intimp.2024.112937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Cardiomyopathy is particularly common in septic patients. Our previous studies have shown that activation of the alpha 1 adrenergic receptor (α1-AR) on cardiomyocytes inhibits sepsis-induced myocardial dysfunction. However, the role of cardiac endothelial α1-AR in septic cardiomyopathy has not been determined. Here, we identified α1-AR expression in mouse and human endothelial cells and showed that activation of α1-AR with phenylephrine (PE) improved cardiac function and survival by preventing cardiac endothelial injury in septic mice. Mechanistically, activating α1-AR with PE decreased the expression of ICAM-1, VCAM-1, iNOS, E-selectin, and p-p38MAPK, while promoting PKC and ERK1/2 phosphorylation in LPS-treated endothelial cells. These effects were abolished by a PKC inhibitor or α1-AR antagonist. PE also reduced p65 nuclear translocation, but this suppression is not blocked by PKC inhibition. Treatment with U0126 (a specific ERK1/2 inhibitor) reversed the effects of PE on p38MAPK phosphorylation. Our results demonstrate that cardiac endothelial α1-AR activation prevents sepsis-induced myocardial dysfunction in mice by inhibiting the endothelial injury via PKC-ERK/p38MAPK signaling pathway and a PKC-independent inhibition of p65 nuclear translocation. These findings offer a new perspective for septic patients with cardiac dysfunction by inhibiting cardiac endothelial cell injury through α1-AR activation.
Collapse
Affiliation(s)
- Tian Tian
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Qing Yu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Duomeng Yang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xue Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chanjuan Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jianling Li
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuxiu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongmei Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
3
|
Garduno A, Martín-Loeches I. Targeting Sepsis: Disease Tolerance, Immune Resilience, and Compartmentalized Immunity. Biomedicines 2024; 12:2420. [PMID: 39594987 PMCID: PMC11592085 DOI: 10.3390/biomedicines12112420] [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: 09/18/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
Introduction: Sepsis remains a major contributor to critical care mortality and morbidity worldwide. Despite advances in understanding its complex immunopathology, the compartmentalized nature of immune responses across different organs has yet to be fully translated into targeted therapies. This review explores the burden of sepsis on organ-specific immune dysregulation, immune resilience, and epigenetic reprogramming, emphasizing translational challenges and opportunities. Methods: We implemented a systematic literature search strategy, incorporating data from studies published between 2010 and 2024, to evaluate the role of molecular profiling techniques, including transcriptomics and epigenetic markers, in assessing the feasibility of targeted therapies. Results: Sepsis-induced immune dysregulation manifests differently in various organs, with lung, heart, liver, and kidney responses driven by unique local immune environments. Organ-specific biomarkers, such as the Spns2/S1P axis in lung macrophages, mitochondrial dysfunction in the heart, proenkephalin for early acute kidney injury (AKI), and adrenomedullin for predicting multi-organ failure, offer promising avenues for timely intervention. Furthermore, immune resilience, particularly through regulatory T-cell modulation and cytokine targeting (e.g., IL-18), is crucial for long-term recovery. Epigenetic mechanisms, including histone modification and trained immunity, present opportunities for reprogramming immune responses but require more precision to avoid unintended inflammatory sequelae. Conclusions: A deeper understanding of compartmentalized immune responses and the dynamic immune landscape in sepsis is critical for developing precision therapies. Real-time immune monitoring and organ-targeted interventions could revolutionize sepsis management, although significant barriers remain in clinical translation. Further research is required to establish biomarkers and treatment timing that optimize therapeutic efficacy while minimizing systemic risks.
Collapse
Affiliation(s)
- Alexis Garduno
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James’ Hospital, D08 NHY1 Dublin, Ireland;
| | - Ignacio Martín-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James’ Hospital, D08 NHY1 Dublin, Ireland;
- Hospital Clinic, Universitat de Barcelona, IDIBAPS, CIBERES, 08036 Barcelona, Spain
| |
Collapse
|
4
|
Feng J, Liu L, Liu J, Wang J. Immunological alterations in the endothelial barrier: a new predictive and therapeutic paradigm for sepsis. Expert Rev Clin Immunol 2024; 20:1205-1217. [PMID: 38850066 DOI: 10.1080/1744666x.2024.2366301] [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: 03/12/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Despite the fact incidence and mortality vary widely among regions, sepsis remains a major cause of morbidity and cost worldwide. The importance of the endothelial barrier in sepsis and infectious diseases is increasingly recognized; however, the underlying pathophysiology of the endothelial barrier in sepsis remains poorly understood. AREAS COVERED Here we review the advances in basic and clinical research for relevant papers in PubMed database. We attempt to provide an updated overview of immunological alterations in endothelial dysfunction, discussing the central role of endothelial barrier involved in sepsis to provide new predictive and therapeutic paradigm for sepsis. EXPERT OPINION Given its physiological and immunological functions in infectious diseases, the endothelial barrier has been dramatically altered in sepsis, suggesting that endothelial dysfunction may play a critical role in the pathogenesis of sepsis. Although many reliable biomarkers have been investigated to monitor endothelial activation and injury in an attempt to find diagnostic and therapeutic tools, there are no specific therapies to treat sepsis due to its complex pathophysiology. Since sepsis is initiated by both hyperinflammation and immunoparalysis occurring simultaneously, a 'one-treatment-fits-all' strategy for sepsis-induced immune injury and immunoparalysis is bound to fail, and an individualized 'precision medicine' approach is required.
Collapse
Affiliation(s)
- Jun Feng
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lina Liu
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junya Liu
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Gonçalves Pereira J, Fernandes J, Mendes T, Gonzalez FA, Fernandes SM. Artificial Intelligence to Close the Gap between Pharmacokinetic/Pharmacodynamic Targets and Clinical Outcomes in Critically Ill Patients: A Narrative Review on Beta Lactams. Antibiotics (Basel) 2024; 13:853. [PMID: 39335027 PMCID: PMC11428226 DOI: 10.3390/antibiotics13090853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial dosing can be a complex challenge. Although a solid rationale exists for a link between antibiotic exposure and outcome, conflicting data suggest a poor correlation between pharmacokinetic/pharmacodynamic targets and infection control. Different reasons may lead to this discrepancy: poor tissue penetration by β-lactams due to inflammation and inadequate tissue perfusion; different bacterial response to antibiotics and biofilms; heterogeneity of the host's immune response and drug metabolism; bacterial tolerance and acquisition of resistance during therapy. Consequently, either a fixed dose of antibiotics or a fixed target concentration may be doomed to fail. The role of biomarkers in understanding and monitoring host response to infection is also incompletely defined. Nowadays, with the ever-growing stream of data collected in hospitals, utilizing the most efficient analytical tools may lead to better personalization of therapy. The rise of artificial intelligence and machine learning has allowed large amounts of data to be rapidly accessed and analyzed. These unsupervised learning models can apprehend the data structure and identify homogeneous subgroups, facilitating the individualization of medical interventions. This review aims to discuss the challenges of β-lactam dosing, focusing on its pharmacodynamics and the new challenges and opportunities arising from integrating machine learning algorithms to personalize patient treatment.
Collapse
Affiliation(s)
- João Gonçalves Pereira
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Serviço de Medicina Intensiva, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Joana Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Centro Hospitalar de Trás-os-Montes e Alto Douro, 5000-508 Vila Real, Portugal
| | - Tânia Mendes
- Serviço de Medicina Interna, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Filipe André Gonzalez
- Serviço de Medicina Intensiva, Hospital Garcia De Orta, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Susana M Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Hospital Santa Maria, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
6
|
He RR, Yue GL, Dong ML, Wang JQ, Cheng C. Sepsis Biomarkers: Advancements and Clinical Applications-A Narrative Review. Int J Mol Sci 2024; 25:9010. [PMID: 39201697 PMCID: PMC11354379 DOI: 10.3390/ijms25169010] [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: 07/22/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
Sepsis is now defined as a life-threatening syndrome of organ dysfunction triggered by a dysregulated host response to infection, posing significant challenges in critical care. The main objective of this review is to evaluate the potential of emerging biomarkers for early diagnosis and accurate prognosis in sepsis management, which are pivotal for enhancing patient outcomes. Despite advances in supportive care, traditional biomarkers like C-reactive protein and procalcitonin have limitations, and recent studies have identified novel biomarkers with increased sensitivity and specificity, including circular RNAs, HOXA distal transcript antisense RNA, microRNA-486-5p, protein C, triiodothyronine, and prokineticin 2. These emerging biomarkers hold promising potential for the early detection and prognostication of sepsis. They play a crucial role not only in diagnosis but also in guiding antibiotic therapy and evaluating treatment effectiveness. The introduction of point-of-care testing technologies has brought about a paradigm shift in biomarker application, enabling swift and real-time patient evaluation. Despite these advancements, challenges persist, notably concerning biomarker variability and the lack of standardized thresholds. This review summarizes the latest advancements in sepsis biomarker research, spotlighting the progress and clinical implications. It emphasizes the significance of multi-biomarker strategies and the feasibility of personalized medicine in sepsis management. Further verification of biomarkers on a large scale and their integration into clinical practice are advocated to maximize their efficacy in future sepsis treatment.
Collapse
Affiliation(s)
- Rong-Rong He
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (R.-R.H.); (G.-L.Y.)
| | - Guo-Li Yue
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (R.-R.H.); (G.-L.Y.)
| | - Mei-Ling Dong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China;
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Jia-Qi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Chen Cheng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China;
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| |
Collapse
|
7
|
Cazanga V, Riquelme J, Cornejo D, Jeldres JA, Palma C, Pérez-Fernández R. Influence of Escherichia coli lipopolysaccharide-induced endotoxemia on plasma and tissue disposition of florfenicol after intramuscular administration in rabbits. Res Vet Sci 2024; 170:105187. [PMID: 38422840 DOI: 10.1016/j.rvsc.2024.105187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
To assess the effects of the acute inflammatory response (AIR) induced by Escherichia coli lipopolysaccharide (LPS) on plasma and tissue disposition of florfenicol (FFC) and its metabolite florfenicol amine (FFC-a), after its intramuscular (IM) administration, twenty-two New Zealand rabbits were randomly distributed in two experimental groups: Group 1 (LPS) was treated with three intravenous doses of 2 μg LPS/kg bw, before an intramuscular dose of 20 mg/kg FFC twenty-four h after the first LPS or SS injection; Group 2 (Control) was treated with saline solution (SS) in equivalent volumes as LPS-treated group. Blood samples were collected before (T0) and at different times after FFC administration. Acute inflammatory response was assessed in a parallel study where significant increases in body temperature, C-reactive protein concentrations and leukopenia were observed in the group treated with LPS. In another two groups of rabbits, 4 h after FFC treatment, rabbits were euthanized and tissue samples were collected for analysis of FFC and FFC-a concentrations. Pharmacokinetic parameters of FFC that showed significantly higher values in LPS-treated rabbits compared with control rabbits were absorption half-life, area under the curve, mean residence time and clearance /F (Cl/F). Elimination half-life and mean residence time of FFC-a were significantly higher in LPS-treated rabbits, whereas the metabolite ratio of FFC-a decreased significantly. Significant differences in tissue distribution of FFC and FFC-a were observed in rabbits treated with LPS. Modifications in plasma and tissue disposition of FFC and FFC-a were attributed mainly to haemodynamic modifications induced by the AIR through LPS administration.
Collapse
Affiliation(s)
- Victoria Cazanga
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile.
| | - José Riquelme
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile
| | - Diego Cornejo
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile
| | - Jessie-Ana Jeldres
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile
| | - Cristina Palma
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile
| | - Rubén Pérez-Fernández
- Laboratorio de Farmacología, Departamento de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile.
| |
Collapse
|
8
|
Wang P, Liu S, Sun T, Yang J. Daily fluid intake as a novel covariate affecting the population pharmacokinetics of polymyxin B in patients with sepsis. Int J Antimicrob Agents 2024; 63:107099. [PMID: 38280575 DOI: 10.1016/j.ijantimicag.2024.107099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/08/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Polymyxin B dosing in patients with sepsis is difficult because pathophysiological changes and supportive therapies alter drug pharmacokinetics (PK). This study aimed to investigate the impact of fluid management and renal function on the PK of polymyxin B and to propose alternative dosing regimens. METHODS Patients (aged ≥ 18 y) with sepsis and receiving intravenous polymyxin B for ≥ 96 h were enrolled. Blood samples were collected at steady state. Plasma concentrations were measured by liquid chromatography-tandem mass spectrometry and subjected to population PK modelling. Monte Carlo simulations were used to optimise dosage regimens. RESULTS Eighty-three patients with a median (range) daily fluid intake of 4.2 (1.3-8.4) L and a creatinine clearance (CrCL) of 87.5 (17.3-309.7) mL/min were included. Polymyxin B PK was adequately characterised by a two-compartment model. The PK covariate analysis revealed daily fluid intake statistically significantly affected central volume of distribution and central compartment clearance (CL), and CrCL influenced CL. Simulation indicated that a decreased dosing would be suitable for patients with renal dysfunction (CrCL < 40 mL/min), and therapeutic drug monitoring is recommended to avoid exposure fluctuation when patients have fluid overload. CONCLUSIONS Fluid management as well as renal function are essential factors affecting polymyxin B PK for patients with sepsis, which can help optimise dosage regimens.
Collapse
Affiliation(s)
- Peile Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China; Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Shaohua Liu
- Department of General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongwen Sun
- Department of General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China; Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
9
|
Cortellini S, DeClue AE, Giunti M, Goggs R, Hopper K, Menard JM, Rabelo RC, Rozanski EA, Sharp CR, Silverstein DC, Sinnott-Stutzman V, Stanzani G. Defining sepsis in small animals. J Vet Emerg Crit Care (San Antonio) 2024; 34:97-109. [PMID: 38351524 DOI: 10.1111/vec.13359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE To discuss the definitions of sepsis in human and veterinary medicine. DESIGN International, multicenter position statement on the need for consensus definitions of sepsis in veterinary medicine. SETTING Veterinary private practice and university teaching hospitals. ANIMALS Dogs and cats. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Sepsis is a life-threatening condition associated with the body's response to an infection. In human medicine, sepsis has been defined by consensus on 3 occasions, most recently in 2016. In veterinary medicine, there is little uniformity in how sepsis is defined and no consensus on how to identify it clinically. Most publications rely on modified criteria derived from the 1991 and 2001 human consensus definitions. There is a divergence between the human and veterinary descriptions of sepsis and no consensus on how to diagnose the syndrome. This impedes research, hampers the translation of pathophysiology insights to the clinic, and limits our abilities to optimize patient care. It may be time to formally define sepsis in veterinary medicine to help the field move forward. In this narrative review, we present a synopsis of prior attempts to define sepsis in human and veterinary medicine, discuss developments in our understanding, and highlight some criticisms and shortcomings of existing schemes. CONCLUSIONS This review is intended to serve as the foundation of current efforts to establish a consensus definition for sepsis in small animals and ultimately generate evidence-based criteria for its recognition in veterinary clinical practice.
Collapse
Affiliation(s)
- Stefano Cortellini
- Department of Clinical Science and Services, The Royal Veterinary College, University of London, Hatfield, UK
| | - Amy E DeClue
- Fetch Specialty and Emergency Veterinary Center, Greenville, South Carolina, USA
| | - Massimo Giunti
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Robert Goggs
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Kate Hopper
- Department of Veterinary Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Julie M Menard
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Elizabeth A Rozanski
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Claire R Sharp
- School of Veterinary Medicine, Murdoch University, Perth, Western Australia, Australia
| | - Deborah C Silverstein
- Department of Clinical Studies and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
10
|
Wang M, Feng J, Zhou D, Wang J. Bacterial lipopolysaccharide-induced endothelial activation and dysfunction: a new predictive and therapeutic paradigm for sepsis. Eur J Med Res 2023; 28:339. [PMID: 37700349 PMCID: PMC10498524 DOI: 10.1186/s40001-023-01301-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Lipopolysaccharide, a highly potent endotoxin responsible for severe sepsis, is the major constituent of the outer membrane of gram-negative bacteria. Endothelial cells participate in both innate and adaptive immune responses as the first cell types to detect lipopolysaccharide or other foreign debris in the bloodstream. Endothelial cells are able to recognize the presence of LPS and recruit specific adaptor proteins to the membrane domains of TLR4, thereby initiating an intracellular signaling cascade. However, lipopolysaccharide binding to endothelial cells induces endothelial activation and even damage, manifested by the expression of proinflammatory cytokines and adhesion molecules that lead to sepsis. MAIN FINDINGS LPS is involved in both local and systemic inflammation, activating both innate and adaptive immunity. Translocation of lipopolysaccharide into the circulation causes endotoxemia. Endothelial dysfunction, including exaggerated inflammation, coagulopathy and vascular leakage, may play a central role in the dysregulated host response and pathogenesis of sepsis. By discussing the many strategies used to treat sepsis, this review attempts to provide an overview of how lipopolysaccharide induces the ever more complex syndrome of sepsis and the potential for the development of novel sepsis therapeutics. CONCLUSIONS To reduce patient morbidity and mortality, preservation of endothelial function would be central to the management of sepsis.
Collapse
Affiliation(s)
- Min Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Jun Feng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| | - Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
11
|
De Sutter PJ, De Cock P, Johnson TN, Musther H, Gasthuys E, Vermeulen A. Predictive Performance of Physiologically Based Pharmacokinetic Modelling of Beta-Lactam Antibiotic Concentrations in Adipose, Bone, and Muscle Tissues. Drug Metab Dispos 2023; 51:499-508. [PMID: 36639242 DOI: 10.1124/dmd.122.001129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models consist of compartments representing different tissues. As most models are only verified based on plasma concentrations, it is unclear how reliable associated tissue profiles are. This study aimed to assess the accuracy of PBPK-predicted beta-lactam antibiotic concentrations in different tissues and assess the impact of using effect site concentrations for evaluation of target attainment. Adipose, bone, and muscle concentrations of five beta-lactams (piperacillin, cefazolin, cefuroxime, ceftazidime, and meropenem) in healthy adults were collected from literature and compared with PBPK predictions. Model performance was evaluated with average fold errors (AFEs) and absolute AFEs (AAFEs) between predicted and observed concentrations. In total, 26 studies were included, 14 of which reported total tissue concentrations and 12 unbound interstitial fluid (uISF) concentrations. Concurrent plasma concentrations, used as baseline verification of the models, were fairly accurate (AFE: 1.14, AAFE: 1.50). Predicted total tissue concentrations were less accurate (AFE: 0.68, AAFE: 1.89). A slight trend for underprediction was observed but none of the studies had AFE or AAFE values outside threefold. Similarly, predictions of microdialysis-derived uISF concentrations were less accurate than plasma concentration predictions (AFE: 1.52, AAFE: 2.32). uISF concentrations tended to be overpredicted and two studies had AFEs and AAFEs outside threefold. Pharmacodynamic simulations in our case showed only a limited impact of using uISF concentrations instead of unbound plasma concentrations on target attainment rates. The results of this study illustrate the limitations of current PBPK models to predict tissue concentrations and the associated need for more accurate models. SIGNIFICANCE STATEMENT: Clinical inaccessibility of local effect site concentrations precipitates a need for predictive methods for the estimation of tissue concentrations. This is the first study in which the accuracy of PBPK-predicted tissue concentrations of beta-lactam antibiotics in humans were assessed. Predicted tissue concentrations were found to be less accurate than concurrent predicted plasma concentrations. When using PBPK models to predict tissue concentrations, this potential relative loss of accuracy should be acknowledged when clinical tissue concentrations are unavailable to verify predictions.
Collapse
Affiliation(s)
- Pieter-Jan De Sutter
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Pieter De Cock
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Trevor N Johnson
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Helen Musther
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - Elke Gasthuys
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Department of Bioanalysis, Faculty of Pharmaceutical Sciences (P-J.DS., E.G., A.V.), Department of Basic and Applied Medical Science, Faculty of Medicine and Health Sciences (P.D-C), Ghent University, Ghent, Belgium; Department of Pharmacy and Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium (P.D-C.); and Certara UK Limited, Sheffield, United Kingdom (T.N.J., H.M.)
| |
Collapse
|
12
|
Codina MS, Bozkir HÖ, Jorda A, Zeitlinger M. Individualised antimicrobial dose optimisation: a systematic review and meta-analysis of randomised controlled trials. Clin Microbiol Infect 2023:S1198-743X(23)00134-9. [PMID: 36965694 DOI: 10.1016/j.cmi.2023.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/27/2023]
Abstract
BACKGROUND Therapeutic drug management (TDM) and model-informed precision dosing (MIPD) allow dose individualisation to increase drug effectivity and reduce toxicity. OBJECTIVES Evaluate the available evidence on the clinical efficacy of individualised antimicrobial dosing optimisation. METHODS Data sources: Pubmed, Embase, Web of Science, and Cochrane Library databases from database inception to the 11th of November 2022. STUDY ELIGIBILITY CRITERIA Published peer-reviewed Randomised Controlled Trials (RCTs). PARTICIPANTS Human subjects aged ≥18 years receiving an antibiotic or antifungal drug. INTERVENTIONS Patients receiving individualised antimicrobial dose adjustment. Assessment of risk of bias: Cochrane risk-of-bias tool for randomised trials (RoB2). Methods of data synthesis: Primary outcome was the risk of mortality. Secondary outcomes included target attainment, treatment failure, clinical and microbiological cure, length of stay, treatment duration and adverse events. Effect sizes were pooled using a random-effects model. Statistical heterogeneity was assessed by inconsistency testing (I2). RESULTS Ten RCTs were included in the meta-analysis (1,241 participants; n= 624 in the TDM group, n = 617 in the control group). Individualised antimicrobial dose optimisation was associated with a numerical decrease in mortality (RR = 0.86; 95% CI 0.71-1.05), without reaching statistical significance. Moreover, it was associated with significantly higher target attainment rates (RR = 1.41; 95% CI, 1.13-1.76) and a significant decrease in treatment failure (RR = 0.70; 95% CI, 0.54-0.92). Individualised antimicrobial dose optimisation was also associated with improvement, but not significant in clinical cure (RR = 1.33; 95% CI, 0.94-1.33) and microbiological outcome (RR = 1.25; CI, 1.00-1.57), as well as with a significant decrease in the risk of nephrotoxicity (RR = 0.55; 95% CI, 0.31-0.97). CONCLUSIONS This meta-analysis demonstrates that target attainment, treatment failure, and nephrotoxicity were significantly improved in patients who underwent individualised antimicrobial dose optimisation. However, it did not show a significant decrease in mortality, clinical cure or microbiological outcome.
Collapse
Affiliation(s)
- Maria Sanz Codina
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Haktan Övul Bozkir
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
13
|
Lee CW, You BH, Yim S, Han SY, Chae HS, Bae M, Kim SY, Yu JE, Jung J, Nhoek P, Kim H, Choi HS, Chin YW, Kim HW, Choi YH. Change of metformin concentrations in the liver as a pharmacological target site of metformin after long-term combined treatment with ginseng berry extract. Front Pharmacol 2023; 14:1148155. [PMID: 36998615 PMCID: PMC10043734 DOI: 10.3389/fphar.2023.1148155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Metformin as an oral glucose-lowering drug is used to treat type 2 diabetic mellitus. Considering the relatively high incidence of cardiovascular complications and other metabolic diseases in diabetic mellitus patients, a combination of metformin plus herbal supplements is a preferrable way to improve the therapeutic outcomes of metformin. Ginseng berry, the fruit of Panax ginseng Meyer, has investigated as a candidate in metformin combination mainly due to its anti-hyperglycemic, anti-hyperlipidemic, anti-obesity, anti-hepatic steatosis and anti-inflammatory effects. Moreover, the pharmacokinetic interaction of metformin via OCTs and MATEs leads to changes in the efficacy and/or toxicity of metformin. Thus, we assessed how ginseng berry extract (GB) affects metformin pharmacokinetics in mice, specially focusing on the effect of the treatment period (i.e., 1-day and 28-day) of GB on metformin pharmacokinetics. In 1-day and 28-day co-treatment of metformin and GB, GB did not affect renal excretion as a main elimination route of metformin and GB therefore did not change the systemic exposure of metformin. Interestingly, 28-day co-treatment of GB increased metformin concentration in the livers (i.e., 37.3, 59.3% and 60.9% increases versus 1-day metformin, 1-day metformin plus GB and 28-day metformin groups, respectively). This was probably due to the increased metformin uptake via OCT1 and decreased metformin biliary excretion via MATE1 in the livers. These results suggest that co-treatment of GB for 28 days (i.e., long-term combined treatment of GB) enhanced metformin concentration in the liver as a pharmacological target tissue of metformin. However, GB showed a negligible impact on the systemic exposure of metformin in relation to its toxicity (i.e., renal and plasma concentrations of metformin).
Collapse
Affiliation(s)
- Choong Whan Lee
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Byoung Hoon You
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Sreymom Yim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Seung Yon Han
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hee-Sung Chae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS, United States
| | - Mingoo Bae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Seo-Yeon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jeong-Eun Yu
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jieun Jung
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Piseth Nhoek
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk-University Ilsan Oriental Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Han Seok Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Woo Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young Hee Choi
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang-si, Gyeonggi-do, Republic of Korea
- *Correspondence: Young Hee Choi,
| |
Collapse
|
14
|
Thu VTA, Dat LD, Jayanti RP, Trinh HKT, Hung TM, Cho YS, Long NP, Shin JG. Advancing personalized medicine for tuberculosis through the application of immune profiling. Front Cell Infect Microbiol 2023; 13:1108155. [PMID: 36844400 PMCID: PMC9950414 DOI: 10.3389/fcimb.2023.1108155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
While early and precise diagnosis is the key to eliminating tuberculosis (TB), conventional methods using culture conversion or sputum smear microscopy have failed to meet demand. This is especially true in high-epidemic developing countries and during pandemic-associated social restrictions. Suboptimal biomarkers have restricted the improvement of TB management and eradication strategies. Therefore, the research and development of new affordable and accessible methods are required. Following the emergence of many high-throughput quantification TB studies, immunomics has the advantages of directly targeting responsive immune molecules and significantly simplifying workloads. In particular, immune profiling has been demonstrated to be a versatile tool that potentially unlocks many options for application in TB management. Herein, we review the current approaches for TB control with regard to the potentials and limitations of immunomics. Multiple directions are also proposed to hopefully unleash immunomics' potential in TB research, not least in revealing representative immune biomarkers to correctly diagnose TB. The immune profiles of patients can be valuable covariates for model-informed precision dosing-based treatment monitoring, prediction of outcome, and the optimal dose prediction of anti-TB drugs.
Collapse
Affiliation(s)
- Vo Thuy Anh Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Ly Da Dat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Rannissa Puspita Jayanti
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Hoang Kim Tu Trinh
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh, Ho Chi Minh City, Vietnam
| | - Tran Minh Hung
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Yong-Soon Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea,*Correspondence: Jae-Gook Shin, ; Nguyen Phuoc Long,
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea,Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, Republic of Korea,Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea,*Correspondence: Jae-Gook Shin, ; Nguyen Phuoc Long,
| |
Collapse
|
15
|
He YF, Hu XM, Khan MA, Yu BY, Sheng YC, Xiao XZ, Wan XX, Tan SP, Xiong K. HSF1 Alleviates Brain Injury by Inhibiting NLRP3-Induced Pyroptosis in a Sepsis Model. Mediators Inflamm 2023; 2023:2252255. [PMID: 36741074 PMCID: PMC9897924 DOI: 10.1155/2023/2252255] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/15/2022] [Accepted: 09/16/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Sepsis, which could cause a systemic inflammatory response, is a life-threatening disease with a high morbidity and mortality rate. There is evidence that brain injury may be related to severe systemic infection induced by sepsis. The brain injury caused by sepsis could increase the risk of mortality in septic patients, which seriously affects the septic patient's prognosis of survival. Although there remains a focus on sepsis research, clinical measures to prevent and treat brain injury in sepsis are not yet available, and the high mortality rate is still a big health burden. Therefore, it is necessary to investigate the new molecules or regulated pathways that can effectively inhibit the progress of sepsis. OBJECTIVE NLR family pyrin domain-containing 3 (NLRP3) increased in the procession of sepsis and functioned as the key regulator of pyroptosis. Heat shock factor 1 (HSF1) can protect organs from multiorgan dysfunction syndrome induced by lipopolysaccharides in mice, and NLRP3 could be inhibited by HSF1 in many organs. However, whether HSF1 regulated NLRP3 in sepsis-induced brain injury, as well as the detailed mechanism of HSF1 in brain injury, remains unknown in the sepsis model. In this research, we try to explore the relationship between HSF1 and NLRP3 in a sepsis model and try to reveal the mechanism of HSF1 inhibiting the process of brain injury. METHODS In this study, we used wild-type mice and hsf1 -/- mice for in vivo research and PC12 cells for in vitro research. Real-time PCR and Western blot were used to analyze the expression of HSF1, NLRP3, cytokines, and pyrolytic proteins. EthD-III staining was chosen to detect the pyroptosis of the hippocampus and PC12 cells. RESULTS The results showed that HSF1 is negatively related to pyroptosis. The pyroptosis in cells of brain tissue was significantly increased in the hsf1 -/- mouse model compared to hsf1 +/+ mice. In PC12 cells, hsf1 siRNA can upregulate pyroptosis while HSF1-transfected plasmid could inhibit the pyroptosis. HSF1 could negatively regulate the NLRP3 pathway in PC12 cells, while hsf1 siRNA enhanced the pyroptosis in PC12 cells, which could be reversed by nlrp3 siRNA. CONCLUSION These results imply that HSF1 could alleviate sepsis-induced brain injury by inhibiting pyroptosis through the NLRP3-dependent pathway in brain tissue and PC12 cells, suggesting HSF1 as a potential molecular target for treating brain injury in sepsis clinical studies.
Collapse
Affiliation(s)
- Yi-fu He
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xi-min Hu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Md. Asaduzzaman Khan
- The Research Centre for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Bo-yao Yu
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Yi-cun Sheng
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xian-zhong Xiao
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xin-xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Si-pin Tan
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
16
|
Association of Procalcitonin with the Patient’s Infection Characteristics and Prognosis after Hematopoietic Stem Cell Transplantation. DISEASE MARKERS 2022; 2022:9157396. [PMID: 36148158 PMCID: PMC9489411 DOI: 10.1155/2022/9157396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/08/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022]
Abstract
Objective To study whether procalcitonin (PCT) is an important indicator of infection with or without agranulocytosis and to reveal whether PCT can distinguish between infected sites and affect prognosis after hematopoietic stem cell transplantation (HSCT). Method In the present study, 682 patients with HSCT were enrolled, and their clinical characteristics were noted. Their blood culture and inflammatory and biochemical indicators were studied. The patients were divided into respective groups according to the degree of agranulocytosis, type of bacterial infection, infected sites, and prognosis. Results The PCT, CRP, and D-dimer levels were significantly improved in patients with positive blood culture results compared to the case for those with negative blood culture results. The PCT level was the highest in the gram-negative group. The levels of PCT and D-dimer were significantly elevated in patients with infection and agranulocytosis after HSCT compared to those in the nonagranulocytosis cohort. Interestingly, no significant difference in the PCT level was observed among any of the eight foci. Lower PCT levels were associated with higher survival in patients with infection after HSCT. Conclusion Among patients that underwent HSCT, PCT levels were significantly elevated in those with infection and agranulocytosis, with the levels being specifically high in the gram-negative group. Moreover, lower PCT levels were associated with higher survival in patients with infection after HSCT.
Collapse
|
17
|
Relationship between Pharmacokinetic/Pharmacodynamic Target Attainment and Microbiological Outcome in Critically Ill COVID-19 Patients with Documented Gram-Negative Superinfections Treated with TDM-Guided Continuous-Infusion Meropenem. Pharmaceutics 2022; 14:pharmaceutics14081585. [PMID: 36015211 PMCID: PMC9412264 DOI: 10.3390/pharmaceutics14081585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 12/13/2022] Open
Abstract
Objectives: The objective of this study was to explore the relationship between pharmacokinetic/pharmacodynamic (PK/PD) target attainment of continuous-infusion (CI) meropenem and microbiological outcome in critical COVID-19 patients with documented Gram-negative superinfections. Methods: Patients receiving CI meropenem for documented Gram-negative infections at the COVID ICU of the IRCCS Azienda Ospedaliero-Universitaria di Bologna and undergoing therapeutic drug monitoring from January 2021 to February 2022 were retrospectively assessed. Average steady-state meropenem concentrations (Css) were calculated and the Css/MIC ratio was selected as a pharmacodynamic parameter of meropenem efficacy. The Css/MIC ratio was defined as optimal if ≥4, quasi-optimal if between 1 and 4, and suboptimal if <1. The relationship between Css/MIC and microbiological outcome was assessed. Results: Overall, 43 critical COVID-19 patients with documented Gram-negative infections were retrieved. Combination therapy was implemented in 26 cases. Css/MIC ratios were optimal in 27 (62.8%), quasi-optimal in 7 (16.3%), and suboptimal in 9 cases (20.9%). Microbiological failure occurred in 21 patients (48.8%), with no difference between monotherapy and combination therapy (43.8% vs. 53.8%; p = 0.53). The microbiological failure rate was significantly lower in patients with an optimal Css/MIC ratio compared to those with a quasi-optimal or suboptimal Css/MIC ratio (33.3% vs. 75.0%; p = 0.01). Conclusion: Suboptimal attainment of meropenem PK/PD targets may be a major determinant impacting on microbiological failure in critical COVID-19 patients with Gram-negative superinfections.
Collapse
|
18
|
Ngougni Pokem P, Wittebole X, Collienne C, Rodriguez-Villalobos H, Tulkens PM, Elens L, Van Bambeke F, Laterre PF. Population Pharmacokinetics of Temocillin Administered by Continuous Infusion in Patients with Septic Shock Associated with Intra-Abdominal Infection and Ascitic Fluid Effusion. Antibiotics (Basel) 2022; 11:898. [PMID: 35884152 PMCID: PMC9311849 DOI: 10.3390/antibiotics11070898] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/16/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Temocillin is active against Gram-negative bacteria, including many extended-spectrum β-lactamase (ESBL)-producing Enterobacterales. We studied its pharmacokinetics in plasma and ascitic fluid after intravenous administration of a loading dose of 2 g over 30 min, followed by continuous infusion of 6 g/24 h, to 19 critically-ill patients with septic shock associated with complicated intra-abdominal infection. We established a pharmacokinetic model describing unbound temocillin concentrations in plasma and ascitic fluid and performed Monte-Carlo simulations to evaluate the probability of target attainment (PTA) of unbound concentrations (100% fT > MIC, i.e., unbound concentrations remaining above the MIC during 100% of the time) for the applied and hypothetical dosing regimens. The temocillin AUC in ascitic fluid was 46% of the plasma AUC. Plasma unbound concentrations were best described by a two-compartment model, and an additional compartment was added to describe unbound concentration in ascitic fluid, with renal clearance as a covariate. Dosing simulations showed that 90% PTA was achieved in the plasma with the current dosing regimen for MIC ≤ 16 mg/L (EUCAST susceptibility breakpoint) but not in the ascitic fluid if renal clearance was ≥40 mL/min. Hypothetical dosing with a higher (a) loading dose or (b) infused dose allowed to reach target concentrations in ascitic fluid (a) more rapidly or (b) sustainably, but these simulations need to be evaluated in the clinics for safety and efficacy.
Collapse
Affiliation(s)
- Perrin Ngougni Pokem
- Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.N.P.); (P.M.T.)
- Integrated PharmacoMetrics, PharmacoGenomics and PharmacoKinetics, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Xavier Wittebole
- Department of Critical Care Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium; (X.W.); (C.C.); (P.-F.L.)
| | - Christine Collienne
- Department of Critical Care Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium; (X.W.); (C.C.); (P.-F.L.)
| | | | - Paul M. Tulkens
- Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.N.P.); (P.M.T.)
| | - Laure Elens
- Integrated PharmacoMetrics, PharmacoGenomics and PharmacoKinetics, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Françoise Van Bambeke
- Pharmacologie Cellulaire et Moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium; (P.N.P.); (P.M.T.)
| | - Pierre-François Laterre
- Department of Critical Care Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium; (X.W.); (C.C.); (P.-F.L.)
| |
Collapse
|