1
|
Jorda A, Pracher L, Eberl S, Nussbaumer-Pröll A, Sarhan M, Weber M, Wahrmann M, Al Jalali V, Bergmann F, Prager M, Leutzendorff A, Sanz-Codina M, Tegrovsky L, Pecho T, Jilma B, Müller L, Spittler A, Rocha-Hasler M, Eckl-Dorna J, Kusienicka A, Farlik M, Zeitlinger M. Stability of cytokine, cellular and clinical response to the intravenous LPS challenge repeated after one year: a healthy volunteer trial. Med Microbiol Immunol 2025; 214:14. [PMID: 40047923 PMCID: PMC11885351 DOI: 10.1007/s00430-025-00823-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Whether the magnitude of individual cytokine, cellular, and clinical responses to the intravenous lipopolysaccharide (LPS) challenge is constant in individuals over extended time periods is unknown. Nine healthy volunteers received an intravenous LPS injection of 2 ng/kg bodyweight twice at intervals of at least one year. Circulating cytokines and leukocyte subsets were quantified using a multiplex immunoassay and cytometry by time-of-flight, respectively. Self-reported symptoms and vital signs were also assessed. We observed moderate to strong intra-individual correlations in the responsiveness of most cytokines (IL-6 [AUC0 - 10]: R = 0.93, p < 0.001; CRP [mg/dL]: R = 0.88, p = 0.004; IL-8 [AUC0 - 10]: R = 0.71, p = 0.031; TNF-alpha [AUC0 - 10]: R = 0.67, p = 0.047; IL-10 [AUC0 - 10]: R = 0.42, p = 0.26) and cellular subsets (CD8 T lymphocytes: R = 0.9, p = 0.002; B lymphocytes [G/L]: R = 0.89, p = 0.003; CD4 T lymphocytes: R = 0.84, p = 0.001; neutrophils: R = 0.80, p = 0.017; monocytes: R = 0.16, p = 0.710) between the 1st and 2nd LPS challenges. Vital signs and symptoms were not reproducible. While the average cellular and clinical response was similar between the two LPS challenges, we found a significantly attenuated AUC0 - 10 of IL-6 (percent difference, -41.9% [95% CI -73.0 - -10.7]) and TNF-alpha (percent difference, -35.7% [95% CI -70.0 - -1.6]) at the 2nd LPS challenge. Individual cytokine and cellular responses to intravenous LPS showed a significant degree of correlation when measured more than one year apart. These correlations did not translate to the reproducibility of clinical symptoms and vital signs, which showed greater variability and were not constant over time. The partly reduced cytokine release in the 2nd LPS challenge might be interpreted as an indicator of a long-lasting tolerance to endotoxin.
Collapse
Affiliation(s)
- Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lena Pracher
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Sabine Eberl
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Alina Nussbaumer-Pröll
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Vienna, Austria
| | - Valentin Al Jalali
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Felix Bergmann
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Marlene Prager
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Amelie Leutzendorff
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine III, Vienna, Austria
| | - Maria Sanz-Codina
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lara Tegrovsky
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Theresa Pecho
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lena Müller
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Research Laboratories, Core Facility Flow Cytometry & Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Anna Kusienicka
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria.
| |
Collapse
|
2
|
Carcillo JA, Shakoory B. Cytokine Storm and Sepsis-Induced Multiple Organ Dysfunction Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:441-457. [PMID: 39117832 DOI: 10.1007/978-3-031-59815-9_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
There is extensive overlap of clinical features among familial or primary HLH (pHLH), reactive or secondary hemophagocytic lymphohistiocytosis (sHLH) [including macrophage activation syndrome (MAS) related to rheumatic diseases], and hyperferritinemic sepsis-induced multiple organ dysfunction syndrome (MODS); however, the distinctive pathobiology that causes hyperinflammatory process in each condition requires careful considerations for therapeutic decision-making. pHLH is defined by five or more of eight HLH-2004 criteria [1], where genetic impairment of natural killer (NK) cells or CD8+ cytolytic T cells results in interferon gamma (IFN-γ)-induced hyperinflammation regardless of triggering factors. Cytolytic treatments (e.g., etoposide) or anti-IFN-γ monoclonal antibody (emapalumab) has been effectively used to bridge the affected patients to hematopoietic stem cell transplant. Secondary forms of HLH also have normal NK cell number with decreased cytolytic function of varying degrees depending on the underlying and triggering factors. Although etoposide was uniformly used in sHLH/MAS in the past, the treatment strategy in different types of sHLH/MAS is increasingly streamlined to reflect the triggering/predisposing conditions, severity/progression, and comorbidities. Accordingly, in hyperferritinemic sepsis, the combination of hepatobiliary dysfunction (HBD) and disseminated intravascular coagulation (DIC) reflects reticuloendothelial system dysfunction and defines sepsis-associated MAS. It is demonstrated that as the innate immune response to infectious organism prolongs, it results in reduction in T cells and NK cells with subsequent lymphopenia even though normal cytolytic activity continues (Figs. 30.1, 30.2, 30.3, and 30.4). These changes allow free hemoglobin and pathogens to stimulate inflammasome activation in the absence of interferon-γ (IFN-γ) production that often responds to source control, intravenous immunoglobulin (IVIg), plasma exchange, and interleukin 1 receptor antagonist (IL-1Ra), similar to non-EBV, infection-induced HLH.
Collapse
Affiliation(s)
- Joseph A Carcillo
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Bita Shakoory
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Fedorka CE, Ball BA, Walker OF, McCormick ME, Scoggin KE, Kennedy LA, Squires EL, Troedsson MHT. Alterations of Circulating Biomarkers During Late Term Pregnancy Complications in the Horse Part I: Cytokines. J Equine Vet Sci 2021; 99:103425. [PMID: 33781421 DOI: 10.1016/j.jevs.2021.103425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023]
Abstract
Equine abortions are attributed to both infectious and noninfectious causes. Clinical extrapolations are often made from the experimental model for ascending placentitis towards other causes of fetal compromise, including various markers of inflammation, including the cytokines IL-2, 5, IL-6, IL-10, IFNγ, and TNF. It is unknown if these cytokine changes are noted under field conditions, or if they increase preceding other pregnancy related complications. To assess this, Thoroughbred mares (n = 702) had weekly blood obtained beginning in December 2013 and continuing until parturition. Fetal membranes were submitted to the UKVDL for complete gross and pathologic assessment and classified as either ascending placentitis (n = 6), focal mucoid placentitis (n = 6), idiopathic abortion (n = 6) or control (n = 20). Weekly serum samples were analyzed via immunoassay for concentrations of IL-2, IL-5, IL-6, IL-10, IFNγ, and TNF. For both focal mucoid placentitis and ascending placentitis, an increase (P < .05) in the concentrations of IL-2, IL-5, IL-6, IL-10, IFNγ, and TNF was noted preceding parturition in comparison to controls. Cytokine profiles preceding idiopathic abortion did not differ from controls. In conclusion, serum cytokines may be considered potential biomarkers for the prediction of placental infection, while no changes in cytokine profiles were noted when noninfectious causes of abortion occurred. Additionally, this is the first study to report an increase in cytokines during the disease process of focal mucoid placentitis, the etiology of which includes Nocardioform placentitis.
Collapse
Affiliation(s)
- C E Fedorka
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY
| | - B A Ball
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY.
| | - O F Walker
- Lincoln Memorial University, College of Veterinary Medicine, Harrogate, TN
| | - M E McCormick
- Rhode Island College, College of Nursing. Providence, RI; Rhode Island Department of Health. Providence, RI
| | - K E Scoggin
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY
| | - L A Kennedy
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY
| | - E L Squires
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY
| | - M H T Troedsson
- University of Kentucky, Department of Veterinary Sciences, Lexington, KY
| |
Collapse
|
4
|
Brooks D, Barr LC, Wiscombe S, McAuley DF, Simpson AJ, Rostron AJ. Human lipopolysaccharide models provide mechanistic and therapeutic insights into systemic and pulmonary inflammation. Eur Respir J 2020; 56:13993003.01298-2019. [PMID: 32299854 DOI: 10.1183/13993003.01298-2019] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
Inflammation is a key feature in the pathogenesis of sepsis and acute respiratory distress syndrome (ARDS). Sepsis and ARDS continue to be associated with high mortality. A key contributory factor is the rudimentary understanding of the early events in pulmonary and systemic inflammation in humans, which are difficult to study in clinical practice, as they precede the patient's presentation to medical services. Lipopolysaccharide (LPS), a constituent of the outer membrane of Gram-negative bacteria, is a trigger of inflammation and the dysregulated host response in sepsis. Human LPS models deliver a small quantity of LPS to healthy volunteers, triggering an inflammatory response and providing a window to study early inflammation in humans. This allows biological/mechanistic insights to be made and new therapeutic strategies to be tested in a controlled, reproducible environment from a defined point in time. We review the use of human LPS models, focussing on the underlying mechanistic insights that have been gained by studying the response to intravenous and pulmonary LPS challenge. We discuss variables that may influence the response to LPS before considering factors that should be considered when designing future human LPS studies.
Collapse
Affiliation(s)
- Daniel Brooks
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Laura C Barr
- Dept of Respiratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Sarah Wiscombe
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Daniel F McAuley
- School of Medicine, Dentistry and Biomedical Sciences, Institute for Health Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - A John Simpson
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Anthony J Rostron
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
5
|
Coakley JD, Breen EP, Moreno-Olivera A, Al-Harbi AI, Melo AM, O’Connell B, McManus R, Doherty DG, Ryan T. Dysregulated T helper type 1 (Th1) and Th17 responses in elderly hospitalised patients with infection and sepsis. PLoS One 2019; 14:e0224276. [PMID: 31658288 PMCID: PMC6816565 DOI: 10.1371/journal.pone.0224276] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The role of Th1 and Th17 lymphocyte responses in human infection and sepsis of elderly patients has yet to be clarified. DESIGN A prospective observational study of patients with sepsis, infection only and healthy controls. SETTING The acute medical wards and intensive care units in a 1000 bed university hospital. PATIENTS 32 patients with sepsis, 20 patients with infection, and 20 healthy controls. Patients and controls were older than 65 years of age. Patients with recognised underlying immune compromise were excluded. METHODS Phenotype, differentiation status and cytokine production by T lymphocytes were determined by flow cytometry. MEASUREMENTS The differentiation states of circulating CD3+, CD4+, and CD8+ T cells were characterised as naive (CD45RA+, CD197+), central memory (CD45RA-, CD197+), effector memory (CD45RA-, CD197-), or terminally differentated (CD45RA+, CD197-). Expression of IL-12 and IL-23 receptors, and the transcription factors T-bet and RORγt, was analysed in circulating T lymphocytes. Expression of interferon- γ and IL-17A were analysed following stimulation in vitro. RESULTS CD4+ T cells from patients with infection predominantly expressed effector-memory or terminally differentiated phenotypes but CD4+ T cells from patients with severe sepsis predominantly expressed naive phenotypes (p<0.0001). CD4+ T cells expressing IL-23 receptor were lower in patients with sepsis compared to patients with infection alone (p = 0.007). RORγt expression by CD4+ T cells was less frequent in patients with sepsis (p<0.001), whereas T-bet expressing CD8+ T cells that do not express RORγt was lower in the sepsis patients. HLA-DR expression by monocytes was lower in patients with sepsis. In septic patients fewer monocytes expressed IL-23. CONCLUSION Persistent failure of T cell activation was observed in patients with sepsis. Sepsis was associated with attenuated CD8+Th1 and CD4+Th17 based lymphocyte response.
Collapse
Affiliation(s)
- John D. Coakley
- Department of Intensive Care Medicine, St James’s Hospital, Dublin, Ireland
- * E-mail:
| | - Eamon P. Breen
- Trinity Translational Medicine Institute, St James’s Hospital, Dublin, Ireland
| | - Ana Moreno-Olivera
- Department of Immunology, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Alhanouf I. Al-Harbi
- Department of Immunology, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Ashanty M. Melo
- Department of Immunology, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Brian O’Connell
- Department of Clinical Microbiology, St James’s Hospital, Dublin, Ireland
| | - Ross McManus
- Department of Clinical Medicine and Genetics, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Derek G. Doherty
- Department of Immunology, Trinity Translational Medicine Institute, Dublin, Ireland
| | - Thomas Ryan
- Department of Intensive Care Medicine, St James’s Hospital, Dublin, Ireland
| |
Collapse
|
6
|
B-cell dynamics during experimental endotoxemia in humans. Biosci Rep 2019; 39:BSR20182347. [PMID: 30962268 PMCID: PMC6522728 DOI: 10.1042/bsr20182347] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 03/28/2019] [Accepted: 04/05/2019] [Indexed: 01/05/2023] Open
Abstract
Recently, B cells with regulatory functions suppressing T-cell immunity were identified. Inflammation in the context of sepsis is characterized by a profound immune dysfunction increasing the patient’s risk for additional infections. The impact of endotoxemia on B-cell dynamics, regulatory B cells (Breg) and its contribution to immune dysfunction is unknown. It is the aim of the present study to characterize the dynamics of the B-cell compartment and Breg in an experimental human endotoxemia model. In this randomized placebo-controlled cross-over study, 20 healthy males received an intravenous injection of endotoxin (Escherichia coli lipopolysaccharide, LPS, 0.8 ng/kg body weight) or placebo (saline 0.9%) on two otherwise identical study days. B cells were analyzed by flow cytometry at baseline and repeatedly up to 72 h after endotoxin/placebo injection. Absolute CD19+ B cells counts showed a significant decrease 3 h after endotoxin injection. Memory B cells were partially depleted from the circulation; the total number of Breg was significantly diminished 3 h after LPS challenge. Production of anti-inflammatory interleukin (IL)-10 (IL-10) by Breg was unaltered after LPS challenge. Systemic B-cell activating factor (BAFF) levels were significantly increased with a maximum after 24 h and remained increased up to 72 h post-injection. Endotoxemia causes a transient depletion of memory B cells and Breg from the circulation. However, the functional capacity of B cells to produce IL-10 is not impaired.
Collapse
|
7
|
Brinkhoff A, Sieberichs A, Engler H, Dolff S, Benson S, Korth J, Schedlowski M, Kribben A, Witzke O, Wilde B. Pro-Inflammatory Th1 and Th17 Cells Are Suppressed During Human Experimental Endotoxemia Whereas Anti-Inflammatory IL-10 Producing T-Cells Are Unaffected. Front Immunol 2018; 9:1133. [PMID: 29868038 PMCID: PMC5968108 DOI: 10.3389/fimmu.2018.01133] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/04/2018] [Indexed: 01/05/2023] Open
Abstract
Objective Sepsis is one of the leading causes of the deaths in hospitals. During sepsis, patients are exposed to endotoxemia, which may contribute to the dysregulation of the immune system frequently observed in sepsis. This dysregulation leads to impaired pro-inflammatory responses and may increase the risk for secondary infections in sepsis. The experimental human endotoxemia model is widely used as a model system to study the acute effects of endotoxemia. Under physiological circumstances, the immune system is tightly regulated. Effector T-cells exert pro-inflammatory function and are restrained by regulatory T-cells (Tregs), which modulate pro-inflammatory effector responses. Endotoxemia may induce inadequate Treg activity or render effector T-cells dysfunctional. It was the aim of the study to investigate effector T-cell and Treg responses in an experimental human endotoxemia model. Methods In a cross-over designed placebo-controlled study, 20 healthy male volunteers received an intravenous injection of either lipopolysaccharide (LPS) (0.8 ng/kg body weight) or a placebo (saline 0.9%). CD3+ T-cells, CD4+ T-cells, CD8+ T-cells, and intracellular cytokine profiles were measured with flow cytometry at baseline and at repeated points after LPS/placebo injection. Complete blood cell counts were obtained with an automated hematology analyzer and cytokines were quantified by ELISA. Results Circulating neutrophils were significantly increased 2 h after LPS injection (p < 0.001) while absolute number of CD3+ T-cells, CD4+ T-cells, and CD8+ T-cells decreased (p < 0.001). Effector T-helper-cells (THs) showed a significant—but transient—decrease of pro-inflammatory IFNγ, interleukin (IL)-2, TNFα, and IL-17A production after LPS injection (p < 0.001). In contrast, the frequency of Treg and the capacity to produce IL-10 were unchanged (p = 0.21). Conclusion Effector THs fail to produce pro-inflammatory Th1-/Th17-associated cytokines after LPS challenge. In contrast, IL-10 production by Treg is not affected. Thus, endotoxemia-induced suppression of pro-inflammatory THs might be considered as a contributing factor to immunoparalysis in sepsis.
Collapse
Affiliation(s)
- Alexandra Brinkhoff
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annette Sieberichs
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sebastian Dolff
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sven Benson
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Korth
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
8
|
Villois P, Grimaldi D, Spadaro S, Shinotsuka CR, Fontana V, Scolletta S, Franchi F, Vincent JL, Creteur J, Taccone FS. Lymphopaenia in cardiac arrest patients. Ann Intensive Care 2017; 7:85. [PMID: 28808927 PMCID: PMC5555958 DOI: 10.1186/s13613-017-0308-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/06/2017] [Indexed: 12/29/2022] Open
Abstract
Background A decrease in circulating lymphocytes has been described as a marker of poor prognosis after septic shock; however, scarce data are available after cardiac arrest (CA). The aim of this study was to evaluate the impact of lymphopaenia after successful cardiopulmonary resuscitation. Methods This is a retrospective analysis of an institutional database including all adult CA patients admitted to the intensive care unit (ICU) between January 2007 and December 2014 who survived for at least 24 h. Demographic, CA-related data and ICU mortality were recorded as was lymphocyte count on admission and for the first 48 h. A cerebral performance category score of 3–5 at 3 months was considered as an unfavourable neurological outcome. Results Data from 377 patients were analysed (median age: 62 [IQRs: 52–75] years). Median time to return of spontaneous circulation (ROSC) was 15 [8–25] min and 232 (62%) had a non-shockable initial rhythm. ICU mortality was 58% (n = 217) and 246 (65%) patients had an unfavourable outcome at 3 months. The median lymphocyte count on admission was 1208 [700–2350]/mm3 and 151 (40%) patients had lymphopaenia (lymphocyte count <1000/mm3). Predictors of lymphopaenia on admission were older age, a shorter time to ROSC, prior use of corticosteroid therapy and high C-reactive protein levels on admission. ICU non-survivors had lower lymphocyte counts on admission than survivors (1100 [613–2317] vs. 1316 [891–2395]/mm3; p = 0.05) as did patients with unfavourable compared to those with favourable neurological outcomes (1100 [600–2013] vs. 1350 [919–2614]/mm3; p = 0.003). However, lymphopaenia on admission was not an independent predictor of poor outcomes in the entire population, but only among OHCA patients. Conclusions A low lymphocyte count is common in CA survivors and is associated with poor outcome after OHCA. Electronic supplementary material The online version of this article (doi:10.1186/s13613-017-0308-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola Villois
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - David Grimaldi
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Savino Spadaro
- Department of Morphological Surgery and Experimental Medicine, Arcispedale Sant'Anna, Università di Ferrara, Via AldoMoro, 8, 44121, Ferrara, Italy
| | - Claudia Righy Shinotsuka
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Vito Fontana
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Sabino Scolletta
- Department of Anesthesia and Intensive Care, Policlinico Santa Maria alle Scotte, Universitá di Siena, Viale Bracci, 14, 53100, Siena, Italy
| | - Federico Franchi
- Department of Anesthesia and Intensive Care, Policlinico Santa Maria alle Scotte, Universitá di Siena, Viale Bracci, 14, 53100, Siena, Italy
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Brussels, Belgium.
| |
Collapse
|
9
|
McGowan DC, Herschke F, Pauwels F, Stoops B, Smyej I, Last S, Pieters S, Embrechts W, Khamlichi MD, Thoné T, Van Schoubroeck B, Mostmans W, Wuyts D, Verstappen D, Scholliers A, De Pooter D, Dhuyvetter D, Borghys H, Tuefferd M, Arnoult E, Hong J, Fanning G, Bollekens J, Urmaliya V, Teisman A, Horton H, Jonckers THM, Raboisson P. Identification and Optimization of Pyrrolo[3,2-d]pyrimidine Toll-like Receptor 7 (TLR7) Selective Agonists for the Treatment of Hepatitis B. J Med Chem 2017; 60:6137-6151. [PMID: 28671847 DOI: 10.1021/acs.jmedchem.7b00365] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Pyrrolo[3,2-d]pyrimidines were identified as a new series of potent and selective TLR7 agonists. Compounds were optimized for their activity and selectivity over TLR8. This presents an advantage over recently described scaffolds that have residual TLR8 activity, which may be detrimental to the tolerability of the candidate drug. Oral administration of the lead compound 54 effectively induced a transient interferon stimulated gene (ISG) response in mice and cynomolgus monkeys. We aimed for a high first pass effect, limiting cytokine induction systemically, and demonstrated the potential for the immunotherapy of viral hepatitis.
Collapse
Affiliation(s)
- David C McGowan
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Florence Herschke
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Frederik Pauwels
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Bart Stoops
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Ilham Smyej
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Stefaan Last
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Serge Pieters
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Werner Embrechts
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Mourad Daoubi Khamlichi
- Villapharma Research S.L. , Parque Tecnológico de Fuente Álamo, Ctra. El Estrecho-Lobosillo, Km. 2.5-Av. Azul, 30320 Fuente Álamo de Murcia, Murcia, Spain
| | - Tine Thoné
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Wendy Mostmans
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Debbie Wuyts
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Dorien Verstappen
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Annick Scholliers
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Dorien De Pooter
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Herman Borghys
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marianne Tuefferd
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Eric Arnoult
- Janssen Research & Development L.L.C. , 1400 McKean Road, Spring House, Pennsylvania 19454, United States
| | - Jin Hong
- Alios Biopharma, Inc. , 260 East Grand Avenue, South San Francisco, California 94080, United States
| | - Gregory Fanning
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Jacques Bollekens
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Vijay Urmaliya
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Ard Teisman
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Helen Horton
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Tim H M Jonckers
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Pierre Raboisson
- Janssen Pharmaceutica , N. V. Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|