Skip to main content
Erschienen in: Critical Care 1/2023

Open Access 01.12.2023 | Research

Defining critical illness using immunological endotypes in patients with and without sepsis: a cohort study

verfasst von: Jeremy A. Balch, Uan-I Chen, Oliver Liesenfeld, Petr Starostik, Tyler J. Loftus, Philip A. Efron, Scott C. Brakenridge, Timothy E. Sweeney, Lyle L. Moldawer

Erschienen in: Critical Care | Ausgabe 1/2023

Abstract

Background

Sepsis is a heterogenous syndrome with limited therapeutic options. Identifying immunological endotypes through gene expression patterns in septic patients may lead to targeted interventions. We investigated whether patients admitted to a surgical intensive care unit (ICU) with sepsis and with high risk of mortality express similar endotypes to non-septic, but still critically ill patients using two multiplex transcriptomic metrics obtained both on admission to a surgical ICU and at set intervals.

Methods

We analyzed transcriptomic data from 522 patients in two single-site, prospective, observational cohorts admitted to surgical ICUs over a 5-year period ending in July 2020. Using an FDA-cleared analytical platform (nCounter FLEX®, NanoString, Inc.), we assessed a previously validated 29-messenger RNA transcriptomic classifier for likelihood of 30-day mortality (IMX-SEV-3) and a 33-messenger RNA transcriptomic endotype classifier. Clinical outcomes included all-cause mortality, development of chronic critical illness, and secondary infections. Univariate and multivariate analyses were performed to assess for true effect and confounding.

Results

Sepsis was associated with a significantly higher predicted and actual hospital mortality. At enrollment, the predominant endotype for both septic and non-septic patients was adaptive, though with significantly different distributions. Inflammopathic and coagulopathic septic patients, as well as inflammopathic non-septic patients, showed significantly higher frequencies of secondary infections compared to those with adaptive endotypes (p < 0.01). Endotypes changed during ICU hospitalization in 57.5% of patients. Patients who remained adaptive had overall better prognosis, while those who remained inflammopathic or coagulopathic had worse overall outcomes. For severity metrics, patients admitted with sepsis and a high predicted likelihood of mortality showed an inflammopathic (49.6%) endotype and had higher rates of cumulative adverse outcomes (67.4%). Patients at low mortality risk, whether septic or non-septic, almost uniformly presented with an adaptive endotype (100% and 93.4%, respectively).

Conclusion

Critically ill surgical patients express different and evolving immunological endotypes depending upon both their sepsis status and severity of their clinical course. Future studies will elucidate whether endotyping critically ill, septic patients can identify individuals for targeted therapeutic interventions to improve patient management and outcomes.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13054-023-04571-x.
Jeremy A. Balch and Uan-I Chen contributed equally.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALC
Absolute lymphocyte count
APACHE II
Acute physiology and chronic health evaluation
CCI
Chronic critical illness
ICU
Intensive care unit
SD
Standard deviation
SOFA
Sequential organ failure assessment
WBC
White blood cell
IL-6
Interleukin-6

Introduction

Sepsis remains one of the most common causes of mortality and morbidity in critically ill patients, affecting as many as 50 million individuals annually with case mortality rates as high as 40% [1]. Earlier recognition and near-universal implementation of sepsis protocols have improved in-hospital clinical outcomes; however, targeted therapies remain elusive [24].
Sepsis is defined as a dysregulated host immune response to infection resulting in life-threatening organ dysfunction [5, 6]. However, inherent to this definition is a wide range of insults and trajectories of physiologic decline. This disease heterogeneity likely explains the lack of efficacy in previous randomized controlled trials employing immune modulating therapeutics [711]. To address this heterogeneity, efforts have been made to classify patients based on constellations of observable characteristics and commonly available laboratory values, also called phenotypes [1215]. However, phenotypes based on these clinical variables may not accurately discriminate differences in the underlying disease mechanisms, also called endotypes. Thus, efforts at phenotyping have not led to substantial changes in patient care or outcomes [16, 17].
Multiplex metabolomics, proteomics, and transcriptomics offer the potential to reveal a spectrum of sepsis endotypes, both illuminating common underlying mechanisms for immunological dyscrasia and providing potential therapeutic targets. Semantically, we choose the term “endotype” to highlight subphenotypes with distinct functional or pathobiological mechanisms amenable to targeted interventions and to contrast against clinically observable phenotypes. While the present classification schema has not been proven to be linked to treatment effect, it does align with previous research by our group and others [7, 1820]. Previous research has identified 2–5 endotypes in diagnosed sepsis, though they vary with regards to domains, data sources, classification algorithms, statistical methodology, duration of observations, and stated goals [18, 2126]. Cumulatively, however, these studies have sparked interest in re-defining aspects of critical illness in terms of underlying physiologic perturbations rather than phenotypic syndromes [9, 27].
In this study, we apply 29- and 33-gene transcriptomic signatures to simultaneously classify severity and endotype, respectively, within both septic and non-septic critically ill patients [25, 2830]. These transcriptomic signatures were originally validated in non-surgical patients with bacterial or viral sepsis, and were classified into adaptative, inflammopathic, and coagulopathic endotypes based on gene ontology analysis [25]. We investigate whether patients admitted to a surgical ICU with sepsis and with high risk of mortality would express similar endotypes to non-septic, but still critically ill patients at-risk of developing sepsis. We hypothesize that patients admitted to a surgical ICU with sepsis and with high risk of mortality would express similar endotypes to non-septic, but still critically ill patients. In addition, we compare differences in endotype on admission between patients with predicted high severity by the transcriptomic metric versus the ground truth of those who clinically developed adverse outcomes. We also examine how these endotypes evolve over time in critically ill patients, tracking those who either rapidly recover versus those who experience adverse outcomes–defined as all-cause (in-hospital, 30-, 90-day) mortality, development or absence of chronic critical illness (CCI), secondary infections, and poor discharge disposition.

Materials and methods

Study designs

This post hoc study performed transcriptomic analyses on samples from two single-site, prospective, observational cohorts that enrolled a total of 522 patients admitted to non-cardiac, surgical ICUs and were classified as either (1) critically ill patients with a diagnosis of sepsis (septic) or (2) non-septic critically ill patients, at high risk of subsequently developing sepsis (at risk or non-septic; Fig. 1) [28, 29]. As a post hoc analysis, it was not powered for any specific outcome. Data and additional samples were obtained from the University of Florida CTSA Biorepository, a resource available to the scientific community [31]. In the first study (INF-05) [29], the parent cohort included 363 patients admitted to a surgical ICU between January 2015 and January 2020 with a diagnosis of sepsis (NCT02276417). Sepsis cohort inclusion criteria were: (1) age greater than or equal to 18 years, (2) clinical diagnosis of sepsis as defined by 2001 consensus guidelines, and (3) entrance into the electronic health record (EHR)-based sepsis clinical management protocol. Although prospectively enrolled using 2001 sepsis consensus criteria, these patients were retrospectively re-adjudicated and reclassified using Sepsis-3 consensus definitions [5, 32]. Detailed descriptions of the inclusion and exclusion criteria are contained in Additional file 1: Supplemental Materials: Methods.
The second prospective diagnostic and prognostic study (INF-06) was conducted between July 2020 and July 2021 [28] and enrolled two cohorts of critically ill patients at the time of surgical ICU admission (NCT04414189). Comparisons between INF-05 and INF-06 are shown in Additional file 1: Table S1. One cohort included patients with a suspected diagnosis of sepsis admitted to the ICU for protocolized sepsis management, as in the aforementioned study. Sepsis was defined according to Sepsis-3 criteria. The second cohort included critically ill patients without sepsis (e.g., severely injured trauma patients, post-operative patients, patients admitted directly to ICU from emergency department, see Additional file 1: Table S2). Inclusion and exclusion criteria, study design, and cohort flow are contained in Fig. 1 with a more detailed flow diagram shown in Additional file 1: Fig. S1.
In both studies, all enrolled subjects underwent post hoc adjudication by physician-investigators within one week of cohort enrollment to confirm sepsis diagnosis, severity, and source. Hospital-acquired secondary infections were adjudicated by physician-investigators during primary data/chart review utilizing current United States Centers for Disease Control definitions and guidelines [10].
Individual clinical outcome variables included all-cause (in-hospital, 30-, 90-day) mortality, development or absence of chronic critical illness (CCI), secondary infections, and poor discharge disposition. Inpatient clinical trajectory was defined as “early death,” “rapid recovery,” or “CCI.” CCI was defined as an ICU length of stay greater than or equal to 14 days with evidence of persistent organ dysfunction (SOFA score ≥ 2) [33, 34]. Hospitalized patients who died after an ICU length of stay greater than 14 days from the index hospitalization were also classified as CCI. Rapid recovery patients were those discharged from the ICU within 14 days following resolution of organ dysfunction. Poor disposition was defined as discharge to a skilled nursing facility, long-term acute care facility, or hospice. Our study was performed in accordance with the STROBE guidelines.

Sample collections

Blood samples were collected in PAXgene™ blood RNA tubes within 12–24 h of ICU admission and were stored at  − 80 °C for subsequent analysis. In the second study, additional blood samples were also collected on ICU days 4 and 7 and weekly thereafter during ICU stay (Additional file 1: Methods). RNA was extracted with the RNeasy® Plus Micro Kit (QIAGEN, Germantown, MD). The IMX-SEV-3 severity and the 33-mRNA endotyping classifiers were quantitated simultaneously from 200 ng of RNA input using the 510(k)-cleared NanoString nCounter FLEX™ profiler (NanoString, Seattle, WA) according to a validated standard operating protocol in a Clinical Laboratory Improvement Amendments-certified diagnostic laboratory (UF Health Medical Laboratories at Rocky Point, Gainesville, FL) by licensed laboratory technicians.

Severity and endotype classification

The probability of an adverse clinical outcome (in-hospital, 30-, and 90-day mortality, development of CCI and discharge disposition) was estimated by a 29 host-messenger RNA (mRNA) test (IMX-SEV-3, Inflammatix Inc., Sunnyvale, CA) that uses a machine learning algorithm to report results as both a continuous variable and stratified “risk bands” to meet clinically actionable performance thresholds: “low,” “moderate,” and “high” likelihood of 30-day mortality (see Additional file 1: Fig. S2) [35]. Severity classification was performed using supervised multi-layer perceptron (MLP) models as previously described [30].
Classification into three endotypes was computed from the whole blood expression of 33 host immune mRNAs using a previously published classifier [25, 28, 36]. These endotypes—adaptive, inflammopathic, and coagulopathic—were derived from the difference of geometric means of gene expression for each of three modules. The inflammopathic module comprises the expression of ARG1, LCN2, LTF, OLFM4, and HLA-DMB; the coagulopathic module comprises KCNMB4, CRISP2, HTRA1, PPL, RHBDF2, ZCCHC4, YKT6, DDX6, SENP5, RAPGEF1, DTX2, and RELB, and the adaptive module comprises YKT6, PDE4B, TWISTNB, BTN2A2, ZBTB33, PSMB9, CAMK4, TMEM19, SLC12A7, TP53BP1, PLEKHO1, SLC25A22, FRS2, GADD45A, CD24, S100A12, and STX1A expression. An overall endotype assignment for each subject was calculated using a 3-class logistic regression model which takes as input the three modules and generates a probability of endotype assignment {for each subject, the total probability [p(Inflammopathic) + p(Adaptive) + p(Coagulopathic)] sums to 1}. Each sample is assigned an endotype according to the highest probability. Numerical values are presented in the Additional file 1: Table S2 [25].
Total leukocyte and absolute lymphocyte counts (ALCs) were determined at the University of Florida Health Clinical and Diagnostic Laboratories. Plasma IL-6 levels were determined using the Luminex MagPix® platform (Austin, TX).

Statistical analysis

Descriptive data are presented as frequencies and percentages or means and standard deviations (SD). The Fisher exact test or Pearson's Chi-squared test and t-test were used for comparison of categorical and continuous variables, respectively. All significance tests were two sided, with a raw p ≤ 0.05 considered statistically significant. Significance levels 0.05 > p > 0.01 are reported precisely; 0.01 > p > 0.001 are reported as ‘p < 0.01,’ and lower values are all reported as ‘p < 0.001’. Univariable and multivariable logistic regressions were performed, controlling for age, sex, WBC, IL-6, SOFA, endotype, Charlson Comorbidity Index, and septic status. Analyses were performed using the R Project statistical package, version 4.2.0 (R Project for Statistical Computing).

Study approval

Ethics approvals were obtained from the University of Florida Institutional Review Board (IRB#201400611 and IRB#201702261). Informed consent was obtained from each subject or their surrogate decision-maker. Self-reported or proxy-reported race and ethnicity category data were collected as per National Institutes of Health reporting guidelines and requirements.

Results

Septic and non-septic cohorts

The overall analytic cohort consisted of 522 critically ill patients from the two consecutive, prospective observational studies (Fig. 1). Prediction of sepsis severity and endotype analyses were conducted on 377 septic and 145 non-septic patients within 24 h of ICU admission (Table 1). A subset of septic (N = 51) and all non-septic (N = 145) patients had repeat blood sampling at designated intervals over their ICU stay. Three hundred and twenty-six (86%) septic patients were drawn from the initial cohort (INF-05), and all non-septic patients were drawn from the second cohort (INF-06) [28, 29]. Demographics of included patients are shown in Table 1, while Table 2 shows outcomes, endotypes, and severity predictions of the two critically ill cohorts.
Table 1
Patient demographics at enrollment
Variable
At enrollment
p valueb
Septic (N = 377)a
Non-septic (N = 145)a
Age (yr)
58.9 (15.4)
57.4 (19.4)
0.39
Male
202 (53.6%)
92 (63.5%)
0.04
Race
 African American
38 (10.1%)
10 (7%)
0.02
 Asian
2 (0.5%)
0 (0%)
 
 Other
2 (0.5%)
6 (4.2%)
 
 White
333 (88.8%)
127 (88.8%)
 
 Missing
2
2
 
WBC (× 1000/mm3)
17.9 (8.5)
12.9 (5.5)
 < 0.001
 Missing
1
6
 
Neutrophils (%)
80.7 (12.9)
76.8 (15.3)
0.12
 Missing
30
102
 
Lymphocytes (%)
5.1 (4)
11.7 (8.1)
 < 0.001
 Missing
30
102
 
Lymphocytes (× 1000/mm3)
0.8 (0.5)
1.2 (0.8)
 < 0.001
 Missing
30
102
 
IL-6 (pg/mL)c
738.6 (1807)
148.7 (313.7)
 < 0.001
 Missing
3
2
 
SOFA Score
6 (4)
3 (3.2)
 < 0.001
 Missing
3
0
 
Charlson Comorbidity Index
3.2 (2.7)
2.7 (2.5)
0.03
 Missing
2
1
 
aMean (SD); n (%)
bWelch two-sample t-test; Pearson's Chi-squared test; Fisher's exact test
cvalues represent samples obtained within 24 h post enrollment
Table 2
Clinical outcomes, endotypes, and severity predictions
Variable
Septic (N = 377)a
Non-septic (N = 145)a
p valueb
Secondary infection
114 (30.2%)
12 (8.3%)
 < 0.001
CCI
122 (32.4%)
10 (6.9%)
 < 0.001
Adverse outcome
213 (56.7%)
40 (27.8%)
 < 0.001
 Missing
1
1
 
Poor discharge disposition
152 (40.4%)
23 (16%)
 < 0.001
 Missing
1
1
 
In-hospital mortality
28 (7.4%)
3 (2.1%)
0.02
30-day mortality
38 (10.2%)
6 (4.1%)
0.03
 Missing
4
0
 
90-day mortality
61 (16.8%)
8 (5.5%)
 < 0.001
 Missing
13
0
 
Endotype
  
 < 0.001
 Adaptive
151 (40.1%)
74 (51%)
 
 Coagulopathic
97 (25.7%)
48 (33.1%)
 
 Inflammopathic
129 (34.2%)
23 (15.9%)
 
IMX-SEV severity risk band
  
 < 0.001
 Low
38 (10.1%)
16 (11%)
 
 Moderate
250 (66.3%)
124 (85.5%)
 
 High
89 (23.6%)
5 (3.5%)
 
CCI chronic critical illness, adverse outcome is defined as cumulative incidence of in-hospital, 30-, and 90-day mortality, development of CCI, and poor discharge disposition
an (%)
bPearson's Chi-squared test
As expected, critically ill patients admitted to the ICU with sepsis had significantly higher SOFA and Charlson Comorbidity scores compared to the non-septic cohort, indicating more severe organ dysfunction and greater number of comorbidities. As shown in Table 2, poorer outcomes were observed among the septic cohort, including a higher incidence of secondary infection (30.2 vs. 8.3%, p < 0.001), development of CCI (32.4 vs. 6.9%, p < 0.001), poor discharge disposition (40.4 vs. 16.0%, p < 0.001), in-hospital mortality (7.4 vs. 2.1%, p = 0.02), 30-day (10.2 vs. 4.1%, p = 0.03), and 90-day (16.8 vs. 5.5%, p < 0.01) mortality.

Endotype distributions and outcomes

Endotype distributions were significantly different between septic and non-septic groups (Table 2). In both septic and non-septic cohorts, the adaptive endotype was most frequent, although it was more common in non-septic patients (40.1% vs. 51%). The inflammopathic endotype was second most common in septic patients and third in non-septic patients (34.2 vs. 15.9%) (Table 2). However, septic patients (n = 377) had different clinical outcomes depending upon their endotype at admission (Table 3). Inflammopathic and coagulopathic septic patients had a significantly higher frequency of secondary infections (37% each) compared to septic patients with an adaptive endotype (20%, p < 0.01). Similar increases in the frequency of secondary infections were seen in the inflammopathic non-septic patients (26%) versus patients with coagulopathic (4%) or adaptive (5%) endotypes (p < 0.01). Thirty-day mortality, CCI, and adverse discharge disposition did not reach statistical significance.
Table 3
Endotypes and outcomes on ICU admission
Variable
Septic (N = 377)
Non-septic (N = 145)
Adaptive (N = 151)a
Coagulopathic (N = 97)a
Inflammopathic (N = 129)a
p valueb
Adaptive (N = 74)a
Coagulopathic (N = 48)a
Inflammopathic (N = 23)a
p valueb
Secondary infection
30 (19.9%)
36 (37.1%)
48 (37.2%)
 < 0.01
4 (5.4%)
2 (4.2%)
6 (26.1%)
 < 0.01
CCI
40 (26.5%)
38 (39.2%)
44 (34.1%)
0.1
2 (2.7%)
2 (4.2%)
6 (26.1%)
 < 0.01
Adverse outcome
63 (41.7%)
63 (65.6%)
87 (67.4%)
 < 0.001
18 (24.7%)
10 (20.8%)
12 (52.2%)
0.02
 Missing
0
1
0
 
1
0
0
 
Poor discharge disposition
49 (32.5%)
42 (43.8%)
61 (47.3%)
0.03
12 (16.4%)
7 (14.6%)
4 (17.4%)
0.95
 Missing
0
1
0
 
1
0
0
 
In-hospital mortality
7 (4.6%)
6 (6.2%)
15 (11.6%)
0.07
2 (2.7%)
1 (2.1%)
0 (0%)
 > 0.99
30-day mortality
11 (7.4%)
9 (9.5%)
18 (14%)
0.19
4 (5.4%)
1 (2.1%)
1 (4.4%)
0.74
 Missing
2
2
0
     
90-day mortality
22 (15.2%)
16 (17%)
23 (18.4%)
0.78
5 (6.8%)
2 (4.2%)
1 (4.4%)
0.89
 Missing
6
3
4
     
IMX-SEV severity risk band
   
 < 0.001
   
 < 0.001
 Low
38 (25.2%)
0 (0%)
0 (0.00%)
 
15 (20.3%)
1 (2.1%)
0 (0%)
 
 Moderate
113 (74.8%)
72 (74.2%)
65 (50.4%)
 
59 (79.7%)
47 (97.9%)
18 (78.3%)
 
 High
0 (0.0%)
25 (25.8%)
64 (49.6%)
 
0 (0%)
0 (0%)
5 (21.7%)
 
CCI chronic critical illness, adverse outcome is defined as cumulative incidence of in-hospital, 30-, and 90-day mortality, development of CCI, and poor discharge disposition
aMean (SD); n (%)
bKruskal-Wallis rank sum test; Pearson's Chi-squared test; Fisher's exact test
To examine whether endotype at baseline is associated with different patient outcomes, a multivariable logistic regression was conducted by including endotypes and other clinically relevant factors into the model. Of interest, patients with inflammopathic (OR 2.4, 95% CI 1.4–4.1, p < 0.001) and coagulopathic endotypes (OR 1.9, 95% CI 1.1–3.1, p = 0.014) had higher odds of having an adverse outcome compared to those with the adaptive endotype (Additional file 1: Table S3 and Fig. S3).

Endotype transitions

Figure 2 illustrates endotype distributions and transitions over time until death or hospital discharge. Measurements for both the septic (n = 52, Group 1) and non-septic patients (n = 145, Group 2) were obtained only from the second cohort (INF-06). 61 patients had at least one missing value, with 20% of data missing secondary to declined blood draw and 7% due to inadequate samples, labeling errors, or staff unavailability. Endotypes changed in 57.5% of patients during their hospitalization; of the remaining, 19% remained adaptative, 4% inflammopathic, and 3% coagulopathic.
We assessed pooled outcome data between septic and non-septic patients obtained after the last collected endotype measurement (Table 4). Based on similar clinical behavior and worse overall clinical outcomes, we also pooled inflammopathic and coagulopathic endotypes. In most cases, the final endotype assessment was drawn on day 7 or 10 of ICU admission. There were significant differences in prognosis among the classes depending on endotype trajectory; patients who remained adaptive (N = 60) had the best nominal outcomes across all endpoints measured, though these were not individually tested. There were non-significant differences between those who transitioned either to or from adaptive endotype.
Table 4
Change in endotypes over time in ICU and subsequent outcomes
Variable
Endotype change
p valuee
Adaptive to adaptive (N = 60)a
Adaptive to I/C* (N = 24)b
I/C to adaptive (N = 38)c
I/C to I/C (N = 74)d
In-hospital mortality
0 (0%)
2 (8.3%)
1 (2.6%)
3 (4.1%)
0.15
30-day mortality
1 (1.7%)
4 (16.7%)
2 (5.3%)
7 (9.5%)
0.06
90-day mortality
2 (3.3%)
5 (20.8%)
3 (7.9%)
9 (12.2%)
0.07
CCI
1 (1.7%)
1 (4.2%)
5 (13.2%)
10 (13.5%)
0.04
Poor discharge disposition
7 (11.7%)
7 (30.4%)
8 (21.1%)
16 (21.9%)
0.2
 Missing
0
1
0
1
 
Total ICU LOS (days)
2 (1, 4)
6.5 (2, 8)
4 (2, 8)
5 (2, 11)
 < 0.001
 Missing
0
2
0
0
 
aFirst and last endotypes are both adaptive
bFirst endotype was adaptive; last endotype was inflammopathic or coagulopathic
cFirst endotype was inflammopathic or coagulopathic; last endotype was adaptive
dFirst and last endotypes are both inflammopathic and coagulopathic
eFisher’s exact test; Kruskal–Wallis rank sum test
*I/C Inflammopathic/coagulopathic, ICU intensive care unit, LOS length of stay

Endotypes and predicted severity

To better control for disease severity when comparing endotypes, we employed the severity transcriptomic metric (IMX-SEV-3) and found that endotypes were imbalanced across severity metrics (Fig. 3). Patients predicted to be low severity (n = 54), independent of their ICU admission cause, were near universally adaptive (98%): only one patient expressed a coagulopathic endotype while the patients with moderate severity prediction by IMX-SEV-3 continued to favor adaptive versus inflammopathic and coagulopathic endotypes (septic cohort: 45 vs. 26 vs. 29%, respectively; non-septic cohort: 48 vs. 15 vs. 38%, respectively). In contrast, those patients with high severity prediction based on IMX-SEV-3 were inflammopathic or coagulopathic in the septic (72% vs. 28%) and inflammopathic in the non-septic (100% vs. 0%) cohorts. We noted that inflammopathic (n = 69) and coagulopathic patients (n = 25) with a high risk of predicted mortality by IMX-SEV-3 appeared clinically similar, with nonsignificant differences in SOFA score, secondary infection, CCI, adverse outcomes, or mortality. The only noted difference was that inflammopathic patients demonstrated significantly higher plasma IL-6 concentrations than their coagulopathic counterparts (1870 vs. 642 pg/ml, p < 0.01; Additional file 1: Table S4).

Discussion

Key findings

This post hoc analysis of a septic and non-septic cohorts of critically ill surgical patients showed similar endotype profiles regardless of Sepsis-3 criteria, with an inflammopathic endotype on admission corresponding to high severity and worse outcomes by composite measure. Endotypes transitions occurred frequently during hospital admission. We found no statistically significant differences in mortality.

Context

Recent advances in sepsis endotyping research benefit from standard comparisons between studies, as advocated by De Merle et al. [7]. By observing endotypes in other septic and non-septic patients, we join efforts to redefine sepsis nosology as a heterogenous condition that shares characteristics across the spectrum of critical illness [9].
Our study shares commonalities and differences with others. The concept of expression-based sepsis subclasses dates to 2009 [37], though the last five years have witnessed an explosion of interest in this field: the MARS consortium investigated and validated patient endotypes in sepsis and identified four groups (MARS1-4) from the expression of 140 genes from 787 septic patients [24]; research by Davenport et al. [38] identified two distinct response signatures reflecting degree of immunosuppression in sepsis patients; and a study by Baghela et al. [18] validated five distinct gene expression profiles across several hospital systems, clustering patients into neutrophilic-suppressive, inflammatory, innate host defense, interferon, and adaptive. These authors assessed mortality using values drawn at a single time point within 24 h of admission and examined the biological plausibility of the identified genes known to cytokine signaling, cell proliferation, and lymphocyte and metabolic pathways, among others. Selected genes varied between studies, possibly due to differences in cohorts or in the classification techniques used to derive the groups [12, 25]. While these expression profiles carried prognostic significance, these groups did not examine changes in gene expression over the hospital course of illness, nor was there follow-up beyond 14 days.
Several studies, however, have analyzed gene expression profiles at different points during admission and following discharge. In patients expected to require at least 72 h of mechanical ventilation, a follow-up study of the PREVAIL trial assessed samples at days 1, 3, 6, 14, 21, and 28 to differentiate septic and non-septic patients using a novel scoring mechanism [23]. While they demonstrated changes in gene expression profiles through patient admission, they did not analyze outcomes. Similarly, Burnham et al. [39] showed that 46% of patients with community-acquired pneumonia and fecal peritonitis had changes to their gene expression profile on days 1, 3, and 5 of admission. Moreover, while patients who transitioned to the more critically ill group had nominally higher mortality rates, this is was not specifically analyzed. However, Cano-Gamez et al. [40], from the same institution, did demonstrate that patients with the largest decrease in genomic sepsis response had the lowest mortality rate. In a pediatric population, Wong et al. [41] showed that 42% of patients transitioned endotypes, and that those who remained in the more severe class had increased odds of mortality with administration of steroids. Finally, Kwok et al. [19] also examined expression patterns in convalescent samples 6 months after the septic event, finding persistent granulocytic dysfunction.

Current work

We were able to both identify and track changes in gene expression profile and severity scores over the course of acute critical illness in an adult, critically ill, surgical patient population. While admission endotype appeared to be the strongest predictor of outcomes, the extensive crossover noted between days 2 and 7 suggests that it may be valuable to continue assessing gene expression profiles, rather than focus on a single timepoint. We also noted that very few patients remained inflammatory or coagulopathic throughout their stay, suggesting a transitory maladaptation. This permits monitoring for resolution of immunologic dyscrasia, severity of condition, as well as possible responses to therapy.
Our study recapitulates observations about the 33-mRNA endotypes shown in previous investigations [22, 25, 42]. In contrast with previous investigations, however, we found that inflammopathic and coagulopathic patients had more similarities than differences, perhaps representing a single endotype. When pooled together, we showed that patients who presented with inflammopathic or coagulopathic endotype had increased incidence of adverse outcomes and secondary infections, though differences in in-hospital, 30-day, and 90-day mortality did not reach statistical significance.
In addition to analyzing septic patients, we also included a non-septic, critically ill cohort. While inflammopathic patterns had higher rates of secondary infections regardless of sepsis status, there were no overall changes in mortality or poor discharge disposition. Interestingly, both inflammopathic and coagulopathic subjects in the high-severity risk category had similar outcomes. These results may contribute to the understanding of sepsis as a part of a spectrum of critical illness rather than a separate entity.
Finally, this study applied the endotyping signature in a surgical cohort, while prior evaluations have mostly been in medical, bacterial sepsis, or COVID-19 patients [9, 18, 24, 37, 39, 41]. A recent report suggested the potential for endotypes to underpin different forms of critical illness [9]: a possibility that an ‘inflammopathic’ COVID-19 patient may be similar to an ‘inflammopathic’ surgical sepsis patient in molecular pathophysiology, further contributing to the idea of sepsis as a critical illness subtype.

Limitations

We note several limitations to our study. First, this study was performed at a single institution with a predominately Caucasian patient population and may lack generalizability. However, both the IMX-SEV-3 severity and the endotyping classifier have been validated multiple times in external hospitals with similar results [25, 36, 42]. Second, our non-septic cohort was broadly defined and with lower overall APACHE II scores. Age, gender, and Charlson comorbidity index were similar between the cohorts. However, when controlling for high-severity risk, we noted similar demographic and patient characteristics between the cohorts. Third, the majority (86%) of septic patients were derived from the initial cohort. These patients generally had higher SOFA scores and rates of CCI, with similar discharge disposition, complications, and mortality to the septic patients recruited in the second cohort. Fourth, as the first cohort was recruited from 2015 until 2020, there is the possibility of data drift, though standard of care for septic patients did not change during that period for our institution. Fifth, the multiple time series population contained only 196 patients, limiting our ability to draw conclusions based on trends and outcomes; and, as common to the literature in transcriptomics, this study is limited as a post-hoc analysis of an existing dataset and may not be powered for a specific outcome, though we have shown significance in several areas. Another important caveat is that with three endotypes, two cohorts, and multiple outcomes measures, we present numerous hypotheses in this manuscript, and we chose not to apply a multiple-hypothesis correction for ease of readership. Larger prospective studies are needed. Sixth, our findings regarding outcomes in final endotype measurements may not be representative of their endotype closer to the outcome measure, as day 10 measurements may have less impact on 30- and 90-day mortality. Finally, this paper did not seek to investigate the biological underpinnings of the mRNAs used in the two classifiers and their relation to pathophysiology; this has been done elsewhere [21, 25].

Future directions

Results from this study and others could assist in paving the way for personalization of sepsis treatment. By monitoring heterogenous, pathophysiologic responses to therapy, clinicians and researchers may be able to “divide and conquer” the sepsis syndrome and perhaps redefine sepsis along a spectrum of critical illness rather than as a separate entity. Current work into both immunosuppressant and immunostimulant therapies would benefit from targeting specific endotypes. The results of this study may be incorporated into randomized controlled trials or advanced causal analysis techniques employing observational data. From a prognostic standpoint, the conduct of similar endotyping on patients following discharge could also inform our clinical outreach efforts in diverting resources to those with greater follow-up needs.

Conclusion

Critically ill surgical patients with and without sepsis express different immunological endotypes. These endotypes are dynamic across a patient’s admission and are associated with distinct outcomes, and transitions between them may inform patient prognosis and care. Having identified differences among the patient groups using an endotyping classifier, future prospective studies are needed evaluate differences in therapeutic response between the classes.

Acknowledgements

The authors wish to acknowledge the staff of the Sepsis and Critical Illness Research Center and the UF Health Medical Laboratory at Rocky Point whose professional conduct of the two studies was essential to the success of the program. In addition, the authors wish to thank the patients and their families for their willingness to participate in this institutionally approved research with the hope that these findings could lead to improved outcomes in sepsis. We thank Purvesh Khatri, Ph.D. for his external review of the manuscript prior to publication. We appreciate expert statistical and scientific support from Drs. James Wacker and Florian Uhle.

Declarations

Ethics approval was obtained from the University of Florida Institutional Review Board (#201702261 and #201400611). Informed consent was obtained from each subject or their surrogate decision-maker. Self-reported or proxy-reported race and ethnicity category data were collected as per National Institutes of Health reporting guidelines and requirements.
Not applicable.

Competing interests

Dr. Balch reported receiving Grants from the National Institute of General Medical Sciences during the conduct of the study. Drs. Chen, Liesenfield, and Sweeney reported being an employee and stock option holder of Inflammatix Inc during the conduct of the study, and outside the submitted work. Dr. Starostik reported funding from the U.S. Army Medical Research Acquisition. Drs. Loftus and Efron reported receiving Grants from the National Institute of General Medical Sciences during the conduct of the study. Dr. Brakenridge reported receiving joint academic-industry Department of Health and Human Services, Biomedical Advanced Research and Development Authority (BARDA) funding from Inflammatix Inc during the conduct of the study. Dr. Sweeney reported receiving Grants from BARDA Division of Research, Innovation, and Ventures (DRIVe); being an employee and shareholder during the conduct of the study and outside the submitted work; and being the licensed inventor on several patents pending and issued that cover the IMX-SEV metric. Dr. Moldawer reported a subcontract on BARDA Grant to Inflammatix Inc during the conduct of the study and receiving grants from the National Institute of General Medical Sciences. No other disclosures were reported.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the global burden of disease study. Lancet. 2020;395(10219):200–11.PubMedPubMedCentralCrossRef Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the global burden of disease study. Lancet. 2020;395(10219):200–11.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87.PubMedCrossRef Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87.PubMedCrossRef
3.
Zurück zum Zitat Herrán-Monge R, Muriel-Bombín A, García-García MM, Merino-García PA, Cítores-González R, Fernández-Ratero JA, Albalá N, Carriedo D, Moradillo-González S, Álvarez-Martínez B, et al. mortality reduction and long-term compliance with surviving sepsis campaign: a nationwide multicenter study. Shock. 2016;45(6):598–606.PubMedCrossRef Herrán-Monge R, Muriel-Bombín A, García-García MM, Merino-García PA, Cítores-González R, Fernández-Ratero JA, Albalá N, Carriedo D, Moradillo-González S, Álvarez-Martínez B, et al. mortality reduction and long-term compliance with surviving sepsis campaign: a nationwide multicenter study. Shock. 2016;45(6):598–606.PubMedCrossRef
4.
Zurück zum Zitat van Zanten AR, Brinkman S, Arbous MS, Abu-Hanna A, Levy MM, de Keizer NF. Guideline bundles adherence and mortality in severe sepsis and septic shock. Crit Care Med. 2014;42(8):1890–8.PubMedCrossRef van Zanten AR, Brinkman S, Arbous MS, Abu-Hanna A, Levy MM, de Keizer NF. Guideline bundles adherence and mortality in severe sepsis and septic shock. Crit Care Med. 2014;42(8):1890–8.PubMedCrossRef
5.
Zurück zum Zitat Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, Rubenfeld G, Kahn JM, Shankar-Hari M, Singer M, et al. Assessment of clinical criteria for sepsis: for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):762–74.PubMedPubMedCentralCrossRef Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, Rubenfeld G, Kahn JM, Shankar-Hari M, Singer M, et al. Assessment of clinical criteria for sepsis: for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):762–74.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M. Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):775–87.PubMedPubMedCentralCrossRef Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M. Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):775–87.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat DeMerle KM, Angus DC, Baillie JK, Brant E, Calfee CS, Carcillo J, Chang CH, Dickson R, Evans I, Gordon AC, et al. Sepsis subclasses: a framework for development and interpretation. Crit Care Med. 2021;49(5):748–59.PubMedPubMedCentralCrossRef DeMerle KM, Angus DC, Baillie JK, Brant E, Calfee CS, Carcillo J, Chang CH, Dickson R, Evans I, Gordon AC, et al. Sepsis subclasses: a framework for development and interpretation. Crit Care Med. 2021;49(5):748–59.PubMedPubMedCentralCrossRef
8.
9.
Zurück zum Zitat Maslove DM, Tang B, Shankar-Hari M, Lawler PR, Angus DC, Baillie JK, Baron RM, Bauer M, Buchman TG, Calfee CS, et al. Redefining critical illness. Nat Med. 2022;28(6):1141–8.PubMedCrossRef Maslove DM, Tang B, Shankar-Hari M, Lawler PR, Angus DC, Baillie JK, Baron RM, Bauer M, Buchman TG, Calfee CS, et al. Redefining critical illness. Nat Med. 2022;28(6):1141–8.PubMedCrossRef
11.
Zurück zum Zitat Vincent JL, van der Poll T, Marshall JC. The end of “one size fits all” sepsis therapies: toward an individualized approach. Biomedicines. 2022;10:9.CrossRef Vincent JL, van der Poll T, Marshall JC. The end of “one size fits all” sepsis therapies: toward an individualized approach. Biomedicines. 2022;10:9.CrossRef
12.
Zurück zum Zitat Baek MS, Kim JH, Kwon YS. Cluster analysis integrating age and body temperature for mortality in patients with sepsis: a multicenter retrospective study. Sci Rep. 2022;12(1):1090.PubMedPubMedCentralCrossRef Baek MS, Kim JH, Kwon YS. Cluster analysis integrating age and body temperature for mortality in patients with sepsis: a multicenter retrospective study. Sci Rep. 2022;12(1):1090.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Xu Z, Mao C, Su C, Zhang H, Siempos I, Torres LK, Pan D, Luo Y, Schenck EJ, Wang F. Sepsis subphenotyping based on organ dysfunction trajectory. Crit Care. 2022;26(1):197.PubMedPubMedCentralCrossRef Xu Z, Mao C, Su C, Zhang H, Siempos I, Torres LK, Pan D, Luo Y, Schenck EJ, Wang F. Sepsis subphenotyping based on organ dysfunction trajectory. Crit Care. 2022;26(1):197.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Yehya N, Fitzgerald JC, Hayes K, Zhang D, Bush J, Koterba N, Chen F, Tuluc F, Teachey DT, Balamuth F, et al. Temperature trajectory sub-phenotypes and the immuno-inflammatory response in pediatric sepsis. Shock. 2022;57(5):645–51.PubMedPubMedCentralCrossRef Yehya N, Fitzgerald JC, Hayes K, Zhang D, Bush J, Koterba N, Chen F, Tuluc F, Teachey DT, Balamuth F, et al. Temperature trajectory sub-phenotypes and the immuno-inflammatory response in pediatric sepsis. Shock. 2022;57(5):645–51.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Zhao H, Kennedy JN, Wang S, Brant EB, Bernard GR, DeMerle K, Chang CH, Angus DC, Seymour CW. Revising host phenotypes of sepsis using microbiology. Front Med. 2021;8:775511.CrossRef Zhao H, Kennedy JN, Wang S, Brant EB, Bernard GR, DeMerle K, Chang CH, Angus DC, Seymour CW. Revising host phenotypes of sepsis using microbiology. Front Med. 2021;8:775511.CrossRef
16.
Zurück zum Zitat Seymour CW, Kennedy JN, Wang S, Chang CH, Elliott CF, Xu Z, Berry S, Clermont G, Cooper G, Gomez H, et al. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA. 2019;321(20):2003–17.PubMedPubMedCentralCrossRef Seymour CW, Kennedy JN, Wang S, Chang CH, Elliott CF, Xu Z, Berry S, Clermont G, Cooper G, Gomez H, et al. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA. 2019;321(20):2003–17.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Sweeney TE, Perumal TM, Henao R, Nichols M, Howrylak JA, Choi AM, Bermejo-Martin JF, Almansa R, Tamayo E, Davenport EE, et al. A community approach to mortality prediction in sepsis via gene expression analysis. Nat Commun. 2018;9(1):694.PubMedPubMedCentralCrossRef Sweeney TE, Perumal TM, Henao R, Nichols M, Howrylak JA, Choi AM, Bermejo-Martin JF, Almansa R, Tamayo E, Davenport EE, et al. A community approach to mortality prediction in sepsis via gene expression analysis. Nat Commun. 2018;9(1):694.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Baghela A, Pena OM, Lee AH, Baquir B, Falsafi R, An A, Farmer SW, Hurlburt A, Mondragon-Cardona A, Rivera JD, et al. Predicting sepsis severity at first clinical presentation: the role of endotypes and mechanistic signatures. EBioMedicine. 2022;75:103776.PubMedPubMedCentralCrossRef Baghela A, Pena OM, Lee AH, Baquir B, Falsafi R, An A, Farmer SW, Hurlburt A, Mondragon-Cardona A, Rivera JD, et al. Predicting sepsis severity at first clinical presentation: the role of endotypes and mechanistic signatures. EBioMedicine. 2022;75:103776.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Kwok AJ, Allcock A, Ferreira RC, Cano-Gamez E, Smee M, Burnham KL, Zurke YX, McKechnie S, Mentzer AJ, Monaco C, et al. Neutrophils and emergency granulopoiesis drive immune suppression and an extreme response endotype during sepsis. Nat Immunol. 2023;24(5):767–79.PubMedCrossRef Kwok AJ, Allcock A, Ferreira RC, Cano-Gamez E, Smee M, Burnham KL, Zurke YX, McKechnie S, Mentzer AJ, Monaco C, et al. Neutrophils and emergency granulopoiesis drive immune suppression and an extreme response endotype during sepsis. Nat Immunol. 2023;24(5):767–79.PubMedCrossRef
21.
Zurück zum Zitat He YD, Wohlford EM, Uhle F, Buturovic L, Liesenfeld O, Sweeney TE. The optimization and biological significance of a 29-host-immune-mRNA panel for the diagnosis of acute infections and sepsis. J Pers Med. 2021;11:8.CrossRef He YD, Wohlford EM, Uhle F, Buturovic L, Liesenfeld O, Sweeney TE. The optimization and biological significance of a 29-host-immune-mRNA panel for the diagnosis of acute infections and sepsis. J Pers Med. 2021;11:8.CrossRef
22.
Zurück zum Zitat Iglesias J, Vassallo AV, Liesenfeld O, Levine JS, Patel VV, Sullivan JB, Cavanaugh JB, Elbaga Y, Sweeney TE. A 33-mRNA classifier is able to produce inflammopathic, adaptive, and coagulopathic endotypes with prognostic significance: the outcomes of metabolic resuscitation using ascorbic acid, thiamine, and glucocorticoids in the early treatment of sepsis (ORANGES) trial. J Pers Med. 2020;11:1.CrossRef Iglesias J, Vassallo AV, Liesenfeld O, Levine JS, Patel VV, Sullivan JB, Cavanaugh JB, Elbaga Y, Sweeney TE. A 33-mRNA classifier is able to produce inflammopathic, adaptive, and coagulopathic endotypes with prognostic significance: the outcomes of metabolic resuscitation using ascorbic acid, thiamine, and glucocorticoids in the early treatment of sepsis (ORANGES) trial. J Pers Med. 2020;11:1.CrossRef
23.
Zurück zum Zitat Maslove DM, Shapira T, Tyryshkin K, Veldhoen RA, Marshall JC, Muscedere J. Validation of diagnostic gene sets to identify critically ill patients with sepsis. J Crit Care. 2019;49:92–8.PubMedCrossRef Maslove DM, Shapira T, Tyryshkin K, Veldhoen RA, Marshall JC, Muscedere J. Validation of diagnostic gene sets to identify critically ill patients with sepsis. J Crit Care. 2019;49:92–8.PubMedCrossRef
24.
Zurück zum Zitat Scicluna BP, van Vught LA, Zwinderman AH, Wiewel MA, Davenport EE, Burnham KL, Nürnberg P, Schultz MJ, Horn J, Cremer OL, et al. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir Med. 2017;5(10):816–26.PubMedCrossRef Scicluna BP, van Vught LA, Zwinderman AH, Wiewel MA, Davenport EE, Burnham KL, Nürnberg P, Schultz MJ, Horn J, Cremer OL, et al. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir Med. 2017;5(10):816–26.PubMedCrossRef
25.
Zurück zum Zitat Sweeney TE, Azad TD, Donato M, Haynes WA, Perumal TM, Henao R, Bermejo-Martin JF, Almansa R, Tamayo E, Howrylak JA, et al. Unsupervised analysis of transcriptomics in bacterial sepsis across multiple datasets reveals three robust clusters. Crit Care Med. 2018;46(6):915–25.PubMedPubMedCentralCrossRef Sweeney TE, Azad TD, Donato M, Haynes WA, Perumal TM, Henao R, Bermejo-Martin JF, Almansa R, Tamayo E, Howrylak JA, et al. Unsupervised analysis of transcriptomics in bacterial sepsis across multiple datasets reveals three robust clusters. Crit Care Med. 2018;46(6):915–25.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Bodinier M, Monneret G, Casimir M, Fleurie A, Conti F, Venet F, Cazalis MA, Cerrato E, Peronnet E, Rimmelé T, et al. Identification of a sub-group of critically ill patients with high risk of intensive care unit-acquired infections and poor clinical course using a transcriptomic score. Crit Care. 2023;27(1):158.PubMedPubMedCentralCrossRef Bodinier M, Monneret G, Casimir M, Fleurie A, Conti F, Venet F, Cazalis MA, Cerrato E, Peronnet E, Rimmelé T, et al. Identification of a sub-group of critically ill patients with high risk of intensive care unit-acquired infections and poor clinical course using a transcriptomic score. Crit Care. 2023;27(1):158.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Heijnen NFL, Hagens LA, Smit MR, Cremer OL, Ong DSY, van der Poll T, van Vught LA, Scicluna BP, Schnabel RM, van der Horst ICC, et al. Biological subphenotypes of acute respiratory distress syndrome show prognostic enrichment in mechanically ventilated patients without acute respiratory distress syndrome. Am J Respir Crit Care Med. 2021;203(12):1503–11.PubMedCrossRef Heijnen NFL, Hagens LA, Smit MR, Cremer OL, Ong DSY, van der Poll T, van Vught LA, Scicluna BP, Schnabel RM, van der Horst ICC, et al. Biological subphenotypes of acute respiratory distress syndrome show prognostic enrichment in mechanically ventilated patients without acute respiratory distress syndrome. Am J Respir Crit Care Med. 2021;203(12):1503–11.PubMedCrossRef
28.
Zurück zum Zitat Brakenridge SC, Chen UI, Loftus T, Ungaro R, Dirain M, Kerr A, Zhong L, Bacher R, Starostik P, Ghita G, et al. Evaluation of a multivalent transcriptomic metric for diagnosing surgical sepsis and estimating mortality among critically Ill patients. JAMA Netw Open. 2022;5(7):e2221520.PubMedPubMedCentralCrossRef Brakenridge SC, Chen UI, Loftus T, Ungaro R, Dirain M, Kerr A, Zhong L, Bacher R, Starostik P, Ghita G, et al. Evaluation of a multivalent transcriptomic metric for diagnosing surgical sepsis and estimating mortality among critically Ill patients. JAMA Netw Open. 2022;5(7):e2221520.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Brakenridge SC, Starostik P, Ghita G, Midic U, Darden D, Fenner B, Wacker J, Efron PA, Liesenfeld O, Sweeney TE, et al. A transcriptomic severity metric that predicts clinical outcomes in critically Ill surgical sepsis patients. Crit Care Explor. 2021;3(10):e0554.PubMedPubMedCentralCrossRef Brakenridge SC, Starostik P, Ghita G, Midic U, Darden D, Fenner B, Wacker J, Efron PA, Liesenfeld O, Sweeney TE, et al. A transcriptomic severity metric that predicts clinical outcomes in critically Ill surgical sepsis patients. Crit Care Explor. 2021;3(10):e0554.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Mayhew MB, Buturovic L, Luethy R, Midic U, Moore AR, Roque JA, Shaller BD, Asuni T, Rawling D, Remmel M, et al. A generalizable 29-mRNA neural-network classifier for acute bacterial and viral infections. Nat Commun. 2020;11(1):1177.PubMedPubMedCentralCrossRef Mayhew MB, Buturovic L, Luethy R, Midic U, Moore AR, Roque JA, Shaller BD, Asuni T, Rawling D, Remmel M, et al. A generalizable 29-mRNA neural-network classifier for acute bacterial and viral infections. Nat Commun. 2020;11(1):1177.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G. 2001 SCCM/ESICM/ACCP/ATS/SIS international sepsis definitions conference. Crit Care Med. 2003;31(4):1250–6.PubMedCrossRef Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, Cohen J, Opal SM, Vincent JL, Ramsay G. 2001 SCCM/ESICM/ACCP/ATS/SIS international sepsis definitions conference. Crit Care Med. 2003;31(4):1250–6.PubMedCrossRef
33.
Zurück zum Zitat Stortz JA, Cox MC, Hawkins RB, Ghita GL, Brumback BA, Mohr AM, Moldawer LL, Efron PA, Brakenridge SC, Moore FA. Phenotypic heterogeneity by site of infection in surgical sepsis: a prospective longitudinal study. Crit Care. 2020;24(1):203.PubMedPubMedCentralCrossRef Stortz JA, Cox MC, Hawkins RB, Ghita GL, Brumback BA, Mohr AM, Moldawer LL, Efron PA, Brakenridge SC, Moore FA. Phenotypic heterogeneity by site of infection in surgical sepsis: a prospective longitudinal study. Crit Care. 2020;24(1):203.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Stortz JA, Mira JC, Raymond SL, Loftus TJ, Ozrazgat-Baslanti T, Wang Z, Ghita GL, Leeuwenburgh C, Segal MS, Bihorac A, et al. Benchmarking clinical outcomes and the immunocatabolic phenotype of chronic critical illness after sepsis in surgical intensive care unit patients. J Trauma Acute Care Surg. 2018;84(2):342–9.PubMedPubMedCentralCrossRef Stortz JA, Mira JC, Raymond SL, Loftus TJ, Ozrazgat-Baslanti T, Wang Z, Ghita GL, Leeuwenburgh C, Segal MS, Bihorac A, et al. Benchmarking clinical outcomes and the immunocatabolic phenotype of chronic critical illness after sepsis in surgical intensive care unit patients. J Trauma Acute Care Surg. 2018;84(2):342–9.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Ducharme J, Self WH, Osborn TM, Ledeboer NA, Romanowsky J, Sweeney TE, Liesenfeld O, Rothman RE. A Multi-mRNA host-response molecular blood test for the diagnosis and prognosis of acute infections and sepsis: proceedings from a clinical advisory panel. J Pers Med. 2020;10:4.CrossRef Ducharme J, Self WH, Osborn TM, Ledeboer NA, Romanowsky J, Sweeney TE, Liesenfeld O, Rothman RE. A Multi-mRNA host-response molecular blood test for the diagnosis and prognosis of acute infections and sepsis: proceedings from a clinical advisory panel. J Pers Med. 2020;10:4.CrossRef
36.
Zurück zum Zitat Galtung N, Diehl-Wiesenecker E, Lehmann D, Markmann N, Bergström WH, Wacker J, Liesenfeld O, Mayhew M, Buturovic L, Luethy R, et al. Prospective validation of a transcriptomic severity classifier among patients with suspected acute infection and sepsis in the emergency department. Eur J Emerg Med. 2022;29(5):357–65.PubMedPubMedCentralCrossRef Galtung N, Diehl-Wiesenecker E, Lehmann D, Markmann N, Bergström WH, Wacker J, Liesenfeld O, Mayhew M, Buturovic L, Luethy R, et al. Prospective validation of a transcriptomic severity classifier among patients with suspected acute infection and sepsis in the emergency department. Eur J Emerg Med. 2022;29(5):357–65.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Wong HR, Wheeler DS, Tegtmeyer K, Poynter SE, Kaplan JM, Chima RS, Stalets E, Basu RK, Doughty LA. Toward a clinically feasible gene expression-based subclassification strategy for septic shock: proof of concept. Crit Care Med. 2010;38(10):1955–61.PubMedPubMedCentralCrossRef Wong HR, Wheeler DS, Tegtmeyer K, Poynter SE, Kaplan JM, Chima RS, Stalets E, Basu RK, Doughty LA. Toward a clinically feasible gene expression-based subclassification strategy for septic shock: proof of concept. Crit Care Med. 2010;38(10):1955–61.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Davenport EE, Burnham KL, Radhakrishnan J, Humburg P, Hutton P, Mills TC, Rautanen A, Gordon AC, Garrard C, Hill AV, et al. Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir Med. 2016;4(4):259–71.PubMedPubMedCentralCrossRef Davenport EE, Burnham KL, Radhakrishnan J, Humburg P, Hutton P, Mills TC, Rautanen A, Gordon AC, Garrard C, Hill AV, et al. Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir Med. 2016;4(4):259–71.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Burnham KL, Davenport EE, Radhakrishnan J, Humburg P, Gordon AC, Hutton P, Svoren-Jabalera E, Garrard C, Hill AVS, Hinds CJ, et al. Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia. Am J Respir Crit Care Med. 2017;196(3):328–39.PubMedPubMedCentralCrossRef Burnham KL, Davenport EE, Radhakrishnan J, Humburg P, Gordon AC, Hutton P, Svoren-Jabalera E, Garrard C, Hill AVS, Hinds CJ, et al. Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia. Am J Respir Crit Care Med. 2017;196(3):328–39.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Cano-Gamez E, Burnham KL, Goh C, Allcock A, Malick ZH, Overend L, Kwok A, Smith DA, Peters-Sengers H, Antcliffe D, et al. An immune dysfunction score for stratification of patients with acute infection based on whole-blood gene expression. Sci Transl Med. 2022;14(669):eabq44339.CrossRef Cano-Gamez E, Burnham KL, Goh C, Allcock A, Malick ZH, Overend L, Kwok A, Smith DA, Peters-Sengers H, Antcliffe D, et al. An immune dysfunction score for stratification of patients with acute infection based on whole-blood gene expression. Sci Transl Med. 2022;14(669):eabq44339.CrossRef
41.
Zurück zum Zitat Wong HR, Cvijanovich NZ, Anas N, Allen GL, Thomas NJ, Bigham MT, Weiss SL, Fitzgerald JC, Checchia PA, Meyer K, et al. Endotype transitions during the acute phase of pediatric septic shock reflect changing risk and treatment response. Crit Care Med. 2018;46(3):e242–9.PubMedPubMedCentralCrossRef Wong HR, Cvijanovich NZ, Anas N, Allen GL, Thomas NJ, Bigham MT, Weiss SL, Fitzgerald JC, Checchia PA, Meyer K, et al. Endotype transitions during the acute phase of pediatric septic shock reflect changing risk and treatment response. Crit Care Med. 2018;46(3):e242–9.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Sweeney TE, Liesenfeld O, Wacker J, He YD, Rawling D, Remmel M, Coyle S, Midic U, Kotsaki A, Kanavou A, et al. Validation of inflammopathic, adaptive, and coagulopathic sepsis endotypes in coronavirus disease 2019. Crit Care Med. 2021;49(2):e170–8.PubMedCrossRef Sweeney TE, Liesenfeld O, Wacker J, He YD, Rawling D, Remmel M, Coyle S, Midic U, Kotsaki A, Kanavou A, et al. Validation of inflammopathic, adaptive, and coagulopathic sepsis endotypes in coronavirus disease 2019. Crit Care Med. 2021;49(2):e170–8.PubMedCrossRef
Metadaten
Titel
Defining critical illness using immunological endotypes in patients with and without sepsis: a cohort study
verfasst von
Jeremy A. Balch
Uan-I Chen
Oliver Liesenfeld
Petr Starostik
Tyler J. Loftus
Philip A. Efron
Scott C. Brakenridge
Timothy E. Sweeney
Lyle L. Moldawer
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2023
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-023-04571-x

Weitere Artikel der Ausgabe 1/2023

Critical Care 1/2023 Zur Ausgabe

Blutdrucksenkung schon im Rettungswagen bei akutem Schlaganfall?

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.