A Molecular Host Response Assay to Discriminate Between Sepsis and Infection-Negative Systemic Inflammation in Critically Ill Patients: Discovery and Validation in Independent Cohorts

Leo McHugh, Therese A Seldon, Roslyn A Brandon, James T Kirk, Antony Rapisarda, Allison J Sutherland, Jeffrey J Presneill, Deon J Venter, Jeffrey Lipman, Mervyn R Thomas, Peter M C Klein Klouwenberg, Lonneke van Vught, Brendon Scicluna, Marc Bonten, Olaf L Cremer, Marcus J Schultz, Tom van der Poll, Thomas D Yager, Richard B Brandon, Leo McHugh, Therese A Seldon, Roslyn A Brandon, James T Kirk, Antony Rapisarda, Allison J Sutherland, Jeffrey J Presneill, Deon J Venter, Jeffrey Lipman, Mervyn R Thomas, Peter M C Klein Klouwenberg, Lonneke van Vught, Brendon Scicluna, Marc Bonten, Olaf L Cremer, Marcus J Schultz, Tom van der Poll, Thomas D Yager, Richard B Brandon

Abstract

Background: Systemic inflammation is a whole body reaction having an infection-positive (i.e., sepsis) or infection-negative origin. It is important to distinguish between these two etiologies early and accurately because this has significant therapeutic implications for critically ill patients. We hypothesized that a molecular classifier based on peripheral blood RNAs could be discovered that would (1) determine which patients with systemic inflammation had sepsis, (2) be robust across independent patient cohorts, (3) be insensitive to disease severity, and (4) provide diagnostic utility. The goal of this study was to identify and validate such a molecular classifier.

Methods and findings: We conducted an observational, non-interventional study of adult patients recruited from tertiary intensive care units (ICUs). Biomarker discovery utilized an Australian cohort (n = 105) consisting of 74 cases (sepsis patients) and 31 controls (post-surgical patients with infection-negative systemic inflammation) recruited at five tertiary care settings in Brisbane, Australia, from June 3, 2008, to December 22, 2011. A four-gene classifier combining CEACAM4, LAMP1, PLA2G7, and PLAC8 RNA biomarkers was identified. This classifier, designated SeptiCyte Lab, was validated using reverse transcription quantitative PCR and receiver operating characteristic (ROC) curve analysis in five cohorts (n = 345) from the Netherlands. Patients for validation were selected from the Molecular Diagnosis and Risk Stratification of Sepsis study (ClinicalTrials.gov, NCT01905033), which recruited ICU patients from the Academic Medical Center in Amsterdam and the University Medical Center Utrecht. Patients recruited from November 30, 2012, to August 5, 2013, were eligible for inclusion in the present study. Validation cohort 1 (n = 59) consisted entirely of unambiguous cases and controls; SeptiCyte Lab gave an area under curve (AUC) of 0.95 (95% CI 0.91-1.00) in this cohort. ROC curve analysis of an independent, more heterogeneous group of patients (validation cohorts 2-5; 249 patients after excluding 37 patients with an infection likelihood of "possible") gave an AUC of 0.89 (95% CI 0.85-0.93). Disease severity, as measured by Sequential Organ Failure Assessment (SOFA) score or Acute Physiology and Chronic Health Evaluation (APACHE) IV score, was not a significant confounding variable. The diagnostic utility of SeptiCyte Lab was evaluated by comparison to various clinical and laboratory parameters available to a clinician within 24 h of ICU admission. SeptiCyte Lab was significantly better at differentiating cases from controls than all tested parameters, both singly and in various logistic combinations, and more than halved the diagnostic error rate compared to procalcitonin in all tested cohorts and cohort combinations. Limitations of this study relate to (1) cohort compositions that do not perfectly reflect the composition of the intended use population, (2) potential biases that could be introduced as a result of the current lack of a gold standard for diagnosing sepsis, and (3) lack of a complete, unbiased comparison to C-reactive protein.

Conclusions: SeptiCyte Lab is a rapid molecular assay that may be clinically useful in managing ICU patients with systemic inflammation. Further study in population-based cohorts is needed to validate this assay for clinical use.

Conflict of interest statement

The authors have read the journal’s policy, and declare the following: AJS JL PKK LVV BS MB OC MJS TVDP have no competing interests; TAS LM AR JTK TDY RAB RBB DJV MRT are employees and/or shareholders of Immunexpress, which holds the intellectual property in SeptiCyte Lab; JJP has no competing interests except for this: "In 2010 Immunexpress Inc. paid to the Adult Intensive Care Unit, Mater Health Services, Brisbane, Australia a sum of AUS $38,509 (thirty-eight thousand, five hundred and nine Australian dollars) as half-time temporary salary support for an ICU research coordinator to facilitate patient identification, recruitment and sample collection for 6 months from 20 May 2010.”

Figures

Fig 1. Flow diagram for selection of…
Fig 1. Flow diagram for selection of patients composing the validation cohorts.
From the MARS study, only patients admitted to the ICU between the dates of November 30, 2012, and August 5, 2013, were eligible for possible inclusion in the present study. A detailed description of the inclusion and exclusion criteria, and the classification algorithm, is given in S2 Text. In the last step of the cohort selection process as described in this figure, 14 patients were reassigned as controls because the attending physicians had retrospectively adjudicated the patients to have an infection likelihood of none for their sepsis events. S.I., systemic inflammation.
Fig 2. Analysis of behavior of PLAC8…
Fig 2. Analysis of behavior of PLAC8, LAMP1, PLA2G7, and CEACAM4 in the discovery cohort.
(A) Heat map representation of the discovery cohort (74 cases, 31 controls). Normalized expression levels of the individual genes comprising the SeptiCyte Lab classifier (color) are plotted versus disease status (dendrogram position) using unsupervised clustering with equally weighted Euclidean distance. The normalization scale (expression level Z-score) for up-regulation (red) or down-regulation (green) is shown in the insert at the left of the heat map. (The Z-score is the number of standard deviations a value lies away from the mean. Higher absolute Z-scores correspond to lower p-values. A Z-score of ±1.96 equates to a p-value of 0.05 in a two-tailed test.) In the cases (sepsis), two genes are predominately up-regulated (PLAC8 and LAMP1), whilst two are predominantly down regulated (PLA2G7 and CEACAM4). (B) Scatterplot representation of microarray expression levels for individual genes in the SeptiCyte Lab classifier, for the discovery cohort. The expression level on log2 scale (y-axis) is presented for PLAC8, CEACAM4, LAMP1, and PLA2G7 in individual patients (red for cases, black for controls). Each gene contributes to the ability of the SeptiCyte Lab classifier to separate the cases and controls. S.I., systemic inflammation.
Fig 3. Performance of SeptiCyte Lab in…
Fig 3. Performance of SeptiCyte Lab in validation cohort 1.
Left: Scatterplot of SeptiScores for 24 cases (sepsis; red) versus 35 controls (infection-negative systemic inflammation [S.I.]; black). The blue dashed lines denote SeptiScore values of 3.1, 4.0, 6.0, and 9.0, which are used for subsequent calculations. Right: ROC curve. The grey shading denotes the 95% confidence area.
Fig 4. Performance of SeptiCyte Lab in…
Fig 4. Performance of SeptiCyte Lab in validation cohorts 3, 4, and 5.
In each panel, the left side presents a scatterplot of SeptiCyte Lab scores for cases (sepsis; red), controls (infection-negative systemic inflammation [S.I.]; black), and individuals with an infection likelihood of possible (green), and the right side presents the corresponding ROC curve. In the scatterplots, the blue dashed lines denote SeptiScore values of 3.1, 4.0, 6.0, and 9.0, which are used for subsequent calculations. On the right, the grey shading denotes the 95% confidence area for the ROC curve. Patients with an infection likelihood of possible have been excluded from the calculation of ROC curves. (A) Validation cohort 3 (29 cases, 77 controls). (B) Validation cohort 4 (20 cases, 47 controls), consisting of patients sequentially admitted to the ICU. (C) Validation cohort 5 (21 cases, 25 controls), consisting of black and Asian patients sequentially admitted to the ICU.
Fig 5. Test for disease severity as…
Fig 5. Test for disease severity as a potential confounding variable.
Validation cohorts 1–5 (excluding patients with an infection likelihood of possible) were combined, and then stratified on APACHE IV score or SOFA score. (A) ROC curve analysis was performed on the combined dataset. (B) Separate ROC curve analyses were performed over sub-ranges of the APACHE IV score (left) or SOFA score (right). The error bars indicate the 95% CI for the AUC as estimated by resampling. The results for the various strata of APACHE IV and SOFA score indicate that disease severity has a relatively weak influence (AUC = 0.52–0.65) on the discrimination of cases from controls, while SeptiCyte Lab has a much stronger influence (AUC = 0.87–0.91). Additional details of the calculations are given in S5 Data.
Fig 6. Performance of PCT, clinical parameters,…
Fig 6. Performance of PCT, clinical parameters, and SeptiCyte Lab for the discrimination of sepsis versus infection-negative systemic inflammation.
This comparative analysis used the largest set of patients (n = 157) for which values were available for all comparisons. Patients having an infection likelihood of possible were excluded. Performance is reported as AUC. The error bars indicate the 95% CI for the AUC, obtained from a 50 × 2 cross-validation with training and test samples selected in a 1:1 ratio. The significance levels (p-value by t-test) for pairwise comparisons to SeptiCyte Lab are indicated in the figure. Details of the analysis are given in S7 Data. NS, not significant; Top 3, top three clinical parameters; Top 5, top five clinical parameters.

References

    1. Hsu AA, Fenton K, Weinstein S, Carpenter J, Dalton H, Bell MJ. Neurological injury markers in children with septic shock. Pediatr Crit Care Med. 2008;9:245–251. 10.1097/PCC.0b013e3181727b22
    1. Winters BD, Eberlein M, Leung J, Needham DM, Pronovost PJ, Sevransky JE. Long-term mortality and quality of life in sepsis: a systematic review. Crit Care Med. 2010;38:1276–1283. 10.1097/CCM.0b013e3181d8cc1d
    1. Bauer M, Reinhart K. Molecular diagnostics of sepsis—where are we today? Int J Med Microbiol. 2010;300:411–413. 10.1016/j.ijmm.2010.04.006
    1. Calandra T, Cohen J. The International Sepsis Forum consensus conference on definitions of infection in the intensive care unit. Crit Care Med. 2005;33:1538–1548.
    1. Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock. Crit Care Med. 2008;36:296–327.
    1. Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2012. Intensive Care Med. 2013;39:165–228. 10.1007/s00134-012-2769-8
    1. Coburn B, Morris AM, Tomlinson G, Detsky AS. Does this adult patient with suspected bacteremia require blood cultures? JAMA 2012;308:502–511. 10.1001/jama.2012.8262
    1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369:840–851. 10.1056/NEJMra1208623
    1. Forner L, Larsen T, Kilian M, Holmstrup P. Incidence of bacteremia after chewing, tooth brushing and scaling in individuals with periodontal inflammation. J Clin Periodontol. 2006;33:401–407.
    1. Lamy B, Roy P, Carret G, Flandrois J- P, Delignette-Muller ML. What is the relevance of obtaining multiple blood samples for culture? A comprehensive model to optimize the strategy for diagnosing bacteremia. Clin Infect Dis. 2002;35:842–850.
    1. Aronson MD, Bor DH. Blood cultures. Ann Intern Med. 1987;106:246–253.
    1. Uzzan B, Cohen R, Nicolas P, Cucherat M, Perret G-Y. Procalcitonin as a diagnostic test for sepsis in critically ill adults and after surgery or trauma: a systematic review and meta-analysis. Crit Care Med. 2006;34:1996–2003.
    1. Meynaar IA, Droog W, Batstra M, Vreede R, Herbrink P. In critically ill patients, serum procalcitonin is more useful in differentiating between sepsis and SIRS than CRP, Il-6, or LBP. Crit Care Res Pract. 2011;2011:594645 10.1155/2011/594645
    1. Tang BM, McLean AS, Dawes IW, Huang SJ, Lin RC. The use of gene-expression profiling to identify candidate genes in human sepsis. Am J Respir Crit Care Med. 2007;176:676–684.
    1. Cruciani M. Meta-analyses of diagnostic tests in infectious diseases: how helpful are they in the intensive care setting? HSR Proc Intensive Care Cardiovasc Anesth. 2011;3:103–108.
    1. Hoenigl M, Raggam RB, Wagner J, Prueller F, Grisold AJ, Leitner E, et al. Procalcitonin fails to predict bacteremia in SIRS patients: a cohort study. Int J Clin Pract. 2014;68:1278–1281. 10.1111/ijcp.12474
    1. Reinhart K, Bauer M, Riedemann NC, Hartog CS. New approaches to sepsis: molecular diagnostics and biomarkers. Clin Microbiol Rev. 2012;25:609–634. 10.1128/CMR.00016-12
    1. Shapiro NI, Trzeciak S, Hollander JE, Birkhahn R, Otero R, Osborn TM, et al. A prospective, multicenter derivation of a biomarker panel to assess risk of organ dysfunction, shock, and death in emergency department patients with suspected sepsis. Crit Care Med. 2009;37:96–104. 10.1097/CCM.0b013e318192fd9d
    1. Gibot S, Bene MC, Noel R, Massin F, Guy J, Cravoisy A, et al. Combination biomarkers to diagnose sepsis in the critically ill patient. Am J Resp Crit Care Med. 2012;186:65–71. 10.1164/rccm.201201-0037OC
    1. Venter D, Thomas M, Lipman J, Tang B, McLean A, Pascoe R, et al. A novel molecular biomarker diagnostic for the early detection of sepsis. Crit Care 2010;14 (Suppl 2):P9 10.1186/cc9112
    1. Sutherland A, Thomas M, Brandon RA, Brandon RB, Lipman J, Tang B, et al. Development and validation of a novel molecular biomarker diagnostic test for the early detection of sepsis. Crit Care 2011;15:R149 10.1186/cc10274
    1. Brandon RB, Thomas M, Brandon RA, Venter D, Presneill J, Lipman J, et al. A limited set of molecular biomarkers may provide superior diagnostic outcomes to procalcitonin in sepsis. Crit Care 2012;16 (Suppl 3):P40 10.1186/cc11727
    1. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, et al. 2001 SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med. 2003;31:1250–1256.
    1. Klein Klouwenberg PM, Ong DS, Bos LD, de Beer FM, van Hooijdonk RT, Huson MA, et al. Interobserver agreement of centers for disease control and prevention criteria for classifying infections in critically ill patients. Crit Care Med. 2013;41:2373–2378. 10.1097/CCM.0b013e3182923712
    1. Klein Klouwenberg PM, van Mourik MS, Ong DS, Horn J, Schultz MJ, Cremer OL, et al. Electronic implementation of a novel surveillance paradigm for ventilator-associated events. Feasibility and validation. Am J Respir Crit Care Med. 2014;189:947–955. 10.1164/rccm.201307-1376OC
    1. Garner JS, Jarvis WR, Emori TG, Horan TC, Hughes JM. CDC definitions for nosocomial infections, 1988. Am J Infect Control 1988;16:128–140.
    1. Lockstone HE. Exon array data analysis using Affymetrix power tools and R statistical software. Brief Bioinform. 2011;12.6:634–644.
    1. Huber PJ. Robust statistics New York: John Wiley and Sons; 1981.
    1. Guyon I, Weston J, Barnhill S, Vapnik V. Gene selection for cancer classification using support vector machines. Mach Learn. 2002;46:389–422. 10.1023/A:1012487302797
    1. Díaz-Uriarte R, Alvarez de Andrés S. Gene selection and classification of microarray data using random forest. BMC Bioinformatics 2006;7:3
    1. Helm D, Labischinski H, Schallehn G, Naumann D. Classification and identification of bacteria by Fourier-transform infrared spectroscopy. J Gen Microbiol. 1991;137:69–79.
    1. Clinical and Laboratory Standards Institute. Assessment of the diagnostic accuracy of laboratory tests using receiver operating characteristic curves; approved guideline 2nd edition. EP24-A2. Wayne (Pennsylvania): Clinical and Laboratory Standards Institute; 2011.
    1. Huang SH. Dimensionality reduction in automatic knowledge acquisition: a simple greedy search approach. IEEE Trans Knowl Data Eng. 2003;15.6:1364–1373. 10.1109/TKDE.2003.1245278
    1. Venkatraman ES. A permutation test to compare receiver operating characteristic curves. Biometrics 2000;56:1134–1138.
    1. Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 2011;12:77–93. 10.1186/1471-2105-12-77
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics 1988;44:837–845.
    1. Gustafson P, Le Nhu D. Comparing the effects of continuous and discrete covariate mismeasurement, with emphasis on the dichotomization of mismeasured predictors. Biometrics 2002;58:878–887.
    1. Royston P, Altman DG, Sauerbrei W. Dichotomizing continuous predictors in multiple regression: a bad idea. Stat Med. 2006;25:127–141.
    1. Naggara O, Raymond J, Guilbert F, Roy D, Weill A, Altman DG. Analysis by categorizing or dichotomizing continuous variables is inadvisable: an example from the natural history of unruptured aneurysms. Am J Neuroradiol. 2011;32:437–440. 10.3174/ajnr.A2425
    1. Caille A, Leyrat C, Giraudeau B. Dichotomizing a continuous outcome in cluster randomized trials: impact on power. Stat Med. 2012;31:2822–2832. 10.1002/sim.5409
    1. Gu W, Pepe MS. Estimating the capacity for improvement in risk prediction with a marker. Biostatistics 2009;10:172–186. 10.1093/biostatistics/kxn025
    1. Pencina MJ, D’Agostino RB Sr, D’Agostino RB Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27:157–172.
    1. Kerr KF, Wang Z, Janes H, McClelland RL, Psaty BM, Pepe MS. Net reclassification indices for evaluating risk prediction instruments: a critical review. Epidemiology 2014;25:114–121. 10.1097/EDE.0000000000000018
    1. Leisenring W, Alonzo T, Pepe MS. Comparisons of predictive values of binary medical diagnostic tests for paired designs. Biometrics 2000;56:345–351.
    1. Bossuyt PM, Reitsma JB, Bruns DE, Gatsonis CA, Glasziou PP, Irwig LM, et al. Towards complete and accurate reporting of studies of diagnostic accuracy: the STARD initiative. Clin Chem. 2003;49:1–6.
    1. Bossuyt PM, Reitsma JB, Bruns DE, Gatsonis CA, Glasziou PP, Irwig LM, et al. The STARD statement for reporting studies of diagnostic accuracy: explanation and elaboration. Clin Chem. 2003;49:7–18.
    1. Takahashi W, Watanabe E, Fujimura L, Watanabe-Takano H, Yoshidome H, Swanson PE, et al. Kinetics and protective role of autophagy in a mouse cecal ligation and puncture-induced sepsis. Crit Care 2013;17:R160 10.1186/cc12839
    1. Huynh KK, Eskelinen EL, Scott CC, Malevanets A, Saftig P, Grinstein S. LAMP proteins are required for fusion of lysosomes with phagosomes. EMBO J 2007;26:313–324.
    1. Lin L, Ayala P, Larson J, Mulks M, Fukuda M, Carlsson SR, et al. The Neisseria type 2 IgA1 protease cleaves LAMP1 and promotes survival of bacteria within epithelial cells. Mol Microbiol. 1997;24:1083–1094.
    1. Endo S, Inada K, Yamashita H, Takakuwa T, Nakae H, Kasai T, et al. Platelet-activating factor (PAF) acetylhydrolase activity, type II phospholipase A2, and cytokine levels in patients with sepsis. Res Commun Chem Pathol Pharmacol. 1994;83:289–295.
    1. Yang J, Xu J, Chen X, Zhang Y, Jiang X, Guo X, et al. Decrease of plasma platelet-activating factor acetylhydrolase activity in lipopolysaccharide induced Mongolian gerbil sepsis model. PLoS ONE 2010;5:e9190 10.1371/journal.pone.0009190
    1. Gomes RN, Bozza FA, Amancio RT, Japiassu AM, Vianna RC, Larangeira AP, et al. Exogenous platelet-activating factor acetylhydrolase reduces mortality in mice with systemic inflammatory response syndrome and sepsis. Shock 2006;26:41–49.
    1. Takakuwa T, Endo S, Nakae H, Suzuki T, Inada K, Yoshida M, et al. Relationships between plasma levels of type-II phospholipase A2, PAF-acetylhydrolase, leukotriene B4, complements, endothelin-1, and thrombomodulin in patients with sepsis. Res Commun Chem Pathol Pharmacol. 1994;84:271–281.
    1. Schuster DP, Metzler M, Opal S, Lowry S, Balk R, Abraham E, et al. Recombinant platelet-activating factor acetylhydrolase to prevent acute respiratory distress syndrome and mortality in severe sepsis: phase IIb, multicenter, randomized, placebo-controlled, clinical trial. Crit Care Med. 2003;31:1612–1619.
    1. Opal S, Laterre P-F, Abraham E, Francois B, Wittebole X, Lowry S, et al. Recombinant human platelet-activating factor acetylhydrolase for treatment of severe sepsis: results of a phase III, multicenter, randomized, double-blind, placebo-controlled, clinical trial. Crit Care Med. 2004;32:332–341.
    1. Pankla R, Buddhisa S, Berry M, Blankenship DM, Bancroft GJ, Banchereau J, et al. Genomic transcriptional profiling identifies a candidate blood biomarker signature for the diagnosis of septicemic melioidosis. Genome Biol. 2009;10:R127 10.1186/gb-2009-10-11-r127
    1. Ledford JG, Kovarova M, Koller BH. Impaired host defense in mice lacking ONZIN. J Immunol. 2007;178:5132–5143.
    1. Johnson RM, Kerr MS, Slaven JE. Perforin is detrimental to controlling C. muridarum replication in vitro, but not in vivo. PLoS ONE 2013;8:e63340 10.1371/journal.pone.0063340
    1. Johnson RM, Kerr MS, Slaven JE. Plac8-dependent and inducible NO synthase-dependent mechanisms clear Chlamydia muridarum infections from the genital tract. J Immunol. 2012;188:1896–1904. 10.4049/jimmunol.1102764
    1. Wieland S, Thimme R, Purcell RH, Chisari FV. Genomic analysis of the host response to hepatitis B virus infection. Proc Natl Acad Sci U S A 2004;101:6669–6674.
    1. Kuroki M, Arakawa F, Matsuo Y, Oikawa S, Misumi Y, Nakazato H, et al. Molecular cloning of nonspecific cross-reacting antigens in human granulocytes. J Biol Chem. 1991;266:11810–11817.
    1. Tascón JD, Adrian J, Kopp K, Scholz P, Tschan MP, Kuespert K, et al. The granulocyte orphan receptor CEACAM4 is able to trigger phagocytosis of bacteria. J Leukocyte Biol. 2015;97:521–531. 10.1189/jlb.2AB0813-449RR
    1. Bos MP, Grunert F, Belland RJ. Differential recognition of members of the carcinoembryonic antigen family by Opa variants of Neisseria gonorrhoeae. Infect Immun. 1997;65:2353–2361.
    1. Klein SL. Sex differences in prophylaxis and therapeutic treatments for viral diseases. Handb Exp Pharmacol. 2012;214:499–522. 10.1007/978-3-642-30726-3_22
    1. Pennell LM, Galligan CL, Fish EN. Sex affects immunity. J Autoimmun. 2012;38:J282–J291. 10.1016/j.jaut.2011.11.013
    1. Gelfand EW. Development of asthma is determined by the age-dependent host response to respiratory virus infection: therapeutic implications. Curr Opin Immunol. 2012;24:713–719. 10.1016/j.coi.2012.08.011
    1. Stoecklein VM, Osuka A, Lederer JA. Trauma equals danger—damage control by the immune system. J Leukocyte Biol. 2012;92:539–551. 10.1189/jlb.0212072
    1. Lang PO, Aspinall R. Immunosenescence and herd immunity: with an ever increasing aging population do we need to rethink vaccine schedules? Expert Rev Vaccines 2012;11:167–176. 10.1586/erv.11.187
    1. Fulop T, Larbi A, Kotb R, de Angelis AF, Pawelec G. Aging, immunity, and cancer. Discov Med 2011;11:537–550.
    1. Ransohoff DF, Feinstein AR. Problems of spectrum and bias in evaluating the efficacy of diagnostic tests. N Engl J Med. 1978;299:926–930.
    1. Lijmer JG, Mol BW, Heisterkamp S, Bonsel GJ, Prins MH, van der Meulen JH, et al. Empirical evidence of design-related bias in studies of diagnostic tests. J Am Med Assoc. 1999;282:1061–1066.
    1. Mulherin SA, Miller WC. Spectrum bias or spectrum effect? Subgroup variation in diagnostic test evaluation. Ann Intern Med. 2002;137:598–602.
    1. Willis BH. Spectrum bias—why clinicians need to be cautious when applying diagnostic test studies. Fam Pract. 2008;25:390–396. 10.1093/fampra/cmn051
    1. Castelli GP, Pognani C, Meisner M, Stuani A, Bellomi D, Sgarbi L. Procalcitonin and C-reactive protein during systemic inflammatory response syndrome, sepsis and organ dysfunction. Crit Care. 2004;8:R234–R242. 10.1186/cc2877
    1. Luzzani A, Polati E, Dorizzi R, Rungatscher A, Pavan R, Merlini A. Comparison of procalcitonin and C-reactive protein as markers of sepsis. Crit Care Med. 2003;31:1737–1741.
    1. Rey C, Los Arcos M, Concha A, Medina A, Prieto S, Martinez P, et al. Procalcitonin and C-reactive protein as markers of systemic inflammatory response syndrome severity in critically ill children. Intensive Care Med. 2007;33:477–484.
    1. van Gestel A, Bakker J, Veraart CP, van Hout BA. Prevalence and incidence of severe sepsis in Dutch intensive care units. Crit Care. 2004;8:R153–R162. 10.1186/cc2858
    1. Pewsner D, Battaglia M, Minder C, Marx A, Bucher HC, Egger M. Ruling a diagnosis in or out with “SpPIn” and “SnNOut”: a note of caution. BMJ. 2004;329:209–213.
    1. Knaus WA, Wagner DP, Draper EA, Zimmerman JE, Bergner M, Bastos PG, et al. The APACHE III prognostic system. Risk prediction of hospital mortality for critically ill hospitalized adults. Chest 1991;100:1619–1636.
    1. Moreno R, Vincent J-L, Matos R, Mendonca A, Cantraine F, Thijs L, et al. The use of maximum SOFA score to quantify organ dysfunction/failure in intensive care. Results of a prospective, multicentre study. Working Group on Sepsis related Problems of the ESICM. Intensive Care Med. 1999;25:686–696.
    1. Cook R, Cook DJ, Tilley J, Lee K, Marshall J. Multiple organ dysfunction: baseline and serial component scores. Crit Care Med. 2001;29:2046–2050.
    1. Braykov NP, Morgan DJ, Schweizer ML, Uslan DZ, Kelesidis T, Weisenberg SA, et al. Assessment of empirical antibiotic therapy optimisation in six hospitals: an observational cohort study. Lancet Infect Dis. 2014;14:1220–1227. 10.1016/S1473-3099(14)70952-1
    1. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AA, Knaus WA, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest 1992;101:1644–1655.
    1. Nathan C. Points of control in inflammation. Nature 2002;420:846–852.
    1. Nathan C, Ding A. Non-resolving inflammation. Cell 2010;140:871–882. 10.1016/j.cell.2010.02.029

Source: PubMed

3
S'abonner