Prognostic and predictive enrichment in sepsis

Natalja L Stanski, Hector R Wong, Natalja L Stanski, Hector R Wong

Abstract

Sepsis is a heterogeneous disease state that is both common and consequential in critically ill patients. Unfortunately, the heterogeneity of sepsis at the individual patient level has hindered advances in the field beyond the current therapeutic standards, which consist of supportive care and antibiotics. This complexity has prompted attempts to develop a precision medicine approach, with research aimed towards stratifying patients into more homogeneous cohorts with shared biological features, potentially facilitating the identification of new therapies. Several investigators have successfully utilized leukocyte-derived mRNA and discovery-based approaches to subgroup patients on the basis of biological similarities defined by transcriptomic signatures. A critical next step is to develop a consensus sepsis subclassification system, which includes transcriptomic signatures as well as other biological and clinical data. This goal will require collaboration among various investigative groups, and validation in both existing data sets and prospective studies. Such studies are required to bring precision medicine to the bedside of critically ill patients with sepsis.

Conflict of interest statement

H.R.W. and the Cincinnati Children’s Hospital Research Foundation hold United States Patents for the PERSEVERE biomarkers and paediatric endotyping strategies described in the manuscript. N.L.S. declares no competing interests.

Figures

Fig. 1. Simplified diagram illustrating a generic…
Fig. 1. Simplified diagram illustrating a generic approach to prognostic and predictive enrichment of sepsis.
A heterogeneous cohort of patients with sepsis is first divided using prognostic enrichment into those at a low and those at a high risk of mortality. Patients who are at a low mortality risk are treated using standard care. The high-risk patients can be further divided using predictive enrichment and treated with targeted therapies based on their underlying, subgroup-defining biology.
Fig. 2. Network of candidate genes for…
Fig. 2. Network of candidate genes for predicting patient mortality identified through prognostic enrichment strategies.
The community approach to mortality risk prediction in a sepsis study identified 65 risk-predicting genes based on publicly available gene expression data from adults, children and neonate with sepsis. Similarly, during the initial discovery phase of the PERSEVERE biomarkers used for prognostic enrichment, 80 genes associated with poor outcome were selected based on transcriptomic data from a cohort of paediatric patients with septic shock. An analysis of the genes identified in these two studies revealed 11 genes in common: adhesion G protein-coupled receptor E3 (ADGRE3), CD24 molecule (CD24), carcinoembryonic antigen-related cell adhesion molecule 8 (CEACAM8), CDC28 protein kinase regulatory subunit 2 (CKS2), C-X3-C motif chemokine receptor 1 (CX3CR1), DNA damage inducible transcript 4 (DDIT4), G0/G1 switch 2 (G0S2), C-X-C motif chemokine ligand 8 (CXCL8, also known as IL-8), MAF bZIP transcription factor F (MAFF), regulator of G protein signalling 1 (RGS1) and transforming growth factor beta induced (TGFBI). The network was generated by uploading these 11 genes to the Ingenuity Pathway Analysis platform. Solid lines represent direct interactions and dashed lines represent indirect interactions; solid arrowheads can represent activation, modification and phosphorylation among others (for more details see Ingenuity Pathway Analysis legend), open arrowhead represents translocation and bar-headed arrow indicates that the gene inhibits and acts on its target. The gene network contains three highly connected central nodes relevant to the pathobiology of sepsis and inflammation: tumour necrosis factor (TNF), CXCL8 and tumour protein p53 (TP53). APOBEC3B, apolipoprotein B mRNA editing enzyme catalytic subunit 3B; CCRL2, C-C motif chemokine receptor like 2; CD300C, CMRF35-like molecule 6; DCSTAMP, dendrocyte expressed seven transmembrane protein; EHD1, EH domain containing 1; FFAR2, free fatty acid receptor 2; HRG, histidine-rich glycoprotein; HTR4, 5-hydroxytryptamine receptor 4; IL36RN, interleukin 36 receptor antagonist; LAT2, linker for activation of T cells family member 2; LILRA5, leukocyte immunoglobulin-like receptor A5; LSS, lanosterol synthase; MIR142, microRNA 142; NINJ1, ninjurin 1; PRIM1, DNA primase subunit 1; RGS13, regulator of G protein signalling 13; SCO2, SCO cytochrome c oxidase assembly protein 2; SQLE, squalene epoxidase; TGFB1, transforming growth factor beta 1; TGM2, transglutaminase 2; TREM1, triggering receptor expressed on myeloid cells 1. aRefers to the mature sequence miR-125b-2–3p, encoded in the micro RNA-125b-2 (MIR125B2) stem loop sequence, and includes other microRNAs with the seed sequence CAAGUCA.

References

    1. Hotchkiss RS, et al. Sepsis and septic shock. Nat. Rev. Dis. Prim. 2016;2:16045.
    1. Singer E, et al. Neutrophil gelatinase-associated lipocalin: pathophysiology and clinical applications. Acta Physiol. 2013;207:663–672.
    1. Marshall JC. Why have clinical trials in sepsis failed? Trends Mol. Med. 2014;20:195–203.
    1. Collins FS, Varmus H. A new initiative on precision medicine. N. Engl. J. Med. 2015;372:793–795.
    1. Wong HR. Intensive care medicine in 2050: precision medicine. Intensive Care Med. 2017;43:1507–1509.
    1. Tsimberidou A-M. Initiative for molecular profiling and advanced cancer therapy and challenges in the implementation of precision medicine. Curr. Probl. Cancer. 2017;41:176–181.
    1. Coyne GO, Takebe N, Chen AP. Defining precision: The precision medicine initiative trials NCI-MPACT and NCI-MATCH. Curr. Probl. Cancer. 2017;41:182–193.
    1. Seibel NL, et al. Pediatric oncology enters an era of precision medicine. Curr. Probl. Cancer. 2017;41:194–200.
    1. The White House President Barack Obama. The precision medicine initiative. The White House (2015).
    1. Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu VX. Toward smarter lumping and smarter splitting: rethinking strategies for sepsis and acute respiratory distress syndrome clinical trial design. Am. J. Respir. Crit. Care Med. 2016;194:147–155.
    1. Center for Drug Evaluation and Research. Enrichment strategies for clinical trials to support approval of human drugs and biological products (FDA, 2019).
    1. CONSENSUS Trial Study Group Effects of enalapril on mortality in severe congestive heart failure. results of the cooperative north scandinavian enalapril survival study (CONSENSUS) N. Engl. J. Med. 1987;316:1429–1435.
    1. Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 2005;353:1659–1672.
    1. Zhang Z, Zhang G, Goyal H, Mo L, Hong Y. Identification of subclasses of sepsis that showed different clinical outcomes and responses to amount of fluid resuscitation: a latent profile analysis. Crit. Care. 2018;22:347.
    1. Gårdlund B, et al. Six subphenotypes in septic shock: latent class analysis of the PROWESS shock study. J. Crit. Care. 2018;47:70–79.
    1. Bhavani SV, et al. Identifying novel sepsis subphenotypes using temperature trajectories. Am J. Respir. Crit. Care Med. 2019;200:327–335.
    1. Seymour CW, et al. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA. 2019;321:2003–2017.
    1. Wong HR, et al. The pediatric sepsis biomarker risk model. Crit. Care. 2012;16:R174.
    1. Wong HR, et al. Testing the prognostic accuracy of the updated pediatric sepsis biomarker risk model. PLOS ONE. 2014;9:e86242.
    1. Wong HR, et al. Pediatric sepsis biomarker risk model-ii: redefining the pediatric sepsis biomarker risk model with septic shock phenotype. Crit. Care Med. 2016;44:2010–2017.
    1. Wong HR, et al. A multibiomarker-based outcome risk stratification model for adult septic shock*. Crit. Care Med. 2014;42:781–789.
    1. Wong HR, et al. Improved risk stratification in pediatric septic shock using both protein and mrna biomarkers. PERSEVERE-XP. Am. J. Respir. Crit. Care Med. 2017;196:494–501.
    1. Scicluna BP, et al. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir. Med. 2017;5:816–826.
    1. Sweeney TE, et al. Unsupervised analysis of transcriptomics in bacterial sepsis across multiple datasets reveals three robust clusters. Crit. Care Med. 2018;46:915–925.
    1. Wong HR, et al. Identification of pediatric septic shock subclasses based on genome-wide expression profiling. BMC Med. 2009;7:34.
    1. Wong HR, et al. Toward a clinically feasible gene expression-based subclassification strategy for septic shock: proof of concept. Crit. Care Med. 2010;38:1955–1961.
    1. Wong HR, et al. Validation of a gene expression-based subclassification strategy for pediatric septic shock. Crit. Care Med. 2011;39:2511–2517.
    1. Wong HR, et al. Developing a clinically feasible personalized medicine approach to pediatric septic shock. Am. J. Respir. Crit. Care Med. 2015;191:309–315.
    1. Langley RJ, et al. An integrated clinico-metabolomic model improves prediction of death in sepsis. Sci. Transl. Med. 2013;5:195ra95.
    1. Sweeney TE, et al. A community approach to mortality prediction in sepsis via gene expression analysis. Nat. Commun. 2018;9:694.
    1. Davenport EE, et al. Genomic landscape of the individual host response and outcomes in sepsis: a prospective cohort study. Lancet Respir. Med. 2016;4:259–271.
    1. Kaplan JM, Wong HR. Biomarker discovery and development in pediatric critical care medicine. Pediatr. Crit. Care Med. 2011;12:165–173.
    1. Meyer NJ, et al. A functional synonymous coding variant in the IL1RN gene is associated with survival in septic shock. Am. J. Respir. Crit. Care Med. 2014;190:656–664.
    1. Wong HR, et al. Endotype transitions during the acute phase of pediatric septic shock reflect changing risk and treatment response. Crit. Care Med. 2018;46:e242–e249.
    1. Burnham KL, et al. Shared and distinct aspects of the sepsis transcriptomic response to fecal peritonitis and pneumonia. Am. J. Respir. Crit. Care Med. 2017;196:328–339.
    1. Antcliffe DB, et al. Transcriptomic signatures in sepsis and a differential response to steroids: from the VANISH randomized trial. Am. J. Respir. Crit. Care Med. 2018;199:980–986.
    1. Wong HR, Sweeney TE, Hart KW, Khatri P, Lindsell CJ. Pediatric sepsis endotypes among adults with sepsis. Crit. Care Med. 2017;45:e1289–e1291.
    1. Wynn JL, et al. Defective innate immunity predisposes murine neonates to poor sepsis outcome but is reversed by TLR agonists. Blood. 2008;112:1750–1758.
    1. Wynn JL, et al. The influence of developmental age on the early transcriptomic response of children with septic shock. Mol. Med. 2011;17:1146–1156.
    1. Wynn JL, et al. Postnatal age is a critical determinant of the neonatal host response to sepsis. Mol. Med. 2015;21:496–504.
    1. Raymond SL, et al. Unique transcriptomic response to sepsis is observed among patients of different age groups. PLOS ONE. 2017;12,:e0184159.
    1. Atkinson SJ, et al. Matrix metalloproteinase-8 augments bacterial clearance in a juvenile sepsis model. Mol. Med. 2016;22:455–463.
    1. Cernada M, et al. Genome-wide expression profiles in very low birth weight infants with neonatal sepsis. Pediatrics. 2014;133:e1203–e1211.
    1. Gentile LF, et al. Protective immunity and defects in the neonatal and elderly immune response to sepsis. J. Immunol. 2014;192:3156–3165.
    1. Annane D, et al. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA. 2002;288:862–871.
    1. Sprung CL, et al. Hydrocortisone therapy for patients with septic shock. N. Engl. J. Med. 2008;358:111–124.
    1. Panacek EA, et al. Efficacy and safety of the monoclonal anti-tumor necrosis factor antibody F(ab’)2 fragment afelimomab in patients with severe sepsis and elevated interleukin-6 levels. Crit. Care Med. 2004;32:2173–2182.
    1. Janz DR, et al. Randomized, placebo-controlled trial of acetaminophen for the reduction of oxidative injury in severe sepsis: the acetaminophen for the reduction of oxidative injury in severe sepsis trial. Crit. Care Med. 2015;43:534–541.
    1. Dellinger RP, et al. Effect of targeted polymyxin b hemoperfusion on 28-day mortality in patients with septic shock and elevated endotoxin level: the EUPHRATES randomized clinical trial. JAMA. 2018;320:1455–1463.
    1. Vincent J-L, et al. Effect of a recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy: the SCARLET randomized clinical trial. JAMA. 2019;321:1993–2002.
    1. Hawchar F, et al. Extracorporeal cytokine adsorption in septic shock: a proof of concept randomized, controlled pilot study. J. Crit. Care. 2019;49:172–178.
    1. Annane D, et al. Hydrocortisone plus fludrocortisone for adults with septic shock. N. Engl. J. Med. 2018;378:809–818.
    1. Venkatesh B, et al. Adjunctive glucocorticoid therapy in patients with septic shock. N. Engl. J. Med. 2018;378:797–808.
    1. Casserly B, et al. Low-dose steroids in adult septic shock: results of the surviving sepsis campaign. Intensive Care Med. 2012;38:1946–1954.
    1. Menon S, et al. Urinary biomarker incorporation into the renal angina index early in intensive care unit admission optimizes acute kidney injury prediction in critically ill children: a prospective cohort study. Nephrol. Dial. Transplant. 2016;31:586–594.
    1. Zimmerman JJ, Williams MD. Adjunctive corticosteroid therapy in pediatric severe sepsis: observations from the RESOLVE study. Pediatr Crit. Care Med. 2011;12:2–8.
    1. Funk D, et al. Low-dose corticosteroid treatment in septic shock: a propensity-matching study. Crit. Care Med. 2014;42:2333–2341.
    1. Atkinson SJ, et al. Corticosteroids and pediatric septic shock outcomes: a risk stratified analysis. PLOS ONE. 2014;9:e112702.
    1. Wong HR, et al. Combining prognostic and predictive enrichment strategies to identify children with septic shock responsive to corticosteroids. Crit. Care Med. 2016;44:e1000–e1003.
    1. US National Library of Medicine. (2019).
    1. Alobaidi R, et al. Association between fluid balance and outcomes in critically ill children. JAMA Pediatr. 2018;172:257–268.
    1. Abulebda K, et al. Post-ICU admission fluid balance and pediatric septic shock outcomes: a risk-stratified analysis. Crit. Care Med. 2014;42:397–403.
    1. Bernard GR, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N. Engl. J. Med. 2001;344:699–709.
    1. Dhainaut J-F, et al. Treatment effects of drotrecogin alfa (activated) in patients with severe sepsis with or without overt disseminated intravascular coagulation. J. Thromb. Haemost. 2004;2:1924–1933.
    1. Vincent J-L, et al. A randomized, double-blind, placebo-controlled, Phase 2b study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Crit. Care Med. 2013;41:2069–2079.
    1. Fisher CJ, et al. Initial evaluation of human recombinant interleukin-1 receptor antagonist in the treatment of sepsis syndrome: a randomized, open-label, placebo-controlled multicenter trial. Crit. Care Med. 1994;22:12–21.
    1. Fisher CJ, et al. Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group. JAMA. 1994;271:1836–1843.
    1. Opal SM, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. The Interleukin-1 Receptor Antagonist Sepsis Investigator Group. Crit. Care Med. 1997;25:1115–1124.
    1. Meyer NJ, et al. Mortality benefit of recombinant human interleukin-1 receptor antagonist for sepsis varies by initial interleukin-1 receptor antagonist plasma concentration. Crit. Care Med. 2018;46:21–28.
    1. Klein DJ, et al. Polymyxin B hemoperfusion in endotoxemic septic shock patients without extreme endotoxemia: a post hoc analysis of the EUPHRATES trial. Intensive Care Med. 2018;44:2205–2212.
    1. Calfee CS, et al. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir. Med. 2014;2:611–620.
    1. Delucchi K, et al. Stability of ARDS subphenotypes over time in two randomised controlled trials. Thorax. 2018;73:439–445.
    1. Calfee CS, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir. Med. 2018;6:691–698.
    1. Famous KR, et al. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am. J. Respir. Crit. Care Med. 2017;195:331–338.
    1. Yehya N, Thomas NJ, Wong HR. Evidence of endotypes in pediatric acute hypoxemic respiratory failure caused by sepsis. Pediatr. Crit. Care Med. 2019;20:110–112.
    1. Uchino S, et al. Acute renal failure in critically ill patients: a multinational, multicenter Study. JAMA. 2005;294:813–818.
    1. Bouchard J, et al. A Prospective international multicenter study of AKI in the intensive Care Unit. Clin. J. Am. Soc. Nephrol. 2015;10:1324–1331.
    1. Alobaidi R, Basu RK, Goldstein SL, Bagshaw SM. Sepsis-associated acute kidney injury. Semin. Nephrol. 2015;35:2–11.
    1. Bagshaw SM, et al. Acute kidney injury in septic shock: clinical outcomes and impact of duration of hypotension prior to initiation of antimicrobial therapy. Intensive Care Med. 2009;35:871–881.
    1. Kellum JA, Prowle JR. Paradigms of acute kidney injury in the intensive care setting. Nat. Rev. Nephrol. 2018;14:217.
    1. Gomez H, et al. A unified theory of sepsis-induced acute kidney injury: inflammation, microcirculatory dysfunction, bioenergetics, and the tubular cell adaptation to injury. Shock. 2014;41:3–11.
    1. Schrier RW, Wang W. Acute renal failure and sepsis. N. Engl. J. Med. 2004;351:159–169.
    1. Bagshaw SM, et al. Septic acute kidney injury in critically ill patients: clinical characteristics and outcomes. Clin. J. Am. Soc. Nephrol. 2007;2:431–439.
    1. Bagshaw SM, George C, Bellomo R, ANZICS Database Management Committee Early acute kidney injury and sepsis: a multicentre evaluation. Crit. Care. 2008;12:R47.
    1. Drey M, et al. C-terminal agrin fragment (CAF) reflects renal function in patients suffering from severe sepsis or septic shock. Clin. Lab. 2015;61:69–76.
    1. Yuan ZK, et al. [Value of urine soluble triggering receptor expressed on myeloid cells-1 in the early diagnosis of sepsis associated acute kidney injury] Zhonghua Er Ke Za Zhi. 2018;56:342–346.
    1. Kim Ji Hoon, Park Yoo Seok, Yoon Chang-Yun, Lee Hye Sun, Kim Sinae, Lee Jong Wook, Kong Taeyoung, You Je Sung, Park Jong Woo, Chung Sung Phil. Delta Neutrophil Index for the Prediction of the Development of Sepsis-Induced Acute Kidney Injury in the Emergency Department. SHOCK. 2019;52(4):414–422.
    1. Wheeler DS, et al. Serum neutrophil gelatinase-associated lipocalin (NGAL) as a marker of acute kidney injury in critically ill children with septic shock. Crit. Care Med. 2008;36:1297–1303.
    1. Jiang D, et al. Serum heart-type fatty acid-binding protein as a predictor for the development of sepsis-associated acute kidney injury. Expert Rev. Mol. Diagn. 2019;17:1–9.
    1. Tu Y, et al. Urinary netrin-1 and KIM-1 as early biomarkers for septic acute kidney injury. Ren. Fail. 2014;36:1559–1563.
    1. Honore PM, et al. Urinary tissue inhibitor of metalloproteinase-2 and insulin-like growth factor-binding protein 7 for risk stratification of acute kidney injury in patients with sepsis. Crit. Care Med. 2016;44:1851–1860.
    1. Kim S, et al. Is plasma neutrophil gelatinase-associated lipocalin a predictive biomarker for acute kidney injury in sepsis patients? A systematic review and meta-analysis. J. Crit. Care. 2016;33:213–223.
    1. Wong HR, et al. A multibiomarker-based model for estimating the risk of septic acute kidney injury. Crit. Care Med. 2015;43:1646–1653.
    1. Basu RK, et al. Identification of candidate serum biomarkers for severe septic shock-associated kidney injury via microarray. Crit. Care. 2011;15:R273.
    1. Song J, et al. Value of the combination of renal resistance index and central venous pressure in the early prediction of sepsis-induced acute kidney injury. J. Crit. Care. 2018;45:204–208.
    1. Lee C-W, et al. A combination of SOFA score and biomarkers gives a better prediction of septic AKI and in-hospital mortality in critically ill surgical patients: a pilot study. World J. Emerg. Surg. 2018;13:41.
    1. Zhou J, et al. A simple risk score for prediction of sepsis associated-acute kidney injury in critically ill patients. J. Nephrol. 2019 doi: 10.1007/s40620-019-00625-y.
    1. Dai X, et al. Diagnostic value of neutrophil gelatinase-associated lipocalin, cystatin C, and soluble triggering receptor expressed on myeloid cells-1 in critically ill patients with sepsis-associated acute kidney injury. Crit. Care. 2015;19:223.
    1. Iwaki T, et al. PPAR α contributes to protection against metabolic and inflammatory derangements associated with acute kidney injury in experimental sepsis. Physiol. Rep. 2019;7:e14078.
    1. Guinney J, et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015;21:1350–1356.
    1. Maslove DM, Wong HR. Gene expression profiling in sepsis: timing, tissue, and translational considerations. Trends Mol. Med. 2014;20:204–213.
    1. Wong HR, Sweeney TE, Lindsell CJ. Simplification of a septic shock endotyping strategy for clinical application. Am. J. Respir. Crit. Care Med. 2017;195:263–265.
    1. Tang BMP, McLean AS, Dawes IW, Huang SJ, Lin RCY. Gene-expression profiling of peripheral blood mononuclear cells in sepsis. Crit. Care Med. 2009;37:882–888.
    1. Tang BMP, McLean AS, Dawes IW, Huang SJ, Lin RCY. The use of gene-expression profiling to identify candidate genes in human sepsis. Am. J. Respir. Crit. Care Med. 2007;176:676–684.
    1. Vartanian K, et al. Gene expression profiling of whole blood: comparison of target preparation methods for accurate and reproducible microarray analysis. BMC Genomics. 2009;10:2.
    1. Hedlund E, Deng Q. Single-cell RNA sequencing: technical advancements and biological applications. Mol. Aspects Med. 2018;59:36–46.
    1. National Institute of Diabetes and Digestive and Kidney Diseases. Kidney precision medicine project NIDDK, 2018)
    1. Kidney Precision Medicine Project. Changing the way we understand and treat kidney disease. KPMP (2019)
    1. Talwar S, et al. Gene expression profiles of peripheral blood leukocytes after endotoxin challenge in humans. Physiol. Genomics. 2006;25:203–215.
    1. Kwan A, Hubank M, Rashid A, Klein N, Peters MJ. Transcriptional instability during evolving sepsis may limit biomarker based risk stratification. PLOS ONE. 2013;8,:e60501.
    1. McDunn JE, et al. Plasticity of the systemic inflammatory response to acute infection during critical illness: development of the riboleukogram. PLOS ONE. 2008;3,:e1564.
    1. Polpitiya AD, McDunn JE, Burykin A, Ghosh BK, Cobb JP. Using systems biology to simplify complex disease: immune cartography. Crit. Care Med. 2009;37:S16–S21.
    1. Zhou B, et al. Analysis of factorial time-course microarrays with application to a clinical study of burn injury. Proc. Natl. Acad. Sci. USA. 2010;107:9923–9928.
    1. Storey JD, Xiao W, Leek JT, Tompkins RG, Davis RW. Significance analysis of time course microarray experiments. Proc. Natl. Acad. Sci. USA. 2005;102:12837–12842.
    1. Aerts J-M, Haddad WM, An G, Vodovotz Y. From data patterns to mechanistic models in acute critical illness. J. Crit. Care. 2014;29:604–610.
    1. Vodovotz Y, et al. Evidence-based modeling of critical illness: an initial consensus from the society for complexity in acute illness. J. Crit. Care. 2007;22:77–84.

Source: PubMed

3
Prenumerera