An immune dysfunction score for stratification of patients with acute infection based on whole-blood gene expression

Eddie Cano-Gamez, Katie L Burnham, Cyndi Goh, Alice Allcock, Zunaira H Malick, Lauren Overend, Andrew Kwok, David A Smith, Hessel Peters-Sengers, David Antcliffe, GAinS Investigators, Stuart McKechnie, Brendon P Scicluna, Tom van der Poll, Anthony C Gordon, Charles J Hinds, Emma E Davenport, Julian C Knight, Nigel Webster, Helen Galley, Jane Taylor, Sally Hall, Jenni Addison, Sian Roughton, Heather Tennant, Achyut Guleri, Natalia Waddington, Dilshan Arawwawala, John Durcan, Alasdair Short, Karen Swan, Sarah Williams, Susan Smolen, Christine Mitchell-Inwang, Tony Gordon, Emily Errington, Maie Templeton, Pyda Venatesh, Geraldine Ward, Marie McCauley, Simon Baudouin, Charley Higham, Jasmeet Soar, Sally Grier, Elaine Hall, Stephen Brett, David Kitson, Robert Wilson, Laura Mountford, Juan Moreno, Peter Hall, Jackie Hewlett, Stuart McKechnie, Christopher Garrard, Julian Millo, Duncan Young, Paula Hutton, Penny Parsons, Alex Smiths, Roser Faras-Arraya, Jasmeet Soar, Parizade Raymode, Jonathan Thompson, Sarah Bowrey, Sandra Kazembe, Natalie Rich, Prem Andreou, Dawn Hales, Emma Roberts, Simon Fletcher, Melissa Rosbergen, Georgina Glister, Jeronimo Moreno Cuesta, Julian Bion, Joanne Millar, Elsa Jane Perry, Heather Willis, Natalie Mitchell, Sebastian Ruel, Ronald Carrera, Jude Wilde, Annette Nilson, Sarah Lees, Atul Kapila, Nicola Jacques, Jane Atkinson, Abby Brown, Heather Prowse, Anton Krige, Martin Bland, Lynne Bullock, Donna Harrison, Gary Mills, John Humphreys, Kelsey Armitage, Shond Laha, Jacqueline Baldwin, Angela Walsh, Nicola Doherty, Stephen Drage, Laura Ortiz-Ruiz de Gordoa, Sarah Lowes, Charley Higham, Helen Walsh, Verity Calder, Catherine Swan, Heather Payne, David Higgins, Sarah Andrews, Sarah Mappleback, Charles Hind, Chris Garrard, D Watson, Eleanor McLees, Alice Purdy, Martin Stotz, Adaeze Ochelli-Okpue, Stephen Bonner, Iain Whitehead, Keith Hugil, Victoria Goodridge, Louisa Cawthor, Martin Kuper, Sheik Pahary, Geoffrey Bellingan, Richard Marshall, Hugh Montgomery, Jung Hyun Ryu, Georgia Bercades, Susan Boluda, Andrew Bentley, Katie Mccalman, Fiona Jefferies, Julian Knight, Emma Davenport, Katie Burnham, Narelle Maugeri, Jayachandran Radhakrishnan, Yuxin Mi, Alice Allcock, Cyndi Goh, Eddie Cano-Gamez, Katie L Burnham, Cyndi Goh, Alice Allcock, Zunaira H Malick, Lauren Overend, Andrew Kwok, David A Smith, Hessel Peters-Sengers, David Antcliffe, GAinS Investigators, Stuart McKechnie, Brendon P Scicluna, Tom van der Poll, Anthony C Gordon, Charles J Hinds, Emma E Davenport, Julian C Knight, Nigel Webster, Helen Galley, Jane Taylor, Sally Hall, Jenni Addison, Sian Roughton, Heather Tennant, Achyut Guleri, Natalia Waddington, Dilshan Arawwawala, John Durcan, Alasdair Short, Karen Swan, Sarah Williams, Susan Smolen, Christine Mitchell-Inwang, Tony Gordon, Emily Errington, Maie Templeton, Pyda Venatesh, Geraldine Ward, Marie McCauley, Simon Baudouin, Charley Higham, Jasmeet Soar, Sally Grier, Elaine Hall, Stephen Brett, David Kitson, Robert Wilson, Laura Mountford, Juan Moreno, Peter Hall, Jackie Hewlett, Stuart McKechnie, Christopher Garrard, Julian Millo, Duncan Young, Paula Hutton, Penny Parsons, Alex Smiths, Roser Faras-Arraya, Jasmeet Soar, Parizade Raymode, Jonathan Thompson, Sarah Bowrey, Sandra Kazembe, Natalie Rich, Prem Andreou, Dawn Hales, Emma Roberts, Simon Fletcher, Melissa Rosbergen, Georgina Glister, Jeronimo Moreno Cuesta, Julian Bion, Joanne Millar, Elsa Jane Perry, Heather Willis, Natalie Mitchell, Sebastian Ruel, Ronald Carrera, Jude Wilde, Annette Nilson, Sarah Lees, Atul Kapila, Nicola Jacques, Jane Atkinson, Abby Brown, Heather Prowse, Anton Krige, Martin Bland, Lynne Bullock, Donna Harrison, Gary Mills, John Humphreys, Kelsey Armitage, Shond Laha, Jacqueline Baldwin, Angela Walsh, Nicola Doherty, Stephen Drage, Laura Ortiz-Ruiz de Gordoa, Sarah Lowes, Charley Higham, Helen Walsh, Verity Calder, Catherine Swan, Heather Payne, David Higgins, Sarah Andrews, Sarah Mappleback, Charles Hind, Chris Garrard, D Watson, Eleanor McLees, Alice Purdy, Martin Stotz, Adaeze Ochelli-Okpue, Stephen Bonner, Iain Whitehead, Keith Hugil, Victoria Goodridge, Louisa Cawthor, Martin Kuper, Sheik Pahary, Geoffrey Bellingan, Richard Marshall, Hugh Montgomery, Jung Hyun Ryu, Georgia Bercades, Susan Boluda, Andrew Bentley, Katie Mccalman, Fiona Jefferies, Julian Knight, Emma Davenport, Katie Burnham, Narelle Maugeri, Jayachandran Radhakrishnan, Yuxin Mi, Alice Allcock, Cyndi Goh

Abstract

Dysregulated host responses to infection can lead to organ dysfunction and sepsis, causing millions of global deaths each year. To alleviate this burden, improved prognostication and biomarkers of response are urgently needed. We investigated the use of whole-blood transcriptomics for stratification of patients with severe infection by integrating data from 3149 samples from patients with sepsis due to community-acquired pneumonia or fecal peritonitis admitted to intensive care and healthy individuals into a gene expression reference map. We used this map to derive a quantitative sepsis response signature (SRSq) score reflective of immune dysfunction and predictive of clinical outcomes, which can be estimated using a 7- or 12-gene signature. Last, we built a machine learning framework, SepstratifieR, to deploy SRSq in adult and pediatric bacterial and viral sepsis, H1N1 influenza, and COVID-19, demonstrating clinically relevant stratification across diseases and revealing some of the physiological alterations linking immune dysregulation to mortality. Our method enables early identification of individuals with dysfunctional immune profiles, bringing us closer to precision medicine in infection.

Conflict of interest statement

Competing interests

ACG has received consulting fees as part of a Data Monitoring Committee from 30 Respiratory paid to his institution. All remaining authors declare that they have no competing interests.

Figures

Fig. 1. Construction of a reference map…
Fig. 1. Construction of a reference map of gene expression in sepsis using data from three different platforms.
(A) CCA analysis of GAinS samples with RNA-seq and microarray data available. Histograms represent marginal SRS1 (red) and SRS2 (blue) distributions. R = Pearson correlation; p = correlation p value. (B) Contribution of each gene to CC1, ranked increasingly. (C) CC1 contribution of each microarray (X axis) and RNA-seq (Y axis) feature. Black and red dots indicate genes in the Davenport signature and amongst the top 1% highest CC1 contributors, respectively. (D) Correlation of microarray/RNA-seq (X axis) and qRT-PCR (Y axis) measurements. Best linear fits are shown. R = Pearson correlation; p = correlation p value. (E) A reference map of sepsis based on the Davenport signature (PCA visualization). Dots represent samples, with shapes indicating profiling platform and colors SRS group.
Fig. 2. Stratification of patients with sepsis…
Fig. 2. Stratification of patients with sepsis based on whole blood gene expression.
(A) Receiver operating characteristic (ROC) curves showing cross-validation performance. AUROCs = area under the ROC curve. (B) UpSet plot showing prediction agreement between modalities. Colors indicate SRS classes (horizontal) and cross-modality agreement (vertical). Gray bars indicate samples with only one modality available. The heatmap (top) shows the level of cross-modality agreement (Jaccard index). (C) Volcano plot showing SRS1/SRS2 differential gene expression. Red indicates upregulation in SRS1 and blue upregulation in SRS2. (D) Correlation between SRS-associated log-fold changes from microarray and RNA-seq. The identity line is shown as a reference. Cor = Pearson correlation; p = correlation p value. (E) Cell count distribution per SRS group. p = T-test (top) or Kruskal-Wallis (bottom) p value. (F) SOFA score distribution per SRS group at the latest available time point. p = T-test (left) or Kruskal-Wallis (right) p value. (G) Kaplan-Meier curves of 28-day survival per SRS group, defined at the latest available time point. Shades indicate 95% confidence intervals. p = log-rank test p value.
Fig. 3. A quantitative score reflective of…
Fig. 3. A quantitative score reflective of immune dysfunction severity.
(A) Diffusion map estimated using the Extended gene signature. Colors indicate SRS group; shapes indicate profiling platforms. (B) Distribution of SRSq across cohorts. p = Kruskal-Wallis test p value. (C) Association between SRSq and mortality in GAinS, as determined using a sliding window approach. Shades represent 95% confidence intervals. (D) Estimated hazard ratios and 95% confidence intervals. (E) SRSq values stratified by ICU-acquired infection score (ICU-AI). β = regression coefficient; p = regression p value. (F) Kaplan-Meier curves of 28-day survival in patients sampled at multiple time points. Patients were stratified into quartiles based on their rate of SRSq reduction over time. Shades indicate 95% confidence intervals. p = log-rank test p value. (G) Association between rate of SRSq reduction and mortality, as determined using a sliding window. Shades represent 95% confidence intervals. (H) Causal model assumed for mediation analysis. Arrows represent causal directions. (I) Mediation effects. Lines indicate 95% confidence intervals, with solid and dotted lines corresponding to the treatment (high SRSq) and control (low SRSq) conditions. ACME = Average Causal Mediation Effect; ADE = Average Direct Effect; p = mediation p value.
Fig. 4. SepstratifieR’s construction and application to…
Fig. 4. SepstratifieR’s construction and application to new data.
Schematic representation of how SepstratifieR was built (top panel) and how it is applied to new data (bottom panel). Publicly available data (5, 6) were used to construct sepsis reference maps based on small gene signatures. Next, random forest models were trained to predict SRS and SRSq. When applying SepstratifieR to new samples, genes in the signature of interest are extracted and used to align new samples to the reference map. After alignment, SRS and SRSq were predicted using pre-trained models.
Fig. 5. Stratification of patients with pediatric…
Fig. 5. Stratification of patients with pediatric sepsis by SRSq.
(A) PCA plots based on whole blood transcriptomes. Samples are colored by illness severity (top), SRS (middle), and SRSq (bottom). (B) UpSet plot showing the agreement between SRS predictions and disease severity. Bar colors indicate SRS groups (top) and clinical phenotypes (bottom). The heatmap (top) quantifies the extent of this agreement (Jaccard indices). (C) SRSq distribution by clinical phenotype; p = Wilcoxon test p value. (D) SRSq distribution by time point and clinical phenotype. p = Wilcoxon test p value. (E) Correlation between SRSq-associated gene expression changes in adult (GAinS) and pediatric sepsis. Cor = Pearson correlation; p = correlation p value. (F) Immune-relevant pathways positively (left) or negatively (right) enriched in SRSq-associated genes.
Fig. 6. SRSq predicts oxygen requirement and…
Fig. 6. SRSq predicts oxygen requirement and reveals temporal immune dynamics in influenza.
(A) PCA plots based on whole blood transcriptomes. Samples are colored by oxygen requirement (top), SRS (middle), and SRSq (bottom). (B) Volcano plot showing genes differentially expressed along SRSq. Red indicates positive and blue negative associations with SRSq. The scatter plot (right) compares log-fold changes in sepsis (GAinS) and Influenza. Cor = Pearson correlation; p = correlation p value. (C) Top genes positively (top) and negatively (bottom) associated with SRSq. Samples are colored by SRS group. (D) SRSq stratified by supplemental oxygen requirement; p = Kruskal-Wallis test p value, *** = adjusted Dunn’s post-hoc test p < 0.01. (E) SRSq stratified by time since admission and oxygen requirement. Samples are colored by SRS group. p = Kruskal-Wallis test p value. (F) Line plot showing changes of SRSq over time. Line colors indicate SRS group assignment at recruitment.
Fig. 7. SRSq predicts severity of illness…
Fig. 7. SRSq predicts severity of illness and pinpoints mediators of COVID-19 mortality.
(A) PCA based on whole blood transcriptomes. Samples are colored by clinical severity. (B) Heatmap showing the overlap (as indicated by Jaccard index) between SRS and clinical severity groups. (C) SRSq stratified by clinical severity. p = Kruskal-Wallis test p value. (D) Association between SRSq and clinical variables. Samples are colored by SRS group. Lines indicate best linear fits. Cor = Pearson correlation; p = correlation p value. (E) Association between SRSq and mortality. (F) Estimated hazard ratios and 95% confidence intervals. (G) Causal model used for mediation analysis. Arrows represent causal directions. (H) Results from mediation analysis, with SOFA (left) and P/F ratios (right) as mediators. Lines indicate 95% confidence intervals. Solid and dotted lines represent estimates for the treatment (high SRSq) and control (low SRSq) conditions. ACME = Average Causal Mediation Effect; ADE = Average Direct Effect; p = mediation p value. (I) Correlation between SRSq-associated mRNA (x axis) and protein (y axis) changes. Dark red indicates the protein is significantly associated with SRSq. Cor = Pearson correlation; p = correlation p value.

References

    1. GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396:1204–1222.
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. 2017;17:407–420.
    1. Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, Fleischmann-Struzek C, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395:200–211.
    1. Sandhu C, Qureshi A, Emili A. Panomics for Precision Medicine. Trends Mol Med. 2018;24:85–101.
    1. Davenport EE, Burnham KL, Radhakrishnan J, Humburg P, Hutton P, Mills TC, Rautanen A, Gordon AC, Garrard C, Hill AVS, Hinds CJ, 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. Burnham KL, Davenport EE, Radhakrishnan J, Humburg P, Gordon AC, Hutton P, Svoren-Jabalera E, Garrard C, Hill AVS, Hinds CJ, Knight JC. 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. Scicluna BP, van Vught LA, Zwinderman AH, Wiewel MA, Davenport EE, Burnham KL, NÜrnberg P, Schultz MJ, Horn J, Cremer OL, Bonten MJ, et al. MARS consortium, Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir Med. 2017;5:816–826.
    1. Sweeney TE, Perumal TM, Henao R, Nichols M, Howrylak JA, Choi AM, Bermejo-Martin JF, Almansa R, Tamayo E, Davenport EE, Burnham KL, et al. A community approach to mortality prediction in sepsis via gene expression analysis. Nat Commun. 2018;9:694.
    1. Baghela A, Pena OM, Lee AH, Baquir B, Falsafi R, An A, Farmer SW, Hurlburt A, Mondragon-Cardona A, Rivera JD, Baker A, et al. Predicting sepsis severity at first clinical presentation: The role of endotypes and mechanistic signatures. EBioMedicine. 2022:103776.
    1. Seymour CW, Kennedy JN, Wang S, Chang CCH, Elliott CF, Xu Z, Berry S, Clermont G, Cooper G, Gomez H, Huang DT, et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA. 2019;321:2003–2017.
    1. Wong HR, Cvijanovich N, Allen GL, Lin R, Anas N, Meyer K, Freishtat RJ, Monaco M, Odoms K, Sakthivel B, Shanley TP. Genomics of Pediatric SIRS/Septic Shock Investigators, Genomic expression profiling across the pediatric systemic inflammatory response syndrome, sepsis, and septic shock spectrum. Crit Care Med. 2009;37:1558–1566.
    1. Banerjee S, Mohammed A, Wong HR, Palaniyar N, Kamaleswaran R. Machine Learning Identifies Complicated Sepsis Course and Subsequent Mortality Based on 20 Genes in Peripheral Blood Immune Cells at 24 H Post-ICU Admission. Front Immunol. 2021;12:592303.
    1. Wong HR, Caldwell JT, Cvijanovich NZ, Weiss SL, Fitzgerald JC, Bigham MT, Jain PN, Schwarz A, Lutfi R, Nowak J, Allen GL, et al. Prospective clinical testing and experimental validation of the Pediatric Sepsis Biomarker Risk Model. Sci Transl Med. 2019;11
    1. Antcliffe DB, Burnham KL, Al-Beidh F, Santhakumaran S, Brett SJ, Hinds CJ, Ashby D, Knight JC, Gordon AC. Transcriptomic Signatures in Sepsis and a Differential Response to Steroids. From the VANISH Randomized Trial. Am J Respir Crit Care Med. 2019;199:980–986.
    1. Haghverdi L, Lun ATL, Morgan MD, Marioni JC. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat Biotechnol. 2018;36:421–427.
    1. Rangel-Frausto MS, Pittet D, Costigan M, Hwang T, Davis CS, Wenzel RP. The natural history of the systemic inflammatory response syndrome (SIRS) A prospective study JAMA. 1995;273:117–123.
    1. Rhee C, Klompas M. Elucidating the Spectrum of Disease Severity Encompassed by Sepsis. JAMA Netw Open. 2022;5:e2147888.
    1. Pearl J. Interpretation and identification of causal mediation. Psychol Methods. 2014;19:459–481.
    1. Imai K, Keele L, Tingley D. A general approach to causal mediation analysis. Psychol Methods. 2010;15:309–334.
    1. COMBAT Consortium. A blood atlas of COVID-19 defines hallmarks of disease severity and specificity. Cell. 2022;185:916–938.
    1. Parnell GP, Tang BM, Nalos M, Armstrong NJ, Huang SJ, Booth DR, Mc AS. Identifying key regulatory genes in the whole blood of septic patients to monitor underlying immune dysfunctions. Shock. 2013;40:166–174.
    1. Gajic O, Ahmad SR, Wilson ME, Kaufman DA. Outcomes of critical illness: what is meaningful. Curr Opin Crit Care. 2018;24:394–400.
    1. Dunning J, Blankley S, Hoang LT, Cox M, Graham CM, James PL, Bloom CI, Chaussabel D, Banchereau J, Brett SJ, Moffatt MF, et al. Progression of whole-blood transcriptional signatures from interferon-induced to neutrophil-associated patterns in severe influenza. Nat Immunol. 2018;19:625–635.
    1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche J-D, Coopersmith CM, Hotchkiss RS, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) JAMA. 2016;315:801–810.
    1. Aschenbrenner AC, Mouktaroudi M, Krämer B, Oestreich M, Antonakos N, Nuesch-Germano M, Gkizeli K, Bonaguro L, Reusch N, Baßler K, Saridaki M, Knoll R, et al. German COVID-19 Omics Initiative (DeCOI), Disease severity-specific neutrophil signatures in blood transcriptomes stratify COVID-19 patients. Genome Med. 2021;13:7.
    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. Olwal CO, Nganyewo NN, Tapela K, Djomkam Zune AL, Owoicho O, Bediako Y, Duodu S. Parallels in Sepsis and COVID-19 Conditions: Implications for Managing Severe COVID-19. Front Immunol. 2021;12:602848.
    1. Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med. 2014;20:195–203.
    1. Clermont G, Bartels J, Kumar R, Constantine G, Vodovotz Y, Chow C. In silico design of clinical trials: a method coming of age. Crit Care Med. 2004;32:2061–2070.
    1. Cockrell RC, An G. Examining the controllability of sepsis using genetic algorithms on an agent-based model of systemic inflammation. PLoS Comput Biol. 2018;14:e1005876.
    1. Kwok AJ, Allcock A, Ferreira RC, Smee M, Cano-Gamez E, Burnham KL, Zurke YX, McKechnie S, Monaco C, Udalova I, Hinds CJ, et al. Identification of deleterious neutrophil states and altered granulopoiesis in sepsis. medRxiv. 2022
    1. Lelubre C, Vincent J-L. Mechanisms and treatment of organ failure in sepsis. Nat Rev Nephrol. 2018;14:417–427.
    1. Inouye M, Silander K, Hamalainen E, Salomaa V, Harald K, Jousilahti P, Männistö S, Eriksson JG, Saarela J, Ripatti S, Perola M, et al. An Immune Response Network Associated with Blood Lipid Levels. PLoS Genet. 2010;6:e1001113.
    1. Mayerle J, den Hoed CM, Schurmann C, Stolk L, Homuth G, Peters MJ, Capelle LG, Zimmermann K, Rivadeneira F, Gruska S, Völzke H, et al. Identification of genetic loci associated with Helicobacter pylori serologic status. JAMA. 2013;309:1912–1920.
    1. Westra H-J, Peters MJ, Esko T, Yaghootkar H, Schurmann C, Kettunen J, Christiansen MW, Fairfax BP, Schramm K, Powell JE, Zhernakova A, et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet. 2013;45:1238–1243.
    1. Aguirre-Gamboa R, de Klein N, di Tommaso J, Claringbould AM, van der Wijst G, de Vries D, Brugge H, Oelen R, Võsa U, Zorro MM, Chu X, et al. Deconvolution of bulk blood eQTL effects into immune cell subpopulations. BMC Bioinformatics. 2020;21:243.
    1. Netea MG, Joosten LAB, Li Y, Kumar V, Oosting M, Smeekens S, Jaeger M, Ter Horst R, Schirmer M, Vlamakis H, Notebaart R, et al. Understanding human immune function using the resources from the Human Functional Genomics Project. Nat Med. 2016;22:831–833.
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    1. Fang H, Knezevic B, Burnham KL, Knight JC. XGR software for enhanced interpretation of genomic summary data, illustrated by application to immunological traits. Genome Med. 2016;8:129.

Source: PubMed

3
Iratkozz fel