Preferential inhibition of adaptive immune system dynamics by glucocorticoids in patients after acute surgical trauma

Edward A Ganio, Natalie Stanley, Viktoria Lindberg-Larsen, Jakob Einhaus, Amy S Tsai, Franck Verdonk, Anthony Culos, Sajjad Ghaemi, Kristen K Rumer, Ina A Stelzer, Dyani Gaudilliere, Eileen Tsai, Ramin Fallahzadeh, Benjamin Choisy, Henrik Kehlet, Nima Aghaeepour, Martin S Angst, Brice Gaudilliere, Edward A Ganio, Natalie Stanley, Viktoria Lindberg-Larsen, Jakob Einhaus, Amy S Tsai, Franck Verdonk, Anthony Culos, Sajjad Ghaemi, Kristen K Rumer, Ina A Stelzer, Dyani Gaudilliere, Eileen Tsai, Ramin Fallahzadeh, Benjamin Choisy, Henrik Kehlet, Nima Aghaeepour, Martin S Angst, Brice Gaudilliere

Abstract

Glucocorticoids (GC) are a controversial yet commonly used intervention in the clinical management of acute inflammatory conditions, including sepsis or traumatic injury. In the context of major trauma such as surgery, concerns have been raised regarding adverse effects from GC, thereby necessitating a better understanding of how GCs modulate the immune response. Here we report the results of a randomized controlled trial (NCT02542592) in which we employ a high-dimensional mass cytometry approach to characterize innate and adaptive cell signaling dynamics after a major surgery (primary outcome) in patients treated with placebo or methylprednisolone (MP). A robust, unsupervised bootstrap clustering of immune cell subsets coupled with random forest analysis shows profound (AUC = 0.92, p-value = 3.16E-8) MP-induced alterations of immune cell signaling trajectories, particularly in the adaptive compartments. By contrast, key innate signaling responses previously associated with pain and functional recovery after surgery, including STAT3 and CREB phosphorylation, are not affected by MP. These results imply cell-specific and pathway-specific effects of GCs, and also prompt future studies to examine GCs' effects on clinical outcomes likely dependent on functional adaptive immune responses.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Study workflow.
Fig. 1. Study workflow.
a In a double-blind study, patients were randomized to receive a single preoperative dose of 125 mg methylprednisolone (MP, n = 28 patients) or saline placebo (n = 30 patients). Peripheral blood and clinical outcomes data were collected prior to surgery (baseline) and at the indicated time points after surgery. After erythrocyte lysis, peripheral immune cells were barcoded, stained with cell-phenotyping and intracellular cell-signaling antibodies, and analyzed by mass cytometry. Unsupervised bootstrapped clustering of immune cell subsets followed by random forest analysis was performed to identify differential immune cell dynamics in MP vs. control groups. b A non-linear dimensional reduction algorithm (Isomap) showing individual patients’ immunological trajectories after surgery along the innate (X) and adaptive (Y) axes (MP in red, control in black). Snapshots are shown for the 1 h, 6 h, 24 h, 48 h and 2 wk time points (animated trajectories can be found in Supplementary Movie 1). Right panel. Overview of median trajectories for the MP and control groups are shown in red and black.
Fig. 2. A high-resolution atlas detailing peripheral…
Fig. 2. A high-resolution atlas detailing peripheral immune cell alterations by MP after surgery.
a Immune cells were clustered based on the expression of all phenotypic markers using an unsupervised bootstrapped clustering algorithm. The clusters were projected into two dimensions and major immune cell compartments were identified based on phenotypic marker expression (contoured in orange/green for innate/adaptive immune compartments, respectively). b Univariate p-values (two-sided Wilcoxon Rank Sum Test) were computed for each cluster at each time point to quantify the difference in functional marker expression or cell frequency between samples in the control (n = 30 patients) and MP (n = 28 patients) groups. At each time point, clusters were colored by the best univariate p-value observed for cell frequency and functional marker expression. c A Random Forest model was trained to classify patients in the control or MP group at each timepoint based on cluster-derived cell frequency and intracellular signaling responses. The boxplot depicts the probability predicted by the Random Forest model that samples from patients in the control (gray) or MP (blue) group were allocated to the MP group. The model revealed that samples from placebo- or MP-treated patients were distinguishable at 1 h (AUC = 0.91, p = 1.03E−7), 6 h (AUC = 0.92, p = 3.16E−8), 24 h (AUC = 0.85, p = 3.81E−6), and 48 h (AUC = 0.76, p = 3.2E−3) after surgery (two-sided Wilcoxon rank-sum test, p-values calculated for each unique model). All boxplots show median values, interquartile range, whiskers of 1.5 times interquartile range.
Fig. 3. Alteration of innate and adaptive…
Fig. 3. Alteration of innate and adaptive immune cell frequencies by MP.
a Immune cell atlas depicting differences in cell frequency between the MP and control groups at 1, 6 and 24 h after surgery (expressed as % of CD45+ mononuclear cells, with the exception of neutrophils, which are expressed as % of total live cells). Cell clusters are color-coded according to the directional differences between the control (n = 30 patients) and MP (n = 28 patients) group. Directional differences were computed using a two-sided Wilcoxon rank-sum test (sign(r) -log(p-value), blue/red indicating an increased/decreased frequency in the MP group; arrows point at cell clusters that differ most significantly between the patient groups). In cell clusters of the adaptive immune compartment (contoured in green), MP treatment resulted in decreased frequencies of CD4+Tmem at 1 and 6 h and CD4+Tnaïve cells at 6 h, but no significant changes in CD8+T or B cell frequencies. In cell clusters of the innate immune compartment (contoured in orange), MP treatment resulted in decreased frequencies of cMCs and M-MDSCs at 1 h, and ncMCs and mDCs at 1 and 6 h. In contrast, MP resulted in increased frequencies of CD56loCD16+NK cells at 1 h and mDCs at 24 h. b, c Box-plots depicting the frequency of manually gated immune cell subsets corroborating observations contained in the immune cell atlas. Immune cell subsets for which MP’s effect on adaptive (b) and innate (c) immune cell frequencies were most pronounced (CD4+ Tmem, CD4+ Tnaive, cMCs) are shown. Neutrophil frequencies (not included in the immune cell atlas) are also shown. Box plots for all manually gated immune cells are available in Supplementary Fig. 4. All boxplots show median values, interquartile range, whiskers of 1.5 times interquartile range. (Two-sided Wilcoxon rank-sum test, *p < 0.01, **p < 0.001, ***p < 0.0001). Exact p-values are available in Supplementary Table 2.
Fig. 4. Alteration of intracellular pSTAT3 responses…
Fig. 4. Alteration of intracellular pSTAT3 responses by MP.
a Immune cell atlas depicting differences of the phospho-(p)STAT3 response (arcsinh ratio) between the control (n = 30 patients) and MP (n = 28 patients) group at 1, 6, and 24 h after surgery relative to the preoperative time point. Blue/red cluster colors indicate increased/decreased signaling in the MP group, respectively (two-sided Wilcoxon rank-sum test). Arrows point at cell clusters that differ most significantly between the patient groups. In cell clusters of the adaptive compartment (contoured in green), MP treatment resulted in a sustained attenuation of pSTAT3 responses in CD4+T cells (first in Tbet+CD4+T cells at 1 h, then in CD4+ Tnaive and CD4+ Tmem cells at 6 and 24 h. In contrast, in clusters of the innate compartment (contoured in orange) MP treatment resulted in no significant changes in the pSTAT3 signal in monocyte subsets (including cMCs, intMCs, and M-MDSCs) and in only a modest increase in the pSTAT3 signal in CD56loCD16+NK cells at 1 and 6 h and ncMCs at 24 h. b, c Box plots depict the pSTAT3 signal in manually gated immune cell subsets corroborating observations contained in the immune atlas. b MP’s attenuation of the pSTAT3 signal was most pronounced in CD4+ T cell subsets. c MP does not attenuate the pSTAT3 signal in neutrophils (signal is increased at 24 h) or cMCs. All boxplots show median values, interquartile range, whiskers of 1.5 times interquartile range (two-sided Wilcoxon rank-sum test, *p < 0.01, **p < 0.001, ***p < 0.0001). Exact p-values are available in Supplementary Table 2.
Fig. 5. Alteration of total IκBα by…
Fig. 5. Alteration of total IκBα by MP.
a Immune cell atlas depicting differences in total IκBα (arcsinh ratio) between the control (n = 30 patients) and MP (n = 28 patients) group at 1, 6, and 24 h after surgery. Blue/red cluster colors indicate increased/decreased signaling in the MP group, respectively (two-sided Wilcoxon rank-sum test). MP treatment resulted in increased total IκBα in both the adaptive (CD4+Tmem, CD4+Tnaive, CD8+Tnaive and CD8+Tmem, contoured in green) and the innate compartment (CD56loCD16+ NK cells, mDCs, cMCs, ncMCs, M-MDSCs and DCs, contoured in orange, which was most prominent at 6 and 24 h after surgery. b, c Box-plots depict total IκBα in manually gated immune cell subsets corroborating observations contained in the immune atlas. Select immune cell subsets for which MP’s effect on total IκBα was most pronounced (CD8+ Tnaive and CD8+ Tmem, cMCs, and neutrophils) are shown. All boxplots show median values, interquartile range, and whiskers of 1.5 times interquartile range. (Two-sided Wilcoxon rank-sum test, *p < 0.01, **p < 0.001, ***p < 0.0001). Exact p-values are available in Supplementary Table 2.
Fig. 6. Clinical recovery outcome measures between…
Fig. 6. Clinical recovery outcome measures between control and MP treatment.
All boxplots show median values, interquartile range, whiskers of 1.5 times interquartile range of clinical recovery parameters (a) pain, (b) fatigue and daily functioning, and (c) functional impairment of the operated hip over the course of 28 days after surgery between control (n = 30 patients) and MP (n = 28 patients) groups (two-sided Wilcoxon rank sum test). Pain and functional impairment of the hip were assessed with an adapted version of the Western Ontario and McMaster Universities Arthritis Index (pain 0 to 40 = none to worst; function 0 to 60 = no to most severe impairment). Postoperative fatigue and daily functioning were assessed with the Surgical Recovery Scale (17 to 100 = worst to none).

References

    1. Lunn TH, Kehlet H. Perioperative glucocorticoids in hip and knee surgery - benefit vs. harm? A review of randomized clinical trials. Acta Anaesthesiol. Scand. 2013;57:823–834.
    1. Asehnoune K, Futier E, Feuillet F, Roquilly A, group, P. PACMAN trial protocol, Perioperative Administration of Corticotherapy on Morbidity and mortality After Non-cardiac major surgery: a randomised, multicentre, double-blind, superiority study. BMJ Open. 2019;9:e021262.
    1. Chatham WW, Kimberly RP. Treatment of lupus with corticosteroids. Lupus. 2001;10:140–147.
    1. Salerno A, Hermann R. Efficacy and safety of steroid use for postoperative pain relief. Update and review of the medical literature. J. Bone Jt. Surg. Am. 2006;88:1361–1372.
    1. Clark AR. Anti-inflammatory functions of glucocorticoid-induced genes. Mol. Cell Endocrinol. 2007;275:79–97.
    1. Coutinho AE, Chapman KE. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell Endocrinol. 2011;335:2–13.
    1. Toner AJ, Ganeshanathan V, Chan MT, Ho KM, Corcoran TB. Safety of perioperative glucocorticoids in elective noncardiac surgery: a systematic review and meta-analysis. Anesthesiology. 2017;126:234–248.
    1. Kehlet H, Lindberg-Larsen V. High-dose glucocorticoid before hip and knee arthroplasty: To use or not to use-that’s the question. Acta Orthopaedica. 2018;89:477–479.
    1. Ho CM, Ho ST, Wang JJ, Tsai SK, Chai CY. Dexamethasone has a central antiemetic mechanism in decerebrated cats. Anesthesia Analgesia. 2004;99:734–739.
    1. De Oliveira GS, Jr, Almeida MD, Benzon HT, McCarthy RJ. Perioperative single dose systemic dexamethasone for postoperative pain: a meta-analysis of randomized controlled trials. Anesthesiology. 2011;115:575–588.
    1. Holte K, Kehlet H. Perioperative single-dose glucocorticoid administration: pathophysiologic effects and clinical implications. J. Am. Coll. Surg. 2002;195:694–712.
    1. Anstead GM. Steroids, retinoids, and wound healing. Adv. Wound Care. 1998;11:277–285.
    1. Freeman, J. et al. Effect of perioperative lidocaine, propofol and steroids on pulmonary metastasis in a murine model of breast cancer surgery. Cancers10.3390/cancers11050613 (2019).
    1. Cain DW, Cidlowski JA. Immune regulation by glucocorticoids. Nat. Rev. Immunol. 2017;17:233–247.
    1. Fillinger MP, et al. Glucocorticoid effects on the inflammatory and clinical responses to cardiac surgery. J. Cardiothorac. Vasc. Anesthesia. 2002;16:163–169.
    1. Corcoran T, et al. Intraoperative dexamethasone alters immune cell populations in patients undergoing elective laparoscopic gynaecological surgery. Br. J. Anaesth. 2017;119:221–230.
    1. Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014;6:a016295.
    1. Yue C, Wei R, Liu Y. Perioperative systemic steroid for rapid recovery in total knee and hip arthroplasty: a systematic review and meta-analysis of randomized trials. J. Orthop. Surg. Res. 2017;12:100.
    1. Roquilly A, et al. Hydrocortisone prevents immunosuppression by interleukin-10+ natural killer cells after trauma-hemorrhage. Crit. Care Med. 2014;42:e752–e761.
    1. Seshadri A, et al. Phenotyping the immune response to trauma: a multiparametric systems immunology approach. Crit. care Med. 2017;45:1523–1530.
    1. Bendall SC, Nolan GP, Roederer M, Chattopadhyay PK. A deep profiler’s guide to cytometry. Trends Immunol. 2012;33:323–332.
    1. Bjornson ZB, Nolan GP, Fantl WJ. Single-cell mass cytometry for analysis of immune system functional states. Curr. Opin. Immunol. 2013;25:484–494.
    1. Gaudilliere B, et al. Clinical recovery from surgery correlates with single-cell immune signatures. Sci. Transl. Med. 2014;6:255ra131.
    1. Fragiadakis GK, et al. Patient-specific immune states before surgery are strong correlates of surgical recovery. Anesthesiology. 2015;123:1241–1255.
    1. Angele MK, Faist E. Clinical review: immunodepression in the surgical patient and increased susceptibility to infection. Crit. Care. 2002;6:298–305.
    1. Stoecklein VM, Osuka A, Lederer JA. Trauma equals danger–damage control by the immune system. J. Leukoc. Biol. 2012;92:539–551.
    1. Hall GM, Peerbhoy D, Shenkin A, Parker CJ, Salmon P. Relationship of the functional recovery after hip arthroplasty to the neuroendocrine and inflammatory responses. Br. J. Anaesth. 2001;87:537–542.
    1. Kehlet H, Wilmore DW. Evidence-based surgical care and the evolution of fast-track surgery. Ann. Surg. 2008;248:189–198.
    1. Ni Choileain N, Redmond HP. Cell response to surgery. Arch. Surg. 2006;141:1132–1140.
    1. Giannoudis PV, Dinopoulos H, Chalidis B, Hall GM. Surgical stress response. Injury. 2006;37(Suppl 5):S3–S9.
    1. Marik PE, Flemmer M. The immune response to surgery and trauma: implications for treatment. J. Trauma Acute Care Surg. 2012;73:801–808.
    1. Dabrowska AM, Slotwinski R. The immune response to surgery and infection. Cent. Eur. J. Immunol. 2014;39:532–537.
    1. Raymond, S. L. et al. Prospective validation of a transcriptomic metric in severe trauma. Annals Surgery, 10.1097/SLA.0000000000003204 (2019).
    1. Xiao W, et al. A genomic storm in critically injured humans. J. Exp. Med. 2011;208:2581–2590.
    1. Tarnok A, et al. Preoperative prediction of postoperative edema and effusion in pediatric cardiac surgery by altered antigen expression patterns on granulocytes and monocytes. Cytometry. 2001;46:247–253.
    1. Lindberg-Larsen V, Petersen PB, Jans O, Beck T, Kehlet H. Effect of pre-operative methylprednisolone on orthostatic hypotension during early mobilization after total hip arthroplasty. Acta Anaesthesiologica Scandinavica. 2018;62:882–892.
    1. Lindberg-Larsen V, Kehlet H, Bagger J, Madsbad S. Preoperative high-dose methylprednisolone and glycemic control early after total hip and knee arthroplasty: a randomized, double-blind, placebo-controlled trial. Anesthesia Analgesia. 2018;127:906–913.
    1. Aghaeepour, N. et al. Deep immune profiling of an arginine-enriched nutritional intervention in patients undergoing surgery. J. Immunol. 10.4049/jimmunol.1700421 (2017).
    1. Lindberg-Larsen V, et al. Preoperative methylprednisolone does not reduce loss of knee-extension strength after total knee arthroplastyA randomized, double-blind, placebo-controlled trial of 61 patients. Acta Orthopaedica. 2017;88:543–549.
    1. Lunn TH, et al. Effect of high-dose preoperative methylprednisolone on pain and recovery after total knee arthroplasty: a randomized, placebo-controlled trial. Br. J. Anaesth. 2011;106:230–238.
    1. Koh IJ, Chang CB, Lee JH, Jeon YT, Kim TK. Preemptive low-dose dexamethasone reduces postoperative emesis and pain after TKA: a randomized controlled study. Clin. Orthop. Relat. Res. 2013;471:3010–3020.
    1. Kardash KJ, Sarrazin F, Tessler MJ, Velly AM. Single-dose dexamethasone reduces dynamic pain after total hip arthroplasty. Anesthesia Analgesia. 2008;106:1253–1257.
    1. Bergeron SG, Kardash KJ, Huk OL, Zukor DJ, Antoniou J. Perioperative dexamethasone does not affect functional outcome in total hip arthroplasty. Clin. Orthop. Relat. Res. 2009;467:1463–1467.
    1. Zhu J, Paul WE. CD4 T cells: fates, functions, and faults. Blood. 2008;112:1557–1569.
    1. O’Shea JJ, et al. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu. Rev. Med. 2015;66:311–328.
    1. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat. Rev. Cancer. 2009;9:798–809.
    1. Halle S, Halle O, Forster R. Mechanisms and dynamics of T cell-mediated cytotoxicity in vivo. Trends Immunol. 2017;38:432–443.
    1. Leoni G, Neumann PA, Sumagin R, Denning TL, Nusrat A. Wound repair: role of immune-epithelial interactions. Mucosal Immunol. 2015;8:959–968.
    1. Nosbaum A, et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J. Immunol. 2016;196:2010–2014.
    1. Krall, J. A. et al. The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci. Transl. Med.10.1126/scitranslmed.aan3464 (2018).
    1. Jorgensen, C. C., Pitter, F. T., Kehlet, H. & Lundbeck Foundation Center for Fast-track Hip and Knee Replacement Collaborative Group. Safety aspects of preoperative high-dose glucocorticoid in primary total knee replacement. Br. J. Anaesth. 119, 267–275 (2017).
    1. Olnes MJ, et al. Effects of systemically administered hydrocortisone on the human immunome. Sci. Rep. 2016;6:23002.
    1. van de Garde MD, et al. Chronic exposure to glucocorticoids shapes gene expression and modulates innate and adaptive activation pathways in macrophages with distinct changes in leukocyte attraction. J. Immunol. 2014;192:1196–1208.
    1. Fay ME, et al. Cellular softening mediates leukocyte demargination and trafficking, thereby increasing clinical blood counts. Proc. Natl Acad. Sci. USA. 2016;113:1987–1992.
    1. Ronchetti, S., Ricci, E., Migliorati, G., Gentili, M. & Riccardi, C. How glucocorticoids affect the neutrophil life. Int. J. Mol. Sci. 10.3390/ijms19124090 (2018).
    1. Chen L, Jondal M, Yakimchuk K. Regulatory effects of dexamethasone on NK and T cell immunity. Inflammopharmacology. 2018;26:1331–1338.
    1. Herold MJ, McPherson KG, Reichardt HM. Glucocorticoids in T cell apoptosis and function. Cell. Mol. Life Sci. 2006;63:60–72.
    1. Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M. Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science. 1995;270:286–290.
    1. Oh KS, et al. Anti-inflammatory chromatinscape suggests alternative mechanisms of glucocorticoid receptor action. Immunity. 2017;47:298–309.e295.
    1. Christian, F., Smith, E. L. & Carmody, R. J. The regulation of NF-kappaB subunits by phosphorylation. Cells10.3390/cells5010012 (2016).
    1. Weikum ER, Knuesel MT, Ortlund EA, Yamamoto KR. Glucocorticoid receptor control of transcription: precision and plasticity via allostery. Nat. Rev. Mol. Cell Biol. 2017;18:159–174.
    1. Luecke HF, Yamamoto KR. The glucocorticoid receptor blocks P-TEFb recruitment by NFkappaB to effect promoter-specific transcriptional repression. Genes Dev. 2005;19:1116–1127.
    1. Petta I, et al. The Interactome of the glucocorticoid receptor and its influence on the actions of glucocorticoids in combatting inflammatory and infectious diseases. Microbiol. Mol. Biol. Rev. 2016;80:495–522.
    1. Murray PJ. The JAK-STAT signaling pathway: input and output integration. J. Immunol. 2007;178:2623–2629.
    1. Mozo L, Suarez A, Gutierrez C. Glucocorticoids up-regulate constitutive interleukin-10 production by human monocytes. Clin. Exp. Allergy. 2004;34:406–412.
    1. Nikolich-Zugich J. The twilight of immunity: emerging concepts in aging of the immune system. Nat. Immunol. 2018;19:10–19.
    1. Goronzy JJ, Weyand CM. Mechanisms underlying T cell ageing. Nat. Rev. Immunol. 2019;19:573–583.
    1. Kehlet, H. & Joshi, G. P. The systematic review/meta-analysis epidemic: a tale of glucocorticoid therapy in total knee arthroplasty. Anaesthesia, 10.1111/anae.14946 (2019).
    1. Jorgensen CC, Kehlet H. Role of patient characteristics for fast-track hip and knee arthroplasty. Br. J. Anaesth. 2013;110:972–980.
    1. Paddison JS, Sammour T, Kahokehr A, Zargar-Shoshtari K, Hill AG. Development and validation of the Surgical Recovery Scale (SRS) J. Surgical Res. 2011;167:e85–e91.
    1. Bellamy N, Buchanan WW, Goldsmith CH, Campbell J, Stitt LW. Validation study of WOMAC: a health status instrument for measuring clinically important patient relevant outcomes to antirheumatic drug therapy in patients with osteoarthritis of the hip or knee. J. Rheumatol. 1988;15:1833–1840.
    1. Bodenmiller B, et al. Multiplexed mass cytometry profiling of cellular states perturbed by small-molecule regulators. Nat. Biotechnol. 2012;30:858–867.
    1. Zunder ER, et al. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat. Protoc. 2015;10:316–333.
    1. Finck R, et al. Normalization of mass cytometry data with bead standards. Cytom. Part A: J. Int. Soc. Anal. Cytol. 2013;83:483–494.
    1. Maecker HT, McCoy JP, Nussenblatt R. Standardizing immunophenotyping for the Human Immunology Project. Nat. Rev. Immunol. 2012;12:191–200.
    1. Balasubramanian M, Schwartz EL. The isomap algorithm and topological stability. Science. 2002;295:7.
    1. Bruggner RV, Bodenmiller B, Dill DL, Tibshirani RJ, Nolan GP. Automated identification of stratifying signatures in cellular subpopulations. Proc. Natl Acad. Sci. USA. 2014;111:E2770–E2777.
    1. Van Gassen S, et al. FlowSOM: using self-organizing maps for visualization and interpretation of cytometry data. Cytom. Part A: J. Int. Soc. Anal. Cytol. 2015;87:636–645.
    1. Levine JH, et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell. 2015;162:184–197.
    1. Ho, T. K. Random decision forests. In Proceedings of 3rd international conference on document analysis and recognition Vol. 1, 278 Montreal, Quebec, Canada. (IEEE Computer Society, 1995).
    1. Stanley, N. et al. VoPo leverages cellular heterogeneity for predictive modeling of single-cell data. Nat. Commun. 10.1038/s41467-020-17569-8 (2020).

Source: PubMed

3
Iratkozz fel