Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 1.
Published in final edited form as: Curr Opin Crit Care. 2021 Dec 1;27(6):717–725. doi: 10.1097/MCC.0000000000000883

Measuring the human immune response to surgery: multiomics for the prediction of postoperative outcomes

Franck Verdonk 1, Jakob Einhaus 1, Amy S Tsai 1, Julien Hedou 1, Benjamin Choisy 1, Dyani Gaudilliere 2, Cindy Kin 2, Nima Aghaeepour 1,3,4, Martin S Angst 1, Brice Gaudilliere 1
PMCID: PMC8585713  NIHMSID: NIHMS1739262  PMID: 34545029

Abstract

Purpose of review:

Postoperative complications including infections, cognitive impairment, and protracted recovery occur in one-third of the 300 million surgeries performed annually worldwide. Complications cause personal suffering along with a significant economic burden on our healthcare system. However, the accurate prediction of postoperative complications and patient-targeted interventions for their prevention remain as major clinical challenges.

Recent findings:

While multifactorial in origin, the dysregulation of immunological mechanisms that occur in response to surgical trauma is a key determinant of postoperative complications. Prior research, primarily focusing on inflammatory plasma markers, has provided important clues regarding their pathogenesis. However, the recent advent of high-content, single-cell transcriptomic and proteomic technologies has considerably improved our ability to characterize the immune response to surgery, thereby providing new means to understand the immunological basis of postoperative complications and to identify prognostic biological signatures.

Summary

The comprehensive and single-cell characterization of the human immune response to surgery has significantly advanced our ability to predict the risk of postoperative complications. Multiomic modeling of patients’ immune states holds promise for the discovery of preoperative predictive biomarkers, ultimately providing patients and surgeons with actionable information to improve surgical outcomes. While recent studies have generated a wealth of knowledge, laying the foundation for a single-cell atlas of the human immune response to surgery, larger-scale multiomic studies are required to derive robust, scalable, and sufficiently powerful models to accurately predict the risk of postoperative complications in individual patients.

Keywords: Immunology, Multiomics, Surgery, Complication, Trajectory

Introduction

More than 300 million surgeries are performed annually worldwide [1]. Postoperative complications, such as infections, cognitive impairment, or prolonged recovery occur in approximately 30% of surgeries [24], resulting in personal suffering, longer hospital stays, mortality, and significant socioeconomic burden [5,6]. Sufficiently powerful paradigms predicting the risk of postoperative complications would allow for patient-tailored preoperative clinical interventions [7,8], optimization of surgery timing, and selection of the most appropriate surgical approach to mitigate such risk [9,10]. However, current metrics and tools for risk estimation, such as the American Society of Anesthesiologists (ASA) classification, the American College of Surgeons (ACS) NSQIP, or the POSSUM scoring system, show poor to moderate performance as indicated by an area under the receiver operating curve of less than 0.75 [11]. Importantly, these metrics on clinical, demographic, or routine laboratory data do not objectively capture biological factors that drive surgical recovery. In particular, key characteristics of a patient’s immune system, which is at the intersection of most, if not all, biological mechanisms that drive surgical recovery [12], are not included in current prediction metrics and algorithms.

Surgery is associated with significant tissue trauma, triggering a complex inflammatory response that engages the innate and adaptive branches of the immune system [13,14]. Complications, including end-organ damage, infections, and protracted recovery, arise as pro-inflammatory and immunosuppressive responses tilt out of balance [15]. While conventional biological research has primarily focused on the analyses of soluble plasma factors, the development of new technologies that provide high-content data, such as proteomics (proteins) [16], transcriptomics (RNA-sequencing) [17], metabolomics (metabolites) [18], and cytomics (single-cell transcriptomic or proteomic analyses) [19], has created new and powerful avenues for the system-level assessment of the human immune response to surgery and the identification of biological events that drive, and are predictive of, surgical complications.

Here, we will summarize the current understanding of perioperative immunology. We will first discuss key innate and adaptive immune mechanisms implicated in the immune response to surgical trauma and summarize prior evidence of immune dysregulation driving complications. Finally, we will highlight recent technological advances that offer a promising approach for biomarker discovery and the development of accurate algorithms for predicting and, ultimately, preventing postoperative complications.

The immune response to surgical injury, a finely tuned immune balance

In the early postoperative period, tissue injury and other surgical trauma evoke a physiological local and systemic inflammatory response that is essential for pathogen defense as well as wound healing. The immune response to surgery is a finely-tuned balance between proinflammatory immune cell responses, including humoral factors (e.g. cytokines and the complement system) and immunosuppressive mechanisms engaged at the local and systemic level. Dysregulation in either direction increases the risk of postoperative complications: overt proinflammatory immune responses that lead to systemic inflammatory response syndrome (SIRS) are associated with severe morbidity [20] while profound postoperative immunosuppression may predispose a patient to postsurgical wound infection and bacterial sepsis [13,15,21,22].

Physiologically, protein and non-protein components of intracellular compartments and extracellular matrix act as biochemical alarmins for the immune system and are recognized as damage-associated molecular patterns (DAMPs) by pattern recognition receptors (PRRs), such as toll-like receptors (TLRs), expressed primarily on innate immune cell types (e.g. macrophages, monocytes, dendritic cells, and neutrophils) [23,24]. Endogenous ligands, including high-mobility group box 1 protein (HMGB1), adenosine triphosphate (ATP), heat-shock proteins (HSP), or hyaluronic acid, are released from the surgical injury site and bind to TLR2, TLR4, the receptor for advanced glycation end products (RAGE), and modulating receptors that amplify PRR signaling, such as triggering receptor expressed on myeloid cells 1 (TREM-1) [2527]. Upon activation by DAMPs, TLRs signal mainly through a MyD88-dependent pathway via IRAK1/4 and TRAF6, and, to a lesser extent, via the alternative adaptor protein TRIF, which targets the transcription factor IRF [23]. Downstream of MyD88-dependent signaling, mitogen-activated protein kinases (MAPKs) and IκB kinase (IKK) ultimately promote NF-κB and AP-1 mediated transcription [28] (Figure 1). Subsequently, proinflammatory cytokines IL-1β, IL-6, IL-8, IL-12, and TNFα, chemokines, growth factors, enzymes (e.g. metalloproteases, cyclooxygenases, or nitric oxide synthase), and other mediators of inflammation are released to kickstart an immediate response of the innate immune system, endothelial cells, and tissue adjacent to the surgical trauma in order to remove damaged tissue, to initiate wound healing, and to prevent pathogen intrusion [29].

Figure 1. Balance of proinflammatory and immunosuppressive features in the physiological immune response to surgery.

Figure 1

Alarmins/danger associated molecular patterns (DAMPs) such as HMGB1, ATP, heat shock proteins or hyaluronic acid are released from damaged tissue at the surgery site and recognized by pattern recognition receptors on immune cells. Proinflammatory activity of neutrophils, monocytes, macrophages and dendritic cells with the production of IL-1β, IL-6, IL-8, IL-12, and TNFα is counterbalanced by immunosuppressive processes, such as the induction of Tregs and promotion of Th2 cell immunity, and humoral factors (e.g. IL-10, TGF-β and IL-4).

Simultaneously, postoperative inflammatory signals are accompanied by anti-inflammatory and immunosuppressive adaptations. Numerically and functionally, adaptive immune cells, particularly T lymphocytes, are suppressed in the early postoperative period [30]. While multiple subsets of the T cell population, including Th1 cells and cytotoxic T cells, have been found to be decreased, activity and survival of immunosuppressive regulatory T cells (Tregs) increase upon DAMP signaling [3133]. Interestingly, postoperative downregulation of Th1 activity, but not Th2 activity, results in a shift towards a Th2-emphasized immune polarization with increased production of immunosuppressive cytokines, such as IL-4 and IL-10 [34,35]. The suppression of T cell function may be linked to the rise of myeloid-derived suppressor cells (MDSCs) in the postoperative period: the induction of arginase 1 (ARG1) in this innate immune cell subset with powerful immunosuppressive capacity leads to an arginine-deficient state inhibitory to normal T cell function [22,36]. MDSCs are induced in a STAT3-dependent manner by inflammatory mediators in the early postoperative period, as their marked expansion profoundly dampens CD4+ and CD8+ T cell responses, promotes the activity of Tregs, and leads to a rise in anti-inflammatory cytokine levels. Paradoxically, MDSCs have been shown in animal models of acute inflammation, such as burns and sepsis, to promote the activation of certain innate immune cell subsets, suggesting that inhibition of MDSCs could be detrimental to surgical recovery [37,38].

Humoral immune factors as drivers of postoperative complications, do they tell the whole story?

The dysregulation of perioperative immune mechanisms that drive recovery after surgery is an essential pathophysiological component of postoperative complications, such as infection, protracted recovery, or cognitive decline. For the past decades, many studies have focused on the assessment of circulating humoral factors, such as cytokines and other serum or plasma inflammatory mediators to identify dysregulated immune responses associated with surgical complications. As humoral factors often exert pleiotropic cellular effects, these studies have provided important insight into pro- and anti-inflammatory mechanisms associated with postoperative complications. For example, high levels of IL-6 induce a Th17/Treg imbalance, as IL-6 promotes specific differentiation of naïve CD4+ T cells into Th17 cells [39], inhibits TGF-β-induced Treg differentiation [40], and can contribute to insufficient immune response to infection [41]. An early postoperative increase in IL-10 is also well known to induce immunosuppression by altering the monocyte activation through STAT3-dependent expression of SOCS3 [42] and sequestering of HLA-DR within cells [43]. A recent study identified a regulatory loop in which IL-10 directly restricts CD8+ T cell activation thereby contributing to the development of infections [44]. Many clinical and experimental studies implicated IL-10 as an inhibitor of the deleterious effects of the innate pro-inflammatory immune response in a Th1-dominated environment unless the balance is shifted towards a Th2 response [45].

Circulating humoral factors have also long been targets in the search for diagnostic or predictive biomarkers of infection or sepsis following surgery. Postoperative cytokine levels correlate with the duration and the extent of surgery [46]. Logically, several circulating cytokines (such as IL-6, IL-1α/β, and IL-10) have been suggested as useful markers for predicting the development of postoperative complications [47]. Elevated levels of these cytokines on postoperative day one have been associated with infection following colorectal surgery [4850] and bone tumor surgery [51], overall postoperative complications in patients with gastric cancer [52], and longer ICU and hospital stays. In a study of genetic predisposition to postoperative sepsis, patients with an AA homozygous genotype of a TNF-α−308 G/A polymorphism in the TNF-α-gene were more likely to develop sepsis compared to non-AA homozygous individuals; the authors attributed this association to an increased capacity to produce proinflammatory TNF-α, IL-6, and IL-8 [53] (Figure 2).

Figure 2. Pathogenesis of adverse surgical outcomes.

Figure 2

Preoperative factors, such as individual susceptibility due to genetic or lifestyle factors, surgical triggers, such as laparoscopic or open approach, or duration of surgery, influence the degree of tissue damage. Following surgical injury, innate immune cells are activated to secrete cytokines, triggering a cascade of immunological events, such as Treg/Th17 imbalance, CD8+ T cell activation, and HLA-DR sequestration. The nuanced individual differences in each of these pre- and perioperative factors and processes in turn determine whether patients will suffer from postoperative complications.

Another promising biomarker of postoperative complications is HMGB1, which is both passively released into the circulation from the nucleus of traumatized necrotic cells and rapidly secreted by stimulated monocytes, macrophages, dendritic cells, and natural killer cells [54] in response to surgery. In observational clinical studies, high serum HMGB1 concentrations within the first hours after surgery was associated with a significantly increased risk of developing complications, including long-term cognitive impairment [55] and acute lung injury [56]. HMGB1 triggers the nuclear translocation of NF-κB in monocytes, which activates the release of pro-inflammatory cytokines, including IL-6 and IL-1β [57]. Multiple preclinical studies have shown that systemic anti-HMGB1 antibody treatment exerts neuroprotective effects by lowering systemic and hippocampal inflammatory responses to surgery and, hence, prevents the development of cognitive impairment [58,59].

While the postoperative patterns of multiple cytokines (e.g. IL-1β, IL-6, IL-10) and alarmins (HMGB1) have been associated with adverse postoperative outcomes, only IL-6 levels have been associated with surgical complications when measured before surgery: elevated IL-6 plasma levels before surgery were observed to forecast adverse postoperative outcomes, including delirium onset [60], survival [61], and acute kidney injury [62]. Interestingly, the COVID-19 pandemic provided an illustration of the consequences of preoperative immune alterations on postoperative outcomes. When invading its target cells, SARS-CoV2 induces an interferon (IFN)-like response and release of key pro-inflammatory cytokines, the “cytokine storm”, such as IFNɣ, IL-1β, IL-33, and IL-6 [63], which are associated with sustained functional changes in circulating immune cells [6466]. In a large international prospective cohort study, a greater risk of postoperative morbidity and mortality was observed in patients diagnosed with SARS-CoV-2 infection within six weeks prior to surgery (odds ratio > 2.8) [67]. This finding emphasized the link between the preoperative immune environment and postoperative prognosis.

Together, human studies of plasma factors have highlighted important immune dysfunctions that differentiate patients with and without postoperative complications. However, measurement of these factors alone has yielded only weak to moderate associations with clinical outcomes (e.g. AUC < 0.73) [68]. As such, few discoveries have translated into clinically relevant predictive tools. A major impediment has been the lack of high-content, functional assays that can integrate the assessment of plasma factors with single-cell assessment of the complex, multi-cellular immune response to surgery.

Towards an integrated, multiomic, predictive model of postoperative complications

High-content -omic technologies, together with recent computational advances that integrate transcriptomic, proteomic, and metabolomic data have enabled the immune system-wide characterization of human diseases with unprecedented depth and single-cell resolution. Application of these technologies to study the human immune response to surgery is increasingly recognized as a promising approach for the identification of patient-specific immune states predictive of postoperative complications (Figure 3).

Figure 3. Multiomic prediction of postoperative outcomes.

Figure 3

Surgical patients are recruited from major medical centers. 1) Whole blood samples are collected and processed for immunome, transcriptome, proteome, and metabolome analysis. 2) Machine learning algorithms applied to individual data layers predict surgical outcomes. The combined multiomic model shows increased predictive power (black) when compared to individual -omics. 3) Patients are stratified into poor or favorable surgical outcomes based on model output.

Single-cell technologies, such as mass cytometry, or single-cell RNA sequencing are particularly valuable, as the data generated holds substantially more predictive information than the clinical immunological laboratory results often collected before surgery. For example, mass cytometry can quantify over 50 proteomic parameters on a cell-by-cell basis, with a throughput allowing for the single-cell analysis of millions of cells per sample [69]. As such, mass cytometry allows in-depth characterization of the entire immune system both phenotypically and functionally, including the simultaneous identification of multiple immune cell subsets with quantification of activity (endogenous) and reactivity (response to proinflammatory stimuli) of key signaling pathways to produce a set of biological features predictive of the postoperative immune response [13].

Recent studies in patients undergoing total hip arthroplasties have applied this technology to identify perioperative immune signatures of surgical recovery. Specifically, increased immunosuppressive cellular activity preoperatively, including elevated pSTAT3 and pCREB activity within myeloid (M)-MDSCs, was predictive of longer-lasting functional impairment, fatigue, and pain after surgery [13,70]. Mass cytometry is also a valuable tool to profile the effects of perioperative therapeutic interventions on multiple immune cell subsets. As an example, preoperative administration of steroids is common to decrease postoperative nausea and vomiting; its effectiveness to improve surgical recovery, however, remains controversial. In a randomized controlled trial of methylprednisolone administration, mass cytometry analysis revealed major immune cell signaling alterations postoperatively that were predominantly confined to the adaptive immune compartment, while innate immune markers previously associated with surgical recovery were largely preserved in patients who received methylprednisolone [71,72].

While mass cytometry and other single-cell proteomic approaches require the a priori selection of a restricted set of analytes, transcriptomic platforms can offer untargeted analyses. RNA-sequencing (RNAseq) allows for a transcriptome-wide analysis of immune cell activity by detection of messenger, micro, transfer, and ribosomal RNA from the bulk analysis of pooled (bulk RNAseq) or single (single-cell RNAseq) peripheral immune cells. In fully unsupervised analyses of immune cell transcriptional activity in the context of surgery, RNAseq effectively predicted patients who developed pouchitis after ileal pouch–anal anastomosis [73], as well as individuals who developed postoperative recurrence of Crohn’s disease [74].

Circulating proteins and metabolites released from immune and non-immune cells are components of the regulatory network connecting immune cells with other biological systems implicated in the physiological response to surgery (e.g. neuro-endocrine and cardiovascular systems). Therefore, examination of the plasma proteome and metabolome are essential elements of the multiomic analysis of local and systemic immune responses predictive of postoperative complications. Recent developments in antibody-based (Olink [75]) or aptamer-based (Somalogic [76]) platforms have revolutionized the field of proteomics, allowing for analysis of up to 7000 plasma proteins. Application of high-content proteomic platforms to analyze patient samples collected before and after surgery has enabled the identification of predictive signatures of postoperative complications, increased length of stay, and risk of discharge to post-acute facility [77]. In addition, targeted and untargeted metabolomic platforms profile the molecular end-products of cell biological processes and provide a readout of cellular activity on a biochemical level. As such, a recent metabolomic study characterizing altered metabolite levels of anaerobic glycolysis as well as amino acid, nitrogenous, lipid, and bile acid metabolism after surgery identified metabolic signatures of postoperative complications following liver transplantation [78].

Integrating the knowledge garnered from individual -omic technologies to generate a holistic understanding of all immunological processes implicated in the pathogenesis of postoperative complications creates fundamental challenges in the bioinformatics field. Combining transcriptomic, proteomic, metabolomic, and mass cytometry features into multiomic predictive models requires the development of new statistical tools specifically adapted to high-dimensional datasets, which often contain more features than the number of samples collected: a computational dilemma referred to as “the curse of dimensionality” [79]. Regularized regression methods such as the Elastic Net (EN) algorithm are useful for the identification of predictive models of clinical outcomes and selection of key predictive features while accounting for the dimensionality of the dataset. Application of an EN algorithm was highlighted in a multiomic study that combined mass cytometry with plasma proteomics analysis of peripheral blood samples to characterize the immunological properties of a nutritional intervention known to reduce the incidence of postoperative complications (an Arginine-rich supplement) in patients undergoing abdominal surgery [80]. In addition, recent application of a stacked generalization algorithm that combines multiple regularized regression models built on individual -omic datasets has developed as a valuable approach for -omic data integration, while improving overall predictive performance [81]. The stacked generalization algorithm and other multiomic data integration methods have been successfully implemented in several recent studies for the prediction of various clinical outcomes, including the development of insulin resistance [82], the onset of spontaneous labor [83], preterm birth [84], survival in pancreatic cancer [85], and COVID-19 severity [86]. Cost and access to technology are clear limitations of multiomic approaches which have thus far often restricted the study sample size. However, multiomic approaches provide the means to cast an exploratory net and “find the needle in a haystack” for the validation of reduced predictive models that contain a limited number of features in large prospective studies.

Conclusion

A detailed understanding of the human immune response to surgery is a necessary first step towards identification of mechanistic biomarkers for the accurate prediction of postoperative complications. Recent technological advances allowing for an immune system-wide assessment of single-cell events in patients undergoing surgery form the foundation for a comprehensive map of the pre- and postoperative immune states predictive of surgical recovery. Multiomic modeling in studies integrating mass cytometry, plasma proteomic, metabolomic, and transcriptomic data hold promise for the discovery of pre-operative signatures predictive of postoperative outcomes. However, large-scale multiomic studies, as well as innovative machine learning approaches [87], are urgently needed to test the generalizability of identified signatures and to improve the performance of predictive models. Importantly, as most surgeries are non-urgent and have flexibility in timing and approach, the accurate prediction of postoperative complications can guide clinical decision making and the implementation of pre-operative interventions, such as prehabilitation programs or targeted immune therapeutics, to improve patient outcomes after surgery.

Key points:

  • Surgery induces significant tissue trauma that triggers a complex immune reaction with a finely-tuned balance of pro- and anti-inflammatory response mechanisms; however, imbalanced immune activity after surgery is associated with postoperative complications.

  • Postoperative patterns of multiple humoral factors have been associated with adverse postoperative outcomes, but only IL-6 levels have been associated with surgical complications when measured before surgery.

  • Innovative single-cell technologies (e.g. mass cytometry and single-cell RNA sequencing) that allow in-depth characterization of the entire immune system recently demonstrated an association between increased preoperative immunosuppressive cellular activity and longer-lasting functional impairment, fatigue, and pain after surgery.

  • Recent developments in high-content proteomic platforms that analyze patient samples collected before and after surgery have enabled the identification of predictive signatures of postoperative complications.

  • Large multiomic analyses are needed for the characterization of integrated biological signatures that can predict poor surgical outcomes and ultimately guide clinical decisions, including the implementation of preoperative interventions to improve surgical outcomes.

Financial support and sponsorship

This work was supported by the National Institute of Health (NIH) R35GM137936, the Fondation des Gueules Cassées, the Société Française d’Anesthésie-Réanimation (SFAR), the France-Stanford Center For Interdisciplinary Studies, the Doris Duke Charitable Foundation (2018100), the Center for Human Systems Immunology; the Stanford Maternal and Child Health Research Institute; R35GM138353; AG058417, HL13984401, NS114926, DA050960, AG065744

Footnotes

Conflicts of interest

The authors declare no conflicts of interest.

References

  • 1.Meara JG, Leather AJM, Hagander L, Alkire BC, Alonso N, Ameh EA, Bickler SW, Conteh L, Dare AJ, Davies J, et al. : Global Surgery 2030: Evidence and solutions for achieving health, welfare, and economic development. Lancet 2015, doi: 10.1016/S0140-6736(15)60160-X. [DOI] [PubMed] [Google Scholar]
  • 2.Healey MA, Shackford SR, Osler TM, Rogers FB, Burns E: Complications in surgical patients. In Archives of Surgery.. 2002. [DOI] [PubMed] [Google Scholar]
  • 3.Storesund A, Haugen AS, Hjortås M, Nortvedt MW, Flaatten H, Eide GE, Boermeester MA, Sevdalis N, Søfteland E: Accuracy of surgical complication rate estimation using ICD-10 codes. Br J Surg 2019, doi: 10.1002/bjs.10985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Spence J, LeManach Y, Chan MTV, Wang CY, Sigamani A, Xavier D, Pearse R, Alonso-Coello P, Garutti I, Srinathan SK, et al. : Association between complications and death within 30 days after noncardiac surgery. CMAJ 2019, doi: 10.1503/cmaj.190221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Healy MA, Mullard AJ, Campbell DA, Dimick JB: Hospital and payer costs associated with surgical complications. In JAMA Surgery.. 2016. [DOI] [PubMed] [Google Scholar]
  • 6.Gou RY, Hshieh TT, Marcantonio ER, Cooper Z, Jones RN, Travison TG, Fong TG, Abdeen A, Lange J, Earp B, et al. : One-Year Medicare Costs Associated with Delirium in Older Patients Undergoing Major Elective Surgery. JAMA Surg 2021, doi: 10.1001/jamasurg.2020.7260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Waterland JL, McCourt O, Edbrooke L, Granger CL, Ismail H, Riedel B, Denehy L: Efficacy of Prehabilitation Including Exercise on Postoperative Outcomes Following Abdominal Cancer Surgery: A Systematic Review and Meta-Analysis. Front Surg 2021, doi: 10.3389/fsurg.2021.628848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Humeidan ML, Reyes JPC, Mavarez-Martinez A, Roeth C, Nguyen CM, Sheridan E, Zuleta-Alarcon A, Otey A, Abdel-Rasoul M, Bergese SD: Effect of Cognitive Prehabilitation on the Incidence of Postoperative Delirium among Older Adults Undergoing Major Noncardiac Surgery: The Neurobics Randomized Clinical Trial. JAMA Surg 2021, doi: 10.1001/jamasurg.2020.4371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Caroff DA, Chan C, Kleinman K, Calderwood MS, Wolf R, Wick EC, Platt R, Huang S: Association of Open Approach vs Laparoscopic Approach With Risk of Surgical Site Infection After Colon Surgery. JAMA Netw open 2019, doi: 10.1001/jamanetworkopen.2019.13570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Miller LE, Gondusky JS, Bhattacharyya S, Kamath AF, Boettner F, Wright J: Does Surgical Approach Affect Outcomes in Total Hip Arthroplasty Through 90 Days of Follow-Up? A Systematic Review With Meta-Analysis. J Arthroplasty 2018, doi: 10.1016/j.arth.2017.11.011. [DOI] [PubMed] [Google Scholar]
  • 11.Eamer G, Al-Amoodi MJH, Holroyd-Leduc J, Rolfson DB, Warkentin LM, Khadaroo RG: Review of risk assessment tools to predict morbidity and mortality in elderly surgical patients. Am J Surg 2018, doi: 10.1016/j.amjsurg.2018.04.006. [DOI] [PubMed] [Google Scholar]
  • 12.Linn BS, Jensen J: Age and Immune Response to a Surgical Stress. Arch Surg 1983, doi: 10.1001/archsurg.1983.01390040017004. [DOI] [PubMed] [Google Scholar]
  • 13.Gaudilliere B, Fragiadakis GK, Bruggner R V., Nicolau M, Finck R, Tingle M, Silva J, Ganio EA, Yeh CG, Maloney WJ, et al. : Clinical recovery from surgery correlates with single-cell immune signatures. Sci Transl Med 2014, 6:255ra131–255ra131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Choileain NN, Redmond HP: Cell response to surgery. Arch Surg 2006, doi: 10.1001/archsurg.141.11.1132. [DOI] [PubMed] [Google Scholar]
  • 15.Angele MK, Faist E: Clinical review: Immunodepression in the surgical patient and increased susceptibility to infection. Crit Care 2002, doi: 10.1186/cc1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tomic V, Russwurm S, Möller E, Claus RA, Blaess M, Brunkhorst F, Bruegel M, Bode K, Bloos F, Wippermann J, et al. : Transcriptomic and proteomic patterns of systemic inflammation in on-pump and off-pump coronary artery bypass grafting. Circulation 2005, doi: 10.1161/CIRCULATIONAHA.104.531152. [DOI] [PubMed] [Google Scholar]
  • 17.Poitou C, Perret C, Mathieu F, Truong V, Blum Y, Durand H, Alili R, Chelghoum N, Pelloux V, Aron-Wisnewsky J, et al. : Bariatric surgery induces disruption in inflammatory signaling pathways mediated by immune cells in adipose tissue: A RNA-seq study. PLoS One 2015, doi: 10.1371/journal.pone.0125718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Parent BA, Seaton M, Sood RF, Gu H, Djukovic D, Raftery D, O’Keefe GE: Use of metabolomics to trend recovery and therapy after injury in critically ill trauma patients. JAMA Surg 2016, doi: 10.1001/jamasurg.2016.0853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gaudilliere B, Angst MS, Hotchkiss RS: Deep Immune Profiling in Trauma and Sepsis: Flow Is the Way to Go! Crit Care Med 2017, doi: 10.1097/CCM.0000000000002594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sato Y, Kotani T: Systemic inflammatory response syndrome; SIRS. Respir Circ 2008, doi: 10.4161/viru.27135. [DOI] [Google Scholar]
  • 21.White M, Martin-Loeches I, Lawless MW, O’Dwyer MJ, Doherty DG, Young V, Kelleher D, McManus R, Ryan T: Hospital-acquired pneumonia after lung resection surgery is associated with characteristic cytokine gene expression. Chest 2011, doi: 10.1378/chest.10-0016. [DOI] [PubMed] [Google Scholar]
  • 22.Marik PE, Flemmer M: The immune response to surgery and trauma: Implications for treatment. J Trauma Acute Care Surg 2012, doi: 10.1097/TA.0b013e318265cf87. [DOI] [PubMed] [Google Scholar]
  • 23.Midwood KS, Piccinini AM: DAMPening inflammation by modulating TLR signalling. Mediators Inflamm 2010, doi: 10.1155/2010/672395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Gong T, Liu L, Jiang W, Zhou R: DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol 2020, doi: 10.1038/s41577-019-0215-7. ** A review highlighting the recent discoveries concerning the roles of DAMP-sensing receptors in sterile inflammation and in diseases resulting from dysregulated sterile inflammation.
  • 25.Scaffidi P, Misteli T, Bianchi ME: Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
  • 26.Chan JK, Roth J, Oppenheim JJ, Tracey KJ, Vogl T, Feldmann M, Horwood N, Nanchahal J: Alarmins: awaiting a clinical response. J Clin Invest 2012, 122:2711–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Denning NL, Aziz M, Gurien SD, Wang P: Damps and nets in sepsis. Front Immunol 2019, doi: 10.3389/fimmu.2019.02536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Akira S, Takeda K: Toll-like receptor signalling. Nat Rev Immunol 2004, doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 29.Huber-Lang M, Lambris JD, Ward PA: Innate immune responses to trauma. Nat Immunol 2018, doi: 10.1038/s41590-018-0064-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hogan B V, Peter MB, Shenoy HG, Horgan K, Hughes TA: Surgery induced immunosuppression. Surgeon 2011, doi: 10.1016/j.surge.2010.07.011. [DOI] [PubMed] [Google Scholar]
  • 31.Ogawa K, Hirai M, Katsube T, Murayama M, Hamaguchi K, Shimakawa T, Naritake Y, Hosokawa T, Kajiwara T: Suppression of cellular immunity by surgical stress. Surgery 2000, doi: 10.1067/msy.2000.103498. [DOI] [PubMed] [Google Scholar]
  • 32.MacConmara MP, Maung AA, Fujimi S, McKenna AM, Delisle A, Lapchak PH, Rogers S, Lederer JA, Mannick JA: Increased CD4+ CD25+ T regulatory cell activity in trauma patients depresses protective Th1 immunity. Ann Surg 2006, doi: 10.1097/01.sla.0000239031.06906.1f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wild CA, Bergmann C, Fritz G, Schuler P, Hoffmann TK, Lotfi R, Westendorf A, Brandau S, Lang S: HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. Int Immunol 2012, doi: 10.1093/intimm/dxs051. [DOI] [PubMed] [Google Scholar]
  • 34.O’Sullivan ST, Lederer JA, Horgan AF, Chin DHL, Mannick JA, Rodrick ML, Meakins JL, Baker CC, Pruitt BA, Galandiuk S: Major injury leads to predominance of the T helper-2 lymphocyte phenotype and diminished interleukin-12 production associated with decreased resistance to infection. In Annals of Surgery.. 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Brune IB, Wilke W, Hensler T, Holzmann B, Siewert JR: Downregulation of T helper type 1 immune response and altered pro- inflammatory and anti-inflammatory T cell cytokine balance following conventional but not laparoscopic surgery. Am J Surg 1999, doi: 10.1016/S0002-9610(98)00299-2. [DOI] [PubMed] [Google Scholar]
  • 36.Zhu X, Herrera G, Ochoa JB: Immunosupression and infection after major surgery: A nutritional deficiency. Crit Care Clin 2010, doi: 10.1016/j.ccc.2010.04.004. [DOI] [PubMed] [Google Scholar]
  • 37.Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, Blosser RL, Tam AJ, Bruno T, Zhang H, et al. : STAT3 regulates arginase-i in myeloid-derived suppressor cells from cancer patients. J Clin Invest 2013, doi: 10.1172/JCI60083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cuenca AG, Delano MJ, Kelly-Scumpia KM, Moreno C, Scumpia PO, LaFace DM, Heyworth PG, Efron PA, Moldawer LL: A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol Med 2011, 17:281–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Korn T, Bettelli E, Oukka M, Kuchroo VK: IL-17 and Th17 cells. Annu Rev Immunol 2009, doi: 10.1146/annurev.immunol.021908.132710. [DOI] [PubMed] [Google Scholar]
  • 40.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK: Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 41.Zgodziński W, Grywalska E, Siwicka-Gieroba D, Surdacka A, Zinkiewicz K, Szczepanek D, Wallner G, Roliński J: The clinical importance of changes in Treg and Th17 lymphocyte subsets in splenectomized patients after spleen injury. Adv Clin Exp Med 2018, doi: 10.17219/acem/73734. [DOI] [PubMed] [Google Scholar]
  • 42.Williams LM, Ricchetti G, Sarma U, Smallie T, Foxwell BMJ: Interleukin-10 suppression of myeloid cell activation - A continuing puzzle. Immunology 2004, doi: 10.1111/j.1365-2567.2004.01988.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fumeaux T, Pugin J: Role of interleukin-10 in the intracellular sequestration of human leukocyte antigen-DR in monocytes during septic shock. Am J Respir Crit Care Med 2002, doi: 10.1164/rccm.200203-217OC. [DOI] [PubMed] [Google Scholar]
  • 44.Smith LK, Boukhaled GM, Condotta SA, Mazouz S, Guthmiller JJ, Vijay R, Butler NS, Bruneau J, Shoukry NH, Krawczyk CM, et al. : Interleukin-10 Directly Inhibits CD8+ T Cell Function by Enhancing N-Glycan Branching to Decrease Antigen Sensitivity. Immunity 2018, doi: 10.1016/j.immuni.2018.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mingomataj E, Bakiri AH: Regulator Versus Effector Paradigm: Interleukin-10 as Indicator of the Switching Response. Clin Rev Allergy Immunol 2016, doi: 10.1007/s12016-015-8514-7. [DOI] [PubMed] [Google Scholar]
  • 46.Cruickshank AM, Fraser WD, Burns HJG, Van Damme J, Shenkin A: Response of serum interleukin-6 in patients undergoing elective surgery of varying severity. Clin Sci 1990, doi: 10.1042/cs0790161. [DOI] [PubMed] [Google Scholar]
  • 47.Oka Y, Murata A, Nishijima J, Yasuda T, Hiraoka N, Ohmachi Y, Kitagawa K, Yasuda T, Toda H, Tanaka N, et al. : Circulating interleukin 6 as a useful marker for predicting postoperative complications. Cytokine 1992, doi: 10.1016/1043-4666(92)90070-8. [DOI] [PubMed] [Google Scholar]
  • 48.Boersema GSA, Wu Z, Menon AG, Kleinrensink GJ, Jeekel J, Lange JF: Systemic inflammatory cytokines predict the infectious complications but not prolonged postoperative ileus after colorectal surgery. Mediators Inflamm 2018, doi: 10.1155/2018/7141342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Xie T, Zhao C, Ding C, Zhang L, Cheng M, Chun C, Yu W, Gong J, Zhu W: Postoperative Interleukin-6 Predicts Intra-abdominal Septic Complications at an Early Stage after Elective Intestinal Operation for Crohn’s Disease Patients. Inflamm Bowel Dis 2018, doi: 10.1093/ibd/izy090. [DOI] [PubMed] [Google Scholar]
  • 50.Rettig TCD, Verwijmeren L, Dijkstra IM, Boerma D, Van De Garde EMW, Noordzij PG: Postoperative interleukin-6 level and early detection of complications after elective major abdominal surgery. Ann Surg 2016, doi: 10.1097/SLA.0000000000001342. [DOI] [PubMed] [Google Scholar]
  • 51.Chen G: Dynamic relationship between postoperative infection and CRP, IL-6, and Livin in patients with bone tumors. Minerva Chir 2019, doi: 10.23736/S0026-4733.17.07321-7. [DOI] [PubMed] [Google Scholar]
  • 52.Szczepanik AM, Scislo L, Scully T, Walewska E, Siedlar M, Kolodziejczyk P, Lenart M, Rutkowska M, Galas A, Czupryna A, et al. : IL-6 serum levels predict postoperative morbidity in gastric cancer patients. Gastric Cancer 2011, doi: 10.1007/s10120-011-0039-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Baghel K, Srivastava RN, Chandra A, Goel SK, Agrawal J, Kazmi HR, Raj S: TNF-α, IL-6, and IL-8 Cytokines and Their Association with TNF-α−308 G/A Polymorphism and Postoperative Sepsis. J Gastrointest Surg 2014, doi: 10.1007/s11605-014-2574-5. [DOI] [PubMed] [Google Scholar]
  • 54.Ranzato E, Martinotti S, Patrone M: Emerging roles for HMGB1 protein in immunity, inflammation, and cancer. ImmunoTargets Ther 2015, doi: 10.2147/itt.s58064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lin GX, Wang T, Chen MH, Hu ZH, Ouyang W: Serum high-mobility group box 1 protein correlates with cognitive decline after gastrointestinal surgery. Acta Anaesthesiol Scand 2014, doi: 10.1111/aas.12320. [DOI] [PubMed] [Google Scholar]
  • 56.Yamaguchi K, Nakao S, Iwamoto H, Kagimoto A, Handa Y, Sakamoto S, Horimasu Y, Masuda T, Mimae T, Miyamoto S, et al. : Predictive role of circulatory HMGB1 in postoperative acute exacerbation of interstitial lung disease in lung cancer patients. Sci Rep 2021, doi: 10.1038/s41598-021-89663-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vacas S, Degos V, Tracey KJ, Maze M: High-mobility group box 1 protein initiates postoperative cognitive decline by engaging bone marrow-derived macrophages. Anesthesiology 2014, doi: 10.1097/ALN.0000000000000045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Terrando N, Yang T, Wang X, Fang J, Cao M, Andersson U, Harris HE, Ouyang W, Tong J: Systemic HMGB1 neutralization prevents postoperative neurocognitive dysfunction in aged rats. Front Immunol 2016, doi: 10.3389/fimmu.2016.00441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Terrando N, Eriksson LI, Kyu Ryu J, Yang T, Monaco C, Feldmann M, Jonsson Fagerlund M, Charo IF, Akassoglou K, Maze M: Resolving postoperative neuroinflammation and cognitive decline. Ann Neurol 2011, doi: 10.1002/ana.22664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Capri M, Yani SL, Chattat R, Fortuna D, Bucci L, Lanzarini C, Morsiani C, Catena F, Ansaloni L, Adversi M, et al. : Pre-Operative, High-IL-6 Blood Level is a Risk Factor of Post-Operative Delirium Onset in Old Patients. Front Endocrinol (Lausanne) 2014, 5:173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schuettfort VM, Pradere B, Trinh Q-D, D’Andrea D, Quhal F, Mostafaei H, Laukhtina E, Mori K, Sari Motlagh R, Rink M, et al. : Impact of preoperative plasma levels of interleukin 6 and interleukin 6 soluble receptor on disease outcomes after radical cystectomy for bladder cancer. Cancer Immunol Immunother 2021, doi: 10.1007/s00262-021-02953-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhang WR, Garg AX, Coca SG, Devereaux PJ, Eikelboom J, Kavsak P, McArthur E, Thiessen-Philbrook H, Shortt C, Shlipak M, et al. : Plasma IL-6 and IL-10 concentrations predict AKI and long-term mortality in adults after cardiac surgery. J Am Soc Nephrol 2015, doi: 10.1681/ASN.2014080764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, Ruiz C, Melguizo-Rodríguez L: SARS-CoV-2 infection: The role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev 2020, doi: 10.1016/j.cytogfr.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Arunachalam PS, Wimmers F, Mok CKP, Perera RAPM, Scott M, Hagan T, Sigal N, Feng Y, Bristow L, Tsang OTY, et al. : Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science (80- ) 2020, doi: 10.1126/SCIENCE.ABC6261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Carlson DE, Chiu WC, Fiedler SM, Hoffman GE: Central neural distribution of immunoreactive Fos and CRH in relation to plasma ACTH and corticosterone during sepsis in the rat. Exp Neurol 2007, 205:485–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wilk AJ, Rustagi A, Zhao NQ, Roque J, Martínez-Colón GJ, McKechnie JL, Ivison GT, Ranganath T, Vergara R, Hollis T, et al. : A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med 2020, doi: 10.1038/s41591-020-0944-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Timing of surgery following SARS-CoV-2 infection: an international prospective cohort study. Anaesthesia 2021, doi: 10.1111/anae.15458. *Important clinical study in COVID-19 setting showing that pre-operative SARS-CoV-2 infection increases the risk of postoperative mortality and pulmonary complications
  • 68.Lv X-C, Lin Y, Wu Q, Wang L, Hou Y, Dong Y, Chen L: Plasma interleukin-6 is a potential predictive biomarker for postoperative delirium among acute type a aortic dissection patients treated with open surgical repair. J Cardiothorac Surg 2021, 16:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hartmann FJ, Bendall SC: Immune monitoring using mass cytometry and related high-dimensional imaging approaches. Nat Rev Rheumatol 2020, doi: 10.1038/s41584-019-0338-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Fragiadakis GK, Gaudillière B, Ganio EA, Aghaeepour N, Tingle M, Nolan GP, Angst MS: Patient-specific Immune States before Surgery Are Strong Correlates of Surgical Recovery. Anesthesiology 2015, 123:1241–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Ganio EA, Stanley N, Lindberg-Larsen V, Einhaus J, Tsai AS, Verdonk F, Culos A, Gahemi S, Rumer KK, Stelzer IA, et al. : Preferential inhibition of adaptive immune system dynamics by glucocorticoids in patients after acute surgical trauma. Nat Commun 2020, 11:1–12. ** A randomized controlled trial in which the authors employ an original high-dimensional mass cytometry approach to characterize innate and adaptive cell signaling dynamics after a major surgery.
  • 72.Stanley N, Stelzer IA, Tsai AS, Fallahzadeh R, Ganio E, Becker M, Phongpreecha T, Nassar H, Ghaemi S, Maric I, et al. : VoPo leverages cellular heterogeneity for predictive modeling of single-cell data. Nat Commun 2020, doi: 10.1038/s41467-020-17569-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Devlin JC, Axelrad J, Hine AM, Chang S, Sarkar S, Lin J Da, Ruggles KV, Hudesman D, Cadwell K, Loke P: Single-Cell Transcriptional Survey of Ileal-Anal Pouch Immune Cells From Ulcerative Colitis Patients. Gastroenterology 2021, doi: 10.1053/j.gastro.2020.12.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cushing KC, McLean R, McDonald KG, Gustafsson JK, Knoop KA, Kulkarni DH, Sartor RB, Newberry RD: Predicting risk of postoperative disease recurrence in Crohn’s disease: Patients with indolent Crohn’s disease have distinct whole transcriptome profiles at the time of first surgery. Inflamm Bowel Dis 2019, doi: 10.1093/ibd/izy228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kvedaraite E, Hertwig L, Sinha I, Ponzetta A, Myrberg IH, Lourda M, Dzidic M, Akber M, Klingström J, Folkesson E, et al. : Major alterations in the mononuclear phagocyte landscape associated with COVID-19 severity. Proc Natl Acad Sci U S A 2021, doi: 10.1073/pnas.2018587118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Williams SA, Kivimaki M, Langenberg C, Hingorani AD, Casas JP, Bouchard C, Jonasson C, Sarzynski MA, Shipley MJ, Alexander L, et al. : Plasma protein patterns as comprehensive indicators of health. Nat Med 2019, doi: 10.1038/s41591-019-0665-2. * Proof-of-concept study demonstrating that protein expression patterns reliably encode for many different health issues.
  • 77.Fong TG, Chan NY, Dillon ST, Zhou W, Tripp B, Ngo LH, Otu HH, Inouye SK, Vasunilashorn SM, Cooper Z, et al. : Identification of Plasma Proteome Signatures Associated With Surgery Using SOMAscan. Ann Surg 2021, doi: 10.1097/SLA.0000000000003283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Attard JA, Dunn WB, Mergental H, Mirza DF, Afford SC, Perera MTPR: Systematic Review: Clinical Metabolomics to Forecast Outcomes in Liver Transplantation Surgery. Omi A J Integr Biol 2019, doi: 10.1089/omi.2019.0086. [DOI] [PubMed] [Google Scholar]
  • 79.Keogh E, Mueen A: Curse of Dimensionality. In Encyclopedia of Machine Learning and Data Mining.. 2017. [Google Scholar]
  • 80.Aghaeepour N, Kin C, Ganio EA, Jensen KP, Gaudilliere DK, Tingle M, Tsai A, Lancero HL, Choisy B, McNeil LS, et al. : Deep Immune Profiling of an Arginine-Enriched Nutritional Intervention in Patients Undergoing Surgery. J Immunol 2017, 199:2171–2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ghaemi MS, DiGiulio DB, Contrepois K, Callahan B, Ngo TTM, Lee-Mcmullen B, Lehallier B, Robaczewska A, McIlwain D, Rosenberg-Hasson Y, et al. : Multiomics modeling of the immunome, transcriptome, microbiome, proteome and metabolome adaptations during human pregnancy. Bioinformatics 2019, doi: 10.1093/bioinformatics/bty537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Schüssler-Fiorenza Rose SM, Contrepois K, Moneghetti KJ, Zhou W, Mishra T, Mataraso S, Dagan-Rosenfeld O, Ganz AB, Dunn J, Hornburg D, et al. : A longitudinal big data approach for precision health. Nat Med 2019, doi: 10.1038/s41591-019-0414-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Stelzer IA, Ghaemi MS, Han X, Ando K, Hédou JJ, Feyaerts D, Peterson LS, Rumer KK, Tsai ES, Ganio EA, et al. : Integrated trajectories of the maternal metabolome, proteome, and immunome predict labor onset. Sci Transl Med 2021, doi: 10.1126/scitranslmed.abd9898. ** Study laying the groundwork for developing blood-based methods for predicting medical endpoints such as the day of labor, anchored in physiological mechanisms.
  • 84.Jehan F, Sazawal S, Baqui AH, Nisar MI, Dhingra U, Khanam R, Ilyas M, Dutta A, Mitra DK, Mehmood U, et al. : Multiomics Characterization of Preterm Birth in Low-and Middle-Income Countries. JAMA Netw Open 2020, doi: 10.1001/jamanetworkopen.2020.29655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Baek B, Lee H: Prediction of survival and recurrence in patients with pancreatic cancer by integrating multi-omics data. Sci Rep 2020, doi: 10.1038/s41598-020-76025-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Feyaerts D, Hedou J, Gillard J, Chen H, Tsai ES, Peterson LS, Ando K, Manohar M, Do E, Dhondalay GKR, et al. : Integrated plasma proteomic and single-cell immune signaling network signatures demarcate mild, moderate, and severe COVID-19. bioRxiv Prepr Serv Biol 2021, doi: 10.1101/2021.02.09.430269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Culos A, Tsai AS, Stanley N, Becker M, Ghaemi MS, McIlwain DR, Fallahzadeh R, Tanada A, Nassar H, Espinosa C, et al. : Integration of mechanistic immunological knowledge into a machine learning pipeline improves predictions. Nat Mach Intell 2020, doi: 10.1038/s42256-020-00232-8. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES