Early immune markers of clinical, virological, and immunological outcomes in patients with COVID-19: a multi-omics study

Background: The great majority of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections are mild and uncomplicated, but some individuals with initially mild COVID-19 progressively develop more severe symptoms. Furthermore, there is substantial heterogeneity in SARS-CoV-2-specific memory immune responses following infection. There remains a critical need to identify host immune biomarkers predictive of clinical and immunological outcomes in SARS-CoV-2-infected patients. Methods: Leveraging longitudinal samples and data from a clinical trial (N=108) in SARS-CoV-2-infected outpatients, we used host proteomics and transcriptomics to characterize the trajectory of the immune response in COVID-19 patients. We characterized the association between early immune markers and subsequent disease progression, control of viral shedding, and SARS-CoV-2-specific T cell and antibody responses measured up to 7 months after enrollment. We further compared associations between early immune markers and subsequent T cell and antibody responses following natural infection with those following mRNA vaccination. We developed machine-learning models to predict patient outcomes and validated the predictive model using data from 54 individuals enrolled in an independent clinical trial. Results: We identify early immune signatures, including plasma RIG-I levels, early IFN signaling, and related cytokines (CXCL10, MCP1, MCP-2, and MCP-3) associated with subsequent disease progression, control of viral shedding, and the SARS-CoV-2-specific T cell and antibody response measured up to 7 months after enrollment. We found that several biomarkers for immunological outcomes are shared between individuals receiving BNT162b2 (Pfizer–BioNTech) vaccine and COVID-19 patients. Finally, we demonstrate that machine-learning models using 2–7 plasma protein markers measured early within the course of infection are able to accurately predict disease progression, T cell memory, and the antibody response post-infection in a second, independent dataset. Conclusions: Early immune signatures following infection can accurately predict clinical and immunological outcomes in outpatients with COVID-19 using validated machine-learning models. Funding: Support for the study was provided from National Institute of Health/National Institute of Allergy and Infectious Diseases (NIH/NIAID) (U01 AI150741-01S1 and T32-AI052073), the Stanford’s Innovative Medicines Accelerator, National Institutes of Health/National Institute on Drug Abuse (NIH/NIDA) DP1DA046089, and anonymous donors to Stanford University. Peginterferon lambda provided by Eiger BioPharmaceuticals.

[1]  J. Andrews,et al.  TNF-α+ CD4+ T cells dominate the SARS-CoV-2 specific T cell response in COVID-19 outpatients and are associated with durable antibodies , 2022, Cell Reports Medicine.

[2]  Mark M. Davis,et al.  Early non-neutralizing, afucosylated antibody responses are associated with COVID-19 severity , 2022, Science Translational Medicine.

[3]  H. Hedlin,et al.  Favipiravir for Treatment of Outpatients With Asymptomatic or Uncomplicated Coronavirus Disease 2019: A Double-Blind, Randomized, Placebo-Controlled, Phase 2 Trial , 2021, medRxiv.

[4]  Michael I. Mandel,et al.  Waning Immunity after the BNT162b2 Vaccine in Israel , 2021, The New England journal of medicine.

[5]  Mark M. Davis,et al.  Systems vaccinology of the BNT162b2 mRNA vaccine in humans , 2021, Nature.

[6]  L. Nolen,et al.  COVID-19 Vaccine Breakthrough Infections Reported to CDC — United States, January 1–April 30, 2021 , 2021, MMWR. Morbidity and mortality weekly report.

[7]  Lucas T. Graybuck,et al.  Longitudinal immune dynamics of mild COVID-19 define signatures of recovery and persistence , 2021, bioRxiv.

[8]  Shenmin Zhang,et al.  Divergent early antibody responses define COVID-19 disease trajectories , 2021, bioRxiv.

[9]  A. Takaoka,et al.  RIG-I triggers a signaling-abortive anti-SARS-CoV-2 defense in human lung cells , 2021, Nature Immunology.

[10]  Frances E. Muldoon,et al.  Single-cell multi-omics analysis of the immune response in COVID-19 , 2021, Nature Medicine.

[11]  M. Davenport,et al.  Evolution of immune responses to SARS-CoV-2 in mild-moderate COVID-19 , 2021, Nature communications.

[12]  Xiang Wang Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. , 2021, The New England journal of medicine.

[13]  W. Lau,et al.  Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19 , 2021, Cell.

[14]  J. Glenn,et al.  Peginterferon lambda for the treatment of outpatients with COVID-19: a phase 2, placebo-controlled randomised trial , 2021, The Lancet Respiratory Medicine.

[15]  Bjoern Peters,et al.  Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection , 2021, Science.

[16]  K. Blennow,et al.  Proteomic blood profiling in mild, severe and critical COVID-19 patients , 2020, Scientific Reports.

[17]  T. Tabuchi,et al.  Coronavirus Disease , 2021, Encyclopedia of the UN Sustainable Development Goals.

[18]  Aaron J. Wilk,et al.  Multi-omic profiling reveals widespread dysregulation of innate immunity and hematopoiesis in COVID-19 , 2020, bioRxiv.

[19]  J. Casanova,et al.  Life-Threatening COVID-19: Defective Interferons Unleash Excessive Inflammation , 2020, Med.

[20]  L. Notarangelo,et al.  An immune-based biomarker signature is associated with mortality in COVID-19 patients , 2020, JCI insight.

[21]  H. Hedlin,et al.  Peginterferon Lambda-1a for treatment of outpatients with uncomplicated COVID-19: a randomized placebo-controlled trial , 2020, Nature Communications.

[22]  L. Carter,et al.  Functional SARS-CoV-2-Specific Immune Memory Persists after Mild COVID-19 , 2020, Cell.

[23]  S. Ladhani,et al.  Robust SARS-CoV-2-specific T-cell immunity is maintained at 6 months following primary infection , 2020, bioRxiv.

[24]  A. Casto,et al.  Dynamics of Neutralizing Antibody Titers in the Months After Severe Acute Respiratory Syndrome Coronavirus 2 Infection , 2020, The Journal of infectious diseases.

[25]  Barbara B. Shih,et al.  Genetic mechanisms of critical illness in COVID-19 , 2020, Nature.

[26]  K. To,et al.  COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing , 2020, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[27]  Madeleine K. D. Scott,et al.  Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans , 2020, Science.

[28]  Alexander Sczyrba,et al.  Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment , 2020, Cell.

[29]  Eric Song,et al.  Longitudinal analyses reveal immunological misfiring in severe COVID-19 , 2020, Nature.

[30]  Yang Wu,et al.  SARS-CoV-2 infection induces sustained humoral immune responses in convalescent patients following symptomatic COVID-19 , 2020, Nature Communications.

[31]  Laura J. Simpson,et al.  A single-cell atlas of the peripheral immune response in patients with severe COVID-19 , 2020, Nature Medicine.

[32]  Akiko Iwasaki,et al.  Type I and Type III Interferons – Induction, Signaling, Evasion, and Application to Combat COVID-19 , 2020, Cell Host & Microbe.

[33]  G. Gao,et al.  Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19 , 2020, Journal of Allergy and Clinical Immunology.

[34]  Nathaniel Hupert,et al.  Clinical Characteristics of Covid-19 in New York City , 2020, The New England journal of medicine.

[35]  Zunyou Wu,et al.  Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese Center for Disease Control and Prevention. , 2020, JAMA.

[36]  A. Butte,et al.  xCell: digitally portraying the tissue cellular heterogeneity landscape , 2017, bioRxiv.

[37]  A. Egli,et al.  Interferon Lambda: Modulating Immunity in Infectious Diseases , 2017, Front. Immunol..

[38]  K. Rajarathnam,et al.  Chemokine CXCL1 mediated neutrophil recruitment: Role of glycosaminoglycan interactions , 2016, Scientific Reports.

[39]  Alexey Sergushichev,et al.  An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation , 2016 .

[40]  G. Sutter,et al.  CCL2 expression is mediated by type I IFN receptor and recruits NK and T cells to the lung during MVA infection , 2016, Journal of leukocyte biology.

[41]  Lior Pachter,et al.  Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.

[42]  J. Erjefält,et al.  The neutrophil-recruiting chemokine GCP-2/CXCL6 is expressed in cystic fibrosis airways and retains its functional properties after binding to extracellular DNA , 2015, Mucosal Immunology.

[43]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[44]  Sandra Romero-Steiner,et al.  Molecular signatures of antibody responses derived from a systems biological study of 5 human vaccines , 2013, Nature Immunology.

[45]  Herwig P. Moll,et al.  The differential activity of interferon-α subtypes is consistent among distinct target genes and cell types , 2011, Cytokine.

[46]  Christopher J. Obara,et al.  Chemokine Receptor Ccr2 Is Critical for Monocyte Accumulation and Survival in West Nile Virus Encephalitis , 2011, The Journal of Immunology.

[47]  T. Matsumiya,et al.  Function and regulation of retinoic acid-inducible gene-I. , 2010, Critical reviews in immunology.

[48]  Yi Li,et al.  Type I interferon modulates monocyte recruitment and maturation in chronic inflammation. , 2009, The American journal of pathology.

[49]  E. Pamer Tipping the balance in favor of protective immunity during influenza virus infection , 2009, Proceedings of the National Academy of Sciences.

[50]  S. Dower,et al.  Acceleration of Human Neutrophil Apoptosis by TRAIL1 , 2003, The Journal of Immunology.

[51]  Ming Xu,et al.  DNA fragmentation in apoptosis , 2000, Cell Research.

[52]  T. Ley,et al.  DFF45/ICAD can be directly processed by granzyme B during the induction of apoptosis. , 2000, Immunity.

[53]  R. Bravo,et al.  Defects in Macrophage Recruitment and Host Defense in Mice Lacking the CCR2 Chemokine Receptor , 1997, The Journal of experimental medicine.

[54]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .