1
|
Patel N, Surman SL, Jones BG, Penkert RR, Ringwald-Smith K, DeLuca K, Richardson J, Zheng Y, Tang L, Hurwitz JL. Randomized Controlled Clinical Trial of Pediatric Pneumococcus and Hepatitis A Vaccinations With or Without a High-Dose Oral Vitamin A Supplement. Biomolecules 2025; 15:540. [PMID: 40305237 PMCID: PMC12024622 DOI: 10.3390/biom15040540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Previous studies have shown that high-dose vitamin supplements can improve vaccine-induced immune responses and pathogen protection in the context of vitamin deficiencies. To further elucidate the influence of vitamin supplements on immune responses toward pediatric vaccines, we performed a randomized controlled clinical trial (PCVIT) of 20 healthy children 1-4 years of age in Memphis, Tennessee. Study participants received a booster vaccine for pneumococcus and a primary vaccine for hepatitis A virus with or without a high-dose, oral, liquid supplement of 10,000 IU retinyl palmitate. We found that the children enrolled in PCVIT had higher baseline vitamin levels than previously described older children and adults living in Memphis. Only one child in PCVIT had a serum retinol level of less than 0.3 µg/mL. The children frequently consumed milk and baby foods that were likely vitamin-fortified, providing an explanation for the relatively high vitamin levels. Most children in PCVIT responded well to pneumococcus and hepatitis A vaccines by pathogen-specific antibody upregulation. The one child with a serum retinol level below 0.3 µg/mL did not receive a vitamin supplement and exhibited the lowest fold-change in antibody responses toward pneumococcal serotypes. A correlation matrix encompassing demographics, vitamin levels, vaccine-induced immune responses, C-reactive protein, and total serum immunoglobulin isotypes, including IgG2 and IgA, identified variables associated with vaccination outcomes. Perhaps because children were predominantly retinol-sufficient at baseline, the high-dose vitamin A supplement exhibited no benefit to vaccine-induced immune responses. In fact, when vitamin supplemented and vitamin unsupplemented groups were compared among participants with the highest baseline retinol levels, there was a trend toward weaker vaccine-induced immune responses in the vitamin supplemented group. Results encourage the performance of larger clinical studies before high-dose vitamin supplements are recommended for populations that are otherwise vitamin-replete.
Collapse
Affiliation(s)
- Nehali Patel
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (N.P.); (S.L.S.); (B.G.J.); (K.D.)
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (N.P.); (S.L.S.); (B.G.J.); (K.D.)
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (N.P.); (S.L.S.); (B.G.J.); (K.D.)
| | - Rhiannon R. Penkert
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA;
| | - Karen Ringwald-Smith
- Department of Clinical Nutrition, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Kim DeLuca
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (N.P.); (S.L.S.); (B.G.J.); (K.D.)
| | - Julie Richardson
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Ying Zheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Z.); (L.T.)
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Z.); (L.T.)
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (N.P.); (S.L.S.); (B.G.J.); (K.D.)
| |
Collapse
|
2
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Shinde P, Willemsen L, Anderson M, Aoki M, Basu S, Burel JG, Cheng P, Ghosh Dastidar S, Dunleavy A, Einav T, Forschmiedt J, Fourati S, Garcia J, Gibson W, Greenbaum JA, Guan L, Guan W, Gygi JP, Ha B, Hou J, Hsiao J, Huang Y, Jansen R, Kakoty B, Kang Z, Kobie JJ, Kojima M, Konstorum A, Lee J, Lewis SA, Li A, Lock EF, Mahita J, Mendes M, Meng H, Neher A, Nili S, Olsen LR, Orfield S, Overton JA, Pai N, Parker C, Qian B, Rasmussen M, Reyna J, Richardson E, Safo S, Sorenson J, Srinivasan A, Thrupp N, Tippalagama R, Trevizani R, Ventz S, Wang J, Wu CC, Ay F, Grant B, Kleinstein SH, Peters B. Putting computational models of immunity to the test-An invited challenge to predict B.pertussis vaccination responses. PLoS Comput Biol 2025; 21:e1012927. [PMID: 40163550 PMCID: PMC11978014 DOI: 10.1371/journal.pcbi.1012927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 04/08/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Systems vaccinology studies have been used to build computational models that predict individual vaccine responses and identify the factors contributing to differences in outcome. Comparing such models is challenging due to variability in study designs. To address this, we established a community resource to compare models predicting B. pertussis booster responses and generate experimental data for the explicit purpose of model evaluation. We here describe our second computational prediction challenge using this resource, where we benchmarked 49 algorithms from 53 scientists. We found that the most successful models stood out in their handling of nonlinearities, reducing large feature sets to representative subsets, and advanced data preprocessing. In contrast, we found that models adopted from literature that were developed to predict vaccine antibody responses in other settings performed poorly, reinforcing the need for purpose-built models. Overall, this demonstrates the value of purpose-generated datasets for rigorous and open model evaluations to identify features that improve the reliability and applicability of computational models in vaccine response prediction.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lisa Willemsen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Michael Anderson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Minori Aoki
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Saonli Basu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Julie G. Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Peng Cheng
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Souradipto Ghosh Dastidar
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aidan Dunleavy
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Jamie Forschmiedt
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, Illinois, United States of America
| | - Javier Garcia
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - William Gibson
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, Maryland, United States of America
| | - Jason A. Greenbaum
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Weikang Guan
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Jeremy P. Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Brendan Ha
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jason Hsiao
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rick Jansen
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bhargob Kakoty
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zhiyu Kang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James J. Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mari Kojima
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Anna Konstorum
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Laboratory for Systems Biology, University of Florida, Gainesville, Florida, United States of America
| | - Jiyeun Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sloan A. Lewis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eric F. Lock
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Marcus Mendes
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Aidan Neher
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Somayeh Nili
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lars Rønn Olsen
- Department of Immunology and Microbiology, LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Shelby Orfield
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | | | - Nidhi Pai
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cokie Parker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Brian Qian
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Mikkel Rasmussen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joaquin Reyna
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Eve Richardson
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josey Sorenson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aparna Srinivasan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nicola Thrupp
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Raphael Trevizani
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Fundação Oswaldo Cruz, Fiocruz - Ceará, Brazil
| | - Steffen Ventz
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jiuzhou Wang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cheng-Chang Wu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ferhat Ay
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Steven H. Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
4
|
Lane A, Quach HQ, Ovsyannikova IG, Kennedy RB, Ross TM, Einav T. Characterizing the Short- and Long-Term Temporal Dynamics of Antibody Responses to Influenza Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.26.25322965. [PMID: 40061340 PMCID: PMC11888507 DOI: 10.1101/2025.02.26.25322965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Most influenza vaccine studies evaluate acute antibody responses 1 month post-vaccination, leaving long-term immunity poorly understood. Here, we performed a combined analysis of 14 large-scale vaccine studies and conducted two new studies mapping antibody responses in high resolution from their inception out to 1 year post-vaccination. Vaccine antibody responses were classified as weak (<4x fold-change at 1 month and 1 year), transient (≥4x at 1 month, <4x at 1 year), or durable (≥4x at 1 month and 1 year). Surprisingly, >50% of vaccine recipients were weak across seasons, age groups, sexes, pre-vaccination titers, and high or standard vaccine doses. Peak fold-change at 1 month post-vaccination was strongly associated with the long-term response, with most transient responders achieving a maximum fold-change of 4x, while most durable responders reached ≥16x, with both groups maintaining these titers for 2 months (10-75 days post-vaccination). Using the weak, transient, and durable trajectories, a single time point early in the response (days 7-8 or 21) predicted an individual's response out to 1 year post-vaccination. These results demonstrate that influenza vaccine responses range from little-to-no response to eliciting strong-and-durable immunity, highlighting the stark heterogeneity that is consistently seen across influenza seasons.
Collapse
Affiliation(s)
- Aaron Lane
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Huy Q Quach
- Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Ramos I. Predictive signatures of immune response to vaccination and implications of the immune setpoint remodeling. mSphere 2025; 10:e0050224. [PMID: 39853092 PMCID: PMC11852852 DOI: 10.1128/msphere.00502-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
In 2020, I featured two articles in the "mSphere of Influence" commentary series that had profound implications for the field of immunology and helped shape my research perspective. These articles were "Global Analyses of Human Immune Variation Reveal Baseline Predictors of Postvaccination Responses" by Tsang et al. (Cell 157:499-513, 2014, https://doi.org/10.1016/j.cell.2014.03.031) and "A crowdsourced analysis to identify ab initio molecular signatures predictive of susceptibility to viral infection" by Fourati et al. (Nat Commun 9:4418, 2018, https://doi.org/10.1038/s41467-018-06735-8). From these topics, the identification of signatures predictive of immune responses to vaccination has greatly advanced and pivoted our understanding of how the immune state at the time of vaccination predicts (and potentially determines) vaccination outcomes. While most of this work has been done using influenza vaccination as a model, pan-vaccine signatures have been also identified. The key implications are their potential use to predict who will respond to vaccinations and to inform strategies for fine-tuning the immune setpoint to enhance immune responses. In addition, investigations in this area led us to understand that immune perturbations, such as acute infections and vaccinations, can remodel the baseline immune state and alter immune responses to future exposures, expanding this exciting field of research. These processes are likely epigenetically encoded, and some examples have already been identified and are discussed in this minireview. Therefore, further research is essential to gain a deeper understanding of how immune exposures modify the epigenome and transcriptome, influence the immune setpoint in response to vaccination, and define its exposure-specific characteristics.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
6
|
Kumar A, Dixit S, Srinivasan K, M D, Vincent PMDR. Personalized cancer vaccine design using AI-powered technologies. Front Immunol 2024; 15:1357217. [PMID: 39582860 PMCID: PMC11581883 DOI: 10.3389/fimmu.2024.1357217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 09/24/2024] [Indexed: 11/26/2024] Open
Abstract
Immunotherapy has ushered in a new era of cancer treatment, yet cancer remains a leading cause of global mortality. Among various therapeutic strategies, cancer vaccines have shown promise by activating the immune system to specifically target cancer cells. While current cancer vaccines are primarily prophylactic, advancements in targeting tumor-associated antigens (TAAs) and neoantigens have paved the way for therapeutic vaccines. The integration of artificial intelligence (AI) into cancer vaccine development is revolutionizing the field by enhancing various aspect of design and delivery. This review explores how AI facilitates precise epitope design, optimizes mRNA and DNA vaccine instructions, and enables personalized vaccine strategies by predicting patient responses. By utilizing AI technologies, researchers can navigate complex biological datasets and uncover novel therapeutic targets, thereby improving the precision and efficacy of cancer vaccines. Despite the promise of AI-powered cancer vaccines, significant challenges remain, such as tumor heterogeneity and genetic variability, which can limit the effectiveness of neoantigen prediction. Moreover, ethical and regulatory concerns surrounding data privacy and algorithmic bias must be addressed to ensure responsible AI deployment. The future of cancer vaccine development lies in the seamless integration of AI to create personalized immunotherapies that offer targeted and effective cancer treatments. This review underscores the importance of interdisciplinary collaboration and innovation in overcoming these challenges and advancing cancer vaccine development.
Collapse
Affiliation(s)
- Anant Kumar
- School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Shriniket Dixit
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Kathiravan Srinivasan
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Dinakaran M
- School of Computer Science and Engineering, Vellore Institute of Technology, Chennai, India
| | - P. M. Durai Raj Vincent
- School of Computer Science Engineering and Information Systems, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
7
|
Cameron CM, Raghu V, Richardson B, Zagore LL, Tamilselvan B, Golden J, Cartwright M, Schoen RE, Finn OJ, Benos PV, Cameron MJ. Pre-vaccination transcriptomic profiles of immune responders to the MUC1 peptide vaccine for colon cancer prevention. Front Immunol 2024; 15:1437391. [PMID: 39450169 PMCID: PMC11499122 DOI: 10.3389/fimmu.2024.1437391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Self-antigens abnormally expressed on tumors, such as MUC1, have been targeted by therapeutic cancer vaccines. We recently assessed in two clinical trials in a preventative setting whether immunity induced with a MUC1 peptide vaccine could reduce high colon cancer risk in individuals with a history of premalignant colon adenomas. In both trials, there were immune responders and non-responders to the vaccine. Methods Here we used PBMC pre-vaccination and 2 weeks after the first vaccine of responders and non-responders selected from both trials to identify early biomarkers of immune response involved in long-term memory generation and prevention of adenoma recurrence. We performed flow cytometry, phosflow, and differential gene expression analyses on PBMCs collected from MUC1 vaccine responders and non-responders pre-vaccination and two weeks after the first of three vaccine doses. Results MUC1 vaccine responders had higher frequencies of CD4 cells pre-vaccination, increased expression of CD40L on CD8 and CD4 T-cells, and a greater increase in ICOS expression on CD8 T-cells. Differential gene expression analysis revealed that iCOSL, PI3K AKT MTOR, and B-cell signaling pathways are activated early in response to the MUC1 vaccine. We identified six specific transcripts involved in elevated antigen presentation, B-cell activation, and NF-κB1 activation that were directly linked to finding antibody response at week 12. Finally, a model using these transcripts was able to predict non-responders with accuracy. Discussion These findings suggest that individuals who can be predicted to respond to the MUC1 vaccine, and potentially other vaccines, have greater readiness in all immune compartments to present and respond to antigens. Predictive biomarkers of MUC1 vaccine response may lead to more effective vaccines tailored to individuals with high risk for cancer but with varying immune fitness.
Collapse
Affiliation(s)
- Cheryl M. Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Vineet Raghu
- Department of Computer Science, University of Pittsburgh, Pittsburgh, PA, United States
- Massachusetts General Hospital, Harvard Medical School, Cambridge, MA, United States
| | - Brian Richardson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Leah L. Zagore
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Banumathi Tamilselvan
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, United States
| | - Jackelyn Golden
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Michael Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Robert E. Schoen
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Olivera J. Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Panayiotis V. Benos
- Department of Epidemiology, University of Florida, Gainesville, FL, United States
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark J. Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
8
|
Shinde P, Willemsen L, Anderson M, Aoki M, Basu S, Burel JG, Cheng P, Dastidar SG, Dunleavy A, Einav T, Forschmiedt J, Fourati S, Garcia J, Gibson W, Greenbaum JA, Guan L, Guan W, Gygi JP, Ha B, Hou J, Hsiao J, Huang Y, Jansen R, Kakoty B, Kang Z, Kobie JJ, Kojima M, Konstorum A, Lee J, Lewis SA, Li A, Lock EF, Mahita J, Mendes M, Meng H, Neher A, Nili S, Olsen LR, Orfield S, Overton JA, Pai N, Parker C, Qian B, Rasmussen M, Reyna J, Richardson E, Safo S, Sorenson J, Srinivasan A, Thrupp N, Tippalagama R, Trevizani R, Ventz S, Wang J, Wu CC, Ay F, Grant B, Kleinstein SH, Peters B. Putting computational models of immunity to the test - an invited challenge to predict B. pertussis vaccination outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611290. [PMID: 39282381 PMCID: PMC11398469 DOI: 10.1101/2024.09.04.611290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Systems vaccinology studies have been used to build computational models that predict individual vaccine responses and identify the factors contributing to differences in outcome. Comparing such models is challenging due to variability in study designs. To address this, we established a community resource to compare models predicting B. pertussis booster responses and generate experimental data for the explicit purpose of model evaluation. We here describe our second computational prediction challenge using this resource, where we benchmarked 49 algorithms from 53 scientists. We found that the most successful models stood out in their handling of nonlinearities, reducing large feature sets to representative subsets, and advanced data preprocessing. In contrast, we found that models adopted from literature that were developed to predict vaccine antibody responses in other settings performed poorly, reinforcing the need for purpose-built models. Overall, this demonstrates the value of purpose-generated datasets for rigorous and open model evaluations to identify features that improve the reliability and applicability of computational models in vaccine response prediction.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Lisa Willemsen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Michael Anderson
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Minori Aoki
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Saonli Basu
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Julie G Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Peng Cheng
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Aidan Dunleavy
- School of Statistics, University of Minnesota, Minneapolis, MN, USA
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Jamie Forschmiedt
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, IL, USA
| | - Javier Garcia
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - William Gibson
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, MD, USA
| | - Jason A Greenbaum
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Weikang Guan
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Jeremy P Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Brendan Ha
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jason Hsiao
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Rick Jansen
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Bhargob Kakoty
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Zhiyu Kang
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - James J Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mari Kojima
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Anna Konstorum
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Laboratory for Systems Biology, University of Florida
| | - Jiyeun Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Sloan A Lewis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Marcus Mendes
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Aidan Neher
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Somayeh Nili
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Shelby Orfield
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Nidhi Pai
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Cokie Parker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Brian Qian
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mikkel Rasmussen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Joaquin Reyna
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, San Diego, CA, USA
| | - Eve Richardson
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Sandra Safo
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Josey Sorenson
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Aparna Srinivasan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Nicky Thrupp
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Raphael Trevizani
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Fundação Oswaldo Cruz, Fiocruz - Ceará, Brazil
| | - Steffen Ventz
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Jiuzhou Wang
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Cheng-Chang Wu
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Ferhat Ay
- Department of Medicine, University of California San Diego, San Diego, CA, USA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Steven H Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
9
|
Sparks R, Rachmaninoff N, Lau WW, Hirsch DC, Bansal N, Martins AJ, Chen J, Liu CC, Cheung F, Failla LE, Biancotto A, Fantoni G, Sellers BA, Chawla DG, Howe KN, Mostaghimi D, Farmer R, Kotliarov Y, Calvo KR, Palmer C, Daub J, Foruraghi L, Kreuzburg S, Treat JD, Urban AK, Jones A, Romeo T, Deuitch NT, Moura NS, Weinstein B, Moir S, Ferrucci L, Barron KS, Aksentijevich I, Kleinstein SH, Townsley DM, Young NS, Frischmeyer-Guerrerio PA, Uzel G, Pinto-Patarroyo GP, Cudrici CD, Hoffmann P, Stone DL, Ombrello AK, Freeman AF, Zerbe CS, Kastner DL, Holland SM, Tsang JS. A unified metric of human immune health. Nat Med 2024; 30:2461-2472. [PMID: 38961223 DOI: 10.1038/s41591-024-03092-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
Immunological health has been challenging to characterize but could be defined as the absence of immune pathology. While shared features of some immune diseases and the concept of immunologic resilience based on age-independent adaptation to antigenic stimulation have been developed, general metrics of immune health and its utility for assessing clinically healthy individuals remain ill defined. Here we integrated transcriptomics, serum protein, peripheral immune cell frequency and clinical data from 228 patients with 22 monogenic conditions impacting key immunological pathways together with 42 age- and sex-matched healthy controls. Despite the high penetrance of monogenic lesions, differences between individuals in diverse immune parameters tended to dominate over those attributable to disease conditions or medication use. Unsupervised or supervised machine learning independently identified a score that distinguished healthy participants from patients with monogenic diseases, thus suggesting a quantitative immune health metric (IHM). In ten independent datasets, the IHM discriminated healthy from polygenic autoimmune and inflammatory disease states, marked aging in clinically healthy individuals, tracked disease activities and treatment responses in both immunological and nonimmunological diseases, and predicted age-dependent antibody responses to immunizations with different vaccines. This discriminatory power goes beyond that of the classical inflammatory biomarkers C-reactive protein and interleukin-6. Thus, deviations from health in diverse conditions, including aging, have shared systemic immune consequences, and we provide a web platform for calculating the IHM for other datasets, which could empower precision medicine.
Collapse
Affiliation(s)
- Rachel Sparks
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Nicholas Rachmaninoff
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Graduate Program in Biological Sciences, University of Maryland, College Park, MD, USA
| | - William W Lau
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Dylan C Hirsch
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Andrew J Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jinguo Chen
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Candace C Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Laura E Failla
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Angelique Biancotto
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Giovanna Fantoni
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Brian A Sellers
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Daniel G Chawla
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Katherine N Howe
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Darius Mostaghimi
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Rohit Farmer
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Yuri Kotliarov
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA
| | - Katherine R Calvo
- Hematology Section, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Cindy Palmer
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Janine Daub
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Ladan Foruraghi
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Samantha Kreuzburg
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Jennifer D Treat
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Amanda K Urban
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Anne Jones
- Inflammatory Disease Section, NHGRI, NIH, Bethesda, MD, USA
| | - Tina Romeo
- Inflammatory Disease Section, NHGRI, NIH, Bethesda, MD, USA
| | | | | | | | - Susan Moir
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, NIA, Baltimore, MD, USA
| | - Karyl S Barron
- Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | | | - Steven H Kleinstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | | - Neal S Young
- Hematology Branch, NHLBI, NIH, Bethesda, MD, USA
| | | | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | | | | | | | | | | | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | | | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA.
- NIH Center for Human Immunology, Inflammation, and Autoimmunity, NIAID, NIH, Bethesda, MD, USA.
- Center for Systems and Engineering Immunology, Departments of Immunobiology and Biomedical Engineering, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Sugrue JA, Duffy D. Systems vaccinology studies - achievements and future potential. Microbes Infect 2024; 26:105318. [PMID: 38460935 DOI: 10.1016/j.micinf.2024.105318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Human immune responses to vaccination are variable both within and between populations. Systems vaccinology, which is the application of multi-omics technologies to vaccine studies, seeks to understand such variation and predict responses to optimise vaccine strategies. Here, we outline new approaches to systems vaccinology, focusing on the incorporation of additional cohorts, endpoints and technologies.
Collapse
Affiliation(s)
- Jamie A Sugrue
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France.
| |
Collapse
|
11
|
Page L, Dennehy K, Mueller K, Girl P, Loell E, Buijze H, Classen JM, Messmann H, Roemmele C, Hoffmann R, Wurster S, Fuchs A. Antigen-specific T helper cells and cytokine profiles predict intensity and longevity of cellular and humoral responses to SARS-CoV-2 booster vaccination. Front Immunol 2024; 15:1423766. [PMID: 39267758 PMCID: PMC11390417 DOI: 10.3389/fimmu.2024.1423766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Pre-existent pools of coronavirus-specific or cross-reactive T cells were shown to shape the development of cellular and humoral immune responses after primary mRNA vaccination against SARS-CoV-2. However, the cellular determinants of responses to booster vaccination remain incompletely understood. Therefore, we phenotypically and functionally characterized spike antigen-specific T helper (Th) cells in healthy, immunocompetent individuals and correlated the results with cellular and humoral immune responses to BNT162b2 booster vaccination over a six-month period. Methods Blood of 30 healthy healthcare workers was collected before, 1, 3, and 6 months after their 3rd BNT162b2 vaccination. Whole blood was stimulated with spike peptides and analyzed using flow cytometry, a 13-plex cytokine assay, and nCounter-based transcriptomics. Results Spike-specific IgG levels at 1 month after booster vaccination correlated with pre-existing CD154+CD69+IFN-γ+CD4+ effector memory cells as well as spike-induced IL-2 and IL-17A secretion. Early post-booster (1-month) spike IgG levels (r=0.49), spike-induced IL‑2 (r=0.58), and spike-induced IFN‑γ release (r=0.43) correlated moderately with their respective long-term (6-month) responses. Sustained robust IgG responses were significantly associated with S-specific (CD69+±CD154+±IFN-γ+) Th-cell frequencies before booster vaccination (p=0.038), especially double/triple-positive type-1 Th cells. Furthermore, spike IgG levels, spike-induced IL‑2 release, and spike-induced IFN‑γ release after 6 months were significantly associated with increased IL‑2 & IL‑4, IP‑10 & MCP1, and IFN‑γ & IP‑10 levels at 1 month post-booster, respectively. On the transcriptional level, induction of pathways associated with both T-cell proliferation and antigen presentation was indicative of sustained spike-induced cytokine release and spike-specific IgG production 6 months post-booster. Using support vector machine models, pre-booster spike-specific T-cell frequencies and early post-booster cytokine responses predicted sustained (6-month) responses with F1 scores of 0.80-1.00. Discussion In summary, spike-specific Th cells and T-cellular cytokine signatures present before BNT162b2 booster vaccination shape sustained adaptive cellular and humoral responses post-booster. Functional T-cell assays might facilitate early identification of potential non-responders.
Collapse
Affiliation(s)
- Lukas Page
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Kevin Dennehy
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | | | - Philipp Girl
- Bundeswehr Institute of Microbiology, Munich, Germany
- Central Institute of the Bundeswehr Medical Service, Munich, Germany
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Eva Loell
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Hellen Buijze
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Johanna-Maria Classen
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Helmut Messmann
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Christoph Roemmele
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Reinhard Hoffmann
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Sebastian Wurster
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andre Fuchs
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| |
Collapse
|
12
|
Papadatou I, Geropeppa M, Piperi C, Spoulou V, Adamopoulos C, Papavassiliou AG. Deciphering Immune Responses to Immunization via Transcriptional Analysis: A Narrative Review of the Current Evidence towards Personalized Vaccination Strategies. Int J Mol Sci 2024; 25:7095. [PMID: 39000206 PMCID: PMC11240890 DOI: 10.3390/ijms25137095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The development of vaccines has drastically reduced the mortality and morbidity of several diseases. Despite the great success of vaccines, the immunological processes involved in protective immunity are not fully understood and several issues remain to be elucidated. Recently, the advent of high-throughput technologies has enabled a more in-depth investigation of the immune system as a whole and the characterization of the interactions of numerous components of immunity. In the field of vaccinology, these tools allow for the exploration of the molecular mechanisms by which vaccines can induce protective immune responses. In this review, we aim to describe current data on transcriptional responses to vaccination, focusing on similarities and differences of vaccine-induced transcriptional responses among vaccines mostly in healthy adults, but also in high-risk populations, such as the elderly and children. Moreover, the identification of potential predictive biomarkers of vaccine immunogenicity, the effect of age on transcriptional response and future perspectives for the utilization of transcriptomics in the field of vaccinology will be discussed.
Collapse
Affiliation(s)
- Ioanna Papadatou
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
- First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Geropeppa
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.P.); (M.G.); (V.S.)
- First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.P.); (A.G.P.)
| |
Collapse
|
13
|
Cameron CM, Raghu V, Richardson B, Zagore LL, Tamilselvan B, Golden J, Cartwright M, Schoen RE, Finn OJ, Benos PV, Cameron MJ. Pre-vaccination transcriptomic profiles of immune responders to the MUC1 peptide vaccine for colon cancer prevention. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.09.24305336. [PMID: 38766010 PMCID: PMC11100921 DOI: 10.1101/2024.05.09.24305336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Self-antigens abnormally expressed on tumors, such as MUC1, have been targeted by therapeutic cancer vaccines. We recently assessed in two clinical trials in a preventative setting whether immunity induced with a MUC1 peptide vaccine could reduce high colon cancer risk in individuals with a history of premalignant colon adenomas. In both trials, there were immune responders and non-responders to the vaccine. Here we used PBMC pre-vaccination and 2 weeks after the first vaccine of responders and non-responders selected from both trials to identify early biomarkers of immune response involved in long-term memory generation and prevention of adenoma recurrence. We performed flow cytometry, phosflow, and differential gene expression analyses on PBMCs collected from MUC1 vaccine responders and non-responders pre-vaccination and two weeks after the first of three vaccine doses. MUC1 vaccine responders had higher frequencies of CD4 cells pre-vaccination, increased expression of CD40L on CD8 and CD4 T-cells, and a greater increase in ICOS expression on CD8 T-cells. Differential gene expression analysis revealed that iCOSL, PI3K AKT MTOR, and B-cell signaling pathways are activated early in response to the MUC1 vaccine. We identified six specific transcripts involved in elevated antigen presentation, B-cell activation, and NF-kB1 activation that were directly linked to finding antibody response at week 12. Finally, a model using these transcripts was able to predict non-responders with accuracy. These findings suggest that individuals who can be predicted to respond to the MUC1 vaccine, and potentially other vaccines, have greater readiness in all immune compartments to present and respond to antigens. Predictive biomarkers of MUC1 vaccine response may lead to more effective vaccines tailored to individuals with high risk for cancer but with varying immune fitness.
Collapse
Affiliation(s)
- Cheryl M Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH
| | - Vineet Raghu
- Department of Computer Science, University of Pittsburgh, Pittsburgh, PA
- Massachusetts General Hospital, Harvard Medical School, Cambridge, MA
| | - Brian Richardson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| | - Leah L Zagore
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| | | | - Jackelyn Golden
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| | - Michael Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| | - Robert E Schoen
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| | - Panayiotis V Benos
- Department of Epidemiology, University of Florida, Gainesville, FL
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
14
|
Riccomi A, Trombetta CM, Dorrucci M, Di Placido D, Sanarico N, Farchi F, Giuseppetti R, Villano U, Marcantonio C, Marchi S, Ciaramella A, Pezzotti P, Montomoli E, Valdarchi C, Ciccaglione AR, Vendetti S. Effects of Influenza Vaccine on the Immune Responses to SARS-CoV-2 Vaccination. Vaccines (Basel) 2024; 12:425. [PMID: 38675807 PMCID: PMC11054385 DOI: 10.3390/vaccines12040425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
A number of studies have suggested that influenza vaccination can provide protection against COVID-19, but the underlying mechanisms that could explain this association are still unclear. In this study, the effect of the 2021/2022 seasonal influenza vaccination on the immune response to the booster dose of anti-SARS-CoV-2 vaccination was evaluated in a cohort of healthy individuals. A total of 113 participants were enrolled, 74 of whom had no prior COVID-19 diagnosis or significant comorbidities were considered for the analysis. Participants received the anti-influenza tetravalent vaccine and the booster dose of the anti-SARS-CoV-2 vaccine or the anti-SARS-CoV-2 vaccine alone. Blood was collected before and 4 weeks after each vaccination and 12 weeks after SARS-CoV-2 vaccination and analyzed for anti-flu and anti-spike-specific antibody titers and for in vitro influenza and SARS-CoV-2 neutralization capacity. Results indicated an increased reactivity in subjects who received both influenza and SARS-CoV-2 vaccinations compared to those who received only the SARS-CoV-2 vaccine, with sustained anti-spike antibody titers up to 12 weeks post-vaccination. Immune response to the influenza vaccine was evaluated, and individuals were stratified as high or low responders. High responders showed increased antibody titers against the SARS-CoV-2 vaccine both after 4 and 12 weeks post-vaccination. Conversely, individuals classified as low responders were less responsive to the SARS-CoV-2 vaccine. These data indicate that both external stimuli, such as influenza vaccination, and the host's intrinsic ability to respond to stimuli play a role in the response to the vaccine.
Collapse
Affiliation(s)
- A. Riccomi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - C. M. Trombetta
- Department of Molecular and Development Medicine, University of Siena, 53100 Siena, Italy (S.M.)
- VisMederi Research Srl, 53100 Siena, Italy
| | - M. Dorrucci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - D. Di Placido
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - N. Sanarico
- Center for Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - F. Farchi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - R. Giuseppetti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - U. Villano
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - C. Marcantonio
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - S. Marchi
- Department of Molecular and Development Medicine, University of Siena, 53100 Siena, Italy (S.M.)
| | - A. Ciaramella
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - P. Pezzotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - E. Montomoli
- Department of Molecular and Development Medicine, University of Siena, 53100 Siena, Italy (S.M.)
- VisMederi Research Srl, 53100 Siena, Italy
- VisMederi Srl, 53100 Siena, Italy
| | - C. Valdarchi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - A. R. Ciccaglione
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| | - S. Vendetti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy (M.D.); (D.D.P.); (F.F.); (U.V.)
| |
Collapse
|
15
|
Shannon CP, Lee AH, Tebbutt SJ, Singh A. A Commentary on Multi-omics Data Integration in Systems Vaccinology. J Mol Biol 2024; 436:168522. [PMID: 38458605 DOI: 10.1016/j.jmb.2024.168522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Affiliation(s)
| | - Amy Hy Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Vancouver, Canada; Department of Medicine, The University of British Columbia, Vancouver, Canada; Centre for Heart Lung Innovation, Vancouver, Canada
| | - Amrit Singh
- Centre for Heart Lung Innovation, Vancouver, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, Canada.
| |
Collapse
|
16
|
Dobbs KR, Atieli HE, Valim C, Beeson JG. Previous Malaria Exposures and Immune Dysregulation: Developing Strategies To Improve Malaria Vaccine Efficacy in Young Children. Am J Trop Med Hyg 2024; 110:627-630. [PMID: 38442424 PMCID: PMC10993830 DOI: 10.4269/ajtmh.23-0696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 03/07/2024] Open
Abstract
After several decades in development, two malaria vaccines based on the same antigen and with very similar constructs and adjuvants, RTS,S/AS01 (RTS,S) and R21/Matrix-M (R21), were recommended by the WHO for widespread vaccination of children. These vaccines are much-needed additions to malaria control programs that, when used in conjunction with other control measures, will help to accelerate reductions in malaria morbidity and mortality. Although R21 is not yet available, RTS,S is currently being integrated into routine vaccine schedules in some areas. However, the efficacy of RTS,S is partial, short-lived, and varies widely according to age and geographic location. It is not clear why RTS,S induces protection in some individuals and not others, what the immune mechanisms are that favor protective immunity with RTS,S, and how immune mechanisms are influenced by host and environmental factors. Several studies suggest that higher levels of previous malaria exposure negatively impact RTS,S clinical efficacy. In this article, we summarize data suggesting that previous malaria exposures negatively impact the efficacy of RTS,S and other malaria vaccine candidates. We highlight recent evidence suggesting that increasing malaria exposure impairs the generation of functional antibody responses to RTS,S. Finally, we discuss how investigation of clinical and immune factors associated with suboptimal responses to RTS,S can be used to develop strategies to optimize RTS,S, which will remain relevant to R21 and next-generation vaccines.
Collapse
Affiliation(s)
| | | | - Clarissa Valim
- Boston University School of Public Health, Boston, Massachusetts
| | | |
Collapse
|
17
|
Haralambieva IH, Chen J, Quach HQ, Ratishvili T, Warner ND, Ovsyannikova IG, Poland GA, Kennedy RB. Early B cell transcriptomic markers of measles-specific humoral immunity following a 3 rd dose of MMR vaccine. Front Immunol 2024; 15:1358477. [PMID: 38633249 PMCID: PMC11021587 DOI: 10.3389/fimmu.2024.1358477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
B cell transcriptomic signatures hold promise for the early prediction of vaccine-induced humoral immunity and vaccine protective efficacy. We performed a longitudinal study in 232 healthy adult participants before/after a 3rd dose of MMR (MMR3) vaccine. We assessed baseline and early transcriptional patterns in purified B cells and their association with measles-specific humoral immunity after MMR vaccination using two analytical methods ("per gene" linear models and joint analysis). Our study identified distinct early transcriptional signatures/genes following MMR3 that were associated with measles-specific neutralizing antibody titer and/or binding antibody titer. The most significant genes included: the interleukin 20 receptor subunit beta/IL20RB gene (a subunit receptor for IL-24, a cytokine involved in the germinal center B cell maturation/response); the phorbol-12-myristate-13-acetate-induced protein 1/PMAIP1, the brain expressed X-linked 2/BEX2 gene and the B cell Fas apoptotic inhibitory molecule/FAIM, involved in the selection of high-affinity B cell clones and apoptosis/regulation of apoptosis; as well as IL16 (encoding the B lymphocyte-derived IL-16 ligand of CD4), involved in the crosstalk between B cells, dendritic cells and helper T cells. Significantly enriched pathways included B cell signaling, apoptosis/regulation of apoptosis, metabolic pathways, cell cycle-related pathways, and pathways associated with viral infections, among others. In conclusion, our study identified genes/pathways linked to antigen-induced B cell proliferation, differentiation, apoptosis, and clonal selection, that are associated with, and impact measles virus-specific humoral immunity after MMR vaccination.
Collapse
Affiliation(s)
- Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tamar Ratishvili
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Nathaniel D. Warner
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
18
|
van Dorst MMAR, Pyuza JJ, Nkurunungi G, Kullaya VI, Smits HH, Hogendoorn PCW, Wammes LJ, Everts B, Elliott AM, Jochems SP, Yazdanbakhsh M. Immunological factors linked to geographical variation in vaccine responses. Nat Rev Immunol 2024; 24:250-263. [PMID: 37770632 DOI: 10.1038/s41577-023-00941-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Vaccination is one of medicine's greatest achievements; however, its full potential is hampered by considerable variation in efficacy across populations and geographical regions. For example, attenuated malaria vaccines in high-income countries confer almost 100% protection, whereas in low-income regions these same vaccines achieve only 20-50% protection. This trend is also observed for other vaccines, such as bacillus Calmette-Guérin (BCG), rotavirus and yellow fever vaccines, in terms of either immunogenicity or efficacy. Multiple environmental factors affect vaccine responses, including pathogen exposure, microbiota composition and dietary nutrients. However, there has been variable success with interventions that target these individual factors, highlighting the need for a better understanding of their downstream immunological mechanisms to develop new ways of modulating vaccine responses. Here, we review the immunological factors that underlie geographical variation in vaccine responses. Through the identification of causal pathways that link environmental influences to vaccine responsiveness, it might become possible to devise modulatory compounds that can complement vaccines for better outcomes in regions where they are needed most.
Collapse
Affiliation(s)
- Marloes M A R van Dorst
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeremia J Pyuza
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Linda J Wammes
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
19
|
Ye X, Yang S, Tu J, Xu L, Wang Y, Chen H, Yu R, Huang P. Leveraging baseline transcriptional features and information from single-cell data to power the prediction of influenza vaccine response. Front Cell Infect Microbiol 2024; 14:1243586. [PMID: 38384303 PMCID: PMC10879619 DOI: 10.3389/fcimb.2024.1243586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
Introduction Vaccination is still the primary means for preventing influenza virus infection, but the protective effects vary greatly among individuals. Identifying individuals at risk of low response to influenza vaccination is important. This study aimed to explore improved strategies for constructing predictive models of influenza vaccine response using gene expression data. Methods We first used gene expression and immune response data from the Immune Signatures Data Resource (IS2) to define influenza vaccine response-related transcriptional expression and alteration features at different time points across vaccination via differential expression analysis. Then, we mapped these features to single-cell resolution using additional published single-cell data to investigate the possible mechanism. Finally, we explored the potential of these identified transcriptional features in predicting influenza vaccine response. We used several modeling strategies and also attempted to leverage the information from single-cell RNA sequencing (scRNA-seq) data to optimize the predictive models. Results The results showed that models based on genes showing differential expression (DEGs) or fold change (DFGs) at day 7 post-vaccination performed the best in internal validation, while models based on DFGs had a better performance in external validation than those based on DEGs. In addition, incorporating baseline predictors could improve the performance of models based on days 1-3, while the model based on the expression profile of plasma cells deconvoluted from the model that used DEGs at day 7 as predictors showed an improved performance in external validation. Conclusion Our study emphasizes the value of using combination modeling strategy and leveraging information from single-cell levels in constructing influenza vaccine response predictive models.
Collapse
Affiliation(s)
- Xiangyu Ye
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sheng Yang
- Department of Biostatistics, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junlan Tu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, China
| | - Hongbo Chen
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, China
| | - Rongbin Yu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, National Vaccine Innovation Platform, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Odak I, Riemann L, Sandrock I, Cossmann A, Ramos GM, Hammerschmidt SI, Ritter C, Friedrichsen M, Hassan A, Dopfer-Jablonka A, Stankov MV, Weskamm LM, Addo MM, Ravens I, Willenzon S, Schimrock A, Ristenpart J, Janssen A, Barros-Martins J, Hansen G, Falk C, Behrens GMN, Förster R. Systems biology analysis reveals distinct molecular signatures associated with immune responsiveness to the BNT162b COVID-19 vaccine. EBioMedicine 2024; 99:104947. [PMID: 38160529 PMCID: PMC10792461 DOI: 10.1016/j.ebiom.2023.104947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Human immune responses to COVID-19 vaccines display a large heterogeneity of induced immunity and the underlying immune mechanisms for this remain largely unknown. METHODS Using a systems biology approach, we longitudinally profiled a unique cohort of female high and low responders to the BNT162b vaccine, who were known from previous COVID-19 vaccinations to develop maximum and minimum immune responses to the vaccine. We utilized high dimensional flow cytometry, bulk and single cell mRNA sequencing and 48-plex serum cytokine analyses. FINDINGS We revealed early, transient immunological and molecular signatures that distinguished high from low responders and correlated with B and T cell responses measured 14 days later. High responders featured a distinct transcriptional activity of interferon-driven genes and genes connected to enhanced antigen presentation. This was accompanied by a robust cytokine response related to Th1 differentiation. Both transcriptome and serum cytokine signatures were confirmed in two independent confirmatory cohorts. INTERPRETATION Collectively, our data contribute to a better understanding of the immunogenicity of mRNA-based COVID-19 vaccines, which might lead to the optimization of vaccine designs for individuals with poor vaccine responses. FUNDING German Center for Infection Research, German Center for Lung Research, German Research Foundation, Excellence Strategy EXC 2155 "RESIST" and European Regional Development Fund.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Germany
| | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Germany; Clinician Scientist Program TITUS, Else-Kröner-Fresenius Foundation, Hannover Medical School, Germany
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Germany
| | - Anne Cossmann
- Department for Rheumatology and Immunology, Hannover Medical School, Germany
| | - Gema Morillas Ramos
- Department for Rheumatology and Immunology, Hannover Medical School, Germany
| | | | | | | | - Ahmed Hassan
- Institute of Immunology, Hannover Medical School, Germany
| | - Alexandra Dopfer-Jablonka
- Department for Rheumatology and Immunology, Hannover Medical School, Germany; German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Germany
| | - Metodi V Stankov
- Department for Rheumatology and Immunology, Hannover Medical School, Germany
| | - Leonie M Weskamm
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development (IIRVD), University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany; First Department of Medicine, Division of Infectious Diseases, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Inga Ravens
- Institute of Immunology, Hannover Medical School, Germany
| | | | - Anja Schimrock
- Institute of Immunology, Hannover Medical School, Germany
| | | | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Germany
| | | | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Germany; Clinician Scientist Program TITUS, Else-Kröner-Fresenius Foundation, Hannover Medical School, Germany; German Center of Lung Research (DZL), BREATH, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Germany
| | - Christine Falk
- Institute for Transplantation Immunology, Hannover Medical School, Hannover, Germany
| | - Georg M N Behrens
- Department for Rheumatology and Immunology, Hannover Medical School, Germany; German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Germany; Centre for Individualized Infection Medicine (CiiM), Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Germany; Clinician Scientist Program TITUS, Else-Kröner-Fresenius Foundation, Hannover Medical School, Germany; German Centre for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany; German Center of Lung Research (DZL), BREATH, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Germany.
| |
Collapse
|
21
|
Gonzalez Dias Carvalho PC, Dominguez Crespo Hirata T, Mano Alves LY, Moscardini IF, do Nascimento APB, Costa-Martins AG, Sorgi S, Harandi AM, Ferreira DM, Vianello E, Haks MC, Ottenhoff THM, Santoro F, Martinez-Murillo P, Huttner A, Siegrist CA, Medaglini D, Nakaya HI. Baseline gene signatures of reactogenicity to Ebola vaccination: a machine learning approach across multiple cohorts. Front Immunol 2023; 14:1259197. [PMID: 38022684 PMCID: PMC10663260 DOI: 10.3389/fimmu.2023.1259197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The rVSVDG-ZEBOV-GP (Ervebo®) vaccine is both immunogenic and protective against Ebola. However, the vaccine can cause a broad range of transient adverse reactions, from headache to arthritis. Identifying baseline reactogenicity signatures can advance personalized vaccinology and increase our understanding of the molecular factors associated with such adverse events. Methods In this study, we developed a machine learning approach to integrate prevaccination gene expression data with adverse events that occurred within 14 days post-vaccination. Results and Discussion We analyzed the expression of 144 genes across 343 blood samples collected from participants of 4 phase I clinical trial cohorts: Switzerland, USA, Gabon, and Kenya. Our machine learning approach revealed 22 key genes associated with adverse events such as local reactions, fatigue, headache, myalgia, fever, chills, arthralgia, nausea, and arthritis, providing insights into potential biological mechanisms linked to vaccine reactogenicity.
Collapse
Affiliation(s)
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leandro Yukio Mano Alves
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - André G. Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Artificial Intelligence and Analytics Department, Institute for Technological Research, São Paulo, Brazil
| | - Sara Sorgi
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Daniela M. Ferreira
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Angela Huttner
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Infectious Diseases Service, Geneva University Hospitals, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Helder I. Nakaya
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
22
|
Shinde P, Soldevila F, Reyna J, Aoki M, Rasmussen M, Willemsen L, Kojima M, Ha B, Greenbaum JA, Overton JA, Guzman-Orozco H, Nili S, Orfield S, Gygi JP, da Silva Antunes R, Sette A, Grant B, Olsen LR, Konstorum A, Guan L, Ay F, Kleinstein SH, Peters B. A systems vaccinology resource to develop and test computational models of immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555193. [PMID: 37693565 PMCID: PMC10491180 DOI: 10.1101/2023.08.28.555193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Computational models that predict an individual's response to a vaccine offer the potential for mechanistic insights and personalized vaccination strategies. These models are increasingly derived from systems vaccinology studies that generate immune profiles from human cohorts pre- and post-vaccination. Most of these studies involve relatively small cohorts and profile the response to a single vaccine. The ability to assess the performance of the resulting models would be improved by comparing their performance on independent datasets, as has been done with great success in other areas of biology such as protein structure predictions. To transfer this approach to system vaccinology studies, we established a prototype platform that focuses on the evaluation of Computational Models of Immunity to Pertussis Booster vaccinations (CMI-PB). A community resource, CMI-PB generates experimental data for the explicit purpose of model evaluation, which is performed through a series of annual data releases and associated contests. We here report on our experience with the first such 'dry run' for a contest where the goal was to predict individual immune responses based on pre-vaccination multi-omic profiles. Over 30 models adopted from the literature were tested, but only one was predictive, and was based on age alone. The performance of new models built using CMI-PB training data was much better, but varied significantly based on the choice of pre-vaccination features used and the model building strategy. This suggests that previously published models developed for other vaccines do not generalize well to Pertussis Booster vaccination. Overall, these results reinforced the need for comparative analysis across models and datasets that CMI-PB aims to achieve. We are seeking wider community engagement for our first public prediction contest, which will open in early 2024.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ferran Soldevila
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Joaquin Reyna
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA, USA
| | - Minori Aoki
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mikkel Rasmussen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lisa Willemsen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mari Kojima
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Brendan Ha
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jason A Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - James A Overton
- Knocean Inc., 107 Quebec Ave. Toronto, Ontario, M6P 2T3, Canada
| | - Hector Guzman-Orozco
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Somayeh Nili
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shelby Orfield
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jeremy P. Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anna Konstorum
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Ferhat Ay
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Steven H. Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
23
|
Liu YE, Darrah PA, Zeppa JJ, Kamath M, Laboune F, Douek DC, Maiello P, Roederer M, Flynn JL, Seder RA, Khatri P. Blood transcriptional correlates of BCG-induced protection against tuberculosis in rhesus macaques. Cell Rep Med 2023; 4:101096. [PMID: 37390827 PMCID: PMC10394165 DOI: 10.1016/j.xcrm.2023.101096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/29/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023]
Abstract
Blood-based correlates of vaccine-induced protection against tuberculosis (TB) are urgently needed. Here, we analyze the blood transcriptome of rhesus macaques immunized with varying doses of intravenous (i.v.) BCG followed by Mycobacterium tuberculosis (Mtb) challenge. We use high-dose i.v. BCG recipients for "discovery" and validate our findings in low-dose recipients and in an independent cohort of macaques receiving BCG via different routes. We identify seven vaccine-induced gene modules, including an innate module (module 1) enriched for type 1 interferon and RIG-I-like receptor signaling pathways. Module 1 on day 2 post-vaccination highly correlates with lung antigen-responsive CD4 T cells at week 8 and with Mtb and granuloma burden following challenge. Parsimonious signatures within module 1 at day 2 post-vaccination predict protection following challenge with area under the receiver operating characteristic curve (AUROC) ≥0.91. Together, these results indicate that the early innate transcriptional response to i.v. BCG in peripheral blood may provide a robust correlate of protection against TB.
Collapse
Affiliation(s)
- Yiran E Liu
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA; PhD Program in Epidemiology and Clinical Research, Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph J Zeppa
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Megha Kamath
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Farida Laboune
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Data Sciences, Stanford University, Stanford, CA 94305, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Siddiqa A, Wang Y, Thapa M, Martin DE, Cadar AN, Bartley JM, Li S. A pilot metabolomic study of drug interaction with the immune response to seasonal influenza vaccination. NPJ Vaccines 2023; 8:92. [PMID: 37308481 PMCID: PMC10261085 DOI: 10.1038/s41541-023-00682-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
Many human diseases, including metabolic diseases, are intertwined with the immune system. The understanding of how the human immune system interacts with pharmaceutical drugs is still limited, and epidemiological studies only start to emerge. As the metabolomics technology matures, both drug metabolites and biological responses can be measured in the same global profiling data. Therefore, a new opportunity presents itself to study the interactions between pharmaceutical drugs and immune system in the high-resolution mass spectrometry data. We report here a double-blinded pilot study of seasonal influenza vaccination, where half of the participants received daily metformin administration. Global metabolomics was measured in the plasma samples at six timepoints. Metformin signatures were successfully identified in the metabolomics data. Statistically significant metabolite features were found both for the vaccination effect and for the drug-vaccine interactions. This study demonstrates the concept of using metabolomics to investigate drug interaction with the immune response in human samples directly at molecular levels.
Collapse
Affiliation(s)
- Amnah Siddiqa
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Yating Wang
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Maheshwor Thapa
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Dominique E Martin
- Department of Immunology and Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Andreia N Cadar
- Department of Immunology and Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Jenna M Bartley
- Department of Immunology and Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| | - Shuzhao Li
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA.
- Department of Immunology and Center on Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
25
|
Kelkar NS, Morrison KS, Ackerman ME. Foundations for improved vaccine correlate of risk analysis using positive-unlabeled learning. Hum Vaccin Immunother 2023:2204020. [PMID: 37133899 DOI: 10.1080/21645515.2023.2204020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
Insights into mechanisms of protection afforded by vaccine efficacy field trials can be complicated by both low rates of exposure and protection. However, these barriers do not preclude the discovery of correlates of reduced risk (CoR) of infection, which are a critical first step in defining correlates of protection (CoP). Given the significant investment in large-scale human vaccine efficacy trials and immunogenicity data collected to support CoR discovery, novel approaches for analyzing efficacy trials to optimally support discovery of CoP are critically needed. By simulating immunological data and evaluating several machine learning approaches, this study lays the groundwork for deploying Positive/Unlabeled (P/U) learning methods, which are designed to differentiate between two groups in cases where only one group has a definitive label and the other remains ambiguous. This description applies to case-control analysis designs for field trials of vaccine efficacy: infected subjects, or cases, are by definition unprotected, whereas uninfected subjects, or controls, may have been either protected or unprotected but simply never exposed. Here, we investigate the value of applying P/U learning to classify study subjects using model immunogenicity data based on predicted protection status in order to support new insights into mechanisms of vaccine-mediated protection from infection. We demonstrate that P/U learning methods can reliably infer protection status, supporting the discovery of simulated CoP that are not observed in conventional comparisons of infection status cases and controls, and we propose next steps necessary for the practical deployment of this novel approach to correlate discovery.
Collapse
Affiliation(s)
- Natasha S Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Kyle S Morrison
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
26
|
Hirota M, Tamai M, Yukawa S, Taira N, Matthews MM, Toma T, Seto Y, Yoshida M, Toguchi S, Miyagi M, Mori T, Tomori H, Tamai O, Kina M, Sakihara E, Yamashiro C, Miyagi M, Tamaki K, Wolf M, Collins MK, Kitano H, Ishikawa H. Human immune and gut microbial parameters associated with inter-individual variations in COVID-19 mRNA vaccine-induced immunity. Commun Biol 2023; 6:368. [PMID: 37081096 PMCID: PMC10119155 DOI: 10.1038/s42003-023-04755-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
COVID-19 mRNA vaccines induce protective adaptive immunity against SARS-CoV-2 in most individuals, but there is wide variation in levels of vaccine-induced antibody and T-cell responses. However, the mechanisms underlying this inter-individual variation remain unclear. Here, using a systems biology approach based on multi-omics analyses of human blood and stool samples, we identified several factors that are associated with COVID-19 vaccine-induced adaptive immune responses. BNT162b2-induced T cell response is positively associated with late monocyte responses and inversely associated with baseline mRNA expression of activation protein 1 (AP-1) transcription factors. Interestingly, the gut microbial fucose/rhamnose degradation pathway is positively correlated with mRNA expression of AP-1, as well as a gene encoding an enzyme producing prostaglandin E2 (PGE2), which promotes AP-1 expression, and inversely correlated with BNT162b2-induced T-cell responses. These results suggest that baseline AP-1 expression, which is affected by commensal microbial activity, is a negative correlate of BNT162b2-induced T-cell responses.
Collapse
Affiliation(s)
- Masato Hirota
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Miho Tamai
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Sachie Yukawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
- Integrated Open Systems Unit, OIST, Onna-son, Okinawa, Japan
| | - Naoyuki Taira
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | | | - Takeshi Toma
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Yu Seto
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Makiko Yoshida
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Sakura Toguchi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Mio Miyagi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Tomoari Mori
- Research Support Division, Occupational Health and Safety, OIST, Onna-son, Okinawa, Japan
| | | | | | | | - Eishin Sakihara
- Health Care Center of the Naha Medical Association, Naha-city, Okinawa, Japan
| | - Chiaki Yamashiro
- Yamashiro Orthopedic Surgery Ophthalmology Clinic, Naha-city, Okinawa, Japan
| | | | - Kentaro Tamaki
- Naha-Nishi Clinic, Department of Breast Surgery, Naha-city, Okinawa, Japan
| | - Matthias Wolf
- Molecular Cryo-Electron Microscopy Unit, OIST, Onna-son, Okinawa, Japan
| | - Mary K Collins
- Research Support Division, Office of the Provost, OIST, Onna-son, Okinawa, Japan
| | - Hiroaki Kitano
- Integrated Open Systems Unit, OIST, Onna-son, Okinawa, Japan
| | - Hiroki Ishikawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan.
| |
Collapse
|
27
|
Mulè MP, Martins AJ, Cheung F, Farmer R, Sellers B, Quiel JA, Jain A, Kotliarov Y, Bansal N, Chen J, Schwartzberg PL, Tsang JS. Multiscale integration of human and single-cell variations reveals unadjuvanted vaccine high responders are naturally adjuvanted. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.20.23287474. [PMID: 37090674 PMCID: PMC10120791 DOI: 10.1101/2023.03.20.23287474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Advances in multimodal single cell analysis can empower high-resolution dissection of human vaccination responses. The resulting data capture multiple layers of biological variations, including molecular and cellular states, vaccine formulations, inter- and intra-subject differences, and responses unfolding over time. Transforming such data into biological insight remains a major challenge. Here we present a systematic framework applied to multimodal single cell data obtained before and after influenza vaccination without adjuvants or pandemic H5N1 vaccination with the AS03 adjuvant. Our approach pinpoints responses shared across or unique to specific cell types and identifies adjuvant specific signatures, including pro-survival transcriptional states in B lymphocytes that emerged one day after vaccination. We also reveal that high antibody responders to the unadjuvanted vaccine have a distinct baseline involving a rewired network of cell type specific transcriptional states. Remarkably, the status of certain innate immune cells in this network in high responders of the unadjuvanted vaccine appear "naturally adjuvanted": they resemble phenotypes induced early in the same cells only by vaccination with AS03. Furthermore, these cell subsets have elevated frequency in the blood at baseline and increased cell-intrinsic phospho-signaling responses after LPS stimulation ex vivo in high compared to low responders. Our findings identify how variation in the status of multiple immune cell types at baseline may drive robust differences in innate and adaptive responses to vaccination and thus open new avenues for vaccine development and immune response engineering in humans.
Collapse
Affiliation(s)
- Matthew P. Mulè
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- NIH-Oxford-Cambridge Scholars Program; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Andrew J. Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Foo Cheung
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Rohit Farmer
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Brian Sellers
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Juan A. Quiel
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Arjun Jain
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Yuri Kotliarov
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jinguo Chen
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Pamela L. Schwartzberg
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Cell Signaling and Immunity Section, NIAID, NIH, Bethesda, MD, USA
| | - John S. Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| |
Collapse
|
28
|
Tyagi N, Mehla K, Gupta D. Deciphering novel common gene signatures for rheumatoid arthritis and systemic lupus erythematosus by integrative analysis of transcriptomic profiles. PLoS One 2023; 18:e0281637. [PMID: 36928613 PMCID: PMC10019710 DOI: 10.1371/journal.pone.0281637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/24/2023] [Indexed: 03/18/2023] Open
Abstract
Rheumatoid Arthritis (RA) and Systemic Lupus Erythematosus (SLE) are the two highly prevalent debilitating and sometimes life-threatening systemic inflammatory autoimmune diseases. The etiology and pathogenesis of RA and SLE are interconnected in several ways, with limited knowledge about the underlying molecular mechanisms. With the motivation to better understand shared biological mechanisms and determine novel therapeutic targets, we explored common molecular disease signatures by performing a meta-analysis of publicly available microarray gene expression datasets of RA and SLE. We performed an integrated, multi-cohort analysis of 1088 transcriptomic profiles from 14 independent studies to identify common gene signatures. We identified sixty-two genes common among RA and SLE, out of which fifty-nine genes (21 upregulated and 38 downregulated) had similar expression profiles in the diseases. However, antagonistic expression profiles were observed for ACVR2A, FAM135A, and MAPRE1 genes. Thirty genes common between RA and SLE were proposed as robust gene signatures, with persistent expression in all the studies and cell types. These gene signatures were found to be involved in innate as well as adaptive immune responses, bone development and growth. In conclusion, our analysis of multicohort and multiple microarray datasets would provide the basis for understanding the common mechanisms of pathogenesis and exploring these gene signatures for their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Neetu Tyagi
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Kusum Mehla
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
29
|
Takahama S, Ishige K, Nogimori T, Yasutomi Y, Appay V, Yamamoto T. Model for predicting age-dependent safety and immunomodulatory effects of STING ligands in non-human primates. Mol Ther Methods Clin Dev 2022; 28:99-115. [PMID: 36620070 PMCID: PMC9813482 DOI: 10.1016/j.omtm.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Stimulator of interferon genes (STING) is a cytoplasmic dinucleotide sensor used as an immunomodulatory agent for cancer treatment. The efficacy of the STING ligand (STING-L) against various tumors has been evaluated in mouse models; however, its safety and efficacy in non-human primates have not been reported. We examined the effects of escalating doses of cyclic-di-adenosine monophosphate (c-di-AMP) or cyclic [G (3',5')pA (3',5'p] (3'-3'-cGAMP) administered intramuscularly or intravenously to cynomolgus macaques. Both ligands induced transient local and systemic inflammatory responses and systemic immunomodulatory responses, including the upregulation of interferon-α (IFN-α) and IFN-γ expression and the activation of multiple immunocompetent cell subsets. Better immunological responses were observed in animals that received c-di-AMP compared with those that received 3'-3'-cGAMP. Multi-parameter analysis using a dataset obtained before administering the ligands predicted the efficacy outcome partially. Importantly, the efficacy of these ligands was reduced in older macaques. We propose that 0.5 mg/kg c-di-AMP via intramuscular administration should be the optimal starting point for clinical studies. Our study is the first to demonstrate the age-dependent safety and efficacy of STING-L in non-human primates and supports the potential of STING-L use as a direct immunomodulator in vivo.
Collapse
Affiliation(s)
- Shokichi Takahama
- Laboratory of Immunosenescence, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Kazuya Ishige
- Biochemicals Division, Yamasa Corporation, Chiba 288-0056, Japan
| | - Takuto Nogimori
- Laboratory of Immunosenescence, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
| | - Victor Appay
- Laboratory of Immunosenescence, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan,Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Takuya Yamamoto
- Laboratory of Immunosenescence, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan,Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan,Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan,Laboratory of Translational Cancer Immunology and Biology, Next-generation Precision Medicine Research Center, Osaka International Cancer Institute, Osaka 541-8567, Japan,Corresponding author: Takuya Yamamoto, Laboratory of Immunosenescence, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan.
| |
Collapse
|
30
|
Fourati S, Tomalin LE, Mulè MP, Chawla DG, Gerritsen B, Rychkov D, Henrich E, Miller HER, Hagan T, Diray-Arce J, Dunn P, Levy O, Gottardo R, Sarwal MM, Tsang JS, Suárez-Fariñas M, Pulendran B, Kleinstein SH, Sékaly RP. Pan-vaccine analysis reveals innate immune endotypes predictive of antibody responses to vaccination. Nat Immunol 2022; 23:1777-1787. [PMID: 36316476 PMCID: PMC9747610 DOI: 10.1038/s41590-022-01329-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/12/2022] [Indexed: 11/05/2022]
Abstract
Several studies have shown that the pre-vaccination immune state is associated with the antibody response to vaccination. However, the generalizability and mechanisms that underlie this association remain poorly defined. Here, we sought to identify a common pre-vaccination signature and mechanisms that could predict the immune response across 13 different vaccines. Analysis of blood transcriptional profiles across studies revealed three distinct pre-vaccination endotypes, characterized by the differential expression of genes associated with a pro-inflammatory response, cell proliferation, and metabolism alterations. Importantly, individuals whose pre-vaccination endotype was enriched in pro-inflammatory response genes known to be downstream of nuclear factor-kappa B showed significantly higher serum antibody responses 1 month after vaccination. This pro-inflammatory pre-vaccination endotype showed gene expression characteristic of the innate activation state triggered by Toll-like receptor ligands or adjuvants. These results demonstrate that wide variations in the transcriptional state of the immune system in humans can be a key determinant of responsiveness to vaccination.
Collapse
Affiliation(s)
- Slim Fourati
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Lewis E Tomalin
- Center for Biostatistics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew P Mulè
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID and Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
- NIH-Oxford-Cambridge Scholars Program, Cambridge University, Cambridge, UK
| | | | | | - Dmitry Rychkov
- Division of Transplant Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Evan Henrich
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Thomas Hagan
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patrick Dunn
- ImmPort Curation Team, NG Health Solutions, Rockville, MD, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Raphael Gottardo
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Biomedical Data Science Center, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Minnie M Sarwal
- Division of Transplant Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID and Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | - Mayte Suárez-Fariñas
- Center for Biostatistics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bali Pulendran
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Rafick-Pierre Sékaly
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
31
|
Hampton BK, Plante KS, Whitmore AC, Linnertz CL, Madden EA, Noll KE, Boyson SP, Parotti B, Xenakis JG, Bell TA, Hock P, Shaw GD, de Villena FPM, Ferris MT, Heise MT. Forward genetic screen of homeostatic antibody levels in the Collaborative Cross identifies MBD1 as a novel regulator of B cell homeostasis. PLoS Genet 2022; 18:e1010548. [PMID: 36574452 PMCID: PMC9829176 DOI: 10.1371/journal.pgen.1010548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/09/2023] [Accepted: 11/28/2022] [Indexed: 12/28/2022] Open
Abstract
Variation in immune homeostasis, the state in which the immune system is maintained in the absence of stimulation, is highly variable across populations. This variation is attributed to both genetic and environmental factors. However, the identity and function of specific regulators have been difficult to identify in humans. We evaluated homeostatic antibody levels in the serum of the Collaborative Cross (CC) mouse genetic reference population. We found heritable variation in all antibody isotypes and subtypes measured. We identified 4 quantitative trait loci (QTL) associated with 3 IgG subtypes: IgG1, IgG2b, and IgG2c. While 3 of these QTL map to genome regions of known immunological significance (major histocompatibility and immunoglobulin heavy chain locus), Qih1 (associated with variation in IgG1) mapped to a novel locus on Chromosome 18. We further associated this locus with B cell proportions in the spleen and identify Methyl-CpG binding domain protein 1 under this locus as a novel regulator of homeostatic IgG1 levels in the serum and marginal zone B cells (MZB) in the spleen, consistent with a role in MZB differentiation to antibody secreting cells.
Collapse
Affiliation(s)
- Brea K. Hampton
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kenneth S. Plante
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Alan C. Whitmore
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Colton L. Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Emily A. Madden
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kelsey E. Noll
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Samuel P. Boyson
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Breantie Parotti
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - James G. Xenakis
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Timothy A. Bell
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Pablo Hock
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ginger D. Shaw
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mark T. Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
32
|
Wu S, Pushalkar S, Maity S, Pressler M, Rendleman J, Vitrinel B, Carlock M, Ross T, Choi H, Vogel C. Proteomic Signatures of the Serological Response to Influenza Vaccination in a Large Human Cohort Study. Viruses 2022; 14:v14112479. [PMID: 36366577 PMCID: PMC9696600 DOI: 10.3390/v14112479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
The serological response to the influenza virus vaccine is highly heterogeneous for reasons that are not entirely clear. While the impact of demographic factors such as age, body mass index (BMI), sex, prior vaccination and titer levels are known to impact seroconversion, they only explain a fraction of the response. To identify signatures of the vaccine response, we analyzed 273 protein levels from 138 serum samples of influenza vaccine recipients (2019-2020 season). We found that levels of proteins functioning in cholesterol transport were positively associated with seroconversion, likely linking to the known impact of BMI. When adjusting seroconversion for the demographic factors, we identified additional, unexpected signatures: proteins regulating actin cytoskeleton dynamics were significantly elevated in participants with high adjusted seroconversion. Viral strain specific analysis showed that this trend was largely driven by the H3N2 strain. Further, we identified complex associations between adjusted seroconversion and other factors: levels of proteins of the complement system associated positively with adjusted seroconversion in younger participants, while they were associated negatively in the older population. We observed the opposite trends for proteins of high density lipoprotein remodeling, transcription, and hemostasis. In sum, careful integrative modeling can extract new signatures of seroconversion from highly variable data that suggest links between the humoral response as well as immune cell communication and migration.
Collapse
Affiliation(s)
- Shaohuan Wu
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Correspondence: (S.W.); (C.V.)
| | - Smruti Pushalkar
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Shuvadeep Maity
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Birla Institute of Technology and Science (BITS)-Pilani (Hyderabad Campus), Hyderabad 500078, India
| | - Matthew Pressler
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Justin Rendleman
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Burcu Vitrinel
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Michael Carlock
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Ted Ross
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Christine Vogel
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
- Correspondence: (S.W.); (C.V.)
| |
Collapse
|
33
|
Forst CV, Chung M, Hockman M, Lashua L, Adney E, Hickey A, Carlock M, Ross T, Ghedin E, Gresham D. Vaccination History, Body Mass Index, Age, and Baseline Gene Expression Predict Influenza Vaccination Outcomes. Viruses 2022; 14:2446. [PMID: 36366544 PMCID: PMC9697051 DOI: 10.3390/v14112446] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Seasonal influenza is a primary public health burden in the USA and globally. Annual vaccination programs are designed on the basis of circulating influenza viral strains. However, the effectiveness of the seasonal influenza vaccine is highly variable between seasons and among individuals. A number of factors are known to influence vaccination effectiveness including age, sex, and comorbidities. Here, we sought to determine whether whole blood gene expression profiling prior to vaccination is informative about pre-existing immunological status and the immunological response to vaccine. We performed whole transcriptome analysis using RNA sequencing (RNAseq) of whole blood samples obtained prior to vaccination from 275 participants enrolled in an annual influenza vaccine trial. Serological status prior to vaccination and 28 days following vaccination was assessed using the hemagglutination inhibition assay (HAI) to define baseline immune status and the response to vaccination. We find evidence that genes with immunological functions are increased in expression in individuals with higher pre-existing immunity and in those individuals who mount a greater response to vaccination. Using a random forest model, we find that this set of genes can be used to predict vaccine response with a performance similar to a model that incorporates physiological and prior vaccination status alone. A model using both gene expression and physiological factors has the greatest predictive power demonstrating the potential utility of molecular profiling for enhancing prediction of vaccine response. Moreover, expression of genes that are associated with enhanced vaccination response may point to additional biological pathways that contribute to mounting a robust immunological response to the seasonal influenza vaccine.
Collapse
Affiliation(s)
- Christian V. Forst
- Department of Genetics and Genomic Sciences, Department of Microbiology, Icahn School of Medicine at Mt Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029-6574, USA
| | - Matthew Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20894, USA
| | - Megan Hockman
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Lauren Lashua
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Emily Adney
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Angela Hickey
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| | - Michael Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Ted Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20894, USA
| | - David Gresham
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, USA
| |
Collapse
|
34
|
Chou C, Mohanty S, Kang HA, Kong L, Avila‐Pacheco J, Joshi SR, Ueda I, Devine L, Raddassi K, Pierce K, Jeanfavre S, Bullock K, Meng H, Clish C, Santori FR, Shaw AC, Xavier RJ. Metabolomic and transcriptomic signatures of influenza vaccine response in healthy young and older adults. Aging Cell 2022; 21:e13682. [PMID: 35996998 PMCID: PMC9470889 DOI: 10.1111/acel.13682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/25/2023] Open
Abstract
Seasonal influenza causes mild to severe respiratory infections and significant morbidity, especially in older adults. Transcriptomic analysis in populations across multiple flu seasons has provided insights into the molecular determinants of vaccine response. Still, the metabolic changes that underlie the immune response to influenza vaccination remain poorly characterized. We performed untargeted metabolomics to analyze plasma metabolites in a cohort of younger and older subjects before and after influenza vaccination to identify vaccine-induced molecular signatures. Metabolomic and transcriptomic data were combined to define networks of gene and metabolic signatures indicative of high and low antibody response in these individuals. We observed age-related differences in metabolic baselines and signatures of antibody response to influenza vaccination and the abundance of α-linolenic and linoleic acids, sterol esters, fatty-acylcarnitines, and triacylglycerol metabolism. We identified a metabolomic signature associated with age-dependent vaccine response, finding increased tryptophan and decreased polyunsaturated fatty acids (PUFAs) in young high responders (HRs), while fatty acid synthesis and cholesteryl esters accumulated in older HRs. Integrated metabolomic and transcriptomic analysis shows that depletion of PUFAs, which are building blocks for prostaglandins and other lipid immunomodulators, in young HR subjects at Day 28 is related to a robust immune response to influenza vaccination. Increased glycerophospholipid levels were associated with an inflammatory response in older HRs to flu vaccination. This multi-omics approach uncovered age-related molecular markers associated with influenza vaccine response and provides insight into vaccine-induced metabolic responses that may help guide development of more effective influenza vaccines.
Collapse
Affiliation(s)
- Chih‐Hung Chou
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Subhasis Mohanty
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | | | - Lingjia Kong
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | | | - Samit R. Joshi
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Ikuyo Ueda
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Lesley Devine
- Department of Laboratory MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Khadir Raddassi
- Department of NeurologyYale School of MedicineNew HavenConnecticutUSA
| | - Kerry Pierce
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | | | - Kevin Bullock
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Hailong Meng
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Clary Clish
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Fabio R. Santori
- Center for Molecular MedicineUniversity of GeorgiaAthensGeorgiaUSA
| | - Albert C. Shaw
- Section of Infectious Diseases, Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
| | - Ramnik J. Xavier
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Klarman Cell ObservatoryBroad Institute of Harvard and MITCambridgeMassachusettsUSA
- Center for Computational and Integrative Biology and Department of Molecular BiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
35
|
Tomalka JA, Suthar MS, Diamond MS, Sekaly RP. Innate antiviral immunity: how prior exposures can guide future responses. Trends Immunol 2022; 43:696-705. [PMID: 35907675 DOI: 10.1016/j.it.2022.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
Abstract
Innate immunity is an intrinsic baseline defense in cells, with its earliest origins in bacteria, and with key roles in defense against pathogens and in the activation of B and T cell responses. In mammals, the efficacy of innate immunity in initiating the cascades that lead to pathogen control results from the interplay of transcriptomic, epigenomic, and proteomic responses regulating immune activation and long-lived pathogen-specific memory responses. Recent studies suggest that intrinsic innate immunity is modulated by individual exposure histories - prior infections, vaccinations, and metabolites of microbial origin - and this promotes, or impairs, the development of efficacious innate immune responses. Understanding how environmental factors regulate innate immunity and boost protection from infection or response to vaccination could be a valuable tool for pandemic preparedness.
Collapse
Affiliation(s)
- Jeffrey A Tomalka
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Pediatrics, Emory National Primate Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Rafick P Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
36
|
Zhang Y, Sun H, Mandava A, Aevermann BD, Kollmann TR, Scheuermann RH, Qiu X, Qian Y. FastMix: a versatile data integration pipeline for cell type-specific biomarker inference. Bioinformatics 2022; 38:4735-4744. [PMID: 36018232 PMCID: PMC9801972 DOI: 10.1093/bioinformatics/btac585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 01/07/2023] Open
Abstract
MOTIVATION Flow cytometry (FCM) and transcription profiling are the two widely used assays in translational immunology research. However, there is no data integration pipeline for analyzing these two types of assays together with experiment variables for biomarker inference. Current FCM data analysis mainly relies on subjective manual gating analysis, which is difficult to be directly integrated with other automated computational methods. Existing deconvolutional analysis of bulk transcriptomics relies on predefined marker genes in the transcriptomics data, which are unavailable for novel cell types and does not utilize the FCM data that provide canonical phenotypic definitions of the cell types. RESULTS We developed a novel analytics pipeline-FastMix-for computational immunology, which integrates flow cytometry, bulk transcriptomics and clinical covariates for identifying cell type-specific gene expression signatures and biomarker genes. FastMix addresses the 'large p, small n' problem in the gene expression and flow cytometry integration analysis via a linear mixed effects model (LMER) for both cross-sectional and longitudinal studies. Its novel moment-based estimator not only reduces bias in parameter estimation but also is more efficient than iterative optimization. The FastMix pipeline also includes a cutting-edge flow cytometry data analysis method-DAFi-for identifying cell populations of interest and their characteristics. Simulation studies showed that FastMix produced smaller type I/II errors than competing methods. Validation using real data of two vaccine studies showed that FastMix identified a consistent set of signature genes as in independent single-cell RNA-seq analysis, producing additional interesting findings. AVAILABILITY AND IMPLEMENTATION Source code of FastMix is publicly available at https://github.com/terrysun0302/FastMix. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | | | - Aishwarya Mandava
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Brian D Aevermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Tobias R Kollmann
- Systems Vaccinology, Telethon Kids Institute, Perth Children’s Hospital, University of Western Australia, Nedlands, WA 6009, Australia
| | - Richard H Scheuermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA 92037, USA,Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xing Qiu
- To whom correspondence should be addressed. or
| | - Yu Qian
- To whom correspondence should be addressed. or
| |
Collapse
|
37
|
Lipsit S, Facciuolo A, Scruten E, Wilkinson J, Plastow G, Kusalik A, Napper S. Signaling differences in peripheral blood mononuclear cells of high and low vaccine responders prior to, and following, vaccination in piglets. Vaccine X 2022; 11:100167. [PMID: 35692279 PMCID: PMC9175112 DOI: 10.1016/j.jvacx.2022.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 10/28/2022] Open
Abstract
Individual variability in responses to vaccination can result in vaccinated subjects failing to develop a protective immune response. Vaccine non-responders can remain susceptible to infection and may compromise efforts to achieve herd immunity. Biomarkers of vaccine unresponsiveness could aid vaccine research and development as well as strategically improve vaccine administration programs. We previously vaccinated piglets (n = 117) against a commercial Mycoplasma hyopneumoniae vaccine (RespiSure-One) and observed in low vaccine responder piglets, as defined by serum IgG antibody titers, differential phosphorylation of peptides involved in pro-inflammatory cytokine signaling within peripheral blood mononuclear cells (PBMCs) prior to vaccination, elevated plasma interferon-gamma concentrations, and lower birth weight compared to high vaccine responder piglets. In the current study, we use kinome analysis to investigate signaling events within PBMCs collected from the same high and low vaccine responders at 2 and 6 days post-vaccination. Furthermore, we evaluate the use of inflammatory plasma cytokines, birthweight, and signaling events as biomarkers of vaccine unresponsiveness in a validation cohort of high and low vaccine responders. Differential phosphorylation events (FDR < 0.05) within PBMCs are established between high and low responders at the time of vaccination and at six days post-vaccination. A subset of these phosphorylation events were determined to be consistently differentially phosphorylated (p < 0.05) in the validation cohort of high and low vaccine responders. In contrast, there were no differences in birth weight (p > 0.5) and plasma IFNγ concentrations at the time of vaccination (p > 0.6) between high and low responders within the validation cohort. The results in this study suggest, at least within this study population, phosphorylation biomarkers are more robust predictors of vaccine responsiveness than other physiological markers.
Collapse
Affiliation(s)
- Sean Lipsit
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Antonio Facciuolo
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - Erin Scruten
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada
| | - James Wilkinson
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Graham Plastow
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Anthony Kusalik
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Scott Napper
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
38
|
Xie X, Xie B, Xiong D, Hou M, Zuo J, Wei G, Chevallier J. New theoretical ISM-K2 Bayesian network model for evaluating vaccination effectiveness. JOURNAL OF AMBIENT INTELLIGENCE AND HUMANIZED COMPUTING 2022; 14:1-17. [PMID: 35813275 PMCID: PMC9253264 DOI: 10.1007/s12652-022-04199-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/15/2022] [Indexed: 05/30/2023]
Abstract
Aiming at the difficulty in obtaining a complete Bayesian network (BN) structure directly through search-scoring algorithms, authors attempted to incorporate expert judgment and historical data to construct an interpretive structural model with an ISM-K2 algorithm for evaluating vaccination effectiveness (VE). By analyzing the influenza vaccine data provided by Hunan Provincial Center for Disease Control and Prevention, risk factors influencing VE in each link in the process of "Transportation-Storage-Distribution-Inoculation" were systematically investigated. Subsequently, an evaluation index system of VE and an ISM-K2 BN model were developed. Findings include: (1) The comprehensive quality of the staff handling vaccines has a significant impact on VE; (2) Predictive inference and diagnostic reasoning through the ISM-K2 BN model are stable, effective, and highly interpretable, and consequently, the post-production supervision of vaccines is enhanced. The study provides a theoretical basis for evaluating VE and a scientific tool for tracking the responsibility of adverse events of ineffective vaccines, which has the value of promotion in improving VE and reducing the transmission rate of infectious diseases.
Collapse
Affiliation(s)
- Xiaoliang Xie
- School of Mathematics and Statistics, Hunan University of Technology and Busin Ess, Changsha, 410205 China
- Key Laboratory of Hunan Province for Statistical Learning and Intelligent Computation, Hunan University of Technology and Business, Changsha, 410205 Hunan China
| | - Bingqi Xie
- School of Mathematics and Statistics, Hunan University of Technology and Busin Ess, Changsha, 410205 China
- Institute of Big Data and Internet Innovation, Hunan University of Technology and Business, Changsha, 410205 China
| | - Dan Xiong
- School of Mathematics and Statistics, Central South University, Changsha, 410083 China
| | - Muzhou Hou
- School of Mathematics and Statistics, Central South University, Changsha, 410083 China
| | - Jinxia Zuo
- School of Mathematics and Statistics, Hunan University of Technology and Busin Ess, Changsha, 410205 China
- Institute of Big Data and Internet Innovation, Hunan University of Technology and Business, Changsha, 410205 China
| | - Guo Wei
- Department of Mathematics and Computer Science, University of North Carolina at Pembroke, Pembroke, NC 28372 USA
| | - Julien Chevallier
- IPAG Business School (IPAG Lab), 184 boulevard Saint-Germain, 75006 Paris, France
- University Paris 8 (LED), 2 rue de la Liberté, 93526 Saint-Denis, France
| |
Collapse
|
39
|
Walsh CJ, Batt J, Herridge MS, Mathur S, Bader GD, Hu P, Khatri P, Dos Santos CC. Comprehensive multi-cohort transcriptional meta-analysis of muscle diseases identifies a signature of disease severity. Sci Rep 2022; 12:11260. [PMID: 35789175 PMCID: PMC9253003 DOI: 10.1038/s41598-022-15003-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
Muscle diseases share common pathological features suggesting common underlying mechanisms. We hypothesized there is a common set of genes dysregulated across muscle diseases compared to healthy muscle and that these genes correlate with severity of muscle disease. We performed meta-analysis of transcriptional profiles of muscle biopsies from human muscle diseases and healthy controls. Studies obtained from public microarray repositories fulfilling quality criteria were divided into six categories: (i) immobility, (ii) inflammatory myopathies, (iii) intensive care unit (ICU) acquired weakness (ICUAW), (iv) congenital muscle diseases, (v) chronic systemic diseases, (vi) motor neuron disease. Patient cohorts were separated in discovery and validation cohorts retaining roughly equal proportions of samples for the disease categories. To remove bias towards a specific muscle disease category we repeated the meta-analysis five times by removing data sets corresponding to one muscle disease class at a time in a "leave-one-disease-out" analysis. We used 636 muscle tissue samples from 30 independent cohorts to identify a 52 gene signature (36 up-regulated and 16 down-regulated genes). We validated the discriminatory power of this signature in 657 muscle biopsies from 12 additional patient cohorts encompassing five categories of muscle diseases with an area under the receiver operating characteristic curve of 0.91, 83% sensitivity, and 85.3% specificity. The expression score of the gene signature inversely correlated with quadriceps muscle mass (r = -0.50, p-value = 0.011) in ICUAW and shoulder abduction strength (r = -0.77, p-value = 0.014) in amyotrophic lateral sclerosis (ALS). The signature also positively correlated with histologic assessment of muscle atrophy in ALS (r = 0.88, p-value = 1.62 × 10-3) and fibrosis in muscular dystrophy (Jonckheere trend test p-value = 4.45 × 10-9). Our results identify a conserved transcriptional signature associated with clinical and histologic muscle disease severity. Several genes in this conserved signature have not been previously associated with muscle disease severity.
Collapse
Affiliation(s)
- C J Walsh
- Keenan Research Center for Biomedical Science, Saint Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - J Batt
- Keenan Research Center for Biomedical Science, Saint Michael's Hospital, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - M S Herridge
- Interdepartmental Division of Critical Care, University Health Network, University of Toronto, Toronto, ON, Canada
| | - S Mathur
- Department of Physical Therapy, University of Toronto, Toronto, ON, Canada
| | - G D Bader
- The Donnelly Center, University of Toronto, Toronto, ON, Canada
| | - P Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - P Khatri
- Stanford Institute for Immunity, Transplantation and Infection (ITI), Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, USA
| | - C C Dos Santos
- Keenan Research Center for Biomedical Science, Saint Michael's Hospital, Toronto, ON, Canada. .,Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
40
|
Wu S, Ross TM, Carlock MA, Ghedin E, Choi H, Vogel C. Evaluation of determinants of the serological response to the quadrivalent split-inactivated influenza vaccine. Mol Syst Biol 2022; 18:e10724. [PMID: 35514207 PMCID: PMC9073386 DOI: 10.15252/msb.202110724] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/20/2022] Open
Abstract
The seasonal influenza vaccine is only effective in half of the vaccinated population. To identify determinants of vaccine efficacy, we used data from > 1,300 vaccination events to predict the response to vaccination measured as seroconversion as well as hemagglutination inhibition (HAI) titer levels one year after. We evaluated the predictive capabilities of age, body mass index (BMI), sex, race, comorbidities, vaccination history, and baseline HAI titers, as well as vaccination month and vaccine dose in multiple linear regression models. The models predicted the categorical response for > 75% of the cases in all subsets with one exception. Prior vaccination, baseline titer level, and age were the major determinants of seroconversion, all of which had negative effects. Further, we identified a gender effect in older participants and an effect of vaccination month. BMI had a surprisingly small effect, likely due to its correlation with age. Comorbidities, vaccine dose, and race had negligible effects. Our models can generate a new seroconversion score that is corrected for the impact of these factors which can facilitate future biomarker identification.
Collapse
Affiliation(s)
- Shaohuan Wu
- Center for Genomics and Systems BiologyNew York UniversityNYUSA
| | - Ted M Ross
- Department of Infectious DiseasesCollege of Veterinary MedicineUniversity of GeorgiaAthensGAUSA
- Center for Vaccines and ImmunologyUniversity of GeorgiaAthensGAUSA
| | - Michael A Carlock
- Department of Infectious DiseasesCollege of Veterinary MedicineUniversity of GeorgiaAthensGAUSA
- Center for Vaccines and ImmunologyUniversity of GeorgiaAthensGAUSA
| | - Elodie Ghedin
- Center for Genomics and Systems BiologyNew York UniversityNYUSA
- Systems Genomics SectionLaboratory of Parasitic DiseasesNIAID, NIHBethesdaMDUSA
| | - Hyungwon Choi
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| | - Christine Vogel
- Center for Genomics and Systems BiologyNew York UniversityNYUSA
| |
Collapse
|
41
|
Borgognone A, Noguera-Julian M, Oriol B, Noël-Romas L, Ruiz-Riol M, Guillén Y, Parera M, Casadellà M, Duran C, Puertas MC, Català-Moll F, De Leon M, Knodel S, Birse K, Manzardo C, Miró JM, Clotet B, Martinez-Picado J, Moltó J, Mothe B, Burgener A, Brander C, Paredes R. Gut microbiome signatures linked to HIV-1 reservoir size and viremia control. MICROBIOME 2022; 10:59. [PMID: 35410461 PMCID: PMC9004083 DOI: 10.1186/s40168-022-01247-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/16/2022] [Indexed: 05/28/2023]
Abstract
BACKGROUND The potential role of the gut microbiome as a predictor of immune-mediated HIV-1 control in the absence of antiretroviral therapy (ART) is still unknown. In the BCN02 clinical trial, which combined the MVA.HIVconsv immunogen with the latency-reversing agent romidepsin in early-ART treated HIV-1 infected individuals, 23% (3/13) of participants showed sustained low-levels of plasma viremia during 32 weeks of a monitored ART pause (MAP). Here, we present a multi-omics analysis to identify compositional and functional gut microbiome patterns associated with HIV-1 control in the BCN02 trial. RESULTS Viremic controllers during the MAP (controllers) exhibited higher Bacteroidales/Clostridiales ratio and lower microbial gene richness before vaccination and throughout the study intervention when compared to non-controllers. Longitudinal assessment indicated that the gut microbiome of controllers was enriched in pro-inflammatory bacteria and depleted in butyrate-producing bacteria and methanogenic archaea. Functional profiling also showed that metabolic pathways related to fatty acid and lipid biosynthesis were significantly increased in controllers. Fecal metaproteome analyses confirmed that baseline functional differences were mainly driven by Clostridiales. Participants with high baseline Bacteroidales/Clostridiales ratio had increased pre-existing immune activation-related transcripts. The Bacteroidales/Clostridiales ratio as well as host immune-activation signatures inversely correlated with HIV-1 reservoir size. CONCLUSIONS The present proof-of-concept study suggests the Bacteroidales/Clostridiales ratio as a novel gut microbiome signature associated with HIV-1 reservoir size and immune-mediated viral control after ART interruption. Video abstract.
Collapse
Affiliation(s)
- Alessandra Borgognone
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain.
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Bruna Oriol
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- Universitat Autonoma de Barcelona (UAB), Barcelona, Catalonia, Spain
| | - Laura Noël-Romas
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
| | - Yolanda Guillén
- Institut Mar d'Investigacions mediques (IMIM), CIBERONC, Barcelona, Catalonia, Spain
| | - Mariona Parera
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
| | - Maria Casadellà
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
| | - Clara Duran
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- Universitat Autonoma de Barcelona (UAB), Barcelona, Catalonia, Spain
| | - Maria C Puertas
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
| | - Francesc Català-Moll
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
| | - Marlon De Leon
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Samantha Knodel
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kenzie Birse
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christian Manzardo
- Infectious Diseases Service, Hospital Clinic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - José M Miró
- CIBERINFEC, Madrid, Spain
- Infectious Diseases Service, Hospital Clinic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
- Universitat Autonoma de Barcelona (UAB), Barcelona, Catalonia, Spain
- Fight AIDS Foundation, Infectious Diseases Department, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - José Moltó
- CIBERINFEC, Madrid, Spain
- Fight AIDS Foundation, Infectious Diseases Department, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
- Fight AIDS Foundation, Infectious Diseases Department, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain
| | - Adam Burgener
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain
- CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Barcelona, Catalonia, Spain.
- CIBERINFEC, Madrid, Spain.
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain.
- Universitat Autonoma de Barcelona (UAB), Barcelona, Catalonia, Spain.
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
- Fight AIDS Foundation, Infectious Diseases Department, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain.
- Department of Infectious Diseases Service, Germans Trias i Pujol University Hospital, Barcelona, Catalonia, Spain.
| |
Collapse
|
42
|
Moncunill G, Carnes J, Chad Young W, Carpp L, De Rosa S, Campo JJ, Nhabomba A, Mpina M, Jairoce C, Finak G, Haas P, Muriel C, Van P, Sanz H, Dutta S, Mordmüller B, Agnandji ST, Díez-Padrisa N, Williams NA, Aponte JJ, Valim C, Neafsey DE, Daubenberger C, McElrath MJ, Dobaño C, Stuart K, Gottardo R. Transcriptional correlates of malaria in RTS,S/AS01-vaccinated African children: a matched case–control study. eLife 2022; 11:70393. [PMID: 35060479 PMCID: PMC8782572 DOI: 10.7554/elife.70393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background: In a phase 3 trial in African infants and children, the RTS,S/AS01 vaccine (GSK) showed moderate efficacy against clinical malaria. We sought to further understand RTS,S/AS01-induced immune responses associated with vaccine protection. Methods: Applying the blood transcriptional module (BTM) framework, we characterized the transcriptomic response to RTS,S/AS01 vaccination in antigen-stimulated (and vehicle control) peripheral blood mononuclear cells sampled from a subset of trial participants at baseline and month 3 (1-month post-third dose). Using a matched case–control study design, we evaluated which of these ‘RTS,S/AS01 signature BTMs’ associated with malaria case status in RTS,S/AS01 vaccinees. Antigen-specific T-cell responses were analyzed by flow cytometry. We also performed a cross-study correlates analysis where we assessed the generalizability of our findings across three controlled human malaria infection studies of healthy, malaria-naive adult RTS,S/AS01 recipients. Results: RTS,S/AS01 vaccination was associated with downregulation of B-cell and monocyte-related BTMs and upregulation of T-cell-related BTMs, as well as higher month 3 (vs. baseline) circumsporozoite protein-specific CD4+ T-cell responses. There were few RTS,S/AS01-associated BTMs whose month 3 levels correlated with malaria risk. In contrast, baseline levels of BTMs associated with dendritic cells and with monocytes (among others) correlated with malaria risk. The baseline dendritic cell- and monocyte-related BTM correlations with malaria risk appeared to generalize to healthy, malaria-naive adults. Conclusions: A prevaccination transcriptomic signature associates with malaria in RTS,S/AS01-vaccinated African children, and elements of this signature may be broadly generalizable. The consistent presence of monocyte-related modules suggests that certain monocyte subsets may inhibit protective RTS,S/AS01-induced responses. Funding: Funding was obtained from the NIH-NIAID (R01AI095789), NIH-NIAID (U19AI128914), PATH Malaria Vaccine Initiative (MVI), and Ministerio de Economía y Competitividad (Instituto de Salud Carlos III, PI11/00423 and PI14/01422). The RNA-seq project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under grant number U19AI110818 to the Broad Institute. This study was also supported by the Vaccine Statistical Support (Bill and Melinda Gates Foundation award INV-008576/OPP1154739 to R.G.). C.D. was the recipient of a Ramon y Cajal Contract from the Ministerio de Economía y Competitividad (RYC-2008-02631). G.M. was the recipient of a Sara Borrell–ISCIII fellowship (CD010/00156) and work was performed with the support of Department of Health, Catalan Government grant (SLT006/17/00109). This research is part of the ISGlobal’s Program on the Molecular Mechanisms of Malaria which is partially supported by the Fundación Ramón Areces and we acknowledge support from the Spanish Ministry of Science and Innovation through the ‘Centro de Excelencia Severo Ochoa 2019–2023’ Program (CEX2018-000806-S), and support from the Generalitat de Catalunya through the CERCA Program.
Collapse
Affiliation(s)
- Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - William Chad Young
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Lindsay Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Stephen De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | | | - Augusto Nhabomba
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | | | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Paige Haas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - Carl Muriel
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Phu Van
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Héctor Sanz
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | | | - Benjamin Mordmüller
- CIBER de Enfermedades Infecciosas
- Institute of Tropical Medicine and German Center for Infection Research
| | - Selidji T Agnandji
- Institute of Tropical Medicine and German Center for Infection Research
- Centre de Recherches Médicales de Lambaréné (CERMEL), BP 242
| | | | | | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | - Clarissa Valim
- Department of Global Health, Boston University School of Public Health
| | - Daniel E Neafsey
- Broad Institute of Massachusetts Institute of Technology and Harvard
- Harvard T.H. Chan School of Public Health
| | | | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Departments of Laboratory Medicine and Medicine, University of Washington
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Department of Pediatrics, University of Washington
- Department of Global Health, University of Washington
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- University of Lausanne and Centre Hospitalier Universitaire Vaudois
| |
Collapse
|
43
|
Giacomelli Cao R, Christian L, Xu Z, Jaramillo L, Smith B, Karlsson EA, Schultz-Cherry S, Mejias A, Ramilo O. Early Changes in Interferon Gene Expression and Antibody Responses Following Influenza Vaccination in Pregnant Women. J Infect Dis 2022; 225:341-351. [PMID: 34197595 PMCID: PMC8915434 DOI: 10.1093/infdis/jiab345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/29/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Influenza immunization during pregnancy provides protection to the mother and the infant. Studies in adults and children with inactivated influenza vaccine have identified changes in immune gene expression that were correlated with antibody responses. The current study was performed to define baseline blood transcriptional profiles and changes induced by inactivated influenza vaccine in pregnant women and to identify correlates with antibody responses. METHODS Pregnant women were immunized with inactivated influenza vaccine during the 2013-2014 and 2014-2015 seasons. Blood samples were collected on day 0 (before vaccination) and on days 1 and 7 after vaccination for transcriptional profile analyses, and on days 0 and 30, along with delivery and cord blood samples, to measure antibody titers. RESULTS Transcriptional analysis demonstrated overexpression of interferon-stimulated genes (ISGs) on day 1 and of plasma cell genes on day 7. Prevaccination ISG expression and ISGs overexpressed on day 1 were significantly correlated with increased H3N2, B Yamagata, and B Victoria antibody titers. Plasma cell gene expression on day 7 was correlated with increased B Yamagata and B Victoria antibody titers. Compared with women who were vaccinated during the previous influenza season, those who were not showed more frequent significant correlations between ISGs and antibody titers. CONCLUSIONS Influenza vaccination in pregnant women resulted in enhanced expression of ISGs and plasma cell genes correlated with antibody responses. Brief summary: This study identified gene expression profiles of interferon-stimulated genes and plasma cells before vaccination and early after vaccination that were correlated with antibody responses in pregnant women vaccinated for influenza.
Collapse
Affiliation(s)
- Raquel Giacomelli Cao
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Lisa Christian
- Institute for Behavioral Medical Research, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Psychiatry, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Zhaohui Xu
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University, Columbus, Ohio, USA
| | - Lisa Jaramillo
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Bennett Smith
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Erik A Karlsson
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Current affiliation: Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Division of Infectious Diseases, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University, Columbus, Ohio, USA
| | - Octavio Ramilo
- Center for Vaccines and Immunity, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Division of Infectious Diseases, Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
44
|
Preimmunization correlates of protection shared across malaria vaccine trials in adults. NPJ Vaccines 2022; 7:5. [PMID: 35031601 PMCID: PMC8760258 DOI: 10.1038/s41541-021-00425-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/02/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying preimmunization biological characteristics that promote an effective vaccine response offers opportunities for illuminating the critical immunological mechanisms that confer vaccine-induced protection, for developing adjuvant strategies, and for tailoring vaccination regimens to individuals or groups. In the context of malaria vaccine research, studying preimmunization correlates of protection can help address the need for a widely effective malaria vaccine, which remains elusive. In this study, common preimmunization correlates of protection were identified using transcriptomic data from four independent, heterogeneous malaria vaccine trials in adults. Systems-based analyses showed that a moderately elevated inflammatory state prior to immunization was associated with protection against malaria challenge. Functional profiling of protection-associated genes revealed the importance of several inflammatory pathways, including TLR signaling. These findings, which echo previous studies that associated enhanced preimmunization inflammation with protection, illuminate common baseline characteristics that set the stage for an effective vaccine response across diverse malaria vaccine strategies in adults.
Collapse
|
45
|
Wang G, Lv C, Liu C, Shen W. Neutrophil-to-lymphocyte ratio as a potential biomarker in predicting influenza susceptibility. Front Microbiol 2022; 13:1003380. [PMID: 36274727 PMCID: PMC9583527 DOI: 10.3389/fmicb.2022.1003380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Human population exposed to influenza viruses exhibited wide variation in susceptibility. The ratio of neutrophils to lymphocytes (NLR) has been examined to be a marker of systemic inflammation. We sought to investigate the relationship between influenza susceptibility and the NLR taken before influenza virus infection. METHODS We investigated blood samples from five independent influenza challenge cohorts prior to influenza inoculation at the cellular level by using digital cytometry. We used multi-cohort gene expression analysis to compare the NLR between the symptomatic infected (SI) and asymptomatic uninfected (AU) subjects. We then used a network analysis approach to identify host factors associated with NLR and influenza susceptibility. RESULTS The baseline NLR was significantly higher in the SI group in both discovery and validation cohorts. The NLR achieved an AUC of 0.724 on the H3N2 data, and 0.736 on the H1N1 data in predicting influenza susceptibility. We identified four key modules that were not only significantly correlated with the baseline NLR, but also differentially expressed between the SI and AU groups. Genes within these four modules were enriched in pathways involved in B cell-mediated immune responses, cellular metabolism, cell cycle, and signal transduction, respectively. CONCLUSIONS This study identified the NLR as a potential biomarker for predicting disease susceptibility to symptomatic influenza. An elevated NLR was detected in susceptible hosts, who may have defects in B cell-mediated immunity or impaired function in cellular metabolism, cell cycle or signal transduction. Our work can serve as a comparative model to provide insights into the COVID-19 susceptibility.
Collapse
Affiliation(s)
- Guoyun Wang
- Department of Bioinformatics, Shantou University Medical College, Shantou, China
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Cheng Lv
- Department of Bioinformatics, Shantou University Medical College, Shantou, China
| | - Cheng Liu
- Department of Computer Science, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou, China
| | - Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou, China
- *Correspondence: Wenjun Shen
| |
Collapse
|
46
|
Human immune diversity: from evolution to modernity. Nat Immunol 2021; 22:1479-1489. [PMID: 34795445 DOI: 10.1038/s41590-021-01058-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
The extreme diversity of the human immune system, forged and maintained throughout evolutionary history, provides a potent defense against opportunistic pathogens. At the same time, this immune variation is the substrate upon which a plethora of immune-associated diseases develop. Genetic analysis suggests that thousands of individually weak loci together drive up to half of the observed immune variation. Intense selection maintains this genetic diversity, even selecting for the introgressed Neanderthal or Denisovan alleles that have reintroduced variation lost during the out-of-Africa migration. Variations in age, sex, diet, environmental exposure, and microbiome each potentially explain the residual variation, with proof-of-concept studies demonstrating both plausible mechanisms and correlative associations. The confounding interaction of many of these variables currently makes it difficult to assign definitive contributions. Here, we review the current state of play in the field, identify the key unknowns in the causality of immune variation, and identify the multidisciplinary pathways toward an improved understanding.
Collapse
|
47
|
Aevermann BD, Shannon CP, Novotny M, Ben-Othman R, Cai B, Zhang Y, Ye JC, Kobor MS, Gladish N, Lee AHY, Blimkie TM, Hancock RE, Llibre A, Duffy D, Koff WC, Sadarangani M, Tebbutt SJ, Kollmann TR, Scheuermann RH. Machine Learning-Based Single Cell and Integrative Analysis Reveals That Baseline mDC Predisposition Correlates With Hepatitis B Vaccine Antibody Response. Front Immunol 2021; 12:690470. [PMID: 34777332 PMCID: PMC8588842 DOI: 10.3389/fimmu.2021.690470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/25/2021] [Indexed: 01/23/2023] Open
Abstract
Vaccination to prevent infectious disease is one of the most successful public health interventions ever developed. And yet, variability in individual vaccine effectiveness suggests that a better mechanistic understanding of vaccine-induced immune responses could improve vaccine design and efficacy. We have previously shown that protective antibody levels could be elicited in a subset of recipients with only a single dose of the hepatitis B virus (HBV) vaccine and that a wide range of antibody levels were elicited after three doses. The immune mechanisms responsible for this vaccine response variability is unclear. Using single cell RNA sequencing of sorted innate immune cell subsets, we identified two distinct myeloid dendritic cell subsets (NDRG1-expressing mDC2 and CDKN1C-expressing mDC4), the ratio of which at baseline (pre-vaccination) correlated with the immune response to a single dose of HBV vaccine. Our results suggest that the participants in our vaccine study were in one of two different dendritic cell dispositional states at baseline – an NDRG2-mDC2 state in which the vaccine elicited an antibody response after a single immunization or a CDKN1C-mDC4 state in which the vaccine required two or three doses for induction of antibody responses. To explore this correlation further, genes expressed in these mDC subsets were used for feature selection prior to the construction of predictive models using supervised canonical correlation machine learning. The resulting models showed an improved correlation with serum antibody titers in response to full vaccination. Taken together, these results suggest that the propensity of circulating dendritic cells toward either activation or suppression, their “dispositional endotype” at pre-vaccination baseline, could dictate response to vaccination.
Collapse
Affiliation(s)
- Brian D Aevermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
| | - Casey P Shannon
- Prevention of Organ Failure (PROOF) Centre of Excellence, St. Paul's Hospital, Vancouver, BC, Canada.,The University of British Columbia (UBC) Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Mark Novotny
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
| | - Rym Ben-Othman
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Telethon Kids Institute, Perth Children's Hospital, University of Western Australia, Nedlands, WA, Australia
| | - Bing Cai
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Yun Zhang
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States
| | - Jamie C Ye
- Prevention of Organ Failure (PROOF) Centre of Excellence, St. Paul's Hospital, Vancouver, BC, Canada.,The University of British Columbia (UBC) Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Michael S Kobor
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Gladish
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Amy Huei-Yi Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Travis M Blimkie
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Robert E Hancock
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Alba Llibre
- Translational Immunology Lab, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Translational Immunology Lab, Institut Pasteur, Paris, France
| | - Wayne C Koff
- Human Vaccines Project, New York, NY, United States
| | - Manish Sadarangani
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Scott J Tebbutt
- Prevention of Organ Failure (PROOF) Centre of Excellence, St. Paul's Hospital, Vancouver, BC, Canada.,The University of British Columbia (UBC) Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R Kollmann
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Telethon Kids Institute, Perth Children's Hospital, University of Western Australia, Nedlands, WA, Australia
| | - Richard H Scheuermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States.,Department of Pathology, University of California, San Diego, San Diego, CA, United States.,Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| |
Collapse
|
48
|
Sakaram S, Hasin-Brumshtein Y, Khatri P, He YD, Sweeney TE. A Multi-mRNA Prognostic Signature for Anti-TNFα Therapy Response in Patients with Inflammatory Bowel Disease. Diagnostics (Basel) 2021; 11:1902. [PMID: 34679598 PMCID: PMC8534494 DOI: 10.3390/diagnostics11101902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Anti-TNF-alpha (anti-TNFα) therapies have transformed the care and management of inflammatory bowel disease (IBD). However, they are expensive and ineffective in greater than 50% of patients, and they increase the risk of infections, liver issues, arthritis, and lymphoma. With 1.6 million Americans suffering from IBD and global prevalence on the rise, there is a critical unmet need in the use of anti-TNFα therapies: a test for the likelihood of therapy response. Here, as a proof-of-concept, we present a multi-mRNA signature for predicting response to anti-TNFα treatment to improve the efficacy and cost-to-benefit ratio of these biologics. METHODS We surveyed public data repositories and curated four transcriptomic datasets (n = 136) from colonic and ileal mucosal biopsies of IBD patients (pretreatment) who were subjected to anti-TNFα therapy and subsequently adjudicated for response. We applied a multicohort analysis with a leave-one-study-out (LOSO) approach, MetaIntegrator, to identify significant differentially expressed (DE) genes between responders and non-responders and then used a greedy forward search to identify a parsimonious gene signature. We then calculated an anti-TNFα response (ATR) score based on this parsimonious gene signature to predict responder status and assessed discriminatory performance via an area-under-receiver operating-characteristic curve (AUROC). RESULTS We identified 324 significant DE genes between responders and non-responders. The greedy forward search yielded seven genes that robustly distinguish anti-TNFα responders from non-responders, with an AUROC of 0.88 (95% CI: 0.70-1). The Youden index yielded a mean sensitivity of 91%, mean specificity of 76%, and mean accuracy of 86%. CONCLUSIONS Our findings suggest that there is a robust transcriptomic signature for predicting anti-TNFα response in mucosal biopsies from IBD patients prior to treatment initiation. This seven-gene signature should be further investigated for its potential to be translated into a predictive test for clinical use.
Collapse
Affiliation(s)
- Suraj Sakaram
- Inflammatix, Inc., 863 Mitten Rd., Suite 104, Burlingame, CA 94010, USA; (S.S.); (Y.H.-B.)
| | | | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA 94305, USA;
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yudong D. He
- Inflammatix, Inc., 863 Mitten Rd., Suite 104, Burlingame, CA 94010, USA; (S.S.); (Y.H.-B.)
| | - Timothy E. Sweeney
- Inflammatix, Inc., 863 Mitten Rd., Suite 104, Burlingame, CA 94010, USA; (S.S.); (Y.H.-B.)
| |
Collapse
|
49
|
Bukhari SAC, Pawar S, Mandell J, Kleinstein SH, Cheung KH. LinkedImm: a linked data graph database for integrating immunological data. BMC Bioinformatics 2021; 22:105. [PMID: 34433410 PMCID: PMC8385794 DOI: 10.1186/s12859-021-04031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 11/23/2022] Open
Abstract
Background Many systems biology studies leverage the integration of multiple data types (across different data sources) to offer a more comprehensive view of the biological system being studied. While SQL (Structured Query Language) databases are popular in the biomedical domain, NoSQL database technologies have been used as a more relationship-based, flexible and scalable method of data integration. Results We have created a graph database integrating data from multiple sources. In addition to using a graph-based query language (Cypher) for data retrieval, we have developed a web-based dashboard that allows users to easily browse and plot data without the need to learn Cypher. We have also implemented a visual graph query interface for users to browse graph data. Finally, we have built a prototype to allow the user to query the graph database in natural language. Conclusion We have demonstrated the feasibility and flexibility of using a graph database for storing and querying immunological data with complex biological relationships. Querying a graph database through such relationships has the potential to discover novel relationships among heterogeneous biological data and metadata.
Collapse
Affiliation(s)
- Syed Ahmad Chan Bukhari
- Division of Computer Science, Mathematics and Science, Collins College of Professional Studies, St. John's University, New York, NY, USA
| | - Shrikant Pawar
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Jeff Mandell
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT, USA
| | - Steven H Kleinstein
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kei-Hoi Cheung
- Program in Computational Biology and Bioinformatics, Yale School of Medicine, New Haven, CT, USA. .,Department of Emergency Medicine, Yale School of Medicine, New Haven, CT, USA. .,Yale Center for Medical Informatics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
50
|
Wimmers F, Donato M, Kuo A, Ashuach T, Gupta S, Li C, Dvorak M, Foecke MH, Chang SE, Hagan T, De Jong SE, Maecker HT, van der Most R, Cheung P, Cortese M, Bosinger SE, Davis M, Rouphael N, Subramaniam S, Yosef N, Utz PJ, Khatri P, Pulendran B. The single-cell epigenomic and transcriptional landscape of immunity to influenza vaccination. Cell 2021; 184:3915-3935.e21. [PMID: 34174187 PMCID: PMC8316438 DOI: 10.1016/j.cell.2021.05.039] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/15/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
Emerging evidence indicates a fundamental role for the epigenome in immunity. Here, we mapped the epigenomic and transcriptional landscape of immunity to influenza vaccination in humans at the single-cell level. Vaccination against seasonal influenza induced persistently diminished H3K27ac in monocytes and myeloid dendritic cells (mDCs), which was associated with impaired cytokine responses to Toll-like receptor stimulation. Single-cell ATAC-seq analysis revealed an epigenomically distinct subcluster of monocytes with reduced chromatin accessibility at AP-1-targeted loci after vaccination. Similar effects were observed in response to vaccination with the AS03-adjuvanted H5N1 pandemic influenza vaccine. However, this vaccine also stimulated persistently increased chromatin accessibility at interferon response factor (IRF) loci in monocytes and mDCs. This was associated with elevated expression of antiviral genes and heightened resistance to the unrelated Zika and Dengue viruses. These results demonstrate that vaccination stimulates persistent epigenomic remodeling of the innate immune system and reveal AS03's potential as an epigenetic adjuvant.
Collapse
Affiliation(s)
- Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michele Donato
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alex Kuo
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tal Ashuach
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shakti Gupta
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mai Dvorak
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mariko Hinton Foecke
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sarah E Chang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Hagan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sanne E De Jong
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Peggie Cheung
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven E Bosinger
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark Davis
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|