Posters - Schedules

Posters Home

View Posters By Category

Monday, July 11 and Tuesday, July 12 between 12:30 PM CDT and 2:30 PM CDT
Wednesday July 13 between 12:30 PM CDT and 2:30 PM CDT
Session A Poster Set-up and Dismantle Session A Posters set up:
Monday, July 11 between 7:30 AM CDT - 10:00 AM CDT
Session A Posters dismantle:
Tuesday, July 12 at 6:00 PM CDT
Session B Poster Set-up and Dismantle Session B Posters set up:
Wednesday, July 13 between 7:30 AM - 10:00 AM CDT
Session B Posters dismantle:
Thursday. July 14 at 2:00 PM CDT
Virtual: Bioinformatics analysis for single-cell immune profiling of Q fever vaccination in mice
COSI: ssci
  • Li Su, University of Missouri at Columbia, United States
  • Muhammad Alam, The University of Texas at San Antonio, United States
  • Venkatesh Kumaresan, The University of Texas at San Antonio, United States
  • Guoquan Zhang, The University of Texas at San Antonio, United States
  • Dong Xu, University of Missouri at Columbia, United States


Presentation Overview: Show

Human Q fever is a worldwide zoonotic disease caused by the bacterium Coxiella burnetii with high infectious rate. However, there is no FDA-approved vaccine. A previous study found that the formalin-inactivated PI vaccine (PIV) was more protective than the PII vaccine (PIIV) in guinea pigs. Our research objective is to explore the mechanisms of vaccine-induced protective immunity against C. burnetii infection. For this purpose, we utilized 10x Genomics immune profiling technology to simultaneously assay T cell, B cell receptor sequences, transcriptional profiles, and surface protein expression at the single-cell level. Our bioinformatics analysis leveraged the Seurat Weighted Nearest Neighbor algorithm to integrate transcriptional profiles and surface protein expressions to perform cell clustering. After annotating cell types, we focused on B cells and CD4+ T cells to study adaptive immunity in different vaccination groups. To identify specific vaccine-responding clonotypes, we performed B cells trajectory inference, TCR and BCR V(D)J usage, and clonotype analysis. This study helps understand the mechanism of vaccine-induced innate and adaptive immunity against C. burnetii infection and facilitates the discovery of safe and effective vaccines for Q fever.

Virtual: Deciphering the underlying dynamics during injury
COSI: ssci
  • Ranojoy Chatterjee, The George Washington University, United States
  • Brett Shook, The George Washington University, United States
  • Ali Rahnavard, The George Washington University, United States


Presentation Overview: Show

Skin tissue repair and recovery after injury is important to the healing process and organ homeostasis. The outermost skin layer, the epidermis, is comprised of stem cells which are key players in the recovery process. This repair process is hallmarked by multiple complex biological pathways that involve coordination between growth factors, cytokines, and different cell types including resident immune cells, fibroblasts, keratinocytes, and melanocytes. However, there is limited understanding of the interplay of these cells and secreted products and their relationships dictating the recovery process. The recent development of single-cell RNA-sequencing has provided the power to extract transcriptomic, spatial, and lineage information for each individual cell and affords a unique opportunity to study the interactions occurring during skin wound healing. In this study, we elucidate the precise cell populations and their changes in gene expression caused by injury to the skin using non-injured tissue as a comparator. Using a Seurat workflow, we integrated and analyzed injured and non-injured samples for differentially expressed genes across both conditions using the tweediverse R package. We also investigated cell-cell communications occurring during injury and identified key genes marking biological changes due to injury.

Virtual: On the Best Strategy for Generation of Personalized Neoantigens for Cancer Immunotherapy
COSI: ssci
  • Laurie Prelot, Department of Computer Science, ETH Zürich, Switzerland
  • Matthias Hüser, Department of Computer Science, ETH Zürich, Switzerland
  • Jiayu Chen, Department of Computer Science, ETH Zürich, Switzerland
  • Andre Kahles, Department of Computer Science, ETH Zürich, Switzerland
  • Kjong Lehmann, Department of Computer Science, ETH Zürich, Switzerland
  • Gunnar Rätsch, Department of Computer Science, ETH Zürich, Switzerland


Presentation Overview: Show

In immuno-oncology, cancer vaccines train the immune system to recognize proteins presented by tumors (neoantigens). The search-space for neoantigens is immensely large and the cost of developing personalized vaccines correspondingly high, therefore systematic and reliable ways of finding promising candidates are needed. We further developed our novel software ImmunoPepper for identifying cancer-specific antigens from complex splicing graphs with patient-specific mutations. It allows for the generation of mutation and splicing-derived peptides. Moreover, ImmunoPepper enables several filtering steps. Finally, ImmunoPepper supports in-silico prediction of binding to the MHC-I complex. We test the ImmunoPepper software on five BRCA samples, and make recommendations for best proteomic validation of the predicted neoantigens. We first build splicing graphs of BRCA and GTEX cohorts and generate splicing-derived neoantigens from the samples. Then, we filter the candidates against a cohort of normal samples, and we set a threshold of RNA expression support in the sample, as well as a threshold for recurrence in a cohort of 1102 BRCA samples. We combine the latter filters, leading to experimental sets of candidates. We search the candidates against MS spectra and conclude about the validation rate of the putative neoantigens generated by ImmunoPepper as a function of the filtering strategy.

Virtual: Using Deep Learning in Lyme Disease Diagnosis
COSI: ssci
  • Tejaswi Koduru, Thomas Jefferson High School for Science and Technology, United States
  • Tejaswi Koduru, Thomas Jefferson High School for Science and Technology, United States


Presentation Overview: Show

Untreated lyme disease can lead to neurological, cardiac, and dermatological complications. Rapid diagnosis of the erythema migrans (EM) rash, a characteristic symptom of Lyme disease, is therefore crucial to early diagnosis and treatment. In this study, we aim to utilize deep learning frameworks including Tensorflow and Keras to create deep convolutional neural networks (DCNN) to detect images of acute Lyme Disease from images of erythema migrans. This study uses a custom database of erythema migrans images of varying quality to train a DCNN capable of classifying images of EM rashes vs non-EM rashes. Images from publicly available sources were mined to create an initial database. Machine based removal of duplicate images was then performed, followed by a thorough examination of all images by a clinician. The resulting database was combined with images of confounding rashes and regular skin, resulting in a total of 683 images. This database was then used to create a DCNN with an accuracy of 93% when classifying images of rashes as EM vs non EM. Finally, this model was converted into a web and mobile application to allow for rapid diagnosis of EM rashes by both patients and clinicians.

H-001: Identification of disease-relevant cell types by integrating GWAS signals with epigenetic and scRNA-seq annotations for atopic dermatitis
COSI: ssci
  • Fanying Tang, AbbVie, United States
  • Corneliu Bodea, Abbvie, United States
  • Bridget Riley-Gillis, AbbVie, United States
  • Jozsef Karman, AbbVie, United States
  • Michael Macoritto, AbbVie, United States
  • Fedik Rahimov, AbbVie, United States
  • Wade Davis, AbbVie, United States
  • Kathleen Smith, AbbVie, United States
  • Josue Samayao, AbbVie, United States


Presentation Overview: Show

The causal cell types in many autoimmune diseases, including atopic dermatitis (AD), are still unclear. Identifying these disease-relevant cell types is critical for the choice of proper experimental model system, understanding the disease etiology and facilitating the development of targeted therapies.

To answer this question, we utilize GWAS SNP enrichment analysis which connects GWAS signals with cell-type specific functional annotations to prioritize the cell types. To be more specific, we applied LDSC-SEG with epigenetic annotations of immune cells isolated from healthy donors, on GWAS summary statistics to identify the disease relevant cells in AD. The results indicate variants associated with AD are mainly enriched with T cells including Th2 cells.

Moreover, we constructed cell-type programs and disease-progression programs for each cell type using scRNA-seq data from the skin samples of healthy controls and AD patients, and developed MAGMA_sc, a methodology built on MAGMA that is tailored for scRNA-seq data, to combine these RNA expression-based programs and GWAS summary statistics. The results revealed additional enrichment of keratinocytes during disease progression in AD.

Together these results demonstrate GWAS SNP enrichment analysis is a powerful bioinformatic strategy to leverage epigenetic and transcriptomic data for interpreting GWAS signals and identifying disease-relevant cell types.

H-002: Defining immune cells expression profiles with single-cell RNAseq in Rheumatoid Arthritis patients naive to treatment
COSI: ssci
  • Jean Vencic, Universite de Sherbrooke, Canada
  • Sophie Roux, Universite de Sherbrooke, Canada
  • Michelle Scott, Universite de Sherbrooke, Canada
  • Hugues Allard-Chamard, Universite de Sherbrooke, Canada


Presentation Overview: Show

Rheumatoid arthritis (RA) is a chronic, inflammatory and autoimmune disease affecting 1% of the worldwide population. It is characterized by symptomatic flares during which significant inflammation and destruction of the joints appear. The pathophysiology of the disease remains poorly understood and the available treatments can only alleviate its symptoms and rarely induce long-term remission. We currently lack effective tools to predict the course and response of RA to treatments.

RA is a heterogeneous entity resulting in not all patients responding to the same treatments. We posit that this difference is due to a plurality of immune alterations causing distinct immune cell expression profiles namely the RA endophenotypes.

To conduct this study, single-cell RNA sequencing data have been generated from blood mononuclear cell samples of patients presenting with RA, prior to the initiation of treatments. Using bioinformatics methods we aim to discriminate and define specific RA endophenotypes.

The long-term objectives of the project will be to study the possible correlation between immune endophenotypes and response to treatment. Developing a tool capable of linking a new expression profile to a characterized endophenotype (e.i. aggressiveness or response to treatment) would pave the way for personalized medicine in RA.

H-003: The T cell receptor repertoire reflects the dynamics of the immune response to vaccination
COSI: ssci
  • Ravi Sachidanandam, Girihlet Inc., United States


Presentation Overview: Show

The T cell receptor (TCR) provides an early, high-resolution metric to ascertain the immune response to vaccinations, with the complete TCR repertoire reflecting the T cells present in an individual. To this end, we developed Tseek, an unbiased and accurate method for profiling the TCR repertoire and used it to explore the T cell response to both the COVID-19 mRNA vaccine (n=9) and the seasonal inactivated Influenza vaccine (n=5) at several time points. The COVID-19 vaccine elicited a broad T cell response involving multiple expanded clones, whereas the Influenza vaccine elicited a narrower response involving fewer clones. Many distinct T cell clones responded at each sample, providing temporal details lacking in the antibody measurements. Neutralizing antibody titers were also measured in the covid vaccine samples, and the TCR responses broadly presaged the antibody responses. The physical symptoms following vaccinations did not reflect the TCR/antibody responses. The TCR repertoire is an individual fingerprint: donors of blood samples taken years apart could be identified solely based upon their TCR repertoire. These results demonstrate the promise of non-invasive (PBMCs) monitoring of the TCR repertoire, as an early, sensitive measure of the adaptive immune response to vaccination, to help various aspects of vaccine design.

H-004: scPerformer: Single-Cell Classification of Immune Cell Subsets Using Transformers
COSI: ssci
  • Abbas Roayaei Ardakany, La Jolla Institute for Immunology, United States
  • Ferhat Ay, La Jolla Institute for Immunology, United States
  • Sourya Bhattacharyya , La Jolla Institute for Immunology, United States


Presentation Overview: Show

Single-cell RNA sequencing (scRNA-seq) allows us to distinguish clusters/subsets of cells with distinct functions. Given the growing size and complexity of the produced datasets, powerful and robust models are needed to identify clusters across different platforms and experimental settings. A new deep learning architecture named Transformers are particularly powerful in learning meaningful and interpretable representations (encodings) of data. Here, we present scPerformer that utilizes Transformers for classifying cell types from scRNA-seq data. To evaluate our method, we first showed that it has comparable performance to state-of-the-art models on smaller datasets but tends to outperform them as the number of cells and/or cell types increases. We then tested our model on a large dataset (~800k cells) of activated CD4+ T cells pre-sorted into different memory subsets. scPerformer distinguished fine-resolution T-cell subsets accurately. Next, we applied scPerformer on two large-scale CITE-seq datasets of circulating immune cells from COVID-19 patients showing that it recapitulates clusters obtained using surface protein expression using only gene expression. Lastly, by attributing the prediction of the scPerformer model to its input features (genes) using the Integrated Gradients, we were able to extract the important genes that drive the correct classification and distinction between different cell subsets.

H-005: Ultrafast TCR sequence alignment for disease classification
COSI: ssci
  • Bo Li, UT Southwestern Medical Center, United States
  • Hongyi Zhang, UT Southwestern Medical Center, United States


Presentation Overview: Show

Similarity in T-cell receptor (TCR) sequences implies shared antigen specificity between receptors, and could be used to discover novel therapeutic targets. However, existing methods cluster clustering T-cell receptor sequences by similarity are computationally inefficient, making them impractical to use on the ever-expanding datasets of the immune repertoire. Here, we developed GIANA (Geometric Isometry based TCR AligNment Algorithm) a computationally efficient tool for this task that provides the same level of clustering specificity as TCRdist at 600 times its speed, and without sacrificing accuracy. GIANA also allows the rapid query of large reference cohorts within minutes. Using GIANA to cluster large-scale TCR datasets provides candidate disease-specific receptors, and provides a new solution to repertoire classification. Querying unseen TCR-seq samples against an existing reference differentiates samples from patients across various cohorts associated with cancer, infectious and autoimmune disease. Our results demonstrate how GIANA could be used as the basis for a TCR-based non-invasive multi-disease diagnostic platform.

H-006: Full-length transcriptome analysis with ‘Nexons’ reveals the regulation of poison exons in splicing factors in human germinal centre B cells.
COSI: ssci
  • Ozge Gizlenci, Immunology Programme, The Babraham Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Louise Matheson, Immunology Programme, The Babraham Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Simon Andrews, Bioinformatics Group, The Babraham Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Laura Biggins, Bioinformatics Group, The Babraham Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Jingyu Chen, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Rebecca Berrens, University of Oxford, Oxford, UK, United Kingdom
  • Daniel J. Hodson, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK, United Kingdom
  • Martin Turner, Immunology Programme, The Babraham Institute, University of Cambridge, Cambridge, UK, United Kingdom


Presentation Overview: Show

Alternative splicing (AS) plays a major role in the differentiation of immune cells during an immune response as 29% of AS genes are specific to the immune system. Although the role of AS is extensively investigated in T cells, its role in B cell activation is less characterised. We sought to develop a long-read technology ONT workflow to understand post-transcriptional regulation at both gene and isoform levels of human germinal centre (GC) B cells. As one of the challenges of ONT is the accurate computational analysis of isoforms, we developed ‘Nexons’ pipeline to identify the differentially spliced transcript variants using long-read sequencing. An in-depth analysis of splicing regulators with the Nexons revealed the differential regulation of the poison exon (PE) in splicing regulators (e.g. SRSF3) in GC B cells. In GC B cells, PEs of the splicing factors were preferentially spliced out whereas naïve B cells expressed isoforms carrying PE, leading to nonsense-mediated mRNA decay. Moreover, we identified novel spliced variants of these genes, which were undetectable due to the limitations of short-read data. Altogether, our findings validate the combination of Nexons with Smart-seq2 adapted ONT RNA-sequencing workflow as a suitable method for the identification and quantification of complex isoforms.

H-007: mvTCR - Integrating T-cell receptor and transcriptome for large-scale single-cell immune profiling analysis
COSI: ssci
  • Felix Drost, Institute of Computational Biology, Helmholtz Zentrum München, Germany
  • Yang An, Institute of Computational Biology, Helmholtz Zentrum München, Germany
  • Fabian J. Theis, Institute of Computational Biology, Helmholtz Zentrum München, Germany
  • Mohammad Lotfollahi, Institute of Computational Biology, Helmholtz Zentrum München, Germany
  • Benjamin Schubert, Institute of Computational Biology, Helmholtz Zentrum München, Germany


Presentation Overview: Show

T cells are a pivotal part of the adaptive immunity to control infectious diseases and cancer through the recognition of pathogenic peptides by the T-cell receptor (TCR). Yet, their phenotypic and cellular differentiation in the context of the cell’s TCR, and therefore the fate of the T cell is still poorly understood. Rapidly developing single-cell technologies enable the measurement of multiple biological modalities, including transcriptome, and TCR sequences at the same time offering unprecedented insights into the molecular behavior of T cells. Even though both modalities have been shown to be interlinked, they are often analyzed separately, potentially missing the interdependency between function and cell state. Therefore, we developed mvTCR - a multiview Variational Autoencoder capable of fusing information of both modalities from paired single-cell data. mvTCR captures T-cell function and state better than unimodal models, while seamlessly scaling to atlas-level dataset sizes. We showcased mvTCR on a dataset containing SARS-CoV-2 patients finding separated clusters of expanded clonotypes with similar receptors and gene profiles, which were unobservable at the transcriptomic level. Based on these results, we envision that mvTCR will greatly benefit the analysis of large-scale T-cell datasets and thereby help to deepen our understanding of adaptive immunity.

H-008: A genomic region-aware CNN to identify the regulatory factors that control changes in gene expression across stimulated immunocytes
COSI: ssci
  • Alexander Sasse, University of Washington, United States
  • Sara Mostafavi, University of Washington, United States
  • Immunological Genome Consortium, Harvard Medical School, United States


Presentation Overview: Show

Cytokines of the common gamma chain (CGC) family play fundamental roles in the life cycle and the signaling of immunocytes. To dissect the CGC cytokine signaling cascade across immunocytes, we systematically measured the primary transcriptional responses of fourteen immunocytes to the six main cytokines. It is well known that CGC cytokine signaling is routed through the common g-chain receptor, as well as specific receptor subunits which, as a result, activate transcription of specific and common target genes in different cell types. Unexpectedly though, we also observed extensive transcript down-regulation for all combinations of cytokines and immocytes in our data.
To determine the underlying factors that orchestrate these contrasting responses to cytokine signaling, we trained a multi-task convolutional neural network (CNN) on sequences from different genomic regions (upstream-flanking, 5’UTR, exons, introns, 3’UTR, downstream-flanking). Interestingly, coding sequences and the 3’UTR contained the most predictive signals across all models, and adding other regions only improved the performance slightly, suggesting that changes in gene expression upon cytokine stimuli are largely regulated on the post-transcriptional level. Importance scoring of the convolutional kernels identified commonly learned kernels across different folds, initializations, and frequently also for cytokines across different cell types, consistent with their shared regulatory cascades.

H-009: Characterizing Circulating Tumor Reactive T Cells In Metastatic Melanoma via Advanced Differential Abundance Analyses
COSI: ssci
  • Wesley R. Lewis, Yale University, United States
  • Yuval Kluger, Yale University, United States
  • David A. Hafler, Yale University, United States
  • Benjamin Y. Lu, Yale University, United States
  • Harriet Kluger, Yale University, United States
  • Wei Wei, Yale University, United States
  • Liliana Lucca, Yale University, United States
  • Shuangge Ma, Yale University, United States


Presentation Overview: Show

Various candidates have been suggested for peripheral monitoring of the tumor microenvironment (TME). Examples include circulating tumor DNA and circulating tumor cells. Paired transcriptome and TCRαβ repertoire sequencing enable characterization of peripheral T cells alongside tumor infiltrating lymphocytes (TILs). A recent study in human metastatic melanoma demonstrated that effector and cytotoxic signatures from blood T cells mirror concurrently sequenced TILs, suggesting tumor-matched clonotypes as a candidate for peripheral monitoring of the TME. We hypothesize that viability of this cell subpopulation for monitoring cancers depends on consistent characterization and association with dynamics of the TME. Here, we analyze a transcriptome and TCRαβ repertoire dataset from a novel metastatic melanoma cohort. We identify differentially abundant cell populations in matched clonotypes, exposing phenotypically separable subpopulations. Next, we examine splicing profiles of tumor-matched blood T cells (TMB-Ts), uncovering upregulated and downregulated genes dynamically associated with clonal expansion of TMB-Ts. We investigate cell surface markers expressed within TMB-Ts, leveraging gene regulatory pathways within co-regulatory networks of cell surface markers. We further these aims using novel methods in differential abundance analysis. These results clarify the immunological relatedness between the tumor and systemic immune environments.

H-010: Identifying Immune Cell Gene Regulation Patterns Related to T Cell Exhaustion in Cancer via Systematic Analysis of scRNA-seq Data
COSI: ssci
  • Christopher Klocke, Oregon Health & Science University, United States
  • Amy Moran, Oregon Health & Science University, United States
  • Andrew Adey, Oregon Health & Science University, United States
  • Shannon McWeeney, Oregon Health & Science University, United States
  • Guanming Wu, Oregon Health & Science University, United States


Presentation Overview: Show

T cell exhaustion in the tumor immune microenvironment presents a challenge to the efficacy of immune checkpoint inhibitor therapies used in various late-stage cancers. These treatments are highly effective in a subset of patients but fail for others, via mechanisms that are not fully understood. Identifying gene expression and regulation patterns in immune cell subpopulations within the tumor immune microenvironment that contribute to T cell exhaustion poses an analytical challenge, even with the availability of high-resolution expression profiles from scRNA-seq data. We have constructed an analysis framework to order samples by degree of exhaustion in tumor-infiltrating CD8 T cells and identify immune cell type-specific gene regulatory network patterns associated with this trajectory. In this ongoing work, we apply this framework to scRNA-seq data from human skin tumor samples. Furthermore, we will compare the T cell exhaustion trajectory in tumors to an analogous trajectory in chronic viral infection to identify shared and distinct immune cell activity associated with T cell exhaustion under these two conditions. Considering the behavior of other immune cell types, in addition to tumor cells and the CD8 T cells themselves, will expand our current understanding of this biological process that underpins the effective treatment of late-stage cancers.

H-011: Data-driven prediction of human leukocyte antigen (HLA) epitopes using oscillations of physicochemical properties
COSI: ssci
  • Hyeju Song, Auburn University, United States
  • Chris Kieslich, Auburn University, United States


Presentation Overview: Show

Identifying T-cell epitopes is important for improving our understanding of cellular immunity and assisting in the development of peptide-based vaccines, therapeutics, and diagnostics. Human T-cell immunogenicity requires the binding of peptides derived from antigens to HLA molecules, and numerous data-driven prediction tools, have been developed for binding prediction. However, it remains challenging due to the polymorphic nature of HLA class II molecules and variations in peptide length.
The presented work tests the performance of support vector machine (SVM) models trained for allele-specific binding prediction with a comprehensive dataset downloaded from the IEDB database. The SVM models aim to classify binders and non-binders based on their peptide sequences and derived features. In developing the SVM model, we take advantage of underlying periodicities in physicochemical properties along the sequence of a peptide that have been shown to be predictive. Once the physicochemical descriptors are generated, Fourier transforms are then applied to be able to encode peptide sequences of varying lengths. In training and testing the model, cross validation and grid search are applied across multiple train and test datasets, and a feature selection algorithm is also incorporated into the model development to identify an essential set of predictive features.

H-012: Systematic Evaluation of the Predictive Gene Expression Signatures of Immune Checkpoint Inhibitors in Metastatic Melanoma
COSI: ssci
  • Samuel Coleman, Moffitt Cancer Center, United States
  • Mengyu Xie, Moffitt Cancer Center, United States
  • Aik Choon Tan, Moffitt Cancer Center, United States
  • Ahmad Tarhini, Moffitt Cancer Center, United States


Presentation Overview: Show

Advances in immunotherapy and immune checkpoint inhibitors (ICIs) have transformed the standard of care in many cancer types including melanoma. While ICIs have improved the overall outcome of a melanoma patient, a proportion develop primary or secondary tumor resistance. Therefore, a need exists to develop predictive biomarkers to better select patients for ICI therapy. Biomarkers based on genomics and transcriptomics have been investigated, however, most have not been systematically evaluated across different cohorts to determine their reproducibility in metastatic melanoma. We evaluated 28 published biomarkers of ICIs based on gene expression signatures in 8 published studies with available RNA-sequencing data. We found that gene expression-based signatures developed from IFN-γ-responsive genes and T-cell markers in the tumor immune microenvironment are generally predictive of ICIs responders in these patients. Additionally, we identified that these biomarkers have higher predictive values in on-treatment samples as compared to pre-treatment samples in metastatic melanoma. From gene set enrichment analysis, the responders were enriched with inflammation, immune activation, and infiltrated cytotoxic T-cells. Conversely, non-responders were enriched with stromal related cell types and T helper 17 cells. In summary, future biomarker development in ICIs could benefit from integrating multi-omics data to realize personalized therapeutic approach for melanoma patients.

H-013: In Silico Analysis of Antibody Sequences from a Humanized Mouse to Improve PfCSP Directed Antibody
COSI: ssci
  • Kathrin H. Kirsch, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Peter D. Kwong, Vaccine Research Center, NIH, United States
  • Facundo D. Batista, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Robert A. Seder, Vaccine Research Center, NIH, United States
  • Azza H. Idris, Vaccine Research Center, NIH, United States
  • Joe R. Francica, Vaccine Research Center, NIH, United States
  • Yongping Yang, Vaccine Research Center, NIH, United States
  • John Warner, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Michael T. Waring, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Kareen Seignon, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Mateo Reveiz, Vaccine Research Center, NIH, United States
  • Reda Rawi, Vaccine Research Center, NIH, United States
  • Marie Pancera, Vaccine Research Center, NIH, United States
  • Patience K. Kiyuka, Vaccine Research Center, NIH, United States
  • Neville K. Kisalu, Vaccine Research Center, NIH, United States
  • Chen-Hsiang Shen, Vaccine Research Center, NIH, United States
  • Barbara J. Flynn, Vaccine Research Center, NIH, United States
  • Brian Bonilla, Vaccine Research Center, NIH, United States
  • Marlon Dillon, Vaccine Research Center, NIH, United States
  • Baoshan Zhang, Vaccine Research Center, NIH, United States
  • Arne Schön, Department of Biology, Johns Hopkins University, United States
  • Eleonora Melzi, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Johan Arnold, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Gwo-Yu Chuang, Vaccine Research Center, NIH, United States
  • Usha Nair, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Lais Da Silva Pereira, Vaccine Research Center, NIH, United States
  • Kai Xu, Vaccine Research Center, NIH, United States
  • Ying-Cing Lin, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Sven Kratochvil, The Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, United States
  • Prabhanshu Tripathi, Vaccine Research Center, NIH, United States


Presentation Overview: Show

Malaria, mainly caused by Plasmodium falciparum (Pf) species, is a major public health burden with over 400,000 deaths in 2019. The Pf circumsporozoite protein (PfCSP) is the most prevalent sporozoite
antigen, contains a multiply repeated ‘NANP’-tetrapeptide, and uses both sequence degeneracy and structural diversity to evade the immune response. Infusion of PfCSP-directed antibody, CIS43, is effective at preventing malaria infection for up to 9 months. Improved CIS43 are eagerly sought, but the molecular basis mechanism to improve the repeat-targeting antibodies has been unclear. Here, a humanized mouse model where B cells expressed inferred germline CIS43 (iGL-CIS43) antibody was created and used to immunize and to gain informatic insights leading to antibody variants with improved protective efficacy. Based upon informatic sieving of over a hundred mouse derived variants, variants were designed that incorporated somatic hypermutations from the best mouse derived variants. One such improved antibody, iGL-CIS43.D13, exhibited ~7-fold improvement over CIS43 and appears to be the current best in class antibody. Structural and in Silico analysis of antibodies elicited by junctional peptide immunization revealed specific details of improved protective efficacy. Thus, informatics combined antibodies from this mouse model can design highly potent anti-malarial antibodies with improved therapeutic potential.

H-014: Defining the Epigenetic Landscape of Dynamic Pathogenic Transcriptional Cell States in Rheumatoid Arthritis
COSI: ssci
  • Kathryn Weinand, Harvard University, United States
  • Saori Sakaue, Brigham and Women's Hospital, United States
  • Helena Jonsson, Brigham and Women's Hospital, United States
  • Gerald Watts, Brigham and Women's Hospital, United States
  • Aparna Nathan, Harvard University, United States
  • Fan Zhang, University of Colorado, United States
  • Amp Ra Sle Consortium, AMP RA/SLE Consortium, United States
  • Jennifer H. Anolik, University of Rochester School of Medicine and Dentistry, United States
  • Laura T. Donlin, Hospital for Special Surgery, United States
  • Kevin Wei, Brigham and Women's Hospital, United States
  • Michael B. Brenner, Brigham and Women's Hospital, United States
  • Soumya Raychaudhuri, Brigham and Women's Hospital, United States


Presentation Overview: Show

Rheumatoid Arthritis (RA) is an autoimmune disease causing synovial tissue inflammation. Recent work by the AMP-RA consortium (Zhang et al BioRxiv) has defined 77 RA tissue fine-grain cell states using transcriptional and proteomic data. Many states have been shown to play a pathogenic role in mediating inflammation with implications for treatment response and prognosis. The epigenetic relationship between and regulation of these states remains undefined. We used 12 single nucleus ATAC/RNA multiome and 18 scATAC-seq datasets from disaggregated RA/OA patient synovial tissue to characterize their epigenetic cell states. Our study suggested that broad cell types are generally concordant between modalities, but fine-grain transcriptional cell states are hard to distinguish at the epigenetic level. For example, while 9 fibroblast cell states have been observed transcriptionally, only 4 epigenetically defined cell states were seen. We defined combinations of TFs associated with these broader cell states that may explain the regulation underlying the discrepancy between modalities. Within the AMP-RA known transcriptional cell states, we see instances of both 1-1 and 1-many relationships with epigenetic cell states. These relationships can help explain variable RA inflammation between patients and may suggest treatment plans that target either cell states or exogenous factors (e.g., TFs, cytokines).

H-015: Automated Gating and Antigen-Specific T cell Subset Discovery using FAUST and COMPASS on ICS data
COSI: ssci
  • Malisa Smith, Ozette Technologies, United States
  • Evan Greene, Ozette Technologies, United States


Presentation Overview: Show

Intracellular Cytokine Staining (ICS) is used to assess the nature and magnitude of the immune system’s response to antigens. Most ICS experiments are still analyzed manually.
We used Full Annotation Using Shape-constrained Trees (FAUST) (https://doi.org/10.1016/j.patter.2021.100372) to perform automated gating of protein markers in an ICS study of human T cell response to Mycobacterium tuberculosis antigens. This approach accounted for batch effects in this study by identifying antigen-specific cell subsets on a per-sample basis and also used information from the different stimulation conditions to obtain appropriate cytokine gates. We then used FAUST’s cell subset discovery feature to identify candidate antigen-specific combinations of markers which appear consistently across samples in the experiment. As a parallel approach, we ran Combinatorial Polyfunctionality analysis of Antigen-Specific T-cell Subsets (COMPASS) (https://doi.org/10.1038/nbt.3187) on the FAUST-gated results.
We compare our findings to those of the original publication which used manual gating, and we show that we are able to validate and extend the published findings. Thus, we provide an example of using FAUST as an automated analytical tool for identifying meaningful cell populations in ICS data.