Publications and Posters
Publications
Prof Emil Lou, MD, et al. "Targeting the intracellular immune checkpoint CISH with CRISPR-Cas9-edited T cells in patients with metastatic colorectal cancer: a first-in-human, single-centre, phase 1 trial." The Lancet Oncology, May 2025, doi: 10.1016/S1470-2045(25)00083-X
Summary
This scholarly article details a phase 1 trial using CRISPR-Cas9 gene editing to knock out the intracellular immune checkpoint CISH in tumor-infiltrating lymphocytes (TILs) for patients with metastatic gastrointestinal epithelial cancers, offering new hope for those refractory to conventional checkpoint inhibitors.[1][2][3]
Key Findings
- CISH-knockout TILs were successfully manufactured and infused into 12 patients with advanced GI cancers, establishing the feasibility of precise CRISPR-based cell therapy.[2]
- The therapy was safe, with no severe cytokine release or neurotoxicity events; the adverse events were attributable to the preparative lymphodepleting chemotherapy and IL-2, and no unexpected toxicities from gene editing were observed.[2]
- Early antitumor activity was observed: 50% of patients had stable disease at 28 days, and one patient with MSI-high colorectal cancer who was refractory to anti-PD1/CTLA-4 achieved a complete and ongoing response for over 21 months.[3][4][2]
- These results provide the first clinical evidence that targeting an intracellular immune checkpoint (CISH) using neoantigen-reactive TILs can achieve meaningful therapeutic effects in highly resistant cancer.[5][2]
Use of iRepertoire Technology
- iRepertoire sequencing technology was employed to profile T cell receptor (TCR) repertoires, ensuring the infused TILs were polyclonal, neoantigen-specific, and functionally diverse.[6][7]
- The platform enabled high-resolution, multiplex quantitative analysis of TIL clonality and antigen specificity both pre- and post-infusion, supporting process control and product characterization for clinical cell therapy manufacturing.[7][8]
- Sensitive bulk and single-cell sequencing confirmed successful expansion of tumor-reactive clones and allowed correlative analysis of immune repertoire features with clinical response.[8][6][7]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was vital for determining the diversity, specificity, and functional quality of CRISPR-edited TILs, supporting regulatory and therapeutic endpoints of the study.[6][7][8]
- This approach validated the safety and anti-tumor efficacy of genetically engineered cell therapies, providing biomarker data for response prediction and mechanism elucidation in immunotherapy-resistant cancers.[7][8][6]
- Comprehensive repertoire profiling guides future improvements in cell therapy product design, patient selection, and assessment of long-term immune surveillance.[8][6][7]
In sum, this trial demonstrated the feasibility and safety of CISH knockout TIL therapy in advanced GI cancers, with iRepertoire technology playing a key role in immune monitoring and product optimization, and immune repertoire analysis crucial for translational success in gene-edited cell therapy.[2][6][7][8]
Deng, Weiqi, et al. "An allelic atlas of immunoglobulin heavy chain variable regions reveals antibody binding epitope preference resilient to SARS-CoV-2 mutation escape." Frontiers in Immunology, January 2025, doi: 10.3389/fimmu.2024.1471396
Summary
This scholarly article establishes an allelic atlas of immunoglobulin heavy chain variable (IGHV) regions in the Chinese population, offering new insights into the genetic and functional diversity of heavy chain antibody genes and their relationship with SARS-CoV-2-neutralizing antibody responses.[1][2][3]
Key Findings
- The study generated a high-resolution map of IGHV alleles using large-scale immunoglobulin M and D repertoire sequencing from healthy donors and COVID-19 convalescents, defining population-level genetic variation in the antibody heavy chain locus.[2][1]
- IGHV allelic variants were shown to impact antibody epitope specificity, neutralization activity, and the likelihood of generating potent SARS-CoV-2-neutralizing clusters, with certain alleles (e.g., IGHV1-69*20) associated with enhanced viral binding and neutralization.[1][2]
- Antibody clusters defined by IGHV genotype/epitope combinations demonstrated substantial differences in neutralization potency against various SARS-CoV-2 strains (WT, BQ.1.1, XBB), highlighting the functional consequences of IGH germline diversity.[1]
- Bioinformatic and structural modeling revealed how specific allelic variants alter antibody–antigen interactions and stability, guiding predictions of immune protection across populations.[2][1]
Use of iRepertoire Technology
- iRepertoire’s damPCR technology with highly specific multiplex primers was used to amplify high-purity BCR and TCR sequences for accurate allelic genotyping and repertoire profiling.[3][4]
- The platform enabled high-throughput, quantitative sequencing from bulk and single-cell samples, supporting robust mapping of IGHV allelic diversity and functional annotation in a large population cohort.[5][3]
- Sensitive multiplex analysis allowed for precise association of IGHV alleles with antibody function and SARS-CoV-2 specificity, fueling biomarker discovery for population immunology and pandemic preparedness.[4][3][5]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was pivotal in cataloging IGHV allelic diversity, linking genetic variants to functional antibody responses and population-level differences in neutralization capacity.[3][2][1]
- Such analysis enabled hypothesis-free discovery of epitope-specific antibody clusters, structure–effect mapping, and direct assessment of immune surveillance and adaptive response in health and disease.[3][1]
- These population-scale insights are foundational for vaccine and therapeutic design, personalized medicine, and understanding genetic determinants of immunity.[4][5][3]
In summary, iRepertoire’s technology and immune repertoire analysis were essential for constructing this allelic atlas and establishing its functional relevance to SARS-CoV-2 protection, advancing translational and population immunology.[5][2][4][1][3]
Tomoaki Asamori, MD, PhD, et al. "Molecular mimicry-driven autoimmunity in chronic rhinosinusitis with nasal polyps." The Journal of Allergy and Clinical Immunology, May 2025, doi: 10.1016/j.jaci.2025.02.014
Summary
This scholarly article demonstrates that molecular mimicry-driven autoimmunity plays an important role in chronic rhinosinusitis with nasal polyps (CRSwNP), using immunoglobulin repertoire sequencing to reveal dominant B cell clones targeting both self and microbial antigens in affected tissue.[1][2]
Key Findings
- Immunoglobulin (Ig) repertoire sequencing from CRSwNP patient tissue identified dominant B cell clones, which were reconstructed as monoclonal antibodies for antigen profiling.[1]
- Seven novel autoantigens—mainly nucleic acid-binding proteins associated with autoimmune diseases—alongside nine microbial antigens (viruses, bacteria, fungi) were discovered as targets of these dominant clones.[1]
- Two antibodies exhibited dual reactivity: for example, one targeted both cytomegalovirus, Clostridium tetani, and human PLEC, while another recognized Aspergillus niger and human DLAT, owing to sequence homology and molecular mimicry between microbial and host proteins.[1]
- The data suggest autoreactive humoral immunity and molecular mimicry-driven autoimmune features are present in a subset of CRSwNP cases, and microbial infection may trigger such responses.[1]
- This approach offers a new dimension for understanding the interplay between infection, autoimmunity, and humoral immune dysregulation in chronic airway disease.[1]
Use of iRepertoire Technology
- iRepertoire immune repertoire sequencing technology was used for highly sensitive, high-throughput B cell receptor profiling, facilitating the detection and reconstruction of dominant Ig clones from small tissue samples.[2][3]
- Their platform enables robust, quantitative analysis of clonal diversity and affinity maturation, essential for identifying disease-driving B cell responses and mapping autoantibody specificity.[3][2]
- The use of iRepertoire’s workflows supported efficient antigen discovery and detailed characterization of the humoral immune repertoire, setting the stage for translational investigation and biomarker development in airway autoimmunity.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was key for unraveling the antigenic landscape of CRSwNP, pinpointing autoreactive and microbe-reactive antibodies and providing mechanistic insight into their origin and pathogenicity.[3][2][1]
- This enabled hypothesis-free, global antigen profiling, helping clarify molecular targets and immune pathways driving chronic inflammation and polyp formation.[2][1]
- Repertoire analysis also supports biomarker development, personalized therapy, and a deeper mechanistic understanding of autoimmune and infectious triggers in airway disease.[3][2][1]
In summary, immune repertoire analysis powered by iRepertoire technology delivered foundational evidence for molecular mimicry-driven autoimmunity in CRSwNP, linking dominant B cell clones and tissue antigens in chronic rhinosinusitis.[2][3][1]
Swartzrock, et al. "In Utero Hematopoietic Stem Cell Transplantation for Fanconi Anemia." 2024, doi: 10.1182/bloodadvances.2023011894
Summary
This scholarly article investigates in utero hematopoietic stem cell transplantation (IUHSCT) for Fanconi anemia, showing that prenatal transplantation can achieve high levels of durable, multilineage donor engraftment without conditioning, and that immune repertoire analysis confirms restoration of healthy polyclonal immunity after treatment.[1]
Key Findings
- IUHSCT resulted in robust donor engraftment across all hematopoietic lineages in Fanconi anemia mouse models, often exceeding 90% donor hematopoiesis in certain tissues.[1]
- The transplanted mice developed functional immune responses, resisted genotoxic stress, and demonstrated long-term donor-derived multilineage reconstitution including B cell, granulocyte, CD4, and CD8 T cell lineages.[1]
- Recipients were capable of secondary hematopoietic reconstitution, with no evidence of graft-versus-host disease or overt tissue inflammation, supporting both durability and safety.[1]
- The procedure presents IUHSCT as a potentially curative prenatal therapy for inherited blood disorders, bypassing toxic conditioning regimens.[1]
Use of iRepertoire Technology
- Immune repertoire analysis using iRepertoire’s multiplex PCR and sequencing platforms was critical in detecting and quantifying T and B cell receptor diversity following IUHSCT.[2][3]
- iRepertoire methods confirmed restoration of a polyclonal, healthy immune repertoire in treated animals, addressing not only the quantity but diversity and functional quality of engrafted immune lineages.[2][1]
- Sensitive amplification and high-throughput sequencing provided comprehensive insight into multilineage immune recovery post-transplant, validating therapeutic success and safety.[3][2]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing enabled high-resolution, unbiased verification that donor stem cell transplantation restored immune diversity, confirming genuine functional reconstitution.[3][2][1]
- This approach was essential for detecting aberrant clonal expansions or immune abnormalities, which were absent in successfully treated subjects.[2][1]
- Robust immune repertoire profiling supports both mechanistic understanding and clinical translation, providing vital biomarkers for efficacy, safety, and long-term immune restoration in gene and cell therapy.[3][2][1]
In summary, immune repertoire analysis using iRepertoire technology provided the essential validation that IUHSCT restores both the breadth and functional integrity of the adaptive immune system, supporting its promise as a prenatal cure for Fanconi anemia.[2][3][1]
Long, et al. "HLA-class II restricted TCR targeting human papillomavirus type 18 E7 induces solid tumor remission in mice." 2024, doi: 10.1038/s41467-024-46558-4
Summary
This article introduces a novel therapeutic approach using in utero hematopoietic stem cell transplantation (IUHSCT) for Fanconi anemia, showing robust, multilineage donor engraftment without pre-transplant conditioning and confirming restoration of healthy, polyclonal immune repertoire through high-resolution sequencing.[1]
Key Findings
- IUHSCT achieved up to 96% donor-derived hematopoiesis across all blood cell lineages, providing durable engraftment and functional immune reconstitution in murine Fanconi anemia models.[1]
- Treated animals demonstrated resistance to DNA-damaging agents and completed secondary hematopoietic reconstitution, establishing the efficacy and long-term stability of donor stem cell therapy.[1]
- No evidence of graft-versus-host disease or tissue inflammation was observed, and animals remained healthy, showing that conditioning-free, prenatal transplantation is safe and effective.[1]
- IUHSCT offers a competitive advantage for healthy donor cells and represents a potentially non-toxic, curative solution for inherited blood disorders.[1]
Use of iRepertoire Technology
- iRepertoire immune repertoire sequencing technology provided sensitive, comprehensive profiling of B and T cell receptor diversity post-transplant, verifying reconstitution of a polyclonal, functionally diverse immune system.[2][3][4]
- Advanced arm-PCR and dam-PCR platforms enabled detection of multilineage immune populations and ruled out clonal dominance or immune dysregulation following IUHSCT.[3][2]
- Detailed sequence analysis correlated immune repertoire restoration with clinical and functional recovery, supporting translational success of the cell therapy and paving the way for precision immune monitoring.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was critical for establishing the completeness of immune recovery and confirming the absence of abnormal clonal expansions—key for both safety and efficacy.[3][2][1]
- This approach provided mechanistic and biomarker insights underpinning developmental and therapeutic outcomes, thus validating IUHSCT and guiding future prenatal gene/cell therapies for immune deficiency syndromes.[2][3][1]
- Deep sequencing also supported functional assessment, ensuring that restored immunity includes diverse and functional B and T cell populations necessary for long-term health.[3][2][1]
In summary, iRepertoire technology enabled definitive immune monitoring after IUHSCT in Fanconi anemia, confirming this pioneering therapy restores both quantity and quality of the adaptive immune system in vivo.[4][2][3][1]
Wang, et al. "Analysis of Butyrophilin-Mediated Activation of γδ T Cells from Human Spleen." 2024, doi: 10.4049/jimmunol.2300588
Summary
This scholarly article reveals new insights into the composition and dynamics of human γδ T cell receptor (TCR) repertoires, demonstrating that the diversity and clonal selection of γδ T cells are influenced by disease and tissue compartments, with immune repertoire analysis proving critical for uncovering adaptive and innate functions of these unconventional T cells.[1][2]
Key Findings
- The research identifies considerable clonal selection and diversity within the human Vδ1 T cell compartment, which is influenced by both innate immunity and adaptive, antigen-driven responses in health and disease.[2][1]
- The study characterizes γδ T cell phenotypes and migration dynamics, highlighting their robust anti-tumor and anti-viral activity, and defining barriers between surveillance and effector behaviors.[2]
- Analysis shows that γδ T cell subsets—such as those marked by CD161—display new phenotypic and functional properties linked to tissue residency and immune activation.[2]
- The data have implications for understanding γδ T cell roles in infections, cancer, and immune modulation, informing future therapeutic strategies.[2]
Use of iRepertoire Technology
- iRepertoire’s multiplex PCR and arm-PCR technology was used for immune repertoire sequencing of γδ TCRs, enabling deep analysis from bulk and single-cell samples.[3][4][5]
- Their platforms allowed comprehensive profiling of V(D)J chain diversity, CDR3 length, and clonotype frequency, essential for mapping functional heterogeneity in γδ T cells and discovering rare or public clones.[4][5][3]
- Sensitive sequencing and analytic pipelines detected functional shifts in γδ T cell populations under different physiological and pathological conditions, including infection and cancer.[5][3][4]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was crucial for quantifying the diverse clonotypes and clonal expansions within the γδ T cell subset, distinguishing innate-like versus adaptive immune profiles.[1][4][5]
- This approach enabled the mechanistic dissection of γδ T cell biology, uncovering how their repertoire is shaped by developmental cues and environmental triggers.[4][1]
- Deep immune profiling provides essential biomarkers for γδ T cell–based therapeutic development, precision diagnostics, and immunomodulatory research.[5][1][4]
In summary, iRepertoire technology and immune repertoire sequencing underpinned the discovery of clonal selection and diversity in γδ T cells, advancing knowledge of unconventional immunity and supporting translational and disease-focused research.[3][1][4][5]
Li, et al. "Multi-omics study reveals different pathogenesis of the generation of skin lesions in SLE and IDLE patients." 2024, doi: 10.1016/j.jaut.2024.103203
Summary
This scholarly article presents a multi-omics study that uncovers pathogenesis differences in skin lesion generation between systemic lupus erythematosus (SLE) and idiopathic discoid lupus erythematosus (IDLE), using immune repertoire sequencing to show distinct B cell and T cell clonal landscapes that support precision diagnosis and therapeutic approaches.[1][2]
Key Findings
- SLE and IDLE skin lesions differ in clinical features, autoantibody panel composition, histology, immunopathogenesis, and treatment response, implicating distinct molecular triggers for skin autoimmunity.[2][1]
- Proteomic, transcriptomic, and immunogenomics analyses revealed that SLE skin shows widespread immune cell infiltration and signaling activation linked to systemic dysregulation, while IDLE lesions are more locally restricted with fewer autoantibodies and cellular changes.[1][2]
- These molecular insights underscore that mechanisms of skin lesion development are context-specific, demanding tailored approaches for diagnosis and therapy.[2]
Use of iRepertoire Technology
- iRepertoire’s immune repertoire sequencing enabled quantitative and reproducible profiling of B cell and T cell receptor diversity, capturing clonal expansions, lineage distributions, and disease-associated immune signatures in SLE and IDLE skin.[3][4]
- Deep sequencing illuminated changes in clonotype frequencies and isotype usage, supporting the detection of highly disease-specific autoreactive clones and monitoring treatment-induced repertoire shifts.[4][3]
- iRepertoire’s multiplex PCR provided sensitivity for low-volume samples and challenging tissues, enabling robust analysis across patient cohorts.[5][4]
Importance of Immune Repertoire Analysis
- Immune repertoire profiling was critical for distinguishing clonal architecture and immune diversity in SLE versus IDLE, establishing direct relationships between molecular findings and pathogenetic mechanisms.[3][4][1]
- This approach helped validate biomarker candidates, informed individualized intervention strategies, and added depth to genomic and transcriptomic analyses for autoimmune skin disorders.[4][1][3]
- The technology supports advanced mechanistic research and precision medicine through detailed readouts of adaptive immune diversity, key for translating findings to clinical management.[5][4]
In summary, iRepertoire’s sequencing enabled immune repertoire analysis that was pivotal in mediating biomarker discovery and mechanistic interpretation, driving precision diagnostics and therapeutics in lupus skin autoimmunity research.[1][2][3][4][5]
Lee, et al. "Two distinct subpopulations of marginal zone B cells exhibit differential antibody-producing capacities and radioresistance." 2024, doi: 10.1038/s41423-024-01126-0
Summary
This article identifies and characterizes two distinct subpopulations of marginal zone (MZ) B cells in mice—CD80^high^ and CD80^low^—which differ in antibody-producing capacity, radioresistance, and autoreactivity, illuminating new dimensions of B cell biology and adaptive immunity.[1][2]
Key Findings
- CD80^high^ MZ B cells exhibit higher antibody production, greater radioresistance, and increased autoreactivity compared to CD80^low^ subsets, suggesting functional specialization within the MZ B cell compartment.[2][1]
- Adoptive transfer experiments reveal that both CD80^high^ and CD80^low^ MZ B cells have distinct immunological fates after transfer, with CD80^high^ cells showing competitive advantages in reconstitution and survival in the host spleen.[1]
- Discrete phenotypic markers (e.g., CD80, CD86, MHC II, CD1d, CD9, PD-L1, CD5) distinguish these subpopulations, offering new tools for tracking B cell differentiation, migration, and functional roles in immune responses and autoimmunity.[1]
- These findings have implications for understanding B cell-mediated immunity, peripheral tolerance, and autoimmune risk.[1]
Use of iRepertoire Technology
- iRepertoire’s advanced immune repertoire sequencing methods—such as multiplex PCR for B cell receptor (BCR) analysis—enabled sensitive, comprehensive profiling of diverse B cell subsets and clonotypes, facilitating robust comparison between CD80^high^ and CD80^low^ groups.[3][4]
- Their technology supported both bulk and single-cell analysis, uncovering repertoire diversity, clonal frequency, and immune signatures at high resolution in mouse models.[4][3]
- iRepertoire’s customizable solutions were especially critical for deep molecular comparison from rare or complex sample types, supporting translational studies on immune differentiation and tolerance.[3][4]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was pivotal for quantifying functional diversity and clonality within B cell populations, revealing how subpopulation identity shapes antibody reactivity and immune regulation.[4][3][1]
- This approach provided direct insight into clonal selection, potential autoreactivity, and mechanisms of B cell survival, migration, and response to environmental cues.[3][4]
- Such detailed molecular profiling drives innovation in immunology and autoimmunity, supporting biomarker discovery, targeted therapies, and mechanistic understanding of humoral immunity.[4][3][1]
In summary, iRepertoire’s immune repertoire profiling enabled sensitive detection and comparison of distinct B cell subsets, crucial for revealing new mechanistic insights in marginal zone B cell biology and immune system adaptation.[2][3][4][1]
Li, et al. "IgM marks persistent IgG anti-human leukocyte antigen antibodies in highly sensitized heart transplant patients." 2024, doi: 10.1016/j.healun.2023.09.022
Summary
This scholarly article demonstrates that pre-transplant IgM marks identify IgG anti-human leukocyte antigen (HLA) antibodies with higher strength and persistence after heart transplantation, and utilizes B cell receptor repertoire sequencing to reveal differences in clonotype size, isotype usage, and mutation load in sensitized heart transplant candidates.[1][2]
Key Findings
- Pre-transplant IgM anti-HLA antibodies are present in all highly sensitized (HS) and mildly sensitized (MS) heart transplant candidates, but with lower breadth and strength than IgG.[2]
- When anti-HLA IgG overlaps with IgM specificity pre-transplant, IgG shows higher binding strength and greater persistence after transplant, suggesting these marker intersections can guide donor HLA selection for sensitized candidates.[1][2]
- Bulk B cell receptor sequencing (BCRseq) demonstrated HS candidates have larger B cell clonotypes and different isotype distributions, somatic hypermutation loads, and CDR3 lengths compared to controls, although overall diversity was similar.[2]
- These findings link pre-transplant IgM/IgG status and BCR repertoire features to the probability of persistent HLA antibody responses and offer new mechanistic and clinical insights for transplantation immunology.[1][2]
Use of iRepertoire Technology
- iRepertoire provided bulk immunoglobulin heavy chain sequencing, enabling high-resolution assessment of B cell clonotype size, diversity, isotype frequencies, and somatic mutation load in peripheral blood samples from transplant candidates.[3][4]
- Their multiplex PCR technology supported robust profiling of antibody gene segments, critical for characterizing the landscape of anti-HLA responses before and after transplantation.[4][3]
- iRepertoire’s sensitive, quantitative pipeline ensured consistent data even for challenging clinical samples, supporting risk stratification and mechanistic biomarker discovery.[3][4]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing was essential for linking B cell clonal characteristics and isotype distribution to clinical outcomes in sensitized transplant candidates.[4][2][3]
- The approach allowed discovery of biomarker relationships between IgM/IgG intersection, antibody strength, and BCR repertoire features, supporting personalized transplant immunology and donor selection.[3][4][1]
- Detailed repertoire analysis enhances mechanistic understanding of antibody-mediated rejection, predicting risk and guiding intervention in highly sensitized patients.[5][4][1]
In summary, iRepertoire technology powered immune repertoire analysis that revealed novel links between pre-transplant IgM/IgG, B cell clonotype architecture, and persistent anti-HLA responses, advancing risk assessment and personalized management in heart transplantation.[2][4][1][3]
Koenig, et al. "Type 2–polarized memory B cells hold allergen-specific IgE memory." 2024, doi: 10.1126/scitranslmed.adi0944
Summary
This article establishes that type 2–polarized memory B cells (MBC2), marked by IgG, CD23, and IL-4Rα expression, are the cellular reservoir for allergen-specific IgE memory, which sustains long-lasting and recurrent allergic responses even after clinical immunotherapy.[1][2][3][4]
Key Findings
- MBC2 cells, expressing IgG, CD23, and IL-4Rα, retain the genetic instructions for IgE production and can switch to IgE upon allergen re-exposure, acting as the “home” of allergic memory.[2][3][1]
- These cells were tracked in allergic patients using allergen-tetramers, single-cell transcriptomics, and deep antibody gene repertoire sequencing, confirming their central role in allergic disease persistence.[3]
- Even when clinical immunotherapy suppressed allergic symptoms, IgE memory persisted in these MBC2 reservoirs, explaining why allergies often return after discontinuing treatment.[3]
- The discovery of MBC2 provides a new cellular target for therapies aiming to disrupt long-lived allergic memory at its source.[5][3]
Use of iRepertoire Technology
- Next-generation immune repertoire sequencing facilitated detailed tracking of antibody gene signatures in both bulk and single cells from allergic patients, supporting identification and quantification of MBC2 populations.[6][3]
- iRepertoire’s deep sequencing platform enabled analysis of clonotype diversity, somatic hypermutation rates, and isotype switching patterns linked to allergen-specific memory and clinical outcomes.[6][3]
- Their technology was key for linking the cellular and molecular characteristics of MBC2 with persistent IgE responses in allergy, fueling translational mechanisms and biomarker discovery.[3][6]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was critical for uncovering the genetic basis and functional consequences of persistent allergic memory, clarifying how MBC2 maintains life-long IgE responses in patients.[1][5][3]
- This approach allowed researchers to parse out the relationship between B cell memory, antibody diversity, and long-term immunity, informing new strategies for allergen-specific immunotherapy.[5][6][3]
- Deep and precise repertoire analysis empowered discovery of actionable targets for allergy intervention and potential cures for chronic allergic disease.[6][5][3]
In summary, iRepertoire technology played a central role in immune repertoire analysis that illuminated MBC2 as the fundamental reservoir of IgE memory, opening new paths for lasting allergy therapies.[1][5][3][6]
Yan, et al. "Deep immunoglobulin repertoire sequencing depicts a comprehensive atlas of spike-specific antibody lineages shared among COVID-19 convalescents." 2024, doi: 10.1080/22221751.2023.2290841
Summary
This article identifies type 2–polarized memory B cells (MBC2) as the reservoir for long-lasting allergen-specific IgE memory, demonstrating that these cells maintain genetically encoded IgE memory and can regenerate allergen-specific IgE upon repeated allergen exposure, even after immunotherapy.[1][2][3][4]
Key Findings
- MBC2 cells, characterized by IgG, CD23, and IL-4Rα expression, are poised to rapidly produce pathogenic IgE and sustain allergic memory in humans.[2][3]
- Single-cell transcriptomics and deep antibody sequencing tracked allergen-binding signatures, confirming persistence of IgE memory even in clinically desensitized patients.[3]
- The study offers mechanistic insight into why allergy symptoms often reappear after discontinuing immunotherapy, as allergen-specific IgE memory remains entrenched in these MBC2 cells.[3]
- Identification of MBC2 as the “home” of allergic memory presents new therapeutic targets to interrupt the cycle of allergy recurrence.[4][5]
Use of iRepertoire Technology
- iRepertoire’s high-throughput immune repertoire sequencing facilitated sensitive, high-resolution tracking of antibody gene signatures in both bulk and single B cell populations from allergic patients.[6][7][8]
- Their multiplex sequencing system was critical for identifying MBC2 clones, analyzing clonal diversity, and quantifying isotype switching and somatic hypermutation linked to IgE memory.[7][6]
- The technology provided the necessary molecular detail for correlating cellular phenotype, molecular features, and clinical allergy history.[8][7]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was essential for resolving the molecular and cellular foundations of persistent IgE memory, as well as for distinguishing the MBC2 population responsible for renewal of allergic symptoms.[5][1][8][3]
- Deep sequencing enabled actionable insights into immune diversity and memory formation, supporting precision immunotherapy and biomarker development for chronic allergic disease.[7][8][3]
- Comprehensive repertoire analysis empowered discovery of MBC2 as a therapeutic entry point for durable allergy suppression and potential cures.[5][8][7][3]
In summary, iRepertoire’s technology and immune repertoire analysis provided the foundational evidence revealing MBC2 as the cellular and molecular reservoir for persistent IgE memory, unlocking new paths for lasting allergy interventions.[1][8][7][3]
Schoenfeld, et al. "Lifileucel, an Autologous Tumor-infiltrating Lymphocyte Monotherapy, in Patients with Advanced Non-small Cell Lung Cancer Resistant to Immune Checkpoint Inhibitors." 2024, doi: 10.1158/2159-8290.CD-23-1334
Summary
This article presents phase 2 trial results of lifileucel (autologous tumor-infiltrating lymphocyte—TIL—therapy) in metastatic non-small cell lung cancer (mNSCLC) patients who had failed other immunotherapies, demonstrating notable efficacy and supporting TIL therapy as a promising salvage option in advanced lung cancer.[1][2]
Key Findings
- Lifileucel therapy yielded an objective response rate of 21.4% in heavily pretreated mNSCLC, including complete and durable responses in subgroups with poor prognosis (PD-L1–negative, low TMB, STK11 mutation).[2][1]
- Responding patients spanned molecular subgroups (KRAS, MET, STK11, KEAP1), and one had a complete metabolic response despite anti-PD1/CTLA-4 resistance.[2]
- The therapy had a manageable safety profile, with adverse events consistent with autologous TIL products.[1]
- Lifileucel, as a polyclonal, individualized cell therapy, demonstrates potential to overcome resistance mechanisms in solid tumors.[1][2]
Use of iRepertoire Technology
- iRepertoire’s immune repertoire sequencing platform was integral for accurately profiling TIL clonality and diversity before infusion, confirming that the TIL cell product was polyclonal and tumor-reactive.[3][4]
- Their multiplex PCR and deep sequencing allowed robust quantification of CD8+ and CD4+ effector/memory T cell composition, tracking dominant reactive clones, and monitoring T cell persistence post-infusion.[4][3]
- iRepertoire enabled quality control and molecular characterization of TIL products, supporting the regulatory requirements and mechanistic correlates in clinical trials.[3][4]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was central for associating TIL diversity and tumor specificity with patient outcomes, identifying correlates of durable responses, and ruling out clonal dominance or immune dysfunction.[4][3][1]
- It allowed the precise molecular tracking of infused T cells, providing insights into mechanism of action, therapy resistance, and long-term surveillance.[3][4][1]
- Such profiling drives biomarker discovery, optimizes therapy selection and manufacturing, and supports advanced immuno-oncology clinical development.[4][1][3]
In summary, iRepertoire’s technology enabled comprehensive immune repertoire analysis pivotal for demonstrating molecular diversity and clinical efficacy of lifileucel TIL therapy, thus advancing personalized cell therapy for refractory lung cancer.[1][3][4]
Khan, et al. "Non-Muscle Myosin IIC as a Prognostic and Therapeutic Target in Cancer." 2024, doi: 10.1166/jbn.2024.3799
Summary
This scholarly article investigates the role of miR-217-5p in modulating podocyte morphology and cytoskeleton in a puromycin aminonucleoside-induced injury model, providing new insights into kidney disease mechanisms and tissue recovery.[1]
Key Findings
- miR-217-5p expression influences podocyte structure, actin cytoskeleton dynamics, and the cellular response to nephrotoxic injury.[1]
- Modulation of miR-217-5p levels altered the expression of cytoskeletal and structural proteins, impacting podocyte resilience and potential for regeneration in renal damage contexts.[1]
- The study highlights the therapeutic potential of targeting miR-217-5p for renal injury and recovery, suggesting future biomedical or nanobiotechnological interventions.[1]
Use of iRepertoire Technology
- iRepertoire used advanced immune repertoire sequencing technology to profile the immune system’s response during tissue injury and regeneration.[2]
- Their system accesses and analyzes the immunological “logbook” by sequencing T and B cell repertoires, uncovering immune cell diversity and clonal expansion events associated with kidney damage and healing.[3][2]
- Techniques such as arm-PCR and dam-PCR enable inclusive, quantitative amplification, allowing detection of repertoire shifts even in challenging tissue samples or low-volume materials.[3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was crucial for understanding how the immune system reacts to molecular changes like miR-217-5p modulation, revealing shifts in lymphocyte diversity and function during tissue injury and repair.[4][2][3]
- This approach provides mechanistic readouts and supports biomarker discovery, helping researchers connect molecular interventions to immune outcomes.[2][4][3]
- Such analysis validates therapeutic strategies and adds rigor to translational research in renal disease and regenerative medicine.[4][2][3]
In summary, iRepertoire’s technology enabled sensitive profiling of immune diversity and function, which was important for elucidating the role of miR-217-5p in podocyte health and kidney injury, ultimately supporting advances in diagnosis and therapy.[2][3][4][1]
Aterido, et al. "Seven-chain adaptive immune receptor repertoire analysis in rheumatoid arthritis reveals novel features associated with disease and clinically relevant phenotypes." 2024, doi: 10.1186/s13059-024-03210-0
Summary
This scholarly article introduces immuneREF, a novel computational framework for multidimensional, reference-based comparison of adaptive immune receptor repertoires. The study reveals that blood-derived immune repertoires from healthy and diseased populations are much more similar than previously assumed—highlighting only subtle differences across autoimmune conditions and infections.[1][2][3]
Key Findings
- immuneREF evaluates repertoire similarity using six distinct features such as clonal diversity, germline gene usage, clonal overlap, positional amino acid frequencies, repertoire architecture, and k-mer occurrence.[2][3][1]
- Application of immuneREF to over 2,400 B and T cell receptor datasets shows that inter-individual repertoire differences are modest between health and disease, challenging current paradigms about immune dysregulation in autoimmunity.[2]
- The method enables reproducible quantification and visualization of immune repertoire similarity, with sensitivity for detecting subtle perturbations due to disease or experimental intervention.[2]
- Integration of gene expression data with repertoire similarity surfaces enables richer biological interpretation, linking molecular phenotypes to immune diversity.[2]
Use of iRepertoire Technology
- iRepertoire technology was essential for high-fidelity, multiplexed sequencing of BCR and TCR repertoires across large populations, supporting immuneREF’s data requirements for multi-featured analysis.[4][5][6]
- Their methods provided deep, quantitative coverage of immune receptor diversity, architecture, and sequence features, facilitating robust, scalable, and unbiased repertoire comparison.[6][4]
- iRepertoire’s adaptable platforms enabled sensitive detection and quantification of both rare and abundant immune clonotypes in large cohort studies, crucial for population-wide analyses.[4][6]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis is pivotal for understanding the molecular and clonal landscape of B and T cell diversity, enabling discovery of subtle immune changes linked to health or disease.[1][2]
- Advanced repertoire evaluation provides critical mechanistic insights, supports biomarker discovery, and underpins translational research in immunology—demonstrating that most immune diversity is stable even during disease states.[6][4]
- The scalable, sensitive characterization enabled by iRepertoire technology powers multidimensional research and innovation in systems immunology, precision medicine, and population health.[4][6]
Overall, the article demonstrates that immuneREF—enabled by iRepertoire’s deep sequencing platforms—offers a powerful and nuanced approach to reference-based, cohort-scale immune repertoire analysis, redefining understanding of immune similarity and difference across populations.[5][1][6][4][2]
Winkler, et al. "Adoptive transfer of donor-B lymphocytes: a phase I/IIa study for patients after allogeneic stem cell transplantation." 2024, doi: 10.1182/bloodadvances.2023012305
Summary
This scholarly article reports a phase 1/2a study evaluating the adoptive transfer of donor B lymphocytes after allogeneic stem cell transplantation (allo-HSCT), demonstrating dose-dependent safety, tolerability, and enhanced vaccine-specific immune responses without increased incidence of chronic graft-versus-host disease (cGVHD).[1]
Key Findings
- Patients who received higher doses of donor B lymphocytes (DL3/4 group) exhibited significantly greater frequencies of vaccine-specific antibody-secreting cells (ASCs) and vaccine-responding patients compared to lower dose groups (DL1/2), establishing a clear dose effect.[1]
- Allo-HSCT recipients treated with donor B cell transfer showed durable B cell reconstitution and improved responses to tetanus and diphtheria booster vaccinations.[1]
- The rate of acute or chronic GVHD did not increase, likely due to highly purified B cell products with minimal T cell contamination, indicating a favorable safety profile for B cell therapy in this setting.[1]
- The data suggest that timely adoptive B cell transfer post-allo-HSCT can restore humoral immunity and enhance vaccine responsiveness in immunocompromised patients.[1]
iRepertoire Technology Usage
- iRepertoire’s advanced multiplex PCR technologies—such as arm-PCR and dam-PCR—were utilized to sensitively and inclusively capture the immune repertoire, enabling quantification of B cell clonal diversity and isotype usage after cell therapy and vaccination.[2]
- Their sequencing approach provided deep coverage for BCR (immunoglobulin) gene segments, allowing for assessment of vaccine-induced clonal expansions and monitoring B cell recovery.[3][2]
- iRepertoire’s methods proved ideal for profiling low-volume and challenging patient samples, supporting robust measurement of immune reconstitution post-transplant.[2]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was pivotal for demonstrating enhanced diversity and responsiveness of B cells following adoptive transfer, helping link clonal data with functional vaccine responses.[2][1]
- The approach revealed the effects of therapy on both frequency and spectrum of antibody-secreting clones, validating dose-dependent benefits and monitoring safety by detecting potential autoreactive expansions.[2][1]
- Such profiling is essential for assessing immune recovery, guiding B cell therapy dosing, and advancing transplantation immunology toward more precise, targeted interventions.[4][2][1]
In summary, iRepertoire technology delivered deep, quantitative immune repertoire data, enabling critical analyses that established safety and efficacy of donor B lymphocyte adoptive transfer post-allo-HSCT and confirmed improved vaccine responsiveness in immunocompromised patients.[3][4][2][1]
Choudhury, et al. "Immune responses to citrullinated and homocitrullinated peptides in healthy donors are not restricted to the HLA SE shared allele and can be selectred into the memory pool." 2023, doi: 10.1111/imm.13645
Summary
This scholarly article demonstrates that immune responses to citrullinated and homocitrullinated peptides occur in both healthy donors and rheumatoid arthritis (RA) patients, showing that these stress-induced post-translational modifications (siPTMs) can prime the adaptive immune system even outside autoimmunity. Notably, responses are not limited to individuals carrying the HLA-SE shared allele and modified peptide-specific T cells can be selected into the memory pool, reflecting environmental and infectious stress triggers rather than solely disease-specific mechanisms.[1]
Key Findings
- The majority of healthy donors exhibited CD4 T cell proliferation to multiple citrullinated and homocitrullinated peptides, not confined to known RA-associated HLA alleles.[1]
- Most responding CD4 T cells were effector memory cells with Th1/cytotoxic characteristics, indicating past antigen exposure and adaptation.[1]
- The memory response originated for specific peptides like Vim28-49cit, while other responses such as Vim415-433cit were naïve, suggesting peptide- and context-specific immunogenicity.[1]
- Experimental models confirmed that immunization with citrullinated peptides evokes durable memory responses, and peptide-specific T cells recognize stress-induced modifications during infection, linking these responses to environmental triggers.[1]
iRepertoire Technology Usage
- iRepertoire sequencing technology was used to quantify and characterize T cell receptor (TCR) diversity and clonality in response to modified peptides, mapping unique clonotypes among healthy and diseased subjects.[2][3]
- The technology’s high-throughput, multiplex approach enabled sensitive and unbiased detection of epitope-specific T cell repertoires, confirming that modified peptide-responsive cells can be selected into the memory pool and are not restricted by HLA-SE status.[3][2]
- iRepertoire allowed for deep quantitative analysis of effector and memory T cell populations, supporting insights into immune memory formation and environmental stress responses.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was pivotal in detecting the breadth and specificity of T cell responses to citrullinated and homocitrullinated peptides, determining how immune memory forms in health and disease.[3][2][1]
- This analysis revealed the true diversity of antigen-specific T cell pools, debunking the idea that these responses are unique to RA or certain HLA backgrounds.[2][3]
- By enabling precise tracking of memory and effector T cell subsets, immune repertoire analysis helped clarify how protein modifications from inflammation, infection, and stress can shape lasting immunity.[3][2][1]
In summary, iRepertoire technology powered robust immune repertoire sequencing, enabling discovery of broad, stress-responsive T cell memory to citrullinated and homocitrullinated peptides and illuminating new dimensions of adaptive immunity in both health and autoimmunity.[2][3][1]
Janarthanam, et al. "Bulk T-cell receptor sequencing confirms clonality in pediatric eosinophilic esophagitis and identifies a food-specific repertoire." 2023, doi: 10.1111/all.15773
Summary
This scholarly article uses bulk T-cell receptor (TCR) sequencing to confirm clonality in pediatric eosinophilic esophagitis (EoE), providing key insight into adaptive immune mechanisms underlying EoE onset and progression.[1][2]
Key Findings
- The study found differences in T cell clonality between pediatric and adult EoE patients, suggesting age-related variations in the immunopathogenesis of EoE.[2][1]
- Pediatric EoE demonstrated distinctive clonal T cell expansions, supporting the hypothesis of antigen-driven T cell responses in the disease’s early stages.[1][2]
- There were no significant differences in clonality measures between adults with EoE and healthy controls, indicating adaptive immune responses may play a more central role in pediatric disease.[2][1]
- These findings underscore a need for further immunological stratification in diagnosing and treating pediatric EoE based on immune repertoire signatures.[1]
Use of iRepertoire Technology
- The study leveraged iRepertoire’s multiplex PCR-based sequencing technology, enabling comprehensive, unbiased analysis of TCR beta chain diversity from bulk patient samples.[3][4]
- iRepertoire provided deep sequencing coverage across the CDR3 region, critical for detecting rare or dominant T cell clonotypes and characterizing immune responses at high resolution.[4][3]
- Their technology supported cost-efficient, sample-multiplexed analysis, facilitating robust quantitative comparison across pediatric and adult cohorts.[3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was essential for identifying and quantifying clonal T cell populations, revealing antigen-driven responses in pediatric EoE and advancing understanding of EoE etiology.[4][3][1]
- This approach enabled the discovery of potential disease biomarkers and therapeutic targets, supporting improved clinical management of EoE in children.[3][4][1]
- By highlighting clonality differences, repertoire sequencing aids in early intervention strategies and personalized medicine initiatives for immune-mediated diseases like EoE.[4][1][3]
In summary, iRepertoire’s technology enabled high-throughput TCR repertoire analysis that illuminated age-dependent clonal expansions in pediatric EoE, providing foundational insight for future diagnostic, therapeutic, and translational research.[1][3][4]
Dirks, et al. "IgD shapes the pre-immune naïve B cell compartment in humans." 2023, doi: 10.3389/fimmu.2023.1096019
Summary
This scholarly article explores how surface IgD expression shapes the pre-immune naïve B cell compartment in humans, utilizing patients with heterozygous IGHD mutations and deep immunoglobulin repertoire sequencing to uncover IgD’s role in B cell maturation and antigen receptor selection.[1]
Key Findings
- IgD-negative naïve B cells, arising from heterozygous IGHD mutations, are phenotypically distinct—showing reduced CD19, CD20, and CD21 expression, lower BAFF-R and integrin-β7, and are less responsive to IgM-BCR, TLR7/9, or CD40 stimulation in vitro.[1]
- IgD-negative B cells are at a selective disadvantage from the T2 transitional stage onwards; mature naïve B cells lack IgD and show lower frequencies of IgM low/– B cells, which are enriched for autoreactive clones.[1]
- High-throughput immune repertoire sequencing revealed altered selection of immunoglobulin VH segments, indicating that IgD expression helps guide which BCR clonotypes populate the pre-immune repertoire; specifically, IgD-negative cells had over- or underrepresentation of certain VH segments and lower species richness and diversity in most cases.[1]
- The study concludes that while IgD is dispensable for naïve B cell generation, its expression is critical for shaping VH segment selection and maintaining diversity of the pre-immune B cell pool.[1]
Use of iRepertoire Technology
- Next-generation immune repertoire sequencing of sorted B cell subsets was performed using iRepertoire technology, which enabled in-depth profiling of VH, DH, and JH gene usage, CDR3 length and composition, and diversity metrics.[1]
- The multiplex PCR and sequencing pipeline allowed sensitive and unbiased detection of major and minor B cell clones, illuminating the fine structure of the naïve repertoire in individuals with and without IgD expression.[1]
- iRepertoire’s bioinformatics suite facilitated quantitative analysis of clonotype frequency, diversity, and genetic selection, directly supporting the discovery of critical differences in VH segment representation tied to IgD status.[1]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was essential for mapping clonal selection events and for quantitatively measuring how loss of IgD expression alters the diversity, richness, and VH segment usage of naïve B cells.[1]
- The approach enabled correlation of functional, phenotypic, and genetic data, clarifying mechanistic links between receptor expression and adaptive immune diversity.[1]
- This level of analysis was crucial for revealing that IgD shapes the pre-immune B cell compartment beyond simply enabling naïve B cell generation, providing new insight into the molecular rules of humoral immunity.[1]
In summary, by integrating iRepertoire sequencing technology and extensive immune repertoire analysis, this paper demonstrates that surface IgD fine-tunes selection and survival of key naïve B cell subsets and their receptor diversity in humans.[1]
Lee, et al. "The endogenous repertoire harbors self-reactive CD4+ T cell clones that adopt a follicular helper T cell-like phenotype at steady state." 2023, doi: 10.1038/s41590-023-01425-0
Summary
This scholarly article characterizes self-reactive CD4+ T cells suppressed by regulatory T cells (Tregs), illuminating how immune tolerance is maintained and how breakdowns in this process can lead to autoimmunity.[1][2]
Key Findings
- The study reveals that the endogenous CD4+ T cell repertoire contains substantial numbers of self-reactive clones, many of which are actively suppressed by Tregs.[2][1]
- Advanced repertoire sequencing and computational analysis identified the phenotypic and transcriptional identities of Treg-suppressed self-reactive CD4+ T cells, providing molecular insight into immune homeostasis.[1]
- Experiments showed that removal of Treg-mediated suppression unlocks these latent self-reactive T cell populations, leading to autoimmune pathology in model systems.[1]
- The findings contribute to understanding the balance between immune tolerance and activation and highlight targets for therapeutic modulation of Tregs in autoimmune disease.[2][1]
Use of iRepertoire Technology
- iRepertoire’s technology facilitated bulk and single-cell TCR repertoire sequencing, enabling detailed mapping of T cell clonal diversity and antigen specificity from complex immune populations.[3][4][5]
- Their multiplex PCR and sequencing workflows allowed for high-resolution detection of rare self-reactive T cell clonotypes and functional characterization of Treg-suppressed subsets.[6][5]
- iRepertoire platforms supported unbiased comparison of TCR β-chain usage and facilitated the tracking of clonal expansion, persistence, and fate in response to experimental manipulation of Treg activity.[4][5][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was critical for the identification and quantification of self-reactive CD4+ T cell clones, clarifying their frequency, phenotype, and dynamics within the immune system.[5][6][1]
- This approach enabled mechanistic dissection of immune tolerance and autoimmunity, providing pivotal data for translational immunology and therapeutic design in autoimmune and regulatory disorders.[5][1]
- The capability to profile single-cell and bulk repertoires at high depth is essential for linking clonal diversity with functional state and responses to regulatory perturbations.[3][6][5]
In summary, iRepertoire repertoire sequencing and analytic platforms were integral to revealing the landscape and biology of self-reactive T cells suppressed by Tregs, advancing understanding and intervention in immune tolerance and autoimmunity.[6][4][3][2][5][1]
Lee, et al. "Alteration of γδ T cell subsets in non-human primates transplanted with GGTA1 gene-deficient porcine blood vessels." 2023, doi: 10.1111/xen.12838
Summary
This scholarly article demonstrates that xenotransplantation in non-human primates significantly alters the γδ T cell receptor (TCR) repertoire, highlighting the critical role of γδ T cells in sustained xenoreactive immune responses and informing future strategies to manage graft rejection.[1]
Key Findings
- Xenotransplantation induces pronounced changes in the composition and clonal diversity of γδ T cell subsets, suggesting that these cells actively participate in the immune response to xenogeneic grafts.[1]
- The altered repertoire correlates with long-term graft survival and immune reactivity, indicating that monitoring γδ T cell dynamics may serve as a biomarker for transplant outcome and immune adaptation.[1]
- The study provides new evidence for γδ T cells as key mediators of xenoreactivity and immune surveillance following transplantation across species barriers.[1]
Use of iRepertoire Technology
- iRepertoire’s multiplex PCR technology was used to perform high-throughput, sensitive sequencing of γδ TCR chains, allowing for comprehensive quantitative analysis of repertoire shifts following xenotransplantation.[2][3]
- Bulk and single-cell sequencing approaches enabled unbiased detection of clonal expansions, rare and dominant TCRs, and identification of functional subset specificities relevant to graft adaptation and immune response.[3][2]
- iRepertoire pipelines provided robust data from small samples and complex tissues, essential for mechanistic and translational studies in transplantation immunology.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was pivotal for characterizing the full extent of TCR diversity and clonal selection in transplanted primates, revealing immune correlates linked to long-term xenograft survival or rejection.[3][2][1]
- The approach shed light on the molecular mechanisms underlying immune adaptation and xenoreactivity, advancing biomarker discovery and therapeutic intervention.[2][3][1]
- This technology supports precision immune monitoring and guides future innovations in xenotransplantation through detailed mapping of adaptive and innate immune function.[3][2]
In summary, iRepertoire technology enabled sensitive and comprehensive immune repertoire analysis essential for revealing the dynamic role of γδ T cells in xenotransplantation, propelling research on immune adaptation and graft success across species barriers.[2][3][1]