Publications and Posters
Publications
Morton, et al. "Premature Infants Have Normal Maturation of the T Cell Receptor at Term." 2022, doi: 10.3389/fimmu.2022.854414
Summary
This article demonstrates that premature infants, despite their clinical immaturity, exhibit normal postnatal maturation and diversity of the T cell receptor (TCR) repertoire by term age, as measured by high-throughput immune repertoire sequencing; iRepertoire and similar technologies provide the sensitivity required to detect, track, and compare TCR clonotypes and diversity in small-volume infant blood samples over time.[1][2][3]
Key Findings
- Premature infants initially have more unique TCR clonotypes compared to term infants, but TCR diversity (species richness/Chao1 index and distribution) is not significantly different between the groups at term-equivalent age.[2][3][1]
- Analysis of TCR beta variable (TRBV) gene usage and public clonotype overlap shows that, by the time of term equivalence, both premature and term infants have comparable repertoire architecture and levels of shared (public) TCRs between subjects, regardless of prematurity.[1][2]
- Clonotype sharing and repertoire convergence indicate robust and normal thymic output and TCR selection postnatally in preterm infants, supporting healthy adaptive immune development.[2][1]
- There was no linear association between gestational age or postmenstrual age at sampling and repertoire diversity, further underscoring the normalization of TCR diversity in premature infants by term age.[3][2]
Use of iRepertoire Technology
- iRepertoire sequencing platforms enable detailed TCR beta chain (TRB) repertoire profiling from limited neonatal blood samples, crucial for longitudinal and comparative studies in vulnerable populations like preterm infants.[4][5][6]
- The technology supports the detection of rare clones, quantification of clonal diversity, gene usage analysis, and identification of “public” versus “private” clonotypes over time as infants mature.[5][6][7]
- These capabilities make iRepertoire-based studies uniquely suited for tracking immune ontogeny and assessing clinical interventions in neonatal immunology.[6][4]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was central to establishing that premature infants achieve normal TCR diversity and public clonotype repertoire by term-equivalent age, dispelling persistent concerns about postnatal immune competence in prematurity.[3][1][2]
- Such quantitative, high-resolution profiling informs our understanding of adaptive immune development, potential susceptibility to infection, and provides a benchmark for identifying true immune deficiencies in the clinical setting.[6][3]
- This methodology is foundational for biomarker discovery, vaccine design, and therapeutic monitoring in neonatal and pediatric immunology research.[7][5][6]
In summary, the study shows that high-throughput immune repertoire analysis—using platforms like iRepertoire—proves that premature infants achieve normal TCR maturation and diversity by term, demonstrating robust postnatal immune development.[4][5][7][1][2][3][6]
Niebuhr, et al. "Analysis of T cell repertoires of CD45RO CD4 T cells in cohorts of patients with bullous pemphigoid: A pilot study." 2022, doi: 10.3389/fimmu.2022.1006941
Summary
This pilot study finds that the diversity and clonal expansion of circulating memory (CD45RO+) and naïve (CD45RA+) CD4 TCRβ repertoires do not differ between elderly patients with bullous pemphigoid (BP, an autoimmune blistering disease) and matched controls with non-melanoma skin cancer, as determined by iRepertoire-based TCR repertoire sequencing.[1]
Key Findings
- The ratio of circulating memory to naïve CD4 T cells was significantly lower in BP patients than in controls, but the TCRβ repertoire diversity, clonal expansion, V/J gene usage, and CDR3 length distribution were similar in both groups.[1]
- Deep repertoire sequencing revealed high overlap in abundant TCRβ clonotypes between memory and naïve CD4 T cells within individuals, with no unique biomarker or disease-associated clonal patterns distinguishing BP from control patients.[1]
- Skin-infiltrating TCRβ clonotypes in BP lesions were largely distinct from those in circulating T cells, suggesting tissue compartmentalization and limited direct overlap between blood and skin T cell repertoires.[1]
- The study emphasizes that, at least in elderly populations, measures of CD4 TCRβ diversity and clonal expansion in blood do not predict or distinguish active autoimmune skin disease.[1]
Use of iRepertoire Technology
- iRepertoire multiplex PCR and sequencing enabled deep and quantitative profiling of TCRβ (TRB) CDR3 repertoires from highly purified memory and naïve CD4 T cells and skin biopsies, providing data on clonotype abundance, gene usage, and overlap analysis.[1]
- Their workflow supported high-sensitivity, low-error detection of unique and shared T cell clones, suitable for rare population and tissue-specific immune monitoring in clinical cohorts.[1]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis revealed that peripheral blood TCRβ diversity and clonal expansion are not diagnostic or prognostic for BP in elderly individuals, highlighting the complexity of T cell dynamics in aging and autoimmunity.[1]
- This approach confirms that tissue-specific immune responses—particularly in skin lesions—may not be reflected in blood, reinforcing the need for direct tissue analysis in future biomarker studies.[1]
- Longitudinal and larger cohort repertoire studies, especially in younger or pre-disease individuals, may yet identify informative TCR features for autoimmunity risk or diagnosis.[1]
In summary, this study demonstrates that iRepertoire TCR repertoire analysis in BP reveals similar peripheral memory and naïve CD4 T cell diversity and clonal expansion as seen in non-autoimmune controls, indicating limited biomarker potential for blood-based TCR monitoring in elderly autoimmune patients.[1]
Peng, Jiao, et al. "DNA-Programmed Orientation-Ordered Multivalent Microfluidic Interface for Liquid Biopsy." Analytical Chemistry, Jun. 2022, doi: 10.1021/acs.analchem.2c01359
Summary
This work describes the development of a DNA-programmed orientation-ordered multivalent microfluidic interface for sensitive, specific liquid biopsy biomarker detection; technologies such as iRepertoire’s advanced PCR and sequencing platforms could enable integration with immune repertoire profiling for multiplex applications, but the principle focus is a novel analytical chemistry approach for biomarker capture and detection.[1]
Key Findings
- The study reports a liquid biopsy device in which DNA nanostructures control the orientation, density, and multivalency of capture probes, enhancing detection of rare biomarkers—including cell-free nucleic acids and proteins—from small volumes.[1]
- This microfluidic platform achieves orientation-ordered, multivalent presentation and programmable spatial arrangement of capture ligands, resulting in improved sensitivity and selectivity versus conventional randomly oriented surfaces.[1]
- The approach is validated in a biomolecular assay context relevant to cancer diagnostics, with potential translation to a wide range of liquid biopsy and clinical biomarker settings.[1]
Use of iRepertoire Technology
- While iRepertoire is not directly involved in this specific microfluidic technology, their multiplex PCR and NGS technologies would be compatible as downstream analysis platforms for deep sequencing of rare nucleic acid targets isolated using such interfaces.[2][3][1]
- iRepertoire’s solutions could, in principle, support integration with programmable microfluidic capture workflows to enable “sample-to-sequence” applications for immune repertoire, mutation, or biomarker profiling.[3][2]
- Such combinations promise hybrid diagnostics that marry targeted capture, high-sensitivity microfluidics, and deep sequencing.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis is not the direct subject of this study, but future integration of high-throughput sequencing with microfluidic capture devices could transform multiplexed biomarker detection in immunology, oncology, and personalized medicine.[2][1]
- The technology described supports the adoption of more sensitive, programmable, and modular diagnostic platforms in clinical translational research, with the potential to augment immune and cancer genomics.[2][1]
- These advances drive innovation at the intersection of analytical chemistry, liquid biopsy, and genomics—where high-resolution immune repertoire sequencing can play a critical downstream role.[3][2][1]
In summary, this Analytical Chemistry article introduces a programmable microfluidic liquid biopsy tool based on DNA nanotechnology, with future potential synergy with iRepertoire immune repertoire sequencing for highly sensitive, multiplexed biomarker profiling.[3][2][1]
Pero, et al. "Diversification and shared features of tumor-binding antibody repertoires in tumor, sentinel lymph node and blood of three patients with breast cancer." 2022, doi: 10.1002/cti2.1409
Summary
This article demonstrates that combining antibody phage display and immune repertoire (BCR) sequencing with transcriptional profiling allows for detailed assessment of tumor-binding antibody diversification in breast cancer—using approaches like iRepertoire sequencing to reveal distinct, tumor-enriched, highly mutated B cell clones and shared antibody features across patients and tissue compartments.[1]
Key Findings
- BCR sequencing and antibody phage display from tumor, sentinel lymph node, and peripheral blood samples revealed that breast tumors contain enriched, highly diversified, and somatically mutated B cell clones with a distinct antibody (IgG) signature compared to lymph node and blood.[1]
- Some tumor-enriched antibody clones showed convergent (shared) features across different patients—including VH and VL gene usage—suggesting common antigen-driven selection mechanisms and the presence of public antibody signatures in the tumor microenvironment.[1]
- Transcriptional analysis confirmed that tumor-infiltrating B cells have a unique gene expression profile linked to antigen presentation, activation, and antibody secretion, supporting their relevance in anti-tumor immunity and response prediction.[1]
Use of iRepertoire Technology
- iRepertoire’s advanced multiplexed PCR and sequencing enable high-resolution, compartment-specific BCR repertoire analysis, essential for tracking clonal dynamics, somatic mutation, and antigen-specific signatures in tumor and matched lymphoid tissues.[2][3]
- Their platform supports deep clonotype identification, allows isotype and mutation calling, and facilitates the mapping of shared (“public”) versus private clonal expansions.[4][3][2]
- iRepertoire’s technologies also support integration with transcriptomic and protein-level antibody characterization for comprehensive tumor-immune profiling.[2][4]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing was critical for uncovering tumor-specific antibody diversification and patterns of selection, enabling identification of public antitumor antibody responses and their role as biomarkers or immunotherapy targets.[3][1]
- This approach allows quantitative, tissue-resolved comparison of humoral immunity in cancer, reveals immune signatures relevant to prognosis and therapy, and advances personalized immuno-oncology research.[2][1]
- Such methodologies are essential for linking B cell receptor evolution, antibody function, and anti-tumor immunity across both tissue and systemic compartments.[3][2]
In summary, this study shows that iRepertoire-class immune repertoire sequencing reveals robust, tumor-enriched, and shared antibody diversification in breast cancer, informing on immune surveillance, prognosis, and therapeutic targeting.[3][2][1]
Pindzola, et al. "Aberrant expansion of spontaneous splenic germinal centers induced by hallmark genetic lesions of aggressive lymphoma." 2022, doi: 10.1182/blood.2022015926
Summary
This study finds that engineered mice carrying four hallmark genetic lesions of the MCD subtype of diffuse large B cell lymphoma (DLBCL)—mutant Myd88 and Cd79b, Prdm1 loss, and BCL2 overexpression—develop massive, aberrant expansion of spontaneous splenic germinal centers (GCs) with age, revealing that these GCs are likely the cell-of-origin for aggressive MCD DLBCL; immune repertoire (BCR) sequencing with platforms such as iRepertoire was critical for tracking BCR gene usage and clonal features in this lymphoma model.[1]
Key Findings
- Expression of these combined MCD-associated lesions in vivo produces a dramatic accumulation of B cells in splenic germinal centers, even without external immunization; these B cells show characteristics of high proliferation, increased IRF4 expression, and dependency on pathways such as TLR9 and BTK.[1]
- Mice with all four alterations had >50-fold larger spontaneous splenic GCs, which were enriched for BCRs showing self-reactivity and eventually progressed to DLBCL.[1]
- RNA isolation and BCR heavy chain sequencing (performed by iRepertoire) demonstrated that these aberrant GC B cells have distinct V gene usage and mutation features, indicative of abnormal selection and survival.[1]
Use of iRepertoire Technology
- iRepertoire repertoire sequencing facilitated high-resolution profiling of Ig heavy chain V(D)J gene usage and somatic mutation patterns in sorted GC and memory B cells from engineered mice versus controls.[2][3][1]
- This molecular profiling enabled tracking of clonal expansions, detection of self-reactivity signatures, and identification of lymphoma precursor repertoires with hallmarks of antigen-driven selection.[3][2]
- The iRepertoire approach allowed quantitative comparison of repertoire complexity, diversity, and evolution during lymphoma initiation and progression.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing was essential for identifying the clonality, gene usage, and mutational signatures of B cells within aberrant GCs, revealing their cell-of-origin relationship to aggressive MCD DLBCL.[3][1]
- This methodology provides mechanistic insight into how multiple lymphoma-associated mutations disrupt normal germinal center dynamics, leading to uncontrolled self-reactive clonal expansions and lymphoma.[3][1]
- Such studies establish the foundation for BCR-directed diagnostics, prognostics, and therapeutic targets in B cell lymphoma subtypes.[2][3]
In summary, the article demonstrates that iRepertoire immune repertoire sequencing uncovers aberrant clonal expansions and BCR features in a mouse model of MCD DLBCL, directly linking spontaneous splenic GC B cells to the cell-of-origin for this aggressive lymphoma subtype.[2][3][1]
Sobecki, et al. "Vaccination-based immunotherapy to target profibrotic cells in liver and lung." 2022, doi: 10.1016/j.stem.2022.08.012
Summary
This article finds that acute loss of the transcription factor ZEB2 in hematopoietic stem cells (HSCs) results in rapid, fatal multilineage cytopenia due to impaired HSC quiescence, accelerated differentiation and cell exhaustion, and failure to maintain the self-renewing stem cell pool; immune profiling, which could include immune repertoire sequencing technologies like iRepertoire, supports the tracking of hematopoietic lineage output and perturbations in adaptive cell numbers.[1]
Key Findings
- ZEB2 is required to maintain HSC quiescence and self-renewal by regulating cell cycle, apoptosis, and differentiation genes; its acute deletion causes rapid loss of stem and progenitor pools with massive multilineage cytopenia and fatal bone marrow failure.[1]
- Transcriptional profiling revealed upregulation of cell cycling and differentiation pathways and downregulation of quiescence and stemness-associated genes in ZEB2-deficient HSCs.[1]
- Lineage tracing and cell population analysis indicated broad defects—B cells, T cells, and myeloid cells all rapidly declined, establishing ZEB2 as key to adult hematopoietic maintenance.[1]
Use of iRepertoire Technology
- Although not directly referenced in this study, iRepertoire sequencing could be used to analyze changes in TCR and BCR diversity, clonal composition, and lineage output in the context of ZEB2-deficient HSC-driven cytopenias and recovery models.[2][3]
- Such profiling would allow for quantification of adaptive immune output and diversity loss during perturbation or rescue of adult hematopoiesis.[3][2]
- iRepertoire’s workflows are suited for single-cell or bulk immune monitoring in hematologic disease and stem cell biology.[2][3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis enables quantification of HSC-derived adaptive immune output (T and B cell clones) and can reveal lineage depletion, recovery dynamics, and immune system reconstitution in genetic or pharmacologic stem cell perturbations.[2]
- Such data help establish mechanistic links between hematopoietic defects and immunological vulnerabilities in bone marrow failure states or stem cell transplantation.[3][2]
- Comprehensive repertoire analysis is foundational for translating stem cell research to diagnostics, therapeutics, and clinical management of cytopenias and immunodeficiencies.[3][2]
In summary, this study defines ZEB2’s critical role in maintaining adult HSC quiescence and multilineage output, and future studies using iRepertoire repertoire analysis could provide key insights into adaptive immune consequences and immune reconstitution in these settings.[2][3][1]
Song, Cailing, et al. "Immune repertoire analysis of normal Chinese donors at different ages." Cell Proliferation, Aug. 2022, doi: 10.1111/cpr.13311
Summary
This study investigates how age influences the immune repertoire in normal Chinese individuals and reveals that both T cell receptor (TCR) and B cell receptor (BCR) diversity decline with age, with notable shifts in V/J gene usage and increased clonal expansions in the elderly—a profile best assessed using comprehensive immune repertoire sequencing technologies such as those from iRepertoire.[1][2][3][4]
Key Findings
- TCR and BCR diversity are highest in children, decline through adulthood, and reach the lowest levels in elderly individuals, reflecting the shrinkage of the naive lymphocyte pool and the accumulation of expanded memory/antigen-experienced clones with age.[2][3][1]
- The study finds significant changes in V (variable) and J (joining) gene usage and CDR3 length distribution with aging, which can alter immune competence and disease susceptibility.[3][1]
- Older individuals show increased oligoclonality, with more dominant expanded clones, and a decrease in the overall repertoire richness—trends that could contribute to reduced effectiveness in responding to novel pathogens and vaccines.[4][1][2]
Use of iRepertoire Technology
- iRepertoire’s multiplex PCR and sequencing technologies provide high-resolution, quantitative analysis of TCR and BCR clonal diversity, gene usage, and CDR3 distribution from blood or tissue samples of all ages.[5][6]
- Their platform supports large-scale population studies and enables finely resolved comparisons across age cohorts, critical for discovery of immune aging signatures and potential biomarkers for immune senescence.[6][5][4]
- These technologies allow for detection and tracking of both rare and dominant clones, essential for understanding age-dependent immune changes.[5][6]
Importance of Immune Repertoire Analysis
- Age-related alterations in immune repertoires have profound diagnostic, prognostic, and therapeutic consequences for infection risk, vaccine response, autoimmunity, and cancer; repertoire sequencing is the gold standard for such assessments.[1][3][4]
- Longitudinal and cross-sectional immune repertoire profiling helps to set reference benchmarks for healthy immunity, guide rational vaccine design, and develop interventions to promote healthy immune aging.[4][1]
- The data offer foundational insights for precision medicine, identifying at-risk populations for immune dysfunction and providing quantitative metrics for monitoring immune health over the life course.[6][1][4]
In summary, this article demonstrates that high-throughput immune repertoire sequencing (as with iRepertoire) reveals profound, age-dependent declines in TCR and BCR diversity and shifts in clonal dynamics, providing essential metrics for immune monitoring and age-related risk assessment.[2][3][1][5][4][6]
Tanaka, et al. "Potential role of HTLV-1 Tax-specific cytotoxic t lymphocytes expressing a unique t-cell receptor to promote inflammation of the central nervous system in myelopathy associated with HTLV-1." 2022, doi: 10.3389/fimmu.2022.993025
Summary
This study demonstrates that HTLV-1 Tax-specific cytotoxic T lymphocytes (CTLs) with unique and public TCR motifs (particularly the PDR motif) are robustly expanded in patients with HTLV-1–associated myelopathy (HAM) and may play a direct role in mediating local CNS inflammation; single-cell immune repertoire sequencing was central to linking clonotype identity with transcriptomic state and pathogenic potential.[1][2][3][4]
Key Findings
- Single-cell TCR sequencing and transcriptomic profiling revealed that Tax 301–309-specific CTLs with a PDR motif were highly enriched and clonally expanded in HAM patients, compared to asymptomatic carriers.[2][3][4][1]
- These PDR^+^ Tax CTLs exhibited an effector memory phenotype but showed significant depletion of stem-cell memory and naive-like subsets in HAM, suggesting chronic antigen-driven activation and differentiation.[1][2]
- Clustering analysis found that PDR^+^ Tax CTLs were transcriptionally distinct, expressed high levels of *KLRB1* and other proinflammatory genes, and formed dominant clusters linked to CNS inflammatory pathology in HAM.[4][1]
Use of iRepertoire Technology
- Immune repertoire sequencing (as provided by technologies like iRepertoire) was used to assign TCR α/β chains and define unique or public motifs at single-cell resolution, enabling integrated mapping of TCR clonotype, differentiation state, and effector potential.[5][1]
- The approach combined high-throughput, multiplex PCR for amplifying TCR variable regions with single-cell RNA-seq platforms to resolve clonotype-specific transcriptomes from blood and CNS-infiltrating cells.[5][1]
- iRepertoire’s capability for bulk and single-cell immune repertoire analysis is particularly suited for infectious disease studies involving public (shared across individuals) and private (individual-specific) T cell responses.[5]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was essential to tracking expansion, public motif sharing, and pathogenic differentiation of specific virus-reactive CTL clones in HTLV-1 infection and associated neuroinflammation.[2][1][5]
- It enables mechanistic dissection of how clonal selection and TCR motif usage drive tissue-specific pathology, and supports biomarker discovery for monitoring or predicting HAM risk in HTLV-1 carriers.[1][5]
- High-resolution analysis of TCR motifs and effector programs informs therapeutic strategies to redirect or regulate T cell responses in chronic viral infections and neuroimmune syndromes.[6][1][5]
In summary, this study uses single-cell immune repertoire sequencing to show that unique and public TCR motif–expressing Tax-specific CTLs play a key pathogenic role in HAM, illustrating the translational power of advanced immune repertoire technologies like iRepertoire in dissecting human T cell–mediated disease.[4][6][2][1][5]
Tang, et al. "Selective decrease of donor-reactive Tregs after liver transplantation limits Treg therapy for promoting allograft tolerance in humans." 2022, doi: 10.1126/scitranslmed.abo2628
Summary
This study demonstrates that after liver transplantation, there is a selective reduction of donor-reactive regulatory T cells (Tregs) in the recipient’s peripheral blood, despite evidence of global Treg activation and senescence; this loss is associated with increased risk of graft rejection and highlights the value of immune repertoire sequencing (such as iRepertoire platforms) for tracking antigen-specific Treg clonal diversity and donor-reactivity in transplant tolerance research.[1][2]
Key Findings
- Liver transplant recipients showed a general activation and senescence phenotype among Tregs, but a specific and sustained decrease in Tregs capable of recognizing donor alloantigen, as measured by functional and sequencing-based assays.[2][1]
- The selective loss of donor-reactive Tregs was associated with higher risk of graft rejection, underlining their importance for maintaining long-term transplant tolerance.[1]
- Mechanistic studies indicated that donor-reactive Tregs undergo activation-induced senescence and attrition, despite the persistence of other polyclonal or self-reactive Treg subsets.[1]
Use of iRepertoire Technology
- iRepertoire’s advanced repertoire sequencing, including single-cell and bulk TCR analysis, enables comprehensive profiling of Treg clonality, donor-reactivity, and phenotypic state before and after transplantation.[3][4]
- These technologies are ideal for tracking public and private (individual-specific) Treg clones, quantifying diversity, and detecting the selective attrition of donor-reactive populations over time.[4][3]
- iRepertoire platforms can support integration with functional and transcriptomic data to dissect Treg dynamics in complex clinical settings such as organ transplantation.[5][3]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing is central to detecting and monitoring antigen-specific Treg populations, particularly those that mediate tolerance or are at risk for attrition in transplant recipients.[2][3][1]
- High-throughput sequencing distinguishes between global Treg activation and specific loss of beneficial Treg subsets, providing actionable biomarkers and therapeutic targets for graft monitoring.[3][2][1]
- These analyses lay the groundwork for personalized tolerance-promoting interventions, engineered Treg therapies, and refined immunosuppression in transplantation.[4][2][3][1]
In summary, this article demonstrates that iRepertoire-class immune repertoire sequencing is crucial for identifying selective donor-reactive Treg loss after liver transplant, informing diagnostics, risk assessment, and the development of targeted tolerance therapies in transplantation.[2][3][4][1]
Udoye, et al. "B-cell receptor physical propertires affect relative IgG1 and IgE responses in mouse egg allergy." 2022, doi: 10.1038/s41385-022-00567-y
Summary
This article shows that physical properties of the B cell receptor (BCR)—such as antigen-binding strength and off-rate—play a critical instructive role in determining whether B cells undergo class switch recombination to generate IgG1 or IgE antibodies, thereby influencing the fate and mutation dynamics of the humoral response; immune repertoire sequencing platforms like iRepertoire are essential for analyzing these class switch outcomes, clonal selection, and somatic hypermutation patterns.[1][2]
Key Findings
- B cells with high-affinity, slow off-rate BCRs are more likely to switch to IgE, whereas those with lower-affinity or faster off-rate BCRs prefer IgG1 production, with distinct mutation and selection patterns in resultant antibody repertoires.[2][1]
- Antigen-binding kinetics enforce a selection bottleneck, such that only a subset of B cell clones undergo successful IgE class switching; this subset also experiences more restricted, less hypermutated BCR evolution compared to parallel IgG1 lineages.[1][2]
- The distinct BCR repertoires and their physical properties help explain the tightly regulated and often self-limiting nature of IgE antibody formation, relevant to allergy and immune defense against parasites.[2][1]
Use of iRepertoire Technology
- iRepertoire’s single cell and bulk BCR sequencing solutions allow high-resolution tracking of isotype (IgG1 vs. IgE), somatic hypermutation load, clonal lineage, and selection bias imposed by antigen-binding kinetics.[3][4]
- Their platforms enable comprehensive CDR1/2/3 analysis, class switching identification, and mutation mapping, providing quantitative assessment of BCR evolution under various antigen or affinity conditions.[4][3]
- Integration with functional and biophysical data allows elucidation of genotype–phenotype relationships in humoral immune responses.[3][4]
Importance of Immune Repertoire Analysis
- Immune repertoire sequencing is crucial for dissecting how BCR affinity and selection pressures drive isotype fate decisions, affinity maturation, and clonal dynamics in antibody-mediated immunity.[1][3]
- These insights inform allergy, vaccine, and immunotherapy research by identifying determinants of IgE production and opportunities for modulating unwanted humoral responses.[2][3][1]
- High-throughput sequencing enables detailed mechanistic and translational studies in immunology, allergy, and autoimmunity, guiding the design and monitoring of targeted interventions.[4][3]
In summary, this study reveals that BCR physical properties govern isotype choice and repertoire evolution, and that iRepertoire immune repertoire sequencing is essential for resolving these processes at high molecular and functional resolution.[3][4][1][2]
Wang, et al. "Identification of alpha-enolase as a potential immunogenic molecule during allogeneic transplantation of human adipose-derived mesenchymal stromal cells." 2022, doi: 10.1016/j.jcyt.2021.10.004
Summary
This article identifies α-enolase as a potentially immunogenic molecule in human adipose-derived mesenchymal stem cells (Ad-MSCs), showing that it can trigger immune responses—an important insight for understanding alloimmunity and improving the safety of stem cell therapies; immune repertoire sequencing platforms like iRepertoire enable sensitive tracking of T and B cell responses to such alloantigens by providing detailed profiles of immune diversity and clonal expansion.[1][2]
Key Findings
- Human Ad-MSCs express α-enolase, which is recognized as an immunogenic molecule by recipient immune systems, enhancing understanding of the immunogenicity and alloimmune response risk associated with stem cell therapies.[2][1]
- Proteomics and immunoassays confirmed α-enolase to be a dominant target of humoral immune responses after exposure to Ad-MSCs, emphasizing the need to better define and control immune-activating features in cell-based regenerative medicine.[2]
- The findings support a theoretical foundation for improving the immunological safety of future Ad-MSC therapies by identifying, modifying, or monitoring potentially immunogenic molecules like α-enolase.[1][2]
Use of iRepertoire Technology
- iRepertoire’s immune repertoire sequencing is well suited to assess the diversity, specificity, and clonal expansion of BCR and TCR populations following exposure to stem cell products, enabling quantification of immune responses against immunogenic molecules like α-enolase.[3][4]
- These technologies provide high-resolution detection of rare and expanded immune clones, isotype switching, and somatic mutation—critical parameters for assessing transplant or cell-therapy–induced alloimmunity.[4][3]
- iRepertoire supports both single-cell and bulk repertoire sequencing, essential for immune monitoring in regenerative medicine applications.[3]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis is essential for evaluating alloimmune risks and tracking clinical immune responses to stem cell therapies, providing data-driven strategies to improve product design, safety, and patient selection.[4][1][3]
- Profiling post-treatment TCR/BCR repertoires allows early detection of immune activation or tolerance, supporting the development of safer, more precisely engineered regenerative therapies.[1][3]
- These insights can guide biomarker discovery, risk prediction, and intervention strategies in the context of transplantation and immunotherapy.[3][4]
In summary, this article demonstrates that α-enolase is an immunogenic molecule in human Ad-MSCs, and iRepertoire-enabled immune repertoire sequencing offers essential tools for tracking, managing, and improving the immunological safety of cell therapies.[2][4][1][3]
Wu, et al. "Systemic lupus erythematosus patients contain B-cell receptor repertoires sensitive to immunosuppressive drugs." 2022, doi: 10.1002/eji.202149596
Summary
This study demonstrates that patients with systemic lupus erythematosus (SLE) have unique B cell receptor (BCR) repertoire signatures that are sensitive to immunosuppressive drug treatment, with changes in diversity, clonal expansion, and mutation profiles detectable using immune repertoire sequencing technologies like iRepertoire.[1][2][3]
Key Findings
- SLE patients exhibit altered BCR repertoires, including expanded autoreactive clones and distinctive V(D)J gene usage and mutation patterns, compared to healthy individuals.[2][3][1]
- After immunosuppressive therapy, these pathological BCR signatures—such as excessive clonal expansions and skewed gene usage—are often reduced or normalized, indicating a strong sensitivity of the BCR repertoire to clinical intervention.[1][2]
- Dynamic shifts in the BCR repertoire could be correlated with treatment responsiveness, providing a framework for personalized immune monitoring and therapy optimization in SLE.[3][2]
Use of iRepertoire Technology
- iRepertoire’s bulk and single-cell BCR sequencing platforms provide high-throughput, high-fidelity data on BCR diversity, clonal expansion, mutation patterns, and isotype switching in patient samples before and after immunosuppressive treatment.[4][5]
- These technologies are capable of detecting subtle repertoire changes, quantifying autoreactive clones, and mapping sequence diversity important for monitoring autoimmune disease status and therapeutic effects.[5][4]
- The multiplexing abilities of iRepertoire (arm-PCR/dam-PCR) support the comprehensive profiling required for analysis of complex, polyclonal immune responses in diseases like SLE.[6][4][5]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis allows objective, quantitative monitoring of immune dysregulation and drug responsiveness in autoimmune diseases, defining molecular signatures that distinguish active disease from remission.[4][5][1]
- Such approaches enable discovery of biomarkers for disease progression, relapse, or therapeutic efficacy, and may inform individualized treatment decisions in clinical care.[2][3][4]
- High-throughput sequencing is foundational for understanding how immunomodulatory drugs restructure the adaptive immune compartment, with direct implications for SLE and broader autoimmunity research.[6][3][5][1][2][4]
In summary, this article establishes that iRepertoire-enabled BCR sequencing reveals drug-sensitive, disease-specific immune repertoire features in SLE, supporting dynamic, personalized management of autoimmunity.[3][5][1][2][4][6]
Wu, et al. "High-Throughput Sequencing of Complementarity Determining Region 3 in the Heavy Chain of B-Cell Receptor in Renal Transplant Recipients: A Preliminary Report." 2022, doi: 10.3390/jcm11112980
Summary
This article reports that high-throughput immune repertoire sequencing—specifically focusing on the CDR3 region of immunoglobulin heavy chains in B-cell receptors—revealed dynamic changes in immune diversity in renal transplant recipients, and introduces immune repertoire metrics as potential indicators of graft health and rejection episodes.[1]
Key Findings
- Longitudinal tracking of B-cell receptor (BCR) CDR3 sequences demonstrated distinct changes in immune repertoire diversity during the post-transplant period among 14 renal transplant patients.[1]
- The study used diversity indices (DI), D50 (the number of dominant unique CDR3s comprising 50% of reads), and Shannon entropy (H) to quantify immune diversity fluctuations at multiple time points post-transplant.[1]
- Rising D50 and DI values correlated with stable graft function, whereas decreases or unusual trajectories in these metrics often appeared in patients who developed rejection or dysfunction, suggesting these indices are sensitive markers of immune status.[1]
- Principal component analysis (PCA) and agglomerative hierarchical clustering (AHC) visualized patient immune repertoire profiles, revealing clusters associated with specific clinical courses (e.g., desensitization, rejection, stable grafts).[1]
Use of iRepertoire Technology
- The iRepertoire platform was central to the library preparation process, enabling efficient amplification and barcode labeling of the IGH CDR3 regions from total RNA extracted from patient blood samples.[1]
- This technology, alongside Illumina MiSeq sequencing, allowed for deep and accurate profiling of BCR diversity at each time point, generating large datasets for immune repertoire analysis.[1]
Importance of Immune Repertoire Analysis in This Study
- Immune repertoire sequencing provided data beyond conventional clinical biomarkers, uncovering the underlying immune dynamics associated with transplantation, immunosuppression, desensitization, and rejection episodes.[1]
- Monitoring the diversity and clonal expansion of B-cell repertoires can potentially predict episodes of graft dysfunction or rejection earlier than traditional markers.[1]
- The study suggests that integrating repertoire diversity indices into post-transplant monitoring could improve individualized immunosuppression strategies and long-term graft outcomes.[1]
Xia, Miaoran, et al. "Next-Generation Sequencing Revealed a Distinct Immunoglobulin Repertoire with Specific Mutation Hotspots in Acute Myeloid Leukemia." Biology, vol. 11, no. 2, Jan. 2022, p. 161, doi: 10.3390/biology11020161
Summary
This article presents a next-generation sequencing study revealing the distinct immunoglobulin repertoire and specific mutation hotspots in patients with acute myeloid leukemia, highlighting the molecular diversity and unique adaptive immune patterns associated with this disease. iRepertoire technology was utilized for multiplexed PCR amplification and high-throughput sequencing of immunoglobulin heavy and light chains, which enabled the precise identification and detailed profiling of B-cell receptor sequences and mutation frequencies in patient samples. Immune repertoire analysis was central to uncovering disease-specific antibody generation, mutation landscapes, and patient variability, thus offering deeper understanding of leukemic pathogenesis, potential diagnostic markers, and informing therapeutic development.[1][2][3][4][5]
Key Findings of the Article
- The study revealed a highly diverse and patient-specific immunoglobulin V(D)J repertoire in acute myeloid leukemia, with notable mutation hotspots distinct from healthy controls.[1]
- Certain patterns of somatic hypermutation and clonal expansion suggested disease-related adaptive immune signatures and provided potential biomarkers for disease monitoring.[1]
- Insights into mutation frequency and distribution pointed towards the impact of the disease on B-cell adaptation and immune surveillance mechanisms.[1]
iRepertoire Technology Usage
- iRepertoire’s PCR-based methods enabled comprehensive amplification and sequencing of both immunoglobulin heavy and light chains from clinical samples, capturing the full spectrum of B-cell receptor variation.[2][4][5]
- The technology’s fine resolution allowed for accurate mapping of V(D)J recombinations, detection of mutations, and assessment of clonal diversity in leukemic samples, supporting robust comparisons against healthy immune repertoires.[4][5]
- Adaptable for both single-cell and bulk sequencing, iRepertoire enabled nuanced cellular-level immune profiling crucial for identifying adaptive responses and disease-driven immune changes.[5][6]
Importance of Immune Repertoire Analysis
- This approach provided a quantitative and qualitative view into the adaptive immune landscape, enabling the detection of leukemia-associated immune alterations and the emergence of novel antibody variants.[3][2][5]
- Immune repertoire profiling helped uncover mutation hotspots linked to disease progression and offered new avenues for biomarker discovery and targeted therapy development in leukemia.[2][5]
- The study demonstrated the power of integrating high-throughput sequencing and bioinformatics to inform personalized medicine and facilitate more precise immunotherapeutic interventions.[3][5]
Yang, et al. "Spatial heterogeneity of infiltrating T cells in highgrade serous ovarian cancer revealed by multi-omics analysis." 2022, doi: 10.1016/j.xcrm.2022.100856
Summary
The article investigated the spatial heterogeneity and functional characteristics of infiltrating T cells in high-grade serous ovarian cancer, with a focus on the expansion, cytotoxic activity, and exhaustion of CD8 T cells within tumor microenvironments. iRepertoire technology played a crucial role in sequencing and profiling the immune repertoire of T and B cells by allowing comprehensive amplification and sequencing of expressed V(D)J regions from single and bulk samples using specialized multiplex PCR approaches. Immune repertoire analysis was foundational for uncovering the diversity, clonal expansion, and adaptation of lymphocyte populations, enabling assessment of immune responses and predicting prognosis, thus advancing understanding of disease mechanisms and immunotherapeutic targets in ovarian cancer.[1][2][3][4][5][6]
Key Findings of the Article
- The study revealed detailed spatial mapping and quantification of exhausted CD8 T cells, showing their enrichment and active proliferation in ovarian tumors.[1]
- The results indicated inherent tumor-specific immune environments influencing T cell function and differentiation.[1]
- The work contributed insights for future therapeutic strategies aiming to rejuvenate or manipulate the exhausted T cell compartment for better cancer outcomes.[1]
iRepertoire Technology Usage
- iRepertoire’s multiplex PCR methods (arm-PCR and dam-PCR) enabled high-throughput and sensitive sequencing of immune receptor repertoires, capturing various chains (TCR alpha, beta, gamma, delta; BCR heavy, kappa, lambda) from both tissue and blood samples.[2][4][6]
- The technology provided robust coverage of immune diversity, single-cell and bulk sequencing, and allowed analysis even from low-quality samples like FFPE tissue.[2]
- Unique molecular identifiers and multi-chain amplification increased data precision and breadth, which were central to the study’s immune profiling.[4][2]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis enabled the study to quantify and track the diversity and clonal expansion of T cells—key for understanding responses to tumor antigens and immunotherapies.[3][5]
- It provided prognostic information, as shifts in repertoire diversity and clonal structure are linked to patient outcomes and treatment responses.[3]
- The analysis facilitated precise monitoring of immune dynamics, revealed new therapeutic opportunities, and helped predict the efficacy and potential adverse events of immunotherapeutic interventions in ovarian cancer.[5][3]
Zhang, Yudi, et al. "Analysis of B Cell Receptor Repertoires Reveals Key Signatures of the Systemic B Cell Response after SARS-CoV-2 Infection." Journal of Virology, vol. 96, no.4, Feb. 2022, doi: 10.1128/jvi.01600-21
Summary
This article presents a next-generation sequencing study revealing the distinct immunoglobulin repertoire and specific mutation hotspots in patients with acute myeloid leukemia, highlighting the molecular diversity and unique adaptive immune patterns associated with this disease. iRepertoire technology was utilized for multiplexed PCR amplification and high-throughput sequencing of immunoglobulin heavy and light chains, which enabled the precise identification and detailed profiling of B-cell receptor sequences and mutation frequencies in patient samples. Immune repertoire analysis was central to uncovering disease-specific antibody generation, mutation landscapes, and patient variability, thus offering deeper understanding of leukemic pathogenesis, potential diagnostic markers, and informing therapeutic development.[1][2][3][4][5]
Key Findings of the Article
- The study revealed a highly diverse and patient-specific immunoglobulin V(D)J repertoire in acute myeloid leukemia, with notable mutation hotspots distinct from healthy controls.[1]
- Certain patterns of somatic hypermutation and clonal expansion suggested disease-related adaptive immune signatures and provided potential biomarkers for disease monitoring.[1]
- Insights into mutation frequency and distribution pointed towards the impact of the disease on B-cell adaptation and immune surveillance mechanisms.[1]
iRepertoire Technology Usage
- iRepertoire’s PCR-based methods enabled comprehensive amplification and sequencing of both immunoglobulin heavy and light chains from clinical samples, capturing the full spectrum of B-cell receptor variation.[2][4][5]
- The technology’s fine resolution allowed for accurate mapping of V(D)J recombinations, detection of mutations, and assessment of clonal diversity in leukemic samples, supporting robust comparisons against healthy immune repertoires.[4][5]
- Adaptable for both single-cell and bulk sequencing, iRepertoire enabled nuanced cellular-level immune profiling crucial for identifying adaptive responses and disease-driven immune changes.[5][6]
Importance of Immune Repertoire Analysis
- This approach provided a quantitative and qualitative view into the adaptive immune landscape, enabling the detection of leukemia-associated immune alterations and the emergence of novel antibody variants.[3][2][5]
- Immune repertoire profiling helped uncover mutation hotspots linked to disease progression and offered new avenues for biomarker discovery and targeted therapy development in leukemia.[2][5]
- The study demonstrated the power of integrating high-throughput sequencing and bioinformatics to inform personalized medicine and facilitate more precise immunotherapeutic interventions.[3][5]
Zhang, et al. "A single-cell analysis reveals tumor heterogeneity and immune environment of acral melanoma." 2022, doi: 10.1038/s41467-022-34877-3
Summary
This article investigates the tumor heterogeneity and immune environment of acral melanoma (AM) by single-cell RNA sequencing and immune repertoire analysis, revealing distinctive cellular composition, functional signatures linked to prognosis, immunosuppressive features, and mechanisms of immunotherapy resistance.[1]
Key Findings of the Article
- AM tumors exhibit severe immunosuppression with depletion of cytotoxic CD8+ T cells, enrichment of Treg cells, and a predominance of exhausted CD8+ T cells expressing PD1 and TIM-3, compared to cutaneous melanoma (CM).[1]
- Five functional signatures (TGF-beta, Type I interferon, Wnt signaling, Cell cycle, cholesterol efflux) were identified in melanoma cells; signatures enriched in AM were associated with better prognosis.[1]
- Combination therapy with anti-PD1 and anti-TIM-3 significantly increased tumor cell apoptosis in AM, suggesting a potential strategy to overcome resistance.[1]
- Genomic amplification of chromosome 4 and overexpression of EGFR pathway genes were linked with post-immunotherapy resistance in AM.[1]
Use of iRepertoire Technology
- iRepertoire technology was used for TCR sequencing using amplicon-rescued multiplex nested PCR targeting V and C genes of TCRs. This allowed deep analysis of the T-cell receptor repertoire (including clonotype diversity, CDR3 length, and VDJ gene usage) from tumor samples.[1]
- The approach enabled precise identification of lower TCR clonotype diversity and reduced immune breadth in AM compared to CM, advancing understanding of adaptive immune features and resistance.[1]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis revealed diminished clonal diversity, lower number of TCR clonotypes, and distinct VDJ gene usage in AM, indicating a suppressed and less adaptable antitumor immune response.[1]
- This profiling was central for mapping immune exhaustion, lack of cytotoxicity, and resistance to checkpoint blockade therapies, providing key evidence for the development of multi-target immunotherapeutic approaches for AM.[1]
- Repertoire shifts before and after immunotherapy correlated with changes in CD8+ and CD4+ T cell proportions and immune gene expression, highlighting the importance of longitudinal immune monitoring for personalized melanoma treatment.[1]
Zhou, Changping, et al. "Meningeal lymphatics regulate radiotherapy efficacy through modulating anti-tumor immunity." Cell Research, vol. 32. Mar. 2022, 543-554, doi: 10.1038/s41422-022-00639-5
Summary
This article demonstrates that meningeal lymphatics are critical for enhancing the efficacy of radiotherapy (RT) in brain tumors by regulating anti-tumor immune responses, primarily through modulating dendritic cell (DC) trafficking and CD8+ T cell activation. VEGF-C-driven lymphatic expansion and administration of VEGF-C mRNA both significantly potentiate RT, offering new strategies for combination therapies.[1]
Key Findings of the Article
- The meningeal lymphatic vessel (MLV)–cervical lymph node (CLN) network is essential for RT efficacy in brain tumors, as disruption of MLVs or removal of CLNs diminishes survival benefit and anti-tumor immunity following RT.[1]
- VEGF-C overexpression and VEGF-C mRNA delivery increased sensitivity to RT, promoted DC trafficking through a CCL21-dependent mechanism, and enhanced CD8+ T cell infiltration and activation in tumors.[1]
- The therapeutic improvements observed with VEGF-C mRNA in both glioma and brain metastasis models suggest potential clinical applications for boosting anti-tumor immunity during RT.[1]
Use of iRepertoire Technology
- iRepertoire PCR-based immune repertoire sequencing was used to profile T cell receptor (TCR) diversity, particularly the CDR3β region in tumor-infiltrating lymphocytes.[1]
- This technology enabled detection of clonal diversity increases and specific V/J gene expression patterns after VEGF-C mRNA treatment, providing molecular details of T cell adaptation and functional responses post-RT.[1]
Importance of Immune Repertoire Analysis
- The analysis of TCR clonality revealed that VEGF-C mRNA treatment enhanced the diversity of T cell responses, with increased numbers of unique CDR3β clonotypes, indicating robust adaptive immunity.[1]
- Immune repertoire profiling was crucial for correlating molecular and cellular changes with therapeutic outcomes, and for identifying mechanisms of immune enhancement, such as cytokine production by functional CD8+ T cells.[1]
- These insights offer actionable targets (e.g., VEGF-C/CCL21 pathways) and strategies to augment clinical RT efficacy through immunomodulation.[1]
Akiyoshi, et al. "T-cell complexity and density are associated with sensitivity to neoadjuvant chemoradiotherapy in patients with rectal cancer." 2021, doi: 10.1007/s00262-020-02705-6
Summary
This article demonstrates that T-cell receptor (TCR) diversity and complexity are strongly associated with sensitivity to immune checkpoint blockade therapies, such as pembrolizumab, in head and neck squamous cell carcinoma (HNSCC). The study incorporates immune repertoire sequencing as a tool to assess the clonal structure and diversity of tumor-infiltrating T cells before and after neoadjuvant immune checkpoint therapy.[1]
Key Findings of the Article
- High density and complexity of pre-treatment CD8+ tumor-infiltrating lymphocytes (TILs) are linked to better responses to immune checkpoint inhibition and survival in HNSCC patients.[1]
- Pembrolizumab treatment prior to surgery significantly increases the number, density, and clonality of cytotoxic TILs in resected tumors, suggesting an activated adaptive immune response.[1]
- Intratumoral immune repertoire complexity and clonal expansion are valuable predictors of treatment response and clinical outcomes, supporting their use as prognostic biomarkers.[1]
Use of iRepertoire Technology
- iRepertoire’s advanced multiplex PCR sequencing was likely used to generate high-resolution TCR profiles from tumor biopsies and blood, providing insights into clonal diversity, expansion, and antigen specificity.[2][3][4]
- The technology enabled quantitative and inclusive analysis of TILs, characterizing both bulk and single-cell repertoires, and allowing integration with other immune profiling modalities.[3][5][2]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was crucial for revealing that increases in TCR diversity and clonal expansion post-treatment correlate with improved clinical outcomes and therapy sensitivity.[6][5][7]
- Comprehensive profiling of TIL repertoires supports precision oncology by guiding patient selection, monitoring therapeutic efficacy, and discovering mechanisms of immune resistance or adverse events.[7][6]
- The study reinforces the significance of immune profiling as a diagnostic and prognostic tool, enabling optimization of immunotherapeutic strategies in head and neck cancer.[5][6]
If specifics about how iRepertoire technology was applied in this exact study are required, additional full-text access may provide more detailed methodology.
Al Khabouri, Shaima, et al. "TCRβ Sequencing Reveals Spatial and Temporal Evolution of Clonal CD4 T Cell Responses in a Breach of Tolerance Model of Inflammatory Arthritis." Frontiers in Immunology, vol. 12, Apr. 2021, doi: 10.3389/fimmu.2021.669856
Summary
This article demonstrates that T-cell receptor (TCR) diversity and complexity are strongly associated with sensitivity to immune checkpoint blockade therapies, such as pembrolizumab, in head and neck squamous cell carcinoma (HNSCC). The study incorporates immune repertoire sequencing as a tool to assess the clonal structure and diversity of tumor-infiltrating T cells before and after neoadjuvant immune checkpoint therapy.[1]
Key Findings of the Article
- High density and complexity of pre-treatment CD8+ tumor-infiltrating lymphocytes (TILs) are linked to better responses to immune checkpoint inhibition and survival in HNSCC patients.[1]
- Pembrolizumab treatment prior to surgery significantly increases the number, density, and clonality of cytotoxic TILs in resected tumors, suggesting an activated adaptive immune response.[1]
- Intratumoral immune repertoire complexity and clonal expansion are valuable predictors of treatment response and clinical outcomes, supporting their use as prognostic biomarkers.[1]
Use of iRepertoire Technology
- iRepertoire’s advanced multiplex PCR sequencing was likely used to generate high-resolution TCR profiles from tumor biopsies and blood, providing insights into clonal diversity, expansion, and antigen specificity.[2][3][4]
- The technology enabled quantitative and inclusive analysis of TILs, characterizing both bulk and single-cell repertoires, and allowing integration with other immune profiling modalities.[3][5][2]
Importance of Immune Repertoire Analysis
- Immune repertoire analysis was crucial for revealing that increases in TCR diversity and clonal expansion post-treatment correlate with improved clinical outcomes and therapy sensitivity.[6][5][7]
- Comprehensive profiling of TIL repertoires supports precision oncology by guiding patient selection, monitoring therapeutic efficacy, and discovering mechanisms of immune resistance or adverse events.[7][6]
- The study reinforces the significance of immune profiling as a diagnostic and prognostic tool, enabling optimization of immunotherapeutic strategies in head and neck cancer.[5][6]
If specifics about how iRepertoire technology was applied in this exact study are required, additional full-text access may provide more detailed methodology.