Abstract Background While highly efficacious for numerous cancers, immune checkpoint inhibitors (ICIs) can cause unpredictable and potentially severe immune-related adverse events (irAEs), underscoring the need to understand irAE biology. Methods We used a multidimensional approach incorporating single-cell RNA sequencing, mass cytometry, multiplex cytokine assay, and antinuclear antibody (ANA) profiling to characterize the peripheral immune landscape of patients receiving ICI therapy according to irAE development. Results Analysis of 162 patients revealed that individuals who developed clinically significant irAEs exhibited a baseline proinflammatory, autoimmune-like state characterized by a significantly higher abundance of CD57^+ T and natural killer (NK) T cells, plasmablasts, proliferating and activated CXCR3^+ lymphocytes, CD8^+ effector and terminal effector memory T cells, along with reduced NK cells and elevated plasma ANA levels. In irAE cases, we identified distinct baseline proinflammatory gene signatures, including markedly higher expression of IL1B and CXCL8 in monocytes and CXCR3, TNF, and IFNG in T/NK cells. TNF signaling was the most enriched pathway, while immunosuppressive genes SIGLEC7 and CXCR4 were downregulated. Following ICI initiation, these patients exhibited an enhanced shift toward an activated and inflammatory immune phenotype, including monocyte reprogramming characterized by upregulation of IL18 and elevated gene expression levels of CXCL10. Conversely, post-treatment levels of CXCL8 were decreased in irAE patients. Notably, in patients who did not develop clinically significant irAE, we identified increased baseline abundance of a TGFBI^high myeloid cluster enriched in immunosuppressive markers such as STAB1. In addition, patients without irAE exhibited upregulation of TNF and AIRE, accompanied by distinct myeloid protumorigenic reprogramming. Conclusions A pre-existing activated, autoimmune-like proinflammatory state drives the development of irAE during ICI therapy through three key axes: increased plasmablast/ANA, heightened interferon-gamma/CXCL10/CXCR3 axis, and amplified TNF signaling. These findings may serve as potential peripheral immune biomarkers for predicting irAE and provide biological insights into the mechanisms governing and mitigating irAE. Keywords: Immune related adverse event - irAE, Immunotherapy, Immune Checkpoint Inhibitor, Autoimmune, Antibody __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * After tens of thousands of patients have been treated with immunotherapy, the only established risk factor for immune-related adverse events (irAE) is pre-existing autoimmune disease. * While several studies have reported on immune parameters associated with irAE development, the underlying biology of irAEs remains elusive, as do clinically useful predictive biomarkers. WHAT THIS STUDY ADDS * This multiomic biomarker analysis identifies a preexisting activated, autoimmune-like proinflammatory state in patients who subsequently develop irAE. * Three key axes contribute to this irAE risk phenotype: increased plasmablasts/antinuclear antibody, heightened interferon (IFN)γ/CXCL10/CXCR3 axis, and amplified TNF signaling. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * Identification of the molecular mechanisms underlying irAE may serve as potential peripheral immune biomarkers for predicting irAE. * These findings also provide possible targets for specific therapeutic interventions affecting T cells, B cells, and IFN-mediated inflammation. Introduction Immune checkpoint inhibitors (ICIs) are highly effective therapies that have revolutionized the treatment of numerous cancers. However, in some cases, these agents cause autoimmune toxicities termed immune-related adverse events (irAEs). In contrast to the predictability and limited scope of chemotherapy and molecularly targeted therapy toxicities, irAEs may occur at any point during—and occasionally even well after—ICI therapy.[71]^1 2 They may affect almost any organ system, including potentially fatal cardiovascular and neurologic events in rare cases.[72]^3 In recent years, ICI indications have expanded from refractory, advanced malignancies to earlier-stage, curable tumors.[73]^4 At the same time, combination ICI regimens, which are associated with greater irAE occurrence and severity, have received approval for melanoma, kidney cancer, lung cancer, liver cancer, and mesothelioma.[74]^5 Taken together, these trends have only heightened the clinical importance of irAEs. Further complicating ICI administration are profound challenges in monitoring and diagnosing irAEs. Although irAEs may affect a plethora of organs, the rarity of certain types such as myocarditis renders longitudinal, extensive multiorgan monitoring costly and impractical. Indeed, expert guidelines generally limit routine surveillance to standard chemistries and thyroid function tests, which may detect kidney, liver, and certain endocrine toxicities but little else.[75]^6 7 Once irAEs do occur, accurate and timely recognition, evaluation, and treatment remain elusive. Like non-ICI-related autoimmune diseases—which may require incorporation of clinical history, physical examination findings, serologies, radiographic studies, and occasionally histologic sampling to render a diagnosis—it appears that irAEs are far more difficult to diagnose than chemotherapy-related or targeted therapy-related toxicities.[76]^8 Failure to detect and treat irAE quickly could result in high-grade events. Although lower-grade irAEs have been associated with ICI efficacy,[77]^9 10 the acute clinical deterioration caused by severe irAEs may overshadow any potential benefit.[78]^11 Given the expanding use of ICI therapy across cancer types and stages, the ability to predict and accurately diagnose irAEs represents a key clinical priority in the field of immuno-oncology. Several studies have reported on immune parameters associated with irAE development, including cytokines, chemokines, pre-existing or induced autoantibodies, T cell receptor (TCR) repertoire, immune cell subsets, and gut microbiomes.[79]12,[80]24 However, the underlying biology of irAEs remains elusive, as do clinically useful predictive biomarkers. To address these challenges, we performed systematic, longitudinal, and comprehensive multidimensional analysis incorporating single-cell RNA sequencing (scRNA-seq), mass cytometry (CyTOF), antinuclear antibody (ANA) profiling, and multiplex cytokine assays of longitudinal blood samples from ICI-treated patients. Materials and methods Study design, clinical data sources, and clinical specimen collection As described previously, we collected clinical data and longitudinal specimens from patients enrolled in an institutional prospective immunotherapy cohort. Eligible patients had any type of cancer planned for but not yet started on ICI (including anti-programmed death 1 (PD1), PD1 ligand (PDL1), and cytotoxic T lymphocyte antigen 4 (CTLA4)) monoclonal antibodies, administered as single-agent therapy or in combinations). At the time of case selection, anti-lymphocyte antigen 3 (LAG3) antibodies had not been Food and Drug Administration approved and were therefore not included in the patient sample. Due to challenges in determining the occurrence, type, timing, and severity of irAE,[81]^8 two oncologists (MSvI, DH) experienced in ICI administration and monitoring reviewed each case for irAE, with discrepancies reviewed and adjudicated by a third experienced clinician (DEG). As performed in other studies,[82]^10 25 we categorized irAE as clinically significant (Common Terminology Criteria for Adverse Events (CTCAE) grade≥2) or not clinically significant (CTCAE grade ≤1). Additional details on data collection, review, and characterization are provided in [83]online supplemental methods. Enrolled patients underwent initial blood collection at pretreatment baseline (BL) immediately prior to ICI therapy, after one treatment cycle (2–4 weeks), and at 6–8 weeks. We used 6–8 weeks as the main post-ICI initiation time point because[84]^1 (1) previous studies suggested that it provided more useful comparison with pretreatment BL than do earlier post-ICI initiation time points,[85]^12 and[86]^2 mosti(2) rAEs occur after this time,[87]^10 26 thereby rendering this time point potentially useful for prediction of future events. Blood samples were centrifuged at 3,000 rpm at 4°C for 15 min to obtain plasma and isolate peripheral blood mononuclear cells (PBMCs). For the selection of cases for analysis by varying laboratory methodologies, we used the following guiding principles: (1) including the greatest number of cases possible within financial constraints; (2) maximizing overlap of cases across methodologies, and (3) minimizing overlap of cases when validating across methodologies. Single-cell RNA sequencing and data analysis We produced scRNA-seq data on a total of ∼400,000 cells across 72 samples. All 72 PBMC patient samples in this study were processed for scRNA-seq on a 10X Chromium instrument. Integrating 72 samples for batch correction to identify shared cell states across samples was performed using Harmony. We then performed normalization and dimensionality reduction analysis using SCTransform on our integrated dataset to eliminate any technical variation and identify different cell states. We performed unsupervised clustering and identified 22 different cell clusters in our dataset across 72 samples. Unsupervised clustering of scRNA-seq data was performed using Seurat packages in R, Cellenics and dseqr. We further used the Azimuth PBMC dataset as a reference to identify immune cell subsets at a much higher resolution using the top 30 principal components and performed pseudobulk differential gene expression analysis using dseqr with false discovery rate (FDR) <0.05. We further performed additional unsupervised sub-clustering analysis of B cells, T/natural killer (NK) and myeloid cells. Additional details on scRNA-seq and data analysis are provided in [88]online supplemental methods. Cytokine/chemokine and ANA analysis Determination of cytokine and chemokine levels was performed using Bio-Plex Pro Human Chemokine 40-plex Panel (Bio-Rad Laboratories, Hercules, California, USA) according to the manufacturer’s instructions using a Luminex 200 System. The list of cytokines and chemokines is provided in [89]online supplemental table 1. Bio-Plex Manager V.6.1 software was used for data analysis. Concentrations of cytokines and chemokines (pg/mL) were determined based on the fit of a standard curve for mean fluorescence intensity versus pg/mL. We assessed the plasma levels of antinuclear IgG antibodies using QUANTA lite ANA assay (INOVA diagnostics). All plasma samples were run in duplicates, and analysis was performed using GraphPad Prism. Results are expressed as the mean±SE. Mass cytometry A panel of 35 antibodies (metal isotope-labeled conjugates, Maxpar Direct Immune Profiling Assay Panel by Fluidigm) was used for staining Cryopreserved PBMCs derived from patients receiving ICI therapy. Cells were analyzed on a Helios mass cytometer (Fluidigm). Data were normalized and analyzed with gating on live, singlet CD45^+ cells using the OptSNE and FlowSOM using OMIQ.ai (Omiq, Santa Clara, California, USA). We then performed unsupervised consensus meta-clustering using Euclidean as a distance metric and Max Equal sampling of all 110 samples. Statistical analysis of cell abundance was performed using EdgeR in OMIQ or GraphPad Prism software. CyTOF data was also analyzed using a manual gating strategy. Statistical analysis Statistical significance was calculated using IBM SPSS Statistics software (V.29) and GraphPad Prism Software (GraphPad Software, La Jolla, California, USA). Student’s t-test, Welch’s t-test, the Mann-Whitney U test, Wilcoxon matched-pairs signed-rank test, and Edge R were used when applicable. For toxicity analyses, because irAEs may occur throughout the course of treatment with ICI, patients were considered not to have developed irAEs only if they had been followed without evidence of toxicity for at least 6 months. Results are expressed as the mean±SE. To perform multimarker analysis, we conducted logistic regression analysis using the ‘glm’ function, and calculated the area under the receiver operating curve using the ‘pROC’package in R (V.4.3.2) programming. Spearman correlation analysis and calculation of Spearman rank-weighted composite scores were performed in R using dplyr, pROC and ggplot packages. Unless specified, for all statistical analysis, the level of significance was set at p<0.05. For all gene expression analysis results presented the level of significance was set to adjusted p<0.05 and minimum fold change ≥1.5. Results Clinical characterization of patients and study design A total of 162 patients were included in the study. [90]Figure 1 outlines the overall experimental strategy and patient cohorts included in each analysis, including cytokines/chemokines (n=146), ANA (n=65), CyTOF (n=55), and scRNA-seq (n=36). The overall numbers and selection of cases for each assay reflect cost/resources, specimen availability, and an effort to maximize the overlap of cases across assays. [91]Table 1 provides demographic, cancer, and treatment information for included cases, of which 93 (57%) had grade ≥2 irAE. Figure 1. Study design. The study included 162 patients with cancer receiving ICI therapy (either anti-PD1, anti-PDL1 or combination anti-PD1 plus anti-CTLA4). Blood was collected at baseline before the initiation of ICI therapy and post-immunotherapy (2–3 weeks and 6–8 weeks). PBMCs were used for single-cell RNA sequencing (scRNA-seq) and mass cytometry assays. Plasma was used to assess levels of antinuclear antibodies (ANA) and 40 cytokines. The number of patients used for each assay is shown in the brackets. BL, baseline; CTLA4, cytotoxic T lymphocyte antigen 4; ICI, immune checkpoint inhibitor; PBMC, peripheral blood mononuclear cell; PD1, programmed death 1; PDL1, programmed death ligand 1; t-SNE, t-distributed stochastic neighbor embedding. [92]Figure 1 [93]Open in a new tab Table 1. Characteristics of patients included in the study (N=162). Characteristic Number (%) Sex  Female 58 (36)  Male 104 (64) Race—ethnicity  Asian 4 (2)  Black or African American 12 (7)  Hispanic white 12 (7)  Non-Hispanic white 126 (78)  Unknown 8 (5) Cancer type  Non-small cell lung cancer 81 (50)  Melanoma 38 (23)  Head and neck 11 (7)  Kidney 9 (6)  Other[94]^* 23 (14) ICI therapy  PD1/PDL1 136 (84)  CTLA4 1 (1)  PD1/PDL1+CTLA4 21 (13)  Other PD1/PDL1 combinations[95]^† 4 (2) [96]Open in a new tab ^* Includes breast cancer (n=1), cholangiocarcinoma (n=2), cutaneous squamous cell carcinoma (n=1), glioblastoma multiforme (n=2), hepatocellular carcinoma (n=2), mesothelioma (n=3), pancreas cancer (n=2), sarcoma (n=1), small cell lung cancer (n=6), and urothelial/bladder cancer (n=2). ^† Includes CSF1R (n=3) and NKG2A (n=1). CTLA4, cytotoxic T lymphocyte antigen 4; ICI, immune checkpoint inhibitor; PD1, programmed death 1; PDL1, programmed death ligand 1. Baseline abundance of immune cell subsets associated with irAEs We performed 5' scRNA-seq using the 10X chromium immune profiling assay on paired BL and 6–8 week samples from 40 patients, of whom 30 had experienced various types of grade ≥2 irAE. After applying quality control filters, 36 matched sample pairs (BL and 6–8 weeks) yielded high-quality transcriptomes, of which four were technical replicates (1 no-irAE and 3 irAE) ([97]figure 2A). Following data integration, normalization, and dimensionality reduction, an initial unsupervised clustering analysis at a lower resolution identified 23 distinct cell clusters across samples. These clusters represented all major immune cell lineages ([98]figure 2B). The proportion of these 23 unsupervised Louvain clusters is shown in [99]figure 2C, and the expression of canonical marker genes is presented in [100]figure 2D. Cell annotations based on the expression of top five marker genes and a heat map of top marker genes for each unsupervised cluster are provided in [101]online supplemental figures 1 and 2. Figure 2. Single-cell RNA sequencing (scRNA-seq) analysis of PBMCs at baseline and post-ICI therapy. (A) scRNA-seq experimental design for 72 blood samples (36 pairs) from patients with cancer at baseline (BL) before the initiation of ICI therapy and 6–8 weeks after ICI initiation. (B) UMAP of 23 cell states identified by unsupervised clustering shown according to cell type. (C) Frequency plot showing the proportion of 23 clusters identified by unsupervised clustering in irAE versus no-irAE group at BL and 6–8 weeks post-ICI therapy. (D) Dot plot showing canonical marker gene expression for 23 unsupervised clusters. Arrows point to cluster 2 (NK) and cluster 18 (Mki67+). (E) The abundance of cluster 18 (Mki67+), shown as a percentage of total PBMCs in two groups at baseline (BL) identified by unsupervised clustering. (F) UMAP of PBMCs annotated by Azimuth Reference shown by major immune cell types and 30 immune cell subsets. The abundance of NK (CD56dim) cells (G), CD4 proliferating T cells (H), plasmablasts (I), and combined proliferating lymphocyte cluster (CD4, CD8 and NK) (J) shown as a percentage of total PBMCs in No irAE versus irAE group at BL guided by Azimuth Reference. ASDC, AXL+SIGLEC6+ dendritic cell; cDC, conventional dendritic cell; CTLA4, cytotoxic T lymphocyte antigen 4; DC, dendritic cell; dnT, double-negative T cell; gdT, gamma delta T cell; HSPC, hematopoietic stem and progenitor cell; ICI, immune checkpoint inhibitor; irAE, immune-related adverse event; MAIT, mucosal-associated invariant T cell; NK, natural killer cell; PBMC, peripheral blood mononuclear cell; PD1, programmed death 1; pDC, plasmacytoid dendritic cell; PDL1, programmed death ligand 1; TCM, central memory T cell; TEM, effector memory T cell; Treg, T regulatory cell; UMAP, uniform manifold approximation and projection. [102]Figure 2 [103]Open in a new tab Frequency calculations as a percentage of total PBMCs revealed a significant increase in the abundance of proliferating lymphocyte cluster (cluster 18, characterized by Mki67^+ cells) ([104]figure 2E). Conversely, the NK cell cluster (cluster 6) was reduced in irAE patients. In addition, cluster 22, which resembled plasma cell/plasmablast-like clusters showed increased frequency in irAE patients at BL. To confirm these findings, we performed a reference-guided cell annotation analysis with Azimuth on the integrated datasets of 72 patient samples ([105]figure 2F). Consistent with the unsupervised clustering results, we identified three cell clusters significantly correlated with the irAE development. The NK cell cluster—specifically CD56^dim CD16^+ NK cells—was significantly less abundant at BL in patients with irAE, whereas CD4 proliferating T cells and plasmablasts were significantly more abundant ([106]figure 2G–I). When combining Azimuth-guided CD4, CD8, and NK proliferating clusters, the BL aggregated proliferating lymphocyte cluster was most strongly correlated with irAE ([107]figure 2J), mirroring the unsupervised clustering data and demonstrating an activated immune state in patients predisposed to irAE. Next, we evaluated whether the BL abundance of this proliferating lymphocyte cell cluster might serve as a candidate biomarker. Receiver operating characteristic (ROC) analysis yielded an area under the curve (AUC) of 0.78 (p<0.0001) for the CD4 proliferating cluster, indicating its potential clinical utility in predicting future irAE occurrence. Mass cytometry analysis reveals distinct immune subsets associated with irAEs We performed CyTOF analysis on PBMC samples from an expanded cohort of 55 patients (43 irAE, 12 no‐irAE) with paired BL and 6–8 weeks post-ICI time points using a panel of 36 antibodies ([108]figure 3A). Metaclustering analysis yielded 30 distinct immune cell clusters—including subsets of B cells, CD4^+ T cells, CD8^+ T cells, NK cells, and myeloid cells ([109]figure 3B,C). A corresponding heat map of cell surface marker expression is shown in [110]figure 3D. Figure 3. Mass cytometry (CyTOF) analysis of PBMC samples at baseline and post-ICI therapy. (A) CyTOF experimental design of 110 blood samples (55 pairs) from 55 patients with cancer (n=12 no-irAE and n=43 irAE) at baseline (BL) before the initiation of ICI therapy and 6–8 weeks post immunotherapy. Opt-SNE of 30 cell clusters identified by unsupervised consensus meta-clustering of CYTOF data using Euclidean as a distance metric is shown according to major immune cell types (B) and 30 immune cell subsets (C). (D) Heatmap showing expression of 35 cell surface immune markers used in the CyTOF panel in 30 metaclusters. (E) Volcano plot of CyTOF data showing significant metaclusters (aqua circles) differentially abundant in irAE versus no-irAE at baseline with (p<0.05). The abundance of significant clusters; K10, plasmablast (F); K2, NK (G); K4, CD8 terminal effector (H); K8, TCRγδT (I) and K13, CD8 terminal effector (J), identified by unsupervised clustering of CyTOF data using Edge R (p<0.05) shown as a percentage of total PBMCs in no-irAE versus irAE cases at BL. The abundance of significant clusters, including NKT (K) and CD57 positive clusters (L) identified by manual gating strategy in irAE versus no-irAE cases. (M) The abundance of CD8+ KLRG1+ CCR7 CD8 effector memory cluster identified by unsupervised T/NK subclustering analysis of scRNA-seq data. Significant increase in abundance of cluster K21 (CD4 memory) in irAE and no-irAE cases postimmunotherapy in all cases (N) and in melanoma cases (O) by Wilcoxon matched-pairs signed-rank test of CyTOF data. CTLA4, cytotoxic T lymphocyte antigen 4; FC, fold change; FDR, false discovery rate; ICI, immune checkpoint inhibitor; irAE, immune-related adverse event; NK, natural killer cell; PBMC, peripheral blood mononuclear cell; PD1, programmed death 1; pDC, plasmacytoid dendritic cell; PDL1, programmed death ligand 1; TCM, central memory T cell; TCR, T cell receptor; scRNA-seq, single-cell RNA sequencing. [111]Figure 3 [112]Open in a new tab We next calculated the abundance of each cell cluster as a percentage of total CD45^+ cells. EdgeR analysis comparing irAE and no‐irAE groups at BL identified 11 significant clusters in OMIQ with p<0.05 ([113]figure 3E). Specifically, seven clusters (K4-CD8, K8-TCRγδ, K9-B cell, K10-plasmablast, K13-CD8, K17-TCRγδ, K25-CD8) were increased in irAE patients, whereas four clusters (K2-NK, K16-CD123^+, K28-CD8, K29-CD4) were decreased. Consistent with our scRNA-seq findings, plasmablasts (cluster K10) were elevated (p=0.01) while NK cells (cluster K2, CD56^+CD57^−) were reduced (p=0.007) in the irAE group ([114]figure 3F,G). Further stratification by cancer and therapy type revealed that clusters K4, K8, and K13 exhibited the most significant differences in irAE patients ([115]figure 3H–J). Clusters K4 (CD8^+CCR7^−CD45RA^+CD27^−CD28^−CD57^+) and K13 (CD8^+CCR7^−CD45RA^+CD27^−CD28^−CD57^−) resembled terminal effector memory CD8^+ T cells, while cluster K8 comprised double-negative TCRγδ^+CD57^+ cells. Extended CyTOF analyses by therapy and cancer type are presented in [116]online supplemental figure 3 (non-small cell lung cancer, melanoma, and PD1/PDL1 therapy). Although the limited number of patients receiving combination immunotherapy precluded detailed analysis of that subgroup, the directionality and magnitude of subgroup findings largely mirrored those of the overall cohort shown in [117]figure 3. For additional rigor, we performed manual gating analysis of the CyTOF data. We identified a greater BL abundance of the NKT cell population ([118]figure 3K) and a striking increase in CD57^+ T cells, including CD57^+CD8^+ and CD8^+CD57^+ NKT populations in irAE patients ([119]figure 3L). The manual gating strategy is shown in [120]online supplemental figure 4. Of note, the two highly correlated clusters identified by metaclustering were CD8 terminal effector cell cluster K4 and TCRγδ cluster K13, which were both CD57^+. As reported by other groups,[121]^13 we also observed a greater abundance of HLA-DR^+CD38^+CD4^+ memory T cells in irAE patients at BL. To validate the increase in CD8^+ effector cells in scRNA-seq data, we performed high-resolution subclustering of CD8^+ and NK cells, which revealed a significantly higher abundance of CD8^+KLRG1^+CCR7^− effector memory clusters in irAE patients ([122]figure 3M). Collectively, our CyTOF analysis at BL revealed a T cell profile skewed toward an activated effector/terminal effector phenotype (CD57^+), increased plasmablasts, and reduced CD56^dim CD16^+ NK cells in patients with irAEs. Next, we investigated treatment-induced changes by comparing the abundance of metaclusters between irAE and no‐irAE groups at 6–8 weeks post-ICI initiation. Following ICI initiation, the abundance of CD8^+ effector clusters (K4 and K13) and TCRγδ cluster K8 remained significantly higher in irAE patients, whereas the abundance of NK cell cluster K2 remained significantly lower. CD57^+ T and NKT cell populations remained significantly higher in the irAE group post-therapy. Using the Wilcoxon matched-pairs signed-rank test, we further assessed longitudinal changes in the abundance of CyTOF metaclusters relative to BL within each group. This analysis revealed a significant increase in the CD4^+ memory cluster (K21, CD4^+CD45RO^+CD45RA^−CD27^+CD28^+CD57^−) exclusively in irAE cases (p=0.003 vs p=0.38 in no‐irAE), particularly in patients with melanoma ([123]figure 3N,O). After ICI initiation, several populations decreased including memory B cells (cluster K6), exhausted T cells (CD57^+PD1^+CD4^+ cluster K22 and CD57^+PD1^+CD8^+ cluster K5), NKT cells (CD8^+CD161^+CD57⁻ cluster K28), and an intermediate naïve/memory CD8^+ cluster (K18) ([124]online supplemental figure 5A–E). Interestingly, these decreases were significant only in irAE cases, suggesting an enhanced activated state post-therapy. In summary, our CyTOF analysis revealed an activated and skewed T cell compartment in irAE patients at BL characterized by an effector/terminal effector phenotype with increased plasmablasts and reduced NK cells. Baseline proinflammatory autoimmune-like state associated with irAEs Because our scRNA-seq and CyTOF analyses revealed a high BL abundance of plasmablasts in patients with irAEs, we further examined the B cell compartment by performing differential gene expression and pathway analysis between irAE and no-irAE groups for naïve and memory B cells but observed no significant changes in major pathways before or after therapy. Consistent with earlier work by Das and colleagues,[125]^17 we also observed a significant increase in plasmablasts in patients with irAEs treated with combination (CTLA4+PD1) therapy. Additionally, we found that increased BL abundance of plasmablasts was most strongly associated with high-grade (grade ≥3) irAEs ([126]figure 4A). Figure 4. Baseline autoimmune-like, proliferative and inflammatory state associated with irAE development. (A) Abundance of plasmablasts as a percentage of total PBMCs at baseline (BL) according to the grade of irAE. (B) Comparison of plasma concentration of antinuclear antibodies (ANA) (N=65 total; n=22 no-irAE and n=43 irAE) at BL and postimmunotherapy (at 2–4 weeks and 6–8 weeks). (C) Comparison of plasma concentration of ANA in 65 patients based on irAE grade at BL and postimmunotherapy. G0-1, Grade 0–1; G2, Grade 2; G3, Grade 3. (D) Abundance of CXCR3+Mki67+ cell cluster as a percentage of total PBMCs at BL based on the grade of irAE. (E) Pseudobulk scRNA-seq differential gene expression analysis of T and NK cells in the irAE group compared with the no-irAE group at baseline is shown as a volcano plot. Significantly upregulated genes are shown in blue dots, significantly downregulated genes in red dots, and genes with no significant change are shown in gray (F) Heat map depicting logFC and p values for the 3-gene signature in T and NK cells comparing irAE versus No irAE group at baseline (adjusted p<0.05 and ≥2 fold change). (G) MsigDB pathway enrichment analysis for T/NK cells at baseline. Red arrow shows the TNFα signaling as the highly enriched pathway in irAEs at baseline. (H) GSEA enrichment analysis for T/NK cells showing TNF signaling as the most enriched pathway at baseline. Abundance of CXCR3+Mki67+ cluster (I) and CXCR3+CD4+ memory cluster (J) identified by high-resolution unsupervised clustering of scRNA-seq data. (K) Abundance of CXCR3+CD8+ memory clusters in the scRNA-seq data at baseline. (L) CXCR3 gene expression in T and NK cells at baseline. Abundance of CXCR3+CD8+ clusters (M) and CXCR3+CD4+ (N) memory clusters at baseline identified in the CyTOF data. FC, fold change; GSEA, gene set enrichment analysis; irAE, immune-related adverse event; NK, natural killer cell; PBMC, peripheral blood mononuclear cell; scRNA-seq, single-cell RNA sequencing. [127]Figure 4 [128]Open in a new tab Given these findings, we next assessed plasma ANA levels—a non-specific clinical biomarker for systemic lupus erythematosus and other autoimmune diseases[129]^27—in 65 patients across time points ([130]figure 4B). BL ANA levels were significantly higher in patients who subsequently developed irAEs compared with those without irAEs, and levels were highest for grade ≥3 irAE cases at all time points ([131]figure 4C). To determine whether this autoimmune-like state correlates with a more proliferative and proinflammatory phenotype, we performed a grade-based analysis of the proliferating lymphocyte cluster (Mki67^+) identified in the scRNA seq data. This highly proliferative lymphocyte cluster, which was CXCR3^+HLADR^+, was most significantly associated with higher-grade irAEs ([132]figure 4D). Next, we evaluated gene expression changes in T and NK cells at BL. Pseudobulk differential analysis between irAE and no irAE cases revealed significant changes (adjusted p<0.05 and fold change ≥1.5) in over 300 genes at BL ([133]figure 4E). Notably, IFNG, TNF, and CXCR3 were among the top upregulated genes in patients who developed irAEs ([134]figure 4F). Gene set enrichment analysis (GSEA) and MsigDB Pathway enrichment identified TNF signaling as the most significantly upregulated pathway in T cells of irAE patients at BL ([135]figure 4G,H). In contrast, genes associated with T cell inhibition and immunosuppression (CXCR4 and SIGLEC7) were downregulated in irAE patients. Interestingly, the autoimmune regulator (AIRE) gene was significantly upregulated (fold change >2; adjusted p=0.04) after ICI treatment only in the no irAE group, suggesting a potential role in enhanced self‐tolerance and prevention of irAEs. Because CXCR3 is a homing receptor for CXCL9, CXCL10, and CXCL11 and is expressed on activated/effector T cells with a propensity to migrate to inflammatory sites, we examined our scRNA-seq data at a higher resolution and performed additional manual gating of the CyTOF data. This higher-resolution unsupervised analysis of PBMC scRNA-seq replicated our earlier finding of an increased Mki67^+CXCR3^+HLADR^+cells in irAE patients and also identified a higher abundance of the CXCR3^+CD4^+IL32^+ memory cluster associated with irAEs, thereby supporting the increased CXCR3 gene expression data ([136]figure 4I,J). Subclustering analysis of CD8/NK/Mki67 clusters also showed an increased abundance of activated CXCR3-expressing CD8 memory clusters expressing CXCR3 in the irAE patients at BL ([137]figure 4K). An overall increase in CXCR3 gene expression in scRNA-seq data at BL is depicted in [138]figure 4L. Manual gating of T cell subsets in the CyTOF data likewise revealed a higher abundance of CXCR3^+CD4^+ and CXCR3^+CD8^+ effector memory cells in irAE patients at BL ([139]figure 4M,N). Distinct features of myeloid cells and cytokine/chemokines associated with irAEs Unsupervised clustering analysis of myeloid cells in PBMC scRNA-seq data ([140]figure 2B) revealed eight distinct myeloid subsets (Clusters 6, 9, 10, 11, 13, 17, 19, and 21) defined by the expression of canonical myeloid markers. These included six monocyte clusters—two CD16^+ clusters (10 and 19), three CD14^+ clusters (6, 9, and 13), one intermediate monocyte cluster,[141]^11 a plasmacytoid dendritic cell (DC) cluster[142]^21 and a conventional DC cluster.[143]^17 Pseudobulk differential single-cell gene expression analysis revealed significant changes in over 250 genes (adjusted p<0.05 and fold change ≥1.5), including upregulation of IL1B and CXCL8 and downregulation of the immunosuppressive gene SIGLEC-10 and complement genes C1QA, C1QB, and C1QC in irAE patients at BL ([144]figure 5A). GSEA and MsigDB pathway enrichment identified TNF signaling and inflammatory response as the two significantly upregulated pathways in irAE patients at BL ([145]figure 5B,C). A heatmap of selected upregulated genes (adjusted p<0.05 and fold change ≥1.5) involved in TNF signaling and inflammatory responses is shown in [146]figure 5D. Figure 5. Single-cell RNA sequencing analysis of myeloid cells in patients with and without irAE. (A) Pseudobulk differential expression analysis of unsupervised myeloid cell clusters of scRNA-seq data in irAE versus no-irAE group at baseline is shown as a volcano plot (adjusted p<0.05 and fold change ≥1.5). Significantly upregulated genes are shown in blue dots, significantly downregulated genes in red dots, and genes with no change are shown in gray. Gene set enrichment analysis (GSEA) in panel B and MsigDB pathway enrichment in panel C of myeloid cells at baseline in irAE patients. The red arrow in panel C shows the TNF signaling and inflammatory response pathways as highly enriched in myeloid cells in the irAE group at baseline. (D) Heatmap of selected upregulated genes in myeloid cells in irAE versus no-irAE at baseline (adjusted p<0.05 and fold change ≥1.5). (E) Gene expression changes of CXCL10, CCL3, CCL4, SPARC and SIGLEC1 in myeloid cells in irAE versus no-irAE cases post-therapy (adjusted p<0.05 and fold change ≥1.5). (F) Gene expression changes of TNF and IRF4 in monocytes in no-irAE cases post-therapy compared with baseline (adjusted p<0.05 and fold change ≥1.5). (G) Heatmap of selected upregulated and downregulated genes in Azimuth-guided CD14+ classical monocytes in irAE versus no-irAE at baseline (adjusted p<0.05 and fold change ≥1.5). (H) Gene Ontology analysis in CD14+ classical monocytes in irAE versus no-irAE at baseline. (I) Heatmap of selected upregulated genes in Azimuth-guided CD14+ and CD16+ classical monocytes in irAE versus no-irAE at post-therapy (adjusted p<0.05 and fold change ≥1.5). (J) Heatmap of selected upregulated genes in Azimuth-guided CD14+ and CD16+ classical monocytes in no-irAE at post-therapy compared with baseline (adjusted p<0.05 and fold change ≥1.5). (K) Unsupervised subclustering analysis of all myeloid cells (without pDCs). (L) Dot plot showing gene expression markers for 16 unsupervised Louvain clusters for myeloid cells. (M) Significantly upregulated cytokines/chemokine ligands and their receptors at baseline in irAE patients compared with no-irAE patients at baseline (adjusted p<0.05 and fold change ≥1.5). (N) Trajectory analysis of myeloid cells using Monocle 3. (O) Continuous embedding depicting the fraction of myeloid cells expressing CXCL8 and IL1B genes and their corresponding gene expression changes between irAE and no-irAE patients at baseline in myeloid cells. (P) Abundance of TGFBIhigh cluster 13 in irAE versus no-irAE patients at baseline shown as a percentage of total myeloid cells. BL, baseline; FC, fold change; IFN, interferon; irAE, immune-related adverse event; pDC, plasmacytoid dendritic cell; scRNA-seq, single-cell RNA sequencing; UMAP, uniform manifold approximation and projection. [147]Figure 5 [148]Open in a new tab At the post-ICI initiation time point, we observed significant upregulation of chemokine genes (CXCL10, CCL3, and CCL4) in CD16^+ monocytes from irAE patients, whereas the immunosuppressive gene SPARC was downregulated ([149]figure 5E). Intriguingly, TNF was significantly upregulated at 6–8 weeks post-therapy compared with BL exclusively in no irAE patients ([150]figure 5F). To further validate these findings and elucidate the role of monocytes, we performed an Azimuth reference-guided analysis of peripheral blood monocytes, which identified two clusters: CD14^+ classical monocytes and CD16^+ non-classical monocytes. Differential single-cell gene expression analysis between irAE and no‐irAE cases in these monocyte clusters revealed significant changes in over 250 genes in classical monocytes at BL, including upregulation of proinflammatory genes IL1B, TNF, and CXCL8. TNF signaling emerged as the top enriched pathway in irAE patients, mirroring our unsupervised clustering results. Gene Ontology analysis further showed significant upregulation of genes involved in inflammatory responses, leukocyte/monocyte chemotaxis, and migration in CD14^+ monocytes, while type I interferon (IFN) signaling and complement genes were downregulated in irAE patients at BL ([151]figure 5G,H). Post-ICI initiation, we detected over 450 differentially expressed genes in classical monocytes and more than 150 genes in non-classical monocytes, including upregulation of several chemokines involved in recruiting T cells and monocytes to sites of organ inflammation (among them CCL3, CCL4, and CXCL10) in irAE patients following ICI treatment. A heatmap depicting significant gene expression between irAE and no irAE patients at BL is shown in [152]figure 5I. Notably, patients without irAE displayed elevated expression of a distinct set of genes involved in macrophage migration/chemotaxis post-ICI treatment including SLAMF8, CCR2, MST1, PTK2, C5, and CCL5 in CD14^+ monocytes, and TNF, HES1, CCL5, KLF13, and NOTCH1 in CD16^+ monocytes, indicative of a pro-tumorigenic myeloid reprogramming ([153]figure 5J). To gain a deeper understanding of myeloid cell involvement in irAEs, we performed higher-resolution unsupervised subclustering analysis of myeloid cells, which revealed 16 distinct clusters ([154]figure 5K,L). As before, the irAE patients showed significant BL upregulation of IL1B and CXCL8, with increased CCL3, CCL4, and CXCL10 post-therapy. Examining all cytokines/chemokine ligands and their receptors at BL revealed IL1B, CCL3L1, CCR2, CXCL2, CXCL3, CXCL8, IL6R, IL6ST, TNFSF14 and TNFRSF10D significantly upregulated in irAE patients ([155]figure 5M). [156]F[157]igure 5N–[158]P displays trajectory analyses, which indicated that (1) CD14^+ monocytes differentiate into three distinct branches: DC-like (C6), monocyte/macrophage-like (C5, C13), and the most differentiated CD16^+CD14^− state (C0, C14, C15); (2) differential expression of IL1B and CXCL8 in myeloid clusters according to irAE development; and greater abundance of the TGFBI^high cluster (C13) in no-irAE patients. Given this enhanced cytokine and chemokine gene expression profile in irAE patients, we next evaluated plasma levels of 40 cytokines and chemokines using a multiplex cytokine panel in 146 patients at three time points: pretreatment BL, 2–3 weeks, and 6–8 weeks post-ICI initiation. As previously reported by our group and others,[159]^12 28 IFNγ-inducible chemokines CXCL9 and CXCL10 were elevated in both irAE and no-irAE groups following ICI therapy relative to BL. However, irAE patients showed a significantly greater increase in plasma CXCL10 at 6–8 weeks compared with the no-irAE group ([160]figure 6A). Similarly, CXCL9 levels displayed a more pronounced fold induction in irAE patients at 6–8 weeks ([161]figure 6B). This heightened induction of CXCL9 and CXCL10 was observed in patients receiving both single-agent and combination therapy ([162]figure 6C–F). In contrast, serum CXCL8 levels at 6–8 weeks post-ICI were significantly lower in irAE patients than in those without irAEs ([163]figure 6G–I). Notably, changes in CXCL8, CXCL9, and CXCL10 in serum reflected their respective gene expression levels in myeloid cells ([164]figure 6J), implying that circulating monocytes may partially account for the source of these serum cytokines. Figure 6. Multiplex cytokine and chemokine analysis in patients with and without irAEs. Comparison of plasma concentrations of chemokines at baseline (BL) and postimmunotherapy (6–8 weeks). CXCL10 (A) and CXCL9 (B) in all patients. CXCL10 (C) and CXCL9 (D) in PD1/PDL1-treated patients. CXCL10 (E) and CXCL9 (F) in combination (CTLA4+PD1)-treated patients. CXCL8 in all patients (G), in PD1/PDL1-treated patients (H) and in combination (CTLA4+PD1)-treated patients (I). Dot plot showing comparison of relative gene expression values of chemokines CXCL10, CXCL9 and CXCL8 at baseline (BL) and postimmunotherapy (6–8 weeks) in myeloid cells in scRNA-seq data (J). Number of patients for comparison of plasma concentration of chemokines: All patients (N=146 total: n=79 irAE and n=67 no-irAE); PD1/PDL1-treated patients (N=122 total: n=59 No irAE and n=63 irAE); patients treated with combination CTLA4+PD1 therapy (N=18 total: n=6 No irAE and n=12 irAE). CTLA4, cytotoxic T lymphocyte antigen 4; irAE, immune-related adverse event; PD1, programmed death 1; PDL1, programmed death ligand 1; scRNAseq, -seq, single-cell RNA sequencing. [165]Figure 6 [166]Open in a new tab Multimarker analysis For both scRNA-seq and CyTOF, we integrated the individual markers with strongest association with irAE and performed logistic regression analysis. Because only 17 patients had both scRNA seq and CyTOF data, we were not able to combine both datasets into a single multimarker analysis. Incorporating (1) CD4-CD8-NK proliferating, (2) NK, and (3) plasmablast markers, the scRNA seq multimarker had an AUC of 0.78 ([167]online supplemental figure 6A). Incorporating CD57+NKT, CD57+CD4, CD57+CD8, K4 (CD8^+CCR7^−CD45RA^+CD27^−CD28^−CD57^+), K8 (double-negative TCRγδ^+CD57^+), and K13 (CD8^+CCR7^−CD45RA^+CD27^−CD28^−CD57^−) populations, the CyTOF multimarker had an AUC of 0.79 ([168]online supplemental figure 6B). For 17 patients that had both ScRNA-seq and CyTOF datasets, we performed Spearman correlation analysis and ranked the immune variables by their Spearman correlation with irAE development. We then calculated Spearman rank-weighted composite score using the immune features that showed the strongest associations with irAEs and ran AUC/ROC analysis. Spearman rank-weighted composite scores combining CD4-CD8-NK proliferating, NK, plasmablast and CD57NKT markers showed AUC of 0.865 and a p value of 0.03 ([169]online supplemental figure 6C,D). Validation in external datasets We accessed publicly available external datasets by Lozano et al and Nuñez et al[170]^13 29 to validate our scRNA seq and CyTOF findings, respectively. Interpreting these analyses requires consideration of their features and limitations. The Lozano dataset is small (n=13), limited to melanoma cases, and observed irAE only in patients that received combination ICI. The Nuñez dataset lacks PBMC scRNA seq data and lacks a CD20 marker in the CyTOF panel (thereby precluding identification of plasmablasts). For scRNA-seq markers, we validated the association of lower NK cells, higher CD4 proliferating cells, and higher CD8 terminal effector cells at BL in patients with severe (grade ≥3) irAEs ([171]online supplemental figure 7A–C). For CyTOF, we validated the association of higher TCRγδ T cells, higher CD8 terminal effector cells, and lower NK cells at BL with severe (grade ≥3) irAEs ([172]online supplemental figure 8A–D). Discussion Despite widespread recognition and attention to irAE, the mechanisms underlying these autoimmune ICI toxicities remain poorly understood, as do reliable means of predicting and even diagnosing them. To address these issues and knowledge gaps, we conducted a comprehensive multidimensional cellular and molecular analysis of the host peripheral immune system in a large and diverse cohort of ICI-treated patients. Using various assays on longitudinal peripheral blood samples, our study revealed several key findings. Patients who eventually developed irAEs had (1) an increased BL abundance of plasmablasts and significantly elevated plasma ANA levels; (2) a skewed BL T cell profile characterized by increased proliferative and activated CXCR3^+ T cells, CD57^+ T cells and NKT cells along with a lower BL abundance of CD56^dim CD16^+ NK cells; (3) distinct upregulated gene signatures at BL in T/NK cells (CXCR3, TNF and IL1B) and in myeloid cells (IL1B and CXCL8) coupled with enhanced TNF signaling and higher fraction of IL1B-positive myeloid cells; and (4) a significant shift post-therapy towards an activated and inflammatory immune state, marked by increased CD4 memory populations, upregulated IL18 and significantly elevated IFN-γ inducible proinflammatory chemokines CXCL9 and CXCL10 coupled with reduced CXCL8 levels in myeloid cells. In contrast, patients without irAE exhibited an immunosuppressive profile characterized by increased BL abundance of a TGFBI^high myeloid cluster and post-treatment myeloid protumorigenic reprogramming. Together, these findings may suggest that underlying subclinical autoimmunity and a proliferative activated proinflammatory immune state predispose patients to irAEs. Our study is notable for the large size of the study cohort (which permitted subset analyses for some assays) and the comprehensive nature of the correlative studies performed (allowing cross-validation of findings across orthogonal methodologies). Additional strengths include the quality of clinical data (particularly irAE occurrence, grade, and type), the use of real-world cases, the focus on pretreatment BL blood samples (the results of which could be incorporated into the upfront selection of patients, treatment regimens, and monitoring strategies), and validation of certain assay results in external datasets. The numerous novel observations in our present study could reflect the relatively large sample size, the diversity of cancer types, and the inclusion of grade 2 irAE (whereas some prior reports incorporated only grade ≥3 toxicities[173]^13). We chose to incorporate grade 2 events because (1) they are medically significant, requiring adjustment of ICI and/or administration of immunosuppression or other therapies; (2) in some cases, they may become permanent; and (3) if not recognized and acted on, they could progress to higher-grade toxicities. Recently, efforts aimed at elucidating the biological underpinnings of irAE have emerged. A scRNA-seq analysis of ICI-treated patients with melanoma identified an activated CD4 effector memory subset in those who developed high-grade irAE.[174]^13 A separate CyTOF study found early on-treatment expansion of Ki67^+CD8^+ T cells and T regulatory cells in patients with lung cancer and melanoma who experienced irAE.[175]^29 The current analysis extends these findings, revealing associations between an increased BL abundance of plasmablasts, proliferating activated Mki67^+CXCR3^+HLADR^+ T cells, and high ANA levels, suggesting a pre-existing autoimmune-like state predisposes patients to irAEs. Multiple previous studies have identified CD8 effector and terminal effector T cells in irAE-affected tissues in colitis,[176]^30 31 arthritis,[177]^32 myocarditis,[178]^33 and thyroiditis.[179]^34 In the present analysis, we found a higher abundance of circulating CD8 effector and terminal effector T cells in irAE patients at BL, raising the possibility that these circulating CD8 T cells may act as precursors to the tissue-infiltrating CD8+ T cell subsets observed in irAE target organs. We also found elevated levels of CD57^+ T cells, NKT cells, and TCRγδ cells in irAE patients prior to ICI initiation. CD57 is a marker of terminally differentiated cells with high cytotoxic potential. Multiple autoimmune diseases, including Graves’ disease, ankylosing spondylitis, and rheumatoid arthritis, are characterized by increased circulating CD57^+ T cells.[180]^35 Furthermore, BL T/NK cells from irAE cases exhibited enrichment for the TNF signaling pathway and displayed a distinct three-gene signature (TNF-CXCR3-IFNG) that could have a potential role in irAE prediction. Conversely, patients without eventual irAE exhibited a more immunosuppressive and immune-evasive T/NK profile at BL, characterized by upregulation of CXCR4 and SIGLEC7.[181]36,[182]38 scRNA-seq and cytokine analyses highlighted the central role that peripheral blood myeloid cells may have in mediating irAEs. In irAE cases, we identified a proinflammatory monocyte phenotype at BL, a significant upregulation of CXCL9 and CXCL10 gene expression in monocytes, and a marked increase in their serum levels following ICI initiation. After ICI initiation, monocytes in irAE cases exhibited upregulated IL18 expression, notable because IL18 activates T and NK cells and potently induces IFNγ production, thereby contributing to hyperinflammation.[183]^39 40 Together, these findings suggest that bidirectional interactions between monocytes and T cells may support irAE pathogenesis. In contrast, a more immunosuppressive and immune evasive phenotype was associated in non-irAE cases, including increased abundance of TGFB1^high myeloid cluster enriched for immunosuppressive markers such as CCR2 and STAB1 at BL and upregulation of immunosuppressive SPARC after ICI initiation. This study has multiple limitations, including the grouping of diverse irAE types into a single category. Although we initially intended to perform scRNA-seq on discrete populations according to organ-specific toxicities, the frequent occurrence of multiple irAE types within a single patient precluded that approach. Indeed, multiple other studies have reported irAE co-occurrence in up to 50% of irAE cases.[184]^41 42 Relatively small case numbers for some analyses also prevent meaningful subset analysis according to irAE type. The inclusion of multiple cancer types, stages, and ICI treatments in the present study prohibits analysis of efficacy outcomes, which have been shown to correlate with irAE.[185]^24 43 Certainly, any link between proposed irAE biomarkers and efficacy would be critical to identify, especially if resulting clinical decisions could include modifying or avoiding ICI therapy. While that represents one approach to reducing irAE risk, other strategies such as personalizing irAE monitoring might mitigate the risk of the most severe autoimmune toxicities while not detracting from potential therapeutic benefits. Other constraints include limited sample sizes in certain cancer groups and among combination ICI-treated patients, as well as a lack of TCR and B cell receptor sequencing data. Additionally, while scRNA-seq and CyTOF provide valuable insights, their complexity and cost limit widespread clinical implementation. Future studies could validate our identified biomarkers, cell subsets and molecular signatures using more accessible methods such as flow cytometry and bulk RNA sequencing. Although ANA testing is clinically available, its sensitivity appears insufficient for standalone risk stratification. Finally, variations in clustering methods, cohort sizes, and cell numbers used for assays may influence results, underscoring the need for further validation across diverse cancer types and treatment regimens in larger patient cohorts to facilitate clinical translation. This multidimensional analysis identified distinct molecular signatures and circulating immunological determinants predictive of irAEs in ICI-treated patients. These include elevated BL ANA levels; increased plasmablasts; greater abundance of activated/proliferative CXCR3^+ lymphocytes, CXCR3^+ T memory, and effector memory cells; and an upregulated 3-gene signature—IFNG, TNF, and CXCR3—in T/NK cells. Overall, patients who developed irAEs exhibited an activated, autoimmune-like proinflammatory state at BL, predisposing them to excessive inflammatory responses after ICI therapy, whereas non-irAE cases exhibited an immunosuppressive immune profile. These novel insights into irAE biology may contribute to future predictive biomarkers, open new avenues for irAE monitoring and mitigation, and provide a valuable resource for the growing research community focused on understanding irAEs. Supplementary material online supplemental figure 1 [186]jitc-13-9-s001.pdf^ (1.7MB, pdf) DOI: 10.1136/jitc-2025-012414 online supplemental file 1 [187]jitc-13-9-s002.docx^ (26.9KB, docx) DOI: 10.1136/jitc-2025-012414 Acknowledgements