Patient samples
Freshly resected, histologically-confirmed tumor tissue and/or blood were collected from patients with cutaneous melanoma. Tumor samples were obtained immediately following surgery and processed, as outlined below. Heparinized blood samples were collected prior to surgery or treatment and immediately processed, as outlined below. Patient sample details, including the tissue site sampled, processing details, sex, age, treatment history, and clinical outcome, are available in table S1.
Tumor dissociation and peripheral mononuclear cell isolation
Single-cell tumor and blood suspensions were prepared, as previously described 7. In brief, freshly resected tumor samples were mechanically dissociated and digested in HBSS medium with collagenase IV (2.5 mg/ml; Roche) and DNase I (0.2 mg/ml; Worthington Biochemical Corporation) at 37°C for 30 min. Dissociated tumor suspensions were then isolated using Lymphoprep gradient centrifugation. Peripheral blood mononuclear cells were isolated from whole blood using Lymphoprep gradient centrifugation. For samples Mel-T-09, Mel-T-11, Mel-UT-03, Mel-UT-05, Mel-UT-08-13, and Mel-UT-17-18, single-cell suspensions were cryopreserved in GemCell human AB serum (Gemini) with 20% DMSO in liquid nitrogen. The single-cell suspensions were then thawed per 10X Genomics protocols CG000233 and CG000447 for tumor and blood samples, respectively, prior to further processing.
Cell sorting
Single-cell suspensions were stained for live cells (Live/Dead Cell Viability Assay; Life Technologies) followed by fluorophore-conjugated anti-human antibodies, as summarized in table S2. Samples were sorted on a BD FACS Aria II with the gating strategies outlined in table S3. At least 30,000 live cells were sorted per sample.
CITEseq antibody staining
Cryopreserved peripheral blood mononuclear cell suspensions were thawed, as above. Lyophilized TotalSeq™-C Human Universal Cocktail V1. 0 (BioLegend) were reconstituted and used for staining, per the manufacturer’s protocols. Each sample was reconstituted to a concentration of 20 million cells/mL and incubated with Human TruStain FcX™ Fc Blocking reagent (BioLegend) for 10 minutes at 4°C. The blocked cells were then stained with the reconstituted TotalSeq-C antibody cocktail and fluorophore conjugated surface antibodies for cell staining (table S2) for 30 minutes at 4°C as washed with Cell Staining Buffer (BioLegend) prior to cell sorting (table S3).
10X library preparation and sequencing
Sorted samples were prepared for single-cell sequencing by the Keck Microarray Shared Resource (KMSR) and sequenced by the Yale Center for Genome Analysis (YCGA) at Yale University. Cells were processed following the recommended protocol with the Chromium Single Cell 5’ Library Construction Kit and Chromium Single Cell V(D)J Enrichment Kit (Human T Cell; Single Cell 5’ Chemistry). Libraries were sequenced on an Illumina NovaSeq S4 instrument. scRNAseq libraries were sequenced at a read length of 26 × 8 × 91 bp and a depth of 300 million reads per sample. CITEseq libraries were sequenced at a read length of 26 × 8 × 91 bp and a depth of 100 million reads per sample. scTCRseq libraries were sequenced at a read length of 150 x 150 bp and a depth of 20 million reads per sample. FASTQ files were generated and analyzed with CellRanger (version 5.0.1) using the GRCh38 human reference genome for alignment.
Whole exome sequencing
Melanoma tumors and adjacent normal tissue were analyzed with whole-exome sequencing, as previously described in-detail 37. Briefly, genomic DNA was extracted from snap-frozen tumors using DNeasy purification kits (Qiagen Inc.). Genomic DNA was sheared, end repaired, ligated with custom adapters (Integrated DNA Technologies), amplified, and size selected. Sample concentrations were normalized to 2nM and sequenced on an Illumina NovaSeq 6000 using 101 bp paired-end sequencing reads, per Illumina protocols. Raw sequencing files were aligned to the GRCh38 human genome.
Bulk RNA sequencing
Bulk RNA sequencing was performed on melanoma tumors, as previously described 37,38. In brief, RNA was extracted from tumors using the RNeasy PowerLyzer Tissue & Cells Kit (QIAGEN) and quality was assessed with a 2100 Bioanalyzer System (Agilent Technologies). Ribosomal RNA was depleted Kapa RNA HyperPrep Kit with RiboErase (Kapa Biosystems, Inc., Cape Town, South Africa). Sequencing libraries were generated using TruSeq RNA sample prep kits (Illumina) and sequenced on an Illumina HiSeq 2500 using 2 × 150 bp paired-end reads with a target of 20–25 million reads per sample. Raw sequencing files were aligned to the GRCh38 human genome.
Immunophenotyping human peripheral blood mononuclear cells
Cryopreserved peripheral blood mononuclear cells were thawed, as above, and rested at 37°C for 16 hours. Cells were then stained for viability with Fixable Near-IR Dead Cell Stain (Life Technologies) at room temperature for 10 minutes in the dark. Samples were then washed with PBS with 2% FBS, prior to staining with a master mix of surface antibodies (table S2) at room temperature for 20 minutes. Cells were then washed with PBS with 2% FBS. Samples undergoing intracellular staining were fixed for 45 minutes with Fixation Buffer, per the manufacturer’s protocol (eBioscience). Cells were washed with Permeabilization Buffer (eBioscience), stained with a master mix of intracellular antibodies (table S2) at 4°C for 16 hours, and resuspended in PBS + 10% FBS + 0.2% EDTA for analysis on a BD LSR Fortessa in the Yale Flow Cytometry Core.
MCR construction
HLA typing was computationally inferred from the single-cell RNA sequencing data from blood cells using the python-based package, arcasHLA 39. MCR libraries were generated by cloning oligonucleotide libraries (Twist Bioscience) into MCR-SCTz retroviral expression vectors (having an IRES-hCD4tr (1-420aa) downstream) for each patient HLA, as described previously 30. For cloning of single peptide-MCR constructs, oligos were purchased from Microsynth AG (Balgach, Switzerland). The libraries cover the full length of selected antigens, as detailed below, by sliding 10-mers shifted by one amino acid. After retroviral production, 16.2X cells were transduced with the MCR libraries. After 2-3 days, transduced cells were sorted based on hCD4 expression (BioLegend) on a BD FACSAria Fusion.
TCR cloning
Retroviral expression vectors with TCRs as a bicistronic TCRα-T2A-TCRβ transcript were produced by Twist Bioscience. 16.2A2 cells (16.2 T cell hybridoma expressing human CD3E, CD3G, and CD3D) were transduced and after 2-3 days, TCR expressing (BioLegend) cells sorted on a BD FACSAria Fusion.
MCR screening
MCR screening was performed as previously described 29,30, with the adaption of pooling of up to five different TCRs per co-culture. TCR-cells and MCR-cells were co-cultured with a 5-fold excess of each TCR compared to MCR-cells. Up to 7 rounds of enrichment were done. When a NFAT-signal was detected, the enriched MCR cells were co-cultured with the individual TCR-expressing cell lines to identify the TCR recognizing the MCR cells. Sorting was performed on BD FACSAria Fusions, flow-cytometry for acquisition only was done on a BD LSRFortessa or Luminex Guava easyCyte. When multiple peptides (due to multiple transduction) were detected in the sequencing of positive single cell clones, the peptides were subcloned and re-expressed as single peptides-MCR-SCTz (e.g. TCR2328). Expanded single cells were harvested, and DNA was isolated (KAPA Express Extract), followed by Sanger sequencing of the linked peptide and the HLA class I heavy chain.
Data Processing and Statistics
Single-cell RNA sequencing processing and analysis
CellRanger output matrices containing gene expression values for each cell were analyzed using the open-source toolkit, Seurat v4.2 40. Cell level quality control was performed for each sample separately. Thresholds for removing low-quality cells were selected based on the distribution of the number of unique genes per cell, the number of genes per unique molecular barcode (novelty score), the percentage of genes that map to the mitochondrial genome, and the expression of housekeeping genes. Library size normalization and variance stabilization was performed using SCTransform to apply a regularized negative binomial regression based on the 3,000 most variable genes 41. Genes encoded by the Y chromosome were removed for downstream analysis. Normalized datasets from tumor and blood samples for each patient were then combined using the FindIntegrationAnchors and IntegrateData functions in Seurat with default parameters 42. Scaled z-scores for each gene were calculated and used for principal component analysis (PCA) based on the integrated dataset.
Cell clustering was performed by first using the shared nearest neighbor (SNN) method based on statistically significant principal components, as applied by the FindNeighbors function. Differentially expressed genes between clusters of interest were then identified using the Wilcoxon Rank Sum test with Bonferroni correction (q < 0.05) 43. For subsequent T cell subset analyses, T cell receptor genes were removed to mitigate the effects of clustering driven strictly by highly expanded clones. Cluster annotation was manually assigned based on the expression of canonical marker genes, differentially expressed genes, overlap coefficients with previously published cell phenotype gene signatures 5,25,44,45, automated reference-based annotations 46, and the proportion of cells with paired T cell receptor CDR3α and CDR3β chains. Clusters with T cells were selected for downstream T cell phenotyping (table S4-S5). CD8A, CD8B, and CD4 expression following Adaptively-thresholded Low Rank Approximation imputation (ALRA, version 1.0.0) 47 were also used to assign CD8+ and CD4+ phenotypes.
T cell receptor repertoire analysis
T cell receptor contigs called with high confidence were extracted from CellRanger VDJ outputs and retained for downstream analyses. Nucleotide and amino acid CDR3α and CDR3β sequences and count matrices of gene expression were matched for each cell based on barcode identities using custom R scripts. Only cells with a single CDR3α and single CDR3β sequence were retained for downstream analyses. Cells with multiple sequences or those with missing sequences were not considered for clonal analyses. Clonal relatedness was defined by cells with exact matching single CDR3α and single CDR3β sequence per patient.
To quantify the degree of clonal expansion, we calculated the proportion of T cell clones occupied by a given clonotype per patient. To predict T cell receptor specificities, we matched clonotype sequences in our dataset with VDJdb, a publicly available database of T cell receptor sequences annotated with experimentally validated specificities 13,48. Human species clonotypes were extracted from VDJdb and matched based on exact amino acid sequences of the CDR3 from the paired a-chain and b-chains. We then calculated the number of tumor antigen-specific and viral-specific clonotypes in each reactivity group.
Additional single-cell T cell receptor repertoire analysis was performed using the scRepertoire package (version 1.12.0) 49. Filtered annotated contig files generated by the CellRanger VDJ pipeline were used as input files. Cells with paired single alpha- and beta-chains were selected for downstream analysis. Clonotypes were defined by using the CDR3 nucleotide and amino acid sequences. Indices of diversity (Shannon entropy, Inverted Simpson) and species richness (Chao, ACE) were calculated using clonalDiversity function. Between group differences were calculated with Kruskal-Wallis one-way analysis of variance testing and Dunn’s multiple comparison testing. Visualization of repertoire metrics were generated using GraphPad Prism (version 10.1.1).
Differential abundance analysis
To unbiasedly identify cellular neighborhoods enriched for tumor matched reactive or unreactive circulating CD8+ T cells, we used an R implementation of Local Two-Sample Testing for differential abundance protocols 15. The first 30 dimensions of principle components and k = 25 were used to construct a k-nearest neighbors (KNN) graph of the data using Euclidian distances. Random walk scan statistics were then calculated (q < 0.05), and differential abundance labels were assigned if q < 0.01. Differentially abundant cells were then grouped into different clusters by employing Seurat SNN clustering with default parameters. All differentially expressed genes were then calculated for each differentially abundant cluster using the FindAllMarkers function in Seurat (table S6).
Single-cell gene set enrichment analyses
Gene set enrichment analysis of the neoTCR scores, the KIR+CD8+ signature, and the conserved KIR+CD8+ signatures were calculated from the cell counts of the single-cell RNA sequencing data using the R-based package AUCell (version 1.24.0) 50. Gene sets were considered active based on the internal global_k1 threshold or through a manual examination of the distribution pattern.
Single-cell gene signature scoring was performed using the R-based package UCell (version 2.1.2) with default parameters 51. Between group differences were calculated using Wilcoxon Rank Sum testing with continuity correction (q < 0.05).
Gene set enrichment analyses between gene lists was performed using the GSEA software (version 4.3.2) 52. Differentially expressed genes from differentially abundant populations were pre-ranked based on the fold-change calculated from Wilcoxon signed-rank testing with Bonferroni correction (q < 0.05). Pre-ranked gene lists were then compared with the top 200 genes upregulated in human KIR+CD8+ T cells from patients with multiple sclerosis 2 and immune signatures from Hallmark gene sets in the Molecular Signatures Database (MSigDB) 53.
Conserved gene signature analysis
Differentially upregulated genes shared between circulating KIR+CD8+ T cells and clonally related cells in the tumor were identified using the FindConservedMarkers function in Seurat. To derive the conserved signature, differentially expressed genes were first identified between KIR+CD8+ T cells and all other CD8+ T cells in the blood and tumor separately based on Wilcoxon Rank Sum testing with Bonferroni testing for multiple-comparison correction. Differentially expressed genes across tissue compartments (maximum p < 0.01), expression in ≥1% of cells, and an average log-fold change ≥ 0.15 were considered conserved and used for downstream analysis (table S7).
Trajectory analyses
To perform trajectory inference analysis, we fit a minimum spanning spanning tree (MST) to the UMAP dimensionality reduction plot using the Slingshot package (version 2.7.0) 54. The piecewise linear trajectory was smoothed using simultaneous principal curves to derive trajectory and pseudotime values. To determine associations between gene expression and pseudotime values, we fit a negative binomial general additive model using the tradeSeq package (version 1.10.0) 55. Pairwise differences in the gene expression pattern between lineages was determined using the Wald test (p < 0.05), as applied by the patternTest function with default parameters (table S8).
Neoantigen prediction
Aligned sequencing reads generated by whole exome sequencing were analyzed for somatic variants by NEXUS Personalized Health Technologies core at ETH Zurich using the SwissMTB workflow 56. Somatic mutations were selected if identified by at least two of three variant callers (Strelka2, MuTect2 57, VarScan 58). Somatic variants were subsequently annotated using Ensembl Variant Effect Predictor 59.
Antigen selection for MCR library
Peptides derived from neoantigens identified above and shared antigens were included for MCR screening. Shared antigens included curated tumor associated antigens, select human endogenous retroviruses (HERVs), previously described driver mutations 60, and previously described bacterial peptides from melanoma tumors 61. Sequences from cytomegalovirus, Epstein Barr virus, and influenza peptides from the Immune Epitope Database were included as controls 62.
Tumor associated antigens were first selected by performing differential expression analysis between 369 metastatic melanoma samples from The Cancer Genome Atlas (TCGA) and 974 normal skin controls from the Genotype-Tissue Expression (GTEx) 63. Genes were filtered for expression of ≥10 transcripts per million (TPM), ≥10% of samples, and ≥3-fold change expression in melanoma tumors were selected. Identified genes were further filtered for a median expression ≥ 5 TPM in the bulk RNA sequencing data generated from 20 tumors from 13 patients in the Yale cohort (table S1), yielding 66 total genes. An additional 29 genes from known and investigational tumor associated antigens were also included for MCR screening.
Human endogenous retroviruses were selected by first aligning raw reads from the bulk RNA sequencing data from the melanoma tumor in the Yale cohort to a previously published reference of pro-viral sequences 64. HERVs pro-viral sequences with expression levels ≥10 TPM in ≥3 tumors and with previously annotated gene identifiers were selected, resulting in 19 genes. An additional 5 genes associated with HERV-driven oncogenesis or eliciting T cell responses were also included.
CITE-seq data processing and analysis
Raw and filtered UMI matrices for antibody-derived tag (ADT) counts generated from the CellRanger pipeline were normalized with the dsb package (version 1.0.3) using default parameters 65. Background droplets were identified by examining the protein library size distribution. The normalized ADT matrices were then added to Seurat objects generated from the RNA count matrices and analyzed, as described above.
Surface protein-encoding gene selection
To identify candidate surface markers where gene expression correlated with protein-level expression, we calculated Spearman’s Rank correlation coefficients between normalized expression in the ADT and RNA assays. Of 137 markers analyzed, 40 genes positively correlated (r > 0.1) with protein-level expression and were therefore included in the surface marker classifier models (table S9).
Cell classification analysis
To further investigate the relationship between surface markers and tumor reactivity, we adopted a Lasso Logistic model 66–68 to analyze normalized count matrices from CD8+ T cells (glmnet version 4.1-4). Training and testing datasets were constructed by randomly splitting cells from the immunotherapy-naive cohort into equal sample sizes. In the training phase, the Lasso Logistic model was fitted to the count matrices for the 40-surface protein-encoded genes, select clinical variables (sex, age, clinical stage, lesion location, technical batch), and predicted reactivity. The classifier was then applied to the testing datasets and the immunotherapy resistant cohorts. Receiving operating characteristics statistics were calculated based on the differential abundance reactivity labels for the immunotherapy naive cohort and neoTCR8 reactivity labels for the immunotherapy resistant cohort and visualized using the R-based package pROC (version 1.18.0).
To facilitate the application of the Lasso Logistic model classifier to clinically applicable techniques such as flow cytometry, we adopted a Decision Tree model to assign hierarchical combinations of markers using samples where simultaneous RNA sequencing and CITE-seq were performed. Testing and training datasets were constructed by randomly splitting cells from the CITEseq cohort. Thresholds to dichotomize protein-level expression of the markers selected by the Lasso Logistic model classifier were determined by examining the pattern of expression in the ADT assay. A Decision Tree was constructed based on the binary protein expression levels to predict tumor reactivity in the training set using R-based package rpart (version 4.1.19).
Flow cytometry data analysis
Flow data was analyzed using FlowJo™ (version 10.9). Gating was determined by comparison to Fluorescence Minus One (FMO) controls using healthy donor human peripheral blood mononuclear cells.
Survival analysis
The association between the frequency of reactive KIR+CD8+ T cells (high, or greater than or equal to the median frequency for that cohort, vs. low, or less than the median frequency for the cohort) as classified by flow cytometry staining (table S10), the Lasso logistic regression classifier (table S11), or the decision tree model and overall survival (OS) time was assessed by Kaplan-Meier analysis. We used the Gehan-Breslow-Wilcoxon test for trend (p < 0.05) to detect significant differences in OS between the high proportion and low proportion groups. All survival analyses were performed using GraphPad Prism (version 10.1.1).
Study approval
This study was approved by the Yale University Institutional Review Board. All participants provided written, informed consent before tissue and/or blood collection.
Data availability
Further information and requests for resources and reagents should be directed to and will be fulfilled by the corresponding authors.