Skip to main content
Advertisement
Main content starts here
Full access
Research Article
CANCER IMMUNOLOGY

Coordinated immune networks in leukemia bone marrow microenvironments distinguish response to cellular therapy

Science Immunology
24 Jan 2025
Vol 10, Issue 103

Editor’s summary

Hematopoietic stem cell transplantation (HSCT) represents the only potential cure for patients with aggressive myeloid leukemia. Donor lymphocyte infusion (DLI) is used to treat patients experiencing leukemia relapse after HSCT, but why some patients respond better to DLI immunotherapy remains unclear. Using single-cell and spatial transcriptomics, Maurer et al. analyzed longitudinal bone marrow samples from patients with relapsed acute myeloid leukemia (AML). Response to DLI was associated with expansion of ZNF683hi cytotoxic T cells, which were mostly derived from the DLI product and formed complex cellular networks with other bone marrow immune cells. Together, these findings provide insight into the features of effective antileukemia immune responses. —Claire Olingy

Abstract

Understanding how intratumoral immune populations coordinate antitumor responses after therapy can guide treatment prioritization. We systematically analyzed an established immunotherapy, donor lymphocyte infusion (DLI), by assessing 348,905 single-cell transcriptomes from 74 longitudinal bone marrow samples of 25 patients with relapsed leukemia; a subset was evaluated by both protein- and transcriptome-based spatial analysis. In acute myeloid leukemia (AML) DLI responders, we identified clonally expanded ZNF683+ CD8+ cytotoxic T lymphocytes with in vitro specificity for patient-matched AML. These cells originated primarily from the DLI product and appeared to coordinate antitumor immune responses through interaction with diverse immune cell types within the marrow microenvironment. Nonresponders lacked this cross-talk and had cytotoxic T lymphocytes with elevated TIGIT expression. Our study identifies recipient bone marrow microenvironment differences as a determinant of an effective antileukemia response and opens opportunities to modulate cellular therapy.

INTRODUCTION

Immunotherapy has transformed cancer treatment, but responses vary widely among tumor types and patients (1). The biological underpinnings of such responses and of resistance are poorly defined, particularly in myeloid malignancies where conventional immunotherapies have had limited success (24). Effective antitumor immunity relies at least in part on coordinated activity among cell types within the tumor microenvironment (TME) (58). However, the specific role and characteristics of tumor-infiltrating immune cells remain unclear (911). Developing a better understanding of the cellular players within the TME may be the key to unlocking effective tumor-directed responses (1214).
An established form of combinatorial chemoimmunotherapy is allogeneic hematopoietic stem cell transplantation (HSCT), the primary modality and only possibility of cure for patients with aggressive myeloid leukemia. Maintenance of remission after HSCT relies on the graft-versus-leukemia (GvL) effect, wherein donor immune cells eliminate residual leukemic recipient cells. Immunological escape by residual leukemia leads to disease relapse after HSCT, but remission can be achieved by donor lymphocyte infusion (DLI), which reinstates immunological control through effective GvL (15). Despite long-standing clinical recognition, the cell types and mechanisms underlying this process remain undefined. Although DLI responsiveness is highly successful for chronic myeloid leukemia (CML; 70 to 80% response rate), it is markedly less effective for acute myeloid leukemia (AML; 15 to 20% response rate) (16), suggesting divergent sensitivity to GvL even among myeloid malignancies. Therefore, DLI represents an informative model system for elucidating determinants of immunological response and resistance within the leukemia bone marrow (BM) microenvironment (1720).
Single-cell transcriptomic and spatial profiling can powerfully enhance our ability to disentangle the complex TME (2123). We previously identified expansion of precursor exhausted T (TPE) cells in CML responders (CML-Rs) to DLI through single-cell RNA sequencing (scRNA-seq) and machine learning methods (11); however, whether this population is associated with response in AML is unknown. We therefore embarked on a comprehensive characterization of the leukemic marrow microenvironment to broadly investigate cross-talk between heterogeneous leukemic states and immune cells shaping GvL with a focus on the high-risk and clinically challenging setting of relapsed AML, for which few therapies exist and outcomes are poor (24). We present integrated scRNA-seq, machine learning, spatial analysis, and in vitro functional validation results that reveal coordinated multicellular interactions driven by a CD8+ cytotoxic T lymphocyte (CTL) population and underlying response to adoptive cellular immunotherapy for AML.

RESULTS

Distinct BM microenvironments among myeloid leukemias

To dissect immune cell states in the BM microenvironment of patients with post-HSCT relapsed AML treated with DLI, we profiled the transcriptomes of individual marrow cells from 30 biopsy samples [five responders (Rs) and four nonresponders (NRs); Fig. 1A and data file S1]. Patients in both groups had similar baseline, transplant, and DLI characteristics; however, there were more female recipients in the R group (fig. S1, A to C, and data file S2). A total of 47,372 viable bone marrow mononuclear cells (BMMCs) were profiled by scRNA-seq, single-cell T cell receptor sequencing (scTCR-seq), and cellular indexing of transcriptomes and epitopes by sequencing (scCITE-seq) for proteomic characterization. We further included seven DLI product samples [43,919 cells from four AML responders (AML-Rs) and three AML nonresponders (AML-NRs)]; two post-HSCT samples from nonrelapsed patients with AML (7617 cells); and, as further controls, six samples (27,131 cells) from three patients with post-HSCT relapsed AML who entered remission after chemotherapy without DLI.
Fig. 1. Single-cell transcriptional analysis of the leukemic BM microenvironment response to DLI.
(A) Schema of BM samples analyzed by scRNA-seq, scTCR-seq, and scCITE-seq. (B) Two-dimensional (2D) Uniform Manifold Approximation and Projection (UMAP) of all 356,070 BM transcriptomes (including doublets) analyzed. Each dot represents a cell and is colored on the basis of 58 clusters denoting distinct cell subsets. Dashed lines indicate the major cell types. (C) Coloring of 2D UMAP for BM myeloid cells on the basis of trajectories defined by Decipher for healthy myeloid differentiation (top), CML evolution (middle), and AML evolution (bottom). Gray: nonmyeloid cells. Heatmaps on right: gene expression profiles of BM myeloid cells sorted by each Decipher component. (D) Coloring of 2D UMAP on the basis of the percentage of cells per cluster from each disease. Red: AML; blue: CML. (E) Coloring of 2D UMAP on the basis of patient response. (F) Top: bar graph normalized by total number of cells per disease type demonstrating the percentage of each cluster from AML-Rs (teal), AML-NRs (pink), CML-Rs (purple), and CML-NRs (orange) and the percentage of each cluster from before DLI (lighter colors) or after DLI (darker colors). Bottom: relative size of clusters and major cell type per cluster. Top: HSCs (gold) and leukemia cells (gray); middle: myeloid (red) and erythroid (orange) cells; and bottom: lymphoid cells [T cells (blue), B cells (pink), and NK cells (purple)].
To expand our BMMC compendium for myeloid malignancies, we merged the AML data with our prior dataset of 42 marrow specimens from 16 patients with CML who received DLI and 2 nonrelapse controls (11) and extended the analysis from T cells to the entire BM microenvironment. After batch correction and clustering (25), we constructed an integrated atlas of 451,453 mononuclear cells (Fig. 1B). Fifty-three distinct cell clusters were annotated using both reference-based and manual annotation (Fig. 1B; fig. S1, D and E; and data files S3 and S4). Expression of lineage-defining genes confirmed major cell type annotations (fig. S1F).
Distinguishing leukemic cells transcriptionally from healthy hematopoietic stem cells (HSCs) and myeloid progenitor cells is challenging because of partial overlap in differentiation trajectories and interpatient heterogeneity (23, 26, 27). We annotated leukemia cells on the basis of a combination of gene expression (fig. S1G) (28), chromosome Y genes for sex-mismatched donor/recipient pairs (fig. S1H), and copy number variants (CNVs) (fig. S1, I and J) (29) to create a composite donor/recipient map of cells (fig. S1K). To enable analysis of shared phenotypes in leukemic clusters specific to patients with AML, we combined myeloid clusters into seven metaclusters (MCs) and then subclustered on the basis of donor (MC1-7) or recipient origin (MC1-7-leukemia) (fig. S1L).
We next asked whether CML and AML had divergent evolutionary trajectories from each other and from normal myeloid development. We applied Decipher (27), a deep generative model for reconstructing and aligning cell trajectories. Decipher revealed three continuous trajectories corresponding to shared normal myeloid differentiation in CML and AML specimens expressing VCAN, FCN1, CD14, and S100A8 (30) and divergent CML and AML leukemic evolutionary paths, with higher expression of CXCL8, RACK1, B2M, TRIM8, and IL18 for AML (3134) and KCNH2, HIST1H4C, and HSPA8 (35) for CML (Fig. 1C and data file S4). Nonleukemia clusters were overall evenly distributed by response (P = 1.0, t test), dataset [P = 1.0, analysis of variance (ANOVA)], and donor/recipient (P = 1.0, t test; Fig. 1, D and E, and figs. S1K and S2, A to C), indicating robust correction for batch and library size. After normalizing for total cell number for AML and CML, few clusters were dominated by cells from one disease [n = 7 (13%) for AML, n = 10 (19%) for CML] (Fig. 1F). Together, construction of this cell state atlas revealed distinct disease phenotypic trajectories and identified both shared and disease-specific immune cell populations.

A cascade of leukemia–immune cell interactions involving CD8+ZNF683Hi cytotoxic T cells in AML-Rs

To identify cell subsets associated with response to DLI, we evaluated individual cluster compositions of marrow cells by disease (AML versus CML), time point (before DLI versus after DLI) and response (Rs versus NRs) (data file S4). We hypothesized that populations expanding after DLI may represent cellular mediators of a GvL response. We also assessed clusters enriched in the BM microenvironment before DLI in Rs or NRs as candidate predictors of treatment response. One population trended toward expansion in Rs but not NRs with CML: T cell C2 (P = 0.06 and P = 0.70, respectively; Fig. 2, A and B). This cluster expressed CD4, SELL, CCR7, TCF7, and IL7R (fig. S2, D to H), aligning with our previously identified TPE cell population (11). This cluster was not expanded in AML-Rs, suggesting involvement of alternate cell types in the GvL response in AML. A tumor-infiltrating natural killer (NK) population (cluster C36) was found to be enriched in CML Rs before DLI (figs. S2, G and H, and S3A).
Fig. 2. Temporal dynamics and interactions of BM-infiltrating immune cell states.
(A) Percentage of C2: CD4 TPE cells among all cells per CML-R sample (n = 15 in R-post, 15 R-pre), CML-NR sample (left; n = 11 NR-pre, 8 NR-post), and AML sample (right; n = 3 NR-post, 7 NR-pre, 11 R-post, 9 R-pre). (B) DIISCO prediction of the percentage of total marrow cells comprising C2 (CD4 TPE cells) and CML1 clusters over time for CML-R samples (top; n = 24 samples) and CML-NR samples (bottom; n = 11 samples). Point sizes are proportional to the sample size. (C) Percentage of total marrow cells comprising C0: CD8+ZNF683Hi CTLs (left), C40: CD8 TEM cells (middle), and C1: cytolytic NK cells (right) per sample. Numbers of patient samples evaluated and statistical testing are same as in (A). P value by t test, P < 0.025 for significance with Bonferroni correction. (D) DIISCO prediction for C0: CD8+ZNF683Hi CTLs, C40: CD8 TEM cells, C1: cytolytic NK cells, and MC1 leukemia clusters for AML-Rs (top; n = 14) and AML-NRs (bottom; n = 10). Dot sizes are proportional to the sample size. (E) DIISCO prediction network for AML-Rs (top) and AML-NRs (bottom). (F) Example interactions between C0: CD8+ZNF683Hi CTLs and MC1 leukemia cells (top) and between C1: cytolytic NK cells and C0: CD8+ZNF683Hi CTLs (bottom). (G) Percentage of CD8 CTL/TEMRA cells among all T cells in peripheral blood of independent patients with post-HSCT relapsed AML who received DLI (n = 29 Rs, 24 NRs, Wilcoxon rank sum) by flow cytometry. n.s., not significant.
AML-Rs were characterized by expansion of T cell clusters C0 (P = 0.003) and C40 (P = 0.04) with a trend toward expansion in T cell C5 (P = 0.07), whereas AML-NRs were characterized by expansion of T cell clusters C21 (P = 0.01) and C26 (P = 0.001) (Fig. 2C, fig. S3B, and data file S5). None of these clusters were expanded in patients with CML. NK cluster C1 was expanded in AML-Rs (P = 0.002) but not in CML-Rs (P = 0.94) or AML-NRs (P = 0.49); however, AML-NRs had a wide range of C1 NK cell proportions across samples. T cell clusters C0 and C40 displayed higher CD8A and effector/cytotoxicity gene expression (e.g., ZNF683/Hobit, GZMB, PRF1, and B3GAT1/CD57; fig. S2, D and E) than other CD8+ T cell clusters. Compared with C40, the C0 cluster had even higher expression of ZNF683 and CD45RA by scCITE-seq (P = 0.0004, Kolmogorov-Smirnov test; fig. S2F), consistent with a CTL or T effector memory reexpressing CD45RA (TEMRA) cell profile (36, 37), and was thus designated as CD8+ZNF683Hi CTLs. The profile of T cell cluster C40 was most consistent with CD8+ T effector memory (TEM) cells.
NK cluster C1 displayed high expression of B3GAT1, FCGR3A, and GZMB and higher scCITE-seq expression of CD57 compared with other NK clusters, suggesting a cytolytic phenotype (fig. S2, G and H) (38). The C0: CD8+ZNF683Hi CTL, C1: cytolytic NK cell, and C40: CD8 TEM cell clusters did not expand in chemotherapy-only (no DLI) controls (fig. S3C), and their median proportions were similar to that of nonrelapse controls. To account for possible confounding variables that could overrepresent expansion of C0: CD8+ZNF683Hi CTLs, we used several additional analyses. Expansion of C0: CD8+ZNF683Hi CTLs was observed even after (i) calculating their proportion from only lymphocytes to account for possible higher leukemia burden in NRs (P = 0.007; fig. S3D), (ii) limiting samples to the first 300 days after DLI to account for longer follow-ups in Rs (P = 0.003; fig. S3E); and (iii) down-sampling cells to account for a greater number of cells in R samples than NR samples (P = 0.002; fig. S3F). Given that our prior CML cohort (11) was treated with CD8-depleted DLI, we additionally evaluated pre- and post-DLI samples from four patients with CML treated with conventional (i.e., CD8-replete) DLI (Fig. 1A). Similar to patients with CML treated with CD8-depleted DLI (39), we did not detect post-DLI expansion of C0: CD8+ZNF683Hi CTLs in Rs (fig. S3, G and H).
We asked whether any cell types were enriched in pre-DLI samples. Erythroid [C6: CML-enriched (P = 0.01) and C7: CML-enriched (P = 0.03)] and immature populations [C16: myeloerythroid progenitors (MEPs) (P = 0.03) and C37: MEPs (P = 0.02)] were elevated in AML-NRs compared with AML-Rs before DLI. No cell types were enriched in AML-Rs before DLI. Together, these analyses support the notion that multiple immune cell subtypes in the AML BM microenvironment undergo dynamic changes after DLI.
We reasoned that the dynamic patterns of response might arise from interactions and cellular cross-talk within the leukemic BM microenvironment over time. However, current computational methods for inferring cell-cell interactions from scRNA-seq data generate a static model of predicted interactions. We therefore applied DIISCO (dynamic intercellular interactions in single-cell transciptomics) (40), our newly devised Bayesian model, which uses a Gaussian process regression network (41, 42), to identify dynamic networks of cell-cell interactions within the BM microenvironment as “interactomes,” incorporating time relative to treatment as a variable (fig. S4A). In CD8-depleted CML DLI, DIISCO identified a central role for expanding C2: CD4+ TPE cells in Rs after DLI, predicted to inhibit CML1 cells and HSCs (C15), through coordination with C13: mature NKs, C0: CD8+ZNF683Hi CTLs, and C20: plasma B cells (Fig. 2B and fig. S4, B to D). This C2-centralized inhibitory role was not observed in CML nonresponders (CML-NRs) (fig. S4D). Predicted ligand:receptor complexes correlated with inferred interactions between C2: CD4+ TPE cells and CML1 cells included the protein products encoded by TNF:LTBR and CD226:NECTIN2, which have been associated with effective T cell responses (fig. S4E) (43, 44). These predicted networks support the central role of C2: CD4+ TPE cells in mediating GvL in CML.
For AML-Rs, the DIISCO analysis, in contrast, revealed a cascading response with multiple cell types expanding and interacting after DLI at different time points centering around C0: CD8+ZNF683Hi CTLs, C40: CD8 TEMs, and C5: CD8+ exhausted T (TEX) cells and including C1: cytolytic NK cells (Fig. 2, C to E, and fig. S5A). A strong inhibitory interaction between C0: CD8+ZNF683Hi CTLs and AML (MC1-leukemia) was observed in Rs (Fig. 2E), consistent with a productive GvL-driven contraction of MC1-leukemia cells. DIISCO identified positive interactions among C0: CD8+ZNF683Hi CTLs and C40: CD8 TEM cells along with C1: cytolytic NK cells, supporting the notion that these cell subsets formed a coordinated immune network supporting enhanced GvL activity (movie S1). To confirm that these predictions were not primarily attributable to increased immune infiltration, we retrained DIISCO on only immune clusters, resulting in recapitulation of this coordinated immune network (fig. S5B). These analyses reinforced the role of C0: CD8+ZNF683Hi CTLs, C40: CD8 TEM cells, and C1: cytolytic NK cells in the DLI response in AML.
Given that the proportion of C0: CD8+ZNF683Hi CTLs varied inversely with the proportion of leukemia cells in AML-Rs over time (Fig. 2D and data file S2), we postulated that this cluster might interact with leukemia, leading to T cell activation in AML-Rs but T cell inhibition in AML-NRs. We observed inhibitory signals from C0: CD8+ZNF683Hi CTLs toward AML-MC1 (e.g., GZMA:F2R, CCL4:SLC7A1) associated with CD8 T cell cytotoxicity and tumor suppression (Fig. 2F) (4547). Intramarrow interactions in AML-Rs after DLI were driven predominantly by C0: CD8+ZNF683Hi CTLs (P < 0.0001, Mann-Whitney U) (fig. S5B). Beyond lymphoid cells, DIISCO revealed inhibitory links in Rs between C0: CD8+ZNF683Hi CTLs and myeloid MC2 (dendritic cells and classical monocytes; Fig. 2E, fig. S5B, and data file S4). Conversely, in AML-NRs, DIISCO revealed the absence of notable activating interactions between T cells and other immune cell types after DLI.
We further asked whether CD8+ZNF683Hi CTLs could also be identified in peripheral blood. Analysis of patient R3501, whose scRNA-seq and scTCR-seq data from date-matched peripheral blood mononuclear cells (PBMCs) and BMMCs revealed T cell clonotypes in common, suggested that at least some T cell clonotypes were detectable in both compartments (fig. S5C). Moreover, the phenotype of the circulating post-DLI PBMC T cells was consistent with BMMC C0: CD8+ZNF683Hi CTLs (high expression of ZNF683 and GZMH and scCITE-seq expression of CD45RA). We therefore evaluated the proportion of circulating CD8+ TEMRA cells in PBMCs from 53 independent patients similarly treated with DLI for post-HSCT relapsed AML (29 Rs and 24 NRs). Multiparameter flow cytometry of PBMCs from this larger cohort had been performed in real time before and after DLI at our institution (data file S6). We detected marked expansion of the CD8+ CTL (TEMRA) population after DLI in Rs (fold change: 2.56 after DLI versus before DLI, P < 0.001, Wilcoxon rank sum) but not NRs (fold change: 1.3, P = 0.47, Wilcoxon rank sum) (Fig. 2G and fig. S5D). Excluding patients on active therapy (i.e., receiving systemic antileukemia therapy within 30 days of the analyzed sample) confirmed this finding (fig. S5E). A similar increase in AML-R after DLI was also seen in a naive B cell population (fig. S4, E and F). In our scRNA-seq dataset, C3: naive B cells skewed toward expansion in four of five AML-Rs (figs. S3D and S5, F and G). This independent PBMC confirmation of our BM scRNA-seq findings in a larger independent validation cohort supported a key role of CD8+ CTLs in effective GvL in AML. Together, application of DIISCO confirmed key roles of distinct T cell clusters in CML and AML and postulated a model of coordinated immune interactions driving effective GvL in AML centered around C0: CD8+ZNF683Hi CTLs.

Distinct spatial relationships of BM immune and leukemia cells between AML-Rs and AML-NRs

Organized networks of immune cells within solid TMEs have been linked to improved outcomes after immunotherapy (57, 4850). We hypothesized that the BM microenvironment of Rs might be spatially organized into lymphoid networks involving C0: CD8+ZNF683Hi CTLs, potentially promoting GvL. To confirm our DIISCO predictions that CD8+ CTLs function as a central hub for response to DLI in AML, we used codetection by indexing (CODEX) (51) to spatially assess the organization of immune cells at single-cell resolution. This was applied to six pre-DLI (four R and two NR) and six post-DLI (three R and three NR) BM core biopsy specimens obtained concomitantly with aspirates analyzed by scRNA-seq (data file S1). Upon quantifying the proportion of TEMRA cells (coexpressing CD3, CD57, and granzyme B) in five R and two NR samples containing >50 segmented cells, we confirmed TEMRA cell expansion relative to all cells in Rs (Fig. 3A).
Fig. 3. Spatial mapping of coordinated immune cell types in BM biopsies.
(A) Proportion of TEMRA cells (CD3+ and CD57+ or granzyme B+) among all cells. (B) Schema for defining niche types for R samples (n = 7 samples; 75,350 cells) and NR samples (n = 5 samples; 22,780 cells). (C and D) Niche types identified in Rs (C) and NRs (D) sorted by the ratio of cells before and after DLI. Bar plots: distribution of niche types across samples (top) and cell types (middle) after normalizing by the total number of cells annotated per sample. Niche type diversity (middle): entropy of distribution of cell types in each niche. Niche type density (bottom): cellular density of each niche. (E and F) Expression of CODEX markers for T cells in each niche identified in Rs (E) and NRs (F). Gray: niches with <50 annotated T cells. Boxes: signatures of exhausted, effector memory, and effector cells. n = 61,103 T cells for Rs, n = 19,579 cells for NRs. (G) Example window of a pre-DLI R sample (R3507) enriched for niche type r7. Contour line reflects spatial locations with a similar distribution of niche type. Smaller distance between contours: steeper change in niche density. Cells colored by cell type (top) and select markers (bottom). (H) Same as (G) for niche type r8 in a post-DLI R sample (R3509). (I) Same as (G) for niche type nr7 in a post-DLI NR sample (NR3510). P values by Mann-Whitney U test.
To systematically evaluate the immune networks defining response and resistance, we identified shared spatial neighborhood patterns across samples. Cells were aggregated across all R samples (four pre-DLI and three post-DLI) and separately across all NR (two pre-DLI and three post-DLI) samples and then clustered by composition of major cell types in their neighborhoods. Given that our marker panel (data file S7) was designed to resolve T cell subsets and other major immune cell types rather than leukemia cells, we focused on defining niches according to immune cell colocalization. After filtering, 98,130 cells (75,350 for Rs and 22,780 for NRs) were assigned to one of four major cell types (i.e., T and NK cells, B cells, HSC or leukemia cells, or myeloid cells), and the number of cells assigned to each major cell type within its neighborhood was quantified. Recurring colocalization patterns were designated as “niche types” using Phenograph (Fig. 3B). Together, we identified 37 distinct niche types in Rs (excluding two sample-specific niches, r0 and r1) and 19 distinct niche types in NRs (data file S8).
The R niche types (i.e., r2 to r37) typically comprised diverse immune cell populations over time (Fig. 3C), whereas the NR niche types (nr1 to nr19) lost diversity after DLI (quantified by Shannon entropy, P = 0.01, Mann-Whitney U; Fig. 3, C and D). We observed a trend toward higher cellular density (P = 0.06, Mann-Whitney U) in pre-DLI–enriched niches in Rs compared with NRs, suggesting preexisting diverse immunologic neighborhoods as a distinguishing feature of response. In addition, we observed higher abundance of T and B cells dominating pre-DLI R niche types versus NR niche types (P = 0.004 and P = 0.04, respectively; Mann-Whitney U), whereas myeloid cells exhibited higher proportions in NRs versus Rs before DLI (P = 0.002; Fig. 3, C and D). T cell abundance was further increased in Rs after DLI versus before DLI in parallel with decreased niche diversity (P = 0.0004; Fig. 3D).
Deep annotation of T cell phenotypic states across time identified that expanded T cells in Rs shifted phenotype from inhibitory [expressing T cell immunoglobulin and mucin-domain containing-3 (TIM3) and cytotoxic T lymphocyte associated protein 4 (CTLA4)] to effector [expressing lymphocyte-activation gene 3 (LAG3), granzyme B, CD57, and OX40] after DLI (Fig. 3E). Post-DLI niches in Rs had higher T cell expression of CD57 and OX40 (Mann-Whitney U test, P < 1 × 10−15), whereas NRs displayed enrichment of niches expressing the inhibitory marker T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains (TIGIT) (Fig. 3F). Although CD57 is sometimes considered a marker of proliferative senescence, numerous studies have demonstrated effector function of CD8+CD57+ T cells (5255). Down-sampling analysis to account for differences in cell numbers between Rs and NRs confirmed these marker expression differences (fig. S6, A and B). This transition to effector and cytotoxic T cell states in the BM microenvironment of Rs complemented the expansion of C0: CD8+ZNF683Hi CTLs and C40: CD8 TEM cells found in our temporal scRNA-seq analysis with DIISCO. Spatial mapping of pre-DLI R cells further underscored the colocalization of TEX cells with other immune cells, including B cells and myeloid and leukemia cells (Fig. 3G). Niche types dominating Rs after DLI included CD8+ CD57+ CTLs colocalized with other cytotoxic and TEM cells and myeloid cells enriched in dendritic cell marker CD11c (Fig. 3, E and H, and fig. S6C). In contrast, niche types in NRs reflected low diversity and dominance of myeloid cell types (Fig. 3I).
Our CODEX analysis was limited to a protein marker panel designed to capture T cell subsets. To extend the phenotypic analysis of spatially resolved marrow-resident immune cell subpopulations, we performed targeted spatial transcriptomic profiling (153 transcripts; Fig. 4A; fig. S7, A and B; and data file S9) paired with eight protein surface markers (56) on a subset of formalin-fixed paraffin-embedded BM samples from our CODEX discovery cohort and additional samples from our flow cytometry validation cohort (data file S6). Cells were clustered and manually annotated using differentially expressed gene (DEG) analysis (Fig. 4B and fig. S7C). T and NK cells were subclustered and finely annotated to identify CD8+ TEMRA cells (Fig. 4C and fig. S7D). In this manner, we confirmed that R samples after DLI globally had higher cellular diversity than NR samples after DLI (Shannon diversity index, P = 0.04, t test; Fig. 4D). In addition, we observed a higher diversity in the local neighborhood of CD8+ TEMRA cells and other immune cell types in Rs after DLI than in NRs (P < 0.05; Fig. 4E and fig. S7E), corroborating the paradigm of coordinated immune networks driving GvL. The higher diversity in CD8+ TEMRA neighborhoods in Rs was independent of the choice of local neighborhood radius (fig. S7F).
Fig. 4. In situ transcript and protein detection in BM biopsies.
(A) Schema of in situ transcript and protein profiling platform (Singular Genomics). (B) 2D UMAP of 193,651 cells analyzed by spatial transcriptomics and surface protein profiling with clusters manually annotated by DEG analysis. (C) 2D UMAP of 24,554 T and NK cells, subclustered by manual annotation, with CD8 TEMRA cells indicated (green). (D) Shannon diversity of the distribution of cell types computed for each sample (n = 6 Rs before DLI, 6 Rs after DLI, 4 NRs before DLI, 5 NRs after DLI). Entropy for each response group (Shannon diversity). P = 0.043 by t test. (E) Shannon diversity for distribution of cell types within the local neighborhood of cells, grouped by cell types and response group. P values by t test. Dots: mean diversity index for that cell type per sample. Abbreviations: Mye., myeloid progenitor cells; CD4 T, CD4+ T cells; CD8 Mem., CD8+ memory T cells; Ery., erythroid/precursor cells; Fib., fibroblasts; Mac., macrophages; Mega., megakaryocyte/megakaryocyte progenitor cells; Mono., monocytes. (F) Niche type composition across all samples (n = 5 Rs before DLI, 6 Rs after DLI, 4 NRs before DLI, 4 NRs after DLI). (G) Sample-level entropy on niche composition for each sample. P value by t test. (H) Niche 3 cell type composition (top) and sample fraction (bottom). (I) Niche types enriched after DLI in Rs (niches 2 and 13) or NRs (niches 10 and 6). Pie charts: niche composition by cell type. Box plots: fraction of niche in each sample stratified by response. (J) Niche 15 enriched in CD8+ TEMRA cells. Pie chart: cell type composition; box plot: fraction of niche in each sample stratified by response. P value by t test.
To further examine local immune composition, 16 distinct cellular niche types were identified across both R and NR samples using an unsupervised neighborhood analysis approach similar to that used for our CODEX data (Fig. 4F). R samples had greater niche type diversity than NR samples after DLI (Shannon diversity, P = 0.003; Fig. 4G). Niches 2, 3, and 13 contained higher fractions of CD8+ TEMRA cells and were enriched in Rs versus NRs (Fig. 4, H and I). Conversely, niches 6 and 10 were more enriched in NRs after DLI than Rs and exhibited a higher fraction of AML and myeloid cells (Fig. 4I). Niche 15 contained the highest fraction of CD8+ TEMRA cells and showed an increase in fraction in Rs after DLI, but statistical significance could not be established because of the sample size (P = 0.32; Fig. 4J). The spatial analyses of samples from both the discovery and validation cohorts thus provided orthogonal confirmation of post-DLI expansion of CD8+ TEMRA cells and enhanced cellular diversity of immune niches in the AML-R BM microenvironment.

AML-R T cells span a trajectory associated with more effector function and less T cell inhibition than in AML-NR T cells

Our multimodal analyses demonstrated that expansion of effector T cells is a central feature in the AML response to DLI. To better characterize the T cell phenotypes in AML-Rs and AML-NRs associated with response and resistance, we once again used Decipher (27) to map trajectories spanning all T cell clusters. We identified one key latent factor describing a trajectory toward terminally differentiated states (fig. S8A) and a second discriminating between CD4 and CD8 subsets (Fig. 5A). When projecting cells on these latent factors, we observed a trajectory recapitulating T cell differentiation on the basis of gene expression profiling within our CD8+ T cell clusters. T cells from AML-Rs skewed toward effector states (enriched in GZMH, CCL5, and NKG7; Fig. 5B and fig. S8, B and C), whereas those of AML-NRs skewed toward terminal differentiation with higher expression of coinhibitory molecules (TIGIT, KLRG1, and TCF7) (distribution shift in terminal differentiation trajectory, P = 2.2 × 10−11, Kolmogorov-Smirnov test) (Fig. 5, A and B, and fig. S8D).
Fig. 5. Characterization of CD8 ZNF683Hi CTLs in comparison with other T cells.
(A) 2D scatterplot of Decipher components for terminal differentiation (x axis) and CD4 versus CD8 (y axis) (bottom). Distribution of T cells along the terminal differentiation axis, Rs versus NRs, P value by Kolmogorov-Smirnov test (n = 16,182 cells) (top). Dots: cells colored by phenotypic cluster. Numbers: alphanumeric cluster identifier to which the cells belong. Ellipses were manually drawn to separate the CD4 and CD8 clusters. (B) Examples of activation gene expression (ZNF683 and GZMB) and coinhibitory genes (TIGIT and GZMK) along the terminal differentiation axis. (C) Comparison of gene expression profiles of the CD8 T and NK cell subsets from the current study (columns) versus other published datasets (37, 57, 58). Dot size: percentage of cells in each cluster aligned with the expression profile from the external dataset. Dot color: probability of similar expression to the external dataset cluster. (D) 2D UMAP with cells from HSCT-naive individuals (blue) and post-HSCT individuals (pink) (left) from (60). Coloring of UMAP showing ZNF683 expression (right). (E) Percentage of CD8 ZNF683Hi T cells of all CD8 T cells in HSCT-naive (blue, n = 10) and post-HSCT (pink, n = 8) patients (t test).
Comparison of our T cell clusters with those in previous TME studies confirmed the CD8+CTL and TEMRA characteristics of C0: CD8+ZNF683Hi CTLs (Fig. 5C). Gene expression signatures of our six CD8+ clusters compared with T cell MCs from a large scRNA-seq pan-cancer atlas of tumor-infiltrating CD8 T cells curated from 316 patients across 21 cancer types (57) showed that C0: CD8+ZNF683Hi CTLs most closely aligned with the CD8 MC signature c06.Temra.CX3CR1 (Fig. 5C, left). A key difference between this MC and C0: CD8+ZNF683Hi CTLs, however, was the high expression of ZNF683 in the latter (57). We also evaluated profiles of marrow-derived T and NK cells from healthy volunteers and from patients with AML treated with immune checkpoint blockade (ICB) (58). C0: CD8+ZNF683Hi CTLs most closely aligned with CD8+ CTL populations from the marrow of patients with AML but not healthy marrow T cells (Fig. 5C, middle). Last, we compared our T cell populations with a recent study of Richter syndrome, which identified a population of ZNF683Hi CD8+ T cells associated with response to PD-1 ICB (37). C0: CD8+ZNF683Hi CTLs were most transcriptionally similar to the ZNF683-intermediate (ZNF683Int) CD8+ T cell population, followed by the ZNF683Hi CD8+ subset (Fig. 5C, right).
Examination of our C0: CD8+ZNF683Hi CTLs demonstrated expression of some genes more commonly associated with NK cells (e.g., NKG7, B3GAT1, and FCGR3A), raising the possibility that this cluster had NK-like properties. A recent report identified an NK-like population of CD8+ T cells, expanded by cytomegalovirus (CMV) exposure and defined by low BCL11B expression, with effector function against leukemia cell lines (59). To determine whether our C0: CD8+ZNF683Hi CTLs were compatible with this previously described NK-like population, we compared our NK and T cell clusters with that of the NK-like population but did not find concordance between expression profiles (fig. S8E), confirming C0: CD8+ZNF683Hi CTLs as distinct from the previously described CMV-reactive NK-like CD8 population.
We asked whether C0: CD8+ZNF683Hi CTLs were enriched in the post-HSCT setting rather than other AML treatment settings and therefore reanalyzed two scRNA-seq datasets generated from marrow and blood. We reclustered marrow-derived T cells collected from patients with post-HSCT–relapsed or HSCT-naive–relapsed myelodysplastic syndrome (MDS) or AML after treatment with the DNA methyltransferase inhibitor decitabine and ipilimumab and identified four clusters with ZNF683Hi expression (cluster IDs 2, 3, 4, and 10) similar to C0: CD8+ZNF683Hi CTLs (fig. S8, F to H) (60).These cells were more prevalent in post-HSCT patients than in non-HSCT patients (P = 0.01, Mann-Whitney U), supporting the notion that they arise in the setting of immune reconstitution after HSCT (Fig. 5, D and E). We then reanalyzed the BM T cells from the aforementioned ICB-treated AML cohort (58) and identified a subset of ZNF683Hi CD8+ T cells, most consistent with C0: CD8+ZNF683Hi CTLs (fig. S8, I and J). We detected a small increase in this population in Rs after ICB (P = 0.27) but not NRs (P = 0.85, paired t test; fig. S8K).
Given that C0 was present in both AML-Rs and AML-NRs, we hypothesized that distinct properties of the cells might explain their association with response. DEG analysis among C0: CD8+ZNF683Hi CTLs identified ZNF683 as highly differentially expressed in AML-Rs versus AML-NRs (P < 0.0001; Fig. 6A and data file S10). AML-R C0: CD8+ZNF683Hi CTLs also displayed higher expression of activation genes (NKG7, STAT1, JUNB, and IFNG). In contrast, AML-NR cells highly expressed KLRB1 (encoding CD161), an inhibitor of tumor-infiltrating lymphocyte (TIL) cytotoxicity (61), and multiple metallothionein pathway genes (MT1X, MT2A, MT1F, and MT1E) associated with TIL dysfunction (Fig. 6A) (62).
Fig. 6. Phenotypic and functional characterization of CD8+ZNF683Hi CTLs.
(A) Top DEGs of AML C0: CD8+ZNF683Hi CTLs in Rs (right) versus NRs (left). (B) CellPhoneDB networks visualized using Cytoscape (85) with interactions between T cell clusters C0: CD8+ZNF683Hi CTLs and C40 (blue nodes) with AML-enriched clusters C10, C15, and C42 (gray nodes). (C and E) Representative flow cytometry plots of CD137 (C) or IFN-γ (E) expression in CD8 TEMRA cells alone from Rs (top left) or NRs (top right) and CD8 TEMRA cells cocultured with patient-matched AML cells from Rs (bottom left) or NRs (bottom right). (D and F) Quantification of CD137 expression (D) or IFN-γ (F) across six AML-R and three AML-NR PBMC samples cultured with or without patient-matched AML cells. Lines connect samples from the same participant. P values by t test, corrected for multiple comparisons with Benjamini-Hochberg. (G) Violin plot of CD8 TEMRA signature scores of cells sorted for CD8+CD137+ (left; n = 2265) versus CD8+CD137 (right; n = 19,990 cells) after in vitro coculture with patient-matched leukemia cells aggregated across three independent Rs. P value by Mann-Whitney U test, P < 0.000001 for significance after Bonferroni correction. (H) Percentage of C0: CD8+ZNF683Hi CTLs of all DLI product cells for Rs versus NRs. P value by t test.
To better define potential pathways of interest in AML-R versus AML-NR C0: CD8+ZNF683Hi CTLs and leukemia cells, we applied CellPhoneDB (63) and CellChat (64) to identify possible receptor-ligand (R-L) pairs between these clusters (data file S11). AML-NRs demonstrated inferred interactions between the coinhibitory marker TIGIT on CD8+ CTLs and NECTIN2 on AML leukemia cells (Fig. 6B and data file S11) (6567). The TIGIT axis is a well-described immune checkpoint pathway for T and NK cell activation (with CD96 or CD226) and T cell inhibition [with Nectin-2 or poliovirus receptor (PVR)] (44, 68). Accordingly, potential NECTIN2 or PVR interactions with CD226 but not TIGIT were inferred by CellPhoneDB in AML-Rs, corroborating a more activated profile in Rs (Fig. 6B). Thus, C0: CD8+ZNF683Hi CTLs from AML-Rs are predicted to be more highly activated and cytotoxic after DLI compared with cells from AML-NRs that are more inhibited and dysfunctional, consistent with their potential roles in GvL response and resistance, respectively.

CD8+ZNF683Hi CTLs define a CD8+ T cell population with leukemia-specific activity

To test whether CD8+ CTLs had functional antileukemic activity, we devised an in vitro coculture assay to evaluate leukemia-specific T cell activation of CD8+ CTLs after encounter with leukemia cells from the same patient (fig. S9A). CD8+ TEMRA CTLs from six AML-Rs were more activated and cytotoxic, expressing higher CD137 (P = 0.024, t test, corrected with Benjamini-Hochberg) and interferon-γ (IFN-γ) (P = 0.003, t test) after coculture with primary patient-matched leukemia cells than CD8+ TEMRA cells from three AML-NRs (Fig. 6, C to F). CD8+ CTLs cultured in the absence of leukemia cells (Fig. 6, C to F) or in the presence of patient-matched (recipient) BM fibroblasts (fig. S9, B and C) did not show elevated expression of either CD137 or IFN-γ, suggesting leukemia-specific activation in Rs but not NRs. This activity was not observed in CD8+ T cell subsets aside from TEMRA cells (fig. S9, D and E), indicating leukemia-specific activation of CD8+ CTLs, consistent with a potential functional role of these cells in AML DLI Rs.
To determine whether the activated and cytotoxic-appearing in vitro population was consistent with the C0: CD8+ZNF683Hi CTLs implicated in GvL by scRNA-seq, we performed the same coculture experiment on samples from three additional Rs. Cells were then sorted by flow cytometry into CD8+CD137+ and CD8+CD137 populations and analyzed by scRNA-seq and scTCR-seq (fig. S9, F and G). The CD8+CD137+ population was enriched for a CD8+ TEMRA/CTL gene expression signature compared with the CD8+CD137 population (fig. S9, H and I), demonstrating similarity to C0: CD8+ZNF683Hi CTLs (Fig. 6G and fig. S9J). Together, these characterizations provided independent support for a likely role of ZNF683-expressing CD8+ CTLs in mediating antileukemia responses.

CD8+ CTLs from the DLI product undergo clonal expansion in AML-Rs

Because expansion of the C0: CD8+ZNF683Hi CTL population is central to the antileukemia response, we asked whether the increased proportion of C0: CD8+ZNF683Hi CTLs in AML-Rs after DLI was due to a higher proportion in the DLI product. Analysis of cell type proportions within the DLI products of four AML-Rs and three AML-NRs revealed no differences in C0: CD8+ZNF683Hi CTL percentages (P = 0.88) in other expanding BM clusters (Fig. 6H and fig. S10A) or in other major cell types (fig. S10B). Alternatively, CD8+ZNF683Hi CTLs may have undergone greater clonal expansion after infusion because of increased support from a more immunologically diverse BM microenvironment. In support of this hypothesis, scTCR-seq analysis of T cell clonotypes within BM and DLI products of AML-Rs demonstrated marked clonal expansion primarily in C0: CD8+ZNF683Hi CTLs (Fig. 7, A to C). Most (63%) clonally expanded cells derived from the post-DLI BM, with 78% of the post-BM expanded clones found in C0: CD8+ZNF683Hi CTLs. Expanded clones expressed more ZNF683, whereas unexpanded clones expressed more TIGIT, corroborating our R versus NR DEGs (Fig. 7D). The C28: CD8 TUnconv cluster was noted to be highly clonal. Examination of this cluster revealed that 91.7% of the cluster (609 of 664 cells) comprised mucosal-associated invariant T (MAIT) cells, explaining their clonal/invariant nature. To determine whether clonal expansion correlated with T cell activation or inhibition, we analyzed the fold change of genes expressed in C0: CD8+ZNF683Hi CTLs versus C5: CD8 TEX cells and their relationship with clonal expansion (Fig. 7E). Activation genes (e.g., ZNF683, GZMB, PRF1, and CX3CR1) were more highly expressed in C0: CD8+ZNF683Hi CTLs, associating with increased clonal expansion, whereas coinhibitory markers (e.g., TIGIT, LTB, and GZMK) were more highly expressed in C5: CD8 TEX cells, anticorrelated to clonality. These data corroborate our in vitro findings that C0: CD8+ CTLs become activated and clonally expanded in response to AML cells.
Fig. 7. Donor CD8+ZNF683Hi CTLs undergo clonal expansion in AML-Rs.
(A) 2D UMAP of all BM (blue) or DLI product (orange) T cells from patients with AML. (B) Coloring of 2D UMAPs of BM (left) and DLI product (right) T cells by cluster. (C) Coloring of 2D UMAP by degree of clonal expansion for each T cell clone for all T cells (n = 27,159, left), BM T cells (n = 10,199, middle), and DLI product T cells (n = 16,960, right). Yellow: greater clonal expansion; blue: clones with a single member. (D) 2D UMAPs demonstrating average expression of ZNF683 and TIGIT. Higher gene expression indicated by green or yellow. (E) Correlation plot of increasing clonality (x axis) with greater expression of genes in C0: CD8+ZNF683Hi CTLs versus C5: CD8 TEX cells. (F) Bar plot: number of expanded clonotypes shared between DLI product and post-DLI BM (blue) versus those shared between pre-DLI BM and post-DLI BM (red) or found in all three (green) for each participant. Stars indicate clonally expanded TCRs specific to known viral epitopes. (G) Schema of the proposed model.
We next examined whether the C0: CD8+ZNF683Hi CTLs originated from the recipient’s BM or the DLI product on the basis of T cell receptor (TCR) clonotype tracking. Clonal expansion and activation of C0: CD8+ZNF683Hi CTLs were primarily due to expansion of T cells from the DLI product, not the pre-DLI BM (Fig. 7F and fig. S10, C to E) (11, 17, 69). This finding was corroborated by down-sampling analysis to control for the variable number of T cells profiled (Fig. 7F, fig. S10F, and data file S12). Furthermore, higher TCR diversity was observed in the post-DLI BM of Rs compared with NRs (P = 0.01, t test on Gini coefficient; fig. S10G). TCR diversity within DLI products or the pre-DLI BM did not differ between AML-Rs and AML-NRs (P = 0.59 and P = 0.6, respectively, t test; fig. S10G), suggesting that the phenotypic state of T cells and their communication with other immune cells in the BM microenvironment were the primary driver of C0: CD8+ZNF683Hi CTL activation and clonal expansion after DLI, rather than differences in the T cell clonality or composition of the DLI product. Thus, our findings support the notion that DLI infusion in the less exhausted, more immunologically diverse R BM microenvironment promotes clonal expansion of highly activated effector C0: CD8+ZNF683Hi CTLs with GvL cytotoxic activity (Fig. 7G). Conversely, in NRs, infusion into the exhausted, less-diverse BM microenvironment (i.e., TIGIT-high) prevents expansion and cytotoxicity of C0: CD8+ZNF683Hi CTLs, leading to TIL dysfunction and therapy resistance.

DISCUSSION

Highly coordinated immune cell networks enriched in cytotoxic T cells, in complex with B and myeloid cells, have been identified as key features across multiple solid cancers that respond to immunotherapy (57, 4850, 70). For myeloid malignancies, such immune networks have been only minimally evaluated (71). AML responses to ICB in clinical trials have been disappointing (3, 4), and defining the role of immune cell cross-talk could advance our understanding of effective antileukemia response. Using integrated single-cell analysis of deeply clinically annotated patients with AML treated with immunotherapy, time-resolved cell-cell interaction prediction, transcriptional and protein-level confirmation in an independent cohort, and functional analysis, we identified previously unappreciated cell types linked to effective GvL. Our machine learning frameworks for orthogonal spatial and temporal analysis in complex clinical multiomics data also provide a roadmap for reverse-engineering impacts of therapies. In particular, the adaptation and application of the Bayesian model DIISCO establish its ability to infer dynamic cell cross-talk that may be associated with response to therapy, using heterogeneous patient specimens with variable timing.
Our first major insight was the notably different organization of cellular populations within the marrow of AML-Rs versus AML-NRs. Rs demonstrated coordinated dynamic expansion of T and NK cells with predicted direct interactions with leukemia cells, whereas NRs revealed relative “neglect” from infiltrating immune cell populations. This concept is reminiscent of the “hot” and “cold” tumor paradigm in ICB response (2, 72, 73). Our predictions were confirmed spatially by CODEX, which further revealed colocalization of T and B cells within immunological communities in the AML-R BM microenvironment and a preexisting preponderance of colocalization with myeloid cells in AML-NRs. Although our discovery analysis has limitations, including a small cohort, disparity in timing of post-DLI samples between Rs and NRs, and some unpaired samples in CODEX, our findings illuminate cellular and molecular mechanisms of effective GvL, which are supported by orthogonal confirmation through single-cell spatial transcriptomic profiling. Together, our analyses support a central role for higher diversity of immune cell types locally interacting and coordinating with an evolving profile of T cells to promote effector T cell activity in AML-Rs, whereas the inhibitory AML-NR BM microenvironment prevents effective GvL.
Even as we observed multiple immune cell populations creating a network of activity, these appeared to converge upon a distinct CD8+CTL population as the key feature distinguishing response. Thus, our second major insight was identifying CD8+ZNF683Hi CTLs as a central hub associated with GvL in AML, validated by IFN-γ and CD137 up-regulation in coculture and flow cytometry confirmation of expansion of this population in an independent cohort. Our work aligns with other recent studies that point to ZNF683-expressing CD8+ T cells as mediators of antitumor responses (37, 7480). Conversely, AML-NR CD8+ CTLs were characterized by relatively lower expression of ZNF683 and high expression of the inhibitory/terminal differentiation markers TIGIT and KLRG1, suggesting a less diverse BM microenvironment with impaired effector function as a contributor of resistance to GvL.
Third, our work demonstrated that the C0: CD8+ZNF683Hi CTL population of leukemia-reactive T cells in AML-Rs originated primarily from the DLI product, on the basis of TCR clone tracking, despite similar DLI product composition between AML-Rs and AML-NRs. We therefore inferred that, whereas the major cellular effectors of GvL (the C0: CD8+ZNF683Hi CTL population) in AML derive from the DLI product, their subsequent activation, expansion, and sustained immune coordination depend on support by a recipient BM microenvironment that is more diverse and less immune inhibitory. This is in marked contrast with our prior findings in CML, where DLI rather provided “immunologic help” to preexisting marrow-resident TEX cells, further supported by our findings in patients with CML receiving conventional CD8-replete DLI. Thus, our results in AML raise the notion of disease-specific mechanisms by which GvL is mediated and hence the ability to tailor adoptive cellular therapy on the basis of disease context. Another hypothesis that is not mutually exclusive with the role of a more diverse BM microenvironment is that expansion of the C0: CD8+ZNF683Hi CTLs is facilitated by encounter with cognate antigen–bearing cells (either leukemia cells or other antigen-presenting cells) in Rs, but these antigens are absent in NRs.
Our work not only presents opportunities for translational impact but also provides a roadmap for inquiry. We observed high TIGIT expression associated with GvL resistance, suggesting possible interactions with PVR or NECTIN2 on leukemia cells, although the existence of these interactions requires further experimental validation. Given the relative resistance of AML to ICB (3, 4), TIGIT may be a potential target for a subset of patients with AML. The intrinsic antileukemia activity of CD8+ZNF683Hi CTLs makes them an interesting candidate for cellular therapy for AML. That these cells derive primarily from the DLI product provides the opportunity for graft optimization and engineering. Given the increasingly evident role for CD8+ZNF683Hi CTLs in association with immunotherapy response in other cancers (37, 76, 78), understanding the mechanisms of antitumor activity in this population has broad implications for improving immunotherapy across tumor types. An area of interest is discerning the antigen specificity of these cells because the expression of cognate antigen(s) most likely drives the clonal expansion of this population in AML-Rs. Recent approaches to high-throughput prediction of immunogenic epitopes, including tumor-associated antigens, neoantigens, and minor histocompatibility antigens (81), raise the possibility that the genomic features of relapsed leukemia could also contribute to the fate of this T cell population once infused into the recipient.
As the forebear to modern day adoptive cellular therapy, the study of DLI enables the ready ability to address key questions in this rapidly evolving field, namely, identifying cell populations essential for effective antitumor activity, their interactions with malignant and other immune cells, and their kinetics over time. Elucidating the mechanism underpinning GvL has broad implications for better understanding the drivers of immunotherapy response in cancer.

MATERIALS AND METHODS

Study design

The primary aim of this study was to identify a cell population(s) associated with the response to DLI and effective GvL for relapsed myeloid malignancy after HSCT. We performed scRNA-seq, scTCR-seq, and scCITE-seq on cryopreserved BM biopsy specimens from nine patients with relapsed AML after HSCT who received DLI (five Rs and four NRs) and merged these data with previously generated data from similarly treated patients with relapsed CML after HSCT. Patients with AML had similar leukemia burden at relapse and at the time of DLI and were similar in other disease and treatment characteristics. We further confirmed our major findings with spatial profiling using CODEX and spatial transcriptomic profiling, as well as by flow cytometry in a larger independent cohort. All newly sequenced patient samples were obtained from patients receiving standard of care therapy with conventional CD8-replete DLI (i.e., not in a clinical trial). All samples analyzed were obtained from patients who had provided written informed consent for use of clinical data and samples on a biobanking protocol approved by the Institutional Review Board (protocol number 01206) at the Dana-Farber Cancer Institute/Harvard Cancer Center. These studies were conducted in accordance with the Declaration of Helsinki.

Patient DLI characteristics

The patients with AML in this cohort were treated with DLI at the Dana-Farber Cancer Institute, Boston, between 2004 and 2021. Patients had similar baseline characteristics including HLA (human leukocyte antigen) match of donor, recipient and donor sex, time from HSCT to relapse, time from HSCT to DLI, and timing of pre-DLI samples (fig. S1, A and B, and data file S2). Rs had a longer duration of post-DLI marrow sampling because of longer survival after DLI. Patients in both groups had comparable disease burden at relapse, defined as percentage of BM blasts (fig. S1C). Three of five Rs and two of four NRs received cytoreducing therapy between relapse and DLI in an attempt to achieve remission before DLI (data file S2). Two Rs and one NR had minimal disease (i.e., <5% blasts but marrow dysplasia and/or progressive cytopenias and/or recurrence of mutations by next-generation sequencing) at relapse and did not receive therapy before DLI given the minimal disease burden. All patients except one NR had <5% blasts at the time of DLI. The exception was NR3512, whose disease at relapse was considered as smoldering, and therefore DLI was pursued without prior therapy despite 16% marrow blasts.
All patients received between one and three doses of DLI (median of 1 for Rs and 1.5 for NRs), with a median dose of 1 × 107 CD3+ cells/kg for Rs versus 1.6 × 107 CD3+ cells/kg for NRs (data file S2). Response was defined as durable morphologic and molecular remission for at least 1 year after DLI, whereas NRs lacked posttreatment reduction in disease burden. A median of three (range two to six) serial marrow biopsy specimens collected before and after DLI were evaluated per patient (Fig. 1A and data file S1). Response was defined as durable morphologic and molecular (i.e., cytogenetic and/or next-generation sequencing detection of disease-associated mutations) remission for at least 1 year after DLI, whereas nonresponse was defined as a lack of post-DLI BM blast reduction. Two patients who never relapsed were included as nonrelapse controls. Pre– and post–chemotherapy treatment samples from three patients who had post-HSCT relapsed AML and entered durable (>1 year) remission with chemotherapy but did not subsequently receive DLI were also included as chemotherapy-only (no DLI) controls. The presence of acute and chronic graft-versus-host disease (GVHD) was graded by standard consensus criteria (82); grades 0 and 1 acute GVHD were considered clinically equivalent (83). Characteristics of patients with CML were previously described (11).

Sample collection

BM aspirates and peripheral blood samples were collected and banked before and after DLI. BMMCs or PBMCs were isolated via Ficoll-Hypaque density gradient centrifugation, cryopreserved with 10% dimethyl sulfoxide, and stored in vapor-phase liquid nitrogen until the time of sample processing. For DLI products, 1 ml of the cryopreserved DLI apheresis product was obtained from quality control (QC) vials stored at the time of apheresis.

Sample processing and library preparation for scRNA-seq, scTCR-seq, and scCITE-seq

CML data were obtained from previously described sequencing runs, and all cells were analyzed for the current study, whereas we previously reported only the subset of data originating from T cells (11). For preparation of AML samples, cryopreserved primary BMMCs or PBMCs (for the matched sample from R3501) were thawed on the day of sequencing at 37°C and dispensed into a warmed solution of 10% fetal bovine serum (FBS) and 10% DNase I (StemCell Technologies, catalog no. 07900) in phosphate-buffered saline (PBS). The cell suspension was centrifuged at 200g for 10 min at room temperature. Viable cells were negatively selected using the MACS Dead Cell Removal Kit (Miltenyi Biotec, catalog no. 130-090-101). Collected live cells were resuspended in warmed cell staining buffer (BioLegend). Cells were stained with TotalSeq C hashtag and CITE-seq antibodies (data file S7) according to the manufacturer’s instructions. For batch AML1, after washing three times, cells from four samples were combined into one pool (data file S3, “Sequencing Run” column) with ~20,000 cells from each sample and diluted to a final concentration of 1000 cells/μl in 0.04% UltraPure bovine serum albumin (BSA) (Thermo Fisher Scientific). Seven total pools were processed in this way. Subsequent samples [i.e., batch AML2, DLI products, and control samples (data file S3)] were run individually. Cells were then taken immediately for scRNA-seq, scTCR-seq, and scCITE-seq. Approximately 50,000 cells from pooled samples or 10,000 cells from individual samples were loaded into one lane of a 10X Genomics Chromium instrument according to the manufacturer’s instructions. scRNA-seq libraries were processed using a Chromium Single Cell 5′ Library and gel bead v2 kit (10X Genomics). Coupled scTCR-seq libraries were obtained using a Chromium Single Cell V(D)J Enrichment Kit (human T cell) (10X Genomics), and coupled scCITE-seq libraries were obtained using a Chromium Single Cell Feature Barcode Kit. QC for amplified cDNA libraries and final sequencing libraries was performed using the Bioanalyzer High Sensitivity DNA Kit (Agilent). The scRNA-seq, scTCR-seq, and scCITE-seq libraries were normalized to a 4-nM concentration and pooled in a volume ratio of 4:1. The pooled libraries were sequenced on an Illumina NovaSeq S4 platform. The sequencing parameters were as follows: read 1 of 26 base pairs (bp), read 2 of 90 bp, index 1 of 10 bp, and index 2 of 10 bp. The sequencing data were demultiplexed and processed as described below.

CODEX overview

We imaged seven R samples (four pre-DLI and three post-DLI) from four R and five NR samples (two pre-DLI and three post-DLI) from four NRs (data file S1). Four total samples (two from Rs and two from NRs) were imaged with a confocal microscope only, whereas all other samples were imaged with confocal and wide-field microscopy as well to better capture a low signal in channel 3 (750 nm). Each sample was imaged over 18 cycles, with 33 markers across three channels, excluding 4′,6-diamidino-2-phenylindole (DAPI), which was used in channel 0 (405 nm) for every cycle. Channel 1 (561 nm) markers included CD14, CD34, granzyme B, CD44, CD38, CD21, myeloperoxidase (MPO), CD19, TIGIT, CD8, and Ki67. Channel 2 (647 nm) markers included OX40 [tumor necrosis factor receptor superfamily, member 4 (TNFRSF4)], CD11A, CD152, S100A4, CD74, CD11C, CD68, inducible T cell costimulator (ICOS), CD4, forkhead box P3 (FOXP3), CD3E, LAG3, programmed death-ligand 1 (PDL1), CD34, TIM3, and CD11B. Channel 3 (750 nm) markers included galectin-9, CD57, CD31, CD47, CD20, and vimentin. The entire area of the BM biopsy specimen was considered in the analysis.

In vitro experiments

From six AML DLI Rs (two from the scRNA-seq discovery cohort and four from the flow cytometry validation cohort) and three NRs (one from the scRNA-seq discovery cohort and two from the flow cytometry validation cohort), we obtained peripheral blood after DLI (in remission for Rs) and isolated T cells from PBMCs (human Pan T Cell Isolation Kit, Miltenyi). A total of 1 × 105 T cells were cultured with 1 × 105 leukemia cells from peripheral blood or BM at the time of relapse after HSCT at an effector-to-target ratio of 1:1 or with T cells alone as a negative control. Equal numbers of cells were used for both Rs and NRs. After 18 hours of in vitro coculture, cells were stained for 20 min at 4°C in a cell staining buffer (BioLegend) with surface antibodies (CD34, CD3, CD4, CD8, CD62L, and CD45RA) (data file S7) at a 1:100 dilution. T cell functional state was assessed by IFN-γ catch assay according to the manufacturer’s instructions (Miltenyi). In a separate aliquot of cells from the same well, cells were stained with the same surface antibody cocktail as before along with CD137 to assess for antigen-specific T cell activation (70, 84). Cells were then washed three times before resuspending in a fluorescence-activated cell sorting (FACS) buffer (5% v/v FBS in PBS), and data were acquired using a BD Fortessa II instrument and then analyzed using the FlowJo (Tree Star) software. Cell subsets were defined as follows: CD8+ TEMRA/CTL (CD3+CD8+CD62LCD45RA+) and CD8+ non-TEMRA/CTL (CD3+CD8+CD62LCD45RA and CD3+CD8+CD62L+CD45RA+/−) (fig. S9A). For coculture experiments using BM fibroblasts, cryopreserved BM samples for three R patients were thawed and plated on tissue culture flasks in a medium consisting of Dulbecco’s modified Eagle’s medium (Gibco) supplemented with 20% heat-inactivated FBS, 1% penicillin/streptomycin, 1% Hepes buffer, 1% nonessential amino acids, and 1% sodium pyruvate. The medium was changed the next day to remove nonadherent cells. Adherent cells (fibroblasts) were allowed to grow until confluent, with the medium changed every 3 days. On the day of the coculture experiments, cells were lifted with 0.05% trypsin and plated into three wells of a 96-well plate at a density of 1 × 105 cells per well for each patient. Post-DLI T cells were isolated from peripheral blood by negative selection (Miltenyi) and added to wells containing fibroblasts (or without fibroblasts, for the T cell–only negative control). The coculture experiment was then performed as described above. For the single-cell analysis of CD137+ versus CD137 sorted cells, peripheral blood T cells from three Rs in remission after DLI were obtained and cocultured with patient-matched leukemia cells using the same protocol described above. After 18 hours of coculture, cells were washed in a sterile FACS buffer, stained with CD8 and CD137 antibodies along with a viability dye (Zombie Aqua) for 20 min, and then washed three times and taken for flow cytometry sorting. Live cells were collected from the CD8+CD137+ fraction and the CD8+CD137 fraction. There were too few cells after sorting to perform scCITE-seq staining. Cells were then washed once and resuspended in 0.04% BSA and then taken immediately for scRNA-seq and scTCR-seq using the 10X Genomics instrument and protocol described above.

Statistical analysis

Statistical and graphical analyses were performed using Python (3.8.3) and scipy packages (ttest_ind, mannwhitneyu, f_oneway, and kstest) and the GraphPad Prism software (version 10.0). ANOVA and paired t test were used for analyzing normally distributed data and comparisons with few data points. Mann-Whitney U and Kolmogorov-Smirnov tests were used for nonparametric data. When available, two-sided tests were used. For Fig. 6G and fig. S2F, Bonferroni correction was used to perform multiple-testing correction for determining statistical significance, with alpha set to 0.000001 and 0.001, respectively. For all other analyses, results were considered statistically significant for P < 0.05. For DIISCO analysis, the area represents 1 SD (16th to 84th percentile). For box plots of scRNA-seq data, boxes denote lower and upper quartiles, and whiskers show the full range of data. Outliers are shown as dots and were determined with the seaborn package using a method based on the interquartile range.

Acknowledgments

We thank D. Hearsey and all of the staff at the Ted and Eileen Pasquarello Tissue Bank in Hematologic Malignancies for excellent technical support with banking of clinical samples. We thank the patients for contribution of research samples for this study. We are thankful to D. Pe’er, D. Knowles, N. Beltran-Velez, S. He, L. Shi, M. Pressler, and M. Zhang for helpful discussions and feedback.
Funding: K.M. is supported by the Lubin Family Foundation Scholar Award. S.L. is supported by the National Institutes of Health, National Cancer Institute Research Specialist Award R50CA251956. C.Y.P. is supported by the Columbia University Kaganov Fellowship. L.P. is supported by the American Society of Hematology Scholar Award, the BIH Charité Digital Clinician Scientist Program, Deutsche Forschungsgemeinschaft, the Charité—Universitätsmedizin Berlin, the Berlin Institute of Health at Charité (BIH), the Max-Eder program of the German Cancer Aid (Deutsche Krebshilfe), and the Else Kröner-Fresenius-Stiftung (2023_EKEA.102). P.B. is supported by the CPRIT Scholar in Cancer Research Award, the Amy Strelzer Manasevit Scholar Award from the Be The Match Foundation, and the NIH NCI grant 1K08CA248458-01. C.J.W. is supported by the Lavine Family Foundation. C.J.W., E.A., K.M., and R.J.S. are supported by the Leukemia and Lymphoma Society grant SCOR-22937-22. E.A. is supported by NIH NCI R00CA230195 and NHGRI R01HG012875.
Author contributions: Conceptualization: K.M., C.Y.P., E.A., and C.J.W. Methodology: K.M., C.Y.P., E.A., C.J.W., S.L., J.S., W.L., H.L., K.J.L., and P.B. DIISCO design and analysis: E.A., S.M., and C.Y.P. CODEX design and assay performance: K.M., D.A., C.M., S.K., and S.L.F. CODEX analysis: C.Y.P., E.A., Y.J., J.Y.Z., and C.S. Spatial transcriptomics and protein detection design and assay performance: E.N.G., M.F., M.J.L., and K.M. Spatial transcriptomic analysis: F.R., K.H.G. III, C.Y.P., E.A., and K.M. Statistical analysis design and interpretation: E.A., C.Y.P., and D.S.N. Single-cell and flow cytometry analysis: K.M., E.A., C.Y.P., M.B., L.P., J.R.B., and L.R.O. DLI clinical cohort design, patient care, sample collection, and provision of clinical data: R.J.S. and J.R. Writing—original draft: K.M., C.Y.P., E.A., and C.J.W. Writing—review and editing: K.M., C.Y.P., S.M., M.B., L.P., Y.J., J.Y.Z., C.S., J.R.B., J.S., S.K., W.L., H.L., D.A., C.M., L.R.O., D.S.N., P.B., S.L.F., S.L., K.J.L., J.R., R.J.S., C.J.W., E.A., F.R., K.H.G. III, M.J.L., M.F., and E.N.G.
Competing interests: K.M., C.Y.P, E.A., and C.J.W. are inventors on a licensed, pending international patent application, having serial number USP Serial No. 63/586,686, filed by the Dana-Farber Cancer Institute, directed to certain subject matter related to the CD8+ZNF683Hi CTL population described in this manuscript. C.J.W. is an equity holder of BioNTech Inc. and receives research funding from Pharmacyclics. P.B. reports equity in Agenus, Amgen, Johnson & Johnson, Exelixis, and BioNTech and receives research support from Allogene Therapeutics. D.N. has stock ownership in Madrigal Pharmaceuticals. F.R., K.H.G. III, M.J.L., M.F., and E.N.G. are employees of Singular Genomics. J.R. receives research funding from Kite/Gilead, Novartis, and Oncternal Therapeutics and serves on advisory boards for Clade Therapeutics, Garuda Therapeutics, LifeVault Bio, Novartis, and Smart Immune. K.J.L. reports equity in Standard BioTools Inc. and serves on the scientific advisory board for MBQ Pharma Inc. R.J.S. serves on the board of directors for Be the Match–National Marrow Donor Program and DSMB for BMS and reports personal fees from Vor Biopharma, Smart Immune, Neovii, Astellas, Amgen, Bluesphere Bio, and Jasper. The remaining authors declare that they have no competing interests.
Data and materials availability: Single-cell transcriptome, CITE, and TCR data are publicly available in NCBI’s Database of Genotypes and Phenotype under accession number phs003630.v1.p1 (dbGaP; http://ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs003630.v1.p1) and in the National Center for Biotechnology Information’s Gene Expression Omnibus under accession number GSE255530. The DIISCO method is available at http://github.com/azizilab/DIISCO_public. A permanent archive of the version used in this manuscript is available at 10.5281/zenodo.13823393. All codes used for producing the results and figures in this manuscript are available at http://github.com/azizilab/dli_reproducibility. Tabulated data underlying the figures are provided in data file S13. All other data needed to support the conclusions in the paper are present in the paper or the Supplementary Materials.

Supplementary Materials

The PDF file includes:

Materials and Methods
Figs. S1 to S10
Legends for data files S1 to S13
Legend for movie S1
References (86121)

Other Supplementary Material for this manuscript includes the following:

Data files S1 to S13
Movie S1
MDAR Reproducibility Checklist

REFERENCES AND NOTES

1
H. N. Bell, W. Zou, Beyond the barrier: Unraveling the mechanisms of immunotherapy resistance. Annu. Rev. Immunol. 42, 521–550 (2024).
2
J. Vadakekolathu, S. Rutella, Escape from T cell targeting immunotherapies in acute myeloid leukemia. Blood 143, 2689–2700 (2024).
3
S. Rutella, J. Vadakekolathu, F. Mazziotta, S. Reeder, T.-O. Yau, R. Mukhopadhyay, B. Dickins, H. Altmann, M. Kramer, H. A. Knaus, B. R. Blazar, V. Radojcic, J. F. Zeidner, A. Arruda, B. Wang, H. A. Abbas, M. D. Minden, S. K. Tasian, M. Bornhäuser, I. Gojo, L. Luznik, Immune dysfunction signatures predict outcomes and define checkpoint blockade-unresponsive microenvironments in acute myeloid leukemia. J. Clin. Invest. 132, e159579 (2022).
4
Y. Abaza, A. M. Zeidan, Immune checkpoint inhibition in acute myeloid leukemia and myelodysplastic syndromes. Cells 11, 2249 (2022).
5
K. Pelka, M. Hofree, J. H. Chen, S. Sarkizova, J. D. Pirl, V. Jorgji, A. Bejnood, D. Dionne, W. H. Ge, K. H. Xu, S. X. Chao, D. R. Zollinger, D. J. Lieb, J. W. Reeves, C. A. Fuhrman, M. L. Hoang, T. Delorey, L. T. Nguyen, J. Waldman, M. Klapholz, I. Wakiro, O. Cohen, J. Albers, C. S. Smillie, M. S. Cuoco, J. Wu, M.-J. Su, J. Yeung, B. Vijaykumar, A. M. Magnuson, N. Asinovski, T. Moll, M. N. Goder-Reiser, A. S. Applebaum, L. K. Brais, L. K. DelloStritto, S. L. Denning, S. T. Phillips, E. K. Hill, J. K. Meehan, D. T. Frederick, T. Sharova, A. Kanodia, E. Z. Todres, J. Jané-Valbuena, M. Biton, B. Izar, C. D. Lambden, T. E. Clancy, R. Bleday, N. Melnitchouk, J. Irani, H. Kunitake, D. L. Berger, A. Srivastava, J. L. Hornick, S. Ogino, A. Rotem, S. Vigneau, B. E. Johnson, R. B. Corcoran, A. H. Sharpe, V. K. Kuchroo, K. Ng, M. Giannakis, L. T. Nieman, G. M. Boland, A. J. Aguirre, A. C. Anderson, O. Rozenblatt-Rosen, A. Regev, N. Hacohen, Spatially organized multicellular immune hubs in human colorectal cancer. Cell 184, 4734–4752.e20 (2021).
6
W. H. Fridman, M. Meylan, G. Pupier, A. Calvez, I. Hernandez, C. Sautès-Fridman, Tertiary lymphoid structures and B cells: An intratumoral immunity cycle. Immunity 56, 2254–2269 (2023).
7
B. A. Helmink, S. M. Reddy, J. Gao, S. Zhang, R. Basar, R. Thakur, K. Yizhak, M. Sade-Feldman, J. Blando, G. Han, V. Gopalakrishnan, Y. Xi, H. Zhao, R. N. Amaria, H. A. Tawbi, A. P. Cogdill, W. Liu, V. S. LeBleu, F. G. Kugeratski, S. Patel, M. A. Davies, P. Hwu, J. E. Lee, J. E. Gershenwald, A. Lucci, R. Arora, S. Woodman, E. Z. Keung, P.-O. Gaudreau, A. Reuben, C. N. Spencer, E. M. Burton, L. E. Haydu, A. J. Lazar, R. Zapassodi, C. W. Hudgens, D. A. Ledesma, S. Ong, M. Bailey, S. Warren, D. Rao, O. Krijgsman, E. A. Rozeman, D. Peeper, C. U. Blank, T. N. Schumacher, L. H. Butterfield, M. A. Zelazowska, K. M. McBride, R. Kalluri, J. Allison, F. Petitprez, W. H. Fridman, C. Sautès-Fridman, N. Hacohen, K. Rezvani, P. Sharma, M. T. Tetzlaff, L. Wang, J. A. Wargo, B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).
8
J. H. Chen, L. T. Nieman, M. Spurrell, V. Jorgji, L. Elmelech, P. Richieri, K. H. Xu, R. Madhu, M. Parikh, I. Zamora, A. Mehta, C. S. Nabel, S. S. Freeman, J. D. Pirl, C. Lu, C. B. Meador, J. L. Barth, M. Sakhi, A. L. Tang, S. Sarkizova, C. Price, N. F. Fernandez, G. Emanuel, J. He, K. Van Raay, J. W. Reeves, K. Yizhak, M. Hofree, A. Shih, M. Sade-Feldman, G. M. Boland, K. Pelka, M. J. Aryee, M. Mino-Kenudson, J. F. Gainor, I. Korsunsky, N. Hacohen, Human lung cancer harbors spatially organized stem-immunity hubs associated with response to immunotherapy. Nat. Immunol. 25, 644–658 (2024).
9
I. Tirosh, B. Izar, S. M. Prakadan, M. H. Wadsworth II, D. Treacy, J. J. Trombetta, A. Rotem, C. Rodman, C. Lian, G. Murphy, M. Fallahi-Sichani, K. Dutton-Regester, J.-R. Lin, O. Cohen, P. Shah, D. Lu, A. S. Genshaft, T. K. Hughes, C. G. K. Ziegler, S. W. Kazer, A. Gaillard, K. E. Kolb, A.-C. Villani, C. M. Johannessen, A. Y. Andreev, E. M. Van Allen, M. Bertagnolli, P. K. Sorger, R. J. Sullivan, K. T. Flaherty, D. T. Frederick, J. Jané-Valbuena, C. H. Yoon, O. Rozenblatt-Rosen, A. K. Shalek, A. Regev, L. A. Garraway, Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).
10
E. Azizi, A. J. Carr, G. Plitas, A. E. Cornish, C. Konopacki, S. Prabhakaran, J. Nainys, K. Wu, V. Kiseliovas, M. Setty, K. Choi, R. M. Fromme, P. Dao, P. T. McKenney, R. C. Wasti, K. Kadaveru, L. Mazutis, A. Y. Rudensky, D. Pe’er, Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308.e36 (2018).
11
P. Bachireddy, E. Azizi, C. Burdziak, V. N. Nguyen, C. S. Ennis, K. Maurer, C. Y. Park, Z.-N. Choo, S. Li, S. H. Gohil, N. G. Ruthen, Z. Ge, D. B. Keskin, N. Cieri, K. J. Livak, H. T. Kim, D. S. Neuberg, R. J. Soiffer, J. Ritz, E. P. Alyea, D. Pe’er, C. J. Wu, Mapping the evolution of T cell states during response and resistance to adoptive cellular therapy. Cell Rep. 37, 109992 (2021).
12
M. Binnewies, E. W. Roberts, K. Kersten, V. Chan, D. F. Fearon, M. Merad, L. M. Coussens, D. I. Gabrilovich, S. Ostrand-Rosenberg, C. C. Hedrick, R. H. Vonderheide, M. J. Pittet, R. K. Jain, W. Zou, T. K. Howcroft, E. C. Woodhouse, R. A. Weinberg, M. F. Krummel, Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).
13
A. I. Kraemer, C. Chong, F. Huber, H. Pak, B. J. Stevenson, M. Müller, J. Michaux, E. R. Altimiras, S. Rusakiewicz, L. Simó-Riudalbas, E. Planet, M. Wiznerowicz, J. Dagher, D. Trono, G. Coukos, S. Tissot, M. Bassani-Sternberg, The immunopeptidome landscape associated with T cell infiltration, inflammation and immune editing in lung cancer. Nat. Cancer 4, 608–628 (2023).
14
T. Tang, X. Huang, G. Zhang, Z. Hong, X. Bai, T. Liang, Advantages of targeting the tumor immune microenvironment over blocking immune checkpoint in cancer immunotherapy. Signal Transduct. Target. Ther. 6, 72 (2021).
15
D. L. Porter, E. P. Alyea, J. H. Antin, M. DeLima, E. Estey, J. H. F. Falkenburg, N. Hardy, N. Kroeger, J. Leis, J. Levine, D. G. Maloney, K. Peggs, J. M. Rowe, A. S. Wayne, S. Giralt, M. R. Bishop, K. van Besien, NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: Report from the committee on treatment of relapse after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 16, 1467–1503 (2010).
16
H. J. Kolb, A. Schattenberg, J. M. Goldman, B. Hertenstein, N. Jacobsen, W. Arcese, P. Ljungman, A. Ferrant, L. Verdonck, D. Niederwieser, F. van Rhee, J. Mittermueller, T. de Witte, E. Holler, H. Ansari, European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia, Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood 86, 2041–2050 (1995).
17
R. J. Soiffer, E. P. Alyea, J. Ritz, Immunomodulatory effects of donor lymphocyte infusions following allogeneic bone marrow transplantation. J. Clin. Apher. 10, 139–143 (1995).
18
M. Bleakley, S. R. Riddell, Molecules and mechanisms of the graft-versus-leukaemia effect. Nat. Rev. Cancer 4, 371–380 (2004).
19
N. Cieri, K. Maurer, C. J. Wu, 60 years young: The evolving role of allogeneic hematopoietic stem cell transplantation in cancer immunotherapy. Cancer Res. 81, 4373–4384 (2021).
20
S. Nikiforow, E. P. Alyea, Maximizing GVL in allogeneic transplantation: Role of donor lymphocyte infusions. Hematology Am. Soc. Hematol. Educ. Program 2014, 570–575 (2014).
21
S. Liu, J. B. Iorgulescu, S. Li, M. Borji, I. A. Barrera-Lopez, V. Shanmugam, H. Lyu, J. W. Morriss, Z. N. Garcia, E. Murray, D. A. Reardon, C. H. Yoon, D. A. Braun, K. J. Livak, C. J. Wu, F. Chen, Spatial maps of T cell receptors and transcriptomes reveal distinct immune niches and interactions in the adaptive immune response. Immunity 55, 1940–1952.e5 (2022).
22
A. Rao, D. Barkley, G. S. França, I. Yanai, Exploring tissue architecture using spatial transcriptomics. Nature 596, 211–220 (2021).
23
P. van Galen, V. Hovestadt, M. H. Wadsworth Ii, T. K. Hughes, G. K. Griffin, S. Battaglia, J. A. Verga, J. Stephansky, T. J. Pastika, J. Lombardi Story, G. S. Pinkus, O. Pozdnyakova, I. Galinsky, R. M. Stone, T. A. Graubert, A. K. Shalek, J. C. Aster, A. A. Lane, B. E. Bernstein, Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell 176, 1265–1281.e24 (2019).
24
N. Bejanyan, D. J. Weisdorf, B. R. Logan, H.-L. Wang, S. M. Devine, M. de Lima, D. W. Bunjes, M.-J. Zhang, Survival of patients with acute myeloid leukemia relapsing after allogeneic hematopoietic cell transplantation: A center for international blood and marrow transplant research study. Biol. Blood Marrow Transplant. 21, 454–459 (2015).
25
R. Lopez, B. Li, H. Keren-Shaul, P. Boyeau, M. Kedmi, D. Pilzer, A. Jelinski, I. Yofe, E. David, A. Wagner, C. Ergen, Y. Addadi, O. Golani, F. Ronchese, M. I. Jordan, I. Amit, N. Yosef, DestVI identifies continuums of cell types in spatial transcriptomics data. Nat. Biotechnol. 40, 1360–1369 (2022).
26
J. H. Levine, E. F. Simonds, S. C. Bendall, K. L. Davis, E.-A. D. Amir, M. D. Tadmor, O. Litvin, H. G. Fienberg, A. Jager, E. R. Zunder, R. Finck, A. L. Gedman, I. Radtke, J. R. Downing, D. Pe’er, G. P. Nolan, Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).
27
A. Nazaret, J. L. Fan, V.-P. Lavallée, A. E. Cornish, V. Kiseliovas, I. Masilionis, J. Chun, R. L. Bowman, S. E. Eisman, J. Wang, L. Shi, R. L. Levine, L. Mazutis, D. Blei, D. Pe’er, E. Azizi, Deep generative model deciphers derailed trajectories in acute myeloid leukemia. bioRxiv 566719 [Preprint] (2023). http://doi.org/10.1101/2023.11.11.566719.
28
A. Giustacchini, S. Thongjuea, N. Barkas, P. S. Woll, B. J. Povinelli, C. A. G. Booth, P. Sopp, R. Norfo, A. Rodriguez-Meira, N. Ashley, L. Jamieson, P. Vyas, K. Anderson, Å. Segerstolpe, H. Qian, U. Olsson-Strömberg, S. Mustjoki, R. Sandberg, S. E. W. Jacobsen, A. J. Mead, Single-cell transcriptomics uncovers distinct molecular signatures of stem cells in chronic myeloid leukemia. Nat. Med. 23, 692–702 (2017).
29
T. Gao, R. Soldatov, H. Sarkar, A. Kurkiewicz, E. Biederstedt, P.-R. Loh, P. V. Kharchenko, Haplotype-aware analysis of somatic copy number variations from single-cell transcriptomes. Nat. Biotechnol. 41, 417–426 (2023).
30
A.-C. Villani, R. Satija, G. Reynolds, S. Sarkizova, K. Shekhar, J. Fletcher, M. Griesbeck, A. Butler, S. Zheng, S. Lazo, L. Jardine, D. Dixon, E. Stephenson, E. Nilsson, I. Grundberg, D. McDonald, A. Filby, W. Li, P. L. De Jager, O. Rozenblatt-Rosen, A. A. Lane, M. Haniffa, A. Regev, N. Hacohen, Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).
31
J. D. Burton, R. Stein, A. Chandra, S. Chen, N. Mishra, T. Shah, D. M. Goldenberg, Expression of CD74 by AML blasts and cell lines, and enhanced in vitro cytotoxicity of anti-CD74 antibody after interferon-gamma (IFN-γ) treatment. J. Clin. Oncol. 28, 6576–6576 (2010).
32
H. Zhang, B. Cui, Y. Zhou, X. Wang, W. Wu, Z. Wang, Z. Dai, Q. Cheng, K. Yang, B2M overexpression correlates with malignancy and immune signatures in human gliomas. Sci. Rep. 11, 5045 (2021).
33
J. Lei, Q. Li, Y. Gao, L. Zhao, Y. Liu, Increased PKCα activity by Rack1 overexpression is responsible for chemotherapy resistance in T cell acute lymphoblastic leukemia-derived cell line. Sci. Rep. 6, 33717 (2016).
34
Y. Li, J. Cheng, Y. Li, Y. Jiang, J. Ma, Q. Li, T. Pang, CXCL8 is associated with the recurrence of patients with acute myeloid leukemia and cell proliferation in leukemia cell lines. Biochem. Biophys. Res. Commun. 499, 524–530 (2018).
35
V. P. Čokić, S. Mojsilović, A. Jauković, N. Kraguljac-Kurtović, S. Mojsilović, D. Šefer, O. Mitrović Ajtić, V. Milošević, A. Bogdanović, D. Đikić, P. Milenković, R. K. Puri, Gene expression profile of circulating CD34+ cells and granulocytes in chronic myeloid leukemia. Blood Cells Mol. Dis. 55, 373–381 (2015).
36
N. A. M. Kragten, F. M. Behr, F. A. Vieira Braga, E. B. M. Remmerswaal, T. H. Wesselink, A. E. Oja, P. Hombrink, A. Kallies, R. A. W. van Lier, R. Stark, K. P. J. M. van Gisbergen, Blimp-1 induces and Hobit maintains the cytotoxic mediator granzyme B in CD8 T cells. Eur. J. Immunol. 48, 1644–1662 (2018).
37
E. M. Parry, C. K. Lemvigh, S. Deng, N. Dangle, N. Ruthen, B. A. Knisbacher, J. Broséus, S. Hergalant, R. Guièze, S. Li, W. Zhang, C. Johnson, J. M. Long, S. Yin, L. Werner, A. Anandappa, N. Purroy, S. Gohil, G. Oliveira, P. Bachireddy, S. A. Shukla, T. Huang, J. D. Khoury, B. Thakral, M. Dickinson, C. Tam, K. J. Livak, G. Getz, D. Neuberg, P. Feugier, P. Kharchenko, W. Wierda, L. R. Olsen, N. Jain, C. J. Wu, ZNF683 marks a CD8 T cell population associated with anti-tumor immunity following anti-PD-1 therapy for Richter syndrome. Cancer Cell 41, 1803–1816.e8 (2023).
38
S. Lopez-Vergès, J. M. Milush, S. Pandey, V. A. York, J. Arakawa-Hoyt, H. Pircher, P. J. Norris, D. F. Nixon, L. L. Lanier, CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood 116, 3865–3874 (2010).
39
E. P. Alyea, R. J. Soiffer, C. Canning, D. Neuberg, R. Schlossman, C. Pickett, H. Collins, Y. Wang, K. C. Anderson, J. Ritz, Toxicity and efficacy of defined doses of CD4+ donor lymphocytes for treatment of relapse after allogeneic bone marrow transplant. Blood 91, 3671–3680 (1998).
40
C. Y. Park, S. Mani, N. Beltran-Velez, K. Maurer, T. Huang, S. Li, S. Gohil, K. J. Livak, D. A. Knowles, C. J. Wu, E. Azizi, A Bayesian framework for inferring dynamic intercellular interactions from time-series single-cell data. Genome Res. 34, 1384–1396 (2024).
41
A. G. Wilson, D. A. Knowles, Z. Ghahramani, Gaussian process regression networks. arXiv:1110.4411 [stat.ML] (2011).
42
S. Li, W. Xing, M. Kirby, S. Zhe, Scalable variational Gaussian process regression networks. arXiv:2003.11489 [cs.LG] (2020).
43
S.-J. Park, M.-J. Kim, H.-B. Kim, C.-D. Kang, S.-H. Kim, Sensitization of imatinib-resistant CML cells to TRAIL-induced apoptosis is mediated through down-regulation of Bcr-Abl as well as c-FLIP. Biochem. J. 420, 73–81 (2009).
44
S. Tahara-Hanaoka, K. Shibuya, Y. Onoda, H. Zhang, S. Yamazaki, A. Miyamoto, S.-I. Honda, L. L. Lanier, A. Shibuya, Functional characterization of DNAM-1 (CD226) interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112). Int. Immunol. 16, 533–538 (2004).
45
Y. Gao, Q. Xu, X. Li, Y. Guo, B. Zhang, Y. Jin, C. Zhu, Y. Shen, P. Yang, Y. Shi, R. Jin, D. Liu, Y. Ouyang, X. Liu, W. Wang, D. Chen, T. Yang, Heterogeneity induced GZMA-F2R communication inefficient impairs antitumor immunotherapy of PD-1 mAb through JAK2/STAT1 signal suppression in hepatocellular carcinoma. Cell Death Dis. 13, 213 (2022).
46
S. Goyama, M. Shrestha, J. Schibler, L. Rosenfeldt, W. Miller, E. O’Brien, B. Mizukawa, T. Kitamura, J. S. Palumbo, J. C. Mulloy, Protease-activated receptor-1 inhibits proliferation but enhances leukemia stem cell activity in acute myeloid leukemia. Oncogene 36, 2589–2598 (2017).
47
N. Mukaida, S.-I. Sasaki, T. Baba, CCL4 signaling in the tumor microenvironment. Adv. Exp. Med. Biol. 1231, 23–32 (2020).
48
J. H. Chen, L. T. Nieman, M. Spurrell, V. Jorgji, P. Richieri, K. H. Xu, R. Madhu, M. Parikh, I. Zamora, A. Mehta, C. S. Nabel, S. S. Freeman, J. D. Pirl, C. Lu, C. B. Meador, J. L. Barth, M. Sakhi, A. L. Tang, S. Sarkizova, C. Price, N. F. Fernandez, G. Emanuel, J. He, K. Van Raay, J. W. Reeves, K. Yizhak, M. Hofree, A. Shih, M. Sade-Feldman, G. M. Boland, K. Pelka, M. Aryee, I. Korsunsky, M. Mino-Kenudson, J. F. Gainor, N. Hacohen, Spatial analysis of human lung cancer reveals organized immune hubs enriched for stem-like CD8 T cells and associated with immunotherapy response. bioRxiv 535379 [Preprint] (2023). http://doi.org/10.1101/2023.04.04.535379.
49
G. Gaglia, M. L. Burger, C. C. Ritch, D. Rammos, Y. Dai, G. E. Crossland, S. Z. Tavana, S. Warchol, A. M. Jaeger, S. Naranjo, S. Coy, A. J. Nirmal, R. Krueger, J.-R. Lin, H. Pfister, P. K. Sorger, T. Jacks, S. Santagata, Lymphocyte networks are dynamic cellular communities in the immunoregulatory landscape of lung adenocarcinoma. Cancer Cell 41, 871–886.e10 (2023).
50
C. M. Schürch, S. S. Bhate, G. L. Barlow, D. J. Phillips, L. Noti, I. Zlobec, P. Chu, S. Black, J. Demeter, D. R. McIlwain, S. Kinoshita, N. Samusik, Y. Goltsev, G. P. Nolan, Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 183, 838 (2020).
51
Y. Goltsev, N. Samusik, J. Kennedy-Darling, S. Bhate, M. Hale, G. Vazquez, S. Black, G. P. Nolan, Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981.e15 (2018).
52
J. G. Casado, R. Soto, O. DelaRosa, E. Peralbo, M. del Carmen Muñoz-Villanueva, L. Rioja, J. Peña, R. Solana, R. Tarazona, CD8 T cells expressing NK associated receptors are increased in melanoma patients and display an effector phenotype. Cancer Immunol. Immunother. 54, 1162–1171 (2005).
53
B. Huang, R. Liu, P. Wang, Z. Yuan, J. Yang, H. Xiong, N. Zhang, Q. Huang, X. Fu, W. Sun, L. Li, CD8+CD57+ T cells exhibit distinct features in human non-small cell lung cancer. J. Immunother. Cancer 8, e000639 (2020).
54
Y. Le Priol, D. Puthier, C. Lécureuil, C. Combadière, P. Debré, C. Nguyen, B. Combadière, High cytotoxic and specific migratory potencies of senescent CD8+ CD57+ cells in HIV-infected and uninfected individuals. J. Immunol. 177, 5145–5154 (2006).
55
E. Bandrés, J. Merino, B. Vázquez, S. Inogés, C. Moreno, M. L. Subirá, A. Sánchez-Ibarrola, The increase of IFN-γ production through aging correlates with the expanded CD8+highCD28CD57+ subpopulation. Clin. Immunol. 96, 230–235 (2000).
56
M. Lawson, Y. Ishitsuka, A. Pawlowski, Z. Hong, J. Koh, K. Gouin, N. Ing, R. Que, Y. Youn, S. Ding, T. Facchini, R. Costello, K. Nelson, J. Stover, H. Lee, S. Duggal, T. Plaziak, K. Cameron, M. Abdu, D. Witters, M. Fabani, E. Glezer, Abstract 6169: High throughput in situ spatial sequencing of proteins and RNA in FFPE tissue. Cancer Res. 84, 6169–6169 (2024).
57
L. Zheng, S. Qin, W. Si, A. Wang, B. Xing, R. Gao, X. Ren, L. Wang, X. Wu, J. Zhang, N. Wu, N. Zhang, H. Zheng, H. Ouyang, K. Chen, Z. Bu, X. Hu, J. Ji, Z. Zhang, Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).
58
H. A. Abbas, D. Hao, K. Tomczak, P. Barrodia, J. S. Im, P. K. Reville, Z. Alaniz, W. Wang, R. Wang, F. Wang, G. Al-Atrash, K. Takahashi, J. Ning, M. Ding, H. C. Beird, J. T. Mathews, L. Little, J. Zhang, S. Basu, M. Konopleva, M. L. Marques-Piubelli, L. M. Solis, E. R. Parra, W. Lu, A. Tamegnon, G. Garcia-Manero, M. R. Green, P. Sharma, J. P. Allison, S. M. Kornblau, K. Rai, L. Wang, N. Daver, A. Futreal, Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat. Commun. 12, 6071 (2021).
59
R. Sottile, M. K. Panjwani, C. M. Lau, A. F. Daniyan, K. Tanaka, J. N. Barker, R. J. Brentjens, J. C. Sun, J.-B. Le Luduec, K. C. Hsu, Human cytomegalovirus expands a CD8+ T cell population with loss of BCL11B expression and gain of NK cell identity. Sci. Immunol. 6, eabe6968 (2021).
60
L. Penter, Y. Liu, J. O. Wolff, L. Yang, L. Taing, A. Jhaveri, J. Southard, M. Patel, N. M. Cullen, K. L. Pfaff, N. Cieri, G. Oliveira, S. Kim-Schulze, S. Ranasinghe, R. Leonard, T. Robertson, E. A. Morgan, H. X. Chen, M. H. Song, M. Thurin, S. Li, S. J. Rodig, C. Cibulskis, S. Gabriel, P. Bachireddy, J. Ritz, H. Streicher, D. S. Neuberg, F. S. Hodi, M. S. Davids, S. Gnjatic, K. J. Livak, J. Altreuter, F. Michor, R. J. Soiffer, J. S. Garcia, C. J. Wu, Mechanisms of response and resistance to combined decitabine and ipilimumab for advanced myeloid disease. Blood 141, 1817–1830 (2023).
61
N. D. Mathewson, O. Ashenberg, I. Tirosh, S. Gritsch, E. M. Perez, S. Marx, L. Jerby-Arnon, R. Chanoch-Myers, T. Hara, A. R. Richman, Y. Ito, J. Pyrdol, M. Friedrich, K. Schumann, M. J. Poitras, P. C. Gokhale, L. N. Gonzalez Castro, M. E. Shore, C. M. Hebert, B. Shaw, H. L. Cahill, M. Drummond, W. Zhang, O. Olawoyin, H. Wakimoto, O. Rozenblatt-Rosen, P. K. Brastianos, X. S. Liu, P. S. Jones, D. P. Cahill, M. P. Frosch, D. N. Louis, G. J. Freeman, K. L. Ligon, A. Marson, E. A. Chiocca, D. A. Reardon, A. Regev, M. L. Suvà, K. W. Wucherpfennig, Inhibitory CD161 receptor identified in glioma-infiltrating T cells by single-cell analysis. Cell 184, 1281–1298.e26 (2021).
62
M. Singer, C. Wang, L. Cong, N. D. Marjanovic, M. S. Kowalczyk, H. Zhang, J. Nyman, K. Sakuishi, S. Kurtulus, D. Gennert, J. Xia, J. Y. H. Kwon, J. Nevin, R. H. Herbst, I. Yanai, O. Rozenblatt-Rosen, V. K. Kuchroo, A. Regev, A. C. Anderson, A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell 166, 1500–1511.e9 (2016).
63
M. Efremova, M. Vento-Tormo, S. A. Teichmann, R. Vento-Tormo, CellPhoneDB: Inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).
64
S. Jin, C. F. Guerrero-Juarez, L. Zhang, I. Chang, R. Ramos, C.-H. Kuan, P. Myung, M. V. Plikus, Q. Nie, Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).
65
J. D. Worboys, K. N. Vowell, R. K. Hare, A. R. Ambrose, M. Bertuzzi, M. A. Conner, F. P. Patel, W. H. Zammit, J. Gali-Moya, K. S. Hazime, K. L. Jones, C. Rey, S. Jonjic, T. L. Rovis, G. M. Tannahill, G. D. S. C. De Matos, J. D. Waight, D. M. Davis, TIGIT can inhibit T cell activation via ligation-induced nanoclusters, independent of CD226 co-stimulation. Nat. Commun. 14, 5016 (2023).
66
J. Yeo, M. Ko, D.-H. Lee, Y. Park, H.-S. Jin, TIGIT/CD226 axis regulates anti-tumor immunity. Pharmaceuticals 14, 200 (2021).
67
K. L. Banta, X. Xu, A. S. Chitre, A. Au-Yeung, C. Takahashi, W. E. O’Gorman, T. D. Wu, S. Mittman, R. Cubas, L. Comps-Agrar, A. Fulzele, E. J. Bennett, J. L. Grogan, E. Hui, E. Y. Chiang, I. Mellman, Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8 T cell responses. Immunity 55, 512–526.e9 (2022).
68
E. Y. Chiang, P. E. de Almeida, D. E. de Almeida Nagata, K. H. Bowles, X. Du, A. S. Chitre, K. L. Banta, Y. Kwon, B. McKenzie, S. Mittman, R. Cubas, K. R. Anderson, S. Warming, J. L. Grogan, CD96 functions as a co-stimulatory receptor to enhance CD8+ T cell activation and effector responses. Eur. J. Immunol. 50, 891–902 (2020).
69
P. Bachireddy, U. Hainz, M. Rooney, O. Pozdnyakova, J. Aldridge, W. Zhang, X. Liao, F. S. Hodi, K. O’Connell, W. N. Haining, N. R. Goldstein, C. M. Canning, R. J. Soiffer, J. Ritz, N. Hacohen, E. P. Alyea III, H. T. Kim, C. J. Wu, Reversal of in situ T cell exhaustion during effective human antileukemia responses to donor lymphocyte infusion. Blood 123, 1412–1421 (2014).
70
G. Oliveira, K. Stromhaug, S. Klaeger, T. Kula, D. T. Frederick, P. M. Le, J. Forman, T. Huang, S. Li, W. Zhang, Q. Xu, N. Cieri, K. R. Clauser, S. A. Shukla, D. Neuberg, S. Justesen, G. MacBeath, S. A. Carr, E. F. Fritsch, N. Hacohen, M. Sade-Feldman, K. J. Livak, G. M. Boland, P. A. Ott, D. B. Keskin, C. J. Wu, Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 596, 119–125 (2021).
71
J. B. Koedijk, I. van der Werf, L. Penter, M. A. Vermeulen, F. Barneh, A. Perzolli, J. I. Meesters-Ensing, M. Fiocco, H. A. de Groot-Kruseman, R. Moeniralam, K. B. Christensen, B. Porter, K. Pfaff, J. S. Garcia, S. J. Rodig, C. J. Wu, H. Hasle, S. Nierkens, M. E. Belderbos, C. M. Zwaan, O. Heidenreich, A multidimensional analysis reveals distinct immune phenotypes and tertiary lymphoid structure-like aggregates in the bone marrow of pediatric acute myeloid leukemia. medRxiv 23286485 [Preprint] (2023). http://doi.org/10.1101/2023.03.03.23286485.
72
J. Galon, D. Bruni, Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18, 197–218 (2019).
73
J. Davidson-Moncada, E. Viboch, S. E. Church, S. E. Warren, S. Rutella, Dissecting the immune landscape of acute myeloid leukemia. Biomedicines 6, 110 (2018).
74
P. A. Szabo, H. M. Levitin, M. Miron, M. E. Snyder, T. Senda, J. Yuan, Y. L. Cheng, E. C. Bush, P. Dogra, P. Thapa, D. L. Farber, P. A. Sims, Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun. 10, 4706 (2019).
75
C. V. Rao, C. Xu, Y. Zhang, A. S. Asch, H. Y. Yamada, Genomic instability genes in lung and colon adenocarcinoma indicate organ specificity of transcriptomic impact on copy number alterations. Sci. Rep. 12, 11739 (2022).
76
M. Kitakaze, M. Uemura, T. Hara, R. Chijimatsu, D. Motooka, T. Hirai, M. Konno, D. Okuzaki, Y. Sekido, T. Hata, T. Ogino, H. Takahashi, N. Miyoshi, K. Ofusa, T. Mizushima, H. Eguchi, Y. Doki, H. Ishii, Cancer-specific tissue-resident memory T cells express ZNF683 in colorectal cancer. Br. J. Cancer 128, 1828–1837 (2023).
77
Y.-C. Lu, L. Jia, Z. Zheng, E. Tran, P. F. Robbins, S. A. Rosenberg, Single-cell transcriptome analysis reveals gene signatures associated with T cell persistence following adoptive cell therapy. Cancer Immunol. Res. 7, 1824–1836 (2019).
78
G. Oliveira, A. M. Egloff, A. B. Afeyan, J. O. Wolff, Z. Zeng, R. D. Chernock, L. Zhou, C. Messier, P. Lizotte, K. L. Pfaff, K. Stromhaug, L. Penter, R. I. Haddad, G. J. Hanna, J. D. Schoenfeld, L. A. Goguen, D. J. Annino, V. Jo, P. Oppelt, P. Pipkorn, R. Jackson, S. V. Puram, R. C. Paniello, J. T. Rich, J. Webb, J. P. Zevallos, M. Mansour, J. Fu, G. P. Dunn, S. J. Rodig, J. Ley, L. G. T. Morris, L. Dunn, C. P. Paweletz, D. Kallogjeri, J. F. Piccirillo, D. R. Adkins, C. J. Wu, R. Uppaluri, Preexisting tumor-resident T cells with cytotoxic potential associate with response to neoadjuvant anti-PD-1 in head and neck cancer. Sci. Immunol. 8, eadf4968 (2023).
79
F. M. Behr, L. Parga-Vidal, N. A. M. Kragten, T. J. P. van Dam, T. H. Wesselink, B. S. Sheridan, R. Arens, R. A. W. van Lier, R. Stark, K. P. J. M. van Gisbergen, Tissue-resident memory CD8+ T cells shape local and systemic secondary T cell responses. Nat. Immunol. 21, 1070–1081 (2020).
80
S. L. Park, S. N. Christo, A. C. Wells, L. C. Gandolfo, A. Zaid, Y. O. Alexandre, T. N. Burn, J. Schröder, N. Collins, S.-J. Han, S. M. Guillaume, M. Evrard, C. Castellucci, B. Davies, M. Osman, A. Obers, K. M. McDonald, H. Wang, S. N. Mueller, G. Kannourakis, S. P. Berzins, L. A. Mielke, F. R. Carbone, A. Kallies, T. P. Speed, Y. Belkaid, L. K. Mackay, Divergent molecular networks program functionally distinct CD8+ skin-resident memory T cells. Science 382, 1073–1079 (2023).
81
N. Cieri, N. Hookeri, K. Stromhaug, L. Li, J. Keating, P. Díaz-Fernández, V. Gómez-García de Soria, J. Stevens, R. Kfuri-Rubens, Y. Shao, K. A. Kooshesh, K. Powell, H. Ji, G. M. Hernandez, J. Abelin, S. Klaeger, C. Forman, K. R. Clauser, S. Sarkizova, D. A. Braun, L. Penter, H. T. Kim, W. J. Lane, G. Oliveira, L. S. Kean, S. Li, K. J. Livak, S. A. Carr, D. B. Keskin, C. Muñoz-Calleja, V. T. Ho, J. Ritz, R. J. Soiffer, D. Neuberg, C. Stewart, G. Getz, C. J. Wu, Systematic identification of minor histocompatibility antigens predicts outcomes of allogeneic hematopoietic cell transplantation. Nat. Biotechnol. 2024, 10.1038/s41587-024-02348-3 (2024).
82
D. Przepiorka, D. Weisdorf, P. Martin, H. G. Klingemann, P. Beatty, J. Hows, E. D. Thomas, 1994 Consensus conference on acute GVHD grading. Bone Marrow Transplant. 15, 825–828 (1995).
83
A. Gratwohl, J. Hermans, J. Apperley, W. Arcese, A. Bacigalupo, G. Bandini, P. di Bartolomeo, M. Boogaerts, A. Bosi, E. Carreras, A. Devergie, A. Ferrant, W. E. Fibbe, F. Frassoni, G. Gahrton, J. Goldman, A. Iriondo, N. Jacobsen, H. J. Kolb, H. Link, M. Michallet, H. G. Prentice, J. Reiffers, F. V. Rhee, T. Ruutu, H. Schwaighofer, J. P. Vernant, T. de Witte, D.Niederwieser for the Working Party Chronic Leukemia of the European Group for Blood and Marrow Transplantation, Acute graft-versus-host disease: Grade and outcome in patients with chronic myelogenous leukemia. Working Party Chronic Leukemia of the European Group for Blood and Marrow Transplantation. Blood 86, 813–818 (1995).
84
M. Wolfl, J. Kuball, W. Y. Ho, H. Nguyen, T. J. Manley, M. Bleakley, P. D. Greenberg, Activation-induced expression of CD137 permits detection, isolation, and expansion of the full repertoire of CD8+ T cells responding to antigen without requiring knowledge of epitope specificities. Blood 110, 201–210 (2007).
85
P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang, D. Ramage, N. Amin, B. Schwikowski, T. Ideker, Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).
86
P. Bankhead, M. B. Loughrey, J. A. Fernández, Y. Dombrowski, D. G. McArt, P. D. Dunne, S. McQuaid, R. T. Gray, L. J. Murray, H. G. Coleman, J. A. James, M. Salto-Tellez, P. W. Hamilton, QuPath: Open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).
87
K. Maurer, H. T. Kim, T. M. Kuczmarski, H. M. Garrity, A. Weber, C. G. Reynolds, D. Liney, C. Cutler, J. H. Antin, J. Koreth, J. Ritz, R. M. Shapiro, R. Romee, C. J. Wu, R. J. Soiffer, S. Nikiforow, V. T. Ho, M. Gooptu, Impact of cryopreservation and transit times of allogeneic grafts on hematopoietic and immune reconstitution. Blood Adv. 5, 5140–5149 (2021).
88
Y. Huang, D. J. McCarthy, O. Stegle, Vireo: Bayesian demultiplexing of pooled single-cell RNA-seq data without genotype reference. Genome Biol. 20, 273 (2019).
89
G. X. Y. Zheng, J. M. Terry, P. Belgrader, P. Ryvkin, Z. W. Bent, R. Wilson, S. B. Ziraldo, T. D. Wheeler, G. P. McDermott, J. Zhu, M. T. Gregory, J. Shuga, L. Montesclaros, J. G. Underwood, D. A. Masquelier, S. Y. Nishimura, M. Schnall-Levin, P. W. Wyatt, C. M. Hindson, R. Bharadwaj, A. Wong, K. D. Ness, L. W. Beppu, H. J. Deeg, C. McFarland, K. R. Loeb, W. J. Valente, N. G. Ericson, E. A. Stevens, J. P. Radich, T. S. Mikkelsen, B. J. Hindson, J. H. Bielas, Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).
90
Y. Kotliarov, R. Sparks, A. J. Martins, M. P. Mulè, Y. Lu, M. Goswami, L. Kardava, R. Banchereau, V. Pascual, A. Biancotto, J. Chen, P. L. Schwartzberg, N. Bansal, C. C. Liu, F. Cheung, S. Moir, J. S. Tsang, Broad immune activation underlies shared set point signatures for vaccine responsiveness in healthy individuals and disease activity in patients with lupus. Nat. Med. 26, 618–629 (2020).
91
R. Lopez, J. Regier, M. B. Cole, M. I. Jordan, N. Yosef, Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).
92
C. Domínguez Conde, C. Xu, L. B. Jarvis, D. B. Rainbow, S. B. Wells, T. Gomes, S. K. Howlett, O. Suchanek, K. Polanski, H. W. King, L. Mamanova, N. Huang, P. A. Szabo, L. Richardson, L. Bolt, E. S. Fasouli, K. T. Mahbubani, M. Prete, L. Tuck, N. Richoz, Z. K. Tuong, L. Campos, H. S. Mousa, E. J. Needham, S. Pritchard, T. Li, R. Elmentaite, J. Park, E. Rahmani, D. Chen, D. K. Menon, O. A. Bayraktar, L. K. James, K. B. Meyer, N. Yosef, M. R. Clatworthy, P. A. Sims, D. L. Farber, K. Saeb-Parsy, J. L. Jones, S. A. Teichmann, Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376, eabl5197 (2022).
93
S. Zundler, E. Becker, M. Spocinska, M. Slawik, L. Parga-Vidal, R. Stark, M. Wiendl, R. Atreya, T. Rath, M. Leppkes, K. Hildner, R. López-Posadas, S. Lukassen, A. B. Ekici, C. Neufert, I. Atreya, K. P. J. M. van Gisbergen, M. F. Neurath, Hobit- and Blimp-1-driven CD4+ tissue-resident memory T cells control chronic intestinal inflammation. Nat. Immunol. 20, 288–300 (2019).
94
L. Parga-Vidal, F. M. Behr, N. A. M. Kragten, B. Nota, T. H. Wesselink, I. Kavazović, L. E. Covill, M. B. P. Schuller, Y. T. Bryceson, F. M. Wensveen, R. A. W. van Lier, T. J. P. van Dam, R. Stark, K. P. J. M. van Gisbergen, Hobit identifies tissue-resident memory T cell precursors that are regulated by Eomes. Sci. Immunol. 6, eabg3533 (2021).
95
J. Li, J. Wilhelmy, M. M. Davis, Characterization of KIR+CD8+ regulatory T cells in humans by scRNA- and TCR-seq. Methods Mol. Biol. 2574, 41–121 (2022).
96
D. A. Braun, K. Street, K. P. Burke, D. L. Cookmeyer, T. Denize, C. B. Pedersen, S. H. Gohil, N. Schindler, L. Pomerance, L. Hirsch, Z. Bakouny, Y. Hou, J. Forman, T. Huang, S. Li, A. Cui, D. B. Keskin, J. Steinharter, G. Bouchard, M. Sun, E. M. Pimenta, W. Xu, K. M. Mahoney, B. A. McGregor, M. S. Hirsch, S. L. Chang, K. J. Livak, D. F. McDermott, S. A. Shukla, L. R. Olsen, S. Signoretti, A. H. Sharpe, R. A. Irizarry, T. K. Choueiri, C. J. Wu, Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell 39, 632–648.e8 (2021).
97
S. M. Kaech, W. Cui, Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).
98
B. C. Miller, D. R. Sen, R. Al Abosy, K. Bi, Y. V. Virkud, M. W. LaFleur, K. B. Yates, A. Lako, K. Felt, G. S. Naik, M. Manos, E. Gjini, J. R. Kuchroo, J. J. Ishizuka, J. L. Collier, G. K. Griffin, S. Maleri, D. E. Comstock, S. A. Weiss, F. D. Brown, A. Panda, M. D. Zimmer, R. T. Manguso, F. S. Hodi, S. J. Rodig, A. H. Sharpe, W. N. Haining, Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).
99
A. Marçais, M. Tomkowiak, T. Walzer, C.-A. Coupet, A. Ravel-Chapuis, J. Marvel, Maintenance of CCL5 mRNA stores by post-effector and memory CD8 T cells is dependent on transcription and is coupled to increased mRNA stability. Eur. J. Immunol. 36, 2745–2754 (2006).
100
J. Bastid, A. Cottalorda-Regairaz, G. Alberici, N. Bonnefoy, J.-F. Eliaou, A. Bensussan, ENTPD1/CD39 is a promising therapeutic target in oncology. Oncogene 32, 1743–1751 (2013).
101
K. A. Mullan, N. de Vrij, S. Valkiers, P. Meysman, Current annotation strategies for T cell phenotyping of single-cell RNA-seq data. Front. Immunol. 14, 1306169 (2023).
102
A. Stewart, J. C.-F. Ng, G. Wallis, V. Tsioligka, F. Fraternali, D. K. Dunn-Walters, Single-cell transcriptomic analyses define distinct peripheral B cell subsets and discrete development pathways. Front. Immunol. 12, 602539 (2021).
103
P. Nieto, M. Elosua-Bayes, J. L. Trincado, D. Marchese, R. Massoni-Badosa, M. Salvany, A. Henriques, J. Nieto, S. Aguilar-Fernández, E. Mereu, C. Moutinho, S. Ruiz, P. Lorden, V. T. Chin, D. Kaczorowski, C.-L. Chan, R. Gallagher, A. Chou, E. Planas-Rigol, C. Rubio-Perez, I. Gut, J. M. Piulats, J. Seoane, J. E. Powell, E. Batlle, H. Heyn, A single-cell tumor immune atlas for precision oncology. Genome Res. 31, 1913–1926 (2021).
104
A. M. Abel, C. Yang, M. S. Thakar, S. Malarkannan, Natural killer cells: Development, maturation, and clinical utilization. Front. Immunol. 9, 1869 (2018).
105
L. Lajoie, N. Congy-Jolivet, A. Bolzec, G. Thibault, Gradual increase of FcγRIIIa/CD16a expression and shift toward IFN-γ secretion during differentiation of CD56dim natural killer cells. Front. Immunol. 8, 1556 (2017).
106
L. F. de Andrade, Y. Lu, A. Luoma, Y. Ito, D. Pan, J. W. Pyrdol, C. H. Yoon, G.-C. Yuan, K. W. Wucherpfennig, Discovery of specialized NK cell populations infiltrating human melanoma metastases. JCI Insight 4, e133103 (2019).
107
Y. Huang, huangyh09/cellSNP: Release v0.1.6 (Zenodo, 2019); http://zenodo.org/record/3516629.
108
L. McInnes, J. Healy, J. Melville, UMAP: Uniform manifold approximation and projection for dimension reduction. arXiv:1802.03426 [stat.ML] (2018).
109
N. Novershtern, A. Subramanian, L. N. Lawton, R. H. Mak, W. N. Haining, M. E. McConkey, N. Habib, N. Yosef, C. Y. Chang, T. Shay, G. M. Frampton, A. C. B. Drake, I. Leskov, B. Nilsson, F. Preffer, D. Dombkowski, J. W. Evans, T. Liefeld, J. S. Smutko, J. Chen, N. Friedman, R. A. Young, T. R. Golub, A. Regev, B. L. Ebert, Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144, 296–309 (2011).
110
S. L. Wolock, R. Lopez, A. M. Klein, Scrublet: Computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).
111
D. Türei, T. Korcsmáros, J. Saez-Rodriguez, OmniPath: Guidelines and gateway for literature-curated signaling pathway resources. Nat. Methods 13, 966–967 (2016).
112
F. A. Wolf, P. Angerer, F. J. Theis, SCANPY: Large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).
113
ANTs Contributors, ANTsPy documentation—ANTsPy master documentation; http://antspyx.readthedocs.io/en/latest/index.html#.
114
T. Peng, K. Thorn, T. Schroeder, L. Wang, F. J. Theis, C. Marr, N. Navab, A BaSiC tool for background and shading correction of optical microscopy images. Nat. Commun. 8, 14836 (2017).
115
J. Markel, The SIFT algorithm for fundamental frequency estimation. IEEE Trans. Audio Electroacoust. 20, 367–377 (1972).
116
N. F. Greenwald, G. Miller, E. Moen, A. Kong, A. Kagel, T. Dougherty, C. C. Fullaway, B. J. McIntosh, K. X. Leow, M. S. Schwartz, C. Pavelchek, S. Cui, I. Camplisson, O. Bar-Tal, J. Singh, M. Fong, G. Chaudhry, Z. Abraham, J. Moseley, S. Warshawsky, E. Soon, S. Greenbaum, T. Risom, T. Hollmann, S. C. Bendall, L. Keren, W. Graf, M. Angelo, D. Van Valen, Whole-cell segmentation of tissue images with human-level performance using large-scale data annotation and deep learning. Nat. Biotechnol. 40, 555–565 (2022).
117
S. Dicks, P. A., J. Pintar, T. Korten, A. Kumar, I. Virshup, P. Metzger, Scverse/rapids_singlecell: v0.10.6 (Zenodo, 2024); http://zenodo.org/doi/10.5281/zenodo.12533399.
118
I. Korsunsky, N. Millard, J. Fan, K. Slowikowski, F. Zhang, K. Wei, Y. Baglaenko, M. Brenner, P.-R. Loh, S. Raychaudhuri, Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
119
V. A. Traag, L. Waltman, N. J. van Eck, From Louvain to Leiden: Guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).
120
D. van Dijk, R. Sharma, J. Nainys, K. Yim, P. Kathail, A. J. Carr, C. Burdziak, K. R. Moon, C. L. Chaffer, D. Pattabiraman, B. Bierie, L. Mazutis, G. Wolf, S. Krishnaswamy, D. Pe’er, Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).
121
Y. Hao, S. Hao, E. Andersen-Nissen, W. M. Mauck III, S. Zheng, A. Butler, M. J. Lee, A. J. Wilk, C. Darby, M. Zager, P. Hoffman, M. Stoeckius, E. Papalexi, E. P. Mimitou, J. Jain, A. Srivastava, T. Stuart, L. M. Fleming, B. Yeung, A. J. Rogers, J. M. McElrath, C. A. Blish, R. Gottardo, P. Smibert, R. Satija, Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Neither embedded figures nor equations with special characters can be submitted, and we discourage the use of figures and equations within eLetters in general. If a figure or equation is essential, please include within the text of the eLetter a link to the figure, equation, or full text with special characters at a public repository with versioning, such as Zenodo. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science Immunology
Volume 10 | Issue 103
January 2025

Article versions

Submission history

Received: 13 June 2024
Accepted: 18 December 2024

Permissions

Request permissions for this article.

Acknowledgments

We thank D. Hearsey and all of the staff at the Ted and Eileen Pasquarello Tissue Bank in Hematologic Malignancies for excellent technical support with banking of clinical samples. We thank the patients for contribution of research samples for this study. We are thankful to D. Pe’er, D. Knowles, N. Beltran-Velez, S. He, L. Shi, M. Pressler, and M. Zhang for helpful discussions and feedback.
Funding: K.M. is supported by the Lubin Family Foundation Scholar Award. S.L. is supported by the National Institutes of Health, National Cancer Institute Research Specialist Award R50CA251956. C.Y.P. is supported by the Columbia University Kaganov Fellowship. L.P. is supported by the American Society of Hematology Scholar Award, the BIH Charité Digital Clinician Scientist Program, Deutsche Forschungsgemeinschaft, the Charité—Universitätsmedizin Berlin, the Berlin Institute of Health at Charité (BIH), the Max-Eder program of the German Cancer Aid (Deutsche Krebshilfe), and the Else Kröner-Fresenius-Stiftung (2023_EKEA.102). P.B. is supported by the CPRIT Scholar in Cancer Research Award, the Amy Strelzer Manasevit Scholar Award from the Be The Match Foundation, and the NIH NCI grant 1K08CA248458-01. C.J.W. is supported by the Lavine Family Foundation. C.J.W., E.A., K.M., and R.J.S. are supported by the Leukemia and Lymphoma Society grant SCOR-22937-22. E.A. is supported by NIH NCI R00CA230195 and NHGRI R01HG012875.
Author contributions: Conceptualization: K.M., C.Y.P., E.A., and C.J.W. Methodology: K.M., C.Y.P., E.A., C.J.W., S.L., J.S., W.L., H.L., K.J.L., and P.B. DIISCO design and analysis: E.A., S.M., and C.Y.P. CODEX design and assay performance: K.M., D.A., C.M., S.K., and S.L.F. CODEX analysis: C.Y.P., E.A., Y.J., J.Y.Z., and C.S. Spatial transcriptomics and protein detection design and assay performance: E.N.G., M.F., M.J.L., and K.M. Spatial transcriptomic analysis: F.R., K.H.G. III, C.Y.P., E.A., and K.M. Statistical analysis design and interpretation: E.A., C.Y.P., and D.S.N. Single-cell and flow cytometry analysis: K.M., E.A., C.Y.P., M.B., L.P., J.R.B., and L.R.O. DLI clinical cohort design, patient care, sample collection, and provision of clinical data: R.J.S. and J.R. Writing—original draft: K.M., C.Y.P., E.A., and C.J.W. Writing—review and editing: K.M., C.Y.P., S.M., M.B., L.P., Y.J., J.Y.Z., C.S., J.R.B., J.S., S.K., W.L., H.L., D.A., C.M., L.R.O., D.S.N., P.B., S.L.F., S.L., K.J.L., J.R., R.J.S., C.J.W., E.A., F.R., K.H.G. III, M.J.L., M.F., and E.N.G.
Competing interests: K.M., C.Y.P, E.A., and C.J.W. are inventors on a licensed, pending international patent application, having serial number USP Serial No. 63/586,686, filed by the Dana-Farber Cancer Institute, directed to certain subject matter related to the CD8+ZNF683Hi CTL population described in this manuscript. C.J.W. is an equity holder of BioNTech Inc. and receives research funding from Pharmacyclics. P.B. reports equity in Agenus, Amgen, Johnson & Johnson, Exelixis, and BioNTech and receives research support from Allogene Therapeutics. D.N. has stock ownership in Madrigal Pharmaceuticals. F.R., K.H.G. III, M.J.L., M.F., and E.N.G. are employees of Singular Genomics. J.R. receives research funding from Kite/Gilead, Novartis, and Oncternal Therapeutics and serves on advisory boards for Clade Therapeutics, Garuda Therapeutics, LifeVault Bio, Novartis, and Smart Immune. K.J.L. reports equity in Standard BioTools Inc. and serves on the scientific advisory board for MBQ Pharma Inc. R.J.S. serves on the board of directors for Be the Match–National Marrow Donor Program and DSMB for BMS and reports personal fees from Vor Biopharma, Smart Immune, Neovii, Astellas, Amgen, Bluesphere Bio, and Jasper. The remaining authors declare that they have no competing interests.
Data and materials availability: Single-cell transcriptome, CITE, and TCR data are publicly available in NCBI’s Database of Genotypes and Phenotype under accession number phs003630.v1.p1 (dbGaP; http://ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs003630.v1.p1) and in the National Center for Biotechnology Information’s Gene Expression Omnibus under accession number GSE255530. The DIISCO method is available at http://github.com/azizilab/DIISCO_public. A permanent archive of the version used in this manuscript is available at 10.5281/zenodo.13823393. All codes used for producing the results and figures in this manuscript are available at http://github.com/azizilab/dli_reproducibility. Tabulated data underlying the figures are provided in data file S13. All other data needed to support the conclusions in the paper are present in the paper or the Supplementary Materials.

Authors

Affiliations

Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing - original draft, and Writing - review & editing.
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing - original draft, and Writing - review & editing.
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Computer Science, Columbia University, New York, NY 10027, USA.
Roles: Data curation, Formal analysis, Methodology, Software, Validation, Visualization, Writing - original draft, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Data curation, Formal analysis, Methodology, Software, Visualization, and Writing - original draft.
Florian Raths
Singular Genomics, San Diego, CA 92121, USA.
Roles: Data curation, Formal analysis, Software, Visualization, and Writing - review & editing.
Singular Genomics, San Diego, CA 92121, USA.
Roles: Data curation, Formal analysis, Software, Visualization, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany.
Roles: Methodology, Resources, and Software.
Yinuo Jin
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
Role: Software.
Jia Yi Zhang
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
Roles: Data curation, Software, and Writing - review & editing.
Crystal Shin
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
Roles: Methodology and Software.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Roles: Formal analysis, Visualization, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Role: Investigation.
Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Investigation, Writing - original draft, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Role: Investigation.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Role: Investigation.
Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA.
Role: Investigation.
Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Investigation, Validation, and Writing - review & editing.
Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
Roles: Formal analysis and Writing - review & editing.
Singular Genomics, San Diego, CA 92121, USA.
Roles: Investigation, Methodology, Resources, Supervision, Visualization, and Writing - review & editing.
Martin Fabani
Singular Genomics, San Diego, CA 92121, USA.
Roles: Project administration, Resources, and Supervision.
Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Roles: Formal analysis, Methodology, and Writing - review & editing.
Department of Hematopoietic Biology & Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA.
Roles: Methodology and Writing - review & editing.
Eli N. Glezer
Singular Genomics, San Diego, CA 92121, USA.
Roles: Conceptualization, Methodology, Resources, Supervision, Validation, Visualization, and Writing - review & editing.
Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Project administration, Resources, Supervision, Validation, and Writing - review & editing.
Shuqiang Li
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Roles: Data curation, Investigation, and Resources.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Roles: Methodology and Supervision.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Roles: Conceptualization, Funding acquisition, Methodology, Resources, Supervision, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Roles: Conceptualization, Data curation, Funding acquisition, Project administration, Resources, Supervision, Visualization, and Writing - review & editing.
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
Harvard Medical School, Boston, MA 02115, USA.
Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
Roles: Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing - original draft, and Writing - review & editing.
Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA.
Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
Department of Computer Science, Columbia University, New York, NY 10027, USA.
Roles: Conceptualization, Funding acquisition, Methodology, Project administration, Software, Supervision, Writing - original draft, and Writing - review & editing.

Funding Information

National Cancer Institute: 1K08CA248458-01
NIH: R50CA251956
LLS: SCOR-22937-22

Notes

These authors contributed equally to this work.
*
Corresponding author. Email: [email protected] (E.A.); [email protected] (C.J.W.)

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. In the Right Place and the Right State: Spatial Cross-Talk and Immune State Dictate Leukemia Response to Immunotherapy, Cancer Research, 85, 9, (1574-1576), (2025).http://doi.org/10.1158/0008-5472.CAN-25-1018
    Crossref
Loading...

View Options

View options

PDF format

Download this article as a PDF file

Download PDF

Log in to view the full text

AAAS ID LOGIN

Loading institution options

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

More options

Purchase access to this article

Download and print this article within 24 hours for your personal scholarly, research, and educational use.

Figures

Tables

Multimedia

Share

Share

Copy the article link

Share on social media

ScienceAdviser

Get Science’s award-winning newsletter with the latest news, commentary, and research, free to your inbox daily.