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 (
2–
4). Effective antitumor immunity relies at least in part on coordinated activity among cell types within the tumor microenvironment (TME) (
5–
8). However, the specific role and characteristics of tumor-infiltrating immune cells remain unclear (
9–
11). Developing a better understanding of the cellular players within the TME may be the key to unlocking effective tumor-directed responses (
12–
14).
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 (
17–
20).
Single-cell transcriptomic and spatial profiling can powerfully enhance our ability to disentangle the complex TME (
21–
23). We previously identified expansion of precursor exhausted T (T
PE) 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.
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 (
5–
7,
48–
50,
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,
74–
80). 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 × 10
7 CD3
+ cells/kg for Rs versus 1.6 × 10
7 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 200
g 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 × 10
5 T cells were cultured with 1 × 10
5 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
+ T
EMRA/CTL (CD3
+CD8
+CD62L
−CD45RA
+) and CD8
+ non-T
EMRA/CTL (CD3
+CD8
+CD62L
−CD45RA
− 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 × 10
5 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.