Immune dysfunction signatures predict outcomes and define checkpoint blockade-unresponsive microenvironments in acute myeloid leukemia

Sergio Rutella, Jayakumar Vadakekolathu, Francesco Mazziotta, Stephen Reeder, Tung-On Yau, Rupkatha Mukhopadhyay, Benjamin Dickins, Heidi Altmann, Michael Kramer, Hanna A Knaus, Bruce R Blazar, Vedran Radojcic, Joshua F Zeidner, Andrea Arruda, Bofei Wang, Hussein A Abbas, Mark D Minden, Sarah K Tasian, Martin Bornhäuser, Ivana Gojo, Leo Luznik, Sergio Rutella, Jayakumar Vadakekolathu, Francesco Mazziotta, Stephen Reeder, Tung-On Yau, Rupkatha Mukhopadhyay, Benjamin Dickins, Heidi Altmann, Michael Kramer, Hanna A Knaus, Bruce R Blazar, Vedran Radojcic, Joshua F Zeidner, Andrea Arruda, Bofei Wang, Hussein A Abbas, Mark D Minden, Sarah K Tasian, Martin Bornhäuser, Ivana Gojo, Leo Luznik

Abstract

BackgroundImmune exhaustion and senescence are dominant dysfunctional states of effector T cells and major hurdles for the success of cancer immunotherapy. In the current study, we characterized how acute myeloid leukemia (AML) promotes the generation of senescent-like CD8+ T cells and whether they have prognostic relevance.METHODSWe analyzed NanoString, bulk RNA-Seq and single-cell RNA-Seq data from independent clinical cohorts comprising 1,896 patients treated with chemotherapy and/or immune checkpoint blockade (ICB).ResultsWe show that senescent-like bone marrow CD8+ T cells were impaired in killing autologous AML blasts and that their proportion negatively correlated with overall survival (OS). We defined what we believe to be new immune effector dysfunction (IED) signatures using 2 gene expression profiling platforms and reported that IED scores correlated with adverse-risk molecular lesions, stemness, and poor outcomes; these scores were a more powerful predictor of OS than 2017-ELN risk or leukemia stem cell (LSC17) scores. IED expression signatures also identified an ICB-unresponsive tumor microenvironment and predicted significantly shorter OS.ConclusionThe IED scores provided improved AML-risk stratification and could facilitate the delivery of personalized immunotherapies to patients who are most likely to benefit.TRIAL REGISTRATIONClinicalTrials.gov; NCT02845297.FUNDINGJohn and Lucille van Geest Foundation, Nottingham Trent University's Health & Wellbeing Strategic Research Theme, NIH/NCI P01CA225618, Genentech-imCORE ML40354, Qatar National Research Fund (NPRP8-2297-3-494).

Keywords: Cancer immunotherapy; Cellular senescence; Hematology; Leukemias.

Figures

Figure 1. Study workflow.
Figure 1. Study workflow.
Immune Signature Data Base (100); IED, immune effector dysfunction; NES, normalized enrichment score; FDR, false discovery rate; SOC, standard of care.
Figure 2. Markers of T cell senescence…
Figure 2. Markers of T cell senescence correlate with impaired T cell killing and poor clinical outcomes.
(A) Flow-sorted AML blasts were cocultured with autologous, patient-derived CD8+ T cells (n = 13 patients) for 5 days. Data were compared using the Kruskal-Wallis test. TW = Transwell insert; BL = AML blasts; Mono = monocytes. (B) Flow-sorted healthy-donor monocytes were cocultured with patient-derived CD8+ T cells (n = 9 patients) for 5 days. (C) In vitro killing of primary CD33+ CD34+ AML blasts (n = 10 patients) after 48-hour culture with autologous, flow-sorted T cells in the presence of anti-CD33/CD3 and control bispecific T cell engager (BiTE) antibody constructs (effector/target ratio = 1:5). T cell cytotoxicity was determined by flow cytometry, as detailed in the Supplemental Methods. (D) Kaplan-Meier estimates of OS in patients (JHU1 cohort, n = 43 patients) with senescent T cells above and below the optimal cut point, which was computed using the maxstat package in R. Survival curves were compared using a log-rank test. Median OS is indicated (color-coded by the optimal cut point of the proportion of CD3+CD8+CD57+KLRG1+ T cells). (E and F) Correlograms showing coexpression of NK and T cell markers in (E) TCGA-AML and (F) Beat-AML cases. The correlation matrix was reordered using the hclust function. Rectangles were drawn based on the results of hierarchical clustering (Euclidean distance, complete linkage). Inhibitory receptors (CD244, BTLA, CD160, TIGIT, LAG3, and PDCD1) are highlighted in red. NK cell, T cell, monocyte-macrophage (CD14, CD68, and CD163), and AML-associated markers (CD34, IL3RA, KIT, and THY1) were selected by integrating knowledge from multiple publications (10, 25, 101).
Figure 3. Signatures of immune effector dysfunction…
Figure 3. Signatures of immune effector dysfunction correlate with immune infiltration and with adverse-risk molecular features in the TCGA-AML and Beat-AML cohorts.
(A) Overlap between the IED172 signature genes from this study and published signatures that predict chemotherapy refractoriness as well as response to bispecific T cell engagers (2). IFN, interferon; IED, Immune effector dysfunction. (B) Semantic similarity between the IED172 genes in the context of their chromosomal location (XGR [eXploring Genomic Relations] web tool [ref. 102]). The degree of similarity between genes is visualized by the color of the links, with light yellow representing a low degree of similarity and red representing more. The chromosomal locus of each gene is indicated by the numbers and colors along the outer rim of the diagram. GO:MF, gene ontology molecular functions. (C) Correlation between the IED172 score and previously published immune traits (n = 45) (2, 42) in TCGA-AML (n = 157 patients). Signature scores are available through the original publications. (D) Correlation between the IED172 score and previously published immune traits and PARADIGM scores (n = 68; downloaded from the UCSC Xena data portal [https://xenabrowser.net/datapages/]; refs. 40, 44). The principal component analysis (PCA) plot was generated using the ggfortify and ggplot2 R packages. The top contributors to the first and second PC (n = 20) are shown as a bar graph. The dotted reference line in the bar graph indicates the expected value if the contribution were uniform. Any feature above the reference line can be considered as important in contributing to the dimension. (E) IED172 scores and percentage of blasts at diagnosis in TCGA-AML cases. Data were compared using the Mann-Whitney U test for unpaired determinations. PB = peripheral blood. (F) IED172 scores and leukemia burden at diagnosis in Beat-AML cases (n = 264). Data were compared using the Mann-Whitney U test for unpaired determinations. (G) Stacked bar graph showing the proportion of IED172hi and IED172lo cases harboring mutations of TP53, RUNX1, ASXL1, DNMT3A, NPM1, and FLT3–internal tandem duplication (ITD). Mut, mutated.
Figure 4. Covarying gene programs of immune…
Figure 4. Covarying gene programs of immune effector dysfunction and stemness in the TCGA-AML cohort.
(A) Pairwise correlation between transcriptomic traits of immune infiltration and LSC17/IED172 scores. Nonsignificant P values are shown as blank boxes. Modules, including traits that are densely connected (hubs), are identified based on hierarchical clustering (Euclidean distance, complete linkage) and are shown in black boxes. The IED172 and LSC17 scores are highlighted in red. IED, Immune effector dysfunction. (B) LSC17 score in IED172hi and IED172lo TCGA cases (n = 157; median split). Data were compared using the Mann-Whitney U test for unpaired determinations. (C) Inferred relative frequency of hematopoietic stem cells (HSCs) in patients with IED172hi or IED172lo, as estimated by xCELL (54). Precalculated TCGA scores were downloaded from https://xcell.ucsf.edu/ (D) Kaplan-Meier estimates of overall survival (OS) in patients with IED172hi with above-median and below-median LSC17 scores. Survival curves were compared using a log-rank test. HR, hazard ratio. (E) Kaplan-Meier estimates of OS in patients with IED172lo with above-median and below-median LSC17 scores.
Figure 5. Predictive ability of immune effector…
Figure 5. Predictive ability of immune effector dysfunction gene programs in the TCGA-AML cohort.
(A) Area under the receiver operating characteristic (AUROC) curve measuring the predictive ability of the prognostic index (PI24) genes for overall survival (n = 157 TCGA cases). (B) Forest plot (ggforest function in survminer package in R) of pretreatment features (WBC count at diagnosis, percentage of bone marrow blasts, FLT3-ITD and NPM1 mutational status, patient age at diagnosis), and RNA-based scores associated with survival in multivariate Cox proportional hazard analyses (PI24, LSC17, and IFN scores; refs. 2, 47, 52). HR = hazard ratio for death. (C) Kaplan-Meier estimates of relapse-free survival (RFS) in patients with TCGA-AML with above-median and below-median PI24 scores, which were calculated using β values from Cox regression analyses of gene expression and patient survival (56). Survival curves were compared using the log-rank test. (D) Kaplan-Meier estimates of OS in TCGA-AML patients with above-median and below-median PI24 scores. Survival curves were compared using the log-rank test.
Figure 6. Immune effector dysfunction scores correlate…
Figure 6. Immune effector dysfunction scores correlate with immune infiltration, stemness, primary induction failure, and patient outcome in an external AML cohort.
(A) Bubble plot depicting enriched REACTOME pathways (https://reactome.org/) in IED172 and IED68 signature genes (clusterProfiler package in R), which were ranked based on the gene ratio (gene count divided by set size). IED, immune effector dysfunction. (B) Correlation between the IED68 score and previously published immune traits (n = 45; refs. 2, 42) in the PMCC cohort (n = 290 patients). Signature scores are available in the original publications. (C) Correlation between IED68 scores and leukemia burden at diagnosis in the PMCC cohort. Data were compared using the Mann-Whitney U test for unpaired determinations. BM, bone marrow; PB, peripheral blood. (D) Response to induction chemotherapy in patients with above-median and below-median prognostic index (PI20) scores in the PMCC cohort. PIF, primary induction failure following a standard 1 or 2 cycles of induction chemotherapy. CR, complete remission (defined as <5% BM blasts). (E) Kaplan-Meier estimates of relapse-free survival (RFS) in PMCC patients with above-median and below-median PI20 scores. Survival curves were compared using a log-rank test. HR, hazard ratio. (F) Kaplan-Meier estimates of overall survival (OS) in PMCC patients with higher than median and lower-than-median PI20 scores. Survival curves were compared using a log-rank test. (G) Area under the receiver operating characteristic (AUROC) curve measuring the predictive ability of the PI20 and the ELN cytogenetic risk classifier for OS. CI, confidence interval.
Figure 7. Immune effector dysfunction scores correlate…
Figure 7. Immune effector dysfunction scores correlate with immune infiltration and separate survival in pediatric AML cohorts.
(A) Leukemia burden in COG-TARGET AML cases (n = 145) with above-median and below-median IED172 scores. Data were compared using the Mann-Whitney U test for unpaired determinations. BM, bone marrow; PB, peripheral blood; IED, immune effector dysfunction. (B) WBC count at diagnosis in COG-TARGET AML cases with above-median and below-median IED172 scores. Data were compared using the Mann-Whitney U test for unpaired determinations. (C) IED172 scores at time of AML diagnosis and response assessment (bulk RNA-Seq data from matched BM samples available in 31 COG-TARGET AML cases). (D) Kaplan-Meier estimate of relapse-free survival (RFS) in patients from the COG-TARGET AML cohort with above-median and below-median prognostic index (PI24) scores. Survival curves were compared using a log-rank test (survminer package in R). HR, hazard ratio. (E) Kaplan-Meier estimate of overall survival (OS) in patients from the COG-TARGET AML cohort with above-median and below-median PI24 scores. (F) Correlation between the IED68 score and previously published immune traits (n = 45) in the CHOP AML series (n = 40). Signature scores are available through the original publications (2, 42). (G) IED68 scores in samples from the CHOP AML series collected at time of diagnosis and response assessment (n = 14 matched BM samples). Data were compared using the Wilcoxon’s matched-pairs signed-rank test. (H) Kaplan-Meier estimates of RFS in patients from the CHOP AML cohort with above-median and below-median PI20 scores. (I) Kaplan-Meier estimate of OS in patients from the CHOP cohort with above-median and below-median PI20 scores.
Figure 8. Immune effector dysfunction scores increase…
Figure 8. Immune effector dysfunction scores increase at time of response assessment and predict outcomes in additional external AML cohorts.
(A) Expression of the IED68 genes in patients from the SAL and JHU cohorts (n = 183 BM samples from 90 patients). The heatmap annotation track shows sample collection time points (baseline and post-chemotherapy response assessment). (B) IED68 score at time of diagnosis and response assessment (Kruskal-Wallis test with correction for multiple comparisons). Nonsignificant P values are not shown. CR = complete remission; PIF = primary induction failure; ER = early relapse (<6 months after the achievement of CR); LR = late relapse (>6) months after the achievement of CR); IED = immune effector dysfunction. (C) Kaplan-Meier estimate of relapse-free survival (RFS; data available in 56 subjects) in higher-than-median and lower-than-median PI20 groups. HR = hazard ratio. (D) Kaplan-Meier estimate of overall survival (OS; data available in 90 subjects) in higher-than-median and lower-than-median PI20 groups. (E) Volcano plot showing differentially expressed genes (DEGs) between samples collected at baseline and post-chemotherapy (post-CT) response assessment (EnhancedVolcano package in R). Genes discussed in the paper are named. (F) Graphical summary of over-representation analysis (clusterProfiler package in R) showing the overlap between DEGs (post-chemotherapy versus baseline) and curated cell type signature gene sets (C8 collection), which were retrieved from the MSigDB (http://www.gsea-msigdb.org/gsea/index.jsp). Gene ratio = gene count divided by set size.
Figure 9. Immune effector dysfunction scores predict…
Figure 9. Immune effector dysfunction scores predict response to AZA+Pembro in clinical trial NCT02845297.
(A) Differentially expressed genes (DEGs) at baseline associated with complete response (CResp) to AZA+Pembro (n = 33 patients). The heatmap annotation track shows the prognostic index (PI20) group and response status (complete remission [CR] and nonresponder [NR]) after 2 cycles of azacitidine and pembrolizumab. Complete response was defined as CR, CR with partial hematologic recovery (CRh), CR with incomplete hematologic recovery (CRi), or morphological leukemia-free state (MLFS) at the end of cycle 2. Patients with partial response (PR; >50% decrease in bone marrow blasts from baseline to 5%–25% at the end of cycle 1) were categorized as NRs. C, cluster. (B) Area under the receiver operating characteristic (AUROC) curve measuring the predictive ability of IED68 genes for response to AZA+Pembro. CI, confidence interval. (C) Kaplan-Meier estimate of overall survival (OS) in patients with above-median and below-median PI20. Survival curves were compared using the Gehan-Breslow-Wilcoxon’s test, a generalization of the Wilcoxon’s rank-sum test that attributes more weight to deaths at early time points. HR, hazard ratio. (D) Kaplan-Meier estimate of OS in patients with above-median and below-median IFN scores, which were computed as previously published (2). Survival curves were compared using the Gehan-Breslow-Wilcoxon’s test. (E) Volcano plot showing DEGs between baseline and end-of-cycle 2 (EO2) bone marrow samples. The top 20 DEGs are named. (F) The overlap between DEGs post-reatment versus baseline in the chemotherapy (CT; SAL and JHU2) and AZA+Pembro patient series is shown as a Venn diagram. Nonredundant, enriched gene ontologies in DEGs between the CT and AZA+Pembro cohorts were visualized as a network diagram (cnetplot) with color nodes using the cnetplot function of the GOSemSim package in R (67).
Figure 10. Immune effector dysfunction (IED) scores…
Figure 10. Immune effector dysfunction (IED) scores predict immunotherapy response in melanoma.
(A) Progression-free survival (PFS) in 427 patients with melanoma from the TCGA Pan-Cancer Atlas profiling project. Participants were stratified based on an optimal cut point of the prognostic index (PI24) (value, 0.862). Survival curves were compared using a log-rank test. RNA-Seq and outcomes data were retrieved through the cBioPortal for Cancer Genomics (https://www.cbioportal.org/). HR, hazard ratio. (B) Overall survival (OS) in patients with melanoma from the TCGA Pan-Cancer Atlas cohort. (C) Volcano plot showing differentially expressed genes (DEGs) between patients with PI24hi or PI24lo in the PRJEB23709 immunotherapy cohort (73 participants with melanoma treated with standard-of-care single-agent nivolumab or pembrolizumab (n = 41) or combination anti–PD-1 + anti–CTLA-4 (n = 32; Supplemental Table 10). RNA-Seq and outcome data were retrieved through the original publication (72) and the Tumor Immune Dysfunction and Exclusion (TIDE) portal (http://tide.dfci.harvard.edu/login/) (58). The top 15 DEGs are named. (D) Number of responders and nonresponders with above-median and below-median PI24 scores in the PRJEB23709 immunotherapy cohort. Fisher’s exact test. CR = complete response; PR, partial response. In the original publication (72), responders are defined as individuals with complete response, partial response, or stable disease of greater than 6 months with no progression, whereas nonresponders are defined as progressive disease or stable disease for less than or equal to 6 months before disease progression. (E) AUROC curve measuring the predictive ability of PI24 genes for response to ICB-based therapies in the PRJEB23709 cohort. CI, confidence interval. (F) PFS and OS in patients with melanoma in the PRJEB23709 immunotherapy cohort. Patients were dichotomized based on an optimal cut point of PI24 values (0.12 and 0.344 for PFS and OS, respectively).

References

    1. Dohner H, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–447. doi: 10.1182/blood-2016-08-733196.
    1. Vadakekolathu J, et al. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci Transl Med. 2020;12(546):eaaz0463. doi: 10.1126/scitranslmed.aaz0463.
    1. Vago L, Gojo I. Immune escape and immunotherapy of acute myeloid leukemia. J Clin Invest. 2020;130(4):1552–1564. doi: 10.1172/JCI129204.
    1. Zeidner JF, et al. Immunomodulation with pomalidomide at early lymphocyte recovery after induction chemotherapy in newly diagnosed AML and high-risk MDS. Leukemia. 2020;34(6):1563–1576. doi: 10.1038/s41375-019-0693-4.
    1. Daver N, et al. Efficacy, safety, and biomarkers of response to azacitidine and nivolumab in relapsed/refractory acute myeloid leukemia: a nonrandomized, open-label, phase II study. Cancer Discov. 2019;9(3):370–383. doi: 10.1158/-18-0774.
    1. Mathew NR, et al. Sorafenib promotes graft-versus-leukemia activity in mice and humans through IL-15 production in FLT3-ITD-mutant leukemia cells. Nat Med. 2018;24(3):282–291. doi: 10.1038/nm.4484.
    1. Zeidner JF, et al. Phase II trial of pembrolizumab after high-dose cytarabine in relapsed/refractory acute myeloid leukemia. Blood Cancer Discov. 2021;2(6):616–629. doi: 10.1158/2643-3230.BCD-21-0070.
    1. Uy GL, et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood. 2021;137(6):751–762. doi: 10.1182/blood.2020007732.
    1. Geissler K, et al. Immune signature of tumor infiltrating immune cells in renal cancer. Oncoimmunology. 2015;4(1):e985082. doi: 10.4161/2162402X.2014.985082.
    1. Knaus HA, et al. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight. 2018;3(21):e120974. doi: 10.1172/jci.insight.120974.
    1. Liu X, et al. Senescent T cells within suppressive tumor microenvironments: emerging target for tumor immunotherapy. J Clin Invest. 2020;130(3):1073–1083. doi: 10.1172/JCI133679.
    1. Philip M, Schietinger A. CD8+ T cell differentiation and dysfunction in cancer. Nat Rev Immunol. 2022;22(4):209–223. doi: 10.1038/s41577-021-00574-3.
    1. Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat Rev Immunol. 2011;11(4):289–295. doi: 10.1038/nri2959.
    1. Liu X, et al. Reprogramming lipid metabolism prevents effector T cell senescence and enhances tumor immunotherapy. Sci Transl Med. 2021;13(587):eaaz6314. doi: 10.1126/scitranslmed.aaz6314.
    1. Henson SM, et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J Clin Invest. 2014;124(9):4004–4016. doi: 10.1172/JCI75051.
    1. Robert C, et al. Immunotherapy discontinuation - how, and when? Data from melanoma as a paradigm. Nat Rev Clin Oncol. 2020;17(11):707–715. doi: 10.1038/s41571-020-0399-6.
    1. Larkin J, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019;381(16):1535–1546. doi: 10.1056/NEJMoa1910836.
    1. Mussai F, et al. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood. 2013;122(5):749–758. doi: 10.1182/blood-2013-01-480129.
    1. Deng M, et al. LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature. 2018;562(7728):605–609. doi: 10.1038/s41586-018-0615-z.
    1. Pyzer AR, et al. MUC1-mediated induction of myeloid-derived suppressor cells in patients with acute myeloid leukemia. Blood. 2017;129(13):1791–1801. doi: 10.1182/blood-2016-07-730614.
    1. Williams P, et al. The distribution of T cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer. 2019;125(9):1470–1481. doi: 10.1002/cncr.31896.
    1. Krupka C, et al. CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T cell-engaging antibody AMG 330. Blood. 2014;123(3):356–365. doi: 10.1182/blood-2013-08-523548.
    1. Laszlo GS, et al. Cellular determinants for preclinical activity of a novel CD33/CD3 bispecific T cell engager (BiTE) antibody, AMG 330, against human AML. Blood. 2014;123(4):554–561. doi: 10.1182/blood-2013-09-527044.
    1. Harrington KH, et al. The broad anti-AML activity of the CD33/CD3 BiTE antibody construct, AMG 330, is impacted by disease stage and risk. PLoS One. 2015;10(8):e0135945. doi: 10.1371/journal.pone.0135945.
    1. Bagaev A, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021;39(6):845–865. doi: 10.1016/j.ccell.2021.04.014.
    1. Thorsson V, et al. The immune landscape of cancer. Immunity. 2018;48(4):812–830. doi: 10.1016/j.immuni.2018.03.023.
    1. Pereira BI, et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat Immunol. 2020;21(6):684–694. doi: 10.1038/s41590-020-0643-3.
    1. Good CR, et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell. 2021;184(25):6081–6100. doi: 10.1016/j.cell.2021.11.016.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. doi: 10.1073/pnas.0506580102.
    1. Dufva O, et al. Immunogenomic landscape of hematological malignancies. Cancer Cell. 2020;38(3):380–399. doi: 10.1016/j.ccell.2020.06.002.
    1. van Galen P, et al. Single-Cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019;176(6):1265–1281. doi: 10.1016/j.cell.2019.01.031.
    1. Yang C, et al. Heterogeneity of human bone marrow and blood natural killer cells defined by single -cell transcriptome. Nat Commun. 2019;10(1):3931. doi: 10.1038/s41467-019-11947-7.
    1. Martinez-Zamudio RI, et al. Senescence-associated β-galactosidase reveals the abundance of senescent CD8+ T cells in aging humans. Aging Cell. 2021;20(5):e13344.
    1. Guo X, et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat Med. 2018;24(7):978–985. doi: 10.1038/s41591-018-0045-3.
    1. Gueguen P, et al. Contribution of resident and circulating precursors to tumor-infiltrating CD8+ T cell populations in lung cancer. Sci Immunol. 2021;6(55):eabd5778. doi: 10.1126/sciimmunol.abd5778.
    1. Abbas HA, et al. Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat Commun. 2021;12(1):6071. doi: 10.1038/s41467-021-26282-z.
    1. Vignard V, et al. MicroRNAs in tumor exosomes drive immune escape in melanoma. Cancer Immunol Res. 2020;8(2):255–267. doi: 10.1158/2326-6066.CIR-19-0522.
    1. Su H, et al. miR-9-5p regulates immunometabolic and epigenetic pathways in beta-glucan-trained immunity via IDH3alpha. JCI Insight. 2021;6(9):e144260. doi: 10.1172/jci.insight.144260.
    1. Pu J, et al. M2 macrophage-derived extracellular vesicles facilitate CD8+T cell exhaustion in hepatocellular carcinoma via the miR-21-5p/YOD1/YAP/β-catenin pathway. Cell Death Discov. 2021;7(1):182. doi: 10.1038/s41420-021-00556-3.
    1. Goldman MJ, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38(6):675–678. doi: 10.1038/s41587-020-0546-8.
    1. Ayers M, et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127(8):2930–2940. doi: 10.1172/JCI91190.
    1. Danaher P, et al. Gene expression markers of tumor infiltrating leukocytes. J Immunother Cancer. 2017;5:18. doi: 10.1186/s40425-017-0215-8.
    1. Campbell JD, et al. Genomic, pathway network, and immunologic features distinguishing squamous carcinomas. Cell Rep. 2018;23(1):194–212. doi: 10.1016/j.celrep.2018.03.063.
    1. Vaske CJ, et al. Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics. 2010;26(12):i237–i245. doi: 10.1093/bioinformatics/btq182.
    1. Hoadley KA, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 2014;158(4):929–944. doi: 10.1016/j.cell.2014.06.049.
    1. Tyner JW, et al. Functional genomic landscape of acute myeloid leukaemia. Nature. 2018;562(7728):526–531. doi: 10.1038/s41586-018-0623-z.
    1. Vadakekolathu J, et al. TP53 abnormalities correlate with immune infiltration and associate with response to flotetuzumab immunotherapy in AML. Blood Adv. 2020;4(20):5011–5024. doi: 10.1182/bloodadvances.2020002512.
    1. Sallman DA, et al. TP53 mutations in myelodysplastic syndromes and secondary AML confer an immunosuppressive phenotype. Blood. 2020;136(24):2812–2823. doi: 10.1182/blood.2020006158.
    1. Miranda A, et al. Cancer stemness, intratumoral heterogeneity, and immune response across cancers. Proc Natl Acad Sci U S A. 2019;116(18):9020–9029. doi: 10.1073/pnas.1818210116.
    1. Sturm G, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics. 2019;35(14):i436–i445. doi: 10.1093/bioinformatics/btz363.
    1. Bill M, et al. Mutations associated with a 17-gene leukemia stem cell score and the score’s prognostic relevance in the context of the European LeukemiaNet classification of acute myeloid leukemia. Haematologica. 2020;105(3):721–729. doi: 10.3324/haematol.2019.225003.
    1. Ng SW, et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature. 2016;540(7633):433–437. doi: 10.1038/nature20598.
    1. Ott PA, et al. T cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318–327. doi: 10.1200/JCO.2018.78.2276.
    1. Aran D, et al. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017;18(1):220. doi: 10.1186/s13059-017-1349-1.
    1. Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16(4):385–395. doi: 10.1002/(SICI)1097-0258(19970228)16:4<385::AID-SIM380>;2-3.
    1. Wagner S, et al. A parsimonious 3-gene signature predicts clinical outcomes in an acute myeloid leukemia multicohort study. Blood Adv. 2019;3(8):1330–1346. doi: 10.1182/bloodadvances.2018030726.
    1. Herold T, et al. A 29-gene and cytogenetic score for the prediction of resistance to induction treatment in acute myeloid leukemia. Haematologica. 2018;103(3):456–465. doi: 10.3324/haematol.2017.178442.
    1. Jiang P, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018;24(10):1550–1558. doi: 10.1038/s41591-018-0136-1.
    1. Vadakekolathu J, et al. An immune senescence and exhaustion-related RNA profile predicts clinical outcomes in acute myeloid leukemia. Blood. 2020;136(suppl 1):26–27. doi: 10.1182/blood-2020-134779.
    1. Willier S, et al. CLEC12A and CD33 coexpression as a preferential target for pediatric AML combinatorial immunotherapy. Blood. 2021;137(8):1037–1049. doi: 10.1182/blood.2020006921.
    1. Bolouri H, et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med. 2018;24(1):103–112. doi: 10.1038/nm.4439.
    1. de Rooij JD, et al. Pediatric AML: from biology to clinical management. J Clin Med. 2015;4(1):127–149. doi: 10.3390/jcm4010127.
    1. Tarlock K, Meshinchi S. Pediatric acute myeloid leukemia: biology and therapeutic implications of genomic variants. Pediatr Clin North Am. 2015;62(1):75–93. doi: 10.1016/j.pcl.2014.09.007.
    1. Conneely SE, Stevens AM. Acute myeloid leukemia in children: emerging paradigms in genetics and new approaches to therapy. Curr Oncol Rep. 2021;23(2):16. doi: 10.1007/s11912-020-01009-3.
    1. Carrasco E, et al. The role of T cells in age-related diseases. Nat Rev Immunol. 2022;22(2):97–111. doi: 10.1038/s41577-021-00557-4.
    1. Mittelbrunn M, Kroemer G. Hallmarks of T cell aging. Nat Immunol. 2021;22(6):687–698. doi: 10.1038/s41590-021-00927-z.
    1. Yu G, et al. GOSemSim: an R package for measuring semantic similarity among GO terms and gene products. Bioinformatics. 2010;26(7):976–978. doi: 10.1093/bioinformatics/btq064.
    1. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell. 2015;161(7):1681–1696. doi: 10.1016/j.cell.2015.05.044.
    1. Saltz J, et al. Spatial organization and molecular correlation of tumor-infiltrating lymphocytes using deep learning on pathology images. Cell Rep. 2018;23(1):181–193. doi: 10.1016/j.celrep.2018.03.086.
    1. Kalaora S, et al. Mechanisms of immune activation and regulation: lessons from melanoma. Nat Rev Cancer. 2022;22(4):195–207. doi: 10.1038/s41568-022-00442-9.
    1. Tirosh I, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352(6282):189–196. doi: 10.1126/science.aad0501.
    1. Gide TN, et al. Distinct immune cell populations define response to Anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 combined therapy. Cancer Cell. 2019;35(2):238–255. doi: 10.1016/j.ccell.2019.01.003.
    1. SadeFeldman M, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018;175(4):998–1013. doi: 10.1016/j.cell.2018.10.038.
    1. Rutella S, et al. Capturing the complexity of the immune microenvironment of acute myeloid leukemia with 3D biology technology. J Clin Oncol. 2018;36(5 suppl):50. doi: 10.1200/JCO.2018.36.5_suppl.50.
    1. Liu X, et al. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat Commun. 2018;9(1):249. doi: 10.1038/s41467-017-02689-5.
    1. Montes CL, et al. Tumor-induced senescent T cells with suppressor function: a potential form of tumor immune evasion. Cancer Res. 2008;68(3):870–879. doi: 10.1158/0008-5472.CAN-07-2282.
    1. Milanovic M, et al. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018;553(7686):96–100. doi: 10.1038/nature25167.
    1. Duy C, et al. Chemotherapy induces senescence-like resilient cells capable of initiating AML recurrence. Cancer Discov. 2021;11(6):1542–1561. doi: 10.1158/-20-1375.
    1. Mosteiro L, et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354(6315):aa4445. doi: 10.1126/science.aaf4445.
    1. Ritschka B, et al. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev. 2017;31(2):172–183. doi: 10.1101/gad.290635.116.
    1. Abdul-Aziz AM, et al. Acute myeloid leukemia induces protumoral p16INK4a-driven senescence in the bone marrow microenvironment. Blood. 2019;133(5):446–456. doi: 10.1182/blood-2018-04-845420.
    1. Lee S, Schmitt CA. The dynamic nature of senescence in cancer. Nat Cell Biol. 2019;21(1):94–101. doi: 10.1038/s41556-018-0249-2.
    1. Akbar AN, et al. Senescence of T Lymphocytes: Implications for Enhancing Human Immunity. Trends Immunol. 2016;37(12):866–876. doi: 10.1016/j.it.2016.09.002.
    1. Mondal AM, et al. p53 isoforms regulate aging- and tumor-associated replicative senescence in T lymphocytes. J Clin Invest. 2013;123(12):5247–5257. doi: 10.1172/JCI70355.
    1. Miller BC, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20(3):326–336. doi: 10.1038/s41590-019-0312-6.
    1. Ravandi F, et al. Updated results from phase I dose-escalation study of AMG 330, a bispecific T cell engager molecule, in patients with relapsed/refractory acute myeloid leukemia (R/R AML) J Clin Oncol. 2020;38(15_suppl):7508. doi: 10.1200/JCO.2020.38.15_suppl.7508.
    1. Zeidan AM, et al. A multi-center phase I Trial of Ipilimumab in patients with myelodysplastic syndromes following hypomethylating agent failure. Clin Cancer Res. 2018;24(15):3519–3527. doi: 10.1158/1078-0432.CCR-17-3763.
    1. Zeidan AM, et al. A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for older patients with AML. Blood Adv. 2022;6(7):2219–2229. doi: 10.1182/bloodadvances.2021006138.
    1. Daver N, et al. T cell-based immunotherapy of acute myeloid leukemia: current concepts and future developments. Leukemia. 2021;35(7):1843–1863. doi: 10.1038/s41375-021-01253-x.
    1. Prat A, et al. Immune-related gene expression profiling after PD-1 blockade in non-small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 2017;77(13):3540–3550. doi: 10.1158/0008-5472.CAN-16-3556.
    1. Huang AC, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25(3):454–461. doi: 10.1038/s41591-019-0357-y.
    1. Fairfax BP, et al. Peripheral CD8+ T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat Med. 2020;26(2):193–199. doi: 10.1038/s41591-019-0734-6.
    1. Bruck O, et al. Immune profiles in acute myeloid leukemia bone marrow associate with patient age, T cell receptor clonality, and survival. Blood Adv. 2020;4(2):274–286. doi: 10.1182/bloodadvances.2019000792.
    1. Dohner H, et al. Toward precision medicine for AML. Nat Rev Clin Oncol. 2021;18(9):577–590. doi: 10.1038/s41571-021-00509-w.
    1. Morita K, et al. Clonal evolution of acute myeloid leukemia revealed by high-throughput single-cell genomics. Nat Commun. 2020;11(1):5327. doi: 10.1038/s41467-020-19119-8.
    1. Horibata S, et al. Heterogeneity in refractory acute myeloid leukemia. Proc Natl Acad Sci U S A. 2019;116(21):10494–10503. doi: 10.1073/pnas.1902375116.
    1. Toffalori C, et al. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nat Med. 2019;25(4):603–611. doi: 10.1038/s41591-019-0400-z.
    1. Miles LA, et al. Single-cell mutation analysis of clonal evolution in myeloid malignancies. Nature. 2020;587(7834):477–482. doi: 10.1038/s41586-020-2864-x.
    1. Im SJ, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537(7620):417–421. doi: 10.1038/nature19330.
    1. Godec J, et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity. 2016;44(1):194–206. doi: 10.1016/j.immuni.2015.12.006.
    1. Ehninger A, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014;4:e218. doi: 10.1038/bcj.2014.39.
    1. Fang H, et al. XGR software for enhanced interpretation of genomic summary data, illustrated by application to immunological traits. Genome Med. 2016;8(1):129. doi: 10.1186/s13073-016-0384-y.

Source: PubMed

3
Prenumerera