Pembrolizumab and decitabine for refractory or relapsed acute myeloid leukemia

Meghali Goswami, Gege Gui, Laura W Dillon, Katherine E Lindblad, Julie Thompson, Janet Valdez, Dong-Yun Kim, Jack Y Ghannam, Karolyn A Oetjen, Christin B Destefano, Dana M Smith, Hanna Tekleab, Yeusheng Li, Pradeep Dagur, Thomas Hughes, Jennifer L Marté, Jaydira Del Rivero, Joanna Klubo-Gwiezdzinksa, James L Gulley, Katherine R Calvo, Catherine Lai, Christopher S Hourigan, Meghali Goswami, Gege Gui, Laura W Dillon, Katherine E Lindblad, Julie Thompson, Janet Valdez, Dong-Yun Kim, Jack Y Ghannam, Karolyn A Oetjen, Christin B Destefano, Dana M Smith, Hanna Tekleab, Yeusheng Li, Pradeep Dagur, Thomas Hughes, Jennifer L Marté, Jaydira Del Rivero, Joanna Klubo-Gwiezdzinksa, James L Gulley, Katherine R Calvo, Catherine Lai, Christopher S Hourigan

Abstract

Background: The powerful 'graft versus leukemia' effect thought partly responsible for the therapeutic effect of allogeneic hematopoietic cell transplantation in acute myeloid leukemia (AML) provides rationale for investigation of immune-based therapies in this high-risk blood cancer. There is considerable preclinical evidence for potential synergy between PD-1 immune checkpoint blockade and the hypomethylating agents already commonly used for this disease.

Methods: We report here the results of 17 H-0026 (PD-AML, NCT02996474), an investigator sponsored, single-institution, single-arm open-label 10-subject pilot study to test the feasibility of the first-in-human combination of pembrolizumab and decitabine in adult patients with refractory or relapsed AML (R-AML).

Results: In this cohort of previously treated patients, this novel combination of anti-PD-1 and hypomethylating therapy was feasible and associated with a best response of stable disease or better in 6 of 10 patients. Considerable immunological changes were identified using T cell receptor β sequencing as well as single-cell immunophenotypic and RNA expression analyses on sorted CD3+ T cells in patients who developed immune-related adverse events (irAEs) during treatment. Clonal T cell expansions occurred at irAE onset; single-cell sequencing demonstrated that these expanded clones were predominately CD8+ effector memory T cells with high cell surface PD-1 expression and transcriptional profiles indicative of activation and cytotoxicity. In contrast, no such distinctive immune changes were detectable in those experiencing a measurable antileukemic response during treatment.

Conclusion: Addition of pembrolizumab to 10-day decitabine therapy was clinically feasible in patients with R-AML, with immunological changes from PD-1 blockade observed in patients experiencing irAEs.

Keywords: adaptive immunity; immunotherapy; investigational; lymphocyte activation; therapies; translational medical research.

Conflict of interest statement

Competing interests: CSH receives research support from Merck Sharpe & Dohme and SELLAS Life Sciences Group AG. CL is on the Speakers’ Bureau for Astellas, Jazz Pharma and serves in a consulting or advisory role for Daiichi, Jazz Pharma, Amgen, Abbvie, Macrogenics, and Agios. The remaining authors declare no competing interests.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
PD-AML clinical trial schema, sampling intervals, and survival curve. (A) Pembrolizumab was administered on day 1 of every cycle for eight cycles, and decitabine was given on days 8–12 and 15–19 of every other cycle. Bone marrow aspirate and peripheral blood was collected before the start of treatment (day 0), on cycle 1 day 8 (C1D8) after one dose of pembrolizumab, and at the end of every other cycle (EOC2, EOC4, EOC6, EOC8); clinical responses were also assessed at these timepoints. (B) Summary of patients treated on NCT02996474 and responses. These patients have been reported previously. (C) Kaplan-Meier survival curve with 95% CI. (D) Summary of grades 3 and 4 adverse events and irAEs that occurred during treatment. AML, acute myeloid leukemia; EOC2, end of 2 cycles of treatment; EOC4, end of 4 cycles of treatment; EOC6, end of 6 cycles of treatment; EOC8, end of 8 cycles of treatment.
Figure 2
Figure 2
Development of irAEs is associated with clonal T cell expansion. The number of significantly expanded or contracted clones at timepoints sampled during treatment compared to baseline were calculated in (A) peripheral blood (PB) and (B) bone marrow (BM) for patients developing irAEs. Time of irAE indicated by black arrows. TSH levels and significantly expanded T cells at EOC2 and EOC4 in PB that were

Figure 3

Unique expression signatures in clonally…

Figure 3

Unique expression signatures in clonally expanded T cells at time of irAE. Cells…

Figure 3
Unique expression signatures in clonally expanded T cells at time of irAE. Cells harboring the unique TCRβ CDR3 from differentially abundant clones identified by bulk TCRβ-seq were grouped and differentially expressed genes (DEG) and cell surface proteins (DEP) were identified by supervised analyses compared with top 10 abundant T cell clones in the sample identified by bulk TCRβ-seq. (A) DEG and (B) DEP of expanded CD8+ T cell clones of interest in PD-AML two at hypothyroidism diagnosis vs other abundant CD8+ T cell clonotypes. (C) Frequencies from TCRβ-seq data of clonotypes coexpressing cell surface PD-1, TIM-3, and CD27 (four clones, top panel) and those not expressing this signature (two clones, bottom panel). (D) DEG and (E) DEP of expanded CD8+ T cell clones of interest in PD-AML three at time of diagnosis of central diabetes insipidus versus other abundant CD8+ T cell clonotypes. (F) Frequencies from TCRβ-seq data of clonotypes coexpressing cell surface PD-1, TIM-3, 4-1BB, HLA-DR, and CD27 (six clones, top panel) and those not expressing this signature (three clones, bottom panel). Heatmaps were created using the mean expression of all cells within each clone; differentially expressed markers were ordered by adjusted p values and fold changes. Adjusted p values are based on Wilcoxon rank sum test with Bonferroni corrections using all features in the dataset. ‘Difference **’ denotes adjusted p

Figure 4

Antileukemic response to combination immunotherapy…

Figure 4

Antileukemic response to combination immunotherapy was not associated with clonal T cell expansion.…

Figure 4
Antileukemic response to combination immunotherapy was not associated with clonal T cell expansion. Number of significantly expanded or contracted clones at timepoints sampled during treatment compared with baseline were calculated in (A) PB and (B) BM for two patients achieving CR by eight cycles of treatment. Decreasing blast percentage in the BM and few to no significantly expanded clones at EOC2 or EOC4 in BM that were low in frequency (

Figure 5

Stable T cell immunophenotypes in…

Figure 5

Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization…

Figure 5
Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization of T cell immunophenotyping based on cell surface expression CD4, CD8, CCR7, CD45RA, CD127 and CD25. (B) Quantification and proportion of CD4+ and CD8+ T cell subset frequencies, as a percentage of CD3+. (C) Differential gene expression analysis among different cell types with scaled log normalization. For panels A-C, T cell subsets indicated in legend. (D) Frequencies of CD4+ and CD8+ effector memory (EM) and terminal effector (TE) T cells at Day0, EOC2, and EOC4. (E) Per cent change in frequencies of CD4+ and CD8+ EM and TE cells at EOC2 and EOC4 compared with day 0. (F) T cell subset clonality in T cell subsets identified by immunophenotyping. Patients and time points indicated in key. (G) Cells expressing exhaustion (purple) and cytotoxicity (orange) predefined gene sets overlaid on integrated UMAP with exhaustion and cytotoxicity scores for CD4+ and CD8+ T cells. Scores were calculated for each cell based on mean expression of genes in the gene set and subtracting the background. For panels D–F, individual patients indicated in legend.

Figure 6

No signal of immune evasion…

Figure 6

No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq…

Figure 6
No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq on unsorted BMMCs, clustering was based on the cell surface protein expression and clusters were annotated by their top three or four most highly differentially expressed markers. Clusters were classified as putative leukemia using on protein expression of known leukemia-associated and myeloid markers based on the patients’ previous clinical flow cytometry records. Relative expression of HLA molecules and PD-L1 on leukemic blasts at baseline (top) and at relapse (bottom) in (A) PD-AML 1 and (B) PD-AML 5 visualized using ridge plots. Single-cell DNA and antibody-oligonucleotide sequencing of unsorted BMMCs in (C) PD-AML 1 and (D) PD-AML five shows the genomic and immunophenotypic features of the leukemic (AML, red line) and preceding/distinct CHIP (clonal hematopoiesis, gray lines) clones present at relapse as compared with day 0 (adapted from41). Left: Genomic subclones with wild type (WT), heterozygous (HET), present (Gain/+) or absent (−) features. Right: Cell surface protein expression for each subclone.
Similar articles
Cited by
References
    1. Breems DA, Van Putten WLJ, Huijgens PC, et al. . Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol 2005;23:1969–78. 10.1200/JCO.2005.06.027 - DOI - PubMed
    1. Ramos NR, Mo CC, Karp JE, et al. . Current approaches in the treatment of relapsed and refractory acute myeloid leukemia. J Clin Med 2015;4:665–95. 10.3390/jcm4040665 - DOI - PMC - PubMed
    1. Ferrara JLM, Levine JE, Reddy P, et al. . Graft-versus-host disease. Lancet 2009;373:1550–61. 10.1016/S0140-6736(09)60237-3 - DOI - PMC - PubMed
    1. van Bergen CAM, van Luxemburg-Heijs SAP, de Wreede LC, et al. . Selective graft-versus-leukemia depends on magnitude and diversity of the alloreactive T cell response. J Clin Invest 2017;127:517–29. 10.1172/JCI86175 - DOI - PMC - PubMed
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350–5. 10.1126/science.aar4060 - DOI - PMC - PubMed
Show all 76 references
Publication types
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3
Figure 3
Unique expression signatures in clonally expanded T cells at time of irAE. Cells harboring the unique TCRβ CDR3 from differentially abundant clones identified by bulk TCRβ-seq were grouped and differentially expressed genes (DEG) and cell surface proteins (DEP) were identified by supervised analyses compared with top 10 abundant T cell clones in the sample identified by bulk TCRβ-seq. (A) DEG and (B) DEP of expanded CD8+ T cell clones of interest in PD-AML two at hypothyroidism diagnosis vs other abundant CD8+ T cell clonotypes. (C) Frequencies from TCRβ-seq data of clonotypes coexpressing cell surface PD-1, TIM-3, and CD27 (four clones, top panel) and those not expressing this signature (two clones, bottom panel). (D) DEG and (E) DEP of expanded CD8+ T cell clones of interest in PD-AML three at time of diagnosis of central diabetes insipidus versus other abundant CD8+ T cell clonotypes. (F) Frequencies from TCRβ-seq data of clonotypes coexpressing cell surface PD-1, TIM-3, 4-1BB, HLA-DR, and CD27 (six clones, top panel) and those not expressing this signature (three clones, bottom panel). Heatmaps were created using the mean expression of all cells within each clone; differentially expressed markers were ordered by adjusted p values and fold changes. Adjusted p values are based on Wilcoxon rank sum test with Bonferroni corrections using all features in the dataset. ‘Difference **’ denotes adjusted p

Figure 4

Antileukemic response to combination immunotherapy…

Figure 4

Antileukemic response to combination immunotherapy was not associated with clonal T cell expansion.…

Figure 4
Antileukemic response to combination immunotherapy was not associated with clonal T cell expansion. Number of significantly expanded or contracted clones at timepoints sampled during treatment compared with baseline were calculated in (A) PB and (B) BM for two patients achieving CR by eight cycles of treatment. Decreasing blast percentage in the BM and few to no significantly expanded clones at EOC2 or EOC4 in BM that were low in frequency (

Figure 5

Stable T cell immunophenotypes in…

Figure 5

Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization…

Figure 5
Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization of T cell immunophenotyping based on cell surface expression CD4, CD8, CCR7, CD45RA, CD127 and CD25. (B) Quantification and proportion of CD4+ and CD8+ T cell subset frequencies, as a percentage of CD3+. (C) Differential gene expression analysis among different cell types with scaled log normalization. For panels A-C, T cell subsets indicated in legend. (D) Frequencies of CD4+ and CD8+ effector memory (EM) and terminal effector (TE) T cells at Day0, EOC2, and EOC4. (E) Per cent change in frequencies of CD4+ and CD8+ EM and TE cells at EOC2 and EOC4 compared with day 0. (F) T cell subset clonality in T cell subsets identified by immunophenotyping. Patients and time points indicated in key. (G) Cells expressing exhaustion (purple) and cytotoxicity (orange) predefined gene sets overlaid on integrated UMAP with exhaustion and cytotoxicity scores for CD4+ and CD8+ T cells. Scores were calculated for each cell based on mean expression of genes in the gene set and subtracting the background. For panels D–F, individual patients indicated in legend.

Figure 6

No signal of immune evasion…

Figure 6

No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq…

Figure 6
No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq on unsorted BMMCs, clustering was based on the cell surface protein expression and clusters were annotated by their top three or four most highly differentially expressed markers. Clusters were classified as putative leukemia using on protein expression of known leukemia-associated and myeloid markers based on the patients’ previous clinical flow cytometry records. Relative expression of HLA molecules and PD-L1 on leukemic blasts at baseline (top) and at relapse (bottom) in (A) PD-AML 1 and (B) PD-AML 5 visualized using ridge plots. Single-cell DNA and antibody-oligonucleotide sequencing of unsorted BMMCs in (C) PD-AML 1 and (D) PD-AML five shows the genomic and immunophenotypic features of the leukemic (AML, red line) and preceding/distinct CHIP (clonal hematopoiesis, gray lines) clones present at relapse as compared with day 0 (adapted from41). Left: Genomic subclones with wild type (WT), heterozygous (HET), present (Gain/+) or absent (−) features. Right: Cell surface protein expression for each subclone.
Similar articles
Cited by
References
    1. Breems DA, Van Putten WLJ, Huijgens PC, et al. . Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol 2005;23:1969–78. 10.1200/JCO.2005.06.027 - DOI - PubMed
    1. Ramos NR, Mo CC, Karp JE, et al. . Current approaches in the treatment of relapsed and refractory acute myeloid leukemia. J Clin Med 2015;4:665–95. 10.3390/jcm4040665 - DOI - PMC - PubMed
    1. Ferrara JLM, Levine JE, Reddy P, et al. . Graft-versus-host disease. Lancet 2009;373:1550–61. 10.1016/S0140-6736(09)60237-3 - DOI - PMC - PubMed
    1. van Bergen CAM, van Luxemburg-Heijs SAP, de Wreede LC, et al. . Selective graft-versus-leukemia depends on magnitude and diversity of the alloreactive T cell response. J Clin Invest 2017;127:517–29. 10.1172/JCI86175 - DOI - PMC - PubMed
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350–5. 10.1126/science.aar4060 - DOI - PMC - PubMed
Show all 76 references
Publication types
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Antileukemic response to combination immunotherapy was not associated with clonal T cell expansion. Number of significantly expanded or contracted clones at timepoints sampled during treatment compared with baseline were calculated in (A) PB and (B) BM for two patients achieving CR by eight cycles of treatment. Decreasing blast percentage in the BM and few to no significantly expanded clones at EOC2 or EOC4 in BM that were low in frequency (

Figure 5

Stable T cell immunophenotypes in…

Figure 5

Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization…

Figure 5
Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization of T cell immunophenotyping based on cell surface expression CD4, CD8, CCR7, CD45RA, CD127 and CD25. (B) Quantification and proportion of CD4+ and CD8+ T cell subset frequencies, as a percentage of CD3+. (C) Differential gene expression analysis among different cell types with scaled log normalization. For panels A-C, T cell subsets indicated in legend. (D) Frequencies of CD4+ and CD8+ effector memory (EM) and terminal effector (TE) T cells at Day0, EOC2, and EOC4. (E) Per cent change in frequencies of CD4+ and CD8+ EM and TE cells at EOC2 and EOC4 compared with day 0. (F) T cell subset clonality in T cell subsets identified by immunophenotyping. Patients and time points indicated in key. (G) Cells expressing exhaustion (purple) and cytotoxicity (orange) predefined gene sets overlaid on integrated UMAP with exhaustion and cytotoxicity scores for CD4+ and CD8+ T cells. Scores were calculated for each cell based on mean expression of genes in the gene set and subtracting the background. For panels D–F, individual patients indicated in legend.

Figure 6

No signal of immune evasion…

Figure 6

No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq…

Figure 6
No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq on unsorted BMMCs, clustering was based on the cell surface protein expression and clusters were annotated by their top three or four most highly differentially expressed markers. Clusters were classified as putative leukemia using on protein expression of known leukemia-associated and myeloid markers based on the patients’ previous clinical flow cytometry records. Relative expression of HLA molecules and PD-L1 on leukemic blasts at baseline (top) and at relapse (bottom) in (A) PD-AML 1 and (B) PD-AML 5 visualized using ridge plots. Single-cell DNA and antibody-oligonucleotide sequencing of unsorted BMMCs in (C) PD-AML 1 and (D) PD-AML five shows the genomic and immunophenotypic features of the leukemic (AML, red line) and preceding/distinct CHIP (clonal hematopoiesis, gray lines) clones present at relapse as compared with day 0 (adapted from41). Left: Genomic subclones with wild type (WT), heterozygous (HET), present (Gain/+) or absent (−) features. Right: Cell surface protein expression for each subclone.
Figure 5
Figure 5
Stable T cell immunophenotypes in patients with anti-leukemic responses. (A) Integrated UMAP visualization of T cell immunophenotyping based on cell surface expression CD4, CD8, CCR7, CD45RA, CD127 and CD25. (B) Quantification and proportion of CD4+ and CD8+ T cell subset frequencies, as a percentage of CD3+. (C) Differential gene expression analysis among different cell types with scaled log normalization. For panels A-C, T cell subsets indicated in legend. (D) Frequencies of CD4+ and CD8+ effector memory (EM) and terminal effector (TE) T cells at Day0, EOC2, and EOC4. (E) Per cent change in frequencies of CD4+ and CD8+ EM and TE cells at EOC2 and EOC4 compared with day 0. (F) T cell subset clonality in T cell subsets identified by immunophenotyping. Patients and time points indicated in key. (G) Cells expressing exhaustion (purple) and cytotoxicity (orange) predefined gene sets overlaid on integrated UMAP with exhaustion and cytotoxicity scores for CD4+ and CD8+ T cells. Scores were calculated for each cell based on mean expression of genes in the gene set and subtracting the background. For panels D–F, individual patients indicated in legend.
Figure 6
Figure 6
No signal of immune evasion in responders at relapse post-treatment. Through 3’v3 scRNA-seq on unsorted BMMCs, clustering was based on the cell surface protein expression and clusters were annotated by their top three or four most highly differentially expressed markers. Clusters were classified as putative leukemia using on protein expression of known leukemia-associated and myeloid markers based on the patients’ previous clinical flow cytometry records. Relative expression of HLA molecules and PD-L1 on leukemic blasts at baseline (top) and at relapse (bottom) in (A) PD-AML 1 and (B) PD-AML 5 visualized using ridge plots. Single-cell DNA and antibody-oligonucleotide sequencing of unsorted BMMCs in (C) PD-AML 1 and (D) PD-AML five shows the genomic and immunophenotypic features of the leukemic (AML, red line) and preceding/distinct CHIP (clonal hematopoiesis, gray lines) clones present at relapse as compared with day 0 (adapted from41). Left: Genomic subclones with wild type (WT), heterozygous (HET), present (Gain/+) or absent (−) features. Right: Cell surface protein expression for each subclone.

References

    1. Breems DA, Van Putten WLJ, Huijgens PC, et al. . Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol 2005;23:1969–78. 10.1200/JCO.2005.06.027
    1. Ramos NR, Mo CC, Karp JE, et al. . Current approaches in the treatment of relapsed and refractory acute myeloid leukemia. J Clin Med 2015;4:665–95. 10.3390/jcm4040665
    1. Ferrara JLM, Levine JE, Reddy P, et al. . Graft-versus-host disease. Lancet 2009;373:1550–61. 10.1016/S0140-6736(09)60237-3
    1. van Bergen CAM, van Luxemburg-Heijs SAP, de Wreede LC, et al. . Selective graft-versus-leukemia depends on magnitude and diversity of the alloreactive T cell response. J Clin Invest 2017;127:517–29. 10.1172/JCI86175
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350–5. 10.1126/science.aar4060
    1. Mueller SN, Ahmed R. High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proc Natl Acad Sci U S A 2009;106:8623–8. 10.1073/pnas.0809818106
    1. Wherry EJ, Blattman JN, Murali-Krishna K, et al. . Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol 2003;77:4911–27. 10.1128/jvi.77.8.4911-4927.2003
    1. Topalian SL, Hodi FS, Brahmer JR, et al. . Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. 10.1056/NEJMoa1200690
    1. Hamid O, Robert C, Daud A, et al. . Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013;369:134–44. 10.1056/NEJMoa1305133
    1. Barber DL, Wherry EJ, Masopust D, et al. . Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439:682–7. 10.1038/nature04444
    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov 2018;8:1069–86. 10.1158/-18-0367
    1. Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood 2009;114:1545–52. 10.1182/blood-2009-03-206672
    1. Zhou Q, Munger ME, Highfill SL, et al. . Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood 2010;116:2484–93. 10.1182/blood-2010-03-275446
    1. Goswami M, Lindblad KE, Ramraj R, et al. . B cell deficiency in patients with relapsed and refractory acute myeloid leukemia. Clin Hematol Int 2020;2:125–8. 10.2991/chi.k.200712.001
    1. Williams P, Basu S, Garcia-Manero G, 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;125:1470–81. 10.1002/cncr.31896
    1. Jia B, Wang L, Claxton DF, et al. . Bone marrow CD8 T cells express high frequency of PD-1 and exhibit reduced anti-leukemia response in newly diagnosed AML patients. Blood Cancer J 2018;8:34. 10.1038/s41408-018-0069-4
    1. Le Dieu R, Taussig DC, Ramsay AG, et al. . Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood 2009;114:3909–16. 10.1182/blood-2009-02-206946
    1. Knaus HA, Berglund S, Hackl H, et al. . Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight 2018;310.1172/jci.insight.120974
    1. Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. . T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol 2015;8:93. 10.1186/s13045-015-0189-2
    1. Lindblad KE, Goswami M, Hourigan CS, et al. . Immunological effects of hypomethylating agents. Expert Rev Hematol 2017;10:745–52. 10.1080/17474086.2017.1346470
    1. Yang H, Bueso-Ramos C, DiNardo C, et al. . Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2014;28:1280–8. 10.1038/leu.2013.355
    1. Ørskov AD, Treppendahl MB, Skovbo A, et al. . Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: a rationale for combined targeting of PD-1 and DNA methylation. Oncotarget 2015;6:9612–26. 10.18632/oncotarget.3324
    1. Goodyear O, Agathanggelou A, Novitzky-Basso I, et al. . Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood 2010;116:1908–18. 10.1182/blood-2009-11-249474
    1. Srivastava P, Paluch BE, Matsuzaki J, et al. . Induction of cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monotherapy. Oncotarget 2016;7:12840–56. 10.18632/oncotarget.7326
    1. Chiappinelli KB, Strissel PL, Desrichard A, et al. . Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 2015;162:974–86. 10.1016/j.cell.2015.07.011
    1. Roulois D, Loo Yau H, Singhania R, et al. . DNA-Demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 2015;162:961–73. 10.1016/j.cell.2015.07.056
    1. Jiang TT, Martinov T, Xin L, et al. . Programmed Death-1 Culls Peripheral Accumulation of High-Affinity Autoreactive CD4 T Cells to Protect against Autoimmunity. Cell Rep 2016;17:1783–94. 10.1016/j.celrep.2016.10.042
    1. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 2010;236:219–42. 10.1111/j.1600-065X.2010.00923.x
    1. Tallman MS, Wang ES, Altman JK, et al. . Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2019;17:721-749. 10.6004/jnccn.2019.0028
    1. Welch JS, Petti AA, Miller CA, et al. . TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med 2016;375:2023–36. 10.1056/NEJMoa1605949
    1. Oetjen KA, Lindblad KE, Goswami M, et al. . Human bone marrow assessment by single-cell RNA sequencing, mass cytometry, and flow cytometry. JCI Insight 2018;310.1172/jci.insight.124928
    1. Carlson CS, Emerson RO, Sherwood AM, et al. . Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun 2013;4:2680. 10.1038/ncomms3680
    1. Robins HS, Campregher PV, Srivastava SK, et al. . Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 2009;114:4099–107. 10.1182/blood-2009-04-217604
    1. Lefranc M-P, Giudicelli V, Ginestoux C, et al. . IMGT, the International immunogenetics information system. Nucleic Acids Res 2009;37:D1006–12. 10.1093/nar/gkn838
    1. DeWitt WS, Emerson RO, Lindau P, et al. . Dynamics of the cytotoxic T cell response to a model of acute viral infection. J Virol 2015;89:4517–26. 10.1128/JVI.03474-14
    1. Rytlewski J, Deng S, Xie T, et al. . Model to improve specificity for identification of clinically-relevant expanded T cells in peripheral blood. PLoS One 2019;14:e0213684. 10.1371/journal.pone.0213684
    1. Dillon LW, Ghannam J, Nosiri C, et al. . Personalized single-cell Proteogenomics to distinguish acute myeloid leukemia from non-malignant clonal hematopoiesis. Blood Cancer Discov 2021;2:319–25. 10.1158/2643-3230.BCD-21-0046
    1. Hopkins AC, Yarchoan M, Durham JN, et al. . T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight 2018;310.1172/jci.insight.122092
    1. Hogan SA, Courtier A, Cheng PF, et al. . Peripheral blood TCR repertoire profiling may facilitate patient stratification for immunotherapy against melanoma. Cancer Immunol Res 2019;7:77–85. 10.1158/2326-6066.CIR-18-0136
    1. Lee H, Hodi FS, Giobbie-Hurder A, et al. . Characterization of thyroid disorders in patients receiving immune checkpoint inhibition therapy. Cancer Immunol Res 2017;5:1133–40. 10.1158/2326-6066.CIR-17-0208
    1. de Filette J, Jansen Y, Schreuer M, et al. . Incidence of thyroid-related adverse events in melanoma patients treated with pembrolizumab. J Clin Endocrinol Metab 2016;101:4431–9. 10.1210/jc.2016-2300
    1. McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr Rev 2014;35:59–105. 10.1210/er.2013-1055
    1. Cohen NR, Brennan PJ, Shay T, et al. . Shared and distinct transcriptional programs underlie the hybrid nature of iNKT cells. Nat Immunol 2013;14:90–9. 10.1038/ni.2490
    1. Cha E, et al. . Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med 2014;6:ra270.
    1. Zhang J-Y, Wang X-M, Xing X, et al. . Single-Cell landscape of immunological responses in patients with COVID-19. Nat Immunol 2020;21:1107–18. 10.1038/s41590-020-0762-x
    1. Christopher MJ, Petti AA, Rettig MP, et al. . Immune escape of relapsed AML cells after allogeneic transplantation. N Engl J Med 2018;379:2330–41. 10.1056/NEJMoa1808777
    1. Vago L, Perna SK, Zanussi M, et al. . Loss of mismatched HLA in leukemia after stem-cell transplantation. N Engl J Med 2009;361:478–88. 10.1056/NEJMoa0811036
    1. Daver N, Garcia-Manero G, Basu S, 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;9:370–83. 10.1158/-18-0774
    1. Ritchie EK, Feldman EJ, Christos PJ, et al. . Decitabine in patients with newly diagnosed and relapsed acute myeloid leukemia. Leuk Lymphoma 2013;54:2003–7. 10.3109/10428194.2012.762093
    1. Khan N, Hantel A, Knoebel RW, et al. . Efficacy of single-agent decitabine in relapsed and refractory acute myeloid leukemia. Leuk Lymphoma 2017;58:1–7. 10.1080/10428194.2017.1289524
    1. Bouligny IM, Mehta V, Isom S, et al. . Efficacy of 10-day decitabine in acute myeloid leukemia. Leuk Res 2021;103:106524. 10.1016/j.leukres.2021.106524
    1. Lessi F, Laurino M, Papayannidis C, et al. . Hypomethylating agents (HMAs) as salvage therapy in relapsed or refractory AML: an Italian multicentric retrospective study. Biomedicines 2021;9. 10.3390/biomedicines9080972. [Epub ahead of print: 06 08 2021].
    1. Stahl M, DeVeaux M, Montesinos P, et al. . Hypomethylating agents in relapsed and refractory AML: outcomes and their predictors in a large international patient cohort. Blood Adv 2018;2:923–32. 10.1182/bloodadvances.2018016121
    1. Zeidan AM, Cavenagh J, Voso MT, et al. . Efficacy and safety of azacitidine (aza) in combination with the anti-PD-L1 Durvalumab (durva) for the front-line treatment of older patients (PTS) with acute myeloid leukemia (AML) who are unfit for intensive chemotherapy (IC) and PTS with higher-risk myelodysplastic syndromes (HR-MDS): results from a large, international, randomized phase 2 study. Blood 2019;134:829. 10.1182/blood-2019-122896
    1. Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. . Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol 2018;4:173–82. 10.1001/jamaoncol.2017.3064
    1. Zhao C, Tella SH, Del Rivero J, et al. . Anti-PD-L1 treatment induced central diabetes insipidus. J Clin Endocrinol Metab 2018;103:365–9. 10.1210/jc.2017-01905
    1. Wang AY, Kline J, Stock W, et al. . Unexpected toxicities when nivolumab was given as maintenance therapy following allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2020;26:1025–7. 10.1016/j.bbmt.2020.01.021
    1. Gesiotto QJ, Swoboda DM, Shallis RM, et al. . Evaluating predictors of immune-related adverse events and response to checkpoint inhibitors in myeloid malignancies. Clin Lymphoma Myeloma Leuk 2021;21:421–4. 10.1016/j.clml.2021.01.010
    1. Oh DY, Cham J, Zhang L, et al. . Immune toxicities Elicted by CTLA-4 blockade in cancer patients are associated with early diversification of the T-cell repertoire. Cancer Res 2017;77:1322–30. 10.1158/0008-5472.CAN-16-2324
    1. Subudhi SK, Aparicio A, Gao J, et al. . Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci U S A 2016;113:11919–24. 10.1073/pnas.1611421113
    1. Luoma AM, Suo S, Williams HL, et al. . Molecular pathways of colon inflammation induced by cancer immunotherapy. Cell 2020;182:655–71. 10.1016/j.cell.2020.06.001
    1. Johnson DB, McDonnell WJ, Gonzalez-Ericsson PI, et al. . A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat Med 2019;25:1243–50. 10.1038/s41591-019-0523-2
    1. Kong Y, Zhang J, Claxton DF, et al. . PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J 2015;5:e330. 10.1038/bcj.2015.58
    1. Blackburn SD, Shin H, Haining WN, et al. . Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 2009;10:29–37. 10.1038/ni.1679
    1. Vadakekolathu J, Minden MD, Hood T, et al. . Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci Transl Med 2020;1210.1126/scitranslmed.aaz0463
    1. Abbas HA, et al. . Single-cell characterization of acute myeloid leukemia and its microenvironment following PD-1 blockade based therapy. Biorxiv 2020.
    1. Kurtulus S, Madi A, Escobar G, et al. . Checkpoint blockade immunotherapy induces dynamic changes in PD-1-CD8+ tumor-infiltrating T cells. Immunity 2019;50:181–94. 10.1016/j.immuni.2018.11.014
    1. Yost KE, Satpathy AT, Wells DK, et al. . Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 2019;25:1251–9. 10.1038/s41591-019-0522-3
    1. Miller BC, Sen DR, Al Abosy R, et al. . Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019;20:326–36. 10.1038/s41590-019-0312-6
    1. Thommen DS, Koelzer VH, Herzig P, et al. . A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 2018;24:994–1004. 10.1038/s41591-018-0057-z
    1. Daver N, Boddu P, Garcia-Manero G, et al. . Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Leukemia 2018;32:1094–105. 10.1038/s41375-018-0070-8
    1. Ghoneim HE, Fan Y, Moustaki A, et al. . De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 2017;170:142–57. 10.1016/j.cell.2017.06.007
    1. Pauken KE, Sammons MA, Odorizzi PM, et al. . Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016;354:1160–5. 10.1126/science.aaf2807
    1. Strauss L, Mahmoud MAA, Weaver JD, et al. . Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 2020;5:1. 10.1126/sciimmunol.aay1863
    1. Krieg C, Nowicka M, Guglietta S, et al. . High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med 2018;24:144–53. 10.1038/nm.4466
    1. Çelik H, Lindblad KE, Popescu B, et al. . Highly multiplexed proteomic assessment of human bone marrow in acute myeloid leukemia. Blood Adv 2020;4:367–79. 10.1182/bloodadvances.2019001124

Source: PubMed

3
Abonneren