Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Haematologica ; 109(4): 1082-1094, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37941406

ABSTRACT

Oral azacitidine (oral-Aza) treatment results in longer median overall survival (OS) (24.7 vs. 14.8 months in placebo) in patients with acute myeloid leukemia (AML) in remission after intensive chemotherapy. The dosing schedule of oral-Aza (14 days/28-day cycle) allows for low exposure of Aza for an extended duration thereby facilitating a sustained therapeutic effect. However, the underlying mechanisms supporting the clinical impact of oral-Aza in maintenance therapy remain to be fully understood. In this preclinical work, we explore the mechanistic basis of oral-Aza/extended exposure to Aza through in vitro and in vivo modeling. In cell lines, extended exposure to Aza results in sustained DNMT1 loss, leading to durable hypomethylation, and gene expression changes. In mouse models, extended exposure to Aza, preferentially targets immature leukemic cells. In leukemic stem cell (LSC) models, the extended dose of Aza induces differentiation and depletes CD34+CD38- LSC. Mechanistically, LSC differentiation is driven in part by increased myeloperoxidase (MPO) expression. Inhibition of MPO activity either by using an MPO-specific inhibitor or blocking oxidative stress, a known mechanism of MPO, partly reverses the differentiation of LSC. Overall, our preclinical work reveals novel mechanistic insights into oral-Aza and its ability to target LSC.


Subject(s)
Azacitidine , Leukemia, Myeloid, Acute , Animals , Mice , Humans , Azacitidine/pharmacology , Azacitidine/therapeutic use , Antigens, CD34/metabolism , Leukemia, Myeloid, Acute/genetics , Peroxidase , Stem Cells/metabolism
2.
Proc Natl Acad Sci U S A ; 117(25): 14331-14341, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513686

ABSTRACT

Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with approximately four new cases per 100,000 persons per year. Standard treatment for AML consists of induction chemotherapy with remission achieved in 50 to 75% of cases. Unfortunately, most patients will relapse and die from their disease, as 5-y survival is roughly 29%. Therefore, other treatment options are urgently needed. In recent years, immune-based therapies have led to unprecedented rates of survival among patients with some advanced cancers. Suppression of T cell function in the tumor microenvironment is commonly observed and may play a role in AML. We found that there is a significant association between T cell infiltration in the bone marrow microenvironment of newly diagnosed patients with AML and increased overall survival. Functional studies aimed at establishing the degree of T cell suppression in patients with AML revealed impaired T cell function in many patients. In most cases, T cell proliferation could be restored by blocking the immune checkpoint molecules PD-1, CTLA-4, or TIM3. Our data demonstrate that AML establishes an immune suppressive environment in the bone marrow, in part through T cell checkpoint function.


Subject(s)
Bone Marrow/metabolism , Leukemia, Myeloid, Acute/metabolism , T-Lymphocytes/metabolism , Tumor Microenvironment/physiology , Bone Marrow/immunology , CTLA-4 Antigen/metabolism , Cell Proliferation , Cytokines/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology
3.
Blood ; 133(6): 588-599, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30425048

ABSTRACT

To identify new therapeutic targets in acute myeloid leukemia (AML), we performed small-molecule and small-interfering RNA (siRNA) screens of primary AML patient samples. In 23% of samples, we found sensitivity to inhibition of colony-stimulating factor 1 (CSF1) receptor (CSF1R), a receptor tyrosine kinase responsible for survival, proliferation, and differentiation of myeloid-lineage cells. Sensitivity to CSF1R inhibitor GW-2580 was found preferentially in de novo and favorable-risk patients, and resistance to GW-2580 was associated with reduced overall survival. Using flow cytometry, we discovered that CSF1R is not expressed on the majority of leukemic blasts but instead on a subpopulation of supportive cells. Comparison of CSF1R-expressing cells in AML vs healthy donors by mass cytometry revealed expression of unique cell-surface markers. The quantity of CSF1R-expressing cells correlated with GW-2580 sensitivity. Exposure of primary AML patient samples to a panel of recombinant cytokines revealed that CSF1R inhibitor sensitivity correlated with a growth response to CSF1R ligand, CSF1, and other cytokines, including hepatocyte growth factor (HGF). The addition of CSF1 increased the secretion of HGF and other cytokines in conditioned media from AML patient samples, whereas adding GW-2580 reduced their secretion. In untreated cells, HGF levels correlated significantly with GW-2580 sensitivity. Finally, recombinant HGF and HS-5-conditioned media rescued cell viability after GW-2580 treatment in AML patient samples. Our results suggest that CSF1R-expressing cells support the bulk leukemia population through the secretion of HGF and other cytokines. This study identifies CSF1R as a novel therapeutic target of AML and provides a mechanism of paracrine cytokine/growth factor signaling in this disease.


Subject(s)
Anisoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Paracrine Communication/drug effects , Pyrimidines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Cell Differentiation , Cell Survival , Culture Media, Conditioned/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Prognosis , Survival Rate , Tumor Cells, Cultured
4.
Nature ; 488(7413): 656-9, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22763442

ABSTRACT

Mutations in the IDH1 and IDH2 genes encoding isocitrate dehydrogenases are frequently found in human glioblastomas and cytogenetically normal acute myeloid leukaemias (AML). These alterations are gain-of-function mutations in that they drive the synthesis of the 'oncometabolite' R-2-hydroxyglutarate (2HG). It remains unclear how IDH1 and IDH2 mutations modify myeloid cell development and promote leukaemogenesis. Here we report the characterization of conditional knock-in (KI) mice in which the most common IDH1 mutation, IDH1(R132H), is inserted into the endogenous murine Idh1 locus and is expressed in all haematopoietic cells (Vav-KI mice) or specifically in cells of the myeloid lineage (LysM-KI mice). These mutants show increased numbers of early haematopoietic progenitors and develop splenomegaly and anaemia with extramedullary haematopoiesis, suggesting a dysfunctional bone marrow niche. Furthermore, LysM-KI cells have hypermethylated histones and changes to DNA methylation similar to those observed in human IDH1- or IDH2-mutant AML. To our knowledge, our study is the first to describe the generation and characterization of conditional IDH1(R132H)-KI mice, and also the first report to demonstrate the induction of a leukaemic DNA methylation signature in a mouse model. Our report thus sheds light on the mechanistic links between IDH1 mutation and human AML.


Subject(s)
Epigenesis, Genetic/genetics , Hematopoietic Stem Cells/cytology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Aging , Animals , Bone Marrow/pathology , Cell Lineage , CpG Islands/genetics , DNA Methylation , Disease Models, Animal , Female , Gene Knock-In Techniques , Glioma/pathology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mutant Proteins/genetics , Myeloid Cells/cytology , Myeloid Cells/metabolism , Spleen/pathology
5.
J Immunol ; 195(10): 4632-40, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26447227

ABSTRACT

TLR-induced maturation of dendritic cells (DCs) leads to the production of proinflammatory cytokines as well as the upregulation of various molecules involved in T cell activation. These are believed to be the critical events that account for the induction of the adaptive immune response. In this study, we have examined the role of miR-155 in DC function and the induction of immunity. Using a model in which the transfer of self-Ag-pulsed, TLR-matured DCs can induce a functional CD8 T cell response and autoimmunity, we find that DCs lacking miR-155 have an impaired ability to break immune tolerance. Importantly, transfer of self- Ag-pulsed DCs overexpressing miR-155 was sufficient to break tolerance in the absence of TLR stimuli. Although these unstimulated DCs induced T cell function in vivo, there was no evidence for the upregulation of costimulatory ligands or cytokine secretion. Further analysis showed that miR-155 influenced the level of the phosphatase SHIP1 in DCs and that the lack of SHIP1 in DCs was sufficient to break T cell tolerance in vivo, again in the absence of TLR-induced DC maturation. Our study demonstrates that the overexpression of miR-155 in DCs is a critical event that is alone sufficient to break self-tolerance and promote a CD8-mediated autoimmune response in vivo. This process is independent of the induction of conventional DC maturation markers, indicating that miR-155 regulation of SHIP represents a unique axis that regulates DC function in vivo.


Subject(s)
Dendritic Cells/metabolism , Immune Tolerance/immunology , MicroRNAs/biosynthesis , Phosphoric Monoester Hydrolases/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Immune Tolerance/genetics , Inositol Polyphosphate 5-Phosphatases , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Up-Regulation
6.
Eur J Immunol ; 44(1): 11-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24571026

ABSTRACT

The activation of T cells is a tightly regulated process that has evolved to maximize protective immune responses to pathogens while minimizing damage to self-tissues. A delicate balance of cell-intrinsic, costimulatory, and transcriptional pathways as well as micro-environmental cues such as local cytokines controls the magnitude and nature of T-cell responses in vivo. The discovery of functional small noncoding RNAs called micro-RNAs (miRNAs) has introduced new mechanisms that contribute to the regulation of protein translation and cellular responses to stimuli. miRNAs are short (approximately 22 bp) RNA species, which bind to mRNAs and suppress translation. Due to their short length and imperfect base pairing requirements, each miRNA has the potential to regulate various pathways through the translational inhibition of multiple mRNAs. The human and mouse genomes each encode hundreds of miRNAs, and studying the function of miRNAs has led to the realization that they play important roles in diverse biological processes from development and cancer to immunity. This review focuses on the function of mir-155 in T cells and the impact of this miRNA on autoimmunity, tumor immunity, and pathogen-induced immunity.


Subject(s)
Infections/immunology , MicroRNAs/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Autoimmunity/genetics , Genome/immunology , Humans , Immunity, Cellular/genetics , Immunomodulation , Infections/genetics , Mice , Neoplasms/genetics , T-Lymphocytes/microbiology
7.
J Immunol ; 190(3): 1210-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275599

ABSTRACT

Recent studies have begun to define the role of micro-RNAs in regulating the immune response. Micro-RNA155 (mir-155) has been shown to play a role in germinal center formation, T cell inflammation, and regulatory T cell development. In this study, we evaluated the role of mir-155 in cytotoxic T cell function. We report in this study that mice lacking mir-155 have impaired CD8(+) T cell responses to infections with lymphocytic choriomeningitis virus and the intracellular bacteria Listeria monocytogenes. We show by a series of adoptive transfer studies that the impaired CD8(+) T cell response to L. monocytogenes is T cell intrinsic. In addition, we observed that CD8(+) T cells lacking mir-155 have impaired activation of the prosurvival Akt pathway after TCR cross-linking. These data suggest that mir-155 may be a good target for therapies aimed at modulating immune responses.


Subject(s)
Listeriosis/immunology , Lymphocytic Choriomeningitis/immunology , MicroRNAs/physiology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Bacterial/immunology , Apoptosis/immunology , Clonal Selection, Antigen-Mediated , Cytotoxicity, Immunologic , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Peptide Fragments/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/transplantation
8.
Front Immunol ; 15: 1297338, 2024.
Article in English | MEDLINE | ID: mdl-38495876

ABSTRACT

Dendritic cells (DC) are mediators between innate and adaptive immune responses to pathogens and tumors. DC development is determined by signaling through the receptor tyrosine kinase Fms-like tyrosine kinase 3 (FLT3) in bone marrow myeloid progenitors. Recently the naming conventions for DC phenotypes have been updated to distinguish between "Conventional" DCs (cDCs) and plasmacytoid DCs (pDCs). Activating mutations of FLT3, including Internal Tandem Duplication (FLT3-ITD), are associated with poor prognosis for acute myeloid leukemia (AML) patients. Having a shared myeloid lineage it can be difficult to distinguish bone fide DCs from AML tumor cells. To date, there is little information on the effects of FLT3-ITD in DC biology. To further elucidate this relationship we utilized CITE-seq technology in combination with flow cytometry and multiplex immunoassays to measure changes to DCs in human and mouse tissues. We examined the cDC phenotype and frequency in bone marrow aspirates from patients with AML to understand the changes to cDCs associated with FLT3-ITD. When compared to healthy donor (HD) we found that a subset of FLT3-ITD+ AML patient samples have overrepresented populations of cDCs and disrupted phenotypes. Using a mouse model of FLT3-ITD+ AML, we found that cDCs were increased in percentage and number compared to control wild-type (WT) mice. Single cell RNA-seq identified FLT3-ITD+ cDCs as skewed towards a cDC2 T-bet- phenotype, previously shown to promote Th17 T cells. We assessed the phenotypes of CD4+ T cells in the AML mice and found significant enrichment of both Treg and Th17 CD4+ T cells in the bone marrow and spleen compartments. Ex vivo stimulation of CD4+ T cells also showed increased Th17 phenotype in AML mice. Moreover, co-culture of AML mouse-derived DCs and naïve OT-II cells preferentially skewed T cells into a Th17 phenotype. Together, our data suggests that FLT3-ITD+ leukemia-associated cDCs polarize CD4+ T cells into Th17 subsets, a population that has been shown to be negatively associated with survival in solid tumor contexts. This illustrates the complex tumor microenvironment of AML and highlights the need for further investigation into the effects of FLT3-ITD mutations on DC phenotypes and their downstream effects on Th polarization.


Subject(s)
Leukemia, Myeloid, Acute , fms-Like Tyrosine Kinase 3 , Animals , Humans , Mice , Dendritic Cells/pathology , fms-Like Tyrosine Kinase 3/genetics , Homeostasis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Tumor Microenvironment/genetics
9.
Proc Natl Acad Sci U S A ; 107(6): 2622-7, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133759

ABSTRACT

Metastasis leads to the death of most cancer patients, and basal breast cancer is the most aggressive breast tumor type. Metastasis involves a complex cell migration process dependent on cytoskeletal remodeling such that targeting such remodeling in tumor cells could be clinically beneficial. Here we show that Hormonally Up-regulated Neu-associated Kinase (HUNK) is dramatically down-regulated in tumor samples and cell lines derived from basal breast cancers. Reconstitution of HUNK expression in basal breast cancer cell lines blocked actin polymerization and reduced cell motility, resulting in decreased metastases in two in vivo murine cancer models. Mechanistically, HUNK overexpression sustained the constitutive phosphorylation and inactivation of cofilin-1 (CFL-1), thereby blocking the incorporation of new actin monomers into actin filaments. HUNK reconstitution in basal breast cancer cell lines prevented protein phosphatase 2-A (PP2A), a phosphatase putatively acting on CFL-1, from binding to CFL-1. Our investigation of HUNK suggests that the interaction between PP2A and CFL-1 may be a target for antimetastasis therapy, particularly for basal breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Cofilin 1/metabolism , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Actins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cofilin 1/genetics , Cytoskeleton/metabolism , Female , Humans , Immunoblotting , Immunoprecipitation , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Protein Binding , Protein Kinases/genetics , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Transplantation, Heterologous
10.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37781631

ABSTRACT

Dendritic cells (DC) are mediators of adaptive immune responses to pathogens and tumors. DC development is determined by signaling through the receptor tyrosine kinase Fms-like tyrosine kinase 3 (FLT3) in bone marrow myeloid progenitors. Recently the naming conventions for DC phenotypes have been updated to distinguish between "Conventional" DCs (cDCs) and plasmacytoid DCs (pDCs). Activating mutations of FLT3, including Internal Tandem Duplication (FLT3-ITD), are associated with poor prognosis for leukemia patients. To date, there is little information on the effects of FLT3-ITD in DC biology. We examined the cDC phenotype and frequency in bone marrow aspirates from patients with acute myeloid leukemia (AML) to understand the changes to cDCs associated with FLT3-ITD. When compared to healthy donor (HD) we found that a subset of FLT3-ITD+ AML patient samples have overrepresented populations of cDCs and disrupted phenotypes. Using a mouse model of FLT3-ITD+ AML, we found that cDCs were increased in percentage and number compared to control wild-type (WT) mice. Single cell RNA-seq identified FLT3-ITD+ cDCs as skewed towards a cDC2 T-bet - phenotype, previously shown to promote Th17 T cells. We assessed the phenotypes of CD4+ T cells in the AML mice and found significant enrichment of both Treg and Th17 CD4+ T cells. Furthermore, co-culture of AML mouse- derived DCs and naïve OT-II cells preferentially skewed T cells into a Th17 phenotype. Together, our data suggests that FLT3-ITD+ leukemia-associated cDCs polarize CD4+ T cells into Th17 subsets, a population that has been shown to be negatively associated with survival in solid tumor contexts. This illustrates the complex tumor microenvironment of AML and highlights the need for further investigation into the effects of FLT3-ITD mutations on DC phenotypes.

11.
Leukemia ; 37(3): 580-592, 2023 03.
Article in English | MEDLINE | ID: mdl-36681742

ABSTRACT

Many acute myeloid leukemia (AML) patients exhibit hallmarks of immune exhaustion, such as increased myeloid-derived suppressor cells, suppressive regulatory T cells and dysfunctional T cells. Similarly, we have identified the same immune-related features, including exhausted CD8+ T cells (TEx) in a mouse model of AML. Here we show that inhibitors that target bromodomain and extra-terminal domain (BET) proteins affect tumor-intrinsic factors but also rescue T cell exhaustion and ICB resistance. Ex vivo treatment of cells from AML mice and AML patients with BET inhibitors (BETi) reversed CD8+ T cell exhaustion by restoring proliferative capacity and expansion of the more functional precursor-exhausted T cells. This reversal was enhanced by combined BETi and anti-PD1 treatment. BETi synergized with anti-PD1 in vivo, resulting in the reduction of circulating leukemia cells, enrichment of CD8+ T cells in the bone marrow, and increase in expression of Tcf7, Slamf6, and Cxcr5 in CD8+ T cells. Finally, we profiled the epigenomes of in vivo JQ1-treated AML-derived CD8+ T cells by single-cell ATAC-seq and found that JQ1 increases Tcf7 accessibility specifically in Tex cells, suggesting that BETi likely acts mechanistically by relieving repression of progenitor programs in Tex CD8+ T cells and maintaining a pool of anti-PD1 responsive CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Leukemia, Myeloid, Acute , Animals , Mice , Leukemia, Myeloid, Acute/metabolism , T-Lymphocytes, Regulatory
12.
Nature ; 442(7106): 997-1002, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16921386

ABSTRACT

Contrary to the proinflammatory role of mast cells in allergic disorders, the results obtained in this study establish that mast cells are essential in CD4+CD25+Foxp3+ regulatory T (T(Reg))-cell-dependent peripheral tolerance. Here we confirm that tolerant allografts, which are sustained owing to the immunosuppressive effects of T(Reg) cells, acquire a unique genetic signature dominated by the expression of mast-cell-gene products. We also show that mast cells are crucial for allograft tolerance, through the inability to induce tolerance in mast-cell-deficient mice. High levels of interleukin (IL)-9--a mast cell growth and activation factor--are produced by activated T(Reg) cells, and IL-9 production seems important in mast cell recruitment to, and activation in, tolerant tissue. Our data indicate that IL-9 represents the functional link through which activated T(Reg) cells recruit and activate mast cells to mediate regional immune suppression, because neutralization of IL-9 greatly accelerates allograft rejection in tolerant mice. Finally, immunohistochemical analysis clearly demonstrates the existence of this novel T(Reg)-IL-9-mast cell relationship within tolerant allografts.


Subject(s)
Immune Tolerance/immunology , Mast Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Count , Gene Expression Regulation/genetics , Graft Rejection/immunology , Interleukin-9/immunology , Interleukin-9/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred Strains , Skin/cytology , Skin/immunology , Skin Transplantation/immunology , Time Factors , Transplantation, Homologous/immunology
13.
Cancer Cell ; 40(8): 850-864.e9, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35868306

ABSTRACT

Acute myeloid leukemia (AML) is a cancer of myeloid-lineage cells with limited therapeutic options. We previously combined ex vivo drug sensitivity with genomic, transcriptomic, and clinical annotations for a large cohort of AML patients, which facilitated discovery of functional genomic correlates. Here, we present a dataset that has been harmonized with our initial report to yield a cumulative cohort of 805 patients (942 specimens). We show strong cross-cohort concordance and identify features of drug response. Further, deconvoluting transcriptomic data shows that drug sensitivity is governed broadly by AML cell differentiation state, sometimes conditionally affecting other correlates of response. Finally, modeling of clinical outcome reveals a single gene, PEAR1, to be among the strongest predictors of patient survival, especially for young patients. Collectively, this report expands a large functional genomic resource, offers avenues for mechanistic exploration and drug development, and reveals tools for predicting outcome in AML.


Subject(s)
Leukemia, Myeloid, Acute , Cell Differentiation , Cohort Studies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Receptors, Cell Surface/genetics , Transcriptome
14.
Leukemia ; 35(1): 156-168, 2021 01.
Article in English | MEDLINE | ID: mdl-32203139

ABSTRACT

Novel targeted agents used in therapy of lymphoid malignancies, such as inhibitors of B-cell receptor-associated kinases, are recognized to have complex immune-mediated effects. NEDD8-activating enzyme (NAE) has been identified as a tractable target in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma. We and others have shown that pevonedistat (TAK-924), a small-molecule inhibitor of NAE, abrogates NF-κB signaling in malignant B cells. However, NF-κB pathway activity is indispensable in immune response, and T-cell function is altered in patients with CLL. Using T cells derived from patients with CLL, we demonstrate that although targeting NAE results in markedly differential expression of NF-κB-regulated genes and downregulation of interleukin (IL)-2 signaling during T-cell activation, T cells evade apoptosis. Meanwhile, NAE inhibition favorably modulates polarization of T cells in vitro, with decreased Treg differentiation and a shift toward TH1 phenotype, accompanied by increased interferon-γ production. These findings were recapitulated in vivo in immunocompetent mouse models. T cells exposed to pevonedistat in washout experiments, informed by its human pharmacokinetic profile, recover NAE activity, and maintain their response to T-cell receptor stimulation and cytotoxic potential. Our data shed light on the potential immune implications of targeting neddylation in CLL and lymphoid malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , Immunomodulation/drug effects , Leukemia, Prolymphocytic, T-Cell/immunology , Leukemia, Prolymphocytic, T-Cell/metabolism , NEDD8 Protein/antagonists & inhibitors , NEDD8 Protein/metabolism , Pyrimidines/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclopentanes/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Leukemia, Prolymphocytic, T-Cell/drug therapy , Leukemia, Prolymphocytic, T-Cell/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Models, Biological , Pyrimidines/therapeutic use , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
15.
Blood Adv ; 3(20): 3038-3051, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31648326

ABSTRACT

Acute myeloid leukemia (AML) remains difficult to treat due to mutational heterogeneity and the development of resistance to therapy. Targeted agents, such as MEK inhibitors, may be incorporated into treatment; however, the impact of MEK inhibitors on the immune microenvironment in AML is not well understood. A greater understanding of the implications of MEK inhibition on immune responses may lead to a greater understanding of immune evasion and more rational combinations with immunotherapies. This study describes the impact of trametinib on both T cells and AML blast cells by using an immunosuppressive mouse model of AML and primary patient samples. We also used a large AML database of functional drug screens to understand characteristics of trametinib-sensitive samples. In the mouse model, trametinib increased T-cell viability and restored T-cell proliferation. Importantly, we report greater proliferation in the CD8+CD44+ effector subpopulation and impaired activation of CD8+CD62L+ naive cells. Transcriptome analysis revealed that trametinib-sensitive samples have an inflammatory gene expression profile, and we also observed increased programmed cell death ligand 1 (PD-L1) expression on trametinib-sensitive samples. Finally, we found that trametinib consistently reduced PD-L1 and PD-L2 expression in a dose-dependent manner on the myeloid population. Altogether, our data present greater insight into the impact of trametinib on the immune microenvironment and characteristics of trametinib-sensitive patient samples.


Subject(s)
Immunomodulation , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Profiling/methods , Humans , Immunomodulation/drug effects , Immunophenotyping , Leukemia, Myeloid, Acute/pathology , Lymphocyte Activation/immunology , MAP Kinase Signaling System/drug effects , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
16.
Front Oncol ; 8: 213, 2018.
Article in English | MEDLINE | ID: mdl-29951373

ABSTRACT

Immunotherapies, such as chimeric antigen receptor T cells, bispecific antibodies, and immune checkpoint inhibitors, have emerged as promising modalities in multiple hematologic malignancies. Despite the excitement surrounding immunotherapy, it is currently not possible to predict which patients will respond. Within solid tumors, the status of the immune microenvironment provides valuable insight regarding potential responses to immune therapies. Much less is known about the immune microenvironment within hematologic malignancies but the characteristics of this environment are likely to serve a similar predictive role. Acute myeloid leukemia (AML) is the most common hematologic malignancy in adults, and only 25% of patients are alive 5 years following their diagnosis. There is evidence that manipulation of the immune microenvironment by leukemia cells may play a role in promoting therapy resistance and disease relapse. In addition, it has long been documented that through modulation of the immune system following allogeneic bone marrow transplant, AML can be cured, even in patients with the highest risk disease. These concepts, along with the poor prognosis associated with this disease, have encouraged many groups to start exploring the utility of novel immune therapies in AML. While the implementation of these therapies into clinical trials for AML has been supported by preclinical rationale, many questions still exist surrounding their efficacy, tolerability, and the overall optimal approach. In this review, we discuss what is known about the immune microenvironment within AML with a specific focus on T cells and checkpoints, along with their implications for immune therapies.

17.
J Immunol Methods ; 453: 44-52, 2018 02.
Article in English | MEDLINE | ID: mdl-29175391

ABSTRACT

A hallmark of the development of cancer is its ability to avoid detection and elimination by the immune system. There are many identified mechanisms of this immune evasion that can be measured both phenotypically and functionally. Functional studies directly show the ability of the tumor microenvironment to suppress immune responses, typically measured as lymphocyte proliferation, cytokine production or killing ability. While a direct measurement of function is ideal, these assays require ex vivo activation which may not accurately mimic in vivo conditions. Phenotypic assays can directly measure the distribution and activation of immune cell types rapidly after isolation, preserving the conditions present in the patient. While conventional flow cytometry is a rapid and well established assay, it currently allows for measurement of only 12-14 parameters. Mass spectrometry-based flow cytometry, or CyTOF, offers the ability to measure 3-fold more parameters than conventional optical-based modalities providing an advantage in depth of analysis that can be crucial for precious human samples. The goal of this report is to describe the system our group has developed to measure both the phenotype and function of immune cells in the bone marrow of patients with acute myeloid leukemia. We hope to explain our system in the context of previous studies aimed at measuring immune status in tumors and to inform the reader as to some experimental approaches our group has found useful in developing the basic data required to rationally pursue immune-based therapies for patients with cancer.


Subject(s)
Flow Cytometry/methods , Leukemia, Myeloid, Acute/immunology , Mass Spectrometry/methods , Animals , Cell Separation , Humans , Immune Tolerance , Immunophenotyping , Leukemia, Myeloid, Acute/diagnosis , Monitoring, Immunologic , Tumor Microenvironment
18.
Cell Rep ; 24(5): 1123-1135, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067970

ABSTRACT

Successful anti-viral response requires the sustained activation and expansion of CD8+ T cells for periods that far exceed the time limit of physical T cell interaction with antigen-presenting cells (APCs). The expanding CD8+ T cell pool generates the effector and memory cell populations that provide viral clearance and long-term immunity, respectively. Here, we demonstrate that 3BP2 is recruited in cytoplasmic microclusters and nucleates a signaling complex that facilitates MHC:peptide-independent activation of signaling pathways downstream of the TCR. We show that induction of the adaptor molecule 3BP2 is a sensor of TCR signal strength and is critical for sustaining CD8+ T cell proliferation and regulating effector and memory differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation , Cells, Cultured , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/immunology , Signal Transduction
19.
Immunol Lett ; 109(2): 93-100, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17374402

ABSTRACT

Long-term humoral immunity elicited by pathogens and vaccines alike relies upon the generation of both memory B cells (B(mem)) and long-lived plasma cells (PCs). Virtually all vaccine formulations induce the concomitant emergence of both B(mem) and PCs, suggesting that the emergence of these two differentiated B cells subsets is commonly controlled. Evidence presented shows specific Toll-like receptor (TLR) agonists coupled with soluble protein antigen (sAg) can selectively induce the expansion of antigen specific B(mem) in the absence of PC generation. The co-administration of either TLR 3 or 9 agonists with sAg induced germinal centre (GC) formation, antigen-specific B(mem), but failed to substantively induce the generation of long-lived bone marrow (BM) PCs. Upon re-challenge, high levels of PCs were induced with concomitant high titres of antigen-specific serum IgG. Hence, vaccines can be developed that can prime and protect the host to subsequent infectious agents without initial, high levels of antibody production. Furthermore, these studies suggest that the signals that govern the expansion and differentiation of B(mem) can be uncoupled from those that induce long-lived BM PCs.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Toll-Like Receptors/agonists , Vaccines/immunology , Animals , Antigens/immunology , Epitopes, B-Lymphocyte/immunology , Freund's Adjuvant/immunology , Germinal Center/immunology , Lipids/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasma Cells/immunology
20.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27424808

ABSTRACT

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/enzymology , Isocitrate Dehydrogenase/genetics , Proto-Oncogene Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Down-Regulation , Hematopoietic Stem Cells/cytology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Proto-Oncogene Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL