Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Leuk Res ; 129: 107075, 2023 06.
Article in English | MEDLINE | ID: mdl-37079999

ABSTRACT

PURPOSE: Despite advances in the treatment of B-cell acute lymphoblastic leukemia (B-ALL), outcomes for relapsed/refractory (R/R) disease remain poor. Preclinical studies suggest that the combination of the CDK4/6 inhibitor palbociclib and dexamethasone may be effective in targeting leukemic cell growth. We conducted a phase I study of escalating doses of palbociclib in combination with dexamethasone in adults with R/R B-ALL. METHODS: Cycle 1 consisted of single agent palbociclib given for 7 days and continued for 28 additional days in combination with dexamethasone 20 mg daily. Palbociclib dosing began at 100 mg daily. Patients with a response were eligible for maintenance consisting of 1 week of palbociclib plus dexamethasone (20 mg daily × 2 days, 16 mg daily × 2 days, 12 mg daily × 2 days, 6 mg daily × 1 day), followed by 3 weeks of palbociclib alone. Safety, efficacy, and the expression of phospho-RB and c-MYB/BCL-2 were measured. CONCLUSIONS: Seven patients were treated on study before it was closed early due to slow accrual. No dose limiting toxicities were identified. One patient had a complete response with incomplete hematologic recovery, suggesting possible efficacy of the treatment. Reduction in CD34+ cells, p-RB, c-MYB, and BCL-2 expression also suggested on-target therapy effects.


Subject(s)
Lymphoma, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Humans , Lymphoma, B-Cell/drug therapy , Pyridines/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Dexamethasone , Antineoplastic Combined Chemotherapy Protocols/adverse effects
2.
Sci Rep ; 12(1): 14173, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35986080

ABSTRACT

To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.


Subject(s)
N-Acetylneuraminic Acid , Neuraminidase , Apoptosis , Cell Differentiation , Humans , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Neutrophils/metabolism
3.
Blood Cancer Discov ; 1(1): 48-67, 2020 07.
Article in English | MEDLINE | ID: mdl-32974613

ABSTRACT

Persistence of drug-resistant quiescent leukemic stem cells (LSC) and impaired natural killer (NK) cell immune response account for relapse of chronic myelogenous leukemia (CML). Inactivation of protein phosphatase 2A (PP2A) is essential for CML-quiescent LSC survival and NK cell antitumor activity. Here we show that MIR300 has antiproliferative and PP2A-activating functions that are dose dependently differentially induced by CCND2/CDK6 and SET inhibition, respectively. MIR300 is upregulated in CML LSCs and NK cells by bone marrow microenvironment (BMM) signals to induce quiescence and impair immune response, respectively. Conversely, BCR-ABL1 downregulates MIR300 in CML progenitors to prevent growth arrest and PP2A-mediated apoptosis. Quiescent LSCs escape apoptosis by upregulating TUG1 long noncoding RNA that uncouples and limits MIR300 function to cytostasis. Genetic and pharmacologic MIR300 modulation and/or PP2A-activating drug treatment restore NK cell activity, inhibit BMM-induced growth arrest, and selectively trigger LSC apoptosis in vitro and in patient-derived xenografts; hence, the importance of MIR300 and PP2A activity for CML development and therapy.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MicroRNAs , Humans , Killer Cells, Natural , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , MicroRNAs/genetics , Neoplastic Stem Cells , Protein Kinase Inhibitors/metabolism , Protein Phosphatase 2/genetics , Tumor Microenvironment/genetics
4.
Haematologica ; 103(12): 2016-2025, 2018 12.
Article in English | MEDLINE | ID: mdl-30049824

ABSTRACT

The fusion oncoprotein BCR-ABL1 exhibits aberrant tyrosine kinase activity and it has been proposed that it deregulates signaling networks involving both transcription factors and non-coding microRNAs that result in chronic myeloid leukemia (CML). Previously, microRNA expression profiling showed deregulated expression of miR-150 and miR-155 in CML. In this study, we placed these findings into the broader context of the MYC/miR-150/MYB/miR-155/PU.1 oncogenic network. We propose that up-regulated MYC and miR-155 in CD34+ leukemic stem and progenitor cells, in concert with BCR-ABL1, impair the molecular mechanisms of myeloid differentiation associated with low miR-150 and PU.1 levels. We revealed that MYC directly occupied the -11.7 kb and -0.35 kb regulatory regions in the MIR150 gene. MYC occupancy was markedly increased through BCR-ABL1 activity, causing inhibition of MIR150 gene expression in CML CD34+ and CD34- cells. Furthermore, we found an association between reduced miR-150 levels in CML blast cells and their resistance to tyrosine kinase inhibitors (TKIs). Although TKIs successfully disrupted BCR-ABL1 kinase activity in proliferating CML cells, this treatment did not efficiently target quiescent leukemic stem cells. The study presents new evidence regarding the MYC/miR-150/MYB/miR-155/PU.1 leukemic network established by aberrant BCR-ABL1 activity. The key connecting nodes of this network may serve as potential druggable targets to overcome resistance of CML stem and progenitor cells.


Subject(s)
Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Genes, myc/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MicroRNAs/genetics , Adult , Aged , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology
5.
Clin Cancer Res ; 24(1): 234-247, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29074603

ABSTRACT

Purpose:fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) is present in 30% of acute myeloid leukemia (AML), and these patients have short disease-free survival. FLT3 inhibitors have limited and transient clinical activity, and concurrent treatment with inhibitors of parallel or downstream signaling may improve responses. The oncogenic serine/threonine kinase Pim-1 is upregulated downstream of FLT3-ITD and also promotes its signaling in a positive feedback loop, suggesting benefit of combined Pim and FLT3 inhibition.Experimental Design: Combinations of clinically active Pim and FLT3 inhibitors were studied in vitro and in vivoResults: Concurrent treatment with the pan-Pim inhibitor AZD1208 and FLT3 inhibitors at clinically applicable concentrations abrogated in vitro growth of FLT3-ITD, but not wild-type FLT3 (FLT3-WT), cell lines. AZD1208 cotreatment increased FLT3 inhibitor-induced apoptosis of FLT3-ITD, but not FLT3-WT, cells measured by sub-G1 fraction, annexin V labeling, mitochondrial membrane potential, and PARP and caspase-3 cleavage. Concurrent treatment with AZD1208 and the FLT3 inhibitor quizartinib decreased growth of MV4-11 cells, with FLT3-ITD, in mouse xenografts, and prolonged survival, enhanced apoptosis of FLT3-ITD primary AML blasts, but not FLT3-WT blasts or remission marrow cells, and decreased FLT3-ITD AML blast colony formation. Mechanistically, AZD1208 and quizartinib cotreatment decreased expression of the antiapoptotic protein Mcl-1. Decrease in Mcl-1 protein expression was abrogated by treatment with the proteasome inhibitor MG132, and was preceded by downregulation of the Mcl-1 deubiquitinase USP9X, a novel mechanism of Mcl-1 regulation in AML.Conclusions: The data support clinical testing of Pim and FLT3 inhibitor combination therapy for FLT3-ITD AML. Clin Cancer Res; 24(1); 234-47. ©2017 AACR.


Subject(s)
Apoptosis/genetics , Gene Duplication , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Membrane Potential, Mitochondrial , Mice , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Proteolysis , Proteome/metabolism , Reactive Oxygen Species/metabolism
6.
Curr Drug Targets ; 18(4): 377-388, 2017.
Article in English | MEDLINE | ID: mdl-27307150

ABSTRACT

The use of imatinib, second and third generation ABL tyrosine kinase inhibitors (TKI) (i.e. dasatinib, nilotinib, bosutinib and ponatinib) made CML a clinically manageable and, in a small percentage of cases, a cured disease. TKI therapy also turned CML blastic transformation into a rare event; however, disease progression still occurs in those patients who are refractory, not compliant with TKI therapy or develop resistance to multiple TKIs. In the past few years, it became clear that the BCRABL1 oncogene does not operate alone to drive disease emergence, maintenance and progression. Indeed, it seems that bone marrow (BM) microenvironment-generated signals and cell autonomous BCRABL1 kinase-independent genetic and epigenetic alterations all contribute to: i. persistence of a quiescent leukemic stem cell (LSC) reservoir, ii. innate or acquired resistance to TKIs, and iii. progression into the fatal blast crisis stage. Herein, we review the intricate leukemic network in which aberrant, but finely tuned, survival, mitogenic and self-renewal signals are generated by leukemic progenitors, stromal cells, immune cells and metabolic microenvironmental conditions (e.g. hypoxia) to promote LSC maintenance and blastic transformation.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Disease Progression , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Protein Kinase Inhibitors/therapeutic use , Tumor Microenvironment/drug effects
7.
Oncotarget ; 7(30): 48280-48295, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27374090

ABSTRACT

Internal tandem duplication of fms-like tyrosine kinase-3 (FLT3-ITD) is frequent (30 percent) in acute myeloid leukemia (AML), and is associated with short disease-free survival following chemotherapy. The serine threonine kinase Pim-1 is a pro-survival oncogene transcriptionally upregulated by FLT3-ITD that also promotes its signaling in a positive feedback loop. Thus inhibiting Pim-1 represents an attractive approach in targeting FLT3-ITD cells. Indeed, co-treatment with the pan-Pim kinase inhibitor AZD1208 or expression of a kinase-dead Pim-1 mutant sensitized FLT3-ITD cell lines to apoptosis triggered by chemotherapy drugs including the topoisomerase 2 inhibitors daunorubicin, etoposide and mitoxantrone, but not the nucleoside analog cytarabine. AZD1208 sensitized primary AML cells with FLT3-ITD to topoisomerase 2 inhibitors, but did not sensitize AML cells with wild-type FLT3 or remission bone marrow cells, supporting a favorable therapeutic index. Mechanistically, the enhanced apoptosis observed with AZD1208 and topoisomerase 2 inhibitor combination treatment was associated with increased DNA double-strand breaks and increased levels of reactive oxygen species (ROS), and co-treatment with the ROS scavenger N-acetyl cysteine rescued FLT3-ITD cells from AZD1208 sensitization to topoisomerase 2 inhibitors. Our data support testing of Pim kinase inhibitors with topoisomerase 2 inhibitors, but not with cytarabine, to improve treatment outcomes in AML with FLT3-ITD.


Subject(s)
DNA Damage , Leukemia, Myeloid, Acute/drug therapy , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacology , Cytarabine/pharmacology , Drug Synergism , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Thiazolidines/administration & dosage , Thiazolidines/pharmacology , Topoisomerase II Inhibitors/administration & dosage
8.
J Immunol ; 194(4): 1832-40, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595786

ABSTRACT

Human NK cells are characterized by their ability to initiate an immediate and direct cytolytic response to virally infected or malignantly transformed cells. Within human peripheral blood, the more mature CD56(dim) NK cell efficiently kills malignant targets at rest, whereas the less mature CD56(bright) NK cells cannot. In this study, we show that resting CD56(bright) NK cells express significantly more phosphatase and tensin homolog deleted on chromosome 10 (PTEN) protein when compared with CD56(dim) NK cells. Consistent with this, forced overexpression of PTEN in NK cells resulted in decreased cytolytic activity, and loss of PTEN in CD56(bright) NK cells resulted in elevated cytolytic activity. Comparable studies in mice showed PTEN overexpression did not alter NK cell development or NK cell-activating and inhibitory receptor expression yet, as in humans, did decrease expression of downstream NK activation targets MAPK and AKT during early cytolysis of tumor target cells. Confocal microscopy revealed that PTEN overexpression disrupts the NK cell's ability to organize immunological synapse components including decreases in actin accumulation, polarization of the microtubule organizing center, and the convergence of cytolytic granules. In summary, our data suggest that PTEN normally works to limit the NK cell's PI3K/AKT and MAPK pathway activation and the consequent mobilization of cytolytic mediators toward the target cell and suggest that PTEN is among the active regulatory components prior to human NK cells transitioning from the noncytolytic CD56(bright) NK cell to the cytolytic CD56(dim) NK cells.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , PTEN Phosphohydrolase/immunology , Animals , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , PTEN Phosphohydrolase/metabolism , Real-Time Polymerase Chain Reaction
9.
Cell Rep ; 8(1): 150-62, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24953655

ABSTRACT

Accumulating evidence indicates that human natural killer (NK) cells develop in secondary lymphoid tissue (SLT) through a so-called "stage 3" developmental intermediate minimally characterized by a CD34(-)CD117(+)CD94(-) immunophenotype that lacks mature NK cell function. This stage 3 population is heterogeneous, potentially composed of functionally distinct innate lymphoid cell (ILC) types that include interleukin-1 receptor (IL-1R1)-positive, IL-22-producing ILC3s. Whether human ILC3s are developmentally related to NK cells is a subject of ongoing investigation. Here, we show that antagonism of the aryl hydrocarbon receptor (AHR) or silencing of AHR gene expression promotes the differentiation of tonsillar IL-22-producing IL-1R1(hi) human ILC3s to CD56(bright)CD94(+) interferon (IFN)-γ-producing cytolytic mature NK cells expressing eomesodermin (EOMES) and T-Box Protein 21 (TBX21 or TBET). Hence, we demonstrate the lineage plasticity of human ILCs by identifying AHR as a transcription factor that prevents IL-1R1(hi) ILC3s from differentiating into NK cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Killer Cells, Natural/immunology , Lymphocytes/immunology , Receptors, Aryl Hydrocarbon/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , CD56 Antigen/genetics , CD56 Antigen/metabolism , Cell Lineage , Cells, Cultured , Humans , Interleukins/genetics , Interleukins/metabolism , Killer Cells, Natural/cytology , Lymphocytes/cytology , Palatine Tonsil/cytology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Interleukin-22
10.
Eur J Immunol ; 43(10): 2750-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23722894

ABSTRACT

Activation of the fibromyalgia syndrome-like tyrosine kinase 3 (FLT3) by its ligand, FLT3 ligand (FL), strongly augments the development of natural killer (NK) cells from human CD34⁺ hematopoietic progenitor cells (HPCs) in the presence of IL-15, compared with NK-cell development in the presence of IL-15 alone. In this study, we observed that blocking the receptor tyrosine kinase Axl/Gas6 pathway with a soluble Axl-IgG1 Fc fusion protein (Axl-Fc) in the presence of FL significantly diminished the absolute number of CD3⁻ CD56⁺ NK cells derived from human CD34⁺ HPCs. Axl-Fc reduced the expression levels of the IL-2/15 receptor ß chain (CD122) and γ chain (CD132) induced by activation of FLT3 and consequently reduced the frequency of NK precursor cells responding to IL-15. Furthermore, Axl-Fc diminished FL-induced FLT3 phosphorylation and impeded the physical interaction between Axl and FLT3 in CD34⁺ HPCs. Collectively, our data suggest that the Axl/Gas6 pathway contributes to normal human NK-cell development at least in part via its positive regulatory effect on FLT3 signaling in CD34⁺ HPCs.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunoglobulin Fc Fragments/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Antigens, CD34/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Hematopoietic Stem Cells/drug effects , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-15/immunology , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Axl Receptor Tyrosine Kinase
11.
Blood ; 121(16): 3126-34, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23422749

ABSTRACT

It is known that microRNAs (miRs) are involved in lymphocyte development, homeostasis, activation, and occasionally malignant transformation. In this study, a miR-155 transgene (tg) was driven to be overexpressed off of the lck promoter in order to assess its effects on natural killer (NK) cell biology in vivo. miR-155 tg mice have an increase in NK-cell number with an excess of the CD11b(low)CD27(high) NK subset, indicative of a halt in terminal NK-cell differentiation that proved to be intrinsic to the cell itself. The increase in NK cells results, in part, from improved survival in medium alone and enhanced expansion with endogenous or exogenous interleukin 15. Phenotypic and functional data from miR-155 tg NK cells showed constitutive activation and enhanced target cell conjugation, resulting in more potent antitumor activity in vitro and improved survival of lymphoma-bearing mice in vivo when compared with wild type NK cells. The enhanced NK-cell survival, expansion, activation, and tumor control that result from overexpression of miR-155 in NK cells could be explained, in part, via diminished expression of the inositol phosphatase SHIP1 and increased activation of ERK and AKT kinases. Thus, the regulation of miR-155 is important for NK-cell development, homeostasis, and activation.


Subject(s)
Killer Cells, Natural/immunology , Lymphoma/immunology , MicroRNAs/genetics , Up-Regulation , Animals , Cell Count , Cell Differentiation , Cell Line, Tumor , Cell Survival , Cells, Cultured , Down-Regulation , Inositol Polyphosphate 5-Phosphatases , Interferon-gamma/immunology , Interleukin-15/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphoma/genetics , Lymphoma/pathology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , MicroRNAs/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/immunology , Transgenes
12.
J Immunol ; 190(6): 2702-11, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418626

ABSTRACT

CD20 is a widely validated, B cell-specific target for therapy in B cell malignancies. Rituximab is an anti-CD20 Ab that prolongs survival of chronic lymphocytic leukemia (CLL) patients when combined with chemotherapy. Ofatumumab and GA101 (obinutuzumab) are CD20-directed Abs currently being developed as alternative agents to rituximab in CLL based upon different properties of enhanced direct cell death, NK cell-mediated Ab-dependent cellular cytotoxicity, or complement-dependent cytotoxicity. Despite widespread study, ofatumumab and GA101 have not been compared with each other, nor studied for their interactions with monocytes and macrophages which are critical for the efficacy of anti-CD20 Abs in murine models. In CLL cells, we show that direct cell death and complement-dependent cytotoxicity are greatest with GA101 and ofatumumab, respectively. GA101 promotes enhanced NK cell activation and Ab-dependent cellular cytotoxicity at high Ab concentrations. Ofatumumab elicits superior Ab-dependent cellular phagocytosis with monocyte-derived macrophages. GA101 demonstrated reduced activation of monocytes with diminished pERK, TNF-α release, and FcγRIIa recruitment to lipid rafts. These data demonstrate that GA101 and ofatumumab are both superior to rituximab against CLL cells via different mechanisms of potential tumor elimination. These findings bear relevance to potential combination strategies with each of these anti-CD20 Abs in the treatment of CLL.


Subject(s)
Antibodies, Neoplasm/toxicity , Antigens, CD20/immunology , Drug Delivery Systems/methods , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Macrophages/immunology , Monocytes/immunology , Antibodies, Neoplasm/therapeutic use , Antigens, CD20/metabolism , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Macrophages/metabolism , Macrophages/pathology , Monocytes/metabolism , Monocytes/pathology , Tumor Cells, Cultured
13.
Blood ; 119(15): 3478-85, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22378844

ABSTRACT

MicroRNAs (miRs) are small, noncoding RNA molecules with important regulatory functions whose role in regulating natural killer (NK) cell biology is not well defined. Here, we show that miR-155 is synergistically induced in primary human NK cells after costimulation with IL-12 and IL-18, or with IL-12 and CD16 clustering. Over-expression of miR-155 enhanced induction of IFN-γ by IL-12 and IL-18 or CD16 stimulation, whereas knockdown of miR-155 or its disruption suppressed IFN-γ induction in monokine and/or CD16-stimulated NK cells. These effects on the regulation of NK cell IFN-γ expression were found to be mediated at least in part via miR-155's direct effects on the inositol phosphatase SHIP1. Consistent with this, we observed that modulation of miR-155 overrides IL-12 and IL-18-mediated regulation of SHIP1 expression in NK cells. Collectively, our data indicate that miR-155 expression is regulated by stimuli that strongly induce IFN-γ in NK cells such as IL-12, IL-18, and CD16 activation, and that miR-155 functions as a positive regulator of IFN-γ production in human NK cells, at least in part via down-regulating SHIP1. These findings may have clinical relevance for targeting miR-155 in neoplastic disease.


Subject(s)
Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , MicroRNAs/physiology , Animals , Cells, Cultured , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/physiology , Gene Expression Regulation , HEK293 Cells , Humans , Inositol Polyphosphate 5-Phosphatases , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Interleukin-12/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, IgG/physiology
14.
J Clin Invest ; 121(4): 1456-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21364281

ABSTRACT

IL-15 may have a role in the development of T cell large granular lymphocyte (T-LGL) or NKT leukemias. However, the mechanisms of action and the identity of the cell subset that undergoes leukemic transformation remain elusive. Here we show that in both mice and humans, NKp46 expression marks a minute population of WT NKT cells with higher activity and potency to become leukemic. Virtually 100% of T-LGL leukemias in IL-15 transgenic mice expressed NKp46, as did a majority of human T-LGL leukemias. The minute NKp46+ NKT population, but not the NKp46⁻ NKT population, was selectively expanded by overexpression of endogenous IL-15. Importantly, IL-15 transgenic NKp46⁻ NKT cells did not become NKp46+ in vivo, suggesting that NKp46+ T-LGL leukemia cells were the malignant counterpart of the minute WT NKp46+ NKT population. Mechanistically, NKp46+ NKT cells possessed higher responsiveness to IL-15 in vitro and in vivo compared with that of their NKp46⁻ NKT counterparts. Furthermore, interruption of IL-15 signaling using a neutralizing antibody could prevent LGL leukemia in IL-15 transgenic mice. Collectively, our data demonstrate that NKp46 identifies a functionally distinct NKT subset in mice and humans that appears to be directly susceptible to leukemic transformation when IL-15 is overexpressed. Thus, IL-15 signaling and NKp46 may be useful targets in the treatment of patients with T-LGL or NKT leukemia.


Subject(s)
Antigens, Ly/metabolism , Cell Transformation, Neoplastic/immunology , Leukemia, Large Granular Lymphocytic/etiology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Ly/genetics , Cell Transformation, Neoplastic/genetics , Humans , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-2 Receptor beta Subunit/antagonists & inhibitors , Leukemia, Large Granular Lymphocytic/genetics , Leukemia, Large Granular Lymphocytic/immunology , Leukemia, Large Granular Lymphocytic/therapy , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Killer T-Cells/classification , Signal Transduction/immunology , T-Lymphocyte Subsets/classification
15.
Blood ; 117(8): 2378-84, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21156847

ABSTRACT

The ability of natural killer (NK) cells to kill malignant or infected cells depends on the integration of signals from different families of cell surface receptors, including cytokine receptors. How such signals then regulate NK-cell cytotoxicity is incompletely understood. Here we analyzed an endogenous inhibitor of protein phosphatase 2A (PP2A) activity called SET, and its role in regulating human NK-cell cytotoxicity and its mechanism of action in human NK cells. RNAi-mediated suppression of SET down-modulates NK-cell cytotoxicity, whereas ectopic overexpression of SET enhances cytotoxicity. SET knockdown inhibits both mRNA and protein granzyme B expression, as well as perforin expression, whereas SET overexpression enhances granzyme B expression. Treatment of NK cells with the PP2A activator 1,9-dideoxy-forskolin also inhibits both granzyme B expression and cytotoxicity. In addition, pretreatment with the PP2A inhibitor okadaic acid rescues declining granzyme B mRNA levels in SET knockdown cells. Down-modulation of SET expression or activation of PP2A also decreases human NK-cell antibody-dependent cellular cytotoxicity. Finally, the induction of granzyme B gene expression by interleukin-2 and interleukin-15 is inhibited by SET knockdown. These data provide evidence that granzyme B gene expression and therefore human NK-cell cytotoxicity can be regulated by the PP2A-SET interplay.


Subject(s)
Granzymes/genetics , Histone Chaperones/physiology , Killer Cells, Natural/metabolism , Protein Phosphatase 2/physiology , Transcription Factors/physiology , Cytotoxicity, Immunologic , DNA-Binding Proteins , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Granzymes/biosynthesis , Humans , Killer Cells, Natural/immunology , Protein Phosphatase 2/antagonists & inhibitors , RNA, Small Interfering/pharmacology
16.
Blood ; 115(6): 1204-13, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19965644

ABSTRACT

CD19 is a B cell-specific antigen expressed on chronic lymphocytic leukemia (CLL) cells but to date has not been effectively targeted with therapeutic monoclonal antibodies. XmAb5574 is a novel engineered anti-CD19 monoclonal antibody with a modified constant fragment (Fc)-domain designed to enhance binding of FcgammaRIIIa. Herein, we demonstrate that XmAb5574 mediates potent antibody-dependent cellular cytotoxicity (ADCC), modest direct cytotoxicity, and antibody-dependent cellular phagocytosis but not complement-mediated cytotoxicity against CLL cells. Interestingly, XmAb5574 mediates significantly higher ADCC compared with both the humanized anti-CD19 nonengineered antibody it is derived from and also rituximab, a therapeutic antibody widely used in the treatment of CLL. The XmAb5574-dependent ADCC is mediated by natural killer (NK) cells through a granzyme B-dependent mechanism. The NK cell-mediated cytolytic and secretory function with XmAb5574 compared with the nonengineered antibody is associated with enhanced NK-cell activation, interferon production, extracellular signal-regulated kinase phosphorylation downstream of Fcgamma receptor, and no increased NK-cell apoptosis. Notably, enhanced NK cell-mediated ADCC with XmAb5574 was enhanced further by lenalidomide. These findings provide strong support for further clinical development of XmAb5574 as both a monotherapy and in combination with lenalidomide for the therapy of CLL and related CD19(+) B-cell malignancies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/physiology , Antigens, CD19/immunology , Immunoglobulin Fc Fragments/genetics , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Apoptosis , Blotting, Western , Cytotoxicity, Immunologic/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Granzymes/metabolism , Humans , Immunoglobulin Fc Fragments/immunology , Leukemia, B-Cell/genetics , Leukemia, B-Cell/immunology , Leukemia, B-Cell/therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Phagocytosis , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Blood ; 115(2): 274-81, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19897577

ABSTRACT

Human CD56(bright) natural killer (NK) cells possess little or no killer immunoglobulin-like receptors (KIRs), high interferon-gamma (IFN-gamma) production, but little cytotoxicity. CD56(dim) NK cells have high KIR expression, produce little IFN-gamma, yet display high cytotoxicity. We hypothesized that, if human NK maturation progresses from a CD56(bright) to a CD56(dim) phenotype, an intermediary NK cell must exist, which demonstrates more functional overlap than these 2 subsets, and we used CD94 expression to test our hypothesis. CD94(high)CD56(dim) NK cells express CD62L, CD2, and KIR at levels between CD56(bright) and CD94(low)CD56(dim) NK cells. CD94(high)CD56(dim) NK cells produce less monokine-induced IFN-gamma than CD56(bright) NK cells but much more than CD94(low)CD56(dim) NK cells because of differential interleukin-12-mediated STAT4 phosphorylation. CD94(high)CD56(dim) NK cells possess a higher level of granzyme B and perforin expression and CD94-mediated redirected killing than CD56(bright) NK cells but lower than CD94(low)CD56(dim) NK cells. Collectively, our data suggest that the density of CD94 surface expression on CD56(dim) NK cells identifies a functional and likely developmental intermediary between CD56(bright) and CD94(low)CD56(dim) NK cells. This supports the notion that, in vivo, human CD56(bright) NK cells progress through a continuum of differentiation that ends with a CD94(low)CD56(dim) phenotype.


Subject(s)
CD56 Antigen/immunology , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Cells, Cultured , Humans , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/cytology , L-Selectin/immunology , Lymphocyte Subsets/cytology , Phosphorylation/immunology , STAT4 Transcription Factor/immunology
18.
J Immunol ; 183(8): 4968-74, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19801519

ABSTRACT

Understanding of heterogeneous NK subsets is important for the study of NK cell biology and development, and for the application of NK cell-based therapies in the treatment of disease. Here we demonstrate that the surface expression of CD94 can distinctively divide mouse NK cells into two approximately even CD94(low) and CD94(high) subsets in all tested organs and tissues. The CD94(high) NK subset has significantly greater capacity to proliferate, produce IFN-gamma, and lyse target cells than does the CD94(low) subset. The CD94(high) subset has exclusive expression of NKG2A/C/E, higher expression of CD117 and CD69, and lower expression of Ly49D (activating) and Ly49G2 (inhibitory). In vivo, purified mouse CD94(low) NK cells become CD94(high) NK cells, but not vice versa. Collectively, our data suggest that CD94 is an Ag that can be used to identify functionally distinct NK cell subsets in mice and could also be relevant to late-stage mouse NK cell development.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , NK Cell Lectin-Like Receptor Subfamily D/immunology , Adoptive Transfer , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Female , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lectins, C-Type , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Proto-Oncogene Proteins c-kit/immunology
19.
Blood ; 114(7): 1374-82, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19520806

ABSTRACT

We showed that Emicro-MiR-155 transgenic mice develop acute lymphoblastic leukemia/high-grade lymphoma. Most of these leukemias start at approximately 9 months irrespective of the mouse strain. They are preceded by a polyclonal pre-B-cell proliferation, have variable clinical presentation, are transplantable, and develop oligo/monoclonal expansion. In this study, we show that in these transgenic mice the B-cell precursors have the highest MiR-155 transgene expression and are at the origin of the leukemias. We determine that Src homology 2 domain-containing inositol-5-phosphatase (SHIP) and CCAAT enhancer-binding protein beta (C/EBPbeta), 2 important regulators of the interleukin-6 signaling pathway, are direct targets of MiR-155 and become gradually more down-regulated in the leukemic than in the preleukemic mice. We hypothesize that miR-155, by down-modulating Ship and C/EBPbeta, initiates a chain of events that leads to the accumulation of large pre-B cells and acute lymphoblastic leukemia/high-grade lymphoma.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cell Transformation, Neoplastic/metabolism , MicroRNAs/biosynthesis , Phosphoric Monoester Hydrolases/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Gene Expression Regulation, Leukemic/genetics , Inositol Polyphosphate 5-Phosphatases , Interleukin-6/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Mice , Mice, Transgenic , MicroRNAs/genetics , Phosphoric Monoester Hydrolases/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/genetics
20.
Blood ; 113(22): 5558-67, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19329776

ABSTRACT

Aberrant methylation of tumor suppressor genes can lead to their silencing in many cancers. TSC-22 is a gene silenced in several solid tumors, but its function and the mechanism(s) responsible for its silencing are largely unknown. Here we demonstrate that the TSC-22 promoter is methylated in primary mouse T or natural killer (NK) large granular lymphocyte (LGL) leukemia and this is associated with down-regulation or silencing of TSC-22 expression. The TSC-22 deregulation was reversed in vivo by a 5-aza-2'-deoxycytidine therapy of T or NK LGL leukemia, which significantly increased survival of the mice bearing this disease. Ectopic expression of TSC-22 in mouse leukemia or lymphoma cell lines resulted in delayed in vivo tumor formation. Targeted disruption of TSC-22 in wild-type mice enhanced proliferation and in vivo repopulation efficiency of hematopoietic precursor cells (HPCs). Collectively, our data suggest that TSC-22 normally contributes to the regulation of HPC function and is a putative tumor suppressor gene that is hypermethylated and silenced in T or NK LGL leukemia.


Subject(s)
Cell Movement/genetics , Cell Proliferation , Hematopoietic Stem Cells/physiology , Leukemia, Large Granular Lymphocytic/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Cells, Cultured , DNA Methylation , Epigenesis, Genetic/physiology , Gene Expression Regulation, Leukemic , Gene Silencing/physiology , Genes, Tumor Suppressor , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...