Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
EBioMedicine ; 71: 103559, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34461601

ABSTRACT

BACKGROUND: The most common B-cell cancers, chronic lymphocytic leukemia/lymphoma (CLL), follicular and diffuse large B-cell (FL, DLBCL) lymphomas, have distinct clinical courses, yet overlapping "cell-of-origin". Dynamic changes to the epigenome are essential regulators of B-cell differentiation. Therefore, we reasoned that these distinct cancers may be driven by shared mechanisms of disruption in transcriptional circuitry. METHODS: We compared purified malignant B-cells from 52 patients with normal B-cell subsets (germinal center centrocytes and centroblasts, naïve and memory B-cells) from 36 donor tonsils using >325 high-resolution molecular profiling assays for histone modifications, open chromatin (ChIP-, FAIRE-seq), transcriptome (RNA-seq), transcription factor (TF) binding, and genome copy number (microarrays). FINDINGS: From the resulting data, we identified gains in active chromatin in enhancers/super-enhancers that likely promote unchecked B-cell receptor signaling, including one we validated near the immunoglobulin superfamily receptors FCMR and PIGR. More striking and pervasive was the profound loss of key B-cell identity TFs, tumor suppressors and their super-enhancers, including EBF1, OCT2(POU2F2), and RUNX3. Using a novel approach to identify transcriptional feedback, we showed that these core transcriptional circuitries are self-regulating. Their selective gain and loss form a complex, iterative, and interactive process that likely curbs B-cell maturation and spurs proliferation. INTERPRETATION: Our study is the first to map the transcriptional circuitry of the most common blood cancers. We demonstrate that a critical subset of B-cell TFs and their cognate enhancers form self-regulatory transcriptional feedback loops whose disruption is a shared mechanism underlying these diverse subtypes of B-cell lymphoma. FUNDING: National Institute of Health, Siteman Cancer Center, Barnes-Jewish Hospital Foundation, Doris Duke Foundation.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Leukemia, B-Cell/etiology , Lymphoma, B-Cell/etiology , Transcription, Genetic , Adult , Aged , Aged, 80 and over , B-Lymphocytes/immunology , Biomarkers , Cell Transformation, Neoplastic/metabolism , Chromatin Immunoprecipitation Sequencing , Computational Biology/methods , DNA Copy Number Variations , Enhancer Elements, Genetic , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Immunophenotyping , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/metabolism , Male , Middle Aged , Models, Biological , Oncogenes , Signal Transduction , Transcription Factors/metabolism
2.
Front Immunol ; 11: 581119, 2020.
Article in English | MEDLINE | ID: mdl-33240268

ABSTRACT

Congenital defects of the immune system called primary immunodeficiency disorders (PID) describe a group of diseases characterized by a decrease, an absence, or a malfunction of at least one part of the immune system. As a result, PID patients are more prone to develop life-threatening complications, including cancer. PID currently include over 400 different disorders, however, the variety of PID-related cancers is narrow. We discuss here reasons for this clinical phenotype. Namely, PID can lead to cell intrinsic failure to control cell transformation, failure to activate tumor surveillance by cytotoxic cells or both. As the most frequent tumors seen among PID patients stem from faulty lymphocyte development leading to leukemia and lymphoma, we focus on the extensive genomic alterations needed to create the vast diversity of B and T lymphocytes with potential to recognize any pathogen and why defects in these processes lead to malignancies in the immunodeficient environment of PID patients. In the second part of the review, we discuss PID affecting tumor surveillance and especially membrane trafficking defects caused by altered exocytosis and regulation of the actin cytoskeleton. As an impairment of these membrane trafficking pathways often results in dysfunctional effector immune cells, tumor cell immune evasion is elevated in PID. By considering new anti-cancer treatment concepts, such as transfer of genetically engineered immune cells, restoration of anti-tumor immunity in PID patients could be an approach to complement standard therapies.


Subject(s)
Leukemia, B-Cell/etiology , Lymphoma, B-Cell/etiology , Primary Immunodeficiency Diseases/complications , Actin Cytoskeleton/genetics , Actin Cytoskeleton/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , DNA Repair/genetics , DNA Repair/immunology , Exocytosis/genetics , Exocytosis/immunology , Genomic Instability , Humans , Immunological Synapses/genetics , Leukemia, B-Cell/genetics , Leukemia, B-Cell/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Models, Immunological , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Risk Factors , Tumor Escape/genetics
3.
Mol Cancer Ther ; 19(12): 2432-2444, 2020 12.
Article in English | MEDLINE | ID: mdl-33051362

ABSTRACT

The IRE-1 kinase/RNase splices the mRNA of the XBP-1 gene, resulting in the spliced XBP-1 (XBP-1s) mRNA that encodes the functional XBP-1s transcription factor that is critically important for the growth and survival of B-cell leukemia, lymphoma, and multiple myeloma (MM). Several inhibitors targeting the expression of XBP-1s have been reported; however, the cytotoxicity exerted by each inhibitor against cancer cells is highly variable. To design better therapeutic strategies for B-cell cancer, we systematically compared the ability of these compounds to inhibit the RNase activity of IRE-1 in vitro and to suppress the expression of XBP-1s in mouse and human MM cell lines. Tricyclic chromenone-based inhibitors B-I09 and D-F07, prodrugs harboring an aldehyde-masking group, emerged as the most reliable inhibitors for potent suppression of XBP-1s expression in MM cells. The cytotoxicity of B-I09 and D-F07 against MM as well as chronic lymphocytic leukemia and mantle cell lymphoma could be further enhanced by combination with inhibitors of the PI3K/AKT pathway. Because chemical modifications of the salicylaldehyde hydroxy group could be used to tune 1,3-dioxane prodrug stability, we installed reactive oxygen species-sensitive structural cage groups onto these inhibitors to achieve stimuli-responsive activities and improve tumor-targeting efficiency.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Development , Endoribonucleases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Development/methods , Drug Screening Assays, Antitumor/methods , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/metabolism , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Mice , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism
4.
Front Immunol ; 11: 1538, 2020.
Article in English | MEDLINE | ID: mdl-32793211

ABSTRACT

B-cell malignancies are a heterogeneous group of hematological neoplasms derived from cells at different stages of B-cell development. Recent studies revealed that dysregulated redox metabolism is one of the factors contributing to the pathogenesis and progression of B-cell malignancies. Elevated levels of oxidative stress markers usually correlate with the advanced stage of various B-cell malignancies. In the complex tumor microenvironment, reactive oxygen species affect not only malignant cells but also bystander cells, including immune cells. Importantly, malignant cells, due to genetic dysregulation, are able to adapt to the increased demands for energy and reducing equivalents via metabolic reprogramming and upregulation of antioxidants. The immune cells, however, are more sensitive to oxidative imbalance. This may cause their dysfunction, leading to immune evasion and tumor progression. On the other hand, the already imbalanced redox homeostasis renders malignant B-cells particularly sensitive to further elevation of reactive oxygen species. Indeed, targeting antioxidant systems has already presented anti-leukemic efficacy in preclinical models. Moreover, the prooxidant treatment that triggers immunogenic cell death has been utilized to generate autologous anti-leukemic vaccines. In this article, we review novel research on the dual role of the reactive oxygen species in B-cell malignancies. We highlight the mechanisms of maintaining redox homeostasis by malignant B-cells along with the antioxidant shield provided by the microenvironment. We summarize current findings regarding therapeutic targeting of redox metabolism in B-cell malignancies. We also discuss how the oxidative stress affects antitumor immune response and how excessive reactive oxygens species influence anticancer prooxidant treatments and immunotherapies.


Subject(s)
Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Reactive Oxygen Species/metabolism , Animals , Disease Susceptibility , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Humans , Immunomodulation , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Oxidation-Reduction , Oxidative Stress , Signal Transduction , Stromal Cells/metabolism , Tumor Microenvironment
5.
Int J Mol Sci ; 21(6)2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32210016

ABSTRACT

Lineage commitment and differentiation of hematopoietic cells takes place in well-defined microenvironmental surroundings. Communication with other cell types is a vital prerequisite for the normal functions of the immune system, while disturbances in this communication support the development and progression of neoplastic disease. Integrins such as the integrin very late antigen-4 (VLA-4; CD49d/CD29) control the localization of healthy as well as malignant B cells within the tissue, and thus determine the patterns of organ infiltration. Malignant B cells retain some key characteristics of their normal counterparts, with B cell receptor (BCR) signaling and integrin-mediated adhesion being essential mediators of tumor cell homing, survival and proliferation. It is thus not surprising that targeting the BCR pathway using small molecule inhibitors has proved highly effective in the treatment of B cell malignancies. Attenuation of BCR-dependent lymphoma-microenvironment interactions was, in this regard, described as a main mechanism critically contributing to the efficacy of these agents. Here, we review the contribution of VLA-4 to normal B cell differentiation on the one hand, and to the pathophysiology of B cell malignancies on the other hand. We describe its impact as a prognostic marker, its interplay with BCR signaling and its predictive role for novel BCR-targeting therapies, in chronic lymphocytic leukemia and beyond.


Subject(s)
Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Cell Differentiation/genetics , Cellular Microenvironment/genetics , Disease Management , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Hematopoiesis/genetics , Humans , Integrins/genetics , Integrins/metabolism , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Molecular Targeted Therapy , Signal Transduction
6.
Clin Cancer Res ; 26(13): 3307-3318, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32220889

ABSTRACT

PURPOSE: Children with Down syndrome (constitutive trisomy 21) that develop acute lymphoblastic leukemia (DS-ALL) have a 3-fold increased likelihood of treatment-related mortality coupled with a higher cumulative incidence of relapse, compared with other children with B-cell acute lymphoblastic leukemia (B-ALL). This highlights the lack of suitable treatment for Down syndrome children with B-ALL. EXPERIMENTAL DESIGN: To facilitate the translation of new therapeutic agents into clinical trials, we built the first preclinical cohort of patient-derived xenograft (PDX) models of DS-ALL, comprehensively characterized at the genetic and transcriptomic levels, and have proven its suitability for preclinical studies by assessing the efficacy of drug combination between the MEK inhibitor trametinib and conventional chemotherapy agents. RESULTS: Whole-exome and RNA-sequencing experiments revealed a high incidence of somatic alterations leading to RAS/MAPK pathway activation in our cohort of DS-ALL, as well as in other pediatric B-ALL presenting somatic gain of the chromosome 21 (B-ALL+21). In murine and human B-cell precursors, activated KRASG12D functionally cooperates with trisomy 21 to deregulate transcriptional networks that promote increased proliferation and self renewal, as well as B-cell differentiation blockade. Moreover, we revealed that inhibition of RAS/MAPK pathway activation using the MEK1/2 inhibitor trametinib decreased leukemia burden in several PDX models of B-ALL+21, and enhanced survival of DS-ALL PDX in combination with conventional chemotherapy agents such as vincristine. CONCLUSIONS: Altogether, using novel and suitable PDX models, this study indicates that RAS/MAPK pathway inhibition represents a promising strategy to improve the outcome of Down syndrome children with B-cell precursor leukemia.


Subject(s)
Down Syndrome/complications , Down Syndrome/genetics , Down Syndrome/metabolism , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/etiology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Computational Biology/methods , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Humans , Immunophenotyping , Leukemia, B-Cell/therapy , Mice , Mice, Transgenic , Oncogenes , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects
7.
Immunol Rev ; 295(1): 39-53, 2020 05.
Article in English | MEDLINE | ID: mdl-32185805

ABSTRACT

In response to mitogenic stimulation, B cells activate different pro-anabolic signaling pathways such as c-Myc- and mTORC1-dependent networks to satisfy the energetic demands of biomass synthesis and proliferation. In order to preserve viability and function, cell growth cannot progress unchecked and must be adjusted according to the availability of nutrients. Nutrient-sensing proteins such as AMPK antagonize mTORC1 activity in response to starvation. If pro-anabolic signaling pathways are aberrantly activated, B cells may lack the metabolic capacity to accommodate their energetic needs, which can lead to cell death. On the other hand, metabolic hyperactivation is a salient feature of cancer cells, suggesting that mechanisms exist, which allow B cells to cope with metabolic stress. The aim of this review is to discuss how B cells respond to a mismatch between energy supply and demand and what the consequences are of metabolic dysregulation in normal and malignant B cells.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic , Energy Metabolism , Immune Checkpoint Proteins/metabolism , Lymphocyte Activation/immunology , Animals , B-Lymphocytes/cytology , Biomarkers , Cell Differentiation/immunology , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Cellular Senescence/immunology , Germinal Center/immunology , Germinal Center/metabolism , Homeostasis , Humans , Immune Checkpoint Proteins/genetics , Immunologic Memory , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Plasma Cells/immunology , Plasma Cells/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism
8.
EBioMedicine ; 52: 102625, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31981978

ABSTRACT

BACKGROUND: DuoBody®-CD3xCD20 (GEN3013) is a full-length human IgG1 bispecific antibody (bsAb) recognizing CD3 and CD20, generated by controlled Fab-arm exchange. Its Fc domain was silenced by introduction of mutations L234F L235E D265A. METHODS: T-cell activation and T-cell-mediated cytotoxicity were measured by flow cytometry following co-culture with tumour cells. Anti-tumour activity of DuoBody-CD3xCD20 was assessed in humanized mouse models in vivo. Non-clinical safety studies were performed in cynomolgus monkeys. FINDINGS: DuoBody-CD3xCD20 induced highly potent T-cell activation and T-cell-mediated cytotoxicity towards malignant B cells in vitro. Comparison of DuoBody-CD3xCD20 to CD3 bsAb targeting alternative B-cell antigens, or to CD3xCD20 bsAb generated using alternative CD20 Ab, emphasized its exceptional potency. In vitro comparison with other CD3xCD20 bsAb in clinical development showed that DuoBody-CD3xCD20 was significantly more potent than three other bsAb with single CD3 and CD20 binding regions and equally potent as a bsAb with a single CD3 and two CD20 binding regions. DuoBody-CD3xCD20 showed promising anti-tumour activity in vivo, also in the presence of excess levels of a CD20 Ab that competes for binding. In cynomolgus monkeys, DuoBody-CD3xCD20 demonstrated profound and long-lasting B-cell depletion from peripheral blood and lymphoid organs, which was comparable after subcutaneous and intravenous administration. Peak plasma levels of DuoBody-CD3xCD20 were lower and delayed after subcutaneous administration, which was associated with a reduction in plasma cytokine levels compared to intravenous administration, while bioavailability was comparable. INTERPRETATION: Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of subcutaneous DuoBody-CD3xCD20 in patients with B-cell malignancies. FUNDING: Genmab.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, CD20/metabolism , CD3 Complex/metabolism , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Antibody Specificity/immunology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Macaca fascicularis , Mice , Mutation , Recombinant Proteins , Xenograft Model Antitumor Assays
9.
Leukemia ; 33(12): 2767-2778, 2019 12.
Article in English | MEDLINE | ID: mdl-31690821

ABSTRACT

The approval of tisagenlecleucel and axicabtagene ciloleucel represents a breakthrough in the field of immune and cellular therapy for hematologic malignancies. These anti-CD19 chimeric antigen receptor-T cells (CAR) proved to be highly effective in the treatment of relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) and specific histologic subtypes of B-cell non-Hodgkin lymphomas. This expert review aims to summarize the current available research evidence in this field, with a special focus on the different challenges faced by treating physicians, and we also provide future perspectives.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antigens, Neoplasm/immunology , Biomarkers, Tumor , Clinical Trials as Topic , Genetic Engineering , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/etiology , Leukemia, B-Cell/mortality , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/mortality , Receptors, Chimeric Antigen/genetics , Treatment Outcome
10.
Cancer J ; 25(6): 386-393, 2019.
Article in English | MEDLINE | ID: mdl-31764119

ABSTRACT

Bruton tyrosine kinase (BTK) is a nonreceptor tyrosine kinase that plays a central role in the signal transduction of the B-cell antigen receptor and other cell surface receptors, both in normal and malignant B lymphocytes. B-cell antigen receptor signaling is activated in secondary lymphatic organs and drives the proliferation of malignant B cells, including chronic lymphocytic leukemia (CLL) cells. During the last 10 years, BTK inhibitors (BTKis) are increasingly replacing chemotherapy-based regimen, especially in patients with CLL and mantle cell lymphoma (MCL). Bruton tyrosine kinase inhibitors are particularly active in patients with CLL and MCL, but also received approval for Waldenström macroglobulinemia, small lymphocytic lymphoma, marginal zone lymphoma, and chronic graft-versus-host disease. Current clinical practice is continuous long-term administration of BTKi, which can be complicated by adverse effects or the development of drug resistance. Alternatives to long-term use of BTKi are being developed, such as combination therapies, permitting for limited duration therapy. Second-generation BTKis are under development, which differ from ibrutinib, the first-in-class BTKi, in their specificity for BTK, and therefore may differentiate themselves from ibrutinib in terms of adverse effects or efficacy.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arrhythmias, Cardiac/etiology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Clinical Trials as Topic , Drug Resistance, Neoplasm/genetics , Humans , Invasive Fungal Infections/etiology , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Treatment Outcome
11.
Mol Cancer ; 17(1): 57, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29455639

ABSTRACT

Bruton's tyrosine kinase (BTK) is a non-receptor kinase that plays a crucial role in oncogenic signaling that is critical for proliferation and survival of leukemic cells in many B cell malignancies. BTK was initially shown to be defective in the primary immunodeficiency X-linked agammaglobulinemia (XLA) and is essential both for B cell development and function of mature B cells. Shortly after its discovery, BTK was placed in the signal transduction pathway downstream of the B cell antigen receptor (BCR). More recently, small-molecule inhibitors of this kinase have shown excellent anti-tumor activity, first in animal models and subsequently in clinical studies. In particular, the orally administered irreversible BTK inhibitor ibrutinib is associated with high response rates in patients with relapsed/refractory chronic lymphocytic leukemia (CLL) and mantle-cell lymphoma (MCL), including patients with high-risk genetic lesions. Because ibrutinib is generally well tolerated and shows durable single-agent efficacy, it was rapidly approved for first-line treatment of patients with CLL in 2016. To date, evidence is accumulating for efficacy of ibrutinib in various other B cell malignancies. BTK inhibition has molecular effects beyond its classic role in BCR signaling. These involve B cell-intrinsic signaling pathways central to cellular survival, proliferation or retention in supportive lymphoid niches. Moreover, BTK functions in several myeloid cell populations representing important components of the tumor microenvironment. As a result, there is currently a considerable interest in BTK inhibition as an anti-cancer therapy, not only in B cell malignancies but also in solid tumors. Efficacy of BTK inhibition as a single agent therapy is strong, but resistance may develop, fueling the development of combination therapies that improve clinical responses. In this review, we discuss the role of BTK in B cell differentiation and B cell malignancies and highlight the importance of BTK inhibition in cancer therapy.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/metabolism , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Agammaglobulinaemia Tyrosine Kinase/chemistry , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/pathology , Biomarkers, Tumor , Bone Marrow , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Lymphopoiesis/ethics , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Treatment Outcome , Tumor Microenvironment
12.
J Vet Diagn Invest ; 29(4): 529-535, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28467276

ABSTRACT

Acute leukemia is rare in horses. Herein we describe historical, clinicopathologic, and postmortem findings in 6 horses with acute leukemia. Medical records of horses with >20% bone marrow blasts and cytochemical or immunophenotyping results were reviewed. Affected horses were 2-8 y of age and of different breeds and sex. Horses were presented acutely with nonspecific signs (e.g., fever, lethargy). Characteristic hemogram findings were bi- or pancytopenia with low blast numbers. Histologic examination revealed extramedullary infiltrates, especially in lymph nodes, spleen, kidney, liver, and lungs. Leukemias were classified as B-cell ( n = 3) and acute myeloid leukemia (AML) ( n = 3). Tumors in 4 cases expressed multiple lineage markers, which complicated classification. Acute leukemia should be suspected in horses with moderate-to-severe bi- or pancytopenia. Blood smears should be reviewed for neoplastic cells, and bone marrow examination is required for diagnosis. Leukemia classification is best achieved using combined morphologic, cytochemical, and immunophenotyping results.


Subject(s)
Horse Diseases/pathology , Leukemia, B-Cell/veterinary , Leukemia, Myeloid, Acute/veterinary , Animals , Biomarkers/analysis , Female , Horse Diseases/etiology , Horses , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Male , New York
13.
Leuk Lymphoma ; 58(3): 528-539, 2017 03.
Article in English | MEDLINE | ID: mdl-27557932

ABSTRACT

Increasing evidence implicates a role of estrogens in hematological malignancies. We reviewed current knowledge on the emerging role of estrogens and estrogen receptors in normal B-cell function, chronic lymphocytic leukemia, and B-cell lymphoma. Data support that (1) normal human peripheral blood cells (mononuclear cells, total lymphocytes, T as well as B lymphocytes, and NK cells) express both estrogen receptor subtypes (ERα and ERß), (2) B-cell malignancies express mainly ERß while selective ERß agonists inhibit cell growth and induce apoptosis, (3) estrogens regulate, via an ER-mediated pathway, gene expression of cyclins, kinases, bcl-2 proto-oncogene, activation-induced deaminase (AID), and transcription factors, associated with changes in BCR signaling and B cell tumorigenesis. In conclusion, estrogen receptors play an important role in normal B-cell function and B-cell tumorigenesis; however, further investigations are required to delineate the role of estrogens and estrogen receptors in the etiopathogenesis and therapy of B-cell malignancies.


Subject(s)
Estrogens/metabolism , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Leukemia, B-Cell/mortality , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Mitochondria/genetics , Mitochondria/metabolism , Prognosis , Proto-Oncogene Mas , Receptors, Estrogen/metabolism , Signal Transduction
14.
Haematologica ; 101(5): 531-40, 2016 05.
Article in English | MEDLINE | ID: mdl-27132279

ABSTRACT

The tumor microenvironment is the cellular and molecular environment in which the tumor exists and with which it continuously interacts. In B-cell lymphomas, this microenvironment is intriguing in that it plays critical roles in the regulation of tumor cell survival and proliferation, fostering immune escape as well as the development of treatment resistance. The purpose of this review is to summarize the proceedings of the Second Annual Summit on the Immune Microenvironment in Hematologic Malignancies that took place on September 11-12, 2014 in Dublin, Ireland. We provide a timely overview of the composition and biological relevance of the cellular and molecular microenvironment interface and discuss the role of interactions between the microenvironment and neoplastic cells in a variety of B-cell lymphomas. In addition, we focus on various novel therapeutic strategies that target the tumor microenvironment, including agents that modulate B-cell receptor pathways and immune-checkpoints, chimeric antigen receptor T cells and immunomodulatory agents.


Subject(s)
B-Lymphocytes/pathology , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Tumor Microenvironment , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/therapy , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Staging , Signal Transduction , Tumor Escape/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
15.
Virology ; 494: 67-77, 2016 07.
Article in English | MEDLINE | ID: mdl-27085068

ABSTRACT

Mucosal lymphocytes support latent infections of species C adenoviruses. Because infected lymphocytes resist re-infection with adenovirus, we sought to identify changes in cellular gene expression that could inhibit the infectious process. The expression of over 30,000 genes was evaluated by microarray in persistently infected B-and T-lymphocytic cells. BBS9, BNIP3, BTG3, CXADR, SLFN11 and SPARCL1 were the only genes differentially expressed between mock and infected B cells. Most of these genes are associated with oncogenesis or cancer progression. Histone deacetylase and DNA methyltransferase inhibitors released the repression of some of these genes. Cellular and viral gene expression was compared among leukemic cell lines following adenovirus infection. Childhood leukemic B-cell lines resist adenovirus infection and also show reduced expression of CXADR and SPARCL. Thus adenovirus induces limited changes to infected B-cell lines that are similar to changes observed in childhood leukemic cell lines.


Subject(s)
Adenovirus Infections, Human/complications , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , Gene Expression Regulation, Leukemic , Leukemia/etiology , Virus Latency , Adult , Age Factors , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Line, Tumor , Child , Cluster Analysis , Gene Expression Profiling , Humans , Leukemia, B-Cell/etiology
16.
Clin Rev Allergy Immunol ; 50(3): 301-11, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26066671

ABSTRACT

B lymphocytes are generally recognized as the essential component of humoral immunity and also a regulator of innate immunity. The development of B cells is precisely regulated by a variety of factors via different mechanisms, including cytokine/cytokine receptors, signal transduction molecules, and transcription factors. Recent findings suggest that epigenetic factors, such as DNA methylation, histone modification, and non-coding RNA, play critical roles in establishing B cell lineage-specific gene expression profiles to define and sustain B cell identity and function. Epigenetic modifications are also sensitive to external stimuli and might bridge genetic and environmental factors in the pathogenesis or control of B-cell-related immune disorders, such as autoimmune diseases, lymphoma, and leukemia. Better understanding of the epigenetic mechanisms for regulating B cell development and involving B cell abnormal differentiation and function will shed light on the design of new therapeutic approaches to B-cell-related diseases, and potential candidates of epigenetic modulators may be identified to target epigenetic pathways to prevent or treat B cell disorders. We summarize the relevance of epigenetic marks and landscapes in the stages of B cell development, discuss the interaction of the transcriptional networks and epigenetic changes, and review the involvement of epigenetic risk in the pathogenesis of B-cell-related diseases. Understanding how specific epigenetic alterations contribute to the development of B-cell-related autoimmunity and malignancies is instrumental to control B cell disorders.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Immune System Diseases/etiology , Immune System Diseases/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphopoiesis/genetics , Transcription Factors/metabolism
17.
Blood ; 124(9): 1502-12, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25006123

ABSTRACT

The oncomir microRNA-125b (miR-125b) is upregulated in a variety of human neoplastic blood disorders and constitutive upregulation of miR-125b in mice can promote myeloid and B-cell leukemia. We found that miR-125b promotes myeloid and B-cell neoplasm by inducing tumorigenesis in hematopoietic progenitor cells. Our study demonstrates that miR-125b induces myeloid leukemia by enhancing myeloid progenitor output from stem cells as well as inducing immortality, self-renewal, and tumorigenesis in myeloid progenitors. Through functional and genetic analyses, we demonstrated that miR-125b induces myeloid and B-cell leukemia by inhibiting interferon regulatory factor 4 (IRF4) but through distinct mechanisms; it induces myeloid leukemia through repressing IRF4 at the messenger RNA (mRNA) level without altering the genomic DNA and induces B-cell leukemia via genetic deletion of the gene encoding IRF4.


Subject(s)
Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, B-Cell/genetics , Leukemia, B-Cell/metabolism , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Female , Gene Deletion , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Interferon Regulatory Factors/antagonists & inhibitors , Leukemia, B-Cell/etiology , Leukemia, Myeloid/etiology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Up-Regulation
18.
Nat Commun ; 5: 3273, 2014.
Article in English | MEDLINE | ID: mdl-24549417

ABSTRACT

The cell of origin and triggering events for leukaemia are mostly unknown. Here we show that the bone marrow contains a progenitor that expresses renin throughout development and possesses a B-lymphocyte pedigree. This cell requires RBP-J to differentiate. Deletion of RBP-J in these renin-expressing progenitors enriches the precursor B-cell gene programme and constrains lymphocyte differentiation, facilitated by H3K4me3 activating marks in genes that control the pre-B stage. Mutant cells undergo neoplastic transformation, and mice develop a highly penetrant B-cell leukaemia with multi-organ infiltration and early death. These renin-expressing cells appear uniquely vulnerable as other conditional models of RBP-J deletion do not result in leukaemia. The discovery of these unique renin progenitors in the bone marrow and the model of leukaemia described herein may enhance our understanding of normal and neoplastic haematopoiesis.


Subject(s)
Bone Marrow Cells/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Leukemia, B-Cell/etiology , Leukemia, Experimental/etiology , Renin/metabolism , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow/pathology , Bone Marrow Cells/pathology , Epigenesis, Genetic , Female , Hematopoiesis , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Spleen/pathology , Young Adult
19.
Leuk Lymphoma ; 55(7): 1551-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24033110

ABSTRACT

The observation of binucleated lymphocytes (BNLs) on a peripheral blood smear is essential to the diagnosis of persistent polyclonal B-cell lymphocytosis (PPBL). Only a few case reports have mentioned their presence in other contexts, mainly mature B-cell neoplasms such as chronic lymphocytic leukemia or reactive circumstances such as multiple sclerosis undergoing natalizumab treatment. We sought to investigate whether any particular morphologic features of BNL were more specific to PPBL than other diseases. We reviewed peripheral blood smears of a series of patients affected by PPBL or harboring BNLs whatever the diagnosis. We found that BNLs in PPBL were heterogeneous, but mostly medium-sized with a moderately abundant basophilic cytoplasm, sometimes vacuolated. The chromatin was mature in an asymmetrically bilobed nucleus showing one to two nucleoli. Though mainly observed in patients with PPBL, all these criteria remained non-specific. Conversely, the presence of either monocytoid or hyperbasophilic cells had clinical value to confirm PPBL. We conclude that a substantial percentage of BNLs possessing the morphologic features described and observed in a context of monocytoid and/or hyperbasophilic lymphocytes is predictive of the diagnosis of PPBL.


Subject(s)
B-Lymphocytes/pathology , Leukemia, B-Cell/diagnosis , Lymphocytosis/diagnosis , Lymphoma, B-Cell/diagnosis , Arthritis, Rheumatoid/pathology , Epstein-Barr Virus Infections/pathology , Humans , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Lymphocytosis/etiology , Lymphocytosis/pathology , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Multiple Sclerosis/pathology , Neoplasm Grading
20.
PLoS One ; 7(6): e38265, 2012.
Article in English | MEDLINE | ID: mdl-22701616

ABSTRACT

Constitutive activation of FGFR1, through rearrangement with various dimerization domains, leads to atypical myeloproliferative disorders where, although T cell lymphoma are common, the BCR-FGFR1 chimeric kinase results in CML-like leukemia. As with the human disease, mouse bone marrow transduction/transplantation with BCR-FGFR1 leads to CML-like myeloproliferation as well as B-cell leukemia/lymphoma. The murine disease described in this report is virtually identical to the human disease in that both showed bi-lineage involvement of myeloid and B-cells, splenomegaly, leukocytosis and bone marrow hypercellularity. A CD19(+) IgM(-) CD43(+) immunophenotype was seen both in primary tumors and two cell lines derived from these tumors. In all primary tumors, subpopulations of these CD19(+) IgM(-) CD43(+) were also either B220(+) or B220(-), suggesting a block in differentiation at the pro-B cell stage. The B220(-) phenotype was retained in one of the cell lines while the other was B220(+). When the two cell lines were transplanted into syngeneic mice, all animals developed the same B-lymphoblastic leukemia within 2-weeks. Thus, the murine model described here closely mimics the human disease with bilineage myeloid and B-cell leukemia/lymphoma which provides a representative model to investigate therapeutic intervention and a better understanding of the etiology of the disease.


Subject(s)
Cell Line, Tumor , Cell Lineage/physiology , Leukemia, B-Cell/physiopathology , Myeloproliferative Disorders/physiopathology , Precursor Cells, B-Lymphoid/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antigens, CD19/immunology , Blotting, Western , Bone Marrow Transplantation , Cell Lineage/immunology , Comparative Genomic Hybridization , DNA Primers/genetics , Flow Cytometry , Histological Techniques , Immunoglobulin M/immunology , Immunophenotyping , Karyotyping , Leukemia, B-Cell/etiology , Leukocyte Common Antigens/immunology , Leukosialin/immunology , Mice , Mice, Inbred BALB C , Myeloproliferative Disorders/etiology , Precursor Cells, B-Lymphoid/immunology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL