Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 42(5): 833-849.e12, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38701792

ABSTRACT

Glucocorticoids have been used for decades to treat lymphomas without an established mechanism of action. Using functional genomic, proteomic, and chemical screens, we discover that glucocorticoids inhibit oncogenic signaling by the B cell receptor (BCR), a recurrent feature of aggressive B cell malignancies, including diffuse large B cell lymphoma and Burkitt lymphoma. Glucocorticoids induce the glucocorticoid receptor (GR) to directly transactivate genes encoding negative regulators of BCR stability (LAPTM5; KLHL14) and the PI3 kinase pathway (INPP5D; DDIT4). GR directly represses transcription of CSK, a kinase that limits the activity of BCR-proximal Src-family kinases. CSK inhibition attenuates the constitutive BCR signaling of lymphomas by hyperactivating Src-family kinases, triggering their ubiquitination and degradation. With the knowledge that glucocorticoids disable oncogenic BCR signaling, they can now be deployed rationally to treat BCR-dependent aggressive lymphomas and used to construct mechanistically sound combination regimens with inhibitors of BTK, PI3 kinase, BCL2, and CSK.


Subject(s)
Glucocorticoids , Receptors, Antigen, B-Cell , Humans , Glucocorticoids/pharmacology , Receptors, Antigen, B-Cell/metabolism , Animals , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Mice , Cell Line, Tumor , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Molecular Targeted Therapy/methods , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects
2.
Sci Transl Med ; 15(702): eabo3826, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37379367

ABSTRACT

Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) show potent efficacy in several ALK-driven tumors, but the development of resistance limits their long-term clinical impact. Although resistance mechanisms have been studied extensively in ALK-driven non-small cell lung cancer, they are poorly understood in ALK-driven anaplastic large cell lymphoma (ALCL). Here, we identify a survival pathway supported by the tumor microenvironment that activates phosphatidylinositol 3-kinase γ (PI3K-γ) signaling through the C-C motif chemokine receptor 7 (CCR7). We found increased PI3K signaling in patients and ALCL cell lines resistant to ALK TKIs. PI3Kγ expression was predictive of a lack of response to ALK TKI in patients with ALCL. Expression of CCR7, PI3Kγ, and PI3Kδ were up-regulated during ALK or STAT3 inhibition or degradation and a constitutively active PI3Kγ isoform cooperated with oncogenic ALK to accelerate lymphomagenesis in mice. In a three-dimensional microfluidic chip, endothelial cells that produce the CCR7 ligands CCL19/CCL21 protected ALCL cells from apoptosis induced by crizotinib. The PI3Kγ/δ inhibitor duvelisib potentiated crizotinib activity against ALCL lines and patient-derived xenografts. Furthermore, genetic deletion of CCR7 blocked the central nervous system dissemination and perivascular growth of ALCL in mice treated with crizotinib. Thus, blockade of PI3Kγ or CCR7 signaling together with ALK TKI treatment reduces primary resistance and the survival of persister lymphoma cells in ALCL.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lymphoma, Large-Cell, Anaplastic , Humans , Animals , Mice , Crizotinib/pharmacology , Crizotinib/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Receptors, CCR7/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases , Lung Neoplasms/drug therapy , Protein-Tyrosine Kinases , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/pathology , Cell Line, Tumor , Tumor Microenvironment
3.
Cancers (Basel) ; 12(12)2020 Dec 21.
Article in English | MEDLINE | ID: mdl-33371292

ABSTRACT

Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a CD30-positive, anaplastic lymphoma kinase-negative T-cell lymphoma. Where implant history is known, all confirmed cases to date have occurred in patients with exposure to textured implants. There is a spectrum of disease presentation, with the most common occurring as a seroma with an indolent course. A less common presentation occurs as locally advanced or, rarely, as metastatic disease. Here we review the immunological characteristics of BIA-ALCL and potential triggers leading to its development. BIA-ALCL occurs in an inflammatory microenvironment with significant lymphocyte and plasma cell infiltration and a prominent Th1/Th17 phenotype in advanced disease. Genetic lesions affecting the JAK/STAT signaling pathway are commonly present. Proposed triggers for the development of malignancy include mechanical friction, silicone implant shell particulates, silicone leachables, and bacteria. Of these, the bacterial hypothesis has received significant attention, supported by a plausible biologic model. In this model, bacteria form an adherent biofilm in the favorable environment of the textured implant surface, producing a bacterial load that elicits a chronic inflammatory response. Bacterial antigens, primarily of Gram-negative origin, may trigger innate immunity and induce T-cell proliferation with subsequent malignant transformation in genetically susceptible individuals. Although much remains to be elucidated regarding the multifactorial origins of BIA-ALCL, future research should focus on prevention and treatment strategies, recognizing susceptible populations, and whether decreasing the risk of BIA-ALCL is possible.

5.
Nat Med ; 26(7): 1114-1124, 2020 07.
Article in English | MEDLINE | ID: mdl-32483360

ABSTRACT

In many areas of oncology, we lack sensitive tools to track low-burden disease. Although cell-free DNA (cfDNA) shows promise in detecting cancer mutations, we found that the combination of low tumor fraction (TF) and limited number of DNA fragments restricts low-disease-burden monitoring through the prevailing deep targeted sequencing paradigm. We reasoned that breadth may supplant depth of sequencing to overcome the barrier of cfDNA abundance. Whole-genome sequencing (WGS) of cfDNA allowed ultra-sensitive detection, capitalizing on the cumulative signal of thousands of somatic mutations observed in solid malignancies, with TF detection sensitivity as low as 10-5. The WGS approach enabled dynamic tumor burden tracking and postoperative residual disease detection, associated with adverse outcome. Thus, we present an orthogonal framework for cfDNA cancer monitoring via genome-wide mutational integration, enabling ultra-sensitive detection, overcoming the limitation of cfDNA abundance and empowering treatment optimization in low-disease-burden oncology care.


Subject(s)
Biomarkers, Tumor/genetics , Circulating Tumor DNA/blood , DNA, Neoplasm/genetics , Neoplasms/blood , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , DNA Copy Number Variations/genetics , DNA, Neoplasm/blood , Disease-Free Survival , Female , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Male , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Burden/genetics , Whole Genome Sequencing
6.
Nat Genet ; 52(4): 388-400, 2020 04.
Article in English | MEDLINE | ID: mdl-32203470

ABSTRACT

Differences in three-dimensional (3D) chromatin architecture can influence the integrity of topologically associating domains (TADs) and rewire specific enhancer-promoter interactions, impacting gene expression and leading to human disease. Here we investigate the 3D chromatin architecture in T cell acute lymphoblastic leukemia (T-ALL) by using primary human leukemia specimens and examine the dynamic responses of this architecture to pharmacological agents. Systematic integration of matched in situ Hi-C, RNA-seq and CTCF ChIP-seq datasets revealed widespread differences in intra-TAD chromatin interactions and TAD boundary insulation in T-ALL. Our studies identify and focus on a TAD 'fusion' event associated with absence of CTCF-mediated insulation, enabling direct interactions between the MYC promoter and a distal super-enhancer. Moreover, our data also demonstrate that small-molecule inhibitors targeting either oncogenic signal transduction or epigenetic regulation can alter specific 3D interactions found in leukemia. Overall, our study highlights the impact, complexity and dynamic nature of 3D chromatin architecture in human acute leukemia.


Subject(s)
Chromatin/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , T-Lymphocytes/physiology , Animals , CCCTC-Binding Factor/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/genetics , Humans , Jurkat Cells , Mice , Promoter Regions, Genetic/genetics
7.
Sci Transl Med ; 11(511)2019 09 25.
Article in English | MEDLINE | ID: mdl-31554741

ABSTRACT

CAR T cells targeting CD19 provide promising options for treatment of B cell malignancies. However, tumor relapse from antigen loss can limit efficacy. We developed humanized, second-generation CAR T cells against another B cell-specific marker, B cell activating factor receptor (BAFF-R), which demonstrated cytotoxicity against human lymphoma and acute lymphoblastic leukemia (ALL) lines. Adoptively transferred BAFF-R-CAR T cells eradicated 10-day preestablished tumor xenografts after a single treatment and retained efficacy against xenografts deficient in CD19 expression, including CD19-negative variants within a background of CD19-positive lymphoma cells. Four relapsed, primary ALLs with CD19 antigen loss obtained after CD19-directed therapy retained BAFF-R expression and activated BAFF-R-CAR, but not CD19-CAR, T cells. BAFF-R-CAR, but not CD19-CAR, T cells also demonstrated antitumor effects against an additional CD19 antigen loss primary patient-derived xenograft (PDX) in vivo. BAFF-R is amenable to CAR T cell therapy, and its targeting may prevent emergence of CD19 antigen loss variants.


Subject(s)
Antigens, CD19/metabolism , B-Cell Activation Factor Receptor/metabolism , Immunotherapy, Adoptive , Leukemia, B-Cell/therapy , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Leukemia, B-Cell/immunology , Lymphocyte Activation/immunology , Mice , T-Lymphocytes/immunology
8.
Leukemia ; 33(3): 696-709, 2019 03.
Article in English | MEDLINE | ID: mdl-30131584

ABSTRACT

TYK2 is a member of the JAK family of tyrosine kinases that is involved in chromosomal translocation-induced fusion proteins found in anaplastic large cell lymphomas (ALCL) that lack rearrangements activating the anaplastic lymphoma kinase (ALK). Here we demonstrate that TYK2 is highly expressed in all cases of human ALCL, and that in a mouse model of NPM-ALK-induced lymphoma, genetic disruption of Tyk2 delays the onset of tumors and prolongs survival of the mice. Lymphomas in this model lacking Tyk2 have reduced STAT1 and STAT3 phosphorylation and reduced expression of Mcl1, a pro-survival member of the BCL2 family. These findings in mice are mirrored in human ALCL cell lines, in which TYK2 is activated by autocrine production of IL-10 and IL-22 and by interaction with specific receptors expressed by the cells. Activated TYK2 leads to STAT1 and STAT3 phosphorylation, activated expression of MCL1 and aberrant ALCL cell survival. Moreover, TYK2 inhibitors are able to induce apoptosis in ALCL cells, regardless of the presence or absence of an ALK-fusion. Thus, TYK2 is a dependency that is required for ALCL cell survival through activation of MCL1 expression. TYK2 represents an attractive drug target due to its essential enzymatic domain, and TYK2-specific inhibitors show promise as novel targeted inhibitors for ALCL.


Subject(s)
Lymphoma, Large-Cell, Anaplastic/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , STAT1 Transcription Factor/genetics , TYK2 Kinase/genetics , Anaplastic Lymphoma Kinase/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Lymphoma, Large-Cell, Anaplastic/drug therapy , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Translocation, Genetic/drug effects , Translocation, Genetic/genetics
9.
Sci Rep ; 7(1): 11301, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900149

ABSTRACT

T-cell clonality of peripheral T-cell lymphoma (PTCL) is routinely evaluated with a PCR-based method using genomic DNA. However, there are limitations with this approach. The purpose of this study was to determine the utility of RNA-seq for assessing T-cell clonality and T-cell antigen receptor (TCR) repertoire of the neoplastic T-cells in 108 PTCL samples. TCR transcripts, including complementarity-determining region 3 (CDR3) sequences, were assessed. In normal T cells, the CDR3 sequences were extremely diverse, without any clonotype representing more than 2% of the overall TCR population. Dominant clones could be identified in 65 out of 76 PTCL cases (86%) with adequate TCR transcript expression. In monoclonal cases, the dominant clone varied between 11% and 99% of TCRß transcripts. No unique Vα or Vß usage was observed. Small T-cell clones were often observed in T- and NK-cell tumors in a percentage higher than observed in reactive conditions. γ chain expression was very low in tumors expressing TCRαß, but its expression level was high and clonality was detected in a TCRγδ expressing tumor. NK cell lymphoma (NKCL) did not express significant levels of TCR Vß or Vγ genes. RNA-seq is a useful tool for detecting and characterizing clonal TCR rearrangements in PTCL.


Subject(s)
Clonal Evolution/genetics , Lymphoma, T-Cell, Peripheral/genetics , Receptors, Antigen, T-Cell/genetics , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Lymphoma, T-Cell, Peripheral/pathology , Mutation , Sequence Analysis, RNA , Transcriptome
10.
Cancer Cell ; 31(1): 110-126, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27989801

ABSTRACT

Tumor-associated endothelial cells (TECs) regulate tumor cell aggressiveness. However, the core mechanism by which TECs confer stem cell-like activity to indolent tumors is unknown. Here, we used in vivo murine and human tumor models to identify the tumor-suppressive checkpoint role of TEC-expressed insulin growth factor (IGF) binding protein-7 (IGFBP7/angiomodulin). During tumorigenesis, IGFBP7 blocks IGF1 and inhibits expansion and aggresiveness of tumor stem-like cells (TSCs) expressing IGF1 receptor (IGF1R). However, chemotherapy triggers TECs to suppress IGFBP7, and this stimulates IGF1R+ TSCs to express FGF4, inducing a feedforward FGFR1-ETS2 angiocrine cascade that obviates TEC IGFBP7. Thus, loss of IGFBP7 and upregulation of IGF1 activates the FGF4-FGFR1-ETS2 pathway in TECs and converts naive tumor cells to chemoresistant TSCs, thereby facilitating their invasiveness and progression.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/physiology , Neoplastic Stem Cells/drug effects , Animals , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Endothelial Cells/physiology , Fibroblast Growth Factor 4/physiology , Humans , Insulin-Like Growth Factor I/physiology , Mice , Proto-Oncogene Protein c-ets-2/physiology , Receptor, IGF Type 1/physiology
11.
Nat Commun ; 6: 6921, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25891015

ABSTRACT

The contribution of epigenomic alterations to tumour progression and relapse is not well characterized. Here we characterize an association between disease progression and DNA methylation in diffuse large B-cell lymphoma (DLBCL). By profiling genome-wide DNA methylation at single-base pair resolution in thirteen DLBCL diagnosis-relapse sample pairs, we show that DLBCL patients exhibit heterogeneous evolution of tumour methylomes during relapse. We identify differentially methylated regulatory elements and determine a relapse-associated methylation signature converging on key pathways such as transforming growth factor-ß (TGF-ß) receptor activity. We also observe decreased intra-tumour methylation heterogeneity from diagnosis to relapsed tumour samples. Relapse-free patients display lower intra-tumour methylation heterogeneity at diagnosis compared with relapsed patients in an independent validation cohort. Furthermore, intra-tumour methylation heterogeneity is predictive of time to relapse. Therefore, we propose that epigenomic heterogeneity may support or drive the relapse phenotype and can be used to predict DLBCL relapse.


Subject(s)
Biological Evolution , Epigenomics , Lymphoma, Large B-Cell, Diffuse/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Recurrence , Transcriptome
12.
Curr Biol ; 13(21): 1858-66, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14588241

ABSTRACT

BACKGROUND: Bam32/DAPP1 is a B cell adaptor composed of both a PH and an SH2 domain. Previous studies in cell culture and chicken DT40 cells have indicated that Bam32 is critical for normal signaling downstream of the B cell receptor (BCR). RESULTS: We now study the function of Bam32 in mice in which Bam32 has been disrupted by a viral gene trap approach. Although B and T cell development is normal in Bam32(-/-) mice, B cell proliferation is reduced by about 50% after BCR crosslinking when compared with Bam32(+/+) mice. Differences in the activation of Erk, Jnk and p38 Map kinases, PLCgamma, and Ca(2+) flux do not account for the defect in proliferation as activation was similar in Bam32(+/+) and Bam32(-/-) B cells. Interestingly, whereas antibody response to T-dependent (TD) and T-independent (TI)-I antigens was similar between Bam32(+/+) and Bam32(-/-) mice, TI-II responses were defective in Bam32(-/-) mice; Bam32(-/-) mice failed to undergo isotype class switch recombination (CSR) to produce IgG3 antibodies due to a cell-autonomous defect in generation of IgG3 germline transcripts. The defect in TI-II antigen response led to an impaired antibody response to immunization with type 3 Streptococcus pneumoniae capsular polyschaccharide (PS), resulting in a markedly increased susceptibility to infection by Streptococcus pneumoniae. CONCLUSIONS: These findings indicate that Bam32 specifically couples an upstream signal to the IgG3 isotype heavy chain CSR and suggest that defects in Bam32 may account for the increased susceptibility to encapusulated organisms in a subset of immunodeficient patients.


Subject(s)
Adaptor Proteins, Signal Transducing , Antigens, T-Independent/metabolism , B-Lymphocytes/immunology , Carrier Proteins/metabolism , Lipoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , src Homology Domains/physiology , Animals , Antibody Formation , Antigens, T-Independent/immunology , B-Lymphocytes/physiology , Carrier Proteins/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Lipoproteins/immunology , Membrane Proteins/immunology , Mice , Mice, Mutant Strains , Molecular Probe Techniques , Precipitin Tests , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...