Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38252421

ABSTRACT

PURPOSE: Develop a novel therapeutic strategy for patients with subtypes of mature T-cell and NK-cell neoplasms. EXPERIMENTAL DESIGN: Primary specimens, cell lines, patient-derived xenograft models, commercially available and proprietary anti-KLRG1 antibodies were used for screening, target, and functional validation. RESULTS: Here we demonstrate that surface KLRG1 is highly expressed on tumor cells in subsets of patients with extranodal NK/T-cell lymphoma (ENKTCL), T-prolymphocytic leukemia (T-PLL) and gamma/delta T-cell lymphoma (G/D TCL). The majority of the CD8+/CD57+ or CD3-/CD56+ leukemic cells derived from patients with T- and NK-large granular lymphocytic leukemia (T-LGLL and NK-LGLL) respectively expressed surface KLRG1. The humanized afucosylated anti-KLRG1 monoclonal antibody (mAb208) optimized for mouse in vivo use depleted KLRG1+ TCL cells by mechanisms of ADCC, ADCP and CDC rather than apoptosis. mAb208 induced ADCC and ADCP of T-LGLL patient-derived CD8+/CD57+ cells ex vivo. mAb208 effected ADCC of subsets of healthy donor-derived KLRG1+ NK, CD4+, CD8+ Tem and TemRA cells while sparing KLRG1- naive and CD8+ Tcm cells. Treatment of cell line and TCL patient-derived xenografts with mAb208 or anti-CD47 mAb alone and in combination with the PI3K-δ/γ inhibitor, duvelisib extended survival. The depletion of macrophages in vivo antagonized mAb208 efficacy. CONCLUSIONS: Our findings suggest the potential benefit of a broader treatment strategy combining therapeutic antibodies with PI3Ki for the treatment of patients with mature T-cell and NK-cell neoplasms.

2.
Cureus ; 10(8): e3227, 2018 Aug 29.
Article in English | MEDLINE | ID: mdl-30410833

ABSTRACT

Enterococcal empyema is a rare complication of pneumonia. We report the case of a 67-year-old asplenic man with pneumonia complicated by respiratory failure and empyema requiring decortication and prolonged chest tube drainage. Cultures of the empyema were initially negative, but later grew vancomycin-resistant Enterococcus faecium (VRE), which was successfully treated with linezolid. To our knowledge, this is only the second reported case of an empyema caused by VRE that was not associated with an intra-abdominal infection. We suspect superinfection due to airway or chest tube contamination as the most likely mechanism of infection. Physicians should consider multi-drug resistant organisms such as VRE in patients with empyema that fail to resolve with chest tube drainage and broad-spectrum antibiotics.

3.
Sci Transl Med ; 8(363): 363ra147, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807282

ABSTRACT

Multiple myeloma (MM) remains an incurable disease, with a treatment-refractory state eventually developing in all patients. Constant clonal evolution and genetic heterogeneity of MM are a likely explanation for the emergence of drug-resistant disease. Monitoring of MM genomic evolution on therapy by serial bone marrow biopsy is unfortunately impractical because it involves an invasive and painful procedure. We describe how noninvasive and highly sensitive isolation and characterization of circulating tumor cells (CTCs) from peripheral blood at single-cell resolution recapitulate MM in the bone marrow. We demonstrate that CTCs provide the same genetic information as bone marrow MM cells and even reveal mutations with greater sensitivity than bone marrow biopsies in some cases. Single CTC RNA sequencing enables classification of MM and quantitative assessment of genes that are relevant for prognosis. We propose that the genomic characterization of CTCs should be included in clinical trials to follow the emergence of resistant subclones after MM therapy.


Subject(s)
Bone Marrow/pathology , Genetic Heterogeneity , Multiple Myeloma/genetics , Neoplastic Cells, Circulating/pathology , DNA Mutational Analysis , Feasibility Studies , Gene Expression Profiling , Genotype , Humans , Loss of Heterozygosity , Multiple Myeloma/blood , Mutation , Plasma Cells/metabolism , Prognosis , Proof of Concept Study , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic , Tumor Burden
4.
Nat Genet ; 48(3): 265-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829750

ABSTRACT

Translocation events are frequent in cancer and may create chimeric fusions or 'regulatory rearrangements' that drive oncogene overexpression. Here we identify super-enhancer translocations that drive overexpression of the oncogenic transcription factor MYB as a recurrent theme in adenoid cystic carcinoma (ACC). Whole-genome sequencing data and chromatin maps highlight distinct chromosomal rearrangements that juxtapose super-enhancers to the MYB locus. Chromosome conformation capture confirms that the translocated enhancers interact with the MYB promoter. Remarkably, MYB protein binds to the translocated enhancers, creating a positive feedback loop that sustains its expression. MYB also binds enhancers that drive different regulatory programs in alternate cell lineages in ACC, cooperating with TP63 in myoepithelial cells and a Notch program in luminal epithelial cells. Bromodomain inhibitors slow tumor growth in ACC primagraft models in vivo. Thus, our study identifies super-enhancer translocations that drive MYB expression and provides insight into downstream MYB functions in alternate ACC lineages.


Subject(s)
Carcinoma, Adenoid Cystic/genetics , Enhancer Elements, Genetic , Oncogene Proteins v-myb/biosynthesis , Translocation, Genetic , Carcinoma, Adenoid Cystic/pathology , Cell Line, Tumor , Cell Lineage/genetics , Chromatin/genetics , Gene Expression Regulation, Neoplastic , Humans , Oncogene Proteins v-myb/genetics , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Transcription Factors/biosynthesis , Tumor Suppressor Proteins/biosynthesis
5.
Mol Cell ; 61(1): 170-80, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26687680

ABSTRACT

Genome-wide profiling of histone modifications can provide systematic insight into the regulatory elements and programs engaged in a given cell type. However, conventional chromatin immunoprecipitation and sequencing (ChIP-seq) does not capture quantitative information on histone modification levels, requires large amounts of starting material, and involves tedious processing of each individual sample. Here, we address these limitations with a technology that leverages DNA barcoding to profile chromatin quantitatively and in multiplexed format. We concurrently map relative levels of multiple histone modifications across multiple samples, each comprising as few as a thousand cells. We demonstrate the technology by monitoring dynamic changes following inhibition of p300, EZH2, or KDM5, by linking altered epigenetic landscapes to chromatin regulator mutations, and by mapping active and repressive marks in purified human hematopoietic stem cells. Hence, this technology enables quantitative studies of chromatin state dynamics across rare cell types, genotypes, environmental conditions, and drug treatments.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation/methods , Chromatin/metabolism , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing/methods , Histones/metabolism , Leukemia/metabolism , Multiplex Polymerase Chain Reaction/methods , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , DNA Barcoding, Taxonomic , Epigenesis, Genetic/drug effects , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Histones/genetics , Humans , K562 Cells , Leukemia/genetics , Mutation
6.
Nature ; 523(7559): 221-5, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25924064

ABSTRACT

Inflammation is a beneficial host response to infection but can contribute to inflammatory disease if unregulated. The Th17 lineage of T helper (Th) cells can cause severe human inflammatory diseases. These cells exhibit both instability (they can cease to express their signature cytokine, IL-17A) and plasticity (they can start expressing cytokines typical of other lineages) upon in vitro re-stimulation. However, technical limitations have prevented the transcriptional profiling of pre- and post-conversion Th17 cells ex vivo during immune responses. Thus, it is unknown whether Th17 cell plasticity merely reflects change in expression of a few cytokines, or if Th17 cells physiologically undergo global genetic reprogramming driving their conversion from one T helper cell type to another, a process known as transdifferentiation. Furthermore, although Th17 cell instability/plasticity has been associated with pathogenicity, it is unknown whether this could present a therapeutic opportunity, whereby formerly pathogenic Th17 cells could adopt an anti-inflammatory fate. Here we used two new fate-mapping mouse models to track Th17 cells during immune responses to show that CD4(+) T cells that formerly expressed IL-17A go on to acquire an anti-inflammatory phenotype. The transdifferentiation of Th17 into regulatory T cells was illustrated by a change in their signature transcriptional profile and the acquisition of potent regulatory capacity. Comparisons of the transcriptional profiles of pre- and post-conversion Th17 cells also revealed a role for canonical TGF-ß signalling and consequently for the aryl hydrocarbon receptor (AhR) in conversion. Thus, Th17 cells transdifferentiate into regulatory cells, and contribute to the resolution of inflammation. Our data suggest that Th17 cell instability and plasticity is a therapeutic opportunity for inflammatory diseases.


Subject(s)
Cell Transdifferentiation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , Helminthiasis/immunology , Male , Mice , Nippostrongylus/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology
7.
Nat Genet ; 46(4): 364-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584072

ABSTRACT

The identification of activating NOTCH1 mutations in T cell acute lymphoblastic leukemia (T-ALL) led to clinical testing of γ-secretase inhibitors (GSIs) that prevent NOTCH1 activation. However, responses to these inhibitors have been transient, suggesting that resistance limits their clinical efficacy. Here we modeled T-ALL resistance, identifying GSI-tolerant 'persister' cells that expand in the absence of NOTCH1 signaling. Rare persisters are already present in naive T-ALL populations, and the reversibility of their phenotype suggests an epigenetic mechanism. Relative to GSI-sensitive cells, persister cells activate distinct signaling and transcriptional programs and exhibit chromatin compaction. A knockdown screen identified chromatin regulators essential for persister viability, including BRD4. BRD4 binds enhancers near critical T-ALL genes, including MYC and BCL2. The BRD4 inhibitor JQ1 downregulates expression of these targets and induces growth arrest and apoptosis in persister cells, at doses well tolerated by GSI-sensitive cells. Consistently, the GSI-JQ1 combination was found to be effective against primary human leukemias in vivo. Our findings establish a role for epigenetic heterogeneity in leukemia resistance that may be addressed by incorporating epigenetic modulators in combination therapy.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Chromatin/genetics , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic/genetics , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Histones/metabolism , Humans , Indoles , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Real-Time Polymerase Chain Reaction , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...