Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 124(9): 1478-1490, 2021 04.
Article in English | MEDLINE | ID: mdl-33723398

ABSTRACT

The transcriptional upregulation of oncogenes is a driving force behind the progression of many tumours. However, until a decade ago, the concept of 'switching off' these oncogenic pathways represented a formidable challenge. Research has revealed that members of the bromo- and extra-terminal domain (BET) motif family are key activators of oncogenic networks in a spectrum of cancers; their function depends on their recruitment to chromatin through two bromodomains (BD1 and BD2). The advent of potent inhibitors of BET proteins (BETi), which target either one or both bromodomains, represents an important step towards the goal of suppressing oncogenic networks within tumours. Here, we discuss the biology of BET proteins, advances in BETi design and highlight potential biomarkers predicting their activity. We also outline the logic of incorporating BETi into combination therapies to enhance its efficacy. We suggest that understanding mechanisms of activity, defining predictive biomarkers and identifying potent synergies represents a roadmap for clinical success using BETi.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Humans , Neoplasms/metabolism , Neoplasms/pathology , Prognosis
2.
Mol Cancer Ther ; 20(1): 64-75, 2021 01.
Article in English | MEDLINE | ID: mdl-33087508

ABSTRACT

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but often lethal cancer that is diagnosed at a median age of 24 years. Optimal management of patients is not well defined, and current treatment remains challenging, necessitating the discovery of novel therapeutic approaches. The identification of SMARCA4-inactivating mutations invariably characterizing this type of cancer provided insights facilitating diagnostic and therapeutic measures against this disease. We show here that the BET inhibitor OTX015 acts in synergy with the MEK inhibitor cobimetinib to repress the proliferation of SCCOHT in vivo Notably, this synergy is also observed in some SMARCA4-expressing ovarian adenocarcinoma models intrinsically resistant to BETi. Mass spectrometry, coupled with knockdown of newly found targets such as thymidylate synthase, revealed that the repression of a panel of proteins involved in nucleotide synthesis underlies this synergy both in vitro and in vivo, resulting in reduced pools of nucleotide metabolites and subsequent cell-cycle arrest. Overall, our data indicate that dual treatment with BETi and MEKi represents a rational combination therapy against SCCOHT and potentially additional ovarian cancer subtypes.


Subject(s)
Epigenesis, Genetic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nucleotides/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Azetidines/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Synergism , Epigenesis, Genetic/drug effects , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Ovarian Neoplasms/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/therapeutic use , S Phase/drug effects , Xenograft Model Antitumor Assays
3.
Cell Metab ; 31(2): 250-266.e9, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023446

ABSTRACT

Epigenetic modifications on DNA and histones regulate gene expression by modulating chromatin accessibility to transcription machinery. Here we identify methionine as a key nutrient affecting epigenetic reprogramming in CD4+ T helper (Th) cells. Using metabolomics, we showed that methionine is rapidly taken up by activated T cells and serves as the major substrate for biosynthesis of the universal methyl donor S-adenosyl-L-methionine (SAM). Methionine was required to maintain intracellular SAM pools in T cells. Methionine restriction reduced histone H3K4 methylation (H3K4me3) at the promoter regions of key genes involved in Th17 cell proliferation and cytokine production. Applied to the mouse model of multiple sclerosis (experimental autoimmune encephalomyelitis), dietary methionine restriction reduced the expansion of pathogenic Th17 cells in vivo, leading to reduced T cell-mediated neuroinflammation and disease onset. Our data identify methionine as a key nutritional factor shaping Th cell proliferation and function in part through regulation of histone methylation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Epigenesis, Genetic/drug effects , Histones/metabolism , Methionine , Multiple Sclerosis , Th17 Cells/metabolism , Animals , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , HEK293 Cells , Humans , Methionine/metabolism , Methionine/pharmacology , Methylation , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Th17 Cells/cytology
4.
Cancer Res ; 79(10): 2761-2774, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30877105

ABSTRACT

The antitumor activity of bromodomain and extraterminal motif protein inhibitors (BETi) has been demonstrated across numerous types of cancer. As such, these inhibitors are currently undergoing widespread clinical evaluation. However, predictive biomarkers allowing the stratification of tumors into responders and nonresponders to BETi are lacking. Here, we showed significant antiproliferative effects of low dosage BETi in vitro and in vivo against aggressive ovarian and lung cancer models lacking SMARCA4 and SMARCA2, key components of SWI/SNF chromatin remodeling complexes. Restoration of SMARCA4 or SMARCA2 promoted resistance to BETi in these models and, conversely, knockdown of SMARCA4 sensitized resistant cells to BETi. Transcriptomic analysis revealed that exposure to BETi potently downregulated a network of genes involved in receptor tyrosine kinase (RTK) signaling in SMARCA4/A2-deficient cells, including the oncogenic RTK HER3. Repression of signaling downstream of HER3 was found to be an important determinant of response to BETi in SMARCA4/A2-deficient cells. Overall, we propose that BETi represent a rational therapeutic strategy in poor-prognosis, SMARCA4/A2-deficient cancers. SIGNIFICANCE: These findings address an unmet clinical need by identifying loss of SMARCA4/A2 as biomarkers of hypersensitivity to BETi.


Subject(s)
DNA Helicases/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Chromatin Assembly and Disassembly , Female , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Signal Transduction
5.
Biochim Biophys Acta ; 1865(2): 275-88, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27085853

ABSTRACT

Cancer constitutes a set of diseases with heterogeneous molecular pathologies. However, there are a number of universal aberrations common to all cancers, one of these being the epigenetic silencing of tumor suppressor genes (TSGs). The silencing of TSGs is thought to be an early, driving event in the oncogenic process. With this in consideration, great efforts have been made to develop small molecules aimed at the restoration of TSGs in order to limit tumor cell proliferation and survival. However, the molecular forces that drive the broad epigenetic reprogramming and transcriptional repression of these genes remain ill-defined. Undoubtedly, understanding the molecular underpinnings of transcriptionally silenced TSGs will aid us in our ability to reactivate these key anti-cancer targets. Here, we describe what we consider to be the five most logical molecular mechanisms that may account for this widely observed phenomenon: 1) ablation of transcription factor binding, 2) overexpression of DNA methyltransferases, 3) disruption of CTCF binding, 4) elevation of EZH2 activity, 5) aberrant expression of long non-coding RNAs. The strengths and weaknesses of each proposed mechanism is highlighted, followed by an overview of clinical efforts to target these processes.


Subject(s)
Epigenesis, Genetic , Gene Silencing , Genes, Tumor Suppressor , Animals , DNA (Cytosine-5-)-Methyltransferases/physiology , Enhancer of Zeste Homolog 2 Protein , Histone Deacetylase Inhibitors/pharmacology , Humans , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/physiology , RNA, Untranslated/physiology , Transcription, Genetic , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL
...