Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 175(6): 1665-1678.e18, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30343896

ABSTRACT

Low-grade gliomas almost invariably progress into secondary glioblastoma (sGBM) with limited therapeutic option and poorly understood mechanism. By studying the mutational landscape of 188 sGBMs, we find significant enrichment of TP53 mutations, somatic hypermutation, MET-exon-14-skipping (METex14), PTPRZ1-MET (ZM) fusions, and MET amplification. Strikingly, METex14 frequently co-occurs with ZM fusion and is present in ∼14% of cases with significantly worse prognosis. Subsequent studies show that METex14 promotes glioma progression by prolonging MET activity. Furthermore, we describe a MET kinase inhibitor, PLB-1001, that demonstrates remarkable potency in selectively inhibiting MET-altered tumor cells in preclinical models. Importantly, this compound also shows blood-brain barrier permeability and is subsequently applied in a phase I clinical trial that enrolls MET-altered chemo-resistant glioma patients. Encouragingly, PLB-1001 achieves partial response in at least two advanced sGBM patients with rarely significant side effects, underscoring the clinical potential for precisely treating gliomas using this therapy.


Subject(s)
Brain Neoplasms , Exons , Glioblastoma , Mutation , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Drug Delivery Systems , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445128

ABSTRACT

The WNT (Wingless/Integrated) signaling pathway is implicated in various stages of glioblastoma, which is an aggressive brain tumor for which therapeutic options are limited. WNT has been recognized as a hallmark of therapeutic challenge due to its context-dependent role and critical function in healthy tissue homeostasis. In this review, we deeply scrutinize the WNT signaling pathway and its involvement in the genesis of glioblastoma as well as its acquired therapy resistance. We also provide an analysis of the WNT pathway in terms of its therapeutic importance in addition to an overview of the current targeted therapies under clinical investigation.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Wnt Signaling Pathway/genetics , Animals , Humans
3.
Proc Natl Acad Sci U S A ; 111(43): 15532-7, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313037

ABSTRACT

X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) is a tumor suppressor that is frequently inactivated in many human cancers. However, the molecular mechanism underlying its growth-inhibitory function remains largely unknown. Here, we report that XAF1 forms a positive feedback loop with p53 and acts as a molecular switch in p53-mediated cell-fate decisions favoring apoptosis over cell-cycle arrest. XAF1 binds directly to the N-terminal proline-rich domain of p53 and thus interferes with E3 ubiquitin ligase MDM2 binding and ubiquitination of p53. XAF1 stimulates homeodomain-interacting protein kinase 2 (HIPK2)-mediated Ser-46 phosphorylation of p53 by blocking E3 ubiquitin ligase Siah2 interaction with and ubiquitination of HIPK2. XAF1 also steps up the termination of p53-mediated cell-cycle arrest by activating zinc finger protein 313 (ZNF313), a p21(WAF1)-targeting ubiquitin E3 ligase. XAF1 interacts with p53, Siah2, and ZNF313 through the zinc finger domains 5, 6, and 7, respectively, and truncated XAF1 isoforms preferentially expressed in cancer cells fail to form a feedback loop with p53. Together, this study uncovers a novel role for XAF1 in p53 stress response, adding a new layer of complexity to the mechanisms by which p53 determines cell-fate decisions.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Biological , Neoplasm Proteins/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Isoforms/metabolism , Protein Stability/drug effects , Protein Structure, Tertiary , Proteolysis/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Remission Induction , Signal Transduction/drug effects , Tumor Suppressor Protein p53/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
4.
Med Image Anal ; 70: 101995, 2021 05.
Article in English | MEDLINE | ID: mdl-33640720

ABSTRACT

In this paper, we propose a novel microscopy image translation method for transforming a bright-field microscopy image into three different fluorescence images to observe the apoptosis, nuclei, and cytoplasm of cells, which visualize dead cells, nuclei of cells, and cytoplasm of cells, respectively. These biomarkers are commonly used in high-content drug screening to analyze drug response. The main contribution of the proposed work is the automatic generation of three fluorescence images from a conventional bright-field image; this can greatly reduce the time-consuming and laborious tissue preparation process and improve throughput of the screening process. Our proposed method uses only a single bright-field image and the corresponding fluorescence images as a set of image pairs for training an end-to-end deep convolutional neural network. By leveraging deep convolutional neural networks with a set of image pairs of bright-field and corresponding fluorescence images, our proposed method can produce synthetic fluorescence images comparable to real fluorescence microscopy images with high accuracy. Our proposed model uses multi-task learning with adversarial losses to generate more accurate and realistic microscopy images. We assess the efficacy of the proposed method using real bright-field and fluorescence microscopy image datasets from patient-driven samples of a glioblastoma, and validate the method's accuracy with various quality metrics including cell number correlation (CNC), peak signal-to-noise ratio (PSNR), structural similarity index measure (SSIM), cell viability correlation (CVC), error maps, and R2 correlation.


Subject(s)
Image Processing, Computer-Assisted , Neural Networks, Computer , Humans , Microscopy, Fluorescence , Signal-To-Noise Ratio
5.
Cancers (Basel) ; 13(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498427

ABSTRACT

(1) Background: Recent advances in precision oncology research rely on indicating specific genetic alterations associated with treatment sensitivity. Developing ex vivo systems to identify cancer patients who will respond to a specific drug remains important. (2) Methods: cells from 12 patients with glioblastoma were isolated, cultured, and subjected to high-content screening. Multi-parameter analyses assessed the c-Met level, cell viability, apoptosis, cell motility, and migration. A drug repurposing screen and large-scale drug sensitivity screening data across 59 cancer cell lines and patient-derived cells were obtained from 125 glioblastoma samples. (3) Results: High-content analysis of patient-derived cells provided robust and accurate drug responses to c-Met-targeted agents. Only the cells of one glioblastoma patient (PDC6) showed elevated c-Met level and high susceptibility to the c-Met inhibitors. Multi-parameter image analysis also reflected a decreased c-Met expression and reduced cell growth and motility by a c-Met-targeting antibody. In addition, a drug repurposing screen identified Abemaciclib as a distinct CDK4/6 inhibitor with a potent c-Met-inhibitory function. Consistent with this, we present large-scale drug sensitivity screening data showing that the Abemaciclib response correlates with the response to c-Met inhibitors. (4) Conclusions: Our study provides a new insight into high-content screening platforms supporting drug sensitivity prediction and novel therapeutics screening.

6.
Cell Cycle ; 19(13): 1590-1601, 2020 07.
Article in English | MEDLINE | ID: mdl-32423265

ABSTRACT

p97 has recently emerged as a therapeutic target for cancer due to its essential functions in protein homeostasis. CB-5083 is a first-in-class, potent and selective ATP-competitive p97 inhibitor that induces proteotoxic stress in cancer cells. Potential mechanisms regulating the sensitivity of cells to p97 inhibition remain poorly studied. Here, we demonstrate that Thrombospondin-1 (THBS1) is a CB-5083-upregulated gene that helps confer resistance of HCT116 cells to CB-5083. Our immunoblotting and immunofluorescence data showed that CB-5083 significantly increases the steady-state abundance of THBS1. Blockade of THBS1 induction sensitized cells to CB-5083-mediated growth inhibition. Suppression of THBS1 caused an increase of CB-5083-induced sub-G1 population and caspase 3/7 activity suggesting that its function is linked to the survival of cancer cells in response to p97 inhibition. Altogether our data provide new evidence that THBS1 is important for the susceptibility of cells to p97 inhibition.


Subject(s)
Colonic Neoplasms/metabolism , Indoles/pharmacology , Pyrimidines/pharmacology , Thrombospondin 1/metabolism , Valosin Containing Protein/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Thrombospondin 1/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Valosin Containing Protein/metabolism
7.
Biotechniques ; 68(4): 180-184, 2020 04.
Article in English | MEDLINE | ID: mdl-32040335

ABSTRACT

CRISPR-Cas9 has proven to be a versatile tool for the discovery of essential genetic elements involved in various disease states. CRISPR-assisted dense mutagenesis focused on therapeutically challenging protein complexes allows us to systematically perturb protein-coding sequences in situ and correlate them with functional readouts. Such perturbations can mimic targeting by therapeutics and serve as a foundation for the discovery of highly specific modulators. However, translation of such genomics data has been challenging due to the missing link for proteomics under the physiological state of the cell. We present a method based on cellular thermal shift assays to easily interrogate proteomic shifts generated by CRISPR-assisted dense mutagenesis, as well as a case focused on NuRD epigenetic complex.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Mutagenesis, Insertional/genetics , Proteome/genetics , Proteomics/methods , Cell Line , Humans , Proteome/analysis
8.
ACS Omega ; 5(40): 25798-25809, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33073104

ABSTRACT

c-Met, as a receptor expressed on the cell membrane, contributes to the growth and metastasis of tumors, as well as angiogenesis, mainly through the hepatocyte growth factor (HGF)/c-Met axis during tumor progression. Although several c-Met inhibitors, including small molecules and monoclonal antibody inhibitors, are currently being investigated, their clinical outcomes have not been promising. Development of an antibody-drug conjugate (ADC) against c-Met could be an attractive therapeutic strategy that would provide superior antitumor efficacy with broad-spectrum c-Met expression levels. In the present study, site-specific drug-conjugate technology was applied to develop an ADC using the human-mouse cross-reactive c-Met antibody and a prodrug pyrrolobenzodiazepine (PBD). The toxin payload was uniformly conjugated to the light-chain C-terminus of the native cIRCR201 antibody (drug-to-antibody ratio = 2), as confirmed using LC-MS. Using a high-throughput screening system, we found that cIRCR201-dPBD exhibited varying sensitivities depending on the expression levels of c-Met, and it induced receptor-mediated endocytosis and toxin-mediated apoptosis in 47 different cancer cell lines. cIRCR201-dPBD also showed significant antitumor activity on the MET-amplified cancer cells using in vivo xenograft models. Therefore, cIRCR201-dPBD could be a promising therapeutic strategy for tumors with c-Met expression.

9.
Sci Rep ; 10(1): 8096, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415084

ABSTRACT

Abnormal regulation of ß-catenin initiates an oncogenic program that serves as a main driver of many cancers. Albeit challenging, ß-catenin is an attractive drug target due to its role in maintenance of cancer stem cells and potential to eliminate cancer relapse. We have identified C2, a novel ß-catenin inhibitor, which is a small molecule that binds to a novel allosteric site on the surface of ß-catenin. C2 selectively inhibits ß-catenin, lowers its cellular load and significantly reduces viability of ß-catenin-driven cancer cells. Through direct binding to ß-catenin, C2 renders the target inactive that eventually activates proteasome system for its removal. Here we report a novel pharmacologic approach for selective inhibition of ß-catenin via targeting a cryptic allosteric modulation site. Our findings may provide a new perspective for therapeutic targeting of ß-catenin.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Allosteric Regulation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Genome Med ; 12(1): 17, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32070411

ABSTRACT

BACKGROUND: Gastric cancer is among the most lethal human malignancies. Previous studies have identified molecular aberrations that constitute dynamic biological networks and genomic complexities of gastric tumors. However, the clinical translation of molecular-guided targeted therapy is hampered by challenges. Notably, solid tumors often harbor multiple genetic alterations, complicating the development of effective treatments. METHODS: To address such challenges, we established a comprehensive dataset of molecularly annotated patient derivatives coupled with pharmacological profiles for 60 targeted agents to explore dynamic pharmacogenomic interactions in gastric cancers. RESULTS: We identified lineage-specific drug sensitivities based on histopathological and molecular subclassification, including substantial sensitivities toward VEGFR and EGFR inhibition therapies in diffuse- and signet ring-type gastric tumors, respectively. We identified potential therapeutic opportunities for WNT pathway inhibitors in ALK-mutant tumors, a significant association between PIK3CA-E542K mutation and AZD5363 response, and transcriptome expression of RNF11 as a potential predictor of response to gefitinib. CONCLUSIONS: Collectively, our results demonstrate the feasibility of drug screening combined with tumor molecular characterization to facilitate personalized therapeutic regimens for gastric tumors.


Subject(s)
Drug Resistance, Neoplasm , Pharmacogenomic Variants , Stomach Neoplasms/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gefitinib/pharmacology , Gefitinib/therapeutic use , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/genetics , Stomach Neoplasms/drug therapy , Transcriptome , Tumor Cells, Cultured , Wnt Signaling Pathway/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL