Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neuro Oncol ; 21(9): 1150-1163, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31111916

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is one of the most frequent malignant brain tumors of children, and a large set of these tumors is characterized by aberrant activation of the sonic hedgehog (SHH) pathway. While some tumors initially respond to inhibition of the SHH pathway component Smoothened (SMO), tumors ultimately recur due to downstream resistance mechanisms, indicating a need for novel therapeutic options. METHODS: Here we performed a targeted small-molecule screen on a stable, SHH-dependent murine MB cell line (SMB21). Comprehensive isotype profiling of histone deacetylase (HDAC) inhibitors was performed, and effects of HDAC inhibition were evaluated in cell lines both sensitive and resistant to SMO inhibition. Lastly, distinct mouse models of SHH MB were used to demonstrate pharmacologic efficacy in vivo. RESULTS: A subset of the HDAC inhibitors tested significantly inhibit tumor growth of SMB21 cells by preventing SHH pathway activation. Isotype profiling of HDAC inhibitors, together with genetic approaches suggested that concerted inhibition of multiple class I HDACs is necessary to achieve pathway inhibition. Of note, class I HDAC inhibitors were also efficacious in suppressing growth of diverse SMO inhibitor‒resistant clones of SMB21 cells. Finally, we show that the novel HDAC inhibitor quisinostat targets multiple class I HDACs, is well tolerated in mouse models, and robustly inhibits growth of SHH MB cells in vivo as well as in vitro. CONCLUSIONS: Our data provide strong evidence that quisinostat or other class I HDAC inhibitors might be therapeutically useful for patients with SHH MB, including those resistant to SMO inhibition.


Subject(s)
Cell Survival/drug effects , Cerebellar Neoplasms/drug therapy , Hedgehog Proteins/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Medulloblastoma/drug therapy , Anilides , Animals , Biphenyl Compounds , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/metabolism , High-Throughput Screening Assays , Inhibitory Concentration 50 , Medulloblastoma/metabolism , Mice , Proteins/genetics , Pyridines , Repressor Proteins/genetics , Signal Transduction , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism
2.
Cancer Discov ; 7(12): 1436-1449, 2017 12.
Article in English | MEDLINE | ID: mdl-28923910

ABSTRACT

Drug resistance poses a great challenge to targeted cancer therapies. In Hedgehog pathway-dependent cancers, the scope of mechanisms enabling resistance to SMO inhibitors is not known. Here, we performed a transposon mutagenesis screen in medulloblastoma and identified multiple modes of resistance. Surprisingly, mutations in ciliogenesis genes represent a frequent cause of resistance, and patient datasets indicate that cilia loss constitutes a clinically relevant category of resistance. Conventionally, primary cilia are thought to enable oncogenic Hedgehog signaling. Paradoxically, we find that cilia loss protects tumor cells from susceptibility to SMO inhibitors and maintains a "persister" state that depends on continuous low output of the Hedgehog program. Persister cells can serve as a reservoir for further tumor evolution, as additional alterations synergize with cilia loss to generate aggressive recurrent tumors. Together, our findings reveal patterns of resistance and provide mechanistic insights for the role of cilia in tumor evolution and drug resistance.Significance: Using a transposon screen and clinical datasets, we identified mutations in ciliogenesis genes as a new class of resistance to SMO inhibitors. Mechanistically, cilia-mutant tumors can either grow slowly in a "persister" state or evolve and progress rapidly in an "aggressive" state. Cancer Discov; 7(12); 1436-49. ©2017 AACR.See related commentary by Goranci-Buzhala et al., p. 1374This article is highlighted in the In This Issue feature, p. 1355.


Subject(s)
Cilia/genetics , Hedgehog Proteins/genetics , Retroelements/genetics , Smoothened Receptor/antagonists & inhibitors , Animals , Humans , Mice , Signal Transduction
3.
Dev Cell ; 38(4): 333-44, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27554855

ABSTRACT

The Hedgehog (Hh) signaling pathway governs complex developmental processes, including proliferation and patterning within diverse tissues. These activities rely on a tightly regulated transduction system that converts graded Hh input signals into specific levels of pathway activity. Uncontrolled activation of Hh signaling drives tumor initiation and maintenance. However, recent entry of pathway-specific inhibitors into the clinic reveals mixed patient responses and thus prompts further exploration of pathway activation and inhibition. In this review, we share emerging insights into regulated and oncogenic Hh signaling, supplemented with updates on the development and use of Hh pathway-targeted therapies.


Subject(s)
Carcinoma, Basal Cell/pathology , Cell Transformation, Neoplastic/pathology , Hedgehog Proteins/metabolism , Medulloblastoma/pathology , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Humans , Mice
4.
Cancer Res ; 75(17): 3623-35, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26130651

ABSTRACT

Aberrant Shh signaling promotes tumor growth in diverse cancers. The importance of Shh signaling is particularly evident in medulloblastoma and basal cell carcinoma (BCC), where inhibitors targeting the Shh pathway component Smoothened (Smo) show great therapeutic promise. However, the emergence of drug resistance limits long-term efficacy, and the mechanisms of resistance remain poorly understood. Using new medulloblastoma models, we identify two distinct paradigms of resistance to Smo inhibition. Sufu mutations lead to maintenance of the Shh pathway in the presence of Smo inhibitors. Alternatively activation of the RAS-MAPK pathway circumvents Shh pathway dependency, drives tumor growth, and enhances metastatic behavior. Strikingly, in BCC patients treated with Smo inhibitor, squamous cell cancers with RAS/MAPK activation emerged from the antecedent BCC tumors. Together, these findings reveal a critical role of the RAS-MAPK pathway in drug resistance and tumor evolution of Shh pathway-dependent tumors.


Subject(s)
Carcinoma, Basal Cell/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Receptors, G-Protein-Coupled/biosynthesis , ras Proteins/genetics , Anilides/administration & dosage , Animals , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Smoothened Receptor , Xenograft Model Antitumor Assays , ras Proteins/biosynthesis
6.
Nat Med ; 19(11): 1518-23, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24076665

ABSTRACT

In glioblastoma, phosphatidylinositol 3-kinase (PI3K) signaling is frequently activated by loss of the tumor suppressor phosphatase and tensin homolog (PTEN). However, it is not known whether inhibiting PI3K represents a selective and effective approach for treatment. We interrogated large databases and found that sonic hedgehog (SHH) signaling is activated in PTEN-deficient glioblastoma. We demonstrate that the SHH and PI3K pathways synergize to promote tumor growth and viability in human PTEN-deficient glioblastomas. A combination of PI3K and SHH signaling inhibitors not only suppressed the activation of both pathways but also abrogated S6 kinase (S6K) signaling. Accordingly, targeting both pathways simultaneously resulted in mitotic catastrophe and tumor apoptosis and markedly reduced the growth of PTEN-deficient glioblastomas in vitro and in vivo. The drugs tested here appear to be safe in humans; therefore, this combination may provide a new targeted treatment for glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Hedgehog Proteins/metabolism , PTEN Phosphohydrolase/deficiency , Phosphatidylinositol 3-Kinases/metabolism , Aminopyridines/administration & dosage , Animals , Biphenyl Compounds/administration & dosage , Brain Neoplasms/genetics , Cell Line, Tumor , Enzyme Inhibitors/administration & dosage , Glioblastoma/genetics , Hedgehog Proteins/antagonists & inhibitors , Humans , Mice , Mice, Nude , Morpholines/administration & dosage , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Pyridines/administration & dosage , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...