Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Int J Mol Sci ; 18(6)2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28621712

ABSTRACT

Eukaryotes use autophagy as a mechanism for maintaining cellular homeostasis by degrading and recycling organelles and proteins. This process assists in the proliferation and survival of advanced cancers. There is mounting preclinical evidence that targeting autophagy can enhance the efficacy of many cancer therapies. Hydroxychloroquine (HCQ) is the only clinically-approved autophagy inhibitor, and this systematic review focuses on HCQ use in cancer clinical trials. Preclinical trials have shown that HCQ alone and in combination therapy leads to enhancement of tumor shrinkage. This has provided the base for multiple ongoing clinical trials involving HCQ alone and in combination with other treatments. However, due to its potency, there is still a need for more potent and specific autophagy inhibitors. There are multiple autophagy inhibitors in the pre-clinical stage at various stages of development. Additional studies on the mechanism of HCQ and other autophagy inhibitors are still required to answer questions surrounding how these agents will eventually be used in the clinic.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Hydroxychloroquine/therapeutic use , Neoplasms/drug therapy , Animals , Antimalarials/therapeutic use , Clinical Trials as Topic , Humans , Neoplasms/pathology
2.
Cancer Discov ; 9(3): 396-415, 2019 03.
Article in English | MEDLINE | ID: mdl-30563872

ABSTRACT

Resistance to BRAF and MEK inhibitors (BRAFi + MEKi) in BRAF-mutant tumors occurs through heterogeneous mechanisms, including ERK reactivation and autophagy. Little is known about the mechanisms by which ERK reactivation or autophagy is induced by BRAFi + MEKi. Here, we report that in BRAF-mutant melanoma cells, BRAFi + MEKi induced SEC61-dependent endoplasmic reticulum (ER) translocation of the MAPK pathway via GRP78 and KSR2. Inhibition of ER translocation prevented ERK reactivation and autophagy. Following ER translocation, ERK exited the ER and was rephosphorylated by PERK. Reactivated ERK phosphorylated ATF4, which activated cytoprotective autophagy. Upregulation of GRP78 and phosphorylation of ATF4 were detected in tumors of patients resistant to BRAFi + MEKi. ER translocation of the MAPK pathway was demonstrated in therapy-resistant patient-derived xenografts. Expression of a dominant-negative ATF4 mutant conferred sensitivity to BRAFi + MEKi in vivo. This mechanism reconciles two major targeted therapy resistance pathways and identifies druggable targets, whose inhibition would likely enhance the response to BRAFi + MEKi. SIGNIFICANCE: ERK reactivation and autophagy are considered distinct resistance pathways to BRAF + MEK inhibition (BRAFi + MEKi) in BRAF V600E cancers. Here, we report BRAFi + MEKi-induced ER translocation of the MAPK pathway is necessary for ERK reactivation, which drives autophagy. The ER translocation mechanism is a major druggable driver of resistance to targeted therapy.This article is highlighted in the In This Issue feature, p. 305.


Subject(s)
Endoplasmic Reticulum/metabolism , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Animals , Autophagy , Cell Line, Tumor , Drug Resistance, Neoplasm , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Protein Transport , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Discov ; 9(2): 220-229, 2019 02.
Article in English | MEDLINE | ID: mdl-30442709

ABSTRACT

Clinical trials repurposing lysosomotropic chloroquine (CQ) derivatives as autophagy inhibitors in cancer demonstrate encouraging results, but the underlying mechanism of action remains unknown. Here, we report a novel dimeric CQ (DC661) capable of deacidifying the lysosome and inhibiting autophagy significantly better than hydroxychloroquine (HCQ). Using an in situ photoaffinity pulldown strategy, we identified palmitoyl-protein thioesterase 1 (PPT1) as a molecular target shared across monomeric and dimeric CQ derivatives. HCQ and Lys05 also bound to and inhibited PPT1 activity, but only DC661 maintained activity in acidic media. Knockout of PPT1 in cancer cells using CRISPR/Cas9 editing abrogates autophagy modulation and cytotoxicity of CQ derivatives, and results in significant impairment of tumor growth similar to that observed with DC661. Elevated expression of PPT1 in tumors correlates with poor survival in patients in a variety of cancers. Thus, PPT1 represents a new target in cancer that can be inhibited with CQ derivatives. SIGNIFICANCE: This study identifies PPT1 as the previously unknown lysosomal molecular target of monomeric and dimeric CQ derivatives. Genetic suppression of PPT1 impairs tumor growth, and PPT1 levels are elevated in cancer and associated with poor survival. These findings provide a strong rationale for targeting PPT1 in cancer. This article is highlighted in the In This Issue feature, p. 151.


Subject(s)
Antimalarials/pharmacology , Biomarkers, Tumor/metabolism , Chloroquine/pharmacology , Membrane Proteins/metabolism , Neoplasms/pathology , Thiolester Hydrolases/metabolism , Aminoquinolines/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Membrane Proteins/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Polyamines/pharmacology , Prognosis , Survival Rate , Thiolester Hydrolases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Discov ; 7(11): 1266-1283, 2017 11.
Article in English | MEDLINE | ID: mdl-28899863

ABSTRACT

Lysosomes serve dual roles in cancer metabolism, executing catabolic programs (i.e., autophagy and macropinocytosis) while promoting mTORC1-dependent anabolism. Antimalarial compounds such as chloroquine or quinacrine have been used as lysosomal inhibitors, but fail to inhibit mTOR signaling. Further, the molecular target of these agents has not been identified. We report a screen of novel dimeric antimalarials that identifies dimeric quinacrines (DQ) as potent anticancer compounds, which concurrently inhibit mTOR and autophagy. Central nitrogen methylation of the DQ linker enhances lysosomal localization and potency. An in situ photoaffinity pulldown identified palmitoyl-protein thioesterase 1 (PPT1) as the molecular target of DQ661. PPT1 inhibition concurrently impairs mTOR and lysosomal catabolism through the rapid accumulation of palmitoylated proteins. DQ661 inhibits the in vivo tumor growth of melanoma, pancreatic cancer, and colorectal cancer mouse models and can be safely combined with chemotherapy. Thus, lysosome-directed PPT1 inhibitors represent a new approach to concurrently targeting mTORC1 and lysosomal catabolism in cancer.Significance: This study identifies chemical features of dimeric compounds that increase their lysosomal specificity, and a new molecular target for these compounds, reclassifying these compounds as targeted therapies. Targeting PPT1 blocks mTOR signaling in a manner distinct from catalytic inhibitors, while concurrently inhibiting autophagy, thereby providing a new strategy for cancer therapy. Cancer Discov; 7(11); 1266-83. ©2017 AACR.See related commentary by Towers and Thorburn, p. 1218This article is highlighted in the In This Issue feature, p. 1201.


Subject(s)
Lysosomes/drug effects , Melanoma/drug therapy , Membrane Proteins/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Thiolester Hydrolases/antagonists & inhibitors , Animals , Antimalarials/administration & dosage , Antineoplastic Agents/administration & dosage , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/administration & dosage , Humans , Lysosomes/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Proteolysis/drug effects , Signal Transduction/drug effects , Thiolester Hydrolases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL