Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
Clin Cancer Res ; 28(20): 4551-4564, 2022 10 14.
Article En | MEDLINE | ID: mdl-35920742

PURPOSE: The addition of immune checkpoint blockade (ICB) to platinum/etoposide chemotherapy changed the standard of care for small cell lung cancer (SCLC) treatment. However, ICB addition only modestly improved clinical outcomes, likely reflecting the high prevalence of an immunologically "cold" tumor microenvironment in SCLC, despite high mutational burden. Nevertheless, some patients clearly benefit from ICB and recent reports have associated clinical responses to ICB in SCLC with (i) decreased neuroendocrine characteristics and (ii) activation of NOTCH signaling. We previously showed that inhibition of the lysine-specific demethylase 1a (LSD1) demethylase activates NOTCH and suppresses neuroendocrine features of SCLC, leading us to investigate whether LSD1 inhibition would enhance the response to PD-1 inhibition in SCLC. EXPERIMENTAL DESIGN: We employed a syngeneic immunocompetent model of SCLC, derived from a genetically engineered mouse model harboring Rb1/Trp53 inactivation, to investigate combining the LSD1 inhibitor bomedemstat with anti-PD-1 therapy. In vivo experiments were complemented by cell-based studies in murine and human models. RESULTS: Bomedemstat potentiated responses to PD-1 inhibition in a syngeneic model of SCLC, resulting in increased CD8+ T-cell infiltration and strong tumor growth inhibition. Bomedemstat increased MHC class I expression in mouse SCLC tumor cells in vivo and augmented MHC-I induction by IFNγ and increased killing by tumor-specific T cells in cell culture. CONCLUSIONS: LSD1 inhibition increased MHC-I expression and enhanced responses to PD-1 inhibition in vivo, supporting a new clinical trial to combine bomedemstat with standard-of-care PD-1 axis inhibition in SCLC.


Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Cell Death , Enzyme Inhibitors/therapeutic use , Etoposide/therapeutic use , Histone Demethylases/metabolism , Humans , Immune Checkpoint Inhibitors , Lung Neoplasms/pathology , Lysine/therapeutic use , Mice , Platinum/therapeutic use , Small Cell Lung Carcinoma/pathology , Tumor Microenvironment
2.
Genes Dev ; 35(11-12): 870-887, 2021 06.
Article En | MEDLINE | ID: mdl-34016692

Small cell lung carcinoma (SCLC) is among the most lethal of all solid tumor malignancies. In an effort to identify novel therapeutic approaches for this recalcitrant cancer type, we applied genome-scale CRISPR/Cas9 inactivation screens to cell lines that we derived from a murine model of SCLC. SCLC cells were particularly sensitive to the deletion of NEDD8 and other neddylation pathway genes. Genetic suppression or pharmacological inhibition of this pathway using MLN4924 caused cell death not only in mouse SCLC cell lines but also in patient-derived xenograft (PDX) models of pulmonary and extrapulmonary small cell carcinoma treated ex vivo or in vivo. A subset of PDX models were exceptionally sensitive to neddylation inhibition. Neddylation inhibition suppressed expression of major regulators of neuroendocrine cell state such as INSM1 and ASCL1, which a subset of SCLC rely upon for cell proliferation and survival. To identify potential mechanisms of resistance to neddylation inhibition, we performed a genome-scale CRISPR/Cas9 suppressor screen. Deletion of components of the COP9 signalosome strongly mitigated the effects of neddylation inhibition in small cell carcinoma, including the ability of MLN4924 to suppress neuroendocrine transcriptional program expression. This work identifies neddylation as a regulator of neuroendocrine cell state and potential therapeutic target for small cell carcinomas.


Carcinoma, Small Cell/therapy , Cyclopentanes , Lung Neoplasms/therapy , NEDD8 Protein/metabolism , Pyrimidines , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , COP9 Signalosome Complex/genetics , Carcinoma, Small Cell/physiopathology , Cell Death/drug effects , Cell Line, Tumor , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lung Neoplasms/physiopathology , Mice , NEDD8 Protein/genetics , Neuroendocrine Cells/cytology , Neuroendocrine Cells/drug effects , Proteins/genetics , Proteins/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Repressor Proteins/genetics , Sequence Deletion
3.
Genes Dev ; 34(17-18): 1210-1226, 2020 09 01.
Article En | MEDLINE | ID: mdl-32820040

Small cell lung cancer (SCLC) is an aggressive neuroendocrine cancer characterized by initial chemosensitivity followed by emergence of chemoresistant disease. To study roles for MYCN amplification in SCLC progression and chemoresistance, we developed a genetically engineered mouse model of MYCN-overexpressing SCLC. In treatment-naïve mice, MYCN overexpression promoted cell cycle progression, suppressed infiltration of cytotoxic T cells, and accelerated SCLC. MYCN overexpression also suppressed response to cisplatin-etoposide chemotherapy, with similar findings made upon MYCL overexpression. We extended these data to genetically perturb chemosensitive patient-derived xenograft (PDX) models of SCLC. In chemosensitive PDX models, overexpression of either MYCN or MYCL also conferred a switch to chemoresistance. To identify therapeutic strategies for MYCN-overexpressing SCLC, we performed a genome-scale CRISPR-Cas9 sgRNA screen. We identified the deubiquitinase USP7 as a MYCN-associated synthetic vulnerability. Pharmacological inhibition of USP7 resensitized chemoresistant MYCN-overexpressing PDX models to chemotherapy in vivo. Our findings show that MYCN overexpression drives SCLC chemoresistance and provide a therapeutic strategy to restore chemosensitivity.


Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , N-Myc Proto-Oncogene Protein/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Heterografts , Humans , Lung Neoplasms/enzymology , Mice , N-Myc Proto-Oncogene Protein/genetics , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/genetics
4.
Cell Rep ; 3(5): 1465-75, 2013 May 30.
Article En | MEDLINE | ID: mdl-23684613

Lipid droplets (LDs) are the major fat storage organelles in eukaryotic cells, but how their size is regulated is unknown. Using genetic screens in C. elegans for LD morphology defects in intestinal cells, we found that mutations in atlastin, a GTPase required for homotypic fusion of endoplasmic reticulum (ER) membranes, cause not only ER morphology defects, but also a reduction in LD size. Similar results were obtained after depletion of atlastin or expression of a dominant-negative mutant, whereas overexpression of atlastin had the opposite effect. Atlastin depletion in Drosophila fat bodies also reduced LD size and decreased triglycerides in whole animals, sensitizing them to starvation. In mammalian cells, co-overexpression of atlastin-1 and REEP1, a paralog of the ER tubule-shaping protein DP1/REEP5, generates large LDs. The effect of atlastin-1 on LD size correlates with its activity to promote membrane fusion in vitro. Our results indicate that atlastin-mediated fusion of ER membranes is important for LD size regulation.


Caenorhabditis elegans Proteins/metabolism , Cytoplasmic Vesicles/chemistry , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Membrane Proteins/metabolism , Animals , COS Cells , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Chlorocebus aethiops , Cytoplasmic Vesicles/metabolism , Drosophila/metabolism , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/genetics , GTP-Binding Proteins/genetics , Humans , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation , RNA Interference , RNA, Small Interfering/metabolism
...