Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell ; 185(1): 169-183.e19, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34963055

ABSTRACT

Non-small cell lung cancers (NSCLCs) harboring KEAP1 mutations are often resistant to immunotherapy. Here, we show that KEAP1 targets EMSY for ubiquitin-mediated degradation to regulate homologous recombination repair (HRR) and anti-tumor immunity. Loss of KEAP1 in NSCLC induces stabilization of EMSY, producing a BRCAness phenotype, i.e., HRR defects and sensitivity to PARP inhibitors. Defective HRR contributes to a high tumor mutational burden that, in turn, is expected to prompt an innate immune response. Notably, EMSY accumulation suppresses the type I interferon response and impairs innate immune signaling, fostering cancer immune evasion. Activation of the type I interferon response in the tumor microenvironment using a STING agonist results in the engagement of innate and adaptive immune signaling and impairs the growth of KEAP1-mutant tumors. Our results suggest that targeting PARP and STING pathways, individually or in combination, represents a therapeutic strategy in NSCLC patients harboring alterations in KEAP1.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Interferon Type I/metabolism , Lung Neoplasms/immunology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Recombinational DNA Repair/genetics , Repressor Proteins/metabolism , Tumor Escape/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunity, Innate/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
3.
Cancer Res ; 80(8): 1693-1706, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32054769

ABSTRACT

A significant therapeutic challenge for patients with cancer is resistance to chemotherapies such as taxanes. Overexpression of LIN9, a transcriptional regulator of cell-cycle progression, occurs in 65% of patients with triple-negative breast cancer (TNBC), a disease commonly treated with these drugs. Here, we report that LIN9 is further elevated with acquisition of taxane resistance. Inhibiting LIN9 genetically or by suppressing its expression with a global BET inhibitor restored taxane sensitivity by inducing mitotic progression errors and apoptosis. While sustained LIN9 is necessary to maintain taxane resistance, there are no inhibitors that directly repress its function. Hence, we sought to discover a druggable downstream transcriptional target of LIN9. Using a computational approach, we identified NIMA-related kinase 2 (NEK2), a regulator of centrosome separation that is also elevated in taxane-resistant cells. High expression of NEK2 was predictive of low survival rates in patients who had residual disease following treatment with taxanes plus an anthracycline, suggesting a role for this kinase in modulating taxane sensitivity. Like LIN9, genetic or pharmacologic blockade of NEK2 activity in the presence of paclitaxel synergistically induced mitotic abnormalities in nearly 100% of cells and completely restored sensitivity to paclitaxel, in vitro. In addition, suppressing NEK2 activity with two distinct small molecules potentiated taxane response in multiple in vivo models of TNBC, including a patient-derived xenograft, without inducing toxicity. These data demonstrate that the LIN9/NEK2 pathway is a therapeutically targetable mediator of taxane resistance that can be leveraged to improve response to this core chemotherapy. SIGNIFICANCE: Resistance to chemotherapy is a major hurdle for treating patients with cancer. Combining NEK2 inhibitors with taxanes may be a viable approach for improving patient outcomes by enhancing mitotic defects induced by taxanes alone.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Mitosis/drug effects , NIMA-Related Kinases/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Paclitaxel/pharmacology , Taxoids/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Suppressor Proteins/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cellular Senescence , Centrosome/enzymology , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Heterografts , Humans , Mitosis/genetics , NIMA-Related Kinases/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Paclitaxel/administration & dosage , Survival Rate , Taxoids/administration & dosage , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Tumor Stem Cell Assay , Tumor Suppressor Proteins/metabolism , Up-Regulation
4.
J Biol Chem ; 294(3): 875-886, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30482844

ABSTRACT

Inhibitors of bromodomain and extra-terminal proteins (BETi) suppress oncogenic gene expression and have been shown to be efficacious in many in vitro and murine models of cancer, including triple-negative breast cancer (TNBC), a highly aggressive disease. However, in most cancer models, responses to BETi can be highly variable. We previously reported that TNBC cells either undergo senescence or apoptosis in response to BETi, but the specific mechanisms dictating these two cell fates remain unknown. Using six human TNBC cell lines, we show that the terminal response of TNBC cells to BETi is dictated by the intrinsic expression levels of the anti-apoptotic protein B-cell lymphoma-extra large (BCL-xL). BCL-xL levels were higher in cell lines that senesce in response to BETi compared with lines that primarily die in response to these drugs. Moreover, BCL-xL expression was further reduced in cells that undergo BETi-mediated apoptosis. Forced BCL-xL overexpression in cells that normally undergo apoptosis following BETi treatment shifted them to senescence without affecting the reported mechanism of action of BETi in TNBC, that is, mitotic catastrophe. Most importantly, pharmacological or genetic inhibition of BCL-xL induced apoptosis in response to BETi, and inhibiting BCL-xL, even after BETi-induced senescence had already occurred, still induced cell death. These results indicate that BCL-xL provides a senescent cell death-inducing or senolytic target that may be exploited to improve therapeutic outcomes of TNBC in response to BETi. They also suggest that the basal levels of BCL-xL should be predictive of tumor responses to BETi in current clinical trials.


Subject(s)
Apoptosis , Cellular Senescence , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , bcl-X Protein/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Female , Humans , Nuclear Proteins/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , bcl-X Protein/genetics
5.
Pharmacol Res ; 129: 156-176, 2018 03.
Article in English | MEDLINE | ID: mdl-29154989

ABSTRACT

Breast cancer is a collection of distinct tumor subtypes that are driven by unique gene expression profiles. These transcriptomes are controlled by various epigenetic marks that dictate which genes are expressed and suppressed. During carcinogenesis, extensive restructuring of the epigenome occurs, including aberrant acetylation, alteration of methylation patterns, and accumulation of epigenetic readers at oncogenes. As epigenetic alterations are reversible, epigenome-modulating drugs could provide a mechanism to silence numerous oncogenes simultaneously. Here, we review the impact of inhibitors of the Bromodomain and Extraterminal (BET) family of epigenetic readers in breast cancer. These agents, including the prototypical BET inhibitor JQ1, have been shown to suppress a variety of oncogenic pathways while inducing minimal, if any, toxicity in models of several subtypes of breast cancer. BET inhibitors also synergize with multiple approved anti-cancer drugs, providing a greater response in breast cancer cell lines and mouse models than either single agent. The combined findings of the studies discussed here provide an excellent rationale for the continued investigation of the utility of BET inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Female , Humans , Molecular Targeted Therapy , Proteins/chemistry , Proteins/metabolism
6.
Oncoscience ; 4(9-10): 128-130, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29142903
7.
Cancer Res ; 77(19): 5395-5408, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28807940

ABSTRACT

Triple-negative breast cancers (TNBC) are highly aggressive, lack FDA-approved targeted therapies, and frequently recur, making the discovery of novel therapeutic targets for this disease imperative. Our previous analysis of the molecular mechanisms of action of bromodomain and extraterminal protein inhibitors (BETi) in TNBC revealed these drugs cause multinucleation, indicating BET proteins are essential for efficient mitosis and cytokinesis. Here, using live cell imaging, we show that BET inhibition prolonged mitotic progression and induced mitotic cell death, both of which are indicative of mitotic catastrophe. Mechanistically, the mitosis regulator LIN9 was a direct target of BET proteins that mediated the effects of BET proteins on mitosis in TNBC. Although BETi have been proposed to function by dismantling super-enhancers (SE), the LIN9 gene lacks an SE but was amplified or overexpressed in the majority of TNBCs. In addition, its mRNA expression predicted poor outcome across breast cancer subtypes. Together, these results provide a mechanism for cancer selectivity of BETi that extends beyond modulation of SE-associated genes and suggest that cancers dependent upon LIN9 overexpression may be particularly vulnerable to BETi. Cancer Res; 77(19); 5395-408. ©2017 AACR.


Subject(s)
Mitosis/drug effects , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Biol Chem ; 291(45): 23756-23768, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27650498

ABSTRACT

Bromodomain and extraterminal (BET) proteins are epigenetic "readers" that recognize acetylated histones and mark areas of the genome for transcription. BRD4, a BET family member protein, has been implicated in a number of types of cancer, and BET protein inhibitors (BETi) are efficacious in many preclinical cancer models. However, the drivers of response to BETi vary depending on tumor type, and little is known regarding the target genes conveying BETi activity in triple-negative breast cancer (TNBC). Here, we show that BETi repress growth of multiple in vitro and in vivo models of TNBC by inducing two terminal responses: apoptosis and senescence. Unlike in other cancers, response to BETi in TNBC is not dependent upon suppression of MYC Instead, both end points are preceded by the appearance of polyploid cells caused by the suppression of Aurora kinases A and B (AURKA/B), which are critical mediators of mitosis. In addition, AURKA/B inhibitors phenocopy the effects of BETi. These results indicate that Aurora kinases play an important role in the growth suppressive activity of BETi in TNBC. Elucidating the mechanism of response to BETi in TNBC should 1) facilitate the prediction of how distinct TNBC tumors will respond to BETi and 2) inform the rational design of drug combination therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Breast/drug effects , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Transcription Factors/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinase A/metabolism , Aurora Kinase B/metabolism , Breast/metabolism , Breast/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...