Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nature ; 595(7869): 730-734, 2021 07.
Article in English | MEDLINE | ID: mdl-34290403

ABSTRACT

Hepatocellular carcinoma (HCC)-the most common form of liver cancer-is an aggressive malignancy with few effective treatment options1. Lenvatinib is a small-molecule inhibitor of multiple receptor tyrosine kinases that is used for the treatment of patients with advanced HCC, but this drug has only limited clinical benefit2. Here, using a kinome-centred CRISPR-Cas9 genetic screen, we show that inhibition of epidermal growth factor receptor (EGFR) is synthetic lethal with lenvatinib in liver cancer. The combination of the EGFR inhibitor gefitinib and lenvatinib displays potent anti-proliferative effects in vitro in liver cancer cell lines that express EGFR and in vivo in xenografted liver cancer cell lines, immunocompetent mouse models and patient-derived HCC tumours in mice. Mechanistically, inhibition of fibroblast growth factor receptor (FGFR)  by lenvatinib treatment leads to feedback activation of the EGFR-PAK2-ERK5 signalling axis, which is blocked by EGFR inhibition. Treatment of 12 patients with advanced HCC who were unresponsive to lenvatinib treatment with the combination of lenvatinib plus gefitinib (trial identifier NCT04642547) resulted in meaningful clinical responses. The combination therapy identified here may represent a promising strategy for the approximately 50% of patients with advanced HCC who have high levels of EGFR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gefitinib/pharmacology , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Fibroblast Growth Factor , Signal Transduction , Xenograft Model Antitumor Assays
2.
Cancer Cell ; 12(4): 395-402, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936563

ABSTRACT

A large-scale RNA interference screen to discover genes involved in trastuzumab resistance in breast cancer identified only PTEN as a modulator of drug sensitivity. Oncogenic mutants of PIK3CA (activator of the same pathway and frequently mutated in breast cancer) also conferred resistance to trastuzumab in cell culture. In a cohort of 55 breast cancer patients, activation of the PI3K pathway, as judged by the presence of oncogenic PIK3CA mutations or low PTEN expression, was associated with poor prognosis after trastuzumab therapy, and the combined analysis of PTEN and PIK3CA identified twice as many patients at increased risk for progression compared to PTEN alone. Thus, assessment of PI3K pathway activation may provide a biomarker to identify patients unlikely to respond to trastuzumab-based therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Cohort Studies , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Middle Aged , Mutation , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Patient Selection , Phosphatidylinositol 3-Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Transduction, Genetic , Trastuzumab , Treatment Outcome
3.
Mol Cancer Ther ; 21(12): 1862-1874, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36198031

ABSTRACT

Low-grade serous ovarian cancer (LGSOC) is a rare subtype of epithelial ovarian cancer with high fatality rates in advanced stages due to its chemoresistant properties. LGSOC is characterized by activation of MAPK signaling, and recent clinical trials indicate that the MEK inhibitor (MEKi) trametinib may be a good treatment option for a subset of patients. Understanding MEKi-resistance mechanisms and subsequent identification of rational drug combinations to suppress resistance may greatly improve LGSOC treatment strategies. Both gain-of-function and loss-of-function CRISPR-Cas9 genome-wide libraries were used to screen LGSOC cell lines to identify genes that modulate the response to MEKi. Overexpression of MAML2 and loss of MAP3K1 were identified, both leading to overexpression of the NOTCH target HES1, which has a causal role in this process as its knockdown reversed MEKi resistance. Interestingly, increased HES1 expression was also observed in selected spontaneous trametinib-resistant clones, next to activating MAP2K1 (MEK1) mutations. Subsequent trametinib synthetic lethality screens identified SHOC2 downregulation as being synthetic lethal with MEKis. Targeting SHOC2 with pan-RAF inhibitors (pan-RAFis) in combination with MEKi was effective in parental LGSOC cell lines, in MEKi-resistant derivatives, in primary ascites cultures from patients with LGSOC, and in LGSOC (cell line-derived and patient-derived) xenograft mouse models. We found that the combination of pan-RAFi with MEKi downregulated HES1 levels in trametinib-resistant cells, providing an explanation for the synergy that was observed. Combining MEKis with pan-RAFis may provide a promising treatment strategy for patients with LGSOC, which warrants further clinical validation.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Peritoneal Neoplasms , Female , Humans , Mice , Animals , Drug Resistance, Neoplasm/genetics , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Signal Transduction , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Peritoneal Neoplasms/drug therapy , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/pharmacology
4.
Nature ; 428(6981): 431-7, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-15042092

ABSTRACT

RNA interference (RNAi) is a powerful new tool with which to perform loss-of-function genetic screens in lower organisms and can greatly facilitate the identification of components of cellular signalling pathways. In mammalian cells, such screens have been hampered by a lack of suitable tools that can be used on a large scale. We and others have recently developed expression vectors to direct the synthesis of short hairpin RNAs (shRNAs) that act as short interfering RNA (siRNA)-like molecules to stably suppress gene expression. Here we report the construction of a set of retroviral vectors encoding 23,742 distinct shRNAs, which target 7,914 different human genes for suppression. We use this RNAi library in human cells to identify one known and five new modulators of p53-dependent proliferation arrest. Suppression of these genes confers resistance to both p53-dependent and p19ARF-dependent proliferation arrest, and abolishes a DNA-damage-induced G1 cell-cycle arrest. Furthermore, we describe siRNA bar-code screens to rapidly identify individual siRNA vectors associated with a specific phenotype. These new tools will greatly facilitate large-scale loss-of-function genetic screens in mammalian cells.


Subject(s)
Gene Library , RNA Interference , Tumor Suppressor Protein p53/metabolism , Cell Division , Cell Line, Tumor , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Down-Regulation , Fibroblasts , Genetic Vectors/genetics , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reproducibility of Results , Retroviridae/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics
5.
Oncogene ; 37(33): 4611-4625, 2018 08.
Article in English | MEDLINE | ID: mdl-29760405

ABSTRACT

Current treatment for advanced stage ovarian clear cell cancer is severely hampered by a lack of effective systemic therapy options, leading to a poor outlook for these patients. Sequencing studies revealed that ARID1A is mutated in over 50% of ovarian clear cell carcinomas. To search for a rational approach to target ovarian clear cell cancers with ARID1A mutations, we performed kinome-centered lethality screens in a large panel of ovarian clear cell carcinoma cell lines. Using the largest OCCC cell line panel established to date, we show here that BRD2 inhibition is predominantly lethal in ARID1A mutated ovarian clear cell cancer cells. Importantly, small molecule inhibitors of the BET (bromodomain and extra terminal domain) family of proteins, to which BRD2 belongs, specifically inhibit proliferation of ARID1A mutated cell lines, both in vitro and in ovarian clear cell cancer xenografts and patient-derived xenograft models. BET inhibitors cause a reduction in the expression of multiple SWI/SNF members including ARID1B, providing a potential explanation for the observed lethal interaction with ARID1A loss. Our data indicate that BET inhibition may represent a novel treatment strategy for a subset of ARID1A mutated ovarian clear cell carcinomas.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/genetics , Mutation/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Proteins/antagonists & inhibitors , Transcription Factors/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins , Female , Humans , Mice , Ovary/drug effects
6.
Clin Cancer Res ; 24(16): 3928-3940, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29685880

ABSTRACT

Purpose: Advanced-stage ovarian clear cell carcinoma (OCCC) is unresponsive to conventional platinum-based chemotherapy. Frequent alterations in OCCC include deleterious mutations in the tumor suppressor ARID1A and activating mutations in the PI3K subunit PIK3CA In this study, we aimed to identify currently unknown mutated kinases in patients with OCCC and test druggability of downstream affected pathways in OCCC models.Experimental Design: In a large set of patients with OCCC (n = 124), the human kinome (518 kinases) and additional cancer-related genes were sequenced, and copy-number alterations were determined. Genetically characterized OCCC cell lines (n = 17) and OCCC patient-derived xenografts (n = 3) were used for drug testing of ERBB tyrosine kinase inhibitors erlotinib and lapatinib, the PARP inhibitor olaparib, and the mTORC1/2 inhibitor AZD8055.Results: We identified several putative driver mutations in kinases at low frequency that were not previously annotated in OCCC. Combining mutations and copy-number alterations, 91% of all tumors are affected in the PI3K/AKT/mTOR pathway, the MAPK pathway, or the ERBB family of receptor tyrosine kinases, and 82% in the DNA repair pathway. Strong p-S6 staining in patients with OCCC suggests high mTORC1/2 activity. We consistently found that the majority of OCCC cell lines are especially sensitive to mTORC1/2 inhibition by AZD8055 and not toward drugs targeting ERBB family of receptor tyrosine kinases or DNA repair signaling. We subsequently demonstrated the efficacy of mTORC1/2 inhibition in all our unique OCCC patient-derived xenograft models.Conclusions: These results propose mTORC1/2 inhibition as an effective treatment strategy in OCCC. Clin Cancer Res; 24(16); 3928-40. ©2018 AACR.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Ovarian Neoplasms/genetics , Adenocarcinoma, Clear Cell/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Morpholines/pharmacology , Mutation/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/pathology , Signal Transduction/genetics , Transcription Factors/genetics
7.
Clin Cancer Res ; 22(21): 5238-5248, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27172896

ABSTRACT

PURPOSE: Despite the substantial progress in the development of targeted anticancer drugs, treatment failure due to primary or acquired resistance is still a major hurdle in the effective treatment of most advanced human cancers. Understanding these resistance mechanisms will be instrumental to improve personalized cancer treatment. EXPERIMENTAL DESIGN: Genome-wide loss-of-function genetic screens were performed to identify genes implicated in resistance to HER2/PI3K/mTOR targeting agents in HER2+ breast cancer cell lines. Expression and adjuvant trastuzumab response data from the HER2+ breast cancer trials FinHer and Responsify were used to validate our findings in patient series. RESULTS: We find that reduced ARID1A expression confers resistance to several drugs that inhibit the HER2/PI3K/mTOR signaling cascade at different levels. We demonstrate that ARID1A loss activates annexin A1 (ANXA1) expression, which is required for drug resistance through its activation of AKT. We find that the AKT inhibitor MK2206 restores sensitivity of ARID1A knockdown breast cancer cells to both the mTOR kinase inhibitor AZD8055 and trastuzumab. Consistent with these in vitro data, we find in two independent HER2+ breast cancer patient series that high ANXA1 expression is associated with resistance to adjuvant trastuzumab-based therapy. CONCLUSIONS: Our findings provide a rationale for why tumors accumulate ARID1A mutations and identify high ANXA1 expression as a predictive biomarker for trastuzumab-based treatment. Our findings also suggest strategies to treat breast cancers with elevated ANXA1 expression. Clin Cancer Res; 22(21); 5238-48. ©2016 AACR.


Subject(s)
Annexin A1/metabolism , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Trastuzumab/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , MCF-7 Cells , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Oncogene ; 23(30): 5138-50, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15133492

ABSTRACT

The E2F family of transcription factors play an important role in regulating cell cycle progression. We report here the characterization and functional properties of a new member of the human E2F family, referred to as E2F-7. E2F-7 has two separate DNA-binding domains, a feature that distinguishes E2F-7 from other mammalian E2F proteins, but resembling the organization of recently isolated E2F-like proteins from Arabidopsis. E2F-7 binds to DNA independently of a DP partner and delays cell cycle progression. Interestingly, E2F-7 modulates the transcription properties of other E2F proteins. A mutational analysis indicates that the integrity of both DNA-binding domains is required for cell cycle delay and transcriptional modulation. Biochemical results and protein modelling studies suggest that in binding to DNA interactions occur between the two DNA-binding domains, most probably as a homodimer, thereby mimicking the organization of an E2F/DP heterodimer. These structural and functional properties of E2F-7 imply a unique role in regulating cellular proliferation.


Subject(s)
DNA-Binding Proteins/genetics , Protein Structure, Tertiary , Repressor Proteins/genetics , Transcription Factors/genetics , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Cycle , Cell Line, Tumor , Cell Nucleus/chemistry , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , E2F7 Transcription Factor , Genes, Reporter , HeLa Cells , Humans , Luciferases/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Precipitin Tests , Protein Binding , Repressor Proteins/chemistry , Repressor Proteins/isolation & purification , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Transcription Factors/metabolism
9.
Cancer Res ; 74(14): 3810-20, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25028367

ABSTRACT

Tamoxifen is one of the most widely used endocrine agents for the treatment of estrogen receptor α (ERα)-positive breast cancer. Although effective in most patients, resistance to tamoxifen is a clinically significant problem and the mechanisms responsible remain elusive. To address this problem, we performed a large scale loss-of-function genetic screen in ZR-75-1 luminal breast cancer cells to identify candidate resistance genes. In this manner, we found that loss of function in the deubiquitinase USP9X prevented proliferation arrest by tamoxifen, but not by the ER downregulator fulvestrant. RNAi-mediated attenuation of USP9X was sufficient to stabilize ERα on chromatin in the presence of tamoxifen, causing a global tamoxifen-driven activation of ERα-responsive genes. Using a gene signature defined by their differential expression after USP9X attenuation in the presence of tamoxifen, we were able to define patients with ERα-positive breast cancer experiencing a poor outcome after adjuvant treatment with tamoxifen. The signature was specific in its lack of correlation with survival in patients with breast cancer who did not receive endocrine therapy. Overall, our findings identify a gene signature as a candidate biomarker of response to tamoxifen in breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Tamoxifen/therapeutic use , Ubiquitin Thiolesterase/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Chromatin/metabolism , Cluster Analysis , Down-Regulation , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Protein Binding , RNA, Small Interfering/genetics
10.
J Biol Chem ; 281(31): 21582-21587, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16737956

ABSTRACT

The helix-loop-helix transcription factor TFE3 has been suggested to play a role in the control of cell growth by acting as a binding partner of transcriptional regulators such as E2F3, SMAD3, and LEF-1. Furthermore, translocations/TFE3 fusions have been directly implicated in tumorigenesis. Surprisingly, however, a direct functional role for TFE3 in the regulation of proliferation has not been reported. By screening retroviral cDNA expression libraries to identify cDNAs that confer resistance to a pRB-induced proliferation arrest, we have found that TFE3 overrides a growth arrest in Rat1 cells induced by pRB and its upstream regulator p16(INK4A). In addition, TFE3 expression blocks the anti-mitogenic effects of TGF-beta in rodent and human cells. We provide data supporting a role for endogenous TFE3 in the direct regulation of CYCLIN E expression in an E2F3-dependent manner. These observations establish TFE3 as a functional regulator of proliferation and offer a potential mechanism for its involvement in cancer.


Subject(s)
Cell Proliferation , E2F3 Transcription Factor/physiology , Retinoblastoma Protein/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cells, Cultured , Cyclin E/genetics , DNA, Complementary , E2F3 Transcription Factor/genetics , Epithelial Cells/cytology , Fibroblasts/cytology , Gene Expression Regulation , Gene Library , Humans , Mink , Rats , Retinoblastoma Protein/physiology , Transfection , Transforming Growth Factor beta/physiology
11.
Genes Dev ; 19(3): 376-86, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15687260

ABSTRACT

The Wnt signaling cascade is a central regulator of cell fate determination during embryonic development, whose deregulation contributes to oncogenesis. Naked cuticle is the first Wnt-induced antagonist found in this pathway, establishing a negative-feedback loop that limits the Wnt signal required for early segmentation. In addition, Naked cuticle is proposed to function as a switch, acting to restrict classical Wnt signaling and to activate a second Wnt signaling pathway that controls planar cell polarity during gastrulation movements in vertebrates. Little is known about the biochemical function of Naked cuticle or its regulation. Here we report that PR72, a Protein Phosphatase type 2A regulatory subunit of unknown function, interacts both physically and functionally with Naked cuticle. We show that PR72, like Naked cuticle, acts as a negative regulator of the classical Wnt signaling cascade, establishing PR72 as a novel regulator of the Wnt signaling pathway. Our data provide evidence that the inhibitory effect of Naked cuticle on Wnt signaling depends on the presence of PR72, both in mammalian cell culture and in Xenopus embryos. Moreover, PR72 is required during early embryonic development to regulate cell morphogenetic movements during body axis formation.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction/physiology , Xenopus Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cloning, Molecular , Cytoskeletal Proteins/metabolism , Dishevelled Proteins , Embryo, Nonmammalian/metabolism , Eye/embryology , Eye/metabolism , Gastrula/metabolism , Humans , Phosphoproteins/metabolism , Protein Phosphatase 2 , Trans-Activators/metabolism , Wnt Proteins , Xenopus , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL