Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Chem Biol ; 30(9): 1156-1168.e7, 2023 09 21.
Article de Anglais | MEDLINE | ID: mdl-37689063

RÉSUMÉ

A challenge for screening new anticancer drugs is that efficacy in cell culture models is not always predictive of efficacy in patients. One limitation of standard cell culture is a reliance on non-physiological nutrient levels, which can influence cell metabolism and drug sensitivity. A general assessment of how physiological nutrients affect cancer cell response to small molecule therapies is lacking. To address this, we developed a serum-derived culture medium that supports the proliferation of diverse cancer cell lines and is amenable to high-throughput screening. We screened several small molecule libraries and found that compounds targeting metabolic enzymes were differentially effective in standard compared to serum-derived medium. We exploited the differences in nutrient levels between each medium to understand why medium conditions affected the response of cells to some compounds, illustrating how this approach can be used to screen potential therapeutics and understand how their efficacy is modified by available nutrients.


Sujet(s)
Techniques de culture cellulaire , Tests de criblage à haut débit , Humains , Lignée cellulaire , Bibliothèques de petites molécules/pharmacologie
2.
bioRxiv ; 2023 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-36909640

RÉSUMÉ

A challenge for screening new candidate drugs to treat cancer is that efficacy in cell culture models is not always predictive of efficacy in patients. One limitation of standard cell culture is a reliance on non-physiological nutrient levels to propagate cells. Which nutrients are available can influence how cancer cells use metabolism to proliferate and impact sensitivity to some drugs, but a general assessment of how physiological nutrients affect cancer cell response to small molecule therapies is lacking. To enable screening of compounds to determine how the nutrient environment impacts drug efficacy, we developed a serum-derived culture medium that supports the proliferation of diverse cancer cell lines and is amenable to high-throughput screening. We used this system to screen several small molecule libraries and found that compounds targeting metabolic enzymes were enriched as having differential efficacy in standard compared to serum-derived medium. We exploited the differences in nutrient levels between each medium to understand why medium conditions affected the response of cells to some compounds, illustrating how this approach can be used to screen potential therapeutics and understand how their efficacy is modified by available nutrients.

3.
Mol Cancer Ther ; 19(10): 2186-2195, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32747420

RÉSUMÉ

Uveal melanoma is a rare and aggressive cancer that originates in the eye. Currently, there are no approved targeted therapies and very few effective treatments for this cancer. Although activating mutations in the G protein alpha subunits, GNAQ and GNA11, are key genetic drivers of the disease, few additional drug targets have been identified. Recently, studies have identified context-specific roles for the mammalian SWI/SNF chromatin remodeling complexes (also known as BAF/PBAF) in various cancer lineages. Here, we find evidence that the SWI/SNF complex is essential through analysis of functional genomics screens and further validation in a panel of uveal melanoma cell lines using both genetic tools and small-molecule inhibitors of SWI/SNF. In addition, we describe a functional relationship between the SWI/SNF complex and the melanocyte lineage-specific transcription factor Microphthalmia-associated Transcription Factor, suggesting that these two factors cooperate to drive a transcriptional program essential for uveal melanoma cell survival. These studies highlight a critical role for SWI/SNF in uveal melanoma, and demonstrate a novel path toward the treatment of this cancer.


Sujet(s)
Chromatine/métabolisme , Mélanome/génétique , Tumeurs de l'uvée/génétique , Animaux , Lignée cellulaire tumorale , Protéines chromosomiques nonhistones , Humains , Souris , Facteurs de transcription
4.
Mol Cancer Ther ; 18(12): 2368-2380, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31439712

RÉSUMÉ

KRAS, an oncogene mutated in nearly one third of human cancers, remains a pharmacologic challenge for direct inhibition except for recent advances in selective inhibitors targeting the G12C variant. Here, we report that selective inhibition of the protein tyrosine phosphatase, SHP2, can impair the proliferation of KRAS-mutant cancer cells in vitro and in vivo using cell line xenografts and primary human tumors. In vitro, sensitivity of KRAS-mutant cells toward the allosteric SHP2 inhibitor, SHP099, is not apparent when cells are grown on plastic in 2D monolayer, but is revealed when cells are grown as 3D multicellular spheroids. This antitumor activity is also observed in vivo in mouse models. Interrogation of the MAPK pathway in SHP099-treated KRAS-mutant cancer models demonstrated similar modulation of p-ERK and DUSP6 transcripts in 2D, 3D, and in vivo, suggesting a MAPK pathway-dependent mechanism and possible non-MAPK pathway-dependent mechanisms in tumor cells or tumor microenvironment for the in vivo efficacy. For the KRASG12C MIAPaCa-2 model, we demonstrate that the efficacy is cancer cell intrinsic as there is minimal antiangiogenic activity by SHP099, and the effects of SHP099 is recapitulated by genetic depletion of SHP2 in cancer cells. Furthermore, we demonstrate that SHP099 efficacy in KRAS-mutant models can be recapitulated with RTK inhibitors, suggesting RTK activity is responsible for the SHP2 activation. Taken together, these data reveal that many KRAS-mutant cancers depend on upstream signaling from RTK and SHP2, and provide a new therapeutic framework for treating KRAS-mutant cancers with SHP2 inhibitors.


Sujet(s)
Tumeurs/traitement médicamenteux , Tumeurs/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protéines proto-oncogènes p21(ras)/génétique , Tachykinines/antagonistes et inhibiteurs , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Humains , Souris , Tumeurs/anatomopathologie , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
5.
ACS Med Chem Lett ; 9(7): 746-751, 2018 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-30034612

RÉSUMÉ

Mutant isocitrate dehydrogenase 1 (IDH1) is an attractive therapeutic target for the treatment of various cancers such as AML, glioma, and glioblastoma. We have evaluated 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors that bind to an allosteric, induced pocket of IDH1R132H. This Letter describes SAR exploration focused on improving both the in vitro and in vivo metabolic stability of the compounds, leading to the identification of 19 as a potent and selective mutant IDH1 inhibitor that has demonstrated brain penetration and excellent oral bioavailability in rodents. In a preclinical patient-derived IDH1 mutant xenograft tumor model study, 19 efficiently inhibited the production of the biomarker 2-HG.

6.
ACS Med Chem Lett ; 8(10): 1116-1121, 2017 Oct 12.
Article de Anglais | MEDLINE | ID: mdl-29057061

RÉSUMÉ

Inhibition of mutant IDH1 is being evaluated clinically as a promising treatment option for various cancers with hotspot mutation at Arg132. Having identified an allosteric, induced pocket of IDH1R132H, we have explored 3-pyrimidin-4-yl-oxazolidin-2-ones as mutant IDH1 inhibitors for in vivo modulation of 2-HG production and potential brain penetration. We report here optimization efforts toward the identification of clinical candidate IDH305 (13), a potent and selective mutant IDH1 inhibitor that has demonstrated brain exposure in rodents. Preclinical characterization of this compound exhibited in vivo correlation of 2-HG reduction and efficacy in a patient-derived IDH1 mutant xenograft tumor model. IDH305 (13) has progressed into human clinical trials for the treatment of cancers with IDH1 mutation.

7.
Cell ; 170(3): 577-592.e10, 2017 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-28753431

RÉSUMÉ

Elucidation of the mutational landscape of human cancer has progressed rapidly and been accompanied by the development of therapeutics targeting mutant oncogenes. However, a comprehensive mapping of cancer dependencies has lagged behind and the discovery of therapeutic targets for counteracting tumor suppressor gene loss is needed. To identify vulnerabilities relevant to specific cancer subtypes, we conducted a large-scale RNAi screen in which viability effects of mRNA knockdown were assessed for 7,837 genes using an average of 20 shRNAs per gene in 398 cancer cell lines. We describe findings of this screen, outlining the classes of cancer dependency genes and their relationships to genetic, expression, and lineage features. In addition, we describe robust gene-interaction networks recapitulating both protein complexes and functional cooperation among complexes and pathways. This dataset along with a web portal is provided to the community to assist in the discovery and translation of new therapeutic approaches for cancer.


Sujet(s)
Tumeurs/génétique , Tumeurs/anatomopathologie , Interférence par ARN , Lignée cellulaire tumorale , Banque de gènes , Réseaux de régulation génique , Humains , Complexes multiprotéiques/métabolisme , Tumeurs/métabolisme , Oncogènes , Petit ARN interférent , Transduction du signal , Facteurs de transcription/métabolisme
8.
Structure ; 25(3): 506-513, 2017 03 07.
Article de Anglais | MEDLINE | ID: mdl-28132785

RÉSUMÉ

Oncogenic IDH1 and IDH2 mutations contribute to cancer via production of R-2-hydroxyglutarate (2-HG). Here, we characterize two structurally distinct mutant- and isoform-selective IDH1 inhibitors that inhibit 2-HG production. Both bind to an allosteric pocket on IDH1, yet shape it differently, highlighting the plasticity of this site. Oncogenic IDH1R132H mutation destabilizes an IDH1 "regulatory segment," which otherwise restricts compound access to the allosteric pocket. Regulatory segment destabilization in wild-type IDH1 promotes inhibitor binding, suggesting that destabilization is critical for mutant selectivity. We also report crystal structures of oncogenic IDH2 mutant isoforms, highlighting the fact that the analogous segment of IDH2 is not similarly destabilized. This intrinsic stability of IDH2 may contribute to observed inhibitor IDH1 isoform selectivity. Moreover, discrete residues in the IDH1 allosteric pocket that differ from IDH2 may also guide IDH1 isoform selectivity. These data provide a deeper understanding of how IDH1 inhibitors achieve mutant and isoform selectivity.


Sujet(s)
Antienzymes/pharmacologie , Isocitrate dehydrogenases/composition chimique , Isocitrate dehydrogenases/génétique , Tumeurs/génétique , Bibliothèques de petites molécules/pharmacologie , Régulation allostérique , Site allostérique , Cristallographie aux rayons X , Glutarates/métabolisme , Humains , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Liaison aux protéines , Conformation des protéines , Isoformes de protéines/composition chimique , Isoformes de protéines/génétique
9.
Cancer Res ; 76(23): 6950-6963, 2016 12 01.
Article de Anglais | MEDLINE | ID: mdl-27659046

RÉSUMÉ

Like classical chemotherapy regimens used to treat cancer, targeted therapies will also rely upon polypharmacology, but tools are still lacking to predict which combinations of molecularly targeted drugs may be most efficacious. In this study, we used image-based proliferation and apoptosis assays in colorectal cancer cell lines to systematically investigate the efficacy of combinations of two to six drugs that target critical oncogenic pathways. Drug pairs targeting key signaling pathways resulted in synergies across a broad spectrum of genetic backgrounds but often yielded only cytostatic responses. Enhanced cytotoxicity was observed when additional processes including apoptosis and cell cycle were targeted as part of the combination. In some cases, where cell lines were resistant to paired and tripled drugs, increased expression of antiapoptotic proteins was observed, requiring a fourth-order combination to induce cytotoxicity. Our results illustrate how high-order drug combinations are needed to kill drug-resistant cancer cells, and they also show how systematic drug combination screening together with a molecular understanding of drug responses may help define optimal cocktails to overcome aggressive cancers. Cancer Res; 76(23); 6950-63. ©2016 AACR.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs colorectales/traitement médicamenteux , Animaux , Prolifération cellulaire , Tumeurs colorectales/génétique , Femelle , Humains , Souris , Transduction du signal
10.
Nat Med ; 20(1): 87-92, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24362935

RÉSUMÉ

Mantle cell lymphoma (MCL) is an aggressive malignancy that is characterized by poor prognosis. Large-scale pharmacological profiling across more than 100 hematological cell line models identified a subset of MCL cell lines that are highly sensitive to the B cell receptor (BCR) signaling inhibitors ibrutinib and sotrastaurin. Sensitive MCL models exhibited chronic activation of the BCR-driven classical nuclear factor-κB (NF-κB) pathway, whereas insensitive cell lines displayed activation of the alternative NF-κB pathway. Transcriptome sequencing revealed genetic lesions in alternative NF-κB pathway signaling components in ibrutinib-insensitive cell lines, and sequencing of 165 samples from patients with MCL identified recurrent mutations in TRAF2 or BIRC3 in 15% of these individuals. Although they are associated with insensitivity to ibrutinib, lesions in the alternative NF-κB pathway conferred dependence on the protein kinase NIK (also called mitogen-activated protein 3 kinase 14 or MAP3K14) both in vitro and in vivo. Thus, NIK is a new therapeutic target for MCL treatment, particularly for lymphomas that are refractory to BCR pathway inhibitors. Our findings reveal a pattern of mutually exclusive activation of the BCR-NF-κB or NIK-NF-κB pathways in MCL and provide critical insights into patient stratification strategies for NF-κB pathway-targeted agents.


Sujet(s)
Lymphome à cellules du manteau/traitement médicamenteux , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Pyrroles/pharmacologie , Quinazolines/pharmacologie , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Adénine/analogues et dérivés , Protéine-3 contenant des répétitions IAP baculovirales , Séquence nucléotidique , Technique de Western , Protéines adaptatrices de signalisation CARD/métabolisme , Lignée cellulaire , Survie cellulaire , Amorces ADN/génétique , Guanylate cyclase/métabolisme , Humains , Protéines IAP/génétique , Protéines IAP/métabolisme , Mesures de luminescence , Analyse sur microréseau , Données de séquences moléculaires , Pipéridines , Protein-Serine-Threonine Kinases/génétique , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Interférence par ARN , Réaction de polymérisation en chaine en temps réel , Récepteurs pour l'antigène des lymphocytes B/antagonistes et inhibiteurs , Analyse de séquence d'ARN , Facteur-2 associé aux récepteurs de TNF/génétique , Facteur-2 associé aux récepteurs de TNF/métabolisme , Facteur-3 associé aux récepteurs de TNF/métabolisme , Bleu de trypan , Ubiquitin-protein ligases ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE