Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Anal Chem ; 95(11): 4834-4839, 2023 03 21.
Article En | MEDLINE | ID: mdl-36876898

The growing opportunities recognized for covalent drug inhibitors, like KRAS G12C inhibitors, are driving the need for mass spectrometry methods that can quickly and robustly measure therapeutic drug activity in vivo for drug discovery research and development. Effective front-end sample preparation is critical for proteins extracted from tumors but is generally labor intensive and impractical for large sample numbers typical in pharmacodynamic (PD) studies. Herein, we describe an automated and integrated sample preparation method for the measurement of activity levels of KRAS G12C drug inhibitor alkylation from complex tumor samples involving high throughput detergent removal and preconcentration followed by quantitation using mass spectrometry. We introduce a robust assay with an average intra-assay coefficient of variation (CV) of 4% and an interassay CV of 6% obtained from seven studies, enabling us to understand the relationship between KRAS G12C target occupancy and the therapeutic PD effect from mouse tumor samples. Further, the data demonstrated that the drug candidate GDC-6036, a KRAS G12C covalent inhibitor, shows dose-dependent target inhibition (KRAS G12C alkylation) and MAPK pathway inhibition, which correlate with high antitumor potency in the MIA PaCa-2 pancreatic xenograft model.


Antineoplastic Agents , Proto-Oncogene Proteins p21(ras) , Humans , Animals , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor , Mutation , Antineoplastic Agents/pharmacology , Disease Models, Animal
2.
Nat Immunol ; 23(4): 532-542, 2022 04.
Article En | MEDLINE | ID: mdl-35332327

The use of lipid-formulated RNA vaccines for cancer or COVID-19 is associated with dose-limiting systemic inflammatory responses in humans that were not predicted from preclinical studies. Here, we show that the 'interleukin 1 (IL-1)-interleukin 1 receptor antagonist (IL-1ra)' axis regulates vaccine-mediated systemic inflammation in a host-specific manner. In human immune cells, RNA vaccines induce production of IL-1 cytokines, predominantly IL-1ß, which is dependent on both the RNA and lipid formulation. IL-1 in turn triggers the induction of the broad spectrum of pro-inflammatory cytokines (including IL-6). Unlike humans, murine leukocytes respond to RNA vaccines by upregulating anti-inflammatory IL-1ra relative to IL-1 (predominantly IL-1α), protecting mice from cytokine-mediated toxicities at >1,000-fold higher vaccine doses. Thus, the IL-1 pathway plays a key role in triggering RNA vaccine-associated innate signaling, an effect that was unexpectedly amplified by certain lipids used in vaccine formulations incorporating N1-methyl-pseudouridine-modified RNA to reduce activation of Toll-like receptor signaling.


Inflammation , Interleukin 1 Receptor Antagonist Protein , Interleukin-1 , Animals , COVID-19 , Inflammation/immunology , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1/genetics , Interleukin-1/immunology , Lipids , Mice , RNA , Vaccines, Synthetic , mRNA Vaccines/adverse effects , mRNA Vaccines/metabolism
3.
Clin Cancer Res ; 27(4): 1162-1173, 2021 02 15.
Article En | MEDLINE | ID: mdl-33023953

PURPOSE: Lung adenocarcinomas comprise the largest fraction of non-small cell lung cancer, which is the leading cause of cancer-related deaths. Seventy-five percent of adenocarcinomas lack targeted therapies because of scarcity of druggable drivers. Here, we classified tumors on the basis of signaling similarities and discovered subgroups within this unmet patient population. EXPERIMENTAL DESIGN: We leveraged transcriptional data from >800 early- and advanced-stage patients. RESULTS: We identified three robust subtypes dubbed mucinous, proliferative, and mesenchymal with respective pathway phenotypes. These transcriptional states lack discrete and causative mutational etiology as evidenced by similarly distributed oncogenic drivers, including KRAS and EGFR. The subtypes capture heterogeneity even among tumors lacking known oncogenic drivers. Paired multi-regional intratumoral biopsies demonstrated unified subtypes despite divergently evolved prooncogenic mutations, indicating subtype stability during selective pressure. Heterogeneity among in vitro and in vivo preclinical models is expounded by the human lung adenocarcinoma subtypes and can be leveraged to discover subtype-specific vulnerabilities. As proof of concept, we identified differential subtype response to MEK pathway inhibition in a chemical library screen of 89 lung cancer cell lines, which reproduces across model systems and a clinical trial. CONCLUSIONS: Our findings support forward translational relevance of transcriptional subtypes, where further exploration therein may improve lung adenocarcinoma treatment.See related commentary by Skoulidis, p. 913.


Adenocarcinoma of Lung/drug therapy , Biomarkers, Tumor/genetics , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Clinical Trials as Topic , Datasets as Topic , Female , Genetic Heterogeneity , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Neoplasm Staging , Protein Kinase Inhibitors/pharmacology , RNA-Seq , Transcriptome/genetics , Xenograft Model Antitumor Assays
4.
Neuro Oncol ; 22(6): 819-829, 2020 06 09.
Article En | MEDLINE | ID: mdl-32383735

BACKGROUND: Studies evaluating the CNS penetration of a novel tyrosine kinase inhibitor, entrectinib, proved challenging, particularly due to discrepancies across earlier experiments regarding P-glycoprotein (P-gp) interaction and brain distribution. To address this question, we used a novel "apical efflux ratio" (AP-ER) model to assess P-gp interaction with entrectinib, crizotinib, and larotrectinib, and compared their brain-penetration properties. METHODS: AP-ER was designed to calculate P-gp interaction with the 3 drugs in vitro using P-gp-overexpressing cells. Brain penetration was studied in rat plasma, brain, and cerebrospinal fluid (CSF) samples after intravenous drug infusion. Unbound brain concentrations were estimated through kinetic lipid membrane binding assays and ex vivo experiments, while the antitumor activity of entrectinib was evaluated in a clinically relevant setting using an intracranial tumor mouse model. RESULTS: Entrectinib showed lower AP-ER (1.1-1.15) than crizotinib and larotrectinib (≥2.8). Despite not reaching steady-state brain exposures in rats after 6 hours, entrectinib presented a more favorable CSF-to-unbound concentration in plasma (CSF/Cu,p) ratio (>0.2) than crizotinib and larotrectinib at steady state (both: CSF/Cu,p ~0.03). In vivo experiments validated the AP-ER approach. Entrectinib treatment resulted in strong tumor inhibition and full survival benefit in the intracranial tumor model at clinically relevant systemic exposures. CONCLUSIONS: Entrectinib, unlike crizotinib and larotrectinib, is a weak P-gp substrate that can sustain CNS exposure based on our novel in vitro and in vivo experiments. This is consistent with the observed preclinical and clinical efficacy of entrectinib in neurotrophic tropomyosin receptor kinase (NTRK) and ROS1 fusion-positive CNS tumors and secondary CNS metastases.


ATP Binding Cassette Transporter, Subfamily B, Member 1 , Protein-Tyrosine Kinases , ATP Binding Cassette Transporter, Subfamily B , Animals , Benzamides , Cell Differentiation , Indazoles , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins , Rats
5.
J Exp Med ; 217(4)2020 04 06.
Article En | MEDLINE | ID: mdl-31940002

Tumor-specific mutations can generate neoantigens that drive CD8 T cell responses against cancer. Next-generation sequencing and computational methods have been successfully applied to identify mutations and predict neoantigens. However, only a small fraction of predicted neoantigens are immunogenic. Currently, predicted peptide binding affinity for MHC-I is often the major criterion for prioritizing neoantigens, although little progress has been made toward understanding the precise functional relationship between affinity and immunogenicity. We therefore systematically assessed the immunogenicity of peptides containing single amino acid mutations in mouse tumor models and divided them into two classes of immunogenic mutations. The first comprises mutations at a nonanchor residue, for which we find that the predicted absolute binding affinity is predictive of immunogenicity. The second involves mutations at an anchor residue; here, predicted relative affinity (compared with the WT counterpart) is a better predictor. Incorporating these features into an immunogenicity model significantly improves neoantigen ranking. Importantly, these properties of neoantigens are also predictive in human datasets, suggesting that they can be used to prioritize neoantigens for individualized neoantigen-specific immunotherapies.


Antigens, Neoplasm/immunology , Mutation , Neoplasms/genetics , Neoplasms/immunology , Amino Acids/genetics , Animals , Antibody Affinity , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class I/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , Peptides/genetics , Peptides/immunology , RNA-Seq , Exome Sequencing
6.
Sci Signal ; 11(547)2018 09 11.
Article En | MEDLINE | ID: mdl-30206136

The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.


DNA-Binding Proteins/genetics , Inflammation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Hippo Signaling Pathway , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transplantation, Heterologous
7.
Clin Cancer Res ; 21(13): 2916-23, 2015 Jul 01.
Article En | MEDLINE | ID: mdl-25838394

Tumors consist of a heterogeneous mixture of functionally distinct cancer cells. These functional differences can be caused by varying levels of receptor activity, differentiation, and distinct metabolic and epigenetic states. Intratumoral heterogeneity can lead to interdependence among different subpopulations of cells for sustained tumor growth. In addition, subpopulations can vary widely in their responses to therapeutic agents. As such, it is believed that intratumoral heterogeneity may underlie incomplete treatment responses, acquired and innate resistance, and disease relapse observed in the clinic in response to conventional chemotherapy and targeted agents.


Neoplasms/drug therapy , Animals , Drug Resistance, Neoplasm , Epigenesis, Genetic , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology
8.
Cancer Cell ; 24(1): 59-74, 2013 Jul 08.
Article En | MEDLINE | ID: mdl-23845442

Sustained tumor progression has been attributed to a distinct population of tumor-propagating cells (TPCs). To identify TPCs relevant to lung cancer pathogenesis, we investigated functional heterogeneity in tumor cells isolated from Kras-driven mouse models of non-small-cell lung cancer (NSCLC). CD24(+)ITGB4(+)Notch(hi) cells are capable of propagating tumor growth in both a clonogenic and an orthotopic serial transplantation assay. While all four Notch receptors mark TPCs, Notch3 plays a nonredundant role in tumor cell propagation in two mouse models and in human NSCLC. The TPC population is enriched after chemotherapy, and the gene signature of mouse TPCs correlates with poor prognosis in human NSCLC. The role of Notch3 in tumor propagation may provide a therapeutic target for NSCLC.


CD24 Antigen/analysis , Carcinoma, Non-Small-Cell Lung/etiology , Integrin beta4/analysis , Lung Neoplasms/etiology , Receptors, Notch/physiology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Receptor, Notch3 , Spheroids, Cellular
9.
Sci Transl Med ; 5(171): 171ra18, 2013 Feb 06.
Article En | MEDLINE | ID: mdl-23390248

Although standard chemotherapies are commonly used to treat most types of solid tumors, such treatment often results in inadequate response to, or relapse after, therapy. This is particularly relevant for lung cancer because most patients are diagnosed with advanced-stage disease and are treated with frontline chemotherapy. By studying the residual tumor cells that remain after chemotherapy in several in vivo non-small cell lung cancer models, we found that these cells have increased levels of human epidermal growth factor receptor (HER) signaling due, in part, to the enrichment of a preexisting NRG1(HI) subpopulation. Neuregulin 1 (NRG1) signaling in these models can be mediated by either the HER3 or HER4 receptor, resulting in the differential activation of downstream effectors. Inhibition of NRG1 signaling inhibits primary tumor growth and enhances the magnitude and duration of the response to chemotherapy. Moreover, we show that inhibition of ligand-mediated Her4 signaling impedes disease relapse in cases where NRG1 inhibition is insufficient. These findings demonstrate that ligand-dependent Her4 signaling plays an important role in disease relapse.


Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Neuregulin-1/antagonists & inhibitors , Signal Transduction , Animals , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Autocrine Communication/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans , Ligands , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm, Residual/drug therapy , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neuregulin-1/metabolism , Receptor, ErbB-4 , Signal Transduction/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
10.
Chromosome Res ; 15(3): 299-314, 2007.
Article En | MEDLINE | ID: mdl-17406994

Regulation of histone methylation is critical for proper gene expression and chromosome function. Suppressor of Zeste 12 (SUZ12) is a requisite member of the EED/EZH2 histone methyltransferase complexes, and is required for full activity of these complexes in vitro. In mammals and flies, SUZ12/Su(z)12 is necessary for trimethylation of histone H3 on lysine 27 (H3K27me3) on facultative heterochromatin. However, Su(z)12 is unique among Polycomb Group Proteins in that Su(z)12 mutant flies exhibit gross defects in position effect variegation, suggesting a role for Su(z)12 in constitutive heterochromatin formation. We investigated the role of Suz12 in constitutive heterochromatin and discovered that Suz12 is required for histone H3 lysine 9 tri-methylation (H3K9me3) in differentiated but not undifferentiated mouse embryonic stem cells. Knockdown of SUZ12 in human cells caused a reduction in H3K27me3 and H3K9me3, and altered the distribution of HP1 alpha. In contrast, EZH2 knockdown caused loss of H3K27me3 but not H3K9me3, indicating that SUZ12 regulates H3-K9 methylation in an EZH2-independent fashion. This work uncovers a role for SUZ12 in H3-K9 methylation.


Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , Histones/metabolism , Repressor Proteins/physiology , Animals , Carrier Proteins/physiology , Chromobox Protein Homolog 5 , DNA-Binding Proteins/physiology , Enhancer of Zeste Homolog 2 Protein , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase , Humans , Lysine/metabolism , Mice , Neoplasm Proteins , Nuclear Proteins/physiology , Polycomb Repressive Complex 2 , Proteins/physiology , Tissue Distribution , Transcription Factors/physiology
11.
Cell ; 128(5): 1003-12, 2007 Mar 09.
Article En | MEDLINE | ID: mdl-17350582

Histone lysine residues can be mono-, di-, or trimethylated. These posttranslational modifications regulate the affinity of effector proteins and may also impact chromatin structure independent of their role as adaptors. In order to study histone lysine methylation, particularly in the context of chromatin, we have developed a chemical approach to install analogs of methyl lysine into recombinant proteins. This approach allows for the rapid generation of large quantities of histones in which the site and degree of methylation can be specified. We demonstrate that these methyl-lysine analogs (MLAs) are functionally similar to their natural counterparts. These methylated histones were used to examine the influence of specific lysine methylation on the binding of effecter proteins and the rates of nucleosome remodeling. This simple method of introducing site-specific and degree-specific methylation into recombinant histones provides a powerful tool to investigate the biochemical mechanisms by which lysine methylation influences chromatin structure and function.


Histones/chemistry , Lysine/analogs & derivatives , Xenopus Proteins/chemistry , Animals , Cysteine/chemistry , Histones/genetics , Histones/metabolism , Methylation , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
12.
Chromosoma ; 114(3): 183-92, 2005 Aug.
Article En | MEDLINE | ID: mdl-15986205

Chromatin modifications are among the epigenetic alterations essential for genetic reprogramming during development. The Polycomb group (PcG) gene family mediates chromatin modifications that contribute to developmentally regulated transcriptional silencing. Trimethylation of histone H3 on lysine 27, mediated by a PcG protein complex consisting of Eed, Ezh2, and Suz12, is integral in differentiation, stem cell self-renewal, and tumorigenesis. Eed and Ezh2 are also implicated in the developmentally regulated silencing of the inactive X chromosome, as they are transiently enriched on the inactive X chromosome when X chromosome silencing is initiated. Here we analyze the dynamic localization of Suz12 during cellular differentiation and X-inactivation. Though Suz12 is a requisite member of the Eed/Ezh2 complexes, we found that Suz12 exhibits a notable difference from Ezh2 and Eed: while Ezh2 and Eed levels decrease during stem cell differentiation, Suz12 levels remain constant. Despite the differential regulation in abundance of Suz12 and Eed/Ezh2, Suz12 is also transiently enriched on the Xi during early stages of X-inactivation, and this accumulation is Xist RNA dependent. These results suggest that Suz12 may have a function that is not mediated by its association with Eed and Ezh2, and that this additional function is not involved in the regulation of X-inactivation.


Carrier Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , X Chromosome Inactivation , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Differentiation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Neoplasm Proteins , Nuclear Proteins , Polycomb Repressive Complex 2 , Protein Methyltransferases , RNA, Long Noncoding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Transfection , Trophoblasts/cytology , Trophoblasts/metabolism
13.
Science ; 300(5616): 131-5, 2003 Apr 04.
Article En | MEDLINE | ID: mdl-12649488

The Polycomb group (PcG) protein Eed is implicated in regulation of imprinted X-chromosome inactivation in extraembryonic cells but not of random X inactivation in embryonic cells. The Drosophila homolog of the Eed-Ezh2 PcG protein complex achieves gene silencing through methylation of histone H3 on lysine 27 (H3-K27), which suggests a role for H3-K27 methylation in imprinted X inactivation. Here we demonstrate that transient recruitment of the Eed-Ezh2 complex to the inactive X chromosome (Xi) occurs during initiation of X inactivation in both extraembryonic and embryonic cells and is accompanied by H3-K27 methylation. Recruitment of the complex and methylation on the Xi depend on Xist RNA but are independent of its silencing function. Together, our results suggest a role for Eed-Ezh2-mediated H3-K27 methylation during initiation of both imprinted and random X inactivation and demonstrate that H3-K27 methylation is not sufficient for silencing of the Xi.


Blastocyst/physiology , Dosage Compensation, Genetic , Histones/metabolism , Stem Cells/physiology , Trophoblasts/physiology , X Chromosome/metabolism , Animals , Blastocyst/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cells, Cultured , Female , Fluorescent Antibody Technique , Genomic Imprinting , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Lysine/metabolism , Male , Methylation , Mice , Mutation , Polycomb Repressive Complex 2 , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Transgenes
...