Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters








Publication year range
2.
NPJ Precis Oncol ; 8(1): 90, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627602

ABSTRACT

In this review, we cover the current understanding of BRAF mutations and associated clinical characteristics in patients with metastatic NSCLC, approved and emerging treatment options, BRAF sequencing approaches, and unmet needs. The BRAFV600E mutation confers constitutive activity of the MAPK pathway, leading to enhanced growth, proliferation, and survival of tumor cells. Testing for BRAF mutations enables patients to be treated with therapies that directly target BRAFV600E and the MAPK pathway, but BRAF testing lags behind other oncogene testing in metastatic NSCLC. Additional therapies targeting BRAFV600E mutations provide options for patients with metastatic NSCLC. Emerging therapies and combinations under investigation could potentially overcome issues of resistance and target non-V600E mutations. Therefore, because targeted therapies with enhanced efficacy are on the horizon, being able to identify BRAF mutations in metastatic NSCLC may become even more important.

3.
Article in English | MEDLINE | ID: mdl-37788883

ABSTRACT

Animal models, particularly genetically engineered mouse models (GEMMs), continue to have a transformative impact on our understanding of the initiation and progression of hematological malignancies and solid tumors. Furthermore, GEMMs have been employed in the design and optimization of potent anticancer therapies. Increasingly, drug responses are assessed in mouse models either prior, or in parallel, to the implementation of precision medical oncology, in which groups of patients with genetically stratified cancers are treated with drugs that target the relevant oncoprotein such that mechanisms of drug sensitivity or resistance may be identified. Subsequently, this has led to the design and preclinical testing of combination therapies designed to forestall the onset of drug resistance. Indeed, mouse models of human lung cancer represent a paradigm for how a wide variety of GEMMs, driven by a variety of oncogenic drivers, have been generated to study initiation, progression, and maintenance of this disease as well as response to drugs. These studies have now expanded beyond targeted therapy to include immunotherapy. We highlight key aspects of the relationship between mouse models and the evolution of therapeutic approaches, including oncogene-targeted therapies, immunotherapies, acquired drug resistance, and ways in which successful antitumor strategies improve on efficiently translating preclinical approaches into successful antitumor strategies in patients.


Subject(s)
Lung Neoplasms , Animals , Mice , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Genetic Engineering , Disease Models, Animal
4.
Cancer Res ; 82(22): 4261-4273, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36112789

ABSTRACT

Mutationally activated BRAF is detected in approximately 7% of human lung adenocarcinomas, with BRAFT1799A serving as a predictive biomarker for treatment of patients with FDA-approved inhibitors of BRAFV600E oncoprotein signaling. In genetically engineered mouse (GEM) models, expression of BRAFV600E in the lung epithelium initiates growth of benign lung tumors that, without additional genetic alterations, rarely progress to malignant lung adenocarcinoma. To identify genes that cooperate with BRAFV600E for malignant progression, we used Sleeping Beauty-mediated transposon mutagenesis, which dramatically accelerated the emergence of lethal lung cancers. Among the genes identified was Rbms3, which encodes an RNA-binding protein previously implicated as a putative tumor suppressor. Silencing of RBMS3 via CRISPR/Cas9 gene editing promoted growth of BRAFV600E lung organoids and promoted development of malignant lung cancers with a distinct micropapillary architecture in BRAFV600E and EGFRL858R GEM models. BRAFV600E/RBMS3Null lung tumors displayed elevated expression of Ctnnb1, Ccnd1, Axin2, Lgr5, and c-Myc mRNAs, suggesting that RBMS3 silencing elevates signaling through the WNT/ß-catenin signaling axis. Although RBMS3 silencing rendered BRAFV600E-driven lung tumors resistant to the effects of dabrafenib plus trametinib, the tumors were sensitive to inhibition of porcupine, an acyltransferase of WNT ligands necessary for their secretion. Analysis of The Cancer Genome Atlas patient samples revealed that chromosome 3p24, which encompasses RBMS3, is frequently lost in non-small cell lung cancer and correlates with poor prognosis. Collectively, these data reveal the role of RBMS3 as a lung cancer suppressor and suggest that RBMS3 silencing may contribute to malignant NSCLC progression. SIGNIFICANCE: Loss of RBMS3 cooperates with BRAFV600E to induce lung tumorigenesis, providing a deeper understanding of the molecular mechanisms underlying mutant BRAF-driven lung cancer and potential strategies to more effectively target this disease.


Subject(s)
Adenocarcinoma of Lung , Carcinogenesis , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Proto-Oncogene Proteins B-raf , RNA-Binding Proteins , Trans-Activators , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation , Lung/pathology , Lung Neoplasms/genetics , Mutagenesis , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/genetics , Trans-Activators/metabolism , Wnt Signaling Pathway , Carcinogenesis/genetics
5.
Cell Rep ; 32(5): 107994, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755586

ABSTRACT

NTRK1 gene fusions are actionable drivers of numerous human malignancies. Here, we show that expression of the TPR-NTRK1 fusion kinase in immortalized mouse pancreatic ductal epithelial (IMPE) (pancreas) or mouse lung epithelial (MLE-12) cells is sufficient to promote rapidly growing tumors in mice. Both tumor models are exquisitely sensitive to targeted inhibition with entrectinib, a tropomyosin-related kinase A (TRKA) inhibitor. Initial regression of NTRK1-driven tumors is driven by induced expression of BIM, such that BIM silencing leads to a diminished response to entrectinib in vivo. However, the emergence of drug-resistant disease limits the long-term durability of responses. Based on the reactivation of RAF>MEK>ERK signaling observed in entrectinib-treated tumors, we show that the combination of entrectinib plus the MEK1/2 inhibitor cobimetinib dramatically forestalls the onset of drug resistance in vivo. Collectively, these data provide a mechanistic rationale for rapid clinical deployment of combined inhibition of TRKA plus MEK1/2 in NTRK1-driven cancers.


Subject(s)
Benzamides/pharmacology , Drug Resistance, Neoplasm/drug effects , Indazoles/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Models, Biological , Neoplasms/enzymology , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor, trkA/metabolism , Animals , Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Mice, Inbred NOD , Mitogen-Activated Protein Kinase Kinases/metabolism , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology
6.
Cancer Res ; 77(13): 3551-3563, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28428274

ABSTRACT

Oncogenic kinase fusions of ALK, ROS1, RET, and NTRK1 act as drivers in human lung and other cancers. Residual tumor burden following treatment of ALK or ROS1+ lung cancer patients with oncogene-targeted therapy ultimately enables the emergence of drug-resistant clones, limiting the long-term effectiveness of these therapies. To determine the signaling mechanisms underlying incomplete tumor cell killing in oncogene-addicted cancer cells, we investigated the role of EGFR signaling in drug-naïve cancer cells harboring these oncogene fusions. We defined three distinct roles for EGFR in the response to oncogene-specific therapies. First, EGF-mediated activation of EGFR blunted fusion kinase inhibitor binding and restored fusion kinase signaling complexes. Second, fusion kinase inhibition shifted adaptor protein binding from the fusion oncoprotein to EGFR. Third, EGFR enabled bypass signaling to critical downstream pathways such as MAPK. While evidence of EGFR-mediated bypass signaling has been reported after ALK and ROS1 blockade, our results extended this effect to RET and NTRK1 blockade and uncovered the other additional mechanisms in gene fusion-positive lung cancer cells, mouse models, and human clinical specimens before the onset of acquired drug resistance. Collectively, our findings show how EGFR signaling can provide a critical adaptive survival mechanism that allows cancer cells to evade oncogene-specific inhibitors, providing a rationale to cotarget EGFR to reduce the risks of developing drug resistance. Cancer Res; 77(13); 3551-63. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Oncogene Proteins, Fusion/metabolism , Small Molecule Libraries/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Oncogene Proteins, Fusion/genetics , Signal Transduction , Xenograft Model Antitumor Assays
7.
Nat Med ; 21(9): 1038-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26301689

ABSTRACT

One strategy for combating cancer-drug resistance is to deploy rational polytherapy up front that suppresses the survival and emergence of resistant tumor cells. Here we demonstrate in models of lung adenocarcinoma harboring the oncogenic fusion of ALK and EML4 that the GTPase RAS-mitogen-activated protein kinase (MAPK) pathway, but not other known ALK effectors, is required for tumor-cell survival. EML4-ALK activated RAS-MAPK signaling by engaging all three major RAS isoforms through the HELP domain of EML4. Reactivation of the MAPK pathway via either a gain in the number of copies of the gene encoding wild-type K-RAS (KRAS(WT)) or decreased expression of the MAPK phosphatase DUSP6 promoted resistance to ALK inhibitors in vitro, and each was associated with resistance to ALK inhibitors in individuals with EML4-ALK-positive lung adenocarcinoma. Upfront inhibition of both ALK and the kinase MEK enhanced both the magnitude and duration of the initial response in preclinical models of EML4-ALK lung adenocarcinoma. Our findings identify RAS-MAPK dependence as a hallmark of EML4-ALK lung adenocarcinoma and provide a rationale for the upfront inhibition of both ALK and MEK to forestall resistance and improve patient outcomes.


Subject(s)
Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinases/physiology , Oncogene Proteins, Fusion/physiology , ras Proteins/physiology , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Drug Resistance, Neoplasm , Dual Specificity Phosphatase 6/physiology , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oncogene Proteins, Fusion/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , ras Proteins/genetics
8.
Cancer Discov ; 5(10): 1049-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216294

ABSTRACT

UNLABELLED: Oncogenic TRK fusions induce cancer cell proliferation and engage critical cancer-related downstream signaling pathways. These TRK fusions occur rarely, but in a diverse spectrum of tumor histologies. LOXO-101 is an orally administered inhibitor of the TRK kinase and is highly selective only for the TRK family of receptors. Preclinical models of LOXO-101 using TRK-fusion-bearing human-derived cancer cell lines demonstrate inhibition of the fusion oncoprotein and cellular proliferation in vitro, and tumor growth in vivo. The tumor of a 41-year-old woman with soft-tissue sarcoma metastatic to the lung was found to harbor an LMNA-NTRK1 gene fusion encoding a functional LMNA-TRKA fusion oncoprotein as determined by an in situ proximity ligation assay. In a phase I study of LOXO-101 (ClinicalTrials.gov no. NCT02122913), this patient's tumors underwent rapid and substantial tumor regression, with an accompanying improvement in pulmonary dyspnea, oxygen saturation, and plasma tumor markers. SIGNIFICANCE: TRK fusions have been deemed putative oncogenic drivers, but their clinical significance remained unclear. A patient with a metastatic soft-tissue sarcoma with an LMNA-NTRK1 fusion had rapid and substantial tumor regression with a novel, highly selective TRK inhibitor, LOXO-101, providing the first clinical evidence of benefit from inhibiting TRK fusions.


Subject(s)
Antineoplastic Agents/therapeutic use , Oncogene Proteins, Fusion/genetics , Oncogene Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Sarcoma/drug therapy , Sarcoma/genetics , Adult , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Dose-Response Relationship, Drug , Female , Humans , Lamin Type A/genetics , Neoplasm Staging , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sarcoma/diagnosis , Tomography, X-Ray Computed , Treatment Outcome
9.
Cancer Discov ; 5(1): 25-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25527197

ABSTRACT

UNLABELLED: The use of high-throughput next-generation sequencing techniques in multiple tumor types during the last few years has identified NTRK1, 2, and 3 gene rearrangements encoding novel oncogenic fusions in 19 different tumor types to date. These recent developments have led us to revisit an old oncogene, Trk (originally identified as OncD), which encodes the TPM3-NTRK1 gene fusion and was one of the first transforming chromosomal rearrangements identified 32 years ago. However, no drug has yet been approved by the FDA for cancers harboring this oncogene. This review will discuss the biology of the TRK family of receptors, their role in human cancer, the types of oncogenic alterations, and drugs that are currently in development for this family of oncogene targets. SIGNIFICANCE: Precision oncology approaches have accelerated recently due to advancements in our ability to detect oncogenic mutations in tumor samples. Oncogenic alterations, most commonly gene fusions, have now been detected for the genes encoding the TRKA, TRKB, and TRKC receptor tyrosine kinases across multiple tumor types. The scientific rationale for the targeting of the TRK oncogene family will be discussed here.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Alternative Splicing , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Molecular Targeted Therapy , Multigene Family , Mutation , Neoplasms/drug therapy , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins, Fusion , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
10.
PLoS One ; 8(12): e82236, 2013.
Article in English | MEDLINE | ID: mdl-24349229

ABSTRACT

The targeting of oncogenic 'driver' kinases with small molecule inhibitors has proven to be a highly effective therapeutic strategy in selected non-small cell lung cancer (NSCLC) patients. However, acquired resistance to targeted therapies invariably arises and is a major limitation to patient care. ROS1 fusion proteins are a recently described class of oncogenic driver, and NSCLC patients that express these fusions generally respond well to ROS1-targeted therapy. In this study, we sought to determine mechanisms of acquired resistance to ROS1 inhibition. To accomplish this, we analyzed tumor samples from a patient who initially responded to the ROS1 inhibitor crizotinib but eventually developed acquired resistance. In addition, we generated a ROS1 inhibition-resistant derivative of the initially sensitive NSCLC cell line HCC78. Previously described mechanisms of acquired resistance to tyrosine kinase inhibitors including target kinase-domain mutation, target copy number gain, epithelial-mesenchymal transition, and conversion to small cell lung cancer histology were found to not underlie resistance in the patient sample or resistant cell line. However, we did observe a switch in the control of growth and survival signaling pathways from ROS1 to EGFR in the resistant cell line. As a result of this switch, ROS1 inhibition-resistant HCC78 cells became sensitive to EGFR inhibition, an effect that was enhanced by co-treatment with a ROS1 inhibitor. Our results suggest that co-inhibition of ROS1 and EGFR may be an effective strategy to combat resistance to targeted therapy in some ROS1 fusion-positive NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Signal Transduction , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Crizotinib , Drug Resistance, Neoplasm/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Gene Amplification/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/drug effects
11.
Development ; 140(10): 2160-71, 2013 May.
Article in English | MEDLINE | ID: mdl-23578928

ABSTRACT

FoxA transcription factors play major roles in organ-specific gene expression, regulating, for example, glucagon expression in the pancreas, GLUT2 expression in the liver, and tyrosine hydroxylase expression in dopaminergic neurons. Organ-specific gene regulation by FoxA proteins is achieved through cooperative regulation with a broad array of transcription factors with more limited expression domains. Fork head (Fkh), the sole Drosophila FoxA family member, is required for the development of multiple distinct organs, yet little is known regarding how Fkh regulates tissue-specific gene expression. Here, we characterize Sage, a bHLH transcription factor expressed exclusively in the Drosophila salivary gland (SG). We show that Sage is required for late SG survival and normal tube morphology. We find that many Sage targets, identified by microarray analysis, encode SG-specific secreted cargo, transmembrane proteins, and the enzymes that modify these proteins. We show that both Sage and Fkh are required for the expression of Sage target genes, and that co-expression of Sage and Fkh is sufficient to drive target gene expression in multiple cell types. Sage and Fkh drive expression of the bZip transcription factor Senseless (Sens), which boosts expression of Sage-Fkh targets, and Sage, Fkh and Sens colocalize on SG chromosomes. Importantly, expression of Sage-Fkh target genes appears to simply add to the tissue-specific gene expression programs already established in other cell types, and Sage and Fkh cannot alter the fate of most embryonic cell types even when expressed early and continuously.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , RNA-Binding Proteins/metabolism , Salivary Proteins and Peptides/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosomes/ultrastructure , Crosses, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Forkhead Transcription Factors , Gene Expression Profiling , In Situ Hybridization , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Organ Specificity , Salivary Glands/metabolism , Salivary Proteins and Peptides/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL