Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38775153

ABSTRACT

Small cell lung cancer (SCLC) is the most aggressive lung cancer entity with an extremely limited therapeutic outcome. Most patients are diagnosed at an extensive stage. However, the molecular mechanisms driving SCLC invasion and metastasis remain largely elusive. We used an autochthonous SCLC mouse model and matched samples from patients with primary and metastatic SCLC to investigate the molecular characteristics of tumor metastasis. We demonstrate that tumor cell invasion and liver metastasis in SCLC are triggered by an Angiopoietin-2 (ANG-2)/Integrin ß-1-dependent pathway in tumor cells, mediated by focal adhesion kinase/Src kinase signaling. Strikingly, CRISPR-Cas9 KO of Integrin ß-1 or blocking Integrin ß-1 signaling by an anti-ANG-2 treatment abrogates liver metastasis formation in vivo. Interestingly, analysis of a unique collection of matched samples from patients with primary and metastatic SCLC confirmed a strong increase of Integrin ß-1 in liver metastasis in comparison with the primary tumor. We further show that ANG-2 blockade combined with PD-1-targeted by anti-PD-1 treatment displays synergistic treatment effects in SCLC. Together, our data demonstrate a fundamental role of ANG-2/Integrin ß-1 signaling in SCLC cells for tumor cell invasion and liver metastasis and provide a potentially new effective treatment strategy for patients with SCLC.


Subject(s)
Angiopoietin-2 , Integrin beta1 , Liver Neoplasms , Lung Neoplasms , Signal Transduction , Small Cell Lung Carcinoma , Animals , Female , Humans , Male , Mice , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Cell Line, Tumor , Integrin beta1/metabolism , Integrin beta1/genetics , Liver Neoplasms/secondary , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/drug therapy
2.
Pathol Oncol Res ; 27: 596522, 2021.
Article in English | MEDLINE | ID: mdl-34257546

ABSTRACT

Purpose: Abrogation of Notch signaling, which is pivotal for lung development and pulmonary epithelial cell fate decisions was shown to be involved in the aggressiveness and the differentiation of lung carcinomas. Additionally, the transcription factors YAP and TAZ which are involved in the Hippo pathway, were recently shown to be tightly linked with Notch signaling and to regulate the cell fate in epidermal stem cells. Thus, we aim to elucidate the effects of conditional Notch1 deficiency on carcinogenesis and TAZ expression in lung cancer. Methods: We investigated the effect of conditional Cre-recombinase mediated Notch1 knock-out on lung cancer cells in vivo using an autochthonous mouse model of lung adenocarcinomas driven by Kras LSL-G12V and comprehensive immunohistochemical analysis. In addition, we analyzed clinical samples and human lung cancer cell lines for TAZ expression and supported our findings by publicly available data from The Cancer Genome Atlas (TCGA). Results: In mice, we found induction of papillary adenocarcinomas and protrusions of tumor cells from the bronchiolar lining upon Notch1 deficiency. Moreover, the mutated Kras driven lung tumors with deleted Notch1 showed increased TAZ expression and focal nuclear translocation which was frequently observed in human pulmonary adenocarcinomas and squamous cell carcinomas of the lung, but not in small cell lung carcinomas. In addition, we used data from TCGA to show that putative inactivating NOTCH1 mutations co-occur with KRAS mutations and genomic amplifications in lung adenocarcinomas. Conclusion: Our in vivo study provides evidence that Notch1 deficiency in mutated Kras driven lung carcinomas contributes to lung carcinogenesis in a subgroup of patients by increasing TAZ expression who might benefit from TAZ signaling blockade.


Subject(s)
Acyltransferases/metabolism , Bronchi/pathology , Disease Models, Animal , Lung Neoplasms/pathology , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Notch1/physiology , Acyltransferases/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bronchi/metabolism , Carcinogenesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Prognosis , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Cells, Cultured
3.
Cell Rep ; 31(4): 107568, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348765

ABSTRACT

Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF)-VEGFR2 signaling has shown limited efficacy in lung cancer patients. Here, we demonstrate that inhibition of VEGFR2 in tumor cells, expressed in ∼20% of non-squamous non-small cell lung cancer (NSCLC) patients, leads to a pro-invasive phenotype. Drug-induced inhibition of tumor VEGFR2 interferes with the formation of the EphA2/VEGFR2 heterocomplex, thereby allowing RSK to interact with Serine 897 of EphA2. Inhibition of RSK decreases phosphorylation of Serine 897 EphA2. Selective genetic modeling of Serine 897 of EphA2 or inhibition of EphA2 abrogates the formation of metastases in vivo upon VEGFR2 inhibition. In summary, these findings demonstrate that VEGFR2-targeted therapy conditions VEGFR2-positive NSCLC to Serine 897 EphA2-dependent aggressive tumor growth and metastasis. These data shed light on the molecular mechanisms explaining the limited efficacy of VEGFR2-targeted anti-angiogenic treatment in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Receptor, EphA2/metabolism , Serine/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Oncogene ; 37(42): 5682-5693, 2018 10.
Article in English | MEDLINE | ID: mdl-29970903

ABSTRACT

FGFR1 amplification has been found in 15% of patients with breast cancer and has been postulated as a promising marker to predict response against FGFR inhibitors. However, early phase clinical trials of selective FGFR inhibitors demonstrated only limited efficacy in FGFR1-amplified breast cancer patients. We found that BGJ398, an FGFR inhibitor, effectively inhibited phosphorylation of FGFR1 and MEK/ERK signaling in FGFR1-amplified breast cancer without affecting tumor cell proliferation. However, FGFR1 knockout inhibited tumor angiogenesis in vivo. We unraveled that FGFR1 regulates the secretion of the proangiogenic vascular endothelial growth factor (VEGF) in a MAPK-dependent manner. We further found that FGF-FGFR1 signaling induces an autocrine activation of VEGF-VEGFR1 pathway that again amplifies VEGF secretion via VEGF-VEGFR1-AKT signaling. Targeting both VEGFR1 and FGFR1 resulted in synergistic anti-angiogenic treatment effects in vivo. We thus postulate synergistic treatment effects in FGFR1/VEGFR1-positive breast cancer patients by dual targeting of FGFR and VEGFR.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Synergism , Female , Heterografts , Humans , Mice , Mice, Inbred NOD , Naphthalenes/pharmacology , Neovascularization, Pathologic , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Quinolines/pharmacology
5.
Mol Cell ; 70(4): 730-744.e6, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706538

ABSTRACT

Processes like cellular senescence are characterized by complex events giving rise to heterogeneous cell populations. However, the early molecular events driving this cascade remain elusive. We hypothesized that senescence entry is triggered by an early disruption of the cells' three-dimensional (3D) genome organization. To test this, we combined Hi-C, single-cell and population transcriptomics, imaging, and in silico modeling of three distinct cells types entering senescence. Genes involved in DNA conformation maintenance are suppressed upon senescence entry across all cell types. We show that nuclear depletion of the abundant HMGB2 protein occurs early on the path to senescence and coincides with the dramatic spatial clustering of CTCF. Knocking down HMGB2 suffices for senescence-induced CTCF clustering and for loop reshuffling, while ectopically expressing HMGB2 rescues these effects. Our data suggest that HMGB2-mediated genomic reorganization constitutes a primer for the ensuing senescent program.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Genome, Human , HMGB2 Protein/metabolism , CCCTC-Binding Factor/genetics , Cell Proliferation , Cellular Senescence , Chromatin/genetics , HMGB2 Protein/genetics , Human Umbilical Vein Endothelial Cells , Humans
6.
Oncogene ; 37(20): 2746-2756, 2018 05.
Article in English | MEDLINE | ID: mdl-29503447

ABSTRACT

LIN28B is a RNA-binding protein regulating predominantly let-7 microRNAs with essential functions in inflammation, wound healing, embryonic stem cells, and cancer. LIN28B expression is associated with tumor initiation, progression, resistance, and poor outcome in several solid cancers, including lung cancer. However, the functional role of LIN28B, especially in non-small cell lung adenocarcinomas, remains elusive. Here, we investigated the effects of LIN28B expression on lung tumorigenesis using LIN28B transgenic overexpression in an autochthonous KRASG12V-driven mouse model. We found that LIN28B overexpression significantly increased the number of CD44+/CD326+ tumor cells, upregulated VEGF-A and miR-21 and promoted tumor angiogenesis and epithelial-to-mesenchymal transition (EMT) accompanied by enhanced AKT phosphorylation and nuclear translocation of c-MYC. Moreover, LIN28B accelerated tumor initiation and enhanced proliferation which led to a shortened overall survival. In addition, we analyzed lung adenocarcinomas of the Cancer Genome Atlas (TCGA) and found LIN28B expression in 24% of KRAS-mutated cases, which underscore the relevance of our model.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , RNA-Binding Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Epithelial Cell Adhesion Molecule/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Survival Analysis
7.
Genome Biol ; 16: 7, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25650807

ABSTRACT

Genomic translocation events frequently underlie cancer development through generation of gene fusions with oncogenic properties. Identification of such fusion transcripts by transcriptome sequencing might help to discover new potential therapeutic targets. We developed TRUP (Tumor-specimen suited RNA-seq Unified Pipeline) (https://github.com/ruping/TRUP), a computational approach that combines split-read and read-pair analysis with de novo assembly for the identification of chimeric transcripts in cancer specimens. We apply TRUP to RNA-seq data of different tumor types, and find it to be more sensitive than alternative tools in detecting chimeric transcripts, such as secondary rearrangements in EML4-ALK-positive lung tumors, or recurrent inactivating rearrangements affecting RASSF8.


Subject(s)
Chromosome Breakpoints , Computational Biology , High-Throughput Nucleotide Sequencing , Oncogene Fusion , Transcriptome , Translocation, Genetic , Base Sequence , Cell Line, Tumor , Cluster Analysis , Computational Biology/methods , Gene Silencing , Genomics , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Tumor Suppressor Proteins/genetics
8.
Cancer Discov ; 4(4): 415-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469108

ABSTRACT

UNLABELLED: We discovered a novel somatic gene fusion, CD74-NRG1, by transcriptome sequencing of 25 lung adenocarcinomas of never smokers. By screening 102 lung adenocarcinomas negative for known oncogenic alterations, we found four additional fusion-positive tumors, all of which were of the invasive mucinous subtype. Mechanistically, CD74-NRG1 leads to extracellular expression of the EGF-like domain of NRG1 III-ß3, thereby providing the ligand for ERBB2-ERBB3 receptor complexes. Accordingly, ERBB2 and ERBB3 expression was high in the index case, and expression of phospho-ERBB3 was specifically found in tumors bearing the fusion (P < 0.0001). Ectopic expression of CD74-NRG1 in lung cancer cell lines expressing ERBB2 and ERBB3 activated ERBB3 and the PI3K-AKT pathway, and led to increased colony formation in soft agar. Thus, CD74-NRG1 gene fusions are activating genomic alterations in invasive mucinous adenocarcinomas and may offer a therapeutic opportunity for a lung tumor subtype with, so far, no effective treatment. SIGNIFICANCE: CD74­NRG1 fusions may represent a therapeutic opportunity for invasive mucinous lung adenocarcinomas, a tumor with no effective treatment that frequently presents with multifocal unresectable disease.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Histocompatibility Antigens Class II/genetics , Lung Neoplasms/genetics , Neuregulin-1/genetics , Oncogene Proteins, Fusion/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adenocarcinoma, Mucinous/pathology , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Molecular Sequence Data , NIH 3T3 Cells , Oncogene Proteins, Fusion/metabolism , Sequence Analysis, DNA , Signal Transduction/genetics
9.
J Clin Invest ; 123(4): 1732-40, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23454747

ABSTRACT

The molecular mechanisms that control the balance between antiangiogenic and proangiogenic factors and initiate the angiogenic switch in tumors remain poorly defined. By combining chemical genetics with multimodal imaging, we have identified an autocrine feed-forward loop in tumor cells in which tumor-derived VEGF stimulates VEGF production via VEGFR2-dependent activation of mTOR, substantially amplifying the initial proangiogenic signal. Disruption of this feed-forward loop by chemical perturbation or knockdown of VEGFR2 in tumor cells dramatically inhibited production of VEGF in vitro and in vivo. This disruption was sufficient to prevent tumor growth in vivo. In patients with lung cancer, we found that this VEGF:VEGFR2 feed-forward loop was active, as the level of VEGF/VEGFR2 binding in tumor cells was highly correlated to tumor angiogenesis. We further demonstrated that inhibition of tumor cell VEGFR2 induces feedback activation of the IRS/MAPK signaling cascade. Most strikingly, combined pharmacological inhibition of VEGFR2 (ZD6474) and MEK (PD0325901) in tumor cells resulted in dramatic tumor shrinkage, whereas monotherapy only modestly slowed tumor growth. Thus, a tumor cell-autonomous VEGF:VEGFR2 feed-forward loop provides signal amplification required for the establishment of fully angiogenic tumors in lung cancer. Interrupting this feed-forward loop switches tumor cells from an angiogenic to a proliferative phenotype that sensitizes tumor cells to MAPK inhibition.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood supply , Feedback, Physiological/drug effects , Lung Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Benzamides , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Diphenylamine/analogs & derivatives , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System , Mice , Mice, Nude , Piperidines , Quinazolines , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
10.
Proc Natl Acad Sci U S A ; 109(42): 17034-9, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23035247

ABSTRACT

Small cell lung cancer (SCLC) accounts for about 15% of all lung cancers. The prognosis of SCLC patients is devastating and no biologically targeted therapeutics are active in this tumor type. To develop a framework for development of specific SCLC-targeted drugs we conducted a combined genomic and pharmacological vulnerability screen in SCLC cell lines. We show that SCLC cell lines capture the genomic landscape of primary SCLC tumors and provide genetic predictors for activity of clinically relevant inhibitors by screening 267 compounds across 44 of these cell lines. We show Aurora kinase inhibitors are effective in SCLC cell lines bearing MYC amplification, which occur in 3-7% of SCLC patients. In MYC-amplified SCLC cells Aurora kinase inhibition associates with G2/M-arrest, inactivation of PI3-kinase (PI3K) signaling, and induction of apoptosis. Aurora dependency in SCLC primarily involved Aurora B, required its kinase activity, and was independent of depletion of cytoplasmic levels of MYC. Our study suggests that a fraction of SCLC patients may benefit from therapeutic inhibition of Aurora B. Thus, thorough chemical and genomic exploration of SCLC cell lines may provide starting points for further development of rational targeted therapeutic intervention in this deadly tumor type.


Subject(s)
Enzyme Inhibitors/pharmacology , G2 Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Benzothiazoles , Cell Line, Tumor , Cell Survival/drug effects , DNA Primers/genetics , Diamines , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Immunoblotting , Organic Chemicals , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Quinolines , Reverse Transcriptase Polymerase Chain Reaction
11.
Nat Genet ; 44(10): 1104-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22941188

ABSTRACT

Small-cell lung cancer (SCLC) is an aggressive lung tumor subtype with poor prognosis. We sequenced 29 SCLC exomes, 2 genomes and 15 transcriptomes and found an extremely high mutation rate of 7.4±1 protein-changing mutations per million base pairs. Therefore, we conducted integrated analyses of the various data sets to identify pathogenetically relevant mutated genes. In all cases, we found evidence for inactivation of TP53 and RB1 and identified recurrent mutations in the CREBBP, EP300 and MLL genes that encode histone modifiers. Furthermore, we observed mutations in PTEN, SLIT2 and EPHA7, as well as focal amplifications of the FGFR1 tyrosine kinase gene. Finally, we detected many of the alterations found in humans in SCLC tumors from Tp53 and Rb1 double knockout mice. Our study implicates histone modification as a major feature of SCLC, reveals potentially therapeutically tractable genomic alterations and provides a generalizable framework for the identification of biologically relevant genes in the context of high mutational background.


Subject(s)
Genome, Human , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Amino Acid Substitution , Animals , CREB-Binding Protein/genetics , Cell Line, Tumor , DNA Copy Number Variations , DNA Mutational Analysis , E1A-Associated p300 Protein/genetics , Gene Expression Profiling , Gene Regulatory Networks , Genome-Wide Association Study , Histone-Lysine N-Methyltransferase , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Models, Molecular , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/genetics , Polymorphism, Single Nucleotide , Protein Processing, Post-Translational/genetics , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/genetics
12.
Clin Cancer Res ; 18(17): 4682-90, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22912387

ABSTRACT

PURPOSE: ALK rearrangement-positive lung cancers can be effectively treated with ALK inhibitors. However, the magnitude and duration of response is heterogeneous. In addition, acquired resistance limits the efficacy of ALK inhibitors, with most upfront resistance mechanisms being unknown. EXPERIMENTAL DESIGN: By making use of the Ba/F3 cell line model, we analyzed the cytotoxic efficacy of ALK kinase inhibitors as a function of different EML4-ALK fusion variants v1, v2, v3a, and v3b as well as of three artificially designed EML4-ALK deletion constructs and the ALK fusion genes KIF5b-ALK and NPM1-ALK. In addition, the intracellular localization, the sensitivity to HSP90 inhibition and the protein stability of ALK fusion proteins were studied. RESULTS: Different ALK fusion genes and EML4-ALK variants exhibited differential sensitivity to the structurally diverse ALK kinase inhibitors crizotinib and TAE684. In addition, differential sensitivity correlated with differences in protein stability in EML4-ALK-expressing cells. Furthermore, the sensitivity to HSP90 inhibition also varied depending on the ALK fusion partner but differed from ALK inhibitor sensitivity patterns. Finally, combining inhibitors of ALK and HSP90 resulted in synergistic cytotoxicity. CONCLUSIONS: Our results might explain some of the heterogeneous responses of ALK-positive tumors to ALK kinase inhibition observed in the clinic. Thus, targeted therapy of ALK-positive lung cancer should take into account the precise ALK genotype. Furthermore, combining ALK and HSP90 inhibitors might enhance tumor shrinkage in EML4-ALK-driven tumors.


Subject(s)
Lung Neoplasms , Oncogene Proteins, Fusion , Protein-Tyrosine Kinases , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Adenocarcinoma , Adenocarcinoma of Lung , Anaplastic Lymphoma Kinase , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Crizotinib , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Kinesins/genetics , Kinesins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , NIH 3T3 Cells , Nucleophosmin , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Stability/drug effects , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects
13.
Clin Cancer Res ; 17(23): 7394-401, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21948233

ABSTRACT

PURPOSE: EML4-ALK fusions define a subset of lung cancers that can be effectively treated with anaplastic lymphoma kinase (ALK) inhibitors. Unfortunately, the duration of response is heterogeneous and acquired resistance limits their ultimate efficacy. Thus, a better understanding of resistance mechanisms will help to enhance tumor control in EML4-ALK-positive tumors. EXPERIMENTAL DESIGN: By applying orthogonal functional mutagenesis screening approaches, we screened for mutations inducing resistance to the aminopyridine PF02341066 (crizotinib) and/or the diaminopyrimidine TAE684. RESULTS: Here, we show that the resistance mutation, L1196M, as well as other crizotinib resistance mutations (F1174L and G1269S), are highly sensitive to the structurally unrelated ALK inhibitor TAE684. In addition, we identified two novel EML4-ALK resistance mutations (L1198P and D1203N), which unlike previously reported mutations, induced resistance to both ALK inhibitors. An independent resistance screen in ALK-mutant neuroblastoma cells yielded the same L1198P resistance mutation but defined two additional mutations conferring resistance to TAE684 but not to PF02341066. CONCLUSIONS: Our results show that different ALK resistance mutations as well as different ALK inhibitors impact the therapeutic efficacy in the setting of EML4-ALK fusions and ALK mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/genetics , Drug Resistance, Neoplasm/genetics , Microtubule-Associated Proteins/genetics , Oncogene Proteins, Fusion/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Serine Endopeptidases/genetics , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Crizotinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Polymorphism, Single Nucleotide , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
14.
Cancer Discov ; 1(1): 78-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22328973

ABSTRACT

UNLABELLED: While genomically targeted therapies have improved outcomes for patients with lung adenocarcinoma, little is known about the genomic alterations which drive squamous cell lung cancer. Sanger sequencing of the tyrosine kinome identified mutations in the DDR2 kinase gene in 3.8% of squamous cell lung cancers and cell lines. Squamous lung cancer cell lines harboring DDR2 mutations were selectively killed by knock-down of DDR2 by RNAi or by treatment with the multi-targeted kinase inhibitor dasatinib. Tumors established from a DDR2 mutant cell line were sensitive to dasatinib in xenograft models. Expression of mutated DDR2 led to cellular transformation which was blocked by dasatinib. A squamous cell lung cancer patient with a response to dasatinib and erlotinib treatment harbored a DDR2 kinase domain mutation. These data suggest that gain-of-function mutations in DDR2 are important oncogenic events and are amenable to therapy with dasatinib. As dasatinib is already approved for use, these findings could be rapidly translated into clinical trials. SIGNIFICANCE: DDR2 mutations are present in 4% of lung SCCs, and DDR2 mutations are associated with sensitivity to dasatinib. These findings provide a rationale for designing clinical trials with the FDA-approved drug dasatinib in patients with lung SCCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Dasatinib , Discoidin Domain Receptors , Erlotinib Hydrochloride , Humans , Lung Neoplasms/enzymology , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , src-Family Kinases/genetics
15.
Sci Transl Med ; 2(62): 62ra93, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21160078

ABSTRACT

Lung cancer remains one of the leading causes of cancer-related death in developed countries. Although lung adenocarcinomas with EGFR mutations or EML4-ALK fusions respond to treatment by epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) inhibition, respectively, squamous cell lung cancer currently lacks therapeutically exploitable genetic alterations. We conducted a systematic search in a set of 232 lung cancer specimens for genetic alterations that were therapeutically amenable and then performed high-resolution gene copy number analyses. We identified frequent and focal fibroblast growth factor receptor 1 (FGFR1) amplification in squamous cell lung cancer (n = 155), but not in other lung cancer subtypes, and, by fluorescence in situ hybridization, confirmed the presence of FGFR1 amplifications in an independent cohort of squamous cell lung cancer samples (22% of cases). Using cell-based screening with the FGFR inhibitor PD173074 in a large (n = 83) panel of lung cancer cell lines, we demonstrated that this compound inhibited growth and induced apoptosis specifically in those lung cancer cells carrying amplified FGFR1. We validated the FGFR1 dependence of FGFR1-amplified cell lines by FGFR1 knockdown and by ectopic expression of an FGFR1-resistant allele (FGFR1(V561M)), which rescued FGFR1-amplified cells from PD173074-mediated cytotoxicity. Finally, we showed that inhibition of FGFR1 with a small molecule led to significant tumor shrinkage in vivo. Thus, focal FGFR1 amplification is common in squamous cell lung cancer and associated with tumor growth and survival, suggesting that FGFR inhibitors may be a viable therapeutic option in this cohort of patients.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/drug therapy , Male , Mice , Mice, Nude , Pyrimidines/therapeutic use , RNA Interference , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Xenograft Model Antitumor Assays
16.
PLoS One ; 5(1): e8919, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20111714

ABSTRACT

Despite the successful introduction of potent anti-cancer therapeutics, most of these drugs lead to only modest tumor-shrinkage or transient responses, followed by re-growth of tumors. Combining different compounds has resulted in enhanced tumor control and prolonged survival. However, methods querying the efficacy of such combinations have been hampered by limited scalability, analytical resolution, statistical feasibility, or a combination thereof. We have developed a theoretical framework modeling cellular viability as a stochastic lifetime process to determine synergistic compound combinations from high-throughput cellular screens. We apply our method to data derived from chemical perturbations of 65 cancer cell lines with two inhibitors. Our analysis revealed synergy for the combination of both compounds in subsets of cell lines. By contrast, in cell lines in which inhibition of one of both targets was sufficient to induce cell death, no synergy was detected, compatible with the topology of the oncogenically activated signaling network. In summary, we provide a tool for the measurement of synergy strength for combination perturbation experiments that might help define pathway topologies and direct clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Models, Theoretical , Cell Line, Tumor , Drug Synergism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Stochastic Processes
17.
Cancer Res ; 70(3): 868-74, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20103621

ABSTRACT

Reversible epidermal growth factor receptor (EGFR) inhibitors are the first class of small molecules to improve progression-free survival of patients with EGFR-mutated lung cancers. Second-generation EGFR inhibitors introduced to overcome acquired resistance by the T790M resistance mutation of EGFR have thus far shown limited clinical activity in patients with T790M-mutant tumors. In this study, we systematically analyzed the determinants of the activity and selectivity of the second-generation EGFR inhibitors. A focused library of irreversible as well as structurally corresponding reversible EGFR-inhibitors was synthesized for chemogenomic profiling involving over 79 genetically defined NSCLC and 19 EGFR-dependent cell lines. Overall, our results show that the growth-inhibitory potency of all irreversible inhibitors against the EGFR(T790M) resistance mutation was limited by reduced target inhibition, linked to decreased binding velocity to the mutant kinase. Combined treatment of T790M-mutant tumor cells with BIBW-2992 and the phosphoinositide-3-kinase/mammalian target of rapamycin inhibitor PI-103 led to synergistic induction of apoptosis. Our findings offer a mechanistic explanation for the limited efficacy of irreversible EGFR inhibitors in EGFR(T790M) gatekeeper-mutant tumors, and they prompt combination treatment strategies involving inhibitors that target signaling downstream of the EGFR.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Afatinib , Amino Acid Substitution , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , ErbB Receptors/metabolism , Furans/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/classification , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects
18.
Proc Natl Acad Sci U S A ; 106(43): 18351-6, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19805051

ABSTRACT

In cancer, genetically activated proto-oncogenes often induce "upstream" dependency on the activity of the mutant oncoprotein. Therapeutic inhibition of these activated oncoproteins can induce massive apoptosis of tumor cells, leading to sometimes dramatic tumor regressions in patients. The PI3K and MAPK signaling pathways are central regulators of oncogenic transformation and tumor maintenance. We hypothesized that upstream dependency engages either one of these pathways preferentially to induce "downstream" dependency. Therefore, we analyzed whether downstream pathway dependency segregates by genetic aberrations upstream in lung cancer cell lines. Here, we show by systematically linking drug response to genomic aberrations in non-small-cell lung cancer, as well as in cell lines of other tumor types and in a series of in vivo cancer models, that tumors with genetically activated receptor tyrosine kinases depend on PI3K signaling, whereas tumors with mutations in the RAS/RAF axis depend on MAPK signaling. However, efficacy of downstream pathway inhibition was limited by release of negative feedback loops on the reciprocal pathway. By contrast, combined blockade of both pathways was able to overcome the reciprocal pathway activation induced by inhibitor-mediated release of negative feedback loops and resulted in a significant increase in apoptosis and tumor shrinkage. Thus, by using a systematic chemo-genomics approach, we identify genetic lesions connected to PI3K and MAPK pathway activation and provide a rationale for combined inhibition of both pathways. Our findings may have implications for patient stratification in clinical trials.


Subject(s)
MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Genotype , Humans , Neoplasms/enzymology , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors
19.
J Clin Invest ; 119(6): 1727-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19451690

ABSTRACT

Somatic genetic alterations in cancers have been linked with response to targeted therapeutics by creation of specific dependency on activated oncogenic signaling pathways. However, no tools currently exist to systematically connect such genetic lesions to therapeutic vulnerability. We have therefore developed a genomics approach to identify lesions associated with therapeutically relevant oncogene dependency. Using integrated genomic profiling, we have demonstrated that the genomes of a large panel of human non-small cell lung cancer (NSCLC) cell lines are highly representative of those of primary NSCLC tumors. Using cell-based compound screening coupled with diverse computational approaches to integrate orthogonal genomic and biochemical data sets, we identified molecular and genomic predictors of therapeutic response to clinically relevant compounds. Using this approach, we showed that v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations confer enhanced Hsp90 dependency and validated this finding in mice with KRAS-driven lung adenocarcinoma, as these mice exhibited dramatic tumor regression when treated with an Hsp90 inhibitor. In addition, we found that cells with copy number enhancement of v-abl Abelson murine leukemia viral oncogene homolog 2 (ABL2) and ephrin receptor kinase and v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (avian) (SRC) kinase family genes were exquisitely sensitive to treatment with the SRC/ABL inhibitor dasatinib, both in vitro and when it xenografted into mice. Thus, genomically annotated cell-line collections may help translate cancer genomics information into clinical practice by defining critical pathway dependencies amenable to therapeutic inhibition.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Profiling , Humans , Magnetic Resonance Imaging , Mice , Models, Molecular , Mutation/genetics , Phenotype , Protein Structure, Tertiary , Substrate Specificity
20.
Cancer Res ; 69(8): 3256-61, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19351834

ABSTRACT

Clinical resistance to epidermal growth factor receptor (EGFR) inhibition in lung cancer has been linked to the emergence of the EGFR T790M resistance mutation or amplification of MET. Additional mechanisms contributing to EGFR inhibitor resistance remain elusive. By applying combined analyses of gene expression, copy number, and biochemical analyses of EGFR inhibitor responsiveness, we identified homozygous loss of PTEN to segregate EGFR-dependent and EGFR-independent cells. We show that in EGFR-dependent cells, PTEN loss partially uncouples mutant EGFR from downstream signaling and activates EGFR, thereby contributing to erlotinib resistance. The clinical relevance of our findings is supported by the observation of PTEN loss in 1 out of 24 primary EGFR-mutant non-small cell lung cancer (NSCLC) tumors. These results suggest a novel resistance mechanism in EGFR-mutant NSCLC involving PTEN loss.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/metabolism , Lung Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Chromosome Aberrations , Cluster Analysis , Drug Resistance, Neoplasm , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Gene Deletion , Gene Expression , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , PTEN Phosphohydrolase/deficiency , Protein Kinase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL