Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Phys Rev Lett ; 125(5): 051302, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32794885

ABSTRACT

Certain anomalies in the CMB bring out a tension between the six-parameter flat ΛCDM model and the CMB data. We revisit the PLANCK analysis with loop quantum cosmology (LQC) predictions and show that LQC alleviates both the large-scale power anomaly and the tension in the lensing amplitude. These differences arise because, in LQC, the primordial power spectrum is scale dependent for small k, with a specific power suppression. We conclude with a prediction of larger optical depth and power suppression in the B-mode polarization power spectrum on large scales.

2.
Phys Rev Lett ; 124(25): 251301, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32639780

ABSTRACT

We introduce an extension of the standard inflationary paradigm on which the big bang singularity is replaced by an anisotropic bounce. Unlike in the big bang model, cosmological perturbations find an adiabatic regime in the past. We show that this scenario accounts for the observed quadrupolar modulation in the temperature anisotropies of the cosmic microwave background, and we make predictions for the polarization angular correlation functions E-E, B-B, and E-B, together with temperature-polarization correlations T-B and T-E, that can be used to test our ideas. We base our calculations on the bounce predicted by loop quantum cosmology, but our techniques and conclusions apply to other bouncing models as well.

3.
Cancer Res ; 75(22): 4937-48, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26490646

ABSTRACT

Patients with lung tumors harboring activating mutations in the EGF receptor (EGFR) show good initial treatment responses to the EGFR tyrosine kinase inhibitors (TKI) erlotinib or gefitinib. However, acquired resistance invariably develops. Applying a focused shRNA screening approach to identify genes whose knockdown can prevent and/or overcome acquired resistance to erlotinib in several EGFR-mutant non-small cell lung cancer (NSCLC) cell lines, we identified casein kinase 1 α (CSNK1A1, CK1α). We found that CK1α suppression inhibits the NF-κB prosurvival signaling pathway. Furthermore, downregulation of NF-κB signaling by approaches independent of CK1α knockdown can also attenuate acquired erlotinib resistance, supporting a role for activated NF-κB signaling in conferring acquired drug resistance. Importantly, CK1α suppression prevented erlotinib resistance in an HCC827 xenograft model in vivo. Our findings suggest that patients with EGFR-mutant NSCLC might benefit from a combination of EGFR TKIs and CK1α inhibition to prevent acquired drug resistance and to prolong disease-free survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Casein Kinase I/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Erlotinib Hydrochloride/pharmacology , Female , Gene Knockdown Techniques , Genes, erbB-1/genetics , Humans , Immunoblotting , Lung Neoplasms/enzymology , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
4.
Nature ; 483(7391): 570-5, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22460902

ABSTRACT

Clinical responses to anticancer therapies are often restricted to a subset of patients. In some cases, mutated cancer genes are potent biomarkers for responses to targeted agents. Here, to uncover new biomarkers of sensitivity and resistance to cancer therapeutics, we screened a panel of several hundred cancer cell lines--which represent much of the tissue-type and genetic diversity of human cancers--with 130 drugs under clinical and preclinical investigation. In aggregate, we found that mutated cancer genes were associated with cellular response to most currently available cancer drugs. Classic oncogene addiction paradigms were modified by additional tissue-specific or expression biomarkers, and some frequently mutated genes were associated with sensitivity to a broad range of therapeutic agents. Unexpected relationships were revealed, including the marked sensitivity of Ewing's sarcoma cells harbouring the EWS (also known as EWSR1)-FLI1 gene translocation to poly(ADP-ribose) polymerase (PARP) inhibitors. By linking drug activity to the functional complexity of cancer genomes, systematic pharmacogenomic profiling in cancer cell lines provides a powerful biomarker discovery platform to guide rational cancer therapeutic strategies.


Subject(s)
Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Genes, Neoplasm/genetics , Genetic Markers/genetics , Genome, Human/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genomics , Humans , Indoles/pharmacology , Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Pharmacogenetics , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
5.
Oncotarget ; 2(6): 518-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21680956

ABSTRACT

STAT transcription factors transduce signals from the cell surface to the nucleus, where they regulate the expression of genes that control proliferation, survival, self-renewal, and other critical cellular functions. Under normal physiological conditions, the activation of STATs is tightly regulated. In cancer, by contrast, STAT proteins, particularly STAT3 and STAT5, become activated constitutively, thereby driving the malignant phenotype of cancer cells. Since these proteins are largely dispensable in the function of normal adult cells, STATs represent a potentially important target for cancer therapy. Although transcription factors have traditionally been viewed as suboptimal targets for pharmacological inhibition, chemical biology approaches have been particularly fruitful in identifying compounds that can modulate this pathway through a variety of mechanisms. STAT inhibitors have notable anti-cancer effects in many tumor systems, show synergy with other therapeutic modalities, and have the potential to eradicate tumor stem cells. Furthermore, STAT inhibitors identified through the screening of chemical libraries can then be employed in large scale analyses such as gene expression profiling, RNA interference screens, or large-scale tumor cell line profiling. Data derived from these studies can then provide key insights into mechanisms of STAT signal transduction, as well as inform the rational design of targeted therapeutic strategies for cancer patients.


Subject(s)
Clinical Trials as Topic/methods , Drug Design , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , STAT Transcription Factors/antagonists & inhibitors , Adult , Humans , Time Factors , Translational Research, Biomedical/methods
6.
Cell Cycle ; 9(18): 3710-4, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20930519

ABSTRACT

Impaired regulation of kinase activity can lead to a variety of diseases, including cancer. Inhibition of kinase activity has, therefore, been considered an attractive anti-cancer therapeutic strategy. The success of targeted therapy with kinase inhibitors has been well documented with BCR-ABL, where imatinib specifically inhibits kinase activity with impressive pharmacological responses in chronic myelogenous leukemia (CML). However, the success of kinase inhibitors as cancer therapeutics is being challenged clinically by the emergence of acquired resistance. Most kinase inhibitors available today are ATP-competitive. There have been efforts to develop kinase inhibitors with new modes of action. In this review, we highlight the development of 'allosteric kinase inhibitors' that inhibit kinase activity by binding to a site remote from the active site of the kinase. We focus on recent efforts directed towards BCR-ABL, for which, significant progress has been made to develop allosteric inhibitors with promising therapeutic activity, especially in the context of overcoming clinically acquired resistance mutations to the first generation of ATP-competitive kinase inhibitors.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Allosteric Regulation , Benzamides , Dasatinib , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/chemistry , Piperazines/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Thiazoles/chemistry , Thiazoles/therapeutic use
7.
Cell ; 141(1): 69-80, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371346

ABSTRACT

Accumulating evidence implicates heterogeneity within cancer cell populations in the response to stressful exposures, including drug treatments. While modeling the acute response to various anticancer agents in drug-sensitive human tumor cell lines, we consistently detected a small subpopulation of reversibly "drug-tolerant" cells. These cells demonstrate >100-fold reduced drug sensitivity and maintain viability via engagement of IGF-1 receptor signaling and an altered chromatin state that requires the histone demethylase RBP2/KDM5A/Jarid1A. This drug-tolerant phenotype is transiently acquired and relinquished at low frequency by individual cells within the population, implicating the dynamic regulation of phenotypic heterogeneity in drug tolerance. The drug-tolerant subpopulation can be selectively ablated by treatment with IGF-1 receptor inhibitors or chromatin-modifying agents, potentially yielding a therapeutic opportunity. Together, these findings suggest that cancer cell populations employ a dynamic survival strategy in which individual cells transiently assume a reversibly drug-tolerant state to protect the population from eradication by potentially lethal exposures.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/pathology , Cell Line, Tumor , Chromatin/metabolism , Chromatin/pathology , DNA Damage , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Neoplasms/metabolism , Receptor, IGF Type 1/metabolism
8.
Biochem Pharmacol ; 80(5): 666-73, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20211150

ABSTRACT

Rational approaches to targeted cancer therapy have begun to predominate the pipelines of oncology drug development. Our rapidly increasing understanding of the "wiring" of tumor cells and the vulnerabilities of such cells that can potentially be exploited through targeted treatments has opened up enormous opportunities for improved therapies. Accumulating evidence suggests that many of these vulnerabilities reflect states of dependency or "addiction" that are unique to cancer cells (versus normal cells). Such addiction can arise due to a strict dependency on a single activated oncogene, a cell lineage-specific factor, or even to a non-oncogene, and identifying these "Achilles' heels" within individual tumors remains an important challenge to the development of targeted therapies. Recent technology advances that facilitate high-throughput genomic analysis of tumor specimens and genome-wide RNA interference screening in cancer cell lines are key among the newly developed tools that are beginning to reveal novel context-dependent therapeutic targets, and the rapidly increasing application of these technologies by a large number of laboratories will undoubtedly lead to more effective cancer therapies in the near future. Here, we review the various forms of cancer cell addiction and their relevance to the discovery of novel therapeutic targets.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Neoplasms/drug therapy , Carcinogens , Humans , Neoplasms/genetics , Neoplasms/pathology
9.
Nat Rev Cancer ; 10(4): 241-53, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20300105

ABSTRACT

Efforts to discover new cancer drugs and predict their clinical activity are limited by the fact that laboratory models to test drug efficacy do not faithfully recapitulate this complex disease. One important model system for evaluating candidate anticancer agents is human tumour-derived cell lines. Although cultured cancer cells can exhibit distinct properties compared with their naturally growing counterparts, recent technologies that facilitate the parallel analysis of large panels of such lines, together with genomic technologies that define their genetic constitution, have revitalized efforts to use cancer cell lines to assess the clinical utility of new investigational cancer drugs and to discover predictive biomarkers.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Treatment Outcome , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/drug effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Drug Synergism , ErbB Receptors/genetics , Gene Expression Profiling , Genomics/methods , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Predictive Value of Tests
10.
Chem Biol ; 17(3): 285-95, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20338520

ABSTRACT

The fibroblast growth factor receptor tyrosine kinases (FGFR1, 2, 3, and 4) represent promising therapeutic targets in a number of cancers. We have developed the first potent and selective irreversible inhibitor of FGFR1, 2, 3, and 4, which we named FIIN-1 that forms a covalent bond with cysteine 486 located in the P loop of the FGFR1 ATP binding site. We demonstrated that the inhibitor potently inhibits Tel-FGFR1-transformed Ba/F3 cells (EC(50) = 14 nM) as well as numerous FGFR-dependent cancer cell lines. A biotin-derivatized version of the inhibitor, FIIN-1-biotin, was shown to covalently label FGFR1 at Cys486. FIIN-1 is a useful probe of FGFR-dependent cellular phenomena and may provide a starting point of the development of therapeutically relevant irreversible inhibitors of wild-type and drug-resistant forms of FGFR kinases.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cysteine/metabolism , Humans , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
11.
Clin Cancer Res ; 16(1): 109-20, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028770

ABSTRACT

PURPOSE: Cyclin-dependent kinases (Cdk) and their associated cyclins are targets for lung cancer therapy and chemoprevention given their frequent deregulation in lung carcinogenesis. This study uncovered previously unrecognized consequences of targeting the cyclin E-Cdk-2 complex in lung cancer. EXPERIMENTAL DESIGN: Cyclin E, Cdk-1, and Cdk-2 were individually targeted for repression with siRNAs in lung cancer cell lines. Cdk-2 was also pharmacologically inhibited with the reversible kinase inhibitor seliciclib. Potential reversibility of seliciclib effects was assessed in washout experiments. Findings were extended to a large panel of cancer cell lines using a robotic-based platform. Consequences of cyclin E-Cdk-2 inhibition on chromosome stability and on in vivo tumorigenicity were explored as were effects of combining seliciclib with different taxanes in lung cancer cell lines. RESULTS: Targeting the cyclin E-Cdk-2 complex, but not Cdk-1, resulted in marked growth inhibition through the induction of multipolar anaphases triggering apoptosis. Treatment with the Cdk-2 kinase inhibitor seliciclib reduced lung cancer formation in a murine syngeneic lung cancer model and decreased immunohistochemical detection of the proliferation markers Ki-67 and cyclin D1 in lung dysplasia spontaneously arising in a transgenic cyclin E-driven mouse model. Combining seliciclib with a taxane resulted in augmented growth inhibition and apoptosis in lung cancer cells. Pharmacogenomic analysis revealed that lung cancer cell lines with mutant ras were especially sensitive to seliciclib. CONCLUSIONS: Induction of multipolar anaphases leading to anaphase catastrophe is a previously unrecognized mechanism engaged by targeting the cyclin E-Cdk-2 complex. This exerts substantial antineoplastic effects in the lung.


Subject(s)
Anaphase/drug effects , Antineoplastic Agents/pharmacology , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Cyclin E/antagonists & inhibitors , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Docetaxel , Drug Delivery Systems , Mice , Mice, Transgenic , Purines/pharmacology , Roscovitine , Taxoids/pharmacology
12.
Cancer Res ; 69(9): 3937-46, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19366796

ABSTRACT

Platelet-derived growth factor (PDGF) receptors (PDGFR) and their ligands play critical roles in several human malignancies. Sunitinib is a clinically approved multitargeted tyrosine kinase inhibitor that inhibits vascular endothelial growth factor receptor, c-KIT, and PDGFR, and has shown clinical activity in various solid tumors. Activation of PDGFR signaling has been described in gastrointestinal stromal tumors (PDGFRA mutations) as well as in chronic myeloid leukemia (BCR-PDGFRA translocation), and sunitinib can yield clinical benefit in both settings. However, the discovery of PDGFR activating mutations or gene rearrangements in other tumor types could reveal additional patient populations who might benefit from treatment with anti-PDGFR therapies, such as sunitinib. Using a high-throughput cancer cell line screening platform, we found that only 2 of 637 tested human tumor-derived cell lines show significant sensitivity to single-agent sunitinib exposure. These two cell lines [a non-small-cell lung cancer (NSCLC) and a rhabdomyosarcoma] showed expression of highly phosphorylated PDGFRA. In the sunitinib-sensitive adenosquamous NSCLC cell line, PDGFRA expression was associated with focal PFGRA gene amplification, which was similarly detected in a small fraction of squamous cell NSCLC primary tumor specimens. Moreover, in this NSCLC cell line, focal amplification of the gene encoding the PDGFR ligand PDGFC was also detected, and silencing PDGFRA or PDGFC expression by RNA interference inhibited proliferation. A similar codependency on PDGFRA and PDGFC was observed in the sunitinib-sensitive rhabdomyosarcoma cell line. These findings suggest that, in addition to gastrointestinal stromal tumors, rare tumors that show PDGFC-mediated PDGFRA activation may also be clinically responsive to pharmacologic PDGFRA or PDGFC inhibition.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Indoles/pharmacology , Lung Neoplasms/drug therapy , Pyrroles/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Rhabdomyosarcoma/drug therapy , Antibodies/immunology , Antibodies/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Gene Amplification , Gene Expression Profiling , Humans , Ligands , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lymphokines/genetics , Lymphokines/immunology , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/immunology , RNA, Small Interfering/genetics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/genetics , Rhabdomyosarcoma/enzymology , Rhabdomyosarcoma/genetics , Sunitinib
13.
Gastroenterology ; 136(5): 1741-9.e6, 2009 May.
Article in English | MEDLINE | ID: mdl-19208345

ABSTRACT

BACKGROUND & AIMS: The Notch signaling pathway is required for the expansion of undifferentiated pancreatic progenitor cells during embryonic development and has been implicated in the progression of pancreatic ductal adenocarcinoma (PDAC). The interaction of Notch ligands with their receptors promotes a gamma-secretase-dependent cleavage of the Notch receptor and release of the Notch intracellular domain, which translocates to the nucleus and activates transcription. We investigated the role of this pathway in PDAC progression. METHODS: We tested the effects of a gamma-secretase inhibitor (GSI) that blocks Notch signaling in PDAC cell lines and a genetically engineered mouse model of PDAC (Kras p53 L/+ mice). RESULTS: Notch signaling was activated in PDAC precursors and advanced tumors. The GSI inhibited the growth of premalignant pancreatic duct-derived cells in a Notch-dependent manner. Additionally, in a panel of over 400 human solid tumor-derived cell lines, PDAC cells, as a group, were more sensitive to the GSI than any other tumor type. Finally, the GSI completely inhibited tumor development in the genetically engineered model of invasive PDAC (P < .005, chi2 test; compared with mice exposed to vehicle). CONCLUSIONS: These results suggest that Notch signaling is required for PDAC progression. Pharmacologic targeting of this pathway offers therapeutic potential in this treatment-refractory malignancy.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Notch/physiology , Signal Transduction , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line , Cell Line, Tumor , Cyclic S-Oxides/pharmacology , Disease Progression , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Thiadiazoles/pharmacology
14.
Exp Cell Res ; 315(4): 557-71, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18721806

ABSTRACT

Lung cancer remains the leading cause of cancer deaths worldwide, and advanced stage disease is largely refractory to conventional chemotherapy. Thus, there is an important need for alternative treatment strategies, and the ErbB proteins have emerged as potentially important therapeutic drug targets in this setting, apparently reflecting a state of "oncogene addiction" in some lung tumors. In this review, we discuss the recent identification of mutations that promote activation of ErbB family proteins in a subset of lung cancers, and the development of selective inhibitors of these proteins that have demonstrated clinical efficacy. We also discuss the problem of drug resistance, which severely limits the clinical utility of such agents, and has prompted intense efforts to better understand molecular mechanisms underlying drug resistance as well as strategies to overcome or prevent such resistance.


Subject(s)
Genes, erbB/genetics , Lung Neoplasms/physiopathology , Drug Delivery Systems , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Signal Transduction
15.
Anal Biochem ; 385(2): 314-20, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19103144

ABSTRACT

Identification of drug targets is a key step in the development of novel pharmaceuticals. To this end, chemical probes or affinity matrices are often used, requiring substantial structure-activity relationship (SAR) studies. Here we report on the development of a novel technique for drug-target identification from total cellular lysate conducted independently of SAR information. This technique relies on binding of a drug to its target inducing a conformational change in target protein, thereby altering its susceptibility to proteolysis and resulting in specific degradation in some cases or in protection of target protein in others. As proof of concept, three drugs with identified targets were used. First, incubation of cellular lysates with okadaic acid elicited a specific protective effect on its target, protein phosphatase 2A catalytic subunit. Second, specific protection from exogenous protease of FKBP12 by FK506 and Hsp90 fragments by radicicol were observed. We then used the method to validate the targets of UCS15A, an Src signaling inhibitor. UCS15A induced proteolysis of a number of proteins, one of which was identified as Sam68. These studies suggest that the technology may be generally useful for identification and validation of drug targets.


Subject(s)
Drug Delivery Systems , Pharmaceutical Preparations/chemistry , Proteins/drug effects , Benzaldehydes/pharmacology , Cells , Macrolides/pharmacology , Okadaic Acid/pharmacology , Peptide Hydrolases/drug effects , Protease Inhibitors , Protein Conformation/drug effects , Protein Phosphatase 2/drug effects , Proteins/chemistry
16.
Clin Cancer Res ; 14(21): 6867-76, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18980981

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) kinase inhibitors induce dramatic clinical responses in a subset of non-small cell lung cancer (NSCLC) patients with advanced disease, and such responses are correlated with the presence of somatic activating mutations within the EGFR kinase domain. Consequently, one of these inhibitors, erlotinib, has been Food and Drug Administration-approved as a second- or third-line treatment for chemotherapy-refractory advanced NSCLC. However, responses are typically relatively short-lived due to acquired drug resistance, prompting studies to determine whether first-line treatment with EGFR inhibitors could provide greater clinical benefit. NSCLC-derived cell lines have provided a powerful system for modeling EGFR mutation-correlated sensitivity to EGFR inhibitors and for modeling mechanisms of acquired drug resistance that are observed clinically. EXPERIMENTAL DESIGN: In a cell culture model of an erlotinib-sensitive EGFR-mutant NSCLC cell line, we tested the hypothesis that prior exposure to platinum agents, a standard component of NSCLC chemotherapy treatment, affects the subsequent response to erlotinib. RESULTS: Indeed, NSCLC cells initially selected for growth in cisplatin exhibit 5-fold reduced sensitivity to erlotinib, even after propagating the cisplatin-treated cells in the absence of cisplatin for several months. This lingering effect of cisplatin exposure appears to reflect changes in PTEN tumor suppressor activity and persistent EGFR-independent signaling through the phosphatidylinositol 3-kinase/AKT survival pathway. CONCLUSIONS: These preclinical findings suggest that first-line chemotherapy treatment of EGFR-mutant NSCLCs may reduce the benefit of subsequent treatment with EGFR kinase inhibitors and should prompt further clinical investigation of these inhibitors as a first-line therapy in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin/administration & dosage , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Quinazolines/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Drug Resistance, Neoplasm , Erlotinib Hydrochloride , Humans , Mutation , Oncogene Protein v-akt , Time Factors , Tumor Cells, Cultured
17.
Cancer Res ; 68(12): 4853-61, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559533

ABSTRACT

Activating BRAF kinase mutations arise in approximately 7% of all human tumors, and preclinical studies have validated the RAF-mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase-ERK signaling cascade as a potentially important therapeutic target in this setting. Selective RAF kinase inhibitors are currently undergoing clinical development, and based on the experience with other kinase-targeted therapeutics, it is expected that clinical responses to these agents, if observed, will lead to the eventual emergence of drug resistance in most cases. Thus, it is important to establish molecular mechanisms underlying such resistance to develop effective therapeutic strategies to overcome or prevent drug resistance. To anticipate potential mechanisms of acquired resistance to RAF inhibitors during the course of treatment, we established drug-resistant clones from a human melanoma-derived cell line harboring the recurrent V600E activating BRAF mutation, which exhibits exquisite sensitivity to AZ628, a selective RAF kinase inhibitor. We determined that elevated CRAF protein levels account for the acquisition of resistance to AZ628 in these cells, associated with a switch from BRAF to CRAF dependency in tumor cells. We also found that elevated CRAF protein levels may similarly contribute to primary insensitivity to RAF inhibition in a subset of BRAF mutant tumor cells. Interestingly, AZ628-resistant cells demonstrating either primary drug insensitivity or acquired drug resistance exhibit exquisite sensitivity to the HSP90 inhibitor geldanamycin. Geldanamycin effectively promotes the degradation of CRAF, thereby revealing a potential therapeutic strategy to overcome resistance to RAF inhibition in a subset of BRAF mutant tumors.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Cell Proliferation/drug effects , Humans , In Situ Hybridization, Fluorescence , MAP Kinase Kinase 1/metabolism , Melanoma/metabolism , Melanoma/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Phosphorylation/drug effects , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , Up-Regulation
18.
Methods Enzymol ; 438: 331-41, 2008.
Article in English | MEDLINE | ID: mdl-18413259

ABSTRACT

Human cancer cell lines that can be propagated and manipulated in culture have proven to be excellent models for studying many aspects of gene function in cancer. In addition, they can provide a powerful system for assessing the molecular determinants of sensitivity to anticancer drugs. They have also been used in recent studies to identify genomic alterations and gene expression patterns that provide important insights into the genetic features that distinguish the properties of tumor cells associated with similar histologies. We have established a large repository of human tumor cell lines (>1000) corresponding to a wide variety of tumor types, and we have developed a methodology for profiling the collection for sensitivity to putative anticancer compounds. The rationale for examining tumor cell lines on this relatively large scale reflects accumulating evidence indicating that there is substantial genetic heterogeneity among human tumor cells-even those derived from tumors of similar histologies. Thus, to develop an accurate picture of the molecular determinants of tumorigenesis and response to therapy, it is essential to study the nature of such heterogeneity in a relatively large sample set. Here, we describe the methodologies used to conduct such screens and we describe a "proof-of-concept" screen using the EGFR kinase inhibitor, erlotinib (Tarceva), with a panel of lung cancer lines to demonstrate a correlation between EGFR mutations and drug sensitivity.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/genetics , Quinazolines/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Erlotinib Hydrochloride , Humans , Lung Neoplasms/drug therapy
19.
Genes Dev ; 21(24): 3214-31, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18079171

ABSTRACT

In pugilistic parlance, the one-two punch is a devastating combination of blows, with the first punch setting the stage and the second delivering the knock-out. This analogy can be extended to molecularly targeted cancer therapies, with oncogene addiction serving to set the stage for tumor cell killing by a targeted therapeutic agent. While in vitro and in vivo examples abound documenting the existence of this phenomenon, the mechanistic underpinnings that govern oncogene addiction are just beginning to emerge. Our current inability to fully exploit this weakness of cancer cells stems from an incomplete understanding of oncogene addiction, which nonetheless represents one of the rare chinks in the formidable armor of cancer cells.


Subject(s)
Neoplasms/therapy , Oncogenes , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology
20.
Proc Natl Acad Sci U S A ; 104(50): 19936-41, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077425

ABSTRACT

Kinase inhibitors constitute an important new class of cancer drugs, whose selective efficacy is largely determined by underlying tumor cell genetics. We established a high-throughput platform to profile 500 cell lines derived from diverse epithelial cancers for sensitivity to 14 kinase inhibitors. Most inhibitors were ineffective against unselected cell lines but exhibited dramatic cell killing of small nonoverlapping subsets. Cells with exquisite sensitivity to EGFR, HER2, MET, or BRAF kinase inhibitors were marked by activating mutations or amplification of the drug target. Although most cell lines recapitulated known tumor-associated genotypes, the screen revealed low-frequency drug-sensitizing genotypes in tumor types not previously associated with drug susceptibility. Furthermore, comparing drugs thought to target the same kinase revealed striking differences, predictive of clinical efficacy. Genetically defined cancer subsets, irrespective of tissue type, predict response to kinase inhibitors, and provide an important preclinical model to guide early clinical applications of novel targeted inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Genotype , Humans , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...