Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
NPJ Breast Cancer ; 7(1): 60, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34040000

ABSTRACT

The effect of scheduling of targeted therapy combinations on drug resistance is underexplored in triple-negative breast cancer (TNBC). TNBC constitutes heterogeneous cancer cell populations the composition of which can change dynamically during treatment resulting in the selection of resistant clones with a fitness advantage. We evaluated crizotinib (ALK/MET inhibitor) and navitoclax (ABT-263; Bcl-2/Bcl-xL inhibitor) combinations in a large design consisting of 696 two-cycle sequential and concomitant treatment regimens with varying treatment dose, duration, and drug holiday length over a 26-day period in MDA-MB-231 TNBC cells and found that patterns of resistance depend on the schedule and sequence in which the drugs are given. Further, we tracked the clonal dynamics and mechanisms of resistance using DNA-integrated barcodes and single-cell RNA sequencing. Our study suggests that longer formats of treatment schedules in vitro screening assays are required to understand the effects of resistance and guide more realistically in vivo and clinical studies.

2.
Mol Syst Biol ; 17(3): e9810, 2021 03.
Article in English | MEDLINE | ID: mdl-33769711

ABSTRACT

Identifying cooperating modules of driver alterations can provide insights into cancer etiology and advance the development of effective personalized treatments. We present Cancer Rule Set Optimization (CRSO) for inferring the combinations of alterations that cooperate to drive tumor formation in individual patients. Application to 19 TCGA cancer types revealed a mean of 11 core driver combinations per cancer, comprising 2-6 alterations per combination and accounting for a mean of 70% of samples per cancer type. CRSO is distinct from methods based on statistical co-occurrence, which we demonstrate is a suboptimal criterion for investigating driver cooperation. CRSO identified well-studied driver combinations that were not detected by other approaches and nominated novel combinations that correlate with clinical outcomes in multiple cancer types. Novel synergies were identified in NRAS-mutant melanomas that may be therapeutically relevant. Core driver combinations involving NFE2L2 mutations were identified in four cancer types, supporting the therapeutic potential of NRF2 pathway inhibition. CRSO is available at https://github.com/mikekleinsgit/CRSO/.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Computer Simulation , DNA Copy Number Variations/genetics , Databases, Genetic , Genes, Neoplasm , Humans
3.
Cancer Res ; 80(3): 524-535, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31690671

ABSTRACT

Acquired resistance to HER2-targeted therapies occurs frequently in HER2+ breast tumors and new strategies for overcoming resistance are needed. Here, we report that resistance to trastuzumab is reversible, as resistant cells regained sensitivity to the drug after being cultured in drug-free media. RNA-sequencing analysis showed that cells resistant to trastuzumab or trastuzumab + pertuzumab in combination increased expression of oxidative phosphorylation pathway genes. Despite minimal changes in mitochondrial respiration, these cells exhibited increased expression of ATP synthase genes and selective dependency on ATP synthase function. Resistant cells were sensitive to inhibition of ATP synthase by oligomycin A, and knockdown of ATP5J or ATP5B, components of ATP synthase complex, rendered resistant cells responsive to a low dose of trastuzumab. Furthermore, combining ATP synthase inhibitor oligomycin A with trastuzumab led to regression of trastuzumab-resistant tumors in vivo. In conclusion, we identify a novel vulnerability of cells with acquired resistance to HER2-targeted antibody therapies and reveal a new therapeutic strategy to overcome resistance. SIGNIFICANCE: These findings implicate ATP synthase as a novel potential target for tumors resistant to HER2-targeted therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligomycins/administration & dosage , Trastuzumab/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncotarget ; 10(47): 4822-4839, 2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31448050

ABSTRACT

Mesenchymal stem-like (MSL) breast cancers are enriched for cells with tumor reconstituting and mesenchymal characteristics. These cancers are often triple-negative and have a poor prognosis. Few effective targeted treatment options exist for patients with these cancers, and even when targeted therapies exist, resistance often arises and tumors recur, due in part to drug-tolerant persisting tumor cells with self-renewal capability. Effective treatment strategies will combine agents that target the bulk-tumor and reconstituting cells. In order to identify such a combination therapy, we conducted an inhibitor screen using 40 targeted agents at three different doses in all pairwise combinations. Checkpoint Kinase 1 (CHK1) inhibitors were identified as potent inhibitors of MSL breast cancers. When combined with a pro-apoptotic agent/B Cell Lymphoma 2 (BCL2) inhibitor, the effectiveness of the combination regimen was super-additive compared to either treatment alone and was selective for MSL cancers. Treatment of MSL breast cancer cells results in DNA damage, cell-cycle defects characterized by a prolonged S-phase, increased apoptosis and decreased colony forming abilities compared to untreated cells. These data suggest that a combination of a CHK1 and BCL2 inhibitor could be an effective treatment for patients with MSL breast cancer. Several other effective drug combinations were also identified.

5.
Pigment Cell Melanoma Res ; 32(2): 292-302, 2019 03.
Article in English | MEDLINE | ID: mdl-30281931

ABSTRACT

This study evaluates the use of HMG-CoA reductase inhibitors, or statins, as an adjunctive to BRAF and MEK inhibition as a treatment in melanomas and other tumors with driver mutations in the MAPK pathway. Experiments used simvastatin in conjunction with vemurafenib and selumetinib in vitro and simvastatin with vemurafenib in vivo to demonstrate additional growth abrogation beyond MAPK blockade alone. Additional studies demonstrated that statin anti-tumor effects appeared to depend on inhibition of isoprenoid synthesis given rescue with add-back of downstream metabolites. Ultimately, we concluded that statins represent a possible useful adjunctive therapy in MAPK-driven tumors when given with current approved targeted therapy.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Prenylation , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Melanoma/enzymology , Melanoma/pathology , Mevalonic Acid/metabolism , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects
6.
Cancer Discov ; 8(1): 20-23, 2018 01.
Article in English | MEDLINE | ID: mdl-29311225

ABSTRACT

Greatest fitness of tumor cell subclones in patients undergoing MAPK-targeting therapies requires just-right levels of MAPK pathway signaling. New therapeutic approaches induce tumor cell death by intensifying MAPK signaling induced by inhibitor withdrawal in combination with DNA damage, or prevent selection of resistant clones with a steep fitness barrier imposed by triple combination of BRAF, MEK, and ERK inhibitors. Cancer Discov; 8(1); 20-3. ©2018 AACRSee related article by Hong et al., p. 74.


Subject(s)
Cell Line, Tumor , Proto-Oncogene Proteins B-raf , Cell Death , Humans , MAP Kinase Signaling System , Protein Kinase Inhibitors , Signal Transduction
7.
J Invest Dermatol ; 137(10): 2187-2196, 2017 10.
Article in English | MEDLINE | ID: mdl-28599981

ABSTRACT

Despite improvements in survival in metastatic melanoma with combined BRAF and mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor treatment, the overwhelming majority of patients eventually acquire resistance to both agents. Consequently, new targets for therapy in resistant tumors are currently being evaluated. Previous studies have identified p90 subfamily of ribosomal S6 kinase (p90RSK) family kinases as key factors for growth and proliferation, as well as protein synthesis via assembly of the 7-methyl-guanosine triphosphate cap-dependent translation complex. We sought to evaluate inhibitors of p90RSK family members: BI-D1870 and BRD7389, for their ability to inhibit both proliferation and protein synthesis in patient-derived melanoma cell lines with acquired resistance to combined treatment with the BRAF inhibitor vemurafenib and the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor selumetinib. We found that the RSK inhibitors blocked cell proliferation and protein synthesis in multiple dual-resistant melanoma lines. In addition, single agent RSK inhibitor treatment was effective in drug-naïve lines, two of which are innately vemurafenib resistant. We also used Reverse Phase Protein Array screening to identify differential protein expression that correlates with BI-D1870 sensitivity, and identified prognostic biomarkers for survival in human melanoma patients. These findings establish p90RSK inhibition as a therapeutic strategy in treatment-resistant melanoma and provide insight into the mechanism of action.


Subject(s)
Drug Resistance, Neoplasm/drug effects , MAP Kinase Kinase 1/biosynthesis , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/biosynthesis , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MAP Kinase Kinase 1/drug effects , Melanoma/drug therapy , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/drug effects , Pteridines , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction
8.
BMC Bioinformatics ; 18(1): 317, 2017 Jun 26.
Article in English | MEDLINE | ID: mdl-28651562

ABSTRACT

BACKGROUND: Personalizing treatment regimes based on gene expression profiles of individual tumors will facilitate management of cancer. Although many methods have been developed to identify pathways perturbed in tumors, the results are often not generalizable across independent datasets due to the presence of platform/batch effects. There is a need to develop methods that are robust to platform/batch effects and able to identify perturbed pathways in individual samples. RESULTS: We present Gene-Ranking Analysis of Pathway Expression (GRAPE) as a novel method to identify abnormal pathways in individual samples that is robust to platform/batch effects in gene expression profiles generated by multiple platforms. GRAPE first defines a template consisting of an ordered set of pathway genes to characterize the normative state of a pathway based on the relative rankings of gene expression levels across a set of reference samples. This template can be used to assess whether a sample conforms to or deviates from the typical behavior of the reference samples for this pathway. We demonstrate that GRAPE performs well versus existing methods in classifying tissue types within a single dataset, and that GRAPE achieves superior robustness and generalizability across different datasets. A powerful feature of GRAPE is the ability to represent individual gene expression profiles as a vector of pathways scores. We present applications to the analyses of breast cancer subtypes and different colonic diseases. We perform survival analysis of several TCGA subtypes and find that GRAPE pathway scores perform well in comparison to other methods. CONCLUSIONS: GRAPE templates offer a novel approach for summarizing the behavior of gene-sets across a collection of gene expression profiles. These templates offer superior robustness across distinct experimental batches compared to existing methods. GRAPE pathway scores enable identification of abnormal gene-set behavior in individual samples using a non-competitive approach that is fundamentally distinct from popular enrichment-based methods. GRAPE may be an appropriate tool for researchers seeking to identify individual samples displaying abnormal gene-set behavior as well as to explore differences in the consensus gene-set behavior of groups of samples. GRAPE is available in R for download at https://CRAN.R-project.org/package=GRAPE .


Subject(s)
Gene Expression Profiling/methods , Transcriptome , User-Computer Interface , Humans , Internet , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Support Vector Machine , Survival Analysis
9.
Mol Cancer Ther ; 16(6): 1041-1053, 2017 06.
Article in English | MEDLINE | ID: mdl-28292938

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is the fourth most common cause of cancer-related death in the United States. PDAC is difficult to manage effectively, with a five-year survival rate of only 5%. PDAC is largely driven by activating KRAS mutations, and as such, cannot be directly targeted with therapeutic agents that affect the activated protein. Instead, inhibition of downstream signaling and other targets will be necessary to effectively manage PDAC. Here, we describe a tiered single-agent and combination compound screen to identify targeted agents that impair growth of a panel of PDAC cell lines. Several of the combinations identified from the screen were further validated for efficacy and mechanism. Combination of the bromodomain inhibitor JQ1 and the neddylation inhibitor MLN4294 altered the production of reactive oxygen species in PDAC cells, ultimately leading to defects in the DNA damage response. Dual bromodomain/neddylation blockade inhibited in vivo growth of PDAC cell line xenografts. Overall, this work revealed novel combinatorial regimens, including JQ1 plus MLN4294, which show promise for the treatment of RAS-driven PDAC. Mol Cancer Ther; 16(6); 1041-53. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Screening Assays, Antitumor/methods , Drug Synergism , High-Throughput Nucleotide Sequencing , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Superoxides , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
10.
PLoS Comput Biol ; 13(1): e1005308, 2017 01.
Article in English | MEDLINE | ID: mdl-28085880

ABSTRACT

A promising alternative to address the problem of acquired drug resistance is to rely on combination therapies. Identification of the right combinations is often accomplished through trial and error, a labor and resource intensive process whose scale quickly escalates as more drugs can be combined. To address this problem, we present a broad computational approach for predicting synergistic combinations using easily obtainable single drug efficacy, no detailed mechanistic understanding of drug function, and limited drug combination testing. When applied to mutant BRAF melanoma, we found that our approach exhibited significant predictive power. Additionally, we validated previously untested synergy predictions involving anticancer molecules. As additional large combinatorial screens become available, this methodology could prove to be impactful for identification of drug synergy in context of other types of cancers.


Subject(s)
Drug Combinations , Drug Discovery/methods , Drug Synergism , Antineoplastic Agents , Cell Line, Tumor , Computational Biology , Humans , Melanoma/drug therapy , Melanoma/genetics , Models, Theoretical , Proto-Oncogene Proteins B-raf/genetics
11.
Cancer Res ; 77(2): 566-578, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27872098

ABSTRACT

Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor/methods , Triple Negative Breast Neoplasms , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Immunoprecipitation , Principal Component Analysis
12.
Sci Rep ; 6: 20567, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26837275

ABSTRACT

Although normal tissue samples adjacent to tumors are sometimes collected from patients in cancer studies, they are often used as normal controls to identify genes differentially expressed between tumor and normal samples. However, it is in general more difficult to obtain and clearly define paired normal samples, and whether these samples should be treated as "normal" due to their close proximity to tumors. In this article, by analyzing the accrued data in The Cancer Genome Atlas (TCGA), we show the surprising results that the paired normal samples are in general more informative on patient survival than tumors. Different lines of evidence suggest that this is likely due to tumor micro-environment instead of tumor cell contamination or field cancerization effect. Pathway analyses suggest that tumor micro-environment may play an important role in cancer patient survival either by boosting the adjacent metabolism or the in situ immunization. Our results suggest the potential benefit of collecting and profiling matched normal tissues to gain more insights on disease etiology and patient progression.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Neoplasms/genetics , Transcriptome , Databases, Genetic , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Survival Analysis , Tumor Microenvironment
13.
Proc Natl Acad Sci U S A ; 113(3): E282-90, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729871

ABSTRACT

In the lactating mammary gland, the plasma membrane calcium ATPase2 (PMCA2) transports milk calcium. Its expression is activated in breast cancers, where high tumor levels predict increased mortality. We find that PMCA2 expression correlates with HER2 levels in breast cancers and that PMCA2 interacts with HER2 in specific actin-rich membrane domains. Knocking down PMCA2 increases intracellular calcium, disrupts interactions between HER2 and HSP-90, inhibits HER2 signaling, and results in internalization and degradation of HER2. Manipulating PMCA2 levels regulates the growth of breast cancer cells, and knocking out PMCA2 inhibits the formation of tumors in mouse mammary tumor virus (MMTV)-Neu mice. These data reveal previously unappreciated molecular interactions regulating HER2 localization, membrane retention, and signaling, as well as the ability of HER2 to generate breast tumors, suggesting that interactions between PMCA2 and HER2 may represent therapeutic targets for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Breast Neoplasms/pathology , Calcium/pharmacology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Endocytosis/drug effects , Female , Fluorescent Antibody Technique , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Intracellular Space/metabolism , Mammary Neoplasms, Animal , Mice , Protein Binding , Protein Transport , Survival Analysis
14.
Sci Signal ; 8(401): ra111, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26535009

ABSTRACT

Cholesterol is a lipid that is critical for steroid hormone production and the integrity of cellular membranes, and, as such, it is essential for cell growth. The epidermal growth factor receptor (EGFR) family member ERBB4, which forms signaling complexes with other EGFR family members, can undergo ligand-induced proteolytic cleavage to release a soluble intracellular domain (ICD) that enters the nucleus to modify transcription. We found that ERBB4 activates sterol regulatory element binding protein-2 (SREBP-2) to enhance low-density lipoprotein (LDL) uptake and cholesterol biosynthesis. Expression of the ERBB4 ICD in mammary epithelial cells or activation of ERBB4 with the ligand neuregulin 1 (NRG1) induced the expression of SREBP target genes involved in cholesterol biosynthesis, including HMGCR and HMGCS1, and lipid uptake, LDLR, which encodes the LDL receptor. Addition of NRG1 increased the abundance of the cleaved, mature form of SREBP-2 through a pathway that was blocked by addition of inhibitors of PI3K (phosphatidylinositol 3-kinase) or dual inhibition of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2, but not by inhibition of AKT or mTORC1. Pharmacological inhibition of the activity of SREBP site 1 protease or of all EGFR family members (with lapatinib), but not EGFR alone (with erlotinib), impaired NRG1-induced expression of cholesterol biosynthesis genes. Collectively, our findings indicated that activation of ERBB4 promotes SREBP-2-regulated cholesterol metabolism. The connections of EGFR and ERBB4 signaling with SREBP-2-regulated cholesterol metabolism are likely to be important in ERBB-regulated developmental processes and may contribute to metabolic remodeling in ERBB-driven cancers.


Subject(s)
Cholesterol/biosynthesis , Lipoproteins, LDL/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-4/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Cell Line, Tumor , Cholesterol/genetics , Female , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipoproteins, LDL/genetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neuregulin-1/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/genetics , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 2/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26485762

ABSTRACT

Targeting anti-apoptotic proteins can sensitize tumor cells to conventional chemotherapies or other targeted agents. Antagonizing the Inhibitor of Apoptosis Proteins (IAPs) with mimetics of the pro-apoptotic protein SMAC is one such approach. We used sensitization compound screening to uncover possible agents with the potential to further sensitize lung adenocarcinoma cells to the SMAC mimetic Debio 1143. Several compounds in combination with Debio 1143, including taxanes, topoisomerase inhibitors, and bromodomain inhibitors, super-additively inhibited growth and clonogenicity of lung adenocarcinoma cells. Co-treatment with Debio 1143 and the bromodomain inhibitor JQ1 suppresses the expression of c-IAP1, c-IAP2, and XIAP. Non-canonical NF-κB signaling is also activated following Debio 1143 treatment, and Debio 1143 induces the formation of the ripoptosome in Debio 1143-sensitive cell lines. Sensitivity to Debio 1143 and JQ1 co-treatment was associated with baseline caspase-8 expression. In vivo treatment of lung adenocarcinoma xenografts with Debio 1143 in combination with JQ1 or docetaxel reduced tumor volume more than either single agent alone. As Debio 1143-containing combinations effectively inhibited both in vitro and in vivo growth of lung adenocarcinoma cells, these data provide a rationale for Debio 1143 combinations currently being evaluated in ongoing clinical trials and suggest potential utility of other combinations identified here.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Azocines/pharmacology , Benzhydryl Compounds/pharmacology , Camptothecin/analogs & derivatives , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Taxoids/pharmacology , Topoisomerase Inhibitors/pharmacology , Triazoles/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Docetaxel , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Irinotecan , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 14(7): 1680-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948295

ABSTRACT

BRAF kinase inhibitors have dramatically affected treatment of BRAF(V600E) (/) (K)-driven metastatic melanoma. Early responses assessed using [(18)F]fluorodeoxyglucose uptake-positron emission tomography (FDG-PET) have shown dramatic reduction of radiotracer signal within 2 weeks of treatment. Despite high response rates, relapse occurs in nearly all cases, frequently at sites of treated metastatic disease. It remains unclear whether initial loss of (18)FDG uptake is due to tumor cell death or other reasons. Here, we provide evidence of melanoma cell volume reduction in a patient cohort treated with BRAF inhibitors. We present data demonstrating that BRAF inhibition reduces melanoma glucose uptake per cell, but that this change is no longer significant following normalization for cell volume changes. We also demonstrate that volume normalization greatly reduces differences in transmembrane glucose transport and hexokinase-mediated phosphorylation. Mechanistic studies suggest that this loss of cell volume is due in large part to decreases in new protein translation as a consequence of vemurafenib treatment. Ultimately, our findings suggest that cell volume regulation constitutes an important physiologic parameter that may significantly contribute to radiographic changes observed in clinic.


Subject(s)
Cell Size , Glucose/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Biological Transport/drug effects , Drug Resistance, Neoplasm , Flow Cytometry , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Glucose/pharmacokinetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , Immunoblotting , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Positron-Emission Tomography , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , Sulfonamides/pharmacology , Vemurafenib
17.
Pigment Cell Melanoma Res ; 28(4): 417-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25854919

ABSTRACT

BRAF inhibitors have revolutionized treatment of mutant BRAF metastatic melanomas. However, resistance develops rapidly following BRAF inhibitor treatment. We have found that BRAF-mutant melanoma cell lines are more sensitive than wild-type BRAF cells to the small molecule tyrosine kinase inhibitor dovitinib. Sensitivity is associated with inhibition of a series of known dovitinib targets. Dovitinib in combination with several agents inhibits growth more effectively than either agent alone. These combinations inhibit BRAF-mutant melanoma and colorectal carcinoma cell lines, including cell lines with intrinsic or selected BRAF inhibitor resistance. Hence, combinations of dovitinib with second agents are potentially effective therapies for BRAF-mutant melanomas, regardless of their sensitivity to BRAF inhibitors.


Subject(s)
Benzimidazoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Indoles/pharmacology , Male , Melanoma/enzymology , Mice, Nude , Neoplasm Proteins/metabolism , Skin Neoplasms , Small Molecule Libraries/pharmacology , Sulfonamides/pharmacology , Vemurafenib , Melanoma, Cutaneous Malignant
18.
Cancer Res ; 75(7): 1399-412, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712345

ABSTRACT

Melanoma development involves members of the AGC kinase family, including AKT, PKC, and, most recently, PDK1, as elucidated recently in studies of Braf::Pten mutant melanomas. Here, we report that PDK1 contributes functionally to skin pigmentation and to the development of melanomas harboring a wild-type PTEN genotype, which occurs in about 70% of human melanomas. The PDK1 substrate SGK3 was determined to be an important mediator of PDK1 activities in melanoma cells. Genetic or pharmacologic inhibition of PDK1 and SGK3 attenuated melanoma growth by inducing G1 phase cell-cycle arrest. In a synthetic lethal screen, pan-PI3K inhibition synergized with PDK1 inhibition to suppress melanoma growth, suggesting that focused blockade of PDK1/PI3K signaling might offer a new therapeutic modality for wild-type PTEN tumors. We also noted that responsiveness to PDK1 inhibition associated with decreased expression of pigmentation genes and increased expression of cytokines and inflammatory genes, suggesting a method to stratify patients with melanoma for PDK1-based therapies. Overall, our work highlights the potential significance of PDK1 as a therapeutic target to improve melanoma treatment.


Subject(s)
Melanoma/enzymology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/enzymology , Animals , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints , Humans , Immediate-Early Proteins/metabolism , Indazoles/pharmacology , Lymphatic Metastasis , Melanoma/drug therapy , Melanoma/secondary , Mice, Knockout , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Skin/enzymology , Skin/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
19.
Breast Cancer Res ; 16(6): 501, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25516216

ABSTRACT

INTRODUCTION: Human Epidermal Growth Factor Receptor (ERBB4/HER4) belongs to the Epidermal Growth Factor receptor/ERBB family of receptor tyrosine kinases. While ERBB1, ERBB2 and ERBB3 are often overexpressed or activated in breast cancer, and are oncogenic, the role of ERBB4 in breast cancer is uncertain. Some studies suggest a tumor suppressor role of ERBB4, while other reports suggest an oncogenic potential. Alternative splicing of ERBB4 yields four major protein products, these spliced isoforms differ in the extracellular juxtamembrane domain (JM-a versus JM-b) and cytoplasmic domain (CYT-1 versus CYT-2). Two of these isoforms, JM-a CYT-1 and JM-a CYT-2, are expressed in the mammary gland. Failure to account for isoform-specific functions in previous studies may account for conflicting reports on the role of ERBB4 in breast cancer. METHODS: We have produced mouse mammary tumour virus (MMTV) -ERBB4 transgenic mice to evaluate potential developmental and carcinogenic changes associated with full length (FL) JM-a ERBB4 CYT-1 versus ERBB4 CYT-2. Mammary tissue was isolated from transgenic mice and sibling controls at various developmental stages for whole mount analysis, RNA extraction, and immunohistochemistry. To maintain maximal ERBB4 expression, transgenic mice were bred continuously for a year after which mammary glands were isolated and analyzed. RESULTS: Overexpressing FL CYT-1 isoform resulted in suppression of mammary ductal morphogenesis which was accompanied by decreased number of mammary terminal end buds (TEBs) and Ki-67 positive cells within TEBs, while FL CYT-2 isoform had no effect on ductal growth in pubescent mice. The suppressive ductal phenotype in CYT-1 mice disappeared after mid-pregnancy, and subsequent developmental stages showed no abnormality in mammary gland morphology or function in CYT-1 or CYT-2 transgenic mice. However, sustained expression of FL CYT-1 isoform resulted in formation of neoplastic mammary lesions, suggesting a potential oncogenic function for this isoform. CONCLUSIONS: Together, we present isoform-specific roles of ERBB4 during puberty and early pregnancy, and reveal a novel oncogenic property of CYT-1 ERBB4. The results may be exploited to develop better therapeutic strategies in breast cancer.


Subject(s)
Carcinogenesis/genetics , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/genetics , Pregnancy/genetics , Protein Isoforms/genetics , Receptor, ErbB-4/genetics , Alternative Splicing , Animals , Carcinogenesis/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Pregnancy/metabolism , Protein Isoforms/metabolism , Receptor, ErbB-4/metabolism
20.
Sci Signal ; 7(355): ra116, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25492965

ABSTRACT

The receptor tyrosine kinase ERBB4, a member of the epidermal growth factor receptor (EGFR) family, is unusual in that ERBB4 can undergo intramembrane proteolysis, releasing a soluble intracellular domain (ICD) that modulates transcription in the nucleus. We found that ERBB4 activated the transcriptional coactivator YAP, which promotes organ and tissue growth and is inhibited by the Hippo tumor-suppressor pathway. Overexpressing ERBB4 in cultured mammary epithelial cells or adding the ERBB4 ligand neuregulin 1 (NRG1) to breast cancer cell cultures promoted the expression of genes regulated by YAP, such as CTGF. Knocking down YAP or ERBB4 prevented the induction of CTGF expression by NRG1, as did treating cells with the ERBB inhibitors lapatinib or erlotinib, which reduced ERBB4 cleavage. NRG1 stimulated YAP activity to an extent comparable to that of EGF (epidermal growth factor) or LPA (lysophosphatidic acid), known activators of YAP. NRG1 stimulated YAP-dependent cell migration in breast cancer cell lines. These observations connect the unusual nuclear function of a growth factor receptor with a mechanosensory pathway and suggest that NRG1-ERBB4-YAP signaling contributes to the aggressive behavior of tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Mechanotransduction, Cellular , Neuregulin-1/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-4/metabolism , Transcription Factors/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Erlotinib Hydrochloride , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Hippo Signaling Pathway , Humans , Lapatinib , Neuregulin-1/genetics , Nuclear Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Quinazolines/pharmacology , Receptor, ErbB-4/antagonists & inhibitors , Receptor, ErbB-4/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...