16 results on '"Rix, Uwe"'
Search Results
2. Abstract C110: Cancer associated fibroblast-derived IGF-binding proteins augment osimertinib activity in EGFR-mutant NSCLC cells
- Author
-
Rix, Lily L. Remsing, primary, Sumi, Natalia J., additional, Bryant, Annamarie T., additional, Li, Xueli, additional, Welsh, Eric A., additional, Fang, Bin, additional, Kuenzi, Brent M., additional, Antonia, Scott J., additional, Marusyk, Andriy, additional, Lovly, Christine M., additional, Koomen, John M., additional, Haura, Eric B., additional, and Rix, Uwe, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract B022: PARP1 complex composition as a predictor of response to PARP inhibitors in BRCA-linked ovarian carcinoma
- Author
-
Deng, Ou, primary, Dash, Sweta, additional, Nepomuceno, Thales, additional, Han, Ming D, additional, Fang, Bin, additional, Marchion, Doug, additional, Monteiro, Alvaro N, additional, and Rix, Uwe, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract 3973: Dabrafenib suppresses the growth ofBRAF-WTcancers through inhibition of novel targets Nek9 and Cdk16
- Author
-
Phadke, Manali S., primary, Rix, Lily, additional, Smalley, Inna, additional, Bryant, Annamarie, additional, Lawrence, Harshani, additional, Schaible, Braydon, additional, Chen, Ann, additional, Rix, Uwe, additional, and Smalley, Keiran, additional
- Published
- 2018
- Full Text
- View/download PDF
5. Abstract 5089: A quantitative proteomic survey reveals mechanisms of the bi-directional signaling between lung fibroblasts and lung cancer cells
- Author
-
DJIDJOU, MARTIAL BOUTCHUENG, primary, KIM, Jae-Young, additional, Han, Ki-Cheol, additional, Wright, Gabriela, additional, Majumder, Anurima, additional, Fang, Bin, additional, Koomen, John, additional, Rix, Lily.L Remsing, additional, Rix, Uwe, additional, and Haura, Eric, additional
- Published
- 2018
- Full Text
- View/download PDF
6. Abstract 2882: Elucidation of the mechanism of action of the multi-kinase inhibitor midostaurin in NSCLC
- Author
-
Ctortecka, Claudia, primary, Kuenzi, Brent, additional, Fang, Bin, additional, Sumi, Natalia, additional, Izumi, Victoria, additional, Kinose, Fumi, additional, Haura, Eric, additional, Koomen, John, additional, and Rix, Uwe, additional
- Published
- 2018
- Full Text
- View/download PDF
7. Abstract 221: Integrated functional proteomics of MET/VEGFR inhibitors reveals complex mechanism of action of foretinib in NSCLC
- Author
-
Sumi, Natalia J., primary, Fang, Bin, additional, Rix, Lily L., additional, Ayaz, Muhammad, additional, Kinose, Fumi, additional, Welsh, Eric A., additional, Eschrich, Steven A., additional, Lawrence, Harshani R., additional, Koomen, John M., additional, Haura, Eric B., additional, and Rix, Uwe, additional
- Published
- 2017
- Full Text
- View/download PDF
8. Abstract 675: Off-target based drug repurposing opportunities for tivantinib in acute myeloid leukemia
- Author
-
Kuenzi, Brent M., primary, Remsing Rix, Lily L., additional, Kunigal, Sateesh S., additional, Kinose, Fumi, additional, Knezevic, Claire E., additional, Wright, Gabriela, additional, Kroeger, Jodi L., additional, Lancet, Jeffrey E., additional, Padron, Eric, additional, and Rix, Uwe, additional
- Published
- 2015
- Full Text
- View/download PDF
9. EGFR Mediates Responses to Small-Molecule Drugs Targeting Oncogenic Fusion Kinases.
- Author
-
Vaishnavi, Aria, Schubert, Laura, Rix, Uwe, Marek, Lindsay A., Le, Anh T., Keysar, Stephen B., Glogowska, Magdalena J., Smith, Matthew A., Kako, Severine, Sumi, Natalia J., Davies, Kurtis D., Ware, Kathryn E., Varella-Garcia, Marileila, Haura, Eric B., Jimeno, Antonio, Heasley, Lynn E., Aisner, Dara L., and Doebele, Robert C.
- Subjects
- *
EPIDERMAL growth factor receptors , *SMALL molecules , *TARGETED drug delivery , *ONCOGENES - Abstract
Oncogenic kinase fusions of ALK, ROS1, RET, and NTRK1 act as drivers in human lung and other cancers. Residual tumor burden following treatment of ALK or ROS1+ lung cancer patients with oncogene-targeted therapy ultimately enables the emergence of drug-resistant clones, limiting the long-term effectiveness of these therapies. To determine the signaling mechanisms underlying incomplete tumor cell killing in oncogene-addicted cancer cells, we investigated the role of EGFR signaling in drug-naïve cancer cells harboring these oncogene fusions. We defined three distinct roles for EGFR in the response to oncogene-specific therapies. First, EGF-mediated activation of EGFR blunted fusion kinase inhibitor binding and restored fusion kinase signaling complexes. Second, fusion kinase inhibition shifted adaptor protein binding from the fusion oncoprotein to EGFR. Third, EGFR enabled bypass signaling to critical downstream pathways such as MAPK. While evidence of EGFR-mediated bypass signaling has been reported after ALK and ROS1 blockade, our results extended this effect to RET and NTRK1 blockade and uncovered the other additional mechanisms in gene fusion-positive lung cancer cells, mouse models, and human clinical specimens before the onset of acquired drug resistance. Collectively, our findings show how EGFR signaling can provide a critical adaptive survival mechanism that allows cancer cells to evade oncogene-specific inhibitors, providing a rationale to cotarget EGFR to reduce the risks of developing drug resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
10. Dissecting gatekeeper BCR-ABL-specific drug synergy by an integrated systems chemical biology approach.
- Author
-
Rix, Uwe, Winter, Georg E., Bennett, Keiryn L., White, Forest M., Superti-Furga, Giulio, Carlson, Scott M., Gleixner, Karoline V., Grebien, Florian, Gridling, Manuela, Mueller, Andre, Bilban, Martin, Colinge, Jacques, and Valent, Peter
- Subjects
- *
MYELOID leukemia , *LEUKEMIA treatment , *GENETIC mutation , *PROTEOMICS , *PHARMACODYNAMICS , *MITOGEN-activated protein kinases - Abstract
The BCR-ABLT315I gatekeeper mutation represents one of the most difficult challenges in the therapy of chronic myeloid leukemia (CML) and although several drug candidates are currently undergoing clinical evaluation, there is no FDA-approved drug that successfully targets this mutation. We therefore performed a focused synergy screen with a panel of established BCR- ABL inhibitors postulating, as most kinase inhibitors are unspecific, the possibility of additional efficacy due to off-target effects. Using the Ba/F3 cell system we detected strong synergy between two multikinase inhibitors, danusertib and bosutinib, that was specific for cells carrying the BCR-ABL gatekeeper mutant over BCR-ABL wild-type expressing cells. This synergy translated also to colony formation assays with BCR-ABLT315I-transduced primary mouse bone marrow cells and ex vivo proliferation assays using BCR-ABLT315I patient cells. In order to elucidate the underlying molecular mechanisms of action, which distinguishes the individual drug effects from the drug combination, we applied a combined systems-level approach comprised of quantitative phosphoproteomics, transcriptomics, and quantitative chemical proteomics, which query the different layers of signaling pathways. Intersecting these orthogonal datasets through pathway mapping revealed previously unappreciated off-targets for both compounds, which impinged on the Mapk signaling cascade downstream of BCR-ABL, along with partial inhibition of BCR-ABLT315I itself. Collectively, these cooperative molecular interventions resulted in impaired activity of c-Myc causing a genome-wide down regulation of c-Myc target genes that represents the dominant mode-of action of the detected synergy. In summary, we here present a comprehensive integration of three different large-scale -omics approaches that has broad potential for elucidating complex cellular mechanisms of drug synergy in general. [ABSTRACT FROM AUTHOR]
- Published
- 2012
11. PTEN Lipid Phosphatase Activity Suppresses Melanoma Formation by Opposing an AKT/mTOR/FRA1 Signaling Axis.
- Author
-
Xu X, Bok I, Jasani N, Wang K, Chadourne M, Mecozzi N, Deng O, Welsh EA, Kinose F, Rix U, and Karreth FA
- Subjects
- Animals, Mice, Signal Transduction genetics, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, TOR Serine-Threonine Kinases metabolism, Cell Proliferation, Lipids, Proto-Oncogene Proteins c-akt metabolism, Melanoma genetics, Melanoma metabolism
- Abstract
Inactivating mutations in PTEN are prevalent in melanoma and are thought to support tumor development by hyperactivating the AKT/mTOR pathway. Conversely, activating mutations in AKT are relatively rare in melanoma, and therapies targeting AKT or mTOR have shown disappointing outcomes in preclinical models and clinical trials of melanoma. This has led to the speculation that PTEN suppresses melanoma by opposing AKT-independent pathways, potentially through noncanonical functions beyond its lipid phosphatase activity. In this study, we examined the mechanisms of PTEN-mediated suppression of melanoma formation through the restoration of various PTEN functions in PTEN-deficient cells or mouse models. PTEN lipid phosphatase activity predominantly inhibited melanoma cell proliferation, invasion, and tumor growth, with minimal contribution from its protein phosphatase and scaffold functions. A drug screen underscored the exquisite dependence of PTEN-deficient melanoma cells on the AKT/mTOR pathway. Furthermore, activation of AKT alone was sufficient to counteract several aspects of PTEN-mediated melanoma suppression, particularly invasion and the growth of allograft tumors. Phosphoproteomics analysis of the lipid phosphatase activity of PTEN validated its potent inhibition of AKT and many of its known targets, while also identifying the AP-1 transcription factor FRA1 as a downstream effector. The restoration of PTEN dampened FRA1 translation by inhibiting AKT/mTOR signaling, and FRA1 overexpression negated aspects of PTEN-mediated melanoma suppression akin to AKT. This study supports AKT as the key mediator of PTEN inactivation in melanoma and identifies an AKT/mTOR/FRA1 axis as a driver of melanomagenesis., Significance: PTEN suppresses melanoma predominantly through its lipid phosphatase function, which when lost, elevates FRA1 levels through AKT/mTOR signaling to promote several aspects of melanomagenesis., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
12. Targeted Therapy Given after Anti-PD-1 Leads to Prolonged Responses in Mouse Melanoma Models through Sustained Antitumor Immunity.
- Author
-
Phadke MS, Chen Z, Li J, Mohamed E, Davies MA, Smalley I, Duckett DR, Palve V, Czerniecki BJ, Forsyth PA, Noyes D, Adeegbe DO, Eroglu Z, Nguyen KT, Tsai KY, Rix U, Burd CE, Chen YA, Rodriguez PC, and Smalley KSM
- Subjects
- Animals, Antineoplastic Agents chemistry, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Disease Models, Animal, Female, Imidazoles chemistry, Immunotherapy, Melanoma genetics, Melanoma immunology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Molecular Targeted Therapy, Monomeric GTP-Binding Proteins genetics, Mutation, Oximes chemistry, Programmed Cell Death 1 Receptor antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Pyridones chemistry, Pyrimidines chemistry, Pyrimidinones chemistry, Skin Neoplasms genetics, Skin Neoplasms immunology, Sulfones chemistry, T-Lymphocytes, Regulatory immunology, Antineoplastic Agents therapeutic use, Melanoma drug therapy, Monomeric GTP-Binding Proteins antagonists & inhibitors, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Skin Neoplasms drug therapy
- Abstract
Immunotherapy (IT) and targeted therapy (TT) are both effective against melanoma, but their combination is frequently toxic. Here, we investigated whether the sequence of IT (anti-PD-1)→ TT (ceritinib-trametinib or dabrafenib-trametinib) was associated with improved antitumor responses in mouse models of BRAF- and NRAS -mutant melanoma. Mice with NRAS- mutant (SW1) or BRAF -mutant (SM1) mouse melanomas were treated with either IT, TT, or the sequence of IT→TT. Tumor volumes were measured, and samples from the NRAS -mutant melanomas were collected for immune-cell analysis, single-cell RNA sequencing (scRNA-seq), and reverse phase protein analysis (RPPA). scRNA-seq demonstrated that the IT→TT sequence modulated the immune environment, leading to increased infiltration of T cells, monocytes, dendritic cells and natural killer cells, and decreased numbers of tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells. Durable responses to the IT→TT sequence were dependent on T-cell activity, with depletion of CD8
+ , but not CD4+ , T cells abrogating the therapeutic response. An analysis of transcriptional heterogeneity in the melanoma compartment showed the sequence of IT→TT enriched for a population of melanoma cells with increased expression of MHC class I and melanoma antigens. RPPA analysis demonstrated that the sustained immune response induced by IT→TT suppressed tumor-intrinsic signaling pathways required for therapeutic escape. These studies establish that upfront IT improves the responses to TT in BRAF- and NRAS -mutant melanoma models., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
13. TRK xDFG Mutations Trigger a Sensitivity Switch from Type I to II Kinase Inhibitors.
- Author
-
Cocco E, Lee JE, Kannan S, Schram AM, Won HH, Shifman S, Kulick A, Baldino L, Toska E, Arruabarrena-Aristorena A, Kittane S, Wu F, Cai Y, Arena S, Mussolin B, Kannan R, Vasan N, Gorelick AN, Berger MF, Novoplansky O, Jagadeeshan S, Liao Y, Rix U, Misale S, Taylor BS, Bardelli A, Hechtman JF, Hyman DM, Elkabets M, de Stanchina E, Verma CS, Ventura A, Drilon A, and Scaltriti M
- Subjects
- Humans, Mutation, Oncogenes, Protein Kinase Inhibitors pharmacology, Neoplasms drug therapy, Neoplasms genetics, Receptor, trkA genetics
- Abstract
On-target resistance to next-generation TRK inhibitors in TRK fusion-positive cancers is largely uncharacterized. In patients with these tumors, we found that TRK xDFG mutations confer resistance to type I next-generation TRK inhibitors designed to maintain potency against several kinase domain mutations. Computational modeling and biochemical assays showed that TRKA
G667 and TRKCG696 xDFG substitutions reduce drug binding by generating steric hindrance. Concurrently, these mutations stabilize the inactive (DFG-out) conformations of the kinases, thus sensitizing these kinases to type II TRK inhibitors. Consistently, type II inhibitors impede the growth and TRK-mediated signaling of xDFG-mutant isogenic and patient-derived models. Collectively, these data demonstrate that adaptive conformational resistance can be abrogated by shifting kinase engagement modes. Given the prior identification of paralogous xDFG resistance mutations in other oncogene-addicted cancers, these findings provide insights into rational type II drug design by leveraging inhibitor class affinity switching to address recalcitrant resistant alterations. SIGNIFICANCE: In TRK fusion-positive cancers, TRK xDFG substitutions represent a shared liability for type I TRK inhibitors. In contrast, they represent a potential biomarker of type II TRK inhibitor activity. As all currently available type II agents are multikinase inhibitors, rational drug design should focus on selective type II inhibitor creation. This article is highlighted in the In This Issue feature, p. 1 ., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
14. MEK Inhibition Modulates Cytokine Response to Mediate Therapeutic Efficacy in Lung Cancer.
- Author
-
Xie M, Zheng H, Madan-Lala R, Dai W, Gimbrone NT, Chen Z, Kinose F, Blackstone SA, Smalley KSM, Cress WD, Haura EB, Rix U, and Beg AA
- Subjects
- Animals, Cell Line, Tumor, Cytokines, Humans, Mice, Protein Kinase Inhibitors, Lung Neoplasms, Proto-Oncogene Proteins B-raf
- Abstract
Activating mutations in BRAF, a key mediator of RAS signaling, are present in approximately 50% of melanoma patients. Pharmacologic inhibition of BRAF or the downstream MAP kinase MEK is highly effective in treating BRAF-mutant melanoma. In contrast, RAS pathway inhibitors have been less effective in treating epithelial malignancies, such as lung cancer. Here, we show that treatment of melanoma patients with BRAF and MEK inhibitors (MEKi) activated tumor NF-κB activity. MEKi potentiated the response to TNFα, a potent activator of NF-κB. In both melanoma and lung cancer cells, MEKi increased cell-surface expression of TNFα receptor 1 (TNFR1), which enhanced NF-κB activation and augmented expression of genes regulated by TNFα and IFNγ. Screening of 289 targeted agents for the ability to increase TNFα and IFNγ target gene expression demonstrated that this was a general activity of inhibitors of MEK and ERK kinases. Treatment with MEKi led to acquisition of a novel vulnerability to TNFα and IFNγ-induced apoptosis in lung cancer cells that were refractory to MEKi killing and augmented cell-cycle arrest. Abolishing the expression of TNFR1 on lung cancer cells impaired the antitumor efficacy of MEKi, whereas the administration of TNFα and IFNγ in MEKi-treated mice enhanced the antitumor response. Furthermore, immunotherapeutics known to induce expression of these cytokines synergized with MEKi in eradicating tumors. These findings define a novel cytokine response modulatory function of MEKi that can be therapeutically exploited. SIGNIFICANCE: Lung cancer cells are rendered sensitive to MEK inhibitors by TNFα and IFNγ, providing a strong mechanistic rationale for combining immunotherapeutics, such as checkpoint blockers, with MEK inhibitor therapy for lung cancer. See related commentary by Havel, p. 5699 ., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
15. Charting Immune Signaling Proteomes En Route to New Therapeutic Strategies.
- Author
-
Haura EB, Beg AA, Rix U, and Antonia S
- Subjects
- Cytokines metabolism, Humans, Mass Spectrometry, Tumor Microenvironment, Immunotherapy methods, Neoplasms therapy, Proteomics, Signal Transduction immunology, T-Lymphocytes immunology
- Abstract
The activation state of an antitumor effector T cell in a tumor depends on the sum of all stimulatory signals and inhibitory signals that it receives in the tumor microenvironment. Accumulating data address the increasing complexity of these signals produced by a myriad of immune checkpoint molecules, cytokines, and metabolites. While reductionist experiments have identified key molecules and their importance in signaling, less clear is the integration of all these signals that allows T cells to guide their responses in health and in disease. Mass spectrometry-based proteomics is well poised to offer such insights, including monitoring emergence of resistance mechanisms to immunotherapeutics during treatments. A major application of this technology is in the discovery and characterization of small-molecule agents capable of enhancing the response to immunotherapeutic agents. Such an approach would reinvigorate small-molecule drug development aimed not at tumor cells but rather at tumor-resident T cells capable of producing dramatic and durable antitumor responses., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
16. Adaptive responses to dasatinib-treated lung squamous cell cancer cells harboring DDR2 mutations.
- Author
-
Bai Y, Kim JY, Watters JM, Fang B, Kinose F, Song L, Koomen JM, Teer JK, Fisher K, Chen YA, Rix U, and Haura EB
- Subjects
- Adaptive Immunity drug effects, Apoptosis drug effects, Cell Line, Tumor, Dasatinib, Discoidin Domain Receptors, Drug Resistance, Neoplasm genetics, Drug Resistance, Neoplasm immunology, Drug Synergism, Humans, Lung Neoplasms immunology, Lung Neoplasms pathology, Mutation, Neoplasms, Squamous Cell immunology, Neoplasms, Squamous Cell pathology, Proto-Oncogene Proteins c-ret metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Mitogen metabolism, Receptors, Somatomedin antagonists & inhibitors, Signal Transduction genetics, Lung Neoplasms drug therapy, Neoplasms, Squamous Cell drug therapy, Pyrimidines administration & dosage, Receptor Protein-Tyrosine Kinases genetics, Receptors, Mitogen genetics, Thiazoles administration & dosage
- Abstract
DDR2 mutations occur in approximately 4% of lung squamous cell cancer (SCC) where the tyrosine kinase inhibitor dasatinib has emerged as a new therapeutic option. We found that ERK and AKT phosphorylation was weakly inhibited by dasatinib in DDR2-mutant lung SCC cells, suggesting that dasatinib inhibits survival signals distinct from other oncogenic receptor tyrosine kinases (RTK) and/or compensatory signals exist that dampen dasatinib activity. To gain better insight into dasatinib's action in these cells, we assessed altered global tyrosine phosphorylation (pY) after dasatinib exposure using a mass spectrometry-based quantitative phosphoproteomics approach. Overlaying protein-protein interaction relationships upon this dasatinib-regulated pY network revealed decreased phosphorylation of Src family kinases and their targets. Conversely, dasatinib enhanced tyrosine phosphorylation in a panel of RTK and their signaling adaptor complexes, including EGFR, MET/GAB1, and IGF1R/IRS2, implicating a RTK-driven adaptive response associated with dasatinib. To address the significance of this observation, these results were further integrated with results from a small-molecule chemical library screen. We found that dasatinib combined with MET and insulin-like growth factor receptor (IGF1R) inhibitors had a synergistic effect, and ligand stimulation of EGFR and MET rescued DDR2-mutant lung SCC cells from dasatinib-induced loss of cell viability. Importantly, we observed high levels of tyrosine-phosphorylated EGFR and MET in a panel of human lung SCC tissues harboring DDR2 mutations. Our results highlight potential RTK-driven adaptive-resistant mechanisms upon DDR2 targeting, and they suggest new, rationale cotargeting strategies for DDR2-mutant lung SCC., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.