45 results on '"Tiedt R"'
Search Results
2. 101P The landscape of MET alterations in European cancer patients
- Author
-
Wolf, J., primary, Ventouras, L-A., additional, Menu, P., additional, Wójtowicz, A., additional, Zou, M., additional, Diallo, S., additional, Chassot Agostinho, A., additional, Tiedt, R., additional, and Mina, M., additional
- Published
- 2021
- Full Text
- View/download PDF
3. The landscape of MET alterations in European cancer patients
- Author
-
Wolf, J., Ventouras, L-A., Menu, P., Wojtowicz, A., Zou, M., Diallo, S., Agostinho, A. Chassot, Tiedt, R., Mina, M., Wolf, J., Ventouras, L-A., Menu, P., Wojtowicz, A., Zou, M., Diallo, S., Agostinho, A. Chassot, Tiedt, R., and Mina, M.
- Published
- 2021
4. P76.03 Efficacy and Safety of Capmatinib Plus Nivolumab in Pretreated Patients with EGFR Wild-Type Non–Small Cell Lung Cancer
- Author
-
Felip, E., primary, Minotti, V., additional, Tan, D., additional, Wolf, J., additional, Mark, M., additional, Boyer, M., additional, Hughes, B., additional, Bearz, A., additional, Moro-Sibilot, D., additional, Le, X., additional, Vazquez, J., additional, Massuti, B., additional, Liu, N., additional, Hao, L., additional, Cheng, Y., additional, Tiedt, R., additional, and Cobo, M., additional
- Published
- 2021
- Full Text
- View/download PDF
5. A phase 1b study of the MET inhibitor capmatinib combined with cetuximab in patients with MET-positive colorectal cancer who had progressed following anti-EGFR monoclonal antibody treatment
- Author
-
Delord J, Argiles G, Fayette J, Wirth L, Kasper S, Siena S, Mesia R, Berardi R, Cervantes A, Dekervel J, Zhao S, Sun Y, Hao H, Tiedt R, Vicente S, Myers A, and Siu L
- Subjects
digestive system diseases - Abstract
Background Overcoming resistance to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs) in patients with KRAS wildtype (WT) metastatic colorectal cancer (mCRC) could help meet the needs of patients with limited treatment options. Methods In this phase 1b study, patients with N/KRAS WT, MET-positive mCRC who had progressed following anti-EGFR mAb treatment received escalating oral doses of capmatinib (150, 300, and 400 mg) twice daily plus weekly intravenous cetuximab (at the approved dose). The primary objective was to establish a recommended dose for expansion (RDE) of capmatinib in combination with cetuximab. Safety, preliminary activity, pharmacokinetics, and pharmacodynamics were also explored. Results Thirteen patients were enrolled. No patients experienced a dose-limiting toxicity at investigated doses; the RDE was established as capmatinib 400 mg twice daily plus cetuximab. All patients experienced adverse events (AEs) suspected to be related to the study treatment. Five patients (38.5%) reported study-drug-related AEs of grade 3/4 in severity. No patients achieved a complete or partial response according to RECIST v1.1; however, tumor shrinkage of 29-44% was observed in 4 patients. Conclusions Capmatinib plus cetuximab was well tolerated. Preliminary signs of activity were observed. Further investigation is warranted to obtain efficacy data and refine predictive biomarkers of response. Clinical trial registration NCT02205398.
- Published
- 2020
6. A Phase Ib/II, open-label, multicenter study of INC280 (capmatinib) alone and in combination with buparlisib (BKM120) in adult patients with recurrent glioblastoma
- Author
-
Bent, M.J. (Martin) van den, Azaro, A. (Analia), Vos, F. (Filip) de, Sepulveda, J. (Juan), Yung, W.K.A. (W. K. Alfred), Wen, P.Y. (Patrick), Lassman, A.B. (Andrew B.), Joerger, M., Tabatabai, G. (Ghazaleh), Rodon, J. (Jordi), Tiedt, R. (Ralph), Zhao, S. (Sylvia), Kirsilae, T. (Tiina), Cheng, Y. (Yi), Vicente, S. (Sergio), Balbin, O.A. (O. Alejandro), Zhang, H. (Hefei), Wick, W. (Wolfgang), Bent, M.J. (Martin) van den, Azaro, A. (Analia), Vos, F. (Filip) de, Sepulveda, J. (Juan), Yung, W.K.A. (W. K. Alfred), Wen, P.Y. (Patrick), Lassman, A.B. (Andrew B.), Joerger, M., Tabatabai, G. (Ghazaleh), Rodon, J. (Jordi), Tiedt, R. (Ralph), Zhao, S. (Sylvia), Kirsilae, T. (Tiina), Cheng, Y. (Yi), Vicente, S. (Sergio), Balbin, O.A. (O. Alejandro), Zhang, H. (Hefei), and Wick, W. (Wolfgang)
- Abstract
Purpose: To estimate the maximum tolerated dose (MTD) and/or identify the recommended Phase II dose (RP2D) for combined INC280 and buparlisib in patients with recurrent glioblastoma with homozygous phosphatase and tensin homolog (PTEN) deletion, mutation or protein loss. Methods: This multicenter, open-label, Phase Ib/II study included adult patients with glioblastoma with mesenchymal-epithelial transcription factor (c-Met) amplification. In Phase Ib, patients received INC280 as capsules or tablets in combination with buparlisib. In Phase II, patients received INC280 only. Response was assessed centrally using Response Assessment in Neuro-Oncology response criteria for high-grade gliomas. All adverse events (AEs) were recorded and graded. Results: 33 patients entered Phase Ib, 32 with altered PTEN. RP2D was not declared due to potential drug–drug interactions, which may have resulted in lack of efficacy; thus, Phase II, including 10 patients, was continued with INC280 monotherapy only. Best response was stable disease in 30% of patients. In the selected patient population, enrollment was halted due to limited activity with INC280 monotherapy. In Phase Ib, the most common treatment-related AEs were fatigue (36.4%), nausea (30.3%) and increased alanine aminotransferase (30.3%). MTD was identified at INC280 Tab 300 mg twice daily + buparlisib 80 mg once daily. In Phase II, the most common AEs were headache (40.0%), constipation (30.0%), fatigue (30.0%) and increased lipase (30.0%). Conclusion: The combination of INC280/buparlisib resulted in no clear activity in patients with recurrent PTEN-deficient glioblastoma. More stringent molecular selection strategies might produce better outcomes. Trial registration: NCT01870726.
- Published
- 2019
- Full Text
- View/download PDF
7. Complementary activities of DOT1L and Menin inhibitors in MLL-rearranged leukemia
- Author
-
Dafflon, C, primary, Craig, V J, additional, Méreau, H, additional, Gräsel, J, additional, Schacher Engstler, B, additional, Hoffman, G, additional, Nigsch, F, additional, Gaulis, S, additional, Barys, L, additional, Ito, M, additional, Aguadé-Gorgorió, J, additional, Bornhauser, B, additional, Bourquin, J-P, additional, Proske, A, additional, Stork-Fux, C, additional, Murakami, M, additional, Sellers, W R, additional, Hofmann, F, additional, Schwaller, J, additional, and Tiedt, R, additional
- Published
- 2016
- Full Text
- View/download PDF
8. A de novo splice donor mutation in the thrombopoietin gene causes hereditary thrombocythemia in a Polish family
- Author
-
Liu, K., primary, Kralovics, R., additional, Rudzki, Z., additional, Grabowska, B., additional, Buser, A. S., additional, Olcaydu, D., additional, Gisslinger, H., additional, Tiedt, R., additional, Frank, P., additional, Okon, K., additional, van der Maas, A. P.C., additional, and Skoda, R. C., additional
- Published
- 2008
- Full Text
- View/download PDF
9. The RING finger protein Siah-1 regulates the level of the transcriptional coactivator OBF-1
- Author
-
Tiedt, R., primary
- Published
- 2001
- Full Text
- View/download PDF
10. P76.03 Efficacy and Safety of Capmatinib Plus Nivolumab in Pretreated Patients with EGFRWild-Type Non–Small Cell Lung Cancer
- Author
-
Felip, E., Minotti, V., Tan, D., Wolf, J., Mark, M., Boyer, M., Hughes, B., Bearz, A., Moro-Sibilot, D., Le, X., Vazquez, J., Massuti, B., Liu, N., Hao, L., Cheng, Y., Tiedt, R., and Cobo, M.
- Published
- 2021
- Full Text
- View/download PDF
11. Increased lysosomal biomass is responsible for the resistance of triple-negative breast cancers to CDK4/6 inhibition
- Author
-
Alice Loo, Piotr Stepien, Avery S. Feit, David M. Sabatini, Johann Bergholz, Walter Michael, Monther Abu-Remaileh, Christopher Thomas Brain, Rinath Jeselsohn, Baishan Jiang, Deborah Butter, Michael D. Cameron, Carmine DeAngelis, Wojciech Michowski, Rachel Schiff, Deborah A. Dillon, Karolina Maria Nowak, Iga Stukan, Bojana Jovanovic, Jean J. Zhao, Tobias Otto, Nathanael S. Gray, Maria Ericsson, Anne Fassl, Piotr Sicinski, Ralph Tiedt, Myles Brown, Kornelia Polyak, Qing Sheng, Fassl, A., Brain, C., Abu-Remaileh, M., Stukan, I., Butter, D., Stepien, P., Feit, A. S., Bergholz, J., Michowski, W., Otto, T., Sheng, Q., Loo, A., Michael, W., Tiedt, R., De Angelis, C., Schiff, R., Jiang, B., Jovanovic, B., Nowak, K., Ericsson, M., Cameron, M., Gray, N., Dillon, D., Zhao, J. J., Sabatini, D. M., Jeselsohn, R., Brown, M., Polyak, K., and Sicinski, P.
- Subjects
endocrine system diseases ,medicine.drug_class ,Antibiotics ,03 medical and health sciences ,0302 clinical medicine ,Chloroquine ,Medicine ,skin and connective tissue diseases ,neoplasms ,Research Articles ,Cancer ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,integumentary system ,biology ,business.industry ,Kinase ,Cyclin-dependent kinase 2 ,Siramesine ,SciAdv r-articles ,Cell Biology ,enzymes and coenzymes (carbohydrates) ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Antidepressant ,Cyclin-dependent kinase 6 ,biological phenomena, cell phenomena, and immunity ,CDK4/6 Inhibition ,business ,Research Article ,medicine.drug - Abstract
This study presents strategies to render triple-negative breast cancers sensitive to CDK4/6 inhibitors., Inhibitors of cyclin-dependent kinases CDK4 and CDK6 have been approved for treatment of hormone receptor–positive breast cancers. In contrast, triple-negative breast cancers (TNBCs) are resistant to CDK4/6 inhibition. Here, we demonstrate that a subset of TNBC critically requires CDK4/6 for proliferation, and yet, these TNBC are resistant to CDK4/6 inhibition due to sequestration of CDK4/6 inhibitors into tumor cell lysosomes. This sequestration is caused by enhanced lysosomal biogenesis and increased lysosomal numbers in TNBC cells. We developed new CDK4/6 inhibitor compounds that evade the lysosomal sequestration and are efficacious against resistant TNBC. We also show that coadministration of lysosomotropic or lysosome-destabilizing compounds (an antibiotic azithromycin, an antidepressant siramesine, an antimalaria compound chloroquine) renders resistant tumor cells sensitive to currently used CDK4/6 inhibitors. Lastly, coinhibition of CDK2 arrested proliferation of CDK4/6 inhibitor-resistant cells. These observations may extend the use of CDK4/6 inhibitors to TNBCs that are refractory to current anti-CDK4/6 therapies.
- Published
- 2020
12. A gain-of-function mutation of JAK2 in myeloproliferative disorders.
- Author
-
Kralovics R, Passamonti F, Buser AS, Teo S, Tiedt R, Passweg JR, Tichelli A, Cazzola M, and Skoda RC
- Published
- 2005
13. Capmatinib plus nivolumab in pretreated patients with EGFR wild-type advanced non-small cell lung cancer.
- Author
-
Felip E, Metro G, Tan DSW, Wolf J, Mark M, Boyer M, Hughes BGM, Bearz A, Moro-Sibilot D, Le X, Puente J, Massuti B, Tiedt R, Wang Y, Xu C, Mardjuadi FI, and Cobo M
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Benzamides therapeutic use, Benzamides administration & dosage, Adult, Proto-Oncogene Proteins c-met genetics, Proto-Oncogene Proteins c-met metabolism, Aged, 80 and over, Acrylamides therapeutic use, Acrylamides administration & dosage, Imidazoles, Triazines, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung mortality, Nivolumab administration & dosage, Nivolumab therapeutic use, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, ErbB Receptors genetics, ErbB Receptors antagonists & inhibitors
- Abstract
Introduction: Dysregulated MET is an established oncogenic driver in non-small cell lung cancer (NSCLC). MET signaling may also suppress anticancer immune responses. Concomitant MET inhibition with capmatinib (a MET inhibitor) synergistically enhanced the efficacy of immunotherapies in murine cancer models, regardless of tumor dependency to MET signaling. Here, we report results of a multicenter, open-label, phase 2 study of capmatinib plus nivolumab (a PD-1 inhibitor) in patients with EGFR wild-type advanced NSCLC, previously treated with platinum-based chemotherapy., Methods: Patients were allocated into high-MET or low-MET groups according to MET expression determined by immunohistochemistry, MET gene copy number as assessed by fluorescence in-situ hybridization, and presence of MET exon 14 skipping mutation, then received capmatinib 400 mg, oral, twice daily in combination with nivolumab 3 mg/kg intravenously every 2 weeks. The primary endpoint was investigator-assessed 6-month progression-free survival (PFS) rate per RECIST v1.1., Results: The primary endpoint was met in both the high-MET (N = 16) and low-MET (N = 30) groups. In the high-MET and low-MET groups, respectively, the estimated mean 6-month PFS rate (95 % credible interval) by Bayesian analysis was 68.9 % (48.5-85.7) and 50.9 % (35.6-66.4). The Kaplan-Meier median PFS (95 % CI) was 6.2 months (3.5-19.2) and 4.2 months (1.8-7.4). The overall response rate (95 % CI) was 25.0 % (7.3-52.4) and 16.7 % (5.6-34.7). Most frequent treatment-related adverse events (≥30 % any grade, N = 46) were nausea (52.2 %), peripheral edema (34.8 %), and increased blood creatinine (30.4 %)., Conclusions: Capmatinib plus nivolumab showed clinical activity and manageable safety in pretreated patients with advanced EGFR wild-type NSCLC, independent of MET status., Trial Registration: ClinicalTrials.gov NCT02323126., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Enriqueta Felip: Consulting or advisory role for Abbvie, Amgen, Astra Zeneca, Bayer, Beigene, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi Sankyo, Eli Lilly, F. Hoffmann – La Roche, Genentech, Gilead, Glaxo Smith Kline, Janssen, Medscape, Merck Serono, Merck Sharp & Dohme, Novartis, Peervoice, Peptomyc, Pfizer, Regeneron, Sanofi, Takeda, Touch Oncology, Turning Point Therapeutics. Giulio Metro: No conflict of interest to declare. Daniel S. W. Tan: Consulting or advisory role for Novartis, Merck, Loxo, AstraZeneca, Roche, Pfizer, Amgen, and Janssen; travel, accommodation, and expenses from Pfizer, Boehringer Ingelheim, and Roche; honoraria from Bristol-Myers Squibb, Takeda, Novartis, Roche, and Pfizer; and research funding (institution) from Novartis, GlaxoSmithKline, Amgen, and AstraZeneca. Juergen Wolf: Advisory boards and lecture fees for Amgen, AstraZeneca, Bayer, Blueprint, BMS, Boehringer-Ingelheim, Chugai, Daiichi Sankyo, Ignyta, Janssen, Lilly, Loxo, MSD, Novartis, Pfizer, Roche, Seattle Genetics, Takeda; research support (to institution) from BMS, Janssen Pharmaceutica, Novartis, Pfizer. Michael Mark: Consulting fees from Amgen, Astra Zeneca, BMS, MSD, Pfizer, Takeda, Roche; travel support from Astra Zeneca, Roche, Takeda. Michael Boyer: Advisory board for Merck Sharpe and Dohme, Astra Zeneca, Amgen, Bristol Myers Squibb, and Roche. Brett G. M. Hughes: Advisory board for Novartis, Merck Sharpe and Dohme, AstraZeneca, Roche, Pfizer, Amgen, Bristol-Myers Squibb, Takeda, Sanofi and Eisai; and research funding (institution) from Amgen. Alessandra Bearz: Consulting/advisory fees from Pfizer, MSD, AstraZeneca, Eli Lilly, Boehringer, Ingelheim, Sanofi. Denis Moro-Sibilot: Consulting/advisory fees from Eli Lilly, AstraZeneca, BMS, Novartis, MSD, Daiichi Sankyo, Janssen, Pfizer, Roche, Abbvie, Takeda, Amgen; research funding to Institution from Eli Lilly, MSD, Astra Zeneca, Pfizer, BMS, Roche; travel Support from Roche, Takeda, MSD. Xiuning Le: Consulting/advisory fees from Eli Lilly, EMD Serono (Merck KGaA), AstraZeneca, Spectrum Pharmaceutics, Novartis, Regeneron, Boehringer Ingelheim, Hengrui Therapeutics, Bayer, Teligene, Taiho, Daiichi Sankyo, Janssen, Blueprint Medicines, Sensei Biotherapeutics, SystImmune, ArriVent, Abion, and Abbvie; research funding to Institution from Eli Lilly, EMD Serono, ArriVent, Dizal, Teligene, Regeneron, Janssen, ThermoFisher, Takeda, and Boehringer Ingelheim; travel support from EMD Serono, Janssen, and Spectrum Pharmaceutics; stock options from BlossomHill. Javier Puente: Research funding from Astellas Pharma, Pfizer; consulting or advisory role from Pfizer, Astellas Pharma, Janssen-Cilag, Merck Sharp & Dohme, Bayer, Roche, Bristol Myers Squibb, Clovis Oncology, Ipsen, Eisai, Sanofi; honoraria from Pfizer, Bristol Myers Squibb, Ipsen, AstraZeneca, Roche, MSD Oncology, Janssen-Cilag, Astellas Pharma, EUSA Pharma, Eisai, Pierre Fabre, Sanofi, Bayer; travel/accommodations/expenses from Pfizer, Roche, Janssen-Cilag, Bristol Myers Squibb, MSD Oncology. Bartomeu Massuti: Consulting fees from AstraZeneca, Roche and Sanofi; payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing or educational events from AstraZeneca, Boehringer Ingelheim, MSD, Roche and Takeda; and support for attending meetings and/or travel from AstraZeneca and MSD. Ralph Tiedt: Past employee of Novartis at the time of the study; current employee of Monte Rosa Therapeutics. Yingying Wang: Employee of Novartis. Chao Xu: Employee of Novartis. Feby I. Mardjuadi: Employee of Novartis. Manuel Cobo: Speaker for Novartis, AstraZeneca, Boehringer-Ingelheim, Roche, BMS, Lilly, MSD, Takeda, Kyowa, Pierre-fabre, Novocure, Sanofi, Jansen; consultant or advisory role for Novartis, AstraZeneca, Boehringer-Ingelheim, Roche, BMS, Lilly, MSD, Takeda, Phyzer, Kyowa, Sanofi, Jansen., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
14. Integrated CRISPR screening and drug profiling identifies combination opportunities for EGFR, ALK, and BRAF/MEK inhibitors.
- Author
-
Tiedt R, King FJ, Stamm C, Niederst MJ, Delach S, Zumstein-Mecker S, Meltzer J, Mulford IJ, Labrot E, Engstler BS, Baltschukat S, Kerr G, Golji J, Wyss D, Schnell C, Ainscow E, Engelman JA, Sellers WR, Barretina J, Caponigro G, and Porta DG
- Subjects
- Humans, Proto-Oncogene Proteins B-raf genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Neoplasm Recurrence, Local genetics, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, ErbB Receptors genetics, Receptor Protein-Tyrosine Kinases genetics, Mitogen-Activated Protein Kinase Kinases genetics, Mutation, Cell Line, Tumor, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology
- Abstract
Anti-tumor efficacy of targeted therapies is variable across patients and cancer types. Even in patients with initial deep response, tumors are typically not eradicated and eventually relapse. To address these challenges, we present a systematic screen for targets that limit the anti-tumor efficacy of EGFR and ALK inhibitors in non-small cell lung cancer and BRAF/MEK inhibitors in colorectal cancer. Our approach includes genome-wide CRISPR screens with or without drugs targeting the oncogenic driver ("anchor therapy"), and large-scale pairwise combination screens of anchor therapies with 351 other drugs. Interestingly, targeting of a small number of genes, including MCL1, BCL2L1, and YAP1, sensitizes multiple cell lines to the respective anchor therapy. Data from drug combination screens with EGF816 and ceritinib indicate that dasatinib and agents disrupting microtubules act synergistically across many cell lines. Finally, we show that a higher-order-combination screen with 26 selected drugs in two resistant EGFR-mutant lung cancer cell lines identified active triplet combinations., Competing Interests: Declaration of interests All authors are or have been employees and shareholders of Novartis. During the preparation of the manuscript, W.R.S. was a Board or SAB member and equity holder in Peloton Therapeutics, Ideaya Biosciences, Civetta Therapeutics, Scorpion Therapeutics, and Bluebird Bio and has consulted for Array, Astex, Dynamo Therapeutics, Ipsen, PearlRiver Bio, Sanofi, and Servier, and receives research funding from Pfizer Pharmaceuticals, Merck, Ideaya Biosciences, Boehringer-Ingelheim, and Deerfield Management., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. BET bromodomain inhibitors regulate keratinocyte plasticity.
- Author
-
Schutzius G, Kolter C, Bergling S, Tortelli F, Fuchs F, Renner S, Guagnano V, Cotesta S, Rueeger H, Faller M, Bouchez L, Salathe A, Nigsch F, Richards SM, Louis M, Gruber V, Aebi A, Turner J, Grandjean F, Li J, Dimitri C, Thomas JR, Schirle M, Blank J, Drueckes P, Vaupel A, Tiedt R, Manley PW, Klopp J, Hemmig R, Zink F, Leroy N, Carbone W, Roma G, Keller CG, Dales N, Beyerbach A, Zimmerlin A, Bonenfant D, Terranova R, Berwick A, Sahambi S, Reynolds A, Jennings LL, Ruffner H, Tarsa P, Bouwmeester T, Driver V, Frederiksen M, Lohmann F, and Kirkland S
- Subjects
- Animals, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins metabolism, Disease Models, Animal, Epidermis metabolism, Epidermis pathology, Fluorescence Resonance Energy Transfer, Gene Expression Regulation, High-Throughput Screening Assays, Humans, Keratinocytes drug effects, Keratinocytes metabolism, Keratinocytes pathology, Male, Mice, Mice, Inbred C57BL, Primary Cell Culture, Protein Isoforms antagonists & inhibitors, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Precursors antagonists & inhibitors, Protein Precursors genetics, Protein Precursors metabolism, Re-Epithelialization genetics, Skin Ulcer genetics, Skin Ulcer metabolism, Skin Ulcer pathology, Small Molecule Libraries chemistry, Structure-Activity Relationship, Transcription Factors antagonists & inhibitors, Transcription Factors metabolism, Transcription, Genetic, Wounds, Nonpenetrating genetics, Wounds, Nonpenetrating metabolism, Wounds, Nonpenetrating pathology, Cell Cycle Proteins genetics, Epidermis drug effects, Re-Epithelialization drug effects, Skin Ulcer drug therapy, Small Molecule Libraries pharmacology, Transcription Factors genetics, Wounds, Nonpenetrating drug therapy
- Abstract
Although most acute skin wounds heal rapidly, non-healing skin ulcers represent an increasing and substantial unmet medical need that urgently requires effective therapeutics. Keratinocytes resurface wounds to re-establish the epidermal barrier by transitioning to an activated, migratory state, but this ability is lost in dysfunctional chronic wounds. Small-molecule regulators of keratinocyte plasticity with the potential to reverse keratinocyte malfunction in situ could offer a novel therapeutic approach in skin wound healing. Utilizing high-throughput phenotypic screening of primary keratinocytes, we identify such small molecules, including bromodomain and extra-terminal domain (BET) protein family inhibitors (BETi). BETi induce a sustained activated, migratory state in keratinocytes in vitro, increase activation markers in human epidermis ex vivo and enhance skin wound healing in vivo. Our findings suggest potential clinical utility of BETi in promoting keratinocyte re-epithelialization of skin wounds. Importantly, this novel property of BETi is exclusively observed after transient low-dose exposure, revealing new potential for this compound class.
- Published
- 2021
- Full Text
- View/download PDF
16. Phase Ib Study of Ribociclib plus Fulvestrant and Ribociclib plus Fulvestrant plus PI3K Inhibitor (Alpelisib or Buparlisib) for HR + Advanced Breast Cancer.
- Author
-
Tolaney SM, Im YH, Calvo E, Lu YS, Hamilton E, Forero-Torres A, Bachelot T, Maur M, Fasolo A, Tiedt R, Nardi L, Stammberger U, Abdelhady AM, Ruan S, and Lee SC
- Subjects
- Aminopyridines adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cyclin-Dependent Kinase 4, Female, Fulvestrant, Humans, Morpholines, Purines, Receptors, Estrogen, Thiazoles, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Phosphatidylinositol 3-Kinases
- Abstract
Purpose: Resistance to treatment with endocrine therapy in patients with HR
+ , HER2- advanced breast cancer (ABC) is common and dual inhibition of CDK4/6 and PI3K pathways may delay the development of resistance. This phase Ib trial evaluates the safety and tolerability of triple and double regimens containing the CDK4/6 inhibitor ribociclib., Patients and Methods: In this open-label, multicenter, phase Ib study, 70 postmenopausal women with HR+ , HER2- ABC were enrolled into one of four treatment combinations: ribociclib (once daily, 3 weeks on, 1 week off) plus fulvestrant; ribociclib (continuous dosing) plus fulvestrant; ribociclib plus alpelisib plus fulvestrant; or ribociclib plus buparlisib plus fulvestrant., Results: The recommended phase II dose (RP2D) of ribociclib was confirmed to be 600 mg (3 weeks on, 1 week off) and 400 mg (continuous dosing) plus fulvestrant 500 mg. For the triple combination with buparlisib, the RP2D was ribociclib 400 mg plus buparlisib 30 mg plus fulvestrant 500 mg. Enrollment for the triple combinations was stopped due to unexpected toxicity. No RP2D was determined for the alpelisib combination. The safety profiles of the ribociclib plus fulvestrant combinations were consistent with those in previous studies. There was no marked difference in ribociclib exposure in the presence of triple-combination partners. The highest overall response rate was seen in the buparlisib triple combination (25.0%; 95% confidence interval, 9.8-46.7)., Conclusions: Ribociclib plus fulvestrant demonstrated safety in the treatment of patients with HR+ , HER2- ABC. Triple combinations with alpelisib or buparlisib plus fulvestrant are not recommended for phase II investigation. See related commentary by Clark et al., p. 371 ., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
17. A phase 1b study of the MET inhibitor capmatinib combined with cetuximab in patients with MET-positive colorectal cancer who had progressed following anti-EGFR monoclonal antibody treatment.
- Author
-
Delord JP, Argilés G, Fayette J, Wirth L, Kasper S, Siena S, Mesia R, Berardi R, Cervantes A, Dekervel J, Zhao S, Sun Y, Hao HX, Tiedt R, Vicente S, Myers A, and Siu LL
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Benzamides adverse effects, Benzamides pharmacokinetics, Cell Line, Tumor, Cetuximab adverse effects, Cetuximab pharmacokinetics, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Disease Progression, Drug Resistance, Neoplasm, ErbB Receptors antagonists & inhibitors, Female, Head and Neck Neoplasms drug therapy, Humans, Imidazoles adverse effects, Imidazoles pharmacokinetics, Male, Maximum Tolerated Dose, Middle Aged, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Proto-Oncogene Proteins c-met metabolism, Response Evaluation Criteria in Solid Tumors, Squamous Cell Carcinoma of Head and Neck drug therapy, Triazines adverse effects, Triazines pharmacokinetics, ras Proteins genetics, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Benzamides administration & dosage, Cetuximab administration & dosage, Colorectal Neoplasms drug therapy, Imidazoles administration & dosage, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins c-met antagonists & inhibitors, Triazines administration & dosage
- Abstract
Background Overcoming resistance to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs) in patients with KRAS wildtype (WT) metastatic colorectal cancer (mCRC) could help meet the needs of patients with limited treatment options. Methods In this phase 1b study, patients with N/KRAS WT, MET-positive mCRC who had progressed following anti-EGFR mAb treatment received escalating oral doses of capmatinib (150, 300, and 400 mg) twice daily plus weekly intravenous cetuximab (at the approved dose). The primary objective was to establish a recommended dose for expansion (RDE) of capmatinib in combination with cetuximab. Safety, preliminary activity, pharmacokinetics, and pharmacodynamics were also explored. Results Thirteen patients were enrolled. No patients experienced a dose-limiting toxicity at investigated doses; the RDE was established as capmatinib 400 mg twice daily plus cetuximab. All patients experienced adverse events (AEs) suspected to be related to the study treatment. Five patients (38.5%) reported study-drug-related AEs of grade 3/4 in severity. No patients achieved a complete or partial response according to RECIST v1.1; however, tumor shrinkage of 29-44% was observed in 4 patients. Conclusions Capmatinib plus cetuximab was well tolerated. Preliminary signs of activity were observed. Further investigation is warranted to obtain efficacy data and refine predictive biomarkers of response. Clinical trial registration NCT02205398.
- Published
- 2020
- Full Text
- View/download PDF
18. Novel inhibitors of the histone methyltransferase DOT1L show potent antileukemic activity in patient-derived xenografts.
- Author
-
Perner F, Gadrey JY, Xiong Y, Hatton C, Eschle BK, Weiss A, Stauffer F, Gaul C, Tiedt R, Perry JA, Armstrong SA, and Krivtsov AV
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Benzimidazoles pharmacokinetics, Drugs, Investigational pharmacokinetics, Drugs, Investigational therapeutic use, Enzyme Inhibitors pharmacokinetics, Humans, Jurkat Cells, K562 Cells, Leukemia pathology, Mice, Treatment Outcome, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Benzimidazoles therapeutic use, Enzyme Inhibitors therapeutic use, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Leukemia drug therapy
- Published
- 2020
- Full Text
- View/download PDF
19. Increased lysosomal biomass is responsible for the resistance of triple-negative breast cancers to CDK4/6 inhibition.
- Author
-
Fassl A, Brain C, Abu-Remaileh M, Stukan I, Butter D, Stepien P, Feit AS, Bergholz J, Michowski W, Otto T, Sheng Q, Loo A, Michael W, Tiedt R, DeAngelis C, Schiff R, Jiang B, Jovanovic B, Nowak K, Ericsson M, Cameron M, Gray N, Dillon D, Zhao JJ, Sabatini DM, Jeselsohn R, Brown M, Polyak K, and Sicinski P
- Abstract
Inhibitors of cyclin-dependent kinases CDK4 and CDK6 have been approved for treatment of hormone receptor-positive breast cancers. In contrast, triple-negative breast cancers (TNBCs) are resistant to CDK4/6 inhibition. Here, we demonstrate that a subset of TNBC critically requires CDK4/6 for proliferation, and yet, these TNBC are resistant to CDK4/6 inhibition due to sequestration of CDK4/6 inhibitors into tumor cell lysosomes. This sequestration is caused by enhanced lysosomal biogenesis and increased lysosomal numbers in TNBC cells. We developed new CDK4/6 inhibitor compounds that evade the lysosomal sequestration and are efficacious against resistant TNBC. We also show that coadministration of lysosomotropic or lysosome-destabilizing compounds (an antibiotic azithromycin, an antidepressant siramesine, an antimalaria compound chloroquine) renders resistant tumor cells sensitive to currently used CDK4/6 inhibitors. Lastly, coinhibition of CDK2 arrested proliferation of CDK4/6 inhibitor-resistant cells. These observations may extend the use of CDK4/6 inhibitors to TNBCs that are refractory to current anti-CDK4/6 therapies., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
20. Correction: The potent and selective cyclin-dependent kinases 4 and 6 inhibitor ribociclib (LEE011) is a versatile combination partner in preclinical cancer models.
- Author
-
Kim S, Tiedt R, Loo A, Horn T, Delach S, Kovats S, Haas K, Engstler BS, Cao A, Pinzon-Ortiz M, Mulford I, Acker MG, Chopra R, Brain C, Tomaso ED, Sellers WR, and Caponigro G
- Abstract
[This corrects the article DOI: 10.18632/oncotarget.26215.]., (Copyright: © 2019 Kim et al.)
- Published
- 2020
- Full Text
- View/download PDF
21. DOT1L inhibition is lethal for multiple myeloma due to perturbation of the endoplasmic reticulum stress pathway.
- Author
-
Dafflon C, Gaulis S, Barys L, Kapur K, Cornacchione V, Schukur L, Bergling S, Traggiai E, Jansky S, Hellmann L, Engstler BS, Kerr G, de Weck A, Ruddy DA, Naumann U, Stauffer F, Gaul C, Lin Y, Billy E, Weiss A, Hofmann F, Ito M, and Tiedt R
- Abstract
The histone 3 lysine 79 (H3K79) methyltransferase (HMT) DOT1L is known to play a critical role for growth and survival of MLL -rearranged leukemia. Serendipitous observations during high-throughput drug screens indicated that the use of DOT1L inhibitors might be expandable to multiple myeloma (MM). Through pharmacologic and genetic experiments, we could validate that DOT1L is essential for growth and viability of a subset of MM cell lines, in line with a recent report from another team. In vivo activity against established MM xenografts was observed with a novel DOT1L inhibitor. In order to understand the molecular mechanism of the dependency in MM, we examined gene expression changes upon DOT1L inhibition in sensitive and insensitive cell lines and discovered that genes belonging to the endoplasmic reticulum (ER) stress pathway and protein synthesis machinery were specifically suppressed in sensitive cells. Whole-genome CRISPR screens in the presence or absence of a DOT1L inhibitor revealed that concomitant targeting of the H3K4me3 methyltransferase SETD1B increases the effect of DOT1L inhibition. Our results provide a strong basis for further investigating DOT1L and SETD1B as targets in MM., Competing Interests: CONFLICTS OF INTEREST All authors are or have been employees of Novartis., (Copyright: © 2020 Dafflon et al.)
- Published
- 2020
- Full Text
- View/download PDF
22. A Phase Ib/II, open-label, multicenter study of INC280 (capmatinib) alone and in combination with buparlisib (BKM120) in adult patients with recurrent glioblastoma.
- Author
-
van den Bent M, Azaro A, De Vos F, Sepulveda J, Yung WKA, Wen PY, Lassman AB, Joerger M, Tabatabai G, Rodon J, Tiedt R, Zhao S, Kirsilae T, Cheng Y, Vicente S, Balbin OA, Zhang H, and Wick W
- Subjects
- Adult, Aged, Aminopyridines administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Benzamides, Brain Neoplasms genetics, Brain Neoplasms pathology, Female, Follow-Up Studies, Glioblastoma genetics, Glioblastoma pathology, Humans, Imidazoles administration & dosage, Male, Maximum Tolerated Dose, Middle Aged, Morpholines administration & dosage, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, PTEN Phosphohydrolase genetics, Prognosis, Proto-Oncogene Proteins c-met genetics, Tissue Distribution, Triazines administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Neoplasm Recurrence, Local drug therapy
- Abstract
Purpose: To estimate the maximum tolerated dose (MTD) and/or identify the recommended Phase II dose (RP2D) for combined INC280 and buparlisib in patients with recurrent glioblastoma with homozygous phosphatase and tensin homolog (PTEN) deletion, mutation or protein loss., Methods: This multicenter, open-label, Phase Ib/II study included adult patients with glioblastoma with mesenchymal-epithelial transcription factor (c-Met) amplification. In Phase Ib, patients received INC280 as capsules or tablets in combination with buparlisib. In Phase II, patients received INC280 only. Response was assessed centrally using Response Assessment in Neuro-Oncology response criteria for high-grade gliomas. All adverse events (AEs) were recorded and graded., Results: 33 patients entered Phase Ib, 32 with altered PTEN. RP2D was not declared due to potential drug-drug interactions, which may have resulted in lack of efficacy; thus, Phase II, including 10 patients, was continued with INC280 monotherapy only. Best response was stable disease in 30% of patients. In the selected patient population, enrollment was halted due to limited activity with INC280 monotherapy. In Phase Ib, the most common treatment-related AEs were fatigue (36.4%), nausea (30.3%) and increased alanine aminotransferase (30.3%). MTD was identified at INC280 Tab 300 mg twice daily + buparlisib 80 mg once daily. In Phase II, the most common AEs were headache (40.0%), constipation (30.0%), fatigue (30.0%) and increased lipase (30.0%)., Conclusion: The combination of INC280/buparlisib resulted in no clear activity in patients with recurrent PTEN-deficient glioblastoma. More stringent molecular selection strategies might produce better outcomes., Trial Registration: NCT01870726.
- Published
- 2020
- Full Text
- View/download PDF
23. A phase II study of the efficacy and safety of the MET inhibitor capmatinib (INC280) in patients with advanced hepatocellular carcinoma.
- Author
-
Qin S, Chan SL, Sukeepaisarnjaroen W, Han G, Choo SP, Sriuranpong V, Pan H, Yau T, Guo Y, Chen M, Ren Z, Xu J, Yen CJ, Lin ZZ, Manenti L, Gu Y, Sun Y, Tiedt R, Hao L, Song W, and Tanwandee T
- Abstract
Background: The objectives of this phase II study were to determine the clinical activity of the MET tyrosine kinase inhibitor capmatinib (INC280) in patients with MET-dysregulated advanced hepatocellular carcinoma (HCC) and to assess the safety, pharmacokinetics, and correlation of biomarkers with the response., Methods: This phase II, open-label, single-arm study evaluated twice daily (BID) oral capmatinib in a dose-determining stage, utilizing a Bayesian Logistic Regression Model (BLRM) subject to Escalation with Overdose Control criteria, safety, pharmacokinetics, and pharmacodynamic information to determine a recommended dose for expansion (RDE) evaluating efficacy in patients with MET-dysregulated HCC., Results: A total of 38 patients received treatment. In the dose-determining stage, patients received capmatinib 300 mg BID capsules ( n = 8), and in the expansion, patients received 600 mg BID capsules ( n = 28) or 400 mg BID tablets ( n = 2) based on the BLRM and other relevant clinical data. No predefined qualifying adverse events (AEs) were observed during the first 28 days of treatment, and the RDE was 600 mg BID capsules (equivalent pharmacokinetics to 400 mg BID tablets). The most common any causality AEs were nausea (42%), vomiting (37%), and diarrhea (34%). In the expansion stage, in a subgroup of 10 patients with MET-high HCC, the overall response rate was 30%, including 1 durable complete response (>600 days) and 2 partial responses [1 durable (>600 days)]., Conclusions: Single agent capmatinib at the RDE is tolerable with a manageable safety profile. Antitumor activity was seen in a subset of patients with MET-dysregulated (MET-high) HCC., Trial Registration: ClinicalTrials.gov: NCT01737827. https://clinicaltrials.gov/ct2/show/NCT01737827., Competing Interests: Conflict of interest statement: Su Pin Choo received funding, nonfinancial support, and honoraria from BMS, nonfinancial support and honoraria from Bayer, and honoraria from Novartis, Shire, Sirtex, Eisai, and Celgene. Virote Sriuranpong has received research support from Novartis. Shukui Qin, Stephen Lam Chan, Wattana Sukeepaisarnjaroen, Guohong Han, Hongming Pan, Thomas Yau, Yabing Guo, Minshan Chen, Zhenggang Ren, Jianming Xu, Chia-Jui Yen, Zhong-Zhe Lin, and Tawesak Tanwandee have no competing financial interests. Yi Gu, Yongjian Sun, Lu Hao, and Wenjie Song are employees of Novartis. Luigi Manenti and Ralph Tiedt are employees of Novartis and hold stock with Novartis., (© The Author(s), 2019.)
- Published
- 2019
- Full Text
- View/download PDF
24. New Potent DOT1L Inhibitors for in Vivo Evaluation in Mouse.
- Author
-
Stauffer F, Weiss A, Scheufler C, Möbitz H, Ragot C, Beyer KS, Calkins K, Guthy D, Kiffe M, Van Eerdenbrugh B, Tiedt R, and Gaul C
- Abstract
In MLL-rearranged cancer cells, disruptor of telomeric silencing 1-like protein (DOT1L) is aberrantly recruited to ectopic loci leading to local hypermethylation of H3K79 and consequently misexpression of leukemogenic genes. A structure-guided optimization of a HTS hit led to the discovery of DOT1L inhibitors with subnanomolar potency, allowing testing of the therapeutic principle of DOT1L inhibition in a preclinical mouse tumor xenograft model. Compounds displaying good exposure in mouse and nanomolar inhibition of target gene expression in cells were obtained and tested in vivo., Competing Interests: The authors declare the following competing financial interest(s): Authors are shareholders of Novartis and/or employees of Novartis., (Copyright © 2019 American Chemical Society.)
- Published
- 2019
- Full Text
- View/download PDF
25. A conditional inducible JAK2V617F transgenic mouse model reveals myeloproliferative disease that is reversible upon switching off transgene expression.
- Author
-
Chapeau EA, Mandon E, Gill J, Romanet V, Ebel N, Powajbo V, Andraos-Rey R, Qian Z, Kininis M, Zumstein-Mecker S, Ito M, Hynes NE, Tiedt R, Hofmann F, Eshkind L, Bockamp E, Kinzel B, Mueller M, Murakami M, Baffert F, and Radimerski T
- Subjects
- Amino Acid Substitution, Animals, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Mutation, Missense, Gene Expression Regulation, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Janus Kinase 2 biosynthesis, Janus Kinase 2 genetics, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Transgenes
- Abstract
Aberrant activation of the JAK/STAT pathway is thought to be the critical event in the pathogenesis of the chronic myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia and primary myelofibrosis. The most frequent genetic alteration in these pathologies is the activating JAK2V617F mutation, and expression of the mutant gene in mouse models was shown to cause a phenotype resembling the human diseases. Given the body of genetic evidence, it has come as a sobering finding that JAK inhibitor therapy only modestly suppresses the JAK2V617F allele burden, despite showing clear benefits in terms of reducing splenomegaly and constitutional symptoms in patients. To gain a better understanding if JAK2V617F is required for maintenance of myeloproliferative disease once it has evolved, we generated a conditional inducible transgenic JAK2V617F mouse model using the SCL-tTA-2S tet-off system. Our model corroborates that expression of JAK2V617F in hematopoietic stem and progenitor cells recapitulates key hallmarks of human myeloproliferative neoplasms, and exhibits gender differences in disease manifestation. The disease was found to be transplantable, and importantly, reversible when transgenic JAK2V617F expression was switched off. Our results indicate that mutant JAK2V617F-specific inhibitors should result in profound disease modification by disabling the myeloproliferative clone bearing mutant JAK2., Competing Interests: Some of the authors are full-time employees of Novartis Pharma AG (Emilie A. Chapeau, Emeline Mandon, Vincent Romanet, Nicolas Ebel, Rita Andraos-Rey, Zhiyan Qian, Miltos Kininis, Sabine Zumstein-Mecker, Ralph Tiedt, Francesco Hofmann, Matthias Mueller, Fabienne Baffert), or have been full-time employees of Novartis Pharma AG (Violetta Powajbo, Moriko Ito, Bernd Kinzel, Masato Murakami, Thomas Radimerski). Novartis Pharma AG had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Ruxolitinib is a marketed product of Incyte, for which Novartis Pharma AG received exclusive development and commercialization rights outside of the United States. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2019
- Full Text
- View/download PDF
26. Capmatinib (INC280) Is Active Against Models of Non-Small Cell Lung Cancer and Other Cancer Types with Defined Mechanisms of MET Activation.
- Author
-
Baltschukat S, Engstler BS, Huang A, Hao HX, Tam A, Wang HQ, Liang J, DiMare MT, Bhang HC, Wang Y, Furet P, Sellers WR, Hofmann F, Schoepfer J, and Tiedt R
- Subjects
- Animals, Benzamides, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Enzyme Activation drug effects, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Hepatocyte Growth Factor genetics, Hepatocyte Growth Factor metabolism, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met genetics, Stomach Neoplasms drug therapy, Stomach Neoplasms genetics, Stomach Neoplasms metabolism, Stomach Neoplasms pathology, Xenograft Model Antitumor Assays, Carcinoma, Non-Small-Cell Lung drug therapy, Drug Evaluation, Preclinical methods, Imidazoles pharmacology, Lung Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met metabolism, Triazines pharmacology
- Abstract
Purpose: The selective MET inhibitor capmatinib is being investigated in multiple clinical trials, both as a single agent and in combination. Here, we describe the preclinical data of capmatinib, which supported the clinical biomarker strategy for rational patient selection., Experimental Design: The selectivity and cellular activity of capmatinib were assessed in large cellular screening panels. Antitumor efficacy was quantified in a large set of cell line- or patient-derived xenograft models, testing single-agent or combination treatment depending on the genomic profile of the respective models., Results: Capmatinib was found to be highly selective for MET over other kinases. It was active against cancer models that are characterized by MET amplification, marked MET overexpression, MET exon 14 skipping mutations, or MET activation via expression of the ligand hepatocyte growth factor (HGF). In cancer models where MET is the dominant oncogenic driver, anticancer activity could be further enhanced by combination treatments, for example, by the addition of apoptosis-inducing BH3 mimetics. The combinations of capmatinib and other kinase inhibitors resulted in enhanced anticancer activity against models where MET activation co-occurred with other oncogenic drivers, for example EGFR activating mutations., Conclusions: Activity of capmatinib in preclinical models is associated with a small number of plausible genomic features. The low fraction of cancer models that respond to capmatinib as a single agent suggests that the implementation of patient selection strategies based on these biomarkers is critical for clinical development. Capmatinib is also a rational combination partner for other kinase inhibitors to combat MET-driven resistance., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
27. The potent and selective cyclin-dependent kinases 4 and 6 inhibitor ribociclib (LEE011) is a versatile combination partner in preclinical cancer models.
- Author
-
Kim S, Tiedt R, Loo A, Horn T, Delach S, Kovats S, Haas K, Engstler BS, Cao A, Pinzon-Ortiz M, Mulford I, Acker MG, Chopra R, Brain C, di Tomaso E, Sellers WR, and Caponigro G
- Abstract
Inhibition of cyclin-dependent kinases 4 and 6 (CDK4/6) is associated with robust antitumor activity. Ribociclib (LEE011) is an orally bioavailable CDK4/6 inhibitor that is approved for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer, in combination with an aromatase inhibitor, and is currently being evaluated in several additional trials. Here, we report the preclinical profile of ribociclib. When tested across a large panel of kinase active site binding assays, ribociclib and palbociclib were highly selective for CDK4, while abemaciclib showed affinity to several other kinases. Both ribociclib and abemaciclib showed slightly higher potency in CDK4 -dependent cells than in CDK6 -dependent cells, while palbociclib did not show such a difference. Profiling CDK4/6 inhibitors in large-scale cancer cell line screens in vitro confirmed that RB1 loss of function is a negative predictor of sensitivity. We also found that routinely used cellular viability assays measuring adenosine triphosphate levels as a proxy for cell numbers underestimated the effects of CDK4/6 inhibition, which contrasts with assays that assess cell number more directly. Robust antitumor efficacy and combination benefit was detected when ribociclib was added to encorafenib, nazartinib, or endocrine therapies in patient-derived xenografts., Competing Interests: CONFLICTS OF INTEREST All authors are current or previous employees and stockholders of Novartis.
- Published
- 2018
- Full Text
- View/download PDF
28. Targeting multiple nodes of MLL complexes to improve leukemia therapy.
- Author
-
Dafflon C, Tiedt R, and Schwaller J
- Published
- 2017
- Full Text
- View/download PDF
29. Project DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Relationships Uncovered by Large-Scale, Deep RNAi Screening.
- Author
-
McDonald ER 3rd, de Weck A, Schlabach MR, Billy E, Mavrakis KJ, Hoffman GR, Belur D, Castelletti D, Frias E, Gampa K, Golji J, Kao I, Li L, Megel P, Perkins TA, Ramadan N, Ruddy DA, Silver SJ, Sovath S, Stump M, Weber O, Widmer R, Yu J, Yu K, Yue Y, Abramowski D, Ackley E, Barrett R, Berger J, Bernard JL, Billig R, Brachmann SM, Buxton F, Caothien R, Caushi JX, Chung FS, Cortés-Cros M, deBeaumont RS, Delaunay C, Desplat A, Duong W, Dwoske DA, Eldridge RS, Farsidjani A, Feng F, Feng J, Flemming D, Forrester W, Galli GG, Gao Z, Gauter F, Gibaja V, Haas K, Hattenberger M, Hood T, Hurov KE, Jagani Z, Jenal M, Johnson JA, Jones MD, Kapoor A, Korn J, Liu J, Liu Q, Liu S, Liu Y, Loo AT, Macchi KJ, Martin T, McAllister G, Meyer A, Mollé S, Pagliarini RA, Phadke T, Repko B, Schouwey T, Shanahan F, Shen Q, Stamm C, Stephan C, Stucke VM, Tiedt R, Varadarajan M, Venkatesan K, Vitari AC, Wallroth M, Weiler J, Zhang J, Mickanin C, Myer VE, Porter JA, Lai A, Bitter H, Lees E, Keen N, Kauffmann A, Stegmeier F, Hofmann F, Schmelzle T, and Sellers WR
- Subjects
- Cell Line, Tumor, Gene Library, Gene Regulatory Networks, Humans, Multiprotein Complexes metabolism, Neoplasms metabolism, Oncogenes, RNA, Small Interfering, Signal Transduction, Transcription Factors metabolism, Neoplasms genetics, Neoplasms pathology, RNA Interference
- Abstract
Elucidation of the mutational landscape of human cancer has progressed rapidly and been accompanied by the development of therapeutics targeting mutant oncogenes. However, a comprehensive mapping of cancer dependencies has lagged behind and the discovery of therapeutic targets for counteracting tumor suppressor gene loss is needed. To identify vulnerabilities relevant to specific cancer subtypes, we conducted a large-scale RNAi screen in which viability effects of mRNA knockdown were assessed for 7,837 genes using an average of 20 shRNAs per gene in 398 cancer cell lines. We describe findings of this screen, outlining the classes of cancer dependency genes and their relationships to genetic, expression, and lineage features. In addition, we describe robust gene-interaction networks recapitulating both protein complexes and functional cooperation among complexes and pathways. This dataset along with a web portal is provided to the community to assist in the discovery and translation of new therapeutic approaches for cancer., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Resistance mechanisms to TP53-MDM2 inhibition identified by in vivo piggyBac transposon mutagenesis screen in an Arf -/- mouse model.
- Author
-
Chapeau EA, Gembarska A, Durand EY, Mandon E, Estadieu C, Romanet V, Wiesmann M, Tiedt R, Lehar J, de Weck A, Rad R, Barys L, Jeay S, Ferretti S, Kauffmann A, Sutter E, Grevot A, Moulin P, Murakami M, Sellers WR, Hofmann F, and Jensen MR
- Subjects
- Allografts, Animals, Antineoplastic Agents pharmacology, Biomarkers, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Disease Models, Animal, Genetic Drift, Humans, Kaplan-Meier Estimate, Mice, Mice, Knockout, Neoplasms genetics, Neoplasms metabolism, Neoplasms mortality, Neoplasms pathology, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 metabolism, bcl-X Protein genetics, bcl-X Protein metabolism, DNA Transposable Elements, Drug Resistance, Neoplasm genetics, Genetic Vectors genetics, Mutagenesis, Insertional, Proto-Oncogene Proteins c-mdm2 genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Inhibitors of double minute 2 protein (MDM2)-tumor protein 53 (TP53) interaction are predicted to be effective in tumors in which the TP53 gene is wild type, by preventing TP53 protein degradation. One such setting is represented by the frequent CDKN2A deletion in human cancer that, through inactivation of p14ARF , activates MDM2 protein, which in turn degrades TP53 tumor suppressor. Here we used piggyBac (PB) transposon insertional mutagenesis to anticipate resistance mechanisms occurring during treatment with the MDM2-TP53 inhibitor HDM201. Constitutive PB mutagenesis in Arf
- /- mice provided a collection of spontaneous tumors with characterized insertional genetic landscapes. Tumors were allografted in large cohorts of mice to assess the pharmacologic effects of HDM201. Sixteen out of 21 allograft models were sensitive to HDM201 but ultimately relapsed under treatment. A comparison of tumors with acquired resistance to HDM201 and untreated tumors identified 87 genes that were differentially and significantly targeted by the PB transposon. Resistant tumors displayed a complex clonality pattern suggesting the emergence of several resistant subclones. Among the most frequent alterations conferring resistance, we observed somatic and insertional loss-of-function mutations in transformation-related protein 53 ( Trp53 ) in 54% of tumors and transposon-mediated gain-of-function alterations in B-cell lymphoma-extra large ( Bcl-xL ), Mdm4 , and two TP53 family members, resulting in expression of the TP53 dominant negative truncations ΔNTrp63 and ΔNTrp73. Enhanced BCL-xL and MDM4 protein expression was confirmed in resistant tumors, as well as in HDM201-resistant patient-derived tumor xenografts. Interestingly, concomitant inhibition of MDM2 and BCL-xL demonstrated significant synergy in p53 wild-type cell lines in vitro. Collectively, our findings identify several potential mechanisms by which TP53 wild-type tumors may escape MDM2-targeted therapy.- Published
- 2017
- Full Text
- View/download PDF
31. Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma.
- Author
-
Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, Lennerz JK, Vu P, Deshpande V, Kambadakone A, Mussolin B, Reyes S, Henderson L, Sun JE, Van Seventer EE, Gurski JM Jr, Baltschukat S, Schacher-Engstler B, Barys L, Stamm C, Furet P, Ryan DP, Stone JR, Iafrate AJ, Getz G, Porta DG, Tiedt R, Bardelli A, Juric D, Corcoran RB, Bardeesy N, and Zhu AX
- Subjects
- Adult, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology, Cell Cycle Proteins, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, Circulating Tumor DNA genetics, Female, Gene Fusion, Humans, Male, Membrane Transport Proteins, Middle Aged, Mutation, Receptor, Fibroblast Growth Factor, Type 2 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 2 chemistry, Receptor, Fibroblast Growth Factor, Type 3 chemistry, Receptor, Fibroblast Growth Factor, Type 3 metabolism, Transcription Factor TFIIIA genetics, Antineoplastic Agents therapeutic use, Bile Duct Neoplasms drug therapy, Cholangiocarcinoma drug therapy, Drug Resistance, Neoplasm genetics, Phenylurea Compounds therapeutic use, Pyrimidines therapeutic use, Receptor, Fibroblast Growth Factor, Type 2 genetics
- Abstract
Genetic alterations in the fibroblast growth factor receptor (FGFR) pathway are promising therapeutic targets in many cancers, including intrahepatic cholangiocarcinoma (ICC). The FGFR inhibitor BGJ398 displayed encouraging efficacy in patients with FGFR2 fusion-positive ICC in a phase II trial, but the durability of response was limited in some patients. Here, we report the molecular basis for acquired resistance to BGJ398 in three patients via integrative genomic characterization of cell-free circulating tumor DNA (cfDNA), primary tumors, and metastases. Serial analysis of cfDNA demonstrated multiple recurrent point mutations in the FGFR2 kinase domain at progression. Accordingly, biopsy of post-progression lesions and rapid autopsy revealed marked inter- and intralesional heterogeneity, with different FGFR2 mutations in individual resistant clones. Molecular modeling and in vitro studies indicated that each mutation led to BGJ398 resistance and was surmountable by structurally distinct FGFR inhibitors. Thus, polyclonal secondary FGFR2 mutations represent an important clinical resistance mechanism that may guide the development of future therapeutic strategies. Significance: We report the first genetic mechanisms of clinical acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive ICC. Our findings can inform future strategies for detecting resistance mechanisms and inducing more durable remissions in ICC and in the wide variety of cancers where the FGFR pathway is being explored as a therapeutic target. Cancer Discov; 7(3); 252-63. ©2016 AACR. See related commentary by Smyth et al., p. 248 This article is highlighted in the In This Issue feature, p. 235 ., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
32. Discovery of Potent, Selective, and Structurally Novel Dot1L Inhibitors by a Fragment Linking Approach.
- Author
-
Möbitz H, Machauer R, Holzer P, Vaupel A, Stauffer F, Ragot C, Caravatti G, Scheufler C, Fernandez C, Hommel U, Tiedt R, Beyer KS, Chen C, Zhu H, and Gaul C
- Abstract
Misdirected catalytic activity of histone methyltransferase Dot1L is believed to be causative for a subset of highly aggressive acute leukemias. Targeting the catalytic domain of Dot1L represents a potential therapeutic approach for these leukemias. In the context of a comprehensive Dot1L hit finding strategy, a knowledge-based virtual screen of the Dot1L SAM binding pocket led to the discovery of 2 , a non-nucleoside fragment mimicking key interactions of SAM bound to Dot1L. Fragment linking of 2 and 3 , an induced back pocket binder identified in earlier studies, followed by careful ligand optimization led to the identification of 7 , a highly potent, selective and structurally novel Dot1L inhibitor.
- Published
- 2017
- Full Text
- View/download PDF
33. Optimization of a Fragment-Based Screening Hit toward Potent DOT1L Inhibitors Interacting in an Induced Binding Pocket.
- Author
-
Scheufler C, Möbitz H, Gaul C, Ragot C, Be C, Fernández C, Beyer KS, Tiedt R, and Stauffer F
- Abstract
Mixed lineage leukemia (MLL) gene rearrangement induces leukemic transformation by ectopic recruitment of disruptor of telomeric silencing 1-like protein (DOT1L), a lysine histone methyltransferase, leading to local hypermethylation of H3K79 and misexpression of genes (including HoxA), which drive the leukemic phenotype. A weak fragment-based screening hit identified by SPR was cocrystallized with DOT1L and optimized using structure-based ligand optimization to yield compound 8 (IC50 = 14 nM). This series of inhibitors is structurally not related to cofactor SAM and is not interacting within the SAM binding pocket but induces a pocket adjacent to the SAM binding site.
- Published
- 2016
- Full Text
- View/download PDF
34. Discovery of Novel Dot1L Inhibitors through a Structure-Based Fragmentation Approach.
- Author
-
Chen C, Zhu H, Stauffer F, Caravatti G, Vollmer S, Machauer R, Holzer P, Möbitz H, Scheufler C, Klumpp M, Tiedt R, Beyer KS, Calkins K, Guthy D, Kiffe M, Zhang J, and Gaul C
- Abstract
Oncogenic MLL fusion proteins aberrantly recruit Dot1L, a histone methyltransferase, to ectopic loci, leading to local hypermethylation of H3K79 and misexpression of HoxA genes driving MLL-rearranged leukemias. Inhibition of the methyltransferase activity of Dot1L in this setting is predicted to reverse aberrant H3K79 methylation, leading to repression of leukemogenic genes and tumor growth inhibition. In the context of our Dot1L drug discovery program, high-throughput screening led to the identification of 2, a weak Dot1L inhibitor with an unprecedented, induced pocket binding mode. A medicinal chemistry campaign, strongly guided by structure-based consideration and ligand-based morphing, enabled the discovery of 12 and 13, potent, selective, and structurally completely novel Dot1L inhibitors.
- Published
- 2016
- Full Text
- View/download PDF
35. Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth.
- Author
-
Harbinski F, Craig VJ, Sanghavi S, Jeffery D, Liu L, Sheppard KA, Wagner S, Stamm C, Buness A, Chatenay-Rivauday C, Yao Y, He F, Lu CX, Guagnano V, Metz T, Finan PM, Hofmann F, Sellers WR, Porter JA, Myer VE, Graus-Porta D, Wilson CJ, Buckler A, and Tiedt R
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, ErbB Receptors metabolism, High-Throughput Screening Assays, Humans, Mice, Mutation, Neoplasms drug therapy, Neoplasms genetics, Neoplasms pathology, Phosphorylation drug effects, Protein Kinase Inhibitors pharmacology, Receptor, ErbB-2 genetics, Signal Transduction drug effects, Transplantation, Heterologous, Neoplasms metabolism, Proto-Oncogene Proteins c-met metabolism, Receptor, ErbB-2 metabolism, Receptor, Fibroblast Growth Factor, Type 2 metabolism, Receptor, Fibroblast Growth Factor, Type 3 metabolism
- Abstract
The overall power of kinase inhibitors is substantially overshadowed by the acquisition of drug resistance. To address this issue, we systematically assessed the potential of secreted proteins to induce resistance to kinase inhibitors. To this end, we developed a high-throughput platform for screening a cDNA library encoding 3,432 secreted proteins in cellular assays. Using cancer cells originally dependent on either MET, FGFR2, or FGFR3, we observed a bypass of dependence through ligand-mediated activation of alternative receptor tyrosine kinases (RTK). Our findings indicate a broad and versatile potential for RTKs from the HER and FGFR families as well as MET to compensate for loss of each other. We further provide evidence that combined inhibition of simultaneously active RTKs can lead to an added anticancer effect.
- Published
- 2012
- Full Text
- View/download PDF
36. Structure--activity relationship and mode of action of N-(6-ferrocenyl-2-naphthoyl) dipeptide ethyl esters: novel organometallic anticancer compounds.
- Author
-
Mooney A, Tiedt R, Maghoub T, O'Donovan N, Crown J, White B, and Kenny PT
- Subjects
- Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Coordination Complexes chemistry, Coordination Complexes pharmacology, DNA Damage drug effects, Dipeptides chemistry, Dipeptides pharmacology, Drug Screening Assays, Antitumor, Esters, Ferrous Compounds chemistry, Ferrous Compounds pharmacology, Guanine chemistry, Humans, Metallocenes, Naphthalenes chemistry, Naphthalenes pharmacology, Oxidation-Reduction, Structure-Activity Relationship, Antineoplastic Agents chemical synthesis, Coordination Complexes chemical synthesis, Dipeptides chemical synthesis, Ferrous Compounds chemical synthesis, Naphthalenes chemical synthesis
- Abstract
In this article, we report the findings of a comprehensive structure-activity relationship study of N-(6-ferrocenyl-2-naphthoyl) dipeptide ethyl esters, in which novel analogues were designed, synthesized, and evaluated in vitro for antiproliferative effect. Two new compounds, 2 and 16, showed potent nanomolar activity in the H1299 NSCLC cell line, with exceptional IC(50) values of 0.13 and 0.14 μM, respectively. These compounds were also found to have significant activity in the Sk-Mel-28 malignant melanoma cell line (IC(50) values of 1.10 and 1.06 μM, respectively). Studies were also conducted to elucidate the mode of action of these novel organometallic anticancer compounds. Cell cycle analysis in the H1299 cell line suggests these compounds induce apoptosis, while guanine oxidation studies confirm that 2 is capable of generating oxidative damage via a ROS-mediated mechanism.
- Published
- 2012
- Full Text
- View/download PDF
37. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition.
- Author
-
Weigert O, Lane AA, Bird L, Kopp N, Chapuy B, van Bodegom D, Toms AV, Marubayashi S, Christie AL, McKeown M, Paranal RM, Bradner JE, Yoda A, Gaul C, Vangrevelinghe E, Romanet V, Murakami M, Tiedt R, Ebel N, Evrot E, De Pover A, Régnier CH, Erdmann D, Hofmann F, Eck MJ, Sallan SE, Levine RL, Kung AL, Baffert F, Radimerski T, and Weinstock DM
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, DNA Primers genetics, Female, Flow Cytometry, Gene Expression Profiling, HSP90 Heat-Shock Proteins metabolism, Humans, Immunoblotting, Immunohistochemistry, Isoxazoles therapeutic use, Janus Kinase 2 metabolism, Leukemia, B-Cell drug therapy, Leukemia, B-Cell genetics, Luciferases, Mice, Mice, Inbred BALB C, Mutagenesis, Mutation, Missense genetics, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Phosphorylation, RNA, Small Interfering genetics, Receptors, Cytokine genetics, Receptors, Cytokine metabolism, Resorcinols therapeutic use, X-Ray Microtomography, HSP90 Heat-Shock Proteins antagonists & inhibitors, Isoxazoles pharmacology, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 genetics, Leukemia, B-Cell enzymology, Myeloproliferative Disorders enzymology, Resorcinols pharmacology, Signal Transduction physiology
- Abstract
Enzymatic inhibitors of Janus kinase 2 (JAK2) are in clinical development for the treatment of myeloproliferative neoplasms (MPNs), B cell acute lymphoblastic leukemia (B-ALL) with rearrangements of the cytokine receptor subunit cytokine receptor-like factor 2 (CRLF2), and other tumors with constitutive JAK2 signaling. In this study, we identify G935R, Y931C, and E864K mutations within the JAK2 kinase domain that confer resistance across a panel of JAK inhibitors, whether present in cis with JAK2 V617F (observed in MPNs) or JAK2 R683G (observed in B-ALL). G935R, Y931C, and E864K do not reduce the sensitivity of JAK2-dependent cells to inhibitors of heat shock protein 90 (HSP90), which promote the degradation of both wild-type and mutant JAK2. HSP90 inhibitors were 100-1,000-fold more potent against CRLF2-rearranged B-ALL cells, which correlated with JAK2 degradation and more extensive blockade of JAK2/STAT5, MAP kinase, and AKT signaling. In addition, the HSP90 inhibitor AUY922 prolonged survival of mice xenografted with primary human CRLF2-rearranged B-ALL further than an enzymatic JAK2 inhibitor. Thus, HSP90 is a promising therapeutic target in JAK2-driven cancers, including those with genetic resistance to JAK enzymatic inhibitors.
- Published
- 2012
- Full Text
- View/download PDF
38. A drug resistance screen using a selective MET inhibitor reveals a spectrum of mutations that partially overlap with activating mutations found in cancer patients.
- Author
-
Tiedt R, Degenkolbe E, Furet P, Appleton BA, Wagner S, Schoepfer J, Buck E, Ruddy DA, Monahan JE, Jones MD, Blank J, Haasen D, Drueckes P, Wartmann M, McCarthy C, Sellers WR, and Hofmann F
- Subjects
- Amino Acid Substitution, Aminopyridines metabolism, Aminopyridines pharmacology, Animals, Antineoplastic Agents metabolism, Bridged Bicyclo Compounds, Heterocyclic metabolism, Cell Line, Transformed, Cell Line, Tumor, Crystallography, X-Ray, DNA Mutational Analysis, DNA, Neoplasm genetics, Enzyme Activation genetics, Humans, Mice, Models, Molecular, Mutagenesis, Neoplasms drug therapy, Neoplasms genetics, Protein Binding, Protein Conformation, Protein Kinase Inhibitors metabolism, Protein Structure, Tertiary, Proto-Oncogene Proteins c-met chemistry, Proto-Oncogene Proteins c-met genetics, Pyrazoles metabolism, Pyrazoles pharmacology, Quinolines metabolism, Receptors, Growth Factor chemistry, Receptors, Growth Factor genetics, Tyrosine metabolism, Antineoplastic Agents pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Drug Resistance, Neoplasm genetics, Mutation, Missense, Point Mutation, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Quinolines pharmacology, Receptors, Growth Factor antagonists & inhibitors
- Abstract
The emergence of drug resistance is a primary concern in any cancer treatment, including with targeted kinase inhibitors as exemplified by the appearance of Bcr-Abl point mutations in chronic myeloid leukemia (CML) patients treated with imatinib. In vitro approaches to identify resistance mutations in Bcr-Abl have yielded mutation spectra that faithfully recapitulated clinical observations. To predict resistance mutations in the receptor tyrosine kinase MET that could emerge during inhibitor treatment in patients, we conducted a resistance screen in BaF3 TPR-MET cells using the novel selective MET inhibitor NVP-BVU972. The observed spectrum of mutations in resistant cells was dominated by substitutions of tyrosine 1230 but also included other missense mutations and partially overlapped with activating MET mutations that were previously described in cancer patients. Cocrystallization of the MET kinase domain in complex with NVP-BVU972 revealed a key role for Y1230 in binding of NVP-BVU972, as previously reported for multiple other selective MET inhibitors. A second resistance screen in the same format with the MET inhibitor AMG 458 yielded a distinct spectrum of mutations rich in F1200 alterations, which is consistent with a different predicted binding mode. Our findings suggest that amino acid substitutions in the MET kinase domain of cancer patients need to be carefully monitored before and during treatment with MET inhibitors, as resistance may preexist or emerge. Compounds binding in the same manner as NVP-BVU972 might be particularly susceptible to the development of resistance through mutations in Y1230, a condition that may be addressed by MET inhibitors with alternative binding modes.
- Published
- 2011
- Full Text
- View/download PDF
39. Pronounced thrombocytosis in transgenic mice expressing reduced levels of Mpl in platelets and terminally differentiated megakaryocytes.
- Author
-
Tiedt R, Coers J, Ziegler S, Wiestner A, Hao-Shen H, Bornmann C, Schenkel J, Karakhanova S, de Sauvage FJ, Jackson CW, and Skoda RC
- Subjects
- Animals, Cell Differentiation physiology, Cells, Cultured, DNA, Complementary, Gene Dosage, Hematopoietic Stem Cells cytology, Megakaryocytes cytology, Megakaryocytes physiology, Mice, Mice, Inbred Strains, Mice, Transgenic, Platelet Count, Promoter Regions, Genetic physiology, Thrombopoietin metabolism, Transgenes physiology, Receptors, Thrombopoietin genetics, Receptors, Thrombopoietin metabolism, Thrombocytosis pathology, Thrombocytosis physiopathology, Thrombopoiesis physiology
- Abstract
We generated mice expressing a full-length Mpl transgene under the control of a 2-kb Mpl promoter in an Mpl(-/-) background, effectively obtaining mice that express full-length Mpl in the absence of other Mpl isoforms. These mice developed thrombocytosis with platelet levels approximately 5-fold higher than wild-type controls and markedly increased megakaryocyte numbers. The reintroduction of one wild-type Mpl allele restored normal platelet counts. We excluded the deletion of Mpl-tr, a dominant-negative isoform, as the underlying molecular cause for thrombocytosis. Instead, we found that transgene expression driven by the 2-kb Mpl promoter fragment was decreased during late megakaryocyte maturation, resulting in strongly diminished Mpl protein expression in platelets. Because platelets exert a negative feedback on thrombopoiesis by binding and consuming Tpo in the circulation through Mpl, we propose that the severe reduction of Mpl protein in platelets in Mpl-transgenic Mpl(-/-) mice shifts the equilibrium of this feedback loop, resulting in markedly elevated levels of megakaryocytes and platelets at steady state. Although the mechanism causing decreased expression of Mpl protein in platelets from patients with myeloproliferative disorders differs from this transgenic model, our results suggest that lowering Mpl protein in platelets could contribute to raising the platelet count.
- Published
- 2009
- Full Text
- View/download PDF
40. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice.
- Author
-
Tiedt R, Hao-Shen H, Sobas MA, Looser R, Dirnhofer S, Schwaller J, and Skoda RC
- Subjects
- Animals, Colony-Forming Units Assay, Granulocytes enzymology, Hematopoiesis, Extramedullary, Humans, Hyperplasia, Integrases metabolism, Megakaryocytes enzymology, Megakaryocytes pathology, Mice, Mice, Transgenic, Phenotype, Polycythemia Vera enzymology, Polycythemia Vera pathology, Primary Myelofibrosis enzymology, Primary Myelofibrosis pathology, Recombination, Genetic, Thrombocythemia, Essential enzymology, Thrombocythemia, Essential pathology, Transgenes, Janus Kinase 2 metabolism, Mutant Proteins metabolism, Myeloproliferative Disorders enzymology, Myeloproliferative Disorders pathology, Phenylalanine genetics, Valine genetics
- Abstract
An acquired somatic mutation in the JAK2 gene (JAK2-V617F) is present in the majority of patients with myeloproliferative disorders (MPDs). Several phenotypic manifestations (polycythemia vera [PV], essential thrombocythemia [ET], and primary myelofibrosis) can be associated with the same mutation. We generated JAK2-V617F transgenic mice using a human JAK2 gene with the sequences encoding the kinase domain placed in the inverse orientation and flanked by antiparallel loxP sites. Crossing mice of one transgenic line (FF1) with transgenic mice expressing Cre-recombinase under the control of the hematopoiesis specific Vav promoter led to expression of JAK2-V617F that was lower than the endogenous wild-type Jak2. These mice developed a phenotype resembling ET with strongly elevated platelet counts and moderate neutrophilia. Induction of the JAK2-V617F transgene with the interferon-inducible MxCre resulted in expression of JAK2-V617F approximately equal to wild-type Jak2 and a PV-like phenotype with increased hemoglobin, thrombocytosis, and neutrophilia. Higher levels of JAK2-V617F in mouse bone marrow by retroviral transduction caused a PV-like phenotype without thrombocytosis. These data are consistent with the hypothesis that the ratio of mutant to wild-type JAK2 is critical for the phenotypic manifestation. A similar correlation was also found in patients with MPD.
- Published
- 2008
- Full Text
- View/download PDF
41. Talin is required for integrin-mediated platelet function in hemostasis and thrombosis.
- Author
-
Petrich BG, Marchese P, Ruggeri ZM, Spiess S, Weichert RA, Ye F, Tiedt R, Skoda RC, Monkley SJ, Critchley DR, and Ginsberg MH
- Subjects
- Animals, Female, Gene Deletion, Homozygote, Integrin alpha2beta1 metabolism, Integrin beta1 metabolism, Integrin beta3 metabolism, Male, Mice, Mice, Inbred C57BL, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Platelet Membrane Glycoprotein IIb biosynthesis, Blood Platelets metabolism, Hemostasis, Integrins metabolism, Talin physiology, Thrombosis
- Abstract
Integrins are critical for hemostasis and thrombosis because they mediate both platelet adhesion and aggregation. Talin is an integrin-binding cytoplasmic adaptor that is a central organizer of focal adhesions, and loss of talin phenocopies integrin deletion in Drosophila. Here, we have examined the role of talin in mammalian integrin function in vivo by selectively disrupting the talin1 gene in mouse platelet precursor megakaryocytes. Talin null megakaryocytes produced circulating platelets that exhibited normal morphology yet manifested profoundly impaired hemostatic function. Specifically, platelet-specific deletion of talin1 led to spontaneous hemorrhage and pathological bleeding. Ex vivo and in vitro studies revealed that loss of talin1 resulted in dramatically impaired integrin alphaIIbbeta3-mediated platelet aggregation and beta1 integrin-mediated platelet adhesion. Furthermore, loss of talin1 strongly inhibited the activation of platelet beta1 and beta3 integrins in response to platelet agonists. These data establish that platelet talin plays a crucial role in hemostasis and provide the first proof that talin is required for the activation and function of mammalian alpha2beta1 and alphaIIbbeta3 integrins in vivo.
- Published
- 2007
- Full Text
- View/download PDF
42. Megakaryocyte-restricted MYH9 inactivation dramatically affects hemostasis while preserving platelet aggregation and secretion.
- Author
-
Léon C, Eckly A, Hechler B, Aleil B, Freund M, Ravanat C, Jourdain M, Nonne C, Weber J, Tiedt R, Gratacap MP, Severin S, Cazenave JP, Lanza F, Skoda R, and Gachet C
- Subjects
- Animals, Bleeding Time, Blood Platelets pathology, Female, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myosin Heavy Chains genetics, Myosin Heavy Chains metabolism, Myosin Heavy Chains physiology, Nonmuscle Myosin Type IIA deficiency, Nonmuscle Myosin Type IIA metabolism, Nonmuscle Myosin Type IIA physiology, Organ Specificity genetics, Thrombocytopenia blood, Thrombocytopenia genetics, Thrombocytopenia pathology, Blood Platelets metabolism, Hemostasis genetics, Megakaryocytes metabolism, Nonmuscle Myosin Type IIA genetics, Platelet Aggregation genetics
- Abstract
Mutations in the MYH9 gene encoding the nonmuscle myosin heavy chain IIA result in bleeding disorders characterized by a macrothrombocytopenia. To understand the role of myosin in normal platelet functions and in pathology, we generated mice with disruption of MYH9 in megakaryocytes. MYH9Delta mice displayed macrothrombocytopenia with a strong increase in bleeding time and absence of clot retraction. However, platelet aggregation and secretion in response to any agonist were near normal despite absence of initial platelet contraction. By contrast, integrin outside-in signaling was impaired, as observed by a decrease in integrin beta3 phosphorylation and PtdIns(3,4)P(2) accumulation following stimulation. Upon adhesion on a fibrinogen-coated surface, MYH9Delta platelets were still able to extend lamellipodia but without stress fiber-like formation. As a consequence, thrombus growth and organization, investigated under flow by perfusing whole blood over collagen, were strongly impaired. Thrombus stability was also decreased in vivo in a model of FeCl(3)-induced injury of carotid arteries. Overall, these results demonstrate that while myosin seems dispensable for aggregation and secretion in suspension, it plays a key role in platelet contractile phenomena and outside-in signaling. These roles of myosin in platelet functions, in addition to thrombocytopenia, account for the strong hemostatic defects observed in MYH9Delta mice.
- Published
- 2007
- Full Text
- View/download PDF
43. Pf4-Cre transgenic mice allow the generation of lineage-restricted gene knockouts for studying megakaryocyte and platelet function in vivo.
- Author
-
Tiedt R, Schomber T, Hao-Shen H, and Skoda RC
- Subjects
- Animals, Chromosomes, Artificial, Bacterial, Integrases analysis, Integrases genetics, Mice, Mice, Transgenic, Recombination, Genetic, Blood Platelets physiology, Cell Lineage, Megakaryocytes physiology, Platelet Factor 4 genetics, Transgenes
- Abstract
To generate transgenic mice that express Cre-recombinase exclusively in the megakaryocytic lineage, we modified a mouse bacterial artificial chromosome (BAC) clone by homologous recombination and replaced the first exon of the platelet factor 4 (Pf4), also called CXCL4, with a codon-improved Cre cDNA. Several strains expressing the transgene were obtained and one strain, Q3, was studied in detail. Crossing Q3 mice with the ROSA26-lacZ reporter strain showed that Cre-recombinase activity was confined to megakaryocytes. These results were further verified by crossing the Q3 mice with a strain containing loxP-flanked integrin beta1. Excision of this conditional allele in megakaryocytes was complete at the DNA level, and platelets were virtually devoid of the integrin beta1 protein. The Pf4-Cre transgenic strain will be a valuable tool to study megakaryopoiesis, platelet formation, and platelet function.
- Published
- 2007
- Full Text
- View/download PDF
44. Altered gene expression in myeloproliferative disorders correlates with activation of signaling by the V617F mutation of Jak2.
- Author
-
Kralovics R, Teo SS, Buser AS, Brutsche M, Tiedt R, Tichelli A, Passamonti F, Pietra D, Cazzola M, and Skoda RC
- Subjects
- Adaptor Proteins, Signal Transducing, Amino Acid Substitution, Biomarkers, Cytoskeletal Proteins, Enzyme Activation genetics, Female, Granulocyte Colony-Stimulating Factor administration & dosage, Granulocyte Colony-Stimulating Factor adverse effects, Granulocytes metabolism, Humans, Janus Kinase 2, Male, Polycythemia Vera complications, Polycythemia Vera drug therapy, Polycythemia Vera metabolism, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins genetics, Sepsis etiology, Sepsis metabolism, Tumor Suppressor Proteins genetics, Gene Expression Regulation genetics, Point Mutation, Polycythemia Vera genetics, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, Signal Transduction
- Abstract
We identified 13 new gene expression markers that were elevated and one marker, ANKRD15, that was down-regulated in patients with polycythemia vera (PV). These 14 markers, as well as the previously described PRV1 and NF-E2, exhibited the same gene expression alterations also in patients with exogenously activated granulocytes due to sepsis or granulocyte colony-stimulating factor (G-CSF) treatment. The recently described V617F mutation in the Janus kinase 2 (JAK2) gene allows defining subclasses of patients with myeloproliferative disorders based on the JAK2 genotype. Patients with PV who were homozygous or heterozygous for JAK2-V617F exhibited higher levels of expression of the 13 new markers, PRV1, and NF-E2 than patients without JAK2-V617F, whereas ANKRD15 was down-regulated in these patients. Our results suggest that the alterations in expression of the markers studied are due to the activation of the Jak/signal transducer and activator of transcription (STAT) pathway through exogenous stimuli (sepsis or G-CSF treatment), or endogenously through the JAK2-V617F mutation.
- Published
- 2005
- Full Text
- View/download PDF
45. Zim17, a novel zinc finger protein essential for protein import into mitochondria.
- Author
-
Burri L, Vascotto K, Fredersdorf S, Tiedt R, Hall MN, and Lithgow T
- Subjects
- Amino Acid Sequence, Arabidopsis Proteins metabolism, Base Sequence, DNA-Binding Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, Membrane Transport Proteins metabolism, Mitochondrial Membrane Transport Proteins metabolism, Mitochondrial Precursor Protein Import Complex Proteins, Mitochondrial Proteins genetics, Molecular Sequence Data, Protein Structure, Tertiary, Protein Transport physiology, RNA-Binding Proteins metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Zinc Fingers genetics, Mitochondria metabolism, Mitochondrial Proteins metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Zinc Fingers physiology
- Abstract
Translocation of precursor proteins across the mitochondrial membranes requires the coordinated action of multisubunit translocases in the outer and inner membrane, and the driving force for translocation across the inner membrane is provided by the matrix-located heat shock protein 70 (mtHsp70). The central components of the protein import machinery are essential. Here we describe Zim17, an essential protein with a zinc finger motif involved in protein import into mitochondria. Comparative genomics suggested a correction to the open reading frame of YNL310c, the gene encoding Zim17 in Saccharomyces cerevisiae. The revised open reading frame codes for a classic mitochondrial targeting signal, which is processed from Zim17 in the mitochondrial matrix. Loss of Zim17 selectively diminishes import of proteins into the matrix of mitochondria, but this loss of Zim17 is partially suppressed by overexpression of the J-protein Pam18/Tim14. We propose that Zim17 functions as an example of a "fractured" J-protein, where a protein like Zim17 contributes a zinc finger domain to Type III J-proteins, in toto providing for substrate loading onto Hsp70.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.