21 results on '"Brandon Antonakos"'
Search Results
2. Supplementary Table III from Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
James D. Griffin, Marion Wiesmann, Sean M. McDonough, Jens U. Wuerthner, Sebastien Jeay, Martin Sattler, Erik Nelson, Kristen Cowens, Ilene Galinsky, Richard Stone, Moriko Ito, Louise Barys, Brandon Antonakos, Jing Yuan, Irene Simkin, Takaomi Sanda, Tao Ren, Atsushi Nonami, Vesselina G. Cooke, Ensar Halilovic, and Ellen Weisberg
- Abstract
Supplementary Table III: AML patient samples tested with HDM2 inhibitor, CGM097: IC50 values for CGM097.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
James D. Griffin, Marion Wiesmann, Sean M. McDonough, Jens U. Wuerthner, Sebastien Jeay, Martin Sattler, Erik Nelson, Kristen Cowens, Ilene Galinsky, Richard Stone, Moriko Ito, Louise Barys, Brandon Antonakos, Jing Yuan, Irene Simkin, Takaomi Sanda, Tao Ren, Atsushi Nonami, Vesselina G. Cooke, Ensar Halilovic, and Ellen Weisberg
- Abstract
The tumor suppressor p53 is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (WT) p53, the HDM2 protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell-cycle arrest. Here, we investigated the ability of the novel HDM2 inhibitor CGM097 to potently and selectively kill WT p53-expressing AML cells. The antileukemic effects of CGM097 were studied using cell-based proliferation assays (human AML cell lines, primary AML patient cells, and normal bone marrow samples), apoptosis, and cell-cycle assays, ELISA, immunoblotting, and an AML patient–derived in vivo mouse model. CGM097 potently and selectively inhibited the proliferation of human AML cell lines and the majority of primary AML cells expressing WT p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against oncogenic FLT3-expressing cells cultured both in the absence as well as the presence of cytoprotective stromal-secreted cytokines, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. These data suggest that CGM097 is a promising treatment for AML characterized as harboring WT p53 as a single agent, as well as in combination with other therapies targeting oncogene-activated pathways that drive AML. Mol Cancer Ther; 14(10); 2249–59. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table I from Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
James D. Griffin, Marion Wiesmann, Sean M. McDonough, Jens U. Wuerthner, Sebastien Jeay, Martin Sattler, Erik Nelson, Kristen Cowens, Ilene Galinsky, Richard Stone, Moriko Ito, Louise Barys, Brandon Antonakos, Jing Yuan, Irene Simkin, Takaomi Sanda, Tao Ren, Atsushi Nonami, Vesselina G. Cooke, Ensar Halilovic, and Ellen Weisberg
- Abstract
Supplementary Table I. AML patient information.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figures 1-7 from Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
James D. Griffin, Marion Wiesmann, Sean M. McDonough, Jens U. Wuerthner, Sebastien Jeay, Martin Sattler, Erik Nelson, Kristen Cowens, Ilene Galinsky, Richard Stone, Moriko Ito, Louise Barys, Brandon Antonakos, Jing Yuan, Irene Simkin, Takaomi Sanda, Tao Ren, Atsushi Nonami, Vesselina G. Cooke, Ensar Halilovic, and Ellen Weisberg
- Abstract
Supplementary Figures 1-7 Supplementary Figure 1. Amplification and sequencing primers used for genotyping of p53 in AML patient samples. Supplementary Figure 2. Apoptosis data corresponding to representative bar graphs shown in Figure 1C. Supplementary Figure 3. Cell cycle analysis data corresponding to representative bar graphs shown in Figure 1D. Supplementary Figure 4. Inhibition of growth of mutant FLT3-positive AML cells by CGM097. Supplementary Figure 5. Potentiation of effects of PKC412 by CGM097 against mutant FLT3-positive AML cells. Supplementary Figure 6. Potentiation of effects of AC220 by CGM097 against mutant FLT3-positive AML cells cultured in the absence and the presence of cytoprotective SCM. Supplementary Figure 7 (A-F). Apoptosis data corresponding to representative bar graphs shown in Figure 5C. Supplementary Figure 7 (G-L). Apoptosis data corresponding to representative bar graphs shown in Figure 5C. Supplementary Figure 7 (M-R). Apoptosis data corresponding to representative bar graphs shown in Figure 5C.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table II from Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
James D. Griffin, Marion Wiesmann, Sean M. McDonough, Jens U. Wuerthner, Sebastien Jeay, Martin Sattler, Erik Nelson, Kristen Cowens, Ilene Galinsky, Richard Stone, Moriko Ito, Louise Barys, Brandon Antonakos, Jing Yuan, Irene Simkin, Takaomi Sanda, Tao Ren, Atsushi Nonami, Vesselina G. Cooke, Ensar Halilovic, and Ellen Weisberg
- Abstract
Supplementary Table II: Cell lines tested with HDM2 inhibitor, CGM097: IC50 values for CGM097.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table 2 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
XLSX file - 243KB, This table summarizes all significant genetic features that differentiate sensitive versus insensitive cell lines for TNKSi/MEKi combination. The first sheet lists all cell lines for each group. The second sheet lists all genetic features that are enriched in the sensitive group, using a False Discovery Rate (FDR) p-value lower than 0.25.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 5 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
PDF file - 68KB, Sensitivity of KRAS mutant cancer cells to TNKSi/MEKi combination. Synergistic cell lines are in red (cut-off of synergy score of 2).
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figures 1 - 6 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
PDF file - 660KB, Supplementary Figure S1. All RAS mutant cell lines have a higher sensitivity to the TNKSi/MEKi combination. Supplementary Figure S2. Validation of TNKSi/MEKi combination in KRAS mutant cancer cells. Supplementary Figure S3. Combinations with MEK inhibitor in the SW480 cell line. Supplementary Figure S4. TNKSi/MEKi combination leads to enhanced inhibition of AKT signaling activity. Supplementary Figure S5. TNKSi potentiates MEKi by releasing a feedback loop on FGFR2 signaling. Supplementary Figure S6. Consequences of combined inhibition of TNKS and MEK on FGFR2 and AKT signaling pathways in KRAS mutant cell lines.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Materials, Figure Legends, Table Legends from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
PDF file - 123KB
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Table 3 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
XLSX file - 234KB, This table shows the features of all 138 cancer cell lines tested for the TNKSi/MEKi combination in the unbiased combination screen. Cell line name, lineage, RAS mutation status and synergy score for the TNKSi/MEKi combination are represented. Cell lines are ranked according to their synergy score.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 1 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
PDF file - 81KB, List of identified synergistic cell lines for TNKSi/MEKi combination in the large-scale combination screen and their KRAS status. KRAS mutants are in red.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Table 4 from Inhibiting Tankyrases Sensitizes KRAS-Mutant Cancer Cells to MEK Inhibitors via FGFR2 Feedback Signaling
- Author
-
Wenlin Shao, Nika N. Danial, William R. Sellers, Mark D. Stump, Jebediah Ledell, Janine Steiger, Vesselina G. Cooke, Brandon Antonakos, Michael D. Jones, Daisy Flemming, Joshua M. Korn, Dmitriy Sonkin, Yuji Mishina, Yan Yan-Neale, Joseph Lehár, Kristen E. Hurov, and Marie Schoumacher
- Abstract
PDF file - 54KB, Chemical inhibitors used in the study.
- Published
- 2023
- Full Text
- View/download PDF
14. 888 Synergistic efficacy of the BRM/BRG1 ATPase inhibitor, FHD-286, and anti-PD-1 antibody in mouse syngeneic tumor models
- Author
-
Kana Ichikawa, Ammar Adam, Hsin-Jung Wu, David Lahr, Lan Xu, Brandon Antonakos, Liv Johannessen, Steven Bellon, Ryan Kruger, Richard Centore, and Martin Hentemann
- Published
- 2022
- Full Text
- View/download PDF
15. Targeted degradation of IKZF2 for cancer immunotherapy
- Author
-
Jonathan Solomon, Simone Bonazzi, Eva d'Hennezel, Rohan Beckwith, Lei Xu, Aleem Fazal, Anna Magracheva, Radha Ramesh, Artiom Cernijenko, Brandon Antonakos, Hyo-eun Bhang, Roxana Garcia Caro, Jennifer Cobb, Elizabeth Ornelas, Xiaolei Ma, Charles Wartchow, Matt Clifton, Ry Forseth, Bethany Fortnam, Hongbo Lu, Alfredo Csibi, Jennifer Tullai, Seth Carbonneau, Noel Thomsen, Jay Larrow, Barbara Chie-Leon, Dominik Hainzl, Yi Gu, Darlene Lu, Matthew Meyer, Dylan Alexander, Jacqueline Kinyamu-Akunda, Catherine Sabatos-Peyton, Natalie Dales, Frederic Zecri, Rishi Jain, Janine Shulok, Y. Karen Wang, Karin Briner, Jeffrey Porter, John Tallarico, Jeffrey Engelman, Glenn Dranoff, Jay Bradner, and Michael Visser
- Abstract
Growing malignant tumors must evade destruction by the immune system, a hurdle some malignancies overcome by attracting immune-suppressive regulatory T-cells (Tregs)1. The IKZF2 (Helios) transcription factor plays a crucial role in maintaining function and stability of Tregs, and IKZF2 deficiency enhances immune responses to tumors in mice2, suggesting IKZF2 may be an attractive target for cancer immunotherapy. Here we describe the discovery and characterization of DKY709, the first molecular glue degrader of IKZF2/4 which spares IKZF1/3. DKY709 was identified through a recruitment-guided medicinal chemistry campaign that redirected the degradation selectivity of CRBN binders towards IKZF2. The IKZF transcription factor selectivity of DKY709 was rationalized by the X-ray structure of the CRBN-DKY709-IKZF2(ZF2) ternary complex. Upon exposure to DKY709, human Tregs showed reduced suppressive activity and exhausted T-effector cells recovered IFNγ production. In vivo, oral treatment with DKY709 drove a rapid and sustained degradation of IKZF2 including in humans and led to delayed tumor growth in mice with humanized immune systems and enhanced immunization responses in monkeys. DKY709 is a first-in-class, potent and selective oral IKZF2/4 degrader currently being investigated in a phase 1 clinical trial as an immune-enhancing agent for cancer immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
16. Discovery and characterization of a selective IKZF2 glue degrader for cancer immunotherapy
- Author
-
Simone Bonazzi, Eva d’Hennezel, Rohan E.J. Beckwith, Lei Xu, Aleem Fazal, Anna Magracheva, Radha Ramesh, Artiom Cernijenko, Brandon Antonakos, Hyo-eun C. Bhang, Roxana García Caro, Jennifer S. Cobb, Elizabeth Ornelas, Xiaolei Ma, Charles A. Wartchow, Matthew C. Clifton, Ry R. Forseth, Bethany Hughes Fortnam, Hongbo Lu, Alfredo Csibi, Jennifer Tullai, Seth Carbonneau, Noel M. Thomsen, Jay Larrow, Barbara Chie-Leon, Dominik Hainzl, Yi Gu, Darlene Lu, Matthew J. Meyer, Dylan Alexander, Jacqueline Kinyamu-Akunda, Catherine A. Sabatos-Peyton, Natalie A. Dales, Frédéric J. Zécri, Rishi K. Jain, Janine Shulok, Y. Karen Wang, Karin Briner, Jeffery A. Porter, John A. Tallarico, Jeffrey A. Engelman, Glenn Dranoff, James E. Bradner, Michael Visser, and Jonathan M. Solomon
- Subjects
Pharmacology ,Clinical Biochemistry ,Drug Discovery ,Molecular Medicine ,Molecular Biology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
17. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases
- Author
-
Palermo Mark G, Timothy Michael Ramsey, Ping Zhu, Shumei Liu, Jay Larrow, Laura R. La Bonte, Rajesh Karki, Chen Christine Hiu-Tung, Kavitha Venkatesan, Jaison Jacob, Pascal D. Fortin, Francois Lenoir, Hui Gao, Guizhi Yang, Matthew J. Meyer, Ji-Hu Zhang, William R. Sellers, Michael Shultz, Denise Grunenfelder, Edmund Price, Jorge Garcia-Fortanet, Feng Fei, Zhouliang Chen, Gang Liu, Vesselina G. Cooke, Jing Yuan, Michelle Fodor, Ping Wang, Minying Pu, Nicholas Keen, Samuel B. Ho, Kathy Hsiao, Markus Warmuth, Travis Stams, Christopher Quinn, Mitsunori Kato, Subarna Shakya, Sarah Williams, Dyuti Majumdar, Peter Fekkes, Michael G. Acker, Cary Fridrich, Joanna Slisz, Huaixiang Hao, Matthew J. LaMarche, Ying-Nan P. Chen, Brandon Antonakos, Jason R. Dobson, Brant Firestone, Lawrence Blas Perez, Zhao B. Kang, Ho Man Chan, and Zhan Deng
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,Cell growth ,Protein tyrosine phosphatase ,Receptor tyrosine kinase ,Immune checkpoint ,Cell biology ,PTPN11 ,03 medical and health sciences ,030104 developmental biology ,Growth factor receptor ,biology.protein ,Signal transduction ,Tyrosine - Abstract
SHP099, a selective inhibitor of signalling meditator SHP2 with drug-like properties, has an allosteric mechanism of action whereby it stabilizes SHP2 in an auto-inhibited conformation, and suppresses RAS–ERK signalling and proliferation in receptor-tyrosine-kinase-driven cancer cell lines and mouse tumour xenograft models. The tyrosine phosphatase SHP2 is a key mediator of receptor tyrosine kinase (RTK) signalling, as well as being important in immune checkpoint pathways. Reduction of SHP2 activity suppresses tumour cell growth, and SHP2 is a potential, but so far elusive, therapeutic target in cancer. Pascal Fortin and colleagues report the development of a selective SHP2 inhibitor with drug-like properties. The inhibitor, SHP099, has an allosteric mechanism of action whereby it stabilizes SHP2 in an auto-inhibited conformation. It also suppresses RAS–ERK signalling to inhibit RTK-driven proliferation in human cancer cell lines and mouse tumour xenograft models. The non-receptor protein tyrosine phosphatase SHP2, encoded by PTPN11, has an important role in signal transduction downstream of growth factor receptor signalling and was the first reported oncogenic tyrosine phosphatase1. Activating mutations of SHP2 have been associated with developmental pathologies such as Noonan syndrome and are found in multiple cancer types, including leukaemia, lung and breast cancer and neuroblastoma1,2,3,4,5. SHP2 is ubiquitously expressed and regulates cell survival and proliferation primarily through activation of the RAS–ERK signalling pathway2,3. It is also a key mediator of the programmed cell death 1 (PD-1) and B- and T-lymphocyte attenuator (BTLA) immune checkpoint pathways6,7. Reduction of SHP2 activity suppresses tumour cell growth and is a potential target of cancer therapy8,9. Here we report the discovery of a highly potent (IC50 = 0.071 μM), selective and orally bioavailable small-molecule SHP2 inhibitor, SHP099, that stabilizes SHP2 in an auto-inhibited conformation. SHP099 concurrently binds to the interface of the N-terminal SH2, C-terminal SH2, and protein tyrosine phosphatase domains, thus inhibiting SHP2 activity through an allosteric mechanism. SHP099 suppresses RAS–ERK signalling to inhibit the proliferation of receptor-tyrosine-kinase-driven human cancer cells in vitro and is efficacious in mouse tumour xenograft models. Together, these data demonstrate that pharmacological inhibition of SHP2 is a valid therapeutic approach for the treatment of cancers.
- Published
- 2016
- Full Text
- View/download PDF
18. Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097
- Author
-
Vesselina G. Cooke, Atsushi Nonami, Takaomi Sanda, Tao Ren, Sébastien Jeay, Irene Simkin, Ilene Galinsky, Sean McDonough, Ellen Weisberg, Erik Nelson, Jens Wuerthner, Richard Stone, Marion Wiesmann, James D. Griffin, Kristen Cowens, Jing Yuan, Louise Barys, Martin Sattler, Moriko Ito, Brandon Antonakos, and Ensar Halilovic
- Subjects
Cancer Research ,Myeloid ,Cell cycle checkpoint ,Cell ,Gene Expression ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,Biology ,Piperazines ,Article ,Inhibitory Concentration 50 ,Mice, Inbred NOD ,Cell Line, Tumor ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Benzothiazoles ,Cell Proliferation ,Phenylurea Compounds ,Wild type ,Drug Synergism ,Proto-Oncogene Proteins c-mdm2 ,Cell cycle ,Isoquinolines ,Staurosporine ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular biology ,Tumor Burden ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Oncology ,Cell culture ,Female ,Tumor Suppressor Protein p53 - Abstract
The tumor suppressor p53 is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (WT) p53, the HDM2 protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell-cycle arrest. Here, we investigated the ability of the novel HDM2 inhibitor CGM097 to potently and selectively kill WT p53-expressing AML cells. The antileukemic effects of CGM097 were studied using cell-based proliferation assays (human AML cell lines, primary AML patient cells, and normal bone marrow samples), apoptosis, and cell-cycle assays, ELISA, immunoblotting, and an AML patient–derived in vivo mouse model. CGM097 potently and selectively inhibited the proliferation of human AML cell lines and the majority of primary AML cells expressing WT p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against oncogenic FLT3-expressing cells cultured both in the absence as well as the presence of cytoprotective stromal-secreted cytokines, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. These data suggest that CGM097 is a promising treatment for AML characterized as harboring WT p53 as a single agent, as well as in combination with other therapies targeting oncogene-activated pathways that drive AML. Mol Cancer Ther; 14(10); 2249–59. ©2015 AACR.
- Published
- 2015
- Full Text
- View/download PDF
19. Abstract 2084: Conformational activation and allosteric inhibition of SHP2 in RTK-driven cancers
- Author
-
Kavitha Venkatesan, Jaison Jacob, Shumei Liu, Fei Feng, Brandon Antonakos, Zhao B. Kang, Jonathan R. LaRochelle, Jason R. Dobson, Hui Gao, Laura R. La Bonte, Huaixiang Hao, Rajesh Karki, Samuel B. Ho, Guizhi Yang, Markus Warmuth, Ping Zhu, Matthew J. LaMarche, Brant Firestone, Matthew J. Meyer, Stephen C. Blacklow, Edmund Price, Kathy Hsiao, Jorge Garcia-Fortanet, Zhuoliang Chen, Chen Christine Hiu-Tung, Palermo Mark G, Vesselina G. Cooke, Cary Fridrich, Jay Larrow, Ping Wang, Sarah Williams, Ying-Nan P. Chen, Subarna Shakya, William R. Sellers, Nicholas Keen, Jing Yuan, Michael Shultz, Gang Liu, Michelle Fodor, Michael G. Acker, Pascal D. Fortin, Ho Man Chan, Timothy Michael Ramsey, Zhan Deng, Ji-Hu Zhang, Mitsunori Kato, Dyuti Majumdar, Peter Fekkes, Minying Pu, and Travis Stams
- Subjects
Cancer Research ,biology ,Philosophy ,Allosteric regulation ,Cancer therapy ,Protein tyrosine phosphatase ,medicine.disease ,Mapk signaling ,Oncology ,Allosteric enzyme ,Neuroblastoma ,Cancer research ,medicine ,biology.protein ,Tumor growth ,Majumdar - Abstract
The non-receptor protein tyrosine phosphatase (PTP) SHP2 is an important component of RTK signaling in response to growth factor stimulus and sits just upstream of the RAS-MAPK signaling cascade. The first oncogenic phosphatase to be identified, SHP2 is dysregulated in multiple human diseases including the developmental disorders Noonan and Leopard syndromes, as well as leukemia, lung cancer and neuroblastoma where aberrant activity of SHP2 leads to uncontrolled MAPK signaling. Cancer-associated activating mutations in SHP2 impart an “auto-on” state of the enzyme, boosting basal activity by shifting the equilibrium away from the auto-inhibited state. Reduction of SHP2 activity through genetic knockdown suppresses tumor growth, validating SHP2 as a target for cancer therapy. SHP099, a recently reported potent and selective allosteric inhibitor of SHP2, stabilizes the auto-inhibited form of SHP2 through interactions with the N-terminal SH2 and C-terminal PTP domains of the protein. SHP099 suppresses MAPK signaling in RTK amplified cancers resulting in suppressed proliferation in vitro and inhibition of tumor growth in mouse tumor xenograft models. Together, these data demonstrate the therapeutic potential of SHP2 inhibition in the treatment of cancer and other RAS/MAPK-linked diseases. Citation Format: Michael G. Acker, Ying-Nan P. Chen, Matthew J. LaMarche, Ho Man Chan, Peter Fekkes, Jorge Garcia-Fortanet, Jonathan R. LaRochelle, Brandon Antonakos, Christine Hiu-Tung Chen, Zhuoliang Chen, Vesselina G. Cooke, Jason R. Dobson, Zhan Deng, Fei Feng, Brant Firestone, Michelle Fodor, Cary Fridrich, Hui Gao, Huai-Xiang Hao, Jaison Jacob, Samuel Ho, Kathy Hsiao, Zhao B. Kang, Rajesh Karki, Mitsunori Kato, Jay Larrow, Laura R. La Bonte, Gang Liu, Shumei Liu, Dyuti Majumdar, Matthew J. Meyer, Mark Palermo, Minying Pu, Edmund Price, Subarna Shakya, Michael D. Shultz, Kavitha Venkatesan, Ping Wang, Markus Warmuth, Sarah Williams, Guizhi Yang, Jing Yuan, Ji-Hu Zhang, Ping Zhu, Stephen C. Blacklow, Timothy Ramsey, Nicholas J. Keen, William R. Sellers, Travis Stams, Pascal D. Fortin. Conformational activation and allosteric inhibition of SHP2 in RTK-driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2084. doi:10.1158/1538-7445.AM2017-2084
- Published
- 2017
- Full Text
- View/download PDF
20. Inhibiting Tankyrases sensitizes KRAS-mutant cancer cells to MEK inhibitors via FGFR2 feedback signaling
- Author
-
Vesselina G. Cooke, Marie Schoumacher, Daisy Flemming, Brandon Antonakos, Dmitriy Sonkin, Kristen E. Hurov, Yan Yan-Neale, Nika N. Danial, Joseph Lehar, Mark Stump, Yuji Mishina, Jebediah Ledell, Wenlin Shao, William R. Sellers, Joshua M. Korn, Janine L. Steiger, and Michael D. Jones
- Subjects
Cancer Research ,Morpholines ,Aminopyridines ,Mice, Nude ,Pyrimidinones ,Biology ,Fibroblast growth factor ,medicine.disease_cause ,Proto-Oncogene Proteins p21(ras) ,Erlotinib Hydrochloride ,Mice ,Tankyrases ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Acetamides ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 2 ,Mitosis ,Protein Kinase Inhibitors ,Feedback, Physiological ,Sulfonamides ,Aniline Compounds ,Wnt signaling pathway ,Drug Synergism ,MAP Kinase Kinase Kinases ,Molecular biology ,Xenograft Model Antitumor Assays ,Thiazoles ,Oncology ,Apoptosis ,Cancer cell ,Mutation ,Cancer research ,Quinazolines ,ras Proteins ,Female ,KRAS ,Signal transduction ,Signal Transduction - Abstract
Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis, and mitosis, offering attractive targets for anticancer treatment. Using unbiased combination screening in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors (MEKi) in KRAS-mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and antitumor activity both in vitro and in vivo than effects observed by previously reported MEKi combinations. Altogether, our results show how a novel combination of TNKS and MEK inhibitors can be highly effective in targeting KRAS-mutant cancers by suppressing a newly discovered resistance mechanism. Cancer Res; 74(12); 3294–305. ©2014 AACR.
- Published
- 2014
21. Abstract LB-107: Inhibiting TNKS sensitizes KRAS mutant cancer cells to MEK inhibitors by suppressing FGFR2 feedback signaling
- Author
-
Joshua M. Korn, Yuji Mishina, Yan Yan-Neale, Kristen Hurov, Brandon Antonakos, Joseph Lehar, Michael D. Jones, Nika N. Danial, Mark Stump, Dmitriy Sonkin, Vessilina Cooke, William R. Sellers, Daisy Flemming, Marie Schoumacher, and Wenlin Shao
- Subjects
Genetics ,Cancer Research ,business.industry ,MEK inhibitor ,Wnt signaling pathway ,Cancer ,medicine.disease ,medicine.disease_cause ,Oncology ,Apoptosis ,Tankyrases ,Cancer cell ,medicine ,Cancer research ,KRAS ,Signal transduction ,business - Abstract
Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis and mitosis, and are therefore considered as attractive targets for anti-cancer treatment. Using unbiased combination screens in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors in KRAS mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and anti-tumor activity both in vitro and in vivo than effects observed by previously reported MEK inhibitor combinations. Altogether, our data provides a strong rationale for combined targeting of TNKS and MEK in KRAS mutant cancers. Citation Format: Wenlin Shao, Marie Schoumacher, Kristen Hurov, Joseph Lehar, Yan Yan-Neale, Yuji Mishina, Dmitriy Sonkin, Joshua Korn, Daisy Flemming, Michael Jones, Brandon Antonakos, Vessilina Cooke, Mark Stump, Nika Danial, William Sellers. Inhibiting TNKS sensitizes KRAS mutant cancer cells to MEK inhibitors by suppressing FGFR2 feedback signaling. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-107. doi:10.1158/1538-7445.AM2014-LB-107
- 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.