96 results on '"Sean Caenepeel"'
Search Results
2. Supplementary Figure S9 from Dual Inhibition of Angiopoietin-TIE2 and MET Alters the Tumor Microenvironment and Prolongs Survival in a Metastatic Model of Renal Cell Carcinoma
- Author
-
Roberto Pili, Angela Coxon, Sean Caenepeel, Karen Rex, Sheng-yu Ku, Swathi Ramakrishnan, Eric Ciamporcero, Sreenivasulu Chintala, Kiersten Marie Miles, Li Shen, Remi Adelaiye-Ogala, Nur P. Damayanti, Ashley R. Orillion, and May Elbanna
- Abstract
Supplementary Figure S9 shows TIE2 and macrophage markers expression.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Data File 1 from In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Angela Coxon, Isabelle Dussault, Richard Kendall, Robert Radinsky, Jean-Christophe Harmange, Deborah M. Choquette, Daniel Baker, Michael A. Damore, Jonathan Werner, Jodi Moriguchi, Paula J. Kaplan-Lefko, Benny Amore, Teresa L. Burgess, Hue T. Kha, Martin A. Broome, Yihong Zhang, Yajing Yang, Sean Caenepeel, Karen Rex, and Paul E. Hughes
- Abstract
AMG 337 KINOME scan binding data
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Exploiting MCL1 Dependency with Combination MEK + MCL1 Inhibitors Leads to Induction of Apoptosis and Tumor Regression in KRAS-Mutant Non–Small Cell Lung Cancer
- Author
-
Aaron N. Hata, Paul E. Hughes, Cyril H. Benes, Angela Coxon, Sean P. Brown, Kristopher A. Sarosiek, Justin F. Gainor, Christopher G. Azzoli, Anna F. Farago, Zofia Piotrowska, Lecia V. Sequist, Colleen Keyes, John H. Shin, Daniel P. Cahill, Kevin A. Raskin, Cameron D. Wright, Michael Lanuti, Lorin A. Ferris, Kristof Vajda, Diamanda Rigas, Cameron Fraser, Chendi Li, Hannah L. Archibald, Maria Gomez-Caraballo, Nicole Phan, Samantha J. Bilton, Daria Timonina, Sean Caenepeel, Faria M. Siddiqui, and Varuna Nangia
- Abstract
BH3 mimetic drugs, which inhibit prosurvival BCL2 family proteins, have limited single-agent activity in solid tumor models. The potential of BH3 mimetics for these cancers may depend on their ability to potentiate the apoptotic response to chemotherapy and targeted therapies. Using a novel class of potent and selective MCL1 inhibitors, we demonstrate that concurrent MEK + MCL1 inhibition induces apoptosis and tumor regression in KRAS-mutant non–small cell lung cancer (NSCLC) models, which respond poorly to MEK inhibition alone. Susceptibility to BH3 mimetics that target either MCL1 or BCL-xL was determined by the differential binding of proapoptotic BCL2 proteins to MCL1 or BCL-xL, respectively. The efficacy of dual MEK + MCL1 blockade was augmented by prior transient exposure to BCL-xL inhibitors, which promotes the binding of proapoptotic BCL2 proteins to MCL1. This suggests a novel strategy for integrating BH3 mimetics that target different BCL2 family proteins for KRAS-mutant NSCLC.Significance:Defining the molecular basis for MCL1 versus BCL-xL dependency will be essential for effective prioritization of BH3 mimetic combination therapies in the clinic. We discover a novel strategy for integrating BCL-xL and MCL1 inhibitors to drive and subsequently exploit apoptotic dependencies of KRAS-mutant NSCLCs treated with MEK inhibitors.See related commentary by Leber et al., p. 1511.This article is highlighted in the In This Issue feature, p. 1494
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data File 2 from In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Angela Coxon, Isabelle Dussault, Richard Kendall, Robert Radinsky, Jean-Christophe Harmange, Deborah M. Choquette, Daniel Baker, Michael A. Damore, Jonathan Werner, Jodi Moriguchi, Paula J. Kaplan-Lefko, Benny Amore, Teresa L. Burgess, Hue T. Kha, Martin A. Broome, Yihong Zhang, Yajing Yang, Sean Caenepeel, Karen Rex, and Paul E. Hughes
- Abstract
Tumor cell line profiling data
- Published
- 2023
- Full Text
- View/download PDF
6. Data from In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Angela Coxon, Isabelle Dussault, Richard Kendall, Robert Radinsky, Jean-Christophe Harmange, Deborah M. Choquette, Daniel Baker, Michael A. Damore, Jonathan Werner, Jodi Moriguchi, Paula J. Kaplan-Lefko, Benny Amore, Teresa L. Burgess, Hue T. Kha, Martin A. Broome, Yihong Zhang, Yajing Yang, Sean Caenepeel, Karen Rex, and Paul E. Hughes
- Abstract
The MET receptor tyrosine kinase is involved in cell growth, survival, and invasion. Clinical studies with small molecule MET inhibitors have shown the role of biomarkers in identifying patients most likely to benefit from MET-targeted therapy. AMG 337 is an oral, small molecule, ATP-competitive, highly selective inhibitor of the MET receptor. Herein, we describe AMG 337 preclinical activity and mechanism of action in MET-dependent tumor models. These studies suggest MET is the only therapeutic target for AMG 337. In an unbiased tumor cell line proliferation screen (260 cell lines), a closely related analogue of AMG 337, Compound 5, exhibited activity in 2 of 260 cell lines; both were MET-amplified. Additional studies examining the effects of AMG 337 on the proliferation of a limited panel of cell lines with varying MET copy numbers revealed that high-level focal MET amplification (>12 copies) was required to confer MET oncogene addiction and AMG 337 sensitivity. One MET-amplified cell line, H1573 (>12 copies), was AMG 337 insensitive, possibly because of a downstream G12A KRAS mutation. Mechanism-of-action studies in sensitive MET-amplified cell lines demonstrated that AMG 337 inhibited MET and adaptor protein Gab-1 phosphorylation, subsequently blocking the downstream PI3K and MAPK pathways. AMG 337 exhibited potency in pharmacodynamic assays evaluating MET signaling in tumor xenograft models; >90% inhibition of Gab-1 phosphorylation was observed at 0.75 mg/kg. These findings describe the preclinical activity and mechanism of action of AMG 337 in MET-dependent tumor models and indicate its potential as a novel therapeutic for the treatment of MET-dependent tumors. Mol Cancer Ther; 15(7); 1568–79. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S2 from In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Angela Coxon, Isabelle Dussault, Richard Kendall, Robert Radinsky, Jean-Christophe Harmange, Deborah M. Choquette, Daniel Baker, Michael A. Damore, Jonathan Werner, Jodi Moriguchi, Paula J. Kaplan-Lefko, Benny Amore, Teresa L. Burgess, Hue T. Kha, Martin A. Broome, Yihong Zhang, Yajing Yang, Sean Caenepeel, Karen Rex, and Paul E. Hughes
- Abstract
High-level focal amplification of MET is associated with sensitivity to AMG 337
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 1 from Preclinical Evaluation of AMG 337, a Highly Selective Small Molecule MET Inhibitor, in Hepatocellular Carcinoma
- Author
-
Mingqiang Zhang, Angela Coxon, Liaoyuan Hu, Eric Huang, Jacqueline Huang, Hui Zhou, Wenge Zhong, Ouhong Wang, En-Tzu Tang, Ying He, Yanni Zhang, Karen Rex, Yuqing Shen, Sean Caenepeel, and Zhiqiang Du
- Abstract
Western blot analysis in responsive PDX model (LI0612) after 14 days treatment. Total and phosphorylated levels of the indicated proteins were evaluated to assess the effects of AMG 337 on MET signaling. Five to twelve PDX samples were processed for each group.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figures from In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Angela Coxon, Isabelle Dussault, Richard Kendall, Robert Radinsky, Jean-Christophe Harmange, Deborah M. Choquette, Daniel Baker, Michael A. Damore, Jonathan Werner, Jodi Moriguchi, Paula J. Kaplan-Lefko, Benny Amore, Teresa L. Burgess, Hue T. Kha, Martin A. Broome, Yihong Zhang, Yajing Yang, Sean Caenepeel, Karen Rex, and Paul E. Hughes
- Abstract
Figure S1. Kinase interaction map for AMG 337; Figure S2. In a large unbiased cancer cell line viability screen only MET-amplified cell lines were sensitive to treatment with an analogue of AMG 337 (Compound 5); Figure S3: AMG 337 inhibits the phosphorylation of MET and but not its downstream effectors in MET-amplified, KRAS mutant NSCLC cell line NCI-H1573; Figure S4. Cell lines harboring MET FISH scores >3 exhibited sensitivity to AMG 337. MET FISH analysis was performed on a subset of cancer cell lines exhibiting elevated MET gene copy number; Figure S5. Selective inhibition of MET exhibits partial effects on the viability of U-87 MG glioblastoma cells harboring an HGF/MET autocrine loop; Figure S6. Increases in MET gene number correlate with high levels of total MET protein; Figure S7. AMG 337 inhibits Gab-1 phosphorylation in a concentration dependent manner in the TPR-MET mouse tumor model.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Dual Inhibition of Angiopoietin-TIE2 and MET Alters the Tumor Microenvironment and Prolongs Survival in a Metastatic Model of Renal Cell Carcinoma
- Author
-
Roberto Pili, Angela Coxon, Sean Caenepeel, Karen Rex, Sheng-yu Ku, Swathi Ramakrishnan, Eric Ciamporcero, Sreenivasulu Chintala, Kiersten Marie Miles, Li Shen, Remi Adelaiye-Ogala, Nur P. Damayanti, Ashley R. Orillion, and May Elbanna
- Abstract
Receptor tyrosine kinase inhibitors have shown clinical benefit in clear cell renal cell carcinoma (ccRCC), but novel therapeutic strategies are needed. The angiopoietin/Tie2 and MET pathways have been implicated in tumor angiogenesis, metastases, and macrophage infiltration. In our study, we used trebananib, an angiopoietin 1/2 inhibitor, and a novel small-molecule MET kinase inhibitor in patient-derived xenograft (PDX) models of ccRCC. Our goal was to assess the ability of these compounds to alter the status of tumor-infiltrating macrophages, inhibit tumor growth and metastases, and prolong survival. Seven-week-old SCID mice were implanted subcutaneously or orthotopically with human ccRCC models. One month postimplantation, mice were treated with angiopoietin 1/2 inhibitor trebananib (AMG 386), MET kinase inhibitor, or combination. In our metastatic ccRCC PDX model, RP-R-02LM, trebananib alone, and in combination with a MET kinase inhibitor, significantly reduced lung metastases and M2 macrophage infiltration (P = 0.0075 and P = 0.0205, respectively). Survival studies revealed that treatment of the orthotopically implanted RP-R-02LM tumors yielded a significant increase in survival in both trebananib and combination groups. In addition, resection of the subcutaneously implanted primary tumor allowed for a significant survival advantage to the combination group compared with vehicle and both single-agent groups. Our results show that the combination of trebananib with a MET kinase inhibitor significantly inhibits the spread of metastases, reduces infiltrating M2-type macrophages, and prolongs survival in our highly metastatic ccRCC PDX model, suggesting a potential use for this combination therapy in treating patients with ccRCC.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data from Exploiting MCL1 Dependency with Combination MEK + MCL1 Inhibitors Leads to Induction of Apoptosis and Tumor Regression in KRAS-Mutant Non–Small Cell Lung Cancer
- Author
-
Aaron N. Hata, Paul E. Hughes, Cyril H. Benes, Angela Coxon, Sean P. Brown, Kristopher A. Sarosiek, Justin F. Gainor, Christopher G. Azzoli, Anna F. Farago, Zofia Piotrowska, Lecia V. Sequist, Colleen Keyes, John H. Shin, Daniel P. Cahill, Kevin A. Raskin, Cameron D. Wright, Michael Lanuti, Lorin A. Ferris, Kristof Vajda, Diamanda Rigas, Cameron Fraser, Chendi Li, Hannah L. Archibald, Maria Gomez-Caraballo, Nicole Phan, Samantha J. Bilton, Daria Timonina, Sean Caenepeel, Faria M. Siddiqui, and Varuna Nangia
- Abstract
Supplementary Figure 1-5, Supplementary Tables 1-3
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
Supplementary Figure 1 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary File from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Tissue expression datasets
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 3 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
Supplementary Figure 3 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
AMG 386 is an investigational first-in-class peptide-Fc fusion protein (peptibody) that inhibits angiogenesis by preventing the interaction of angiopoietin-1 (Ang1) and Ang2 with their receptor, Tie2. Although the therapeutic value of blocking Ang2 has been shown in several models of tumorigenesis and angiogenesis, the potential benefit of Ang1 antagonism is less clear. To investigate the consequences of Ang1 neutralization, we have developed potent and selective peptibodies that inhibit the interaction between Ang1 and its receptor, Tie2. Although selective Ang1 antagonism has no independent effect in models of angiogenesis-associated diseases (cancer and diabetic retinopathy), it induces ovarian atrophy in normal juvenile rats and inhibits ovarian follicular angiogenesis in a hormone-induced ovulation model. Surprisingly, the activity of Ang1 inhibitors seems to be unmasked in some disease models when combined with Ang2 inhibitors, even in the context of concurrent vascular endothelial growth factor inhibition. Dual inhibition of Ang1 and Ang2 using AMG 386 or a combination of Ang1- and Ang2-selective peptibodies cooperatively suppresses tumor xenograft growth and ovarian follicular angiogenesis; however, Ang1 inhibition fails to augment the suppressive effect of Ang2 inhibition on tumor endothelial cell proliferation, corneal angiogenesis, and oxygen-induced retinal angiogenesis. In no case was Ang1 inhibition shown to (a) confer superior activity to Ang2 inhibition or dual Ang1/2 inhibition or (b) antagonize the efficacy of Ang2 inhibition. These results imply that Ang1 plays a context-dependent role in promoting postnatal angiogenesis and that dual Ang1/2 inhibition is superior to selective Ang2 inhibition for suppression of angiogenesis in some postnatal settings. Mol Cancer Ther; 9(10); 2641–51. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Methods from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
File contains Supplementary Methods
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 3 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary Figure 3 Evaluation of DLL3 expression in tumor types other than SCLC.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 6 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary Figure 6 A, Expression of human CD25, CD69, PD-1, and 4-1BB on human CD4+ T cells isolated from mouse lungs harboring SHP-77 tumors 168 hours after treatment with AMG 757 or a control HLE BiTE® molecule. B, Expression of human CD25, CD69, PD-1, and 4-1BB on human CD8+ T cells isolated from mouse lungs harboring SHP-77 tumors 168 hours after treatment with either AMG 757 or a control HLE BiTE® molecule. *P < 0.05; **P < 0.01; *** P< 0.001
- Published
- 2023
- Full Text
- View/download PDF
19. Data from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Purpose:Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a high relapse rate, limited therapeutic options, and poor prognosis. We investigated the antitumor activity of AMG 757, a half-life extended bispecific T-cell engager molecule targeting delta-like ligand 3 (DLL3)—a target that is selectively expressed in SCLC tumors, but with minimal normal tissue expression.Experimental Design:AMG 757 efficacy was evaluated in SCLC cell lines and in orthotopic and patient-derived xenograft (PDX) mouse SCLC models. Following AMG 757 administration, changes in tumor volume, pharmacodynamic changes in tumor-infiltrating T cells (TILs), and the spatial relationship between the appearance of TILs and tumor histology were examined. Tolerability was assessed in nonhuman primates (NHPs).Results:AMG 757 showed potent and specific killing of even those SCLC cell lines with very low DLL3 expression (Conclusions:AMG 757 has a compelling safety and efficacy profile in preclinical studies making it a viable option for targeting DLL3-expressing SCLC tumors in the clinical setting.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Tables from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
This file contains Supplementary Tables 1-5
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Methods, Figure Legends, and References from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
Supplementary Methods, Figure Legends, and References from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 1 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary Figure 1 DLL3 is differentially expressed in SCLC tumors and cell lines compared with normal tissues.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 2 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary Figure 2 Representative photomicrographs of DLL3 immunostaining (brown color) in normal human tissues using immunohistochemistry.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure 5 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary Figure 5 A, DLL3 RNA expression levels in a panel of SCLC patient-derived xenograft (PDX) models compared with positive (NCI-H82) and negative (NCI-H460) control SCLC cell lines. Data are reported as relative quantification normalized to NCI-H460 cell line expression. B, DLL3 protein expression levels in a panel of SCLC PDX models compared with positive (NCI-H82) and negative (NCI-H460) control SCLC cell lines. GAPDH, glyceraldehyde 3-phosphate dehydrogenase.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 2 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
Supplementary Figure 2 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 4 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Author
-
Jonathan D. Oliner, Robert Radinsky, Richard Kendall, Tom Boone, Luke Li, Donald M. McDonald, Beverly L. Falcón, Isaac J. Hayward, Anthony Ndifor, Shao Xiong Wang, Linh Nguyen, Eunju Hurh, Russell Cattley, Grant Shimamoto, Eric Hsu, Mark L. Michaels, Seog Joon Han, Haejin Kim, David Cordover, Paul Hughes, Sean Caenepeel, Karen Rex, Ling Wang, James McCabe, Brad Bolon, Juan Estrada, Ji-Rong Sun, Tani Ann Lee, Dongyin Yu, Juan Leal, Stephen Kaufman, Hosung Min, James Bready, and Angela Coxon
- Abstract
Supplementary Figure 4 from Context-Dependent Role of Angiopoietin-1 Inhibition in the Suppression of Angiogenesis and Tumor Growth: Implications for AMG 386, an Angiopoietin-1/2–Neutralizing Peptibody
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 4 from AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Paul E. Hughes, Julie M. Bailis, Angela Coxon, Jude Canon, Pedro J. Beltran, Jennitte Stevens, Sean Caenepeel, Yajing Yang, Tobias Raum, Joshua T. Pearson, Matthias Friedrich, Oliver Homann, Fei Lee, Siyuan Liu, Jonathan Werner, Christopher M. Murawsky, Melissa Thomas, Petra Deegen, Jinghui Zhan, Juan Estrada, Edward K. Lobenhofer, Keegan Cooke, and Michael J. Giffin
- Abstract
Supplementary FIgure 4 AMG 757 demonstrated antitumor activity in the WM266-4 melanoma xenograft model.
- Published
- 2023
- Full Text
- View/download PDF
28. LKB1 loss rewires JNK-induced apoptotic protein dynamics through NUAKs and sensitizes KRAS-mutant NSCLC to combined KRASG12C + MCL-1 blockade
- Author
-
Aaron Hata, Chendi Li, Mohammed Syed, Yi Shen, Cameron Fraser, Jian Ouyang, Johannes Kreuzer, Sarah Clark, Audris Oh, Makeba Walcott, Robert Morris, Christopher Nabel, Sean Caenepeel, Anne Saiki, Karen Rex, J Lipford, Rebecca Heist, Jessica Lin, Wilhelm Haas, Kristopher Sarosiek, and Paul Hughes
- Abstract
The efficacy of molecularly targeted anti-cancer therapies may be limited by the presence of co-occurring mutations within a tumor.1-3 Conversely, these alterations may confer collateral vulnerabilities that can be leveraged for the development of novel therapeutic approaches. KRAS mutant lung cancers are distinguished by recurrent mutations in tumor suppressor genes such as TP53 and STK11/LKB1.4 However, clinically actionable targets associated with these alterations are largely undefined. BH3 mimetics targeting distinct pro-survival BCL-2 family proteins have been proposed to enhance the efficacy of targeted therapies, but no biomarkers exist that can predict which patients are most likely to respond. Here we show that inhibition of oncogenic signaling in KRAS-LKB1 mutant lung cancer cells creates a specific dependency on the anti-apoptotic protein MCL-1 for survival. We find that loss of the LKB1-NUAK signaling axis derepresses a JNK-mediated stress response to KRAS or MEK inhibition, allowing inhibitory phosphorylation of BCL-XL that alters BH3-protein interactions and makes cells vulnerable to concurrent MCL-1 inhibition. These results uncover a previously unknown role for LKB1 in regulating the mitochondrial apoptotic response of cancer cells independent of its tumor suppressor activity mediated by AMPK5-7 and SIK8,9 kinases and suggest a fundamental role for LKB1-NUAKs in suppressing the cellular stress response. Additionally, our study reveals a therapeutically targetable vulnerability in KRAS-LKB1 lung cancer cells and suggests a genotype-informed strategy for improving the efficacy of KRAS-targeted therapies.
- Published
- 2022
- Full Text
- View/download PDF
29. LKB1 loss rewires JNK-induced apoptotic protein dynamics through NUAKs and sensitizes KRAS-mutant NSCLC to combined KRASG12C + MCL-1 blockade
- Author
-
Chendi Li, Mohammed Usman Syed, Yi Shen, Cameron Fraser, Jian Ouyang, Johannes Kreuzer, Sarah E. Clark, Audris Oh, Makeba Walcott, Robert Morris, Christopher Nabel, Sean Caenepeel, Anne Y. Saiki, Karen Rex, J. Russell Lipford, Rebecca S. Heist, Jessica J. Lin, Wilhelm Haas, Kristopher Sarosiek, Paul E. Hughes, and Aaron N. Hata
- Abstract
The recently approved KRASG12C inhibitor sotorasib induces durable responses of KRASG12C-mutant non-small cell lung cancers (NSCLCs), however, some patients do not derive benefit. Identification of specific vulnerabilities conferred by co-occurring mutations may enable the development of biomarker-driven combination therapies in distinct subsets of patients. We report that co-occurring loss of STK11/LKB1 is associated with a drug-induced vulnerability of KRAS-mutant NSCLCs to MCL-1 inhibition. In LKB1-deficient cells, inhibition of KRAS-MAPK signaling leads to hyperactivated JNK, which phosphorylates BCL-XL and impairs its ability to sequester BIM, thus creating a dependency on MCL-1 for survival. In LKB1-proficient cells, LKB1 suppresses drug-induced JNK hyperactivation in a NUAK-dependent manner. Ex vivo treatment of tumors from LKB1-deficient but not LKB1 wild-type KRAS-mutant NSCLC patients with sotorasib or trametinib increased MCL-1 dependence. These results uncover a novel role for the LKB1-NUAK axis in regulation of apoptotic dependency and suggest a genotype-directed therapeutic approach for KRAS-LKB1 mutant NSCLC.
- Published
- 2022
- Full Text
- View/download PDF
30. AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer
- Author
-
Jinghui Zhan, Christopher Murawsky, Petra Deegen, Jonathan Werner, Oliver Homann, Siyuan Liu, Melissa Thomas, Angela Coxon, Keegan Cooke, Pedro J. Beltran, Joshua T. Pearson, Juan Estrada, Fei Lee, Tobias Raum, Julie M. Bailis, Matthias Friedrich, Jude Canon, Jennitte Stevens, Giffin Michael John, Sean Caenepeel, Yajing Yang, Paul E. Hughes, and Edward K. Lobenhofer
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,T-Lymphocytes ,T cell ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Antibodies, Bispecific ,Tumor Cells, Cultured ,Animals ,Humans ,Medicine ,Potency ,Lung cancer ,Cell Proliferation ,business.industry ,Intracellular Signaling Peptides and Proteins ,Antibodies, Monoclonal ,Membrane Proteins ,Half-life ,medicine.disease ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tolerability ,Cell culture ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Cancer research ,Female ,Non small cell ,business - Abstract
Purpose: Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a high relapse rate, limited therapeutic options, and poor prognosis. We investigated the antitumor activity of AMG 757, a half-life extended bispecific T-cell engager molecule targeting delta-like ligand 3 (DLL3)—a target that is selectively expressed in SCLC tumors, but with minimal normal tissue expression. Experimental Design: AMG 757 efficacy was evaluated in SCLC cell lines and in orthotopic and patient-derived xenograft (PDX) mouse SCLC models. Following AMG 757 administration, changes in tumor volume, pharmacodynamic changes in tumor-infiltrating T cells (TILs), and the spatial relationship between the appearance of TILs and tumor histology were examined. Tolerability was assessed in nonhuman primates (NHPs). Results: AMG 757 showed potent and specific killing of even those SCLC cell lines with very low DLL3 expression ( Conclusions: AMG 757 has a compelling safety and efficacy profile in preclinical studies making it a viable option for targeting DLL3-expressing SCLC tumors in the clinical setting.
- Published
- 2021
- Full Text
- View/download PDF
31. Dual Inhibition of Angiopoietin-TIE2 and MET Alters the Tumor Microenvironment and Prolongs Survival in a Metastatic Model of Renal Cell Carcinoma
- Author
-
Remi Adelaiye-Ogala, Karen Rex, Li Shen, Swathi Ramakrishnan, Roberto Pili, May Elbanna, Ashley Orillion, Eric Ciamporcero, Sean Caenepeel, Sreenivasulu Chintala, Sheng-Yu Ku, Nur P. Damayanti, Kiersten Marie Miles, and Angela Coxon
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Combination therapy ,Mice, SCID ,Article ,Angiopoietin-2 ,Angiopoietin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Carcinoma ,Animals ,Humans ,Neoplasm Metastasis ,Carcinoma, Renal Cell ,Tumor microenvironment ,biology ,business.industry ,medicine.disease ,Survival Analysis ,Primary tumor ,Angiopoietin receptor ,Clear cell renal cell carcinoma ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business - Abstract
Receptor tyrosine kinase inhibitors have shown clinical benefit in clear cell renal cell carcinoma (ccRCC), but novel therapeutic strategies are needed. The angiopoietin/Tie2 and MET pathways have been implicated in tumor angiogenesis, metastases, and macrophage infiltration. In our study, we used trebananib, an angiopoietin 1/2 inhibitor, and a novel small-molecule MET kinase inhibitor in patient-derived xenograft (PDX) models of ccRCC. Our goal was to assess the ability of these compounds to alter the status of tumor-infiltrating macrophages, inhibit tumor growth and metastases, and prolong survival. Seven-week-old SCID mice were implanted subcutaneously or orthotopically with human ccRCC models. One month postimplantation, mice were treated with angiopoietin 1/2 inhibitor trebananib (AMG 386), MET kinase inhibitor, or combination. In our metastatic ccRCC PDX model, RP-R-02LM, trebananib alone, and in combination with a MET kinase inhibitor, significantly reduced lung metastases and M2 macrophage infiltration (P = 0.0075 and P = 0.0205, respectively). Survival studies revealed that treatment of the orthotopically implanted RP-R-02LM tumors yielded a significant increase in survival in both trebananib and combination groups. In addition, resection of the subcutaneously implanted primary tumor allowed for a significant survival advantage to the combination group compared with vehicle and both single-agent groups. Our results show that the combination of trebananib with a MET kinase inhibitor significantly inhibits the spread of metastases, reduces infiltrating M2-type macrophages, and prolongs survival in our highly metastatic ccRCC PDX model, suggesting a potential use for this combination therapy in treating patients with ccRCC.
- Published
- 2020
- Full Text
- View/download PDF
32. Discovery and in Vivo Evaluation of Macrocyclic Mcl-1 Inhibitors Featuring an α-Hydroxy Phenylacetic Acid Pharmacophore or Bioisostere
- Author
-
Gwenaella Rescourio, Ana Z. Gonzalez, Salman Jabri, Brian Belmontes, Gordon Moody, Doug Whittington, Xin Huang, Sean Caenepeel, Mario Cardozo, Alan C. Cheng, David Chow, Hannah Dou, Adrie Jones, Ron C. Kelly, Yihong Li, Mike Lizarzaburu, Mei-Chu Lo, Rommel Mallari, Cesar Meleza, Yosup Rew, Scott Simonovich, Daqing Sun, Simon Turcotte, Xuelei Yan, Simon G. Wong, Evelyn Yanez, Manuel Zancanella, Jonathan Houze, Julio C. Medina, Paul E. Hughes, and Sean P. Brown
- Subjects
Sulfonamides ,Administration, Oral ,Biological Availability ,Mice, Nude ,Antineoplastic Agents ,Hydrogen Bonding ,Crystallography, X-Ray ,Xenograft Model Antitumor Assays ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Drug Stability ,Cell Line, Tumor ,Drug Design ,Drug Discovery ,Animals ,Humans ,Myeloid Cell Leukemia Sequence 1 Protein ,Molecular Medicine ,Female ,Multiple Myeloma ,Phenylacetates - Abstract
Overexpression of the antiapoptotic protein Mcl-1 provides a survival advantage to some cancer cells, making inhibition of this protein an attractive therapeutic target for the treatment of certain types of tumors. Herein, we report our efforts toward the identification of a novel series of macrocyclic Mcl-1 inhibitors featuring an α-hydroxy phenylacetic acid pharmacophore or bioisostere. This work led to the discovery of
- Published
- 2019
- Full Text
- View/download PDF
33. Abstract 2150: LKB1 loss rewires JNK-induced apoptotic protein dynamics through NUAKs and sensitizes KRAS-mutant non-small cell lung cancers to combined KRAS G12C + MCL-1 blockade
- Author
-
Chendi Li, Mohammed Usman Syed, Yi Shen, Audris Oh, Cameron Fraser, Johannes Kreuzer, Christopher Nabel, Kaitlyn Webster, Robert Morris, Sean Caenepeel, Anne Y. Saiki, Karen Rex, J. Russell Lipford, Wilhelm Hass, Kristopher Sarosiek, Paul E. Hughes, and Aaron Hata
- Subjects
Cancer Research ,Oncology - Abstract
The recent approval of the KRAS G12C inhibitor sotorasib (AMG 510) for non-small cell lung cancer (NSCLC) marked a milestone in the development of targeted therapies for KRAS mutant cancers. While sotorasib and other KRAS G12C inhibitors have demonstrated rapid and durable responses in the clinic, some patients do not achieve responses. The identification of specific vulnerabilities conferred by recurrent co-occurring mutations may enable the development of biomarker-driven combination therapies with enhanced activity in distinct subsets of patients. We screened a panel of KRAS-mutant NSCLC cell lines as well as patient-derived xenograft (PDX) mouse models and observed that loss of the tumor suppressor STK11/LKB1 is associated with increased sensitivity to combined MAPK (either the KRAS G12C inhibitor sotorasib or MEK inhibitor trametinib) and MCL-1 inhibition (AMG 176). Restoration of LKB1 expression in LKB1-deficient cell lines and PDX tumors blunted the apoptotic response to MAPK + MCL-1 inhibition; conversely, deletion of LKB1 in LKB1 wild-type models increased sensitivity. Mitochondrial apoptotic cell death is regulated by interactions between pro- (e.g., BIM) and anti-apoptotic (e.g., MCL-1, BCL-XL) BCL-2 family members. MAPK inhibition increases BIM, while MCL-1 inhibition prevents BIM sequestration by MCL-1, resulting in apoptosis. LKB1 deficient cells exhibit increased association of BIM and MCL-1 upon MAPK inhibition, effectively priming cells for death upon inhibition of MCL-1. Mechanistically, LKB1 deficiency and associated loss of NUAK phosphorylation leads to hyperactivation of the JNK phospho-kinase network. JNK phosphorylates MCL-1 at S64 and T163, which enhances BIM: MCL-1 protein-protein interaction. Conversely, JNK phosphorylates BCL-XL at S62 and prevents sequestration of BIM. This series of phosphorylation events increases MCL-1 dependence and creates a specific vulnerability of KRAS-LKB1 tumors to MAPK + MCL-1 inhibition. Consistent with this mechanism, ex vivo treatment of tumor tissue from a KRAS-LKB1 mutant NSCLC patient with sotorasib or trametinib increased MCL-1 dependent priming. These results reveal a novel link between LKB1 and the regulation of BCL-2 family proteins and provide preclinical rationale for evaluation of combined KRAS G12C + MCL-1 inhibitors for KRAS-LKB1 mutant NSCLC. Citation Format: Chendi Li, Mohammed Usman Syed, Yi Shen, Audris Oh, Cameron Fraser, Johannes Kreuzer, Christopher Nabel, Kaitlyn Webster, Robert Morris, Sean Caenepeel, Anne Y. Saiki, Karen Rex, J. Russell Lipford, Wilhelm Hass, Kristopher Sarosiek, Paul E. Hughes, Aaron Hata. LKB1 loss rewires JNK-induced apoptotic protein dynamics through NUAKs and sensitizes KRAS-mutant non-small cell lung cancers to combined KRAS G12C + MCL-1 blockade [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2150.
- Published
- 2022
- Full Text
- View/download PDF
34. Abstract 2779: A live tumor fragment (LTF) platform with real-time imaging for immune response assays
- Author
-
Tomasz Zal, Leung Kau Tang, Laura Hrycyniak, Anura Shrestha, Dinesh Joshi, Pichet Adstamongkonkul, Kelsey Tweed, Wesley A. Cox-Muranami, Sean Caenepeel, Hinco J. Gierman, Kevin Eliceiri, Kristina A. Matkowskyj, Sam J. Lubner, John Rafter, Mike Szulczewski, Maneesh Arora, and Jonathan Oliner
- Subjects
Cancer Research ,Oncology - Abstract
Immuno-Oncology (IO) therapies provide remarkable clinical benefits. However, too few patients respond, and there are no diagnostic tools that predict IO response with high accuracy. Moreover, as more IO drugs and combinations are approved, selecting the best IO-based regimen for each patient will become more complex. To facilitate this selection, we are developing an ex vivo live tumor fragment (LTF) screening platform that retains representation of a patient’s tumor microenvironment (TME) including immune cells, enabling development of deep neural networks to predict IO response and thereby individualize therapy. Here we present preliminary data from our proof-of-concept platform that performs tissue fragmentation/sorting, liquid handling, drug treatment, high-resolution dynamic imaging, and multiplex immuno-assays. Human tumor excisions were obtained from the Univ. of Wisconsin (IRB approved), and CT26 tumors were grown in mice subcutaneously. Live tumors were cut into 300 x 300 µm fragments of 100 - 300 µm thickness, sorted into multi-well plates, and cultured for 48 or 72 h in the presence or absence of anti-PD1 (nivolumab or mouse equivalent), anti-PD1 plus anti-CTLA4 (ipilimumab or mouse equivalent), or concanavalin A (ConA) as a positive control. Cell viability was measured by ATP luminescence assay or flow cytometry. Motility of CD8+ cells was tracked in human LTF using camelid VHH anti-hCD8a-AF594 and 3D multiphoton microscopy for 30 min. Supernatant cytokines were measured using bead immunoassay and T cell markers by flow cytometry. T cells were retained within LTFs, and the proportion of lymphocytes in LTFs was independent of fragment thickness (1.6%/2.1% for 300 µm, 1.4%/2.3% for 200 µm and 1.4%/2.8% for 100 µm thickness, for CD4+ and CD8+ T cells, respectively). Total cell viability and T cell viability exceeded 80% at 48h, and 3D LTF structure remained intact for at least 48 h. We treated LTFs with anti-PD1, anti-PD1 plus anti-CTLA4, or ConA and confirmed the presence of IFN-γ and 10 (mouse) or 16 (human) other cytokines associated with immune activation in both the ConA and anti-PD1 treated samples, but not the control. In human LTFs, cytokine panel upregulation was observed for anti-PD1 vs. control (p=1.1e-5) and anti-PD1 plus anti-CTLA4 vs. anti-PD1 (p=3.7e-9). Using multiphoton microscopy and CD8-binding nanobodies, we observed vigorous CD8+ T cell motility in human LTFs, with a speed of 10 µm/min, which is comparable to that reported in vivo. Our LTF platform has an immuno-competent TME in which we can detect cellular and secreted immune response markers, compare alternative treatments, and track the surveillance activity of infiltrating T cells. Future work will further advance the platform, enabling clinical trials for training and validating deep neural networks to predict response to checkpoint inhibitors and other IO drugs. Citation Format: Tomasz Zal, Leung Kau Tang, Laura Hrycyniak, Anura Shrestha, Dinesh Joshi, Pichet Adstamongkonkul, Kelsey Tweed, Wesley A. Cox-Muranami, Sean Caenepeel, Hinco J. Gierman, Kevin Eliceiri, Kristina A. Matkowskyj, Sam J. Lubner, John Rafter, Mike Szulczewski, Maneesh Arora, Jonathan Oliner. A live tumor fragment (LTF) platform with real-time imaging for immune response assays [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2779.
- Published
- 2022
- Full Text
- View/download PDF
35. Exploiting MCL1 Dependency with Combination MEK + MCL1 Inhibitors Leads to Induction of Apoptosis and Tumor Regression in KRAS-Mutant Non–Small Cell Lung Cancer
- Author
-
Lorin A. Ferris, Kristof Vajda, Hannah L. Archibald, Daniel P. Cahill, Justin F. Gainor, Diamanda Rigas, Sean P. Brown, Anna F. Farago, Aaron N. Hata, Chendi Li, Cyril H. Benes, Kevin A. Raskin, Varuna Nangia, Michael Lanuti, Cameron D. Wright, John H. Shin, Kristopher A. Sarosiek, Lecia V. Sequist, Christopher G. Azzoli, Angela Coxon, Daria Timonina, Faria M. Siddiqui, Zofia Piotrowska, Paul E. Hughes, Sean Caenepeel, Cameron Fraser, Maria Gomez-Caraballo, Nicole Phan, Colleen Keyes, and Samantha J. Bilton
- Subjects
0301 basic medicine ,A549 cell ,Chemotherapy ,Chemistry ,medicine.medical_treatment ,Mutant ,medicine.disease ,medicine.disease_cause ,Article ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Cell culture ,Apoptosis ,medicine ,Cancer research ,MCL1 ,KRAS ,Lung cancer ,neoplasms - Abstract
BH3 mimetic drugs, which inhibit prosurvival BCL2 family proteins, have limited single-agent activity in solid tumor models. The potential of BH3 mimetics for these cancers may depend on their ability to potentiate the apoptotic response to chemotherapy and targeted therapies. Using a novel class of potent and selective MCL1 inhibitors, we demonstrate that concurrent MEK + MCL1 inhibition induces apoptosis and tumor regression in KRAS-mutant non–small cell lung cancer (NSCLC) models, which respond poorly to MEK inhibition alone. Susceptibility to BH3 mimetics that target either MCL1 or BCL-xL was determined by the differential binding of proapoptotic BCL2 proteins to MCL1 or BCL-xL, respectively. The efficacy of dual MEK + MCL1 blockade was augmented by prior transient exposure to BCL-xL inhibitors, which promotes the binding of proapoptotic BCL2 proteins to MCL1. This suggests a novel strategy for integrating BH3 mimetics that target different BCL2 family proteins for KRAS-mutant NSCLC. Significance: Defining the molecular basis for MCL1 versus BCL-xL dependency will be essential for effective prioritization of BH3 mimetic combination therapies in the clinic. We discover a novel strategy for integrating BCL-xL and MCL1 inhibitors to drive and subsequently exploit apoptotic dependencies of KRAS-mutant NSCLCs treated with MEK inhibitors. See related commentary by Leber et al., p. 1511. This article is highlighted in the In This Issue feature, p. 1494
- Published
- 2018
- Full Text
- View/download PDF
36. Targeting MCL-1 in hematologic malignancies: Rationale and progress
- Author
-
Andrew H. Wei, Sean Caenepeel, Khalid Mezzi, Paul E. Hughes, Andrew Spencer, Roland B. Walter, Phuong Khanh Morrow, Zach McIver, David S. Siegel, Andrew W. Roberts, Aaron S. Rosenberg, and Anthony S. Stein
- Subjects
medicine.medical_specialty ,Myeloid ,Chronic lymphocytic leukemia ,Antineoplastic Agents ,Article ,chemistry.chemical_compound ,immune system diseases ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,neoplasms ,Multiple myeloma ,Hematology ,business.industry ,Venetoclax ,Myeloid leukemia ,medicine.disease ,Up-Regulation ,Myeloid Cell Leukemia Sequence 1 Protein ,Gene Expression Regulation, Neoplastic ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,chemistry ,Hematologic Neoplasms ,Cancer research ,business ,Multiple Myeloma ,Signal Transduction - Abstract
Myeloid cell leukemia sequence 1 (MCL-1) is an antiapoptotic protein that plays a key role in promoting cell survival in multiple myeloma (MM), acute myeloid leukemia (AML), and non-Hodgkin lymphoma (NHL). Overexpression of MCL-1 is associated with treatment resistance and poor prognosis; thus, MCL-1 inhibitors are rational therapeutic options for malignancies depending on MCL-1. Several MCL-1 inhibitors have entered clinical trials, including AZD5991, S64315, AMG 176, and AMG 397. A key area of investigation is whether MCL-1 inhibitors will complement the activity of BCL-2 inhibitors, such as venetoclax, and synergistically enhance anti-tumor efficacy when given in combination with other anti-cancer drugs. Another important question is whether a safe therapeutic window can be found for this new class of inhibitors. In summary, inhibition of MCL-1 shows potential as a treatment for hematologic malignancies and clinical evaluation of MCL-1 inhibitors is currently underway.
- Published
- 2019
37. Abstract 1050: Efficacy of AMG 176 in combination with gilteritinib in preclinical models of acute myeloid leukemia
- Author
-
Patricia McElroy, Karen Rex, Paul E. Hughes, Sean Caenepeel, Xiaoyue Chen, Brian Belmontes, and Tao Osgood
- Subjects
Cancer Research ,Oncology ,business.industry ,hemic and lymphatic diseases ,Gilteritinib ,Cancer research ,Myeloid leukemia ,Medicine ,business - Abstract
Acute Myeloid Leukemia (AML) is the most common and aggressive acute leukemia in adults, with a 25% 5-year survival rate. FLT3 is the most frequently mutated gene in AML. About 25% of AML patients harbor FLT3 internal tandem duplication (ITD) mutations, and about 8 % of patients harbor FLT3 tyrosine kinase domain (TKD) mutations. Both FLT3-ITD and FLT3-TKD mutations constitutively activate the protein, which causes poor survival. Gilteritinib is a highly potent and selective oral FLT3 inhibitor recently approved by the FDA. Although gilteritinib showed strong single agent activity in AML patients with FLT3 mutations, the development of gilteritinib resistance limits the long-term efficacy of this treatment, indicating that combination therapy may be advantageous for AML patients with FLT3 mutations. FLT3 mutations are known to be anti-apoptotic. Myeloid cell leukemia-1 (MCL-1), an anti-apoptotic protein, expressed in a large percentage of the AML patient population, plays a critical role in AML cell survival and drug resistance. AMG 176 is a potent, selective and orally bioavailable MCL-1 inhibitor, which induces rapid commitment to apoptosis in AML. Here we demonstrated that AMG 176 and gilteritinib combination treatment synergistically targeted FLT3-ITD mutated AML. A strong synergistic effect (Combination index Citation Format: Xiaoyue Chen, Sean Caenepeel, Brian Belmontes, Patricia L. McElroy, Karen Rex, Tao Osgood, Paul Hughes. Efficacy of AMG 176 in combination with gilteritinib in preclinical models of acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1050.
- Published
- 2021
- Full Text
- View/download PDF
38. Abstract 982: LKB1 loss rewires stress signaling-induced apoptotic protein dynamics and sensitizes KRAS-mutant non-small cell lung cancers to combined MAPK + MCL-1 blockade
- Author
-
Robert Morris, Yi Shen, Chendi Li, Cameron Fraser, Johannes Kreuzer, Audris Oh, Karen Rex, Mohammed Usman Syed, Kaitlyn A. Webster, Paul E. Hughes, Anne Y. Saiki, Aaron N. Hata, Wilhelm Hass, James Russell Lipford, Kristopher A. Sarosiek, and Sean Caenepeel
- Subjects
MAPK/ERK pathway ,Cancer Research ,business.industry ,MEK inhibitor ,Cell ,Cancer ,medicine.disease_cause ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Cell culture ,Apoptosis ,medicine ,Cancer research ,Phosphorylation ,KRAS ,business ,neoplasms - Abstract
Background: There are currently no approved targeted therapies for KRAS-mutant non-small cell lung cancers (NSCLC), which represent 25-30% of lung adenocarcinomas. The development of mutant-specific covalent inhibitors of KRAS G12C has invigorated hope that clinically effective KRAS-targeted therapies are within reach. While these agents have shown activity in early phase clinical trials, identification of specific vulnerabilities conferred by common co-occurring mutations in KRAS-mutant NSCLC may enable development of combination therapies with enhanced activity in distinct subsets of patients. Results: We screened a panel of KRAS-mutant NSCLC cell lines and observed that loss of the tumor suppressor STK11/LKB1 is associated with increased MCL-1 dependence and sensitivity to combined MAPK (either MEK inhibitor or KRAS G12C inhibitor AMG 510) and MCL-1 inhibition (AMG 176). Restoration of LKB1 expression in LKB1-deficient cell lines and mouse xenograft tumors blunted the apoptotic response to MAPK + MCL-1 inhibition; conversely, deletion of LKB1 in LKB1 wild-type models restored the sensitivity. Mechanistically, LKB1 deficiency is associated with an altered phosphoproteome and increased MCL-1-dependent apoptotic priming. LKB1 loss increased cellular stress leading to hyperactivation of JNK1, phosphorylation and stabilization of MCL-1 protein, and increased BIM sequestration by MCL-1. Upon suppression of MAPK signaling, LKB1-deficient cells exhibited greater levels of BIM bound to MCL-1 that could be liberated by AMG 176 to induce apoptosis. Consistent with these results, ex vivo treatment of tumor tissue from a KRAS-LKB1 mutant NSCLC patient with MEK inhibitor or AMG 510 increased MCL-1 dependent priming. Conclusion: These results uncover a novel link between LKB1, cellular stress, and the regulation of MCL-1. LKB1 loss confers a dependency on MCL-1 that can be exploited therapeutically. Moreover, our study provides preclinical rationale for the exploration of combined KRAS G12C + MCL-1 inhibitors, particularly for KRAS-LKB1 mutant patients who respond poorly to standard-of-care checkpoint inhibitor therapy. Citation Format: Chendi Li, Yi Shen, Mohammed Usman Syed, Audris Oh, Cameron Fraser, Johannes Kreuzer, Kaitlyn Webster, Robert Morris, Sean Caenepeel, Anne Y. Saiki, Karen Rex, James Lipford, Wilhelm Hass, Kristopher Sarosiek, Paul E. Hughes, Aaron N. Hata. LKB1 loss rewires stress signaling-induced apoptotic protein dynamics and sensitizes KRAS-mutant non-small cell lung cancers to combined MAPK + MCL-1 blockade [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 982.
- Published
- 2021
- Full Text
- View/download PDF
39. Targeted Therapy and Checkpoint Immunotherapy Combinations for the Treatment of Cancer
- Author
-
Lawren C. Wu, Paul E. Hughes, and Sean Caenepeel
- Subjects
Vascular Endothelial Growth Factor A ,0301 basic medicine ,MAPK/ERK pathway ,Angiogenesis ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,B7-H1 Antigen ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Combined Modality Therapy ,CTLA-4 Antigen ,Molecular Targeted Therapy ,Extracellular Signal-Regulated MAP Kinases ,business.industry ,Antibodies, Monoclonal ,Cancer ,Immunotherapy ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Signal transduction ,business ,Signal Transduction - Abstract
Many advances in the treatment of cancer have been driven by the development of targeted therapies that inhibit oncogenic signaling pathways and tumor-associated angiogenesis, as well as by the recent development of therapies that activate a patient's immune system to unleash antitumor immunity. Some targeted therapies can have effects on host immune responses, in addition to their effects on tumor biology. These immune-modulating effects, such as increasing tumor antigenicity or promoting intratumoral T cell infiltration, provide a rationale for combining these targeted therapies with immunotherapies. Here, we discuss the immune-modulating effects of targeted therapies against the MAPK and VEGF signaling pathways, and how they may synergize with immunomodulatory antibodies that target PD1/PDL1 and CTLA4. We critically examine the rationale in support of these combinations in light of the current understanding of the underlying mechanisms of action of these therapies. We also discuss the available preclinical and clinical data for these combination approaches and their implications regarding mechanisms of action. Insights from these studies provide a framework for considering additional combinations of targeted therapies and immunotherapies for the treatment of cancer.
- Published
- 2016
- Full Text
- View/download PDF
40. Preclinical Evaluation of AMG 337, a Highly Selective Small Molecule MET Inhibitor, in Hepatocellular Carcinoma
- Author
-
Ying He, Ouhong Wang, Yanni Zhang, Zhiqiang Du, En-Tzu Tang, Eric Huang, Hui Zhou, Karen Rex, Wenge Zhong, Yuqing Shen, Mingqiang Zhang, Angela Coxon, Sean Caenepeel, Jacqueline Huang, and Liaoyuan Hu
- Subjects
0301 basic medicine ,Cancer Research ,Carcinoma, Hepatocellular ,Cell Survival ,Pyridones ,Administration, Oral ,Antineoplastic Agents ,Pharmacology ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Carcinoma ,Animals ,Humans ,Viability assay ,Cell Proliferation ,Cell growth ,Chemistry ,Liver Neoplasms ,Gene Amplification ,Cancer ,Proto-Oncogene Proteins c-met ,Triazoles ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Hepatocyte growth factor ,Signal Transduction ,medicine.drug - Abstract
Aberrant hepatocyte growth factor (HGF)/MET signaling has been implicated in hepatocarcinogenesis, suggesting that MET may serve as an attractive therapeutic target in hepatocellular carcinoma. We sought to investigate the in vitro and in vivo antitumor activity of AMG 337, a potent and highly selective small molecule MET kinase inhibitor, in preclinical models of hepatocellular carcinoma. The antiproliferative activity of AMG 337 was evaluated across a panel of hepatocellular carcinoma cell lines in a viability assay. Daily oral administration was used to evaluate the in vivo antitumor activity of AMG 337 in two patient-derived xenograft (PDX) models of hepatocellular carcinoma (LI0612 and LI1078). AMG 337 exerted potent antiproliferative activity against 2 of 40 hepatocellular carcinoma cell lines, namely, MHCC97H (IC50, 0.015 μmol/L) and HCCLM3 (IC50, 0.025 μmol/L). Both sensitive cell lines showed MET amplification (MET/CEN-7 >2.0) assessed by FISH, and high MET expression (3+ IHC) assessed by IHC. AMG 337 potently inhibited p-MET in all cell lines with detectable levels of total MET. However, the dose-dependent inhibition of downstream effectors of HGF/MET signaling, including p-GAB1, p-AKT, and p-ERK, was limited to those cell lines sensitive to AMG 337 in a viability assay (MHCC97H and HCCLM3). AMG 337 significantly inhibited tumor growth at all doses tested in the MET-amplified and MET-high–expressing hepatocellular carcinoma PDX model LI0612 and had no effect on tumor growth in the non-MET–amplified and MET-low–expressing hepatocellular carcinoma PDX model LI1078. AMG 337 represents a promising and novel therapeutic strategy for targeting hepatocellular carcinomas with a dependence on HGF/MET signaling. Mol Cancer Ther; 15(6); 1227–37. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
41. Abstract 4558: Antitumor activity of AMG757, a half-life extended (HLE) bispecific T-cell engager (BiTE®) immune therapy targeting DLL3, in human PDX and orthotopic mouse models of small cell lung cancer (SCLC)
- Author
-
Juan Estrada, Jinghui Zhan, Keegan Cooke, Julie M. Bailis, Jonathan Werner, Jude Canon, Sean Caenepeel, Angela Coxon, M. Giffin, and Paul E. Hughes
- Subjects
Cancer Research ,medicine.diagnostic_test ,biology ,business.industry ,T cell ,CD3 ,medicine.disease ,Flow cytometry ,Metastasis ,medicine.anatomical_structure ,Oncology ,Antigen ,Cancer cell ,medicine ,biology.protein ,Cancer research ,IL-2 receptor ,business ,CD8 - Abstract
SCLC is a highly aggressive neuroendocrine tumor with poor prognosis and limited therapeutic options. Delta-like ligand 3 (DLL3) is a tumor-associated antigen that is highly specific for SCLC; expression is elevated in tumors but minimal and mainly cytoplasmic in normal tissues. AMG 757 is an HLE BiTE® immune therapy that is designed to redirect T-cell cytotoxicity to cancer cells by binding to DLL3 on the surface of cancer cells and CD3 on T cells. AMG 757 is being evaluated in a phase 1 clinical trial in patients with relapsed/refractory SCLC (NCT03319940). We evaluated the efficacy and pharmacodynamic (PD) effects of AMG 757 in three preclinical models of SCLC that express DLL3. We used the LXFS 1129 patient-derived xenograft (PDX) model to assess AMG 757 efficacy in established, subcutaneously implanted tumors similar to human SCLC tumors with their associated stroma. We also developed two orthotopic models of SCLC (SHP-77 and H82) to assess AMG 757 efficacy and PD at biologically relevant sites. In the SHP-77 orthotopic model, cells injected intravenously trafficked to the lungs where they formed diffuse tumors similar to primary SCLC tumors. In the H82 orthotopic model, cells injected intravenously formed discrete metastatic lesions in the liver, mimicking a major site for SCLC metastasis. In each model, mice bearing established tumors received a single infusion of human T cells and were then dosed with AMG 757 or a control HLE BiTE molecule. In the LXFS 1129 PDX model, treatment with AMG 757 induced complete tumor regression in 8 out of 10 mice, whereas treatment with the control HLE BiTE molecule showed no tumor growth inhibition. In the orthotopic SHP-77 model, treatment with AMG 757 led to significant tumor growth inhibition in the lungs relative to treatment with a control HLE BiTE molecule; the bioluminescence (BLI) signal decreased to near the limit of detection 22 days after start of treatment. AMG 757 antitumor activity was associated with increased T-cell trafficking and expansion in tumors; a single dose of AMG 757 significantly increased the number of human CD4+ and CD8+ cells in lungs as assessed by flow cytometry. In the orthotopic H82 model, AMG 757 treatment completely cleared visible lesions from the liver, as assessed by a decrease in BLI to near-background levels and macroscopic analysis of the liver tissue. AMG 757 antitumor activity was associated with increased CD8+ T-cell infiltration as assessed by flow cytometry and IHC and upregulation of the T cell surface activation markers CD25, CD69, PD-1, and 4-1BB as assessed by flow cytometry. Together, these preclinical data demonstrate that AMG 757 can recruit and engage T cells to tumors, consistent with the BiTE® mechanism of action, and that AMG 757-mediated redirected T-cell lysis can drive significant antitumor activity in established SCLC tumor models. Citation Format: Keegan Cooke, Juan Estrada, Jinghui Zhan, Jonathan Werner, Sean Caenepeel, Mike Giffin, Julie M. Bailis, Angela Coxon, Paul E. Hughes, Jude Canon. Antitumor activity of AMG757, a half-life extended (HLE) bispecific T-cell engager (BiTE®) immune therapy targeting DLL3, in human PDX and orthotopic mouse models of small cell lung cancer (SCLC) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4558.
- Published
- 2020
- Full Text
- View/download PDF
42. Abstract 6218: Discovery and preclinical evaluation of AMG 397, a potent, selective and orally bioavailable MCL1 inhibitor
- Author
-
Xiaoyue Chen, Paul E. Hughes, Alla Verlinsky, Jan Wahlstrom, Yajing Yang, Angela Coxon, Jennifer R. Allen, Jude Canon, Sean Caenepeel, Hong Tan, Kexue Li, Brian Belmontes, and Rex Karen
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Venetoclax ,Pharmacology ,Bioavailability ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,chemistry ,Oral administration ,Apoptosis ,In vivo ,030220 oncology & carcinogenesis ,Potency ,Medicine ,business ,IC50 - Abstract
MCL1 inhibitors are an emerging class of therapeutics targeting key protein-protein interactions between MCL1 and BH3 domain containing pro-apoptotic BCL-2 family members. Several MCL1 inhibitors have entered clinical trials including AMG 176, currently in Phase I clinical development for hematologic malignancies. However, all existing clinical stage MCL1 inhibitors are being administered intravenously. Here we describe the discovery and preclinical evaluation of AMG 397, the first orally administered MCL1 inhibitor in clinical development. The pursuit of an orally bioavailable MCL1 inhibitor with an acceptable human clinical dose required improvements to AMG 176 in both potency and pharmacokinetic properties. The chemistry effort to identify AMG 397 began with the observation that there were two conformations in AMG 176 bound to MCL1 with a difference of 2.8 kcal/mol in energy. A structure-guided approach in combination with ligand-based design to attempt to favor the more active conformation provided the advanced lead AM-3106 (Ki=100 pM) with significantly improved cell based potency (OPM2 viability IC50 = 19 nM) compared to AMG 176. Further refinements in potency and PK properties of AM-3106 culminated in the discovery of AMG 397. AMG 397 exhibited picomolar affinities for MCL1 (Ki=15 pM), selectively competing for binding to the BH3-binding groove of MCL1 with pro-apoptotic BCL-2 family members (e.g. BIM). In cells, AMG 397 potently disrupted the interaction between MCL1 and BIM. Treatment of OPM2 cells, an MCL1 dependent multiple myeloma cell line, induced clear increases in Caspase-3/7 activity within one hour. Subsequent effects on viability were observed across a treatment time course with maximal effects observed following 24 hours of continuous exposure (IC50=50 nM). Time course washout viability studies demonstrated that AMG 397 induced a committed step towards apoptosis and suggested that discontinuous dosing schedules would be sufficient to achieve antitumor effects in vivo. Cell lines from hematologic malignancies including AML, multiple myeloma and DLBCL exhibited greatest sensitivity to AMG 397 in a large tumor cell line profiling screen (n=300). In vivo, oral administration of AMG 397 demonstrated rapid and sustained dose dependent increases in activated BAK, cleaved Caspase-3 and cleaved PARP in established OPM2 xenografts. Discontinuous dosing schedules of once or twice weekly at 25 or 50 mg/kg of AMG 397 exhibited significant tumor regressions in mice bearing OPM2 xenografts, with 9 of 10 mice tumor free at the end of the study in both 50 mg/kg groups. AMG 397 was also tested in the MOLM-13 orthotopic model of AML where twice weekly administration at 10, 30 and 60 mg/kg achieved 47% tumor growth inhibition (TGI), 99% TGI and 75% regression respectively. Combination of AMG 397 at 10 mg/kg twice weekly and 50 mg/kg of venetoclax daily achieved 45% regression in the same model. Citation Format: Sean Caenepeel, Rex Karen, Brian Belmontes, Alla Verlinsky, Hong Tan, Yajing Yang, Xiaoyue Chen, Kexue Li, Jennifer Allen, Jan Wahlstrom, Jude Canon, Angela Coxon, Paul Hughes. Discovery and preclinical evaluation of AMG 397, a potent, selective and orally bioavailable MCL1 inhibitor [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6218.
- Published
- 2020
- Full Text
- View/download PDF
43. AMG 176, a Selective MCL1 Inhibitor, Is Effective in Hematologic Cancer Models Alone and in Combination with Established Therapies
- Author
-
Jia-Nan Gong, Angela Coxon, Sean Caenepeel, Cyril H. Benes, Andrew W. Roberts, Tao Osgood, Donia M Moujalled, Elaina Cajulis, Andrew H. Wei, Regina K. Egan, Mario G. Cardozo, Pedro J. Beltran, Liusheng Zhu, Marc Vimolratana, Sean P. Brown, Brian Lucas, Xin Huang, Gordon Moody, Paul E. Hughes, David C.S. Huang, Giovanna Pomilio, Joshua Taygerly, Jan Sun, Danny Chui, Leszek Poppe, Jude Canon, Douglas A. Whittington, Yunxiao Li, Manuel Zancanella, Nick A. Paras, Joseph McClanaghan, Xianghong Wang, Alan C. Cheng, Kathleen S. Keegan, Jonathan B. Houze, Leah J. Damon, Patricia Greninger, and Brian Belmontes
- Subjects
0301 basic medicine ,Apoptosis Inhibitor ,Venetoclax ,business.industry ,Cancer ,Myeloid leukemia ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,Apoptosis ,Cell culture ,Pharmacodynamics ,Cancer research ,medicine ,MCL1 ,business - Abstract
The prosurvival BCL2 family member MCL1 is frequently dysregulated in cancer. To overcome the significant challenges associated with inhibition of MCL1 protein–protein interactions, we rigorously applied small-molecule conformational restriction, which culminated in the discovery of AMG 176, the first selective MCL1 inhibitor to be studied in humans. We demonstrate that MCL1 inhibition induces a rapid and committed step toward apoptosis in subsets of hematologic cancer cell lines, tumor xenograft models, and primary patient samples. With the use of a human MCL1 knock-in mouse, we demonstrate that MCL1 inhibition at active doses of AMG 176 is tolerated and correlates with clear pharmacodynamic effects, demonstrated by reductions in B cells, monocytes, and neutrophils. Furthermore, the combination of AMG 176 and venetoclax is synergistic in acute myeloid leukemia (AML) tumor models and in primary patient samples at tolerated doses. These results highlight the therapeutic promise of AMG 176 and the potential for combinations with other BH3 mimetics. Significance: AMG 176 is a potent, selective, and orally bioavailable MCL1 inhibitor that induces a rapid commitment to apoptosis in models of hematologic malignancies. The synergistic combination of AMG 176 and venetoclax demonstrates robust activity in models of AML at tolerated doses, highlighting the promise of BH3-mimetic combinations in hematologic cancers. See related commentary by Leber et al., p. 1511. This article is highlighted in the In This Issue feature, p. 1494
- Published
- 2018
44. Selective MET Kinase Inhibition in MET-Dependent Glioma Models Alters Gene Expression and Induces Tumor Plasticity
- Author
-
Isabelle Dussault, Arend Heerschap, Angela Coxon, Houshang Amiri, William P.J. Leenders, Tessa J. J. de Bitter, Anna C. Navis, Wiljan Hendriks, Paul N. Span, Pieter Wesseling, Corina N. A. M. van den Heuvel, Karen Rex, Kiek Verrijp, and Sean Caenepeel
- Subjects
0301 basic medicine ,Cancer Research ,Cabozantinib ,Cell Survival ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Aminopyridines ,Biology ,Receptor tyrosine kinase ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Paracrine signalling ,Mice ,0302 clinical medicine ,Growth factor receptor ,In vivo ,Glioma ,Cell Line, Tumor ,Urological cancers Radboud Institute for Molecular Life Sciences [Radboudumc 15] ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,medicine ,Animals ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,Cell Proliferation ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,Brain Neoplasms ,Sequence Analysis, RNA ,Gene Expression Profiling ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Phosphorylation ,Pyrazoles ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,HT29 Cells - Abstract
Contains fulltext : 181795.pdf (Publisher’s version ) (Closed access) The receptor tyrosine kinase (RTK) MET represents a promising tumor target in a subset of glioblastomas. Most RTK inhibitors available in the clinic today, including those inhibiting MET, affect multiple targets simultaneously. Previously, it was demonstrated that treatment with cabozantinib (MET/VEGFR2/RET inhibitor) prolonged survival of mice carrying orthotopic patient-derived xenografts (PDX) of the MET-addicted glioblastoma model E98, yet did not prevent development of recurrent and cabozantinib-resistant tumors. To exclude VEGFR2 inhibition-inflicted blood-brain barrier normalization and diminished tumor distribution of the drug, we have now investigated the effects of the novel MET-selective inhibitor Compound A in the orthotopic E98 xenograft model. In vitro, Compound A proved a highly potent inhibitor of proliferation of MET-addicted cell lines. In line with its target selectivity, Compound A did not restore the leaky blood-brain barrier and was more effective than cabozantinib in inhibiting MET phosphorylation in vivo Compound A treatment significantly prolonged survival of mice carrying E98 tumor xenografts, but did not prevent eventual progression. Contrasting in vitro results, the Compound A-treated xenografts displayed high levels of AKT phosphorylation despite the absence of phosphorylated MET. Profiling by RNA sequencing showed that in vivo transcriptomes differed significantly from those in control xenografts.Implications: Collectively, these findings demonstrate the plasticity of paracrine growth factor receptor signaling in vivo and urge for prudency with in vitro drug-testing strategies to validate monotherapies. Mol Cancer Res; 15(11); 1587-97. (c)2017 AACR.
- Published
- 2017
45. MAPK pathway inhibition induces MET and GAB1 levels, priming BRAF mutant melanoma for rescue by hepatocyte growth factor
- Author
-
Keegan Cooke, Blanca Homet Moreno, Sean Caenepeel, Sarah Wadsworth, Giulia Parisi, Lidia Robert, Angela Coxon, Antoni Ribas, Richard Kendall, Paul E. Hughes, Elaina Cajulis, Guo Huang, and Pedro J. Beltran
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,MAPK/ERK pathway ,Indoles ,medicine.medical_treatment ,Nude ,Drug Resistance ,GAB1 ,Apoptosis ,Targeted therapy ,Mice ,0302 clinical medicine ,HGF ,Cancer ,Sulfonamides ,Tumor ,Hepatocyte Growth Factor ,Melanoma ,Adaptor Proteins ,Dipeptides ,Proto-Oncogene Proteins c-met ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Benzamides ,MET ,Hepatocyte growth factor ,Female ,Development of treatments and therapeutic interventions ,Tyrosine kinase ,Research Paper ,medicine.drug ,Proto-Oncogene Proteins B-raf ,Combination therapy ,Pyridones ,MAP Kinase Signaling System ,Oncology and Carcinogenesis ,Mice, Nude ,Antineoplastic Agents ,Cell Line ,BRAF ,resistance ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,melanoma ,Animals ,Humans ,neoplasms ,Protein Kinase Inhibitors ,Adaptor Proteins, Signal Transducing ,business.industry ,Diphenylamine ,Signal Transducing ,Triazoles ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Vemurafenib ,Drug Resistance, Neoplasm ,Immunology ,Cancer research ,Neoplasm ,business - Abstract
Therapeutic resistance is a major obstacle to achieving durable clinical responses with targeted therapies, highlighting a need to elucidate the underlying mechanisms responsible for resistance and identify strategies to overcome this challenge. An emerging body of data implicates the tyrosine kinase MET in mediating resistance to BRAF inhibitors in BRAFV600E mutant melanoma. In this study we observed a dominant role for the HGF/MET axis in mediating resistance to BRAF and MEK inhibitors in models of BRAFV600E and NRAS mutant melanoma. In addition, we showed that MAPK pathway inhibition induced rapid increases in MET and GAB1 levels, providing novel mechanistic insight into how BRAFV600E mutant melanoma is primed for HGF-mediated rescue. We also determined that tumor-derived HGF, not systemic HGF, may be required to convey resistance to BRAF inhibition in vivo and that resistance could be reversed following treatment with AMG 337, a selective MET inhibitor. In summary, these findings support the clinical evaluation of MET-directed targeted therapy to circumvent resistance to BRAF and MEK inhibitors in BRAFV600E mutant melanoma. In addition, the induction of MET following treatment with BRAF and MEK inhibitors has the potential to serve as a predictive biomarker for identifying patients best suited for MET inhibitor combination therapy.
- Published
- 2017
46. MDM2 antagonists synergize broadly and robustly with compounds targeting fundamental oncogenic signaling pathways
- Author
-
Tao Osgood, Julie A. Lofgren, Marc Payton, Rebecca Robertson, Sean Caenepeel, Dongyin Yu, Jude Canon, Cheng Su, Adrie Jones, Jebediah Ledell, Chetan Deshpande, Jonathan D. Oliner, Xiaoning Zhao, Paul E. Hughes, and Anne Y. Saiki
- Subjects
Drug ,MAPK/ERK pathway ,Cell Survival ,media_common.quotation_subject ,Gene Expression ,synergy ,Apoptosis ,Drug resistance ,Biology ,PI3K ,MDM2 ,Cell Line, Tumor ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Viability assay ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,media_common ,FOXM1 ,Rational design ,Drug Synergism ,Proto-Oncogene Proteins c-mdm2 ,MAPK ,MEK ,Oncology ,Targeted drug delivery ,biology.protein ,Cancer research ,Mdm2 ,Drug Screening Assays, Antitumor ,Signal Transduction ,Priority Research Paper - Abstract
While MDM2 inhibitors hold great promise as cancer therapeutics, drug resistance will likely limit their efficacy as single agents. To identify drug combinations that might circumvent resistance, we screened for agents that could synergize with MDM2 inhibition in the suppression of cell viability. We observed broad and robust synergy when combining MDM2 antagonists with either MEK or PI3K inhibitors. Synergy was not limited to cell lines harboring MAPK or PI3K pathway mutations, nor did it depend on which node of the PI3K axis was targeted. MDM2 inhibitors also synergized strongly with BH3 mimetics, BCR-ABL antagonists, and HDAC inhibitors. MDM2 inhibitor-mediated synergy with agents targeting these mechanisms was much more prevalent than previously appreciated, implying that clinical translation of these combinations could have far-reaching implications for public health. These findings highlight the importance of combinatorial drug targeting and provide a framework for the rational design of MDM2 inhibitor clinical trials.
- Published
- 2014
- Full Text
- View/download PDF
47. Inhibition of Mcl-1 Enhances the Efficacy of Tyrosine Kinase Inhibition in FLT3 Mutated AML and Synergizes with Venetoclax Targeting AML Stem Cells
- Author
-
Phuong Khanh Morrow, Po Yee Mak, Qi Zhang, Jude Canon, Michael Andreeff, Xiangmeng Wang, Marina Konopleva, Vivian Ruvolo, Bing Z. Carter, Wenjing Tao, Sean Caenepeel, Vinitha Mary Kuruvilla, Venkata Lokesh Battula, Paul E. Hughes, and Carli M. Stewart
- Subjects
Sorafenib ,Neuroblastoma RAS viral oncogene homolog ,Oncology ,medicine.medical_specialty ,Venetoclax ,business.industry ,Immunology ,Decitabine ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,Leukemia ,chemistry ,hemic and lymphatic diseases ,Internal medicine ,Ven ,medicine ,Stem cell ,Progenitor cell ,business ,medicine.drug - Abstract
Bcl-2 and Mcl-1 play critical roles in AML stem/progenitor cell survival. Venetoclax (VEN), a highly selective Bcl-2 inhibitor, showed limited clinical efficacy in AML as a single agent. FLT3 is the most frequently mutated gene in AML, resulting in constitutive activation of FLT3 tyrosine kinase and its downstream signaling pathways, which can be targeted by FLT3 tyrosine kinase inhibitors (TKIs). However, patients can adapt to TKI treatment by reactivating the MEK signaling pathway (Bruner JK et al., Cancer Res 2017), which is known to stabilize Mcl-1 levels. Furthermore, deregulated Mcl-1 expression was identified as a novel mechanism of primary TKI resistance in a subset of FLT3-ITD mutated AML patients (Breitenbuecher F et al., Blood 2009). Importantly, Mcl-1 can be induced by VEN treatment and could be a major resistance factor to VEN (Pan R et al., Cancer Discover 2014; Carter BZ et al., ASH 2018). Hence, Mcl-1 inhibition may enhance the efficacy of TKIs in FLT3 mutated AML and synergize with VEN, targeting AML cells and stem/progenitor cells. We treated FLT3-ITD positive AML cells with a selective inhibitor of Mcl-1 (AMG 176) and FLT3 TKIs and found that inhibition of Mcl-1 induced cell death and significantly enhanced the activity of sorafenib or gilteritinib in cell lines including cells acquired resistance to VEN (CI We previously showed that overexpression/knockdown of Mcl-1 greatly protected/sensitized AML cells from VEN induced cell death (Carter BZ, ASH 2018) supporting Mcl-1 as a key VEN resistance factor. We treated primary AML cells (n=5) with VEN (10 nM) or AMG 176 (250 nM) alone, or in combination and found that VEN+AMG 176 synergistically induced cell death in AML blasts and AML stem/progenitor cells even in samples clinically resistant to or relapsed after VEN containing regimen (CI To investigate the antileukemia activity in vivo, we tested combined inhibition of Mcl-1 and Bcl-2 using two PDX models in NSG mice. The first model was developed from a resistant/relapsed patient with FLT3-ITD mutation and complex karyotype. The combination showed the most significant antileukemic activity and extension of survival, followed by AMG 176 and VEN treatment alone (median survival for the combination, 146 d, p=0.004; AMG 176, 137 d, p=0.032; VEN, 102 d, p>0.05 vs. control, 85.5 d; respectively). The second PDX model was developed from a patient who first responded and then became resistant to the combination of VEN and decitabine and harbors FLT3-ITD, NRAS, and GATA2 mutations and complex karyotype. VEN or AMG 176 monotherapies marginally prolonged survival (median survival 127 or 129 vs. control 124 d). The combination was highly effective in this model and greatly decreased circulating blasts (Fig. 1) and leukemia tissue infiltration, measured by flow cytometry and spleen size. CyTOF analysis demonstrated that only the combination strongly reduced blasts as well as the AML stem/progenitor cell populations. Median survival for the combination group currently has not been reached (>325 d) (Fig. 2). Collectively, these data demonstrate that inhibition of Mcl-1 enhances the efficacy of TKIs in FLT3 mutated AML. Furthermore, it synergizes with VEN, targeting not only AML blasts but also AML stem/progenitor cells, both in vitro and in vivo in PDX models with the potential of significantly improving treatment outcome, which warrants clinical evaluation. Disclosures Carter: Amgen: Research Funding; AstraZeneca: Research Funding; Ascentage: Research Funding. Zhang:The University of Texas M.D.Anderson Cancer Center: Employment. Kuruvilla:The University of Texas M.D.Anderson Cancer Center: Employment. Konopleva:Kisoji: Consultancy, Honoraria; Eli Lilly: Research Funding; Forty-Seven: Consultancy, Honoraria; Calithera: Research Funding; Stemline Therapeutics: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria, Research Funding; Cellectis: Research Funding; Reata Pharmaceuticals: Equity Ownership, Patents & Royalties; Amgen: Consultancy, Honoraria; F. Hoffman La-Roche: Consultancy, Honoraria, Research Funding; Genentech: Honoraria, Research Funding; Ascentage: Research Funding; Ablynx: Research Funding; Agios: Research Funding; Astra Zeneca: Research Funding. Caenepeel:Amgen Inc.: Employment. Canon:Amgen Inc.: Employment. Hughes:Amgen Inc.: Employment. Morrow:Amgen Inc.: Employment. Andreeff:Daiichi Sankyo, Inc.: Consultancy, Patents & Royalties: Patents licensed, royalty bearing, Research Funding; Celgene: Consultancy; Jazz Pharmaceuticals: Consultancy; Amgen: Consultancy; AstaZeneca: Consultancy; 6 Dimensions Capital: Consultancy; Reata: Equity Ownership; Aptose: Equity Ownership; Eutropics: Equity Ownership; Senti Bio: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Oncoceutics: Equity Ownership; Oncolyze: Equity Ownership; Breast Cancer Research Foundation: Research Funding; CPRIT: Research Funding; NIH/NCI: Research Funding; Center for Drug Research & Development: Membership on an entity's Board of Directors or advisory committees; Cancer UK: Membership on an entity's Board of Directors or advisory committees; NCI-CTEP: Membership on an entity's Board of Directors or advisory committees; German Research Council: Membership on an entity's Board of Directors or advisory committees; Leukemia Lymphoma Society: Membership on an entity's Board of Directors or advisory committees; NCI-RDCRN (Rare Disease Cliln Network): Membership on an entity's Board of Directors or advisory committees; CLL Foundation: Membership on an entity's Board of Directors or advisory committees; BiolineRx: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2019
- Full Text
- View/download PDF
48. Abstract 2180: AMG 176 exhibits robust antitumor activity in combination with standard of care agents in models of acute myeloid leukemia
- Author
-
Sean Caenepeel, Paul E. Hughes, Elaina Cajulis, Angela Coxon, Brian Belmontes, Tao Osgood, and Jude Canon
- Subjects
Cancer Research ,Anthracycline ,business.industry ,Myeloid leukemia ,Decitabine ,Phases of clinical research ,Oncology ,Hypomethylating agent ,medicine ,Cytarabine ,Cancer research ,MCL1 ,Doxorubicin ,business ,medicine.drug - Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy involving the clonal expansion of abnormal myeloid progenitor cells. The anti-apoptotic BCL-2 protein family member MCL1 has been implicated in AML pathogenesis. AMG 176 is a potent and selective MCL1 inhibitor currently being evaluated in an AML Phase I clinical trial. Standard of care (SOC) treatment for AML involves “7+3” induction therapy with cytarabine and an anthracycline (i.e. doxorubicin). In patient’s ineligible for conventional induction therapy, the hypomethylating agent decitabine is a well-tolerated alternative. Here we investigate the activity of AMG 176 and AM-8621, a closely related analog of AMG 176 with similar potency and selectivity, in combination with SOC therapies in preclinical models of AML. We profiled a panel of AML cell lines (MOLM-13, MV-4-11, GDM-1 and EOL-1), testing AM-8621 in combination with cytarabine, doxorubicin and decitabine in 3-day viability assays. Evidence for synergistic activity was detected with each of the combinations across subsets of the cell lines. A time course of AM-8621 treatment revealed a rapid induction of caspase 3/7 activity, with significant increases observed within the first 2-4 hours of treatment while longer duration treatment (>8 hours) was required to activate caspase 3/7 with SOC agents. Combined treatment with AM-8621 and SOC agents exhibited significant improvements in caspase 3/7 activity beyond either single agent alone. To provide mechanistic insight into the ability of SOC agents to sensitize AML cell lines to AM-8621 treatment, signaling experiments were performed to characterize changes in BCL-2 protein family members following treatment with SOC agents. Clear changes in the levels of anti-apoptotic BCL-2 family members (MCL1, BCL-2 and BCL-XL), BH3-only domain containing proteins (BIM, NOXA, PUMA) and the executioner protein BAK were observed, providing insight into potential mechanisms of priming cells to MCL1 inhibition. Combinations of AMG 176 and each of the SOC therapies were also tested in a MOLM-13 orthotopic xenograft model of AML. Mice were treated with combinations of AMG 176 and cytarabine, decitabine or doxorubicin and compared against the single agent therapies. Significant reductions in tumor burden compared to single agent treatments were observed with the AMG 176 + doxorubicin (30% regression) and AMG 176 + decitabine (98% tumor growth inhibition) combinations. These data highlight the promise of combining AMG 176 with SOC therapies in the setting of AML. Citation Format: Sean Caenepeel, Brian Belmontes, Tao Osgood, Elaina Cajulis, Angela Coxon, Jude Canon, Paul E. Hughes. AMG 176 exhibits robust antitumor activity in combination with standard of care agents in models of acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2180.
- Published
- 2019
- Full Text
- View/download PDF
49. The Role of MDM2 Amplification and Overexpression in Tumorigenesis
- Author
-
Jonathan D. Oliner, Anne Y. Saiki, and Sean Caenepeel
- Subjects
0301 basic medicine ,Single-nucleotide polymorphism ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,law ,Gene duplication ,medicine ,SNP ,Animals ,Humans ,neoplasms ,Mice, Knockout ,Gene Amplification ,Proto-Oncogene Proteins c-mdm2 ,medicine.disease ,030104 developmental biology ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Mdm2 ,Suppressor ,Sarcoma ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Nuclear localization sequence ,Perspectives - Abstract
Mouse double minute 2 (MDM2) is a critical negative regulator of the tumor suppressor p53, playing a key role in controlling its transcriptional activity, protein stability, and nuclear localization. MDM2 expression is up-regulated in numerous cancers, resulting in a loss of p53-dependent activities, such as apoptosis and cell-cycle arrest. Genetic amplification and inheritance of MDM2 promoter single-nucleotide polymorphisms (SNPs) are the two best-studied mechanisms for up-regulating MDM2 activity. This article provides an overview of these events in human cancer, highlighting the frequent occurrence of MDM2 amplification in sarcoma and the role of SNP309 and SNP285 in regulating MDM2 expression and cancer risk. The availability of large-scale genomic profiling datasets, like those from The Cancer Genome Atlas Research Network, have provided the opportunity to evaluate the consequences of MDM2 amplification and SNP inheritance across high-quality tumor samples from diverse cancer indications.
- Published
- 2016
50. Synthesis and structure–activity relationships of dual PI3K/mTOR inhibitors based on a 4-amino-6-methyl-1,3,5-triazine sulfonamide scaffold
- Author
-
Yunxin Bo, Tisha San Miguel, Nobuko Nishimura, Jian Jiang, Douglas A. Whittington, Paul E. Hughes, Sean Caenepeel, Mark H. Norman, John D. McCarter, Erin L. Mullady, Daniel J. Freeman, Kevin Yang, Ryan Wurz, Longbin Liu, Raju Subramanian, Kristin L. Andrews, Ling Wang, Liping H. Pettus, and Nancy Zhang
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Phosphorylation ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Sulfonamides ,Binding Sites ,Phosphoinositide 3-kinase ,biology ,Triazines ,Akt/PKB signaling pathway ,Chemistry ,TOR Serine-Threonine Kinases ,Cellular Assay ,Organic Chemistry ,Rats ,Molecular Docking Simulation ,biology.protein ,Molecular Medicine ,Female ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Phosphoinositide 3-kinase (PI3K) is an important target in oncology due to the deregulation of the PI3K/Akt signaling pathway in a wide variety of tumors. A series of 4-amino-6-methyl-1,3,5-triazine sulfonamides were synthesized and evaluated as inhibitors of PI3K. The synthesis, in vitro biological activities, pharmacokinetic and in vivo pharmacodynamic profiling of these compounds are described. The most promising compound from this investigation (compound 3j) was found to be a pan class I PI3K inhibitor with a moderate (>10-fold) selectivity over the mammalian target of rapamycin (mTOR) in the enzyme assay. In a U87 MG cellular assay measuring phosphorylation of Akt, compound 3j displayed low double digit nanomolar IC(50) and exhibited good oral bioavailability in rats (F(oral)=63%). Compound 3j also showed a dose dependent reduction in the phosphorylation of Akt in a U87 tumor pharmacodynamic model with a plasma EC(50)=193 nM (91 ng/mL).
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.