33 results on '"Jodi Moriguchi"'
Search Results
2. 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
3. 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
4. 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
5. 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
6. 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
7. Supplementary Figure 2 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Isabelle Dussault, Teresa L. Burgess, April Chen, Jasmine Lin, Tae-Seong Kim, Monica Reese, Angela Coxon, Bethany Mattson, Tao Osgood, Jodi Moriguchi, Yajing Yang, Karen Rex, Paula J. Kaplan-Lefko, and Yihong Zhang
- Abstract
Supplementary Figure 2 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 1 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Isabelle Dussault, Teresa L. Burgess, April Chen, Jasmine Lin, Tae-Seong Kim, Monica Reese, Angela Coxon, Bethany Mattson, Tao Osgood, Jodi Moriguchi, Yajing Yang, Karen Rex, Paula J. Kaplan-Lefko, and Yihong Zhang
- Abstract
Supplementary Figure 1 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Isabelle Dussault, Teresa L. Burgess, April Chen, Jasmine Lin, Tae-Seong Kim, Monica Reese, Angela Coxon, Bethany Mattson, Tao Osgood, Jodi Moriguchi, Yajing Yang, Karen Rex, Paula J. Kaplan-Lefko, and Yihong Zhang
- Abstract
Recepteur d'origine nantais (RON) is a receptor tyrosine kinase closely related to c-Met. Both receptors are involved in cell proliferation, migration, and invasion, and there is evidence that both are deregulated in cancer. Receptor overexpression has been most frequently described, but other mechanisms can lead to the oncogenic activation of RON and c-Met. They include activating mutations or gene amplification for c-Met and constitutively active splicing variants for RON. We identified a novel inhibitor of RON and c-Met, compound I, and characterized its in vitro and in vivo activities. Compound I selectively and potently inhibited the kinase activity of RON and c-Met with IC50s of 9 and 4 nmol/L, respectively. Compound I inhibited hepatocyte growth factor–mediated and macrophage-stimulating protein–mediated signaling and cell migration in a dose-dependent manner. Compound I was tested in vivo in xenograft models that either were dependent on c-Met or expressed a constitutively active form of RON (RONΔ160 in HT-29). Compound I caused complete tumor growth inhibition in NIH3T3 TPR-Met and U-87 MG xenografts but showed only partial inhibition in HT-29 xenografts. The effect of compound I in HT-29 xenografts is consistent with the expression of the activating b-Raf V600E mutation, which activates the mitogen-activated protein kinase pathway downstream of RON. Importantly, tumor growth inhibition correlated with the inhibition of c-Met–dependent and RON-dependent signaling in tumors. Taken together, our results suggest that a small-molecule dual inhibitor of RON/c-Met has the potential to inhibit tumor growth and could therefore be useful for the treatment of patients with cancers where RON and/or c-Met are activated. [Cancer Res 2008;68(16):6680–7]
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure Legends 1-3 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Isabelle Dussault, Teresa L. Burgess, April Chen, Jasmine Lin, Tae-Seong Kim, Monica Reese, Angela Coxon, Bethany Mattson, Tao Osgood, Jodi Moriguchi, Yajing Yang, Karen Rex, Paula J. Kaplan-Lefko, and Yihong Zhang
- Abstract
Supplementary Figure Legends 1-3 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 3 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Isabelle Dussault, Teresa L. Burgess, April Chen, Jasmine Lin, Tae-Seong Kim, Monica Reese, Angela Coxon, Bethany Mattson, Tao Osgood, Jodi Moriguchi, Yajing Yang, Karen Rex, Paula J. Kaplan-Lefko, and Yihong Zhang
- Abstract
Supplementary Figure 3 from Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Published
- 2023
- Full Text
- View/download PDF
12. Targeting the Mitotic Kinesin KIF18A in Chromosomally Unstable Cancers: Hit Optimization Toward an In Vivo Chemical Probe
- Author
-
Nuria A. Tamayo, Matthew P. Bourbeau, Jennifer R. Allen, Kate S. Ashton, Jian Jeffrey Chen, Matthew R. Kaller, Thomas T. Nguyen, Nobuko Nishimura, Liping H. Pettus, Mary Walton, Brian Belmontes, Jodi Moriguchi, Kui Chen, John D. McCarter, Kelly Hanestad, Grace Chung, Maria Stefania S. Ninniri, Jan Sun, Leszek Poppe, Chris Spahr, John Hui, Lei Jia, Tian Wu, Upendra P. Dahal, Katheryne Z. Edson, and Marc Payton
- Subjects
Cell Death ,Tubulin ,Neoplasms ,Drug Discovery ,Molecular Medicine ,Humans ,Kinesins ,Mitosis ,Spindle Apparatus - Abstract
Chromosomal instability (CIN) is a hallmark of cancer that results from errors in chromosome segregation during mitosis. Targeting of CIN-associated vulnerabilities is an emerging therapeutic strategy in drug development. KIF18A, a mitotic kinesin, has been shown to play a role in maintaining bipolar spindle integrity and promotes viability of CIN cancer cells. To explore the potential of KIF18A, a series of inhibitors was identified. Optimization of an initial hit led to the discovery of analogues that could be used as chemical probes to interrogate the role of KIF18A inhibition. Compounds
- Published
- 2022
13. Abstract 6246: The MTA-cooperative PRMT5 inhibitor AM-9747 exhibits robust antitumor activity in combination with clinically relevant chemotherapies and targeted agents in MTAP null tumor models
- Author
-
Katherine Slemmons, Brian Belmontes, Siyuan Liu, Jodi Moriguchi, Antonia Policheni, and Paul E. Hughes
- Subjects
Cancer Research ,Oncology - Abstract
Homozygous deletion of chr9p21, containing genes CDKN2A and MTAP, occurs in about 15% of cancers. MTAP loss, a key enzyme in the methionine and adenine salvage pathways, leads to the accumulation of its substrate MTA. MTA competes with the methyl donor SAM for binding to type II arginine methyltransferase PRMT5, placing PRMT5 in a hypomorphic state and vulnerable to further PRMT5 inhibition. MTA-cooperative PRMT5 inhibitors are an emerging class of therapeutics targeting MTAP null tumors. AMG 193, currently in Phase 1 trials, and its representative analog AM-9747 have broad spectrum activity in MTAP null tumor models across hematologic and solid tumor indications. Mechanistically, AM-9747 induces DNA damage, shown by increased phosphorylated H2AX, resulting in cell cycle arrest and senescence. Exploration of clinically relevant therapeutic combinations with standard of care (SOC) chemotherapies or targeted agents that could potentiate this DNA damage or target orthogonal pathways is a rational therapeutic approach. Here, we evaluated AM-9747 in combination with SOC chemotherapies and assessed synergy using the Chou-Talalay Method to generate Combination Index (CI) scores. SOC agents representing a variety of mechanisms of action (paclitaxel, carboplatin, gemcitabine, pemetrexed, irinotecan, 5-FU) were evaluated and results ranged from strongly synergistic (CI Citation Format: Katherine Slemmons, Brian Belmontes, Siyuan Liu, Jodi Moriguchi, Antonia Policheni, Paul E. Hughes. The MTA-cooperative PRMT5 inhibitor AM-9747 exhibits robust antitumor activity in combination with clinically relevant chemotherapies and targeted agents in MTAP null tumor models. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6246.
- Published
- 2023
- Full Text
- View/download PDF
14. Abstract 516: Discovery and preclinical characterization of AMG 650, a first-in-class inhibitor of kinesin KIF18A motor protein with potent activity against chromosomally unstable cancers
- Author
-
Brian Belmontes, Jodi Moriguchi, Grace Chung, Jan Sun, Maria Stefania S. Ninniri, Kelly Hanestad, Kui Chen, John D. McCarter, Upendra P. Dahal, Sudipa Ghimire-Rijal, Yue Hao, Christopher P. Mohr, Xinchao Yu, Matthew G. Rees, Melissa Ronan, Jennifer Roth, Sheroy Minocherhomji, Jennifer R. Allen, Matthew P. Bourbeau, Paul E. Hughes, Nuria A Tamayo, and Marc N. Payton
- Subjects
Cancer Research ,Oncology - Abstract
Chromosomal instability (CIN) is a hallmark of human cancers and is caused by persistent errors in chromosome segregation during mitosis. Aggressive types of human cancer such as high-grade serous ovarian cancer and triple-negative breast cancer have elevated levels of CIN and frequently harbor TP53 gene alterations and are poorly served by current treatment options. These two CIN+ cancer types also share mutually exclusive genetic alterations in BRCA1 and CCNE1 cancer genes. KIF18A is a mitotic kinesin motor protein that localizes to the plus-end tips of kinetochore microtubule (MT) spindle fibers, where it regulates chromosome alignment during cell division. KIF18A is overexpressed in a subset of human cancers, and its elevated expression is associated with tumor aggressiveness. Recent reports provide compelling evidence that genetic ablation of KIF18A reduced the viability of CIN cancer cells (Marquis et al. Nature Com 2021, Quinton et al. Nature 2021, Cohen-Sharir et al. Nature 2021). Here, we report the discovery and preclinical characterization of AMG 650, a potent and selective inhibitor of KIF18A with a compelling anti-cancer profile distinct from other cell cycle and anti-mitotic drug targets. Structural insights gained by cryo-EM illustrate the unique binding mode of AMG 650 to an allosteric pocket at the interface of KIF18A motor α4 and α6 helices and α-tubulin. AMG 650 selectively inhibits KIF18A MT-ATPase motor activity (IC50 = 48 nM) and exhibits specificity against a panel of diverse motor proteins. In cells, AMG 650 is active at double-digit nM concentrations and phenocopies KIF18A genetic KD/KO dependencies across a panel of DNA-barcoded cancer cell lines. AMG 650 selectively activates the mitotic checkpoint resulting in multipolarity and apoptosis in breast and ovarian cancer cell lines enriched with CIN features. Notably, AMG 650 has minimal effects on proliferating human bone marrow mononuclear cells in culture at concentrations active on sensitive cancer cells (>100X window). In vivo, AMG 650 has low clearance, long half-life, and good oral bioavailability across preclinical species. In mice, oral administration of AMG 650 induces a dose-dependent pharmacodynamic response (pH3 mitotic marker) that is sustained for 24 hours in OVCAR-3 tumor model. Importantly, continuous once-daily dosing with AMG 650 shows robust anti-cancer activity with evidence of durable tumor regressions in a subset of human ovarian and breast CDX/PDX tumor models at well-tolerated doses. Lastly, the combination of AMG 650 with PARP inhibitor Olaparib enhances anti-cancer activity relative to single agent alone in BRCA1- and CCNE1-altered tumor models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via AMG 650, a first-in-class KIF18A inhibitor. Citation Format: Brian Belmontes, Jodi Moriguchi, Grace Chung, Jan Sun, Maria Stefania S. Ninniri, Kelly Hanestad, Kui Chen, John D. McCarter, Upendra P. Dahal, Sudipa Ghimire-Rijal, Yue Hao, Christopher P. Mohr, Xinchao Yu, Matthew G. Rees, Melissa Ronan, Jennifer Roth, Sheroy Minocherhomji, Jennifer R. Allen, Matthew P. Bourbeau, Paul E. Hughes, Nuria A Tamayo, Marc N. Payton. Discovery and preclinical characterization of AMG 650, a first-in-class inhibitor of kinesin KIF18A motor protein with potent activity against chromosomally unstable cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 516.
- Published
- 2023
- Full Text
- View/download PDF
15. Abstract LB202: Discovery and preclinical characterization of novel small molecule inhibitors of kinesin KIF18A motor protein with potent activity against chromosomally unstable cancers
- Author
-
Jan Sun, Brain Belmontes, Jodi Moriguchi, Grace Chung, Kui Chen, John D. McCarter, Upendra P. Dahal, Andrew S. Boghossian, Matthew G. Rees, Melissa M. Ronan, Jennifer A. Roth, Sheroy Minocherhomji, Matthew P. Bourbeau, Jennifer R. Allen, Angela Coxon, Paul E. Hughes, Nuria Tamayo, and Marc N. Payton
- Subjects
Cancer Research ,Oncology - Abstract
KIF18A is a mitotic kinesin that localizes to the plus-end tips of kinetochore microtubule (MT) spindle fibers during metaphase, where it regulates chromosome alignment, and promotes the viability of chromosomally unstable cancer cells. KIF18A is overexpressed in a subset of human cancers, and its elevated expression is associated with tumor aggressiveness. Chromosomal instability (CIN) is a hallmark of human cancers and is caused by persistent errors in chromosome segregation during mitosis. Aggressive types of human cancer such as high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have elevated levels of CIN and frequently harbor alterations in TP53 tumor suppressor gene. These two CIN+ cancer subtypes share molecular similarities but have limited treatment options at present. The rationale of pharmacological inhibition of KIF18A motor activity is to selectively target a tumor-specific mitotic spindle vulnerability in CIN+ cancer cells while largely sparing normal diploid dividing somatic cells. Here, we describe the identification of a novel series of potent and selective small molecule inhibitors of KIF18A MT-ATPase motor activity exemplified by AM-1882, that disrupt the mitotic spindle and selectively kill chromosomally unstable cancer cells. Our KIF18A inhibitors phenocopy genetic ablation of KIF18A and trigger spindle assembly checkpoint activation, multipolarity, and apoptosis in sensitive CIN+ cancer cell lines. The sensitivity profile of AM-1882 is focal-in-nature with cell potency in the low double-digit nanomolar range across a panel of breast and ovarian cancer cell lines, including lines that harbor genetic alterations (e.g., TP53, CCNE1, RB1, BRCA1, whole genome doubling) frequently enriched in CIN+ cancers and in HGSOC and TNBC tumor subtypes. Furthermore, the sensitivity profile of AM-1882 is distinct from comparator test agents ispinesib (Eg5, pan cytotoxic) and palbociclib (CDK4/6, focal cytostatic). The combination of AM-1882 with PARP inhibitor olaparib is synergistic in BRCA1-deficient cancer cell lines, with evidence of increased double-strand DNA breaks (p-H2AX) and apoptosis (cl-PARP). Importantly, KIF18A inhibitors have minimal toxicity on normal dividing somatic cell types in vitro, including proliferating human bone marrow mononuclear cells, distinct from paclitaxel and small molecule inhibitors of essential mitotic kinases and kinesins. In vivo, we demonstrate that administration of KIF18A inhibitors AM-1882 and AM-5308 induce a robust pharmacodynamic response (pH3, mitotic marker) and frank tumor regressions in two TP53 mutant human HGSOC xenograft models (OVCAR-3, OVCAR-8) at well-tolerated doses. Collectively, our preclinical data provides the first example of a therapeutic strategy to selectively target CIN+ cancers through inhibition of KIF18A motor protein. Citation Format: Jan Sun, Brain Belmontes, Jodi Moriguchi, Grace Chung, Kui Chen, John D. McCarter, Upendra P. Dahal, Andrew S. Boghossian, Matthew G. Rees, Melissa M. Ronan, Jennifer A. Roth, Sheroy Minocherhomji, Matthew P. Bourbeau, Jennifer R. Allen, Angela Coxon, Paul E. Hughes, Nuria Tamayo, Marc N. Payton. Discovery and preclinical characterization of novel small molecule inhibitors of kinesin KIF18A motor protein with potent activity against chromosomally unstable cancers [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 LB202.
- Published
- 2022
- Full Text
- View/download PDF
16. Abstract 1807: The discovery and preclinical characterization of the MTA cooperative PRMT5 inhibitor AM-9747
- Author
-
Brian Belmontes, Antonia Policheni, Siyuan Liu, Katherine Slemmons, Jodi Moriguchi, Hayley Ma, Daniel Aiello, Yajing Yang, Mikkel Vestergaard, Sanne Cowland, Jan Anderson, Ian Sarvary, Nuria Tamayo, Liping Pettus, Susmith Mukund, Leszek Pope, Jennifer R. Allen, Sanne Glad, Matthew Bourbeau, and Paul E. Hughes
- Subjects
Cancer Research ,Oncology - Abstract
Homozygous deletion of the tumor suppressor gene CDKN2A and the neighboring MTAP gene located at chr9p21 occurs in 10-15% of human cancers. Deletion of MTAP, an enzyme in methionine and adenine salvage pathways, results in accumulation of its substrate MTA, which is structurally similar to SAM, the substrate methyl donor for the type II methyltransferase PRMT5. In MTAP deleted cells, MTA competes with SAM for binding to PRMT5, placing PRMT5 in a partially inhibited or hypormorphic state. Multiple studies using shRNAi knockdown have shown that tumor cell lines harboring MTAP deletions are vulnerable to PRMT5 inhibition. PRMT5 inhibitors that have advanced to clinical studies do not selectively target the MTA-bound form of PRMT5, and the preclinical activity of these molecules is not enriched in MTAP-deleted tumor cells lines. Moreover, the therapeutic window of these molecules is narrow, presumably due to the inhibition of PRMT5 in normal cells. We set out to identify PRMT5 inhibitors that bind cooperatively with MTA, with the goal of selectively targeting PRMT5 in MTAP-deleted tumors. A DNA encoded library screen was conducted to identify small molecules that preferentially bind to PRMT5 in the presence of MTA. The subsequent optimization of screening hits to improve potency, MTA-cooperativity, and pharmacokinetic properties led to the identification of AM-9747. The nature of the MTA cooperativity of AM-9747 was interrogated by multiple biophysical methods and structural biology experiments. Following treatment with AM-9747, the levels of SDMA marks were lower in HCT116 MTAP-deleted cells (IC50 = 0.0002 μM) compared to HCT116 MTAP-WT cells (IC50 = 0.050 μM). AM-9747 selectively inhibited the proliferation of HCT116 MTAP-deleted cells (IC50 = 0.027 μM) compared to HCT116 MTAP-WT cells (IC50 = 0.63 μM). The profiling of AM-9747 in an expanded panel of tumor cell lines demonstrated that AM-9747 inhibited the proliferation of most MTAP-deleted cells, with minimal effects on MTAP-WT cells. In vitro mechanism of action studies demonstrated that treatment with AM-9747 induces DNA damage, as illustrated by increased phosphorylation of H2AX, and an arrest in the G2/M phase of the cell cycle in MTAP-deleted cells. In vivo, oral administration of AM-9747 selectively inhibits SDMA and tumor growth in HCT116 MTAP-deleted tumor xenografts, compared to HCT116 MTAP-WT xenografts. Furthermore, treatment with AM-9747 inhibits the growth of multiple MTAP-deleted tumor xenograft models, including BXPC3 (PDAC) and DOHH2 (DLBCL). AM-9747 was profiled against a panel of over twenty PDX models, with greater than 50% tumor growth inhibition observed in the majority of PDX models harboring deletion of the MTAP gene. Our data with AM-9747 indicates that PRMT5 inhibitors that selectively target PRMT5 in cooperation with MTA may represent a novel and compelling therapeutic strategy for the treatment of MTAP-deleted cancers. Citation Format: Brian Belmontes, Antonia Policheni, Siyuan Liu, Katherine Slemmons, Jodi Moriguchi, Hayley Ma, Daniel Aiello, Yajing Yang, Mikkel Vestergaard, Sanne Cowland, Jan Anderson, Ian Sarvary, Nuria Tamayo, Liping Pettus, Susmith Mukund, Leszek Pope, Jennifer R. Allen, Sanne Glad, Matthew Bourbeau, Paul E. Hughes. The discovery and preclinical characterization of the MTA cooperative PRMT5 inhibitor AM-9747 [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 1807.
- Published
- 2022
- Full Text
- View/download PDF
17. Therapeutic Antibody Targeting Tumor- and Osteoblastic Niche-Derived Jagged1 Sensitizes Bone Metastasis to Chemotherapy
- Author
-
Guangwen Ren, Chadwick T. King, Angela Coxon, Minhong Shen, Jodi Moriguchi, Mark Esposito, Jan Sun, Klaus Pantel, Yangjin Bae, Nikolai Tupitsyn, Min Yuan, Yibin Kang, Sabine Kasimir-Bauer, Helen Toni Jun, Yong Wei, Hanqiu Zheng, Rebecca Tang, Jin Chen, Brendan Lee, Lanjing Zhang, and Wenyang Li
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,medicine.drug_class ,medicine.medical_treatment ,Medizin ,Clone (cell biology) ,Mice, Transgenic ,Antineoplastic Agents ,Bone Neoplasms ,Monoclonal antibody ,Article ,Mice ,03 medical and health sciences ,Breast cancer ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Neutralizing antibody ,Chemotherapy ,Osteoblasts ,biology ,Receptors, Notch ,business.industry ,Antibodies, Monoclonal ,Bone metastasis ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,biology.protein ,business ,Jagged-1 Protein - Abstract
Bone metastasis is a major health threat to breast cancer patients. Tumor-derived Jagged1 represents a central node in mediating tumor-stromal interactions that promote osteolytic bone metastasis. Here, we report the development of a highly effective fully human monoclonal antibody against Jagged1 (clone 15D11). In addition to its inhibitory effect on bone metastasis of Jagged1-expressing tumor cells, 15D11 dramatically sensitizes bone metastasis to chemotherapy, which induces Jagged1 expression in osteoblasts to provide a survival niche for cancer cells. We further confirm the bone metastasis-promoting function of osteoblast-derived Jagged1 using osteoblast-specific Jagged1 transgenic mouse model. These findings establish 15D11 as a potential therapeutic agent for the prevention or treatment of bone metastasis.
- Published
- 2017
18. Response-Derived Input Function Estimation for Dynamic Contrast-Enhanced MRI Demonstrated by Anti-DLL4 Treatment in a Murine U87 Xenograft Model
- Author
-
Angela Coxon, Sharon E. Ungersma, Jodi Moriguchi, H. Toni Jun, Matthew D. Silva, Albert Gomez, and Brittany Yerby
- Subjects
Cancer Research ,medicine.medical_specialty ,Contrast Media ,Mice, Nude ,Expected value ,030218 nuclear medicine & medical imaging ,LTI system theory ,03 medical and health sciences ,Drug treatment ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Tumor perfusion ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Computer Simulation ,Mathematics ,Adaptor Proteins, Signal Transducing ,medicine.diagnostic_test ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Input function ,Membrane Proteins ,Magnetic resonance imaging ,Signal Processing, Computer-Assisted ,Magnetic Resonance Imaging ,Xenograft Model Antitumor Assays ,Oncology ,030220 oncology & carcinogenesis ,Area Under Curve ,Immunoglobulin G ,Dynamic contrast-enhanced MRI ,Female ,Radiology ,Biological system - Abstract
Dynamic contrast-enhanced magnetic resonance imaging (DCE MRI) is an accepted method to evaluate tumor perfusion and permeability and anti-vascular cancer therapies. However, there is no consensus on the vascular input function estimation method, which is critical to kinetic modeling and K trans estimation. This work proposes a response-derived input function (RDIF) estimated from the response of the tumor, modeled as a linear, time-invariant (LTI) system. In an LTI system, an unknown input can be estimated from the system response. If applied to DCE MRI, this method would eliminate need of distal image-derived inputs, model inputs, or reference regions. The RDIF method first determines each tumor pixel’s best-fit input function, and then combines the individual fits into a single input function for the entire tumor. The method was tested with simulations and a xenograft study with anti-vascular drug treatment. Simulations showed successful estimation of input function expected values and good performance in the presence of noise. In vivo, significant reductions in K trans and AUC occurred 2 days following anti-delta-like ligand 4 treatment. The in vivo study results yielded K trans consistent with published data in xenograft models. The RDIF method for DCE analysis offers an alternative, easy-to-implement method for estimating the input function in tumors. The method assumes that during the DCE experiment, the changes observed by MRI result solely from vascular perfusion and permeability kinetics, and that information can be used to model the input function. Importantly, the method is demonstrated in a murine xenograft study to yield K trans results consistent with literature values and suitable for compound studies.
- Published
- 2017
19. Identification of a Novel Recepteur d'Origine Nantais/c-Met Small-Molecule Kinase Inhibitor with Antitumor Activity In vivo
- Author
-
Monica Reese, Yihong Zhang, April Chen, Tao Osgood, Karen Rex, Jasmine Lin, Jodi Moriguchi, Yajing Yang, Bethany Mattson, Angela Coxon, Isabelle Dussault, Paula Kaplan-Lefko, Tae-Seong Kim, and Teresa L. Burgess
- Subjects
Cancer Research ,C-Met ,Blotting, Western ,Mice, Nude ,Receptor tyrosine kinase ,Mice ,chemistry.chemical_compound ,In vivo ,Animals ,Humans ,Immunoprecipitation ,Phosphorylation ,Kinase activity ,Protein kinase A ,Receptor ,Protein Kinase Inhibitors ,Molecular Structure ,biology ,Kinase ,Receptor Protein-Tyrosine Kinases ,Proto-Oncogene Proteins c-met ,Xenograft Model Antitumor Assays ,Molecular biology ,Oncology ,chemistry ,Colonic Neoplasms ,NIH 3T3 Cells ,Quinolines ,biology.protein ,Pyrazoles ,Female ,Signal transduction ,Signal Transduction - Abstract
Recepteur d'origine nantais (RON) is a receptor tyrosine kinase closely related to c-Met. Both receptors are involved in cell proliferation, migration, and invasion, and there is evidence that both are deregulated in cancer. Receptor overexpression has been most frequently described, but other mechanisms can lead to the oncogenic activation of RON and c-Met. They include activating mutations or gene amplification for c-Met and constitutively active splicing variants for RON. We identified a novel inhibitor of RON and c-Met, compound I, and characterized its in vitro and in vivo activities. Compound I selectively and potently inhibited the kinase activity of RON and c-Met with IC50s of 9 and 4 nmol/L, respectively. Compound I inhibited hepatocyte growth factor–mediated and macrophage-stimulating protein–mediated signaling and cell migration in a dose-dependent manner. Compound I was tested in vivo in xenograft models that either were dependent on c-Met or expressed a constitutively active form of RON (RONΔ160 in HT-29). Compound I caused complete tumor growth inhibition in NIH3T3 TPR-Met and U-87 MG xenografts but showed only partial inhibition in HT-29 xenografts. The effect of compound I in HT-29 xenografts is consistent with the expression of the activating b-Raf V600E mutation, which activates the mitogen-activated protein kinase pathway downstream of RON. Importantly, tumor growth inhibition correlated with the inhibition of c-Met–dependent and RON-dependent signaling in tumors. Taken together, our results suggest that a small-molecule dual inhibitor of RON/c-Met has the potential to inhibit tumor growth and could therefore be useful for the treatment of patients with cancers where RON and/or c-Met are activated. [Cancer Res 2008;68(16):6680–7]
- Published
- 2008
- Full Text
- View/download PDF
20. Discovery of a Potent, Selective, and Orally Bioavailable c-Met Inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458)
- Author
-
Mark H. Norman, Yohannes Teffera, Longbin Liu, Isabelle Dussault, Tae-Seong Kim, Roman Shimanovich, Paula Kaplan-Lefko, Ning Xi, Matthew R. Lee, Jodi Moriguchi, Jean-Christophe Harmange, Steven F. Bellon, Yajing Yang, Jasmine Lin, Annette Bak, Aaron C. Siegmund, Loren Berry, April Chen, Liyue Huang, Celia Dominguez, Yihong Zhang, and Karen Rex
- Subjects
Cell Survival ,Stereochemistry ,medicine.drug_class ,Administration, Oral ,Aminopyridines ,Carboxamide ,Pyrazole ,Chemical synthesis ,Receptor tyrosine kinase ,c-Met inhibitor ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Cells, Cultured ,Mice, Inbred BALB C ,Molecular Structure ,Bicyclic molecule ,biology ,Proto-Oncogene Proteins c-met ,chemistry ,Enzyme inhibitor ,Drug Design ,Mutation ,biology.protein ,Pyrazoles ,Molecular Medicine ,Pyrazolones - Abstract
Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.
- Published
- 2008
- Full Text
- View/download PDF
21. Discovery and Optimization of Triazolopyridazines as Potent and Selective Inhibitors of the c-Met Kinase
- Author
-
Steven F. Bellon, Monica Reese, Jay Larrow, Isabelle Dussault, Stephanie Springer, Paula Kaplan-Lefko, Jodi Moriguchi, David Bauer, Yajing Yang, Kavita Shah, Loren Berry, Jean-Christophe Harmange, Christiane Bode, Karen Rex, Brian K. Albrecht, Yihong Zhang, Doug Hoffman, Jasmine Lin, Deborah Choquette, Alessandro Boezio, Alexander M. Long, Roman Shimanovich, Anne B. O’Connor, Aaron C. Siegmund, April Chen, Cary Fridrich, Julia Lohman, Yohannes Teffera, Michele Potashman, and Satoko Hirai
- Subjects
Models, Molecular ,C-Met ,In Vitro Techniques ,Crystallography, X-Ray ,medicine.disease_cause ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Growth factor receptor ,Drug Discovery ,medicine ,Animals ,Phosphorylation ,Molecular Structure ,Oncogene ,Hepatocyte Growth Factor ,Chemistry ,Kinase ,Proto-Oncogene Proteins c-met ,Triazoles ,Rats ,Pyridazines ,Biochemistry ,Microsomes, Liver ,Cancer research ,Molecular Medicine ,Hepatocyte growth factor ,Signal transduction ,Carcinogenesis ,medicine.drug - Abstract
Tumorigenesis is a multistep process in which oncogenes play a key role in tumor formation, growth, and maintenance. MET was discovered as an oncogene that is activated by its ligand, hepatocyte growth factor. Deregulated signaling in the c-Met pathway has been observed in multiple tumor types. Herein we report the discovery of potent and selective triazolopyridazine small molecules that inhibit c-Met activity.
- Published
- 2008
- Full Text
- View/download PDF
22. c-Met Inhibitors with Novel Binding Mode Show Activity against Several Hereditary Papillary Renal Cell Carcinoma-related Mutations
- Author
-
Paul E. Rose, Teresa L. Burgess, Tae-Seong Kim, Jodi Moriguchi, Alexander M. Long, Angela Coxon, Isabelle Dussault, Steven F. Bellon, Paula Kaplan-Lefko, Carol W. Johnson, Andrew Tasker, Anne B. O’Connor, Yihong Zhang, Karen Rex, Yan Gu, and Yajing Yang
- Subjects
Models, Molecular ,Indoles ,C-Met ,Protein Conformation ,Recombinant Fusion Proteins ,Transplantation, Heterologous ,Mutant ,Mice, Nude ,Hereditary Papillary Renal Cell Carcinoma ,Pyrimidinones ,Crystallography, X-Ray ,Biochemistry ,Piperazines ,Receptor tyrosine kinase ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Animals ,Humans ,Transferase ,Binding site ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,Molecular Biology ,Sulfonamides ,Binding Sites ,biology ,Kinase ,Cell Biology ,Proto-Oncogene Proteins c-met ,Small molecule ,Molecular biology ,Kidney Neoplasms ,chemistry ,Drug Design ,Mutation ,Quinolines ,Cancer research ,biology.protein ,Female ,Neoplasm Transplantation - Abstract
c-Met is a receptor tyrosine kinase often deregulated in human cancers, thus making it an attractive drug target. One mechanism by which c-Met deregulation leads to cancer is through gain-of-function mutations. Therefore, small molecules capable of targeting these mutations could offer therapeutic benefits for affected patients. SU11274 was recently described and reported to inhibit the activity of the wild-type and some mutant forms of c-Met, whereas other mutants are resistant to inhibition. We identified a novel series of c-Met small molecule inhibitors that are active against multiple mutants previously identified in hereditary papillary renal cell carcinoma patients. AM7 is active against wild-type c-Met as well as several mutants, inhibits c-Met-mediated signaling in MKN-45 and U-87 MG cells, and inhibits tumor growth in these two models grown as xenografts. The crystal structures of AM7 and SU11274 bound to unphosphorylated c-Met have been determined. The AM7 structure reveals a novel binding mode compared with other published c-Met inhibitors and SU11274. The molecule binds the kinase linker and then extends into a new hydrophobic binding site. This binding site is created by a significant movement of the C-helix and so represents an inactive conformation of the c-Met kinase. Thus, our results demonstrate that it is possible to identify and design inhibitors that will likely be active against mutants found in different cancers.
- Published
- 2008
- Full Text
- View/download PDF
23. In Vitro and In Vivo Activity of AMG 337, a Potent and Selective MET Kinase Inhibitor, in MET-Dependent Cancer Models
- Author
-
Martin A. Broome, Jean-Christophe Harmange, Jodi Moriguchi, Hue T. Kha, Benny Amore, Michael A. Damore, Robert Radinsky, Richard Kendall, Teresa L. Burgess, Angela Coxon, Deborah Choquette, Karen Rex, Yihong Zhang, Daniel M. Baker, Jonathan Werner, Paul E. Hughes, Yajing Yang, Isabelle Dussault, Paula Kaplan-Lefko, and Sean Caenepeel
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Cell Survival ,MAP Kinase Signaling System ,Antineoplastic Agents ,Pharmacology ,03 medical and health sciences ,Mice ,Necrosis ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Receptor ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Dose-Response Relationship, Drug ,Kinase ,Chemistry ,Gene Amplification ,Proto-Oncogene Proteins c-met ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Mechanism of action ,Cell culture ,030220 oncology & carcinogenesis ,Phosphorylation ,Female ,medicine.symptom ,Signal Transduction - 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
- 2015
24. Abstract 2968: A novel bispecific CD3/CDH19 antibody construct (CDH19 BiTE) directs potent killing of melanoma cells in vitro and in vivo and is enhanced by blockade of PD-L1
- Author
-
Shyun Li, Khue Dang, Pedro J. Beltran, Suzanne Coberly, Julie M. Bailis, Ryan Case, Brendon Frank, Amy Gilbert, Fei Lee, James B. Rottman, Kim Merriam, Beth Hinkle, Jodi Moriguchi, and Gordon Moody
- Subjects
Cancer Research ,biology ,business.industry ,Melanoma ,CD3 ,Cancer ,medicine.disease ,In vitro ,Oncology ,In vivo ,Immunology ,Blocking antibody ,Cancer research ,medicine ,biology.protein ,Antibody ,Cytotoxicity ,business - Abstract
CDH19 is an unconventional type 2 cadherin widely expressed in malignant melanoma with limited expression in normal tissues. RNAseq and immunohistochemical analysis of CDH19 in human samples confirmed its over-expression in >60% of melanoma, whereas normal expression primarily occurred in tissues derived from the neural crest such as nerve fibers and autonomic ganglia. CD3 based bi-specific T-cell engagers (BiTEs) were used to determine if redirected lysis of T-cells against CDH19 would drive anti-tumor efficacy. In vitro, the CDH19 BiTEs were high affinity binders to both human and cyno CDH19, and were able to induce specific T-cell activation and cytotoxicity against a panel of melanoma cell lines. In addition, soluble CDH19 levels were detected in human serum and the effects of BiTE cytotoxicity toward melanoma cells in the presence of soluble CDH19 was investigated. In vivo studies were conducted to confirm the specificity and activity of CDH19 BiTEs in xenograft models of melanoma. The CDH19 BiTE AMG-CDH19X was able to cause tumor growth inhibition in models expressing as few as 250 receptors per cell, and inhibition of tumor growth was enhanced by the addition of a blocking antibody against PD-L1. Immunohistochemical analysis of post-treatment xenograft samples suggested that anti-PD-L1 the persistence of tumor reactive T cells, and provided rationale for combining a BiTE against CDH19 with a PD-1 or PD-L1 blocking antibody in melanoma. In summary, targeting CDH19 presents a promising novel opportunity for BiTEs in the treatment of melanoma, both alone and in combination with current standard of care. Citation Format: Gordon E. Moody, Jodi Moriguchi, Shyun Li, Fei Lee, Brendon Frank, Amy Gilbert, Ryan Case, Khue Dang, Beth Hinkle, Suzanne Coberly, James Rottman, Kim Merriam, Julie Bailis, Pedro J. Beltran. A novel bispecific CD3/CDH19 antibody construct (CDH19 BiTE) directs potent killing of melanoma cells in vitro and in vivo and is enhanced by blockade of PD-L1. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2968.
- Published
- 2016
- Full Text
- View/download PDF
25. Conatumumab, a fully human agonist antibody to death receptor 5, induces apoptosis via caspase activation in multiple tumor types
- Author
-
Paula J. Kaplan-Lefko, Jonathan D. Graves, Stephen J. Zoog, Yang Pan, Jason Wall, Daniel G. Branstetter, Jodi Moriguchi, Angela Coxon, Justin N. Huard, Ren Xu, Matthew L. Peach, Gloria Juan, Stephen Kaufman, Qing Chen, Allison Bianchi, Jennifer J. Kordich, Mark Ma, Ian N. Foltz, and Brian C. Gliniak
- Subjects
Agonist ,Cancer Research ,medicine.drug_class ,Apoptosis ,Conatumumab ,chemistry.chemical_compound ,Mice ,In vivo ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Receptor ,Caspase ,Pharmacology ,Caspase 7 ,biology ,Caspase 3 ,Antibodies, Monoclonal ,Molecular biology ,Xenograft Model Antitumor Assays ,Enzyme Activation ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Oncology ,chemistry ,Monoclonal ,Cancer research ,biology.protein ,Molecular Medicine ,Tumor necrosis factor alpha - Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to death receptors 4 and 5 (DR4, DR5) to transduce apoptotic signals. Conatumumab (AMG 655) is an investigational, fully human monoclonal agonist antibody (IgG(1)) to human DR5, which induces apoptosis via caspase activation. In this study, we demonstrate that conatumumab binds to DR5, activating intracellular caspases in vitro in the presence of a cross-linker. We also show that conatumumab has activity in vivo and inhibits tumor growth in colon (Colo205 and HCT-15), lung (H2122) and pancreatic (MiaPaCa2/T2) xenograft models. Conatumumab also enhances the antitumor activity of chemotherapeutics in vivo. Caspase activation in Colo205 tumors is dose-dependent and correlated with serum concentrations of conatumumab. We demonstrate for the first time that increases in serum caspase-3/7 activity and levels of M30 (neoepitope of caspase-cleaved cytokeratin-18) are linked to activation of the extrinsic apoptotic pathway using conatumumab in a preclinical model. These data suggest that conatumumab has potential as a therapeutic agent for treating patients with multiple tumor types, and that serum caspase-3/7 and M30 levels may serve as biomarkers of conatumumab activity.
- Published
- 2010
26. Pharmacodynamic analysis of an agonistic antibody to the costimulatory receptor GITR
- Author
-
Beltran Pedro, Patricia McElroy, Yannick Bulliard, Ji-Rong Sun, Jodi Moriguchi, Gordon Moody, and Hong Tan
- Subjects
Pharmacology ,Cancer Research ,Tumor microenvironment ,TNFRSF18 ,biology ,Effector ,Immunology ,Spleen ,In vitro ,medicine.anatomical_structure ,Oncology ,In vivo ,Poster Presentation ,medicine ,biology.protein ,Molecular Medicine ,Immunology and Allergy ,Antibody ,Receptor - Abstract
GITR/TNFRSF18 is a member of the TNF-receptor superfamily preferentially expressed on regulatory T cells (Tregs) and activated T effector cells. Antibody agonists to GITR claim two distinct mechanisms to overcome the repressive tumor microenvironment and drive anti-tumor efficacy in vivo: receptor agonism (forward signaling) on T effector cells and FcγR-mediated Treg depletion. We sought to better understand the contribution of these two mechanisms using pharmacodynamic readouts relating target coverage, Treg depletion and efficacy using isotypic variants of a surrogate antibody against mouse GITR, DTA-1. First, target coverage was determined in spleen, tumor and draining lymph node following treatment with a single dose of mouse IgG2a DTA-1. In this study, efficacy correlated with doses that covered >90% GITR-expressing intratumoral leukocytes and depleted >90% intratumoral Tregs at 24 hours post-dose. Though displaying equivalent agonistic activity in vitro and achieving a similar level of target coverage, the mouse IgG1 N297A variant of DTA-1 neither depleted Tregs nor displayed anti-tumor activity in vivo, in confirmation of recent literature. To further explore the influence of Fc engagement, additional DTA-1 isotypic variants were generated and tested in vivo. In this study, we confirmed that preferential engagement of Fcγ receptors was necessary for optimal activity, as the mouse IgG1 DTA-1 variant failed to regress tumors. Additionally, we identified a variant with enhanced Treg depletion properties, however, the enhanced depletion did not translate to improved anti-tumor efficacy. Lastly, we sought to understand if mouse IgG2a DTA-1-would enhance the effect of PD-1 / PD-L1 blockade in vivo. Using the MC38 tumor model, we observed synergistic tumor regression in the combination group versus either monotherapy. Given the likely non-overlapping mechanism of the antibodies, the results suggest that an ADCC-enabled agonistic GITR antibody could provide benefit to human cancer patients in combination with, or refractory to, PD-1/PD-L1 inhibitors
- Published
- 2015
27. Abstract 728: AMG 337, a novel, potent and selective MET kinase inhibitor, has robust growth inhibitory activity in MET-dependent cancer models
- Author
-
Jodi Moriguchi, Karen Rex, Sean Caenepeel, Richard Kendall, Paul E. Hughes, Yihong Zhang, Angela Coxon, Robert Radinsky, Yajing Yang, Martin A. Broome, Deborah Choquette, Isabelle Dussault, and Paula Kaplan-Lefko
- Subjects
Cancer Research ,Cell growth ,Kinase ,business.industry ,Cancer ,Cell cycle ,Pharmacology ,medicine.disease ,Oncology ,Apoptosis ,Medicine ,Hepatocyte growth factor ,Kinase activity ,business ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Signaling through the MET receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), promotes cell proliferation, survival, and invasion. Activation of MET signaling is a relatively common hallmark of a diverse range of human cancer types, and as a result, inhibition of MET signaling represents an attractive therapeutic opportunity for the treatment of cancer. In this study, we describe the characterization of AMG 337, a potent and highly selective small molecule ATP-competitive MET kinase inhibitor that demonstrates robust activity in MET-dependent cancer models. In enzymatic assays, AMG 337 inhibited MET kinase activity with an IC50 of < 5nM nM. AMG 337 demonstrated exquisite selectivity for MET when profiled against a diverse panel of over 400 protein and lipid kinases in a competitive binding assay. In cellular assays, AMG 337 inhibited HGF-dependent MET phosphorylation with an IC50 of < 10 nM. To identify predictive genomic markers of response, AMG 337 was profiled in cell viability assays using a diverse panel of over 200 cancer cell lines. Treatment with AMG 337 only affected the viability of two gastric cancer cell lines (SNU-5 and Hs746T), both of which harbor amplification of the MET gene. The AMG 337 IC50 in the two sensitive cell lines was < 50 nM, and > 10 μM in all other tested cell lines. Further studies in an expanded panel of additional cancer cell lines derived from gastric, NSCLC, and esophageal cancer confirmed that the in-vitro anti-proliferative activity of AMG 337 correlated with amplification of MET. In those cell lines, treatment with AMG 337 inhibited downstream PI3K and MAPK signaling pathways, which translated into growth arrest as evidenced by an accumulation of cells in the G1 phase of the cell cycle, a concomitant reduction in DNA synthesis, and the induction of apoptosis. In vivo, oral administration of AMG 337 resulted in robust dose-dependent anti-tumor efficacy in MET amplified gastric cancer xenograft models, with inhibition of tumor growth consistent with the pharmacodynamic modulation of MET signaling. In conclusion, these findings illustrate the potential clinical utility of AMG 337 as a therapeutic agent for the treatment of tumors with evidence of dysregulated MET signaling, including MET amplification. A phase 1 clinical study is currently evaluating the safety, tolerability and pharmacokinectics of AMG 337 in patients with solid tumors. Citation Format: Paul E. Hughes, Yajing Yang, Karen Rex, Yihong Zhang, Paula J. Kaplan-Lefko, Sean Caenepeel, Jodi Moriguchi, Martin Broome, Deborah Choquette, Robert Radinsky, Richard Kendall, Angela Coxon, Isabelle Dussault. AMG 337, a novel, potent and selective MET kinase inhibitor, has robust growth inhibitory activity in MET-dependent cancer models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 728. doi:10.1158/1538-7445.AM2014-728
- Published
- 2014
- Full Text
- View/download PDF
28. Abstract 2991: Triple combination of bevacizumab, anti-DLL4 (delta like ligand 4) and trebananib gives enhanced therapeutic effects in three xenograft tumor models
- Author
-
Jon Oliner, Jodi Moriguchi, Angela Coxon, H. Toni Jun, Dave Cordover, Steve Kaufman, Charlie Starnes, and Bethany Mattson
- Subjects
Cancer Research ,education.field_of_study ,Pathology ,medicine.medical_specialty ,Delta-like ligand 4 ,Bevacizumab ,business.industry ,medicine.drug_class ,Angiogenesis ,Therapeutic effect ,Cancer ,medicine.disease ,Monoclonal antibody ,Oncology ,Cancer research ,Medicine ,U87 ,business ,Ovarian cancer ,education ,medicine.drug - Abstract
Bevacizumab is a monoclonal antibody targeting VEGF that is clinically approved for the treatment of multiple indications in oncology, including colorectal, lung and renal cancer as well as glioblastoma. Clinical activity has also been demonstrated in breast and ovarian cancer as well, however, the time periods of benefit for all of these indications is in months (1-5). Multiple pathways are known to participate in the process of angiogenesis, and the limited effects of bevacizumab may represent the maximum activity of targeting just one of them. The purpose of the present study was to examine the effect of simultaneously targeting multiple angiogenic axes including angiopoietins 1 and 2, Dll4 and VEGF. Trebananib is a recombinant peptide-Fc fusion protein that selectively blocks the Tie2 interactions of both Ang1 and Ang2. Trebananib, as well as a recombinant antibody to Dll4 were produced at Amgen, Inc., Thousand Oaks, CA. Bevacizumab was purchased from a local pharmacy. These reagents were employed in the combination treatment of three different preclinical murine tumor models (U87, glioblastoma; MiaPaca and BxPC3, pancreatic carcinoma), one of which (BxPC3) was specifically selected for its relative resistance to VEGF/KDR inhibition. Harlan Athymic nu/nu female mice at 6-7 weeks of age were injected subcutaneously on the right flank with 5 x 106 tumor cells in the presence of Matrigel and treated when the tumors were approximately 300 mm3. The results, as supported by histomorphometric analysis (percent blood vessel area, viable tumor burden), demonstrate that under conditions of optimal biological dosing, the addition of trebananib to a two-way combination of bevacizumab and anti-Dll4 resulted in a statistically significant enhancement of therapeutic effect for the three-way combination, an observation made consistently over 3 different tumor models and 3 different dose levels of bevacizumab and anti-Dll4 (p = or < 0.0024; RMANOVA). Furthermore, the therapeutic effect in all three models approximated prolonged stasis, the ultimate theoretical goal of antiangiogenesis in the treatment of cancer. 1. NEJM, vol. 350, p. 2335, 2004. 2. NEJM, vol. 355, p. 2542, 2006. 3. Lancet, vol. 370, p. 2103, 2007. 4. NEJM, vol. 357, p. 2666, 2007. 5. J. Clin. Oncol., vol. 30, p. 2039, 2012. Citation Format: Bethany Mattson, Jodi Moriguchi, H. Toni Jun, Angela Coxon, Dave Cordover, Steve Kaufman, Jon Oliner, Charlie Starnes. Triple combination of bevacizumab, anti-DLL4 (delta like ligand 4) and trebananib gives enhanced therapeutic effects in three xenograft tumor models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2991. doi:10.1158/1538-7445.AM2014-2991
- Published
- 2014
- Full Text
- View/download PDF
29. Abstract 5089: Combined treatment of trebananib (AMG 386) with panitumumab in preclinical tumor models
- Author
-
James Bready, Stephen Kaufman, Paula Kaplan-Lefko, Jonathan D. Oliner, Jodi Moriguchi, Marc Payton, Robert Radinsky, and Angela Coxon
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,medicine.diagnostic_test ,Cell growth ,business.industry ,Cancer ,medicine.disease ,Angiopoietin receptor ,Flow cytometry ,Angiopoietin ,Oncology ,Stroma ,Epidermoid carcinoma ,Cancer research ,medicine ,biology.protein ,Panitumumab ,business ,medicine.drug - Abstract
Introduction: Cancer therapies that combine agents directly targeting tumor cells with those impacting the tumor stroma hold the promise of improving patient outcomes. In this preclinical study, we examined the effects of inhibiting both EGF signaling in human tumor cells and angiopoietin signaling in the murine stroma. Using tumor xenograft models, we tested a treatment combination of panitumumab, a fully human monoclonal antibody against human EGF receptor, and trebananib, an investigational peptide-Fc fusion protein that prevents binding of angiopoietins 1 and 2 to their receptor, Tie2. We assessed the effect of this combination on tumor growth and explored mechanisms of enhanced efficacy. Methods: Athymic nude mice were injected SC in the right flank with either 1x107 A431 (human epidermoid carcinoma) or 5x106 DLD-1 (colon adenocarcinoma) cells. When tumors were ∼ 200 mm3, 20 μg panitumumab IP and/or 70 μg trebananib SC were given twice weekly. Controls were isotype antibody or human Fc. In mechanism of action studies, DLD-1 tumors (∼ 300-400 mm3) received 2 treatments over 5 days before harvest and processing for paraffin sectioning. To examine endothelial cell proliferation, treated tumors were enzyme-digested and stained with anti-CD31, anti-CD45, and anti-BrdU antibodies. The percentage of BrdU-positive tumor-associated endothelial cells (CD31high/CD45neg) was determined by flow cytometry. Results: In both tumor models, combined treatment with panitumumab and trebananib resulted in significantly greater tumor growth inhibition (p≤0.014) than treatment with either single agent alone. All treatments were well tolerated; no significant weight loss was observed. Mechanistic studies revealed that combination treatment did not alter the single-agent activity of either treatment: histological analyses showed a significant reduction in estimated blood vessel area within viable tumor after treatment with either trebananib (p=0.0006) or panitumumab (p=0.012) alone; this reduction was not affected by combination treatment. Similarly, BrdU incorporation assays demonstrated that single-agent panitumumab significantly impaired tumor cell proliferation (p=0.0003), whereas single-agent trebananib significantly reduced endothelial cell proliferation (p Conclusions: This study demonstrates that combined treatment with agents targeting EGF signaling in the tumor and angiopoietin signaling in the stroma results in significantly enhanced antitumor activity. Neither agent appears to impact the biochemical activities of the other, but instead gives rise to an orthogonal enhancement of antitumor activities. These findings support further clinical testing of agents that target the tumor stroma in combination with direct tumor-targeting therapies. Citation Format: James V. Bready, Paula Kaplan-Lefko, Jodi Moriguchi, Marc Payton, Stephen Kaufman, Jonathan Oliner, Robert Radinsky, Angela Coxon. Combined treatment of trebananib (AMG 386) with panitumumab in preclinical tumor models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5089. doi:10.1158/1538-7445.AM2013-5089
- Published
- 2013
- Full Text
- View/download PDF
30. Abstract LB-311: Inhibition of angiopoietin and Dll4 signaling in the tumor vasculature leads to increased efficacy in mouse tumor xenograft models
- Author
-
Jodi Moriguchi, Juan Estrada, Jenni Lavallee, Jan Sun, Toni Jun, Paula Kaplan-Lefko, Stephen Kaufman, Teresa L. Burgess, Cindy Park, William Wayne, Taruna Arora, Beth Ziegler, and Angela Coxon
- Subjects
Cancer Research ,biology ,Chemistry ,medicine.drug_class ,Angiogenesis ,medicine.disease ,Monoclonal antibody ,Angiopoietin receptor ,Angiopoietin ,Endothelial stem cell ,medicine.anatomical_structure ,Oncology ,Angiopoietin-1 ,Glioma ,cardiovascular system ,medicine ,biology.protein ,Cancer research ,Blood vessel - Abstract
Inhibition of angiogenesis is a proven cancer treatment strategy as an adequate blood supply is critical for tumor growth. Two of the key pathways regulating tumor angiogenesis are angiopoietin/Tie2 and Dll4/Notch. Inhibition of the angiopoietin/Tie2 pathway has been shown to reduce blood vessel density and inhibit tumor growth in mouse xenograft models. Inhibition of Dll4 also prevents tumor xenograft growth but does so by promoting a non-productive vascular network via excessive endothelial branching and sprouting. In this study, we examined the effects of combined inhibition of these pathways in models of angiogenesis and tumor growth. To inhibit the angiopoietin pathway we used AMG 386, a selective angiopoietin 1/2-neutralizing peptibody that prevents the interaction between these two angiopoietins and the Tie2 receptor. To inhibit Dll4 we used a neutralizing monoclonal antibody. The combination of AMG 386 and anti-Dll4 led to enhanced antitumor activity compared with either agent alone in four xenograft models (U-87 glioma, Calu-6 lung, Colo205 colorectal and MiaPaca pancreatic tumors). All treatments were well tolerated and no weight loss was observed. Histological analysis of U-87 tumors following one week of treatment revealed an increase in vessel area with anti-Dll4 alone and the combined treatment with AMG 386 showed a similar increase. At these same doses, AMG 386 reduced anti-Dll4-induced tumor associated endothelial cell proliferation following 24 hours of treatment. We also examined AMG 386/anti-Dll4 treatment in models of angiogenesis. In the rat cornea, individual anti-Dll4 and AMG 386 treatment had distinct morphological effects on VEGF-induced vessel formation. Vessel number and area were decreased following AMG 386 treatment, whereas vessel cross-branching and terminal tufting were observed following anti-Dll4 treatment. After combined treatment, vessels were truncated but the branching/tufting phenotype was preserved. Similar effects were seen in the mouse neonatal retina, where AMG 386 and anti-Dll4 treatment alone resulted in the expected distinct vascular phenotypes; AMG 386 treatment reduced vessel density and inhibited radial expansion, while anti-Dll4 treatment resulted in marked branching and thickening, and showed full radial expansion. In this model, combined treatment led to clear evidence of both mechanisms of action: increased branching and reduced radial expansion were observed in all retinas examined. Taken together, these data suggest that combined inhibition of the angiopoietin/Tie2 and Dll4/Notch pathways does not interfere with their individual mechanisms of action and furthermore, leads to enhanced efficacy in preclinical models. Therefore, the combination of these agents may have the potential to provide an expanded therapeutic opportunity to inhibit tumor angiogenesis beyond either individual agent alone. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-311. doi:1538-7445.AM2012-LB-311
- Published
- 2012
- Full Text
- View/download PDF
31. Abstract 3558: In vitro and in vivo profiling of class I and class II ATP-competitive c-Met kinase inhibitors defines potential c-Met-specific sensitivity biomarkers
- Author
-
Yajing Yang, Beth Ziegler, Hue Kha, Deborah Choquette, Jodi Moriguchi, Richard T. Lewis, Yihong Zhang, Isabelle Dussault, Paula Kaplan-Lefko, Jasmine Lin, Martin A. Broome, Roman Shimanovich, Karen Rex, Xiaoning Zhao, and Mel Mallari
- Subjects
Cancer Research ,C-Met ,biology ,Kinase ,Small molecule ,Molecular biology ,In vitro ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Oncology ,chemistry ,In vivo ,biology.protein ,Kinase activity ,Autocrine signalling - Abstract
c-Met is a receptor tyrosine kinase with oncogenic potential for which several small molecule inhibitors are currently being tested in clinical trials. There are suggestions in the literature that c-Met inhibitors could have anti-tumor effects under a variety of contexts, including in tumors in which the MET gene is amplified, contains a gain-of-function mutation or in tumors with receptor overexpression. We have developed ATP-competitive c-Met inhibitors that possess different selectivity profiles and fall into two categories. Class I molecules are highly selective for c-Met while class II molecules are multi-kinase inhibitors. These molecules were profiled in vitro and in vivo to identify biomarkers of c-Met dependence. We show that both classes of molecules inhibited c-Met kinase activity in vitro and in vivo. However, class I and class II molecules differed extensively in their spectrum of anti-tumor activity. The anti-proliferative effects of the different c-Met inhibitors were tested in 359 cancer cell lines in vitro. The activity of class I molecules was exclusively restricted to cell lines that harbor MET amplification. Western blotting in sensitive or resistant cell lines showed that c-Met inhibitors effectively block signaling downstream of c-Met only in MET amplified cell lines. In vivo, class I molecules inhibited only the growth of tumor models that are highly dependent on c-Met activity due to MET amplification or due to a hepatocyte growth factor (HGF)-driven autocrine loop. The same models that were sensitive to class I inhibitors were also sensitive to class II inhibitors. However, class II inhibitors had additional anti-tumor activities in vitro and in vivo in models that did not respond to class I inhibitors. The majority of cell lines that responded only to a class II molecule in vitro required much higher drug concentrations than those required to inhibit the growth of MET amplified cell lines. Finally, the growth inhibitory effects observed in vitro and in vivo were consistent with the selectivity profiles of the different c-Met inhibitors. Together our data demonstrate that MET amplification is a clinically identifiable, potential sensitivity biomarker for selective c-Met kinase inhibitors. c-Met expression alone is not a sensitivity biomarker in preclinical models. The spectrum of c-Met inhibitor sensitive models defined in this work could be used to understand whether other small molecule c-Met inhibitors are exquisitely selective for c-Met or harbor additional activities. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3558. doi:10.1158/1538-7445.AM2011-3558
- Published
- 2011
- Full Text
- View/download PDF
32. Discovery of a Potent, Selective, and Orally Bioavailable c-Met Inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458).
- Author
-
Longbin Liu, Aaron Siegmund, Ning Xi, Paula Kaplan-Lefko, Karen Rex, April Chen, Jasmine Lin, Jodi Moriguchi, Loren Berry, Liyue Huang, Yohannes Teffera, Yajing Yang, Yihong Zhang, Steven F. Bellon, Matthew Lee, Roman Shimanovich, Annette Bak, Celia Dominguez, Mark H. Norman, and Jean-Christophe Harmange
- Published
- 2008
- Full Text
- View/download PDF
33. Discovery and Optimization of Triazolopyridazines as Potent and Selective Inhibitors of the c-Met Kinase.
- Author
-
Brian K. Albrecht, Jean-Christophe Harmange, David Bauer, Loren Berry, Christiane Bode, Alessandro A. Boezio, April Chen, Deborah Choquette, Isabelle Dussault, Cary Fridrich, Satoko Hirai, Doug Hoffman, Jay F. Larrow, Paula Kaplan-Lefko, Jasmine Lin, Julia Lohman, Alexander M. Long, Jodi Moriguchi, Anne O’Connor, and Michele H. Potashman
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.