11 results on '"Jasmine Lin"'
Search Results
2. 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
3. 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
4. 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
5. 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
6. 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
7. 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
8. Abstract 2663: Preclinical pharmacokinetic characterizations and human pharmacokinetic prediction of AMG 900, an orally available small molecule inhibitor of aurora kinases
- Author
-
Min-Hwa Jasmine Lin, Wei-Jiang Pan, Liyue Huang, Xuhai Be, Zhiyang Zhao, and Loren Berry
- Subjects
Volume of distribution ,Cancer Research ,Oncology ,Pharmacokinetics ,Kinase ,Chemistry ,Oral administration ,Steady state (chemistry) ,PK Parameters ,Pharmacology ,Small molecule ,Bioavailability - Abstract
Introduction: AMG 900 is a small molecule being developed as an orally administered, highly potent and selective pan-aurora kinase inhibitor. AMG 900 is currently undergoing phase 1 clinical evaluation in patients with advanced solid tumors. The objectives of the studies were to characterize single dose pharmacokinetics (PK) of AMG 900 in preclinical species and use the preclinical PK data to predict its human PK. Methods: Following a single intravenous and oral administration of AMG 900 to mice, rats, dogs, and monkeys, blood samples were collected and processed for plasma by centrifugation. AMG 900 plasma concentrations were determined by LC-MS/MS. Non-compartmental analysis of the concentration - time data was performed to calculate PK parameters of AMG 900. Human PK were predicted based on data from preclinical species using the simple allometric scaling method assuming a human body weight of 60 kg. Results: AMG 900 exhibited low (mouse, monkey and dog) to moderate (rat) clearance in the species evaluated. The volume of distribution at steady state was less than 0.5 L/kg across species. Following intravenous administration, the compound had terminal elimination half-life ranging from 0.6 to 2.4 hours. AMG 900 was well absorbed in fasted animals across species with an oral bioavailability ranging from 31% to 104%. The clearance and volume of distribution at steady state in humans were predicted to be 0.03 L/hr/kg and 0.1 L/kg, respectively, with the predicted half-life to be approximately 2.3 hours. Bioavailability after oral administration of AMG 900 was predicted to be 75%, calculated from the mean values observed in non-clinical species. Conclusion: AMG 900 exhibits good PK properties in preclinical species and it was predicted to have low clearance and good oral bioavailability in human. Available PK results from the on-going phase 1 trial are consistent with the allometric scaling predictions. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2663.
- Published
- 2010
- Full Text
- View/download PDF
9. Abstract 4437: Preclinical characterization of AMG 900, an orally available small molecule inhibitor of aurora kinases in phase 1 clinical trials
- Author
-
Min-Hwa Jasmine Lin, Stephanie D. Geuns-Meyer, Tammy L. Bush, Patricia McElroy, Beth Ziegler, Marc Payton, Patrick Eden, Robert Radinsky, Grace Chung, and Richard Kendall
- Subjects
Cancer Research ,business.industry ,Cell growth ,Kinase ,Pharmacology ,medicine.disease_cause ,chemistry.chemical_compound ,Histone H3 ,Aurora kinase ,Oncology ,Docetaxel ,Paclitaxel ,chemistry ,medicine ,business ,Carcinogenesis ,Mitosis ,medicine.drug - Abstract
Background: The aurora family of serine-threonine kinases (aurora-A, -B, -C) regulate cell-cycle progression in mammalian cells. Aurora-A and aurora-B are essential for proper chromosome congression, segregation, and cytokinesis during mitosis, whereas aurora-C function appears restricted to male meiosis. Aurora-B is responsible for the direct phosphorylation of histone H3 on serine-10. Aurora-A and aurora-B expression is elevated in a variety of human cancers and is associated with advanced clinical staging and poor prognosis. The emergence of aurora kinases as key mitotic regulators and their potential role in tumorigenesis has focused substantial interest in developing selective small molecule inhibitors for the treatment of human cancers. Objective: The aim of this study was to determine the in vitro effects of AMG 900 on a panel of tumor cell lines and profiling its antitumor activity across multiple human xenograft models. Results: AMG 900 is a novel ATP competitive small molecule inhibitor that is potent and highly selective for aurora kinases A, B, and C. In cells, AMG 900 inhibits autophosphorylation of aurora-A and aurora-B as well as the phosphorylation of histone H3. The predominant cellular response of tumor cells to AMG 900 treatment is aborted cell division, which leads to endoreduplication and cell death. The effect of AMG 900 was tested in a panel of 26 cell lines to evaluate its potential to inhibit cell proliferation across multiple tumor types. AMG 900 inhibited the proliferation of tumor cell lines at low nanomolar concentrations (EC50 values 1-6 nM), including a number of multidrug resistant (MDR) cell lines expressing ATP-binding cassette transporters (P-gp and BCRP1). In contrast, paclitaxel and three aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358) showed a loss of potency in these MDR cell lines compared to matched parental cell lines. In nude mice, oral administration of AMG 900 inhibited phosphorylation of histone H3 in tumors in a concentration- and dose-dependent manner. The effect of AMG 900 was tested in a panel of nine human xenografts representing five tumor types (breast, colon, lung, pancreatic, and uterine). Oral administration of AMG 900 at 15 mg/kg BID for two consecutive days per week or 3 mg/kg BID every day inhibited tumor growth (50-97%, P < 0.005) in all nine of the xenograft models. Importantly, three of these xenograft models are resistant to taxanes. Mice treated with efficacious doses of AMG 900 showed a transient loss of body weight and bone marrow cellularity, consistent with its on-mechanism effects on normal proliferating cells. Conclusion: Based on these preclinical activities, AMG 900 has the potential to treat advanced cancers, including tumors resistant to chemotherapeutic agents such as paclitaxel and docetaxel. AMG 900 is currently undergoing phase 1 clinical evaluation in patients with advanced solid tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4437.
- Published
- 2010
- Full Text
- View/download PDF
10. Abstract 5776: Discovery of AMG 900, a highly selective, orally bioavailable inhibitor of aurora kinases with efficacy in preclinical antitumor models and activity against multidrug-resistant cells
- Author
-
Brian L. Hodous, Richard Kendall, Beth Ziegler, Karina Romero, Marc Payton, Philip R. Olivieri, Bingfan Du, Vinod F. Patel, Holly L. Deak, Grace Chung, Tammy L. Bush, Robert Radinsky, Patrick Eden, Min-Hwa Jasmine Lin, Stephanie D. Geuns-Meyer, Hanh Nho Nguyen, Victor J. Cee, Liyue Huang, Laurie B. Schenkel, and Xuhai Be
- Subjects
Cancer Research ,biology ,Chemistry ,Kinase ,Aurora inhibitor ,Pharmacology ,biology.organism_classification ,HeLa ,Histone H3 ,Oncology ,Aurora Kinase C ,Phosphorylation ,Aurora Kinase B ,Cell potency - Abstract
The aurora family of serine/threonine kinases (Aurora-A, -B, -C) regulate cell-cycle progression in mammalian cells. Whereas aurora kinase C function appears restricted to meiosis in males, aurora kinases A and B are essential for proper chromosome congression, segregation, and cytokinesis during mitosis. Aurora kinases A and B have been implicated in tumorigenesis, with overexpression levels correlating to clinical staging of cancers and poor prognosis. Thus, these mitotic kinases have become the subject of much interest as targets for anticancer therapy. N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-phenyl-1-phthalazinamine was a key aurora kinase inhibitor lead, possessing oral bioavailability in rats that was lacking in the anthranilamide compounds from which it was derived. This phthalazine compound possessed a key feature that was deemed important to maintain in a clinical candidate: potency against a model multidrug resistant (MDR) cell line (MES-SA Dx5) commensurate with its activity against a cell line that does not overexpress P-gp (HeLa). Improved in vivo potency was desired, as measured by suppression of the phosphorylation of the aurora kinase B substrate Histone H3 on Ser10 six hours after dosing. SAR from targeting this improvement in in vivo activity uncovered a delicate balance between protein binding, pharmacokinetic parameters, and cell potency in MES-SA Dx5 cells. AMG 900 was identified as a suitable candidate for clinical development based on its low single digit nanomolar potency against MDR cell lines, robust PD response (with complete suppression of Histone H3 phosphorylation at six hours), and high selectivity against other kinases. Oral administration of AMG 900 at a well-tolerated dose of 4 mg/kg BID inhibited tumor growth (83% TGI; p < 0.0001) in an HCT116 xenograft model. AMG 900 is currently undergoing phase 1 clinical evaluation in patients with advanced solid tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5776.
- Published
- 2010
- Full Text
- View/download PDF
11. Abstract 3600: Preclinical pharmacokinetic and biomarker analysis of AMG 900, an orally available small molecule inhibitor of aurora kinases, in human xenograft tumor and surrogate tissues
- Author
-
Stephen J. Zoog, Liyue Huang, Raffi Manoukian, Grace Chung, Greg Friberg, Gloria Juan, Richard Kendall, Tammy L. Bush, Robert Radinsky, Min-Hwa Jasmine Lin, Connie Ma, Marc Payton, Beth Ziegler, and Stephanie D. Geuns-Meyer
- Subjects
Cancer Research ,Kinase ,business.industry ,Aurora B kinase ,Cancer ,Pharmacology ,medicine.disease_cause ,medicine.disease ,Oncology ,In vivo ,medicine ,Aurora Kinase C ,Aurora Kinase B ,Aurora Kinase A ,Carcinogenesis ,business - Abstract
Background: The aurora family of serine-threonine kinases (Aurora-A, -B, -C) regulate cell-cycle progression in mammalian cells. Aurora kinases A and B are essential for proper chromosome congression, segregation, and cytokinesis during mitosis, whereas aurora kinase C function appears restricted to male meiosis. Aurora kinase B is responsible for the direct phosphorylation of histone H3 on serine-10 (p-histone H3). Aurora kinase A and B expression is elevated in a variety of human cancers and is associated with advanced clinical staging and poor prognosis. With the emergence of aurora kinases as key mitotic regulators and their potential role in tumorigenesis, we developed AMG 900, a novel ATP competitive small molecule inhibitor, that is highly potent and selective for aurora kinases A, B, and C. In vivo, AMG 900 inhibits the growth of multiple human tumor xenografts, including multidrug-resistant models. Objective: The aim of this study was to establish a pharmacokinetic-pharmacodynamic (PK-PD) relationship for AMG 900 in human tumor xenografts and surrogate tissues. Results: In nude mice, oral administration of AMG 900 inhibited p-histone H3 in tumors and bone marrow in a concentration- and dose-dependent manner. To further refine the AMG 900 PK-PD relationship, p-histone H3 IC50 values for bone marrow and dissociated COLO 205 tumors from mice were associated with plasma concentrations of 294 and 273 ng/mL, respectively. The duration of target coverage required for anti-tumor efficacy was determined to be > 6 hours per day above the in vivo p-histone H3 IC50 using different dosing schedules. To explore whether skin could be a suitable surrogate tissue for measuring p-histone H3 inhibition, tumor-bearing mice were administered AMG 900 at 3.75, 7.5, and 15 mg/kg for 3 hours. Significant suppression of p-histone H3 in skin was only observed at 15 mg/kg (p < 0.0001), whereas in tumor, a dose-dependent inhibition was achieved at all doses (p < 0.0015). To determine the feasibility of measuring p-histone H3 using fine-needle aspirate (FNA) tumor biopsies, tumor-bearing mice were administered AMG 900 at 15 mg/kg for 3 hours. Treatment with AMG 900 significantly inhibited p-histone H3 (> 99%, p < 0.0001) in COLO 205 tumors, suggesting that FNA biopsies may be a viable approach for assessing AMG 900 PD effects in the clinic. Conclusion: These studies demonstrate that AMG 900 inhibits p-histone H3 in tumors and surrogate tissues, although tissues such as skin may be less sensitive to assess PD effects. The use of p-histone H3 as a biomarker to determine plasma levels of AMG 900 required to inhibit aurora B activity may be useful in clinical studies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3600.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.