31 results on '"Tisha San Miguel"'
Search Results
2. 263 A2B694, an autologous logic-gated cell therapy targeting mesothelin
- Author
-
Julian Molina, Talar Tokatlian, Jason Wang, Shruti Sharma, Diane Manry, Martin Naradikian, Grace Asuelime, Breanna DiAndreth, Aaron Winters, Tisha San Miguel, Armen Mardiros, Sandip Patel, Edward Garon, Diane Simeone, Scott Kopetz, Maria Pia Morelli, Theodore Welling, Mitesh Borad, Kedar Kirtane, Eric Ng, John Welch, David Maloney, William Go, Alexander Kamb, Agi Hamburger, and J Randolph Hecht
- Published
- 2022
- Full Text
- View/download PDF
3. Discovery of a Covalent Inhibitor of KRASG12C (AMG 510) for the Treatment of Solid Tumors
- Author
-
Brian A. Lanman, Jennifer R. Allen, John G. Allen, Albert K. Amegadzie, Kate S. Ashton, Shon K. Booker, Jian Jeffrey Chen, Ning Chen, Michael J. Frohn, Guy Goodman, David J. Kopecky, Longbin Liu, Patricia Lopez, Jonathan D. Low, Vu Ma, Ana E. Minatti, Thomas T. Nguyen, Nobuko Nishimura, Alexander J. Pickrell, Anthony B. Reed, Youngsook Shin, Aaron C. Siegmund, Nuria A. Tamayo, Christopher M. Tegley, Mary C. Walton, Hui-Ling Wang, Ryan P. Wurz, May Xue, Kevin C. Yang, Pragathi Achanta, Michael D. Bartberger, Jude Canon, L. Steven Hollis, John D. McCarter, Christopher Mohr, Karen Rex, Anne Y. Saiki, Tisha San Miguel, Laurie P. Volak, Kevin H. Wang, Douglas A. Whittington, Stephan G. Zech, J. Russell Lipford, and Victor J. Cee
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2019
- Full Text
- View/download PDF
4. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity
- Author
-
Timothy J. Price, Victor J. Cee, Tisha San Miguel, Gerald Steven Falchook, Jonathan Werner, Christopher Mohr, Tao Osgood, James Kuo, Kevin Gaida, Xiaochun Zhu, Karen Rex, John D. McCarter, Brian A. Lanman, H. Henary, Roberto Ortiz, Ramaswamy Govindan, Tara Arvedson, Laurie P. Volak, Wenjun Ouyang, J. Russell Lipford, Tyler Holt, Charles G. Knutson, Neelima Koppada, David S. Hong, Aaron S. Rapaport, Anne Y. Saiki, Bert H. O'Neil, Jude Canon, Ji Rong Sun, Marwan Fakih, Brett E. Houk, Dhanashri Bagal, Keegan Cooke, and Jayesh Desai
- Subjects
0301 basic medicine ,Chemotherapy ,Multidisciplinary ,Oncogene ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease_cause ,medicine.disease ,Acquired immune system ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Antigen ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Medicine ,KRAS ,business - Abstract
KRAS is the most frequently mutated oncogene in cancer and encodes a key signalling protein in tumours1,2. The KRAS(G12C) mutant has a cysteine residue that has been exploited to design covalent inhibitors that have promising preclinical activity3–5. Here we optimized a series of inhibitors, using novel binding interactions to markedly enhance their potency and selectivity. Our efforts have led to the discovery of AMG 510, which is, to our knowledge, the first KRAS(G12C) inhibitor in clinical development. In preclinical analyses, treatment with AMG 510 led to the regression of KRASG12C tumours and improved the anti-tumour efficacy of chemotherapy and targeted agents. In immune-competent mice, treatment with AMG 510 resulted in a pro-inflammatory tumour microenvironment and produced durable cures alone as well as in combination with immune-checkpoint inhibitors. Cured mice rejected the growth of isogenic KRASG12D tumours, which suggests adaptive immunity against shared antigens. Furthermore, in clinical trials, AMG 510 demonstrated anti-tumour activity in the first dosing cohorts and represents a potentially transformative therapy for patients for whom effective treatments are lacking. Treatment of KRASG12C-mutant cancer cells with the KRAS(G12C) inhibitor AMG 510 leads to durable response in mice, and anti-tumour activity in patients suggests that AMG 510 could be effective in patients for whom treatments are currently lacking.
- Published
- 2019
- Full Text
- View/download PDF
5. Facile Modulation of Antibody Fucosylation with Small Molecule Fucostatin Inhibitors and Cocrystal Structure with GDP-Mannose 4,6-Dehydratase
- Author
-
Mirna Mujacic, John B. Jordan, Pranhitha Reddy, Aleksander Swietlow, Vivian S. W. Li, Tisha San Miguel, Paul Kodama, Anne-Marie C Rousseau, Seb Caille, E. Allen Sickmier, Michael J. Frohn, Dhanashri Bagal, Steve Osgood, Christopher H. Fotsch, Ki Won Kim, John D. McCarter, Mike Achmatowicz, Oliver R. Thiel, Alex Pickrell, Yong-Jae Kim, and John G. Allen
- Subjects
Guanosine Diphosphate Mannose ,0301 basic medicine ,medicine.drug_class ,Allosteric regulation ,Mannose ,CHO Cells ,Crystallography, X-Ray ,Monoclonal antibody ,Biochemistry ,Mass Spectrometry ,Fucose ,Small Molecule Libraries ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,Cricetinae ,medicine ,Animals ,Hydro-Lyases ,Fucosylation ,Molecular Structure ,General Medicine ,Surface Plasmon Resonance ,Small molecule ,030104 developmental biology ,chemistry ,Molecular Medicine ,Guanosine diphosphate mannose - Abstract
The efficacy of therapeutic antibodies that induce antibody-dependent cellular cytotoxicity can be improved by reduced fucosylation. Consequently, fucosylation is a critical product attribute of monoclonal antibodies produced as protein therapeutics. Small molecule fucosylation inhibitors have also shown promise as potential therapeutics in animal models of tumors, arthritis, and sickle cell disease. Potent small molecule metabolic inhibitors of cellular protein fucosylation, 6,6,6-trifluorofucose per-O-acetate and 6,6,6-trifluorofucose (fucostatin I), were identified that reduces the fucosylation of recombinantly expressed antibodies in cell culture in a concentration-dependent fashion enabling the controlled modulation of protein fucosylation levels. 6,6,6-Trifluorofucose binds at an allosteric site of GDP-mannose 4,6-dehydratase (GMD) as revealed for the first time by the X-ray cocrystal structure of a bound allosteric GMD inhibitor. 6,6,6-Trifluorofucose was found to be incorporated in place of fucose at low levels (1%) in the glycans of recombinantly expressed antibodies. A fucose-1-phosphonate analog, fucostatin II, was designed that inhibits fucosylation with no incorporation into antibody glycans, allowing the production of afucosylated antibodies in which the incorporation of non-native sugar is completely absent-a key advantage in the production of therapeutic antibodies, especially biosimilar antibodies. Inhibitor structure-activity relationships, identification of cellular and inhibitor metabolites in inhibitor-treated cells, fucose competition studies, and the production of recombinant antibodies with varying levels of fucosylation are described.
- Published
- 2016
- Full Text
- View/download PDF
6. Discovery and in Vivo Evaluation of the Potent and Selective PI3Kδ Inhibitors 2-((1S)-1-((6-Amino-5-cyano-4-pyrimidinyl)amino)ethyl)-6-fluoro-N-methyl-3-(2-pyridinyl)-4-quinolinecarboxamide (AM-0687) and 2-((1S)-1-((6-Amino-5-cyano-4-pyrimidinyl)amino)ethyl)-5-fluoro-N-methyl-3-(2-pyridinyl)-4-quinolinecarboxamide (AM-1430)
- Author
-
Iain D. G. Campuzano, Xiaolin Hao, Brian Lucas, Tisha San Miguel, Deanna Mohn, Kirk Henne, Benjamin Fisher, Minna Bui, Mario G. Cardozo, Robert S. Foti, Ron C. Kelly, Christine Vissinga, Thuy B. Tran, John D. McCarter, Leeanne Zalameda, Daniela Metz, Douglas A. Whittington, Sharon Wannberg, Yi-Ling Hu, John Whoriskey, Julio C. Medina, Felix Gonzalez-Lopez de Turiso, Timothy D. Cushing, Xuxia Zhang, Lawrence R. McGee, Michael G. Johnson, Xiao He, Gang Yu, Michelle C. Dunn, Jason Duquette, and Youngsook Shin
- Subjects
0301 basic medicine ,biology ,Stereochemistry ,Chemistry ,Quinoline ,In vitro ,Bioavailability ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Pharmacokinetics ,In vivo ,Pharmacodynamics ,Drug Discovery ,biology.protein ,Molecular Medicine ,Potency ,Keyhole limpet hemocyanin - Abstract
Optimization of the potency and pharmacokinetic profile of 2,3,4-trisubstituted quinoline, 4, led to the discovery of two potent, selective, and orally bioavailable PI3Kδ inhibitors, 6a (AM-0687) and 7 (AM-1430). On the basis of their improved profile, these analogs were selected for in vivo pharmacodynamic (PD) and efficacy experiments in animal models of inflammation. The in vivo PD studies, which were carried out in a mouse pAKT inhibition animal model, confirmed the observed potency of 6a and 7 in biochemical and cellular assays. Efficacy experiments in a keyhole limpet hemocyanin model in rats demonstrated that administration of either 6a or 7 resulted in a strong dose-dependent reduction of IgG and IgM specific antibodies. The excellent in vitro and in vivo profiles of these analogs make them suitable for further development.
- Published
- 2016
- Full Text
- View/download PDF
7. Synthesis and SAR study of potent and selective PI3Kδ inhibitors
- Author
-
Jamie Wong, Thuy B. Tran, Mario G. Cardozo, Kirk Henne, Tisha San Miguel, Julia Suchomel, Deanna Mohn, Julio C. Medina, Leeanne Zalameda, Daniela Metz, Lawrence R. McGee, Xiao He, Youngsook Shin, Simon Wong, Sharon Wannberg, John D. McCarter, Minna Bui, Xiaolin Hao, and Timothy D. Cushing
- Subjects
Male ,Gene isoform ,Class I Phosphatidylinositol 3-Kinases ,Stereochemistry ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Rats ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Drug Discovery ,Quinolines ,Animals ,Humans ,Protein Isoforms ,Molecular Medicine ,Selectivity ,Protein Kinase Inhibitors ,Molecular Biology - Abstract
2,3,4-Substituted quinolines such as (10a) were found to be potent inhibitors of PI3Kδ in both biochemical and cellular assays with good selectivity over three other class I PI3K isoforms. Some of those analogs showed favorable pharmacokinetic properties.
- Published
- 2015
- Full Text
- View/download PDF
8. Discovery and in Vivo Evaluation of (S)-N-(1-(7-Fluoro-2-(pyridin-2-yl)quinolin-3-yl)ethyl)-9H-purin-6-amine (AMG319) and Related PI3Kδ Inhibitors for Inflammation and Autoimmune Disease
- Author
-
Andrew Tasker, Andreas Reichelt, Yi-Ling Hu, Deanna Mohn, John McCarter, Ben Fisher, Robert M. Rzasa, Yi Chen, Julio C. Medina, Gang Yu, Sharon Wannberg, Brian Lucas, Ron C. Kelly, Jennifer Seganish, Felix Gonzalez-Lopez de Turiso, Xiaolin Hao, Kristin L. Andrews, Douglas A. Whittington, Matthew Frank Brown, Dawei Zhang, Youngsook Shin, Mario G. Cardozo, Jason Duquette, Robert C. Wahl, Leeanne Zalameda, Daniela Metz, Vatee Pattaropong, Michael G. Johnson, Tisha San Miguel, Randall W. Hungate, John Whoriskey, Lawrence R. McGee, Timothy D. Cushing, Kirk Henne, Liping H. Pettus, and Xiao He
- Subjects
Models, Molecular ,Adenosine ,Class I Phosphatidylinositol 3-Kinases ,Stereochemistry ,Mice, Transgenic ,Pharmacology ,Crystallography, X-Ray ,Autoimmune Diseases ,Structure-Activity Relationship ,In vivo ,Drug Discovery ,Sf9 Cells ,medicine ,Animals ,Humans ,Structure–activity relationship ,Protein Kinase Inhibitors ,IC50 ,Cells, Cultured ,Whole blood ,Inflammation ,Autoimmune disease ,Mice, Inbred BALB C ,Molecular Structure ,Drug discovery ,Kinase ,Chemistry ,medicine.disease ,Protein Structure, Tertiary ,Disease Models, Animal ,Models, Chemical ,Rats, Inbred Lew ,Quinolines ,Molecular Medicine ,Female ,Amine gas treating ,Protein Binding - Abstract
The development and optimization of a series of quinolinylpurines as potent and selective PI3Kδ kinase inhibitors with excellent physicochemical properties are described. This medicinal chemistry effort led to the identification of 1 (AMG319), a compound with an IC50 of 16 nM in a human whole blood assay (HWB), excellent selectivity over a large panel of protein kinases, and a high level of in vivo efficacy as measured by two rodent disease models of inflammation.
- Published
- 2014
- Full Text
- View/download PDF
9. Phosphoinositide-3-kinase inhibitors: Evaluation of substituted alcohols as replacements for the piperazine sulfonamide portion of AMG 511
- Author
-
Fang-Tsao Hong, Jian Jiang, Brian A. Lanman, Victor J. Cee, Liping H. Pettus, Paul E. Hughes, Nancy Zhang, Douglas A. Whittington, Tisha San Miguel, Anthony B. Reed, Ling Wang, Erin L. Mullady, Kristin L. Andrews, John D. McCarter, Andrew Tasker, Leeanne Zalameda, Mark H. Norman, Tian Wu, Ryan Wurz, and Raju Subramanian
- Subjects
Male ,Pyridines ,Clinical Biochemistry ,Molecular Conformation ,Mice, Nude ,Pharmaceutical Science ,Alcohol ,Biochemistry ,Piperazines ,Rats, Sprague-Dawley ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Piperazine ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,Phosphoinositide-3 Kinase Inhibitors ,chemistry.chemical_classification ,Sulfonamides ,Phosphoinositide 3-kinase ,biology ,Triazines ,TOR Serine-Threonine Kinases ,Organic Chemistry ,In vitro ,Rats ,Sulfonamide ,Liver ,chemistry ,Alcohols ,biology.protein ,Molecular Medicine ,Phosphorylation ,Female ,Hepatocyte growth factor ,Proto-Oncogene Proteins c-akt ,Half-Life ,Protein Binding ,Signal Transduction ,medicine.drug - Abstract
Replacement of the piperazine sulfonamide portion of the PI3Kα inhibitor AMG 511 (1) with a range of aliphatic alcohols led to the identification of a truncated gem-dimethylbenzylic alcohol analog, 2-(5-(4-amino-6-methyl-1,3,5-triazin-2-yl)-6-((5-fluoro-6-methoxypyridin-3-yl)amino)pyridin-3-yl)propan-2-ol (7). This compound possessed good in vitro efficacy and pharmacokinetic parameters and demonstrated an EC50 of 239 ng/mL in a mouse liver pharmacodynamic model measuring the inhibition of hepatocyte growth factor (HGF)-induced Akt Ser473 phosphorylation in CD1 nude mice 6 h post-oral dosing.
- Published
- 2014
- Full Text
- View/download PDF
10. Abstract 4484: Discovery and in vitro characterization of AMG 510–a potent and selective covalent small-molecule inhibitor of KRASG12C
- Author
-
Pragathi Achanta, J. Russell Lipford, Jude Canon, Brian A. Lanman, Laurie P. Volak, Victor J. Cee, Kevin Gaida, Tisha San Miguel, John D. McCarter, Neelima Koppada, Karen Rex, Anne Y. Saiki, Dhanashri Bagal, and Robert S. Foti
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Cell signaling ,Chemistry ,Wild type ,Context (language use) ,medicine.disease_cause ,Molecular biology ,In vitro ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Phosphorylation ,KRAS - Abstract
Activating mutations in RAS represent the most common oncogenic driver mutation in cancer. The single amino acid substitution of cysteine for glycine at position 12 (KRASG12C) is frequently found in solid malignancies, particularly in lung adenocarcinoma (~13%), colorectal adenocarcinoma (3%), and pancreatic adenocarcinoma (~1%). Recently it has been demonstrated that KRASG12C can be targeted with covalent small molecule inhibitors which react with the mutant cysteine adjacent to the switch II pocket (SIIP), locking KRAS in its inactive GDP-bound state. We describe here the discovery and in vitro characterization of AMG 510, a covalent inhibitor of KRASG12C possessing potent biochemical and cellular activity, as well as robust in vivo efficacy. AMG 510 inhibited SOS1-catalyzed nucleotide exchange of recombinant mutant KRASG12C/C118A but had minimal effect on KRASC118A, which is wildtype at position 12. The observed rate constant (kinact/Ki) of covalent modification of KRASG12C by AMG 510 was determined biochemically by mass spectrometry as well as in the cellular context (kobs/[I]). Cysteine proteome analysis of cells treated with AMG 510 revealed that only the G12C-containing peptide of KRAS was covalently modified. AMG 510 inhibited KRAS signaling as measured by ERK phosphorylation in all KRAS p.G12C cell lines tested, but did not inhibit phosphorylation of ERK in cell lines lacking the KRAS p.G12C mutation. Cellular occupancy of KRASG12C by AMG 510 was determined by mass spectrometry and correlated well with inhibition of ERK phosphorylation. AMG 510 also selectively impaired the viability of KRAS p.G12C mutant lines. Combination treatment of AMG 510 with inhibitors of other cellular signaling pathways exhibited evidence for synergistic effects on cell viability. Treatment of KRAS p.G12C lines with covalent KRASG12C inhibitors increased the expression of HLA. To test the impact of KRASG12C inhibition on immune surveillance in vivo, we generated a syngeneic tumor cell line that is suitable for testing AMG 510 in combination with checkpoint inhibitor therapies and characterized this line in vitro. AMG 510 is currently being evaluated in a Phase I study in patients with solid tumors harboring KRAS p.G12Cmutations. Citation Format: Anne Y. Saiki, Kevin Gaida, Karen Rex, Pragathi Achanta, Tisha San Miguel, Neelima Koppada, Dhanashri Bagal, Brian A. Lanman, Robert S. Foti, John D. McCarter, Laurie P. Volak, Jude Canon, Victor J. Cee, J. Russell Lipford. Discovery and in vitro characterization of AMG 510–a potent and selective covalent small-molecule inhibitor of KRASG12C [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4484.
- Published
- 2019
- Full Text
- View/download PDF
11. Discovery and in Vivo Evaluation of the Potent and Selective PI3Kδ Inhibitors 2-((1S)-1-((6-Amino-5-cyano-4-pyrimidinyl)amino)ethyl)-6-fluoro-N-methyl-3-(2-pyridinyl)-4-quinolinecarboxamide (AM-0687) and 2-((1S)-1-((6-Amino-5-cyano-4-pyrimidinyl)amino)ethyl)-5-fluoro-N-methyl-3-(2-pyridinyl)-4-quinolinecarboxamide (AM-1430)
- Author
-
Felix, Gonzalez-Lopez de Turiso, Xiaolin, Hao, Youngsook, Shin, Minna, Bui, Iain D G, Campuzano, Mario, Cardozo, Michelle C, Dunn, Jason, Duquette, Benjamin, Fisher, Robert S, Foti, Kirk, Henne, Xiao, He, Yi-Ling, Hu, Ron C, Kelly, Michael G, Johnson, Brian S, Lucas, John, McCarter, Lawrence R, McGee, Julio C, Medina, Daniela, Metz, Tisha, San Miguel, Deanna, Mohn, Thuy, Tran, Christine, Vissinga, Sharon, Wannberg, Douglas A, Whittington, John, Whoriskey, Gang, Yu, Leeanne, Zalameda, Xuxia, Zhang, and Timothy D, Cushing
- Subjects
Models, Molecular ,B-Lymphocytes ,Dose-Response Relationship, Drug ,Molecular Structure ,Pyridines ,Class Ia Phosphatidylinositol 3-Kinase ,Mice ,Structure-Activity Relationship ,Drug Discovery ,Quinolines ,Animals ,Humans ,Protein Kinase Inhibitors ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Optimization of the potency and pharmacokinetic profile of 2,3,4-trisubstituted quinoline, 4, led to the discovery of two potent, selective, and orally bioavailable PI3Kδ inhibitors, 6a (AM-0687) and 7 (AM-1430). On the basis of their improved profile, these analogs were selected for in vivo pharmacodynamic (PD) and efficacy experiments in animal models of inflammation. The in vivo PD studies, which were carried out in a mouse pAKT inhibition animal model, confirmed the observed potency of 6a and 7 in biochemical and cellular assays. Efficacy experiments in a keyhole limpet hemocyanin model in rats demonstrated that administration of either 6a or 7 resulted in a strong dose-dependent reduction of IgG and IgM specific antibodies. The excellent in vitro and in vivo profiles of these analogs make them suitable for further development.
- Published
- 2016
12. Synthesis and structure–activity relationships of dual PI3K/mTOR inhibitors based on a 4-amino-6-methyl-1,3,5-triazine sulfonamide scaffold
- Author
-
Yunxin Bo, Tisha San Miguel, Nobuko Nishimura, Jian Jiang, Douglas A. Whittington, Paul E. Hughes, Sean Caenepeel, Mark H. Norman, John D. McCarter, Erin L. Mullady, Daniel J. Freeman, Kevin Yang, Ryan Wurz, Longbin Liu, Raju Subramanian, Kristin L. Andrews, Ling Wang, Liping H. Pettus, and Nancy Zhang
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Phosphorylation ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Sulfonamides ,Binding Sites ,Phosphoinositide 3-kinase ,biology ,Triazines ,Akt/PKB signaling pathway ,Chemistry ,TOR Serine-Threonine Kinases ,Cellular Assay ,Organic Chemistry ,Rats ,Molecular Docking Simulation ,biology.protein ,Molecular Medicine ,Female ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Phosphoinositide 3-kinase (PI3K) is an important target in oncology due to the deregulation of the PI3K/Akt signaling pathway in a wide variety of tumors. A series of 4-amino-6-methyl-1,3,5-triazine sulfonamides were synthesized and evaluated as inhibitors of PI3K. The synthesis, in vitro biological activities, pharmacokinetic and in vivo pharmacodynamic profiling of these compounds are described. The most promising compound from this investigation (compound 3j) was found to be a pan class I PI3K inhibitor with a moderate (>10-fold) selectivity over the mammalian target of rapamycin (mTOR) in the enzyme assay. In a U87 MG cellular assay measuring phosphorylation of Akt, compound 3j displayed low double digit nanomolar IC(50) and exhibited good oral bioavailability in rats (F(oral)=63%). Compound 3j also showed a dose dependent reduction in the phosphorylation of Akt in a U87 tumor pharmacodynamic model with a plasma EC(50)=193 nM (91 ng/mL).
- Published
- 2012
- Full Text
- View/download PDF
13. Selective Class I Phosphoinositide 3-Kinase Inhibitors: Optimization of a Series of Pyridyltriazines Leading to the Identification of a Clinical Candidate, AMG 511
- Author
-
Bradley J. Herberich, Tian Wu, Paul E. Hughes, Claire L. M. Jackson, Nobuko Nishimura, Nancy Zhang, Ryan Wurz, Douglas A. Whittington, Shon Booker, Sean Caenepeel, Tisha San Miguel, Jian Jiang, Anthony B. Reed, Adrian L. Smith, Seifu Tadesse, Brian A. Lanman, Andrew Tasker, Erin L. Mullady, John D. McCarter, Fang-Tsao Hong, Liping H. Pettus, Divesh Aidasani, Victor J. Cee, Ling Wang, Bin Wu, Mark H. Norman, Daniel J. Freeman, Kevin Yang, Longbin Liu, Leeanne Zalameda, Yunxin Y. Bo, Xiaochun Zhu, Kristin L. Andrews, Markian Stec, Nuria A. Tamayo, Raju Subramanian, and Noel D'angelo
- Subjects
Models, Molecular ,Phosphoinositide 3-kinase ,biology ,Triazines ,Chemistry ,Cell growth ,Pharmacology ,Crystallography, X-Ray ,medicine.disease ,chemistry.chemical_compound ,In vivo ,Glioma ,Drug Discovery ,Second messenger system ,medicine ,biology.protein ,Molecular Medicine ,Phosphorylation ,Phosphatidylinositol ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors - Abstract
The phosphoinositide 3-kinase family catalyzes the phosphorylation of phosphatidylinositol-4,5-diphosphate to phosphatidylinositol-3,4,5-triphosphate, a secondary messenger which plays a critical role in important cellular functions such as metabolism, cell growth, and cell survival. Our efforts to identify potent, efficacious, and orally available phosphatidylinositol 3-kinase (PI3K) inhibitors as potential cancer therapeutics have resulted in the discovery of 4-(2-((6-methoxypyridin-3-yl)amino)-5-((4-(methylsulfonyl)piperazin-1-yl)methyl)pyridin-3-yl)-6-methyl-1,3,5-triazin-2-amine (1). In this paper, we describe the optimization of compound 1, which led to the design and synthesis of pyridyltriazine 31, a potent pan inhibitor of class I PI3Ks with a superior pharmacokinetic profile. Compound 31 was shown to potently block the targeted PI3K pathway in a mouse liver pharmacodynamic model and inhibit tumor growth in a U87 malignant glioma glioblastoma xenograft model. On the basis of its excellent in vivo efficacy and pharmacokinetic profile, compound 31 was selected for further evaluation as a clinical candidate and was designated AMG 511.
- Published
- 2012
- Full Text
- View/download PDF
14. Structure-Based Design of a Novel Series of Potent, Selective Inhibitors of the Class I Phosphatidylinositol 3-Kinases
- Author
-
Yunxin Y. Bo, Leeanne Zalameda, Kevin Yang, Fang-Tsao Hong, Longbin Liu, Anthony B. Reed, Kristin L. Andrews, Tisha San Miguel, Brian A. Lanman, Paul E. Hughes, Noel D'angelo, Brad Herberich, Jian Jiang, Victor J. Cee, Claire L. M. Jackson, Ryan Wurz, Nobuko Nishimura, Raju Subramanian, Seifu Tadesse, Nancy Zhang, John D. McCarter, Daniel J. Freeman, Ling Wang, Nuria A. Tamayo, Erin L. Mullady, Mark H. Norman, Liping H. Pettus, Tian Wu, Douglas A. Whittington, Sean Caenepeel, Adrian L. Smith, and Shon Booker
- Subjects
Models, Molecular ,Indazoles ,Class I Phosphatidylinositol 3-Kinases ,Pyridines ,Biological Availability ,Mice, Nude ,Crystallography, X-Ray ,Piperazines ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Sulfones ,Phosphatidylinositol ,PI3K/AKT/mTOR pathway ,Sulfonamides ,Triazines ,Akt/PKB signaling pathway ,Chemistry ,Kinase ,TOR Serine-Threonine Kinases ,Xenograft Model Antitumor Assays ,Rats ,Cell biology ,Pyrimidines ,Biochemistry ,Purines ,Drug Design ,Microsomes, Liver ,Pyrazoles ,Molecular Medicine ,Female ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction ,Phosphoinositide-dependent kinase-1 - Abstract
A highly selective series of inhibitors of the class I phosphatidylinositol 3-kinases (PI3Ks) has been designed and synthesized. Starting from the dual PI3K/mTOR inhibitor 5, a structure-based approach was used to improve potency and selectivity, resulting in the identification of 54 as a potent inhibitor of the class I PI3Ks with excellent selectivity over mTOR, related phosphatidylinositol kinases, and a broad panel of protein kinases. Compound 54 demonstrated a robust PD-PK relationship inhibiting the PI3K/Akt pathway in vivo in a mouse model, and it potently inhibited tumor growth in a U-87 MG xenograft model with an activated PI3K/Akt pathway.
- Published
- 2012
- Full Text
- View/download PDF
15. A Potent and Orally Efficacious, Hydroxyethylamine-Based Inhibitor of β-Secretase
- Author
-
Kui Chen, Daniel La, Michael D. Bartberger, Russell Graceffa, Thomas T. Nguyen, Tisha San Miguel, E. Allen Sickmier, Ryan Brown, Vivian S. W. Li, Stephen J. Wood, Lewis D. Pennington, James Brown, Jianhua Zhang, Craig E. Masse, Holger Monenschein, Patricia Lopez, Dean Hickman, Brian K. Albrecht, May Xue, Michael Croghan, Scott Harried, Safura Babu-Khan, Yuan Cheng, Yi Luo, Martin Citron, Matthew R. Kaller, Patricia Amarante, Wenge Zhong, Paul H. Wen, Bryant Yang, Robert C. Wahl, Matthew Weiss, Chuck Kriemen, Stephen Hitchcock, Vinod F. Patel, Ted Judd, and Toni Williamson
- Subjects
chemistry.chemical_classification ,Enzyme ,biology ,Chemistry ,Cerebral Spinal Fluid ,Organic Chemistry ,Drug Discovery ,β secretase ,Amyloid precursor protein ,biology.protein ,Potency ,Pharmacology ,Biochemistry - Abstract
β-Secretase inhibitors are potentially disease-modifying treatments for Alzheimer's disease. Previous efforts in our laboratory have resulted in hydroxyethylamine-derived inhibitors such as 1 with low nanomolar potency against β-site amyloid precursor protein cleaving enzyme (BACE). When dosed intravenously, compound 1 was also shown to significantly reduce Aβ40 levels in plasma, brain, and cerebral spinal fluid. Herein, we report further optimizations that led to the discovery of inhibitor 16 as a novel, potent, and orally efficacious BACE inhibitor.
- Published
- 2012
- Full Text
- View/download PDF
16. Design and Preparation of a Potent Series of Hydroxyethylamine Containing β-Secretase Inhibitors That Demonstrate Robust Reduction of Central β-Amyloid
- Author
-
Yuan Cheng, Matthew R. Kaller, Steven W. Louie, Martin Citron, Lewis D. Pennington, Daniel S. La, Matthew Weiss, Stephen J. Wood, Wenge Zhong, Michael Croghan, Toni Williamson, Scott Harried, James Brown, Kui Chen, Tisha San Miguel, Ronke Imbeah-Ampiah, Holger Monenschein, Qiufen Xue, Hongbing Huang, Patricia Lopez, Claire Rattan, Thomas Dineen, Daniel B. Horne, E. Allen Sickmier, Dean Hickman, Safura Babu-Khan, Charles Kreiman, Vivian S. W. Li, Paul H. Wen, Michael D. Bartberger, Russell Graceffa, Robert C. Wahl, Thomas T. Nguyen, Bryant Yang, Ted Judd, Joel Esmay, Vinod F. Patel, and Stephen Hitchcock
- Subjects
Male ,Models, Molecular ,Protein Conformation ,Administration, Oral ,Biological Availability ,Stereoisomerism ,Pharmacology ,Crystallography, X-Ray ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Dogs ,Pharmacokinetics ,Oral administration ,Drug Discovery ,Ethylamines ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Structure–activity relationship ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,chemistry.chemical_classification ,Amyloid beta-Peptides ,Brain ,Dioxolanes ,Macaca mulatta ,Peptide Fragments ,Rats ,Bioavailability ,Protein Transport ,Enzyme ,chemistry ,Biochemistry ,Drug Design ,Microsomes, Liver ,Microsome ,Molecular Medicine ,Efflux ,Amyloid Precursor Protein Secretases - Abstract
A series of potent hydroxyethyl amine (HEA) derived inhibitors of β-site APP cleaving enzyme (BACE1) was optimized to address suboptimal pharmacokinetics and poor CNS partitioning. This work identified a series of benzodioxolane analogues that possessed improved metabolic stability and increased oral bioavailability. Subsequent efforts focused on improving CNS exposure by limiting susceptibility to Pgp-mediated efflux and identified an inhibitor which demonstrated robust and sustained reduction of CNS β-amyloid (Aβ) in Sprague-Dawley rats following oral administration.
- Published
- 2012
- Full Text
- View/download PDF
17. Design and Synthesis of Potent, Orally Efficacious Hydroxyethylamine Derived β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE1) Inhibitors
- Author
-
Paul H. Wen, Robert C. Wahl, Michael Croghan, Ronke Imbeah-Ampiah, Paul Acton, Stephen J. Wood, Charles Kreiman, Vivian S. W. Li, Qiufen Xue, Lewis D. Pennington, Safura Babu-Khan, Daniel B. Horne, Thomas Dineen, Wenge Zhong, Scott Harried, Douglas A. Whittington, Dean Hickman, James Brown, Martin Citron, Matthew Weiss, Daniel S. La, Holger Monenschein, Bryant Yang, E. Allen Sickmier, Tisha San Miguel, Robert T. Dunn, Russell Graceffa, Patricia Lopez, Toni Williamson, Hongbing Huang, Yuan Cheng, Kui Chen, Matthew R. Kaller, Michael D. Bartberger, Steven W. Louie, Hugo M. Vargas, Stephen Hitchcock, Thomas T. Nguyen, Vinod F. Patel, Ted Judd, and Joel Esmay
- Subjects
Male ,Models, Molecular ,Protein Conformation ,Swine ,Administration, Oral ,Peptide ,In Vitro Techniques ,Pharmacology ,Crystallography, X-Ray ,Inhibitory postsynaptic potential ,Cell Line ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,Dogs ,Pharmacokinetics ,mental disorders ,Drug Discovery ,Amyloid precursor protein ,Animals ,Aspartic Acid Endopeptidases ,Cytochrome P-450 CYP3A ,Cytochrome P-450 Enzyme Inhibitors ,Humans ,Spiro Compounds ,Dosing ,chemistry.chemical_classification ,Amyloid beta-Peptides ,Cardiovascular safety ,biology ,Brain ,Stereoisomerism ,Blood Proteins ,Peptide Fragments ,Rats ,Bioavailability ,Thiazoles ,Enzyme ,Biochemistry ,chemistry ,Drug Design ,Microsomes, Liver ,biology.protein ,Molecular Medicine ,Amyloid Precursor Protein Secretases ,Protein Binding - Abstract
We have previously shown that hydroxyethylamines can be potent inhibitors of the BACE1 enzyme and that the generation of BACE1 inhibitors with CYP 3A4 inhibitory activities in this scaffold affords compounds (e.g., 1) with sufficient bioavailability and pharmacokinetic profiles to reduce central amyloid-β peptide (Aβ) levels in wild-type rats following oral dosing. In this article, we describe further modifications of the P1-phenyl ring of the hydroxyethylamine series to afford potent, dual BACE1/CYP 3A4 inhibitors which demonstrate improved penetration into the CNS. Several of these compounds caused robust reduction of Aβ levels in rat CSF and brain following oral dosing, and compound 37 exhibited an improved cardiovascular safety profile relative to 1.
- Published
- 2012
- Full Text
- View/download PDF
18. Structure–Activity Relationships of Phosphoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors: Investigations of Various 6,5-Heterocycles to Improve Metabolic Stability
- Author
-
Kristin L. Andrews, Kevin Yang, Markian Stec, Tisha San Miguel, Ling Wang, John McCarter, Mark H. Norman, Shon Booker, Jian Jiang, Nan Zhang, Daniel J. Freeman, Erin L. Mullady, Hongyu Liao, Paul E. Hughes, Leeanne Zalameda, Sean Caenepeel, Raju Subramanian, and Nuria A. Tamayo
- Subjects
Models, Molecular ,Pyridines ,Metabolite ,Transplantation, Heterologous ,Mice, Nude ,Antineoplastic Agents ,Pharmacology ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Oxazoles ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Sulfonamides ,Phosphoinositide 3-kinase ,biology ,Chemistry ,TOR Serine-Threonine Kinases ,Imidazoles ,In vitro ,Rats ,Pyridazines ,Thiazoles ,medicine.anatomical_structure ,Biochemistry ,Benzothiazole ,Acetylation ,Hepatocyte ,Hepatocytes ,biology.protein ,Molecular Medicine ,Female ,Drug Screening Assays, Antitumor ,Neoplasm Transplantation - Abstract
N-(6-(6-Chloro-5-(4-fluorophenylsulfonamido)pyridin-3-yl)benzo[d]thiazol-2-yl)acetamide (1) is a potent and efficacious inhibitor of PI3Kα and mTOR in vitro and in vivo. However, in hepatocyte and in vivo metabolism studies, 1 was found to undergo deacetylation on the 2-amino substituent of the benzothiazole. As an approach to reduce or eliminate this metabolic deacetylation, a variety of 6,5-heterocyclic analogues were examined as an alternative to the benzothiazole ring. Imidazopyridazine 10 was found to have similar in vitro potency and in vivo efficacy relative to 1, while only minimal amounts of the corresponding deacetylated metabolite of 10 were observed in hepatocytes.
- Published
- 2011
- Full Text
- View/download PDF
19. Phospshoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors: Discovery and Structure–Activity Relationships of a Series of Quinoline and Quinoxaline Derivatives
- Author
-
Ling Wang, Leeanne Zalameda, Hongyu Liao, Kristin L. Andrews, Yunxin Bo, Nobuko Nishimura, Paul E. Hughes, John D. McCarter, Erin L. Mullady, Tisha San Miguel, Douglas A. Whittington, Sean Caenepeel, Nancy Zhang, Daniel J. Freeman, Mark H. Norman, Shon Booker, Aaron C. Siegmund, Nuria A. Tamayo, Marian C. Bryan, Raju Subramanian, Kevin Yang, and Longbin Liu
- Subjects
Male ,Models, Molecular ,Protein Conformation ,Biological Availability ,In Vitro Techniques ,Pharmacology ,Crystallography, X-Ray ,PI3K signaling ,Rats, Sprague-Dawley ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,chemistry.chemical_compound ,Quinoxaline ,In vivo ,Quinoxalines ,Drug Discovery ,medicine ,Animals ,Humans ,Phosphorylation ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Kinase ,Chemistry ,TOR Serine-Threonine Kinases ,Quinoline ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,Liver ,Quinolines ,Molecular Medicine ,Protein Binding - Abstract
The phosphoinositide 3-kinase (PI3K) family catalyzes the ATP-dependent phosphorylation of the 3'-hydroxyl group of phosphatidylinositols and plays an important role in cell growth and survival. There is abundant evidence demonstrating that PI3K signaling is dysregulated in many human cancers, suggesting that therapeutics targeting the PI3K pathway may have utility for the treatment of cancer. Our efforts to identify potent, efficacious, and orally available PI3K/mammalian target of rapamycin (mTOR) dual inhibitors resulted in the discovery of a series of substituted quinolines and quinoxalines derivatives. In this report, we describe the structure-activity relationships, selectivity, and pharmacokinetic data of this series and illustrate the in vivo pharmacodynamic and efficacy data for a representative compound.
- Published
- 2011
- Full Text
- View/download PDF
20. Discovery, Optimization, and in Vivo Evaluation of Benzimidazole Derivatives AM-8508 and AM-9635 as Potent and Selective PI3Kδ Inhibitors
- Author
-
Yi-Ling Hu, Leeanne Zalameda, Daniela Metz, Julia Suchomel, Thuy B. Tran, Gang Yu, Jason Duquette, John McCarter, Xuxia Zhang, Youngsook Shin, Simon Wong, Julio C. Medina, Sharon Wannberg, Timothy D. Cushing, Tisha San Miguel, Mario G. Cardozo, Lawrence R. McGee, Ron C. Kelly, Christine Vissinga, Deanna Mohn, John Whoriskey, Douglas A. Whittington, Kirk Henne, and Xiao He
- Subjects
0301 basic medicine ,Models, Molecular ,Benzimidazole ,B-cell receptor ,chemical and pharmacologic phenomena ,Crystallography, X-Ray ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Pharmacokinetics ,In vivo ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,Protein Kinase Inhibitors ,Phosphoinositide-3 Kinase Inhibitors ,B-Lymphocytes ,biology ,Chemistry ,In vitro ,Bioavailability ,Rats ,030104 developmental biology ,Biochemistry ,Immunoglobulin M ,Immunoglobulin G ,Hemocyanins ,biology.protein ,Molecular Medicine ,Benzimidazoles ,Keyhole limpet hemocyanin - Abstract
Lead optimization efforts resulted in the discovery of two potent, selective, and orally bioavailable PI3Kδ inhibitors, 1 (AM-8508) and 2 (AM-9635), with good pharmacokinetic properties. The compounds inhibit B cell receptor (BCR)-mediated AKT phosphorylation (pAKT) in PI3Kδ-dependent in vitro cell based assays. These compounds which share a benzimidazole bicycle are effective when administered in vivo at unbound concentrations consistent with their in vitro cell potency as a consequence of improved unbound drug concentration with lower unbound clearance. Furthermore, the compounds demonstrated efficacy in a Keyhole Limpet Hemocyanin (KLH) study in rats, where the blockade of PI3Kδ activity by inbibitors 1 and 2 led to effective inhibition of antigen-specific IgG and IgM formation after immunization with KLH.
- Published
- 2015
21. High-Throughput Mass Spectrometric Analysis of Covalent Protein-Inhibitor Adducts for the Discovery of Irreversible Inhibitors: A Complete Workflow
- Author
-
Todd Rowe, Tara Arvedson, John D. McCarter, Iain D. G. Campuzano, Tisha San Miguel, Victor J. Cee, and Daniel Onea
- Subjects
0301 basic medicine ,Mass spectrometry ,01 natural sciences ,Biochemistry ,Mass Spectrometry ,Analytical Chemistry ,Adduct ,03 medical and health sciences ,High-Throughput Screening Assays ,Solid phase extraction ,Cysteine ,Acrylamide ,Chemistry ,Lysine ,Solid Phase Extraction ,Proteins ,Combinatorial chemistry ,Fluorescence ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,Covalent bond ,Molecular Medicine ,Time-of-flight mass spectrometry ,Biotechnology - Abstract
We have implemented a solid-phase extraction based time-of-flight mass spectrometer system in combination with novel informatics to rapidly screen and characterize the covalent binding of different irreversible inhibitors to intact proteins. This high-throughput screening platform can be used to accurately detect and quantitate the extent of formation of different covalent protein-inhibitor adducts between electrophilic inhibitors and nucleophilic residues such as cysteine or lysine. For a representative 19.5 kDa protein, the analysis time is approximately 20 s per sample, including an efficient sample loading and desalting step. Accurate protein masses are measured (±0.5 amu of the theoretical molecular weight; measured precision of ±0.02 amu). The fraction of protein reacted with an electrophilic compound is determined relative to an unmodified protein control. A key element of the workflow is the automated identification and quantitation of the expected masses of covalent protein-inhibitor adducts using a custom routine that obviates the need to manually inspect each individual spectrum. Parallel screens were performed on a library of approximately 1000 acrylamide containing compounds (different structures and reactivities) using the solid-phase extraction mass spectrometry based assay and a fluorescence based thiol-reactive probe assay enabling comparison of false positives and false negatives between these orthogonal screening approaches.
- Published
- 2015
22. A telomerase enzymatic assay that does not use polymerase chain reaction, radioactivity, or electrophoresis
- Author
-
Wen Zhou, Murray O. Robinson, Hue Kha, Robert C. Wahl, Brian Rasnow, Karen Kearns, Tisha San Miguel, Alex Mladenovic, Barbara Karan-Tamir, Lisa Zeni, and Kui Chen
- Subjects
Electrophoresis ,Radioisotopes ,Detection limit ,Telomerase ,Lysis ,Base Sequence ,Biophysics ,Enzyme-Linked Immunosorbent Assay ,Cell Biology ,Biology ,Polymerase Chain Reaction ,Biochemistry ,Molecular biology ,Cell Line ,law.invention ,Telomere ,law ,Biotinylation ,Humans ,Primer (molecular biology) ,Molecular Biology ,Polymerase chain reaction ,DNA Primers ,Chemiluminescence - Abstract
A telomerase assay has been developed for high-throughput screening in 96-well microtiter plates. A crude cell lysate which adds telomere repeats to a biotinylated DNA primer is the source of telomerase. The telomerase-extended primer is hybridized to a digoxigenin-labeled telomere antisense DNA probe. The hybrid is further processed by enzyme-linked immunosorbent assay (ELISA) as follows. The biotinylated hybrid is captured on streptavidin-coated microtiter plates. The immobilized hybrid is probed with alkaline phosphatase-antidigoxigenin and detected via chemiluminescent readout. The limit of detection of a chemically synthesized tetra-telomere repeat was about 10 attomoles. Apparent telomerase activity was detected in lysates of 293T cells. The signal to background for the assay (ratio of signal for the complete assay mixture divided by the signal for the assay mixture without primer) was around 10. An automated system that performed unattended runs of up to 17 96-well microtiter plates in 8h was constructed.
- Published
- 2004
- Full Text
- View/download PDF
23. The optimization of aminooxadiazoles as orally active inhibitors of Cdc7
- Author
-
Alexander J. Pickrell, Tisha San Miguel, Kelvin K. C. Sham, Christopher H. Fotsch, Andreas Reichelt, Leeanne Zalameda, Matthew P. Bourbeau, Aaron C. Siegmund, John G. Allen, Heller Scott Francis, Sonia Escobar, Julie M. Bailis, Paul E. Harrington, Tammy L. Bush, Jessica Orf, Helming Tan, Faye Hsieh, Michael J. Frohn, Minqing Rong, and Dean Hickman
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Mice, Nude ,Cell Cycle Proteins ,Pharmacology ,Protein Serine-Threonine Kinases ,Biochemistry ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Pharmacokinetics ,Drug Discovery ,Animals ,Isoquinoline ,Molecular Biology ,IC50 ,Cell potency ,Protein Kinase Inhibitors ,Oxadiazoles ,Molecular Structure ,Kinase ,Organic Chemistry ,Glutathione ,Xenograft Model Antitumor Assays ,Rats ,Disease Models, Animal ,chemistry ,Pharmacodynamics ,Molecular Medicine ,Female ,Selectivity - Abstract
A series of aminooxadiazoles was optimized for inhibition of Cdc7. Early lead isoquinoline 1 suffered from modest cell potency (cellular IC50=0.71 μM measuring pMCM2), low selectivity against structurally related kinases, and high IV clearance in rats (CL=18 L/h/kg). Extensive optimization resulted in azaindole 26 (Cdc7 IC50=1.1 nM, pMCM2 IC50=32 nM) that demonstrated robust lowering of pMCM2 in a mouse pharmacodynamic (PD) model when dosed orally. Modifications to improve the pharmacokinetic profile of this series were guided by trapping experiments with glutathione in rat hepatocytes.
- Published
- 2013
24. N-substituted azaindoles as potent inhibitors of Cdc7 kinase
- Author
-
John G. Allen, Andreas Reichelt, Guomin Yao, Julie M. Bailis, Elizabeth M. Doherty, James R. Falsey, Tisha San Miguel, Leeanne Zalameda, Marian C. Bryan, Michael D. Bartberger, and Mike Frohn
- Subjects
Indoles ,Stereochemistry ,Clinical Biochemistry ,Cancer therapy ,Pharmaceutical Science ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biochemistry ,Structure-Activity Relationship ,Drug Discovery ,Structure–activity relationship ,Molecule ,Animals ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,Indole test ,Aza Compounds ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Kinase ,Organic Chemistry ,DNA replication ,Metabolic stability ,Rats ,Binding conformation ,Molecular Medicine - Abstract
Cdc7 kinase is responsible for the initiation and regulation of DNA replication and has been proposed as a target for cancer therapy. We have identified a class of Cdc7 inhibitors based on a substituted indole core. Synthesis of focused indole and azaindole analogs yielded potent and selective 5-azaindole Cdc7 inhibitors with improved intrinsic metabolic stability (ie 36). In parallel, quantum mechanical conformational analysis helped to rationalize SAR observations, led to a proposal of the preferred binding conformation in the absence of co-crystallography data, and allowed the design of 7-azaindole 37 as a second lead in this series.
- Published
- 2012
25. Discovery and optimization of a series of benzothiazole phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) dual inhibitors
- Author
-
Yang Xu, Leeanne Zalameda, Raju Subramanian, Erin L. Mullady, Michael Schrag, Kevin Yang, Daniel J. Freeman, Jian Jiang, Longbin Liu, Tian Wu, Derin C. D'amico, John D. McCarter, Noel D'angelo, Sean Caenepeel, Peter Yakowec, Ling Wang, Mark H. Norman, Robert C. Wahl, Tisha San Miguel, Jin Tang, Douglas A. Whittington, Paul E. Hughes, Shon Booker, Ning Xi, Tae-Seong Kim, Kui Chen, Nancy Zhang, and Kristin L. Andrews
- Subjects
Models, Molecular ,Transplantation, Heterologous ,Biological Availability ,Mice, Nude ,Antineoplastic Agents ,Crystallography, X-Ray ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,Benzothiazoles ,Phosphorylation ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Sulfonamides ,Phosphoinositide 3-kinase ,Binding Sites ,biology ,Drug discovery ,Chemistry ,Kinase ,TOR Serine-Threonine Kinases ,In vitro ,Rats ,Transplantation ,Biochemistry ,Benzothiazole ,Liver ,biology.protein ,Molecular Medicine ,Female ,Drug Screening Assays, Antitumor ,Proto-Oncogene Proteins c-akt ,Neoplasm Transplantation - Abstract
Phosphoinositide 3-kinase α (PI3Kα) is a lipid kinase that plays a key regulatory role in several cellular processes. The mutation or amplification of this kinase in humans has been implicated in the growth of multiple tumor types. Consequently, PI3Kα has become a target of intense research for drug discovery. Our studies began with the identification of benzothiazole compound 1 from a high throughput screen. Extensive SAR studies led to the discovery of sulfonamide 45 as an early lead, based on its in vitro cellular potency. Subsequent modifications of the central pyrimidine ring dramatically improved enzyme and cellular potency and led to the identification of chloropyridine 70. Further arylsulfonamide SAR studies optimized in vitro clearance and led to the identification of 82 as a potent dual inhibitor of PI3K and mTOR. This molecule exhibited potent enzyme and cell activity, low clearance, and high oral bioavailability. In addition, compound 82 demonstrated tumor growth inhibition in U-87 MG, A549, and HCT116 tumor xenograft models.
- Published
- 2011
26. High capacity homogeneous non-radioactive cortisol detection assays for human 11beta-hydroxysteroid dehydrogenase type 1
- Author
-
Matthew H. Plant, Rebecca Nybo, Tisha San Miguel, Clarence Hale, Minghan Wang, Jamie Baumgartner, Yanyan Tudor, Violeta Yu, David Powers, Robert J.M. Kurzeja, Michelle Chen, Yen Nguyen, and Ki Won Kim
- Subjects
Radioisotope Dilution Technique ,Hydrocortisone ,Dehydrogenase ,High-performance liquid chromatography ,Sensitivity and Specificity ,11β-hydroxysteroid dehydrogenase type 1 ,Drug Discovery ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,medicine ,Electrochemistry ,Fluorescence Resonance Energy Transfer ,Humans ,biology ,Chemistry ,Microchemistry ,Reproducibility of Results ,Thin-layer chromatography ,Förster resonance energy transfer ,Biochemistry ,Luminescent Measurements ,biology.protein ,Molecular Medicine ,Cortisone ,hormones, hormone substitutes, and hormone antagonists ,Glucocorticoid ,medicine.drug - Abstract
11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes the interconversion of inert glucocorticoid (cortisone) to the active glucocorticoid (cortisol) and is enriched in liver and fat tissues. Increasing evidence suggests that selective inhibition of 11beta-HSD1 may reduce the excess glucocorticoid levels that underlie the etiology of many common disorders that constitute the metabolic syndrome. Measurement of 11beta-HSD1 activity has historically involved the detection of cortisol by methods unfavorable for large-scale screening, such as high performance liquid chromatography or thin layer chromatography. Here we describe the development and validation of novel homogeneous time-resolved fluorescence resonance energy transfer (TR-FRET) and electrochemiluminescence assays for the measurement of cortisol. These non-radioactive assays were easy to perform and produced robust results with reference compound values comparable to those obtained by conventional methods. The TR-FRET assay was easily automated and was successfully employed for the high-throughput screening of a large compound library for inhibitors of purified human recombinant 11beta-HSD1.
- Published
- 2007
27. Correction to Selective Class I Phosphoinositide 3-Kinase Inhibitors: Optimization of a Series of Pyridyltriazines Leading to the Identification of a Clinical Candidate, AMG 511
- Author
-
Mark H. Norman, Kristin L. Andrews, Yunxin Y. Bo, Shon K. Booker, Sean Caenepeel, Victor J. Cee, Noel D. D’Angelo, Daniel J. Freeman, Bradley J. Herberich, Fang-Tsao Hong, Claire L. M. Jackson, Jian Jiang, Brian A. Lanman, Longbin Liu, John D. McCarter, Erin L. Mullady, Nobuko Nishimura, Liping H. Pettus, Anthony B. Reed, Tisha San Miguel, Adrian L. Smith, Markian M. Stec, Seifu Tadesse, Andrew Tasker, Divesh Aidasani, Xiaochun Zhu, Raju Subramanian, Nuria A. Tamayo, Ling Wang, Douglas A. Whittington, Bin Wu, Tian Wu, Ryan P. Wurz, Kevin Yang, Leeanne Zalameda, Nancy Zhang, and Paul E. Hughes
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2012
- Full Text
- View/download PDF
28. Abstract 253: Identification of STK33 kinase inhibitors for the validation of a synthetic lethal relationship between STK33 and mutant KRAS
- Author
-
Anke Munzli, Ted Judd, Manory Fernando, Carol Babij, Yihong Zhang, James Zondlo, John C. Robinson, Tisha San Miguel, Ralf Schwandner, John McCarter, Robert J.M. Kurzeja, Isabelle Dussault, and Josette Carnahan
- Subjects
Genetics ,Cancer Research ,Oncology ,Kinase ,Mutant ,medicine ,Identification (biology) ,KRAS ,Biology ,medicine.disease_cause - Abstract
The kinase activity of the serine/threonine kinase STK33 was recently shown to be required for the survival of cancer cell lines dependent on the KRAS oncogene. This suggests that an STK33 kinase inhibitor could be useful to treat cancer patients whose tumors harbor KRAS mutations. Our goal was to identify STK33 kinase inhibitors to explore the role of this kinase in mutant KRAS cancer cell lines. First, full-length wild-type or kinase dead (KD) STK33 proteins were prepared using a baculovirus/insect expression system and purified by chromatography. Preliminary experiments identified two peptides, CREBtide and p70S6K peptides, as potential substrates for STK33. Both peptides were therefore used to track STK33 kinase activity during purification steps. We found that phosphorylation of the p70S6K peptide correlated well with STK33 presence during purification, but the peptide was not phosphorylated by the kinase dead STK33. In contrast, CREBtide phosphorylation showed a poor correlation with STK33 positive fractions and also occurred in the presence of a kinase dead STK33. The p70S6K peptide was thus chosen as the substrate for final optimization of an STK33 kinase assay. Over 400 000 compounds were screened using a high throughput STK33 kinase assay. Multiple hits were identified and IC50s were determined for the top 1043 compounds. Multiple potent STK33 kinase inhibitors were identified, including 69 compounds with IC50s 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 253. doi:10.1158/1538-7445.AM2011-253
- Published
- 2011
- Full Text
- View/download PDF
29. Structure–Activity Relationships of Phosphoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors: Investigations of Various 6,5-Heterocycles to Improve Metabolic Stability.
- Author
-
Markian M. Stec, Kristin L. Andrews, Shon K. Booker, Sean Caenepeel, Daniel J. Freeman, Jian Jiang, Hongyu Liao, John McCarter, Erin L. Mullady, Tisha San Miguel, Raju Subramanian, Nuria Tamayo, Ling Wang, Kevin Yang, Leeanne P. Zalameda, Nancy Zhang, Paul E. Hughes, and Mark H. Norman
- Published
- 2011
- Full Text
- View/download PDF
30. Discovery and Optimization of a Series of Benzothiazole Phosphoinositide 3-Kinase (PI3K)/Mammalian Target of Rapamycin (mTOR) Dual Inhibitors.
- Author
-
Noel D. D’Angelo, Tae-Seong Kim, Kristin Andrews, Shon K. Booker, Sean Caenepeel, Kui Chen, Derin D’Amico, Dan Freeman, Jian Jiang, Longbin Liu, John D. McCarter, Tisha San Miguel, Erin L. Mullady, Michael Schrag, Raju Subramanian, Jin Tang, Robert C. Wahl, Ling Wang, Douglas A. Whittington, and Tian Wu
- Published
- 2011
- Full Text
- View/download PDF
31. High Capacity Homogeneous Non-Radioactive Cortisol Detection Assays for Human 11β-Hydroxysteroid Dehydrogenase Type 1.
- Author
-
Violeta Yu, Yanyan Tudor, Clarence Hale, Matthew Plant, Ki Won Kim, Minghan Wang, Yen Nguyen, Tisha San Miguel, Michelle Chen, Rebecca Nybo, Jamie Baumgartner, Robert J.M. Kurzeja, and David Powers
- Subjects
HYDROCORTISONE ,ADRENOCORTICAL hormones ,DEHYDROGENASES ,CHEMICAL inhibitors ,CHROMATOGRAPHIC analysis ,ETIOLOGY of diseases - Abstract
Abstract11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) catalyzes the interconversion of inert glucocorticoid (cortisone) to the active glucocorticoid (cortisol) and is enriched in liver and fat tissues. Increasing evidence suggests that selective inhibition of 11β-HSD1 may reduce the excess glucocorticoid levels that underlie the etiology of many common disorders that constitute the metabolic syndrome. Measurement of 11β-HSD1 activity has historically involved the detection of cortisol by methods unfavorable for large-scale screening, such as high performance liquid chromatography or thin layer chromatography. Here we describe the development and validation of novel homogeneous time-resolved fluorescence resonance energy transfer (TR-FRET) and electrochemiluminescence assays for the measurement of cortisol. These non-radioactive assays were easy to perform and produced robust results with reference compound values comparable to those obtained by conventional methods. The TR-FRET assay was easily automated and was successfully employed for the high-throughput screening of a large compound library for inhibitors of purified human recombinant 11β-HSD1. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.