101 results on '"Scott A, Biller"'
Search Results
2. Structure and inhibition mechanism of the catalytic domain of human squalene epoxidase
- Author
-
Anil K. Padyana, Stefan Gross, Lei Jin, Giovanni Cianchetta, Rohini Narayanaswamy, Feng Wang, Rui Wang, Cheng Fang, Xiaobing Lv, Scott A. Biller, Lenny Dang, Christopher E. Mahoney, Nelamangala Nagaraja, David Pirman, Zhihua Sui, Janeta Popovici-Muller, and Gromoslaw A. Smolen
- Subjects
Science - Abstract
Squalene epoxidase (SQLE) is a key enzyme in cholesterol biosynthesis and is a target for hypercholesteremia and cancer drug development. Here the authors present the crystal structures of the human SQLE catalytic domain alone and bound with small molecule inhibitors, which will facilitate the development of next-generation SQLE inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
3. Supplementary Data Table S3 from Selective Vulnerability to Pyrimidine Starvation in Hematologic Malignancies Revealed by AG-636, a Novel Clinical-Stage Inhibitor of Dihydroorotate Dehydrogenase
- Author
-
Danielle B. Ulanet, Kevin M. Marks, Josh Murtie, Scott A. Biller, Jonathan Hurov, Georg Lenz, Lenny Dang, Nelamangala Nagaraja, Zhihua Sui, Sreevalsam Gopinath, Thomas Antony, Sunil K. Panigrahi, K. Satish Reddy, Hosahalli Subramanya, Siva Sanjeeva Rao, Kavitha Nellore, Mark Fletcher, Sebastian Hayes, Alan Mann, Tabea Erdmann, Zi-Peng Fan, Charles Locuson, Sebastien Ronseaux, Lei Jin, Anil K. Padyana, Erin Artin, Mya Steadman, Sung Choe, Rohini Narayanaswamy, Kevin Truskowski, John Coco, Victor Chubukov, and Gabrielle McDonald
- Abstract
GO analysis of proteins with at least a 1.5-fold increase in expression in the presence of AG-636 compared to DMSO
- Published
- 2023
4. Supplementary Data from Selective Vulnerability to Pyrimidine Starvation in Hematologic Malignancies Revealed by AG-636, a Novel Clinical-Stage Inhibitor of Dihydroorotate Dehydrogenase
- Author
-
Danielle B. Ulanet, Kevin M. Marks, Josh Murtie, Scott A. Biller, Jonathan Hurov, Georg Lenz, Lenny Dang, Nelamangala Nagaraja, Zhihua Sui, Sreevalsam Gopinath, Thomas Antony, Sunil K. Panigrahi, K. Satish Reddy, Hosahalli Subramanya, Siva Sanjeeva Rao, Kavitha Nellore, Mark Fletcher, Sebastian Hayes, Alan Mann, Tabea Erdmann, Zi-Peng Fan, Charles Locuson, Sebastien Ronseaux, Lei Jin, Anil K. Padyana, Erin Artin, Mya Steadman, Sung Choe, Rohini Narayanaswamy, Kevin Truskowski, John Coco, Victor Chubukov, and Gabrielle McDonald
- Abstract
Supplementary methods, references and figures
- Published
- 2023
5. Data from Selective Vulnerability to Pyrimidine Starvation in Hematologic Malignancies Revealed by AG-636, a Novel Clinical-Stage Inhibitor of Dihydroorotate Dehydrogenase
- Author
-
Danielle B. Ulanet, Kevin M. Marks, Josh Murtie, Scott A. Biller, Jonathan Hurov, Georg Lenz, Lenny Dang, Nelamangala Nagaraja, Zhihua Sui, Sreevalsam Gopinath, Thomas Antony, Sunil K. Panigrahi, K. Satish Reddy, Hosahalli Subramanya, Siva Sanjeeva Rao, Kavitha Nellore, Mark Fletcher, Sebastian Hayes, Alan Mann, Tabea Erdmann, Zi-Peng Fan, Charles Locuson, Sebastien Ronseaux, Lei Jin, Anil K. Padyana, Erin Artin, Mya Steadman, Sung Choe, Rohini Narayanaswamy, Kevin Truskowski, John Coco, Victor Chubukov, and Gabrielle McDonald
- Abstract
Agents targeting metabolic pathways form the backbone of standard oncology treatments, though a better understanding of differential metabolic dependencies could instruct more rationale-based therapeutic approaches. We performed a chemical biology screen that revealed a strong enrichment in sensitivity to a novel dihydroorotate dehydrogenase (DHODH) inhibitor, AG-636, in cancer cell lines of hematologic versus solid tumor origin. Differential AG-636 activity translated to the in vivo setting, with complete tumor regression observed in a lymphoma model. Dissection of the relationship between uridine availability and response to AG-636 revealed a divergent ability of lymphoma and solid tumor cell lines to survive and grow in the setting of depleted extracellular uridine and DHODH inhibition. Metabolic characterization paired with unbiased functional genomic and proteomic screens pointed to adaptive mechanisms to cope with nucleotide stress as contributing to response to AG-636. These findings support targeting of DHODH in lymphoma and other hematologic malignancies and suggest combination strategies aimed at interfering with DNA-damage response pathways.
- Published
- 2023
6. Supplementary Figures 1 - 11 from AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations
- Author
-
Shin-San Michael Su, Scott A. Biller, Virginie Penard-Lacronique, Marion Dorsch, Lenny Dang, Hua Yang, Wei Liu, Lee Silverman, Shengfang Jin, Wentao Wei, Fan Jiang, Cheng Fang, YingXia Xu, Monika Pilichowska, Benoît S. Marteyn, Stéphane de Botton, Olivier A. Bernard, Sophie Broutin, Angelo Paci, Véronique Saada, Olivia Bawa, Paule Opolon, Cyril Quivoron, Francesco G. Salituro, Jeffrey O. Saunders, Giovanni Cianchetta, Zenon Konteatis, Lei Jin, Sung Choe, Raj Nagaraja, Yue Chen, Erica Tobin, Byron DeLaBarre, Stefan Gross, Anil Padyana, Kimberly Straley, Erin Artin, Muriel D. David, Fang Wang, Jeremy Travins, and Katharine Yen
- Abstract
Supplementary Figure 1. Synthesis and pharmacokinetic characterization of AG-221. Supplementary Figure 2. Biochemical attributes of AG-221. Supplementary Figure 3. Biochemistry of AG-221 with respect to substrate and cofactor. Supplementary Figure 4. TF-1 IDH2R140Q cells treated with AG-221. Supplementary Figure 5. AG-221 can reverse the block in EPO-induced differentiation caused by the expression of IDH2R140Q in the TF-1 erythroleukemia cell line. Supplementary Figure 6. Pharmacokinetics/pharmacodynamics of AG-221 in IDH2R140Qmutant U87MG xenograft tumor-bearing mice. Supplementary Figure 7. AG-221 does not affect intrinsic hematological parameters or body weight. Supplementary Figure 8. AG-221 strongly reduces the number of human IDH2R140Q blasts in the liver and spleen in AML-1, AML-2, and AML-3. Supplementary Figure 9. Flow cytometry analyses of bone marrow-derived hCD45+ cells in primary human AML xenograft models. Supplementary Figure 10. Affinity of the IDH2R140Q homodimer for NADPH. Supplementary Figure 11. Electron density map diagrams for bound ligands for IDH2R140Q co-complex structures determined by X-ray crystallography.
- Published
- 2023
7. Supplementary Tables 1 - 8 from AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations
- Author
-
Shin-San Michael Su, Scott A. Biller, Virginie Penard-Lacronique, Marion Dorsch, Lenny Dang, Hua Yang, Wei Liu, Lee Silverman, Shengfang Jin, Wentao Wei, Fan Jiang, Cheng Fang, YingXia Xu, Monika Pilichowska, Benoît S. Marteyn, Stéphane de Botton, Olivier A. Bernard, Sophie Broutin, Angelo Paci, Véronique Saada, Olivia Bawa, Paule Opolon, Cyril Quivoron, Francesco G. Salituro, Jeffrey O. Saunders, Giovanni Cianchetta, Zenon Konteatis, Lei Jin, Sung Choe, Raj Nagaraja, Yue Chen, Erica Tobin, Byron DeLaBarre, Stefan Gross, Anil Padyana, Kimberly Straley, Erin Artin, Muriel D. David, Fang Wang, Jeremy Travins, and Katharine Yen
- Abstract
Supplementary Table 1. Drug metabolism and pharmacokinetic attributes of AG-221. Supplementary Table 2. Selectivity of AG-221 confirmed by testing against a panel of kinases. Supplementary Table 3. Clinical characteristics of patients with IDH2R140Q-mutated AML. Supplementary Table 4. Treated NSG mice (AML-1, AML-2, AML-3) engrafted with human IDH2R140Q mononuclear cells display stable levels of AG-221 in serum. Supplementary Table 5. AG-221 inhibits 2HG production in models AML-1, AML-2, and AML-3. Supplementary Table 6. Summary of pharmacokinetics/pharmacodynamics in primary human acute myeloid leukemia xenograft model (AML-4). Supplementary Table 7. Summary of data collection and refinement statistics. Supplementary Table 8. Percentage of human chimerism in peripheral blood in models AML-1 and AML-2.
- Published
- 2023
8. Supplementary Methods from AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations
- Author
-
Shin-San Michael Su, Scott A. Biller, Virginie Penard-Lacronique, Marion Dorsch, Lenny Dang, Hua Yang, Wei Liu, Lee Silverman, Shengfang Jin, Wentao Wei, Fan Jiang, Cheng Fang, YingXia Xu, Monika Pilichowska, Benoît S. Marteyn, Stéphane de Botton, Olivier A. Bernard, Sophie Broutin, Angelo Paci, Véronique Saada, Olivia Bawa, Paule Opolon, Cyril Quivoron, Francesco G. Salituro, Jeffrey O. Saunders, Giovanni Cianchetta, Zenon Konteatis, Lei Jin, Sung Choe, Raj Nagaraja, Yue Chen, Erica Tobin, Byron DeLaBarre, Stefan Gross, Anil Padyana, Kimberly Straley, Erin Artin, Muriel D. David, Fang Wang, Jeremy Travins, and Katharine Yen
- Abstract
Supplementary Methods
- Published
- 2023
9. MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis
- Author
-
Katya Marjon, Michael J. Cameron, Phong Quang, Michelle F. Clasquin, Everton Mandley, Kaiko Kunii, Michael McVay, Sung Choe, Andrew Kernytsky, Stefan Gross, Zenon Konteatis, Joshua Murtie, Michelle L. Blake, Jeremy Travins, Marion Dorsch, Scott A. Biller, and Kevin M. Marks
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.
- Published
- 2016
- Full Text
- View/download PDF
10. Preclinical Drug Metabolism, Pharmacokinetic, and Pharmacodynamic Profiles of Ivosidenib, an Inhibitor of Mutant Isocitrate Dehydrogenase 1 for Treatment of Isocitrate Dehydrogenase 1-Mutant Malignancies
- Author
-
Janeta Popovici-Muller, Lenny Dang, Scott A. Biller, Nelamangala Nagaraja, Hua Yang, Shinsan M. Su, Yue Chen, Liping Yan, Hyeryun Kim, Lemieux Rene M, Luke Utley, and Bin Fan
- Subjects
IDH1 ,CYP2B6 ,Metabolic Clearance Rate ,Pyridines ,Glycine ,Pharmaceutical Science ,Antineoplastic Agents ,Drug Elimination Routes ,Pharmacology ,Glutarates ,Mice ,Dogs ,Cytochrome P-450 Enzyme System ,Pharmacokinetics ,In vivo ,Animals ,Humans ,Point Mutation ,Drug Interactions ,Dose-Response Relationship, Drug ,CYP3A4 ,Chemistry ,Myeloid leukemia ,Haplorhini ,Xenograft Model Antitumor Assays ,Isocitrate Dehydrogenase ,Rats ,Leukemia, Myeloid, Acute ,Isocitrate dehydrogenase ,Ketoglutaric Acids ,Drug metabolism ,Protein Binding - Abstract
Point mutations in isocitrate dehydrogenase 1 (IDH1) result in conversion of α-ketoglutarate to the oncometabolite, d-2-hydroxyglutarate (2-HG). Ivosidenib is a once daily (QD), orally available, potent, mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor approved for the treatment of patients with relapsed or refractory acute myeloid leukemia (AML) and intensive chemotherapy-ineligible newly diagnosed AML, with a susceptible IDH1 mutation. We characterized the protein binding, metabolism, metabolites, cell permeability, and drug-drug interaction potential of ivosidenib in humans, monkeys, dogs, rats, and/or mice in in vitro experiments. In vivo pharmacokinetic (PK) profiling and assessment of drug distribution and excretion was undertaken in rats, dogs, and monkeys administered single-dose ivosidenib. The PK/pharmacodynamic (PD) relationship between ivosidenib and 2-HG was analyzed in an mIDH1 xenograft mouse model. Ivosidenib was well absorbed, showed low clearance, and moderate to long terminal half-life (5.3–18.5 hours) in rats, dogs, and monkeys. Brain to plasma exposure ratio was low (2.3%), plasma protein binding was high, and oxidative metabolism was the major elimination pathway. Ivosidenib had high cell permeability and was identified as a substrate for P-glycoprotein. There was moderate induction of cytochrome P450 (P450) enzymes CYP3A4 and CYP2B6 but minimal P450 inhibition or autoinduction. Tumor 2-HG reduction appeared to be dose- and drug-exposure–dependent. Ivosidenib showed a favorable PK profile in several animal species, along with a clear PK/PD relationship demonstrating 2-HG inhibition that translated well to patients with AML. SIGNIFICANCE STATEMENT Ivosidenib is a mutant IDH1 (mIDH1) inhibitor approved for the treatment of certain patients with mIDH1 acute myeloid leukemia. In Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys, ivosidenib demonstrated a favorable pharmacokinetic profile, and in female BALB/c mice showed clear dose- and exposure-dependent inhibition of the oncometabolite, d-2-hydroxyglutarate, which is present at abnormal levels in mIDH1 tumors. These findings led to the further development of ivosidenib and are consistent with data from patients with mIDH1 cancers and healthy participants.
- Published
- 2021
11. Discovery of AG-270, a First-in-Class Oral MAT2A Inhibitor for the Treatment of Tumors with Homozygous MTAP Deletion
- Author
-
Joshua Murtie, Anil K. Padyana, Lei Jin, Marc L. Hyer, Zhixiao Liu, Scott A. Biller, Jeremy Travins, Amelia Barnett, Katya Marjon, Brandon Nicolay, Wentao Wei, Raj Nagaraja, Cheng Fang, Yi Gao, Yabo Sun, Ye Zhixiong, Fan Jiang, Peter Kalev, Stefan Gross, Zenon D. Konteatis, Byron DeLaBarre, Zhihua Sui, Kevin Marks, Lenny Dang, Jie Yu, Everton Mandley, and Yue Chen
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Methionine ,Allosteric regulation ,01 natural sciences ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Enzyme ,chemistry ,CDKN2A ,Methionine Adenosyltransferase ,Drug Discovery ,Cancer cell ,Cancer research ,Molecular Medicine ,Structure–activity relationship ,Binding site ,030304 developmental biology - Abstract
The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was recently implicated as a synthetic lethal target in cancers with deletion of the methylthioadenosine phosphorylase (MTAP) gene, which is adjacent to the CDKN2A tumor suppressor and codeleted with CDKN2A in approximately 15% of all cancers. Previous attempts to target MAT2A with small-molecule inhibitors identified cellular adaptations that blunted their efficacy. Here, we report the discovery of highly potent, selective, orally bioavailable MAT2A inhibitors that overcome these challenges. Fragment screening followed by iterative structure-guided design enabled >10 000-fold improvement in potency of a family of allosteric MAT2A inhibitors that are substrate noncompetitive and inhibit release of the product, S-adenosyl methionine (SAM), from the enzyme's active site. We demonstrate that potent MAT2A inhibitors substantially reduce SAM levels in cancer cells and selectively block proliferation of MTAP-null cells both in tissue culture and xenograft tumors. These data supported progressing AG-270 into current clinical studies (ClinicalTrials.gov NCT03435250).
- Published
- 2021
12. Leveraging Structure-Based Drug Design to Identify Next-Generation MAT2A Inhibitors, Including Brain-Penetrant and Peripherally Efficacious Leads
- Author
-
Mingzong Li, Zenon Konteatis, Nelamangala Nagaraja, Yue Chen, Shubao Zhou, Guangning Ma, Stefan Gross, Katya Marjon, Marc L. Hyer, Everton Mandley, Max Lein, Anil K. Padyana, Lei Jin, Shuilong Tong, Rachel Peters, Joshua Murtie, Jeremy Travins, Matthew Medeiros, Peng Liu, Victoria Frank, Evan T. Judd, Scott A. Biller, Kevin M. Marks, Zhihua Sui, and Samuel K. Reznik
- Subjects
S-Adenosylmethionine ,Drug Design ,Neoplasms ,Drug Discovery ,Molecular Medicine ,Brain ,Humans ,Methionine Adenosyltransferase - Abstract
Inhibition of the
- Published
- 2022
13. Vorasidenib (AG-881): A First-in-Class, Brain-Penetrant Dual Inhibitor of Mutant IDH1 and 2 for Treatment of Glioma
- Author
-
Brandon Nicolay, Ingo K. Mellinghoff, Paolo Codega, Hua Yang, Zenon D. Konteatis, Janeta Popovici-Muller, Katharine E. Yen, Rohini Narayaraswamy, Raj Nagaraja, Zhihua Sui, Shinsan M. Su, Erin Artin, Kimberly Straley, Zhenwei Cai, Feng Wang, Shuilong Tong, Lei Jin, Yue Chen, Cui Dawei, Carl Campos, Zhiyong Luo, Cheng Fang, Xiaobing Lv, Lenny Dang, Ding Zhou, Anil K. Padyana, Huachun Tang, and Scott A. Biller
- Subjects
chemistry.chemical_classification ,IDH1 ,Organic Chemistry ,Mutant ,Dual inhibitor ,2-hydroxyglutarate ,medicine.disease ,Biochemistry ,Featured Letter ,Isocitrate dehydrogenase ,vorasidenib ,chemistry.chemical_compound ,Enzyme ,AG-881 ,chemistry ,Glioma ,Drug Discovery ,medicine ,Cancer research ,Binding site ,mutant IDH1/mIDH2 ,Penetrant (biochemical) - Abstract
Inhibitors of mutant isocitrate dehydrogenase (mIDH) 1 and 2 cancer-associated enzymes prevent the accumulation of the oncometabolite d-2-hydroxyglutarate (2-HG) and are under clinical investigation for the treatment of several cancers harboring an IDH mutation. Herein, we describe the discovery of vorasidenib (AG-881), a potent, oral, brain-penetrant dual inhibitor of both mIDH1 and mIDH2. X-ray cocrystal structures allowed us to characterize the compound binding site, leading to an understanding of the dual mutant inhibition. Furthermore, vorasidenib penetrates the brain of several preclinical species and inhibits 2-HG production in glioma tissue by >97% in an orthotopic glioma mouse model. Vorasidenib represents a novel dual mIDH1/2 inhibitor and is currently in clinical development for the treatment of low-grade mIDH glioma.
- Published
- 2020
14. A chemical biology screen identifies a vulnerability of neuroendocrine cancer cells to SQLE inhibition
- Author
-
Lingling Huang, Kevin Marks, Shengfang Jin, Sung Choe, Scott A. Biller, Joshua Murtie, Gromoslaw A. Smolen, Janeta Popovici-Muller, Eric L. Allen, Rohini Narayanaswamy, Stefan Gross, Yingjia Zhang, Wei Liu, Thomas P. Roddy, Gabrielle McDonald, Nelamangala Nagaraja, Marion Dorsch, Anil K. Padyana, Christopher E. Mahoney, Taryn Sleger, Meina Liu, Yue Chen, Stuart Murray, Victor Chubukov, Sebastian Hayes, Giovanni Cianchetta, Ying Chen, Zi Peng Fan, and David Pirman
- Subjects
0301 basic medicine ,Squalene monooxygenase ,Science ,Cell ,Chemical biology ,General Physics and Astronomy ,Antineoplastic Agents ,02 engineering and technology ,Neuroendocrine tumors ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Drug Delivery Systems ,Cell Line, Tumor ,medicine ,Humans ,lcsh:Science ,Regulation of gene expression ,Multidisciplinary ,Cancer ,General Chemistry ,021001 nanoscience & nanotechnology ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Cholesterol ,Squalene Monooxygenase ,Cell culture ,Cancer cell ,Cancer research ,lcsh:Q ,Drug Screening Assays, Antitumor ,0210 nano-technology ,Gene Deletion - Abstract
Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers., Cancer cells are metabolically adaptable and the identification of specific vulnerabilities is challenging. Here the authors identify a subset of neuroendocrine cell lines exquisitely sensitive to inhibition of SQLE, an enzyme in the cholesterol biosynthetic pathway, due to the toxic accumulation of pathway intermediate squalene.
- Published
- 2019
15. Discovery of AG-270, a First-in-Class Oral MAT2A Inhibitor for the Treatment of Tumors with Homozygous
- Author
-
Zenon, Konteatis, Jeremy, Travins, Stefan, Gross, Katya, Marjon, Amelia, Barnett, Everton, Mandley, Brandon, Nicolay, Raj, Nagaraja, Yue, Chen, Yabo, Sun, Zhixiao, Liu, Jie, Yu, Zhixiong, Ye, Fan, Jiang, Wentao, Wei, Cheng, Fang, Yi, Gao, Peter, Kalev, Marc L, Hyer, Byron, DeLaBarre, Lei, Jin, Anil K, Padyana, Lenny, Dang, Joshua, Murtie, Scott A, Biller, Zhihua, Sui, and Kevin M, Marks
- Subjects
S-Adenosylmethionine ,Structure-Activity Relationship ,Binding Sites ,Purine-Nucleoside Phosphorylase ,Drug Design ,Neoplasms ,Homozygote ,Humans ,Methionine Adenosyltransferase ,Enzyme Inhibitors ,Molecular Dynamics Simulation ,Crystallography, X-Ray - Abstract
The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was recently implicated as a synthetic lethal target in cancers with deletion of the methylthioadenosine phosphorylase (
- Published
- 2021
16. The Making of a Drug-Hunting Addict
- Author
-
Scott A. Biller
- Subjects
Drug ,media_common.quotation_subject ,Criminology ,Psychology ,media_common - Published
- 2020
17. Evidence that microsomal triglyceride transfer protein is limiting in the production of apolipoprotein B-containing lipoproteins in hepatic cells
- Author
-
Haris Jamil, Ching-Hsuen Chu, John K. Dickson, Jr., Ying Chen, Mujing Yan, Scott A. Biller, Richard E. Gregg, John R. Wetterau, and David A. Gordon
- Subjects
lipid transfer protein ,protein disulfide isomerase ,photoaffinity ,benzophenone ,HepG2 cells ,McArdle RH-7777 cells ,Biochemistry ,QD415-436 - Abstract
The microsomal triglyceride transfer protein (MTP) is a heterodimeric lipid transfer protein that is required for the assembly and secretion of apolipoprotein B (apoB)-containing lipoproteins. A key unresolved question is whether the MTP-mediated step is rate limiting. To address this, a unique experimental strategy was used that allowed the in situ modulation and measurement of MTP triglyceride transfer activity. In order to accomplish this, an irreversible photoaffinity inhibitor, BMS-192951, was designed and synthesized. When incubated with purified MTP and irradiated with UV light at 360 nm, BMS-192951 inhibits triglyceride transfer by covalently binding to the protein. HepG2 cells were treated with either increasing concentrations of BMS-192951 (0–15 μm) with 5 min of ultraviolet irradiation, or 3.0 μm BMS-192951 with various lengths (0–15 min) of ultraviolet irradiation. Microsomal extracts were prepared exhaustively dialyzed to remove unbound inhibitor, and assayed for MTP-mediated triglyceride transfer activity. BMS-192951 was shown to reduce MTP activity in both a dose- and UV exposure time-dependent fashion. Measurement of apoB concentration in the media showed that apoB secretion was reduced in proportion to the in situ inhibition of MTP activity, while no change was observed in apoA-I secretion. Experiments performed in McArdle RH-7777 rat hepatoma cells and primary rat hepatocytes gave nearly identical results; the decrease in apoB secretion was proportional to the decrease in MTP activity. These results indicate that MTP-mediated lipid transfer is limiting in the assembly and secretion of apoB-containing lipoproteins in hepatic cells under the conditions tested.—Jamil, H., C-H. Chu, J. K. Dickson, Jr., Y. Chen, M. Yan, S. A. Biller, R. E. Gregg, J. R. Wetterau, and D. A. Gordon. Evidence that microsomal triglyceride transfer protein is limiting in the production of apolipoprotein B-containing lipoproteins in hepatic cells. J. Lipid Res. 1998. 39: 1448–1454.
- Published
- 1998
- Full Text
- View/download PDF
18. Selective Vulnerability to Pyrimidine Starvation in Hematologic Malignancies Revealed by AG-636, a Novel Clinical-Stage Inhibitor of Dihydroorotate Dehydrogenase
- Author
-
K. Satish Reddy, Scott A. Biller, Kavitha Nellore, Siva Sanjeeva Rao, Anil K. Padyana, Georg Lenz, Thomas Antony, Charles Locuson, Jonathan Hurov, Mya Steadman, Tabea Erdmann, Mark Fletcher, Zi Peng Fan, Kevin Truskowski, Sreevalsam Gopinath, Alan Mann, Danielle Ulanet, Rohini Narayanaswamy, Sebastien Ronseaux, Gabrielle McDonald, Sung Choe, Zhihua Sui, John Coco, Lenny Dang, Kevin Marks, Victor Chubukov, Erin Artin, Sebastian Hayes, Josh Murtie, Lei Jin, Nelamangala Nagaraja, Hosahalli Subramanya, and Sunil Kumar Panigrahi
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,Oxidoreductases Acting on CH-CH Group Donors ,Cell Survival ,Chemical biology ,Dihydroorotate Dehydrogenase ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,medicine ,Extracellular ,Humans ,Enzyme Inhibitors ,Neoplasm Staging ,Genomics ,medicine.disease ,Uridine ,Lymphoma ,Metabolic pathway ,030104 developmental biology ,Pyrimidines ,Oncology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Hematologic Neoplasms ,Cancer research ,Dihydroorotate dehydrogenase ,DNA Damage - Abstract
Agents targeting metabolic pathways form the backbone of standard oncology treatments, though a better understanding of differential metabolic dependencies could instruct more rationale-based therapeutic approaches. We performed a chemical biology screen that revealed a strong enrichment in sensitivity to a novel dihydroorotate dehydrogenase (DHODH) inhibitor, AG-636, in cancer cell lines of hematologic versus solid tumor origin. Differential AG-636 activity translated to the in vivo setting, with complete tumor regression observed in a lymphoma model. Dissection of the relationship between uridine availability and response to AG-636 revealed a divergent ability of lymphoma and solid tumor cell lines to survive and grow in the setting of depleted extracellular uridine and DHODH inhibition. Metabolic characterization paired with unbiased functional genomic and proteomic screens pointed to adaptive mechanisms to cope with nucleotide stress as contributing to response to AG-636. These findings support targeting of DHODH in lymphoma and other hematologic malignancies and suggest combination strategies aimed at interfering with DNA-damage response pathways.
- Published
- 2020
19. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML
- Author
-
Sung Choe, Zenon D. Konteatis, Chris Bowden, Brandon Nicolay, Courtney D. DiNardo, Bin Wu, Jessica K. Altman, Alice S. Mims, Lenny Dang, Scott A. Biller, Guowen Liu, Daniel A. Pollyea, Parham Nejad, Hongfang Wang, Eyal C. Attar, Richard Stone, Stéphane de Botton, Wei Liu, Vickie Zhang, Eytan M. Stein, Kevin Marks, Justin M. Watts, Gail J. Roboz, Meredith Goldwasser, Lynn Quek, Amir T. Fathi, Hua Liu, Hagop M. Kantarjian, and Martin S. Tallman
- Subjects
0301 basic medicine ,Myeloid ,IDH1 ,Combination therapy ,Pyridines ,Glycine ,IDH2 ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,medicine ,Humans ,Myeloid Neoplasia ,biology ,business.industry ,Myeloid leukemia ,Hematology ,medicine.disease ,Isocitrate Dehydrogenase ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Isocitrate dehydrogenase ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business - Abstract
Isocitrate dehydrogenase (IDH) 1 and 2 mutations result in overproduction of D-2-hydroxyglutarate (2-HG) and impaired cellular differentiation. Ivosidenib, a targeted mutant IDH1 (mIDH1) enzyme inhibitor, can restore normal differentiation and results in clinical responses in a subset of patients with mIDH1 relapsed/refractory (R/R) acute myeloid leukemia (AML). We explored mechanisms of ivosidenib resistance in 174 patients with confirmed mIDH1 R/R AML from a phase 1 trial. Receptor tyrosine kinase (RTK) pathway mutations were associated with primary resistance to ivosidenib. Multiple mechanisms contributed to acquired resistance, particularly outgrowth of RTK pathway mutations and 2-HG–restoring mutations (second-site IDH1 mutations, IDH2 mutations). Observation of multiple concurrent mechanisms in individual patients underscores the complex biology of resistance and has important implications for rational combination therapy design. This trial was registered at www.clinicaltrials.gov as #NCT02074839
- Published
- 2020
20. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations
- Author
-
Justin Taylor, Fabian Correa, Craig B. Thompson, Bo Wang, Gregory A. Petsko, Zenon D. Konteatis, Noushin Farnoud, Maria E. Arcila, Bin Wu, Ariën S. Rustenburg, Sung Choe, Mikhail Roshal, Naofumi Takemoto, Ross L. Levine, Vidushi Durani, Andrew M. Intlekofer, Steven K. Albanese, Hui Liu, Christopher Famulare, Eytan M. Stein, Scott A. Biller, Elli Papaemmanuil, Minal Patel, Justin R. Cross, Abbas Nazir, Martin S. Tallman, Alan H. Shih, and John D. Chodera
- Subjects
Models, Molecular ,0301 basic medicine ,Myeloid ,Glutamine ,Mutant ,cancer metabolism ,Aminopyridines ,Drug resistance ,medicine.disease_cause ,Pathogenesis ,Mice ,AG-221 ,glioma ,Enzyme Inhibitors ,Mutation ,Multidisciplinary ,Clinical Trials, Phase I as Topic ,Triazines ,Chemistry ,leukemia ,Isocitrate Dehydrogenase ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Disease Progression ,enasidenib ,IDH1 ,Female ,IDH2 ,epigenetic ,Allosteric Site ,metabolite ,Enasidenib ,Article ,Glutarates ,AG-120 ,03 medical and health sciences ,Clinical Trials, Phase II as Topic ,medicine ,Animals ,Humans ,Isoleucine ,Alleles ,ivosidenib ,medicine.disease ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,Mutant Proteins ,Protein Multimerization - Abstract
Somatic mutations in the isocitrate dehydrogenase 2 gene (IDH2) contribute to the pathogenesis of acute myeloid leukaemia (AML) through the production of the oncometabolite 2-hydroxyglutarate (2HG)1–8. Enasidenib (AG-221) is an allosteric inhibitor that binds to the IDH2 dimer interface and blocks the production of 2HG by IDH2 mutants9,10. In a phase I/II clinical trial, enasidenib inhibited the production of 2HG and induced clinical responses in relapsed or refractory IDH2-mutant AML11. Here we describe two patients with IDH2-mutant AML who had a clinical response to enasidenib followed by clinical resistance, disease progression, and a recurrent increase in circulating levels of 2HG. We show that therapeutic resistance is associated with the emergence of second-site IDH2 mutations in trans, such that the resistance mutations occurred in the IDH2 allele without the neomorphic R140Q mutation. The in trans mutations occurred at glutamine 316 (Q316E) and isoleucine 319 (I319M), which are at the interface where enasidenib binds to the IDH2 dimer. The expression of either of these mutant disease alleles alone did not induce the production of 2HG; however, the expression of the Q316E or I319M mutation together with the R140Q mutation in trans allowed 2HG production that was resistant to inhibition by enasidenib. Biochemical studies predicted that resistance to allosteric IDH inhibitors could also occur via IDH dimer-interface mutations in cis, which was confirmed in a patient with acquired resistance to the IDH1 inhibitor ivosidenib (AG-120). Our observations uncover a mechanism of acquired resistance to a targeted therapy and underscore the importance of 2HG production in the pathogenesis of IDH-mutant malignancies. A new mechanism of acquired clinical resistance in two patients with acute myeloid leukaemia driven by mutant IDH2 is described, in which a second-site mutation on the wild-type allele induces therapeutic resistance to IDH2 inhibitors.
- Published
- 2018
21. Discovery of AG-120 (Ivosidenib): A First-in-Class Mutant IDH1 Inhibitor for the Treatment of IDH1 Mutant Cancers
- Author
-
Hua Yang, Muriel D. David, Jeffrey O. Saunders, Lemieux Rene M, Gui Yao, Luke Utley, Véronique Saada, Xiaobing Lv, Yue Chen, Fang Wang, Shengfang Jin, Ping Chen, Erin Artin, Stéphane de Botton, Cheng Fang, Hyeryun Kim, Giovanni Cianchetta, Stefan Gross, Lenny Dang, Zhenwei Cai, Scott A. Biller, Andrew J. Olaharski, Erica Tobin, Cui Dawei, Katharine E. Yen, Virginie Penard-Lacronique, Liping Yan, Ding Zhou, Lee Silverman, Cyril Quivoron, Zhiyong Luo, Jeremy Travins, Janeta Popovici-Muller, Shinsan M. Su, Kimberly Straley, Zhiwei Gu, and Francesco G. Salituro
- Subjects
mutant IDH1 ,0301 basic medicine ,Letter ,IDH1 ,ivosidenib ,Cellular differentiation ,Point mutation ,Organic Chemistry ,Mutant ,2-hydroxyglutarate ,Biology ,Biochemistry ,AG-120 ,03 medical and health sciences ,differentiation therapy ,030104 developmental biology ,0302 clinical medicine ,Isocitrate dehydrogenase ,Differentiation therapy ,030220 oncology & carcinogenesis ,Drug Discovery ,Cancer cell ,Cancer research ,isocitrate dehydrogenase ,Ex vivo - Abstract
Somatic point mutations at a key arginine residue (R132) within the active site of the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) confer a novel gain of function in cancer cells, resulting in the production of d-2-hydroxyglutarate (2-HG), an oncometabolite. Elevated 2-HG levels are implicated in epigenetic alterations and impaired cellular differentiation. IDH1 mutations have been described in an array of hematologic malignancies and solid tumors. Here, we report the discovery of AG-120 (ivosidenib), an inhibitor of the IDH1 mutant enzyme that exhibits profound 2-HG lowering in tumor models and the ability to effect differentiation of primary patient AML samples ex vivo. Preliminary data from phase 1 clinical trials enrolling patients with cancers harboring an IDH1 mutation indicate that AG-120 has an acceptable safety profile and clinical activity.
- Published
- 2018
22. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations
- Author
-
Olivia Bawa, Monika Pilichowska, Paule Opolon, Jeffrey O. Saunders, Fang Wang, Cyril Quivoron, Anil K. Padyana, Zenon D. Konteatis, Kimberly Straley, Erica Tobin, Sophie Broutin, Marion Dorsch, Hua Yang, Byron DeLaBarre, Jeremy Travins, Sung Choe, Yue Chen, Lei Jin, Wentao Wei, Virginie Penard-Lacronique, Raj Nagaraja, Wei Liu, Lenny Dang, Shengfang Jin, Cheng Fang, Lee Silverman, Fan Jiang, Katharine E. Yen, Giovanni Cianchetta, Olivier Bernard, Erin Artin, Muriel D. David, Shin-San Michael Su, Stefan Gross, Francesco G. Salituro, Véronique Saada, Stéphane de Botton, Scott A. Biller, Angelo Paci, Benoit S. Marteyn, Yingxia Xu, Agios Pharmaceuticals, Hématopoïèse normale et pathologique (U1170 Inserm), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Institut Gustave Roussy (IGR), Plateforme d’évaluation préclinique (PFEP), Analyse moléculaire, modélisation et imagerie de la maladie cancéreuse (AMMICa), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Pharmacologie, Département de biologie et pathologie médicales [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Laboratoire de thérapie cellulaire, Département de médecine oncologique [Gustave Roussy], Pathogénie microbienne moléculaire, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Tufts Medical Center, ShangPharma, Viva Biotech Ltd., and This work was funded by Agios Pharmaceuticals, Inc., the French National Institute of Health (INSERM-AVIESAN), the National Cancer Institute (INCa-DGOS-Inserm_6043 and INCa 2012-1-RT-09), and the Fondation Association pour la Recherche sur le Cancer (ARC, SL220130607089 Programme Labellisé to V. Penard-Lacronique and S. de Botton). M.D. David is funded by a fellowship from the Institut National du Cancer (INCa-DGOS_5733).
- Subjects
0301 basic medicine ,Myeloid ,IDH1 ,Cellular differentiation ,Myeloid leukemia ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Enasidenib ,medicine.disease ,Molecular biology ,IDH2 ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Isocitrate dehydrogenase ,Oncology ,030220 oncology & carcinogenesis ,medicine - Abstract
Somatic gain-of-function mutations in isocitrate dehydrogenases (IDH) 1 and 2 are found in multiple hematologic and solid tumors, leading to accumulation of the oncometabolite (R)-2-hydroxyglutarate (2HG). 2HG competitively inhibits α-ketoglutarate–dependent dioxygenases, including histone demethylases and methylcytosine dioxygenases of the TET family, causing epigenetic dysregulation and a block in cellular differentiation. In vitro studies have provided proof of concept for mutant IDH inhibition as a therapeutic approach. We report the discovery and characterization of AG-221, an orally available, selective, potent inhibitor of the mutant IDH2 enzyme. AG-221 suppressed 2HG production and induced cellular differentiation in primary human IDH2 mutation–positive acute myeloid leukemia (AML) cells ex vivo and in xenograft mouse models. AG-221 also provided a statistically significant survival benefit in an aggressive IDH2R140Q-mutant AML xenograft mouse model. These findings supported initiation of the ongoing clinical trials of AG-221 in patients with IDH2 mutation–positive advanced hematologic malignancies. Significance: Mutations in IDH1/2 are identified in approximately 20% of patients with AML and contribute to leukemia via a block in hematopoietic cell differentiation. We have shown that the targeted inhibitor AG-221 suppresses the mutant IDH2 enzyme in multiple preclinical models and induces differentiation of malignant blasts, supporting its clinical development. Cancer Discov; 7(5); 478–93. ©2017 AACR. See related commentary by Thomas and Majeti, p. 459. See related article by Shih et al., p. 494. This article is highlighted in the In This Issue feature, p. 443
- Published
- 2017
23. MAT2A Inhibition Blocks the Growth of MTAP-Deleted Cancer Cells by Reducing PRMT5-Dependent mRNA Splicing and Inducing DNA Damage
- Author
-
Rachel Peters, Kate Lizotte, Victoria Frank, Yesim Tuncay, Katie Sellers, Elia Aguado-Fraile, Marc L. Hyer, Everton Mandley, Scott A. Biller, Michelle Clasquin, Katya Marjon, Phong Quang, Jeremy Travins, Joshua E. Goldford, Zenon D. Konteatis, Chi-Chao Chen, Jaclyn Weier, Lenny Dang, Raj Nagaraja, Peter Kalev, Kevin Marks, Wei Liu, Eric Simone, Joshua Murtie, Stefan Gross, Max Lein, Yue Chen, Mark Fletcher, Amelia Barnett, Zhihua Sui, and Sebastian Hayes
- Subjects
0301 basic medicine ,Protein-Arginine N-Methyltransferases ,S-Adenosylmethionine ,Cancer Research ,DNA damage ,RNA Splicing ,Mice, Nude ,Mice, SCID ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,CDKN2A ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,RNA, Messenger ,Enzyme Inhibitors ,Gene ,Cyclin-Dependent Kinase Inhibitor p16 ,Kinase ,Chemistry ,Protein arginine methyltransferase 5 ,Methionine Adenosyltransferase ,HCT116 Cells ,HEK293 Cells ,030104 developmental biology ,Purine-Nucleoside Phosphorylase ,Oncology ,030220 oncology & carcinogenesis ,RNA splicing ,Cancer cell ,Cancer research ,Gene Deletion ,DNA Damage - Abstract
The methylthioadenosine phosphorylase (MTAP) gene is located adjacent to the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor-suppressor gene and is co-deleted with CDKN2A in approximately 15% of all cancers. This co-deletion leads to aggressive tumors with poor prognosis that lack effective, molecularly targeted therapies. The metabolic enzyme methionine adenosyltransferase 2α (MAT2A) was identified as a synthetic lethal target in MTAP-deleted cancers. We report the characterization of potent MAT2A inhibitors that substantially reduce levels of S-adenosylmethionine (SAM) and demonstrate antiproliferative activity in MTAP-deleted cancer cells and tumors. Using RNA sequencing and proteomics, we demonstrate that MAT2A inhibition is mechanistically linked to reduced protein arginine methyltransferase 5 (PRMT5) activity and splicing perturbations. We further show that DNA damage and mitotic defects ensue upon MAT2A inhibition in HCT116 MTAP-/- cells, providing a rationale for combining the MAT2A clinical candidate AG-270 with antimitotic taxanes.
- Published
- 2021
24. A small molecule inhibitor of mutant IDH2 rescues cardiomyopathy in a D-2-hydroxyglutaric aciduria type II mouse model
- Author
-
Gajja S. Salomons, Yong Cang, Scott A. Biller, Hua Yang, Josh Powe, Dongwei Zhu, Stefan Gross, Stuart Murray, Kimberly Straley, Samuel V. Agresta, Karen S. Regan, Katharine E. Yen, Yaguang Si, Mya Steadman, Wei Liu, Lenny Dang, Ana Pop, Yue Chen, Erin Artin, Marion Dorsch, Jeremy Travins, Stephanie Santiago, Shengfang Jin, Muriel D. David, Cyril Quivoron, Andrew Kernytsky, Virginie Penard-Lacronique, Lee Silverman, Chenming Lu, Shin San Michael Su, Zhizhong Lin, Erwin E.W. Jansen, Fang Wang, Eduard A. Struys, Laboratory Medicine, and Amsterdam Neuroscience - Cellular & Molecular Mechanisms
- Subjects
0301 basic medicine ,Mutant ,Cardiomyopathy ,Locus (genetics) ,Biology ,IDH2 ,Germline ,Small Molecule Libraries ,03 medical and health sciences ,Epilepsy ,Mice ,Genetics ,medicine ,Animals ,Genetics(clinical) ,Gene ,Genetics (clinical) ,chemistry.chemical_classification ,Brain Diseases, Metabolic, Inborn ,medicine.disease ,Isocitrate Dehydrogenase ,Disease Models, Animal ,030104 developmental biology ,Enzyme ,chemistry ,Mutation ,Cancer research ,Original Article ,Cardiomyopathies - Abstract
D-2-hydroxyglutaric aciduria (D2HGA) type II is a rare neurometabolic disorder caused by germline gain-of-function mutations in isocitrate dehydrogenase 2 (IDH2), resulting in accumulation of D-2-hydroxyglutarate (D2HG). Patients exhibit a wide spectrum of symptoms including cardiomyopathy, epilepsy, developmental delay and limited life span. Currently, there are no effective therapeutic interventions. We generated a D2HGA type II mouse model by introducing the Idh2R140Q mutation at the native chromosomal locus. Idh2R140Q mice displayed significantly elevated 2HG levels and recapitulated multiple defects seen in patients. AGI-026, a potent, selective inhibitor of the human IDH2R140Q-mutant enzyme, suppressed 2HG production, rescued cardiomyopathy, and provided a survival benefit in Idh2R140Q mice; treatment withdrawal resulted in deterioration of cardiac function. We observed differential expression of multiple genes and metabolites that are associated with cardiomyopathy, which were largely reversed by AGI-026. These findings demonstrate the potential therapeutic benefit of an IDH2R140Q inhibitor in patients with D2HGA type II. Electronic supplementary material The online version of this article (doi:10.1007/s10545-016-9960-y) contains supplementary material, which is available to authorized users.
- Published
- 2016
25. MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis
- Author
-
Scott A. Biller, Michelle L. Blake, Joshua Murtie, Everton Mandley, Kevin Marks, Zenon D. Konteatis, Andrew Kernytsky, Michael J. Cameron, Sung Choe, Marion Dorsch, Jeremy Travins, Kaiko Kunii, Stefan Gross, Michelle Clasquin, Katya Marjon, Phong Quang, and Michael McVay
- Subjects
0301 basic medicine ,Protein-Arginine N-Methyltransferases ,Methyltransferase ,Adenosine ,Purine nucleoside phosphorylase ,Synthetic lethality ,Biology ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,CDKN2A ,Antigens, Neoplasm ,Neoplasms ,Humans ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Thionucleosides ,Protein arginine methyltransferase 5 ,Methylation ,Genomics ,Methionine Adenosyltransferase ,HCT116 Cells ,Molecular biology ,030104 developmental biology ,Purine-Nucleoside Phosphorylase ,lcsh:Biology (General) ,Multiprotein Complexes ,Gene Deletion ,Signal Transduction - Abstract
SummaryHomozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.
- Published
- 2016
- Full Text
- View/download PDF
26. AG-348 enhances pyruvate kinase activity in red blood cells from patients with pyruvate kinase deficiency
- Author
-
Yingxia Xu, Shin-San Michael Su, Zhiwei Gu, Scott A. Biller, Charles Kung, Xiaobing Lv, Cheng Fang, Collin Hill, Yaguang Si, Zhiyong Luo, Penelope A. Kosinski, Francesco G. Salituro, Lenny Dang, Huachun Tang, Hua Yang, Lee Silverman, Kendall Johnson, Janeta Popovici-Muller, Gui Yao, Jeff Hixon, Gavin Histen, Stefan Gross, Yue Chen, Byron DeLaBarre, and Giovanni Cianchetta
- Subjects
Hemolytic anemia ,Erythrocytes ,Anemia ,Immunology ,Allosteric regulation ,Pyruvate Kinase ,Enzyme Activators ,Pharmacology ,Biology ,Pyruvate Metabolism, Inborn Errors ,Biochemistry ,Piperazines ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Red Cells, Iron, and Erythropoiesis ,medicine ,Animals ,Humans ,Glycolysis ,chemistry.chemical_classification ,Sulfonamides ,Cell Biology ,Hematology ,Anemia, Hemolytic, Congenital Nonspherocytic ,medicine.disease ,Molecular biology ,Recombinant Proteins ,Tissue Donors ,Enzyme Activation ,Kinetics ,Enzyme ,chemistry ,030220 oncology & carcinogenesis ,Quinolines ,Pyruvate kinase ,Ex vivo ,030215 immunology ,Pyruvate kinase deficiency - Abstract
Pyruvate kinase (PK) deficiency is a rare genetic disease that causes chronic hemolytic anemia. There are currently no targeted therapies for PK deficiency. Here, we describe the identification and characterization of AG-348, an allosteric activator of PK that is currently in clinical trials for the treatment of PK deficiency. We demonstrate that AG-348 can increase the activity of wild-type and mutant PK enzymes in biochemical assays and in patient red blood cells treated ex vivo. These data illustrate the potential for AG-348 to restore the glycolytic pathway activity in patients with PK deficiency and ultimately lead to clinical benefit.
- Published
- 2017
27. An Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma Cells
- Author
-
Timothy A. Chan, Kaiko Kunii, Thomas G. Graeber, Alicia Pedraza, Barbara Oldrini, Owen Clark, Jennifer Tsoi, Stefanie Schalm, Cameron Brennan, Fang Wang, Yue Chen, Shinsan M. Su, Hua Yang, Alexandra Miller, Scott A. Biller, Sevin Turcan, Lee Silverman, Carl Campos, Marc K. Rosenblum, Wei Liu, Andrew Kernytsky, Evangelia Komisopoulou, Ingo K. Mellinghoff, Katharine E. Yen, Christian Grommes, Nicolaos Palaskas, Dan Rohle, and Janeta Popovici-Muller
- Subjects
Regulation of gene expression ,Multidisciplinary ,biology ,Cellular differentiation ,Mutant ,Molecular biology ,Article ,Cell biology ,Histone ,Isocitrate dehydrogenase ,DNA methylation ,Cancer cell ,biology.protein ,Epigenetics - Abstract
IDHology Among the most exciting drug targets to emerge from cancer genome sequencing projects are two related metabolic enzymes, isocitrate dehydrogenases 1 and 2 (IDH1, IDH2). Mutations in the IDH1 and IDH2 genes are common in certain types of human cancer. Whether inhibition of mutant IDH activity might offer therapeutic benefits is unclear (see the Perspective by Kim and DeBerardinis ). F. Wang et al. (p. 622 , published online 4 April) isolated a small molecule that selectively inhibits mutant IDH2, describe the structural details of its binding to the mutant enzyme, and show that this compound suppresses the growth of patient-derived leukemia cells harboring the IDH2 mutation. Rohle et al. (p. 626 , published online 4 April) show that a small molecule inhibitor of IDH1 selectively slows the growth of patient-derived brain tumor cells with the IDH1 mutation.
- Published
- 2013
28. Targeted Inhibition of Mutant IDH2 in Leukemia Cells Induces Cellular Differentiation
- Author
-
Shunqi Yan, Scott A. Biller, Fang Wang, David P. Schenkein, Marion Dorsch, Shinsan M. Su, Jeffrey O. Saunders, Jeremy Travins, Wentao Wei, Kimberly Straley, Wei Liu, Stefan Gross, Camelia Gliser, Lenny Dang, Elena Mylonas, Hua Yang, Sam Agresta, Erica Hansen, Véronique Saada, Stéphane de Botton, Byron DeLaBarre, Katharine E. Yen, Janeta Popovici-Muller, Stuart Murray, Erin Artin, Stefanie Schalm, Cyril Quivoron, Andrew Kernytsky, Virginie Penard-Lacronique, Yi Gao, and Francesco G. Salituro
- Subjects
Leukemia ,Multidisciplinary ,IDH1 ,Differentiation therapy ,Cellular differentiation ,Allosteric regulation ,Cancer cell ,Mutant ,medicine ,Biology ,Enasidenib ,medicine.disease ,Molecular biology - Abstract
IDHology Among the most exciting drug targets to emerge from cancer genome sequencing projects are two related metabolic enzymes, isocitrate dehydrogenases 1 and 2 (IDH1, IDH2). Mutations in the IDH1 and IDH2 genes are common in certain types of human cancer. Whether inhibition of mutant IDH activity might offer therapeutic benefits is unclear (see the Perspective by Kim and DeBerardinis ). F. Wang et al. (p. 622 , published online 4 April) isolated a small molecule that selectively inhibits mutant IDH2, describe the structural details of its binding to the mutant enzyme, and show that this compound suppresses the growth of patient-derived leukemia cells harboring the IDH2 mutation. Rohle et al. (p. 626 , published online 4 April) show that a small molecule inhibitor of IDH1 selectively slows the growth of patient-derived brain tumor cells with the IDH1 mutation.
- Published
- 2013
29. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic
- Author
-
Katharine, Yen, Jeremy, Travins, Fang, Wang, Muriel D, David, Erin, Artin, Kimberly, Straley, Anil, Padyana, Stefan, Gross, Byron, DeLaBarre, Erica, Tobin, Yue, Chen, Raj, Nagaraja, Sung, Choe, Lei, Jin, Zenon, Konteatis, Giovanni, Cianchetta, Jeffrey O, Saunders, Francesco G, Salituro, Cyril, Quivoron, Paule, Opolon, Olivia, Bawa, Véronique, Saada, Angelo, Paci, Sophie, Broutin, Olivier A, Bernard, Stéphane, de Botton, Benoît S, Marteyn, Monika, Pilichowska, YingXia, Xu, Cheng, Fang, Fan, Jiang, Wentao, Wei, Shengfang, Jin, Lee, Silverman, Wei, Liu, Hua, Yang, Lenny, Dang, Marion, Dorsch, Virginie, Penard-Lacronique, Scott A, Biller, and Shin-San Michael, Su
- Subjects
Leukemia, Myeloid, Acute ,Mice ,Triazines ,Cell Line, Tumor ,Mutation ,Aminopyridines ,Animals ,Humans ,Antineoplastic Agents ,Xenograft Model Antitumor Assays ,Isocitrate Dehydrogenase ,Article - Abstract
AG-221 or enasidenib is a first-in-class selective inhibitor of mutated isocitrate dehydrogenase 2 (IDH2) with early demonstrated clinical efficacy in acute myeloid leukemia as a single agent, yet with persistence of mutant IDH2 clones. Two papers in this issue of Cancer Discovery provide further insight into the biological activity of AG-221 in promoting differentiation of IDH2 mutant cells and reversing aberrant DNA methylation over time, and demonstrating pre-clinical activity in combination with a targeted FLT3 kinase inhibitor to eliminate IDH2 mutant clones.
- Published
- 2016
30. Abstract 3504: A chemical biology screen identifies a unique vulnerability of neuroendocrine cancer cells to SQLE inhibition
- Author
-
Gromoslaw A. Smolen, Stefan Gross, Shengfang Jin, Scott A. Biller, Taryn Sleger, Sung Choe, Rohini Narayanaswamy, Joshua Murtie, Raj Nagaraja, Gabrielle McDonald, Thomas P. Roddy, Yu Chen, David Pirman, Giovanni Cianchetta, Christopher E. Mahoney, Sebastian Hayes, Zi Peng Fan, Anil K. Padyana, Stuart Murray, and Victor Chubukov
- Subjects
Cancer Research ,Oncology ,Neuroendocrine Cancer ,Chemical biology ,Vulnerability ,Computational biology ,Biology - Abstract
Numerous reports have described the differential metabolism of cancer cells as compared to their normal counterparts. However, only relatively few metabolic genes with cancer-specific mutations have been reported and the identification of cancer subsets with particular metabolic vulnerabilities remains a challenge. To explore potential cancer-specific dependencies, we conducted a chemical biology screen utilizing a collection of small molecule inhibitors targeting diverse metabolic pathways in a large panel of cancer cell lines. A subset of neuroendocrine tumors, particularly small cell lung cancers (SCLC), displayed a striking dependence on squalene epoxygenase, SQLE, an enzyme in the cholesterol biosynthetic pathway. To develop further confidence in these findings, we have determined the first three-dimensional SQLE structure and further advanced a pharmacological toolbox for SQLE. Using these tools, we showed that the observed effects are on target and that the patterns of cellular sensitivity observed in vitro display excellent translation to in vivo xenografts studies. Interestingly, using a variety of orthogonal approaches, we demonstrated that SQLE sensitivity appears not to be related to overall inhibition of the cholesterol pathway but rather to specific and toxic accumulation of the SQLE substrate, squalene. Collectively, these findings highlight the utility of chemical biology screens and identify SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly SCLC. Citation Format: Christopher Mahoney, David Pirman, Victor Chubukov, Taryn Sleger, Anil Padyana, Stefan Gross, Sebastian Hayes, Zi Peng Fan, Gabrielle McDonald, Yu Chen, Joshua Murtie, Giovanni Cianchetta, Raj Nagaraja, Rohini Narayanaswamy, Sung Choe, Stuart Murray, Shengfang Jin, Scott Biller, Thomas Roddy, Gromoslaw A. Smolen. A chemical biology screen identifies a unique vulnerability of neuroendocrine cancer cells to SQLE inhibition [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3504.
- Published
- 2018
31. High-Content Micronucleus Assay in Genotoxicity Profiling: Initial-Stage Development and Some Applications in the Investigative/Lead-Finding Studies in Drug Discovery
- Author
-
Thomas H. Marsilje, Alan Collis, Dejan Bojanic, Maria Magnifico, Madhu S. Mondal, Philip Bentley, Claudia Mcginnis, Scott A. Biller, Wilfried Frieauff, Hans-Joerg Martus, Willi Suter, Laszlo Urban, and Joseph Gabriels
- Subjects
Micronucleus Tests ,Drug Industry ,Drug-Related Side Effects and Adverse Reactions ,Drug discovery ,Chemistry ,Gene Expression Profiling ,Chinese hamster ovary cell ,Drug Evaluation, Preclinical ,Gene Expression ,Reproducibility of Results ,CHO Cells ,Computational biology ,Pharmacology ,Toxicology ,medicine.disease_cause ,In vitro ,Gene expression profiling ,Cricetulus ,Cricetinae ,Micronucleus test ,medicine ,Animals ,Cytotoxicity ,Micronucleus ,Genotoxicity - Abstract
This article describes the first step toward full (that includes conditions for both absence and presence of metabolic activation) validation and drug discovery application of a 96-well, automated, high-content micronucleus (HCMN) assay. The current validation tests were performed using Chinese hamster ovary cells, in the absence of metabolic activation, against three distinct sets of drug-like compounds that represent all stages of a drug discovery pipeline. A compound categorization scheme was created based on quantitative relationships between micronucleus (MN) signals, cytotoxicity, and compound solubility. Results from initial validation compounds (n = 38) set the stage for differentiating overall positive and negative MN inducers. To delve deeper into the compound categorization process, a more extensive validation set, consisting of a larger set (n = 370) of "drug-like but less optimized" early-stage compounds, was used for further refinement of positive and negative compound categories. The predictivity and applicability of the assay for clinical stage compounds was ascertained using (n = 168) clinically developed marketed drugs or well-studied compounds. Upon full validation, a detailed analysis of results established five compound categories--NEG (negative), NEG/xx μM (negative up to the solubility limit of xx μM), WPOS (weak positive), POS (positive), and INCON (inconclusive). Furthermore, examples of lead-finding applications and ongoing investigative HCMN activities are described. A proposal is offered on how the HCMN assay can be positioned in parallel to the overall stage gates (e.g., scaffold selection, lead optimization, late-stage preclinical development) of drug discovery programs. Because of its greater throughput, 1-week turnaround time, and a substantially reduced (1-2 mg) requirement for compound consumption, the HCMN assay is appropriate for developing structure-genotoxicity relationships and for mechanistic genotoxicity studies. The assay does not replace the Organization for Economic Cooperation and Development-compliant, non-good laboratory practice in vitro MN test (e.g., slide-based MN test in TK6 lymphoblastoid cells) that is used for full characterization of lead candidates.
- Published
- 2010
32. Abstract B126: AG-881, a brain penetrant, potent, pan-mutant IDH (mIDH) inhibitor for use in mIDH solid and hematologic malignancies
- Author
-
Ingo K. Mellinghoff, Zenon D. Konteatis, Yue Chen, Zhihua Sui, Janeta Popovici-Muller, Erin Artin, Hyeryun Kim, Brandon Nicolay, Camelia Gliser, Carl Campos, Shinsan M. Su, Erica Tobin, Hua Yang, Kimberly Straley, Scott A. Biller, Andrew J. Olaharski, Raj Nagaraja, Katharine E. Yen, Lee Silverman, and Lenny Dang
- Subjects
0301 basic medicine ,Cancer Research ,IDH1 ,Chemistry ,Wild type ,medicine.disease ,IDH2 ,03 medical and health sciences ,030104 developmental biology ,Isocitrate dehydrogenase ,Oncology ,Cell culture ,In vivo ,Glioma ,Cancer research ,medicine ,IC50 - Abstract
AG-881 is an orally available, brain penetrant, potent, small-molecule inhibitor of isocitrate dehydrogenase (IDH) 1 and IDH2 mutant proteins. Small-molecule inhibition of the mutant IDH (mIDH) protein represents a targeted approach to cancer treatment for patients who harbor an IDH1 and/or an IDH2 mutation. Direct inhibition of the gain-of-function activity of the mIDH protein is intended to inhibit the production of the oncogenic metabolite D-2-hydroxyglutarate (2-HG) and induce tumor cell differentiation. Biochemical studies of AG-881 demonstrate that it has low nanomolar potency inhibition (IC50) against multiple mIDH homodimer and heterodimer enzymes. It is a rapid-equilibrium inhibitor of mIDH1-R132H and mIDH2-R172K homodimer enzymes and is a slow-binding inhibitor of mIDH2-R140Q homodimer and wild type (wt) IDH1/mIDH1-R132H, wtIDH2/mIDH2-R140Q, and wtIDH2/mIDH2-R172K heterodimers. The potency against mIDH1 and mIDH2 enzymes has also been shown in cell lines and primary human patient samples. The IC50 range for 2-HG inhibition by AG-881 was 0.04-22 nM in cells expressing mIDH1-R132C, mIDH1-R132G, mIDH1-R132H, or mIDH1-R132S mutations and was 7-14 nM and 130 nM in cells expressing mIDH2-R140Q and mIDH2-R172K mutations, respectively. The treatment of these mIDH cell lines or primary human acute myeloid leukemia samples with AG-881 led to the onset of cellular differentiation. The pharmacokinetics of AG-881 are characterized by rapid oral absorption and low total body plasma clearance in mice (0.406 L/hr/kg) and rats (0.289 L/hr/kg). Because of these favorable properties, a series of in vivo pharmacology studies were conducted with AG-881 in mouse xenograft models. In these studies, dose-response relationships for the reduction in 2-HG in tumor by AG-881 were established. Twice-daily dosing of AG-881 in the HT1080 (mIDH1-R132C) and U87 (mIDH2-R140Q) mouse models reduced tumor 2-HG levels by >96% at doses ≥30 mg/kg. In the orthotopic glioma model (mIDH1-R132H), brain tumor 2-HG levels were reduced by >97% at doses ≥0.1 mg/kg. Based on in vivo exposure-response analyses, plasma AG-881 AUC0-24hr values of 402 hr•ng/mL and 45,200 hr•ng/mL are projected to result in sustained 97% reduction in tumor 2-HG levels in the glioma indication and the non-glioma solid and liquid tumor indications, respectively. AG-881 also exhibits excellent brain penetration, with brain-to-plasma ratios ranging from 0.62 to 1.96 in mice and 1.11 to 1.48 in rats (based on AUC0-24hr), and has an acceptable preclinical safety profile that supports clinical testing. Taken together, these data show that AG-881 is a potent inhibitor of the mIDH1 and mIDH2 proteins and suppresses 2-HG production in enzymatic, cell-based, and in vivo systems. Pharmacology studies support that suppression of 2-HG levels by AG-881 results in alterations of cellular downstream markers, leading to a release from blockage of tumor cell differentiation. AG-881 is currently in phase 1 clinical development in patients with an IDH1 and/or IDH2-mutation who have advanced solid tumors including gliomas (ClinicalTrials.gov NCT02481154), and advanced hematologic malignancies (ClinicalTrials.gov NCT02492737). Citation Format: Katharine Yen, Zenon Konteatis, Zhihua Sui, Erin Artin, Lenny Dang, Kimberly Straley, Erica Tobin, Carl Campos, Hua Yang, Raj Nagaraja, Yue Chen, Hyeryun Kim, Camelia Gliser, Brandon Nicolay, Andrew Olaharski, Lee Silverman, Scott Biller, Shinsan M. Su, Ingo Mellinghoff, Janeta Popovici-Muller. AG-881, a brain penetrant, potent, pan-mutant IDH (mIDH) inhibitor for use in mIDH solid and hematologic malignancies [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr B126.
- Published
- 2018
33. Dapagliflozin, a Selective SGLT2 Inhibitor, Improves Glucose Homeostasis in Normal and Diabetic Rats
- Author
-
John R. Wetterau, Deborah Hagan, William N. Washburn, Wei Meng, Joseph R. Taylor, Songping Han, Scott A. Biller, Jean M. Whaley, and Li Xin
- Subjects
Blood Glucose ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Carbohydrate metabolism ,Sodium-Glucose Transport Proteins ,Cell Line ,chemistry.chemical_compound ,Sodium-Glucose Transporter 1 ,Glucosides ,Sodium-Glucose Transporter 2 ,Reference Values ,Internal medicine ,Diabetes mellitus ,Adipocytes ,Internal Medicine ,Animals ,Humans ,Medicine ,Glucose homeostasis ,Benzhydryl Compounds ,Cloning, Molecular ,Dapagliflozin ,Sodium-Glucose Transporter 2 Inhibitors ,business.industry ,Glucose transporter ,Glucose analog ,medicine.disease ,Rats ,Rats, Zucker ,Renal glucose reabsorption ,Glucose ,Endocrinology ,chemistry ,SGLT2 Inhibitor ,business - Abstract
OBJECTIVE—The inhibition of gut and renal sodium-glucose cotransporters (SGLTs) has been proposed as a novel therapeutic approach to the treatment of diabetes. We have identified dapagliflozin as a potent and selective inhibitor of the renal sodium-glucose cotransporter SGLT2 in vitro and characterized its in vitro and in vivo pharmacology. RESEARCH DESIGN AND METHODS—Cell-based assays measuring glucose analog uptake were used to assess dapagliflozin's ability to inhibit sodium-dependent and facilitative glucose transport activity. Acute and multi-dose studies in normal and diabetic rats were performed to assess the ability of dapagliflozin to improve fed and fasting plasma glucose levels. A hyperinsulinemic-euglycemic clamp study was performed to assess the ability of dapagliflozin to improve glucose utilization after multi-dose treatment. RESULTS—Dapagliflozin potently and selectively inhibited human SGLT2 versus human SGLT1, the major cotransporter of glucose in the gut, and did not significantly inhibit facilitative glucose transport in human adipocytes. In vivo, dapagliflozin acutely induced renal glucose excretion in normal and diabetic rats, improved glucose tolerance in normal rats, and reduced hyperglycemia in Zucker diabetic fatty (ZDF) rats after single oral doses ranging from 0.1 to 1.0 mg/kg. Once-daily dapagliflozin treatment over 2 weeks significantly lowered fasting and fed glucose levels at doses ranging from 0.1 to 1.0 mg/kg and resulted in a significant increase in glucose utilization rate accompanied by a significant reduction in glucose production. CONCLUSIONS—These data suggest that dapagliflozin has the potential to be an efficacious treatment for type 2 diabetes.
- Published
- 2008
34. Discovery, synthesis, and structure–activity studies of tetrazole based growth hormone secretagogues
- Author
-
David A. Gordon, Christa M. Musial, R. Krishna Seethala, Leah Giupponi, Kenneth E.J. Dickinson, Jeffrey A. Robl, Peter T. W. Cheng, Neil Flynn, Mark A. Smith, Dorothy Slusarchyk, Joseph A. Tino, Brian J. Murphy, Stephen G. Swartz, Gary J. Grover, Andres S. Hernandez, R.J. George, Daniel Longhi, and Scott A. Biller
- Subjects
medicine.drug_class ,Clinical Biochemistry ,Tetrazoles ,Pharmaceutical Science ,Carboxamide ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Dogs ,In vivo ,Drug Discovery ,medicine ,Animals ,Potency ,Structure–activity relationship ,Tetrazole ,Molecular Biology ,Chemistry ,Organic Chemistry ,In vitro ,Bioavailability ,Growth Hormone ,Molecular Medicine ,hormones, hormone substitutes, and hormone antagonists - Abstract
A novel class of Growth Hormone Secretagogues (GHS), based on a tetrazole template, has been discovered. In vitro SAR and in vivo potency within this new class of GHS are described. The tetrazole 9q exhibits good oral bioavailability in rats and dogs as well as efficacy following an oral 10 mg/kg dose in dogs. Solution and solid phase protocols for the synthesis of tetrazole based GHS have been developed.
- Published
- 2007
35. 2-Hydroxy-N-arylbenzenesulfonamides as ATP-citrate lyase inhibitors
- Author
-
Bang-Chi Chen, Neil Flynn, R. Michael Lawrence, Rulin Zhao, Ching-Hsuen Chu, James J. Li, Vito G. Sasseville, Scott A. Biller, Luping Chen, Randy Ponticiello, Haixia Wang, Dora M. Schnur, Dong Cheng, Jamil Haris, Jeffrey A. Robl, Kristen Pike, Joseph A. Tino, Mary T. Obermeier, Thomas Harrity, Timothy F. Herpin, and Ramesh Padmanabha
- Subjects
Blood Glucose ,ATP citrate lyase ,Clinical Biochemistry ,Pharmaceutical Science ,ATP Citrate (pro-S)-Lyase ,Biochemistry ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Animals ,Structure–activity relationship ,Enzyme Inhibitors ,Molecular Biology ,Cells, Cultured ,Triglycerides ,chemistry.chemical_classification ,Sulfonamides ,biology ,Triglyceride ,Cholesterol ,Body Weight ,Organic Chemistry ,Benzene ,Lyase ,Rats ,Enzyme ,Adipose Tissue ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Anti-Obesity Agents - Abstract
A novel series of 2-hydroxy-N-arylbenzenesulfonamides were identified to be ATP-citrate lyase (ACL) inhibitors with compound 9 displaying potent in vitro activity (IC(50)=0.13 microM). Chronic oral dosing of compound 9 in high-fat fed mice lowered plasma cholesterol, triglyceride, and glucose, as well as inhibited weight gain.
- Published
- 2007
36. ALDH2(E487K) mutation increases protein turnover and promotes murine hepatocarcinogenesis
- Author
-
Hui Lin, Charles Kung, Scott A. Biller, Zhizhong Lin, Rob A. Cairns, Gavin Histen, Tak W. Mak, Lizao Chen, Shengfang Jin, Shinsan M. Su, Jiang Chen, Hua Yang, Yufei Fu, Stefan Gross, Yiwei Chen, Yuxuan Lu, Yue Chen, Marion Dorsch, Xiujun Cai, Jeffrey Hixon, and Yong Cang
- Subjects
Proteasome Endopeptidase Complex ,Carcinoma, Hepatocellular ,Genotyping Techniques ,DNA damage ,Carcinogenesis ,Mutant ,Biology ,Hyperpigmentation ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Ethanol metabolism ,ALDH2 ,Skin ,Liver injury ,Multidisciplinary ,Polymorphism, Genetic ,Base Sequence ,Ethanol ,Protein Stability ,Aldehyde Dehydrogenase, Mitochondrial ,Liver Neoplasms ,Protein turnover ,Alcohol flush reaction ,Biological Sciences ,Aldehyde Dehydrogenase ,medicine.disease ,Molecular biology ,Immunohistochemistry ,Survival Analysis ,Mice, Inbred C57BL ,Amino Acid Substitution ,Liver ,Mutation ,Hepatocytes ,Mutant Proteins ,Liver cancer ,Alcoholic Intoxication - Abstract
Mitochondrial aldehyde dehydrogenase 2 (ALDH2) in the liver removes toxic aldehydes including acetaldehyde, an intermediate of ethanol metabolism. Nearly 40% of East Asians inherit an inactive ALDH2*2 variant, which has a lysine-for-glutamate substitution at position 487 (E487K), and show a characteristic alcohol flush reaction after drinking and a higher risk for gastrointestinal cancers. Here we report the characterization of knockin mice in which the ALDH2(E487K) mutation is inserted into the endogenous murine Aldh2 locus. These mutants recapitulate essentially all human phenotypes including impaired clearance of acetaldehyde, increased sensitivity to acute or chronic alcohol-induced toxicity, and reduced ALDH2 expression due to a dominant-negative effect of the mutation. When treated with a chemical carcinogen, these mutants exhibit increased DNA damage response in hepatocytes, pronounced liver injury, and accelerated development of hepatocellular carcinoma (HCC). Importantly, ALDH2 protein levels are also significantly lower in patient HCC than in peritumor or normal liver tissues. Our results reveal that ALDH2 functions as a tumor suppressor by maintaining genomic stability in the liver, and the common human ALDH2 variant would present a significant risk factor for hepatocarcinogenesis. Our study suggests that the ALDH2*2 allele-alcohol interaction may be an even greater human public health hazard than previously appreciated.
- Published
- 2015
37. FOCUS--Development of a Global Communication and Modeling Platform for Applied and Computational Medicinal Chemists
- Author
-
Mitsunori Kato, Maxim Totrov, Richard A. Lewis, Peter Gedeck, Scott A. Biller, Eric J. Martin, Lewis Whitehead, Brian Edward Vash, Clayton Springer, Katrin Spiegel, Donovan N. Chin, John H. Van Drie, Tommasi Ruben A, Yongjin Xu, Takanori Kanazawa, Peter Hunt, Valery Polyakov, Christoph L. Buenemann, Mika Lindvall, Nikolaus Stiefl, Eugene Raush, and Ruben Abagyan
- Subjects
business.industry ,Computer science ,General Chemical Engineering ,Chemistry, Pharmaceutical ,Communication ,Interactive 3d ,Computational Biology ,Usability ,General Chemistry ,Library and Information Sciences ,Machine learning ,computer.software_genre ,Ligands ,Computer Science Applications ,Software ,Time frame ,Cheminformatics ,Software deployment ,Drug Discovery ,Computer Simulation ,Artificial intelligence ,Design cycle ,Software engineering ,business ,computer - Abstract
Communication of data and ideas within a medicinal chemistry project on a global as well as local level is a crucial aspect in the drug design cycle. Over a time frame of eight years, we built and optimized FOCUS, a platform to produce, visualize, and share information on various aspects of a drug discovery project such as cheminformatics, data analysis, structural information, and design. FOCUS is tightly integrated with internal services that involve-among others-data retrieval systems and in-silico models and provides easy access to automated modeling procedures such as pharmacophore searches, R-group analysis, and similarity searches. In addition, an interactive 3D editor was developed to assist users in the generation and docking of close analogues of a known lead. In this paper, we will specifically concentrate on issues we faced during development, deployment, and maintenance of the software and how we continually adapted the software in order to improve usability. We will provide usage examples to highlight the functionality as well as limitations of FOCUS at the various stages of the development process. We aim to make the discussion as independent of the software platform as possible, so that our experiences can be of more general value to the drug discovery community.
- Published
- 2015
38. IDH2 mutation-induced histone and DNA hypermethylation is progressively reversed by small-molecule inhibition
- Author
-
Hua Yang, Stefanie Schalm, David P. Schenkein, Shinsan M. Su, Fang Wang, Kimberly Straley, Katharine E. Yen, Andrew Kernytsky, Wei Liu, Camelia Gliser, Marion Dorsch, Erica Hansen, Scott A. Biller, Jeremy Travins, Sam Agresta, and Stuart Murray
- Subjects
Chromatin Immunoprecipitation ,Immunology ,Biology ,medicine.disease_cause ,Biochemistry ,Histones ,Tandem Mass Spectrometry ,Cell Line, Tumor ,Histone H2A ,Histone methylation ,medicine ,Humans ,Cancer epigenetics ,Enzyme Inhibitors ,Principal Component Analysis ,Sulfonamides ,Myeloid Neoplasia ,Reverse Transcriptase Polymerase Chain Reaction ,Phenylurea Compounds ,fungi ,food and beverages ,Cell Biology ,Hematology ,DNA Methylation ,Molecular biology ,Isocitrate Dehydrogenase ,Leukemia, Myeloid, Acute ,Histone ,DNA demethylation ,Histone methyltransferase ,DNA methylation ,Mutation ,biology.protein ,Cancer research ,Carcinogenesis ,Transcriptome ,Chromatography, Liquid - Abstract
Mutations of IDH1 and IDH2, which produce the oncometabolite 2-hydroxyglutarate (2HG), have been identified in several tumors, including acute myeloid leukemia. Recent studies have shown that expression of the IDH mutant enzymes results in high levels of 2HG and a block in cellular differentiation that can be reversed with IDH mutant-specific small-molecule inhibitors. To further understand the role of IDH mutations in cancer, we conducted mechanistic studies in the TF-1 IDH2 R140Q erythroleukemia model system and found that IDH2 mutant expression caused both histone and genomic DNA methylation changes that can be reversed when IDH2 mutant activity is inhibited. Specifically, histone hypermethylation is rapidly reversed within days, whereas reversal of DNA hypermethylation proceeds in a progressive manner over the course of weeks. We identified several gene signatures implicated in tumorigenesis of leukemia and lymphoma, indicating a selective modulation of relevant cancer genes by IDH mutations. As methylation of DNA and histones is closely linked to mRNA expression and differentiation, these results indicate that IDH2 mutant inhibition may function as a cancer therapy via histone and DNA demethylation at genes involved in differentiation and tumorigenesis.
- Published
- 2015
39. Muraglitazar, a Novel Dual (α/γ) Peroxisome Proliferator–Activated Receptor Activator, Improves Diabetes and Other Metabolic Abnormalities and Preserves β-Cell Function in db/db Mice
- Author
-
Narayanan Hariharan, Kasim A. Mookhtiar, Liqun Gu, Jimmy Ren, Chen Sean, Ramakrishna Seethala, Michael Cap, Lori Kunselman, Min Zhou, Peter T. W. Cheng, William Fenderson, Chunning Shao, Cuixia Chu, Scott A. Biller, Hao Zhang, Fucheng Qu, Pratik Devasthale, Dennis Farrelly, Jeon Yoon T, Randolph P. Ponticiello, John R. Wetterau, Wei Wang, Wen-Pin Yang, Thomas Harrity, Aaron Tieman, Denis E. Ryono, and Richard E. Gregg
- Subjects
chemistry.chemical_classification ,medicine.medical_specialty ,Adiponectin ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Fatty liver ,Peroxisome proliferator-activated receptor ,White adipose tissue ,Biology ,medicine.disease ,Muraglitazar ,Endocrinology ,chemistry ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Rosiglitazone ,medicine.drug - Abstract
Muraglitazar, a novel dual (alpha/gamma) peroxisome proliferator-activated receptor (PPAR) activator, was investigated for its antidiabetic properties and its effects on metabolic abnormalities in genetically obese diabetic db/db mice. In db/db mice and normal mice, muraglitazar treatment modulates the expression of PPAR target genes in white adipose tissue and liver. In young hyperglycemic db/db mice, muraglitazar treatment (0.03-50 mg . kg(-1) . day(-1) for 2 weeks) results in dose-dependent reductions of glucose, insulin, triglycerides, free fatty acids, and cholesterol. In older hyperglycemic db/db mice, longer-term muraglitazar treatment (30 mg . kg(-1) . day(-1) for 4 weeks) prevents time-dependent deterioration of glycemic control and development of insulin deficiency. In severely hyperglycemic db/db mice, muraglitazar treatment (10 mg . kg(-1) . day(-1) for 2 weeks) improves oral glucose tolerance and reduces plasma glucose and insulin levels. In addition, treatment increases insulin content in the pancreas. Finally, muraglitazar treatment increases abnormally low plasma adiponectin levels, increases high-molecular weight adiponectin complex levels, reduces elevated plasma corticosterone levels, and lowers elevated liver lipid content in db/db mice. The overall conclusions are that in db/db mice, the novel dual (alpha/gamma) PPAR activator muraglitazar 1) exerts potent and efficacious antidiabetic effects, 2) preserves pancreatic insulin content, and 3) improves metabolic abnormalities such as hyperlipidemia, fatty liver, low adiponectin levels, and elevated corticosterone levels.
- Published
- 2006
40. Synthesis of d3-cerivastatin for use as internal standard in a LC/MS/MS method developed for quantitation of the drug in human serum
- Author
-
Jeffrey A. Robl, Peng Guo, Bang-Chi Chen, Huiping Zhang, Joseph E. Sundeen, Scott A. Biller, Mohammed Jemal, Mark S. Bednarz, Joel C. Barrish, and Balu Balasubramanian
- Subjects
Drug ,Chromatography ,Chemistry ,media_common.quotation_subject ,Organic Chemistry ,Cerivastatin ,Biochemistry ,High-performance liquid chromatography ,Quantitative determination ,Analytical Chemistry ,Drug Discovery ,Lc ms ms ,medicine ,Radiology, Nuclear Medicine and imaging ,Spectroscopy ,medicine.drug ,media_common - Abstract
d3-Cerivastatin was synthesized as an internal standard for use in a LC/MS/MS method developed for the simultaneous quantitative determination of the drug in human serum. d3-Cerivastatin was efficiently prepared on large scale from d3-iodomethane using improved procedures. Copyright © 2006 John Wiley & Sons, Ltd.
- Published
- 2006
41. Abstract PL02-04: IDH mutations and tumorigenicity
- Author
-
Véronique Saada, Katharine E. Yen, Virginie Penard-Lacronique, Frank Salituro, Shinsan Su, Samuel V. Agresta, Marion Dorsch, Scott A. Biller, Jeremy Travins, Kim Straley, Wentao Wei, Erin Artin, David P. Schenkein, Cyril Quivoron, Shunqi Yan, Janeta Popovici-Muller, Yi Gao, Stefan Gross, Fang Wang, Andrew Kernytsky, Elena Mylonas, S. de Botton, Erica Hansen, Stefanie Schalm, Stuart Murray, Byron DeLaBarre, Wei Liu, Jeffrey O. Saunders, Camelia Gliser, Hua Yang, and Lenny Dang
- Subjects
Cancer Research ,Isocitrate dehydrogenase ,Histone ,Oncology ,Cellular differentiation ,DNA methylation ,Mutant ,biology.protein ,Myeloid leukemia ,Epigenetics ,Biology ,Molecular biology ,IDH2 - Abstract
Mutations in the isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) genes are present in ∼20% of acute myeloid leukemia, and cause a neomorphic enzyme activity that results in the production of 2-hydroxyglutarate (2HG). Mutational and epigenetic profiling of a large patient cohort of acute myeloid leukemia (AML) has revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and impaired hematopoietic differentiation. To further investigate the intrinsic effect of 2HG on hematopoietic proliferation and differentiation, we transfected an erythroleukemia cell line (TF-1) with either IDH1 or IDH2 mutant alleles. These cells overexpress the mutant enzyme, have high levels of 2HG, and exhibit GM-CSF independent growth. Consistent with clinical observations, overexpression of the IDH mutant proteins led to hypermethylation of both histones and DNA. These results suggest that mutations in IDH1/2 could lead to epigenetic rewiring of cells that could facilitate the gain of function phenotype. We are currently studying the global and specific effects of IDH1/2 mutant overexpression to gain a broader understanding of the biological consequence of the IDH1/2 gain of function mutations. We have also generated mutation selective molecules that are capable of inhibiting IDHm enzymes. Upon compound treatment in vitro, we are able to reverse hypermethylation of both histones and DNA and induce cellular differentiation in IDHm cell lines and primary human IDHm AML patient samples(1, 2). These data suggest that an inhibitor of IDH1/2 mutations could correct the altered gene expression patterns seen in IDH1/2 mutant AML tumors leading to a profound effect on hematopoietic differentiation, proliferation and tumor growth. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):PL02-04. Citation Format: F. Wang, J. Travins, B. DeLaBarre, V. Penard-Lacronique, S. Schalm, E. Hansen, K. Straley, A. Kernytsky, W. Liu, C. Gliser, H. Yang, S. Gross, E. Artin, V Saada, E. Mylonas, C. Quivoron, J. Popovici-Muller, J. O. Saunders, F. G. Salituro, S. Yan, S. Murray, W. Wei, Y. Gao, L. Dang, M. Dorsch, S. Agresta, D. P. Schenkein, S. A. Biller, S. M. Su, S. de Botton, Katharine E. Yen. IDH mutations and tumorigenicity. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr PL02-04.
- Published
- 2013
42. 5-Carboxamido-1,3,2-dioxaphosphorinanes, potent inhibitors of MTP
- Author
-
John R. Wetterau, Thomas Harrity, Richard B. Sulsky, Jeffrey A. Robl, Kern G. Jolibois, Fergal Connolly, Scott A. Biller, and Lori Kunselman
- Subjects
Male ,medicine.drug_class ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Carboxamide ,Stereoisomerism ,In Vitro Techniques ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cricetinae ,Drug Discovery ,medicine ,Organic chemistry ,Animals ,Humans ,Structure–activity relationship ,Molecular Biology ,Chemistry ,Organic Chemistry ,Biological activity ,General Medicine ,Amides ,Cyclic P-Oxides ,Carrier protein ,Microsome ,Molecular Medicine ,Benzimidazoles ,Piperidine ,Carrier Proteins - Abstract
5-Carboxamido-1,3,2-dioxaphosphorinanes have been identified as potent inhibitors of microsomal triglyceride-transfer protein. The 1,3,2-dioxaphosphorine functionality acted as a neutral and stable replacement for piperidine and piperidine N-oxide.
- Published
- 2004
43. Population Pharmacokinetics and Pharmacodynamics of AG-519, a Pyruvate Kinase Activator for the Treatment of Pyruvate Kinase Deficiency, in Human Healthy Volunteers
- Author
-
Charles Kung, Penelope A. Kosinski, Kha Le, Scott A. Biller, Hua Yang, Marvin Cohen, Elizabeth Merica, and Ann J Barbier
- Subjects
education.field_of_study ,business.industry ,Immunology ,Population ,Cell Biology ,Hematology ,Pharmacology ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Pharmacokinetics ,Oral administration ,Pharmacodynamics ,medicine ,Glycolysis ,education ,business ,Adenosine triphosphate ,Pyruvate kinase ,Pyruvate kinase deficiency - Abstract
INTRODUCTION Pyruvate kinase (PK) deficiency is a life-long chronic hemolytic anemia with a variable clinical presentation, ranging from mild to life-threatening, and is associated with severe, debilitating co-morbidities. PK deficiency is caused by mutations in the PKLR gene, which in the red blood cell (RBC) results in defective pyruvate kinase isoform R (PK-R). PK-R catalyzes the final and irreversible step in glycolysis, the process on which mature RBCs rely almost exclusively to generate the energy carrier molecule, adenosine triphosphate (ATP). PK-R is thus a key enzyme for maintaining RBC energy levels, and it has been proposed that ATP levels are critical for optimally maintaining RBC membrane integrity. PK-deficient RBCs and their progenitors are characterized by changes in metabolism associated with defective glycolysis, including a build-up of phosphoenolpyruvate (PEP) and 2,3-diphosphoglycerate (2,3-DPG), and lowered ATP levels. AG-519 is a potent, highly selective and orally bioavailable PK-R activator shown preclinically to have none of the aromatase inhibitory effects that were observed with AG-348, the first small molecule PK-R activator to enter clinical trials. It is hypothesized that intervention with AG-519 restores glycolytic pathway activity and normalizes RBC metabolism. Treatment of PK-deficient patient RBCs ex vivo with AG-519 results in increased ATP levels, and reductions in 2,3-DPG, consistent with pharmacological activation of PK-R enzyme activity. This analysis integrates the pharmacokinetic and pharmacodynamic (PK/PD) properties of AG-519 in healthy volunteers using population PK/PD modeling and simulation. METHODS PK/PD modeling using a non-linear mixed effects approach was performed to understand the pharmacokinetics of AG-519 and PK/PD relationship of AG-519 to 2,3-DPG, ATP and PK-R enzyme activity in humans. The PK/PD model integrated data from a phase 1, single-center, randomized, double-blind, placebo-controlled study. Data in single and multiple ascending dose escalation arms were included in the analysis, which comprises a total of 88 human volunteers. AG-519 dose levels ranged from 10-1250 mg. Blood was collected from all patients to assess AG-519 pharmacokinetics and levels of ATP and 2,3-DPG in blood, as well as PK-R activity. Population simulations using the final model were performed to examine the dose-exposure-biomarkers relationship. RESULTS AG-519 showed rapid absorption kinetics following oral administration. Area under the concentration-time curve of plasma AG-519 increased in a dose-proportional manner following single and multiple dosing. A three-compartment model with non-linear absorption compartment and linear elimination best described the pharmacokinetics of AG-519. Systemic clearance appeared to be time-invariant and no auto-induction was observed with multiple dosing. The PK/PD relationship between plasma AG-519 and ATP or 2,3-DPG in human volunteers was best fitted by a turnover model where the drug effect was described by an Emax model. A direct Emax model best described the relationship between plasma AG-519 and PK-R enzyme activity. Evaluation of ATP and 2,3-DPG levels and PK-R activity confirmed the potent effect of AG-519 on PK-R. A direct comparison of AG-519 target engagement and biomarker response to AG-348 will also be presented. Population PK/PD model simulations suggested that the doses selected in the phase 1 study covered a large exposure-response range of the three biomarkers, and provided a systematic and integrated framework for the understanding of AG-519 pharmacokinetics and pharmacodynamics, as well as a rationale for dose selection in future trials. CONCLUSIONS This study represents a comprehensive longitudinal PK/PD analysis of AG-519 in humans. This integrated PK/PD model formed the basis for understanding the exposure-response relationship of the ongoing phase 1 study and provided guidance on dose selection to inform the future development of AG-519. Disclosures Le: Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Cohen:Agios Pharmaceuticals, Inc.: Consultancy. Barbier:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Merica:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Kung:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Kosinski:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Biller:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership. Yang:Agios Pharmaceuticals, Inc.: Employment, Equity Ownership.
- Published
- 2016
44. A Novel Series of Highly Potent Benzimidazole-Based Microsomal Triglyceride Transfer Protein Inhibitors
- Author
-
David A. Gordon, Ligaya M. Simpkins, William A. Slusarchyk, David R. Magnin, Chongqing Sun, Prakash Taunk, Jamil Haris, Lori Kunselman, Ying Chen, Scott A. Biller, John R. Wetterau, Fergal Connolly, Jeffrey A. Robl, John K. Dickson, Talal Sabrah, Thomas Harrity, Shih-Jung Lan, Richard B. Sulsky, and Tammy C. Wang
- Subjects
Benzimidazole ,Very low-density lipoprotein ,medicine.drug_class ,Administration, Oral ,Biological Availability ,Carboxamide ,Microsomal triglyceride transfer protein ,Cell Line ,Rats, Sprague-Dawley ,Structure-Activity Relationship ,chemistry.chemical_compound ,In vivo ,Cricetinae ,Microsomes ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Triglycerides ,Apolipoproteins B ,Hypolipidemic Agents ,Fluorenes ,biology ,Cholesterol ,Anticholesteremic Agents ,Rats ,Lipoproteins, LDL ,Macaca fascicularis ,chemistry ,Biochemistry ,biology.protein ,Microsome ,Molecular Medicine ,Benzimidazoles ,lipids (amino acids, peptides, and proteins) ,Carrier Proteins - Abstract
A series of benzimidazole-based analogues of the potent MTP inhibitor BMS-201038 were discovered. Incorporation of an unsubstituted benzimidazole moiety in place of a piperidine group afforded potent inhibitors of MTP in vitro which were weakly active in vivo. Appropriate substitution on the benzimidazole ring, especially with small alkyl groups, led to dramatic increases in potency, both in a cellular assay of apoB secretion and especially in animal models of cholesterol lowering. The most potent in this series, 3g (BMS-212122), was significantly more potent than BMS-201038 in reducing plasma lipids (cholesterol, VLDL/LDL, TG) in both hamsters and cynomolgus monkeys.
- Published
- 2001
45. An MTP Inhibitor That Normalizes Atherogenic Lipoprotein Levels in WHHL Rabbits
- Author
-
Douglas Young, Michael Cap, Olga M. Fryszman, Thomas J. Maccagnan, Mujing Yan, John R. Wetterau, Prakash Taunk, Shih-Jung Lan, Richard B. Sulsky, Michael A. Poss, William A. Slusarchyk, Fergal Connolly, Jeffrey A. Robl, Joseph A. Tino, Kern G. Jolibois, R.J. George, Beverly Ricci, Scott A. Biller, David R. Magnin, Janette V. H. Logan, Thomas Harrity, Lori Kunselman, Ying Chen, R. Michael Lawrence, Jamil Haris, C.M. Arbeeny, John K. Dickson, Christa L. Musial, Ligaya M. Simpkins, Ching-Hsuen Chu, David A. Gordon, and Richard E. Gregg
- Subjects
medicine.medical_specialty ,Apolipoprotein B ,Lipoproteins ,Drug Evaluation, Preclinical ,Hyperlipidemias ,Familial hypercholesterolemia ,Microsomal triglyceride transfer protein ,Hyperlipoproteinemia Type II ,Mice ,chemistry.chemical_compound ,Piperidines ,In vivo ,Cricetinae ,Internal medicine ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Secretion ,Aspartate Aminotransferases ,Triglycerides ,Apolipoproteins B ,Fluorenes ,Multidisciplinary ,Dose-Response Relationship, Drug ,biology ,Triglyceride ,Abetalipoproteinemia ,Alanine Transaminase ,medicine.disease ,Lipids ,Rats ,Disease Models, Animal ,Cholesterol ,Endocrinology ,Liver ,chemistry ,Drug Design ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Rabbits ,Carrier Proteins ,Lipoprotein - Abstract
Patients with abetalipoproteinemia, a disease caused by defects in the microsomal triglyceride transfer protein (MTP), do not produce apolipoprotein B–containing lipoproteins. It was hypothesized that small molecule inhibitors of MTP would prevent the assembly and secretion of these atherogenic lipoproteins. To test this hypothesis, two compounds identified in a high-throughput screen for MTP inhibitors were used to direct the synthesis of a highly potent MTP inhibitor. This molecule (compound 9 ) inhibited the production of lipoprotein particles in rodent models and normalized plasma lipoprotein levels in Watanabe-heritable hyperlipidemic (WHHL) rabbits, which are a model for human homozygous familial hypercholesterolemia. These results suggest that compound 9 , or derivatives thereof, has potential applications for the therapeutic lowering of atherogenic lipoprotein levels in humans.
- Published
- 1998
46. Evidence that microsomal triglyceride transfer protein is limiting in the production of apolipoprotein B-containing lipoproteins in hepatic cells
- Author
-
David A. Gordon, Mujing Yan, John R. Wetterau, Ching-Hsuen Chu, John K. Dickson, Richard E. Gregg, Ying Chen, Jamil Haris, and Scott A. Biller
- Subjects
photoaffinity ,McArdle RH-7777 cells ,Apolipoprotein B ,biology ,Triglyceride ,Chemistry ,Glyceride ,Cell Biology ,lipid transfer protein ,QD415-436 ,protein disulfide isomerase ,Biochemistry ,Microsomal triglyceride transfer protein ,chemistry.chemical_compound ,Endocrinology ,biology.protein ,Hepatic stellate cell ,Microsome ,Secretion ,lipids (amino acids, peptides, and proteins) ,Plant lipid transfer proteins ,HepG2 cells ,benzophenone - Abstract
The microsomal triglyceride transfer protein (MTP) is a heterodimeric lipid transfer protein that is re- quired for the assembly and secretion of apolipoprotein B (apoB)-containing lipoproteins. A key unresolved question is whether the MTP-mediated step is rate limiting. To ad- dress this, a unique experimental strategy was used that al- lowed the in situ modulation and measurement of MTP tri- glyceride transfer activity. In order to accomplish this, an irreversible photoaffinity inhibitor, BMS-192951, was de- signed and synthesized. When incubated with purified MTP and irradiated with UV light at 360 nm, BMS-192951 inhib- its triglyceride transfer by covalently binding to the protein. HepG2 cells were treated with either increasing concentra- tions of BMS-192951 (0-15 m m ) with 5 min of ultraviolet ir- radiation, or 3.0 m m BMS-192951 with various lengths (0-15 min) of ultraviolet irradiation. Microsomal extracts were prepared exhaustively dialyzed to remove unbound inhibi- tor, and assayed for MTP-mediated triglyceride transfer ac- tivity. BMS-192951 was shown to reduce MTP activity in both a dose- and UV exposure time-dependent fashion. Measurement of apoB concentration in the media showed that apoB secretion was reduced in proportion to the in situ inhibition of MTP activity, while no change was observed in apoA-I secretion. Experiments performed in McArdle RH- 7777 rat hepatoma cells and primary rat hepatocytes gave nearly identical results; the decrease in apoB secretion was proportional to the decrease in MTP activity. These re- sults indicate that MTP-mediated lipid transfer is limiting in the assembly and secretion of apoB-containing lipoproteins in hepatic cells under the conditions tested.— Jamil, H., C-H. Chu, J. K. Dickson, Jr., Y. Chen, M. Yan, S. A. Biller, R. E. Gregg, J. R. Wetterau, and D. A. Gordon. Evidence that mi- crosomal triglyceride transfer protein is limiting in the pro- duction of apolipoprotein B-containing lipoproteins in he- patic cells. J. Lipid Res. 1998. 39: 1448-1454.
- Published
- 1998
47. Automated Synthesis and Purification of Amides: Exploitation of Automated Solid Phase Extraction in Organic Synthesis
- Author
-
R. Michael Lawrence, Michael A. Poss, Olga M. Fryszman, and Scott A. Biller
- Subjects
chemistry.chemical_compound ,Ion exchange ,Chemistry ,Organic Chemistry ,Organic synthesis ,Solid phase extraction ,Combinatorial chemistry ,Catalysis - Published
- 1997
48. A solid phase synthesis of miconazole analogs via an iodoetherification reaction
- Author
-
Scott A. Biller and David R. Tortolani
- Subjects
Solid-phase synthesis ,Chemistry ,Organic Chemistry ,Drug Discovery ,medicine ,Miconazole ,Combinatorial synthesis ,Biochemistry ,Combinatorial chemistry ,medicine.drug - Abstract
A procedure for the preparation of various analogs of miconazole on solid support is described. A novel iodoetherification transformation is utilized as the key synthetic step. This approach has been applied to the combinatorial synthesis of 45 analogs.
- Published
- 1996
49. Squalene Synthase Inhibitors
- Author
-
Scott A. Biller, Kent Neuenschwander, Mitree M. Ponpipomt, and C. Dale Poulter
- Subjects
Pharmacology ,Drug Discovery - Abstract
Abstract: Squalene synthase catalyzes the reductive dimerization of famesyl diphosphate (C-15) to form squalene, the C-30 polyisoprene precursor to cholesterol. In recent years, the search for inhibitors of squalene synthase for use as antihypercholesterolemic agents has intensified. The focus on this enzymatic transformation is due to the unique location of squalene synthase at the first step committed to sterols in the isoprene pathway. Selective inhibitors of this enzyme are expected to block cholesterol biosynthesis, without having deleterious effects on the branch pathways of isoprene biosynthesis. Potent inhibitors of squalene synthase have been discovered utilizing three of the major methods for drug discovery: rational design, synthetic chemical screening and natural products screening. These inhibitors have been shown to be potent and effective cholesterol lowering agents in animal models. This article will review the rationale for squalene synthase as a drug discovery target, recent advances in the biology of squalene synthase, and the progress toward clinically useful inhibitors of this key enzyme.
- Published
- 1996
50. Orally Active Squalene Synthase Inhibitors: Bis((acyloxy)alkyl) Prodrugs of the α-Phosphonosulfonic Acid Moiety
- Author
-
J. K. Rinehart, Shih-Jung Lan, Kern G. Jolibois, Richard E. Gregg, Lori Kunselman, John K. Dickson, D. C. Hsieh, Edward W. Petrillo, J. W. Hillyer, Stephen S. Kalinowski, T. W. Harrity, Kasim A. Mookhtiar, D. R. Magnin, Carl P. Ciosek, and Scott A. Biller
- Subjects
chemistry.chemical_classification ,Farnesyl-diphosphate farnesyltransferase ,biology ,Stereochemistry ,Guinea Pigs ,Administration, Oral ,Ether ,Prodrug ,Chemical synthesis ,Rats ,chemistry.chemical_compound ,Squalene ,Farnesyl-Diphosphate Farnesyltransferase ,chemistry ,Enzyme inhibitor ,Drug Discovery ,biology.protein ,Animals ,Molecular Medicine ,Moiety ,Prodrugs ,Enzyme Inhibitors ,Sulfonic Acids ,Alkyl ,Hypolipidemic Agents - Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.