20 results on '"Tatyana Zvyaga"'
Search Results
2. Discovery of BMS-986144, a Third-Generation, Pan-Genotype NS3/4A Protease Inhibitor for the Treatment of Hepatitis C Virus Infection
- Author
-
Nicholas A. Meanwell, Ravi Kumar Trivedi, Tatyana Zvyaga, Richard Rampulla, Sarkunam Kandhasamy, Debarati Mazumder Tagore, Sureshbabu Vishwakrishnan, Arvind Mathur, Sunitha Puttaswamy, Abhijith Rao, Hua Fang, Sankar Sivaprasad, Susan Jenkins, Eric Mull, Kathy Mosure, Kaushik Ghosh, Barbara Zheng, Subba Reddy, Fiona McPhee, Chaoqun Chen, Bowsher Michael S, James Loy, Ying-Kai Wang, Amit Kumar, Ramkumar Rajamani, Kishore Rendunchintala, Paul Michael Scola, Gillis Eric P, Salil D. Desai, Li-Qiang Sun, Rushith Kumar Anumula, Nagalakshmi Pulicharla, Sheldon Hiebert, Venkata Rao Baratam, Paul Falk, Sarmistha Sinha, and Stanley D'andrea
- Subjects
Serine Proteinase Inhibitors ,viruses ,Hepatitis C virus ,CHO Cells ,Hepacivirus ,Microbial Sensitivity Tests ,Tripeptide ,Viral Nonstructural Proteins ,medicine.disease_cause ,Antiviral Agents ,Peptides, Cyclic ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,Cricetulus ,Drug Stability ,Drug Discovery ,Genotype ,medicine ,Animals ,030304 developmental biology ,0303 health sciences ,NS3 ,Molecular Structure ,Chemistry ,virus diseases ,Virology ,digestive system diseases ,Third generation ,Rats ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Microsomes, Liver ,Molecular Medicine ,Ns3 4a protease - Abstract
The discovery of a pan-genotypic hepatitis C virus (HCV) NS3/4A protease inhibitor based on a P1–P3 macrocyclic tripeptide motif is described. The all-carbon tether linking the P1–P3 subsites of 21...
- Published
- 2020
- Full Text
- View/download PDF
3. Identification and Preclinical Evaluation of the Bicyclic Pyrimidine γ-Secretase Modulator BMS-932481
- Author
-
John E. Macor, Yunhui Zhang, James E. Grace, Lorin A. Thompson, Jeremy H. Toyn, Lawrence R. Marcin, Subramaniam Krishnananthan, Jianliang Shi, Dmitry Zuev, Daniel Smith, Jianqing Li, Yong-Jin Wu, Jason M. Guernon, Xiaoliang Zhuo, Tatyana Zvyaga, Xu Li, Arvind Mathur, John Morrison, Jere E. Meredith, Charles F. Albright, S. Roy Kimura, Mendi A. Higgins, Kimberley A. Lentz, Richard E. Olson, Rex Denton, Kenneth M. Boy, Catherine R. Burton, Ashok K. Trehan, and Michael K. Ahlijanian
- Subjects
gamma-secretase modulator ,Letter ,Bicyclic molecule ,Pyrimidine ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,bicyclic pyrimidine ,clinical candidate ,γ secretase modulator ,Pharmacology ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,Cerebrospinal fluid ,In vivo ,Drug Discovery ,Toxicity ,Potency ,Alzheimer’s disease ,Triazine - Abstract
A triazine hit identified from a screen of the BMS compound collection was optimized for potency, in vivo activity, and off-target profile to produce the bicyclic pyrimidine γ-secretase modulator BMS-932481. The compound showed robust reductions of Aβ1–42 and Aβ1–40 in the plasma, brain, and cerebrospinal fluid of mice and rats. Consistent with the γ-secretase modulator mechanism, increases in Aβ1–37 and Aβ1–38 were observed, with no change in the total amount of Aβ1–x produced. No Notch-based toxicity was observed, and the overall preclinical profile of BMS-932481 supported its further evaluation in human clinical trials.
- Published
- 2019
4. Development of BET inhibitors as potential treatments for cancer: A new carboline chemotype
- Author
-
Andrew P. Degnan, Haiquan Fang, Tatyana Zvyaga, Zuzana Haarhoff, Claude A. Quesnelle, Melissa Kramer, Frank Marsilio, Richard A. Westhouse, John S. Tokarski, Shilpa Madari, Jiuqiao Zhao, Amy Wiebesiek, Chunhong Yan, Francis Y. Lee, Michael Sinz, John Morrison, Jean Simmermacher-Mayer, Carolynn Fanslau, Steven Sheriff, Lisa Huang, Ashvinikumar V. Gavai, Chunshan Xie, Matthew D. Hill, and Gerry Everlof
- Subjects
Stereochemistry ,Cell Survival ,Clinical Biochemistry ,Pharmaceutical Science ,Structural diversity ,Administration, Oral ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,Biochemistry ,Polar surface area ,BET inhibitor ,Mice ,Structure-Activity Relationship ,Drug Development ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Biology ,Chemotype ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Cancer ,Mammary Neoplasms, Experimental ,Proteins ,medicine.disease ,In vitro ,Bromodomain ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Carbolines - Abstract
We describe our efforts to introduce structural diversity to a previously described triazole-containing N1-carboline series of bromodomain and extra-terminal (BET) inhibitors. N9 carbolines were designed to retain favorable binding interactions that the N1-carbolines possess. A convergent synthetic route enabled modifications to reduce clearance, enhance physicochemical properties, and improve the overall in vitro profile. This work led to the identification of a potent BET inhibitor, (S)-2‐{8‐fluoro‐5‐[(3‐fluoropyridin‐2‐yl)(oxan‐4‐yl)methyl]‐7‐[4‐(2H3)methyl‐1‐methyl‐1H‐1,2,3‐triazol‐5‐yl]‐5H‐pyrido[3,2‐b]indol‐3‐yl}propan‐2‐ol (10), a compound with enhanced oral exposure in mice. Subsequent evaluation in a mouse triple-negative breast cancer tumor model revealed efficacy at 4 mg/kg of N9-carboline 10.
- Published
- 2021
5. Development of BET inhibitors as potential treatments for cancer: A search for structural diversity
- Author
-
Chunhong Yan, Francis Y. Lee, Ashvinikumar V. Gavai, Melissa Kramer, Frank Marsilio, Eric Shields, Laura Monereau, Zuzana Haarhoff, Richard A. Westhouse, Lisa Zhang, Gerry Everlof, John S. Tokarski, Matthew D. Hill, Tatyana Zvyaga, Asoka Ranasinghe, Carolynn Fanslau, Ching Kim Tye, Steven Sheriff, Christine Huang, Andrew P. Degnan, Krista Menard, John Morrison, and Haiquan Fang
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Structural diversity ,Antineoplastic Agents ,Biochemistry ,BET inhibitor ,Mice ,Structure-Activity Relationship ,Drug Development ,Drug Discovery ,medicine ,Animals ,Humans ,Moiety ,Potency ,Mouse tumor ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Proteins ,Cancer ,Triazoles ,medicine.disease ,Combinatorial chemistry ,Bromodomain ,Molecular Medicine ,Multiple Myeloma ,Carbolines - Abstract
We describe our efforts to identify structurally diverse leads in the triazole-containing N1-carboline series of bromodomain and extra-terminal inhibitors. Replacement of the N5 “cap” phenyl moiety with various heteroaryls, coupled with additional modifications to the carboline core, provided analogs with similar potency, improved pharmacokinetic properties, and increased solubility compared to our backup lead, BMS-986225 (2). Rapid SAR exploration was enabled by a convergent, synthetic route. These efforts provided a potent BET inhibitor, 3-fluoropyridyl 12, that demonstrated robust efficacy in a multiple myeloma mouse tumor model at 1 mg/kg.
- Published
- 2021
- Full Text
- View/download PDF
6. Improving Metabolic Stability with Deuterium: The Discovery of BMT-052, a Pan-genotypic HCV NS5B Polymerase Inhibitor
- Author
-
Dawn D. Parker, Karen Rigat, Kyle J. Eastman, Umesh Hanumegowda, Katharine A. Grant-Young, Nicholas A. Meanwell, Maria Tuttle, Tao Wang, Tatyana Zvyaga, Susan B. Roberts, Hua Fang, Kap-Sun Yeung, Mengping Liu, Julie A. Lemm, Kathy Mosure, Xiaoliang Zhuo, Maria Donoso, Kyle Parcella, Zhongxing Zhang, Matthew G. Soars, Zuzana Haarhoff, Zhiwei Yin, Ying-Kai Wang, John F. Kadow, and Juliang Zhu
- Subjects
0301 basic medicine ,010405 organic chemistry ,Hepatitis C virus ,Organic Chemistry ,Metabolic stability ,Biology ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,Virology ,Ns5b polymerase ,0104 chemical sciences ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Genotype ,medicine ,Primer (molecular biology) - Abstract
Iterative structure–activity analyses in a class of highly functionalized furo[2,3-b]pyridines led to the identification of the second generation pan-genotypic hepatitis C virus NS5B polymerase primer grip inhibitor BMT-052 (14), a potential clinical candidate. The key challenge of poor metabolic stability was overcome by strategic incorporation of deuterium at potential metabolic soft spots. The preclinical profile and status of BMT-052 (14) is described.
- Published
- 2017
- Full Text
- View/download PDF
7. Coupling Laser Diode Thermal Desorption with Acoustic Sample Deposition to Improve Throughput of Mass Spectrometry–Based Screening
- Author
-
Dieter M. Drexler, Andrew Dennis Wagner, Zuzana Haarhoff, Pierre Picard, Tatyana Zvyaga, and Wilson Z. Shou
- Subjects
Analytical chemistry ,Thermal desorption ,Atmospheric-pressure chemical ionization ,Tandem mass spectrometry ,Mass spectrometry ,01 natural sciences ,Biochemistry ,Analytical Chemistry ,law.invention ,Tandem Mass Spectrometry ,law ,Ionization ,Drug Discovery ,Cytochrome P-450 Enzyme Inhibitors ,Solid phase extraction ,Chromatography ,Laser diode ,Chemistry ,Solid Phase Extraction ,010401 analytical chemistry ,Far-infrared laser ,High-Throughput Screening Assays ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Calibration ,Molecular Medicine ,Biological Assay ,Biotechnology - Abstract
The move toward label-free screening in drug discovery has increased the demand for mass spectrometry (MS)-based analysis. Here we investigated the approach of coupling acoustic sample deposition (ASD) with laser diode thermal desorption (LDTD)-tandem mass spectrometry (MS/MS). We assessed its use in a cytochrome P450 (CYP) inhibition assay, where a decrease in metabolite formation signifies CYP inhibition. Metabolite levels for 3 CYP isoforms were measured as CYP3A4-1'-OH-midazolam, CYP2D6-dextrorphan, and CYP2C9-4'-OH-diclofenac. After incubation, samples (100 nL) were acoustically deposited onto a stainless steel 384-LazWell plate, then desorbed by an infrared laser directly from the plate surface into the gas phase, ionized by atmospheric pressure chemical ionization (APCI), and analyzed by MS/MS. Using this method, we achieved a sample analysis speed of 2.14 s/well, with bioanalytical performance comparable to the current online solid-phase extraction (SPE)-based MS method. An even faster readout speed was achieved when postreaction sample multiplexing was applied, where three reaction samples, one for each CYP, were transferred into the same well of the LazWell plate. In summary, LDTD coupled with acoustic sample deposition and multiplexing significantly decreased analysis time to 0.7 s/sample, making this MS-based approach feasible to support high-throughput screening (HTS) assays.
- Published
- 2016
- Full Text
- View/download PDF
8. Development, optimization and implementation of a centralized metabolic soft spot assay
- Author
-
Cheryl Klakouski, Wilson Z. Shou, Yue-Zhong Shu, Ismael Zamora, Benjamin M. Johnson, Anthony A. Paiva, Tatyana Zvyaga, Shu Li, and Jonathan L. Josephs
- Subjects
0301 basic medicine ,Computer science ,Clinical Biochemistry ,Computational biology ,Bioinformatics ,01 natural sciences ,Turnaround time ,Analytical Chemistry ,03 medical and health sciences ,Mice ,Tandem Mass Spectrometry ,Drug Discovery ,Animals ,Humans ,Metabolomics ,General Pharmacology, Toxicology and Pharmaceutics ,Liver microsomes ,Drug discovery ,010401 analytical chemistry ,General Medicine ,Metabolic stability ,0104 chemical sciences ,Rats ,Medical Laboratory Technology ,Kinetics ,030104 developmental biology ,Microsomes, Liver ,Biological Assay ,Software ,Chromatography, Liquid - Abstract
Aim: High clearance is a commonly encountered issue in drug discovery. Here we present a centralized metabolic soft spot identification assay with adequate capacity and turnaround time to support the metabolic optimization needs of an entire discovery organization. Methodology: An integrated quan/qual approach utilizing both an orthogonal sample-pooling methodology and software-assisted structure elucidation was developed to enable the assay. Major metabolic soft spots in liver microsomes (rodent and human) were generated in a batch mode, along with kinetics of parent disappearance and metabolite formation, typically within 1 week of incubation. Results & conclusion: A centralized metabolic soft spot identification assay has been developed and has successfully impacted discovery project teams in mitigating instability and establishing potential structure–metabolism relationships.
- Published
- 2017
9. The Discovery of Asunaprevir (BMS-650032), An Orally Efficacious NS3 Protease Inhibitor for the Treatment of Hepatitis C Virus Infection
- Author
-
Danshi Li, Herbert E. Klei, Sing-Yuen Sit, Piyasena Hewawasam, Ny Sin, Brian Lee Venables, Paul Levesque, Qi Gao, Bilder Donna M, Dennis M. Grasela, Dennis Hernandez, Anthony J. Cocuzza, Amy K. Sheaffer, Richard J. Colonno, Jeffrey Tredup, Kathy Mosure, Stanley D'andrea, Jialong Zhu, Lucy Sun, Fei Yu, Paul Falk, Alan Xiangdong Wang, Steven Levine, Fiona McPhee, Andrew C. Good, Huabin Sun, Jay O. Knipe, Barbara Zheng, Richard Schartman, Nicholas A. Meanwell, Jie Chen, Maria Donoso, James Loy, Guangzhi Zhai, Ramkumar Rajamani, Li-Qiang Sun, Stephen P. Adams, Luciano Mueller, Tatyana Zvyaga, Yong Tu, Hong Shi, Paul Michael Scola, Chaoqun Chen, William Warner, Yan Chen, Yong-Hae Han, Diana Barry, Jacques Friborg, Kevin Kish, and Michael Sinz
- Subjects
Models, Molecular ,Hepatitis C virus ,Phases of clinical research ,Viral Nonstructural Proteins ,Biology ,Pharmacology ,medicine.disease_cause ,Antiviral Agents ,chemistry.chemical_compound ,Dogs ,Drug Discovery ,medicine ,Animals ,Humans ,Protease Inhibitors ,Protease inhibitor (pharmacology) ,Beclabuvir ,Sulfonamides ,NS3 ,Isoquinolines ,Hepatitis C ,Virology ,Rats ,Discontinuation ,Clinical trial ,chemistry ,Molecular Medicine ,Asunaprevir ,Rabbits - Abstract
The discovery of asunaprevir (BMS-650032, 24) is described. This tripeptidic acylsulfonamide inhibitor of the NS3/4A enzyme is currently in phase III clinical trials for the treatment of hepatitis C virus infection. The discovery of 24 was enabled by employing an isolated rabbit heart model to screen for the cardiovascular (CV) liabilities (changes to HR and SNRT) that were responsible for the discontinuation of an earlier lead from this chemical series, BMS-605339 (1), from clinical trials. The structure-activity relationships (SARs) developed with respect to CV effects established that small structural changes to the P2* subsite of the molecule had a significant impact on the CV profile of a given compound. The antiviral activity, preclincial PK profile, and toxicology studies in rat and dog supported clinical development of BMS-650032 (24).
- Published
- 2014
- Full Text
- View/download PDF
10. Discovery and Early Clinical Evaluation of BMS-605339, a Potent and Orally Efficacious Tripeptidic Acylsulfonamide NS3 Protease Inhibitor for the Treatment of Hepatitis C Virus Infection
- Author
-
Alan Xiangdong Wang, Barbara Zheng, Min S. Lee, Dennis Hernandez, Andrew C. Good, Stanley D'andrea, Amy K. Sheaffer, Fiona McPhee, Christoph Gesenberg, Sing-Yuen Sit, Ny Sin, Paul Falk, Danshi Li, Piyasena Hewawasam, Yan Chen, Yong-Hae Han, Paul Levesque, Brian Lee Venables, Nicholas A. Meanwell, Michael Sinz, Wenying Li, Qi Gao, Li-Qiang Sun, Jay O. Knipe, Chaoqun Chen, Caly Chien, Diana Barry, Guangzhi Zhai, Anthony J. Cocuzza, Tatyana Zvyaga, Stephen P. Adams, Jialong Zhu, Fei Yu, Dennis M. Grasela, Richard J. Colonno, Yong Tu, Ramkumar Rajamani, Herbert E. Klei, Jianqing Li, Min Ding, Jan Willem Thuring, Jie Chen, Susan Jenkins, Keith D. Combrink, Jeffrey Allen Campbell, Kenneth S. Santone, Paul Michael Scola, Eric Hughes, and Bilder Donna M
- Subjects
Models, Molecular ,Stereochemistry ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Viral Nonstructural Proteins ,Pharmacology ,Crystallography, X-Ray ,Antiviral Agents ,chemistry.chemical_compound ,Dogs ,Pharmacokinetics ,Drug Discovery ,medicine ,Animals ,Humans ,Moiety ,Potency ,Protease Inhibitors ,Protease inhibitor (pharmacology) ,Isoquinoline ,Sulfonamides ,NS3 ,Protease ,Chemistry ,Drug discovery ,Isoquinolines ,Hepatitis C ,Molecular Medicine - Abstract
The discovery of BMS-605339 (35), a tripeptidic inhibitor of the NS3/4A enzyme, is described. This compound incorporates a cyclopropylacylsulfonamide moiety that was designed to improve the potency of carboxylic acid prototypes through the introduction of favorable nonbonding interactions within the S1' site of the protease. The identification of 35 was enabled through the optimization and balance of critical properties including potency and pharmacokinetics (PK). This was achieved through modulation of the P2* subsite of the inhibitor which identified the isoquinoline ring system as a key template for improving PK properties with further optimization achieved through functionalization. A methoxy moiety at the C6 position of this isoquinoline ring system proved to be optimal with respect to potency and PK, thus providing the clinical compound 35 which demonstrated antiviral activity in HCV-infected patients.
- Published
- 2014
- Full Text
- View/download PDF
11. The discovery of a pan-genotypic, primer grip inhibitor of HCV NS5B polymerase
- Author
-
Katharine A. Grant-Young, Maria Tuttle, Nicholas A. Meanwell, Susan B. Roberts, Maria Donoso, Kap-Sun Yeung, Ying-Kai Wang, Matthew G. Soars, Julie A. Lemm, Vivek Halan, Tatyana Zvyaga, Kyle J. Eastman, Jeffrey Tredup, Zuzana Haarhoff, Karen Rigat, Xiaoliang Zhuo, Umesh Hanumegowda, Steven Sheriff, Kathy Mosure, Kaushik Ghosh, Kyle Parcella, Hua Fang, Adam G. Jardel, Brett R. Beno, Kevin Kish, Tao Wang, Dawn D. Parker, Zhiwei Yin, Zhongxing Zhang, John F. Kadow, and Juliang Zhu
- Subjects
0301 basic medicine ,Pharmacology ,viruses ,Organic Chemistry ,Pharmaceutical Science ,virus diseases ,Metabolic stability ,Biology ,biochemical phenomena, metabolism, and nutrition ,Biochemistry ,Virology ,Ns5b polymerase ,digestive system diseases ,Bioavailability ,body regions ,03 medical and health sciences ,Chemistry ,030104 developmental biology ,Drug Discovery ,Genotype ,Molecular Medicine ,Primer (molecular biology) - Abstract
The development of a series of novel 7-azabenzofurans exhibiting pan-genotype inhibition of HCV NS5B polymerase via binding to the primer grip site is presented. Many challenges, including poor oral bioavailability, high clearance, bioactivation, high human serum shift, and metabolic stability were encountered and overcome through SAR studies. This work culminated in the selection of BMS-986139 (43) as a preclinical candidate.
- Published
- 2016
12. Discovery of a Potent Acyclic, Tripeptidic, Acyl Sulfonamide Inhibitor of Hepatitis C Virus NS3 Protease as a Back-up to Asunaprevir with the Potential for Once-Daily Dosing
- Author
-
Huabin Sun, Hua Fang, Richard Rampulla, Alan Xiangdong Wang, Bilder Donna M, Bang-Chi Chen, Zhao Qian, Chaoqun Chen, Brian Lee Venables, Fiona McPhee, Jie Chen, Maria Donoso, Barbara Zheng, Paul Falk, Ying-Kai Wang, Yan Chen, Nicholas A. Meanwell, Yong-Hae Han, Qi Gao, Richard Schartman, Luciano Mueller, Steven Levine, Herbert E. Klei, Jacques Friborg, Stephen P. Adams, Ramkumar Rajamani, Anthony J. Cocuzza, Theerthagiri Palani, Dennis M. Grasela, Dennis Hernandez, Fei Yu, Sivakumar Ganesan, James Loy, Tatyana Zvyaga, Paul Levesque, Li-Qiang Sun, Amy K. Sheaffer, Eric Mull, Arvind Mathur, Kathy Mosure, Jeffrey Tredup, Stanley D'andrea, Pirama Nayagam Arunachalam, Jialong Zhu, Mark I. Cockett, Hong Shi, Sing-Yuen Sit, Ny Sin, Paul Michael Scola, Jay O. Knipe, Alicia Ng, William Warner, Kevin Kish, Lucy Sun, Michael Sinz, and Danshi Li
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Hepatitis C virus ,medicine.medical_treatment ,Hepacivirus ,Pharmacology ,Viral Nonstructural Proteins ,medicine.disease_cause ,01 natural sciences ,Antiviral Agents ,Drug Administration Schedule ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,Dogs ,Pharmacokinetics ,Drug Discovery ,Drug Resistance, Viral ,medicine ,Structure–activity relationship ,Animals ,Protease inhibitor (pharmacology) ,Dosing ,NS3 ,Sulfonamides ,Protease ,010405 organic chemistry ,Chemistry ,Stereoisomerism ,Isoquinolines ,Virology ,0104 chemical sciences ,Macaca fascicularis ,030104 developmental biology ,Molecular Medicine ,Asunaprevir ,Replicon ,Rabbits ,Oligopeptides - Abstract
The discovery of a back-up to the hepatitis C virus NS3 protease inhibitor asunaprevir (2) is described. The objective of this work was the identification of a drug with antiviral properties and toxicology parameters similar to 2, but with a preclinical pharmacokinetic (PK) profile that was predictive of once-daily dosing. Critical to this discovery process was the employment of an ex vivo cardiovascular (CV) model which served to identify compounds that, like 2, were free of the CV liabilities that resulted in the discontinuation of BMS-605339 (1) from clinical trials. Structure–activity relationships (SARs) at each of the structural subsites in 2 were explored with substantial improvement in PK through modifications at the P1 site, while potency gains were found with small, but rationally designed structural changes to P4. Additional modifications at P3 were required to optimize the CV profile, and these combined SARs led to the discovery of BMS-890068 (29).
- Published
- 2016
13. Generation of 3,8-substituted 1,2,4-triazolopyridines as potent inhibitors of human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD-1)
- Author
-
Haixia Wang, Ramakrishna Seethala, James J. Li, Yi-Xin Li, Lawrence G. Hamann, Tatyana Zvyaga, Rajasree Golla, David A. Gordon, Ligaya M. Simpkins, and Jeffrey A. Robl
- Subjects
Pyridines ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,11β-hydroxysteroid dehydrogenase type 1 ,Amide ,11-beta-Hydroxysteroid Dehydrogenase Type 1 ,Drug Discovery ,Humans ,Enzyme Inhibitors ,Molecular Biology ,chemistry.chemical_classification ,biology ,Bicyclic molecule ,Organic Chemistry ,Triazoles ,Sulfonamide ,Pyrimidines ,Enzyme ,chemistry ,Enzyme inhibitor ,biology.protein ,Molecular Medicine ,Triazolopyridine - Abstract
A series of pyridyl amide/sulfonamide inhibitors of 11β-HSD-1 were modified to incorporate a novel 1,2,4-triazolopyridine scaffold. Optimization of substituents at the 3 and 8 position of the TZP core, with a special focus on enhancing metabolic stability, resulted in the identification of compound 38 as a potent and metabolically stable inhibitor of the enzyme.
- Published
- 2011
- Full Text
- View/download PDF
14. Ultrafast mass spectrometry based bioanalytical method for digoxin supporting an in vitro P-glycoprotein (P-gp) inhibition screen
- Author
-
Timothy V Olah, Tatyana Zvyaga, Janet Kolb, Harold N. Weller, John J. Herbst, Andrew Dennis Wagner, Can C. Özbal, and Wilson Z. Shou
- Subjects
Reproducibility ,Bioanalysis ,Chromatography ,Digoxin ,biology ,Drug discovery ,Chemistry ,Organic Chemistry ,Liquid scintillation counting ,Mass spectrometry ,In vitro ,Analytical Chemistry ,medicine ,biology.protein ,Spectroscopy ,medicine.drug ,P-glycoprotein - Abstract
The evaluation of interactions between drug candidates and transporters such as P-glycoprotein (P-gp) has gained considerable interest in drug discovery and development. Inhibition of P-gp can be assessed by performing bi-directional permeability studies with in vitro P-gp-expressing cellular model systems such as Caco-2 (human colon carcinoma) cells, using digoxin as a substrate probe. Existing methodologies include either assaying 3H-digoxin with liquid scintillation counting (LSC) detection or assaying non-labeled digoxin with liquid chromatography–tandem mass spectrometric (LC-MS/MS) analysis at a speed of several minutes per sample. However, it is not feasible to achieve a throughput high enough using these approaches to sustain an early liability screen that generates more than a thousand samples on a daily basis. To address this challenge, we developed an ultrafast (9 s per sample) bioanalytical method for digoxin analysis using RapidFire™, an on-line solid-phase extraction (SPE) system, with MS/MS detection. A stable isotope labeled analog, d3-digoxin, was used as internal standard to minimize potential ionization matrix effect during the RF-MS/MS analysis. The RF-MS/MS method was more than 16 times faster than the LC-MS/MS method but demonstrated similar sensitivity, selectivity, reproducibility, linearity and robustness. P-gp inhibition results of multiple validation compounds obtained with this RF-MS/MS method were in agreement with those generated by both the LC-MS/MS method and the 3H-radiolabel assay. This method has been successfully deployed to assess P-gp inhibition potential as an important early liability screen for drug–transporter interaction. Copyright © 2011 John Wiley & Sons, Ltd.
- Published
- 2011
- Full Text
- View/download PDF
15. An Automated High Throughput Liquid Chromatography–Mass Spectrometry Process to Assess the Metabolic Stability of Drug Candidates
- Author
-
James V. Belcastro, Colleen A. McNaney, Robert Langish, Dieter M. Drexler, Adrienne A. Tymiak, Kenneth S. Santone, Serhiy Hnatyshyn, Kurt J. Edinger, Petia Shipkova, Mark Sanders, Tatyana Zvyaga, Kenneth E.J. Dickinson, and Jonathan L. Josephs
- Subjects
Quality Control ,Drug ,media_common.quotation_subject ,Drug Evaluation, Preclinical ,Mass spectrometry ,Mass Spectrometry ,Liquid chromatography–mass spectrometry ,Drug Discovery ,Liver microsomes ,NADPH-Ferrihemoprotein Reductase ,media_common ,Chromatography ,Chemistry ,Reproducibility of Results ,Structural integrity ,Robotics ,Metabolic stability ,Pharmaceutical Preparations ,Data Interpretation, Statistical ,Solvents ,Molecular Medicine ,Indicators and Reagents ,Spectrophotometry, Ultraviolet ,Uv detection ,Software ,Chromatography, Liquid - Abstract
An automated high throughput process, termed the MetFast assay, is described to assess in vitro the general microsomal cytochrome P450 beta-nicotinamide adenine dinucleotide phosphate-mediated first-pass metabolic stability of potential drug candidates as a utility for pharmaceutical profiling. Utilizing robotic protocols with a multiprobe liquid handler, compounds are incubated with liver microsomes from different species. Samples are then analyzed by in-line liquid chromatography (LC)-mass spectrometry (MS) to determine the amount of compound remaining after a certain time, which allows calculation of metabolism rates. To quantitatively assess large numbers of structurally diverse compounds by LC-MS, a strategy based on an iterative two-step process was devised. Initially compounds are qualitatively analyzed by LC-ultraviolet (UV)/MS (step 1) to determine purity (UV detection) and structural integrity (MS detection). This step ensures that only correct and verified compounds with sufficient purity are being assayed to obtain reproducible high data quality. In addition, all necessary information is gathered to automatically generate specific quantitative methods for the subsequent bioanalytical analysis of metabolic stability samples by LC-UV/MS (step 2). In-house-developed, highly flexible and sophisticated data management software, termed SmartReport, is utilized for automated qualitative and quantitative LC-MS analysis set-up, data processing, and results reporting. The integration of key aspects, inherent "universal" collision-induced dissociation settings of ion trap mass spectrometers for tandem mass spectrometric scan functions utilized for compound-specific and sensitive quantitative MS methods, generic fast-LC conditions, generic MS instrument settings, and the functionality of SmartReport software resulted in an analytical process that routinely provides reproducible high-quality metabolic stability data on structurally diverse compounds. Described here is the setup of the MetFast assay, and metabolic stability data from assay validation compounds are given.
- Published
- 2007
- Full Text
- View/download PDF
16. A high-speed liquid chromatography/tandem mass spectrometry platform using multiplexed multiple-injection chromatography controlled by single software and its application in discovery ADME screening
- Author
-
Jun, Zhang, Marianne, Vath, Cheryl, Ferraro, Ying, Li, Keeley, Murphy, Tatyana, Zvyaga, Harold, Weller, and Wilson, Shou
- Subjects
Reproducibility of Results ,Propranolol ,Rats ,Mice ,Models, Chemical ,Tandem Mass Spectrometry ,Drug Discovery ,Linear Models ,Animals ,Cytochrome P-450 CYP3A ,Humans ,Caco-2 Cells ,Software ,Chromatography, Liquid - Abstract
Multiplexed liquid chromatography (LC) coupled with multiple-injection-chromatogram acquisition has emerged as the method of choice for high-speed discovery bioanalysis, because it significantly reduces injection-to-injection cycle time while maintaining the chromatography quality. Historically, systems utilizing this approach had been custom built, and therefore relied on custom software tools to communicate with multiple vendor software for system control, which lacked transferability, flexibility and robustness.In this study, we refined a multiplexed bioanalytical system previously reported, by implementing open-deck auto-sampler manifold and multiple-injection-chromatogram acquisition, all on a commercially available system with single software control.As a result of these improvements, the developed LC/tandem mass spectrometry (MS/MS) method on the system was nearly three times faster than the previous method, while demonstrating comparable analytical accuracy, precision and robustness. This system has been evaluated for in vitro ADME screening assays including metabolic stability, CYP inhibition and Caco-2. The biological data generated on the developed system displayed good correlation with those from the previous LC/MS/MS approaches.The developed platform demonstrated applicability to the in vitro screening assays evaluated and has been successfully implemented to support the high-throughput metabolic stability assay, with a significantly improved bioanalytical throughput, capacity and data turnaround.
- Published
- 2012
17. Identification of a potent and metabolically stable series of fluorinated diphenylpyridylethanamine-based cholesteryl ester transfer protein inhibitors
- Author
-
James A. Johnson, Ramakrishna Seethala, Christine Huang, Michael M. Miller, David S. Taylor, Ji Jiang, Ruth R. Wexler, Mark E. Salvati, Jun Zhang, Xiaohong Yin, Yalei Liu, R. Michael Lawrence, Tatyana Zvyaga, David S. Nirschl, Lalgudi S. Harikrishnan, Tara L. Peterson, Michael A. Poss, Alice Ye A. Chen, Leonard P. Adam, Yufeng Wang, and Muthoni G. Kamau
- Subjects
Halogenation ,Pyridines ,Clinical Biochemistry ,Sterol O-acyltransferase ,Pharmaceutical Science ,Biochemistry ,chemistry.chemical_compound ,Mice ,Drug Discovery ,Cholesterylester transfer protein ,Moiety ,Animals ,Humans ,Urea ,Amines ,Molecular Biology ,CETP inhibitor ,biology ,Chemistry ,Organic Chemistry ,Reverse cholesterol transport ,In vitro ,Cholesterol Ester Transfer Proteins ,Rats ,biology.protein ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) - Abstract
A novel series of diphenylpyridylethanamine-based inhibitors of cholesteryl ester transfer protein is described. Optimization of the urea moiety, particularly by incorporation of fluorine, is explored to balance in vitro metabolic stability with CETP potency in the whole plasma assay.
- Published
- 2012
18. Anticipating and Minimizing Drug Interactions in a Drug Discovery and Development Setting: An Industrial Perspective
- Author
-
Punit Marathe, A. David Rodrigues, Ragini Vuppugalla, Yong-Hae Han, Tatyana Zvyaga, Praveen Balimane, and Sean Kim
- Subjects
Polypharmacy ,Drug ,Engineering ,Current age ,business.industry ,Heparg cell ,Drug discovery ,media_common.quotation_subject ,Perspective (graphical) ,Pharmacology ,Risk analysis (engineering) ,New chemical entity ,business ,media_common - Abstract
In the current age of polypharmacy, it is increasingly likely that a new chemical entity (NCE) will be prescribed with a second drug that demonstrates a narrow therapeutic index. As a result, one has to consider interactions involving drug-metabolizing enzymes and transporters. NCEs with drug–drug interaction (DDI) liabilities may have limited marketing potential, as they may alter the pharmacokinetic profile of a co-administered drug resulting in either unwanted side effects or loss of pharmacological activity. Within the current competitive landscape, therefore, it is highly desirable to select candidates with reduced potential for DDIs and most pharmaceutical companies spend considerable resources screening and triaging NCEs for induction and inhibition of drug-metabolizing enzymes (e.g., cytochromes P450) and transporters. Thus, the purpose of the present chapter is to provide an industrial perspective on how the existing strategies are utilized to enable the selection of suitable candidates with reduced DDI risk. Additional emphasis will be placed on in vitro tools and the challenges associated with the prediction of DDIs prior to first in man.
- Published
- 2009
- Full Text
- View/download PDF
19. Synthesis of novel ketoconazole derivatives as inhibitors of the human Pregnane X Receptor (PXR; NR1I2; also termed SXR, PAR)
- Author
-
Michael Sinz, Sean Kim, Eoin C. Power, Sridhar Mani, Jaime Anguiano, Ankanahlli V. Madhukumar, C. Robin Ganellin, Bhaskar C. Das, and Tatyana Zvyaga
- Subjects
Receptors, Steroid ,Stereochemistry ,Chemistry, Pharmaceutical ,Clinical Biochemistry ,Pharmaceutical Science ,Context (language use) ,Biochemistry ,Chemical synthesis ,Catalysis ,Inhibitory Concentration 50 ,Cytochrome P-450 Enzyme System ,Cell Line, Tumor ,Drug Discovery ,medicine ,Cytochrome P-450 CYP3A ,Humans ,Drug Interactions ,Cytotoxicity ,Molecular Biology ,chemistry.chemical_classification ,Pregnane X receptor ,Binding Sites ,Chemistry ,Organic Chemistry ,Pregnane X Receptor ,Biological activity ,Ketoconazole ,Models, Chemical ,Drug Design ,Microsome ,Microsomes, Liver ,Molecular Medicine ,Azole ,medicine.drug - Abstract
PXR, pregnane X receptor, in its activated state, is a validated target for controlling certain drug-drug interactions in humans. In this context, there is a paucity of inhibitors directed toward activated PXR. Using prior observations with ketoconazole as a PXR inhibitor, the target compound 3 was synthesized from (s)-glycidol with overall 56% yield. (+)-Glycidol was reacted with 4-bromophenol and potassium carbonate in DMF to yield the ring opened compound 6. This was then heated to reflux in benzene along with 2', 4'-difluoroacetophenone and catalytic amount of para-toluene sulfonic acid to yield 8. The resultant acetal 8 was then functionalized using Palladium chemistry to yield the target compound 3. The activity of the compound was compared with ketoconazole and UCL2158H. However, in contrast with ketoconazole (IC(50) approximately 0.020 microM; approximately 100% inhibition), 3 has negligible effects on inhibition of microsomal CYP450 (maximum approximately 20% inhibition) at concentrations >40 microM. In vitro, micromolar concentration of ketoconazole is toxic to passaged human cell lines, while 3 does not exhibit cytotoxicity up to concentrations approximately 100 microM (viability >85%). This is the first demonstration of a chemical analog of a PXR inhibitor that retains activity against activated PXR. Furthermore, in contrast with ketoconazole, 3 is less toxic in human cell lines and has negligible CYP450 activity.
- Published
- 2008
20. An automated liquid chromatography-mass spectrometry process to determine metabolic stability half-life and intrinsic clearance of drug candidates by substrate depletion
- Author
-
Serhiy Hnatyshyn, Dieter M. Drexler, Jay O. Knipe, Colleen A. McNaney, James V. Belcastro, Mark Sanders, and Tatyana Zvyaga
- Subjects
Drug ,media_common.quotation_subject ,Drug Evaluation, Preclinical ,Mass spectrometry ,Mass Spectrometry ,Automation ,Mice ,Pharmacokinetics ,Liquid chromatography–mass spectrometry ,Spectrophotometry ,Drug Discovery ,medicine ,Animals ,Chromatography, High Pressure Liquid ,media_common ,Electronic Data Processing ,Chromatography ,medicine.diagnostic_test ,Chemistry ,Half-life ,Metabolic stability ,Rats ,Pharmaceutical Preparations ,Data Interpretation, Statistical ,Microsomes, Liver ,Solvents ,Molecular Medicine ,Indicators and Reagents ,Spectrophotometry, Ultraviolet ,Ion trap ,Software ,Half-Life - Abstract
An automated process is described for the detailed assessment of the in vitro metabolic stability properties of drug candidates in support of pharmaceutical property profiling. Compounds are incubated with liver microsomes using a robotic liquid handler. Aliquots are taken at various time points, and the resulting samples are quantitatively analyzed by liquid chromatography-mass spectrometry utilizing ion trap mass spectrometers to determine the amount of compound remaining. From these data metabolism rates can be calculated. A high degree of automation is achieved through custom software, which is employed for instrument setup, data processing, and results reporting. The assay setup is highly configurable, allowing for any combination of up to six user-selected time points, variable substrate concentration, and microsomes or other biologically active media. The data, based on relative substrate depletion, affords an estimate of metabolic stability through the calculation of half-life (t(1/2)) and intrinsic clearance, which are used to differentiate and rank order drug leads. In general, t(1/2) is the time necessary for the metabolism, following first-order kinetics, of 50% of the initial compound. Intrinsic clearance is the proportionality constant between rate of metabolism of a compound and its concentration at the enzyme site. Described here is the setup of the assay, and data from assay test compounds are presented.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.