20 results on '"Amy Meng"'
Search Results
2. JAK selectivity and the implications for clinical inhibition of pharmacodynamic cytokine signalling by filgotinib, upadacitinib, tofacitinib and baricitinib
- Author
-
Julie A Di Paolo, Amy Meng, Bernard Murray, Paqui G. Través, Federico Campigotto, and René Galien
- Subjects
0301 basic medicine ,Filgotinib ,rheumatoid ,antirheumatic agents ,Pyridines ,medicine.medical_treatment ,Immunology ,immune system diseases ,Rheumatoid Arthritis ,Pharmacology ,Peripheral blood mononuclear cell ,General Biochemistry, Genetics and Molecular Biology ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Piperidines ,medicine ,Immunology and Allergy ,Humans ,Janus Kinase Inhibitors ,Cells, Cultured ,Janus Kinases ,030203 arthritis & rheumatology ,Sulfonamides ,Tofacitinib ,business.industry ,Triazoles ,cytokines ,030104 developmental biology ,Cytokine ,Pyrimidines ,arthritis ,Tyrosine kinase 2 ,Purines ,STAT protein ,Azetidines ,Pyrazoles ,business ,Janus kinase ,Heterocyclic Compounds, 3-Ring ,Ex vivo - Abstract
ObjectiveJanus kinase inhibitors (JAKinibs) are efficacious in rheumatoid arthritis (RA) with variable reported rates of adverse events, potentially related to differential JAK family member selectivity. Filgotinib was compared with baricitinib, tofacitinib and upadacitinib to elucidate the pharmacological basis underlying its clinical efficacy and safety.MethodsIn vitro JAKinib inhibition of signal transducer and activator of transcription phosphorylation (pSTAT) was measured by flow cytometry in peripheral blood mononuclear cells and whole blood from healthy donors and patients with RA following cytokine stimulation of distinct JAK/STAT pathways. The average daily pSTAT and time above 50% inhibition were calculated at clinical plasma drug exposures in immune cells. The translation of these measures was evaluated in ex vivo-stimulated assays in phase 1 healthy volunteers.ResultsJAKinib potencies depended on cytokine stimulus, pSTAT readout and cell type. JAK1-dependent pathways (interferon (IFN)α/pSTAT5, interleukin (IL)-6/pSTAT1) were among the most potently inhibited by all JAKinibs in healthy and RA blood, with filgotinib exhibiting the greatest selectivity for JAK1 pathways. Filgotinib (200 mg once daily) had calculated average daily target inhibition for IFNα/pSTAT5 and IL-6/pSTAT1 that was equivalent to tofacitinib (5 mg two times per day), upadacitinib (15 mg once daily) and baricitinib (4 mg once daily), with the least average daily inhibition for the JAK2-dependent and JAK3-dependent pathways including IL-2, IL-15, IL-4 (JAK1/JAK3), IFNγ (JAK1/JAK2), granulocyte colony stimulating factor, IL-12, IL-23 (JAK2/tyrosine kinase 2) and granulocyte-macrophage colony-stimulating factor (JAK2/JAK2). Ex vivo pharmacodynamic data from phase 1 healthy volunteers clinically confirmed JAK1 selectivity of filgotinib.ConclusionFilgotinib inhibited JAK1-mediated signalling similarly to other JAKinibs, but with less inhibition of JAK2-dependent and JAK3-dependent pathways, providing a mechanistic rationale for its apparently differentiated efficacy:safety profile.
- Published
- 2021
3. Exposure-Response Relationships for Efficacy and Safety of Filgotinib and its metabolite GS-829845 in Subjects with Rheumatoid Arthritis Based on Phase 2 and Phase 3 Studies
- Author
-
Amy Meng, Kacey Anderson, Cara Nelson, Liyun Ni, Shu-Min Chuang, Francesco Bellanti, Peter Chang, Craig Comisar, Brian Kearney, Beatrix Bartok, and Anita Mathias
- Abstract
Aims:Filgotinib is a potent, oral, JAK1-preferential inhibitor for the treatment of rheumatoid arthritis (RA). This report describes exposure-response (ER) analyses of filgotinib for dose confirmation based on three Phase 3 and two Phase 2 studies in moderate to severe RA patients. Methods:The PK exposures used in ER analyses were derived from population pharmacokinetic analysis. The relationship between filgotinib exposures and various efficacy endpoints (ACR20/50/70 and DAS28) was assessed over octile groups of exposures by using combined exposures of filgotinib and GS-829845 (major, active metabolite). For the ER analyses of safety, exposures were examined between subjects who experienced and who did not experience the evaluated safety events, which was conducted separately for filgotinib and GS-829845. Results:Exposure efficacy relationships consistently revealed high response rates across the exposure range for filgotinib 200 mg once daily dose. A trend of increasing response with increasing exposure was observed over the exposure range for the primary and multiple secondary efficacy endpoints, with exposures associated with the 200 mg dose primarily residing on the curve plateau. For exposure-safety analyses, filgotinib and GS-829845 exposures were similar irrespective of presence/absence of the evaluated safety endpoints, indicating no exposure-safety relationship for common TEAEs, common laboratory abnormalities, serious TEAEs, or serious infections. Conclusions:ER analyses confirmed that filgotinib produced more robust therapeutic effects across the exposure range observed at 200 mg once daily compared to lower doses. The positive exposure-efficacy relationship and a lack of exposure-safety relationship on the evaluated safety endpoints supported the 200 mg once daily dose for commercialization.
- Published
- 2021
- Full Text
- View/download PDF
4. Semi-Mechanistic PK/PD Modeling and Simulation of Irreversible BTK Inhibition to Support Dose Selection of Tirabrutinib in Subjects with RA
- Author
-
Chia-Hsiang Hsueh, Ethan Grant, Helen Yu, Thomas Tarnowski, Rita Humeniuk, Franziska Matzkies, Cara H. Nelson, Amy Meng, Ellen Kwan, Andrew N. Billin, Joy Y. Feng, Hoa Truong, and Juliane M. Jürgensmeier
- Subjects
Adult ,Male ,Adolescent ,Anti-Inflammatory Agents ,Pharmacology ,Models, Biological ,Arthritis, Rheumatoid ,Young Adult ,Pharmacokinetics ,hemic and lymphatic diseases ,medicine ,Agammaglobulinaemia Tyrosine Kinase ,Bruton's tyrosine kinase ,Humans ,Pharmacology (medical) ,Computer Simulation ,Drug Dosage Calculations ,Protein Kinase Inhibitors ,PK/PD models ,biology ,Clinical Trials, Phase I as Topic ,business.industry ,Imidazoles ,Middle Aged ,medicine.disease ,Pyrimidines ,Drug development ,Rheumatoid arthritis ,Pharmacodynamics ,biology.protein ,Female ,business ,Tyrosine kinase ,Dose selection - Abstract
Tirabrutinib is an irreversible, small-molecule Bruton's tyrosine kinase (BTK) inhibitor, which was approved in Japan (VELEXBRU) to treat B-cell malignancies and is in clinical development for inflammatory diseases. As an application of model-informed drug development, a semimechanistic pharmacokinetic/pharmacodynamic (PK/PD) model for irreversible BTK inhibition of tirabrutinib was developed to support dose selection in clinical development, based on clinical PK and BTK occupancy data from two phase I studies with a wide range of PK exposures in healthy volunteers and in subjects with rheumatoid arthritis. The developed model adequately described and predicted the PK and PD data. Overall, the model-based simulation supported a total daily dose of at least 40 mg, either q.d. or b.i.d., with adequate BTK occupancy (> 90%) for further development in inflammatory diseases. Following the PK/PD modeling and simulation, the relationship between model-predicted BTK occupancy and preliminary clinical efficacy data was also explored and a positive trend was identified between the increasing time above adequate BTK occupancy and better efficacy in treatment for RA by linear regression.
- Published
- 2021
5. Sofosbuvir/velpatasvir for 12 weeks in hepatitis C virus-infected patients with end-stage renal disease undergoing dialysis
- Author
-
Shariq Haider, Edward Gane, Jose Luis Calleja, Rafael Esteban, Svetlana Markova, Kosh Agarwal, Rafael Bruck, Amy Meng, Stephen D. Shafran, Sergio Borgia, Matthew Foxton, Robert H. Hyland, Javier Ampuero, Bernard Willems, Anu Osinusi, Raymond Fox, David R. Shaw, Conrado M Fernández Rodríguez, Ziv Ben-Ari, Hadas Dvory-Sobol, Ashley Brown, Stephen D. Ryder, Curtis Cooper, Matthew E. Cramp, Brian J. Kirby, Eric M. Yoshida, Yoav Lurie, Sophia Lu, and Janet Dearden
- Subjects
Liver Cirrhosis ,Male ,medicine.medical_specialty ,Sofosbuvir ,Sustained Virologic Response ,medicine.medical_treatment ,Population ,Hepacivirus ,urologic and male genital diseases ,Direct-acting antiviral ,Sofosbuvir/velpatasvir ,Antiviral Agents ,Heterocyclic Compounds, 4 or More Rings ,End stage renal disease ,Peritoneal dialysis ,Renal Dialysis ,Internal medicine ,medicine ,Humans ,HCV, SVR12, drug safety ,ESRD ,education ,Adverse effect ,Dialysis ,education.field_of_study ,Hepatology ,business.industry ,Hepatitis C, Chronic ,Middle Aged ,Chronic hepatitis C infection ,Severe renal impairment ,Drug Combinations ,Treatment Outcome ,Kidney Failure, Chronic ,Female ,Hemodialysis ,Carbamates ,Drug Monitoring ,business ,medicine.drug - Abstract
[Background & Aims] Although off-label use of sofosbuvir-containing regimens occurs regularly in patients with hepatitis C virus (HCV) infection undergoing dialysis for severe renal impairment or end-stage renal disease (ESRD), these regimens are not licensed for this indication, and there is an absence of dosing recommendations in this population. This study evaluated the safety and efficacy of sofosbuvir/velpatasvir in patients with HCV infection with ESRD undergoing dialysis., [Methods] In this phase II, single-arm study, 59 patients with genotype 1–6 HCV infection with ESRD undergoing hemodialysis or peritoneal dialysis received open-label sofosbuvir/velpatasvir (400 mg/100 mg) once daily for 12 weeks. Patients were HCV treatment naive or treatment experienced without cirrhosis or with compensated cirrhosis. Patients previously treated with any HCV NS5A inhibitor were not eligible. The primary efficacy endpoint was the proportion of patients achieving sustained virologic response (SVR) 12 weeks after discontinuation of treatment (SVR12). The primary safety endpoint was the proportion of patients who discontinued study drug due to adverse events., [Results] Overall, 56 of 59 patients achieved SVR12 (95%; 95% CI 86–99%). Of the 3 patients who did not achieve SVR12, 2 patients had virologic relapse determined at post-treatment Week 4 (including 1 who prematurely discontinued study treatment), and 1 patient died from suicide after achieving SVR through post-treatment Week 4. The most common adverse events were headache (17%), fatigue (14%), nausea (14%), and vomiting (14%). Serious adverse events were reported for 11 patients (19%), and all were deemed to be unrelated to sofosbuvir/velpatasvir., [Conclusions] Treatment with sofosbuvir/velpatasvir for 12 weeks was safe and effective in patients with ESRD undergoing dialysis., [Lay summary] Sofosbuvir/velpatasvir is a combination direct-acting antiviral that is approved for treatment of patients with hepatitis C virus (HCV) infection. Despite the lack of dosing recommendations, sofosbuvir-containing regimens (including sofosbuvir/velpatasvir) are frequently used for HCV-infected patients undergoing dialysis. This study evaluated the safety and efficacy of sofosbuvir/velpatasvir for 12 weeks in patients with HCV infection who were undergoing dialysis. Treatment with sofosbuvir/velpatasvir was safe and well tolerated, resulting in a cure rate of 95% in patients with HCV infection and end-stage renal disease., Clinical Trial Number: NCT03036852.
- Published
- 2019
6. THU0017 IN VITRO MECHANISTIC STUDIES DEMONSTRATE FILGOTINIB ACTIVITY THAT HAS POTENTIAL IMPLICATIONS FOR DIFFERENTIATION AMONG JAK INHIBITORS
- Author
-
Nevena Mollova, Julie Di Paolo, Amy Meng, Pei Han, and Yuanjiang Yu
- Subjects
Tofacitinib ,Filgotinib ,biology ,business.industry ,Cell ,Pharmacology ,In vitro ,Proinflammatory cytokine ,medicine.anatomical_structure ,Tyrosine kinase 2 ,Cholesterylester transfer protein ,biology.protein ,Medicine ,business ,Liver X receptor - Abstract
Background Inhibition of the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway has demonstrated efficacy in immune-mediated diseases and has been identified as a therapeutic target for the treatment of rheumatoid arthritis (RA). Differences in JAK inhibitor specificity for JAK1, JAK2, JAK3, and TYK2 may influence their safety profiles, but the mechanism is not known. Selective JAK1 inhibition by filgotinib (FIL) may modulate a subset of proinflammatory cytokines associated with RA pathogenesis and improve the risk-benefit profile by minimizing other non–JAK1-related adverse events. JAK2 inhibition is associated with cytopenias, while JAK3 inhibition has been associated with increased risk for opportunistic infections (eg, tuberculosis and herpes zoster) and chronic low-grade inflammation. In clinical trials, FIL did not negatively impact hemoglobin, LDL/HDL ratios, or natural killer (NK) cell counts.1-3 Objectives To compare the in vitro profile of JAK inhibitors with different JAK selectivity profiles, for effects on erythroid progenitor cell expansion, NK cell proliferation, and liver X receptor (LXR) agonist-induced cholesteryl ester transfer protein (CETP) expression, an enzyme responsible for the conversion of HDL to LDL. Methods JAK inhibitors (FIL, FIL metabolite [GS-829845], baricitinib [BARI], tofacitinib [TOFA], and upadacitinib [UPA]) were evaluated in vitro in human cell-based assays: growth of erythroid progenitors from human cord blood CD34+ cells using a HemaTox™ liquid expansion assay, IL-15–induced NK cell proliferation, and LXR agonist-induced CETP expression in the hepatic cell line (HepG2). Using IC50s generated from these assays and the reported human plasma concentrations of the JAK inhibitors from clinical studies,4-6 we calculated the target coverage for each compound at clinically relevant doses. The activity of FIL in humans was based on a PK-PD modeling algorithm7 of FIL + GS-829845. Results In vitro assay results are described in the table. Based on these results, human exposure data, and modeled PK-PD relationships, FIL 100 mg and FIL 200 mg result in lower calculated cellular inhibition than the other JAK inhibitors at clinical exposures. Notably, FIL 100 mg and FIL 200 mg, but not the other inhibitors, are calculated to reduce CETP expression by 17% and 27%, respectively, while BARI, TOFA, and UPA are not expected to alter CETP levels. a Weak stimulation of LXR agonist-induced CETP expression. Conclusion JAK1 selectivity of FIL and GS-829845 resulted in less inhibition of erythroid progenitor expansion and NK cell proliferation compared with BARI, TOFA, and UPA. FIL also reduced LXR agonist-induced CETP expression, while the other inhibitors did not alter these levels. These results provide a potential mechanistic link to the observed reduction of CETP concentration and activity following FIL treatment, and the observed reduction in LDL:HDL in RA patients.8 References [1] Kavanaugh A, et al. Ann Rheum Dis. 2017;76:1009-1019. [2] Westhovens R, et al. Ann Rheum Dis. 2017;76:998-1008. [3] MC Genovese, et al. ACR 2018. Abstract L06. [4] Shi JG. J Clin Pharmacol. 2014;54:1354-1361. [5] Lamba M. J Clin Pharmacol. 2016;56:1362-1371. [6] Mohamed MF, et al. Clin Pharmacokinet. 2016;55:1547-1558. [7] Tuk B. J Pharmacol Exp Ther. 1999;289:1067-1074. [8] Galien R. Arthritis Rheumatol. 2015;67(suppl 10). Disclosure of Interests Pei Han Shareholder of: Gilead Sciences, Inc., Grant/research support from: Gilead Sciences, Inc., Employee of: Gilead Sciences, Inc., Amy Meng Shareholder of: Gilead Sciences, Inc., Employee of: Gilead Sciences, Inc., Nevena Mollova Shareholder of: Gilead Sciences, Inc., Employee of: Gilead Sciences, Inc., Yuanjiang Yu Grant/research support from: Gilead Sciences, Inc., Employee of: Gilead Sciences, Inc., Julie A. Di Paolo Shareholder of: Gilead Sciences, Inc., Employee of: Gilead Sciences, Inc.
- Published
- 2019
- Full Text
- View/download PDF
7. IDDF2019-ABS-0135 Pharmacokinetics of once-daily sofosbuvir or ledipasvir/sofosbuvir in HCV-infected pediatrics aged 3 to
- Author
-
Amy Meng, Rebecca Begley, Jiang Shao, John Ling, Benedetta Massetto, Christina Sze Man Yip, and Anita Mathias
- Subjects
0301 basic medicine ,education.field_of_study ,medicine.medical_specialty ,Sofosbuvir ,business.industry ,Population ,Cmax ,Gastroenterology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Hcv treatment ,Medicine ,LEDIPASVIR/SOFOSBUVIR ,030211 gastroenterology & hepatology ,Once daily ,business ,education ,medicine.drug - Abstract
Background SOF 400 mg and LDV/SOF 90/400 mg are approved HCV treatment in adults and adolescents. Pharmacokinetic (PK) data indicates that SOF 200 mg and LDV/SOF 45/200 mg are appropriate doses in children 6 to Methods HCV-infected children (3 to Results All but 1subject completed IPK assessments. At baseline, median age and weight for subjects was 5 y and 17 kg in SOF+RBV PK lead-in (N=11) and 5 y and 20 kg in LDV/SOF subjects (N=14). The predefined PK criteria were met as GS-331007 and LDV AUCtau were within the 50–200% boundaries as compared with the adult Phase 2/3 population (table 1). The GS-331007 Cmax (SOF+RBV) was modestly higher; these increases are not considered clinically relevant based on established exposure-safety analyses. SOF concentrations were within the range of those observed in the SOF and LDV/SOF adult Phase 2/3 population (data not shown). Conclusions SOF 200 or 150 mg or LDV/SOF 45/200 or 33.75/150 mg, for subjects ≥17 kg or
- Published
- 2019
- Full Text
- View/download PDF
8. POS0224 SELECTIVITY OF CLINICAL JAK INHIBITORS AND THE IMPACT ON NATURAL KILLER (NK) CELL FUNCTIONAL RESPONSES
- Author
-
L. Simpson, G. Min-Oo, E. Grant, J. A. Di Paolo, P. Gonzalez-Traves, Bernard P. Murray, and Amy Meng
- Subjects
Cell signaling ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Immunology ,Cell ,Population ,Pharmacology ,Peripheral blood mononuclear cell ,General Biochemistry, Genetics and Molecular Biology ,Flow cytometry ,medicine.anatomical_structure ,Rheumatology ,Tyrosine kinase 2 ,Immunology and Allergy ,Medicine ,business ,education ,Janus kinase ,IC50 - Abstract
Background:Janus kinase (JAK) inhibitors (JAKinibs) show similar efficacy in rheumatoid arthritis (RA). However, in vitro studies have shown differences in JAK selectivity profiles for baricitinib (BARI), tofacitinib (TOFA), upadacitinib (UPA) and filgotinib (FIL).1,2 These lead to distinct pharmacologic profiles in cellular signaling assays that may impact clinical efficacy or safety1. NK cells are innate lymphocytes important in anti-pathogen responses and immune surveillance, which function via production of cytokines and cell killing3. NK cell proliferation and IFNγ production are JAK-dependent pathways and may be modulated by JAKinibs. Clinical findings show transient decreases in NK cell numbers in patients treated with JAKinibs, but the link to safety is unclear4Objectives:To extend upon findings in proximal cell signaling assays, we compared the selectivity and potency of clinical JAKinibs on NK cell function by assessing proliferation mediated by IL-15 (JAK1/3) and IFN-γ production driven by IL-12 (JAK2/TYK2)+IL-18.Methods:NK cells were isolated from healthy donor PBMC, incubated in vitro with 8 concentrations of each evaluated JAKinib (TOFA, BARI, FIL, FIL metabolite, UPA) and stimulated with IL-15 for proliferation or IL-12/18 for IFNγ production. Proliferation was assessed by Cell Trace dye dilution after 6 days and IFNγ production by intracellular flow cytometry 4hrs post-stimulation. Half maximal inhibitory concentration (IC50) values were calculated for CD56bright, CD56dim, and total NK cells. Steady-state pharmacologic profile over a clinical dosing interval was modeled using concentration-time profiles from JAKinib population pharmacokinetic data in RA subjects under the therapeutic dose5-7. For each JAKinib, the time above IC50 and average daily inhibition of IFNγ or proliferation were calculated for each NK cell population in each donor.Results:Cellular assays in purified NK cells showed dose-dependent inhibition of IL-15-induced proliferation by all JAKinibs with TOFA showing the highest average inhibition and time above IC50 (35-60% inhibition for 8-15 hrs; TOFA>UPA>BARI≈FIL). The differences between JAKinibs are in line with differences in pSTAT inhibition downstream of IL-151. Interestingly, IL-12/18-induced production of IFNγ, which is mediated via JAK2/TYK2 (IL-12) and non-JAK dependent pathways (IL-18), showed weaker inhibition for all compounds. Moreover, all JAKinibs showed 50 for IFNγ production or pSTAT4 inhibition at clinical doses. CD56dim and CD56bright sub-populations of NK cells are proposed to have distinct functions and unique expression of surface receptors. Analysis of the IC50 for pSTAT4 and IFNγ production showed ~2-10-fold weaker inhibition by JAKinibs in CD56bright NK cells, suggesting less dependence on JAK-dependent signals in CD56bright NK cells than CD56dim NK cells.Conclusion:NK cell proliferation depends on JAK1 and JAK3-mediated signaling and is differentially inhibited at clinical doses of distinct JAKinibs. In contrast, functional responses downstream of JAK2/TYK2-dependent IL-12/18 were not substantially inhibited by any of the JAKinibs studied. Inhibition of functional and proliferative responses in purified NK cells aligned well with proximal pSTAT inhibition. JAKinib modulation of NK cell proliferation, but not response to IL-12, reflects unique pharmacologic profiles of the drugs studied and could be one component underlying clinical safety observations, including increased risk of viral infections or malignancy4.References:[1]Traves PG et al. Ann Rheum Dis 2021 (in press)[2]McInnes IB, et al. Arthritis Res Ther 2019;21:183.[3]Cooper MA, Fehniger TA, Caligiuri MA. Trends Immunol 2001 Nov;22(11):633-40.[4]Winthrop KL. Nat Rev Rheumatol 2017; 13(4):234-243[5]Zhang X, et al. CPT Pharmacometrics Syst Pharmacol 2017;6(12):804-13.[6]CDER. Application Number: 203214Orig1s000. NDA 203214: Tofacitinib.[7]Klunder B et al. Clin Pharmacokinet 2019;58(8):1045-58.Disclosure of Interests:Paqui Gonzalez-Traves Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Laura Simpson Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Bernard Murray Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Amy Meng Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Julie A. Di Paolo Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Ethan Grant Shareholder of: Gilead Sciences, Employee of: Gilead Sciences, Gundula Min-Oo Shareholder of: Gilead Sciences, Employee of: Gilead Sciences
- Published
- 2021
- Full Text
- View/download PDF
9. Studying Gas-Phase Interconversion of Tautomers Using Differential Mobility Spectrometry
- Author
-
W. Scott Hopkins, Luke Melo, Amy Meng-Ci Yang, and J. Larry Campbell
- Subjects
Chemistry ,010401 analytical chemistry ,Analytical chemistry ,Solvation ,Electronic structure ,010402 general chemistry ,Tandem mass spectrometry ,Mass spectrometry ,01 natural sciences ,Tautomer ,0104 chemical sciences ,Ion ,Molecular dynamics ,Structural Biology ,Computational chemistry ,Molecule ,Spectroscopy - Abstract
In this study, we report on the use of differential mobility spectrometry (DMS) as a tool for studying tautomeric species, allowing a more in-depth interrogation of these elusive isomers using ion/molecule reactions and tandem mass spectrometry. As an example, we revisit a case study in which gas-phase hydrogen-deuterium exchange (HDX)-a probe of ion structure in mass spectrometry-actually altered analyte ion structure by tautomerization. For the N- and O-protonated tautomers of 4-aminobenzoic acid, when separated using DMS and subjected to subsequent HDX with trace levels of D2O, the anticipated difference between the exchange rates of the two tautomers is observed. However, when using higher levels of D2O or a more basic reagent, equivalent and almost complete exchange of all labile protons is observed. This second observation is a result of the interconversion of the N-protonated tautomer to the O-protonated form during HDX. We can monitor this transformation experimentally, with support from detailed molecular dynamics and electronic structure calculations. In fact, calculations suggest the onset of bulk solution phase properties for 4-aminobenzoic acid upon solvation with eight CH3OH molecules. These findings also underscore the need for choosing HDX reagents and conditions judiciously when separating interconvertible isomers using DMS. Graphical Abstract ᅟ.
- Published
- 2016
- Full Text
- View/download PDF
10. THU0067 JAK SELECTIVITY AND THE IMPACT ON CYTOKINE SIGNALING INHIBITION AT CLINICAL RHEUMATOID ARTHRITIS DOSES
- Author
-
Bernard P. Murray, J. A. Di Paolo, P. Gonzalez-Traves, F. Campigotto, and Amy Meng
- Subjects
education.field_of_study ,Janus kinase 1 ,business.industry ,medicine.medical_treatment ,Immunology ,Population ,JAK-STAT signaling pathway ,Alpha interferon ,Pharmacology ,General Biochemistry, Genetics and Molecular Biology ,Cytokine ,Rheumatology ,Tyrosine kinase 2 ,STAT protein ,Immunology and Allergy ,Medicine ,education ,business ,CD8 - Abstract
Background:Janus kinase 1 (JAK1) inhibitors are efficacious in rheumatoid arthritis (RA). Despite having similar efficacy, in vitro studies have shown differences in JAK selectivity profiles for the small-molecule JAK inhibitors (JAKi) baricitinib (BARI), tofacitinib (TOFA), and upadacitinib (UPA).1For example, BARI and UPA are JAK1/JAK2 selective, while TOFA is JAK1/JAK3 selective, but each JAKi has some activity against other JAKs. As JAKs form signaling pairs, differences in selectivity could lead to distinct pharmacologic profiles that may impact clinical efficacy and safety.Objectives:As a first step to understand the basis of potential differences at therapeutic doses, we compared the selectivity and potency of filgotinib (FIL) and its major metabolite (MET) to those of BARI, TOFA, and UPA in cytokine-stimulated peripheral blood mononuclear cells (PBMCs) and whole blood (WB).Methods:PBMCs and WB from healthy donors were incubated in vitro with 8 doses of each JAKi, and levels of signal transducer and activator of transcription phosphorylation (pSTAT) were measured following cytokine stimulation. Half maximal inhibitory concentration (IC50) values were calculated in phenotypically sorted leukocyte populations by flow cytometry. Therapeutic dose relevance of the in vitro analyses was assessed using calculated mean concentration-time profiles from JAKi population pharmacokinetic data in RA subjects. For each JAKi, the time above IC50and average daily pSTAT inhibition were calculated for each cytokine/STAT pair in B cells, CD4+ T cells, CD8+ T cells, monocytes, and/or NK cells.Results:Cellular assays in PBMCs and WB showed dose-dependent inhibition of cytokine-induced pSTATs with all JAKi (correlation between the protein-adjusted IC50values from PBMCs and IC50values from WB, r2=0.98). Among the most potently inhibited pathways were JAK1/TYK2-dependent cytokine, interferon alpha (IFNα), and the JAK1/2-dependent cytokine, interleukin (IL)-6. FIL and MET had weaker potencies against JAK2/TYK2 (G-CSF/pSTAT3), JAK1/2 (IFNƴ/pSTAT1), and JAK2/2 (granulocyte-macrophage colony-stimulating factor [GM-CSF])-dependent pathways compared to JAK1/TYK2 (IFNα/pSTAT5). FIL and MET showed the greatest selectivity vs the JAK2/2 pathway (GM-CSF/pSTAT3) in monocytes.The mean concentration-time profiles and time above IC50over 24 hr for each cytokine/STAT pathway showed that JAK1/2 (IL-6/pSTAT1) and JAK1/TYK2 (IFNα/pSTAT1) pathways were strongly modulated with all tested JAKi. FIL (200 mg) showed similar activity in average target coverage and time above IC50to the approved low doses of TOFA (5 mg) and UPA (15 mg); conversely, FIL had reduced mean average inhibition and time above IC50levels against JAK1/2 (IFNƴ/pSTAT1), JAK1/3-dependent cytokines (IL-2, -4, and -15), JAK2/TYK2 (G-CSF/pSTAT3), and JAK2/2 (GM-CSF/pSTAT5)-dependent pathways compared to TOFA and UPA, and in certain cases to BARI (2 mg).Conclusion:Different JAKi modulate distinct cytokine pathways to varying degrees, and no agent potently and continuously inhibited an individual cytokine signaling pathway throughout the dosing interval. FIL (200 mg) showed a similar inhibition profile to TOFA, BARI, and UPA against the JAK1/TYK2- (IFNα/pSTAT1) or JAK1/2-dependent (IL-6/pSTAT1) responses, consistent with the role of these pathways in clinical efficacy.2However, FIL displayed a differentiated pharmacologic profile from the other JAKi, showing biologically reduced activity on the JAK1/2 (IFNγ)-, JAK1/3 (IL-2, -4 and -15)-, JAK2/TYK2 (G-CSF)-, and JAK2/2 (GM-CSF)-dependent pathways, which play important roles in hematopoiesis and immune function. These data suggest that FIL (200 mg) may have less impact on a subset of homeostatic immune functions signaling via JAK2 and JAK3 than those observed at the clinically approved doses of TOFA (5 mg and 10 mg), UPA (15 mg), and BARI (4 mg).References:[1]McInnes IB, et al. Arthritis Res Ther. 2019;21:183.[2]Banerjee S, et al. Drugs. 2017;77:521-546.Disclosure of Interests:Paqui Gonzalez-Traves Employee of: Gilead, Bernard Murray Employee of: Gilead, Federico Campigotto Employee of: Gilead, Amy Meng Shareholder of: Gilead Sciences, Employee of: Gilead, Julie A. Di Paolo Employee of: Gilead
- Published
- 2020
- Full Text
- View/download PDF
11. Tu1903 EVALUATION OF POTENTIAL MECHANISMS UNDERLYING THE SAFETY OBSERVATIONS OF FILGOTINIB IN CLINICAL STUDIES IN RA
- Author
-
Amy Meng, Pei Han, Astrid Clarke, Yuanjiang Yu, Bryan Downie, Julie Di Paolo, and Nevena Mollova
- Subjects
Safety observations ,medicine.medical_specialty ,Filgotinib ,Hepatology ,business.industry ,Gastroenterology ,medicine ,Intensive care medicine ,business - Published
- 2020
- Full Text
- View/download PDF
12. P460 Evaluation of potential mechanisms underlying the safety observations of filgotinib in clinical studies in rheumatoid arthritis
- Author
-
Yuanjiang Yu, Pei Han, B Downie, Astrid S. Clarke, J Di Paolo, Amy Meng, and Nevena Mollova
- Subjects
Safety observations ,medicine.medical_specialty ,Filgotinib ,Tofacitinib ,business.industry ,Baricitinib ,Gastroenterology ,General Medicine ,medicine.disease ,Plasma drug concentration ,Rheumatoid arthritis ,Medicine ,Inhibitory concentration 50 ,Erythroid Progenitor Cells ,business ,Intensive care medicine - Abstract
Background Inhibitors of the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway have demonstrated efficacy in the treatment of rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). Differences in selectivity of JAK inhibitors for JAK1, JAK2, JAK3 and TYK2 may influence their respective safety profiles, and the mechanisms responsible are not currently known. Filgotinib (FIL), a JAK1 inhibitor, did not negatively impact haemoglobin, LDL:HDL ratios or natural killer (NK) cell counts in clinical trials. Here, we compare the in vitro mechanistic profiles of four JAK inhibitors at clinically relevant doses. Methods JAK inhibitors (FIL, FIL metabolite [GS-829845], baricitinib [BARI], tofacitinib [TOFA], and upadacitinib [UPA]) were evaluated in vitro in human-cell-based assays. Growth of erythroid progenitors from human cord blood CD34+ cells was assessed using a HemaTox™ liquid expansion assay, NK cell proliferation was induced by IL-15 and LXR agonist-induced cholesteryl ester transfer protein (CETP) expression was assessed in the hepatic cell line, HepG2. Using assay-generated IC50 values and the reported human plasma concentrations from clinical studies, we calculated the target coverage for each JAK inhibitor at clinically relevant doses. The activity of FIL in humans was based on PK/PD modelling of FIL + GS-829845. Results Inhibition of cellular activity was calculated for each JAK inhibitor based on in vitro dose-response data, human exposure data and modelled PK/PD relationships. At clinically relevant doses, FIL resulted in lower calculated inhibition of NK cell proliferation compared with other JAK inhibitors. FIL 100 mg and 200 mg also reduced CETP expression, whereas other JAK inhibitors had no effect. There was no difference in the effect of FIL vs. other JAK inhibitors on erythroid progenitor cell differentiation or maturation. Conclusion FIL, a JAK1 inhibitor, resulted in less inhibition of NK cell proliferation compared with BARI, TOFA, and UPA. FIL also reduced LXR agonist-induced CETP expression, while the other inhibitors did not alter these levels. These results provide a potential mechanistic link between the observed reduction of CETP concentration following FIL treatment and the previously observed reduction in the LDL:HDL ratio in RA patients.
- Published
- 2020
- Full Text
- View/download PDF
13. Sulfotransferase (SULT) 1A1 polymorphism as a predisposition factor for lung cancer: a case-control analysis
- Author
-
Wang, Yunfei, Spitz, Margaret R., Tsou, Amy Meng-Hsuan, Zhang, Kerang, Makan, Nimisha, and Wu, Xifeng
- Published
- 2002
- Full Text
- View/download PDF
14. Phase 1 First-in-Human, Single- and Multiple-Ascending Dose, and Food Effect Studies to Assess the Safety, Tolerability, and Pharmacokinetics of Presatovir for the Treatment of Respiratory Syncytial Virus Infection
- Author
-
Anita Mathias, Yan Xin, Amy Meng, Jason W. Chien, Polina German, Sandra A Lewis, John Ling, Winnie Weng, and Richard L. Mackman
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,Food Effect Study ,030106 microbiology ,Respiratory disease ,medicine.disease ,Virus ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Pharmacokinetics ,Tolerability ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,Dosing ,Respiratory system ,business ,Respiratory tract - Abstract
Respiratory syncytial virus (RSV)-associated respiratory tract infection is a leading cause of hospitalizations in infants for which no effective treatment exists. RSV infection is also an important cause of respiratory disease in adults and immunocompromised patients. Presatovir (GS-5806) is an orally bioavailable antiviral agent that inhibits fusion of RSV with host cell membranes. Here, results from 2 phase 1 studies that evaluated safety, tolerability, and pharmacokinetics of presatovir in healthy adults following administration of single and multiple (7 days) once- or twice-daily ascending doses (first-in-human study) and in the presence or absence of food (food effect study) are described. Presatovir exhibited favorable safety and pharmacokinetic profiles that supported once-daily dosing. Presatovir exposure increased in an approximately dose-proportional manner across the evaluated dose range (single doses 25-300 mg; multiple doses 10-75 mg once daily for 7 days). Administration of presatovir with a high-fat meal did not alter exposure, supporting administration without regard to a meal in further clinical studies. These data were subsequently used to inform presatovir dosing regimens in a phase 2a challenge study of adults experimentally infected with RSV. Collectively, results from phase 1 evaluations and a phase 2a challenge study support further clinical investigation of presatovir for the treatment of RSV infection.
- Published
- 2017
15. Sulfotransferase (SULT) 1A1 polymorphism as a predisposition factor for lung cancer: a case-control analysis
- Author
-
Margaret R. Spitz, Amy Meng Hsuan Tsou, Nimisha Makan, Kerang Zhang, Yunfei Wang, and Xifeng Wu
- Subjects
Male ,Pulmonary and Respiratory Medicine ,Cancer Research ,Sulfotransferase ,medicine.medical_specialty ,Lung Neoplasms ,Single-nucleotide polymorphism ,Sulfotransferase 1A1 ,Polymerase Chain Reaction ,Sex Factors ,Risk Factors ,Internal medicine ,Genotype ,Odds Ratio ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Lung cancer ,Aged ,DNA Primers ,Genetics ,Polymorphism, Genetic ,business.industry ,Smoking ,Odds ratio ,Middle Aged ,medicine.disease ,Arylsulfotransferase ,Endocrinology ,Oncology ,Female ,Sulfotransferases ,Restriction fragment length polymorphism ,business - Abstract
SULT1A1 enzyme is a member of the sulfotransferase family that alters biological activities of numerous carcinogenic and mutagenic compounds through sulfation. A genetic polymorphism in the coding region of SULT1A1 gene has been associated with modulated enzyme activity. There is a G-->A nucleotide polymorphism in SULT1A1 gene that codes for an Arg-->His substitution, which results in decreased activity and thermal stability of the SULT1A1 enzyme. Utilizing a case-control study design, we hypothesized that the variant allele of the SULT1A1 gene may be associated with lung cancer risk. The PCR-RFLP assay was used to successfully genotype the SULT1A1*2 allele (variant A-allele) in 463 Caucasian lung cancer cases and 485 frequency matched Caucasian controls. There was an overall significant difference between cases and controls when adjusted by sex and smoking status (adjusted OR=1.41, 95% CI: 1.04-1.91). The adjusted OR was higher for females (OR=1.64, 95% CI: 1.06-2.56) than for males (OR=1.23, 95% CI: 0.80-1.88). Furthermore, the risk was significantly higher in current smokers (OR=1.74, 95% CI: 1.08-2.29) and heavy smokers (OR=1.45, 95% CI: 1.05-2.00). Our results support the hypothesis that a genetic polymorphism in the SULT1A1 gene may be associated with increased lung cancer risk.
- Published
- 2002
- Full Text
- View/download PDF
16. For the Dead
- Author
-
Amy Meng
- Subjects
General Medicine - Published
- 2016
- Full Text
- View/download PDF
17. Routine
- Author
-
Amy Meng
- Subjects
General Earth and Planetary Sciences ,General Environmental Science - Published
- 2016
- Full Text
- View/download PDF
18. Very Superstitious, and: Cohabitation
- Author
-
Amy Meng
- Subjects
Cohabitation ,General Earth and Planetary Sciences ,Demographic economics ,Sociology ,General Environmental Science - Published
- 2014
- Full Text
- View/download PDF
19. Novel targets and a cofactor for the Vibrio cholerae quorum sensing transcriptional regulator, HapR
- Author
-
Tsou, Amy Meng-Hsuan and Tsou, Amy Meng-Hsuan
- Abstract
Vibrio cholerae, which causes the severely dehydrating diarrheal disease, cholera, undergoes drastic changes in living conditions as it moves between its aquatic reservoir and its human host. These different living conditions require different gene expression profiles. One method that V. cholerae uses to sense changes in the environment, particularly changes in cell density, is quorum sensing. The bacteria produce small molecules called autoinducers that accumulate with increasing cell density and trigger intracellular signaling pathways to alter gene expression. These intracellular signaling pathways regulate the expression level of the transcriptional regulator, HapR, which controls many phenotypes including virulence factor production and biofilm formation. The studies presented here provide insight into the phenotypes that HapR regulates and the mechanism by which HapR provides this regulation. A bioinformatics-based approach was used to determine consensus binding sequences for HapR and to identify novel targets that are directly regulated by HapR. Many new HapR-regulated promoters were found, and some were activated while others were repressed by HapR. This study led us to investigate HapR's regulation of a hemolysin, HlyA, that had previously been shown to be a potential virulence factor. HapR was found to repress HlyA on two levels: one by directly binding to and repressing transcription from its promoter and another by inducing the transcription of a protease, HapA, which degrades the HlyA protein. HapR is known to repress V. cholerae's two main virulence factors, cholera toxin and the toxin-coregulated pilus, and so its regulation of HlyA represents another mechanism by which quorum sensing represses virulence. The third study identifies a two-component system, VarS/VarA, that is required for full HapR function. A previous report showed that VarS and VarA regulate HapR expression through the quorum sensing system, but we show that in addition to this, VarS
- Published
- 2010
20. Faces and Braces.
- Author
-
Lei, Amy Meng
- Subjects
ORTHODONTICS ,NONFICTION - Abstract
The article reviews the book" Faces and Braces," by William J. Clark.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.